1
|
Cao R, Gao T, Yue J, Sun G, Yang X. Disordered Gut Microbiome and Alterations in Metabolic Patterns Are Associated With Hypertensive Left Ventricular Hypertrophy. J Am Heart Assoc 2024; 13:e034230. [PMID: 39342506 DOI: 10.1161/jaha.123.034230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 08/23/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Left ventricular hypertrophy (LVH) is most common when driven by hypertension, and it is a strong independent risk factor for adverse cardiovascular events and death. Some animal models support a role for gut microbiota and metabolites in the development of LVH, but cohort studies confirming these findings in populations are lacking. METHODS AND RESULTS We investigated the alterations of gut microbiota and metabolites in 30 patients with hypertension, 30 patients with hypertensive LVH, and 30 matched controls on the basis of 16S rDNA and metabolomic analyses. Thirty stool and 90 serum samples were collected in fasting conditions. ANOVA/Kruskal-Wallis/Pearson's χ2/Fisher's exact test and Bonferroni's correction were used (P<0.0167) for comparison among the 3 groups. A regression analysis and subgroup analysis were performed between gut microbiota and left ventricular mass index (LVMI) and metabolites and LVMI, respectively. Spearman correlation analysis was performed between metabolites and flora and metabolites and LVMI. We observed LVH-enriched Faecalitalea (β=6758.55 [95% CI, 2080.92-11436.18]; P=0.009), Turicibacter (β=8424.76 [95% CI, 2494.05-14355.47]; P=0.01), Ruminococcus torques group (β=840.88 [95% CI, 223.1-1458.67]; P=0.013), and Erysipelotrichaceae UCG-003 (β=856.37 [95% CI, 182.76-1529.98]; P=0.019) were positively correlated with LVMI. A total of 1141 (in sera) and 2657 (in feces) metabolites were identified. There was a sex-specific association between metabolites and LVMI. Significant changes in metabolic pathways in LVH were also observed, especially bile acid and lipid metabolism pathways. CONCLUSIONS Our study demonstrated the disordered gut microbiota and microbial metabolite profiles in LVH. This highlights the roles of gut bacteria and metabolite in this disease and could lead to new intervention, diagnostic, or management paradigms for LVH. REGISTRATION URL: https://www.chictr.org.cn; Unique Identifier: ChiCTR2200055603.
Collapse
Affiliation(s)
- Rong Cao
- Department of Cardiovascular Medicine Research Institute of Hypertension, The Second Affiliated Hospital of Baotou Medical College Baotou Inner Mongolia China
- Department of Cardiovascular Medicine The First Affiliated Hospital of Fujian Medical University Fuzhou Fujian China
| | - Ting Gao
- Geriatric Department Baotou Central Hospital Baotou Inner Mongolia China
| | - Jianwei Yue
- Department of Cardiovascular Medicine Research Institute of Hypertension, The Second Affiliated Hospital of Baotou Medical College Baotou Inner Mongolia China
| | - Gang Sun
- Department of Cardiovascular Medicine Research Institute of Hypertension, The Second Affiliated Hospital of Baotou Medical College Baotou Inner Mongolia China
| | - Xiaomin Yang
- General Medicine Sir Run Run Shaw Hospital, Zhejiang University Zhejiang China
| |
Collapse
|
2
|
Stanciu SM, Jinga M, Miricescu D, Stefani C, Nica RI, Stanescu-Spinu II, Vacaroiu IA, Greabu M, Nica S. mTOR Dysregulation, Insulin Resistance, and Hypertension. Biomedicines 2024; 12:1802. [PMID: 39200267 PMCID: PMC11351979 DOI: 10.3390/biomedicines12081802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Worldwide, diabetes mellitus (DM) and cardiovascular diseases (CVDs) represent serious health problems associated with unhealthy diet and sedentarism. Metabolic syndrome (MetS) is characterized by obesity, dyslipidemia, hyperglycemia, insulin resistance (IR) and hypertension. The mammalian target of rapamycin (mTOR) is a serine/threonine kinase with key roles in glucose and lipid metabolism, cell growth, survival and proliferation. mTOR hyperactivation disturbs glucose metabolism, leading to hyperglycemia and further to IR, with a higher incidence in the Western population. Metformin is one of the most used hypoglycemic drugs, with anti-inflammatory, antioxidant and antitumoral properties, having also the capacity to inhibit mTOR. mTOR inhibitors such as rapamycin and its analogs everolimus and temsirolimus block mTOR activity, decrease the levels of glucose and triglycerides, and reduce body weight. The link between mTOR dysregulation, IR, hypertension and mTOR inhibitors has not been fully described. Therefore, the main aim of this narrative review is to present the mechanism by which nutrients, proinflammatory cytokines, increased salt intake and renin-angiotensin-aldosterone system (RAAS) dysregulation induce mTOR overactivation, associated further with IR and hypertension development, and also mTOR inhibitors with higher potential to block the activity of this protein kinase.
Collapse
Affiliation(s)
- Silviu Marcel Stanciu
- Department of Internal Medicine and Gastroenterology, Carol Davila University of Medicine and Pharmacy, Central Military Emergency University Hospital, “Dr. Carol Davila”, 010825 Bucharest, Romania; (S.M.S.); (M.J.)
| | - Mariana Jinga
- Department of Internal Medicine and Gastroenterology, Carol Davila University of Medicine and Pharmacy, Central Military Emergency University Hospital, “Dr. Carol Davila”, 010825 Bucharest, Romania; (S.M.S.); (M.J.)
| | - Daniela Miricescu
- Discipline of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania;
| | - Constantin Stefani
- Department of Family Medicine and Clinical Base, Central Military Emergency University Hospital, “Dr. Carol Davila”, 010825 Bucharest, Romania;
| | - Remus Iulian Nica
- Surgery Department, Central Military Emergency University Hospital, “Dr. Carol Davila”, 010825 Bucharest, Romania;
- Discipline of General Surgery, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanotari Blvd, 054474 Bucharest, Romania
| | - Iulia-Ioana Stanescu-Spinu
- Discipline of Physiology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania
| | - Ileana Adela Vacaroiu
- Department of Nephrology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Maria Greabu
- Discipline of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania;
| | - Silvia Nica
- Emergency Discipline, University Hospital of Bucharest, 050098 Bucharest, Romania;
- Department of Emergency and First Aid, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania
| |
Collapse
|
3
|
Challa AA, Vidal P, Maurya SK, Maurya CK, Baer LA, Wang Y, James NM, Pardeshi PJ, Fasano M, Carley AN, Stanford KI, Lewandowski ED. UCP1-dependent brown adipose activation accelerates cardiac metabolic remodeling and reduces initial hypertrophic and fibrotic responses to pathological stress. FASEB J 2024; 38:e23709. [PMID: 38809700 PMCID: PMC11163965 DOI: 10.1096/fj.202400922r] [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: 04/22/2024] [Revised: 05/06/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
Brown adipose tissue (BAT) is correlated to cardiovascular health in rodents and humans, but the physiological role of BAT in the initial cardiac remodeling at the onset of stress is unknown. Activation of BAT via 48 h cold (16°C) in mice following transverse aortic constriction (TAC) reduced cardiac gene expression for LCFA uptake and oxidation in male mice and accelerated the onset of cardiac metabolic remodeling, with an early isoform shift of carnitine palmitoyltransferase 1 (CPT1) toward increased CPT1a, reduced entry of long chain fatty acid (LCFA) into oxidative metabolism (0.59 ± 0.02 vs. 0.72 ± 0.02 in RT TAC hearts, p < .05) and increased carbohydrate oxidation with altered glucose transporter content. BAT activation with TAC reduced early hypertrophic expression of β-MHC by 61% versus RT-TAC and reduced pro-fibrotic TGF-β1 and COL3α1 expression. While cardiac natriuretic peptide expression was yet to increase at only 3 days TAC, Nppa and Nppb expression were elevated in Cold TAC versus RT TAC hearts 2.7- and 2.4-fold, respectively. Eliminating BAT thermogenic activation with UCP1 KO mice eliminated differences between Cold TAC and RT TAC hearts, confirming effects of BAT activation rather than autonomous cardiac responses to cold. Female responses to BAT activation were blunted, with limited UCP1 changes with cold, partly due to already activated BAT in females at RT compared to thermoneutrality. These data reveal a previously unknown physiological mechanism of UCP1-dependent BAT activation in attenuating early cardiac hypertrophic and profibrotic signaling and accelerating remodeled metabolic activity in the heart at the onset of cardiac stress.
Collapse
Affiliation(s)
- Azariyas A. Challa
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Pablo Vidal
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - Santosh K. Maurya
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Chandan K. Maurya
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Lisa A. Baer
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - Yang Wang
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Natasha Maria James
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - Parth J. Pardeshi
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - Matthew Fasano
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Andrew N. Carley
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Kristin I. Stanford
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - E. Douglas Lewandowski
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| |
Collapse
|
4
|
Chakraborty P, Po SS, Scherlag BJ, Dasari TW. The neurometabolic axis: A novel therapeutic target in heart failure. Life Sci 2023; 333:122122. [PMID: 37774940 DOI: 10.1016/j.lfs.2023.122122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023]
Abstract
Abnormal cardiac metabolism or cardiac metabolic remodeling is reported before the onset of heart failure with reduced ejection fraction (HFrEF) and is known to trigger and maintain the mechanical dysfunction and electrical, and structural abnormalities of the ventricle. A dysregulated cardiac autonomic tone characterized by sympathetic overdrive with blunted parasympathetic activation is another pathophysiological hallmark of HF. Emerging evidence suggests a link between autonomic nervous system activity and cardiac metabolism. Chronic β-adrenergic activation promotes maladaptive metabolic remodeling whereas cholinergic activation attenuates the metabolic aberrations through favorable modulation of key metabolic regulatory molecules. Restoration of sympathovagal balance by neuromodulation strategies is emerging as a novel nonpharmacological treatment strategy in HF. The current review attempts to evaluate the 'neuro-metabolic axis' in HFrEF and whether neuromodulation can mitigate the adverse metabolic remodeling in HFrEF.
Collapse
Affiliation(s)
- Praloy Chakraborty
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sunny S Po
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Benjamin J Scherlag
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tarun W Dasari
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
5
|
Wang H, Liu X, Zhou Q, Liu L, Jia Z, Qi Y, Xu F, Zhang Y. Current status and emerging trends of cardiac metabolism from the past 20 years: A bibliometric study. Heliyon 2023; 9:e21952. [PMID: 38045208 PMCID: PMC10692779 DOI: 10.1016/j.heliyon.2023.e21952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 12/05/2023] Open
Abstract
Background Abnormal cardiac metabolism is a key factor in the development of cardiovascular diseases. Consequently, there has been considerable emphasis on researching and developing drugs that regulate metabolism. This study employed bibliometric methods to comprehensively and objectively analyze the relevant literature, offering insights into the knowledge dynamics in this field. Methods The data source for this study was the Web of Science Core Collection (WoSCC), from which the collected data were imported into bibliometric software for analysis. Results The United States was the leading contributor, accounting for 38.33 % of publications. The University of Washington and Damian J. Tyler were the most active institution and author, respectively. The American Journal of Physiology-Heart and Circulatory Physiology, Journal of Molecular and Cellular Cardiology, Cardiovascular Research, Circulation Research, and American Journal of Physiology-Endocrinology and Metabolism were highly influential journals that published numerous high-quality articles on cardiac metabolism. Common keywords in this research area included heart failure, insulin resistance, skeletal muscle, mitochondria, as well as topic words such as cardiac metabolism, fatty acid oxidation, glucose metabolism, and myocardial metabolism. Co-citation analysis has shown that research on heart failure and in vitro modeling of cardiovascular disease has gained prominence in recent years and making it a research hotspot. Conclusion Research on cardiac metabolism is steadily growing, with a specific focus on heart failure and the interplay between mitochondrial dysfunction, insulin resistance, and cardiac metabolism. An emerging trend in this field involves the enhancement of maturation in human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) through the manipulation of cardiac metabolism.
Collapse
Affiliation(s)
- Hongqin Wang
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolin Liu
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingbing Zhou
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Liu
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Zijun Jia
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Yifei Qi
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengqin Xu
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Zhang
- Institute of Geriatric, Xiyuan Hospital, Beijing, China
- China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Leid J, Gray R, Rakita P, Koenig AL, Tripathy R, Fitzpatrick JAJ, Kaufman C, Solnica-Krezel L, Lavine KJ. Deletion of taf1 and taf5 in zebrafish capitulate cardiac and craniofacial abnormalities associated with TAFopathies through perturbations in metabolism. Biol Open 2023; 12:bio059905. [PMID: 37746814 PMCID: PMC10354717 DOI: 10.1242/bio.059905] [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: 02/27/2023] [Accepted: 05/16/2023] [Indexed: 09/26/2023] Open
Abstract
Intellectual disability is a neurodevelopmental disorder that affects 2-3% of the general population. Syndromic forms of intellectual disability frequently have a genetic basis and are often accompanied by additional developmental anomalies. Pathogenic variants in components of TATA-binding protein associated factors (TAFs) have recently been identified in a subset of patients with intellectual disability, craniofacial hypoplasia, and congenital heart disease. This syndrome has been termed as a TAFopathy and includes mutations in TATA binding protein (TBP), TAF1, TAF2, and TAF6. The underlying mechanism by which TAFopathies give rise to neurodevelopmental, craniofacial, and cardiac abnormalities remains to be defined. Through a forward genetic screen in zebrafish, we have recovered a recessive mutant phenotype characterized by craniofacial hypoplasia, ventricular hypoplasia, heart failure at 96 h post-fertilization and lethality, and show it is caused by a nonsense mutation in taf5. CRISPR/CAS9 mediated gene editing revealed that these defects where phenocopied by mutations in taf1 and taf5. Mechanistically, taf5-/- zebrafish displayed misregulation in metabolic gene expression and metabolism as evidenced by RNA sequencing, respiration assays, and metabolite studies. Collectively, these findings suggest that the TAF complex may contribute to neurologic, craniofacial, and cardiac development through regulation of metabolism.
