1
|
Landim-Vieira M, Nieto Morales PF, ElSafty S, Kahmini AR, Ranek MJ, Solís C. The role of mechanosignaling in the control of myocardial mass. Am J Physiol Heart Circ Physiol 2025; 328:H622-H638. [PMID: 39739566 DOI: 10.1152/ajpheart.00277.2024] [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: 04/30/2024] [Revised: 12/03/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
Regulation of myocardial mass is key for maintaining cardiovascular health. This review highlights the complex and regulatory relationship between mechanosignaling and myocardial mass, influenced by many internal and external factors including hemodynamic and microgravity, respectively. The heart is a dynamic organ constantly adapting to changes in workload (preload and afterload) and mechanical stress exerted on the myocardium, influencing both physiological adaptations and pathological remodeling. Mechanosignaling pathways, such as the mitogen-activated protein kinases (MAPKs) and the phosphoinositide 3-kinases and serine/threonine kinase (PI3K/Akt) pathways, mediate downstream effects on gene expression and play key roles in transducing mechanical cues into biochemical signals, thereby modulating cellular processes, including control of myocardial mass. Dysregulation of these processes can lead to pathological cardiac remodeling, such as hypertrophic cardiomyopathy. Furthermore, recent studies have highlighted the importance of protein quality control mechanisms, such as the ubiquitin-proteasome system, in settings of extreme physiological conditions that alter the heart workload such as pregnancy and microgravity. Overall, this review provides a thorough insight into how mechanical signals are converted into chemical signals to regulate myocardial mass in both healthy and diseased conditions.
Collapse
Affiliation(s)
- Maicon Landim-Vieira
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Paula F Nieto Morales
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Summer ElSafty
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Aida Rahimi Kahmini
- Department of Health, Nutrition, and Food Science, Florida State University, Tallahassee, Florida, United States
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Christopher Solís
- Department of Health, Nutrition, and Food Science, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
2
|
You H, Gou Q, Dong M, Chang F, Xiu J. Exploring the role of iNOS in HFpEF-Related myocardial fibrosis: Involvement of PTEN-PI3K/AKT signaling pathway. Biochem Biophys Res Commun 2024; 734:150589. [PMID: 39245028 DOI: 10.1016/j.bbrc.2024.150589] [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: 07/04/2024] [Revised: 08/06/2024] [Accepted: 08/21/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a challenging condition to treat with myocardial fibrosis being a pivotal pathological component. Previous studies have suggested a role for inducible nitric oxide synthase (iNOS) in the progression of this condition, but the precise mechanisms remain unclear. This study aimed to investigate the role of iNOS in HFpEF-related myocardial fibrosis and identify potential therapeutic targets. METHODS A 'two-hit' mouse model of HFpEF was established, and echocardiography, histopathology and biochemical analyses were performed. In vitro experiments were conducted in mouse cardiac fibroblasts, with iNOS overexpression and application of iNOS or phosphatidylinositol 3 kinase (PI3K) inhibitors. The iNOS-S-nitrosylated phosphatase and TENsin homolog (SNO-PTEN)-phosphorylated-protein kinase B (p-AKT) pathway was investigated, along with the effects on fibrotic markers and cell proliferation and migration. RESULTS HFpEF mice exhibited significant cardiac dysfunction and fibrosis, with increased expression of iNOS, SNO-PTEN, and p-AKT, indicative of the activation of the iNOS-SNO-PTEN-p-AKT pathway. iNOS overexpression in mouse cardiac fibroblasts led to increased SNO-PTEN, decreased PTEN, activated phosphorylated PI3K (p-PI3K) and p-AKT, and enhanced cell proliferation and migration, as well as increased collagen I and III expression. The use of an iNOS inhibitor (L-NIL) or a PI3K inhibitor (LY294002) partially reversed these changes. CONCLUSION Our findings suggest that the iNOS-SNO-PTEN-p-AKT pathway may play a crucial role in HFpEF-related myocardial fibrosis, with iNOS and PI3K inhibitors offering potential therapeutic benefits. These insights may pave the way for the development of effective drug therapies for HFpEF.
Collapse
Affiliation(s)
- Hongjun You
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China; Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Qiling Gou
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Mengya Dong
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Fengjun Chang
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Jiancheng Xiu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
3
|
Okuma H, Tsuchiya K. Tissue-specific activation of insulin signaling as a potential target for obesity-related metabolic disorders. Pharmacol Ther 2024; 262:108699. [PMID: 39111411 DOI: 10.1016/j.pharmthera.2024.108699] [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: 03/06/2024] [Revised: 06/17/2024] [Accepted: 07/31/2024] [Indexed: 09/14/2024]
Abstract
The incidence of obesity is rapidly increasing worldwide. Obesity-associated insulin resistance has long been established as a significant risk factor for obesity-related disorders such as type 2 diabetes and atherosclerosis. Insulin plays a key role in systemic glucose metabolism, with the liver, skeletal muscle, and adipose tissue as the major acting tissues. Insulin receptors and the downstream insulin signaling-related molecules are expressed in various tissues, including vascular endothelial cells, vascular smooth muscle cells, and monocytes/macrophages. In obesity, decreased insulin action is considered a driver for associated disorders. However, whether insulin action has a positive or negative effect on obesity-related disorders depends on the tissue in which it acts. While an enhancement of insulin signaling in the liver increases hepatic fat accumulation and exacerbates dyslipidemia, enhancement of insulin signaling in adipose tissue protects against obesity-related dysfunction of various organs by increasing the capacity for fat accumulation in the adipose tissue and inhibiting ectopic fat accumulation. Thus, this "healthy adipose tissue expansion" by enhancing insulin sensitivity in adipose tissue, but not in the liver, may be an effective therapeutic strategy for obesity-related disorders. To effectively address obesity-related metabolic disorders, the mechanisms of insulin resistance in various tissues of obese patients must be understood and drugs that enhance insulin action must be developed. In this article, we review the potential of interventions that enhance insulin signaling as a therapeutic strategy for obesity-related disorders, focusing on the molecular mechanisms of insulin action in each tissue.
Collapse
Affiliation(s)
- Hideyuki Okuma
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 4093898, Japan
| | - Kyoichiro Tsuchiya
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 4093898, Japan.
| |
Collapse
|
4
|
Caturano A, Galiero R, Vetrano E, Sardu C, Rinaldi L, Russo V, Monda M, Marfella R, Sasso FC. Insulin-Heart Axis: Bridging Physiology to Insulin Resistance. Int J Mol Sci 2024; 25:8369. [PMID: 39125938 PMCID: PMC11313400 DOI: 10.3390/ijms25158369] [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: 07/01/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Insulin signaling is vital for regulating cellular metabolism, growth, and survival pathways, particularly in tissues such as adipose, skeletal muscle, liver, and brain. Its role in the heart, however, is less well-explored. The heart, requiring significant ATP to fuel its contractile machinery, relies on insulin signaling to manage myocardial substrate supply and directly affect cardiac muscle metabolism. This review investigates the insulin-heart axis, focusing on insulin's multifaceted influence on cardiac function, from metabolic regulation to the development of physiological cardiac hypertrophy. A central theme of this review is the pathophysiology of insulin resistance and its profound implications for cardiac health. We discuss the intricate molecular mechanisms by which insulin signaling modulates glucose and fatty acid metabolism in cardiomyocytes, emphasizing its pivotal role in maintaining cardiac energy homeostasis. Insulin resistance disrupts these processes, leading to significant cardiac metabolic disturbances, autonomic dysfunction, subcellular signaling abnormalities, and activation of the renin-angiotensin-aldosterone system. These factors collectively contribute to the progression of diabetic cardiomyopathy and other cardiovascular diseases. Insulin resistance is linked to hypertrophy, fibrosis, diastolic dysfunction, and systolic heart failure, exacerbating the risk of coronary artery disease and heart failure. Understanding the insulin-heart axis is crucial for developing therapeutic strategies to mitigate the cardiovascular complications associated with insulin resistance and diabetes.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Luca Rinaldi
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy;
| | - Vincenzo Russo
- Department of Biology, College of Science and Technology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA;
- Division of Cardiology, Department of Medical Translational Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (A.C.); (R.G.); (E.V.); (C.S.); (R.M.)
| |
Collapse
|
5
|
Achter JS, Vega ET, Sorrentino A, Kahnert K, Galsgaard KD, Hernandez-Varas P, Wierer M, Holst JJ, Wojtaszewski JFP, Mills RW, Kjøbsted R, Lundby A. In-depth phosphoproteomic profiling of the insulin signaling response in heart tissue and cardiomyocytes unveils canonical and specialized regulation. Cardiovasc Diabetol 2024; 23:258. [PMID: 39026321 PMCID: PMC11264841 DOI: 10.1186/s12933-024-02338-4] [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: 03/12/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Insulin signaling regulates cardiac substrate utilization and is implicated in physiological adaptations of the heart. Alterations in the signaling response within the heart are believed to contribute to pathological conditions such as type-2 diabetes and heart failure. While extensively investigated in several metabolic organs using phosphoproteomic strategies, the signaling response elicited in cardiac tissue in general, and specifically in the specialized cardiomyocytes, has not yet been investigated to the same extent. METHODS Insulin or vehicle was administered to male C57BL6/JRj mice via intravenous injection into the vena cava. Ventricular tissue was extracted and subjected to quantitative phosphoproteomics analysis to evaluate the insulin signaling response. To delineate the cardiomyocyte-specific response and investigate the role of Tbc1d4 in insulin signal transduction, cardiomyocytes from the hearts of cardiac and skeletal muscle-specific Tbc1d4 knockout mice, as well as from wildtype littermates, were studied. The phosphoproteomic studies involved isobaric peptide labeling with Tandem Mass Tags (TMT), enrichment for phosphorylated peptides, fractionation via micro-flow reversed-phase liquid chromatography, and high-resolution mass spectrometry measurements. RESULTS We quantified 10,399 phosphorylated peptides from ventricular tissue and 12,739 from isolated cardiomyocytes, localizing to 3,232 and 3,128 unique proteins, respectively. In cardiac tissue, we identified 84 insulin-regulated phosphorylation events, including sites on the Insulin Receptor (InsrY1351, Y1175, Y1179, Y1180) itself as well as the Insulin receptor substrate protein 1 (Irs1S522, S526). Predicted kinases with increased activity in response to insulin stimulation included Rps6kb1, Akt1 and Mtor. Tbc1d4 emerged as a major phosphorylation target in cardiomyocytes. Despite limited impact on the global phosphorylation landscape, Tbc1d4 deficiency in cardiomyocytes attenuated insulin-induced Glut4 translocation and induced protein remodeling. We observed 15 proteins significantly regulated upon knockout of Tbc1d4. While Glut4 exhibited decreased protein abundance consequent to Tbc1d4-deficiency, Txnip levels were notably increased. Stimulation of wildtype cardiomyocytes with insulin led to the regulation of 262 significant phosphorylation events, predicted to be regulated by kinases such as Akt1, Mtor, Akt2, and Insr. In cardiomyocytes, the canonical insulin signaling response is elicited in addition to regulation on specialized cardiomyocyte proteins, such as Kcnj11Y12 and DspS2597. Details of all phosphorylation sites are provided. CONCLUSION We present a first global outline of the insulin-induced phosphorylation signaling response in heart tissue and in isolated adult cardiomyocytes, detailing the specific residues with changed phosphorylation abundances. Our study marks an important step towards understanding the role of insulin signaling in cardiac diseases linked to insulin resistance.
Collapse
Affiliation(s)
- Jonathan Samuel Achter
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Estefania Torres Vega
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Sorrentino
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Konstantin Kahnert
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Douglas Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pablo Hernandez-Varas
- Core Facility for Integrated Microscopy, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Wierer
- Proteomics Research Infrastructure, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen Frank Pind Wojtaszewski
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Robert William Mills
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Kjøbsted
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
6
|
Garmany R, Dasari S, Bos JM, Kim ET, Tester DJ, Dos Remedios C, Maleszewski JJ, Robertson KD, Dearani JA, Ommen SR, Giudicessi JR, Ackerman MJ. Histone Modifications and miRNA Perturbations Contribute to Transcriptional Dysregulation of Hypertrophy in Obstructive Hypertrophic Cardiomyopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593374. [PMID: 38798679 PMCID: PMC11118430 DOI: 10.1101/2024.05.09.593374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background Recently, we demonstrated transcriptional downregulation of hypertrophy pathways in myectomy tissue derived from patients with obstructive hypertrophic cardiomyopathy (HCM) despite translational activation of hypertrophy pathways. The mechanisms and modifiers of this transcriptional dysregulation in HCM remain unexplored. We hypothesized that miRNA and post-translational modifications of histones contribute to transcriptional dysregulation in HCM. Methods First, miRNA-sequencing and chromatin immunoprecipitation sequencing (ChIP-seq) were performed on HCM myectomy tissue and control donor hearts to characterize miRNA and differential histone marks across the genome. Next, the differential miRNA and histone marks were integrated with RNA-sequencing (RNA-seq) data. Finally, the effects of miRNA and histones were removed in silico to determine their necessity for transcriptional dysregulation of pathways. Results miRNA-analysis identified 19 differentially expressed miRNA. ChIP-seq analysis identified 2,912 (7%) differential H3K4me3 peaks, 23,339 (21%) differential H3K9ac peaks, 33 (0.05%) differential H3K9me3 peaks, 58,837 (42%) differential H3K27ac peaks, and 853 (3%) differential H3K27me3 peaks. Univariate analysis of concordance between H3K9ac with RNA-seq data showed activation of cardiac hypertrophy signaling, while H3K27me showed downregulation of cardiac hypertrophy signaling. Similarly, miRNAs were predicted to result in downregulation of cardiac hypertrophy signaling. In silico knock-out that effects either miRNA or histones attenuated transcriptional downregulation while knocking out both abolished downregulation of hypertrophy pathways completely. Conclusion Myectomy tissue from patients with obstructive HCM shows transcriptional dysregulation, including transcriptional downregulation of hypertrophy pathways mediated by miRNA and post-translational modifications of histones. Cardiac hypertrophy loci showed activation via changes in H3K9ac and a mix of activation and repression via H3K27ac.
