1
|
Bakhos JJ, Saliba Y, Hajal J, Achkouty G, Oskaridjian H, Albuquerque M, Azevedo C, Semaan A, Suffee N, Balse E, Hatem SN, Fares N. Inhibiting atrial natriuretic peptide clearance reduces myocardial fibrosis and improves cardiac function in diabetic rats. EUROPEAN HEART JOURNAL OPEN 2025; 5:oeaf031. [PMID: 40201591 PMCID: PMC11977460 DOI: 10.1093/ehjopen/oeaf031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 04/10/2025]
Abstract
Aims Natriuretic peptides (NPs) exert pleiotropic effects through the recruitment of cyclic guanosine monophosphate (cGMP) signalling pathways depending on their bioavailability, which is regulated by clearance receptors and peptidases. Here, we tested the hypothesis that increasing myocardial bioavailability of NP has a beneficial effect on heart failure. We studied the effects of a mutated NP, M-atrial natriuretic peptide (MANP), resistant to neprilysin in a model of diabetic cardiomyopathy characterized by marked myocardial fibrosis. Methods and results Natriuretic peptides as well as sacubitril were delivered via osmotic mini-pumps to high-fat/streptozotocin-induced Type 2 diabetic (T2D) rats. Cardiac function was evaluated by echocardiography. Myocardial remodelling was studied by histological approaches, collagen phenotype, and atrial natriuretic peptide (ANP)/cGMP concentrations. Live-cell cGMP biosensing was conducted on cultured rat cardiac fibroblasts to investigate the biological effects of NP. Cyclic guanosine monophosphate signalling pathway was studied using multiple antibody arrays and biochemical assays in cardiac tissue and cultured fibroblasts. M-atrial natriuretic peptide exhibits superior efficacy than ANP in reducing left ventricular dysfunction and myocardial fibrosis with less extracellular matrix deposition. In vitro, MANP and ANP similarly generated cGMP and activated the protein kinase G (PKG) signalling pathway in cardiac fibroblasts, attenuating Mothers against decapentaplegic homolog 2 (SMAD) activation, collagen secretion, and cell proliferation. Nevertheless, in vivo, MANP specifically enhanced cardiac cGMP accumulation and was more potent than ANP in activating myocardial cGMP/PKG signalling and inhibiting the profibrotic SMAD, extracellular signal-regulated kinases 1/2, and nuclear factor of activated T cells 3 pathways. Endopeptidase inhibition using sacubitril also led to cardiac ANP/cGMP accumulation and reduced myocardial fibrosis. Conclusion Myocardial bioavailability of ANP is a major determinant of peptide efficacy in reducing cardiac fibrosis and improving pump function during diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Jules Joel Bakhos
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Youakim Saliba
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Joelle Hajal
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Guy Achkouty
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Hrag Oskaridjian
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Miguel Albuquerque
- INSERM, Centre de Recherche sur L'inflammation, UMR 1149, Université Paris-Cité, 45 Rue des Saints-Pères 75006 Paris, France
- Service d'Anatomie Pathologique, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, 100 Bd du Général Leclerc, 92110 Clichy, France
| | - Chloé Azevedo
- Institute of Cardiometabolism and Nutrition, IHU ICAN, Sorbonne University, INSERM UMR_S1166 Pitié-Salpêtrière Hospital, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - Albert Semaan
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| | - Nadine Suffee
- Institute of Cardiometabolism and Nutrition, IHU ICAN, Sorbonne University, INSERM UMR_S1166 Pitié-Salpêtrière Hospital, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - Elise Balse
- Institute of Cardiometabolism and Nutrition, IHU ICAN, Sorbonne University, INSERM UMR_S1166 Pitié-Salpêtrière Hospital, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - Stéphane N Hatem
- Institute of Cardiometabolism and Nutrition, IHU ICAN, Sorbonne University, INSERM UMR_S1166 Pitié-Salpêtrière Hospital, 47-83 boulevard de l'Hôpital, 75013 Paris, France
| | - Nassim Fares
- Laboratory of Research in Physiology and Pathophysiology, Faculty of Medicine, Saint-Joseph University of Beirut, 17-5208 - Mar Mikhaël, Beirut 1104 2020, Lebanon
| |
Collapse
|
2
|
Huang J, Shi Z, Huang Z, Lai S. Identification and Verification of Potential Markers Related to Myocardial Fibrosis by Bioinformatics Analysis. Biochem Genet 2024:10.1007/s10528-024-10937-9. [PMID: 39387979 DOI: 10.1007/s10528-024-10937-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
Mounting evidence indicates that myocardial fibrosis (MF) is frequently intertwined with immune and metabolic disorders. This comprehensive review aims to delve deeply into the crucial role of immune-related signature genes in the pathogenesis and progression of MF. This exploration holds significant importance as understanding the underlying mechanisms of MF is essential for developing effective diagnostic and therapeutic strategies. The dataset GSE9735 about myocardial fibrosis and non-fibrosis was downloaded from GEO database. Differentially expressed genes (DEGs) were identified by 'limma' package in R software. Then, the biological function of DEG was determined by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. XCell was used to estimate the composition pattern of matrix and immune cells. Protein-protein interaction (PPI) network was constructed based on STRING analysis software, and Hub genes were screened and functional modules were analyzed. The correlation between hub genes and immune cell subtypes was analyzed. Hub genes with |correlation coefficient|> 0.45 and p-value < 0.05 were used as characteristic biomarkers. Finally, the logistic regression model is used to verify the three markers in the training set and verification set (GSE97358 and GSE225336). A total of 635 DEGs were identified. Functional enrichment analysis shows that inflammation and immune response, extracellular matrix and structural remodeling play an important role in the pathological mechanism of MF. Immune cell infiltration analysis showed that immune cells (Plasma cells, Eosinophils, Chondrocytes and Th2 cells) significantly changed in MF pathological conditions. In PPI network analysis, IL1β, TTN, PTPRC, IGF1, ALDH1A1, CYP26A1, ALDH1A3, MYH11, CSF1R and CD80 were identified as hub genes, among which IL1β, CYP26A1 and GNG2 were regarded as immune-related characteristic markers. The AUC scores of the three biomarkers are all above 0.65, which proves that they have a good discrimination effect in MF. In this study, three immune-related genes were identified as diagnostic biomarkers of MF, which provided a new perspective for exploring the molecular mechanism of MF. This study takes a comprehensive approach to understanding the intricate relationship between myocardial fibrosis and immune metabolism. By identifying key immune-related biomarkers, this study not only reveals the molecular basis of myocardial fibrosis but also paves the way for the development of novel diagnostic tools and therapeutic strategies. These findings are critical for improving patient prognosis and may have broader implications for studying and treating other cardiovascular diseases associated with immune dysregulation.
Collapse
Affiliation(s)
- Jiazhuo Huang
- Department of Cardiology, The First People's Hospital of Zhaoqing City, No.9 Donggang East Road, Zhaoqing, 526040, Guangdong, China
| | - Zhentao Shi
- Department of Cardiology, The First People's Hospital of Zhaoqing City, No.9 Donggang East Road, Zhaoqing, 526040, Guangdong, China
| | - Zhifeng Huang
- Department of Cardiology, The First People's Hospital of Zhaoqing City, No.9 Donggang East Road, Zhaoqing, 526040, Guangdong, China
| | - Shaobin Lai
- Department of Cardiology, The First People's Hospital of Zhaoqing City, No.9 Donggang East Road, Zhaoqing, 526040, Guangdong, China.
| |
Collapse
|
3
|
Dabral S, Noh M, Werner F, Krebes L, Völker K, Maier C, Aleksic I, Novoyatleva T, Hadzic S, Schermuly RT, Perez VADJ, Kuhn M. C-type natriuretic peptide/cGMP/FoxO3 signaling attenuates hyperproliferation of pericytes from patients with pulmonary arterial hypertension. Commun Biol 2024; 7:693. [PMID: 38844781 PMCID: PMC11156916 DOI: 10.1038/s42003-024-06375-3] [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: 07/27/2023] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Pericyte dysfunction, with excessive migration, hyperproliferation, and differentiation into smooth muscle-like cells contributes to vascular remodeling in Pulmonary Arterial Hypertension (PAH). Augmented expression and action of growth factors trigger these pathological changes. Endogenous factors opposing such alterations are barely known. Here, we examine whether and how the endothelial hormone C-type natriuretic peptide (CNP), signaling through the cyclic guanosine monophosphate (cGMP) -producing guanylyl cyclase B (GC-B) receptor, attenuates the pericyte dysfunction observed in PAH. The results demonstrate that CNP/GC-B/cGMP signaling is preserved in lung pericytes from patients with PAH and prevents their growth factor-induced proliferation, migration, and transdifferentiation. The anti-proliferative effect of CNP is mediated by cGMP-dependent protein kinase I and inhibition of the Phosphoinositide 3-kinase (PI3K)/AKT pathway, ultimately leading to the nuclear stabilization and activation of the Forkhead Box O 3 (FoxO3) transcription factor. Augmentation of the CNP/GC-B/cGMP/FoxO3 signaling pathway might be a target for novel therapeutics in the field of PAH.
Collapse
Affiliation(s)
- Swati Dabral
- Institute of Physiology, University of Würzburg, Würzburg, Germany.
| | - Minhee Noh
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Franziska Werner
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Lisa Krebes
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Katharina Völker
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Christopher Maier
- Department of Thoracic and Cardiovascular Surgery, University hospital Würzburg, Würzburg, Germany
| | - Ivan Aleksic
- Department of Thoracic and Cardiovascular Surgery, University hospital Würzburg, Würzburg, Germany
| | - Tatyana Novoyatleva
- Justus-Liebig-University Giessen (JLU), Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Stefan Hadzic
- Justus-Liebig-University Giessen (JLU), Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Ralph Theo Schermuly
- Justus-Liebig-University Giessen (JLU), Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Vinicio A de Jesus Perez
- Divisions of Pulmonary and Critical Care Medicine and Stanford Cardiovascular Institute, Stanford University, California, USA
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
4
|
Giovou AE, Gladka MM, Christoffels VM. The Impact of Natriuretic Peptides on Heart Development, Homeostasis, and Disease. Cells 2024; 13:931. [PMID: 38891063 PMCID: PMC11172276 DOI: 10.3390/cells13110931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
During mammalian heart development, the clustered genes encoding peptide hormones, Natriuretic Peptide A (NPPA; ANP) and B (NPPB; BNP), are transcriptionally co-regulated and co-expressed predominately in the atrial and ventricular trabecular cardiomyocytes. After birth, expression of NPPA and a natural antisense transcript NPPA-AS1 becomes restricted to the atrial cardiomyocytes. Both NPPA and NPPB are induced by cardiac stress and serve as markers for cardiovascular dysfunction or injury. NPPB gene products are extensively used as diagnostic and prognostic biomarkers for various cardiovascular disorders. Membrane-localized guanylyl cyclase receptors on many cell types throughout the body mediate the signaling of the natriuretic peptide ligands through the generation of intracellular cGMP, which interacts with and modulates the activity of cGMP-activated kinase and other enzymes and ion channels. The natriuretic peptide system plays a fundamental role in cardio-renal homeostasis, and its potent diuretic and vasodilatory effects provide compensatory mechanisms in cardiac pathophysiological conditions and heart failure. In addition, both peptides, but also CNP, have important intracardiac actions during heart development and homeostasis independent of the systemic functions. Exploration of the intracardiac functions may provide new leads for the therapeutic utility of natriuretic peptide-mediated signaling in heart diseases and rhythm disorders. Here, we review recent insights into the regulation of expression and intracardiac functions of NPPA and NPPB during heart development, homeostasis, and disease.