Collapse
Affiliation(s)
- Jamison Leid
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ryan Gray
- Departments of Nutritional Sciences, Dell Pediatrics Research Institute, University of Texas at Austin, Austin, TX 78723, USA
| | - Peter Rakita
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew L. Koenig
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rohan Tripathy
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James A. J. Fitzpatrick
- Departments of Neuroscience and Cell Biology, Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Charles Kaufman
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kory J. Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Immunology and Pathology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
7
|
Duan X, Liu X, Zhan Z. Metabolic Regulation of Cardiac Regeneration. Front Cardiovasc Med 2022; 9:933060. [PMID: 35872916 PMCID: PMC9304552 DOI: 10.3389/fcvm.2022.933060] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/13/2022] [Indexed: 12/16/2022] Open
Abstract
The mortality due to heart diseases remains highest in the world every year, with ischemic cardiomyopathy being the prime cause. The irreversible loss of cardiomyocytes following myocardial injury leads to compromised contractility of the remaining myocardium, adverse cardiac remodeling, and ultimately heart failure. The hearts of adult mammals can hardly regenerate after cardiac injury since adult cardiomyocytes exit the cell cycle. Nonetheless, the hearts of early neonatal mammals possess a stronger capacity for regeneration. To improve the prognosis of patients with heart failure and to find the effective therapeutic strategies for it, it is essential to promote endogenous regeneration of adult mammalian cardiomyocytes. Mitochondrial metabolism maintains normal physiological functions of the heart and compensates for heart failure. In recent decades, the focus is on the changes in myocardial energy metabolism, including glucose, fatty acid, and amino acid metabolism, in cardiac physiological and pathological states. In addition to being a source of energy, metabolites are becoming key regulators of gene expression and epigenetic patterns, which may affect heart regeneration. However, the myocardial energy metabolism during heart regeneration is majorly unknown. This review focuses on the role of energy metabolism in cardiac regeneration, intending to shed light on the strategies for manipulating heart regeneration and promoting heart repair after cardiac injury.
Collapse
Affiliation(s)
- Xuewen Duan
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Institute of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xingguang Liu
- Department of Pathogen Biology, Naval Medical University, Shanghai, China
- Xingguang Liu,
| | - Zhenzhen Zhan
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Institute of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Zhenzhen Zhan,
| |
Collapse
|
8
|
Bioactive Peptides and Exercise Modulate the AMPK/SIRT1/PGC-1α/FOXO3 Pathway as a Therapeutic Approach for Hypertensive Rats. Pharmaceuticals (Basel) 2022; 15:ph15070819. [PMID: 35890118 PMCID: PMC9319799 DOI: 10.3390/ph15070819] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/05/2023] Open
Abstract
Peptides are fragments of fundamental protein sequences that may have health benefits in addition to basic dietary benefits. Recently, we have reported on the pharmacological benefits of alcalase potato protein hydrolysate (APPH) and bioactive peptides isolated from APPH. The aim was to evaluate the synergistic effect of exercise along with DIKTNKPVIF (DF) peptides in ameliorating hypertension in spontaneously hypertensive rat (SHR) rats. We examined ECG parameters, lipid profiles, cardiac markers, and histology, and quantified the proteins associated with fibrosis, hypertrophy, apoptosis, mitochondrial biogenesis, and longevity pathways. DF peptide administration, along with exercise, reduced the blood pressure and cardiac marker levels in serum. Furthermore, it also suppressed the expression of fibrosis markers COL1A1, CTGF, and uPA and downregulated cardiac-hypertrophy-associated markers such as calcineurin, NFATC3, GATA4, pGATA4 and BNP. Exercise synergistically increases the expression of IFG1, PI3K, and AKT cell-survival pathway proteins, along with DF administration. Moreover, AMPK/SIRT1/PGC-1α/FOXO3 pathway protein expression was increased with the combinatorial administration of DF and exercise. Our data suggest that exercise, along with DF peptides, act synergistically in alleviating hypertension by activating the mitochondrial biogenesis pathway.
Collapse
|
9
|
Ashraf S, Taegtmeyer H, Harmancey R. Prolonged cardiac NR4A2 activation causes dilated cardiomyopathy in mice. Basic Res Cardiol 2022; 117:33. [PMID: 35776225 PMCID: PMC9249728 DOI: 10.1007/s00395-022-00942-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 06/14/2022] [Accepted: 06/23/2022] [Indexed: 02/03/2023]
Abstract
Transcription factors play a fundamental role in cardiovascular adaptation to stress. Nuclear receptor subfamily 4 group A member 2 (NR4A2; NURR1) is an immediate-early gene and transcription factor with a versatile role throughout many organs. In the adult mammalian heart, and particularly in cardiac myocytes, NR4A2 is strongly up-regulated in response to beta-adrenergic stimulation. The physiologic implications of this increase remain unknown. In this study, we aimed to interrogate the consequences of cardiac NR4A2 up-regulation under normal conditions and in response to pressure overload. In mice, tamoxifen-dependent, cardiomyocyte-restricted overexpression of NR4A2 led to cardiomyocyte hypertrophy, left ventricular dilation, heart failure, and death within 40 days. Chronic NR4A2 induction also precipitated cardiac decompensation during transverse aortic constriction (TAC)-induced pressure overload. Mechanistically, NR4A2 caused adult cardiac myocytes to return to a fetal-like phenotype, with a switch to glycolytic metabolism and disassembly of sarcomeric structures. NR4A2 also re-activated cell cycle progression and stimulated DNA replication and karyokinesis but failed to induce cytokinesis, thereby promoting multinucleation of cardiac myocytes. Activation of cell cycle checkpoints led to induction of an apoptotic response which ultimately resulted in excessive loss of cardiac myocytes and impaired left ventricular contractile function. In summary, myocyte-specific overexpression of NR4A2 in the postnatal mammalian heart results in increased cell cycle re-entry and DNA replication but does not result in cardiac myocyte division. Our findings expose a novel function for the nuclear receptor as a critical regulator in the self-renewal of the cardiac myocyte and heart regeneration.
Collapse
Affiliation(s)
- Sadia Ashraf
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Romain Harmancey
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX, 77030, USA.
| |
Collapse
|
10
|
He X, Zeng H, Cantrell AC, Williams QA, Chen JX. Knockout of TIGAR enhances myocardial phosphofructokinase activity and preserves diastolic function in heart failure. J Cell Physiol 2022; 237:3317-3327. [PMID: 35621078 PMCID: PMC9378637 DOI: 10.1002/jcp.30790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/11/2022] [Indexed: 11/06/2022]
Abstract
Hypertension is an important risk factor in the pathogenesis of diastolic dysfunction. Growing evidence indicates that glucose metabolism plays an essential role in diastolic dysfunction. TP53-induced glycolysis and apoptosis regulator (TIGAR) has been shown to regulate glucose metabolism and heart failure (HF). In the present study, we investigated the role of TIGAR in diastolic function and cardiac fibrosis during pressure overload (PO)-induced HF. WT mice subjected to transverse aortic constriction (TAC), a commonly used method to induce diastolic dysfunction, exhibited diastolic dysfunction as evidenced by increased E/A ratio and E/E' ratio when compared to its sham controls. This was accompanied by increased cardiac interstitial fibrosis. In contrast, the knockout of TIGAR attenuated PO-induced diastolic dysfunction and interstitial fibrosis. Mechanistically, the levels of glucose transporter Glut-1, Glut-4, and key glycolytic enzyme phosphofructokinase 1 (PFK-1) were significantly elevated in TIGAR KO subjected to TAC as compared to that of WT mice. Knockout of TIGAR significantly increased fructose 2,6-bisphosphate levels and phosphofructokinase activity in mouse hearts. In addition, PO resulted in a significant increase in perivascular fibrosis and endothelial activation in the WT mice, but not in the TIGAR KO mice. Our present study suggests a necessary role of TIGAR-mediated glucose metabolism in PO-induced cardiac fibrosis and diastolic dysfunction.
Collapse
Affiliation(s)
- Xiaochen He
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Heng Zeng
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Aubrey C Cantrell
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Quinesha A Williams
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
11
|
Haidar A, Taegtmeyer H. Strategies for Imaging Metabolic Remodeling of the Heart in Obesity and Heart Failure. Curr Cardiol Rep 2022; 24:327-335. [PMID: 35107704 PMCID: PMC9074778 DOI: 10.1007/s11886-022-01650-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Define early myocardial metabolic changes among patients with obesity and heart failure, and to describe noninvasive methods and their applications for imaging cardiac metabolic remodeling. RECENT FINDINGS Metabolic remodeling precedes, triggers, and sustains functional and structural remodeling in the stressed heart. Alterations in cardiac metabolism can be assessed by using a variety of molecular probes. The glucose tracer analog, 18F-FDG, and the labeled tracer 11C-palmitate are still the most commonly used tracers to assess glucose and fatty acid metabolism, respectively. The development of new tracer analogs and imaging agents, including those targeting the peroxisome proliferator-activated receptor (PPAR), provides new opportunities for imaging metabolic activities at a molecular level. While the use of cardiac magnetic resonance spectroscopy in the clinical setting is limited to the assessment of intramyocardial and epicardial fat, new technical improvements are likely to increase its usage in the setting of heart failure. Noninvasive imaging methods are an effective tool for the serial assessment of alterations in cardiac metabolism, either during disease progression, or in response to treatment.
Collapse
Affiliation(s)
- Amier Haidar
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, MSB 1.220, Houston, TX, 77030, USA.
| |
Collapse
|
12
|
An S, Wang D, Ma X, Liu C. Whole body vibration remodels skeletal muscle via autophagy and energy metabolism in diabetic mice. Mol Med Rep 2022; 25:182. [PMID: 35322859 PMCID: PMC8972300 DOI: 10.3892/mmr.2022.12698] [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: 09/04/2020] [Accepted: 03/08/2022] [Indexed: 11/17/2022] Open
Abstract
Hyperglycemia occurs due to a defect in insulin secretion or impaired biological functions, or both. The long-term hyperglycemia during diabetes causes chronic damage and dysfunction of various tissues. Whole body vibration (WBV) has significant effects on lipid and glucose metabolism and endocrine and motor systems. In order to explore the effects of WBV on skeletal muscle, mice trained for 12 weeks with WBV (15 Hz, 30 min) were used as experimental subjects and their skeletal muscle morphology under the pathological state of diabetes was observed. In addition, the blood lipids, blood glucose, gastrocnemius muscle glycogen and mRNA and protein levels of autophagy and glucose metabolism biomarkers were compared among the three groups of mice via western blot and RT-qPCR. The results showed that WBV can significantly reshape skeletal muscle morphology and upregulate high density lipoprotein. The expression of glucose-6-phosphatase (G6P), Beclin1 and Atg7 in the gastrocnemius muscle of the WBV group was significantly increased. Therefore, it can be concluded that WBV promotes skeletal muscle remodeling in diabetic mice. The present study confirmed that WBV can attenuate the development of diabetes melitus (DM) and lead to lower level low density lipoprotein in the blood. In addition, G6P level plays an important role in WBV-treated DM model and may be used to monitor the effect of WBV in patients. The findings of the present study may provide a new molecular basis for WBV to play a therapeutic role in the treatment of diabetes and may have potential clinical applications in the future.
Collapse
Affiliation(s)
- Shanshan An
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Dahao Wang
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Xue Ma
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Chang Liu
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
13
|
Doran S, Arif M, Lam S, Bayraktar A, Turkez H, Uhlen M, Boren J, Mardinoglu A. Multi-omics approaches for revealing the complexity of cardiovascular disease. Brief Bioinform 2021; 22:bbab061. [PMID: 33725119 PMCID: PMC8425417 DOI: 10.1093/bib/bbab061] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/20/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
The development and progression of cardiovascular disease (CVD) can mainly be attributed to the narrowing of blood vessels caused by atherosclerosis and thrombosis, which induces organ damage that will result in end-organ dysfunction characterized by events such as myocardial infarction or stroke. It is also essential to consider other contributory factors to CVD, including cardiac remodelling caused by cardiomyopathies and co-morbidities with other diseases such as chronic kidney disease. Besides, there is a growing amount of evidence linking the gut microbiota to CVD through several metabolic pathways. Hence, it is of utmost importance to decipher the underlying molecular mechanisms associated with these disease states to elucidate the development and progression of CVD. A wide array of systems biology approaches incorporating multi-omics data have emerged as an invaluable tool in establishing alterations in specific cell types and identifying modifications in signalling events that promote disease development. Here, we review recent studies that apply multi-omics approaches to further understand the underlying causes of CVD and provide possible treatment strategies by identifying novel drug targets and biomarkers. We also discuss very recent advances in gut microbiota research with an emphasis on how diet and microbial composition can impact the development of CVD. Finally, we present various biological network analyses and other independent studies that have been employed for providing mechanistic explanation and developing treatment strategies for end-stage CVD, namely myocardial infarction and stroke.