Collapse
|
7
|
Poveda J, González-Lafuente L, Vázquez-Sánchez S, Mercado-García E, Rodríguez-Sánchez E, García-Consuegra I, Sanz AB, Segura J, Fernández-Velasco M, Liaño F, Ruilope LM, Ruiz-Hurtado G. Targeting the TWEAK-Fn14 pathway prevents dysfunction in cardiac calcium handling after acute kidney injury. J Pathol 2023; 261:427-441. [PMID: 37776271 DOI: 10.1002/path.6200] [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: 03/15/2023] [Revised: 07/07/2023] [Accepted: 08/11/2023] [Indexed: 10/02/2023]
Abstract
Heart and kidney have a closely interrelated pathophysiology. Acute kidney injury (AKI) is associated with significantly increased rates of cardiovascular events, a relationship defined as cardiorenal syndrome type 3 (CRS3). The underlying mechanisms that trigger heart disease remain, however, unknown, particularly concerning the clinical impact of AKI on cardiac outcomes and overall mortality. Tumour necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor fibroblast growth factor-inducible 14 (Fn14) are independently involved in the pathogenesis of both heart and kidney failure, and recent studies have proposed TWEAK as a possible therapeutic target; however, its specific role in cardiac damage associated with CRS3 remains to be clarified. Firstly, we demonstrated in a retrospective longitudinal clinical study that soluble TWEAK plasma levels were a predictive biomarker of mortality in patients with AKI. Furthermore, the exogenous application of TWEAK to native ventricular cardiomyocytes induced relevant calcium (Ca2+ ) handling alterations. Next, we investigated the role of the TWEAK-Fn14 axis in cardiomyocyte function following renal ischaemia-reperfusion (I/R) injury in mice. We observed that TWEAK-Fn14 signalling was activated in the hearts of AKI mice. Mice also showed significantly altered intra-cardiomyocyte Ca2+ handling and arrhythmogenic Ca2+ events through an impairment in sarcoplasmic reticulum Ca2+ -adenosine triphosphatase 2a pump (SERCA2a ) and ryanodine receptor (RyR2 ) function. Administration of anti-TWEAK antibody after reperfusion significantly improved alterations in Ca2+ cycling and arrhythmogenic events and prevented SERCA2a and RyR2 modifications. In conclusion, this study establishes the relevance of the TWEAK-Fn14 pathway in cardiac dysfunction linked to CRS3, both as a predictor of mortality in patients with AKI and as a Ca2+ mishandling inducer in cardiomyocytes, and highlights the cardioprotective benefits of TWEAK targeting in CRS3. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jonay Poveda
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Laura González-Lafuente
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Sara Vázquez-Sánchez
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Elisa Mercado-García
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Elena Rodríguez-Sánchez
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Inés García-Consuegra
- Proteomics Unit, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Ana Belén Sanz
- Nephrology Laboratory, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid and REDINREN, Madrid, Spain
| | - Julián Segura
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
- Servicio de Nefrología, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - María Fernández-Velasco
- IdiPAZ Institute for Health Research/Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, CIBER-CV, Madrid, Spain
| | - Fernando Liaño
- Instituto Ramón y Cajal de Investigación Sanitaria (IRyCis), Madrid, Spain
| | - Luis M Ruilope
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
- CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
- School of Doctoral Studies and Research, European University of Madrid, Madrid, Spain
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
- CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
8
|
Salvatori F, D’Aversa E, Serino ML, Singh AV, Secchiero P, Zauli G, Tisato V, Gemmati D. miRNAs Epigenetic Tuning of Wall Remodeling in the Early Phase after Myocardial Infarction: A Novel Epidrug Approach. Int J Mol Sci 2023; 24:13268. [PMID: 37686073 PMCID: PMC10487654 DOI: 10.3390/ijms241713268] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Myocardial infarction (MI) is one of the leading causes of death in Western countries. An early diagnosis decreases subsequent severe complications such as wall remodeling or heart failure and improves treatments and interventions. Novel therapeutic targets have been recognized and, together with the development of direct and indirect epidrugs, the role of non-coding RNAs (ncRNAs) yields great expectancy. ncRNAs are a group of RNAs not translated into a product and, among them, microRNAs (miRNAs) are the most investigated subgroup since they are involved in several pathological processes related to MI and post-MI phases such as inflammation, apoptosis, angiogenesis, and fibrosis. These processes and pathways are finely tuned by miRNAs via complex mechanisms. We are at the beginning of the investigation and the main paths are still underexplored. In this review, we provide a comprehensive discussion of the recent findings on epigenetic changes involved in the first phases after MI as well as on the role of the several miRNAs. We focused on miRNAs function and on their relationship with key molecules and cells involved in healing processes after an ischemic accident, while also giving insight into the discrepancy between males and females in the prognosis of cardiovascular diseases.
Collapse
Affiliation(s)
- Francesca Salvatori
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.S.)
| | - Elisabetta D’Aversa
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.S.)
| | - Maria Luisa Serino
- Centre Haemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
| | - Ajay Vikram Singh
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Paola Secchiero
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.S.)
| | - Giorgio Zauli
- Department of Environmental Science and Prevention, University of Ferrara, 44121 Ferrara, Italy
| | - Veronica Tisato
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.S.)
- LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
- University Centre for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Donato Gemmati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.S.)
- Centre Haemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
- University Centre for Studies on Gender Medicine, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
9
|
Yeh HH, Chang YM, Chang YW, Lu MYJ, Chen YH, Lee CC, Chen CC. Multiomic analyses reveal enriched glycolytic processes in β-myosin heavy chain-expressed cardiomyocytes in early cardiac hypertrophy. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2022; 1:100011. [PMID: 39801720 PMCID: PMC11708374 DOI: 10.1016/j.jmccpl.2022.100011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/24/2022] [Accepted: 06/16/2022] [Indexed: 01/16/2025]
Abstract
Background Cardiac pressure overload induces cardiac hypertrophy and eventually leads to heart failure. One distinct feature of pathological cardiac hypertrophy is fetal-gene re-expression, but not every cardiomyocyte exhibits fetal gene re-expression in the diseased heart. Adult cardiomyocytes are terminally differentiated cells, so we do not know how the heterogeneity is determined and whether the differential fetal-gene reprogramming indicates a different degree of remodeling among cardiomyocytes. We hypothesized that fetal gene-expressed cardiomyocytes show more pathological features in the pressure-overloaded heart. Results We induced pressure overload in mice by transverse aortic constriction (TAC) and observed a cardiomyocyte population with expression of β-myosin heavy chain (βMHC, a fetal gene encoded by Myh7) after TAC for 3 days. On transcriptomic and proteomic analyses, βMHC-expressed cardiomyocytes of 3-day TAC hearts were enriched in genes in cardiomyopathy-associated pathways and glycolytic processes. Moreover, results of immunoblotting and enzyme activity assay suggested higher glycolytic activity in βMHC-expressed than non-expressed cardiomyocytes. When we inhibited the glycolytic flux by 2-deoxy-d-glucose, a widely used glycolysis inhibitor, the number of βMHC-expressed cardiomyocytes was reduced, and the level of TEA domain family member 1 (TEAD1), a transcriptional enhancer, was decreased. Also, our spatial transcriptomic results demonstrated that naïve and 3-day TAC hearts had fetal-gene-rich tissue domains that were enriched in pathways in extracellular matrix organization and tissue remodeling. As well, gene levels of glycolytic enzymes were higher in Myh7-positive than Myh7-negative domains. Conclusions Our data suggest that βMHC-expressed cardiomyocytes progress to pathological remodeling in the early stages of cardiac hypertrophy. In addition, the diverse glycolytic activity among cardiomyocytes might play a role in regulating gene expression via TEAD1 signaling.
Collapse
Affiliation(s)
- Hsiao-hui Yeh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Yao-Ming Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Wang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Mei-Yeh Jade Lu
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-Hua Chen
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Chia-Che Lee
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| |
Collapse
|
10
|
St-Cyr S, Child DD, Giaime E, Smith AR, Pascua CJ, Hahm S, Saiah E, Davidson BL. Huntington’s disease phenotypes are improved via mTORC1 modulation by small molecule therapy. PLoS One 2022; 17:e0273710. [PMID: 36037192 PMCID: PMC9423655 DOI: 10.1371/journal.pone.0273710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
Abstract
Huntington’s Disease (HD) is a dominantly inherited neurodegenerative disease for which the major causes of mortality are neurodegeneration-associated aspiration pneumonia followed by cardiac failure. mTORC1 pathway perturbations are present in HD models and human tissues. Amelioration of mTORC1 deficits by genetic modulation improves disease phenotypes in HD models, is not a viable therapeutic strategy. Here, we assessed a novel small molecule mTORC1 pathway activator, NV-5297, for its improvement of the disease phenotypes in the N171-82Q HD mouse model. Oral dosing of NV-5297 over 6 weeks activated mTORC1, increased striatal volume, improved motor learning and heart contractility. Further, the heart contractility, heart fibrosis, and survival were improved in response to the cardiac stressor isoprenaline when compared to vehicle-treated mice. Cummulatively, these data support mTORC1 activation as a therapeutic target in HD and consolidates NV-5297 as a promising drug candidate for treating central and peripheral HD phenotypes and, more generally, mTORC1-deficit related diseases.
Collapse
Affiliation(s)
- Sophie St-Cyr
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Daniel D. Child
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- The Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, United States of America
| | - Emilie Giaime
- Navitor Pharmaceuticals Inc., Cambridge, MA, United States of America
| | - Alicia R. Smith
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Christine J. Pascua
- Division of Cardiology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Seung Hahm
- Navitor Pharmaceuticals Inc., Cambridge, MA, United States of America
| | - Eddine Saiah
- Navitor Pharmaceuticals Inc., Cambridge, MA, United States of America
- * E-mail: (BLD); (ES)
| | - Beverly L. Davidson
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- The Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail: (BLD); (ES)
| |
Collapse
|
11
|
Walkowski B, Kleibert M, Majka M, Wojciechowska M. Insight into the Role of the PI3K/Akt Pathway in Ischemic Injury and Post-Infarct Left Ventricular Remodeling in Normal and Diabetic Heart. Cells 2022; 11:cells11091553. [PMID: 35563860 PMCID: PMC9105930 DOI: 10.3390/cells11091553] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 02/07/2023] Open
Abstract
Despite the significant decline in mortality, cardiovascular diseases are still the leading cause of death worldwide. Among them, myocardial infarction (MI) seems to be the most important. A further decline in the death rate may be achieved by the introduction of molecularly targeted drugs. It seems that the components of the PI3K/Akt signaling pathway are good candidates for this. The PI3K/Akt pathway plays a key role in the regulation of the growth and survival of cells, such as cardiomyocytes. In addition, it has been shown that the activation of the PI3K/Akt pathway results in the alleviation of the negative post-infarct changes in the myocardium and is impaired in the state of diabetes. In this article, the role of this pathway was described in each step of ischemia and subsequent left ventricular remodeling. In addition, we point out the most promising substances which need more investigation before introduction into clinical practice. Moreover, we present the impact of diabetes and widely used cardiac and antidiabetic drugs on the PI3K/Akt pathway and discuss the molecular mechanism of its effects on myocardial ischemia and left ventricular remodeling.
Collapse
Affiliation(s)
- Bartosz Walkowski
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
| | - Marcin Kleibert
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Correspondence: (M.K.); (M.M.)
| | - Miłosz Majka
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Correspondence: (M.K.); (M.M.)
| | - Małgorzata Wojciechowska
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Invasive Cardiology Unit, Independent Public Specialist Western Hospital John Paul II, Daleka 11, 05-825 Grodzisk Mazowiecki, Poland
| |
Collapse
|
12
|
A Detailed Study to Discover the Trade between Left Atrial Blood Flow, Expression of Calcium-Activated Potassium Channels and Valvular Atrial Fibrillation. Cells 2022; 11:cells11091383. [PMID: 35563689 PMCID: PMC9103658 DOI: 10.3390/cells11091383] [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: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background: The present study aimed to explore the correlation between calcium-activated potassium channels, left atrial flow field mechanics, valvular atrial fibrillation (VAF), and thrombosis. The process of transforming mechanical signals into biological signals has been revealed, which offers new insights into the study of VAF. Methods: Computational fluid dynamics simulations use numeric analysis and algorithms to compute flow parameters, including turbulent shear stress (TSS) and wall pressure in the left atrium (LA). Real-time PCR and western blotting were used to detect the mRNA and protein expression of IKCa2.3/3.1, ATK1, and P300 in the left atrial tissue of 90 patients. Results: In the valvular disease group, the TSS and wall ressure in the LA increased, the wall pressure increased in turn in all disease groups, mainly near the mitral valve and the posterior portion of the LA, the increase in TSS was the most significant in each group near the mitral valve, and the middle and lower part of the back of the LA and the mRNA expression and protein expression levels of IKCa2.3/3.1, AKT1, and P300 increased (p < 0.05) (n = 15). The present study was preliminarily conducted to elucidate whether there might be a certain correlation between IKCa2.3 and LA hemodynamic changes. Conclusions: The TSS and wall pressure changes in the LA are correlated with the upregulation of mRNA and protein expression of IKCa2.3/3.1, AKT1, and P300.
Collapse
|
13
|
Al-Shamasi AA, Elkaffash R, Mohamed M, Rayan M, Al-Khater D, Gadeau AP, Ahmed R, Hasan A, Eldassouki H, Yalcin HC, Abdul-Ghani M, Mraiche F. Crosstalk between Sodium-Glucose Cotransporter Inhibitors and Sodium-Hydrogen Exchanger 1 and 3 in Cardiometabolic Diseases. Int J Mol Sci 2021; 22:12677. [PMID: 34884494 PMCID: PMC8657861 DOI: 10.3390/ijms222312677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 12/14/2022] Open
Abstract
Abnormality in glucose homeostasis due to hyperglycemia or insulin resistance is the hallmark of type 2 diabetes mellitus (T2DM). These metabolic abnormalities in T2DM lead to cellular dysfunction and the development of diabetic cardiomyopathy leading to heart failure. New antihyperglycemic agents including glucagon-like peptide-1 receptor agonists and the sodium-glucose cotransporter-2 inhibitors (SGLT2i) have been shown to attenuate endothelial dysfunction at the cellular level. In addition, they improved cardiovascular safety by exhibiting cardioprotective effects. The mechanism by which these drugs exert their cardioprotective effects is unknown, although recent studies have shown that cardiovascular homeostasis occurs through the interplay of the sodium-hydrogen exchangers (NHE), specifically NHE1 and NHE3, with SGLT2i. Another theoretical explanation for the cardioprotective effects of SGLT2i is through natriuresis by the kidney. This theory highlights the possible involvement of renal NHE transporters in the management of heart failure. This review outlines the possible mechanisms responsible for causing diabetic cardiomyopathy and discusses the interaction between NHE and SGLT2i in cardiovascular diseases.
