1
|
Sun Q, Karwi QG, Wong N, Lopaschuk GD. Advances in myocardial energy metabolism: metabolic remodelling in heart failure and beyond. Cardiovasc Res 2024; 120:1996-2016. [PMID: 39453987 PMCID: PMC11646102 DOI: 10.1093/cvr/cvae231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/28/2024] [Accepted: 07/03/2024] [Indexed: 10/27/2024] Open
Abstract
The very high energy demand of the heart is primarily met by adenosine triphosphate (ATP) production from mitochondrial oxidative phosphorylation, with glycolysis providing a smaller amount of ATP production. This ATP production is markedly altered in heart failure, primarily due to a decrease in mitochondrial oxidative metabolism. Although an increase in glycolytic ATP production partly compensates for the decrease in mitochondrial ATP production, the failing heart faces an energy deficit that contributes to the severity of contractile dysfunction. The relative contribution of the different fuels for mitochondrial ATP production dramatically changes in the failing heart, which depends to a large extent on the type of heart failure. A common metabolic defect in all forms of heart failure [including heart failure with reduced ejection fraction (HFrEF), heart failure with preserved EF (HFpEF), and diabetic cardiomyopathies] is a decrease in mitochondrial oxidation of pyruvate originating from glucose (i.e. glucose oxidation). This decrease in glucose oxidation occurs regardless of whether glycolysis is increased, resulting in an uncoupling of glycolysis from glucose oxidation that can decrease cardiac efficiency. The mitochondrial oxidation of fatty acids by the heart increases or decreases, depending on the type of heart failure. For instance, in HFpEF and diabetic cardiomyopathies myocardial fatty acid oxidation increases, while in HFrEF myocardial fatty acid oxidation either decreases or remains unchanged. The oxidation of ketones (which provides the failing heart with an important energy source) also differs depending on the type of heart failure, being increased in HFrEF, and decreased in HFpEF and diabetic cardiomyopathies. The alterations in mitochondrial oxidative metabolism and glycolysis in the failing heart are due to transcriptional changes in key enzymes involved in the metabolic pathways, as well as alterations in redox state, metabolic signalling and post-translational epigenetic changes in energy metabolic enzymes. Of importance, targeting the mitochondrial energy metabolic pathways has emerged as a novel therapeutic approach to improving cardiac function and cardiac efficiency in the failing heart.
Collapse
Affiliation(s)
- Qiuyu Sun
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John’s, NL A1B 3V6, Canada
| | - Nathan Wong
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| |
Collapse
|
2
|
Park KC, Crump NT, Louwman N, Krywawych S, Cheong YJ, Vendrell I, Gill EK, Gunadasa-Rohling M, Ford KL, Hauton D, Fournier M, Pires E, Watson L, Roseman G, Holder J, Koschinski A, Carnicer R, Curtis MK, Zaccolo M, Hulikova A, Fischer R, Kramer HB, McCullagh JSO, Trefely S, Milne TA, Swietach P. Disrupted propionate metabolism evokes transcriptional changes in the heart by increasing histone acetylation and propionylation. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1221-1245. [PMID: 38500966 PMCID: PMC7615744 DOI: 10.1038/s44161-023-00365-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/15/2023] [Indexed: 03/20/2024]
Abstract
Propiogenic substrates and gut bacteria produce propionate, a post-translational protein modifier. In this study, we used a mouse model of propionic acidaemia (PA) to study how disturbances to propionate metabolism result in histone modifications and changes to gene expression that affect cardiac function. Plasma propionate surrogates were raised in PA mice, but female hearts manifested more profound changes in acyl-CoAs, histone propionylation and acetylation, and transcription. These resulted in moderate diastolic dysfunction with raised diastolic Ca2+, expanded end-systolic ventricular volume and reduced stroke volume. Propionate was traced to histone H3 propionylation and caused increased acetylation genome-wide, including at promoters of Pde9a and Mme, genes related to contractile dysfunction through downscaled cGMP signaling. The less severe phenotype in male hearts correlated with β-alanine buildup. Raising β-alanine in cultured myocytes treated with propionate reduced propionyl-CoA levels, indicating a mechanistic relationship. Thus, we linked perturbed propionate metabolism to epigenetic changes that impact cardiac function.
Collapse
Affiliation(s)
- Kyung Chan Park
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Nicholas T. Crump
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Present Address: Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Niamh Louwman
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Steve Krywawych
- Department of Chemical Pathology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Yuen Jian Cheong
- Epigenetics & Signalling Programmes, Babraham Institute, Cambridge, UK
| | - Iolanda Vendrell
- Nuffield Department of Medicine, Target Discovery Institute, Oxford, UK
- Nuffield Department of Medicine, Chinese Academy for Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Eleanor K. Gill
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | | | - Kerrie L. Ford
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - David Hauton
- Department of Chemistry, University of Oxford, Oxford, UK
| | | | | | - Lydia Watson
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Gerald Roseman
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - James Holder
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Andreas Koschinski
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Ricardo Carnicer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - M. Kate Curtis
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Manuela Zaccolo
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Alzbeta Hulikova
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| | - Roman Fischer
- Nuffield Department of Medicine, Target Discovery Institute, Oxford, UK
- Nuffield Department of Medicine, Chinese Academy for Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Holger B. Kramer
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Sophie Trefely
- Epigenetics & Signalling Programmes, Babraham Institute, Cambridge, UK
| | - Thomas A. Milne
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Pawel Swietach
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Shaul D, Lev-Cohain N, Sapir G, Sosna J, Gomori JM, Joskowicz L, Katz-Brull R. Real-time influence of intracellular acidification and Na + /H + exchanger inhibition on in-cell pyruvate metabolism in the perfused mouse heart: A 31 P-NMR and hyperpolarized 13 C-NMR study. NMR IN BIOMEDICINE 2023; 36:e4993. [PMID: 37424280 DOI: 10.1002/nbm.4993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 07/11/2023]
Abstract
Disruption of acid-base balance is linked to various diseases and conditions. In the heart, intracellular acidification is associated with heart failure, maladaptive cardiac hypertrophy, and myocardial ischemia. Previously, we have reported that the ratio of the in-cell lactate dehydrogenase (LDH) to pyruvate dehydrogenase (PDH) activities is correlated with cardiac pH. To further characterize the basis for this correlation, these in-cell activities were investigated under induced intracellular acidification without and with Na+ /H+ exchanger (NHE1) inhibition by zoniporide. Male mouse hearts (n = 30) were isolated and perfused retrogradely. Intracellular acidification was performed in two ways: (1) with the NH4 Cl prepulse methodology; and (2) by combining the NH4 Cl prepulse with zoniporide. 31 P NMR spectroscopy was used to determine the intracellular cardiac pH and to quantify the adenosine triphosphate and phosphocreatine content. Hyperpolarized [1-13 C]pyruvate was obtained using dissolution dynamic nuclear polarization. 13 C NMR spectroscopy was used to monitor hyperpolarized [1-13 C]pyruvate metabolism and determine enzyme activities in real time at a temporal resolution of a few seconds using the product-selective saturating excitation approach. The intracellular acidification induced by the NH4 Cl prepulse led to reduced LDH and PDH activities (-16% and -39%, respectively). This finding is in line with previous evidence of reduced myocardial contraction and therefore reduced metabolic activity upon intracellular acidification. Concomitantly, the LDH/PDH activity ratio increased with the reduction in pH, as previously reported. Combining the NH4 Cl prepulse with zoniporide led to a greater reduction in LDH activity (-29%) and to increased PDH activity (+40%). These changes resulted in a surprising decrease in the LDH/PDH ratio, as opposed to previous predictions. Zoniporide alone (without intracellular acidification) did not change these enzyme activities. A possible explanation for the enzymatic changes observed during the combination of the NH4 Cl prepulse and NHE1 inhibition may be related to mitochondrial NHE1 inhibition, which likely negates the mitochondrial matrix acidification. This effect, combined with the increased acidity in the cytosol, would result in an enhanced H+ gradient across the mitochondrial membrane and a temporarily higher pyruvate transport into the mitochondria, thereby increasing the PDH activity at the expense of the cytosolic LDH activity. These findings demonstrate the complexity of in-cell cardiac metabolism and its dependence on intracellular acidification. This study demonstrates the capabilities and limitations of hyperpolarized [1-13 C]pyruvate in the characterization of intracellular acidification as regards cardiac pathologies.
