1
|
Li L, Niemann B, Knapp F, Werner S, Mühlfeld C, Schneider JP, Jurida LM, Molenda N, Schmitz ML, Yin X, Mayr M, Schulz R, Kracht M, Rohrbach S. Comparison of the stage-dependent mitochondrial changes in response to pressure overload between the diseased right and left ventricle in the rat. Basic Res Cardiol 2024; 119:587-611. [PMID: 38758338 DOI: 10.1007/s00395-024-01051-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024]
Abstract
The right ventricle (RV) differs developmentally, anatomically and functionally from the left ventricle (LV). Therefore, characteristics of LV adaptation to chronic pressure overload cannot easily be extrapolated to the RV. Mitochondrial abnormalities are considered a crucial contributor in heart failure (HF), but have never been compared directly between RV and LV tissues and cardiomyocytes. To identify ventricle-specific mitochondrial molecular and functional signatures, we established rat models with two slowly developing disease stages (compensated and decompensated) in response to pulmonary artery banding (PAB) or ascending aortic banding (AOB). Genome-wide transcriptomic and proteomic analyses were used to identify differentially expressed mitochondrial genes and proteins and were accompanied by a detailed characterization of mitochondrial function and morphology. Two clearly distinguishable disease stages, which culminated in a comparable systolic impairment of the respective ventricle, were observed. Mitochondrial respiration was similarly impaired at the decompensated stage, while respiratory chain activity or mitochondrial biogenesis were more severely deteriorated in the failing LV. Bioinformatics analyses of the RNA-seq. and proteomic data sets identified specifically deregulated mitochondrial components and pathways. Although the top regulated mitochondrial genes and proteins differed between the RV and LV, the overall changes in tissue and cardiomyocyte gene expression were highly similar. In conclusion, mitochondrial dysfuntion contributes to disease progression in right and left heart failure. Ventricle-specific differences in mitochondrial gene and protein expression are mostly related to the extent of observed changes, suggesting that despite developmental, anatomical and functional differences mitochondrial adaptations to chronic pressure overload are comparable in both ventricles.
Collapse
MESH Headings
- Animals
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Male
- Heart Failure/metabolism
- Heart Failure/physiopathology
- Heart Failure/pathology
- Heart Failure/genetics
- Disease Models, Animal
- Proteomics
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/genetics
- Ventricular Dysfunction, Right/pathology
- Ventricular Function, Right
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Heart Ventricles/pathology
- Rats
- Ventricular Function, Left
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/genetics
- Transcriptome
- Rats, Sprague-Dawley
- Mitochondrial Proteins/metabolism
- Mitochondrial Proteins/genetics
Collapse
Affiliation(s)
- Ling Li
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| | - Bernd Niemann
- Department of Cardiac and Vascular Surgery, Justus Liebig University Giessen, Rudolf-Buchheim-Street. 8, 35392, Giessen, Germany
| | - Fabienne Knapp
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| | - Sebastian Werner
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Christian Mühlfeld
- Hannover Medical School, Institute of Functional and Applied Anatomy, Carl-Neuberg-Street. 1, 30625, Hannover, Germany
| | - Jan Philipp Schneider
- Hannover Medical School, Institute of Functional and Applied Anatomy, Carl-Neuberg-Street. 1, 30625, Hannover, Germany
| | - Liane M Jurida
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Nicole Molenda
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| | - M Lienhard Schmitz
- Institute of Biochemistry, Justus Liebig University Giessen, Friedrichstr. 24, 35392, Giessen, Germany
| | - Xiaoke Yin
- School of Cardiovascular and Metabolic Medicine and Science, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Manuel Mayr
- School of Cardiovascular and Metabolic Medicine and Science, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany
| | - Michael Kracht
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Susanne Rohrbach
- Institute of Physiology, Justus Liebig University Giessen, Aulweg 129, 35392, Giessen, Germany.
| |
Collapse
|
2
|
Hinton A, Claypool SM, Neikirk K, Senoo N, Wanjalla CN, Kirabo A, Williams CR. Mitochondrial Structure and Function in Human Heart Failure. Circ Res 2024; 135:372-396. [PMID: 38963864 PMCID: PMC11225798 DOI: 10.1161/circresaha.124.323800] [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] [Indexed: 07/06/2024]
Abstract
Despite clinical and scientific advancements, heart failure is the major cause of morbidity and mortality worldwide. Both mitochondrial dysfunction and inflammation contribute to the development and progression of heart failure. Although inflammation is crucial to reparative healing following acute cardiomyocyte injury, chronic inflammation damages the heart, impairs function, and decreases cardiac output. Mitochondria, which comprise one third of cardiomyocyte volume, may prove a potential therapeutic target for heart failure. Known primarily for energy production, mitochondria are also involved in other processes including calcium homeostasis and the regulation of cellular apoptosis. Mitochondrial function is closely related to morphology, which alters through mitochondrial dynamics, thus ensuring that the energy needs of the cell are met. However, in heart failure, changes in substrate use lead to mitochondrial dysfunction and impaired myocyte function. This review discusses mitochondrial and cristae dynamics, including the role of the mitochondria contact site and cristae organizing system complex in mitochondrial ultrastructure changes. Additionally, this review covers the role of mitochondria-endoplasmic reticulum contact sites, mitochondrial communication via nanotunnels, and altered metabolite production during heart failure. We highlight these often-neglected factors and promising clinical mitochondrial targets for heart failure.
Collapse
Affiliation(s)
- Antentor Hinton
- Department of Molecular Physiology and Biophysics (A.H., K.N.), Vanderbilt University Medical Center, Nashville
| | - Steven M. Claypool
- Department of Physiology, Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland (S.M.C., N.S.)
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics (A.H., K.N.), Vanderbilt University Medical Center, Nashville
| | - Nanami Senoo
- Department of Physiology, Mitochondrial Phospholipid Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland (S.M.C., N.S.)
| | - Celestine N. Wanjalla
- Department of Medicine, Division of Clinical Pharmacology (C.N.W., A.K.), Vanderbilt University Medical Center, Nashville
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology (C.N.W., A.K.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation (A.K.)
- Vanderbilt Institute for Global Health (A.K.)
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH (C.R.W.)
| |
Collapse
|
3
|
Bornstein MR, Tian R, Arany Z. Human cardiac metabolism. Cell Metab 2024; 36:1456-1481. [PMID: 38959861 PMCID: PMC11290709 DOI: 10.1016/j.cmet.2024.06.003] [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: 01/06/2024] [Revised: 04/12/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024]
Abstract
The heart is the most metabolically active organ in the human body, and cardiac metabolism has been studied for decades. However, the bulk of studies have focused on animal models. The objective of this review is to summarize specifically what is known about cardiac metabolism in humans. Techniques available to study human cardiac metabolism are first discussed, followed by a review of human cardiac metabolism in health and in heart failure. Mechanistic insights, where available, are reviewed, and the evidence for the contribution of metabolic insufficiency to heart failure, as well as past and current attempts at metabolism-based therapies, is also discussed.
Collapse
Affiliation(s)
- Marc R Bornstein
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Zoltan Arany
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Wang Y, Kulkarni VV, Pantaleón García J, Leiva-Juárez MM, Goldblatt DL, Gulraiz F, Vila Ellis L, Chen J, Longmire MK, Donepudi SR, Lorenzi PL, Wang H, Wong LJ, Tuvim MJ, Evans SE. Antimicrobial mitochondrial reactive oxygen species induction by lung epithelial immunometabolic modulation. PLoS Pathog 2023; 19:e1011138. [PMID: 37695784 PMCID: PMC10522048 DOI: 10.1371/journal.ppat.1011138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 09/26/2023] [Accepted: 08/01/2023] [Indexed: 09/13/2023] Open
Abstract
Pneumonia is a worldwide threat, making discovery of novel means to combat lower respiratory tract infection an urgent need. Manipulating the lungs' intrinsic host defenses by therapeutic delivery of certain pathogen-associated molecular patterns protects mice against pneumonia in a reactive oxygen species (ROS)-dependent manner. Here we show that antimicrobial ROS are induced from lung epithelial cells by interactions of CpG oligodeoxynucleotides (ODN) with mitochondrial voltage-dependent anion channel 1 (VDAC1). The ODN-VDAC1 interaction alters cellular ATP/ADP/AMP localization, increases delivery of electrons to the electron transport chain (ETC), increases mitochondrial membrane potential (ΔΨm), differentially modulates ETC complex activities and consequently results in leak of electrons from ETC complex III and superoxide formation. The ODN-induced mitochondrial ROS yield protective antibacterial effects. Together, these studies identify a therapeutic metabolic manipulation strategy to broadly protect against pneumonia without reliance on antibiotics.
Collapse
Affiliation(s)
- Yongxing Wang
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Vikram V. Kulkarni
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| | - Jezreel Pantaleón García
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Miguel M. Leiva-Juárez
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - David L. Goldblatt
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Fahad Gulraiz
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Lisandra Vila Ellis
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jichao Chen
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Michael K. Longmire
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| | - Sri Ramya Donepudi
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Philip L. Lorenzi
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Hao Wang
- Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Lee-Jun Wong
- Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Michael J. Tuvim
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Scott E. Evans
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| |
Collapse
|
5
|
[Mechano-energetic defects in heart failure]. Herz 2023; 48:123-133. [PMID: 36700949 DOI: 10.1007/s00059-022-05161-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 01/27/2023]
Abstract
Heart failure is characterized by defects in excitation-contraction coupling, energetic deficit and oxidative stress. The energy for cardiac contraction and relaxation is provided in mitochondria, whose function is tightly regulated by excitation-contraction coupling in cardiac myocytes. In heart failure with reduced ejection fraction (HFrEF), alterations in the ion balance in cardiac myocytes impair mitochondrial Ca2+ uptake, which is required for activation of the Krebs cycle, causing an energetic deficit and oxidative stress in mitochondria. Recent clinical studies suggest that in heart failure with preserved ejection fraction (HFpEF), in stark contrast to HFrEF, hypercontractility often occurs as an attempt to compensate for a pathological increase in systemic and pulmonary vascular resistance. This hypercontractility increases cardiac energy and oxygen demands at rest and reduces the contractile, diastolic and coronary reserves, preventing an adequate increase in cardiac output during exercise. Moreover, increased contractility causes long-term maladaptive remodeling processes due to oxidative stress and redox-sensitive prohypertrophic signaling pathways. As overweight and diabetes, particularly in the interplay with hemodynamic stress, are important risk factors for the development of HFpEF, interventions targeting metabolism in particular could ameliorate the development and progression of HFpEF.