Collapse
Affiliation(s)
- Stephen Doran
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
| | - Muhammad Arif
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Simon Lam
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
| | - Abdulahad Bayraktar
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jan Boren
- Institute of Medicine, Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital Gothenburg, Sweden
| | - Adil Mardinoglu
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
14
|
Li Q, Li C, Elnwasany A, Sharma G, An YA, Zhang G, Elhelaly WM, Lin J, Gong Y, Chen G, Wang M, Zhao S, Dai C, Smart CD, Liu J, Luo X, Deng Y, Tan L, Lv SJ, Davidson SM, Locasale JW, Lorenzi PL, Malloy CR, Gillette TG, Vander Heiden MG, Scherer PE, Szweda LI, Fu G, Wang ZV. PKM1 Exerts Critical Roles in Cardiac Remodeling Under Pressure Overload in the Heart. Circulation 2021; 144:712-727. [PMID: 34102853 PMCID: PMC8405569 DOI: 10.1161/circulationaha.121.054885] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Metabolic remodeling precedes most alterations during cardiac hypertrophic growth under hemodynamic stress. The elevation of glucose utilization has been recognized as a hallmark of metabolic remodeling. However, its role in cardiac hypertrophic growth and heart failure in response to pressure overload remains to be fully illustrated. Here, we aimed to dissect the role of cardiac PKM1 (pyruvate kinase muscle isozyme 1) in glucose metabolic regulation and cardiac response under pressure overload. METHODS Cardiac-specific deletion of PKM1 was achieved by crossing the floxed PKM1 mouse model with the cardiomyocyte-specific Cre transgenic mouse. PKM1 transgenic mice were generated under the control of tetracycline response elements, and cardiac-specific overexpression of PKM1 was induced by doxycycline administration in adult mice. Pressure overload was triggered by transverse aortic constriction. Primary neonatal rat ventricular myocytes were used to dissect molecular mechanisms. Moreover, metabolomics and nuclear magnetic resonance spectroscopy analyses were conducted to determine cardiac metabolic flux in response to pressure overload. RESULTS We found that PKM1 expression is reduced in failing human and mouse hearts. It is important to note that cardiomyocyte-specific deletion of PKM1 exacerbates cardiac dysfunction and fibrosis in response to pressure overload. Inducible overexpression of PKM1 in cardiomyocytes protects the heart against transverse aortic constriction-induced cardiomyopathy and heart failure. At the mechanistic level, PKM1 is required for the augmentation of glycolytic flux, mitochondrial respiration, and ATP production under pressure overload. Furthermore, deficiency of PKM1 causes a defect in cardiomyocyte growth and a decrease in pyruvate dehydrogenase complex activity at both in vitro and in vivo levels. CONCLUSIONS These findings suggest that PKM1 plays an essential role in maintaining a homeostatic response in the heart under hemodynamic stress.
Collapse
Affiliation(s)
- Qinfeng Li
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chao Li
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Abdallah Elnwasany
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Gaurav Sharma
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yu A. An
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Guangyu Zhang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Waleed M. Elhelaly
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jun Lin
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yingchao Gong
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Guihao Chen
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Meihui Wang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Shangang Zhao
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chongshan Dai
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Charles D. Smart
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Juan Liu
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Xiang Luo
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yingfeng Deng
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shuang-Jie Lv
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shawn M. Davidson
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jason W. Locasale
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Philip L. Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Craig R. Malloy
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas G. Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Matthew G. Vander Heiden
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Luke I. Szweda
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhao V. Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
15
|
Chou C, Chin MT. Pathogenic Mechanisms of Hypertrophic Cardiomyopathy beyond Sarcomere Dysfunction. Int J Mol Sci 2021; 22:ijms22168933. [PMID: 34445638 PMCID: PMC8396307 DOI: 10.3390/ijms22168933] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 01/23/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiovascular disorder, affecting 1 in 500 people in the general population. Although characterized by asymmetric left ventricular hypertrophy, cardiomyocyte disarray, and cardiac fibrosis, HCM is in fact a highly complex disease with heterogenous clinical presentation, onset, and complications. While HCM is generally accepted as a disease of the sarcomere, variable penetrance in families with identical genetic mutations challenges the monogenic origin of HCM and instead implies a multifactorial cause. Furthermore, large-scale genome sequencing studies revealed that many genes previously reported as causative of HCM in fact have little or no evidence of disease association. These findings thus call for a re-evaluation of the sarcomere-centered view of HCM pathogenesis. Here, we summarize our current understanding of sarcomere-independent mechanisms of cardiomyocyte hypertrophy, highlight the role of extracellular signals in cardiac fibrosis, and propose an alternative but integrated model of HCM pathogenesis.
Collapse
Affiliation(s)
- Chun Chou
- Department of Medicine, Tufts University School of Medicine, Boston, MA 02111, USA;
| | - Michael T. Chin
- Department of Medicine, Tufts University School of Medicine, Boston, MA 02111, USA;
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
- Correspondence: ; Tel.: +1-617-636-8776
| |
Collapse
|
16
|
Myocardial Tissue Characterization in Heart Failure with Preserved Ejection Fraction: From Histopathology and Cardiac Magnetic Resonance Findings to Therapeutic Targets. Int J Mol Sci 2021; 22:ijms22147650. [PMID: 34299270 PMCID: PMC8304780 DOI: 10.3390/ijms22147650] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex clinical syndrome responsible for high mortality and morbidity rates. It has an ever growing social and economic impact and a deeper knowledge of molecular and pathophysiological basis is essential for the ideal management of HFpEF patients. The association between HFpEF and traditional cardiovascular risk factors is known. However, myocardial alterations, as well as pathophysiological mechanisms involved are not completely defined. Under the definition of HFpEF there is a wide spectrum of different myocardial structural alterations. Myocardial hypertrophy and fibrosis, coronary microvascular dysfunction, oxidative stress and inflammation are only some of the main pathological detectable processes. Furthermore, there is a lack of effective pharmacological targets to improve HFpEF patients' outcomes and risk factors control is the primary and unique approach to treat those patients. Myocardial tissue characterization, through invasive and non-invasive techniques, such as endomyocardial biopsy and cardiac magnetic resonance respectively, may represent the starting point to understand the genetic, molecular and pathophysiological mechanisms underlying this complex syndrome. The correlation between histopathological findings and imaging aspects may be the future challenge for the earlier and large-scale HFpEF diagnosis, in order to plan a specific and effective treatment able to modify the disease's natural course.
Collapse
|
17
|
Davogustto GE, Salazar RL, Vasquez HG, Karlstaedt A, Dillon WP, Guthrie PH, Martin JR, Vitrac H, De La Guardia G, Vela D, Ribas-Latre A, Baumgartner C, Eckel-Mahan K, Taegtmeyer H. Metabolic remodeling precedes mTORC1-mediated cardiac hypertrophy. J Mol Cell Cardiol 2021; 158:115-127. [PMID: 34081952 DOI: 10.1016/j.yjmcc.2021.05.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 11/17/2022]
Abstract
RATIONALE The nutrient sensing mechanistic target of rapamycin complex 1 (mTORC1) and its primary inhibitor, tuberin (TSC2), are cues for the development of cardiac hypertrophy. The phenotype of mTORC1 induced hypertrophy is unknown. OBJECTIVE To examine the impact of sustained mTORC1 activation on metabolism, function, and structure of the adult heart. METHODS AND RESULTS We developed a mouse model of inducible, cardiac-specific sustained mTORC1 activation (mTORC1iSA) through deletion of Tsc2. Prior to hypertrophy, rates of glucose uptake and oxidation, as well as protein and enzymatic activity of glucose 6-phosphate isomerase (GPI) were decreased, while intracellular levels of glucose 6-phosphate (G6P) were increased. Subsequently, hypertrophy developed. Transcript levels of the fetal gene program and pathways of exercise-induced hypertrophy increased, while hypertrophy did not progress to heart failure. We therefore examined the hearts of wild-type mice subjected to voluntary physical activity and observed early changes in GPI, followed by hypertrophy. Rapamycin prevented these changes in both models. CONCLUSION Activation of mTORC1 in the adult heart triggers the development of a non-specific form of hypertrophy which is preceded by changes in cardiac glucose metabolism.
Collapse
Affiliation(s)
- Giovanni E Davogustto
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rebecca L Salazar
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hernan G Vasquez
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Anja Karlstaedt
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - William P Dillon
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Patrick H Guthrie
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joseph R Martin
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Gina De La Guardia
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Deborah Vela
- Cardiovascular Pathology Research Laboratory, Texas Heart Institute at CHI St. Luke's Health, and the Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Aleix Ribas-Latre
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Corrine Baumgartner
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kristin Eckel-Mahan
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
18
|
Lactate Dehydrogenase A Governs Cardiac Hypertrophic Growth in Response to Hemodynamic Stress. Cell Rep 2021; 32:108087. [PMID: 32877669 PMCID: PMC7520916 DOI: 10.1016/j.celrep.2020.108087] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 07/07/2020] [Accepted: 08/07/2020] [Indexed: 01/06/2023] Open
Abstract
The heart manifests hypertrophic growth in response to high blood pressure, which may decompensate and progress to heart failure under persistent stress. Metabolic remodeling is an early event in this process. However, its role remains to be fully characterized. Here, we show that lactate dehydrogenase A (LDHA), a critical glycolytic enzyme, is elevated in the heart in response to hemodynamic stress. Cardiomyocyte-restricted deletion of LDHA leads to defective cardiac hypertrophic growth and heart failure by pressure overload. Silencing of LDHA in cultured cardiomyocytes suppresses cell growth from pro-hypertrophic stimulation in vitro, while overexpression of LDHA is sufficient to drive cardiomyocyte growth. Furthermore, we find that lactate is capable of rescuing the growth defect from LDHA knockdown. Mechanistically, lactate stabilizes NDRG3 (N-myc downregulated gene family 3) and stimulates ERK (extracellular signal-regulated kinase). Our results together suggest that the LDHA/NDRG3 axis may play a critical role in adaptive cardiomyocyte growth in response to hemodynamic stress. Dai et al. find that LDHA is significantly increased in the heart under hemodynamic stress, and cardiomyocyte-specific deletion of LDHA leads to severe cardiac dysfunction in response to pressure overload. LDHA may govern adaptive growth through elevation of NDRG3 and activation of ERK.
Collapse
|
19
|
He Y, Huang W, Zhang C, Chen L, Xu R, Li N, Wang F, Han L, Yang M, Zhang D. Energy metabolism disorders and potential therapeutic drugs in heart failure. Acta Pharm Sin B 2021; 11:1098-1116. [PMID: 34094822 PMCID: PMC8144890 DOI: 10.1016/j.apsb.2020.10.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/27/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023] Open
Abstract
Heart failure (HF) is a global public health problem with high morbidity and mortality. A large number of studies have shown that HF is caused by severe energy metabolism disorders, which result in an insufficient heart energy supply. This deficiency causes cardiac pump dysfunction and systemic energy metabolism failure, which determine the development of HF and recovery of heart. Current HF therapy acts by reducing heart rate and cardiac preload and afterload, treating the HF symptomatically or delaying development of the disease. Drugs aimed at cardiac energy metabolism have not yet been developed. In this review, we outline the main characteristics of cardiac energy metabolism in healthy hearts, changes in metabolism during HF, and related pathways and targets of energy metabolism. Finally, we discuss drugs that improve cardiac function via energy metabolism to provide new research ideas for the development and application of drugs for treating HF.
Collapse
|
20
|
Therapeutic Manipulation of Myocardial Metabolism: JACC State-of-the-Art Review. J Am Coll Cardiol 2021; 77:2022-2039. [PMID: 33888253 DOI: 10.1016/j.jacc.2021.02.057] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/16/2021] [Indexed: 12/26/2022]
Abstract
The mechanisms responsible for the positive and unexpected cardiovascular effects of sodium-glucose cotransporter-2 inhibitors and glucagon-like peptide-1 receptor agonists in patients with type 2 diabetes remain to be defined. It is likely that some of the beneficial cardiac effects of these antidiabetic drugs are mediated, in part, by altered myocardial metabolism. Common cardiometabolic disorders, including the metabolic (insulin resistance) syndrome and type 2 diabetes, are associated with altered substrate utilization and energy transduction by the myocardium, predisposing to the development of heart disease. Thus, the failing heart is characterized by a substrate shift toward glycolysis and ketone oxidation in an attempt to meet the high energetic demand of the constantly contracting heart. This review examines the metabolic pathways and clinical implications of myocardial substrate utilization in the normal heart and in cardiometabolic disorders, and discusses mechanisms by which antidiabetic drugs and metabolic interventions improve cardiac function in the failing heart.