Collapse
Affiliation(s)
- David Shaul
- Department of Radiology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, Israel
| | - Naama Lev-Cohain
- Department of Radiology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gal Sapir
- Department of Radiology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, Israel
| | - Jacob Sosna
- Department of Radiology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - J Moshe Gomori
- Department of Radiology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Leo Joskowicz
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rachel Katz-Brull
- Department of Radiology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Wohl Institute for Translational Medicine, Hadassah Medical Organization, Jerusalem, Israel
| |
Collapse
|
4
|
Chen K, Shi Y, Zhu H. Analysis of the role of glucose metabolism-related genes in dilated cardiomyopathy based on bioinformatics. J Thorac Dis 2023; 15:3870-3884. [PMID: 37559624 PMCID: PMC10407475 DOI: 10.21037/jtd-23-906] [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: 06/05/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a prevalent condition with diverse etiologies, including viral infection, autoimmune response, and genetic factors. Despite the crucial role of energy metabolism in cardiac function, therapeutic targets for key genes in DCM's energy metabolism remain scarce. METHODS Our study employed the GSE79962 and GSE42955 datasets from the Gene Expression Omnibus (GEO) database for myocardial tissue sample collection and target gene identification via differential gene expression screening. Using various R packages, GSEA software, and the STRING database, we conducted data analysis, gene set enrichment, and protein-protein interaction predictions. The least absolute shrinkage and selection operator (LASSO) and Support Vector Machine (SVM) algorithms aided in feature gene selection, while the predictive model's efficiency was evaluated via the receiver operating characteristic (ROC) curve analysis. We used the non-negative matrix factorization (NMF) method for molecular typing and the cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) algorithm for predicting immune cell infiltration. RESULTS The DLAT and LDHA genes may regulate the immune microenvironment of DCM by influencing activated dendritic cells, activated mast cells, and M0 macrophages, respectively. The BPGM, DLAT, PGM2, ADH1A, ADH1C, LDHA, and PFKM genes may regulate m6A methylation in DCM by affecting the ZC3H13, ALKBH5, RBMX, HNRNPC, METTL3, and YTHDC1 genes. Further regulatory mechanism analysis suggested that PFKM, DLAT, PKLR, PGM2, LDHA, BPGM, ADH1A, and ADH1C could be involved in the development of cardiomyopathy by regulating the Toll-like receptor signaling pathway. CONCLUSIONS PFKM, DLAT, PKLR, PGM2, LDHA, BPGM, ADH1A, and ADH1C may serve as potential targets for guiding the diagnosis, treatment, and follow-up of DCM.
Collapse
Affiliation(s)
- Keping Chen
- Department of Emergency, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yan Shi
- Operating Room, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Haijie Zhu
- Department of Emergency, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
5
|
Sun P, Wu Z, Lin L, Hu G, Zhang X, Wang J. MR-Nucleomics: The study of pathological cellular processes with multinuclear magnetic resonance spectroscopy and imaging in vivo. NMR IN BIOMEDICINE 2023; 36:e4845. [PMID: 36259659 DOI: 10.1002/nbm.4845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 09/28/2022] [Accepted: 10/08/2022] [Indexed: 06/16/2023]
Abstract
Clinical medicine has experienced a rapid development in recent decades, during which therapies targeting specific cellular signaling pathways, or specific cell surface receptors, have been increasingly adopted. While these developments in clinical medicine call for improved precision in diagnosis and treatment monitoring, modern medical imaging methods are restricted mainly to anatomical imaging, lagging behind the requirements of precision medicine. Although positron emission tomography and single photon emission computed tomography have been used clinically for studies of metabolism, their applications have been limited by the exposure risk to ionizing radiation, the subsequent limitation in repeated and longitudinal studies, and the incapability in assessing downstream metabolism. Magnetic resonance spectroscopy (MRS) or spectroscopic imaging (MRSI) are, in theory, capable of assessing molecular activities in vivo, although they are often limited by sensitivity. Here, we review some recent developments in MRS and MRSI of multiple nuclei that have potential as molecular imaging tools in the clinic.
Collapse
Affiliation(s)
- Peng Sun
- Clinical & Technical Support, Philips Healthcare, China
| | - Zhigang Wu
- Clinical & Technical Support, Philips Healthcare, China
| | - Liangjie Lin
- Clinical & Technical Support, Philips Healthcare, China
| | - Geli Hu
- Clinical & Technical Support, Philips Healthcare, China
| | | | - Jiazheng Wang
- Clinical & Technical Support, Philips Healthcare, China
| |
Collapse
|
6
|
Bengel P, Elkenani M, Beuthner BE, Pietzner M, Mohamed BA, Pollok-Kopp B, Krätzner R, Toischer K, Puls M, Fischer A, Binder L, Hasenfuß G, Schnelle M. Metabolomic Profiling in Patients with Different Hemodynamic Subtypes of Severe Aortic Valve Stenosis. Biomolecules 2023; 13:95. [PMID: 36671480 PMCID: PMC9855798 DOI: 10.3390/biom13010095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/16/2022] [Accepted: 12/26/2022] [Indexed: 01/05/2023] Open
Abstract
Severe aortic stenosis (AS) is a common pathological condition in an ageing population imposing significant morbidity and mortality. Based on distinct hemodynamic features, i.e., ejection fraction (EF), transvalvular gradient and stroke volume, four different AS subtypes can be distinguished: (i) normal EF and high gradient, (ii) reduced EF and high gradient, (iii) reduced EF and low gradient, and (iv) normal EF and low gradient. These subtypes differ with respect to pathophysiological mechanisms, cardiac remodeling, and prognosis. However, little is known about metabolic changes in these different hemodynamic conditions of AS. Thus, we carried out metabolomic analyses in serum samples of 40 AS patients (n = 10 per subtype) and 10 healthy blood donors (controls) using ultrahigh-performance liquid chromatography-tandem mass spectroscopy. A total of 1293 biochemicals could be identified. Principal component analysis revealed different metabolic profiles in all of the subgroups of AS (All-AS) vs. controls. Out of the determined biochemicals, 48% (n = 620) were altered in All-AS vs. controls (p < 0.05). In this regard, levels of various acylcarnitines (e.g., myristoylcarnitine, fold-change 1.85, p < 0.05), ketone bodies (e.g., 3-hydroxybutyrate, fold-change 11.14, p < 0.05) as well as sugar metabolites (e.g., glucose, fold-change 1.22, p < 0.05) were predominantly increased, whereas amino acids (e.g., leucine, fold-change 0.8, p < 0.05) were mainly reduced in All-AS. Interestingly, these changes appeared to be consistent amongst all AS subtypes. Distinct differences between AS subtypes were found for metabolites belonging to hemoglobin metabolism, diacylglycerols, and dihydrosphingomyelins. These findings indicate that relevant changes in substrate utilization appear to be consistent for different hemodynamic subtypes of AS and may therefore reflect common mechanisms during AS-induced heart failure. Additionally, distinct metabolites could be identified to significantly differ between certain AS subtypes. Future studies need to define their pathophysiological implications.
Collapse
Affiliation(s)
- Philipp Bengel
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Manar Elkenani
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Bo E. Beuthner
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, UK
- Computational Medicine, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Belal A. Mohamed
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Beatrix Pollok-Kopp
- Department of Transfusion Medicine, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Ralph Krätzner
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Karl Toischer
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Miriam Puls
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Andreas Fischer
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, 37075 Göttingen, Germany
- Division Vascular Signaling and Cancer, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Lutz Binder
- Department of Clinical Chemistry, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Gerd Hasenfuß
- Clinic for Cardiology & Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Moritz Schnelle
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
7
|
Monga S, Valkovič L, Tyler D, Lygate CA, Rider O, Myerson SG, Neubauer S, Mahmod M. Insights Into the Metabolic Aspects of Aortic Stenosis With the Use of Magnetic Resonance Imaging. JACC Cardiovasc Imaging 2022; 15:2112-2126. [PMID: 36481080 PMCID: PMC9722407 DOI: 10.1016/j.jcmg.2022.04.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/25/2022] [Accepted: 04/29/2022] [Indexed: 01/13/2023]
Abstract
Pressure overload in aortic stenosis (AS) encompasses both structural and metabolic remodeling and increases the risk of decompensation into heart failure. A major component of metabolic derangement in AS is abnormal cardiac substrate use, with down-regulation of fatty acid oxidation, increased reliance on glucose metabolism, and subsequent myocardial lipid accumulation. These changes are associated with energetic and functional cardiac impairment in AS and can be assessed with the use of cardiac magnetic resonance spectroscopy (MRS). Proton MRS allows the assessment of myocardial triglyceride content and creatine concentration. Phosphorous MRS allows noninvasive in vivo quantification of the phosphocreatine-to-adenosine triphosphate ratio, a measure of cardiac energy status that is reduced in patients with severe AS. This review summarizes the changes to cardiac substrate and high-energy phosphorous metabolism and how they affect cardiac function in AS. The authors focus on the role of MRS to assess these metabolic changes, and potentially guide future (cellular) metabolic therapy in AS.