Collapse
|
6
|
Wang Y, Kulkarni VV, Pantaleón García J, Leiva-Juárez MM, Goldblatt DL, Gulraiz F, Chen J, Donepudi SR, Lorenzi PL, Wang H, Wong LJ, Tuvim MJ, Evans SE. Antimicrobial mitochondrial reactive oxygen species induction by lung epithelial metabolic reprogramming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524841. [PMID: 36711510 PMCID: PMC9882263 DOI: 10.1101/2023.01.19.524841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Pneumonia is a worldwide threat, making discovery of novel means to combat lower respiratory tract infections an urgent need. We have previously shown that manipulating the lungs' intrinsic host defenses by therapeutic delivery of a unique dyad of pathogen-associated molecular patterns protects mice against pneumonia in a reactive oxygen species (ROS)-dependent manner. Here we show that antimicrobial ROS are induced from lung epithelial cells by interactions of CpG oligodeoxynucleotides (ODNs) with mitochondrial voltage-dependent anion channel 1 (VDAC1) without dependence on Toll-like receptor 9 (TLR9). The ODN-VDAC1 interaction alters cellular ATP/ADP/AMP localization, increases delivery of electrons to the electron transport chain (ETC), enhances mitochondrial membrane potential (Δ Ψm ), and differentially modulates ETC complex activities. These combined effects promote leak of electrons from ETC complex III, resulting in superoxide formation. The ODN-induced mitochondrial ROS yield protective antibacterial effects. Together, these studies identify a therapeutic metabolic manipulation strategy that has the potential to broadly protect patients against pneumonia during periods of peak vulnerability without reliance on currently available antibiotics. Author Summary Pneumonia is a major cause of death worldwide. Increasing antibiotic resistance and expanding immunocompromised populations continue to enhance the clinical urgency to find new strategies to prevent and treat pneumonia. We have identified a novel inhaled therapeutic that stimulates lung epithelial defenses to protect mice against pneumonia in a manner that depends on production of reactive oxygen species (ROS). Here, we report that the induction of protective ROS from lung epithelial mitochondria occurs following the interaction of one component of the treatment, an oligodeoxynucleotide, with the mitochondrial voltage-dependent anion channel 1. This interaction alters energy transfer between the mitochondria and the cytosol, resulting in metabolic reprogramming that drives more electrons into the electron transport chain, then causes electrons to leak from the electron transport chain to form protective ROS. While antioxidant therapies are endorsed in many other disease states, we present here an example of therapeutic induction of ROS that is associated with broad protection against pneumonia without reliance on administration of antibiotics.
Collapse
Affiliation(s)
- Yongxing Wang
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vikram V. Kulkarni
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Jezreel Pantaleón García
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Miguel M. Leiva-Juárez
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David L. Goldblatt
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fahad Gulraiz
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jichao Chen
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sri Ramya Donepudi
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Philip L. Lorenzi
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Hao Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lee-Jun Wong
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael J. Tuvim
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Scott E. Evans
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
7
|
Pathophysiology of heart failure and an overview of therapies. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00025-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
8
|
Mollace V, Rosano GMC, Anker SD, Coats AJS, Seferovic P, Mollace R, Tavernese A, Gliozzi M, Musolino V, Carresi C, Maiuolo J, Macrì R, Bosco F, Chiocchi M, Romeo F, Metra M, Volterrani M. Pathophysiological Basis for Nutraceutical Supplementation in Heart Failure: A Comprehensive Review. Nutrients 2021; 13:257. [PMID: 33477388 PMCID: PMC7829856 DOI: 10.3390/nu13010257] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/25/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
There is evidence demonstrating that heart failure (HF) occurs in 1-2% of the global population and is often accompanied by comorbidities which contribute to increasing the prevalence of the disease, the rate of hospitalization and the mortality. Although recent advances in both pharmacological and non-pharmacological approaches have led to a significant improvement in clinical outcomes in patients affected by HF, residual unmet needs remain, mostly related to the occurrence of poorly defined strategies in the early stages of myocardial dysfunction. Nutritional support in patients developing HF and nutraceutical supplementation have recently been shown to possibly contribute to protection of the failing myocardium, although their place in the treatment of HF requires further assessment, in order to find better therapeutic solutions. In this context, the Optimal Nutraceutical Supplementation in Heart Failure (ONUS-HF) working group aimed to assess the optimal nutraceutical approach to HF in the early phases of the disease, in order to counteract selected pathways that are imbalanced in the failing myocardium. In particular, we reviewed several of the most relevant pathophysiological and molecular changes occurring during the early stages of myocardial dysfunction. These include mitochondrial and sarcoplasmic reticulum stress, insufficient nitric oxide (NO) release, impaired cardiac stem cell mobilization and an imbalanced regulation of metalloproteinases. Moreover, we reviewed the potential of the nutraceutical supplementation of several natural products, such as coenzyme Q10 (CoQ10), a grape seed extract, Olea Europea L.-related antioxidants, a sodium-glucose cotransporter (SGLT2) inhibitor-rich apple extract and a bergamot polyphenolic fraction, in addition to their support in cardiomyocyte protection, in HF. Such an approach should contribute to optimising the use of nutraceuticals in HF, and the effect needs to be confirmed by means of more targeted clinical trials exploring the efficacy and safety of these compounds.
Collapse
Affiliation(s)
- Vincenzo Mollace
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Giuseppe M. C. Rosano
- Cardiology Clinical Academic Group, St George’s Hospitals NHS Trust University of London, London SW17 0QT, UK;
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (A.J.S.C.); (M.V.)
| | - Stefan D. Anker
- Department of Cardiology, Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Andrew J. S. Coats
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (A.J.S.C.); (M.V.)
| | - Petar Seferovic
- Faculty of Medicine, Belgrade University, 11000 Belgrade, Serbia;
| | - Rocco Mollace
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
- Department of Experimental and Applied Medicine, Institute of Cardiology, University of Brescia, 25121 Brescia, Italy;
| | - Annamaria Tavernese
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
- Department of Experimental and Applied Medicine, Institute of Cardiology, University of Brescia, 25121 Brescia, Italy;
| | - Micaela Gliozzi
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Vincenzo Musolino
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Cristina Carresi
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Jessica Maiuolo
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Roberta Macrì
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Francesca Bosco
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Marcello Chiocchi
- Department of Diagnostic Imaging and Interventional Radiology, Policlinico Tor Vergata, 00199 Rome, Italy;
| | - Francesco Romeo
- Department of Experimental Medicine, University of Rome “Tor Vergata”, 00199 Rome, Italy;
| | - Marco Metra
- Department of Experimental and Applied Medicine, Institute of Cardiology, University of Brescia, 25121 Brescia, Italy;
| | - Maurizio Volterrani
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (A.J.S.C.); (M.V.)
| |
Collapse
|
9
|
Lopez R, Marzban B, Gao X, Lauinger E, Van den Bergh F, Whitesall SE, Converso-Baran K, Burant CF, Michele DE, Beard DA. Impaired Myocardial Energetics Causes Mechanical Dysfunction in Decompensated Failing Hearts. FUNCTION 2020; 1:zqaa018. [PMID: 33074265 PMCID: PMC7552914 DOI: 10.1093/function/zqaa018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/11/2020] [Accepted: 09/21/2020] [Indexed: 01/06/2023] Open
Abstract
Cardiac mechanical function is supported by ATP hydrolysis, which provides the chemical-free energy to drive the molecular processes underlying cardiac pumping. Physiological rates of myocardial ATP consumption require the heart to resynthesize its entire ATP pool several times per minute. In the failing heart, cardiomyocyte metabolic dysfunction leads to a reduction in the capacity for ATP synthesis and associated free energy to drive cellular processes. Yet it remains unclear if and how metabolic/energetic dysfunction that occurs during heart failure affects mechanical function of the heart. We hypothesize that changes in phosphate metabolite concentrations (ATP, ADP, inorganic phosphate) that are associated with decompensation and failure have direct roles in impeding contractile function of the myocardium in heart failure, contributing to the whole-body phenotype. To test this hypothesis, a transverse aortic constriction (TAC) rat model of pressure overload, hypertrophy, and decompensation was used to assess relationships between metrics of whole-organ pump function and myocardial energetic state. A multiscale computational model of cardiac mechanoenergetic coupling was used to identify and quantify the contribution of metabolic dysfunction to observed mechanical dysfunction. Results show an overall reduction in capacity for oxidative ATP synthesis fueled by either fatty acid or carbohydrate substrates as well as a reduction in total levels of adenine nucleotides and creatine in myocardium from TAC animals compared to sham-operated controls. Changes in phosphate metabolite levels in the TAC rats are correlated with impaired mechanical function, consistent with the overall hypothesis. Furthermore, computational analysis of myocardial metabolism and contractile dynamics predicts that increased levels of inorganic phosphate in TAC compared to control animals kinetically impair the myosin ATPase crossbridge cycle in decompensated hypertrophy/heart failure.
Collapse
Affiliation(s)
- Rachel Lopez
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Bahador Marzban
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Xin Gao
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Ellen Lauinger
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Françoise Van den Bergh
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Steven E Whitesall
- Frankel Cardiovascular Center Physiology and Phenotyping Core, University of Michigan, Ann Arbor, MI, USA
| | - Kimber Converso-Baran
- Frankel Cardiovascular Center Physiology and Phenotyping Core, University of Michigan, Ann Arbor, MI, USA
| | - Charles F Burant
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA,Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Daniel E Michele
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA,Frankel Cardiovascular Center Physiology and Phenotyping Core, University of Michigan, Ann Arbor, MI, USA
| | - Daniel A Beard
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA,Address correspondence to D.A.B. (e-mail: )
| |
Collapse
|
10
|
Hwang HV, Sandeep N, Paige SL, Ranjbarvaziri S, Hu DQ, Zhao M, Lan IS, Coronado M, Kooiker KB, Wu SM, Fajardo G, Bernstein D, Reddy S. 4HNE Impairs Myocardial Bioenergetics in Congenital Heart Disease-Induced Right Ventricular Failure. Circulation 2020; 142:1667-1683. [PMID: 32806952 DOI: 10.1161/circulationaha.120.045470] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND In patients with complex congenital heart disease, such as those with tetralogy of Fallot, the right ventricle (RV) is subject to pressure overload stress, leading to RV hypertrophy and eventually RV failure. The role of lipid peroxidation, a potent form of oxidative stress, in mediating RV hypertrophy and failure in congenital heart disease is unknown. METHODS Lipid peroxidation and mitochondrial function and structure were assessed in right ventricle (RV) myocardium collected from patients with RV hypertrophy with normal RV systolic function (RV fractional area change, 47.3±3.8%) and in patients with RV failure showing decreased RV systolic function (RV fractional area change, 26.6±3.1%). The mechanism of the effect of lipid peroxidation, mediated by 4-hydroxynonenal ([4HNE] a byproduct of lipid peroxidation) on mitochondrial function and structure was assessed in HL1 murine cardiomyocytes and human induced pluripotent stem cell-derived cardiomyocytes. RESULTS RV failure was characterized by an increase in 4HNE adduction of metabolic and mitochondrial proteins (16 of 27 identified proteins), in particular electron transport chain proteins. Sarcomeric (myosin) and cytoskeletal proteins (desmin, tubulin) also underwent 4HNE adduction. RV failure showed lower oxidative phosphorylation (moderate RV hypertrophy, 287.6±19.75 versus RV failure, 137.8±11.57 pmol/[sec×mL]; P=0.0004), and mitochondrial structural damage. Using a cell model, we show that 4HNE decreases cell number and oxidative phosphorylation (control, 388.1±23.54 versus 4HNE, 143.7±11.64 pmol/[sec×mL]; P<0.0001). Carvedilol, a known antioxidant did not decrease 4HNE adduction of metabolic and mitochondrial proteins and did not improve oxidative phosphorylation. CONCLUSIONS Metabolic, mitochondrial, sarcomeric, and cytoskeletal proteins are susceptible to 4HNE-adduction in patients with RV failure. 4HNE decreases mitochondrial oxygen consumption by inhibiting electron transport chain complexes. Carvedilol did not improve the 4HNE-mediated decrease in oxygen consumption. Strategies to decrease lipid peroxidation could improve mitochondrial energy generation and cardiomyocyte survival and improve RV failure in patients with congenital heart disease.