Collapse
Affiliation(s)
| | | | - Vincent M. Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 1105AZ Amsterdam, The Netherlands; (A.E.G.); (M.M.G.)
| |
Collapse
|
5
|
Yao J, Chen Y, Huang Y, Sun X, Shi X. The role of cardiac microenvironment in cardiovascular diseases: implications for therapy. Hum Cell 2024; 37:607-624. [PMID: 38498133 DOI: 10.1007/s13577-024-01052-3] [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: 11/29/2023] [Accepted: 02/25/2024] [Indexed: 03/20/2024]
Abstract
Due to aging populations and changes in lifestyle, cardiovascular diseases including cardiomyopathy, hypertension, and atherosclerosis, are the leading causes of death worldwide. The heart is a complicated organ composed of multicellular types, including cardiomyocytes, fibroblasts, endothelial cells, vascular smooth muscle cells, and immune cells. Cellular specialization and complex interplay between different cell types are crucial for the cardiac tissue homeostasis and coordinated function of the heart. Mounting studies have demonstrated that dysfunctional cells and disordered cardiac microenvironment are closely associated with the pathogenesis of various cardiovascular diseases. In this paper, we discuss the composition and the homeostasis of cardiac tissues, and focus on the role of cardiac environment and underlying molecular mechanisms in various cardiovascular diseases. Besides, we elucidate the novel treatment for cardiovascular diseases, including stem cell therapy and targeted therapy. Clarification of these issues may provide novel insights into the prevention and potential targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiayu Yao
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuejun Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuqing Huang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| |
Collapse
|
6
|
Miyazawa K, Itoh Y, Fu H, Miyazono K. Receptor-activated transcription factors and beyond: multiple modes of Smad2/3-dependent transmission of TGF-β signaling. J Biol Chem 2024; 300:107256. [PMID: 38569937 PMCID: PMC11063908 DOI: 10.1016/j.jbc.2024.107256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Transforming growth factor β (TGF-β) is a pleiotropic cytokine that is widely distributed throughout the body. Its receptor proteins, TGF-β type I and type II receptors, are also ubiquitously expressed. Therefore, the regulation of various signaling outputs in a context-dependent manner is a critical issue in this field. Smad proteins were originally identified as signal-activated transcription factors similar to signal transducer and activator of transcription proteins. Smads are activated by serine phosphorylation mediated by intrinsic receptor dual specificity kinases of the TGF-β family, indicating that Smads are receptor-restricted effector molecules downstream of ligands of the TGF-β family. Smad proteins have other functions in addition to transcriptional regulation, including post-transcriptional regulation of micro-RNA processing, pre-mRNA splicing, and m6A methylation. Recent technical advances have identified a novel landscape of Smad-dependent signal transduction, including regulation of mitochondrial function without involving regulation of gene expression. Therefore, Smad proteins are receptor-activated transcription factors and also act as intracellular signaling modulators with multiple modes of function. In this review, we discuss the role of Smad proteins as receptor-activated transcription factors and beyond. We also describe the functional differences between Smad2 and Smad3, two receptor-activated Smad proteins downstream of TGF-β, activin, myostatin, growth and differentiation factor (GDF) 11, and Nodal.
Collapse
Affiliation(s)
- Keiji Miyazawa
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.
| | - Yuka Itoh
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Hao Fu
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kohei Miyazono
- Department of Applied Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Laboratory for Cancer Invasion and Metastasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
7
|
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
|
8
|
Zhao Y, Yuan X, Xie Y, Yin X, Liu Y, Sun Y, Gong Y, Liu J, Chen F. Association of Preablation Plasma Corin Levels With Atrial Fibrillation Recurrence After Catheter Ablation: A Prospective Observational Study. J Am Heart Assoc 2024; 13:e031928. [PMID: 38214265 PMCID: PMC10926783 DOI: 10.1161/jaha.123.031928] [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: 08/03/2023] [Accepted: 12/08/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND We assessed the impact of pre- and postprocedural plasma corin levels on the recurrence of atrial fibrillation (AF) after catheter ablation (CA). METHODS AND RESULTS This prospective, single-center, observational study included patients undergoing their first CA of AF. Corin was measured before and 1 day after CA. The primary end point was recurrent AF between 3 and 12 months after ablation. From April 2019 through May 2021, we analyzed 616 patients with AF (59.09% men) with a mean age of 62.86±9.42 years. Overall, 153 patients (24.84%) experienced recurrent AF. In the recurrence group, the pre- and postprocedure corin concentrations were 539.14 (329.24-702.08) and 607.37 (364.50-753.80) pg/mL, respectively, which were significantly higher than the nonrecurrence group's respective concentrations of 369.05 (186.36-489.28) and 489.12 (315.66-629.05) pg/mL (both P<0.0001). A multivariate Cox regression analysis with confounders found that elevated preablation corin levels were significantly associated with an increased risk of AF recurrence after CA. Receiver operating characteristic curve analysis identified that a preablation corin threshold of >494.85 pg/mL predicted AF recurrence at 1 year. An increase of 1 SD in corin concentrations before CA (264.94 pg/mL) increased the risk of recurrent AF by 54.3% after adjusting for confounding variables (hazard ratio, 1.465 [95% CI, 1.282-1.655]; P<0.0001). CONCLUSIONS Plasma corin levels at baseline is a valuable predictor of AF recurrence after CA, independent of established conventional risk factors. Risk stratification before ablation for AF may be useful in selecting treatment regimens for patients.
Collapse
Affiliation(s)
- Yichang Zhao
- Department of CardiologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xiaoyang Yuan
- Department of Laboratory MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yunpeng Xie
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xiaomeng Yin
- Department of CardiologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Ying Liu
- Department of CardiologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yuanjun Sun
- Department of CardiologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yue Gong
- Department of CardiologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Jinqiu Liu
- Department of CardiologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Feifei Chen
- Department of CardiologyThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
9
|
Mangmool S, Duangrat R, Parichatikanond W, Kurose H. New Therapeutics for Heart Failure: Focusing on cGMP Signaling. Int J Mol Sci 2023; 24:12866. [PMID: 37629047 PMCID: PMC10454066 DOI: 10.3390/ijms241612866] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/30/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Current drugs for treating heart failure (HF), for example, angiotensin II receptor blockers and β-blockers, possess specific target molecules involved in the regulation of the cardiac circulatory system. However, most clinically approved drugs are effective in the treatment of HF with reduced ejection fraction (HFrEF). Novel drug classes, including angiotensin receptor blocker/neprilysin inhibitor (ARNI), sodium-glucose co-transporter-2 (SGLT2) inhibitor, hyperpolarization-activated cyclic nucleotide-gated (HCN) channel blocker, soluble guanylyl cyclase (sGC) stimulator/activator, and cardiac myosin activator, have recently been introduced for HF intervention based on their proposed novel mechanisms. SGLT2 inhibitors have been shown to be effective not only for HFrEF but also for HF with preserved ejection fraction (HFpEF). In the myocardium, excess cyclic adenosine monophosphate (cAMP) stimulation has detrimental effects on HFrEF, whereas cyclic guanosine monophosphate (cGMP) signaling inhibits cAMP-mediated responses. Thus, molecules participating in cGMP signaling are promising targets of novel drugs for HF. In this review, we summarize molecular pathways of cGMP signaling and clinical trials of emerging drug classes targeting cGMP signaling in the treatment of HF.
Collapse
Affiliation(s)
- Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.M.); (R.D.)
| | - Ratchanee Duangrat
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.M.); (R.D.)
| | | | - Hitoshi Kurose
- Pharmacology for Life Sciences, Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8505, Japan
| |
Collapse
|
10
|
Liu Z, Zhang X, Wang Y, Tai Y, Yao X, Midgley AC. Emergent Peptides of the Antifibrotic Arsenal: Taking Aim at Myofibroblast Promoting Pathways. Biomolecules 2023; 13:1179. [PMID: 37627244 PMCID: PMC10452577 DOI: 10.3390/biom13081179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Myofibroblasts are the principal effector cells driving fibrosis, and their accumulation in tissues is a fundamental feature of fibrosis. Essential pathways have been identified as being central to promoting myofibroblast differentiation, revealing multiple targets for intervention. Compared with large proteins and antibodies, peptide-based therapies have transpired to serve as biocompatible and cost-effective solutions to exert biomimicry, agonistic, and antagonistic activities with a high degree of targeting specificity and selectivity. In this review, we summarize emergent antifibrotic peptides and their utilization for the targeted prevention of myofibroblasts. We then highlight recent studies on peptide inhibitors of upstream pathogenic processes that drive the formation of profibrotic cell phenotypes. We also briefly discuss peptides from non-mammalian origins that show promise as antifibrotic therapeutics. Finally, we discuss the future perspectives of peptide design and development in targeting myofibroblasts to mitigate fibrosis.
Collapse
Affiliation(s)
- Zhen Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xinyan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yanrong Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yifan Tai
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaolin Yao
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China
| | - Adam C. Midgley
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
11
|
Tao W, Yang X, Zhang Q, Bi S, Yao Z. Optimal treatment for post-MI heart failure in rats: dapagliflozin first, adding sacubitril-valsartan 2 weeks later. Front Cardiovasc Med 2023; 10:1181473. [PMID: 37383701 PMCID: PMC10296765 DOI: 10.3389/fcvm.2023.1181473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/19/2023] [Indexed: 06/30/2023] Open
Abstract
Background Based on previous research, both dapagliflozin (DAPA) and sacubitril-valsartan (S/V) improve the prognosis of patients with heart failure (HF). Our study aims to investigate whether the early initiation of DAPA or the combination of DAPA with S/V in different orders would exert a greater protective effect on heart function than that of S/V alone in post-myocardial infarction HF (post-MI HF). Methods Rats were randomized into six groups: (A) Sham; (B) MI; (C) MI + S/V (1st d); (D) MI + DAPA (1st d); (E) MI + S/V (1st d) + DAPA (14th d); (F) MI + DAPA (1st d) + S/V (14th d). The MI model was established in rats via surgical ligation of the left anterior descending coronary artery. Histology, Western blotting, RNA-seq, and other approaches were used to explore the optimal treatment to preserve the heart function in post-MI HF. A daily dose of 1 mg/kg DAPA and 68 mg/kg S/V was administered. Results The results of our study revealed that DAPA or S/V substantially improved the cardiac structure and function. DAPA and S/V monotherapy resulted in comparable reduction in infarct size, fibrosis, myocardium hypertrophy, and apoptosis. The administration of DAPA followed by S/V results in a superior improvement in heart function in rats with post-MI HF than those in other treatment groups. The administration of DAPA following S/V did not result in any additional improvement in heart function as compared to S/V monotherapy in rats with post-MI HF. Our findings further suggest that the combination of DAPA and S/V should not be administered within 3 days after acute myocardial infarction (AMI), as it resulted in a considerable increase in mortality. Our RNA-Seq data revealed that DAPA treatment after AMI altered the expression of genes related to myocardial mitochondrial biogenesis and oxidative phosphorylation. Conclusions Our study revealed no notable difference in the cardioprotective effects of singular DAPA or S/V in rats with post-MI HF. Based on our preclinical investigation, the most effective treatment strategy for post-MI HF is the administration of DAPA during the 2 weeks, followed by the addition of S/V to DAPA later. Conversely, adopting a therapeutic scheme whereby S/V was administered first, followed by later addition of DAPA, failed to further improve the cardiac function compared to S/V monotherapy.