Collapse
Affiliation(s)
- Shveta Monga
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ladislav Valkovič
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Damian Tyler
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom; Wellcome Centre for Human Genetics, Oxford, United Kingdom
| | - Oliver Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Saul G Myerson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Masliza Mahmod
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
8
|
Yurista SR, Eder RA, Kwon DH, Farrar CT, Yen YF, Tang WHW, Nguyen CT. Magnetic resonance imaging of cardiac metabolism in heart failure: how far have we come? Eur Heart J Cardiovasc Imaging 2022; 23:1277-1289. [PMID: 35788836 PMCID: PMC10202438 DOI: 10.1093/ehjci/jeac121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 11/12/2022] Open
Abstract
As one of the highest energy consumer organs in the body, the heart requires tremendous amount of adenosine triphosphate (ATP) to maintain its continuous mechanical work. Fatty acids, glucose, and ketone bodies are the primary fuel source of the heart to generate ATP with perturbations in ATP generation possibly leading to contractile dysfunction. Cardiac metabolic imaging with magnetic resonance imaging (MRI) plays a crucial role in understanding the dynamic metabolic changes occurring in the failing heart, where the cardiac metabolism is deranged. Also, targeting and quantifying metabolic changes in vivo noninvasively is a promising approach to facilitate diagnosis, determine prognosis, and evaluate therapeutic response. Here, we summarize novel MRI techniques used for detailed investigation of cardiac metabolism in heart failure including magnetic resonance spectroscopy (MRS), hyperpolarized MRS, and chemical exchange saturation transfer based on evidence from preclinical and clinical studies and to discuss the potential clinical application in heart failure.
Collapse
Affiliation(s)
- Salva R Yurista
- Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, 149 13th St, Charlestown, MA 02129, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, 149 13th St, Charlestown, MA 02129, USA
| | - Robert A Eder
- Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, 149 13th St, Charlestown, MA 02129, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, 149 13th St, Charlestown, MA 02129, USA
| | - Deborah H Kwon
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Christian T Farrar
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, 149 13th St, Charlestown, MA 02129, USA
| | - Yi Fen Yen
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, 149 13th St, Charlestown, MA 02129, USA
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Christopher T Nguyen
- Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, 149 13th St, Charlestown, MA 02129, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, 149 13th St, Charlestown, MA 02129, USA
- Division of Health Science Technology, Harvard-Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
- Cardiovascular Innovation Research Center, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
9
|
The Role of Oxidative Stress in the Aging Heart. Antioxidants (Basel) 2022; 11:antiox11020336. [PMID: 35204217 PMCID: PMC8868312 DOI: 10.3390/antiox11020336] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/17/2022] Open
Abstract
Medical advances and the availability of diagnostic tools have considerably increased life expectancy and, consequently, the elderly segment of the world population. As age is a major risk factor in cardiovascular disease (CVD), it is critical to understand the changes in cardiac structure and function during the aging process. The phenotypes and molecular mechanisms of cardiac aging include several factors. An increase in oxidative stress is a major player in cardiac aging. Reactive oxygen species (ROS) production is an important mechanism for maintaining physiological processes; its generation is regulated by a system of antioxidant enzymes. Oxidative stress occurs from an imbalance between ROS production and antioxidant defenses resulting in the accumulation of free radicals. In the heart, ROS activate signaling pathways involved in myocyte hypertrophy, interstitial fibrosis, contractile dysfunction, and inflammation thereby affecting cell structure and function, and contributing to cardiac damage and remodeling. In this manuscript, we review recent published research on cardiac aging. We summarize the aging heart biology, highlighting key molecular pathways and cellular processes that underlie the redox signaling changes during aging. Main ROS sources, antioxidant defenses, and the role of dysfunctional mitochondria in the aging heart are addressed. As metabolism changes contribute to cardiac aging, we also comment on the most prevalent metabolic alterations. This review will help us to understand the mechanisms involved in the heart aging process and will provide a background for attractive molecular targets to prevent age-driven pathology of the heart. A greater understanding of the processes involved in cardiac aging may facilitate our ability to mitigate the escalating burden of CVD in older individuals and promote healthy cardiac aging.
Collapse
|
10
|
Tougaard RS, Laustsen C, Lassen TR, Qi H, Lindhardt JL, Schroeder M, Jespersen NR, Hansen ESS, Ringgaard S, Bøtker HE, Kim WY, Stødkilde-Jørgensen H, Wiggers H. Remodeling after myocardial infarction and effects of heart failure treatment investigated by hyperpolarized [1- 13 C]pyruvate magnetic resonance spectroscopy. Magn Reson Med 2021; 87:57-69. [PMID: 34378800 DOI: 10.1002/mrm.28964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 11/10/2022]
Abstract
PURPOSE Hyperpolarized [1-13 C]pyruvate MRS can measure cardiac metabolism in vivo. We investigated whether [1-13 C]pyruvate MRS could predict left ventricular remodeling following myocardial infarction (MI), long-term left ventricular effects of heart failure medication, and could identify responders to treatment. METHODS Thirty-five rats were scanned with hyperpolarized [1-13 C]pyruvate MRS 3 days after MI or sham surgery. The animals were re-examined after 30 days of therapy with β-blockers and ACE-inhibitors (active group, n = 12), placebo treatment (placebo group, n = 13) or no treatment (sham group, n = 10). Furthermore, heart tissue mitochondrial respiratory capacity was assessed by high-resolution respirometry. Metabolic results were compared between groups, over time and correlated to functional MR data at each time point. RESULTS At 30 ± 0.5 days post MI, left ventricular ejection fraction (LVEF) differed between groups (sham, 77% ± 1%; placebo, 52% ± 3%; active, 63% ± 2%, P < .001). Cardiac metabolism, measured by both hyperpolarized [1-13 C]pyruvate MRS and respirometry, neither differed between groups nor between baseline and follow-up. Three days post MI, low bicarbonate + CO2 /pyruvate ratio was associated with low LVEF. At follow-up, in the active group, a poor recovery of LVEF was associated with high bicarbonate + CO2 /pyruvate ratio, as measured by hyperpolarized MRS. CONCLUSION In a rat model of moderate heart failure, medical treatment improved function, but did not on average influence [1-13 C]pyruvate flux as measured by MRS; however, responders to heart failure medication had reduced capacity for carbohydrate metabolism.
Collapse
Affiliation(s)
- Rasmus Stilling Tougaard
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Christoffer Laustsen
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | | | - Haiyun Qi
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Jakob Lykke Lindhardt
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Marie Schroeder
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | | | | | - Steffen Ringgaard
- MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark
| | - Won Yong Kim
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark.,MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | | | - Henrik Wiggers
- Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark
| |
Collapse
|
11
|
Agger P, Hyldebrandt JA, Hansen ESS, Omann C, Bøgh N, Waziri F, Nielsen PM, Laustsen C. Magnetic resonance hyperpolarization imaging detects early myocardial dysfunction in a porcine model of right ventricular heart failure. Eur Heart J Cardiovasc Imaging 2021; 21:93-101. [PMID: 31329841 PMCID: PMC6923679 DOI: 10.1093/ehjci/jez074] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/24/2019] [Accepted: 04/02/2019] [Indexed: 12/31/2022] Open
Abstract
Aims Early detection of heart failure is important for timely treatment. During the development of heart failure, adaptive intracellular metabolic processes that evolve prior to macro-anatomic remodelling, could provide an early signal of impending failure. We hypothesized that metabolic imaging with hyperpolarized magnetic resonance would detect the early development of heart failure before conventional echocardiography could reveal cardiac dysfunction. Methods and results Five 8.5 kg piglets were subjected to pulmonary banding and subsequently examined by [1-13C]pyruvate hyperpolarization, conventional magnetic resonance imaging, echocardiography, and blood testing, every 4 weeks for 16 weeks. They were compared with a weight matched, healthy control group. Conductance catheter examination at the end of the study showed impaired right ventricular systolic function along with compromised left ventricular diastolic function. After 16 weeks, we saw a significant decrease in the conversion ratio of pyruvate/bicarbonate in the left ventricle from 0.13 (0.04) in controls to 0.07 (0.02) in animals with pulmonary banding, along with a significant increase in the lactate/bicarbonate ratio to 3.47 (1.57) compared with 1.34 (0.81) in controls. N-terminal pro-hormone of brain natriuretic peptide was increased by more than 300%, while cardiac index was reduced to 2.8 (0.95) L/min/m2 compared with 3.9 (0.95) in controls. Echocardiography revealed no changes. Conclusion Hyperpolarization detected a shift towards anaerobic metabolism in early stages of right ventricular dysfunction, as evident by an increased lactate/bicarbonate ratio. Dysfunction was confirmed with conductance catheter assessment, but could not be detected by echocardiography. Hyperpolarization has a promising future in clinical assessment of heart failure in both acquired and congenital heart disease.