Collapse
Affiliation(s)
- HyunTae V Hwang
- Department of Pediatrics (Cardiology) (HT.V.H., N.S., S.L.P., S. Ranjbarvairi, D-Q.H., M.Z., G.F., D.B., S. Reddy), Stanford University, Palo Alto, CA
| | - Nefthi Sandeep
- Department of Pediatrics (Cardiology) (HT.V.H., N.S., S.L.P., S. Ranjbarvairi, D-Q.H., M.Z., G.F., D.B., S. Reddy), Stanford University, Palo Alto, CA
| | - Sharon L Paige
- Department of Pediatrics (Cardiology) (HT.V.H., N.S., S.L.P., S. Ranjbarvairi, D-Q.H., M.Z., G.F., D.B., S. Reddy), Stanford University, Palo Alto, CA
| | - Sara Ranjbarvaziri
- Department of Pediatrics (Cardiology) (HT.V.H., N.S., S.L.P., S. Ranjbarvairi, D-Q.H., M.Z., G.F., D.B., S. Reddy), Stanford University, Palo Alto, CA
| | - Dong-Qing Hu
- Department of Pediatrics (Cardiology) (HT.V.H., N.S., S.L.P., S. Ranjbarvairi, D-Q.H., M.Z., G.F., D.B., S. Reddy), Stanford University, Palo Alto, CA
| | - Mingming Zhao
- Department of Pediatrics (Cardiology) (HT.V.H., N.S., S.L.P., S. Ranjbarvairi, D-Q.H., M.Z., G.F., D.B., S. Reddy), Stanford University, Palo Alto, CA
| | - Ingrid S Lan
- Department of Bioengineering (I.S.L.), Stanford University, Palo Alto, CA
| | | | | | - Sean M Wu
- Department of Medicine (Cardiology) (S.M.W.), Stanford University, Palo Alto, CA
| | - Giovanni Fajardo
- Department of Pediatrics (Cardiology) (HT.V.H., N.S., S.L.P., S. Ranjbarvairi, D-Q.H., M.Z., G.F., D.B., S. Reddy), Stanford University, Palo Alto, CA
| | - Daniel Bernstein
- Department of Pediatrics (Cardiology) (HT.V.H., N.S., S.L.P., S. Ranjbarvairi, D-Q.H., M.Z., G.F., D.B., S. Reddy), Stanford University, Palo Alto, CA
| | - Sushma Reddy
- Department of Pediatrics (Cardiology) (HT.V.H., N.S., S.L.P., S. Ranjbarvairi, D-Q.H., M.Z., G.F., D.B., S. Reddy), Stanford University, Palo Alto, CA
| |
Collapse
|
11
|
Ge Z, Li A, McNamara J, Dos Remedios C, Lal S. Pathogenesis and pathophysiology of heart failure with reduced ejection fraction: translation to human studies. Heart Fail Rev 2020; 24:743-758. [PMID: 31209771 DOI: 10.1007/s10741-019-09806-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Heart failure represents the end result of different pathophysiologic processes, which culminate in functional impairment. Regardless of its aetiology, the presentation of heart failure usually involves symptoms of pump failure and congestion, which forms the basis for clinical diagnosis. Pathophysiologic descriptions of heart failure with reduced ejection fraction (HFrEF) are being established. Most commonly, HFrEF is centred on a reactive model where a significant initial insult leads to reduced cardiac output, further triggering a cascade of maladaptive processes. Predisposing factors include myocardial injury of any cause, chronically abnormal loading due to hypertension, valvular disease, or tachyarrhythmias. The pathophysiologic processes behind remodelling in heart failure are complex and reflect systemic neurohormonal activation, peripheral vascular effects and localised changes affecting the cardiac substrate. These abnormalities have been the subject of intense research. Much of the translational successes in HFrEF have come from targeting neurohormonal responses to reduced cardiac output, with blockade of the renin-angiotensin-aldosterone system (RAAS) and beta-adrenergic blockade being particularly fruitful. However, mortality and morbidity associated with heart failure remains high. Although systemic neurohormonal blockade slows disease progression, localised ventricular remodelling still adversely affects contractile function. Novel therapy targeted at improving cardiac contractile mechanics in HFrEF hold the promise of alleviating heart failure at its source, yet so far none has found success. Nevertheless, there are increasing calls for a proximal, 'cardiocentric' approach to therapy. In this review, we examine HFrEF therapy aimed at improving cardiac function with a focus on recent trials and emerging targets.
Collapse
Affiliation(s)
- Zijun Ge
- Sydney Medical School, University of Sydney, Camperdown, Australia
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Amy Li
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
- Department of Pharmacy and Biomedical Science, La Trobe University, Melbourne, Australia
| | - James McNamara
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Cris Dos Remedios
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Sean Lal
- Sydney Medical School, University of Sydney, Camperdown, Australia.
- Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia.
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia.
- Cardiac Research Laboratory, Discipline of Anatomy and Histology, University of Sydney, Anderson Stuart Building (F13), Camperdown, NSW, 2006, Australia.
| |
Collapse
|
12
|
Zhang J, Abel ED. Effective Metabolic Approaches for the Energy Starved Failing Heart: Bioenergetic Resiliency via Redundancy or Something Else? Circ Res 2019; 123:329-331. [PMID: 30026379 DOI: 10.1161/circresaha.118.313308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Jianyi Zhang
- From the Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham (J.Z.)
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism and Department of Internal Medicine, University of Iowa Carver College of Medicine (E.D.A.)
| |
Collapse
|
13
|
Abstract
Metabolic pathways integrate to support tissue homeostasis and to prompt changes in cell phenotype. In particular, the heart consumes relatively large amounts of substrate not only to regenerate ATP for contraction but also to sustain biosynthetic reactions for replacement of cellular building blocks. Metabolic pathways also control intracellular redox state, and metabolic intermediates and end products provide signals that prompt changes in enzymatic activity and gene expression. Mounting evidence suggests that the changes in cardiac metabolism that occur during development, exercise, and pregnancy as well as with pathological stress (eg, myocardial infarction, pressure overload) are causative in cardiac remodeling. Metabolism-mediated changes in gene expression, metabolite signaling, and the channeling of glucose-derived carbon toward anabolic pathways seem critical for physiological growth of the heart, and metabolic inefficiency and loss of coordinated anabolic activity are emerging as proximal causes of pathological remodeling. This review integrates knowledge of different forms of cardiac remodeling to develop general models of how relationships between catabolic and anabolic glucose metabolism may fortify cardiac health or promote (mal)adaptive myocardial remodeling. Adoption of conceptual frameworks based in relational biology may enable further understanding of how metabolism regulates cardiac structure and function.
Collapse
Affiliation(s)
- Andrew A Gibb
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (A.A.G.)
| | - Bradford G Hill
- the Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville School of Medicine, KY (B.G.H.).
| |
Collapse
|
14
|
Cieslik KA, Sekhar RV, Granillo A, Reddy A, Medrano G, Heredia CP, Entman ML, Hamilton DJ, Li S, Reineke E, Gupte AA, Zhang A, Taffet GE. Improved Cardiovascular Function in Old Mice After N-Acetyl Cysteine and Glycine Supplemented Diet: Inflammation and Mitochondrial Factors. J Gerontol A Biol Sci Med Sci 2019. [PMID: 29538624 DOI: 10.1093/gerona/gly034] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Metabolic, inflammatory, and functional changes occur in cardiovascular aging which may stem from oxidative stress and be remediable with antioxidants. Glutathione, an intracellular antioxidant, declines with aging, and supplementation with glutathione precursors, N-acetyl cysteine (NAC) and glycine (Gly), increases tissue glutathione. Thirty-month old mice were fed diets supplemented with NAC or NAC+Gly and, after 7 weeks, cardiac function and molecular studies were performed. The NAC+Gly supplementation improved diastolic function, increasing peak early filling velocity, and reducing relaxation time, left atrial volume, and left ventricle end diastolic pressure. By contrast, cardiac function did not improve with NAC alone. Both diet supplementations decreased cardiac levels of inflammatory mediators; only NAC+Gly reduced leukocyte infiltration. Several mitochondrial genes reduced with aging were upregulated in hearts by NAC+Gly diet supplementation. These Krebs cycle and oxidative phosphorylation enzymes, suggesting improved mitochondrial function, and permeabilized cardiac fibers from NAC+Gly-fed mice produced ATP from carbohydrate and fatty acid sources, whereas fibers from control old mice were less able to utilize fatty acids. Our data indicate that NAC+Gly supplementation can improve diastolic function in the old mouse and may have potential to prevent important morbidities for older people.
Collapse
Affiliation(s)
- Katarzyna A Cieslik
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Rajagopal V Sekhar
- Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, Texas
| | - Alejandro Granillo
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Anilkumar Reddy
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, Texas.,Indus Instruments, Webster, Texas
| | - Guillermo Medrano
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Celia Pena Heredia
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Mark L Entman
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Dale J Hamilton
- Department of Medicine, Houston Methodist, Texas.,Center for Bioenergetics, Houston Methodist Hospital Research Institute, Texas
| | - Shumin Li
- Center for Bioenergetics, Houston Methodist Hospital Research Institute, Texas
| | - Erin Reineke
- Center for Bioenergetics, Houston Methodist Hospital Research Institute, Texas
| | - Anisha A Gupte
- Department of Medicine, Houston Methodist, Texas.,Center for Bioenergetics, Houston Methodist Hospital Research Institute, Texas
| | - Aijun Zhang
- Department of Medicine, Houston Methodist, Texas.,Center for Bioenergetics, Houston Methodist Hospital Research Institute, Texas
| | - George E Taffet
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Medicine, Houston Methodist, Texas.,Section of Geriatrics, Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
15
|
High-resolution respirometry in human endomyocardial biopsies shows reduced ventricular oxidative capacity related to heart failure. Exp Mol Med 2019; 51:1-10. [PMID: 30765687 PMCID: PMC6376010 DOI: 10.1038/s12276-019-0214-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/17/2018] [Accepted: 10/01/2018] [Indexed: 02/07/2023] Open
Abstract
The lifetime risk of developing heart failure is approximately 20%, and survival rates remain poor. Myocardial mitochondrial function has been suggested to play a pivotal role in heart failure pathophysiology. Human studies on ex vivo mitochondrial function have mostly been limited to atrial tissue obtained during open heart surgery and have provided contradictory results. This study aimed at measuring myocardial mitochondrial function in transcatheter ventricular endomyocardial biopsies and assessing the relationship between oxidative capacity and heart function. We enrolled 40 heart failure patients undergoing ventricular assist device surgery or heart transplantation (34 males, age 57 ± 11 years, body mass index 26.6 ± 4.8 kg/m2) and 29 heart transplant recipients of comparable age and body mass index with normal left ventricular function undergoing surveillance biopsies (23 males, 57 ± 12 years, body mass index 26.2 ± 4.1 kg/m2). High-resolution respirometry was established in the myocardium to measure oxidative capacity ex vivo. The mitochondrial oxidative capacity was 90% higher in ventricular compared to atrial tissues (n = 11, p < 0.01) of explanted hearts. Respiration rates were comparable in ventricular samples of heart failure patients obtained during open heart surgery by standard tissue preparation or ex vivo endomyocardial biopsy (r = 0.9988, p < 0.0001, n = 8), and the mitochondrial oxidative capacity in samples from these patients remained stable for 8 h when stored in either of two common preservation buffers. The oxidative capacity was 44% lower in heart failure than in transplant recipients (67 ± 3 vs. 97 ± 5 pmol/[s mg], p < 0.0001) and correlated positively with heart function (r = 0.49, p < 0.01). High-resolution respirometry of ventricular tissue is feasible in transcatheter biopsies, facilitating clinical studies on myocardial mitochondrial function in patients not undergoing heart surgery. The capacity of mitochondria in heart muscle cells to use oxygen to produce energy correlates with cardiac function. Julia Szendroedi at Heinrich-Heine University, Düsseldorf, Germany, and colleagues have established a technique to reliably evaluate mitochondrial energy metabolism in patients with or recovering from heart failure. They showed that the mitochondrial oxidative capacity of cells in the lower heart chambers (ventricles) was significantly higher than in the upper heart chambers (atria). Moreover, they found that mitochondrial oxidative capacity was reduced by 44% in heart muscle biopsies from patients with heart failure compared with biopsies from heart transplant recipients with normal ventricular function. Stimulating the respiration rate of mitochondria in ventricular heart cells could be a promising strategy for improving cardiac function.