Collapse
Affiliation(s)
- Wenqi Tao
- Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
| | - Xiaoyu Yang
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China
| | - Qing Zhang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuli Bi
- School of Medicine, Nankai University, Tianjin, China
| | - Zhuhua Yao
- Tianjin Union Medical Center, Tianjin Medical University, Tianjin, China
- Department of Cardiology, Tianjin Union Medical Center, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China
| |
Collapse
|
12
|
Suzuki J, Kaji K, Nishimura N, Kubo T, Tomooka F, Shibamoto A, Iwai S, Tsuji Y, Fujinaga Y, Kitagawa K, Namisaki T, Akahane T, Yoshiji H. A Combination of an Angiotensin II Receptor and a Neprilysin Inhibitor Attenuates Liver Fibrosis by Preventing Hepatic Stellate Cell Activation. Biomedicines 2023; 11:1295. [PMID: 37238965 PMCID: PMC10215948 DOI: 10.3390/biomedicines11051295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/12/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
The renin-angiotensin-aldosterone system has gained attention due to its role as a mediator of liver fibrosis and hepatic stellate cell (HSC) activation. Meanwhile, the natriuretic peptide (NP) system, including atrial NP (ANP) and C-type NP (CNP), is a counter-regulatory hormone regulated by neprilysin. Although the combination of an angiotensin receptor and a neprilysin inhibitor (sacubitril/valsartan: SAC/VAL) has shown clinical efficacy in patients with heart failure, its potential effects on hepatic fibrosis have not been clarified. This study assessed the effects of SAC/VAL in carbon tetrachloride (CCl4)-induced murine liver fibrosis as well as the in vitro phenotypes of HSCs. Treatment with SAC and VAL markedly attenuated CCl4-induced liver fibrosis while reducing α-SMA+-HSC expansion and decreasing hepatic hydroxyproline and mRNA levels of pro-fibrogenic markers. Treatment with SAC increased plasma ANP and CNP levels in CCl4-treated mice, and ANP effectively suppressed cell proliferation and TGF-β-stimulated MMP2 and TIMP2 expression in LX-2 cells by activating guanylate cyclase-A/cGMP/protein kinase G signaling. Meanwhile, CNP did not affect the pro-fibrogenic activity of LX-2 cells. Moreover, VAL directly inhibited angiotensin II (AT-II)-stimulated cell proliferation and the expression of TIMP1 and CTGF through the blockade of the AT-II type 1 receptor/protein kinase C pathway. Collectively, SAC/VAL may be a novel therapeutic treatment for liver fibrosis.
Collapse
Affiliation(s)
| | - Kosuke Kaji
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Nara, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Hung MJ, Yeh CT, Kounis NG, Koniari I, Hu P, Hung MY. Coronary Artery Spasm-Related Heart Failure Syndrome: Literature Review. Int J Mol Sci 2023; 24:ijms24087530. [PMID: 37108691 PMCID: PMC10145866 DOI: 10.3390/ijms24087530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Although heart failure (HF) is a clinical syndrome that becomes worse over time, certain cases can be reversed with appropriate treatments. While coronary artery spasm (CAS) is still underappreciated and may be misdiagnosed, ischemia due to coronary artery disease and CAS is becoming the single most frequent cause of HF worldwide. CAS could lead to syncope, HF, arrhythmias, and myocardial ischemic syndromes such as asymptomatic ischemia, rest and/or effort angina, myocardial infarction, and sudden death. Albeit the clinical significance of asymptomatic CAS has been undervalued, affected individuals compared with those with classic Heberden's angina pectoris are at higher risk of syncope, life-threatening arrhythmias, and sudden death. As a result, a prompt diagnosis implements appropriate treatment strategies, which have significant life-changing consequences to prevent CAS-related complications, such as HF. Although an accurate diagnosis depends mainly on coronary angiography and provocative testing, clinical characteristics may help decision-making. Because the majority of CAS-related HF (CASHF) patients present with less severe phenotypes than overt HF, it underscores the importance of understanding risk factors correlated with CAS to prevent the future burden of HF. This narrative literature review summarises and discusses separately the epidemiology, clinical features, pathophysiology, and management of patients with CASHF.
Collapse
Affiliation(s)
- Ming-Jui Hung
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital Keelung, Chang Gung University College of Medicine, Keelung City 24201, Taiwan
| | - Chi-Tai Yeh
- Department of Medical Research and Education, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Continuing Education Program of Food Biotechnology Applications, College of Science and Engineering, National Taitung University, Taitung 95092, Taiwan
| | - Nicholas G Kounis
- Department of Cardiology, University of Patras Medical School, 26221 Patras, Greece
| | - Ioanna Koniari
- Cardiology Department, Liverpool Heart and Chest Hospital, Liverpool L14 3PE, UK
| | - Patrick Hu
- Department of Internal Medicine, School of Medicine, University of California, Riverside, Riverside, CA 92521, USA
- Department of Cardiology, Riverside Medical Clinic, Riverside, CA 92506, USA
| | - Ming-Yow Hung
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, No.291, Zhongzheng Rd., Zhonghe District, New Taipei City 23561, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei City 110301, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, New Taipei City 23561, Taiwan
| |
Collapse
|
14
|
Li Y, Kubo H, Yu D, Yang Y, Johnson JP, Eaton DM, Berretta RM, Foster M, McKinsey TA, Yu J, Elrod JW, Chen X, Houser SR. Combining three independent pathological stressors induces a heart failure with preserved ejection fraction phenotype. Am J Physiol Heart Circ Physiol 2023; 324:H443-H460. [PMID: 36763506 PMCID: PMC9988529 DOI: 10.1152/ajpheart.00594.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/05/2023] [Accepted: 01/18/2023] [Indexed: 02/11/2023]
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is defined as HF with an ejection fraction (EF) ≥ 50% and elevated cardiac diastolic filling pressures. The underlying causes of HFpEF are multifactorial and not well-defined. A transgenic mouse with low levels of cardiomyocyte (CM)-specific inducible Cavβ2a expression (β2a-Tg mice) showed increased cytosolic CM Ca2+, and modest levels of CM hypertrophy, and fibrosis. This study aimed to determine if β2a-Tg mice develop an HFpEF phenotype when challenged with two additional stressors, high-fat diet (HFD) and Nω-nitro-l-arginine methyl ester (l-NAME, LN). Four-month-old wild-type (WT) and β2a-Tg mice were given either normal chow (WT-N, β2a-N) or HFD and/or l-NAME (WT-HFD, WT-LN, WT-HFD-LN, β2a-HFD, β2a-LN, and β2a-HFD-LN). Some animals were treated with the histone deacetylase (HDAC) (hypertrophy regulators) inhibitor suberoylanilide hydroxamic acid (SAHA) (β2a-HFD-LN-SAHA). Echocardiography was performed monthly. After 4 mo of treatment, terminal studies were performed including invasive hemodynamics and organs weight measurements. Cardiac tissue was collected. Four months of HFD plus l-NAME treatment did not induce a profound HFpEF phenotype in FVB WT mice. β2a-HFD-LN (3-Hit) mice developed features of HFpEF, including increased atrial natriuretic peptide (ANP) levels, preserved EF, diastolic dysfunction, robust CM hypertrophy, increased M2-macrophage population, and myocardial fibrosis. SAHA reduced the HFpEF phenotype in the 3-Hit mouse model, by attenuating these effects. The 3-Hit mouse model induced a reliable HFpEF phenotype with CM hypertrophy, cardiac fibrosis, and increased M2-macrophage population. This model could be used for identifying and preclinical testing of novel therapeutic strategies.NEW & NOTEWORTHY Our study shows that three independent pathological stressors (increased Ca2+ influx, high-fat diet, and l-NAME) together produce a profound HFpEF phenotype. The primary mechanisms include HDAC-dependent-CM hypertrophy, necrosis, increased M2-macrophage population, fibroblast activation, and myocardial fibrosis. A role for HDAC activation in the HFpEF phenotype was shown in studies with SAHA treatment, which prevented the severe HFpEF phenotype. This "3-Hit" mouse model could be helpful in identifying novel therapeutic strategies to treat HFpEF.
Collapse
Affiliation(s)
- Yijia Li
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Hajime Kubo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Daohai Yu
- Department of Biomedical Education and Data Science, Center for Biostatistics and Epidemiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Yijun Yang
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Jaslyn P Johnson
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Deborah M Eaton
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Remus M Berretta
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Michael Foster
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Timothy A McKinsey
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Consortium for Fibrosis Research and Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Jun Yu
- Department of Cardiovascular Sciences, Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, Pennsylvania, United States
| | - John W Elrod
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, Pennsylvania, United States
| | - Xiongwen Chen
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Steven R Houser
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
15
|
Sarzani R, Allevi M, Di Pentima C, Schiavi P, Spannella F, Giulietti F. Role of Cardiac Natriuretic Peptides in Heart Structure and Function. Int J Mol Sci 2022; 23:ijms232214415. [PMID: 36430893 PMCID: PMC9697447 DOI: 10.3390/ijms232214415] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Cardiac natriuretic peptides (NPs), atrial NP (ANP) and B-type NP (BNP) are true hormones produced and released by cardiomyocytes, exerting several systemic effects. Together with C-type NP (CNP), mainly expressed by endothelial cells, they also exert several paracrine and autocrine activities on the heart itself, contributing to cardiovascular (CV) health. In addition to their natriuretic, vasorelaxant, metabolic and antiproliferative systemic properties, NPs prevent cardiac hypertrophy, fibrosis, arrhythmias and cardiomyopathies, counteracting the development and progression of heart failure (HF). Moreover, recent studies revealed that a protein structurally similar to NPs mainly produced by skeletal muscles and osteoblasts called musclin/osteocrin is able to interact with the NPs clearance receptor, attenuating cardiac dysfunction and myocardial fibrosis and promoting heart protection during pathological overload. This narrative review is focused on the direct activities of this molecule family on the heart, reporting both experimental and human studies that are clinically relevant for physicians.
Collapse
Affiliation(s)
- Riccardo Sarzani
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
- Correspondence: (R.S.); Tel.: +39-071-5964696
| | - Massimiliano Allevi
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Chiara Di Pentima
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
| | - Paola Schiavi
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Francesco Spannella
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Federico Giulietti
- Internal Medicine and Geriatrics, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Nazionale di Ricovero e Cura per Anziani (IRCCS INRCA), 60127 Ancona, Italy
| |
Collapse
|
16
|
Jones CE, Sharick JT, Sizemore ST, Cukierman E, Strohecker AM, Leight JL. A miniaturized screening platform to identify novel regulators of extracellular matrix alignment. CANCER RESEARCH COMMUNICATIONS 2022; 2:1471-1486. [PMID: 36530465 PMCID: PMC9757767 DOI: 10.1158/2767-9764.crc-22-0157] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/03/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Extracellular matrix alignment contributes to metastasis in a number of cancers and is a known prognostic stromal factor; however, the mechanisms controlling matrix organization remain unclear. Cancer-associated fibroblasts (CAF) play a critical role in this process, particularly via matrix production and modulation of key signaling pathways controlling cell adhesion and contractility. Stroma normalization, as opposed to elimination, is a highly sought strategy, and screening for drugs that effectively alter extracellular matrix (ECM) alignment is a practical way to identify novel CAF-normalizing targets that modulate ECM organization. To meet this need, we developed a novel high-throughput screening platform in which fibroblast-derived matrices were produced in 384-well plates, imaged with automated confocal microscopy, and analyzed using a customized MATLAB script. This platform is a technical advance because it miniaturizes the assay, eliminates costly and time-consuming experimental steps, and streamlines data acquisition and analysis to enable high-throughput screening applications. As a proof of concept, this platform was used to screen a kinase inhibitor library to identify modulators of matrix alignment. A number of novel potential regulators were identified, including several receptor tyrosine kinases (c-MET, tropomyosin receptor kinase 1 (NTRK1), HER2/ERBB2) and the serine/threonine kinases protein kinase A, C, and G (PKA, PKC, and PKG). The expression of these regulators was analyzed in publicly available patient datasets to examine the association between stromal gene expression and patient outcomes.