Collapse
Affiliation(s)
- Peter Agger
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus, Denmark
| | - Janus Adler Hyldebrandt
- Department of Anaesthesiology and Intensive Care, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, Denmark.,Department of Anaesthesiology and Intensive Care, Akershus University Hospital, Sykehusveien 25, Lørenskog, Norway
| | | | - Camilla Omann
- Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, Denmark
| | - Nikolaj Bøgh
- MR Research Centre, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Farhad Waziri
- Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, Denmark
| | - Per Mose Nielsen
- MR Research Centre, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Christoffer Laustsen
- MR Research Centre, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| |
Collapse
|
12
|
Vileigas DF, de Souza SLB, Corrêa CR, Silva CCVDA, de Campos DHS, Padovani CR, Cicogna AC. The effects of two types of Western diet on the induction of metabolic syndrome and cardiac remodeling in obese rats. J Nutr Biochem 2021; 92:108625. [PMID: 33705955 DOI: 10.1016/j.jnutbio.2021.108625] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/13/2022]
Abstract
Metabolic syndrome (MetS) include obesity as a critical feature and is strongly associated with risk of cardiovascular disease (CVD). Insights into mechanisms involved in the pathophysiology of these clinical manifestations are essential for the development of therapeutic strategies. Thus, Western diets (WD) have been widely employed in diet-induced obesity (DIO) model. However, there are variations in fat and sugar proportions of such diets, making comparisons challenging. We aimed to assess the impact of two types of the WD on metabolic status and cardiac remodeling, to achieve a DIO model that better mimics the human pathogenesis of MetS-induced CVD. Male Wistar rats were distributed into three groups: control diet, Western diet fat (WDF), and Western diet sugar (WDS) for 41 weeks. Metabolic and inflammatory parameters and cardiac changes were characterized. WDF and WDS feeding promoted higher serum triglycerides, glucose intolerance, and insulin resistance, while just WDF presented inflammation in adipose tissue. WDF-fed rats showed increased catalase activity and malondialdehyde (MDA) and carbonyl protein levels, suggesting cardiac oxidative stress, while WDS-fed rats only raised MDA. Both WD equally elevated protein expressions involved in lipid metabolism, but only WDF downregulated the glycolysis pathway. Furthermore, the mechanical myocardial function was impaired in obese rats, being more relevant in WDF. In conclusion, both WD effectively triggered MetS features, although inflammation was detected just on the WDF-fed animals. Moreover, the WDF promoted a more pronounced functional, metabolic, and oxidative cardiac disorder, suggesting to be an adequate model for studying CVD in the scenario of MetS.
Collapse
Affiliation(s)
- Danielle Fernandes Vileigas
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil.
| | - Sérgio Luiz Borges de Souza
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Camila Renata Corrêa
- Department of Patology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | | | | | - Carlos Roberto Padovani
- Department of Biostatistics, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Antonio Carlos Cicogna
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil.
| |
Collapse
|
13
|
Shaul D, Azar A, Sapir G, Uppala S, Nardi-Schreiber A, Gamliel A, Sosna J, Gomori JM, Katz-Brull R. Correlation between lactate dehydrogenase/pyruvate dehydrogenase activities ratio and tissue pH in the perfused mouse heart: A potential noninvasive indicator of cardiac pH provided by hyperpolarized magnetic resonance. NMR IN BIOMEDICINE 2021; 34:e4444. [PMID: 33258527 DOI: 10.1002/nbm.4444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 10/05/2020] [Accepted: 10/24/2020] [Indexed: 06/12/2023]
Abstract
Cardiovascular diseases account for more than 30% of all deaths worldwide and many could be ameliorated with early diagnosis. Current cardiac imaging modalities can assess blood flow, heart anatomy and mechanical function. However, for early diagnosis and improved treatment, further functional biomarkers are needed. One such functional biomarker could be the myocardium pH. Although tissue pH is already determinable via MR techniques, and has been since the early 1990s, it remains elusive to use practically. The objective of this study was to explore the possibility to evaluate cardiac pH noninvasively, using in-cell enzymatic rates of hyperpolarized [1-13 C]pyruvate metabolism (ie, moles of product produced per unit time) determined directly in real time using magnetic resonance spectroscopy in a perfused mouse heart model. As a gold standard for tissue pH we used 31 P spectroscopy and the chemical shift of the inorganic phosphate (Pi) signal. The nonhomogenous pH distribution of the perfused heart was analyzed using a multi-parametric analysis of this signal, thus taking into account the heterogeneous nature of this characteristic. As opposed to the signal ratio of hyperpolarized [13 C]bicarbonate to [13 CO2 ], which has shown correlation to pH in other studies, we investigated here the ratio of two intracellular enzymatic rates: lactate dehydrogenase (LDH) and pyruvate dehydrogenase (PDH), by way of determining the production rates of [1-13 C]lactate and [13 C]bicarbonate, respectively. The enzyme activities determined here are intracellular, while the pH determined using the Pi signal may contain an extracellular component, which could not be ruled out. Nevertheless, we report a strong correlation between the tissue pH and the LDH/PDH activities ratio. This work may pave the way for using the LDH/PDH activities ratio as an indicator of cardiac intracellular pH in vivo, in an MRI examination.
Collapse
Affiliation(s)
- David Shaul
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Assad Azar
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Gal Sapir
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Sivaranjan Uppala
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Atara Nardi-Schreiber
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Ayelet Gamliel
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Jacob Sosna
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - J Moshe Gomori
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| | - Rachel Katz-Brull
- Department of Radiology, Hadassah Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Jerusalem, Israel
| |
Collapse
|
14
|
Watson WD, Miller JJJ, Lewis A, Neubauer S, Tyler D, Rider OJ, Valkovič L. Use of cardiac magnetic resonance to detect changes in metabolism in heart failure. Cardiovasc Diagn Ther 2020; 10:583-597. [PMID: 32695639 DOI: 10.21037/cdt.2019.12.13] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The heart has a massive adenosine triphosphate (ATP) requirement, produced from the oxidation of metabolic substrates such as fat and glucose. Magnetic resonance spectroscopy offers a unique opportunity to probe this biochemistry: 31Phosphorus spectroscopy can demonstrate the production of ATP and quantify levels of the transport molecule phosphocreatine while 13Carbon spectroscopy can demonstrate the metabolic fates of glucose in real time. These techniques allow the metabolic deficits in heart failure to be interrogated and can be a potential future clinical tool.
Collapse
Affiliation(s)
- William D Watson
- Oxford Centre for Clinical Magnetic Resonance Research, Clarendon Laboratory, University of Oxford, Oxford, UK
| | - Jack J J Miller
- Oxford Centre for Clinical Magnetic Resonance Research, Clarendon Laboratory, University of Oxford, Oxford, UK.,Department of Physiology, Anatomy and Genetics, Clarendon Laboratory, University of Oxford, Oxford, UK.,Department of Physics, Clarendon Laboratory, University of Oxford, Oxford, UK
| | - Andrew Lewis
- Oxford Centre for Clinical Magnetic Resonance Research, Clarendon Laboratory, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research, Clarendon Laboratory, University of Oxford, Oxford, UK
| | - Damian Tyler
- Oxford Centre for Clinical Magnetic Resonance Research, Clarendon Laboratory, University of Oxford, Oxford, UK.,Department of Physiology, Anatomy and Genetics, Clarendon Laboratory, University of Oxford, Oxford, UK
| | - Oliver J Rider
- Oxford Centre for Clinical Magnetic Resonance Research, Clarendon Laboratory, University of Oxford, Oxford, UK
| | - Ladislav Valkovič
- Oxford Centre for Clinical Magnetic Resonance Research, Clarendon Laboratory, University of Oxford, Oxford, UK.,Department of Imaging Methods, Institute of Measurement Science, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
15
|
Bøgh N, Hansen ESS, Omann C, Lindhardt J, Nielsen PM, Stephenson RS, Laustsen C, Hjortdal VE, Agger P. Increasing carbohydrate oxidation improves contractile reserves and prevents hypertrophy in porcine right heart failure. Sci Rep 2020; 10:8158. [PMID: 32424129 PMCID: PMC7235019 DOI: 10.1038/s41598-020-65098-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/24/2020] [Indexed: 01/16/2023] Open
Abstract
In heart failure, myocardial overload causes vast metabolic changes that impair cardiac energy production and contribute to deterioration of contractile function. However, metabolic therapy is not used in heart failure care. We aimed to investigate the interplay between cardiac function and myocardial carbohydrate metabolism in a large animal heart failure model. Using magnetic resonance spectroscopy with hyperpolarized pyruvate and magnetic resonance imaging at rest and during pharmacological stress, we investigated the in-vivo cardiac pyruvate metabolism and contractility in a porcine model of chronic pulmonary insufficiency causing right ventricular volume overload. To assess if increasing the carbohydrate metabolic reserve improves the contractile reserve, a group of animals were fed dichloroacetate, an activator of pyruvate oxidation. Volume overload caused heart failure with decreased pyruvate dehydrogenase flux and poor ejection fraction reserve. The animals treated with dichloroacetate had a larger contractile response to dobutamine stress than non-treated animals. Further, dichloroacetate prevented myocardial hypertrophy. The in-vivo metabolic data were validated by mitochondrial respirometry, enzyme activity assays and gene expression analyses. Our results show that pyruvate dehydrogenase kinase inhibition improves the contractile reserve and decreases hypertrophy by augmenting carbohydrate metabolism in porcine heart failure. The approach is promising for metabolic heart failure therapy.