Collapse
|
16
|
Yue J, Shen Y, Liang L, Guan X, Zhang X, Xu S, Liang C, Shi W, Xu W. Tracing the molecular dynamics of living mitochondria under phototherapy via surface-enhanced Raman scattering spectroscopy. Analyst 2019; 144:5521-5527. [DOI: 10.1039/c9an01231a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The dynamics of the cell mitochondrion were determined, and the mechanism of action of phototherapy on cancer cells was disclosed by SERS.
Collapse
Affiliation(s)
- Jing Yue
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| | - Yanting Shen
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| | - Lijia Liang
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| | - Xin Guan
- Institute of Frontier Medical Science
- Jilin University
- Changchun 130021
- People's Republic of China
| | - Xue Zhang
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| | - Shuping Xu
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| | - Chongyang Liang
- Institute of Frontier Medical Science
- Jilin University
- Changchun 130021
- People's Republic of China
| | - Wei Shi
- Key Lab for Molecular Enzymology & Engineering of Ministry of Education
- Jilin University
- Changchun 130012
- People's Republic of China
| | - Weiqing Xu
- State Key Laboratory of Supramolecular Structure and Materials
- Institute of Theoretical Chemistry
- College of Chemistry
- Jilin University
- Changchun 130012
| |
Collapse
|
17
|
Bianchi VE. Impact of Nutrition on Cardiovascular Function. Curr Probl Cardiol 2018; 45:100391. [PMID: 30318107 DOI: 10.1016/j.cpcardiol.2018.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/31/2018] [Indexed: 12/11/2022]
Abstract
The metabolic sources of energy for myocardial contractility include mainly free fatty acids (FFA) for 95%, and in lesser amounts for 5% from glucose and minimal contributions from other substrates such lactate, ketones, and amino acids. However, myocardial efficiency is influenced by metabolic condition, overload, and ischemia. During cardiac stress, cardiomyocytes increase glucose oxidation and reduce FFA oxidation. In patients with ischemic coronary disease and heart failure, the low oxygen availability limits myocardial reliance on FFA and glucose utilization must increase. Although glucose uptake is fundamental to cardiomyocyte function, an excessive intracellular glucose level is detrimental. Insulin plays a fundamental role in maintaining myocardial efficiency and in reducing glycemia and inflammation; this is particularly evident in obese and type-2 diabetic patients. An excess of F availability increase fat deposition within cardiomyocytes and reduces glucose oxidation. In patients with high body mass index, a restricted diet or starvation have positive effects on cardiac metabolism and function while, in patients with low body mass index, restrictive diets, or starvation have a deleterious effect. Thus, weight loss in obese patients has positive impacts on ventricular mass and function, whereas, in underweight heart failure patients, such weight reduction adds to the risk of heart damage, predisposing to cachexia. Nutrition plays an essential role in the evolution of cardiovascular disease and should be taken into account. An energy-restricted diet improves myocardial efficiency but can represent a potential risk of heart damage, particularly in patients affected by cardiovascular disease. Micronutrient integration has a marginal effect on cardiovascular efficiency.
Collapse
|
18
|
Inducible Lung Epithelial Resistance Requires Multisource Reactive Oxygen Species Generation To Protect against Viral Infections. mBio 2018; 9:mBio.00696-18. [PMID: 29764948 PMCID: PMC5954225 DOI: 10.1128/mbio.00696-18] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Viral pneumonias cause profound worldwide morbidity, necessitating novel strategies to prevent and treat these potentially lethal infections. Stimulation of intrinsic lung defenses via inhalation of synergistically acting Toll-like receptor (TLR) agonists protects mice broadly against pneumonia, including otherwise-lethal viral infections, providing a potential opportunity to mitigate infectious threats. As intact lung epithelial TLR signaling is required for the inducible resistance and as these cells are the principal targets of many respiratory viruses, the capacity of lung epithelial cells to be therapeutically manipulated to function as autonomous antiviral effectors was investigated. Our work revealed that mouse and human lung epithelial cells could be stimulated to generate robust antiviral responses that both reduce viral burden and enhance survival of isolated cells and intact animals. The antiviral protection required concurrent induction of epithelial reactive oxygen species (ROS) from both mitochondrial and dual oxidase sources, although neither type I interferon enrichment nor type I interferon signaling was required for the inducible protection. Taken together, these findings establish the sufficiency of lung epithelial cells to generate therapeutically inducible antiviral responses, reveal novel antiviral roles for ROS, provide mechanistic insights into inducible resistance, and may provide an opportunity to protect patients from viral pneumonia during periods of peak vulnerability.IMPORTANCE Viruses are the most commonly identified causes of pneumonia and inflict unacceptable morbidity, despite currently available therapies. While lung epithelial cells are principal targets of respiratory viruses, they have also been recently shown to contribute importantly to therapeutically inducible antimicrobial responses. This work finds that lung cells can be stimulated to protect themselves against viral challenges, even in the absence of leukocytes, both reducing viral burden and improving survival. Further, it was found that the protection occurs via unexpected induction of reactive oxygen species (ROS) from spatially segregated sources without reliance on type I interferon signaling. Coordinated multisource ROS generation has not previously been described against viruses, nor has ROS generation been reported for epithelial cells against any pathogen. Thus, these findings extend the potential clinical applications for the strategy of inducible resistance to protect vulnerable people against viral infections and also provide new insights into the capacity of lung cells to protect against infections via novel ROS-dependent mechanisms.
Collapse
|
19
|
Wu S, Zhang A, Li S, Chatterjee S, Qi R, Segura‐Ibarra V, Ferrari M, Gupte A, Blanco E, Hamilton DJ. Polymer Functionalization of Isolated Mitochondria for Cellular Transplantation and Metabolic Phenotype Alteration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1700530. [PMID: 29593955 PMCID: PMC5867055 DOI: 10.1002/advs.201700530] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/26/2017] [Indexed: 05/31/2023]
Abstract
Aberrant mitochondrial energy transfer underlies prevalent chronic health conditions, including cancer, cardiovascular, and neurodegenerative diseases. Mitochondrial transplantation represents an innovative strategy aimed at restoring favorable metabolic phenotypes in cells with dysfunctional energy metabolism. While promising, significant barriers to in vivo translation of this approach abound, including limited cellular uptake and recognition of mitochondria as foreign. The objective is to functionalize isolated mitochondria with a biocompatible polymer to enhance cellular transplantation and eventual in vivo applications. Herein, it is demonstrated that grafting of a polymer conjugate composed of dextran with triphenylphosphonium onto isolated mitochondria protects the organelles and facilitates cellular internalization compared with uncoated mitochondria. Importantly, mitochondrial transplantation into cancer and cardiovascular cells has profound effects on respiration, mediating a shift toward improved oxidative phosphorylation, and reduced glycolysis. These findings represent the first demonstration of polymer functionalization of isolated mitochondria, highlighting a viable strategy for enabling clinical applications of mitochondrial transplantation.
Collapse
Affiliation(s)
- Suhong Wu
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
| | - Aijun Zhang
- Center for BioenergeticsHouston Methodist Research InstituteHoustonTX77030USA
| | - Shumin Li
- Center for BioenergeticsHouston Methodist Research InstituteHoustonTX77030USA
| | - Somik Chatterjee
- Center for BioenergeticsHouston Methodist Research InstituteHoustonTX77030USA
| | - Ruogu Qi
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
| | | | - Mauro Ferrari
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
- Department of MedicineWeill Cornell MedicineNew YorkNY10065USA
| | - Anisha Gupte
- Center for BioenergeticsHouston Methodist Research InstituteHoustonTX77030USA
- Department of PhysiologyWeill Cornell MedicineNew YorkNY10065USA
| | - Elvin Blanco
- Department of NanomedicineHouston Methodist Research InstituteHoustonTX77030USA
| | - Dale J. Hamilton
- Center for BioenergeticsHouston Methodist Research InstituteHoustonTX77030USA
- Department of MedicineWeill Cornell MedicineNew YorkNY10065USA
- Division EndocrinologyDiabetes, and MetabolismDepartment of MedicineHouston Methodist HospitalHoustonTX77030USA
| |
Collapse
|
20
|
Moon SH, Liu X, Cedars AM, Yang K, Kiebish MA, Joseph SM, Kelley J, Jenkins CM, Gross RW. Heart failure-induced activation of phospholipase iPLA 2γ generates hydroxyeicosatetraenoic acids opening the mitochondrial permeability transition pore. J Biol Chem 2017; 293:115-129. [PMID: 29158256 DOI: 10.1074/jbc.ra117.000405] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/08/2017] [Indexed: 11/06/2022] Open
Abstract
Congestive heart failure typically arises from cardiac myocyte necrosis/apoptosis, associated with the pathological opening of the mitochondrial permeability transition pore (mPTP). mPTP opening decreases the mitochondrial membrane potential leading to the activation of Ca2+-independent phospholipase A2γ (iPLA2γ) and the production of downstream toxic metabolites. However, the array of enzymatic mediators and the exact chemical mechanisms responsible for modulating myocardial mPTP opening remain unclear. Herein, we demonstrate that human heart failure activates specific myocardial mitochondrial phospholipases that increase Ca2+-dependent production of toxic hydroxyeicosatetraenoic acids (HETEs) and attenuate the activity of phospholipases that promote the synthesis of protective epoxyeicosatrienoic acids (EETs). Mechanistically, HETEs activated the Ca2+-induced opening of the mPTP in failing human myocardium, and the highly selective pharmacological blockade of either iPLA2γ or lipoxygenases attenuated mPTP opening in failing hearts. In contrast, pharmacological inhibition of cytochrome P450 epoxygenases opened the myocardial mPTP in human heart mitochondria. Remarkably, the major mitochondrial phospholipase responsible for Ca2+-activated release of arachidonic acid (AA) in mitochondria from non-failing hearts was calcium-dependent phospholipase A2ζ (cPLA2ζ) identified by sequential column chromatographies and activity-based protein profiling. In contrast, iPLA2γ predominated in failing human myocardium. Stable isotope kinetics revealed that in non-failing human hearts, cPLA2ζ metabolically channels arachidonic acid into EETs, whereas in failing hearts, increased iPLA2γ activity channels AA into toxic HETEs. These results mechanistically identify the sequelae of pathological remodeling of human mitochondrial phospholipases in failing myocardium. This remodeling metabolically channels AA into toxic HETEs promoting mPTP opening, which induces necrosis/apoptosis leading to further progression of heart failure.