Collapse
|
21
|
Geraets IME, Coumans WA, Strzelecka A, Schönleitner P, Antoons G, Schianchi F, Willemars MMA, Kapsokalyvas D, Glatz JFC, Luiken JJFP, Nabben M. Metabolic Interventions to Prevent Hypertrophy-Induced Alterations in Contractile Properties In Vitro. Int J Mol Sci 2021; 22:ijms22073620. [PMID: 33807195 PMCID: PMC8037191 DOI: 10.3390/ijms22073620] [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: 03/02/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022] Open
Abstract
(1) Background: The exact mechanism(s) underlying pathological changes in a heart in transition to hypertrophy and failure are not yet fully understood. However, alterations in cardiac energy metabolism seem to be an important contributor. We characterized an in vitro model of adrenergic stimulation-induced cardiac hypertrophy for studying metabolic, structural, and functional changes over time. Accordingly, we investigated whether metabolic interventions prevent cardiac structural and functional changes; (2) Methods: Primary rat cardiomyocytes were treated with phenylephrine (PE) for 16 h, 24 h, or 48 h, whereafter hypertrophic marker expression, protein synthesis rate, glucose uptake, and contractile function were assessed; (3) Results: 24 h PE treatment increased expression of hypertrophic markers, phosphorylation of hypertrophy-related signaling kinases, protein synthesis, and glucose uptake. Importantly, the increased glucose uptake preceded structural and functional changes, suggesting a causal role for metabolism in the onset of PE-induced hypertrophy. Indeed, PE treatment in the presence of a PAN-Akt inhibitor or of a GLUT4 inhibitor dipyridamole prevented PE-induced increases in cellular glucose uptake and ameliorated PE-induced contractile alterations; (4) Conclusions: Pharmacological interventions, forcing substrate metabolism away from glucose utilization, improved contractile properties in PE-treated cardiomyocytes, suggesting that targeting glucose uptake, independent from protein synthesis, forms a promising strategy to prevent hypertrophy and hypertrophy-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Ilvy M. E. Geraets
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (I.M.E.G.); (W.A.C.); (A.S.); (F.S.); (M.M.A.W.); (D.K.); (J.F.C.G.); (J.J.F.P.L.)
| | - Will A. Coumans
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (I.M.E.G.); (W.A.C.); (A.S.); (F.S.); (M.M.A.W.); (D.K.); (J.F.C.G.); (J.J.F.P.L.)
| | - Agnieszka Strzelecka
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (I.M.E.G.); (W.A.C.); (A.S.); (F.S.); (M.M.A.W.); (D.K.); (J.F.C.G.); (J.J.F.P.L.)
| | - Patrick Schönleitner
- Departments of Physiology, Maastricht University, 6200-MD Maastricht, The Netherlands; (P.S.); (G.A.)
- CARIM School for Cardiovascular Diseases, Maastricht University, 6200-MD Maastricht, The Netherlands
| | - Gudrun Antoons
- Departments of Physiology, Maastricht University, 6200-MD Maastricht, The Netherlands; (P.S.); (G.A.)
- CARIM School for Cardiovascular Diseases, Maastricht University, 6200-MD Maastricht, The Netherlands
| | - Francesco Schianchi
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (I.M.E.G.); (W.A.C.); (A.S.); (F.S.); (M.M.A.W.); (D.K.); (J.F.C.G.); (J.J.F.P.L.)
| | - Myrthe M. A. Willemars
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (I.M.E.G.); (W.A.C.); (A.S.); (F.S.); (M.M.A.W.); (D.K.); (J.F.C.G.); (J.J.F.P.L.)
| | - Dimitrios Kapsokalyvas
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (I.M.E.G.); (W.A.C.); (A.S.); (F.S.); (M.M.A.W.); (D.K.); (J.F.C.G.); (J.J.F.P.L.)
| | - Jan F. C. Glatz
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (I.M.E.G.); (W.A.C.); (A.S.); (F.S.); (M.M.A.W.); (D.K.); (J.F.C.G.); (J.J.F.P.L.)
- CARIM School for Cardiovascular Diseases, Maastricht University, 6200-MD Maastricht, The Netherlands
| | - Joost J. F. P. Luiken
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (I.M.E.G.); (W.A.C.); (A.S.); (F.S.); (M.M.A.W.); (D.K.); (J.F.C.G.); (J.J.F.P.L.)
- Department of Clinical Genetics, Maastricht University Medical Center, 6200-MD Maastricht, The Netherlands
| | - Miranda Nabben
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200-MD Maastricht, The Netherlands; (I.M.E.G.); (W.A.C.); (A.S.); (F.S.); (M.M.A.W.); (D.K.); (J.F.C.G.); (J.J.F.P.L.)
- CARIM School for Cardiovascular Diseases, Maastricht University, 6200-MD Maastricht, The Netherlands
- Department of Clinical Genetics, Maastricht University Medical Center, 6200-MD Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-43-3881998
| |
Collapse
|
22
|
Pepin ME, Ha CM, Potter LA, Bakshi S, Barchue JP, Haj Asaad A, Pogwizd SM, Pamboukian SV, Hidalgo BA, Vickers SM, Wende AR. Racial and socioeconomic disparity associates with differences in cardiac DNA methylation among men with end-stage heart failure. Am J Physiol Heart Circ Physiol 2021; 320:H2066-H2079. [PMID: 33769919 PMCID: PMC8163657 DOI: 10.1152/ajpheart.00036.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Heart failure (HF) is a multifactorial syndrome that remains a leading cause of worldwide morbidity. Despite its high prevalence, only half of patients with HF respond to guideline-directed medical management, prompting therapeutic efforts to confront the molecular underpinnings of its heterogeneity. In the current study, we examined epigenetics as a yet unexplored source of heterogeneity among patients with end-stage HF. Specifically, a multicohort-based study was designed to quantify cardiac genome-wide cytosine-p-guanine (CpG) methylation of cardiac biopsies from male patients undergoing left ventricular assist device (LVAD) implantation. In both pilot (n = 11) and testing (n = 31) cohorts, unsupervised multidimensional scaling of genome-wide myocardial DNA methylation exhibited a bimodal distribution of CpG methylation found largely to occur in the promoter regions of metabolic genes. Among the available patient attributes, only categorical self-identified patient race could delineate this methylation signature, with African American (AA) and Caucasian American (CA) samples clustering separately. Because race is a social construct, and thus a poor proxy of human physiology, extensive review of medical records was conducted, but ultimately failed to identify covariates of race at the time of LVAD surgery. By contrast, retrospective analysis exposed a higher all-cause mortality among AA (56.3%) relative to CA (16.7%) patients at 2 yr following LVAD placement (P = 0.03). Geocoding-based approximation of patient demographics uncovered disparities in income levels among AA relative to CA patients. Although additional studies are needed, the current analysis implicates cardiac DNA methylation as a previously unrecognized indicator of socioeconomic disparity in human heart failure outcomes. NEW & NOTEWORTHY A bimodal signature of cardiac DNA methylation in heart failure corresponds with racial differences in all-cause mortality following mechanical circulatory support. Racial differences in promoter methylation disproportionately affect metabolic signaling pathways. Socioeconomic factors are associated with racial differences in the cardiac methylome among men with end-stage heart failure. Listen to this article’s corresponding podcast at https://ajpheart.podbean.com/e/racial-socioeconomic-determinants-of-the-cardiac-epigenome/.
Collapse
Affiliation(s)
- Mark E Pepin
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama.,Institute for Experimental Cardiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Chae-Myeong Ha
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Luke A Potter
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sayan Bakshi
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Joseph P Barchue
- Division of Cardiovascular Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ayman Haj Asaad
- Division of Cardiovascular Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Steven M Pogwizd
- Division of Cardiovascular Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Salpy V Pamboukian
- Division of Cardiovascular Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bertha A Hidalgo
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Selwyn M Vickers
- Office of the Dean and Senior Vice President For Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
23
|
Mann PA, Lehrke M. Cardiac substrate utilization in heart failure: Where is the relevance of SGLT2 inhibition? J Thorac Cardiovasc Surg 2021; 164:895-899. [DOI: 10.1016/j.jtcvs.2021.02.092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/14/2022]
|
24
|
Dougherty BV, Rawls KD, Kolling GL, Vinnakota KC, Wallqvist A, Papin JA. Identifying functional metabolic shifts in heart failure with the integration of omics data and a heart-specific, genome-scale model. Cell Rep 2021; 34:108836. [PMID: 33691118 DOI: 10.1016/j.celrep.2021.108836] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 01/07/2021] [Accepted: 02/17/2021] [Indexed: 11/28/2022] Open
Abstract
In diseased states, the heart can shift to use different carbon substrates, measured through changes in uptake of metabolites by imaging methods or blood metabolomics. However, it is not known whether these measured changes are a result of transcriptional changes or external factors. Here, we explore transcriptional changes in late-stage heart failure using publicly available data integrated with a model of heart metabolism. First, we present a heart-specific genome-scale metabolic network reconstruction (GENRE), iCardio. Next, we demonstrate the utility of iCardio in interpreting heart failure gene expression data by identifying tasks inferred from differential expression (TIDEs), which represent metabolic functions associated with changes in gene expression. We identify decreased gene expression for nitric oxide (NO) and N-acetylneuraminic acid (Neu5Ac) synthesis as common metabolic markers of heart failure. The methods presented here for constructing a tissue-specific model and identifying TIDEs can be extended to multiple tissues and diseases of interest.
Collapse
Affiliation(s)
- Bonnie V Dougherty
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Kristopher D Rawls
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Glynis L Kolling
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908, USA
| | - Kalyan C Vinnakota
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD 21702, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD 21702, USA
| | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908, USA; Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
25
|
Sourdon J, Facchin C, Certain A, Viel T, Robin B, Lager F, Marchiol C, Balvay D, Yoganathan T, Favier J, Tharaux PL, Dhaun N, Renault G, Tavitian B. Sunitinib-induced cardiac hypertrophy and the endothelin axis. Am J Cancer Res 2021; 11:3830-3838. [PMID: 33664864 PMCID: PMC7914356 DOI: 10.7150/thno.49837] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022] Open
Abstract
Anti-angiogenics drugs in clinical use for cancer treatment induce cardiotoxic side effects. The endothelin axis is involved in hypertension and cardiac remodelling, and addition of an endothelin receptor antagonist to the anti-angiogenic sunitinib was shown to reduce cardiotoxicity of sunitinib in mice. Here, we explored further the antidote effect of the endothelin receptor antagonist macitentan in sunitinib-treated animals on cardiac remodeling. Methods: Tumor-bearing mice treated per os daily by sunitinib or vehicle were imaged before and after 1, 3 and 6 weeks of treatment by positron emission tomography using [18F]fluorodeoxyglucose and by echocardiography. Non-tumor-bearing animals were randomly assigned to be treated per os daily by vehicle or sunitinib or macitentan or sunitinib+macitentan, and imaged by echocardiography after 5 weeks. Hearts were harvested for histology and molecular analysis at the end of in vivo exploration. Results: Sunitinib treatment increases left ventricular mass and ejection fraction and induces cardiac fibrosis. Sunitinib also induces an early increase in cardiac uptake of [18F]fluorodeoxyglucose, which is significantly correlated with increased left ventricular mass at the end of treatment. Co-administration of macitentan prevents sunitinib-induced hypertension, increase in ejection fraction and cardiac fibrosis, but fails to prevent increase of the left ventricular mass. Conclusion: Early metabolic changes predict sunitinib-induced cardiac remodeling. Endothelin blockade can prevent some but not all cardiotoxic side-effects of sunitinib, in particular left ventricle hypertrophy that appears to be induced by sunitinib through an endothelin-independent mechanism.
Collapse
|
26
|
Cardiac 1H MR spectroscopy: development of the past five decades and future perspectives. Heart Fail Rev 2021; 26:839-859. [PMID: 33409666 DOI: 10.1007/s10741-020-10059-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/23/2020] [Indexed: 01/01/2023]
Abstract
Continued advances in laboratory medicine are required to realize the potential of individualized medicine to impact common cardiovascular diseases. Magnetic resonance imaging (MRI) and spectroscopy (MRS) techniques have advanced over recent years and offer unique, powerful insights into cardiac anatomic and metabolic changes, respectively, occurring in both nascent and advanced heart disease. Although numerous MRI-based in vivo diagnostics are already used in routine clinical practice and more are anticipated, MRS has been less incorporated into routine clinical practice. Given the ability of 1H MRS to identify and quantify specific molecules with high sensitivity and specificity, its potential utility should be successfully transition from "bench-to-bedside" is tantalizing. The present review will highlight the development of 1H MRS techniques for cardiac applications, observations in seminal studies with 1H MRS, and the prospects and challenges for widespread application in patients with cardiovascular disease.
