1
|
Lygate CA. Maintaining energy provision in the heart: the creatine kinase system in ischaemia-reperfusion injury and chronic heart failure. Clin Sci (Lond) 2024; 138:491-514. [PMID: 38639724 DOI: 10.1042/cs20230616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/25/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024]
Abstract
The non-stop provision of chemical energy is of critical importance to normal cardiac function, requiring the rapid turnover of ATP to power both relaxation and contraction. Central to this is the creatine kinase (CK) phosphagen system, which buffers local ATP levels to optimise the energy available from ATP hydrolysis, to stimulate energy production via the mitochondria and to smooth out mismatches between energy supply and demand. In this review, we discuss the changes that occur in high-energy phosphate metabolism (i.e., in ATP and phosphocreatine) during ischaemia and reperfusion, which represents an acute crisis of energy provision. Evidence is presented from preclinical models that augmentation of the CK system can reduce ischaemia-reperfusion injury and improve functional recovery. Energetic impairment is also a hallmark of chronic heart failure, in particular, down-regulation of the CK system and loss of adenine nucleotides, which may contribute to pathophysiology by limiting ATP supply. Herein, we discuss the evidence for this hypothesis based on preclinical studies and in patients using magnetic resonance spectroscopy. We conclude that the correlative evidence linking impaired energetics to cardiac dysfunction is compelling; however, causal evidence from loss-of-function models remains equivocal. Nevertheless, proof-of-principle studies suggest that augmentation of CK activity is a therapeutic target to improve cardiac function and remodelling in the failing heart. Further work is necessary to translate these findings to the clinic, in particular, a better understanding of the mechanisms by which the CK system is regulated in disease.
Collapse
Affiliation(s)
- Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| |
Collapse
|
2
|
Chase D, Eykyn TR, Shattock MJ, Chung YJ. Empagliflozin improves cardiac energetics during ischaemia/reperfusion by directly increasing cardiac ketone utilization. Cardiovasc Res 2023; 119:2672-2680. [PMID: 37819017 PMCID: PMC10730240 DOI: 10.1093/cvr/cvad157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/07/2023] [Accepted: 07/27/2023] [Indexed: 10/13/2023] Open
Abstract
AIMS Empagliflozin (EMPA), a potent inhibitor of the renal sodium-glucose cotransporter 2 and an effective treatment for Type 2 diabetes, has been shown to have cardioprotective effects, independent of improved glycaemic control. Several non-canonical mechanisms have been proposed to explain these cardiac effects, including increasing circulating ketone supply to the heart. This study aims to test whether EMPA directly alters cardiac ketone metabolism independent of supply. METHODS AND RESULTS The direct effects of EMPA on cardiac function and metabolomics were investigated in Langendorff rat heart perfused with buffer containing 5 mM glucose, 4 mM β-hydroxybutyrate (βHb) and 0.4 mM intralipid, subject to low flow ischaemia/reperfusion. Cardiac energetics were monitored in situ using 31P NMR spectroscopy. Steady-state 13C labelling was performed by switching 12C substrates for 13C1 glucose or 13C4 βHb and 13C incorporation into metabolites determined using 2D 1H-13C HSQC NMR spectroscopy. EMPA treatment improved left ventricular-developed pressure during ischaemia and reperfusion compared to vehicle-treated hearts. In EMPA-treated hearts, total adenosine triphosphate (ATP) and phosphocreatine (PCr) levels, and Gibbs free energy for ATP hydrolysis were significantly higher during ischaemia and reperfusion. EMPA treatment did not alter the incorporation of 13C from glucose into glycolytic products lactate or alanine neither during ischaemia nor reperfusion. In ischaemia, EMPA led to a decrease in 13C1 glucose incorporation and a concurrent increase in 13C4 βHb incorporation into tricarboxylic acid (TCA) cycle intermediates succinate, citrate, and glutamate. During reperfusion, the concentration of metabolites originating from 13C1 glucose was similar to vehicle but those originating from 13C4 βHb remained elevated in EMPA-treated hearts. CONCLUSION Our findings indicate that EMPA causes a switch in metabolism away from glucose oxidation towards increased ketone utilization in the rat heart, thereby improving function and energetics both during ischaemia and recovery during reperfusion. This preference of ketone utilization over glucose was observed under conditions of constant supply of substrate, suggesting that EMPA acts directly by modulating cardiac substrate preference, independent of substrate availability. The mechanisms underlying our findings are currently unknown, warranting further study.