Collapse
Affiliation(s)
- Nikolaj Bøgh
- The Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark. .,The MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.
| | - Esben S S Hansen
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Camilla Omann
- The Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Jakob Lindhardt
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Per M Nielsen
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Robert S Stephenson
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.,Institute of Clinical Sciences, College of Medical and Dental Science, The University of Birmingham, Birmingham, United Kingdom
| | - Christoffer Laustsen
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Vibeke E Hjortdal
- The Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Peter Agger
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| |
Collapse
|
16
|
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
|
17
|
Katz MG, Fargnoli AS, Gubara SM, Chepurko E, Bridges CR, Hajjar RJ. Surgical and physiological challenges in the development of left and right heart failure in rat models. Heart Fail Rev 2019; 24:759-777. [PMID: 30903356 PMCID: PMC6698228 DOI: 10.1007/s10741-019-09783-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Rodent surgical animal models of heart failure (HF) are critically important for understanding the proof of principle of the cellular alterations underlying the development of the disease as well as evaluating therapeutics. Robust, reproducible rodent models are a prerequisite to the development of pharmacological and molecular strategies for the treatment of HF in patients. Due to the absence of standardized guidelines regarding surgical technique and clear criteria for HF progression in rats, objectivity is compromised. Scientific publications in rats rarely fully disclose the actual surgical details, and technical and physiological challenges. This lack of reporting is one of the main reasons that the outcomes specified in similar studies are highly variable and associated with unnecessary loss of animals, compromising scientific assessment. This review details rat circulatory and coronary arteries anatomy, the surgical details of rat models that recreate the HF phenotype of myocardial infarction, ischemia/reperfusion, left and right ventricular pressure, and volume overload states, and summarizes the technical and physiological challenges of creating HF. The purpose of this article is to help investigators understand the underlying issues of current HF models in order to reduce variable results and ensure successful, reproducible models of HF.
Collapse
Affiliation(s)
- Michael G Katz
- Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave., Box 1030, New York, NY, 10029-6574, USA.
| | - Anthony S Fargnoli
- Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave., Box 1030, New York, NY, 10029-6574, USA
| | - Sarah M Gubara
- Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave., Box 1030, New York, NY, 10029-6574, USA
| | - Elena Chepurko
- Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave., Box 1030, New York, NY, 10029-6574, USA
| | - Charles R Bridges
- Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave., Box 1030, New York, NY, 10029-6574, USA
| | - Roger J Hajjar
- Cardiovascular Research Center, Department of Cardiology, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave., Box 1030, New York, NY, 10029-6574, USA
| |
Collapse
|
18
|
Le Page LM, Rider OJ, Lewis AJ, Noden V, Kerr M, Giles L, Ambrose LJ, Ball V, Mansor L, Heather LC, Tyler DJ. Assessing the effect of hypoxia on cardiac metabolism using hyperpolarized 13 C magnetic resonance spectroscopy. NMR IN BIOMEDICINE 2019; 32:e4099. [PMID: 31090979 PMCID: PMC6619452 DOI: 10.1002/nbm.4099] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/14/2019] [Accepted: 03/06/2019] [Indexed: 05/03/2023]
Abstract
Hypoxia plays a role in many diseases and can have a wide range of effects on cardiac metabolism depending on the extent of the hypoxic insult. Noninvasive imaging methods could shed valuable light on the metabolic effects of hypoxia on the heart in vivo. Hyperpolarized carbon-13 magnetic resonance spectroscopy (HP 13 C MRS) in particular is an exciting technique for imaging metabolism that could provide such information. The aim of our work was, therefore, to establish whether hyperpolarized 13 C MRS can be used to assess the in vivo heart's metabolism of pyruvate in response to systemic acute and chronic hypoxic exposure. Groups of healthy male Wistar rats were exposed to either acute (30 minutes), 1 week or 3 weeks of hypoxia. In vivo MRS of hyperpolarized [1-13 C] pyruvate was carried out along with assessments of physiological parameters and ejection fraction. Hematocrit was elevated after 1 week and 3 weeks of hypoxia. 30 minutes of hypoxia resulted in a significant reduction in pyruvate dehydrogenase (PDH) flux, whereas 1 or 3 weeks of hypoxia resulted in a PDH flux that was not different to normoxic animals. Conversion of hyperpolarized [1-13 C] pyruvate into [1-13 C] lactate was elevated following acute hypoxia, suggestive of enhanced anaerobic glycolysis. Elevated HP pyruvate to lactate conversion was also seen at the one week timepoint, in concert with an increase in lactate dehydrogenase (LDH) expression. Following three weeks of hypoxic exposure, cardiac metabolism of pyruvate was comparable with that observed in normoxia. We have successfully visualized the effects of systemic hypoxia on cardiac metabolism of pyruvate using hyperpolarized 13 C MRS, with differences observed following 30 minutes and 1 week of hypoxia. This demonstrates the potential of in vivo hyperpolarized 13 C MRS data for assessing the cardiometabolic effects of hypoxia in disease.
Collapse
Affiliation(s)
- Lydia M. Le Page
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
- Department of Physical Therapy and Rehabilitation ScienceUniversity of CaliforniaSan FranciscoSan FranciscoUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoSan FranciscoUSA
| | - Oliver J. Rider
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Andrew J. Lewis
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Victoria Noden
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Matthew Kerr
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Lucia Giles
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Lucy J.A. Ambrose
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Vicky Ball
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Latt Mansor
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Lisa C. Heather
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Damian J. Tyler
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
19
|
Sowton AP, Griffin JL, Murray AJ. Metabolic Profiling of the Diabetic Heart: Toward a Richer Picture. Front Physiol 2019; 10:639. [PMID: 31214041 PMCID: PMC6555155 DOI: 10.3389/fphys.2019.00639] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/06/2019] [Indexed: 01/20/2023] Open
Abstract
The increasing global prevalence of diabetes has been accompanied by a rise in diabetes-related conditions. This includes diabetic cardiomyopathy (DbCM), a progressive form of heart disease that occurs with both insulin-dependent (type-1) and insulin-independent (type-2) diabetes and arises in the absence of hypertension or coronary artery disease. Over time, DbCM can develop into overt heart failure. Like other forms of cardiomyopathy, DbCM is accompanied by alterations in metabolism which could lead to further progression of the pathology, with metabolic derangement postulated to precede functional changes in the diabetic heart. Moreover in the case of type-2 diabetes, underlying insulin resistance is likely to prevent the canonical substrate switch of the failing heart away from fatty acid oxidation toward increased use of glycolysis. Analytical chemistry techniques, collectively known as metabolomics, are useful tools for investigating the condition. In this article, we provide a comprehensive review of those studies that have employed metabolomic techniques, namely chromatography, mass spectrometry and nuclear magnetic resonance spectroscopy, to profile metabolic remodeling in the diabetic heart of human patients and animal models. These studies collectively demonstrate that glycolysis and glucose oxidation are suppressed in the diabetic myocardium and highlight a complex picture regarding lipid metabolism. The diabetic heart typically shows an increased reliance on fatty acid oxidation, yet triacylglycerols and other lipids accumulate in the diabetic myocardium indicating probable lipotoxicity. The application of lipidomic techniques to the diabetic heart has identified specific lipid species that become enriched and which may in turn act as plasma-borne biomarkers for the condition. Metabolomics is proving to be a powerful approach, allowing a much richer analysis of the metabolic alterations that occur in the diabetic heart. Careful physiological interpretation of metabolomic results will now be key in order to establish which aspects of the metabolic derangement are causal to the progression of DbCM and might form the basis for novel therapeutic intervention.
Collapse
Affiliation(s)
- Alice P. Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Julian L. Griffin
- Department of Biochemistry and Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Andrew J. Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
20
|
Onwuli DO, Samuel SF, Sfyri P, Welham K, Goddard M, Abu-Omar Y, Loubani M, Rivero F, Matsakas A, Benoit DM, Wade M, Greenman J, Beltran-Alvarez P. The inhibitory subunit of cardiac troponin (cTnI) is modified by arginine methylation in the human heart. Int J Cardiol 2019; 282:76-80. [PMID: 30772011 DOI: 10.1016/j.ijcard.2019.01.102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 01/29/2019] [Indexed: 02/01/2023]
Abstract
BACKGROUND The inhibitory subunit of cardiac troponin (cTnI) is a gold standard cardiac biomarker and also an essential protein in cardiomyocyte excitation-contraction coupling. The interactions of cTnI with other proteins are fine-tuned by post-translational modification of cTnI. Mutations in cTnI can lead to hypertrophic cardiomyopathy. METHODS AND RESULTS Here we report, for the first time, that cTnI is modified by arginine methylation in human myocardium. Using Western blot, we observed reduced levels of cTnI arginine methylation in human hypertrophic cardiomyopathy compared to dilated cardiomyopathy biopsies. Similarly, using a rat model of cardiac hypertrophy we observed reduced levels of cTnI arginine methylation compared to sham controls. Using mass spectrometry, we identified cTnI methylation sites at R74/R79 and R146/R148 in human cardiac samples. R146 and R148 lie at the boundary between the critical cTnI inhibitory and switch peptides; PRMT1 methylated an extended inhibitory peptide at R146 and R148 in vitro. Mutations at R145 that have been associated with hypertrophic cardiomyopathy hampered R146/R148 methylation by PRMT1 in vitro. H9c2 cardiac-like cells transfected with plasmids encoding for a methylation-deficient R146A/R148A cTnI protein developed cell hypertrophy, with a 32% increase in cell size after 72 h, compared to control cells. DISCUSSION Our results provide evidence for a novel and significant cTnI post-translational modification. Our work opens the door to translational investigations of cTnI arginine methylation as a biomarker of disease, which can include e.g. cardiomyopathies, myocardial infarction and heart failure, and offers a novel way to investigate the effect of cTnI mutations in the inhibitory/switch peptides.