Collapse
Affiliation(s)
- Caitlin E. Jones
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Joe T. Sharick
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
| | - Steven T. Sizemore
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Edna Cukierman
- Cancer Signaling and Epigenetics, The Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, Pennsylvania
| | - Anne Marie Strohecker
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Jennifer L. Leight
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
17
|
Genetic Disruption of Guanylyl Cyclase/Natriuretic Peptide Receptor-A Triggers Differential Cardiac Fibrosis and Disorders in Male and Female Mutant Mice: Role of TGF-β1/SMAD Signaling Pathway. Int J Mol Sci 2022; 23:ijms231911487. [PMID: 36232788 PMCID: PMC9569686 DOI: 10.3390/ijms231911487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 01/01/2023] Open
Abstract
The global targeted disruption of the natriuretic peptide receptor-A (NPRA) gene (Npr1) in mice provokes hypertension and cardiovascular dysfunction. The objective of this study was to determine the mechanisms regulating the development of cardiac fibrosis and dysfunction in Npr1 mutant mice. Npr1 knockout (Npr1-/-, 0-copy), heterozygous (Npr1+/-, 1-copy), and wild-type (Npr1+/+, 2-copy) mice were treated with the transforming growth factor (TGF)-β1 receptor (TGF-β1R) antagonist GW788388 (2 µg/g body weight/day; ip) for 28 days. Hearts were isolated and used for real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blot, and immunohistochemical analyses. The Npr1-/- (0-copy) mice showed a 6-fold induction of cardiac fibrosis and dysfunction with markedly induced expressions of collagen-1α (3.8-fold), monocyte chemoattractant protein (3.7-fold), connective tissue growth factor (CTGF, 5.3-fold), α-smooth muscle actin (α-SMA, 6.1-fold), TGF-βRI (4.3-fold), TGF-βRII (4.7-fold), and phosphorylated small mothers against decapentaplegic (pSMAD) proteins, including pSMAD-2 (3.2-fold) and pSMAD-3 (3.7-fold), compared with wild-type mice. The expressions of phosphorylated extracellular-regulated kinase ERK1/2 (pERK1/2), matrix metalloproteinases-2, -9, (MMP-2, -9), and proliferating cell nuclear antigen (PCNA) were also significantly upregulated in Npr1 0-copy mice. The treatment of mutant mice with GW788388 significantly blocked the expression of fibrotic markers, SMAD proteins, MMPs, and PCNA compared with the vehicle-treated control mice. The treatment with GW788388 significantly prevented cardiac dysfunctions in a sex-dependent manner in Npr1 0-copy and 1-copy mutant mice. The results suggest that the development of cardiac fibrosis and dysfunction in mutant mice is predominantly regulated through the TGF-β1-mediated SMAD-dependent pathway.
Collapse
|
18
|
Smad-dependent pathways in the infarcted and failing heart. Curr Opin Pharmacol 2022; 64:102207. [DOI: 10.1016/j.coph.2022.102207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/10/2022] [Accepted: 02/22/2022] [Indexed: 02/08/2023]
|
19
|
Abramicheva PA, Plotnikov EY. Hormonal Regulation of Renal Fibrosis. Life (Basel) 2022; 12:737. [PMID: 35629404 PMCID: PMC9143586 DOI: 10.3390/life12050737] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Fibrosis is a severe complication of many acute and chronic kidney pathologies. According to current concepts, an imbalance in the synthesis and degradation of the extracellular matrix by fibroblasts is considered the key cause of the induction and progression of fibrosis. Nevertheless, inflammation associated with the damage of tissue cells is among the factors promoting this pathological process. Most of the mechanisms accompanying fibrosis development are controlled by various hormones, which makes humoral regulation an attractive target for therapeutic intervention. In this vein, it is particularly interesting that the kidney is the source of many hormones, while other hormones regulate renal functions. The normal kidney physiology and pathogenesis of many kidney diseases are sex-dependent and thus modulated by sex hormones. Therefore, when choosing therapy, it is necessary to focus on the sex-associated characteristics of kidney functioning. In this review, we considered renal fibrosis from the point of view of vasoactive and reproductive hormone imbalance. The hormonal therapy possibilities for the treatment or prevention of kidney fibrosis are also discussed.
Collapse
Affiliation(s)
- Polina A. Abramicheva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Egor Y. Plotnikov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
- Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| |
Collapse
|
20
|
Phosphodiesterase-1 in the cardiovascular system. Cell Signal 2022; 92:110251. [DOI: 10.1016/j.cellsig.2022.110251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 11/18/2022]
|
21
|
Regional Diversities in Fibrogenesis Weighed as a Key Determinant for Atrial Arrhythmogenesis. Biomedicines 2021; 9:biomedicines9121900. [PMID: 34944715 PMCID: PMC8698388 DOI: 10.3390/biomedicines9121900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 11/18/2022] Open
Abstract
Atrial fibrosis plays a key role in atrial myopathy, resulting in the genesis of atrial fibrillation (AF). The abnormal distribution of fibrotic tissue, electrical coupling, paracrine interactions, and biomechanical–electrical interactions have all been suggested as causes of fibrosis-related arrhythmogenesis. Moreover, the regional difference in fibrogenesis, specifically the left atrium (LA) exhibiting a higher arrhythmogenesis and level of fibrosis than the right atrium (RA) in AF, is a key contributor to atrial arrhythmogenesis. LA fibroblasts have greater profibrotic cellular activities than RA fibroblasts, but knowledge about the regional diversity of atrial regional fibrogenesis remains limited. This article provides a comprehensive review of research findings on the association between fibrogenesis and arrhythmogenesis from laboratory to clinical evidence and updates the current understanding of the potential mechanism underlying the difference in fibrogenesis between the LA and RA.
Collapse
|
22
|
Wu M, Guo Y, Wu Y, Xu K, Lin L. Protective Effects of Sacubitril/Valsartan on Cardiac Fibrosis and Function in Rats With Experimental Myocardial Infarction Involves Inhibition of Collagen Synthesis by Myocardial Fibroblasts Through Downregulating TGF-β1/Smads Pathway. Front Pharmacol 2021; 12:696472. [PMID: 34135764 PMCID: PMC8201773 DOI: 10.3389/fphar.2021.696472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Objectives: To investigate the effect and mechanism of sacubitril/valsartan on myocardial fibrosis in rats following experimental myocardial infarction and in TGF-β1-treated myocardial fibroblasts. Methods: Male Sprague-Dawley (SD) rats were subjected to coronary artery ligation to establish myocardial infarction and intragastrically fed vehicle, valsartan (Val, 32 mg/kg, once-daily) or sacubitril/valsartan (Sac/Val, 68 mg/kg, once-daily) for 4 weeks. In parallel, myocardial fibroblasts (MFs) isolated from neonatal SD rats were exposed to hypoxia and treated with TGF-β1 (5 ng/ml) plus vehicle, Val (107–10–5 M) or Sac/Val (107–105 M). Rat cardiac function and fibrosis were measured by echocardiography and histological method, respectively. MFs viability and collagen synthesis were determined by cell counting kit-8 and enzyme-linked immunosorbent assay, respectively. Protein expressions of TGF-β1, Smad3, phosphorylated Smad3 (p-Smad3), and p-Smad3 subcellular localization were detected by immunoblotting and immunocytochemistry. Results: Sac/Val significantly improved cardiac structure and function in rats after myocardial infarction, including decreased left ventricular end-diastolic diameter and interventricular septal thickness, increased ejection fraction, and reduced myocardial collagen volume fraction and type Ⅰ and type Ⅲ collagen levels, and this effect was superior to that of Val. Besides, Sac/Val inhibited myocardial TGF-β1 and p-Smad3 protein expression better than Val. Mechanically, Sac/Val significantly attenuated TGF-β1-induced proliferation and collagen synthesis of MFs, and inhibit Smad3 phosphorylation and nucleus translocation, and this effect outperformed Val. Overexpression and silencing of Smad3 enhanced and reversed the inhibitory effects of Sac/Val on TGF-β1-induced collagen synthesis by MFs, respectively. Conclusions: Sacubitril/valsartan improves cardiac function and fibrosis in rats after experimental myocardial infarction, and this effect is related to the inhibition of collagen synthesis in myocardial fibroblasts by inhibiting the TGF/Smads signaling pathway.
Collapse
Affiliation(s)
- Meifang Wu
- Department of Cardiology, Affiliated Hospital of Putian University, Southern Medical University, Putian, China
| | - Yanguang Guo
- Department of Cardiology, Affiliated Hospital of Putian University, Southern Medical University, Putian, China
| | - Ying Wu
- Department of Cardiology, Affiliated Hospital of Putian University, Southern Medical University, Putian, China
| | - Kaizu Xu
- Department of Cardiology, Affiliated Hospital of Putian University, Southern Medical University, Putian, China
| | - Liming Lin
- Department of Cardiology, Affiliated Hospital of Putian University, Southern Medical University, Putian, China
| |
Collapse
|
23
|
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
|
24
|
McCarty MF. Nutraceutical, Dietary, and Lifestyle Options for Prevention and Treatment of Ventricular Hypertrophy and Heart Failure. Int J Mol Sci 2021; 22:ijms22073321. [PMID: 33805039 PMCID: PMC8037104 DOI: 10.3390/ijms22073321] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Although well documented drug therapies are available for the management of ventricular hypertrophy (VH) and heart failure (HF), most patients nonetheless experience a downhill course, and further therapeutic measures are needed. Nutraceutical, dietary, and lifestyle measures may have particular merit in this regard, as they are currently available, relatively safe and inexpensive, and can lend themselves to primary prevention as well. A consideration of the pathogenic mechanisms underlying the VH/HF syndrome suggests that measures which control oxidative and endoplasmic reticulum (ER) stress, that support effective nitric oxide and hydrogen sulfide bioactivity, that prevent a reduction in cardiomyocyte pH, and that boost the production of protective hormones, such as fibroblast growth factor 21 (FGF21), while suppressing fibroblast growth factor 23 (FGF23) and marinobufagenin, may have utility for preventing and controlling this syndrome. Agents considered in this essay include phycocyanobilin, N-acetylcysteine, lipoic acid, ferulic acid, zinc, selenium, ubiquinol, astaxanthin, melatonin, tauroursodeoxycholic acid, berberine, citrulline, high-dose folate, cocoa flavanols, hawthorn extract, dietary nitrate, high-dose biotin, soy isoflavones, taurine, carnitine, magnesium orotate, EPA-rich fish oil, glycine, and copper. The potential advantages of whole-food plant-based diets, moderation in salt intake, avoidance of phosphate additives, and regular exercise training and sauna sessions are also discussed. There should be considerable scope for the development of functional foods and supplements which make it more convenient and affordable for patients to consume complementary combinations of the agents discussed here. Research Strategy: Key word searching of PubMed was employed to locate the research papers whose findings are cited in this essay.