Collapse
|
27
|
Gullberg GT, Shrestha UM, Seo Y. PET imaging of glucose and fatty acid metabolism for NAFLD patients. J Nucl Cardiol 2020; 27:1689-1697. [PMID: 30547298 PMCID: PMC8356561 DOI: 10.1007/s12350-018-01532-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Grant T Gullberg
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA.
| | - Uttam M Shrestha
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| |
Collapse
|
28
|
Ghannam M, Yun HJ, Ficaro EP, Ghanbari H, Lazarus JJ, Konerman M, Shah RV, Weinberg R, Corbett JR, Oral H, Murthy VL. Multiparametric assessment of left atrial remodeling using 18F-FDG PET/CT cardiac imaging: A pilot study. J Nucl Cardiol 2020; 27:1547-1562. [PMID: 30191438 PMCID: PMC6411463 DOI: 10.1007/s12350-018-1429-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 08/08/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Left atrial (LA) remodeling is associated with structural, electric, and metabolic LA changes. Integrated evaluation of these features in vivo is lacking. METHODS Patients undergoing 18F-fluorodeoxyglucose (FDG) PET-CT during a hyperinsulinemic-euglycemic clamp were classified into sinus rhythm (SR), paroxysmal AF (PAF), and persistent AF (PerAF). The LA was semiautomatically segmented, and global FDG uptake was quantified using standardized uptake values (SUVmax and SUVmean) in gated, attenuation-corrected images and normalized to LA blood pool activity. Regression was used to relate FDG data to AF burden and critical patient factors. Continuous variables were compared using t-tests or Mann-Whitney tests. RESULTS 117 patients were included (76% men, age 66.4 ± 11.0, ejection fraction (EF) 25[22-35]%) including those with SR (n = 48), PAF (n = 55), and PerAF (n = 14). Patients with any AF had increased SUVmean (2.3[1.5-2.4] vs 2.0[1.5-2.5], P = 0.006), SUVmax (4.4[2.8-6.7] vs 3.2[2.3-4.3], P < 0.001), uptake coefficient of variation (CoV) 0.28[0.22-0.40] vs 0.25[0.2-0.33], P < 0.001), and hypometabolic scar (32%[14%-53%] vs 16.5%[0%-38.5%], P = 0.01). AF burden correlated with increased SUVmean, SUVmax, CoV, and scar independent of age, gender, EF, or LA size (P < 0.03 for all). CONCLUSIONS LA structure and metabolism can be assessed using FDG PET/CT. Greater AF burden correlates with the increased LA metabolism and scar.
Collapse
Affiliation(s)
- Michael Ghannam
- Department of Medicine (Division of Cardiovascular Medicine), University of Michigan, 1500 E. Medical Center Dr. SPC 5873, Ann Arbor, MI, 48109, USA
| | - Hong Jun Yun
- Department of Medicine (Division of Cardiovascular Medicine), University of Michigan, 1500 E. Medical Center Dr. SPC 5873, Ann Arbor, MI, 48109, USA
| | - Edward P Ficaro
- INVIA Medical Imaging Solutions, Ann Arbor, MI, USA
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Hamid Ghanbari
- Department of Medicine (Division of Cardiovascular Medicine), University of Michigan, 1500 E. Medical Center Dr. SPC 5873, Ann Arbor, MI, 48109, USA
| | - John J Lazarus
- Department of Medicine (Division of Cardiovascular Medicine), University of Michigan, 1500 E. Medical Center Dr. SPC 5873, Ann Arbor, MI, 48109, USA
| | - Matthew Konerman
- Department of Medicine (Division of Cardiovascular Medicine), University of Michigan, 1500 E. Medical Center Dr. SPC 5873, Ann Arbor, MI, 48109, USA
| | - Ravi V Shah
- Department of Medicine (Division of Cardiovascular Medicine), Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard Weinberg
- Department of Medicine (Division of Cardiovascular Medicine), University of Michigan, 1500 E. Medical Center Dr. SPC 5873, Ann Arbor, MI, 48109, USA
| | - James R Corbett
- Department of Medicine (Division of Cardiovascular Medicine), University of Michigan, 1500 E. Medical Center Dr. SPC 5873, Ann Arbor, MI, 48109, USA
- INVIA Medical Imaging Solutions, Ann Arbor, MI, USA
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Hakan Oral
- Department of Medicine (Division of Cardiovascular Medicine), University of Michigan, 1500 E. Medical Center Dr. SPC 5873, Ann Arbor, MI, 48109, USA
| | - Venkatesh L Murthy
- Department of Medicine (Division of Cardiovascular Medicine), University of Michigan, 1500 E. Medical Center Dr. SPC 5873, Ann Arbor, MI, 48109, USA.
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
29
|
Geraets IME, Glatz JFC, Luiken JJFP, Nabben M. Pivotal role of membrane substrate transporters on the metabolic alterations in the pressure-overloaded heart. Cardiovasc Res 2020; 115:1000-1012. [PMID: 30938418 DOI: 10.1093/cvr/cvz060] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/04/2019] [Accepted: 03/07/2019] [Indexed: 12/16/2022] Open
Abstract
Cardiac pressure overload (PO), such as caused by aortic stenosis and systemic hypertension, commonly results in cardiac hypertrophy and may lead to the development of heart failure. PO-induced heart failure is among the leading causes of death worldwide, but its pathological origin remains poorly understood. Metabolic alterations are proposed to be an important contributor to PO-induced cardiac hypertrophy and failure. While the healthy adult heart mainly uses long-chain fatty acids (FAs) and glucose as substrates for energy metabolism and to a lesser extent alternative substrates, i.e. lactate, ketone bodies, and amino acids (AAs), the pressure-overloaded heart is characterized by a shift in energy metabolism towards a greater reliance on glycolysis and alternative substrates. A key-governing kinetic step of both FA and glucose fluxes is at the level of their substrate-specific membrane transporters. The relative presence of these transporters in the sarcolemma determines the cardiac substrate preference. Whether the cardiac utilization of alternative substrates is also governed by membrane transporters is not yet known. In this review, we discuss current insight into the role of membrane substrate transporters in the metabolic alterations occurring in the pressure-overloaded heart. Given the increasing evidence of a role for alternative substrates in these metabolic alterations, there is an urgent need to disclose the key-governing kinetic steps in their utilization as well. Taken together, membrane substrate transporters emerge as novel targets for metabolic interventions to prevent or treat PO-induced heart failure.
Collapse
Affiliation(s)
- Ilvy M E Geraets
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, MD Maastricht, The Netherlands
| | - Jan F C Glatz
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, MD Maastricht, The Netherlands
| | - Joost J F P Luiken
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, MD Maastricht, The Netherlands
| | - Miranda Nabben
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, MD Maastricht, The Netherlands
| |
Collapse
|
30
|
Abstract
Heart failure (HF) is a clinical syndrome caused by a decline in cardiac systolic or diastolic function, which leaves the heart unable to pump enough blood to meet the normal physiological requirements of the human body. It is a serious disease burden worldwide affecting nearly 23 million patients. The concept that heart failure is "an engine out of fuel" has been generally accepted and metabolic remodeling has been recognized as an important aspect of this condition; it is characterized by defects in energy production and changes in metabolic pathways involved in the regulation of essential cellular functions such as the process of substrate utilization, the tricarboxylic acid cycle, oxidative phosphorylation, and high-energy phosphate metabolism. Advances in second-generation sequencing, proteomics, and metabolomics have made it possible to perform comprehensive tests on genes and metabolites that are crucial in the process of HF, thereby providing a clearer and comprehensive understanding of metabolic remodeling during HF. In recent years, new metabolic changes such as ketone bodies and branched-chain amino acids were demonstrated as alternative substrates in end-stage HF. This systematic review focuses on changes in metabolic substrate utilization during the progression of HF and the underlying regulatory mechanisms. Accordingly, the conventional concepts of metabolic remodeling characteristics are reviewed, and the latest developments, particularly multi-omics studies, are compiled.
Collapse
Affiliation(s)
- Liang Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital; National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), 167A Beilishi Road, Xi Cheng District, Beijing, 100037, People's Republic of China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital; National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), 167A Beilishi Road, Xi Cheng District, Beijing, 100037, People's Republic of China.
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital; National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), 167A Beilishi Road, Xi Cheng District, Beijing, 100037, People's Republic of China
| |
Collapse
|
31
|
Glatz JFC, Nabben M, Young ME, Schulze PC, Taegtmeyer H, Luiken JJFP. Re-balancing cellular energy substrate metabolism to mend the failing heart. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165579. [PMID: 31678200 PMCID: PMC7586321 DOI: 10.1016/j.bbadis.2019.165579] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/16/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022]
Abstract
Fatty acids and glucose are the main substrates for myocardial energy provision. Under physiologic conditions, there is a distinct and finely tuned balance between the utilization of these substrates. Using the non-ischemic heart as an example, we discuss that upon stress this substrate balance is upset resulting in an over-reliance on either fatty acids or glucose, and that chronic fuel shifts towards a single type of substrate appear to be linked with cardiac dysfunction. These observations suggest that interventions aimed at re-balancing a tilted substrate preference towards an appropriate mix of substrates may result in restoration of cardiac contractile performance. Examples of manipulating cellular substrate uptake as a means to re-balance fuel supply, being associated with mended cardiac function underscore this concept. We also address the molecular mechanisms underlying the apparent need for a fatty acid-glucose fuel balance. We propose that re-balancing cellular fuel supply, in particular with respect to fatty acids and glucose, may be an effective strategy to treat the failing heart.
Collapse
Affiliation(s)
- Jan F C Glatz
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands.
| | - Miranda Nabben
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands
| | - Martin E Young
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - P Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany
| | - Heinrich Taegtmeyer
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joost J F P Luiken
- Department of Genetics & Cell Biology, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
32
|
Barton GP, Macdonald EB, Goss KN, Eldridge MW, Fain SB. Measuring the link between cardiac mechanical function and metabolism during hyperpolarized 13C-pyruvate magnetic resonance experiments. Magn Reson Imaging 2020; 68:9-17. [PMID: 31978518 PMCID: PMC7131884 DOI: 10.1016/j.mri.2020.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/19/2019] [Accepted: 01/19/2020] [Indexed: 10/25/2022]
Abstract
PURPOSE The goal of this study was to develop a methodology to investigate the relationship between contractile function and hyperpolarized (HP) [1-13C]pyruvate metabolism in a small animal model. To achieve sufficient signal from HP 13C compounds, HP 13C MRS/MRSI has required relatively large infusion volumes relative to the total blood volume in small animal models, which may affect cardiac function. METHODS Eight female Sprague Dawley rats were imaged on a 4.7T scanner with a dual tuned 1H/13C volume coil. ECG and respiratory gated k-t spiral MRSI and an IDEAL based reconstruction to determine [1-13C]pyruvate metabolism in the myocardium. This was coupled with 1H cine MRI to determine ventricular volumes and mechanical function pre- and post-infusion of [1-13C]pyruvate. For comparison to the [1-13C]pyruvate experiments, three female Sprague Dawley rats were imaged with 1H cine MRI to determine myocardial function pre- and post-saline infusion. RESULTS We demonstrated significant changes in cardiac contractile function between pre- and post-infusion of [1-13C]pyruvate. Specifically, there was an increase in end-diastolic volume (EDV), stroke volume (SV), and ejection fraction (EF). Additionally, the ventricular vascular coupling ratio (VVCR) showed an improvement after [1-13C]pyruvate infusion, indicating increased systolic performance due to an increased arterial load. There was a moderate to strong relationship between the downstream metabolic conversion of pyruvate to bicarbonate and a strong relationship between the conversion of pyruvate to lactate and the cardiac mechanical function response. CONCLUSION The infusion of [1-13C]pyruvate resulted in demonstrable increases in contractile function which was related to pyruvate conversion to bicarbonate and lactate. The combined effects of the infusion volume and inotropic effects of pyruvate metabolism likely explains the augmentation in myocardial mechanical function seen in these experiments. Given the relationship between pyruvate metabolism and contractile function observed in this study, this methodological approach may be utilized to better understand cardiac metabolic and functional remodeling in heart disease.
Collapse
Affiliation(s)
| | | | - Kara N Goss
- Medicine University of Wisconsin, Madison, WI, USA; Pediatrics University of Wisconsin, Madison, WI, USA
| | - Marlowe W Eldridge
- Pediatrics University of Wisconsin, Madison, WI, USA; Biomedical Engineering, University of Wisconsin, Madison, WI, USA
| | - Sean B Fain
- Medical Physics, University of Wisconsin, Madison, WI, USA; Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Radiology, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
33
|
Sampath S, Parimal AS, Huang W, Manigbas E, Gsell W, Chang MML, Qiu A, Jacobsen K, Evelhoch JL, Chin CL. Quantification of regional myocardial mean intracellular water lifetime: A nonhuman primate study in myocardial stress. NMR IN BIOMEDICINE 2020; 33:e4248. [PMID: 31977123 DOI: 10.1002/nbm.4248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/01/2019] [Accepted: 12/02/2019] [Indexed: 06/10/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is typically associated with early metabolic remodeling. Noninvasive imaging biomarkers that reflect these changes will be crucial in determining responses to early drug interventions in these patients. Mean intracellular water lifetime (τi ) has been shown to be partially inversely related to Na, K-ATPase transporter activity and may thus provide insight into the metabolic status in HFpEF patients. Here, we aim to perform regional quantification of τi using dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) in the nonhuman primate (NHP) heart and evaluate its region-specific variations under conditions of myocardial stress in the context of perturbed myocardial function. Cardiac stress was induced in seven naïve cynomolgus macaques using a dobutamine stepwise infusion protocol. All animals underwent 3 T cardiac dual-bolus DCE and tagging MRI experiments. The shutter-speed model was employed to quantify regional τi from the DCE-MR images. Additionally, τi values were correlated with myocardial strains. During cardiac stress, there was a significant decrease in global τi (192.9 ± 76.3 ms vs 321.6 ± 70 ms at rest, P < 0.05) in the left ventricle, together with an increase in global peak circumferential strain (-15.4% ± 2.7% vs -10.1% ± 2.9% at rest, P < 0.05). Specifically, slice-level analysis further revealed that a greater significant decrease in mean τi was observed in the apical region (ΔτI = 182.4 ms) compared with the basal (Δτi = 113.2 ms) and midventricular regions (Δτi = 108.4 ms). Regional analysis revealed that there was a greater significant decrease in mean τi in the anterior (Δτi = 243.9 ms) and antero-lateral (Δτi = 177.2 ms) regions. In the inferior and infero-septal regions, although a decrease in τi was observed, it was not significant. Whole heart regional quantification of τi is feasible using DCE-MRI. τi is sensitive to regional changes in metabolic state during cardiac stress, and its value correlates with strain.