Collapse
Affiliation(s)
- Sung Ho Moon
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Xinping Liu
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Ari M Cedars
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Kui Yang
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Michael A Kiebish
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Susan M Joseph
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - John Kelley
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Christopher M Jenkins
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Richard W Gross
- Division of Bioorganic Chemistry and Molecular Pharmacology, the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110; Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110; Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110; Department of Chemistry, Washington University, St. Louis, Missouri 63130.
| |
Collapse
|
21
|
Hamilton DJ, Minze LJ, Kumar T, Cao TN, Lyon CJ, Geiger PC, Hsueh WA, Gupte AA. Estrogen receptor alpha activation enhances mitochondrial function and systemic metabolism in high-fat-fed ovariectomized mice. Physiol Rep 2017; 4:4/17/e12913. [PMID: 27582063 PMCID: PMC5027347 DOI: 10.14814/phy2.12913] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/01/2016] [Indexed: 01/12/2023] Open
Abstract
Estrogen impacts insulin action and cardiac metabolism, and menopause dramatically increases cardiometabolic risk in women. However, the mechanism(s) of cardiometabolic protection by estrogen remain incompletely understood. Here, we tested the effects of selective activation of E2 receptor alpha (ERα) on systemic metabolism, insulin action, and cardiac mitochondrial function in a mouse model of metabolic dysfunction (ovariectomy [OVX], insulin resistance, hyperlipidemia, and advanced age). Middle-aged (12-month-old) female low-density lipoprotein receptor (Ldlr)(-/-) mice were subjected to OVX or sham surgery and fed "western" high-fat diet (WHFD) for 3 months. Selective ERα activation with 4,4',4″-(4-Propyl-[1H]-pyrazole-1,3,5-triyl) (PPT), prevented weight gain, improved insulin action, and reduced visceral fat accumulation in WHFD-fed OVX mice. PPT treatment also elevated systemic metabolism, increasing oxygen consumption and core body temperature, induced expression of several metabolic genes such as peroxisome proliferator-activated receptor gamma, coactivator 1 alpha, and nuclear respiratory factor 1 in heart, liver, skeletal muscle, and adipose tissue, and increased cardiac mitochondrial function. Taken together, selective activation of ERα with PPT enhances metabolic effects including insulin resistance, whole body energy metabolism, and mitochondrial function in OVX mice with metabolic syndrome.
Collapse
Affiliation(s)
- Dale J Hamilton
- Center for Metabolic and Bioenergetics Research, Houston Methodist Research Institute and Weill Cornell Medical College, Houston, Texas Houston Methodist Research Institute, Houston, Texas Houston Methodist Department of Medicine, Houston, Texas
| | | | - Tanvi Kumar
- Houston Methodist Research Institute, Houston, Texas
| | - Tram N Cao
- Houston Methodist Research Institute, Houston, Texas
| | | | - Paige C Geiger
- University of Kansas Medical Center, Kansas City, Kansas
| | | | - Anisha A Gupte
- Center for Metabolic and Bioenergetics Research, Houston Methodist Research Institute and Weill Cornell Medical College, Houston, Texas Houston Methodist Research Institute, Houston, Texas
| |
Collapse
|
22
|
Li Z, Gupte AA, Zhang A, Hamilton DJ. Pet Imaging and its Application in Cardiovascular Diseases. Methodist Debakey Cardiovasc J 2017; 13:29-33. [PMID: 28413580 DOI: 10.14797/mdcj-13-1-29] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide and represent a great challenge for modern research and medicine. Despite advances in preventing and treating CVD over the decades, there remains an urgent need to develop sensitive and safe methods for early detection and personalized treatment. With refinements of molecular imaging technologies such as positron emission tomography (PET), noninvasive imaging of CVDs is experiencing impressive progress in both preclinical and clinical settings. In this review, we summarize advances in cardiovascular PET imaging, highlight the latest development of CVD imaging probes, and illustrate the potential for individualized therapy based on metabolic phenotype.
Collapse
Affiliation(s)
- Zheng Li
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| | - Anisha A Gupte
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| | - Anjun Zhang
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| | - Dale J Hamilton
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
23
|
Hamilton DJ. Metabolic Recovery of the Failing Heart: Emerging Therapeutic Options. Methodist Debakey Cardiovasc J 2017; 13:25-28. [PMID: 28413579 DOI: 10.14797/mdcj-13-1-25] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Heart failure has mortality rates that parallel those of breast cancer. Current management strategies include neurohormonal blockade, rate control measures, natriuretic peptide preservation, implantation of mechanical assist devices, and heart transplantation. Despite these strategies, however, the failing myocardium remains energy depleted. New strategies to promote metabolic recovery are being developed to potentially augment current treatment guidelines. For example, an unexpected finding of our own studies showed that mechanical unloading with assist devices in advanced-stage heart failure restored metabolic flux. Unfortunately, at that point it is too late for myocardial recovery. Traditional metabolic therapies addressing hyperglycemia have had limited long-term outcome benefit. Now, new therapeutic options are emerging based on increased understanding of the molecular mechanisms underlying energy depletion. Metabolic cardiac imaging combined with laboratory diagnostics could guide the design of individual therapeutic strategies. To date, agents that show benefit in select individuals include mimetics that stimulate glucagon-like peptide-1, inhibitors of sodium-glucose cotransporter receptors, drugs that limit fatty acid oxidation, and hormonal therapy in select individuals. This review will summarize mechanisms and investigations related to these metabolic approaches to heart failure.
Collapse
Affiliation(s)
- Dale J Hamilton
- Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
24
|
Sheeran FL, Pepe S. Mitochondrial Bioenergetics and Dysfunction in Failing Heart. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:65-80. [PMID: 28551782 DOI: 10.1007/978-3-319-55330-6_4] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Energy insufficiency has been recognized as a key feature of systolic heart failure. Although mitochondria have long been known to sustain myocardial work energy supply, the capacity to therapeutically target mitochondrial bioenergetics dysfunction is hampered by a complex interplay of multiple perturbations that progressively compound causing myocardial failure and collapse. Compared to non-failing human donor hearts, activity rates of complexes I and IV, nicotinamide nucleotide transhydrogenase (NADPH-transhydrogenase, Nnt) and the Krebs cycle enzymes isocitrate dehydrogenase, malate dehydrogenase and aconitase are markedly decreased in end-stage heart failure. Diminished REDOX capacity with lower total glutathione and coenzyme Q10 levels are also a feature of chronic left ventricular failure. Decreased enzyme activities in part relate to abundant and highly specific oxidative, nitrosylative, and hyperacetylation modifications. In this brief review we highlight that energy deficiency in end-stage failing human left ventricle predominantly involves concomitantly impaired activities of key electron transport chain and Krebs cycle enzymes rather than altered expression of respective genes or proteins. Augmented oxidative modification of these enzyme subunit structures, and the formation of highly reactive secondary metabolites, implicates dysfunction due to diminished capacity for management of mitochondrial reactive oxygen species, which contribute further to progressive decreases in bioenergetic capacity and contractile function in human heart failure.
Collapse
Affiliation(s)
- Freya L Sheeran
- Heart Research, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia.,Royal Children's Hospital, Melbourne, Australia
| | - Salvatore Pepe
- Heart Research, Murdoch Children's Research Institute, Melbourne, Australia. .,Department of Paediatrics, University of Melbourne, Melbourne, Australia. .,Royal Children's Hospital, Melbourne, Australia. .,Department of Cardiology, Royal Children's Hospital, 50 Flemington Road, VIC, 3052, Melbourne, Australia.
| |
Collapse
|
25
|
Gupte AA, Hamilton DJ. Mitochondrial Function in Non-ischemic Heart Failure. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:113-126. [PMID: 28551784 DOI: 10.1007/978-3-319-55330-6_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Provision for the continuous demand for energy from the beating heart relies heavily on efficient mitochondrial activity. Non-ischemic cardiomyopathy in which oxygen supply is not limiting results from etiologies such as pressure overload. It is associated with progressive development of metabolic stress culminating in energy depletion and heart failure. The mitochondria from the ventricular walls undergoing non-ischemic cardiomyopathy are subjected to long periods of adaptation to support the changing metabolic milieu, which has been described as mal-adaptation since it ultimately results in loss of cardiac contractile function. While the chronicity of exposure to metabolic stressors, co-morbidities and thereby adaptive changes in mitochondria maybe different between ischemic and non-ischemic heart failure, the resulting pathology is very similar, especially in late stage heart failure. Understanding of the mitochondrial changes in early-stage heart failure may guide the development of mitochondrial-targeted therapeutic options to prevent progression of non-ischemic heart failure. This chapter reviews findings of mitochondrial functional changes in animal models and humans with non-ischemic heart failure. While most animal models of non-ischemic heart failure exhibit cardiac mitochondrial dysfunction, studies in humans have been inconsistent despite confirmed reduction in ATP production. This chapter also reviews the possibility of impairment of substrate supply processes upstream of the mitochondria in heart failure, and discusses potential metabolism-targeted therapeutic options.
Collapse
Affiliation(s)
- Anisha A Gupte
- Center for Metabolism and Bioenergetics Research, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, USA.
| | - Dale J Hamilton
- Center for Metabolism and Bioenergetics Research, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, TX, USA.,Houston Methodist, Department of Medicine, Houston, TX, USA
| |
Collapse
|
26
|
The ‘Goldilocks zone’ of fatty acid metabolism; to ensure that the relationship with cardiac function is just right. Clin Sci (Lond) 2017; 131:2079-2094. [DOI: 10.1042/cs20160671] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 12/25/2022]
Abstract
Fatty acids (FA) are the main fuel used by the healthy heart to power contraction, supplying 60–70% of the ATP required. FA generate more ATP per carbon molecule than glucose, but require more oxygen to produce the ATP, making them a more energy dense but less oxygen efficient fuel compared with glucose. The pathways involved in myocardial FA metabolism are regulated at various subcellular levels, and can be divided into sarcolemmal FA uptake, cytosolic activation and storage, mitochondrial uptake and β-oxidation. An understanding of the critical involvement of each of these steps has been amassed from genetic mouse models, where forcing the heart to metabolize too much or too little fat was accompanied by cardiac contractile dysfunction and hypertrophy. In cardiac pathologies, such as heart disease and diabetes, aberrations in FA metabolism occur concomitantly with changes in cardiac function. In heart failure, FA oxidation is decreased, correlating with systolic dysfunction and hypertrophy. In contrast, in type 2 diabetes, FA oxidation and triglyceride storage are increased, and correlate with diastolic dysfunction and insulin resistance. Therefore, too much FA metabolism is as detrimental as too little FA metabolism in these settings. Therapeutic compounds that rebalance FA metabolism may provide a mechanism to improve cardiac function in disease. Just like Goldilocks and her porridge, the heart needs to maintain FA metabolism in a zone that is ‘just right’ to support contractile function.