Collapse
Affiliation(s)
- Dylan Chase
- British Heart Foundation Centre of Research Excellence, King’s College London, The Rayne Institute, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| | - Thomas R Eykyn
- British Heart Foundation Centre of Research Excellence, King’s College London, The Rayne Institute, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London SE1 7EH, UK
| | - Michael J Shattock
- British Heart Foundation Centre of Research Excellence, King’s College London, The Rayne Institute, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| | - Yu Jin Chung
- British Heart Foundation Centre of Research Excellence, King’s College London, The Rayne Institute, 4th Floor, Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| |
Collapse
|
3
|
Rajanathan R, Riera CVI, Pedersen TM, Staehr C, Bouzinova EV, Nyengaard JR, Thomsen MB, Bøtker HE, Matchkov VV. Hypercontractile Cardiac Phenotype in Mice with Migraine-Associated Mutation in the Na +,K +-ATPase α 2-Isoform. Cells 2023; 12:cells12081108. [PMID: 37190017 DOI: 10.3390/cells12081108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Two α-isoforms of the Na+,K+-ATPase (α1 and α2) are expressed in the cardiovascular system, and it is unclear which isoform is the preferential regulator of contractility. Mice heterozygous for the familial hemiplegic migraine type 2 (FHM2) associated mutation in the α2-isoform (G301R; α2+/G301R mice) have decreased expression of cardiac α2-isoform but elevated expression of the α1-isoform. We aimed to investigate the contribution of the α2-isoform function to the cardiac phenotype of α2+/G301R hearts. We hypothesized that α2+/G301R hearts exhibit greater contractility due to reduced expression of cardiac α2-isoform. Variables for contractility and relaxation of isolated hearts were assessed in the Langendorff system without and in the presence of ouabain (1 µM). Atrial pacing was performed to investigate rate-dependent changes. The α2+/G301R hearts displayed greater contractility than WT hearts during sinus rhythm, which was rate-dependent. The inotropic effect of ouabain was more augmented in α2+/G301R hearts than in WT hearts during sinus rhythm and atrial pacing. In conclusion, cardiac contractility was greater in α2+/G301R hearts than in WT hearts under resting conditions. The inotropic effect of ouabain was rate-independent and enhanced in α2+/G301R hearts, which was associated with increased systolic work.
Collapse
Affiliation(s)
| | - Clàudia Vilaseca I Riera
- Department of Basic Science, School of Medicine and Health Sciences, International University of Catalonia, 08195 Barcelona, Spain
| | | | - Christian Staehr
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | | | - Jens Randel Nyengaard
- Department of Clinical Medicine, Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus University, 8000 Aarhus, Denmark
- Department of Pathology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Morten B Thomsen
- Biomedical Sciences, University of Copenhagen, 1168 Copenhagen, Denmark
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | | |
Collapse
|
4
|
Gupta A. Cardiac 31P MR spectroscopy: development of the past five decades and future vision-will it be of diagnostic use in clinics? Heart Fail Rev 2023; 28:485-532. [PMID: 36427161 DOI: 10.1007/s10741-022-10287-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
In the past five decades, the use of the magnetic resonance (MR) technique for cardiovascular diseases has engendered much attention and raised the opportunity that the technique could be useful for clinical applications. MR has two arrows in its quiver: One is magnetic resonance imaging (MRI), and the other is magnetic resonance spectroscopy (MRS). Non-invasively, highly advanced MRI provides unique and profound information about the anatomical changes of the heart. Excellently developed MRS provides irreplaceable and insightful evidence of the real-time biochemistry of cardiac metabolism of underpinning diseases. Compared to MRI, which has already been successfully applied in routine clinical practice, MRS still has a long way to travel to be incorporated into routine diagnostics. Considering the exceptional potential of 31P MRS to measure the real-time metabolic changes of energetic molecules qualitatively and quantitatively, how far its powerful technique should be waited before a successful transition from "bench-to-bedside" is enticing. The present review highlights the seminal studies on the chronological development of cardiac 31P MRS in the past five decades and the future vision and challenges to incorporating it for routine diagnostics of cardiovascular disease.