Collapse
Affiliation(s)
| | | | - Wei Huang
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, Oregon, United States
| | - Elaine Manigbas
- Imaging, Maccine Pte. Ltd., Singapore
- Comparative Medicine Imaging Facility, National University of Singapore, Singapore
| | - Willy Gsell
- Imaging, Maccine Pte. Ltd., Singapore
- Biomedical MRI, Department of Imaging and Pathology, Molecular Small Imaging Center, Leuven, Belgium
| | | | - Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | | | - Jeffrey L Evelhoch
- Translational Biomarkers, MRL, Merck & Co., Inc, West Point, Pennsylvania
| | | |
Collapse
|
34
|
Li J, Kemp BA, Howell NL, Massey J, Mińczuk K, Huang Q, Chordia MD, Roy RJ, Patrie JT, Davogustto GE, Kramer CM, Epstein FH, Carey RM, Taegtmeyer H, Keller SR, Kundu BK. Metabolic Changes in Spontaneously Hypertensive Rat Hearts Precede Cardiac Dysfunction and Left Ventricular Hypertrophy. J Am Heart Assoc 2020; 8:e010926. [PMID: 30764689 PMCID: PMC6405673 DOI: 10.1161/jaha.118.010926] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Sustained pressure overload leads to changes in cardiac metabolism, function, and structure. Both time course and causal relationships between these changes are not fully understood. Therefore, we studied spontaneously hypertensive rats (SHR) during early hypertension development and compared them to control Wistar Kyoto rats. Methods and Results We serially evaluated myocardial glucose uptake rates (Ki) with dynamic 2‐[18F] fluoro‐2‐deoxy‐D‐glucose positron emission tomography, and ejection fraction and left ventricular mass to body weight ratios with cardiac magnetic resonance imaging in vivo, determined glucose uptake and oxidation rates in isolated perfused hearts, and analyzed metabolites, mammalian target of rapamycin activity and endoplasmic reticulum stress in dissected hearts. When compared with Wistar Kyoto rats, SHR demonstrated increased glucose uptake rates (Ki) in vivo, and reduced ejection fraction as early as 2 months of age when hypertension was established. Isolated perfused SHR hearts showed increased glucose uptake and oxidation rates starting at 1 month. Cardiac metabolite analysis at 2 months of age revealed elevated pyruvate, fatty acyl‐ and branched chain amino acid‐derived carnitines, oxidative stress, and inflammation. Mammalian target of rapamycin activity increased in SHR beginning at 2 months. Left ventricular mass to body weight ratios and endoplasmic reticulum stress were elevated in 5 month‐old SHR. Conclusions Thus, in a genetic hypertension model, chronic cardiac pressure overload promptly leads to increased myocardial glucose uptake and oxidation, and to metabolite abnormalities. These coincide with, or precede, cardiac dysfunction while left ventricular hypertrophy develops only later. Myocardial metabolic changes may thus serve as early diagnostic markers for hypertension‐induced left ventricular hypertrophy.
Collapse
Affiliation(s)
- Jie Li
- 1 Department of Radiology and Medical Imaging University of Virginia Charlottesville VA
| | - Brandon A Kemp
- 2 Division of Endocrinology and Metabolism Department of Medicine University of Virginia Charlottesville VA
| | - Nancy L Howell
- 2 Division of Endocrinology and Metabolism Department of Medicine University of Virginia Charlottesville VA
| | - James Massey
- 1 Department of Radiology and Medical Imaging University of Virginia Charlottesville VA.,3 Department of Biomedical Engineering University of Virginia Charlottesville VA
| | - Krzysztof Mińczuk
- 1 Department of Radiology and Medical Imaging University of Virginia Charlottesville VA
| | - Qiao Huang
- 1 Department of Radiology and Medical Imaging University of Virginia Charlottesville VA
| | - Mahendra D Chordia
- 1 Department of Radiology and Medical Imaging University of Virginia Charlottesville VA
| | - R Jack Roy
- 1 Department of Radiology and Medical Imaging University of Virginia Charlottesville VA
| | - James T Patrie
- 4 Department of Public Health Sciences University of Virginia Charlottesville VA
| | - Giovanni E Davogustto
- 5 McGovern Medical School University of Texas Health Science Center in Houston Houston TX
| | - Christopher M Kramer
- 6 Department of Cardiovascular Medicine University of Virginia Charlottesville VA
| | - Frederick H Epstein
- 3 Department of Biomedical Engineering University of Virginia Charlottesville VA
| | - Robert M Carey
- 2 Division of Endocrinology and Metabolism Department of Medicine University of Virginia Charlottesville VA
| | - Heinrich Taegtmeyer
- 5 McGovern Medical School University of Texas Health Science Center in Houston Houston TX
| | - Susanna R Keller
- 2 Division of Endocrinology and Metabolism Department of Medicine University of Virginia Charlottesville VA
| | - Bijoy K Kundu
- 1 Department of Radiology and Medical Imaging University of Virginia Charlottesville VA.,3 Department of Biomedical Engineering University of Virginia Charlottesville VA.,7 Cardiovascular Research Center University of Virginia Charlottesville VA
| |
Collapse
|
35
|
Pitoulis FG, Terracciano CM. Heart Plasticity in Response to Pressure- and Volume-Overload: A Review of Findings in Compensated and Decompensated Phenotypes. Front Physiol 2020; 11:92. [PMID: 32116796 PMCID: PMC7031419 DOI: 10.3389/fphys.2020.00092] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/27/2020] [Indexed: 12/20/2022] Open
Abstract
The adult human heart has an exceptional ability to alter its phenotype to adapt to changes in environmental demand. This response involves metabolic, mechanical, electrical, and structural alterations, and is known as cardiac plasticity. Understanding the drivers of cardiac plasticity is essential for development of therapeutic agents. This is particularly important in contemporary cardiology, which uses treatments with peripheral effects (e.g., on kidneys, adrenal glands). This review focuses on the effects of different hemodynamic loads on myocardial phenotype. We examine mechanical scenarios of pressure- and volume overload, from the initial insult, to compensated, and ultimately decompensated stage. We discuss how different hemodynamic conditions occur and are underlined by distinct phenotypic and molecular changes. We complete the review by exploring how current basic cardiac research should leverage available cardiac models to study mechanical load in its different presentations.
Collapse
|
36
|
Hill BG, Shiva S, Ballinger S, Zhang J, Darley-Usmar VM. Bioenergetics and translational metabolism: implications for genetics, physiology and precision medicine. Biol Chem 2019; 401:3-29. [PMID: 31815377 PMCID: PMC6944318 DOI: 10.1515/hsz-2019-0268] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 06/24/2019] [Indexed: 12/25/2022]
Abstract
It is now becoming clear that human metabolism is extremely plastic and varies substantially between healthy individuals. Understanding the biochemistry that underlies this physiology will enable personalized clinical interventions related to metabolism. Mitochondrial quality control and the detailed mechanisms of mitochondrial energy generation are central to understanding susceptibility to pathologies associated with aging including cancer, cardiac and neurodegenerative diseases. A precision medicine approach is also needed to evaluate the impact of exercise or caloric restriction on health. In this review, we discuss how technical advances in assessing mitochondrial genetics, cellular bioenergetics and metabolomics offer new insights into developing metabolism-based clinical tests and metabolotherapies. We discuss informatics approaches, which can define the bioenergetic-metabolite interactome and how this can help define healthy energetics. We propose that a personalized medicine approach that integrates metabolism and bioenergetics with physiologic parameters is central for understanding the pathophysiology of diseases with a metabolic etiology. New approaches that measure energetics and metabolomics from cells isolated from human blood or tissues can be of diagnostic and prognostic value to precision medicine. This is particularly significant with the development of new metabolotherapies, such as mitochondrial transplantation, which could help treat complex metabolic diseases.
Collapse
Affiliation(s)
- Bradford G. Hill
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, KY 40202
| | - Sruti Shiva
- Department of Pharmacology & Chemical Biology, Vascular Medicine Institute, Center for Metabolism & Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, PA 15143
| | - Scott Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Veteran Affairs Medical Center, Birmingham, AL 35294
| | - Victor M. Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
37
|
Barton GP, Vildberg L, Goss K, Aggarwal N, Eldridge M, McMillan AB. Simultaneous determination of dynamic cardiac metabolism and function using PET/MRI. J Nucl Cardiol 2019; 26:1946-1957. [PMID: 29717407 PMCID: PMC7851880 DOI: 10.1007/s12350-018-1287-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 04/13/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cardiac metabolic changes in heart disease precede overt contractile dysfunction. However, metabolism and function are not typically assessed together in clinical practice. The purpose of this study was to develop a cardiac positron emission tomography/magnetic resonance (PET/MR) stress test to assess the dynamic relationship between contractile function and metabolism in a preclinical model. METHODS Following an overnight fast, healthy pigs (45-50 kg) were anesthetized and mechanically ventilated. 18F-fluorodeoxyglucose (18F-FDG) solution was administered intravenously at a constant rate of 0.01 mL/s for 60 minutes. A cardiac PET/MR stress test was performed using normoxic gas (FIO2 = .209) and hypoxic gas (FIO2 = .12). Simultaneous cardiac imaging was performed on an integrated 3T PET/MR scanner. RESULTS Hypoxic stress induced a significant increase in heart rate, cardiac output, left ventricular (LV) ejection fraction (EF), and peak torsion. There was a significant decline in arterial SpO2, LV end-diastolic and end-systolic volumes in hypoxia. Increased LV systolic function was coupled with an increase in myocardial FDG uptake (Ki) during hypoxic stress. CONCLUSION PET/MR with continuous FDG infusion captures dynamic changes in both cardiac metabolism and contractile function. This technique warrants evaluation in human cardiac disease for assessment of subtle functional and metabolic abnormalities.
Collapse
Affiliation(s)
- Gregory P Barton
- Department of Pediatrics, UW School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave. H6/551 CSC, Madison, WI, 53792, USA.
- Rankin Laboratory of Pulmonary Medicine, University of Wisconsin-Madison, Madison, USA.
| | - Lauren Vildberg
- Department of Pediatrics, UW School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave. H6/551 CSC, Madison, WI, 53792, USA
- Rankin Laboratory of Pulmonary Medicine, University of Wisconsin-Madison, Madison, USA
| | - Kara Goss
- Department of Pediatrics, UW School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave. H6/551 CSC, Madison, WI, 53792, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, USA
- Rankin Laboratory of Pulmonary Medicine, University of Wisconsin-Madison, Madison, USA
| | - Niti Aggarwal
- Division of Cardiovascular Disease Department of Medicine, University of Wisconsin-Madison, Madison, USA
- Department of Radiology, University of Wisconsin-Madison, Madison, USA
| | - Marlowe Eldridge
- Department of Pediatrics, UW School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave. H6/551 CSC, Madison, WI, 53792, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, USA
- Rankin Laboratory of Pulmonary Medicine, University of Wisconsin-Madison, Madison, USA
| | - Alan B McMillan
- Department of Radiology, University of Wisconsin-Madison, Madison, USA
| |
Collapse
|
38
|
Harhous Z, Booz GW, Ovize M, Bidaux G, Kurdi M. An Update on the Multifaceted Roles of STAT3 in the Heart. Front Cardiovasc Med 2019; 6:150. [PMID: 31709266 PMCID: PMC6823716 DOI: 10.3389/fcvm.2019.00150] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 10/07/2019] [Indexed: 12/18/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a signaling molecule and transcription factor that plays important protective roles in the heart. The protection mediated by STAT3 is attributed to its genomic actions as a transcription factor and other non-genomic roles targeting mitochondrial function and autophagy. As a transcription factor, STAT3 upregulates genes that are anti-oxidative, anti-apoptotic, and pro-angiogenic, but suppresses anti-inflammatory and anti-fibrotic genes. Its suppressive effects on gene expression are achieved through competing with other transcription factors or cofactors. STAT3 is also linked to the modification of mRNA expression profiles in cardiac cells by inhibiting or inducing miRNA. In addition to these genomic roles, STAT3 is suggested to function protectively in mitochondria, where it regulates ROS production, in part by regulating the activities of the electron transport chain complexes, although our recent evidence calls this role into question. Nonetheless, STAT3 is a key player known to be activated in the cardioprotective ischemic conditioning protocols. Through these varied roles, STAT3 participates in various mechanisms that contribute to cardioprotection against different heart pathologies, including myocardial infarction, hypertrophy, diabetic cardiomyopathy, and peripartum cardiomyopathy. Understanding how STAT3 is involved in the protective mechanisms against these different cardiac pathologies could lead to novel therapeutic strategies to treat them.