Collapse
Affiliation(s)
- Al-Anood Al-Shamasi
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Rozina Elkaffash
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Meram Mohamed
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Menatallah Rayan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Dhabya Al-Khater
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Alain-Pierre Gadeau
- INSERM, Biology of Cardiovascular Disease, University of Bordeaux, U1034 Pessac, France;
| | - Rashid Ahmed
- Department of Mechanical and Chemical Engineering, College of Engineering, Qatar University, Doha P.O. Box 2713, Qatar; (R.A.); (A.H.)
- Biomedical Research Centre (BRC), Qatar University, Doha P.O. Box 2713, Qatar;
| | - Anwarul Hasan
- Department of Mechanical and Chemical Engineering, College of Engineering, Qatar University, Doha P.O. Box 2713, Qatar; (R.A.); (A.H.)
- Biomedical Research Centre (BRC), Qatar University, Doha P.O. Box 2713, Qatar;
| | - Hussein Eldassouki
- College of Kinesiology, University of Saskatchewan, Saskatoon, SK S7N 5B5, Canada;
| | | | - Muhammad Abdul-Ghani
- Division of Diabetes, University of Texas Health Science Center at San Antonio, Floyd Curl Drive, San Antonio, TX 7703, USA;
| | - Fatima Mraiche
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
14
|
Okamoto C, Tsukamoto O, Hasegawa T, Hitsumoto T, Matsuoka K, Takashima S, Amaki M, Kanzaki H, Izumi C, Ito S, Kitakaze M. Lower B-type natriuretic peptide levels predict left ventricular concentric remodelling and insulin resistance. ESC Heart Fail 2021; 9:636-647. [PMID: 34786876 PMCID: PMC8787986 DOI: 10.1002/ehf2.13700] [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: 07/29/2021] [Revised: 09/21/2021] [Accepted: 10/29/2021] [Indexed: 01/19/2023] Open
Abstract
Aims Natriuretic peptides have reportedly been associated with cardiac hypertrophy and insulin resistance; however, it has not been established if B‐type natriuretic peptide (BNP) is associated with either insulin resistance or cardiac remodelling in a population with normal plasma BNP levels. We investigated the relationship among plasma BNP levels, insulin resistance, and left ventricular (LV) remodelling in a population with normal physiological plasma BNP levels. Methods and results Among 1632 individuals who participated in annual health checks between 2005 and 2008 in Arita‐cho, Saga, Japan, 675 individuals [median (interquartile range) for age 62 (51–69) years; 227 men (34%)] with LV ejection fraction 50% and BNP level <35 pg/mL were enrolled in this study. Insulin resistance was assessed using homeostatic model assessment of insulin resistance (HOMA‐IR). LV geometry, including LV concentric remodelling, was classified based on relative wall thickness (RWT) and LV mass index values derived from echocardiographic findings. Factors associated with insulin resistance and LV geometry were investigated using multiple logistic regression analysis. Tertiles of BNP were inversely associated with HOMA‐IR [1st tertile, 1.33 (0.76–1.74); 2nd tertile, 1.05 (0.72–1.59); 3rd tertile, 0.95 (0.66–1.58), P = 0.005]. Lower BNP was associated with the prevalence of insulin resistance, defined as HOMA‐IR ≥1.37, even after full multivariate adjustment [1 SD increment in BNP = adjusted odds ratio (aOR) 0.740; 95% confidence interval (CI) 0.601–0.912; P = 0.005]. LV concentric remodelling (RWT >0.42; LV mass index ≤115 g/m2 in men and ≤95 g/m2 in women) was observed in 107 (16%) participants, while normal LV geometry (RWT ≤0.42; LV mass index ≤115 g/m2 in men and ≤95 g/m2 in women) was seen in 423 (63%), and LV hypertrophy (LV mass index >115 g/m2 in men and >95 g/m2 in women) in 145 (21%). Both low BNP level and higher insulin resistance were independently linked to LV concentric remodelling after multivariate adjustment (1 SD increment in BNP = aOR 0.714, 95% CI 0.544–0.938, P = 0.015; HOMA‐IR ≥ 1.37 vs. <1.37: aOR 1.694, 95% CI 1.004–2.857, P = 0.048, respectively). Conclusions Lower BNP levels are linked to either insulin resistance or LV concentric remodelling in a population with normal plasma BNP levels, suggesting that participants with lower natriuretic peptide level might be vulnerable to the development of metabolic disorders and LV morphological abnormalities.
Collapse
Affiliation(s)
- Chisato Okamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Osamu Tsukamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takuya Hasegawa
- Department of Cardiovascular Medicine, Garacia Hospital, Mino, Osaka, Japan
| | - Tatsuro Hitsumoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ken Matsuoka
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Makoto Amaki
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Hideaki Kanzaki
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Chisato Izumi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Shin Ito
- Department of Clinical Medicine and Development, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Masafumi Kitakaze
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Clinical Medicine and Development, National Cerebral and Cardiovascular Center, Osaka, Japan.,Hanwa Daini Senboku Hospital, Sakai, Osaka, 599-8271, Japan
| |
Collapse
|
15
|
Yamamoto M, Hanatani S, Araki S, Izumiya Y, Yamada T, Nakanishi N, Ishida T, Yamamura S, Kimura Y, Arima Y, Nakamura T, Takashio S, Yamamoto E, Sakamoto K, Kaikita K, Matsushita K, Morimoto S, Ito T, Tsujita K. HE4 Predicts Progressive Fibrosis and Cardiovascular Events in Patients With Dilated Cardiomyopathy. J Am Heart Assoc 2021; 10:e021069. [PMID: 34320813 PMCID: PMC8475713 DOI: 10.1161/jaha.120.021069] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Cardiac fibrosis plays a crucial role in the pathogenesis of dilated cardiomyopathy (DCM). HE4 (human epididymis protein 4) is a secretory protein expressed in activated fibroblasts that exacerbates tissue fibrosis. In the present study, we investigated the clinical utility of HE4 measurement in patients with DCM and its pathophysiological role in preclinical experiments in vivo and in vitro. Methods and Results We measured serum HE4 levels of 87 patients with DCM. Endomyocardial biopsy expressed severe fibrosis only in the high HE4 group (P<0.0001). Echocardiography showed that left ventricular end‐diastolic diameter tends to decrease over time (58±7.3 to 51±6.6 mm; P<0.0001) in the low HE4 group (<59.65 pmol/L [median value]). HE4 was significantly associated with risk reduction of mortality and cardiovascular hospitalization in multivariate Cox model. In vivo, HE4 was highly expressed in kidney and lung tissue of mouse, and scarcely expressed in heart. In genetically induced DCM mouse model, HE4 expression increased in kidney but not in heart and lung. In vitro, supernatant from HE4‐transfected human embryonic kidney 293T cells enhanced transdifferentiation of rat neonatal fibroblasts and increased expression of fibrosis‐related genes, and this was accompanied by the activation of extracellular signal‐regulated kinase signaling in cardiac fibroblasts. Treatment with an inhibitor of upstream signal of extracellular signal‐regulated kinase or a neutralizing HE4 antibody canceled the profibrotic properties of HE4. Conclusions HE4 functions as a secretory factor, activating cardiac fibroblasts, thereby inducing cardiac interstitial fibrosis. HE4 could be a promising biomarker for assessing ongoing fibrosis and a novel therapeutic target in DCM. Registration URL: https://upload.umin.ac.jp/cgi‐open‐bin/ctr; Unique identifier: UMIN000043062.
Collapse
Affiliation(s)
- Masahiro Yamamoto
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Shinsuke Hanatani
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Satoshi Araki
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Yasuhiro Izumiya
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan.,Department of Cardiovascular Medicine Osaka City University Graduate School of Medicine Osaka Japan
| | - Toshihiro Yamada
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Nobuhiro Nakanishi
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Toshifumi Ishida
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Satoru Yamamura
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Yuichi Kimura
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Yuichiro Arima
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan.,International Research Center for Medical Sciences Kumamoto University Kumamoto Japan
| | - Taishi Nakamura
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Seiji Takashio
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Kenji Sakamoto
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Koichi Kaikita
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Kenichi Matsushita
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Sachio Morimoto
- Department of Health Sciences Fukuoka International University of Health and Welfare Fukuoka Japan
| | - Takaaki Ito
- Department of Pathology and Experimental Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine Faculty of Life Sciences Kumamoto University Kumamoto Japan.,Center for Metabolic Regulation of Healthy Aging Kumamoto University Kumamoto Japan
| |
Collapse
|
16
|
Zhao X, Ren Y, Ren H, Wu Y, Liu X, Chen H, Ying C. The mechanism of myocardial fibrosis is ameliorated by myocardial infarction-associated transcript through the PI3K/Akt signaling pathway to relieve heart failure. J Int Med Res 2021; 49:3000605211031433. [PMID: 34275376 PMCID: PMC8293849 DOI: 10.1177/03000605211031433] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE This study aimed to investigate the role of long noncoding RNA (LncRNA) myocardial infarction-associated transcript (MIAT) in a heart failure (HF) model in vivo and in vitro by regulating the PI3K/Akt signaling pathway. METHODS We established HF models in vivo and in vitro and evaluated the collagen content of these models and other factors. RESULTS We found that when LncRNA MIAT was silenced, vascular endothelial growth factor, phosphorylated protein kinase B (Akt), and phosphorylated phosphoinositide 3-kinase (PI3K) mRNA and protein levels were significantly downregulated, which suggested that MIAT activated the PI3K/Akt signaling pathway. Akt and PI3K expression was not significantly changed. We also found that when LncRNA MIAT was silenced, collagen expression was significantly downregulated. This finding suggested that MIAT promoted myocardial fibrosis during the development of HF. The levels of inflammatory factors were also significantly reduced with silencing of LncRNA MIAT. This finding suggested that MIAT promoted the expression of inflammatory factors in myocardial fibrosis by activating the PI3K/Akt signaling pathway. CONCLUSION This study indicates that silencing LncRNA MIAT may improve myocardial fibrosis and alleviate HF through the PI3K/Akt signaling pathway, which may be helpful for patients with HF to obtain a better therapeutic effect.
Collapse
Affiliation(s)
- Xingsheng Zhao
- Cardiology Department, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yu Ren
- Cardiology Department, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Hongkun Ren
- Cardiology Department, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yun Wu
- Cardiology Department, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xi Liu
- Cardiology Department, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Hua Chen
- Cardiology Department, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| | - Chun Ying
- Cardiology Department, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia Autonomous Region, China
| |
Collapse
|
17
|
Abstract
Insulin receptors are highly expressed in the heart and vasculature. Insulin signaling regulates cardiac growth, survival, substrate uptake, utilization, and mitochondrial metabolism. Insulin signaling modulates the cardiac responses to physiological and pathological stressors. Altered insulin signaling in the heart may contribute to the pathophysiology of ventricular remodeling and heart failure progression. Myocardial insulin signaling adapts rapidly to changes in the systemic metabolic milieu. What may initially represent an adaptation to protect the heart from carbotoxicity may contribute to amplifying the risk of heart failure in obesity and diabetes. This review article presents the multiple roles of insulin signaling in cardiac physiology and pathology and discusses the potential therapeutic consequences of modulating myocardial insulin signaling.
Collapse
Affiliation(s)
- E Dale Abel
- Division of Endocrinology, Metabolism and Diabetes and Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
18
|
Kaze AD, Santhanam P, Erqou S, Bertoni AG, Ahima RS, Echouffo-Tcheugui JB. Long-term variability of blood pressure and incidence of heart failure among individuals with Type 2 diabetes. ESC Heart Fail 2021; 8:2959-2967. [PMID: 34032375 PMCID: PMC8318432 DOI: 10.1002/ehf2.13385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 01/01/2023] Open
Abstract
AIMS Data on the association of long-term variability of blood pressure (BP) with incident heart failure (HF) in individuals with Type 2 diabetes are scarce. We evaluated this association in a large community-based sample of adults with Type 2 diabetes. METHODS AND RESULTS A total of 4200 participants with Type 2 diabetes who had available BP measurements at four visits (baseline and 12, 24, and 36 months) in the Look AHEAD (Action for Health in Diabetes) study were included. Variability of systolic BP (SBP) and diastolic BP (DBP) across the four visits was assessed using four metrics. Participants free of HF during the first 36 months were followed for HF events. Cox regression was used to generate hazard ratios (HRs) and 95% confidence intervals (CIs) for HF. Of the 4200 participants, the average age was 59 years [standard deviation (SD): 6.8]; 58.5% were women. Over a median follow-up of 6.7 years, 129 developed HF events. After adjusting for relevant confounders, the HR of incident HF for the highest vs. lowest quartile of SD of SBP was 1.77 (95% CI 1.01-3.09); the HR for the highest (vs. lowest) quartile of variability independent of the mean of SBP was 1.29 (95% CI 0.78-2.14). The adjusted HR for participants in the highest (compared with the lowest) quartile of SD of DBP was 1.61 (95% CI 1.01-2.59), and the adjusted HR for variability independent of the mean of DBP was 1.65 (95% CI 1.03-2.65). CONCLUSIONS A greater variability in SBP and DBP is independently associated with greater risk of incident HF in individuals with Type 2 diabetes.