Collapse
Affiliation(s)
- Donatus O Onwuli
- Biomedical Sciences, University of Hull, Cottingham Rd, HU6 7RX Hull, UK
| | | | - Pagona Sfyri
- Hull York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Kevin Welham
- School of Chemistry, University of Hull, Cottingham Rd, HU6 7RX Hull, UK
| | - Martin Goddard
- Papworth Hospital NHS Foundation Trust, Papworth Everard, Cambridge CB23 3RE, UK
| | - Yasir Abu-Omar
- Papworth Hospital NHS Foundation Trust, Papworth Everard, Cambridge CB23 3RE, UK
| | - Mahmoud Loubani
- Hull York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK; Hull and East Yorkshire Hospitals NHS Trust, Castle Rd, Cottingham HU16 5JQ, UK
| | - Francisco Rivero
- Hull York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Antonios Matsakas
- Hull York Medical School, University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - David M Benoit
- School of Chemistry, University of Hull, Cottingham Rd, HU6 7RX Hull, UK
| | - Mark Wade
- Biomedical Sciences, University of Hull, Cottingham Rd, HU6 7RX Hull, UK
| | - John Greenman
- Biomedical Sciences, University of Hull, Cottingham Rd, HU6 7RX Hull, UK
| | | |
Collapse
|
21
|
Loss of LRRC25 accelerates pathological cardiac hypertrophy through promoting fibrosis and inflammation regulated by TGF-β1. Biochem Biophys Res Commun 2018; 506:137-144. [PMID: 30340835 DOI: 10.1016/j.bbrc.2018.09.065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 09/10/2018] [Indexed: 01/07/2023]
Abstract
Despite advances in therapeutic strategies, heart failure-associated mortality rates remain high. Thus, understanding the pathophysiological molecular mechanisms involved in the remodeling process is essential for developing new and effective therapies. LRRs are present various prokaryotic and eukaryotic proteins and important for the innate immune system via regulating protein-protein interactions. LRRC25 is a member of leucine-rich repeat (LRR)-containing protein family. LRRC25 has been shown to negatively modulate nuclear factor κB (NF-κB) activation, a crucial factor related to cardiac hypertrophy. Our aim was to explore the effects of LRRC25 on cardiac hypertrophy. In the present study, LRRC25 levels were decreased in human and mouse hypertrophied hearts. LRRC25 knockout exacerbated cardiac hypertrophy responding to pressure overloading or angiotensin II (Ang II) stimulation. Deletion of LRRC25 accelerated cardiac dysfunction and fibrosis in mice subjected to aortic banding (AB). LRRC25 ablation induced a strong increase in the transcription of both hypertrophy (ANP, BNP, and β-MHC) and fibrosis associated molecules (col1, col3a1, α-SMA and fibronectin). In addition, the expression of transforming growth factor-β1 (TGF-β1), and its down-streaming signals of phosphorylated Smad2/3, was markedly induced by LRRC25 deficiency. LRRC25-knockout mice showed a significantly enhanced inflammation in response to AB surgery by promoting the activation of NF-κB signaling pathway. In mouse cardiomyocytes, LRRC25 deficiency markedly elevated TGF-β1 and NF-κB activation stimulated by Ang II. Treatment with a combination of TGF-β1 or NF-κB inhibitor abolished the effects of LRRC25-knockout on the promotion of cardiac hypertrophy in vitro. Together, our study identified LRRC25 as a critical molecular switch whose down-regulation resulted in cardiac hypertrophy in a TGF-β1- and NF-κB-dependent manner.
Collapse
|
22
|
Karwi QG, Uddin GM, Ho KL, Lopaschuk GD. Loss of Metabolic Flexibility in the Failing Heart. Front Cardiovasc Med 2018; 5:68. [PMID: 29928647 PMCID: PMC5997788 DOI: 10.3389/fcvm.2018.00068] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/18/2018] [Indexed: 12/15/2022] Open
Abstract
To maintain its high energy demand the heart is equipped with a highly complex and efficient enzymatic machinery that orchestrates ATP production using multiple energy substrates, namely fatty acids, carbohydrates (glucose and lactate), ketones and amino acids. The contribution of these individual substrates to ATP production can dramatically change, depending on such variables as substrate availability, hormonal status and energy demand. This "metabolic flexibility" is a remarkable virtue of the heart, which allows utilization of different energy substrates at different rates to maintain contractile function. In heart failure, cardiac function is reduced, which is accompanied by discernible energy metabolism perturbations and impaired metabolic flexibility. While it is generally agreed that overall mitochondrial ATP production is impaired in the failing heart, there is less consensus as to what actual switches in energy substrate preference occur. The failing heart shift toward a greater reliance on glycolysis and ketone body oxidation as a source of energy, with a decrease in the contribution of glucose oxidation to mitochondrial oxidative metabolism. The heart also becomes insulin resistant. However, there is less consensus as to what happens to fatty acid oxidation in heart failure. While it is generally believed that fatty acid oxidation decreases, a number of clinical and experimental studies suggest that fatty acid oxidation is either not changed or is increased in heart failure. Of importance, is that any metabolic shift that does occur has the potential to aggravate cardiac dysfunction and the progression of the heart failure. An increasing body of evidence shows that increasing cardiac ATP production and/or modulating cardiac energy substrate preference positively correlates with heart function and can lead to better outcomes. This includes increasing glucose and ketone oxidation and decreasing fatty acid oxidation. In this review we present the physiology of the energy metabolism pathways in the heart and the changes that occur in these pathways in heart failure. We also look at the interventions which are aimed at manipulating the myocardial metabolic pathways toward more efficient substrate utilization which will eventually improve cardiac performance.
Collapse
Affiliation(s)
| | | | | | - Gary D. Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
23
|
Timm KN, Miller JJ, Henry JA, Tyler DJ. Cardiac applications of hyperpolarised magnetic resonance. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2018; 106-107:66-87. [PMID: 31047602 DOI: 10.1016/j.pnmrs.2018.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/14/2018] [Accepted: 05/29/2018] [Indexed: 05/05/2023]
Abstract
Cardiovascular disease is the leading cause of death world-wide. It is increasingly recognised that cardiac pathologies show, or may even be caused by, changes in metabolism, leading to impaired cardiac energetics. The heart turns over 15 times its own weight in ATP every day and thus relies heavily on the availability of substrates and on efficient oxidation to generate this ATP. A number of old and emerging drugs that target different aspects of metabolism are showing promising results with regard to improved cardiac outcomes in patients. A non-invasive imaging technique that could assess the role of different aspects of metabolism in heart disease, as well as measure changes in cardiac energetics due to treatment, would be valuable in the routine clinical care of cardiac patients. Hyperpolarised magnetic resonance spectroscopy and imaging have revolutionised metabolic imaging, allowing real-time metabolic flux assessment in vivo for the first time. In this review we summarise metabolism in the healthy and diseased heart, give an introduction to the hyperpolarisation technique, 'dynamic nuclear polarisation' (DNP), and review the preclinical studies that have thus far explored healthy cardiac metabolism and different models of human heart disease. We furthermore show what advances have been made to translate this technique into the clinic, what technical challenges still remain and what unmet clinical needs and unexplored metabolic substrates still need to be assessed by researchers in this exciting and fast-moving field.
Collapse
Affiliation(s)
- Kerstin N Timm
- Department of Physiology, Anatomy and Genetics, University of Oxford, UK.
| | - Jack J Miller
- Department of Physiology, Anatomy and Genetics, University of Oxford, UK; Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Oxford, UK; Clarendon Laboratory, Department of Physics, University of Oxford, UK.
| | - John A Henry
- Department of Physiology, Anatomy and Genetics, University of Oxford, UK.
| | - Damian J Tyler
- Department of Physiology, Anatomy and Genetics, University of Oxford, UK; Oxford Centre for Clinical Magnetic Resonance Research, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
24
|
Apps A, Lau J, Peterzan M, Neubauer S, Tyler D, Rider O. Hyperpolarised magnetic resonance for in vivo real-time metabolic imaging. Heart 2018; 104:1484-1491. [PMID: 29703741 PMCID: PMC6161668 DOI: 10.1136/heartjnl-2017-312356] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/20/2018] [Accepted: 03/22/2018] [Indexed: 01/09/2023] Open
Abstract
Although non-invasive perfusion and viability imaging often provide the gateway to coronary revascularisation, current non-invasive imaging methods only report the surrogate markers of inducible hypoperfusion and presence or absence of myocardial scar, rather than actually visualising areas of ischaemia and/or viable myocardium. This may lead to suboptimal revascularisation decisions. Normally respiring (viable) cardiomyocytes convert pyruvate to acetyl-CoA and CO2/bicarbonate (via pyruvate dehydrogenase), but under ischaemic conditions characteristically shift this conversion to lactate (by lactate dehydrogenase). Imaging pyruvate metabolism thus has the potential to improve upon current imaging techniques. Using the novel hyperpolarisation technique of dynamic nuclear polarisation (DNP), the magnetic resonance signal of injected [1-13C]pyruvate can be transiently magnified >10 000 times over that seen in conventional MR spectroscopy, allowing the characteristic metabolic signatures of ischaemia (lactate production) and viability (CO2/bicarbonate production) to be directly imaged. As such DNP imaging of the downstream metabolism of [1-13C]pyruvate could surpass the diagnostic capabilities of contemporary ischaemia and viability testing. Here we review the technique, and with brief reference to the salient biochemistry, discuss its potential applications within cardiology. These include ischaemia and viability testing, and further characterisation of the altered metabolism seen at different stages during the natural history of heart failure.