Collapse
Affiliation(s)
- Zeina Harhous
- Laboratory of Experimental and Clinical Pharmacology, Faculty of Sciences, Doctoral School of Sciences and Technology, Lebanese University, Beirut, Lebanon
- Univ-Lyon, CarMeN Laboratory, INSERM 1060, INRA 1397, University Claude Bernard Lyon1, INSA Lyon, Oullins, France
- IHU OPeRa, Groupement Hospitalier EST, Bron, France
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center, Jackson, MS, United States
| | - Michel Ovize
- Univ-Lyon, CarMeN Laboratory, INSERM 1060, INRA 1397, University Claude Bernard Lyon1, INSA Lyon, Oullins, France
- IHU OPeRa, Groupement Hospitalier EST, Bron, France
| | - Gabriel Bidaux
- Univ-Lyon, CarMeN Laboratory, INSERM 1060, INRA 1397, University Claude Bernard Lyon1, INSA Lyon, Oullins, France
- IHU OPeRa, Groupement Hospitalier EST, Bron, France
| | - Mazen Kurdi
- Laboratory of Experimental and Clinical Pharmacology, Faculty of Sciences, Doctoral School of Sciences and Technology, Lebanese University, Beirut, Lebanon
| |
Collapse
|
39
|
Abstract
Metabolic pathways integrate to support tissue homeostasis and to prompt changes in cell phenotype. In particular, the heart consumes relatively large amounts of substrate not only to regenerate ATP for contraction but also to sustain biosynthetic reactions for replacement of cellular building blocks. Metabolic pathways also control intracellular redox state, and metabolic intermediates and end products provide signals that prompt changes in enzymatic activity and gene expression. Mounting evidence suggests that the changes in cardiac metabolism that occur during development, exercise, and pregnancy as well as with pathological stress (eg, myocardial infarction, pressure overload) are causative in cardiac remodeling. Metabolism-mediated changes in gene expression, metabolite signaling, and the channeling of glucose-derived carbon toward anabolic pathways seem critical for physiological growth of the heart, and metabolic inefficiency and loss of coordinated anabolic activity are emerging as proximal causes of pathological remodeling. This review integrates knowledge of different forms of cardiac remodeling to develop general models of how relationships between catabolic and anabolic glucose metabolism may fortify cardiac health or promote (mal)adaptive myocardial remodeling. Adoption of conceptual frameworks based in relational biology may enable further understanding of how metabolism regulates cardiac structure and function.
Collapse
Affiliation(s)
- Andrew A Gibb
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (A.A.G.)
| | - Bradford G Hill
- the Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville School of Medicine, KY (B.G.H.).
| |
Collapse
|
40
|
Pulinilkunnil T, Kienesberger P, Nagendran J. Editorial: Novel Concepts in Cardiac Energy Metabolism: From Biology to Disease. Front Cardiovasc Med 2019; 6:97. [PMID: 31355212 PMCID: PMC6640113 DOI: 10.3389/fcvm.2019.00097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/26/2019] [Indexed: 12/02/2022] Open
Affiliation(s)
- Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Petra Kienesberger
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Jeevan Nagendran
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
41
|
Affiliation(s)
- Diem H Tran
- 1 Division of Cardiology Department of Internal Medicine University of Texas Southwestern Medical Center Dallas TX
| | - Zhao V Wang
- 1 Division of Cardiology Department of Internal Medicine University of Texas Southwestern Medical Center Dallas TX
| |
Collapse
|
42
|
Wilmanns JC, Pandey R, Hon O, Chandran A, Schilling JM, Forte E, Wu Q, Cagnone G, Bais P, Philip V, Coleman D, Kocalis H, Archer SK, Pearson JT, Ramialison M, Heineke J, Patel HH, Rosenthal NA, Furtado MB, Costa MW. Metformin intervention prevents cardiac dysfunction in a murine model of adult congenital heart disease. Mol Metab 2019; 20:102-114. [PMID: 30482476 PMCID: PMC6358551 DOI: 10.1016/j.molmet.2018.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/06/2018] [Accepted: 11/10/2018] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Congenital heart disease (CHD) is the most frequent birth defect worldwide. The number of adult patients with CHD, now referred to as ACHD, is increasing with improved surgical and treatment interventions. However the mechanisms whereby ACHD predisposes patients to heart dysfunction are still unclear. ACHD is strongly associated with metabolic syndrome, but how ACHD interacts with poor modern lifestyle choices and other comorbidities, such as hypertension, obesity, and diabetes, is mostly unknown. METHODS We used a newly characterized mouse genetic model of ACHD to investigate the consequences and the mechanisms associated with combined obesity and ACHD predisposition. Metformin intervention was used to further evaluate potential therapeutic amelioration of cardiac dysfunction in this model. RESULTS ACHD mice placed under metabolic stress (high fat diet) displayed decreased left ventricular ejection fraction. Comprehensive physiological, biochemical, and molecular analysis showed that ACHD hearts exhibited early changes in energy metabolism with increased glucose dependence as main cardiac energy source. These changes preceded cardiac dysfunction mediated by exposure to high fat diet and were associated with increased disease severity. Restoration of metabolic balance by metformin administration prevented the development of heart dysfunction in ACHD predisposed mice. CONCLUSIONS This study reveals that early metabolic impairment reinforces heart dysfunction in ACHD predisposed individuals and diet or pharmacological interventions can be used to modulate heart function and attenuate heart failure. Our study suggests that interactions between genetic and metabolic disturbances ultimately lead to the clinical presentation of heart failure in patients with ACHD. Early manipulation of energy metabolism may be an important avenue for intervention in ACHD patients to prevent or delay onset of heart failure and secondary comorbidities. These interactions raise the prospect for a translational reassessment of ACHD presentation in the clinic.
Collapse
Affiliation(s)
- Julia C Wilmanns
- Australian Regenerative Medicine Institute, Monash University, Australia; Department of Cardiology and Angiology, Experimental Cardiology, Hannover Medical School, Germany
| | | | | | - Anjana Chandran
- Australian Regenerative Medicine Institute, Monash University, Australia
| | - Jan M Schilling
- VA San Diego Healthcare System and Department of Anesthesiology, University of California San Diego, USA
| | | | - Qizhu Wu
- Monash Biomedical Imaging, Monash University, Australia
| | - Gael Cagnone
- Department of Pharmacology, Research Center of CHU Sainte-Justine, Canada
| | | | | | | | | | - Stuart K Archer
- Monash Bioinformatics Platform, Monash University, Australia; Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Australia
| | - James T Pearson
- Monash Biomedical Imaging, Monash University, Australia; Department of Physiology, Monash University, Australia; National Cerebral & Cardiovascular Center, Suita 565-8565, Japan
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute, Monash University, Australia; Systems Biology Institute, Australia
| | - Joerg Heineke
- Department of Cardiology and Angiology, Experimental Cardiology, Hannover Medical School, Germany
| | - Hemal H Patel
- VA San Diego Healthcare System and Department of Anesthesiology, University of California San Diego, USA
| | - Nadia A Rosenthal
- The Jackson Laboratory, USA; Australian Regenerative Medicine Institute, Monash University, Australia; National Heart and Lung Institute, Imperial College London, W12 0NN, UK
| | - Milena B Furtado
- The Jackson Laboratory, USA; Australian Regenerative Medicine Institute, Monash University, Australia
| | - Mauro W Costa
- The Jackson Laboratory, USA; Australian Regenerative Medicine Institute, Monash University, Australia.
| |
Collapse
|
43
|
Lorén CE, Dahl CP, Do L, Almaas VM, Geiran OR, Mörner S, Hellman U. Low Molecular Mass Myocardial Hyaluronan in Human Hypertrophic Cardiomyopathy. Cells 2019; 8:cells8020097. [PMID: 30699940 PMCID: PMC6406527 DOI: 10.3390/cells8020097] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/11/2019] [Accepted: 01/23/2019] [Indexed: 12/23/2022] Open
Abstract
During the development of hypertrophic cardiomyopathy, the heart returns to fetal energy metabolism where cells utilize more glucose instead of fatty acids as a source of energy. Metabolism of glucose can increase synthesis of the extracellular glycosaminoglycan hyaluronan, which has been shown to be involved in the development of cardiac hypertrophy and fibrosis. The aim of this study was to investigate hyaluronan metabolism in cardiac tissue from patients with hypertrophic cardiomyopathy in relation to cardiac growth. NMR and qRT-PCR analysis of human cardiac tissue from hypertrophic cardiomyopathy patients and healthy control hearts showed dysregulated glucose and hyaluronan metabolism in the patients. Gas phase electrophoresis revealed a higher amount of low molecular mass hyaluronan and larger cardiomyocytes in cardiac tissue from patients with hypertrophic cardiomyopathy. Histochemistry showed high concentrations of hyaluronan around individual cardiomyocytes in hearts from hypertrophic cardiomyopathy patients. Experimentally, we could also observe accumulation of low molecular mass hyaluronan in cardiac hypertrophy in a rat model. In conclusion, the development of hypertrophic cardiomyopathy with increased glucose metabolism affected both hyaluronan molecular mass and amount. The process of regulating cardiomyocyte size seems to involve fragmentation of hyaluronan.
Collapse
Affiliation(s)
- Christina E Lorén
- Cardiology, Heart Centre, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden.
| | - Christen P Dahl
- Department of Cardiology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway.
- Department of Clinical Medicine, UiT, the Arctic University of Norway, 9019 Tromsø, Norway.
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway.
| | - Lan Do
- Cardiology, Heart Centre, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden.
| | - Vibeke M Almaas
- Department of Cardiology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway.
| | - Odd R Geiran
- Faculty of Medicine, University of Oslo, 0318 Oslo, Norway.
- Department of Thoracic and Cardiovascular Surgery, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway.
| | - Stellan Mörner
- Cardiology, Heart Centre, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden.
| | - Urban Hellman
- Cardiology, Heart Centre, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden.
| |
Collapse
|
44
|
Todica A, Beetz NL, Günther L, Zacherl MJ, Grabmaier U, Huber B, Bartenstein P, Brunner S, Lehner S. Monitoring of Cardiac Remodeling in a Mouse Model of Pressure-Overload Left Ventricular Hypertrophy with [ 18F]FDG MicroPET. Mol Imaging Biol 2019; 20:268-274. [PMID: 28852941 DOI: 10.1007/s11307-017-1114-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE This study aims to analyze the left ventricular function parameters, scar load, and hypertrophy in a mouse model of pressure-overload left ventricular (LV) hypertrophy over the course of 8 weeks using 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) micro-positron emission tomography (microPET) imaging. PROCEDURES LV hypertrophy was induced in C57BL/6 mice by transverse aortic constriction (TAC). Myocardial hypertrophy developed after 2-4 weeks. ECG-gated microPET scans with [18F]FDG were performed 4 and 8 weeks after surgery. The extent of fibrosis was measured by histopathologic analysis. LV function parameters and scar load were calculated using QGS®/QPS®. LV metabolic volume (LVMV) and percentage injected dose per gram were estimated by threshold-based analysis. RESULTS The fibrotic tissue volume increased significantly from 4 to 8 weeks after TAC (1.67 vs. 3.91 mm3; P = 0.044). There was a significant increase of the EDV (4 weeks: 54 ± 15 μl, 8 weeks: 79 ± 32 μl, P < 0.01) and LVMV (4 weeks: 222 ± 24 μl, 8 weeks: 276 ± 52 μl, P < 0.01) as well as a significant decrease of the LVEF (4 weeks: 56 ± 17 %, 8 weeks: 44 ± 20 %, P < 0.01). The increase of LVMV had a high predictive value regarding the amount of ex vivo measured fibrotic tissue (R = 0.905, P < 0.001). The myocardial metabolic defects increased within 4 weeks (P = 0.055) but only moderately correlated with the fibrosis volume (R = 0.502, P = 0.021). The increase in end-diastolic volume showed a positive correlation with the fibrosis at 8 weeks (R = 0.763, P = 0.017). CONCLUSIONS [18F]FDG-PET is applicable for serial in vivo monitoring of the TAC mouse model. Myocardial hypertrophy, the dilation of the left ventricle, and the decrease in LVEF could be reliably quantified over time, as well as the developing localized scar. The increase in volume over time is predictive of a high fibrosis load.