Collapse
Affiliation(s)
- Arnaud D Kaze
- Department of Medicine, University of Maryland Medical Center, Baltimore, MD, USA
| | - Prasanna Santhanam
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins School of Medicine, Baltimore, MD, 21224, USA
| | - Sebhat Erqou
- Department of Medicine, Providence VA Medical Center and Alpert Medical School of Brown University, Providence, RI, USA
| | - Alain G Bertoni
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Rexford S Ahima
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins School of Medicine, Baltimore, MD, 21224, USA
| | - Justin B Echouffo-Tcheugui
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins School of Medicine, Baltimore, MD, 21224, USA
| |
Collapse
|
19
|
Cole LK, Zhang M, Chen L, Sparagna GC, Vandel M, Xiang B, Dolinsky VW, Hatch GM. Supplemental Berberine in a High-Fat Diet Reduces Adiposity and Cardiac Dysfunction in Offspring of Mouse Dams with Gestational Diabetes Mellitus. J Nutr 2021; 151:892-901. [PMID: 33484149 DOI: 10.1093/jn/nxaa408] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND There are few evidence-based strategies to attenuate the risk of metabolic syndrome in offspring exposed to gestational diabetes mellitus (GDM). Berberine (BBR) is an isoquinoline alkaloid extracted from Chinese herbs and exhibits glucose lowering properties. OBJECTIVES We hypothesized that dietary BBR would improve health outcomes in the mouse offspring of GDM dams. METHODS Wild-type C57BL/6 female mice were fed either a Lean-inducing low-fat diet (L-LF,10% kcal fat, 35% kcal sucrose) or a GDM-inducing high-fat diet (GDM-HF, 45% kcal fat, 17.5% sucrose) for 6 wk prior to breeding with wild-type C57BL/6 male mice throughout pregnancy and the suckling period. The resulting Lean and GDM-exposed male and female offspring were randomly assigned an LF (10% kcal fat, 35% kcal sucrose), HF (45% kcal fat, 17.5% sucrose), or high-fat berberine (HFB) (45% kcal fat, 17.5% sucrose diet) containing BBR (160 mg/kg/d, HFB) at weaning for 12 wk. The main outcome was to evaluate the effects of BBR on obesity, pancreatic islet function, and cardiac contractility in GDM-exposed HF-fed offspring. Significance between measurements was determined using a 2 (gestational exposure) × 3 (diet) factorial design by a 2- way ANOVA using Tukey post-hoc analysis. RESULTS In the GDM-HF group, body weights were significantly increased (16%) compared with those in baseline (L-LF) animals (P < 0.05). Compared with the L-LF animals, the GDM-HF group had a reduction in pancreatic insulin glucose-stimulated insulin secretion (74%) and increased cardiac isovolumetric contraction time (IVCT; ∼150%) (P < 0.05). Compared with GDM-HF animals, the GDM-HFB group with the dietary addition of BBR had significantly reduced body weight (16%), increased glucose-stimulated insulin secretion from pancreatic islets (254%), and reduced systolic heart function (46% IVCT) (P < 0.05). CONCLUSIONS In a mouse model of GDM, dietary BBR treatment provided protection from obesity and the development of pancreatic islet and cardiac dysfunction.
Collapse
Affiliation(s)
- Laura K Cole
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Department of Pharmacology and Therapeutics, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun, China
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun, China
| | - Genevieve C Sparagna
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Center, Aurora, USA
| | - Marilyne Vandel
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Department of Pharmacology and Therapeutics, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Bo Xiang
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Department of Pharmacology and Therapeutics, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Vernon W Dolinsky
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Department of Pharmacology and Therapeutics, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Grant M Hatch
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Department of Pharmacology and Therapeutics, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Center for Research and Treatment of Atherosclerosis, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
20
|
Jozefczuk E, Szczepaniak P, Guzik TJ, Siedlinski M. Silencing of Sphingosine kinase 1 Affects Maturation Pathways in Mouse Neonatal Cardiomyocytes. Int J Mol Sci 2021; 22:ijms22073616. [PMID: 33807180 PMCID: PMC8037404 DOI: 10.3390/ijms22073616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 12/18/2022] Open
Abstract
Sphingosine kinase-1 (Sphk1) and its product, sphingosine-1-phosphate (S1P) are important regulators of cardiac growth and function. Numerous studies have reported that Sphk1/S1P signaling is essential for embryonic cardiac development and promotes pathological cardiac hypertrophy in adulthood. However, no studies have addressed the role of Sphk1 in postnatal cardiomyocyte (CM) development so far. The present study aimed to assess the molecular mechanism(s) by which Sphk1 silencing might influence CMs development and hypertrophy in vitro. Neonatal mouse CMs were transfected with siRNA against Sphk1 or negative control, and subsequently treated with 1 µM angiotensin II (AngII) or a control buffer for 24 h. The results of RNASeq analysis revealed that diminished expression of Sphk1 significantly accelerated neonatal CM maturation by inhibiting cell proliferation and inducing developmental pathways in the stress (AngII-induced) conditions. Importantly, similar effects were observed in the control conditions. Enhanced maturation of Sphk1-lacking CMs was further confirmed by the upregulation of the physiological hypertrophy-related signaling pathway involving Akt and downstream glycogen synthase kinase 3 beta (Gsk3β) downregulation. In summary, we demonstrated that the Sphk1 silencing in neonatal mouse CMs facilitated their postnatal maturation in both physiological and stress conditions.
Collapse
Affiliation(s)
- Ewelina Jozefczuk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland; (E.J.); (P.S.); (T.J.G.)
| | - Piotr Szczepaniak
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland; (E.J.); (P.S.); (T.J.G.)
| | - Tomasz Jan Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland; (E.J.); (P.S.); (T.J.G.)
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
| | - Mateusz Siedlinski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-121 Cracow, Poland; (E.J.); (P.S.); (T.J.G.)
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
- Correspondence:
| |
Collapse
|
21
|
Schnelle M, Sawyer I, Anilkumar N, Mohamed BA, Richards DA, Toischer K, Zhang M, Catibog N, Sawyer G, Mongue-Din H, Schröder K, Hasenfuss G, Shah AM. NADPH oxidase-4 promotes eccentric cardiac hypertrophy in response to volume overload. Cardiovasc Res 2021; 117:178-187. [PMID: 31821410 PMCID: PMC7797217 DOI: 10.1093/cvr/cvz331] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 11/13/2019] [Accepted: 12/07/2019] [Indexed: 12/15/2022] Open
Abstract
AIMS Chronic pressure or volume overload induce concentric vs. eccentric left ventricular (LV) remodelling, respectively. Previous studies suggest that distinct signalling pathways are involved in these responses. NADPH oxidase-4 (Nox4) is a reactive oxygen species-generating enzyme that can limit detrimental cardiac remodelling in response to pressure overload. This study aimed to assess its role in volume overload-induced remodelling. METHODS AND RESULTS We compared the responses to creation of an aortocaval fistula (Shunt) to induce volume overload in Nox4-null mice (Nox4-/-) vs. wild-type (WT) littermates. Induction of Shunt resulted in a significant increase in cardiac Nox4 mRNA and protein levels in WT mice as compared to Sham controls. Nox4-/- mice developed less eccentric LV remodelling than WT mice (echocardiographic relative wall thickness: 0.30 vs. 0.27, P < 0.05), with less LV hypertrophy at organ level (increase in LV weight/tibia length ratio of 25% vs. 43%, P < 0.01) and cellular level (cardiomyocyte cross-sectional area: 323 µm2 vs. 379 μm2, P < 0.01). LV ejection fraction, foetal gene expression, interstitial fibrosis, myocardial capillary density, and levels of myocyte apoptosis after Shunt were similar in the two genotypes. Myocardial phospho-Akt levels were increased after induction of Shunt in WT mice, whereas levels decreased in Nox4-/- mice (+29% vs. -21%, P < 0.05), associated with a higher level of phosphorylation of the S6 ribosomal protein (S6) and the eIF4E-binding protein 1 (4E-BP1) in WT compared to Nox4-/- mice. We identified that Akt activation in cardiac cells is augmented by Nox4 via a Src kinase-dependent inactivation of protein phosphatase 2A. CONCLUSION Endogenous Nox4 is required for the full development of eccentric cardiac hypertrophy and remodelling during chronic volume overload. Nox4-dependent activation of Akt and its downstream targets S6 and 4E-BP1 may be involved in this effect.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Apoptosis
- Arteriovenous Shunt, Surgical
- Cell Cycle Proteins/metabolism
- Cell Line
- Disease Models, Animal
- Fibrosis
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- NADPH Oxidase 2/genetics
- NADPH Oxidase 2/metabolism
- NADPH Oxidase 4/genetics
- NADPH Oxidase 4/metabolism
- Phosphorylation
- Protein Phosphatase 2/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Ribosomal Protein S6/metabolism
- Signal Transduction
- Ventricular Function, Left
- Ventricular Remodeling
- src-Family Kinases/metabolism
- Mice
Collapse
Affiliation(s)
- Moritz Schnelle
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- Institute for Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Iain Sawyer
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Narayana Anilkumar
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Belal A Mohamed
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Daniel A Richards
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Karl Toischer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Min Zhang
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Norman Catibog
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Greta Sawyer
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Héloïse Mongue-Din
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Ajay M Shah
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
22
|
Takano APC, Senger N, Barreto-Chaves MLM. The endocrinological component and signaling pathways associated to cardiac hypertrophy. Mol Cell Endocrinol 2020; 518:110972. [PMID: 32777452 DOI: 10.1016/j.mce.2020.110972] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 07/14/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023]
Abstract
Although myocardial growth corresponds to an adaptive response to maintain cardiac contractile function, the cardiac hypertrophy is a condition that occurs in many cardiovascular diseases and typically precedes the onset of heart failure. Different endocrine factors such as thyroid hormones, insulin, insulin-like growth factor 1 (IGF-1), angiotensin II (Ang II), endothelin (ET-1), catecholamines, estrogen, among others represent important stimuli to cardiomyocyte hypertrophy. Thus, numerous endocrine disorders manifested as changes in the local environment or multiple organ systems are especially important in the context of progression from cardiac hypertrophy to heart failure. Based on that information, this review summarizes experimental findings regarding the influence of such hormones upon signalling pathways associated with cardiac hypertrophy. Understanding mechanisms through which hormones differentially regulate cardiac hypertrophy could open ways to obtain therapeutic approaches that contribute to prevent or delay the onset of heart failure related to endocrine diseases.
Collapse
Affiliation(s)
| | - Nathalia Senger
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | | |
Collapse
|
23
|
Larsen TS, Jansen KM. Impact of Obesity-Related Inflammation on Cardiac Metabolism and Function. J Lipid Atheroscler 2020; 10:8-23. [PMID: 33537250 PMCID: PMC7838512 DOI: 10.12997/jla.2021.10.1.8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/10/2020] [Accepted: 10/04/2020] [Indexed: 12/11/2022] Open
Abstract
This review focuses on the role of adipose tissue in obese individuals in the development of metabolic diseases, and their consequences for metabolic and functional derangements in the heart. The general idea is that the expansion of adipocytes during the development of obesity gives rise to unhealthy adipose tissue, characterized by low-grade inflammation and the release of proinflammatory adipokines and fatty acids (FAs). This condition, in turn, causes systemic inflammation and elevated FA concentrations in the circulation, which links obesity to several pathologies, including impaired insulin signaling in cardiac muscle and a subsequent shift in myocardial substrate oxidation in favor of FAs and reduced cardiac efficiency. This review also argues that efforts to prevent obesity-related cardiometabolic disease should focus on anti-obesogenic strategies to restore normal adipose tissue metabolism.
Collapse
Affiliation(s)
- Terje S Larsen
- Department of Medical Biology, The Health Sciences Faculty, UiT The Arctic University of Norway, Tromsø, Norway
| | - Kirsten M Jansen
- Department of Medical Biology, The Health Sciences Faculty, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
24
|
Greenig M, Melville A, Huntley D, Isalan M, Mielcarek M. Cross-Sectional Transcriptional Analysis of the Aging Murine Heart. Front Mol Biosci 2020; 7:565530. [PMID: 33102519 PMCID: PMC7545256 DOI: 10.3389/fmolb.2020.565530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease accounts for millions of deaths each year and is currently the leading cause of mortality worldwide. The aging process is clearly linked to cardiovascular disease, however, the exact relationship between aging and heart function is not fully understood. Furthermore, a holistic view of cardiac aging, linking features of early life development to changes observed in old age, has not been synthesized. Here, we re-purpose RNA-sequencing data previously-collected by our group, investigating gene expression differences between wild-type mice of different age groups that represent key developmental milestones in the murine lifespan. DESeq2's generalized linear model was applied with two hypothesis testing approaches to identify differentially-expressed (DE) genes, both between pairs of age groups and across mice of all ages. Pairwise comparisons identified genes associated with specific age transitions, while comparisons across all age groups identified a large set of genes associated with the aging process more broadly. An unsupervised machine learning approach was then applied to extract common expression patterns from this set of age-associated genes. Sets of genes with both linear and non-linear expression trajectories were identified, suggesting that aging not only involves the activation of gene expression programs unique to different age groups, but also the re-activation of gene expression programs from earlier ages. Overall, we present a comprehensive transcriptomic analysis of cardiac gene expression patterns across the entirety of the murine lifespan.
Collapse
Affiliation(s)
- Matthew Greenig
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Andrew Melville
- Department of Mathematics, Imperial College London, London, United Kingdom
| | - Derek Huntley
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Mark Isalan
- Department of Life Sciences, Imperial College London, London, United Kingdom
- Imperial College Center for Synthetic Biology, Imperial College London, London, United Kingdom
| | - Michal Mielcarek
- Department of Life Sciences, Imperial College London, London, United Kingdom
- Imperial College Center for Synthetic Biology, Imperial College London, London, United Kingdom
| |
Collapse
|
25
|
Gitschlag BL, Tate AT, Patel MR. Nutrient status shapes selfish mitochondrial genome dynamics across different levels of selection. eLife 2020; 9:56686. [PMID: 32959778 PMCID: PMC7508553 DOI: 10.7554/elife.56686] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 08/17/2020] [Indexed: 12/23/2022] Open
Abstract
Cooperation and cheating are widespread evolutionary strategies. While cheating confers an advantage to individual entities within a group, competition between groups favors cooperation. Selfish or cheater mitochondrial DNA (mtDNA) proliferates within hosts while being selected against at the level of host fitness. How does environment shape cheater dynamics across different selection levels? Focusing on food availability, we address this question using heteroplasmic Caenorhabditis elegans. We find that the proliferation of selfish mtDNA within hosts depends on nutrient status stimulating mtDNA biogenesis in the developing germline. Interestingly, mtDNA biogenesis is not sufficient for this proliferation, which also requires the stress-response transcription factor FoxO/DAF-16. At the level of host fitness, FoxO/DAF-16 also prevents food scarcity from accelerating the selection against selfish mtDNA. This suggests that the ability to cope with nutrient stress can promote host tolerance of cheaters. Our study delineates environmental effects on selfish mtDNA dynamics at different levels of selection.