Collapse
|
27
|
Ruiz M, Labarthe F, Fortier A, Bouchard B, Thompson Legault J, Bolduc V, Rigal O, Chen J, Ducharme A, Crawford PA, Tardif JC, Des Rosiers C. Circulating acylcarnitine profile in human heart failure: a surrogate of fatty acid metabolic dysregulation in mitochondria and beyond. Am J Physiol Heart Circ Physiol 2017; 313:H768-H781. [PMID: 28710072 DOI: 10.1152/ajpheart.00820.2016] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 07/07/2017] [Accepted: 07/07/2017] [Indexed: 12/19/2022]
Abstract
Heart failure (HF) is associated with metabolic perturbations, particularly of fatty acids (FAs), which remain to be better understood in humans. This study aimed at testing the hypothesis that HF patients with reduced ejection fraction display systemic perturbations in levels of energy-related metabolites, especially those reflecting dysregulation of FA metabolism, namely, acylcarnitines (ACs). Circulating metabolites were assessed using mass spectrometry (MS)-based methods in two cohorts. The main cohort consisted of 72 control subjects and 68 HF patients exhibiting depressed left ventricular ejection fraction (25.9 ± 6.9%) and mostly of ischemic etiology with ≥2 comorbidities. HF patients displayed marginal changes in plasma levels of tricarboxylic acid cycle-related metabolites or indexes of mitochondrial or cytosolic redox status. They had, however, 22-79% higher circulating ACs, irrespective of chain length (P < 0.0001, adjusted for sex, age, renal function, and insulin resistance, determined by shotgun MS/MS), which reflects defective mitochondrial β-oxidation, and were significantly associated with levels of NH2-terminal pro-B-type natriuretic peptide levels, a disease severity marker. Subsequent extended liquid chromatography-tandem MS analysis of 53 plasma ACs in a subset group from the primary cohort confirmed and further substantiated with a comprehensive lipidomic analysis in a validation cohort revealed in HF patients a more complex circulating AC profile. The latter included dicarboxylic-ACs and dihydroxy-ACs as well as very long chain (VLC) ACs or sphingolipids with VLCFAs (>20 carbons), which are proxies of dysregulated FA metabolism in peroxisomes. Our study identified alterations in circulating ACs in HF patients that are independent of biological traits and associated with disease severity markers. These alterations reflect dysfunctional FA metabolism in mitochondria but also beyond, namely, in peroxisomes, suggesting a novel mechanism contributing to global lipid perturbations in human HF.NEW & NOTEWORTHY Mass spectrometry-based profiling of circulating energy metabolites, including acylcarnitines, in two cohorts of heart failure versus control subjects revealed multiple alterations in fatty acid metabolism in peroxisomes in addition to mitochondria, thereby highlighting a novel mechanism contributing to global lipid perturbations in heart failure.Listen to this article's corresponding podcast at http://ajpheart.podbean.com/e/acylcarnitines-in-human-heart-failure/.
Collapse
Affiliation(s)
- Matthieu Ruiz
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada.,Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - François Labarthe
- CHRU de Tours, Université François Rabelais, Institut National de la Santé et de la Recherche Médicale U1069, Nutrition, Croissance et Cancer, Tours, France
| | - Annik Fortier
- Montreal Health Innovations Coordinating Center, Montreal, Quebec, Canada
| | - Bertrand Bouchard
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada.,Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Julie Thompson Legault
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada.,Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Virginie Bolduc
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Odile Rigal
- Laboratoire de Biochimie, Hôpital R. Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jane Chen
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - Anique Ducharme
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Peter A Crawford
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | | | - Christine Des Rosiers
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada; .,Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| |
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
|
Diakos NA, Navankasattusas S, Abel ED, Rutter J, McCreath L, Ferrin P, McKellar SH, Miller DV, Park SY, Richardson RS, Deberardinis R, Cox JE, Kfoury AG, Selzman CH, Stehlik J, Fang JC, Li DY, Drakos SG. Evidence of Glycolysis Up-Regulation and Pyruvate Mitochondrial Oxidation Mismatch During Mechanical Unloading of the Failing Human Heart: Implications for Cardiac Reloading and Conditioning. JACC Basic Transl Sci 2016; 1:432-444. [PMID: 28497127 PMCID: PMC5422992 DOI: 10.1016/j.jacbts.2016.06.009] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
LVAD unloading reverses several but not all aspects of myocardial remodeling and usually leads to incomplete cardiac recovery in a subset of patients with advanced HF. We performed metabolomic analysis and mitochondrial structural and functional characterization in paired human myocardial tissue procured from 31 patients with advanced HF at LVAD implant and at heart transplant plus tissue from 11 normal donors. LVAD unloading induces glycolysis up-regulation without a corresponding increase in glucose oxidation. Lack of post-LVAD improvement in mitochondrial function and volume density could explain the glycolysis-glucose oxidation mismatch. Therapeutic interventions, such as myocardial conditioning, that are known to improve mitochondrial biogenesis, structure, and function might further improve cardiac metabolism and energy production and thereby enhance cardiac recovery with LVAD-induced unloading.
This study sought to investigate the effects of mechanical unloading on myocardial energetics and the metabolic perturbation of heart failure (HF) in an effort to identify potential new therapeutic targets that could enhance the unloading-induced cardiac recovery. The authors prospectively examined paired human myocardial tissue procured from 31 advanced HF patients at left ventricular assist device (LVAD) implant and at heart transplant plus tissue from 11 normal donors. They identified increased post-LVAD glycolytic metabolites without a coordinate increase in early, tricarboxylic acid (TCA) cycle intermediates. The increased pyruvate was not directed toward the mitochondria and the TCA cycle for complete oxidation, but instead, was mainly converted to cytosolic lactate. Increased nucleotide concentrations were present, potentially indicating increased flux through the pentose phosphate pathway. Evaluation of mitochondrial function and structure revealed a lack of post-LVAD improvement in mitochondrial oxidative functional capacity, mitochondrial volume density, and deoxyribonucleic acid content. Finally, post-LVAD unloading, amino acid levels were found to be increased and could represent a compensatory mechanism and an alternative energy source that could fuel the TCA cycle by anaplerosis. In summary, the authors report evidence that LVAD unloading induces glycolysis in concert with pyruvate mitochondrial oxidation mismatch, most likely as a result of persistent mitochondrial dysfunction. These findings suggest that interventions known to improve mitochondrial biogenesis, structure, and function, such as controlled cardiac reloading and conditioning, warrant further investigation to enhance unloading-induced reverse remodeling and cardiac recovery.
Collapse
Affiliation(s)
- Nikolaos A Diakos
- Utah Transplantation Affiliated Hospitals (UTAH) Cardiac Transplant Program, University of Utah Health Sciences Center, Intermountain Medical Center, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| | | | - E Dale Abel
- Division of Endocrinology and Metabolism, Department of Medicine, University of Iowa, Iowa City, Iowa
| | - Jared Rutter
- Department of Biochemistry, University of Utah, Salt Lake City, Utah
| | - Lauren McCreath
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| | - Peter Ferrin
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| | - Stephen H McKellar
- Utah Transplantation Affiliated Hospitals (UTAH) Cardiac Transplant Program, University of Utah Health Sciences Center, Intermountain Medical Center, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| | - Dylan V Miller
- Utah Transplantation Affiliated Hospitals (UTAH) Cardiac Transplant Program, University of Utah Health Sciences Center, Intermountain Medical Center, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| | - Song Y Park
- Department of Exercise and Sport Science, University of Utah, Salt Lake City, Utah
| | - Russell S Richardson
- Department of Exercise and Sport Science, University of Utah, Salt Lake City, Utah
| | - Ralph Deberardinis
- Division of Pediatric Genetics and Metabolism, UT Southwestern, Dallas, Texas
| | - James E Cox
- Department of Biochemistry, University of Utah, Salt Lake City, Utah
| | - Abdallah G Kfoury
- Utah Transplantation Affiliated Hospitals (UTAH) Cardiac Transplant Program, University of Utah Health Sciences Center, Intermountain Medical Center, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| | - Craig H Selzman
- Utah Transplantation Affiliated Hospitals (UTAH) Cardiac Transplant Program, University of Utah Health Sciences Center, Intermountain Medical Center, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| | - Josef Stehlik
- Utah Transplantation Affiliated Hospitals (UTAH) Cardiac Transplant Program, University of Utah Health Sciences Center, Intermountain Medical Center, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| | - James C Fang
- Utah Transplantation Affiliated Hospitals (UTAH) Cardiac Transplant Program, University of Utah Health Sciences Center, Intermountain Medical Center, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| | - Dean Y Li
- Utah Transplantation Affiliated Hospitals (UTAH) Cardiac Transplant Program, University of Utah Health Sciences Center, Intermountain Medical Center, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| | - Stavros G Drakos
- Utah Transplantation Affiliated Hospitals (UTAH) Cardiac Transplant Program, University of Utah Health Sciences Center, Intermountain Medical Center, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| |
Collapse
|
30
|
Haque ZK, Wang DZ. How cardiomyocytes sense pathophysiological stresses for cardiac remodeling. Cell Mol Life Sci 2016; 74:983-1000. [PMID: 27714411 DOI: 10.1007/s00018-016-2373-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 09/01/2016] [Accepted: 09/19/2016] [Indexed: 12/14/2022]
Abstract
In the past decades, the cardiovascular community has laid out the fundamental signaling cascades that become awry in the cardiomyocyte during the process of pathologic cardiac remodeling. These pathways are initiated at the cell membrane and work their way to the nucleus to mediate gene expression. Complexity is multiplied as the cardiomyocyte is subjected to cross talk with other cells as well as a barrage of extracellular stimuli and mechanical stresses. In this review, we summarize the signaling cascades that play key roles in cardiac function and then we proceed to describe emerging concepts of how the cardiomyocyte senses the mechanical and environmental stimuli to transition to the deleterious genetic program that defines pathologic cardiac remodeling. As a highlighting example of these processes, we illustrate the transition from a compensated hypertrophied myocardium to a decompensated failing myocardium, which is clinically manifested as decompensated heart failure.
Collapse
Affiliation(s)
- Zaffar K Haque
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 1260 John F. Enders Research Bldg, 320 Longwood Ave, Boston, MA, 02115, USA.
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 1260 John F. Enders Research Bldg, 320 Longwood Ave, Boston, MA, 02115, USA
| |
Collapse
|
31
|
Hypertrophy induced KIF5B controls mitochondrial localization and function in neonatal rat cardiomyocytes. J Mol Cell Cardiol 2016; 97:70-81. [DOI: 10.1016/j.yjmcc.2016.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/27/2016] [Accepted: 04/12/2016] [Indexed: 11/19/2022]
|
32
|
Emelyanova L, Ashary Z, Cosic M, Negmadjanov U, Ross G, Rizvi F, Olet S, Kress D, Sra J, Tajik AJ, Holmuhamedov EL, Shi Y, Jahangir A. Selective downregulation of mitochondrial electron transport chain activity and increased oxidative stress in human atrial fibrillation. Am J Physiol Heart Circ Physiol 2016; 311:H54-63. [PMID: 27199126 PMCID: PMC4967212 DOI: 10.1152/ajpheart.00699.2015] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 04/27/2016] [Indexed: 12/19/2022]
Abstract
Mitochondria are critical for maintaining normal cardiac function, and a deficit in mitochondrial energetics can lead to the development of the substrate that promotes atrial fibrillation (AF) and its progression. However, the link between mitochondrial dysfunction and AF in humans is still not fully defined. The aim of this study was to elucidate differences in the functional activity of mitochondrial oxidative phosphorylation (OXPHOS) complexes and oxidative stress in right atrial tissue from patients without (non-AF) and with AF (AF) who were undergoing open-heart surgery and were not significantly different for age, sex, major comorbidities, and medications. The overall functional activity of the electron transport chain (ETC), NADH:O2 oxidoreductase activity, was reduced by 30% in atrial tissue from AF compared with non-AF patients. This was predominantly due to a selective reduction in complex I (0.06 ± 0.007 vs. 0.09 ± 0.006 nmol·min(-1)·citrate synthase activity(-1), P = 0.02) and II (0.11 ± 0.012 vs. 0.16 ± 0.012 nmol·min(-1)·citrate synthase activity(-1), P = 0.003) functional activity in AF patients. Conversely, complex V activity was significantly increased in AF patients (0.21 ± 0.027 vs. 0.12 ± 0.01 nmol·min(-1)·citrate synthase activity(-1), P = 0.005). In addition, AF patients exhibited a higher oxidative stress with increased production of mitochondrial superoxide (73 ± 17 vs. 11 ± 2 arbitrary units, P = 0.03) and 4-hydroxynonenal level (77.64 ± 30.2 vs. 9.83 ± 2.83 ng·mg(-1) protein, P = 0.048). Our findings suggest that AF is associated with selective downregulation of ETC activity and increased oxidative stress that can contribute to the progression of the substrate for AF.