Collapse
Affiliation(s)
- Andrei Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Nick L Beetz
- Medical Department I-Cardiology, University Hospital, LMU Munich, Munich, Germany
| | - Lisa Günther
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Mathias J Zacherl
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Ulrich Grabmaier
- Medical Department I-Cardiology, University Hospital, LMU Munich, Munich, Germany
| | - Bruno Huber
- Medical Department I-Cardiology, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Stefan Brunner
- Medical Department I-Cardiology, University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Lehner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.,Ambulatory Healthcare Center Dr. Neumaier & Colleagues, Radiology, Nuclear Medicine, Radiation Therapy, Regensburg, Germany
| |
Collapse
|
45
|
Fulghum K, Hill BG. Metabolic Mechanisms of Exercise-Induced Cardiac Remodeling. Front Cardiovasc Med 2018; 5:127. [PMID: 30255026 PMCID: PMC6141631 DOI: 10.3389/fcvm.2018.00127] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/23/2018] [Indexed: 12/13/2022] Open
Abstract
Exercise has a myriad of physiological benefits that derive in part from its ability to improve cardiometabolic health. The periodic metabolic stress imposed by regular exercise appears fundamental in driving cardiovascular tissue adaptation. However, different types, intensities, or durations of exercise elicit different levels of metabolic stress and may promote distinct types of tissue remodeling. In this review, we discuss how exercise affects cardiac structure and function and how exercise-induced changes in metabolism regulate cardiac adaptation. Current evidence suggests that exercise typically elicits an adaptive, beneficial form of cardiac remodeling that involves cardiomyocyte growth and proliferation; however, chronic levels of extreme exercise may increase the risk for pathological cardiac remodeling or sudden cardiac death. An emerging theme underpinning acute as well as chronic cardiac adaptations to exercise is metabolic periodicity, which appears important for regulating mitochondrial quality and function, for stimulating metabolism-mediated exercise gene programs and hypertrophic kinase activity, and for coordinating biosynthetic pathway activity. In addition, circulating metabolites liberated during exercise trigger physiological cardiac growth. Further understanding of how exercise-mediated changes in metabolism orchestrate cell signaling and gene expression could facilitate therapeutic strategies to maximize the benefits of exercise and improve cardiac health.
Collapse
Affiliation(s)
- Kyle Fulghum
- Department of Medicine, Envirome Institute, Institute of Molecular Cardiology, Diabetes and Obesity Center, Louisville, KY, United States
- Department of Physiology, University of Louisville, Louisville, KY, United States
| | - Bradford G. Hill
- Department of Medicine, Envirome Institute, Institute of Molecular Cardiology, Diabetes and Obesity Center, Louisville, KY, United States
| |
Collapse
|
46
|
Bianchi VE. Impact of Nutrition on Cardiovascular Function. Curr Probl Cardiol 2018; 45:100391. [PMID: 30318107 DOI: 10.1016/j.cpcardiol.2018.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/31/2018] [Indexed: 12/11/2022]
Abstract
The metabolic sources of energy for myocardial contractility include mainly free fatty acids (FFA) for 95%, and in lesser amounts for 5% from glucose and minimal contributions from other substrates such lactate, ketones, and amino acids. However, myocardial efficiency is influenced by metabolic condition, overload, and ischemia. During cardiac stress, cardiomyocytes increase glucose oxidation and reduce FFA oxidation. In patients with ischemic coronary disease and heart failure, the low oxygen availability limits myocardial reliance on FFA and glucose utilization must increase. Although glucose uptake is fundamental to cardiomyocyte function, an excessive intracellular glucose level is detrimental. Insulin plays a fundamental role in maintaining myocardial efficiency and in reducing glycemia and inflammation; this is particularly evident in obese and type-2 diabetic patients. An excess of F availability increase fat deposition within cardiomyocytes and reduces glucose oxidation. In patients with high body mass index, a restricted diet or starvation have positive effects on cardiac metabolism and function while, in patients with low body mass index, restrictive diets, or starvation have a deleterious effect. Thus, weight loss in obese patients has positive impacts on ventricular mass and function, whereas, in underweight heart failure patients, such weight reduction adds to the risk of heart damage, predisposing to cachexia. Nutrition plays an essential role in the evolution of cardiovascular disease and should be taken into account. An energy-restricted diet improves myocardial efficiency but can represent a potential risk of heart damage, particularly in patients affected by cardiovascular disease. Micronutrient integration has a marginal effect on cardiovascular efficiency.
Collapse
|
47
|
Reddy SS, Agarwal H, Barthwal MK. Cilostazol ameliorates heart failure with preserved ejection fraction and diastolic dysfunction in obese and non-obese hypertensive mice. J Mol Cell Cardiol 2018; 123:46-57. [PMID: 30138626 DOI: 10.1016/j.yjmcc.2018.08.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/12/2018] [Accepted: 08/17/2018] [Indexed: 01/02/2023]
Abstract
Cilostazol (Ciloz) a potent Type III phosphodiesterase inhibitor is effective against inflammation, insulin resistance and cardiomyopathy. However, the effect of Ciloz on obesity-associated left ventricular diastolic dysfunction has not been explored yet. Hence, we examined the effect of Ciloz on cardiac remodelling and dysfunction in non-obese and obese-insulin resistant mice infused with AngiotensinII (AngII). Male C57BL/6 J mice were initially subjected to 19 weeks of chow or high fat diet (HFD) regimen and thereafter animals were randomised for AngII (1500 ng/kg/min, s.c) infusion or saline and Ciloz (50 mg/kg, p.o) for another 1 week. Obese and non-obese mice infused with AngII exhibited significant diastolic dysfunction and features of heart failure with preserved ejection fraction (HFpEF) since a decrease in fractional shortening and no change in ejection fraction were observed when compared with respective controls. Administration of AngII and Ciloz in HFD fed mice significantly improved the left ventricular function compared with AngII infused HFD mice as evinced from the echocardiographic data. Further, Ciloz treatment significantly reduced cardiomyocyte area, interstitial and perivascular fibrosis; and collagen deposition. Moreover, Ciloz reduced the inflammatory milieu in the heart as evinced by decreased F4/80+ and CD68+ cells; IL-1β and IL-6 gene transcripts. Quantitative assessment of the expression levels revealed substantial upregulation of MMP-9 (pro- and mature-forms) and α-SMA in the left ventricle of AngII infused HFD-fed mice, which was considerably suppressed by Ciloz regimen. The beneficial effect of Ciloz was associated with the normalization in gene expression of hypertrophic and fibrotic markers. Likewise, Ciloz administration markedly reduced the AngII and HFD induced TGF-β1/SMAD3 and Akt/mTOR signalling. Additionally, AngII administered and HFD-fed mice showed increased glycolytic flux, which was considerably diminished by Ciloz treatment as indicated from suppressed PKM2, HK-2, PDK-2, HIF-1α mRNA and GLUT-1 protein expression. Taken together, Ciloz might be therapeutically exploited against AngII and obesity-associated diastolic dysfunction thereby preventing overt heart failure.
Collapse
Affiliation(s)
- Sukka Santosh Reddy
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), New Delhi 110025, India
| | - Heena Agarwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Manoj Kumar Barthwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
48
|
Changes in the activity of some metabolic enzymes in the heart of SHR rat incurred by transgenic expression of CD36. J Physiol Biochem 2018; 74:479-489. [PMID: 29916179 DOI: 10.1007/s13105-018-0641-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 06/12/2018] [Indexed: 01/12/2023]
Abstract
Hypertension, dyslipidemia, and insulin resistance in the spontaneously hypertensive rat (SHR) can be alleviated by rescuing CD36 fatty acid translocase. The present study investigated whether transgenic rescue of CD36 in SHR could affect mitochondrial function and activity of selected metabolic enzymes in the heart. These analyses were conducted on ventricular preparations derived from SHR and from transgenic strain SHR-Cd36 that expresses a functional wild-type CD36. Our respirometric measurements revealed that mitochondria isolated from the left ventricles exhibited two times higher respiratory activity than those isolated from the right ventricles. Whereas, we did not observe any significant changes in functioning of the mitochondrial respiratory system between both rat strains, enzyme activities of total hexokinase, and both mitochondrial and total malate dehydrogenase were markedly decreased in the left ventricles of transgenic rats, compared to SHR. We also detected downregulated expression of the succinate dehydrogenase subunit SdhB (complex II) and 70 kDa peroxisomal membrane protein in the left ventricles of SHR-Cd36. These data indicate that CD36 may affect in a unique fashion metabolic substrate flexibility of the left and right ventricles.
Collapse
|
49
|
Astragaloside IV inhibits ventricular remodeling and improves fatty acid utilization in rats with chronic heart failure. Biosci Rep 2018; 38:BSR20171036. [PMID: 29301869 PMCID: PMC6048210 DOI: 10.1042/bsr20171036] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 01/02/2018] [Accepted: 01/04/2018] [Indexed: 12/24/2022] Open
Abstract
Chronic heart failure (CHF) is the end-stage of many cardiovascular diseases and severely affects the patients’ lifespan. Inhibiting ventricular remodeling is thus a primary treatment target for CHF patients. Astragaloside IV (AS-IV) can improve cardiac function and protect myocardial cells. The study aims to investigate the effects of AS-IV on ventricular remodeling and explore its role in regulating energy metabolism using a rat CHF model. Sprague–Dawley rats were divided into five groups (n=20 per group): CHF + benazepril hydrochloride (Benazepril HCL), CHF + low-dose (30 mg.kg−1.day−1) AS-IV, CHF + high-dose (60 mg.kg−1.day−1) AS-IV, and a sham control group. After 8 weeks of treatment, the cardiac structure and functional parameters were measured. Morphological changes in the myocardial tissue in five groups were evaluated. Protein and mRNA expression of peroxisome proliferator-activated receptor α (PPARα), medium-chain acyl-CoA dehydrogenase (MCAD), and muscle carnitine palmitoyl transferase-1 (MCPT1) were also analyzed. Our results showed that the left ventricular mass index (LVMI), collagen volume fraction (CVF), and free fatty acid (FFA) concentration of CHF group rats increased when compared with sham control group, while the protein and mRNA expressions of PPARα, MCAD, and MCPT1 decreased in CHF. Importantly, treatment with AS-IV (CHF + AS-IV group) showed improved heart function and structure, increased expression of PPARα, MCAD, and MCPT1 and improved FFA utilization in comparison with CHF group. In conclusion, our study shows that AS-IV inhibits ventricular remodeling, improves cardiac function, and decreases FFA concentration of CHF model rats. Our findings suggest a therapeutic potential of using AS-IV in CHF.
Collapse
|
50
|
Abdurrachim D, Nabben M, Hoerr V, Kuhlmann MT, Bovenkamp P, Ciapaite J, Geraets IME, Coumans W, Luiken JJFP, Glatz JFC, Schäfers M, Nicolay K, Faber C, Hermann S, Prompers JJ. Diabetic db/db mice do not develop heart failure upon pressure overload: a longitudinal in vivo PET, MRI, and MRS study on cardiac metabolic, structural, and functional adaptations. Cardiovasc Res 2018; 113:1148-1160. [PMID: 28549111 DOI: 10.1093/cvr/cvx100] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 05/23/2017] [Indexed: 12/11/2022] Open
Abstract
Aims Heart failure is associated with altered myocardial substrate metabolism and impaired cardiac energetics. Comorbidities like diabetes may influence the metabolic adaptations during heart failure development. We quantified to what extent changes in substrate preference, lipid accumulation, and energy status predict the longitudinal development of hypertrophy and failure in the non-diabetic and the diabetic heart. Methods and results Transverse aortic constriction (TAC) was performed in non-diabetic (db/+) and diabetic (db/db) mice to induce pressure overload. Magnetic resonance imaging, 31P magnetic resonance spectroscopy (MRS), 1H MRS, and 18F-fluorodeoxyglucose-positron emission tomography (PET) were applied to measure cardiac function, energy status, lipid content, and glucose uptake, respectively. In vivo measurements were complemented with ex vivo techniques of high-resolution respirometry, proteomics, and western blotting to elucidate the underlying molecular pathways. In non-diabetic mice, TAC induced progressive cardiac hypertrophy and dysfunction, which correlated with increased protein kinase D-1 (PKD1) phosphorylation and increased glucose uptake. These changes in glucose utilization preceded a reduction in cardiac energy status. At baseline, compared with non-diabetic mice, diabetic mice showed normal cardiac function, higher lipid content and mitochondrial capacity for fatty acid oxidation, and lower PKD1 phosphorylation, glucose uptake, and energetics. Interestingly, TAC affected cardiac function only mildly in diabetic mice, which was accompanied by normalization of phosphorylated PKD1, glucose uptake, and cardiac energy status. Conclusion The cardiac metabolic adaptations in diabetic mice seem to prevent the heart from failing upon pressure overload, suggesting that restoring the balance between glucose and fatty acid utilization is beneficial for cardiac function.
Collapse
Affiliation(s)
- Desiree Abdurrachim
- Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Miranda Nabben
- Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.,Department of Genetics and Cell Biology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Verena Hoerr
- Department of Clinical Radiology, University Hospital of Münster, Münster, Germany.,Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | | | - Philipp Bovenkamp
- Department of Clinical Radiology, University Hospital of Münster, Münster, Germany
| | - Jolita Ciapaite
- Department of Pediatrics and Systems Biology Center for Energy Metabolism and Ageing, Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ilvy M E Geraets
- Department of Genetics and Cell Biology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Will Coumans
- Department of Genetics and Cell Biology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Joost J F P Luiken
- Department of Genetics and Cell Biology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Jan F C Glatz
- Department of Genetics and Cell Biology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Michael Schäfers
- European Institute for Molecular Imaging-EIMI, Münster, Germany.,Cells-in-Motion Cluster of Excellence, University of Münster, Münster, Germany.,Department of Nuclear Medicine, University of Münster, Münster, Germany
| | - Klaas Nicolay
- Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Cornelius Faber
- Department of Clinical Radiology, University Hospital of Münster, Münster, Germany
| | - Sven Hermann
- European Institute for Molecular Imaging-EIMI, Münster, Germany.,Cells-in-Motion Cluster of Excellence, University of Münster, Münster, Germany
| | - Jeanine J Prompers
- Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|