Collapse
Affiliation(s)
- Bryan L Gitschlag
- Department of Biological Sciences, Vanderbilt University, Nashville, United States
| | - Ann T Tate
- Department of Biological Sciences, Vanderbilt University, Nashville, United States
| | - Maulik R Patel
- Department of Biological Sciences, Vanderbilt University, Nashville, United States.,Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, United States.,Diabetes Research and Training Center, Vanderbilt University School of Medicine, Nashville, United States
| |
Collapse
|
26
|
Li S, Lei X, Xiao Z, Xia W, Lin C, Fu X, Fu J, Zhang L, Yu X. Dihydrotanshinone I Ameliorates Cardiac Hypertrophy in Diabetic Mice Induced by Chronic High-Fat Feeding. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20952607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Salvia miltiorrhiza Bge. (Danshen) is widely used to improve blood circulation and the dredge meridian in traditional Chinese medicine. In the present study, we evaluated the effects of dihydrotanshinone I (DHTS), a natural product from Danshen, on chronic high-fat feeding-induced cardiac remodeling and dysfunction. DHTS (25 mg/kg, intraperitoneal) did not affect blood glucose, insulin levels, and glucose intolerance. However, it alleviated diastolic dysfunction induced by the high-fat diet, as indicated by the increase in the ratio of peak early filling velocity to peak late filling velocity of the mitral and suppression of the extension of the isovolumic relaxation phase of the left ventricle. Further investigations revealed that DHTS ameliorated high-fat induced cardiac hypertrophy in mice and suppressed insulin-induced enlargement of cardiomyocytes in vitro. In neonatal cardiomyocytes, DHTS restored insulin-induced suppression of CCAAT/enhancer-binding protein beta-2 isoform (CEBPβ) and the phosphorylation of glycogen synthase kinase-3β (GSK3β) and extracellular signal-regulated kinase (ERK). Taken together, our results indicated that DHTS ameliorated cardiac hypertrophy and diastolic dysfunction in high-fat-fed mice, probably through the inhibition of insulin-induced suppression of CEBPβ and phosphorylation of GSK3β and ERK in cardiomyocytes.
Collapse
Affiliation(s)
- Songpei Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong, P. R. China
| | - Xueping Lei
- Key Laboratory of Molecular Target & Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong, P. R. China
| | - Zekuan Xiao
- Key Laboratory of Molecular Target & Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong, P. R. China
| | - Wenyi Xia
- Key Laboratory of Molecular Target & Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong, P. R. China
| | - Chaojin Lin
- Key Laboratory of Molecular Target & Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong, P. R. China
| | - Xiaomei Fu
- Key Laboratory of Molecular Target & Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong, P. R. China
| | - Jijun Fu
- Key Laboratory of Molecular Target & Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong, P. R. China
| | - Lingmin Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong, P. R. China
| | - Xiyong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology, The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangdong, P. R. China
| |
Collapse
|
27
|
Lin H, Li Y, Zhu H, Wang Q, Chen Z, Chen L, Zhu Y, Zheng C, Wang Y, Liao W, Bin J, Kitakaze M, Liao Y. Lansoprazole alleviates pressure overload-induced cardiac hypertrophy and heart failure in mice by blocking the activation of β-catenin. Cardiovasc Res 2020; 116:101-113. [PMID: 30689763 DOI: 10.1093/cvr/cvz016] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 12/12/2018] [Accepted: 01/17/2019] [Indexed: 02/06/2023] Open
Abstract
AIMS Proton pump inhibitors (PPIs) are widely used in patients receiving percutaneous coronary intervention to prevent gastric bleeding, but whether PPIs are beneficial for the heart is controversial. Here, we investigated the effects of lansoprazole on cardiac hypertrophy and heart failure, as well as the underlying mechanisms. METHODS AND RESULTS Adult male C57 mice were subjected to transverse aortic constriction (TAC) or sham surgery and then were treated with lansoprazole or vehicle for 5 weeks. In addition, cultured neonatal rat ventricular cardiomyocytes and fibroblasts were exposed to angiotensin II in the presence or absence of lansoprazole. At 5 weeks after TAC, the heart weight/body weight ratio was lower in lansoprazole-treated mice than in untreated mice, as was the lung weight/body weight ratio, while left ventricular (LV) fractional shortening and the maximum and minimum rates of change of the LV pressure were higher in lansoprazole-treated mice, along with less cardiac fibrosis. In cultured cardiomyocytes, lansoprazole inhibited angiotensin II-induced protein synthesis and hypertrophy, as well as inhibiting proliferation of fibroblasts. Lansoprazole decreased myocardial levels of phosphorylated Akt, phosphorylated glycogen synthase kinase 3β, and active β-catenin in TAC mice and in angiotensin II-stimulated cardiomyocytes. After overexpression of active β-catenin or knockdown of H+/K+-ATPase α-subunit, lansoprazole still significantly attenuated myocyte hypertrophy. CONCLUSION Lansoprazole inhibits cardiac remodelling by suppressing activation of the Akt/GSK3β/β-catenin pathway independent of H+/K+-ATPase inhibition, and these findings may provide a novel insight into the pharmacological effects of PPIs with regard to alleviation of cardiac remodelling.
Collapse
Affiliation(s)
- Hairuo Lin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Yang Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Hailin Zhu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Qiancheng Wang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Zhenhuan Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Lin Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Yingqi Zhu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Cankun Zheng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Yuegang Wang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Masafumi Kitakaze
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China.,Cardiovascular Division of the Department of Medicine, National Cerebral and Cardiovascular Center, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| |
Collapse
|
28
|
Packer M. Molecular, Cellular, and Clinical Evidence That Sodium-Glucose Cotransporter 2 Inhibitors Act as Neurohormonal Antagonists When Used for the Treatment of Chronic Heart Failure. J Am Heart Assoc 2020; 9:e016270. [PMID: 32791029 PMCID: PMC7660825 DOI: 10.1161/jaha.120.016270] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce the risk of cardiovascular death and hospitalization for heart failure in patients with chronic heart failure. Initially, these drugs were believed to have a profile similar to diuretics or hemodynamically active drugs, but they do not rapidly reduce natriuretic peptides or cardiac filling pressures, and they exert little early benefit on symptoms, exercise tolerance, quality of life, or signs of congestion. Clinically, the profile of SGLT2 inhibitors resembles that of neurohormonal antagonists, whose benefits emerge gradually during sustained therapy. In experimental models, SGLT2 inhibitors produce a characteristic pattern of cellular effects, which includes amelioration of oxidative stress, mitigation of mitochondrial dysfunction, attenuation of proinflammatory pathways, and a reduction in myocardial fibrosis. These cellular effects are similar to those produced by angiotensin converting enzyme inhibitors, β-blockers, mineralocorticoid receptor antagonists, and neprilysin inhibitors. At a molecular level, SGLT2 inhibitors induce transcriptional reprogramming of cardiomyocytes that closely mimics that seen during nutrient deprivation. This shift in signaling activates the housekeeping pathway of autophagy, which clears the cytosol of dangerous cytosolic constituents that are responsible for cellular stress, thereby ameliorating the development of cardiomyopathy. Interestingly, similar changes in cellular signaling and autophagic flux have been seen with inhibitors of the renin-angiotensin system, β-blockers, mineralocorticoid receptor antagonists, and neprilysin inhibitors. The striking parallelism of these molecular, cellular, and clinical profiles supports the premise that SGLT2 inhibitors should be regarded as neurohormonal antagonists when prescribed for the treatment of heart failure with a reduced ejection fraction.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular InstituteBaylor University Medical CenterDallasTX
- Imperial CollegeLondonUnited Kingdom
| |
Collapse
|
29
|
Hynynen H, Mutikainen M, Naumenko N, Shakirzyanova A, Tuomainen T, Tavi P. Short high-fat diet interferes with the physiological maturation of the late adolescent mouse heart. Physiol Rep 2020; 8:e14474. [PMID: 32643294 PMCID: PMC7343666 DOI: 10.14814/phy2.14474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/12/2020] [Accepted: 05/04/2020] [Indexed: 11/24/2022] Open
Abstract
Dietary fats are essential for cardiac function. The metabolites of fats known as fatty acids provide most of the energy for cardiac tissue, serve as building blocks for membranes and regulate important signaling cascades. Despite their importance, excess fat intake can cause cardiac dysfunction. The detrimental effects of high-fat diet (HFD) on cardiac health are widely investigated in long-term studies but the short-term effects of fats have not been thoroughly studied. To elucidate the near-term effects of a HFD on the growth and maturation of late adolescent heart we subjected 11-week-old mice to an 8-week long HFD (42% of calories from fat, 42% from carbohydrate, n = 8) or chow diet (12% of calories from fat, 66% from carbohydrate, n = 7) and assessed their effects on the heart in vivo and in vitro. Our results showed that excessive fat feeding interferes with normal maturation of the heart indicated by the lack of increase in dimensions, volume, and stroke volume of the left ventricles of mice on high fat diet that were evident in mice on chow diet. In addition, differences in regional strain during the contraction cycle between mice on HFD and chow diet were seen. These changes were associated with reduced activity of the growth promoting PI3K-Akt1 signaling cascade and moderate changes in glucose metabolism without changes in calcium signaling. This study suggests that even a short period of HFD during late adolescence hinders cardiac maturation and causes physiological changes that may have an impact on the cardiac health in adulthood.
Collapse
Affiliation(s)
- Heidi Hynynen
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Maija Mutikainen
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Nikolay Naumenko
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | | | - Tomi Tuomainen
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Pasi Tavi
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| |
Collapse
|
30
|
Rodrigues MS, Pieri BLDS, Silveira GDB, Zaccaron RP, Venturini LM, Comin VH, Luiz KD, Silveira PCL. Reduction of oxidative stress improves insulin signaling in cardiac tissue of obese mice. EINSTEIN-SAO PAULO 2020; 18:eAO5022. [PMID: 32215468 PMCID: PMC7069732 DOI: 10.31744/einstein_journal/2020ao5022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 10/23/2019] [Indexed: 01/14/2023] Open
Abstract
Objective To evaluate the effects of oxidative stress on insulin signaling in cardiac tissue of obese mice. Methods Thirty Swiss mice were equally divided (n=10) into three groups: Control Group, Obese Group, and Obese Group Treated with N-acetylcysteine. After obesity and insulin resistance were established, the obese mice were treated with N-acetylcysteine at a dose of 50mg/kg daily for 15 days via oral gavage. Results Higher blood glucose levels and nitrite and carbonyl contents, and lower protein levels of glutathione peroxidase and phosphorylated protein kinase B were observed in the obese group when compared with their respective control. On the other hand, treatment with N-acetylcysteine was effective in reducing blood glucose levels and nitrite and carbonyl contents, and significantly increased protein levels of glutathione peroxidase and phosphorylated protein kinase B compared to the Obese Group. Conclusion Obesity and/or a high-lipid diet may result in oxidative stress and insulin resistance in the heart tissue of obese mice, and the use of N-acetylcysteine as a methodological and therapeutic strategy suggested there is a relation between them.
Collapse
Affiliation(s)
| | | | | | | | | | - Vitor Hugo Comin
- Universidade do Extremo Sul Catarinense , Criciúma , SC , Brazil
| | | | | |
Collapse
|
31
|
Martins Matias A, Murucci Coelho P, Bermond Marques V, dos Santos L, Monteiro de Assis ALE, Valentim Nogueira B, Lima-Leopoldo AP, Soares Leopoldo A. Hypercaloric diet models do not develop heart failure, but the excess sucrose promotes contractility dysfunction. PLoS One 2020; 15:e0228860. [PMID: 32032383 PMCID: PMC7006916 DOI: 10.1371/journal.pone.0228860] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/25/2020] [Indexed: 12/04/2022] Open
Abstract
Several diseases are associated with excess of adipose tissue, and obesity is considered an independent risk factor for the development of cardiac remodeling and heart failure. Dietary aspects have been studied to elucidate the mechanisms involved in these processes. Thus, the purpose was the development and characterization of an obesity experimental model from hypercaloric diets, which resulted in cardiac remodeling and predisposition to heart failure. Thirty- day-old male Wistar rats (n = 52) were randomized into four groups: control (C), high sucrose (HS), high-fat (HF) and high-fat and sucrose (HFHS) for 20 weeks. General characteristics, comorbidities, weights of the heart, left (LV) and right ventricles, atrium, and relationships with the tibia length were evaluated. The LV myocyte cross sectional area and fraction of interstitial collagen were assayed. Cardiac function was determined by hemodynamic analysis and the contractility by cardiomyocyte contractile function. Heart failure was analyzed by pulmonary congestion, right ventricular hypertrophy, and hemodynamic parameters. HF and HFHS models led to obesity by increase in adiposity index (C = 8.3 ± 0.2% vs. HF = 10.9 ± 0.5%, HFHS = 10.2 ± 0.3%). There was no change in the morphological parameters and heart failure signals. HF and HFHS caused a reduction in times to 50% relaxation without cardiomyocyte contractile damage. The HS model presented cardiomyocyte contractile dysfunction visualized by lower shortening (C: 8.34 ± 0.32% vs. HS: 6.91 ± 0.28), as well as the Ca2+ transient amplitude was also increased when compared to HFHS. In conclusion, the experimental diets based on high amounts of sugar, lard or a combination of both did not promote cardiac remodeling with predisposition to heart failure under conditions of obesity or excess sucrose. Nevertheless, excess sucrose causes cardiomyocyte contractility dysfunction associated with alterations in the myocyte sensitivity to intracellular Ca2+.