Collapse
Affiliation(s)
- Larisa Emelyanova
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Zain Ashary
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Milanka Cosic
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Ulugbek Negmadjanov
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Gracious Ross
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Farhan Rizvi
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Susan Olet
- Patient-Centered Research, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin; and
| | - David Kress
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Jasbir Sra
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - A Jamil Tajik
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Ekhson L Holmuhamedov
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Yang Shi
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Arshad Jahangir
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin; Patient-Centered Research, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin; and Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| |
Collapse
|
33
|
Chaggar PS, Williams SG, Yonan N, Fildes J, Venkateswaran R, Shaw SM. Myocardial recovery with mechanical circulatory support. Eur J Heart Fail 2016; 18:1220-1227. [DOI: 10.1002/ejhf.575] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/24/2016] [Accepted: 04/28/2016] [Indexed: 01/18/2023] Open
Affiliation(s)
- Parminder S. Chaggar
- The Transplant Unit; University Hospital of South Manchester; Southmoor Road Manchester M23 9LT UK
- The Manchester Collaborative Centre for Inflammation Research; University of Manchester; Manchester UK
| | - Simon G. Williams
- The Transplant Unit; University Hospital of South Manchester; Southmoor Road Manchester M23 9LT UK
| | - Nizar Yonan
- The Transplant Unit; University Hospital of South Manchester; Southmoor Road Manchester M23 9LT UK
| | - James Fildes
- The Transplant Unit; University Hospital of South Manchester; Southmoor Road Manchester M23 9LT UK
- The Manchester Collaborative Centre for Inflammation Research; University of Manchester; Manchester UK
| | - Rajamiyer Venkateswaran
- The Transplant Unit; University Hospital of South Manchester; Southmoor Road Manchester M23 9LT UK
| | - Steven M. Shaw
- The Transplant Unit; University Hospital of South Manchester; Southmoor Road Manchester M23 9LT UK
| |
Collapse
|
34
|
Lionetti V. Simultaneous exposure to nitric oxide inhibition and angiotensin II overload: is it a murine model of mitochondrial dysfunction in nonischemic heart failure? Am J Physiol Heart Circ Physiol 2016; 310:H1385-7. [DOI: 10.1152/ajpheart.00127.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Vincenzo Lionetti
- Laboratory of Medical Science, Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; and Fondazione Toscana “G. Monasterio,” Pisa, Italy
| |
Collapse
|
35
|
Assessment of Mitochondrial Dysfunction and Monoamine Oxidase Contribution to Oxidative Stress in Human Diabetic Hearts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8470394. [PMID: 27190576 PMCID: PMC4846770 DOI: 10.1155/2016/8470394] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/26/2016] [Accepted: 02/11/2016] [Indexed: 01/08/2023]
Abstract
Mitochondria-related oxidative stress is a pathomechanism causally linked to coronary heart disease (CHD) and diabetes mellitus (DM). Recently, mitochondrial monoamine oxidases (MAOs) have emerged as novel sources of oxidative stress in the cardiovascular system and experimental diabetes. The present study was purported to assess the mitochondrial impairment and the contribution of MAOs-related oxidative stress to the cardiovascular dysfunction in coronary patients with/without DM. Right atrial appendages were obtained from 75 patients randomized into 3 groups: (1) Control (CTRL), valvular patients without CHD; (2) CHD, patients with confirmed CHD; and (3) CHD-DM, patients with CHD and DM. Mitochondrial respiration was measured by high-resolution respirometry and MAOs expression was evaluated by RT-PCR and immunohistochemistry. Hydrogen peroxide (H2O2) emission was assessed by confocal microscopy and spectrophotometrically. The impairment of mitochondrial respiration was substrate-independent in CHD-DM group. MAOs expression was comparable among the groups, with the predominance of MAO-B isoform but no significant differences regarding oxidative stress were detected by either method. Incubation of atrial samples with MAOs inhibitors significantly reduced the H2O2 in all groups. In conclusion, abnormal mitochondrial respiration occurs in CHD and is more severe in DM and MAOs contribute to oxidative stress in human diseased hearts with/without DM.
Collapse
|
36
|
Holzem KM, Vinnakota KC, Ravikumar VK, Madden EJ, Ewald GA, Dikranian K, Beard DA, Efimov IR. Mitochondrial structure and function are not different between nonfailing donor and end-stage failing human hearts. FASEB J 2016; 30:2698-707. [PMID: 27075244 DOI: 10.1096/fj.201500118r] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 04/05/2016] [Indexed: 01/06/2023]
Abstract
During human heart failure, the balance of cardiac energy use switches from predominantly fatty acids (FAs) to glucose. We hypothesized that this substrate shift was the result of mitochondrial degeneration; therefore, we examined mitochondrial oxidation and ultrastructure in the failing human heart by using respirometry, transmission electron microscopy, and gene expression studies of demographically matched donor and failing human heart left ventricular (LV) tissues. Surprisingly, respiratory capacities for failing LV isolated mitochondria (n = 9) were not significantly diminished compared with donor LV isolated mitochondria (n = 7) for glycolysis (pyruvate + malate)- or FA (palmitoylcarnitine)-derived substrates, and mitochondrial densities, assessed via citrate synthase activity, were consistent between groups. Transmission electron microscopy images also showed no ultrastructural remodeling for failing vs. donor mitochondria; however, the fraction of lipid droplets (LDs) in direct contact with a mitochondrion was reduced, and the average distance between an LD and its nearest neighboring mitochondrion was increased. Analysis of FA processing gene expression between donor and failing LVs revealed 0.64-fold reduced transcript levels for the mitochondrial-LD tether, perilipin 5, in the failing myocardium (P = 0.003). Thus, reduced FA use in heart failure may result from improper delivery, potentially via decreased perilipin 5 expression and mitochondrial-LD tethering, and not from intrinsic mitochondrial dysfunction.-Holzem, K. M., Vinnakota, K. C., Ravikumar, V. K., Madden, E. J., Ewald, G. A., Dikranian, K., Beard, D. A., Efimov, I. R. Mitochondrial structure and function are not different between nonfailing donor and end-stage failing human hearts.
Collapse
Affiliation(s)
- Katherine M Holzem
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kalyan C Vinnakota
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Vinod K Ravikumar
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Eli J Madden
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Gregory A Ewald
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Krikor Dikranian
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daniel A Beard
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Igor R Efimov
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA; George Washington University, Washington, D.C., USA
| |
Collapse
|
37
|
Hamilton DJ, Zhang A, Li S, Cao TN, Smith JA, Vedula I, Cordero-Reyes AM, Youker KA, Torre-Amione G, Gupte AA. Combination of angiotensin II and l-NG-nitroarginine methyl ester exacerbates mitochondrial dysfunction and oxidative stress to cause heart failure. Am J Physiol Heart Circ Physiol 2016; 310:H667-80. [PMID: 26747502 DOI: 10.1152/ajpheart.00746.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/22/2015] [Indexed: 11/22/2022]
Abstract
Mitochondrial dysfunction has been implicated as a cause of energy deprivation in heart failure (HF). Herein, we tested individual and combined effects of two pathogenic factors of nonischemic HF, inhibition of nitric oxide synthesis [with l-N(G)-nitroarginine methyl ester (l-NAME)] and hypertension [with angiotensin II (AngII)], on myocardial mitochondrial function, oxidative stress, and metabolic gene expression. l-NAME and AngII were administered individually and in combination to mice for 5 wk. Although all treatments increased blood pressure and reduced cardiac contractile function, the l-NAME + AngII group was associated with the most severe HF, as characterized by edema, hypertrophy, oxidative stress, increased expression of Nppa and Nppb, and decreased expression of Atp2a2 and Camk2b. l-NAME + AngII-treated mice exhibited robust deterioration of cardiac mitochondrial function, as observed by reduced respiratory control ratios in subsarcolemmal mitochondria and reduced state 3 levels in interfibrillar mitochondria for complex I but not for complex II substrates. Cardiac myofibrils showed reduced ADP-supported and oligomycin-inhibited oxygen consumption. Mitochondrial functional impairment was accompanied by reduced mitochondrial DNA content and activities of pyruvate dehydrogenase and complex I but increased H2O2 production and tissue protein carbonyls in hearts from AngII and l-NAME + AngII groups. Microarray analyses revealed the majority of the gene changes attributed to the l-NAME + AngII group. Pathway analyses indicated significant changes in metabolic pathways, such as oxidative phosphorylation, mitochondrial function, cardiac hypertrophy, and fatty acid metabolism in l-NAME + AngII hearts. We conclude that l-NAME + AngII is associated with impaired mitochondrial respiratory function and increased oxidative stress compared with either l-NAME or AngII alone, resulting in nonischemic HF.
Collapse
Affiliation(s)
- Dale J Hamilton
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas; Houston Methodist Department of Medicine, Weill Cornell Medical College, Houston, Texas
| | - Aijun Zhang
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas
| | - Shumin Li
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas
| | - Tram N Cao
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas
| | - Jessie A Smith
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas
| | - Indira Vedula
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas
| | - Andrea M Cordero-Reyes
- Houston Methodist Department of Cardiology, Weill Cornell Medical College, Houston, Texas
| | - Keith A Youker
- Houston Methodist Department of Cardiology, Weill Cornell Medical College, Houston, Texas
| | - Guillermo Torre-Amione
- Houston Methodist Department of Cardiology, Weill Cornell Medical College, Houston, Texas; Catedra de Cardiologia y Medicina Vascular, Tecnologico de Monterrey, Nuevo Leon, Mexico
| | - Anisha A Gupte
- Center for Bioenergetics, Houston Methodist Research Institute, Weill Cornell Medical College, Houston, Texas;
| |
Collapse
|
38
|
Berthiaume J, Kirk J, Ranek M, Lyon R, Sheikh F, Jensen B, Hoit B, Butany J, Tolend M, Rao V, Willis M. Pathophysiology of Heart Failure and an Overview of Therapies. Cardiovasc Pathol 2016. [DOI: 10.1016/b978-0-12-420219-1.00008-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
39
|
Gupte AA, Cordero-Reyes AM, Youker KA, Matsunami RK, Engler DA, Li S, Loebe M, Ashrith G, Torre-Amione G, Hamilton DJ. Differential Mitochondrial Function in Remodeled Right and Nonremodeled Left Ventricles in Pulmonary Hypertension. J Card Fail 2016; 22:73-81. [DOI: 10.1016/j.cardfail.2015.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 08/19/2015] [Accepted: 09/01/2015] [Indexed: 11/26/2022]
|
40
|
Abstract
The current therapy for patients with stable systolic heart failure is largely limited to treatments that interfere with neurohormonal activation. Critical pathophysiological hallmarks of heart failure are an energetic deficit and oxidative stress, and both may be the result of mitochondrial dysfunction. This dysfunction is not (only) the result of defect within mitochondria per se, but is in particular traced to defects in intermediary metabolism and of the regulatory interplay between excitation-contraction coupling and mitochondrial energetics, where defects of cytosolic calcium and sodium handling in failing hearts may play important roles. In the past years, several therapies targeting mitochondria have emerged with promising results in preclinical models. Here, we discuss the mechanisms and results of these mitochondria-targeted therapies, but also of interventions that were not primarily thought to target mitochondria but may have important impact on mitochondrial biology as well, such as iron and exercise. Future research should be directed at further delineating the details of mitochondrial dysfunction in patients with heart failure to further optimize these treatments.