Collapse
Affiliation(s)
- Mark F McCarty
- Catalytic Longevity Foundation, 811 B Nahant Ct., San Diego, CA 92109, USA
| |
Collapse
|
25
|
Quah JX, Dharmaprani D, Tiver K, Lahiri A, Hecker T, Perry R, Selvanayagam JB, Joseph MX, McGavigan A, Ganesan A. Atrial fibrosis and substrate based characterization in atrial fibrillation: Time to move forwards. J Cardiovasc Electrophysiol 2021; 32:1147-1160. [PMID: 33682258 DOI: 10.1111/jce.14987] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/15/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022]
Abstract
Atrial fibrillation (AF) is the most commonly encountered cardiac arrhythmia in clinical practice. However, current therapeutic interventions for atrial fibrillation have limited clinical efficacy as a consequence of major knowledge gaps in the mechanisms sustaining atrial fibrillation. From a mechanistic perspective, there is increasing evidence that atrial fibrosis plays a central role in the maintenance and perpetuation of atrial fibrillation. Electrophysiologically, atrial fibrosis results in alterations in conduction velocity, cellular refractoriness, and produces conduction block promoting meandering, unstable wavelets and micro-reentrant circuits. Clinically, atrial fibrosis has also linked to poor clinical outcomes including AF-related thromboembolic complications and arrhythmia recurrences post catheter ablation. In this article, we review the pathophysiology behind the formation of fibrosis as AF progresses, the role of fibrosis in arrhythmogenesis, surrogate markers for detection of fibrosis using cardiac magnetic resonance imaging, echocardiography and electroanatomic mapping, along with their respective limitations. We then proceed to review the current evidence behind therapeutic interventions targeting atrial fibrosis, including drugs and substrate-based catheter ablation therapies followed by the potential future use of electro phenotyping for AF characterization to overcome the limitations of contemporary substrate-based methodologies.
Collapse
Affiliation(s)
- Jing X Quah
- College of Medicine and Public Health, Flinders University of South Australia, Adelaide, Australia.,Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, Australia
| | - Dhani Dharmaprani
- College of Medicine and Public Health, Flinders University of South Australia, Adelaide, Australia.,College of Science and Engineering, Flinders University of South Australia, Adelaide, Australia
| | - Kathryn Tiver
- Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, Australia
| | - Anandaroop Lahiri
- Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, Australia
| | - Teresa Hecker
- Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, Australia
| | - Rebecca Perry
- Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, Australia.,UniSA Allied Health and Human Performance, University of South Australia, Adelaide, Australia
| | | | - Majo X Joseph
- Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, Australia
| | | | - Anand Ganesan
- College of Medicine and Public Health, Flinders University of South Australia, Adelaide, Australia.,Department of Cardiovascular Medicine, Flinders Medical Centre, Adelaide, Australia
| |
Collapse
|
26
|
Aujla PK, Kassiri Z. Diverse origins and activation of fibroblasts in cardiac fibrosis. Cell Signal 2020; 78:109869. [PMID: 33278559 DOI: 10.1016/j.cellsig.2020.109869] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/21/2022]
Abstract
Cardiac fibroblasts (cFBs) have emerged as a heterogenous cell population. Fibroblasts are considered the main cell source for synthesis of the extracellular matrix (ECM) and as such a dysregulation in cFB function, activity, or viability can lead to disrupted ECM structure or fibrosis. Fibrosis can be initiated in response to different injuries and stimuli, and can be reparative (beneficial) or reactive (damaging). FBs need to be activated to myofibroblasts (MyoFBs) which have augmented capacity in synthesizing ECM proteins, causing fibrosis. In addition to the resident FBs in the myocardium, a number of other cells (pericytes, fibrocytes, mesenchymal, and hematopoietic cells) can transform into MyoFBs, further driving the fibrotic response. Multiple molecules including hormones, cytokines, and growth factors stimulate this process leading to generation of activated MyoFBs. Contribution of different cell types to cFBs and MyoFBs can result in an exponential increase in the number of MyoFBs and an accelerated pro-fibrotic response. Given the diversity of the cell sources, and the array of interconnected signalling pathways that lead to formation of MyoFBs and subsequently fibrosis, identifying a single target to limit the fibrotic response in the myocardium has been challenging. This review article will delineate the importance and relevance of fibroblast heterogeneity in mediating fibrosis in different models of heart failure and will highlight important signalling pathways implicated in myofibroblast activation.
Collapse
Affiliation(s)
- Preetinder K Aujla
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
27
|
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
|
28
|
Hanna A, Humeres C, Frangogiannis NG. The role of Smad signaling cascades in cardiac fibrosis. Cell Signal 2020; 77:109826. [PMID: 33160018 DOI: 10.1016/j.cellsig.2020.109826] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 12/30/2022]
Abstract
Most myocardial pathologic conditions are associated with cardiac fibrosis, the expansion of the cardiac interstitium through deposition of extracellular matrix (ECM) proteins. Although replacement fibrosis plays a reparative role after myocardial infarction, excessive, unrestrained or dysregulated myocardial ECM deposition is associated with ventricular dysfunction, dysrhythmias and adverse prognosis in patients with heart failure. The members of the Transforming Growth Factor (TGF)-β superfamily are critical regulators of cardiac repair, remodeling and fibrosis. TGF-βs are released and activated in injured tissues, bind to their receptors and transduce signals in part through activation of cascades involving a family of intracellular effectors the receptor-activated Smads (R-Smads). This review manuscript summarizes our knowledge on the role of Smad signaling cascades in cardiac fibrosis. Smad3, the best-characterized member of the family plays a critical role in activation of a myofibroblast phenotype, stimulation of ECM synthesis, integrin expression and secretion of proteases and anti-proteases. In vivo, fibroblast Smad3 signaling is critically involved in scar organization and exerts matrix-preserving actions. Although Smad2 also regulates fibroblast function in vitro, its in vivo role in rodent models of cardiac fibrosis seems more limited. Very limited information is available on the potential involvement of the Smad1/5/8 cascade in cardiac fibrosis. Dissection of the cellular actions of Smads in cardiac fibrosis, and identification of patient subsets with overactive or dysregulated myocardial Smad-dependent fibrogenic responses are critical for design of successful therapeutic strategies in patients with fibrosis-associated heart failure.
Collapse
Affiliation(s)
- Anis Hanna
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA
| | - Claudio Humeres
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
Camici PG, Tschöpe C, Di Carli MF, Rimoldi O, Van Linthout S. Coronary microvascular dysfunction in hypertrophy and heart failure. Cardiovasc Res 2020; 116:806-816. [PMID: 31999329 DOI: 10.1093/cvr/cvaa023] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/05/2019] [Accepted: 01/23/2020] [Indexed: 12/12/2022] Open
Abstract
Left ventricular (LV) hypertrophy (LVH) is a growth in left myocardial mass mainly caused by increased cardiomyocyte size. LVH can be a physiological adaptation to physical exercise or a pathological condition either primary, i.e. genetic, or secondary to LV overload. Patients with both primary and secondary LVH have evidence of coronary microvascular dysfunction (CMD). The latter is mainly due to capillary rarefaction and adverse remodelling of intramural coronary arterioles due to medial wall thickening with an increased wall/lumen ratio. An important feature of this phenomenon is the diffuse nature of this remodelling, which generally affects the coronary microvessels in the whole of the left ventricle. Patients with LVH secondary to arterial hypertension can develop both heart failure with preserved ejection fraction (HFpEF) and heart failure with reduced ejection fraction (HFrEF). These patients can develop HFrEF via a 'direct pathway' with an interval myocardial infarction and also in its absence. On the other hand, patients can develop HFpEF that can then progress to HFrEF with or without interval myocardial infarction. A similar evolution towards LV dysfunction and both HFpEF and HFrEF can occur in patients with hypertrophic cardiomyopathy, the most common genetic cardiomyopathy with a phenotype characterized by massive LVH. In this review article, we will discuss both the experimental and clinical studies explaining the mechanisms responsible for CMD in LVH as well as the evidence linking CMD with HFpEF and HFrEF.
Collapse
Affiliation(s)
- Paolo G Camici
- Vita Salute University and San Raffaele Hospital, Milano, Italy
| | - Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany.,Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Marcelo F Di Carli
- Cardiovascular Imaging Program, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ornella Rimoldi
- Vita Salute University and San Raffaele Hospital, Milano, Italy.,CNR IBFM, Segrate, Italy
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
32
|
MacLean J, Pasumarthi KBS. Characterization of primary adult mouse cardiac fibroblast cultures. Can J Physiol Pharmacol 2020; 98:861-869. [PMID: 32721222 DOI: 10.1139/cjpp-2020-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of cardiac fibroblasts (CFs) in disease states has been a focus of cardiovascular research over the past decade. Here, we briefly describe methods for isolation and characterization of CFs from adult mouse ventricles. Primary cultures were stained using antibodies for several marker proteins such as α-smooth muscle actin (αSMA), vimentin, and discoidin domain receptor 2 (DDR2) to confirm the identity of CFs or cardiac myofibroblasts (CMFs). Most cells in primary cultures consisted of CFs, with very low frequencies of endothelial cells, cardiomyocytes, and smooth muscle cells. We compared marker expression between cultures that were not passaged (P0) or passaged for few times (P1-3). When compared with P1-3 cultures, P0 cultures consistently displayed a lower percentage of cells positive for αSMA and DDR2, whereas vimentin expression was significantly higher in P0 cultures compared with P1-3 cultures. P0 cells were also smaller in area than P1-3 cells. Further, P1-3 mouse CFs were found to express both β1 and β2 adrenergic receptors (ARs) and β1ARs were more readily detected on the cell surface compared with β2ARs. In summary, mouse CF cultures underwent phenotype conversion into CMFs after passaging, consistent with what is seen with CF cultures from other species.
Collapse
Affiliation(s)
- Jessica MacLean
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada.,Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Kishore B S Pasumarthi
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada.,Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
33
|
Du Y, Xiao H, Wan J, Wang X, Li T, Zheng S, Feng J, Ye Q, Li J, Li G, Fan Z. Atorvastatin attenuates TGF‑β1‑induced fibrogenesis by inhibiting Smad3 and MAPK signaling in human ventricular fibroblasts. Int J Mol Med 2020; 46:633-640. [PMID: 32468059 PMCID: PMC7307817 DOI: 10.3892/ijmm.2020.4607] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 05/06/2020] [Indexed: 02/06/2023] Open
Abstract
Excessive proliferation and myofibroblasts transformation of cardiac fibroblasts play a critical role in the process of cardiac fibrosis. Atorvastatin (ATV), a 3‑hydroxy‑3‑methyl‑glutaryl‑coenzyme A reductase inhibitor, is commonly used to treat hypercholesterolemia. It has previously been shown that ATV has potential anti‑fibrotic effects. However, the underlying mechanisms of ATV against cardiac fibrosis remain to be fully elucidated, and to the best of our knowledge, there are no reports focusing on the effects of ATV on transforming growth factor‑β1 (TGF‑β1)‑induced human ventricular fibroblasts (hVFs) activation. In the present study, hVFs were stimulated with TGF‑β1 with or without pretreatment with ATV. Subsequently, hVF proliferation, cytotoxicity, myofibroblast differentiation and pro‑fibrotic gene expression were assessed. Canonical and non‑canonical signaling downstream of TGF‑β1, such as Smad3 and mitogen‑activated protein kinase (MAPK) signaling, were investigated by evaluating the phosphorylation levels of Smad3, extracellular signal‑regulated kinase 1/2, p38 MAPK and c‑Jun N‑terminal kinase. The results indicated that ATV significantly prevented TGF‑β1‑induced cell proliferation, myofibroblast differentiation and production of extracellular matrix proteins, such as matrix metalloproteinase‑2, collagen I and collagen III, in hVFs. Furthermore, ATV effectively inhibited TGF‑β1‑induced activation of Smad3 and MAPK signaling in hVFs. In conclusion, the present results demonstrated that ATV prevented TGF‑β1‑induced fibrogenesis in hVFs, at least in part by inhibiting the Smad3 and MAPK signaling pathways. Therefore, these results imply that ATV may be a promising agent to treat myocardial fibrosis.