Collapse
Affiliation(s)
- Amanda Martins Matias
- Postgraduate Program in Nutrition and Health, Center of Health Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Priscila Murucci Coelho
- Postgraduate Program in Nutrition and Health, Center of Health Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Vinícius Bermond Marques
- Center of Health Sciences, Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Leonardo dos Santos
- Center of Health Sciences, Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | | | - Breno Valentim Nogueira
- Center of Health Sciences, Department of Morphology, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Ana Paula Lima-Leopoldo
- Postgraduate Program in Nutrition and Health, Center of Health Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
- Department of Sports, Center of Physical Education and Sports, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - André Soares Leopoldo
- Postgraduate Program in Nutrition and Health, Center of Health Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
- Department of Sports, Center of Physical Education and Sports, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| |
Collapse
|
32
|
Hao Q, Zhang F, Wang Y, Li Y, Qi X. Cardiac Contractility Modulation Attenuates Chronic Heart Failure in a Rabbit Model via the PI3K/AKT Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1625362. [PMID: 31998779 PMCID: PMC6973194 DOI: 10.1155/2020/1625362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/27/2019] [Indexed: 01/01/2023]
Abstract
The Akt plays an important role in regulating cardiac growth, myocardial angiogenesis, and cell death in cardiac myocytes. However, there are few studies to focus on the responses of the Akt pathway to cardiac contractility modulation (CCM) in a chronic heart failure (HF) model. In this study, the effects of CCM on the treatment of HF in a rabbit model were investigated. Thirty six-month-old rabbits were randomly separated into control, HF, and CCM groups. The rabbits in HF and CCM groups were pressure uploaded, which can cause an aortic constriction. Then, CCM was gradually injected to the myocardium of rabbits in the CCM group, and this process lasted for four weeks with six hours per day. Rabbit body weight, heart weight, and heart beating rates were recorded during the experiment. To assess the CCM impacts, rabbit myocardial histology was examined as well. Additionally, western blot analysis was employed to measure the protein levels of Akt, FOXO3, Beclin, Pi3k, mTOR, GSK-3β, and TORC2 in the myocardial histology of rabbits. Results showed that the body and heart weight of rabbits decreased significantly after suffering HF when compared with those in the control group. However, they gradually recovered after CCM application. The CCM significantly decreased collagen volume fraction in myocardial histology of HF rabbits, indicating that CCM therapy attenuated myocardial fibrosis and collagen deposition. The levels of Akt, FOXO3, Beclin, mTOR, GSK-3β, and TORC2 were significantly downregulated, but Pi3k concentration was greatly upregulated after CCM utilization. Based on these findings, it was concluded that CCM could elicit positive effects on HF therapy, which was potentially due to the variation in the Pi3k/Akt signaling pathway.
Collapse
Affiliation(s)
- Qingqing Hao
- School of Graduate, Hebei Medical University, Shijiazhuang, China
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Feifei Zhang
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Yudan Wang
- School of Graduate, Hebei Medical University, Shijiazhuang, China
| | - Yingxiao Li
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| | - Xiaoyong Qi
- School of Graduate, Hebei Medical University, Shijiazhuang, China
- Department of Cardiology Center, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
33
|
Liu J, Liu X, Hui X, Cai L, Li X, Yang Y, Shu S, Wang F, Xia H, Li S. Novel Role for Pleckstrin Homology-Like Domain Family A, Member 3 in the Regulation of Pathological Cardiac Hypertrophy. J Am Heart Assoc 2019; 8:e011830. [PMID: 31426686 PMCID: PMC6759890 DOI: 10.1161/jaha.118.011830] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background Pleckstrin homology-like domain family A, member 3 (PHLDA3), a crucial member of the PHLDA family, is involved in tumor suppression, kidney injury, liver injury, and glucose metabolism. However, the role of PHLDA3 in pathological cardiac hypertrophy and heart failure remains unclear. Methods and Results In the present study, PHLDA3 expression was downregulated in hypertrophic murine hearts and angiotensin II-treated cardiomyocytes. Next, an in vitro study suggested, by using gain- and loss-of-function approaches, that PHLDA3 attenuates Ang II exposure-induced cardiomyocyte hypertrophy. Consistent with the cell phenotype, disruption of PHLDA3 aggravated the effects of pressure overload-induced pathological cardiac hypertrophy, fibrosis, and dysfunction. In contrast, PHLDA3 overexpression resulted in an attenuated hypertrophic phenotype. Molecular analysis revealed that PHLDA3 suppressed the activation of AKT-mTOR-GSK3β-P70S6K signaling in response to hypertrophic stress, and the blockage of AKT activation rescued these adverse pathological effects of PHLDA3 deficiency-induced by AB and Ang II, respectively, in vivo and in vitro. Conclusions Collectively, our data indicated that PHLDA3 could ameliorate pressure overload-induced cardiac remodeling mainly by blocking the AKT signaling pathway, suggesting that PHLDA3 may represent a therapeutic target for the treatment of pathological cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Jia Liu
- Department of Cardiology First Hospital of Jilin University Changchun Jilin China.,Department of Cardiology Cang Zhou People's Hospital Cangzhou Hebei China
| | - Xiaoxiong Liu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China.,Cardiovascular Research Institute Wuhan University Wuhan China.,Hubei Key Laboratory of Cardiology Wuhan China
| | - Xuejun Hui
- Department of Cardiology Second Hospital of Jilin University Changchun Jilin China
| | - Lin Cai
- Zhongnan Hospital of Wuhan University Wuhan China.,Institute of Model Animal of Wuhan University Wuhan China
| | - Xuebo Li
- Department of Cardiology First Hospital of Jilin University Changchun Jilin China
| | - Yang Yang
- Department of Cardiology First Hospital of Jilin University Changchun Jilin China
| | - Shangzhi Shu
- Department of Cardiology First Hospital of Jilin University Changchun Jilin China
| | - Fan Wang
- Department of Cardiology First Hospital of Jilin University Changchun Jilin China
| | - Hao Xia
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China.,Cardiovascular Research Institute Wuhan University Wuhan China.,Hubei Key Laboratory of Cardiology Wuhan China
| | - Shuyan Li
- Department of Cardiology First Hospital of Jilin University Changchun Jilin China
| |
Collapse
|
34
|
Parsanathan R, Maria Joseph A, Karundevi B. Postnatal exposure to di-(2-ethylhexyl)phthalate alters cardiac insulin signaling molecules and GLUT4 Ser488 phosphorylation in male rat offspring. J Cell Biochem 2018; 120:5802-5812. [PMID: 30362281 DOI: 10.1002/jcb.27866] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/19/2018] [Indexed: 01/16/2023]
Abstract
Di-(2-ethylhexyl)phthalate (DEHP), a distinctive endocrine-disrupting chemical, is widely used as a plasticizer in a variety of consumer products. It can easily cross the placenta and enter breast milk and then it is rapidly absorbed by offspring. Since it is generally accepted that individuals are more sensitive to chemical exposure during vital developmental periods, we investigated whether DEHP exposure during lactation affects cardiac insulin signaling and glucose homeostasis in the F1 male rat offspring at postnatal day 22 (PND22). Lactating Wistar rats were administered with DEHP (1, 10, and 100 mg/kg/d) or olive oil from lactation day 1 to 21 by oral gavage. All the male pups were perfused and killed on PND22. On the day before the killing, they were kept for fasting overnight and blood was collected. The cardiac muscle was dissected out, washed in ice-cold physiological saline repeatedly and used for the assay of various parameters. DEHP-exposed offspring had significantly lower body weight than the control. DEHP-exposed offspring showed elevated blood glucose, decreased 14 C-2-deoxyglucose uptake and 14 C-glucose oxidation in cardiac muscle at PND22. The concentration of upstream insulin signaling molecules such as insulin receptor subunit β (InsRβ) and insulin receptor substrate 1 (IRS1) were downregulated in DEHP-exposed offspring. However, no significant alterations were observed in protein kinase B (Akt) and Akt substrate of 160 kDa (AS160). Surprisingly, phosphorylation of IRS1 Tyr632 and Akt Ser473 were diminished. Low levels of glucose transporter type 4 (GLUT4) protein and increased GLUT4 Ser488 phosphorylation which decreases its intrinsic activity and translocation towards plasma membrane were also recorded. Lactational DEHP exposure predisposes F 1 male offspring to cardiac glucometabolic disorders at PND22, which may impair cardiac function.
Collapse
Affiliation(s)
- Rajesh Parsanathan
- Department of Endocrinology, Dr ALM Post Graduate Institute of Basic Medical Sciences University of Madras, Taramani, India
| | - Angelaalincy Maria Joseph
- Department of Endocrinology, Dr ALM Post Graduate Institute of Basic Medical Sciences University of Madras, Taramani, India
| | - Balasubramanian Karundevi
- Department of Endocrinology, Dr ALM Post Graduate Institute of Basic Medical Sciences University of Madras, Taramani, India
| |
Collapse
|
35
|
Ceylan AF, Wang S, Kandadi MR, Chen J, Hua Y, Pei Z, Nair S, Ren J. Cardiomyocyte-specific knockout of endothelin receptor a attenuates obesity cardiomyopathy. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3339-3352. [DOI: 10.1016/j.bbadis.2018.07.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 07/14/2018] [Accepted: 07/16/2018] [Indexed: 12/20/2022]
|
36
|
Uchinaka A, Azuma N, Mizumoto H, Nakano S, Minamiya M, Yoneda M, Aoyama K, Komatsu Y, Yamada Y, Murohara T, Nagata K. Anti-inflammatory effects of heat-killed Lactobacillus plantarum L-137 on cardiac and adipose tissue in rats with metabolic syndrome. Sci Rep 2018; 8:8156. [PMID: 29802339 PMCID: PMC5970162 DOI: 10.1038/s41598-018-26588-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 05/15/2018] [Indexed: 12/17/2022] Open
Abstract
The effects of heat-killed Lactobacillus plantarum L-137 (HK L-137) on chronic inflammation associated with metabolic disorders have remained unknown. We examined the effects of HK L-137 on cardiac and adipose tissue pathophysiology in DahlS.Z-Leprfa/Leprfa (DS/obese) rats as a model of metabolic syndrome. DS/obese rats were treated orally with HK L-137 (2 or 75 mg kg−1 day−1) from 9 to 13 weeks of age. HK L-137 attenuated left ventricular (LV) inflammation and fibrosis as well as adipocyte hypertrophy, inflammation, and up-regulation of sterol regulatory element–binding protein–1c (SREBP-1c) gene expression in visceral and subcutaneous adipose tissue, without affecting body weight gain or hypertension. The low dose of HK L-137 also ameliorated LV diastolic dysfunction, the increase in subcutaneous fat mass, and insulin resistance as well as attenuated the down-regulation of Akt phosphorylation in visceral and subcutaneous adipose tissue, and the elevation of the circulating interleukin-6 concentration. Furthermore, the proportion of regulatory T (Treg) cells among CD4+ T cells in the spleen was increased by HK L-137. These results suggest that the anti-inflammatory effects of HK L-137 on the heart and adipose tissue are related, at least partly, to suppression of systemic inflammation associated with an increase in splenic Treg cell.
Collapse
Affiliation(s)
- Ayako Uchinaka
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoki Azuma
- Department of Medical Technology, Nagoya University School of Health Sciences, Nagoya, Japan
| | - Hisashi Mizumoto
- Department of Medical Technology, Nagoya University School of Health Sciences, Nagoya, Japan
| | - Shiho Nakano
- Department of Medical Technology, Nagoya University School of Health Sciences, Nagoya, Japan
| | - Moeko Minamiya
- Department of Medical Technology, Nagoya University School of Health Sciences, Nagoya, Japan
| | - Mamoru Yoneda
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kiyoshi Aoyama
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Komatsu
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichiro Yamada
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kohzo Nagata
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
37
|
Kimura A, Ishida Y, Furuta M, Nosaka M, Kuninaka Y, Taruya A, Mukaida N, Kondo T. Protective Roles of Interferon-γ in Cardiac Hypertrophy Induced by Sustained Pressure Overload. J Am Heart Assoc 2018; 7:e008145. [PMID: 29555642 PMCID: PMC5907566 DOI: 10.1161/jaha.117.008145] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 02/14/2018] [Indexed: 01/18/2023]
Abstract
BACKGROUND A clear understanding of the molecular mechanisms underlying hemodynamic stress-initiated cardiac hypertrophy is important for preventing heart failure. Interferon-γ (IFN-γ) has been suggested to play crucial roles in various diseases other than immunological disorders by modulating the expression of myriad genes. However, the involvement of IFN-γ in the pathogenesis of cardiac hypertrophy still remains unclear. METHODS AND RESULTS In order to elucidate the roles of IFN-γ in pressure overload-induced cardiac pathology, we subjected Balb/c wild-type (WT) or IFN-γ-deficient (Ifng-/-) mice to transverse aortic constriction (TAC). Three weeks after TAC, Ifng-/- mice developed more severe cardiac hypertrophy, fibrosis, and dysfunction than WT mice. Bone marrow-derived immune cells including macrophages were a source of IFN-γ in hearts after TAC. The activation of PI3K/Akt signaling, a key signaling pathway in compensatory hypertrophy, was detected 3 days after TAC in the left ventricles of WT mice and was markedly attenuated in Ifng-/- mice. The administration of a neutralizing anti-IFN-γ antibody abrogated PI3K/Akt signal activation in WT mice during compensatory hypertrophy, while that of IFN-γ activated PI3K/Akt signaling in Ifng-/- mice. TAC also induced the phosphorylation of Stat5, but not Stat1 in the left ventricles of WT mice 3 days after TAC. Furthermore, IFN-γ induced Stat5 and Akt phosphorylation in rat cardiomyocytes cultured under stretch conditions. A Stat5 inhibitor significantly suppressed PI3K/Akt signaling activation in the left ventricles of WT mice, and aggravated pressure overload-induced cardiac hypertrophy. CONCLUSIONS The IFN-γ/Stat5 axis may be protective against persistent pressure overload-induced cardiac hypertrophy by activating the PI3K/Akt pathway.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Disease Models, Animal
- Fibrosis
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Male
- Mice, Inbred BALB C
- Mice, Knockout
- Myocytes, Cardiac/metabolism
- Phosphatidylinositol 3-Kinase/metabolism
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Rats, Sprague-Dawley
- Receptors, Interferon/genetics
- Receptors, Interferon/metabolism
- STAT5 Transcription Factor/metabolism
- Signal Transduction
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/prevention & control
- Ventricular Function, Left
- Ventricular Remodeling
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Machi Furuta
- Department of Clinical Laboratory Medicine, Wakayama Medical University, Wakayama, Japan
| | - Mizuho Nosaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yumi Kuninaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Akira Taruya
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute Kanazawa University, Kanazawa, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
38
|
Gibb AA, Epstein PN, Uchida S, Zheng Y, McNally LA, Obal D, Katragadda K, Trainor P, Conklin DJ, Brittian KR, Tseng MT, Wang J, Jones SP, Bhatnagar A, Hill BG. Exercise-Induced Changes in Glucose Metabolism Promote Physiological Cardiac Growth. Circulation 2017; 136:2144-2157. [PMID: 28860122 PMCID: PMC5704654 DOI: 10.1161/circulationaha.117.028274] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/25/2017] [Indexed: 12/15/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Exercise promotes metabolic remodeling in the heart, which is associated with physiological cardiac growth; however, it is not known whether or how physical activity–induced changes in cardiac metabolism cause myocardial remodeling. In this study, we tested whether exercise-mediated changes in cardiomyocyte glucose metabolism are important for physiological cardiac growth. Methods: We used radiometric, immunologic, metabolomic, and biochemical assays to measure changes in myocardial glucose metabolism in mice subjected to acute and chronic treadmill exercise. To assess the relevance of changes in glycolytic activity, we determined how cardiac-specific expression of mutant forms of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase affect cardiac structure, function, metabolism, and gene programs relevant to cardiac remodeling. Metabolomic and transcriptomic screenings were used to identify metabolic pathways and gene sets regulated by glycolytic activity in the heart. Results: Exercise acutely decreased glucose utilization via glycolysis by modulating circulating substrates and reducing phosphofructokinase activity; however, in the recovered state following exercise adaptation, there was an increase in myocardial phosphofructokinase activity and glycolysis. In mice, cardiac-specific expression of a kinase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase transgene (GlycoLo mice) lowered glycolytic rate and regulated the expression of genes known to promote cardiac growth. Hearts of GlycoLo mice had larger myocytes, enhanced cardiac function, and higher capillary-to-myocyte ratios. Expression of phosphatase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase in the heart (GlycoHi mice) increased glucose utilization and promoted a more pathological form of hypertrophy devoid of transcriptional activation of the physiological cardiac growth program. Modulation of phosphofructokinase activity was sufficient to regulate the glucose–fatty acid cycle in the heart; however, metabolic inflexibility caused by invariantly low or high phosphofructokinase activity caused modest mitochondrial damage. Transcriptomic analyses showed that glycolysis regulates the expression of key genes involved in cardiac metabolism and remodeling. Conclusions: Exercise-induced decreases in glycolytic activity stimulate physiological cardiac remodeling, and metabolic flexibility is important for maintaining mitochondrial health in the heart.