Collapse
Affiliation(s)
- Andrew Apps
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Justin Lau
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Mark Peterzan
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Damian Tyler
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Oliver Rider
- Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
25
|
Fillmore N, Levasseur JL, Fukushima A, Wagg CS, Wang W, Dyck JRB, Lopaschuk GD. Uncoupling of glycolysis from glucose oxidation accompanies the development of heart failure with preserved ejection fraction. Mol Med 2018; 24:3. [PMID: 30134787 PMCID: PMC6016884 DOI: 10.1186/s10020-018-0005-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 02/13/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Alterations in cardiac energy metabolism contribute to the development and severity of heart failure (HF). In severe HF, overall mitochondrial oxidative metabolism is significantly decreased resulting in a reduced energy reserve. However, despite the high prevalence of HF with preserved ejection fraction (HFpEF) in our society, it is not clear what changes in cardiac energy metabolism occur in HFpEF, and whether alterations in energy metabolism contribute to the development of contractile dysfunction. METHODS We directly assessed overall energy metabolism during the development of HFpEF in Dahl salt-sensitive rats fed a high salt diet (HSD) for 3, 6 and 9 weeks. RESULTS Over the course of 9 weeks, the HSD caused a progressive decrease in diastolic function (assessed by echocardiography assessment of E'/A'). This was accompanied by a progressive increase in cardiac glycolysis rates (assessed in isolated working hearts obtained at 3, 6, and 9 weeks of HSD). In contrast, the subsequent oxidation of pyruvate from glycolysis (glucose oxidation) was not altered, resulting in an uncoupling of glucose metabolism and a significant increase in proton production. Increased glucose transporter (GLUT)1 expression accompanied this elevation in glycolysis. Decreases in cardiac fatty acid oxidation and overall adenosine triphosphate (ATP) production rates were not observed in early HF, but both significantly decreased as HF progressed to HF with reduced EF (i.e. 9 weeks of HSD). CONCLUSIONS Overall, we show that increased glycolysis is the earliest energy metabolic change that occurs during HFpEF development. The resultant increased proton production from uncoupling of glycolysis and glucose oxidation may contribute to the development of HFpEF.
Collapse
Affiliation(s)
- Natasha Fillmore
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute University of Alberta, Edmonton, Canada
| | - Jody L Levasseur
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute University of Alberta, Edmonton, Canada
| | - Arata Fukushima
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute University of Alberta, Edmonton, Canada
| | - Cory S Wagg
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute University of Alberta, Edmonton, Canada
| | - Wei Wang
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute University of Alberta, Edmonton, Canada
| | - Jason R B Dyck
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute University of Alberta, Edmonton, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute University of Alberta, Edmonton, Canada.
| |
Collapse
|
26
|
Steinhauser J, Wespi P, Kwiatkowski G, Kozerke S. Assessing the influence of isoflurane anesthesia on cardiac metabolism using hyperpolarized [1- 13 C]pyruvate. NMR IN BIOMEDICINE 2018; 31. [PMID: 29206326 DOI: 10.1002/nbm.3856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 09/15/2017] [Accepted: 10/02/2017] [Indexed: 05/07/2023]
Abstract
Isoflurane is a frequently used anesthetic in small-animal dissolution dynamic nuclear polarization-magnetic resonance imaging (DNP-MRI) studies. Although the literature suggests interactions with mitochondrial metabolism, the influence of the compound on cardiac metabolism has not been assessed systematically to date. In the present study, the impact of low versus high isoflurane concentration was examined in a crossover experiment in healthy rats. The results revealed that cardiac metabolism is modulated by isoflurane concentration, showing increased [1-13 C]lactate and reduced [13 C]bicarbonate production during high isoflurane relative to low isoflurane dose [average differences: +16% [1-13 C]lactate/total myocardial carbon, -22% [13 C]bicarbonate/total myocardial carbon; +51% [1-13 C]lactate/[13 C]bicarbonate]. These findings emphasize that reproducible anesthesia is important when studying cardiac metabolism. As the depth of anesthesia is difficult to control in an experimental animal setting, careful study design is required to exclude confounding factors.
Collapse
Affiliation(s)
- Jonas Steinhauser
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Patrick Wespi
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Grzegorz Kwiatkowski
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Sebastian Kozerke
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
27
|
Miller JJ, Lau AZ, Tyler DJ. Susceptibility-induced distortion correction in hyperpolarized echo planar imaging. Magn Reson Med 2017; 79:2135-2141. [PMID: 28722201 PMCID: PMC5836862 DOI: 10.1002/mrm.26839] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/02/2017] [Accepted: 06/24/2017] [Indexed: 12/13/2022]
Abstract
Purpose Echo planar imaging is an attractive rapid imaging readout that can image hyperpolarized compounds in vivo. By alternating the sign of the phase encoding gradient waveform, spatial offsets arising from uncertain frequency shifts can be determined. We show here that blip‐reversed echo planar imaging can also be used to correct for susceptibility and B0 inhomogeneity effects that would otherwise produce image‐domain distortion in the heart. Methods Previously acquired blip‐reversed cardiac 3D‐Spectral‐Spatial echo planar imaging volumetric timecourses of hyperpolarized [1‐13C]pyruvate were distortion corrected by a deformation field estimated by reconstructing signal‐to‐noise ratio (SNR)‐weighted progressively subsampled temporally summed images of each metabolite. Results Reconstructing blip‐reversed data as proposed produced volumetric timecourses that overlaid with proton reference images more consistently than without such corrections. Conclusion The method proposed may form an attractive method to correct for image‐domain distortions in hyperpolarized echo planar imaging experiments. Magn Reson Med 79:2135–2141, 2018. © 2017 The Authors Magnetic Resonance in Medicine published by Wiley Periodicals, Inc. on behalf of International Society for Magnetic Resonance in Medicine. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
Collapse
Affiliation(s)
- Jack J Miller
- Department of Physiology, Anatomy & Genetics, Sherrington Building, University of Oxford, Oxford, United Kingdom.,Department of Physics, Clarendon Laboratory, University of Oxford, Oxford, United Kingdom
| | - Angus Z Lau
- Department of Physics, Clarendon Laboratory, University of Oxford, Oxford, United Kingdom.,Health Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Damian J Tyler
- Department of Physiology, Anatomy & Genetics, Sherrington Building, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
28
|
Peterzan MA, Lygate CA, Neubauer S, Rider OJ. Metabolic remodeling in hypertrophied and failing myocardium: a review. Am J Physiol Heart Circ Physiol 2017. [PMID: 28646030 DOI: 10.1152/ajpheart.00731.2016] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The energy starvation hypothesis proposes that maladaptive metabolic remodeling antedates, initiates, and maintains adverse contractile dysfunction in heart failure (HF). Better understanding of the cardiac metabolic phenotype and metabolic signaling could help identify the role metabolic remodeling plays within HF and the conditions known to transition toward HF, including "pathological" hypertrophy. In this review, we discuss metabolic phenotype and metabolic signaling in the contexts of pathological hypertrophy and HF. We discuss the significance of alterations in energy supply (substrate utilization, oxidative capacity, and phosphotransfer) and energy sensing using observations from human and animal disease models and models of manipulated energy supply/sensing. We aim to provide ways of thinking about metabolic remodeling that center around metabolic flexibility, capacity (reserve), and efficiency rather than around particular substrate preferences or transcriptomic profiles. We show that maladaptive metabolic remodeling takes multiple forms across multiple energy-handling domains. We suggest that lack of metabolic flexibility and reserve (substrate, oxidative, and phosphotransfer) represents a final common denominator ultimately compromising efficiency and contractile reserve in stressful contexts.