Collapse
Affiliation(s)
- Yanfei Du
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Haiying Xiao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jun Wan
- Department of Basic Medical Sciences, College of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xinyu Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Shuzhan Zheng
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jian Feng
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Qiang Ye
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jiafu Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Guang Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Zhongcai Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
34
|
Tarbit E, Singh I, Peart JN, Rose'Meyer RB. Biomarkers for the identification of cardiac fibroblast and myofibroblast cells. Heart Fail Rev 2020; 24:1-15. [PMID: 29987445 DOI: 10.1007/s10741-018-9720-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Experimental research has recognized the importance of cardiac fibroblast and myofibroblast cells in heart repair and function. In a normal healthy heart, the cardiac fibroblast plays a central role in the structural, electrical, and chemical aspects within the heart. Interestingly, the transformation of cardiac fibroblast cells to cardiac myofibroblast cells is suspected to play a vital part in the development of heart failure. The ability to differentiate between the two cells types has been a challenge. Myofibroblast cells are only expressed in the stressed or failing heart, so a better understanding of cell function may identify therapies that aid repair of the damaged heart. This paper will provide an outline of what is currently known about cardiac fibroblasts and myofibroblasts, the physiological and pathological roles within the heart, and causes for the transition of fibroblasts into myoblasts. We also reviewed the potential markers available for characterizing these cells and found that there is no single-cell specific marker that delineates fibroblast or myofibroblast cells. To characterize the cells of fibroblast origin, vimentin is commonly used. Cardiac fibroblasts can be identified using discoidin domain receptor 2 (DDR2) while α-smooth muscle actin is used to distinguish myofibroblasts. A known cytokine TGF-β1 is well established to cause the transformation of cardiac fibroblasts to myofibroblasts. This review will also discuss clinical treatments that inhibit or reduce the actions of TGF-β1 and its contribution to cardiac fibrosis and heart failure.
Collapse
Affiliation(s)
- Emiri Tarbit
- School of Medical Sciences, Griffith University, Griffith, QLD, 4222, Australia
| | - Indu Singh
- School of Medical Sciences, Griffith University, Griffith, QLD, 4222, Australia
| | - Jason N Peart
- School of Medical Sciences, Griffith University, Griffith, QLD, 4222, Australia
| | - Roselyn B Rose'Meyer
- School of Medical Sciences, Griffith University, Griffith, QLD, 4222, Australia.
| |
Collapse
|
35
|
Abstract
In eukaryotic cells, about one-third of the synthesized proteins are translocated into the endoplasmic reticulum; they are membrane or lumen resident proteins and proteins direct to the Golgi apparatus. The co-translational translocation takes place through the heterotrimeric protein-conducting channel Sec61 which is associated with the ribosome and many accessory components, such as the heterotetrameric translocon-associated protein (TRAP) complex. Recently, microscopic techniques, such as cryo-electron microscopy and cryo-electron tomography, have enabled the determination of the translocation machinery structure. However, at present, there is a lack of understanding regarding the roles of some of its components; indeed, the TRAP complex function during co-translational translocation needs to be established. In addition, TRAP may play a role during unfolded protein response, endoplasmic-reticulum-associated protein degradation and congenital disorder of glycosylation (ssr4 CDG). In this article, I describe the current understanding of the TRAP complex in the light of its possible function(s).
Collapse
Affiliation(s)
- Antonietta Russo
- Medical Biochemistry and Molecular Biology, UKS, University of Saarland, Homburg, Germany
| |
Collapse
|
36
|
Suffee N, Moore-Morris T, Jagla B, Mougenot N, Dilanian G, Berthet M, Proukhnitzky J, Le Prince P, Tregouet DA, Pucéat M, Hatem SN. Reactivation of the Epicardium at the Origin of Myocardial Fibro-Fatty Infiltration During the Atrial Cardiomyopathy. Circ Res 2020; 126:1330-1342. [PMID: 32175811 DOI: 10.1161/circresaha.119.316251] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
RATIONALE Fibro-fatty infiltration of subepicardial layers of the atrial wall has been shown to contribute to the substrate of atrial fibrillation. OBJECTIVE Here, we examined if the epicardium that contains multipotent cells is involved in this remodeling process. METHODS AND RESULTS One hundred nine human surgical right atrial specimens were evaluated. There was a relatively greater extent of epicardial thickening and dense fibro-fatty infiltrates in atrial tissue sections from patients aged over 70 years who had mitral valve disease or atrial fibrillation when compared with patients aged less than 70 years with ischemic cardiomyopathy as indicated using logistic regression adjusted for age and gender. Cells coexpressing markers of epicardial progenitors and fibroblasts were detected in fibro-fatty infiltrates. Such epicardial remodeling was reproduced in an experimental model of atrial cardiomyopathy in rat and in Wilms tumor 1 (WT1)CreERT2/+;ROSA-tdT+/- mice. In the latter, genetic lineage tracing demonstrated the epicardial origin of fibroblasts within fibro-fatty infiltrates. A subpopulation of human adult epicardial-derived cells expressing PDGFR (platelet-derived growth factor receptor)-α were isolated and differentiated into myofibroblasts in the presence of Ang II (angiotensin II). Furthermore, single-cell RNA-sequencing analysis identified several clusters of adult epicardial-derived cells and revealed their specification from adipogenic to fibrogenic cells in the rat model of atrial cardiomyopathy. CONCLUSIONS Epicardium is reactivated during the formation of the atrial cardiomyopathy. Subsets of adult epicardial-derived cells, preprogrammed towards a specific cell fate, contribute to fibro-fatty infiltration of subepicardium of diseased atria. Our study reveals the biological basis for chronic atrial myocardial remodeling that paves the way of atrial fibrillation.
Collapse
Affiliation(s)
- Nadine Suffee
- From the INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France (N.S., G.D., M.B., J.P., D.A.T.)
| | - Thomas Moore-Morris
- INSERM U 1251, Aix-Marseille University, MMG, France (T.M.-M., M.P.).,IGF, University Montpellier, CNRS, INSERM, Montpellier, France (T.M.-M.)
| | - Bernd Jagla
- Pasteur Institute UtechS CB & Hub de Bioinformatique et Biostatistiques, C3BI, Paris (B.J.)
| | - Nathalie Mougenot
- Sorbonne Universités, INSERM UMR_S28, Faculté de médecine UPMC, Paris, France (N.M.)
| | - Gilles Dilanian
- From the INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France (N.S., G.D., M.B., J.P., D.A.T.)
| | - Myriam Berthet
- From the INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France (N.S., G.D., M.B., J.P., D.A.T.)
| | - Julie Proukhnitzky
- From the INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France (N.S., G.D., M.B., J.P., D.A.T.)
| | - Pascal Le Prince
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Institut de Cardiologie, Hôpital Pitié-Salpêtrière, Paris, France (P.L.P., S.N.H.)
| | - David A Tregouet
- From the INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France (N.S., G.D., M.B., J.P., D.A.T.)
| | - Michel Pucéat
- INSERM U 1251, Aix-Marseille University, MMG, France (T.M.-M., M.P.)
| | - Stéphane N Hatem
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Institut de Cardiologie, Hôpital Pitié-Salpêtrière, Paris, France (P.L.P., S.N.H.)
| |
Collapse
|
37
|
Rahmutula D, Zhang H, Wilson EE, Olgin JE. Absence of natriuretic peptide clearance receptor attenuates TGF-β1-induced selective atrial fibrosis and atrial fibrillation. Cardiovasc Res 2020; 115:357-372. [PMID: 30239604 DOI: 10.1093/cvr/cvy224] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 09/14/2018] [Indexed: 01/04/2023] Open
Abstract
Aims TGF-β1 plays an important role in atrial fibrosis and atrial fibrillation (AF); previous studies have shown that the atria are more susceptible to TGF-β1 mediated fibrosis than the ventricles. Natriuretic peptides (NPs) play an important role in cardiac remodelling and fibrosis, but the role of natriuretic peptide clearance (NPR-C) receptor is largely unknown. We investigated the role of NPR-C in modulating TGF-β1 signalling in the atria. Methods and results MHC-TGF-β1 transgenic (TGF-β1-Tx) mice, which develop isolated atrial fibrosis and AF, were cross-bred with NPR-C knock-out mice (NPR-C-KO). Transverse aortic constriction (TAC) was performed in wild type (Wt) and NPR-C knockout mice to study. Atrial fibrosis and AF inducibility in a pathophysiologic model. Electrophysiology, molecular, and histologic studies were performed in adult mice. siRNA was used to interrogate the interaction between TGF-β1 and NP signalling pathways in isolated atrial and ventricular fibroblasts/myofibroblasts. NPR-C expression level was 17 ± 5.8-fold higher in the atria compared with the ventricle in Wt mice (P = 0.009). Cross-bred mice demonstrated markedly decreased pSmad2 and collagen expression, atrial fibrosis, and AF compared with TGF-β1-Tx mice with intact NPR-C. There was a marked reduction in atrial fibrosis gene expression and AF inducibility in the NPR-C-KO-TAC mice compared with Wt-TAC. In isolated fibroblasts, knockdown of NPR-C resulted in a marked reduction of pSmad2 (56 ± 4% and 24 ± 14% reduction in atrial and ventricular fibroblasts, respectively) and collagen (76 ± 15% and 35 ± 23% reduction in atrial and ventricular fibroblasts/myofibroblasts, respectively) in response to TGF-β1 stimulation. This effect was reversed by simultaneously knocking down NPR-A but not with simultaneous knock down of PKG-1. Conclusion The differential response to TGF-β1 stimulated fibrosis between the atria and ventricle are in part mediated by the abundance of NPR-C receptors in the atria.
Collapse
Affiliation(s)
- Dolkun Rahmutula
- Division of Cardiology, Cardiovascular Research Institute, University of California, San Francisco, 505 Parnassus Avenue, M1182, Box 0124, San Francisco, CA, USA
| | - Hao Zhang
- Division of Cardiology, Cardiovascular Research Institute, University of California, San Francisco, 505 Parnassus Avenue, M1182, Box 0124, San Francisco, CA, USA
| | - Emily E Wilson
- Division of Cardiology, Cardiovascular Research Institute, University of California, San Francisco, 505 Parnassus Avenue, M1182, Box 0124, San Francisco, CA, USA
| | - Jeffrey E Olgin
- Division of Cardiology, Cardiovascular Research Institute, University of California, San Francisco, 505 Parnassus Avenue, M1182, Box 0124, San Francisco, CA, USA
| |
Collapse
|
38
|
ANP/NPRA Inhibits Epithelial-Mesenchymal Transition of Airway by Targeting Smad3 in Asthma. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-018-09804-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
39
|
Maslov MY, Foianini S, Mayer D, Orlov MV, Lovich MA. Interaction Between Sacubitril and Valsartan in Preventing Heart Failure Induced by Aortic Valve Insufficiency in Rats. J Card Fail 2019; 25:921-931. [PMID: 31539619 DOI: 10.1016/j.cardfail.2019.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/03/2019] [Accepted: 09/12/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND Synergistic interactions between neprilysin inhibition (NEPi) with sacubitril and angiotensin receptor type1 blockade (ARB) with valsartan have been implicated in improvement of left ventricular (LV) contractility, relaxation, exercise tolerance, and fibrosis in preexisting heart failure (HF) induced by aortic valve insufficiency (AVI). It is not known whether this pharmacologic synergy can prevent cardiovascular pathology in a similar AVI model. Our aim was to investigate the pharmacology of sacubitril/valsartan in an experimental setting with therapy beginning immediately after creation of AVI. METHODS HF was induced through partial disruption of the aortic valve in rats. Therapy began 3 hours after valve disruption and lasted 8 weeks. Sacubitril/valsartan (68 mg/kg), valsartan (31 mg/kg), sacubitril (31 mg/kg), or vehicle were administered daily via oral gavage (N=8 in each group). Hemodynamic assessments were conducted using Millar technology, and an exercise tolerance test was conducted using a rodent treadmill. RESULTS Only sacubitril/valsartan increased total arterial compliance and ejection fraction (EF). Therapies with sacubitril/valsartan and valsartan similarly improved load-dependent (dP/dtmax) and load independent indices (Ees) of LV contractility, and exercise tolerance, whereas sacubitril did not. None of the therapies improved LV relaxation (dP/dtmin), whereas all reduced myocardial fibrosis. CONCLUSIONS 1) The synergistic interaction between NEPi and ARB in early therapy with sacubitril/valsartan leads to increased total arterial compliance and EF. 2) Improvement in indices of LV contractility, and exercise tolerance with sacubitril/valsartan is likely because of ARB effect of valsartan. 3) All three therapies provided antifibrotic effects, suggesting both ARB and NEPi are capable of reducing myocardial fibrosis.