Collapse
Affiliation(s)
- Andrew A Gibb
- Institute of Molecular Cardiology (A.A.G., Y.Z., L.A.M., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.).,Diabetes and Obesity Center (A.A.G., Y.Z., L.A.M., D.O., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.).,Department of Physiology (A.A.G., B.G.H.)
| | | | | | - Yuting Zheng
- Institute of Molecular Cardiology (A.A.G., Y.Z., L.A.M., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.).,Diabetes and Obesity Center (A.A.G., Y.Z., L.A.M., D.O., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.)
| | - Lindsey A McNally
- Institute of Molecular Cardiology (A.A.G., Y.Z., L.A.M., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.).,Diabetes and Obesity Center (A.A.G., Y.Z., L.A.M., D.O., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.)
| | - Detlef Obal
- Diabetes and Obesity Center (A.A.G., Y.Z., L.A.M., D.O., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.).,Department of Anesthesiology (D.O.)
| | - Kartik Katragadda
- Institute of Molecular Cardiology (A.A.G., Y.Z., L.A.M., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.).,Diabetes and Obesity Center (A.A.G., Y.Z., L.A.M., D.O., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.)
| | - Patrick Trainor
- Institute of Molecular Cardiology (A.A.G., Y.Z., L.A.M., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.).,Diabetes and Obesity Center (A.A.G., Y.Z., L.A.M., D.O., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.)
| | - Daniel J Conklin
- Institute of Molecular Cardiology (A.A.G., Y.Z., L.A.M., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.).,Diabetes and Obesity Center (A.A.G., Y.Z., L.A.M., D.O., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.)
| | - Kenneth R Brittian
- Institute of Molecular Cardiology (A.A.G., Y.Z., L.A.M., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.).,Diabetes and Obesity Center (A.A.G., Y.Z., L.A.M., D.O., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.)
| | | | - Jianxun Wang
- University of Louisville, KY. Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (J.W.)
| | - Steven P Jones
- Institute of Molecular Cardiology (A.A.G., Y.Z., L.A.M., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.).,Diabetes and Obesity Center (A.A.G., Y.Z., L.A.M., D.O., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.)
| | - Aruni Bhatnagar
- Institute of Molecular Cardiology (A.A.G., Y.Z., L.A.M., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.).,Diabetes and Obesity Center (A.A.G., Y.Z., L.A.M., D.O., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.)
| | - Bradford G Hill
- Institute of Molecular Cardiology (A.A.G., Y.Z., L.A.M., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.) .,Diabetes and Obesity Center (A.A.G., Y.Z., L.A.M., D.O., K.K., P.T., D.J.C., K.R.B., S.P.J., A.B., B.G.H.).,Department of Physiology (A.A.G., B.G.H.)
| |
Collapse
|
39
|
Abdul-Ghani M, Suen C, Jiang B, Deng Y, Weldrick JJ, Putinski C, Brunette S, Fernando P, Lee TT, Flynn P, Leenen FHH, Burgon PG, Stewart DJ, Megeney LA. Cardiotrophin 1 stimulates beneficial myogenic and vascular remodeling of the heart. Cell Res 2017; 27:1195-1215. [PMID: 28785017 PMCID: PMC5630684 DOI: 10.1038/cr.2017.87] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/06/2017] [Accepted: 06/21/2017] [Indexed: 12/12/2022] Open
Abstract
The post-natal heart adapts to stress and overload through hypertrophic growth, a process that may be pathologic or beneficial (physiologic hypertrophy). Physiologic hypertrophy improves cardiac performance in both healthy and diseased individuals, yet the mechanisms that propagate this favorable adaptation remain poorly defined. We identify the cytokine cardiotrophin 1 (CT1) as a factor capable of recapitulating the key features of physiologic growth of the heart including transient and reversible hypertrophy of the myocardium, and stimulation of cardiomyocyte-derived angiogenic signals leading to increased vascularity. The capacity of CT1 to induce physiologic hypertrophy originates from a CK2-mediated restraining of caspase activation, preventing the transition to unrestrained pathologic growth. Exogenous CT1 protein delivery attenuated pathology and restored contractile function in a severe model of right heart failure, suggesting a novel treatment option for this intractable cardiac disease.
Collapse
Affiliation(s)
- Mohammad Abdul-Ghani
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Colin Suen
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Baohua Jiang
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada
| | - Yupu Deng
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada
| | - Jonathan J Weldrick
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Charis Putinski
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Steve Brunette
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada
| | - Pasan Fernando
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Biology, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Tom T Lee
- Fate Therapeutics Inc., 3535 General Atomics Court Suite 200, San Diego, CA 92121, USA
| | - Peter Flynn
- Fate Therapeutics Inc., 3535 General Atomics Court Suite 200, San Diego, CA 92121, USA
| | - Frans H H Leenen
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Patrick G Burgon
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Duncan J Stewart
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Lynn A Megeney
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada.,Department of Medicine (Cardiology), Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
40
|
Guo CA, Guo S. Insulin receptor substrate signaling controls cardiac energy metabolism and heart failure. J Endocrinol 2017; 233:R131-R143. [PMID: 28381504 PMCID: PMC9675292 DOI: 10.1530/joe-16-0679] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/05/2017] [Indexed: 12/11/2022]
Abstract
The heart is an insulin-dependent and energy-consuming organ in which insulin and nutritional signaling integrates to the regulation of cardiac metabolism, growth and survival. Heart failure is highly associated with insulin resistance, and heart failure patients suffer from the cardiac energy deficiency and structural and functional dysfunction. Chronic pathological conditions, such as obesity and type 2 diabetes mellitus, involve various mechanisms in promoting heart failure by remodeling metabolic pathways, modulating cardiac energetics and impairing cardiac contractility. Recent studies demonstrated that insulin receptor substrates 1 and 2 (IRS-1,-2) are major mediators of both insulin and insulin-like growth factor-1 (IGF-1) signaling responsible for myocardial energetics, structure, function and organismal survival. Importantly, the insulin receptor substrates (IRS) play an important role in the activation of the phosphatidylinositide-3-dependent kinase (PI-3K) that controls Akt and Foxo1 signaling cascade, regulating the mitochondrial function, cardiac energy metabolism and the renin-angiotensin system. Dysregulation of this branch in signaling cascades by insulin resistance in the heart through the endocrine system promotes heart failure, providing a novel mechanism for diabetic cardiomyopathy. Therefore, targeting this branch of IRS→PI-3K→Foxo1 signaling cascade and associated pathways may provide a fundamental strategy for the therapeutic and nutritional development in control of metabolic and cardiovascular diseases. In this review, we focus on insulin signaling and resistance in the heart and the role energetics play in cardiac metabolism, structure and function.
Collapse
Affiliation(s)
- Cathy A Guo
- Department of Nutrition and Food ScienceCollege of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Shaodong Guo
- Department of Nutrition and Food ScienceCollege of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
41
|
Guan XH, Hong X, Zhao N, Liu XH, Xiao YF, Chen TT, Deng LB, Wang XL, Wang JB, Ji GJ, Fu M, Deng KY, Xin HB. CD38 promotes angiotensin II-induced cardiac hypertrophy. J Cell Mol Med 2017; 21:1492-1502. [PMID: 28296029 PMCID: PMC5542907 DOI: 10.1111/jcmm.13076] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 11/29/2016] [Indexed: 12/17/2022] Open
Abstract
Cardiac hypertrophy is an early hallmark during the clinical course of heart failure and regulated by various signalling pathways. Recently, we observed that mouse embryonic fibroblasts from CD38 knockout mice were significantly resistant to oxidative stress such as H2O2‐induced injury and hypoxia/reoxygenation‐induced injury. In addition, we also found that CD38 knockout mice protected heart from ischaemia reperfusion injury through activating SIRT1/FOXOs‐mediated antioxidative stress pathway. However, the role of CD38 in cardiac hypertrophy is not explored. Here, we investigated the roles and mechanisms of CD38 in angiotensin II (Ang‐II)‐induced cardiac hypertrophy. Following 14 days of Ang‐II infusion with osmotic mini‐pumps, a comparable hypertension was generated in both of CD38 knockout and wild‐type mice. However, the cardiac hypertrophy and fibrosis were much more severe in wild‐type mice compared with CD38 knockout mice. Consistently, RNAi‐induced knockdown of CD38 decreased the gene expressions of atrial natriuretic factor (ANF) and brain natriuretic peptide (BNP) and reactive oxygen species generation in Ang‐II‐stimulated H9c2 cells. In addition, the expression of SIRT3 was elevated in CD38 knockdown H9c2 cells, in which SIRT3 may further activate the FOXO3 antioxidant pathway. The intracellular Ca2+ release induced by Ang‐II markedly decreased in CD38 knockdown H9c2 cells, which might be associated with the decrease of nuclear translocation of NFATc4 and inhibition of ERK/AKT phosphorylation. We concluded that CD38 plays an essential role in cardiac hypertrophy probably via inhibition of SIRT3 expression and activation of Ca2+‐NFAT signalling pathway. Thus, CD38 may be a novel target for treating cardiac hypertrophy.
Collapse
Affiliation(s)
- Xiao-Hui Guan
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Xuan Hong
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Ning Zhao
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Xiao-Hong Liu
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Yun-Fei Xiao
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Ting-Tao Chen
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Li-Bin Deng
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Xiao-Lei Wang
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Jian-Bin Wang
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Guang-Ju Ji
- National Laboratory of Biomacromolecules, Institute of Biophysics Chinese Academy of Sciences, Beijing, China
| | - Mingui Fu
- Department of Basic Medical Science, Shock/Trauma Research Center, School of Medicine, University of Missouri Kansas City, Kansas City, MO, USA
| | - Ke-Yu Deng
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Hong-Bo Xin
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
42
|
Juni RP, Abreu RC, da Costa Martins PA. Regulation of microvascularization in heart failure - an endothelial cell, non-coding RNAs and exosome liaison. Noncoding RNA Res 2017; 2:45-55. [PMID: 30159420 PMCID: PMC6096416 DOI: 10.1016/j.ncrna.2017.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 01/26/2017] [Indexed: 12/22/2022] Open
Abstract
Heart failure is a complex syndrome involving various pathophysiological processes. An increasing body of evidence shows that the myocardial microvasculature is essential for the homeostasis state and that a decompensated heart is associated with microvascular dysfunction as a result of impaired endothelial angiogenic capacity. The intercellular communication between endothelial cells and cardiomyocytes through various signaling molecules, such as vascular endothelial growth factor, nitric oxide, and non-coding RNAs is an important determinant of cardiac microvascular function. Non-coding RNAs are transported from endothelial cells to cardiomyocytes, and vice versa, regulating microvascular properties and angiogenic processes in the heart. Small-exocytosed vesicles, called exosomes, which are secreted by both cell types, can mediate this intercellular communication. The purpose of this review is to highlight the contribution of the microvasculature to proper heart function maintenance by focusing on the interaction between cardiac endothelial cells and myocytes with a specific emphasis on non-coding RNAs (ncRNAs) in this form of cell-to-cell communication. Finally, the potential of ncRNAs as targets for angiogenesis therapy will also be discussed.
Collapse
Affiliation(s)
- Rio P. Juni
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Ricardo C. Abreu
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Paula A. da Costa Martins
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
43
|
Green tea extract intake during lactation modified cardiac macrophage infiltration and AMP-activated protein kinase phosphorylation in weanling rats from undernourished mother during gestation and lactation. J Dev Orig Health Dis 2016; 8:178-187. [PMID: 27919304 DOI: 10.1017/s2040174416000647] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Maternal dietary restriction is often associated with cardiovascular disease in offspring. The aim of this study was to investigate the effect of green tea extract (GTE) intake during lactation on macrophage infiltration, and activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) and serine-threonine kinase Akt (Akt) in the hearts of weanlings exposed to maternal dietary protein restriction. Pregnant Wistar rats were fed control (C) or low-protein diets (LP) throughout gestation. Following delivery, the dams received a control or a GTE-containing control diet during lactation: control diet during gestation and lactation (CC), low-protein diet during gestation and lactation (LPC), low-protein diet during gestation and 0.12% GTE-containing low-protein diet during lactation (LPL), and low-protein diet during gestation and 0.24% GTE-containing low-protein diet during lactation (LPH). The female offspring were sacrificed at day 22. Biochemical parameters in the plasma, macrophage infiltration, degree of fibrosis and expression levels of AMPK and Akt were examined. The plasma insulin level increased in LPH compared with LPC. Percentage of the fibrotic areas and the number of macrophages in LPC were higher than those in CC. Conversely, the fibrotic areas and the macrophage number in LPH were smaller (21 and 56%, respectively) than those in LPC. The levels of phosphorylated AMPK in LPL and LPH, and Akt in LPH were greater than those in LPC. In conclusion, maternal protein restriction may induce macrophage infiltration and the decrease of insulin levels. However, GTE intake during lactation may suppress macrophage infiltration and restore insulin secretion function via upregulation of AMPK and insulin signaling in weanlings.