Collapse
Affiliation(s)
- Mark A Peterzan
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Oliver J Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
29
|
Imaging oxygen metabolism with hyperpolarized magnetic resonance: a novel approach for the examination of cardiac and renal function. Biosci Rep 2017; 37:BSR20160186. [PMID: 27899435 PMCID: PMC5270319 DOI: 10.1042/bsr20160186] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/28/2016] [Accepted: 11/29/2016] [Indexed: 12/24/2022] Open
Abstract
Every tissue in the body critically depends on meeting its energetic demands with sufficient oxygen supply. Oxygen supply/demand imbalances underlie the diseases that inflict the greatest socio-economic burden globally. The purpose of this review is to examine how hyperpolarized contrast media, used in combination with MR data acquisition methods, may advance our ability to assess oxygen metabolism non-invasively and thus improve management of clinical disease. We first introduce the concept of hyperpolarization and how hyperpolarized contrast media have been practically implemented to achieve translational and clinical research. We will then analyse how incorporating hyperpolarized contrast media could enable realization of unmet technical needs in clinical practice. We will focus on imaging cardiac and renal oxygen metabolism, as both organs have unique physiological demands to satisfy their requirements for tissue oxygenation, their dysfunction plays a fundamental role in society’s most prevalent diseases, and each organ presents unique imaging challenges. It is our aim that this review attracts a multi-disciplinary audience and sparks collaborations that utilize an exciting, emergent technology to advance our ability to treat patients adversely affected by an oxygen supply/demand mismatch.
Collapse
|
30
|
Le Page LM, Ball DR, Ball V, Dodd MS, Miller JJ, Heather LC, Tyler DJ. Simultaneous in vivo assessment of cardiac and hepatic metabolism in the diabetic rat using hyperpolarized MRS. NMR IN BIOMEDICINE 2016; 29:1759-1767. [PMID: 27779334 PMCID: PMC5132204 DOI: 10.1002/nbm.3656] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 08/24/2016] [Accepted: 09/12/2016] [Indexed: 05/07/2023]
Abstract
Understanding and assessing diabetic metabolism is vital for monitoring disease progression and improving treatment of patients. In vivo assessments, using MRI and MRS, provide non-invasive and accurate measurements, and the development of hyperpolarized 13 C spectroscopy in particular has been demonstrated to provide valuable metabolic data in real time. Until now, studies have focussed on individual organs. However, diabetes is a systemic disease affecting multiple tissues in the body. Therefore, we have developed a technique to simultaneously measure metabolism in both the heart and liver during a single acquisition. A hyperpolarized 13 C MRS protocol was developed to allow acquisition of metabolic data from the heart and liver during a single scan. This protocol was subsequently used to assess metabolism in the heart and liver of seven control male Wistar rats and seven diabetic rats (diabetes was induced by three weeks of high-fat feeding and a 30 mg/kg injection of streptozotocin). Using our new acquisition, we observed decreased cardiac and hepatic pyruvate dehydrogenase flux in our diabetic rat model. These diabetic rats also had increased blood glucose levels, decreased insulin, and increased hepatic triglycerides. Decreased production of hepatic [1-13 C]alanine was observed in the diabetic group, but this change was not present in the hearts of the same diabetic animals. We have demonstrated the ability to measure cardiac and hepatic metabolism simultaneously, with sufficient sensitivity to detect metabolic alterations in both organs. Further, we have non-invasively observed the different reactions of the heart and liver to the metabolic challenge of diabetes.
Collapse
Affiliation(s)
- Lydia M. Le Page
- Cardiac Metabolism Research Group, Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCAUSA
| | - Daniel R. Ball
- Cardiac Metabolism Research Group, Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Vicky Ball
- Cardiac Metabolism Research Group, Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Michael S. Dodd
- Cardiac Metabolism Research Group, Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Jack J. Miller
- Cardiac Metabolism Research Group, Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Lisa C. Heather
- Cardiac Metabolism Research Group, Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Damian J. Tyler
- Cardiac Metabolism Research Group, Department of Physiology Anatomy and GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
31
|
Németh BT, Mátyás C, Oláh A, Lux Á, Hidi L, Ruppert M, Kellermayer D, Kökény G, Szabó G, Merkely B, Radovits T. Cinaciguat prevents the development of pathologic hypertrophy in a rat model of left ventricular pressure overload. Sci Rep 2016; 6:37166. [PMID: 27853261 PMCID: PMC5112572 DOI: 10.1038/srep37166] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/25/2016] [Indexed: 01/19/2023] Open
Abstract
Pathologic myocardial hypertrophy develops when the heart is chronically pressure-overloaded. Elevated intracellular cGMP-levels have been reported to prevent the development of pathologic myocardial hypertrophy, therefore we investigated the effects of chronic activation of the cGMP producing enzyme, soluble guanylate cyclase by Cinaciguat in a rat model of pressure overload-induced cardiac hypertrophy. Abdominal aortic banding (AAB) was used to evoke pressure overload-induced cardiac hypertrophy in male Wistar rats. Sham operated animals served as controls. Experimental and control groups were treated with 10 mg/kg/day Cinaciguat (Cin) or placebo (Co) p.o. for six weeks, respectively. Pathologic myocardial hypertrophy was present in the AABCo group following 6 weeks of pressure overload of the heart, evidenced by increased relative heart weight, average cardiomyocyte diameter, collagen content and apoptosis. Cinaciguat did not significantly alter blood pressure, but effectively attenuated all features of pathologic myocardial hypertrophy, and normalized functional changes, such as the increase in contractility following AAB. Our results demonstrate that chronic enhancement of cGMP signalling by pharmacological activation of sGC might be a novel therapeutic approach in the prevention of pathologic myocardial hypertrophy.
Collapse
Affiliation(s)
- Balázs Tamás Németh
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Csaba Mátyás
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Attila Oláh
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Árpád Lux
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - László Hidi
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Mihály Ruppert
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Dalma Kellermayer
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Gábor Kökény
- Institute of Pathophysiology, Semmelweis University, Nagyvárad tér 4., 1089 Budapest, Hungary
| | - Gábor Szabó
- Department of Cardiac Surgery, University of Heidelberg, Im Neuenheimer Feld 110., 69210 Heidelberg, Germany
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Városmajor u. 68., 1122 Budapest, Hungary
| |
Collapse
|
32
|
Yoshihara HAI, Bastiaansen JAM, Berthonneche C, Comment A, Schwitter J. An intact small animal model of myocardial ischemia-reperfusion: Characterization of metabolic changes by hyperpolarized 13C MR spectroscopy. Am J Physiol Heart Circ Physiol 2015; 309:H2058-66. [PMID: 26453328 DOI: 10.1152/ajpheart.00376.2015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 10/08/2015] [Indexed: 02/08/2023]
Abstract
Hyperpolarized carbon-13 magnetic resonance spectroscopy ((13)C MRS) enables the sensitive and noninvasive assessment of the metabolic changes occurring during myocardial ischemia-reperfusion. Ischemia-reperfusion models using hyperpolarized (13)C MRS are established in heart preparations ex vivo and in large animals in vivo, but an in vivo model in small animals would be advantageous to allow the study of reperfusion metabolism with neuroendocrine and inflammatory responses intact with the option to perform a greater number of experiments. A novel intact rat model of ischemia-reperfusion is presented that incorporates hyperpolarized (13)C MRS to characterize reperfusion metabolism. Typically, in an in vivo model, a tissue input function (TIF) is required to account for apparent changes in the metabolism of injected hyperpolarized [1-(13)C]pyruvate resulting from changes in perfusion. Whereas the measurement of a TIF by metabolic imaging is particularly challenging in small animals, the ratios of downstream metabolites can be used as an alternative. The ratio of [(13)C]bicarbonate:[1-(13)C]lactate (RatioBic/Lac) measured within 1-2 min after coronary release decreased vs. baseline in ischemic rats (n = 10, 15-min occlusion, controls: n = 10; P = 0.017 for interaction, 2-way ANOVA). The decrease in oxidative pyruvate metabolism [RatioBic/Lac(Ischemia)/RatioBic/Lac(Baseline)] modestly correlated with area at risk (r = 0.66; P = 0.002). Hyperpolarized (13)C MRS was also used to examine alanine production during ischemia, which is observed in ex vivo models, but no significant change was noted; metrics incorporating [1-(13)C]alanine did not substantially improve the discrimination of ischemic-reperfused myocardium from nonischemic myocardium. This intact rat model, which mimics the human situation of reperfused myocardial infarction, could be highly valuable for the testing of new drugs to treat reperfusion injury, thereby facilitating translational research.
Collapse
Affiliation(s)
- Hikari A I Yoshihara
- Division of Cardiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland; Center for Biomedical Imaging (CIBM), Lausanne, Switzerland; Cardiac MR Center, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Jessica A M Bastiaansen
- Institute of Physics of Biological Systems, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland; Center for Biomedical Imaging (CIBM), Lausanne, Switzerland
| | - Corinne Berthonneche
- Cardiovascular Assessment Facility, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Arnaud Comment
- Institute of Physics of Biological Systems, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland; Center for Biomedical Imaging (CIBM), Lausanne, Switzerland
| | - Juerg Schwitter
- Division of Cardiology, Lausanne University Hospital (CHUV), Lausanne, Switzerland; Cardiac MR Center, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
33
|
Bastiaansen JA, Cheng T, Lei H, Gruetter R, Comment A. Direct noninvasive estimation of myocardial tricarboxylic acid cycle flux in vivo using hyperpolarized 13C magnetic resonance. J Mol Cell Cardiol 2015; 87:129-37. [DOI: 10.1016/j.yjmcc.2015.08.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 07/27/2015] [Accepted: 08/12/2015] [Indexed: 11/28/2022]
|