Collapse
Affiliation(s)
- Mikhail Y Maslov
- Steward St. Elizabeth's Medical Center/Tufts University School of Medicine, Department of Anesthesiology, Pain Medicine and Critical Care, Boston, Massachusetts.
| | - Stephan Foianini
- Steward St. Elizabeth's Medical Center/Tufts University School of Medicine, Department of Anesthesiology, Pain Medicine and Critical Care, Boston, Massachusetts
| | - Dita Mayer
- Steward St. Elizabeth's Medical Center/Tufts University School of Medicine, Department of Anesthesiology, Pain Medicine and Critical Care, Boston, Massachusetts
| | - Michael V Orlov
- Steward St. Elizabeth's Medical Center/Tufts University School of Medicine, Department of Cardiology, Boston, Massachusetts
| | - Mark A Lovich
- Steward St. Elizabeth's Medical Center/Tufts University School of Medicine, Department of Anesthesiology, Pain Medicine and Critical Care, Boston, Massachusetts
| |
Collapse
|
40
|
Cruz D, Pinto R, Freitas-Silva M, Nunes JP, Medeiros R. GWAS contribution to atrial fibrillation and atrial fibrillation-related stroke: pathophysiological implications. Pharmacogenomics 2019; 20:765-780. [PMID: 31368859 DOI: 10.2217/pgs-2019-0054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Atrial fibrillation (AF) and stroke are included in a group of complex traits that have been approached regarding of their study by susceptibility genetic determinants. Since 2007, several genome-wide association studies (GWAS) aiming to identify genetic variants modulating AF risk have been conducted. Thus, 11 GWAS have identified 26 SNPs (p < 5 × 10-2), of which 19 reached genome-wide significance (p < 5 × 10-8). From those variants, seven were also associated with cardioembolic stroke and three reached genome-wide significance in stroke GWAS. These associations may shed a light on putative shared etiologic mechanisms between AF and cardioembolic stroke. Additionally, some of these identified variants have been incorporated in genetic risk scores in order to elucidate new approaches of stroke prediction, prevention and treatment.
Collapse
Affiliation(s)
- Diana Cruz
- Molecular Oncology & Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edifício Laboratórios. 4° piso, Rua Dr António Bernardino de Almeida, 4200-4072 Porto, Portugal.,FMUP, Faculty of Medicine, Porto University, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ricardo Pinto
- Molecular Oncology & Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edifício Laboratórios. 4° piso, Rua Dr António Bernardino de Almeida, 4200-4072 Porto, Portugal
| | - Margarida Freitas-Silva
- FMUP, Faculty of Medicine, Porto University, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal.,Department of Medicine, Centro Hospitalar São João, Porto, Portugal
| | - José Pedro Nunes
- FMUP, Faculty of Medicine, Porto University, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal.,Department of Medicine, Centro Hospitalar São João, Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology & Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edifício Laboratórios. 4° piso, Rua Dr António Bernardino de Almeida, 4200-4072 Porto, Portugal.,FMUP, Faculty of Medicine, Porto University, Alameda Prof Hernâni Monteiro, 4200-319 Porto, Portugal.,Research Department, Portuguese League Against Cancer (NRNorte), Estrada Interior da Circunvalação, 6657, 4200-172 Porto, Portugal.,CEBIMED, Faculty of Health Sciences, Fernando Pessoa University, Praça 9 de Abril, 349, 4249-004 Porto, Portugal
| |
Collapse
|
41
|
Gallo G, Presta V, Volpe M, Rubattu S. Molecular and clinical implications of natriuretic peptides in aortic valve stenosis. J Mol Cell Cardiol 2019; 129:266-271. [DOI: 10.1016/j.yjmcc.2019.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/06/2019] [Accepted: 03/11/2019] [Indexed: 11/16/2022]
|
42
|
Crea F, Bairey Merz CN, Beltrame JF, Kaski JC, Ogawa H, Ong P, Sechtem U, Shimokawa H, Camici PG. The parallel tales of microvascular angina and heart failure with preserved ejection fraction: a paradigm shift. Eur Heart J 2019; 38:473-477. [PMID: 27907892 DOI: 10.1093/eurheartj/ehw461] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/26/2016] [Indexed: 01/09/2023] Open
Affiliation(s)
- Filippo Crea
- Institute of Cardiology, Catholic University of the Sacred Heart, Rome, Italy
| | - C Noel Bairey Merz
- Barbra Streisand Women's Heart Center, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - John F Beltrame
- The Queen Elizabeth Hospital Discipline of Medicine, University of Adelaide, Central Adelaide Local Health Network, Adelaide, South Australia, Australia
| | - Juan Carlos Kaski
- Cardiovascular and Cell Sciences Research Institute, St George's, University of London, UK
| | - Hisao Ogawa
- Faculty of Life Science, Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Peter Ong
- Department of Cardiology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Udo Sechtem
- Department of Cardiology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Paolo G Camici
- Vita Salute University and San Raffaele Hospital, Milan, Italy
| | | |
Collapse
|
43
|
Childers RC, Sunyecz I, West TA, Cismowski MJ, Lucchesi PA, Gooch KJ. Role of the cytoskeleton in the development of a hypofibrotic cardiac fibroblast phenotype in volume overload heart failure. Am J Physiol Heart Circ Physiol 2018; 316:H596-H608. [PMID: 30575422 DOI: 10.1152/ajpheart.00095.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hemodynamic load regulates cardiac remodeling. In contrast to pressure overload (increased afterload), hearts subjected to volume overload (VO; preload) undergo a distinct pattern of eccentric remodeling, chamber dilation, and decreased extracellular matrix content. Critical profibrotic roles of cardiac fibroblasts (CFs) in postinfarct remodeling and in response to pressure overload have been well established. Little is known about the CF phenotype in response to VO. The present study characterized the phenotype of primary cultures of CFs isolated from hearts subjected to 4 wk of VO induced by an aortocaval fistula. Compared with CFs isolated from sham hearts, VO CFs displayed a "hypofibrotic" phenotype, characterized by a ~50% decrease in the profibrotic phenotypic markers α-smooth muscle actin, connective tissue growth factor, and collagen type I, despite increased levels of profibrotic transforming growth factor-β1 and an intact canonical transforming growth factor-β signaling pathway. Actin filament dynamics were characterized, which regulate the CF phenotype in response to biomechanical signals. Actin polymerization was determined by the relative amounts of G-actin monomers versus F-actin. Compared with sham CFs, VO CFs displayed ~78% less F-actin and an increased G-actin-to-F-actin ratio (G/F ratio). In sham CFs, treatment with the Rho kinase inhibitor Y-27632 to increase the G/F ratio resulted in recapitulation of the hypofibrotic CF phenotype observed in VO CFs. Conversely, treatment of VO CFs with jasplakinolide to decrease the G/F ratio restored a more profibrotic response (>2.5-fold increase in α-smooth muscle actin, connective tissue growth factor, and collagen type I). NEW & NOTEWORTHY The present study is the first to describe a "hypofibrotic" phenotype of cardiac fibroblasts isolated from a volume overload model. Our results suggest that biomechanical regulation of actin microfilament stability and assembly is a critical mediator of cardiac fibroblast phenotypic modulation.
Collapse
Affiliation(s)
- Rachel C Childers
- Department of Biomedical Engineering, The Ohio State University , Columbus, Ohio.,The Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio.,The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University , Columbus, Ohio
| | - Ian Sunyecz
- Department of Biomedical Engineering, The Ohio State University , Columbus, Ohio.,The Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - T Aaron West
- The Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - Mary J Cismowski
- The Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - Pamela A Lucchesi
- The Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio.,Department of Pediatrics, The Ohio State University , Columbus, Ohio
| | - Keith J Gooch
- Department of Biomedical Engineering, The Ohio State University , Columbus, Ohio.,The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University , Columbus, Ohio
| |
Collapse
|
44
|
Maslov MY, Foianini S, Mayer D, Orlov MV, Lovich MA. Synergy between sacubitril and valsartan leads to hemodynamic, antifibrotic, and exercise tolerance benefits in rats with preexisting heart failure. Am J Physiol Heart Circ Physiol 2018; 316:H289-H297. [PMID: 30461302 DOI: 10.1152/ajpheart.00579.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Simultaneous neprilysin inhibition (NEPi) and angiotensin receptor blockade (ARB) with sacubitril/valsartan improves cardiac function and exercise tolerance in patients with heart failure. However, it is not known whether these therapeutic benefits are primarily due to NEPi with sacubitril or ARB with valsartan or their combination. Therefore, the aim of the present study was to investigate the potential contribution of sacubitril and valsartan to the benefits of the combination therapy on left ventricular (LV) function and exercise tolerance. Heart failure was induced by volume overload via partial disruption of the aortic valve in rats. Therapy began 4 wk after valve disruption and lasted through 8 wk. Drugs were administered daily via oral gavage [sacubitril/valsartan (68 mg/kg), valsartan (31 mg/kg), and sacubitril (31 mg/kg)]. Hemodynamic assessments were conducted using Millar technology, and an exercise tolerance test was conducted using a rodent treadmill. Therapy with sacubitril/valsartan improved load-dependent indexes of LV contractility (dP/d tmax) and relaxation (dP/d tmin), exercise tolerance, and mitigated myocardial fibrosis, whereas monotherapies with valsartan, or sacubitril did not. Both sacubitril/valsartan and valsartan similarly improved a load-independent index of contractility [slope of the end-systolic pressure-volume relationship ( Ees)]. Sacubitril did not improve Ees. First, synergy of NEPi with sacubitril and ARB with valsartan leads to the improvement of load-dependent LV contractility and relaxation, exercise tolerance, and reduction of myocardial collagen content. Second, the improvement in load-independent LV contractility with sacubitril/valsartan appears to be solely due to ARB with valsartan constituent. NEW & NOTEWORTHY Our data suggest the following explanation for the effects of sacubitril/valsartan: 1) synergy of sacubitril and valsartan leads to the improvement of load-dependent left ventricular contractility and relaxation, exercise tolerance, and reduction of myocardial fibrosis and 2) improvement in load-independent left ventricular contractility is solely due to the valsartan constituent. The findings offer a better understanding of the outcomes observed in clinical studies and might facilitate the continuing development of the next generations of angiotensin receptor neprilysin inhibitors.