Collapse
|
44
|
Barry JS, Davidsen ML, Limesand SW, Galan HL, Friedman JE, Regnault TRH, Hay WW. Developmental Changes in Ovine Myocardial Glucose Transporters and Insulin Signaling Following Hyperthermia-Induced Intrauterine Fetal Growth Restriction. Exp Biol Med (Maywood) 2016; 231:566-75. [PMID: 16636305 DOI: 10.1177/153537020623100511] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Developmental changes in ovine myocardial glucose transporters and insulin signaling following hyperthermia-induced intrauterine fetal growth restriction (IUGR) were the focus of our study. Our objective was to test the hypothesis that the fetal ovine myocardium adapts during an IUGR gestation by increasing glucose transporter protein expression, plasma membrane-bound glucose transporter protein concentrations, and insulin signal transduction protein concentrations. Growth measurements and whole heart tissue were obtained at 55 days gestational age (dGA), 90 dGA, and 135 dGA (term = 145 dGA) in fetuses from control (C) and hyperthermic (HT) pregnant sheep. Additionally, in 135 dGA animals, arterial blood was obtained and Doppler ultrasound was used to determine umbilical artery systolic (S) and diastolic (D) flow velocity waveform profiles to calculate pulsatility (S – D/mean) and resistance (S – D/S) indices. Myocardial Glut-1, Glut-4, insulin signal transduction proteins involved in Glut-4 translocation, and glycogen content were measured. Compared to age-matched controls, HT 90-dGA fetal body weights and HT 135-dGA fetal weights and gross heart weights were lower. Heart weights as a percent of body weights were similar between C and HT sheep at 135 dGA. HT 135-dGA animals had (i) lower fetal arterial plasma glucose and insulin concentrations, (ii) lower arterial blood oxygen content and higher plasma lactate concentrations, (iii) higher myocardial Glut-4 plasma membrane (PM) protein and insulin receptor β protein (IRβ) concentrations, (iv) higher myocardial glycogen content, and (v) higher umbilical artery Doppler pulsatility and resistance indices. The HT ovine fetal myocardium adapts to reduced circulating glucose and insulin concentrations by increasing plasma membrane Glut-4 and IRβ protein concentrations. The increased myocardial Glut-4 PM and IRβ protein concentrations likely contribute to or increase the intracellular delivery of glucose and, together with the increased lactate concentrations, enhance glycogen synthesis, which allows for maintained myocardial growth commensurate with fetal body growth.
Collapse
Affiliation(s)
- James S Barry
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, The Children's Hospital, 1056 East 19th Avenue, Box B070, Denver, CO 80218, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Wang Q, Liu Y, Fu Q, Xu B, Zhang Y, Kim S, Tan R, Barbagallo F, West T, Anderson E, Wei W, Abel ED, Xiang YK. Inhibiting Insulin-Mediated β2-Adrenergic Receptor Activation Prevents Diabetes-Associated Cardiac Dysfunction. Circulation 2016; 135:73-88. [PMID: 27815373 DOI: 10.1161/circulationaha.116.022281] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 10/13/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Type 2 diabetes mellitus (DM) and obesity independently increase the risk of heart failure by incompletely understood mechanisms. We propose that hyperinsulinemia might promote adverse consequences in the hearts of subjects with type-2 DM and obesity. METHODS High-fat diet feeding was used to induce obesity and DM in wild-type mice or mice lacking β2-adrenergic receptor (β2AR) or β-arrestin2. Wild-type mice fed with high-fat diet were treated with a β-blocker carvedilol or a GRK2 (G-protein-coupled receptor kinase 2) inhibitor. We examined signaling and cardiac contractile function. RESULTS High-fat diet feeding selectively increases the expression of phosphodiesterase 4D (PDE4D) in mouse hearts, in concert with reduced protein kinase A phosphorylation of phospholamban, which contributes to systolic and diastolic dysfunction. The expression of PDE4D is also elevated in human hearts with DM. The induction of PDE4D expression is mediated by an insulin receptor, insulin receptor substrate, and GRK2 and β-arrestin2-dependent transactivation of a β2AR-extracellular regulated protein kinase signaling cascade. Thus, pharmacological inhibition of β2AR or GRK2, or genetic deletion of β2AR or β-arrestin2, all significantly attenuate insulin-induced phosphorylation of extracellular regulated protein kinase and PDE4D induction to prevent DM-related contractile dysfunction. CONCLUSIONS These studies elucidate a novel mechanism by which hyperinsulinemia contributes to heart failure by increasing PDE4D expression and identify β2AR or GRK2 as plausible therapeutic targets for preventing or treating heart failure in subjects with type 2 DM.
Collapse
MESH Headings
- Animals
- Carbazoles/pharmacology
- Carvedilol
- Cells, Cultured
- Cyclic Nucleotide Phosphodiesterases, Type 4/genetics
- Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism
- Diabetes Mellitus, Type 2/complications
- Diet, High-Fat
- Extracellular Signal-Regulated MAP Kinases/metabolism
- G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors
- G-Protein-Coupled Receptor Kinase 2/metabolism
- Heart Failure/etiology
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardial Contraction/drug effects
- Myocardium/metabolism
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/metabolism
- Obesity/complications
- Propanolamines/pharmacology
- Receptors, Adrenergic, beta-2/deficiency
- Receptors, Adrenergic, beta-2/genetics
- Signal Transduction
- Vasodilator Agents/pharmacology
- beta-Arrestin 2/deficiency
- beta-Arrestin 2/genetics
Collapse
Affiliation(s)
- Qingtong Wang
- Department of Pharmacology, University of California at Davis, CA 95616, USA
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Yongming Liu
- Department of Pharmacology, University of California at Davis, CA 95616, USA
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200000, China
| | - Qin Fu
- Department of Pharmacology, University of California at Davis, CA 95616, USA
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bing Xu
- Department of Pharmacology, University of California at Davis, CA 95616, USA
| | - Yuan Zhang
- Department of Medicine, Division of Endocrinology and Metabolism and Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Sungjin Kim
- Department of Pharmacology, University of California at Davis, CA 95616, USA
| | - Ruensern Tan
- Department of Pharmacology, University of California at Davis, CA 95616, USA
| | - Federica Barbagallo
- Department of Pharmacology, University of California at Davis, CA 95616, USA
| | - Toni West
- Department of Pharmacology, University of California at Davis, CA 95616, USA
| | - Ethan Anderson
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC 27834, USA
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - E Dale Abel
- Department of Medicine, Division of Endocrinology and Metabolism and Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, CA 95616, USA
- VA Northern California Health Care System, Mather, CA 95655, USA
| |
Collapse
|
46
|
Hua G, He C, Lv X, Fan L, Wang C, Remmenga SW, Rodabaugh KJ, Yang L, Lele SM, Yang P, Karpf AR, Davis JS, Wang C. The four and a half LIM domains 2 (FHL2) regulates ovarian granulosa cell tumor progression via controlling AKT1 transcription. Cell Death Dis 2016; 7:e2297. [PMID: 27415427 PMCID: PMC4973349 DOI: 10.1038/cddis.2016.207] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/04/2016] [Accepted: 05/24/2016] [Indexed: 12/14/2022]
Abstract
The four and a half LIM domains 2 (FHL2) has been shown to play important roles in the regulation of cell proliferation, survival, adhesion, motility and signal transduction in a cell type and tissue-dependent manner. However, the function of FHL2 in ovarian physiology and pathology is unclear. The aim of this study was to determine the role and functional mechanism of FHL2 in the progression of ovarian granulosa cell tumors (GCTs). Immunohistochemical analysis indicated that FHL2 was overexpressed in GCT tissues. Cellular localization of FHL2 in GCT cells was cell cycle dependent. Knockdown of FHL2 suppressed GCT cell growth, reduced cell viability and inhibited cell migration. Consistently, ectopic expression of FHL2 in GCT cells with very low endogenous FHL2 promoted cell growth, improved cell viability and enhance cell migration. Importantly, overexpression of FHL2 promoted GCT progression in vivo. Mechanistic studies indicated that FHL2 regulates AKT1 gene expression in vitro and in vivo. Knockdown of FHL2 or AKT1 in GCT cell lines induced very similar phenotypes. Ectopic expression of constitutively active AKT1 rescued FHL2 knockdown-induced arrest of GCT cell growth and reduction of GCT cell viability, suggesting that FHL2 regulates GCT cell growth and viability through controlling AKT1 expression. Finally, co-immunoprecipitation and chromatin immunoprecipitation analyses indicated that FHL2 functions as a co-activator of NFκB and AP-1 to regulate AKT1 gene transcription. In conclusion, results from the present study indicate that FHL2 exerts its oncogenic action in GCT cells via controlling AKT1 gene expression. FHL2 is a promising target for the development of novel drugs against ovarian granulosa cell tumor.
Collapse
Affiliation(s)
- G Hua
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA.,Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei province 430070, China
| | - C He
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA.,Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei province 430070, China
| | - X Lv
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - L Fan
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei province 430070, China
| | - C Wang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei province 430070, China
| | - S W Remmenga
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - K J Rodabaugh
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - L Yang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei province 430070, China
| | - S M Lele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - P Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - A R Karpf
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - J S Davis
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA.,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.,Omaha Veterans Affairs Medical Center, Omaha, NE 68105, USA
| | - C Wang
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA.,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
47
|
Westermeier F, Riquelme JA, Pavez M, Garrido V, Díaz A, Verdejo HE, Castro PF, García L, Lavandero S. New Molecular Insights of Insulin in Diabetic Cardiomyopathy. Front Physiol 2016; 7:125. [PMID: 27148064 PMCID: PMC4828458 DOI: 10.3389/fphys.2016.00125] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/22/2016] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a highly prevalent disease worldwide. Cardiovascular disorders generated as a consequence of T2DM are a major cause of death related to this disease. Diabetic cardiomyopathy (DCM) is characterized by the morphological, functional and metabolic changes in the heart produced as a complication of T2DM. This cardiac disorder is characterized by constant high blood glucose and lipids levels which eventually generate oxidative stress, defective calcium handling, altered mitochondrial function, inflammation and fibrosis. In this context, insulin is of paramount importance for cardiac contractility, growth and metabolism and therefore, an impaired insulin signaling plays a critical role in the DCM development. However, the exact pathophysiological mechanisms leading to DCM are still a matter of study. Despite the numerous questions raised in the study of DCM, there have also been important findings, such as the role of micro-RNAs (miRNAs), which can not only have the potential of being important biomarkers, but also therapeutic targets. Furthermore, exosomes also arise as an interesting variable to consider, since they represent an important inter-cellular communication mechanism and therefore, they may explain many aspects of the pathophysiology of DCM and their study may lead to the development of therapeutic agents capable of improving insulin signaling. In addition, adenosine and adenosine receptors (ARs) may also play an important role in DCM. Moreover, the possible cross-talk between insulin and ARs may provide new strategies to reverse its defective signaling in the diabetic heart. This review focuses on DCM, the role of insulin in this pathology and the discussion of new molecular insights which may help to understand its underlying mechanisms and generate possible new therapeutic strategies.
Collapse
Affiliation(s)
- Francisco Westermeier
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Jaime A Riquelme
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Mario Pavez
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Valeria Garrido
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Ariel Díaz
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Hugo E Verdejo
- Faculty of Medicine, Advanced Center for Chronic Diseases, Pontifical Catholic University of ChileSantiago, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontifical Catholic University of ChileSantiago, Chile
| | - Pablo F Castro
- Faculty of Medicine, Advanced Center for Chronic Diseases, Pontifical Catholic University of ChileSantiago, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontifical Catholic University of ChileSantiago, Chile
| | - Lorena García
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Sergio Lavandero
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of ChileSantiago, Chile; Department of Internal Medicine (Division of Cardiology), University of Texas Southwestern Medical CenterDallas, TX, USA
| |
Collapse
|
48
|
Yoshida Y, Yamanaka S. Understanding Intracellular Signaling Advances Cardiac Reprogramming Technology Toward Clinical Applications. Circ Res 2016; 118:377-8. [PMID: 26846636 DOI: 10.1161/circresaha.115.308075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Yoshinori Yoshida
- From the Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan (Y.Y., S.Y.); and Gladstone Institute of Cardiovascular Disease, San Francisco, CA (S.Y.).
| | - Shinya Yamanaka
- From the Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan (Y.Y., S.Y.); and Gladstone Institute of Cardiovascular Disease, San Francisco, CA (S.Y.)
| |
Collapse
|
49
|
Stoppel WL, Kaplan DL, Black LD. Electrical and mechanical stimulation of cardiac cells and tissue constructs. Adv Drug Deliv Rev 2016; 96:135-55. [PMID: 26232525 DOI: 10.1016/j.addr.2015.07.009] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/16/2015] [Accepted: 07/25/2015] [Indexed: 12/19/2022]
Abstract
The field of cardiac tissue engineering has made significant strides over the last few decades, highlighted by the development of human cell derived constructs that have shown increasing functional maturity over time, particularly using bioreactor systems to stimulate the constructs. However, the functionality of these tissues is still unable to match that of native cardiac tissue and many of the stem-cell derived cardiomyocytes display an immature, fetal like phenotype. In this review, we seek to elucidate the biological underpinnings of both mechanical and electrical signaling, as identified via studies related to cardiac development and those related to an evaluation of cardiac disease progression. Next, we review the different types of bioreactors developed to individually deliver electrical and mechanical stimulation to cardiomyocytes in vitro in both two and three-dimensional tissue platforms. Reactors and culture conditions that promote functional cardiomyogenesis in vitro are also highlighted. We then cover the more recent work in the development of bioreactors that combine electrical and mechanical stimulation in order to mimic the complex signaling environment present in vivo. We conclude by offering our impressions on the important next steps for physiologically relevant mechanical and electrical stimulation of cardiac cells and engineered tissue in vitro.
Collapse
|
50
|
Shende P, Xu L, Morandi C, Pentassuglia L, Heim P, Lebboukh S, Berthonneche C, Pedrazzini T, Kaufmann BA, Hall MN, Rüegg MA, Brink M. Cardiac mTOR complex 2 preserves ventricular function in pressure-overload hypertrophy. Cardiovasc Res 2015; 109:103-14. [DOI: 10.1093/cvr/cvv252] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 11/06/2015] [Indexed: 11/12/2022] Open
|