Collapse
|
41
|
Brazhe NA, Evlyukhin AB, Goodilin EA, Semenova AA, Novikov SM, Bozhevolnyi SI, Chichkov BN, Sarycheva AS, Baizhumanov AA, Nikelshparg EI, Deev LI, Maksimov EG, Maksimov GV, Sosnovtseva O. Probing cytochrome c in living mitochondria with surface-enhanced Raman spectroscopy. Sci Rep 2015; 5:13793. [PMID: 26346634 PMCID: PMC4561893 DOI: 10.1038/srep13793] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 08/03/2015] [Indexed: 11/29/2022] Open
Abstract
Selective study of the electron transport chain components in living mitochondria is essential for fundamental biophysical research and for the development of new medical diagnostic methods. However, many important details of inter- and intramembrane mitochondrial processes have remained in shadow due to the lack of non-invasive techniques. Here we suggest a novel label-free approach based on the surface-enhanced Raman spectroscopy (SERS) to monitor the redox state and conformation of cytochrome c in the electron transport chain in living mitochondria. We demonstrate that SERS spectra of living mitochondria placed on hierarchically structured silver-ring substrates provide exclusive information about cytochrome c behavior under modulation of inner mitochondrial membrane potential, proton gradient and the activity of ATP-synthetase. Mathematical simulation explains the observed enhancement of Raman scattering due to high concentration of electric near-field and large contact area between mitochondria and nanostructured surfaces.
Collapse
Affiliation(s)
- Nadezda A. Brazhe
- Department of Biophysics, Biological Faculty, Moscow State University, Leninskie gory 1/12, Moscow, 119234, Russia
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, Copenhagen University, Blegdamsvej 3, Copenhagen, DK-2200, Denmark
| | - Andrey B. Evlyukhin
- Laser Zentrum Hannover e. V., Hollerihallee 8, D-30419 Hannover, Germany
- Department of Technology and Innovation, University of Southern Denmark, Odense M, DK-5230 Denmark
| | - Eugene A. Goodilin
- Department of Nanomaterials, Faculty of Material Sciences, Moscow State University, Moscow, Leninskie gory 1/73, 119991, Russia
- Department of Inorganic chemistry, Faculty of Chemistry, Moscow State University, Moscow, Leninskie gory 1/3, 119991, Russia
- Kurnakov Institute of General and Inorganic Chemistry of Russian Academy of Sciences, Moscow, Leninskiy prospekt, 119992, Russia
| | - Anna A. Semenova
- Department of Nanomaterials, Faculty of Material Sciences, Moscow State University, Moscow, Leninskie gory 1/73, 119991, Russia
| | - Sergey M. Novikov
- Department of Technology and Innovation, University of Southern Denmark, Odense M, DK-5230 Denmark
| | - Sergey I. Bozhevolnyi
- Department of Technology and Innovation, University of Southern Denmark, Odense M, DK-5230 Denmark
| | - Boris N. Chichkov
- Laser Zentrum Hannover e. V., Hollerihallee 8, D-30419 Hannover, Germany
| | - Asya S. Sarycheva
- Department of Nanomaterials, Faculty of Material Sciences, Moscow State University, Moscow, Leninskie gory 1/73, 119991, Russia
| | - Adil A. Baizhumanov
- Department of Biophysics, Biological Faculty, Moscow State University, Leninskie gory 1/12, Moscow, 119234, Russia
| | - Evelina I. Nikelshparg
- Department of Biophysics, Biological Faculty, Moscow State University, Leninskie gory 1/12, Moscow, 119234, Russia
| | - Leonid I. Deev
- Department of Biophysics, Biological Faculty, Moscow State University, Leninskie gory 1/12, Moscow, 119234, Russia
| | - Eugene G. Maksimov
- Department of Biophysics, Biological Faculty, Moscow State University, Leninskie gory 1/12, Moscow, 119234, Russia
| | - Georgy V. Maksimov
- Department of Biophysics, Biological Faculty, Moscow State University, Leninskie gory 1/12, Moscow, 119234, Russia
| | - Olga Sosnovtseva
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, Copenhagen University, Blegdamsvej 3, Copenhagen, DK-2200, Denmark
| |
Collapse
|
42
|
Monoamine Oxidases as Potential Contributors to Oxidative Stress in Diabetes: Time for a Study in Patients Undergoing Heart Surgery. BIOMED RESEARCH INTERNATIONAL 2015; 2015:515437. [PMID: 26101773 PMCID: PMC4458524 DOI: 10.1155/2015/515437] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 09/01/2014] [Accepted: 09/17/2014] [Indexed: 12/19/2022]
Abstract
Oxidative stress is a pathomechanism causally linked to the progression of chronic cardiovascular diseases and diabetes. Mitochondria have emerged as the most relevant source of reactive oxygen species, the major culprit being classically considered the respiratory chain at the inner mitochondrial membrane. In the past decade, several experimental studies unequivocally demonstrated the contribution of monoamine oxidases (MAOs) at the outer mitochondrial membrane to the maladaptative ventricular hypertrophy and endothelial dysfunction. This paper addresses the contribution of mitochondrial dysfunction to the pathogenesis of heart failure and diabetes together with the mounting evidence for an emerging role of MAO inhibition as putative cardioprotective strategy in both conditions.
Collapse
|
43
|
Rees ML, Subramaniam J, Li Y, Hamilton DJ, Frazier OH, Taegtmeyer H. A PKM2 signature in the failing heart. Biochem Biophys Res Commun 2015; 459:430-6. [PMID: 25735978 PMCID: PMC4380635 DOI: 10.1016/j.bbrc.2015.02.122] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/21/2015] [Indexed: 11/28/2022]
Abstract
A salient feature of the failing heart is metabolic remodeling towards predominant glucose metabolism and activation of the fetal gene program. Sunitinib is a multitargeted receptor tyrosine kinase inhibitor used for the treatment of highly vascularized tumors. In diabetic patients, sunitinib significantly decreases blood glucose. However, a considerable proportion of sunitinib-treated patients develop cardiac dysfunction or failure. We asked whether sunitinib treatment results in shift towards glycolysis in the heart. Glucose uptake by the heart was increased fivefold in mice treated with sunitinib. Transcript analysis by qPCR revealed an induction of genes associated with glycolysis and reactivation of the fetal gene program. Additionally, we observed a shift in the enzyme pyruvate kinase from the adult M1 (PKM1) isoform to the fetal M2 (PKM2) isoform, a hallmark of the Warburg Effect. This novel observation led us to examine whether a similar shift occurs in human heart failure. Examination of tissue from patients with heart failure similarly displayed an induction of PKM2. Moreover, this phenomenon was partially reversed following mechanical unloading. We propose that pyruvate kinase isoform switching represents a novel feature of the fetal gene program in the failing heart.
Collapse
Affiliation(s)
- Meredith L Rees
- Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, 6431 Fannin Street, MSB 1.220, Houston, TX 77030, USA
| | - Janani Subramaniam
- Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, 6431 Fannin Street, MSB 1.220, Houston, TX 77030, USA
| | - Yuanteng Li
- Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, 6431 Fannin Street, MSB 1.220, Houston, TX 77030, USA
| | - Dale J Hamilton
- Department of Medicine, Division of Endocrinology, Bioenergetic Laboratory, Houston Methodist Research Institute, 6550 Fannin Street, #1001, Houston, TX 77030, USA
| | - O Howard Frazier
- Texas Heart Institute, CHI St. Luke's Health - Baylor St. Luke's Medical Center, MC 2-114A, PO Box 20345, Houston, TX 77225, USA
| | - Heinrich Taegtmeyer
- Department of Internal Medicine, Division of Cardiology, The University of Texas Medical School at Houston, 6431 Fannin Street, MSB 1.220, Houston, TX 77030, USA; Texas Heart Institute, CHI St. Luke's Health - Baylor St. Luke's Medical Center, MC 2-114A, PO Box 20345, Houston, TX 77225, USA.
| |
Collapse
|
44
|
Shibayama J, Yuzyuk TN, Cox J, Makaju A, Miller M, Lichter J, Li H, Leavy JD, Franklin S, Zaitsev AV. Metabolic remodeling in moderate synchronous versus dyssynchronous pacing-induced heart failure: integrated metabolomics and proteomics study. PLoS One 2015; 10:e0118974. [PMID: 25790351 PMCID: PMC4366225 DOI: 10.1371/journal.pone.0118974] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/08/2015] [Indexed: 01/08/2023] Open
Abstract
Heart failure (HF) is accompanied by complex alterations in myocardial energy metabolism. Up to 40% of HF patients have dyssynchronous ventricular contraction, which is an independent indicator of mortality. We hypothesized that electromechanical dyssynchrony significantly affects metabolic remodeling in the course of HF. We used a canine model of tachypacing-induced HF. Animals were paced at 200 bpm for 6 weeks either in the right atrium (synchronous HF, SHF) or in the right ventricle (dyssynchronous HF, DHF). We collected biopsies from left ventricular apex and performed comprehensive metabolic pathway analysis using multi-platform metabolomics (GC/MS; MS/MS; HPLC) and LC-MS/MS label-free proteomics. We found important differences in metabolic remodeling between SHF and DHF. As compared to Control, ATP, phosphocreatine (PCr), creatine, and PCr/ATP (prognostic indicator of mortality in HF patients) were all significantly reduced in DHF, but not SHF. In addition, the myocardial levels of carnitine (mitochondrial fatty acid carrier) and fatty acids (12:0, 14:0) were significantly reduced in DHF, but not SHF. Carnitine parmitoyltransferase I, a key regulatory enzyme of fatty acid ß-oxidation, was significantly upregulated in SHF but was not different in DHF, as compared to Control. Both SHF and DHF exhibited a reduction, but to a different degree, in creatine and the intermediates of glycolysis and the TCA cycle. In contrast to this, the enzymes of creatine kinase shuttle were upregulated, and the enzymes of glycolysis and the TCA cycle were predominantly upregulated or unchanged in both SHF and DHF. These data suggest a systemic mismatch between substrate supply and demand in pacing-induced HF. The energy deficit observed in DHF, but not in SHF, may be associated with a critical decrease in fatty acid delivery to the ß-oxidation pipeline, primarily due to a reduction in myocardial carnitine content.
Collapse
Affiliation(s)
- Junko Shibayama
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Tatiana N. Yuzyuk
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
- ARUP Laboratories, Salt Lake City, Utah, United States of America
| | - James Cox
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, Utah, United States of America
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Aman Makaju
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Mickey Miller
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Justin Lichter
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
| | - Hui Li
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Jane D. Leavy
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Sarah Franklin
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Alexey V. Zaitsev
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
45
|
Taegtmeyer H, Lubrano G. Rethinking cardiac metabolism: metabolic cycles to refuel and rebuild the failing heart. F1000PRIME REPORTS 2014; 6:90. [PMID: 25374668 PMCID: PMC4191265 DOI: 10.12703/p6-90] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The heart is a self-renewing biological pump that converts chemical energy into mechanical energy. The entire process of energy conversion is subject to complex regulation at the transcriptional, translational and post-translational levels. Within this system, energy transfer occurs with high efficiency, facilitated by a series of compound-conserved cycles. At the same time, the constituent myocardial proteins themselves are continuously made and degraded in order to adjust to changes in energy demand and changes in the extracellular environment. We recently have identified signals arising from intermediary metabolism that regulate the cycle of myocardial protein turnover. Using a new conceptual framework, we discuss the principle of metabolic cycles and their importance for refueling and for rebuilding the failing heart.
Collapse
|