Collapse
Affiliation(s)
- Mikhail Y Maslov
- Department of Anesthesiology, Pain Medicine and Critical Care, Steward St. Elizabeth's Medical Center/Tufts University School of Medicine , Boston, Massachusetts
| | - Stephan Foianini
- Department of Anesthesiology, Pain Medicine and Critical Care, Steward St. Elizabeth's Medical Center/Tufts University School of Medicine , Boston, Massachusetts
| | - Dita Mayer
- Department of Anesthesiology, Pain Medicine and Critical Care, Steward St. Elizabeth's Medical Center/Tufts University School of Medicine , Boston, Massachusetts
| | - Michael V Orlov
- Department of Cardiology, Steward St. Elizabeth's Medical Center/Tufts University School of Medicine , Boston, Massachusetts
| | - Mark A Lovich
- Department of Anesthesiology, Pain Medicine and Critical Care, Steward St. Elizabeth's Medical Center/Tufts University School of Medicine , Boston, Massachusetts
| |
Collapse
|
45
|
Bolívar S, Santana R, Ayala P, Landaeta R, Boza P, Humeres C, Vivar R, Muñoz C, Pardo V, Fernandez S, Anfossi R, Diaz-Araya G. Lipopolysaccharide Activates Toll-Like Receptor 4 and Prevents Cardiac Fibroblast-to-Myofibroblast Differentiation. Cardiovasc Toxicol 2018; 17:458-470. [PMID: 28220374 DOI: 10.1007/s12012-017-9404-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bacterial lipopolysaccharide (LPS) is a known ligand of Toll-like receptor 4 (TLR4) which is expressed in cardiac fibroblasts (CF). Differentiation of CF to cardiac myofibroblasts (CMF) is induced by transforming growth factor-β1 (TGF-β1), increasing alpha-smooth muscle actin (α-SMA) expression. In endothelial cells, an antagonist effect between LPS-induced signaling and canonical TGF-β1 signaling was described; however, it has not been studied whether in CF and CMF the expression of α-SMA induced by TGF-β1 is antagonized by LPS and the mechanism involved. In adult rat CF and CMF, α-SMA, ERK1/2, Akt, NF-κβ, Smad3, and Smad7 protein levels were determined by western blot, TGF-β isoforms by ELISA, and α-SMA stress fibers by immunocytochemistry. CF and CMF secrete the three TGF-β isoforms, and the secretion levels of TGF-β2 was affected by LPS treatment. In CF, LPS treatment decreased the protein levels of α-SMA, and this effect was prevented by TAK-242 (TLR4 inhibitor) and LY294002 (Akt inhibitor), but not by BAY 11-7082 (NF-κβ inhibitor) and PD98059 (ERK1/2 inhibitor). TGF-β1 increased α-SMA protein levels in CF, and LPS prevented partially this effect. In addition, in CMF α-SMA protein levels were decreased by LPS treatment, which was abolished by TAK-242. Finally, in CF LPS decreased the p-Smad3 phosphorylation and increased the Smad7 protein levels. LPS treatment prevents the CF-to-CMF differentiation and reverses the CMF phenotype induced by TGF-β1, through decreasing p-Smad3 and increasing Smad7 protein levels.
Collapse
Affiliation(s)
- Samir Bolívar
- Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile.,Faculty of Chemistry and Pharmacy, Atlantic University, Barranquilla, Colombia
| | - Roxana Santana
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, University of Chile, 8380492, Santiago, Chile.,Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Pedro Ayala
- Center of Medical Investigations, Catholic University of Chile, Santiago, Chile
| | - Rodolfo Landaeta
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, University of Chile, 8380492, Santiago, Chile.,Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Pía Boza
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, University of Chile, 8380492, Santiago, Chile.,Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Claudio Humeres
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, University of Chile, 8380492, Santiago, Chile.,Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Raúl Vivar
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, University of Chile, 8380492, Santiago, Chile.,Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Claudia Muñoz
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, University of Chile, 8380492, Santiago, Chile.,Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Viviana Pardo
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, University of Chile, 8380492, Santiago, Chile.,Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Samuel Fernandez
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, University of Chile, 8380492, Santiago, Chile.,Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Renatto Anfossi
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, University of Chile, 8380492, Santiago, Chile.,Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Guillermo Diaz-Araya
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, University of Chile, 8380492, Santiago, Chile. .,Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile.
| |
Collapse
|
46
|
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
|
47
|
Grassini DR, Lagendijk AK, De Angelis JE, Da Silva J, Jeanes A, Zettler N, Bower NI, Hogan BM, Smith KA. Nppa and Nppb act redundantly during zebrafish cardiac development to confine AVC marker expression and reduce cardiac jelly volume. Development 2018; 145:dev.160739. [PMID: 29752386 DOI: 10.1242/dev.160739] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 05/02/2018] [Indexed: 12/30/2022]
Abstract
Atrial natriuretic peptide (nppa/anf) and brain natriuretic peptide (nppb/bnp) form a gene cluster with expression in the chambers of the developing heart. Despite restricted expression, a function in cardiac development has not been demonstrated by mutant analysis. This is attributed to functional redundancy; however, their genomic location in cis has impeded formal analysis. Using genome editing, we have generated mutants for nppa and nppb, and found that single mutants were indistinguishable from wild type, whereas nppa/nppb double mutants displayed heart morphogenesis defects and pericardial oedema. Analysis of atrioventricular canal (AVC) markers show expansion of bmp4, tbx2b, has2 and versican expression into the atrium of double mutants. This expanded expression correlates with increased extracellular matrix in the atrium. Using a biosensor for hyaluronic acid to measure the cardiac jelly (cardiac extracellular matrix), we confirmed cardiac jelly expansion in nppa/nppb double mutants. Finally, bmp4 knockdown rescued the expansion of has2 expression and cardiac jelly in double mutants. This definitively shows that nppa and nppb function redundantly during cardiac development to restrict gene expression to the AVC, preventing excessive cardiac jelly synthesis in the atrial chamber.
Collapse
Affiliation(s)
- Daniela R Grassini
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Anne K Lagendijk
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jessica E De Angelis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jason Da Silva
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Angela Jeanes
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Nicole Zettler
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Neil I Bower
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Benjamin M Hogan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Kelly A Smith
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
48
|
Michalska-Kasiczak M, Bielecka-Dabrowa A, von Haehling S, Anker SD, Rysz J, Banach M. Biomarkers, myocardial fibrosis and co-morbidities in heart failure with preserved ejection fraction: an overview. Arch Med Sci 2018; 14:890-909. [PMID: 30002709 PMCID: PMC6040115 DOI: 10.5114/aoms.2018.76279] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 04/24/2018] [Indexed: 02/06/2023] Open
Abstract
The prevalence of heart failure with preserved ejection fraction (HFpEF) is steadily increasing. Its diagnosis remains difficult and controversial and relies mostly on non-invasive echocardiographic detection of left ventricular diastolic dysfunction and elevated filling pressures. The large phenotypic heterogeneity of HFpEF from pathophysiologic al underpinnings to clinical manifestations presents a major obstacle to the development of new therapies targeted towards specific HF phenotypes. Recent studies suggest that natriuretic peptides have the potential to improve the diagnosis of early HFpEF, but they still have significant limitations, and the cut-off points for diagnosis and prognosis in HFpEF remain open to debate. The purpose of this review is to present potential targets of intervention in patients with HFpEF, starting with myocardial fibrosis and methods of its detection. In addition, co-morbidities are discussed as a means to treat HFpEF according to cut-points of biomarkers that are different from usual. Biomarkers and approaches to co-morbidities may be able to tailor therapies according to patients' pathophysiological needs. Recently, soluble source of tumorigenicity 2 (sST2), growth differentiation factor 15 (GDF-15), galectin-3, and other cardiac markers have emerged, but evidence from large cohorts is still lacking. Furthermore, the field of miRNA is a very promising area of research, and further exploration of miRNA may offer diagnostic and prognostic applications and insight into the pathology, pointing to new phenotype-specific therapeutic targets.
Collapse
Affiliation(s)
- Marta Michalska-Kasiczak
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
- Department of Endocrine Disorders and Bone Metabolism, 1 Chair of Endocrinology, Medical University of Lodz, Lodz, Poland
| | - Agata Bielecka-Dabrowa
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
- Department of Cardiology and Congenital Diseases of Adults, Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Göttingen, Germany
| | - Stefan D. Anker
- Division of Cardiology and Metabolism – Heart Failure, Cachexia and Sarcopenia, Department of Cardiology, Campus Virchow-Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
49
|
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
|
50
|
Fu S, Ping P, Wang F, Luo L. Synthesis, secretion, function, metabolism and application of natriuretic peptides in heart failure. J Biol Eng 2018; 12:2. [PMID: 29344085 PMCID: PMC5766980 DOI: 10.1186/s13036-017-0093-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/21/2017] [Indexed: 12/11/2022] Open
Abstract
As a family of hormones with pleiotropic effects, natriuretic peptide (NP) system includes atrial NP (ANP), B-type NP (BNP), C-type NP (CNP), dendroaspis NP and urodilatin, with NP receptor-A (guanylate cyclase-A), NP receptor-B (guanylate cyclase-B) and NP receptor-C (clearance receptor). These peptides are genetically distinct, but structurally and functionally related for regulating circulatory homeostasis in vertebrates. In humans, ANP and BNP are encoded by NP precursor A (NPPA) and NPPB genes on chromosome 1, whereas CNP is encoded by NPPC on chromosome 2. NPs are synthesized and secreted through certain mechanisms by cardiomyocytes, fibroblasts, endotheliocytes, immune cells (neutrophils, T-cells and macrophages) and immature cells (embryonic stem cells, muscle satellite cells and cardiac precursor cells). They are mainly produced by cardiovascular, brain and renal tissues in response to wall stretch and other causes. NPs provide natriuresis, diuresis, vasodilation, antiproliferation, antihypertrophy, antifibrosis and other cardiometabolic protection. NPs represent body's own antihypertensive system, and provide compensatory protection to counterbalance vasoconstrictor-mitogenic-sodium retaining hormones, released by renin-angiotensin-aldosterone system (RAAS) and sympathetic nervous system (SNS). NPs play central roles in regulation of heart failure (HF), and are inactivated through not only NP receptor-C, but also neutral endopeptidase (NEP), dipeptidyl peptidase-4 and insulin degrading enzyme. Both BNP and N-terminal proBNP are useful biomarkers to not only make the diagnosis and assess the severity of HF, but also guide the therapy and predict the prognosis in patients with HF. Current NP-augmenting strategies include the synthesis of NPs or agonists to increase NP bioactivity and inhibition of NEP to reduce NP breakdown. Nesiritide has been established as an available therapy, and angiotensin receptor blocker NEP inhibitor (ARNI, LCZ696) has obtained extremely encouraging results with decreased morbidity and mortality. Novel pharmacological approaches based on NPs may promote a therapeutic shift from suppressing the RAAS and SNS to re-balancing neuroendocrine dysregulation in patients with HF. The current review discussed the synthesis, secretion, function and metabolism of NPs, and their diagnostic, therapeutic and prognostic values in HF.
Collapse
Affiliation(s)
- Shihui Fu
- Department of Geriatric Cardiology, Chinese People’s Liberation Army General Hospital, Beijing, 100853 China
- Department of Cardiology and Hainan Branch, Chinese People’s Liberation Army, General Hospital, Beijing, China
| | - Ping Ping
- Department of Pharmaceutical Care, Chinese People’s, Liberation Army General Hospital, Beijing, China
| | - Fengqi Wang
- Department of Cardiology and Hainan Branch, Chinese People’s Liberation Army, General Hospital, Beijing, China
| | - Leiming Luo
- Department of Geriatric Cardiology, Chinese People’s Liberation Army General Hospital, Beijing, 100853 China
| |
Collapse
|