Collapse
Affiliation(s)
- Ashish Gupta
- Centre of Biomedical Research, SGPGIMS Campus, Lucknow, 226014, India.
| |
Collapse
|
5
|
Galli GM, Strapazzon JV, Marchiori MS, Molosse VL, Deolindo GL, Fracasso M, Copetti PM, Morsch VM, Baldissera MD, Mendes RE, Boiago MM, da Silva AS. Addition of a Blend Based on Zinc Chloride and Lignans of Magnolia in the Diet of Broilers to Substitute for a Conventional Antibiotic: Effects on Intestinal Health, Meat Quality, and Performance. Animals (Basel) 2022; 12:ani12233249. [PMID: 36496770 PMCID: PMC9740953 DOI: 10.3390/ani12233249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
This study aimed to determine whether adding a blend based on zinc chloride and lignans from magnolia to the diet of broilers could replace conventional performance enhancers. For this study, 360 chickens were divided into four groups, with six repetitions per group (n = 15), as follows: CN, without promoter; GPC, control, 50 mg/kg of enramycin growth promoter; T-50, additive blend at a dose of 50 g/ton; and T-100, additive blend at a dose of 100 g/ton. Chickens fed with the additive blend at 50 g/ton showed a production efficiency index equal to that in the GPC group (p < 0.05). At 42 days, the lowest total bacterial count (TBC) was found in the T-100 group, followed by that in the GPC group (p < 0.001). For E. coli, the lowest count was observed in the T-100 group, followed by that in the CP and T-50 groups (p < 0.001). Higher villus/crypt ratios were observed in birds belonging to the T-100 and T-50 groups than in the GPC and NC groups (p < 0.001). Greater water retention was found in the T-50 group than in NC and T-100 groups (p < 0.048). The lowest water loss during cooking was also noted in the T-50 group (p < 0.033). We concluded that adding the antimicrobial blend, primarily at 50 g/ton, maintains the efficiency of the index of production and improves the intestinal health and meat quality of the birds.
Collapse
Affiliation(s)
- Gabriela M. Galli
- Animal Science Graduate Program, Universidade do Estado de Santa Catarina (UDESC), Beloni Trombeta Zanini, n 680-E, Santo Antonio 89815-630, SC, Brazil
| | - João V. Strapazzon
- Animal Science Graduate Program, Universidade do Estado de Santa Catarina (UDESC), Beloni Trombeta Zanini, n 680-E, Santo Antonio 89815-630, SC, Brazil
| | - Maiara S. Marchiori
- Department of Animal Science, Universidade do Estado de Santa Catarina (UDESC), Beloni Trombeta Zanini, n 680-E, Santo Antonio 89815-630, SC, Brazil
| | - Vitor L. Molosse
- Animal Science Graduate Program, Universidade do Estado de Santa Catarina (UDESC), Beloni Trombeta Zanini, n 680-E, Santo Antonio 89815-630, SC, Brazil
| | - Guilherme L. Deolindo
- Animal Science Graduate Program, Universidade do Estado de Santa Catarina (UDESC), Beloni Trombeta Zanini, n 680-E, Santo Antonio 89815-630, SC, Brazil
| | - Mateus Fracasso
- Graduate Program in Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), Av. Roraima, n 1000, Cidade Universitária, Camobi 97105-900, RS, Brazil
| | - Priscila M. Copetti
- Graduate Program in Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), Av. Roraima, n 1000, Cidade Universitária, Camobi 97105-900, RS, Brazil
| | - Vera M. Morsch
- Graduate Program in Toxicological Biochemistry, Universidade Federal de Santa Maria (UFSM), Av. Roraima, n 1000, Cidade Universitária, Camobi 97105-900, RS, Brazil
| | - Matheus D. Baldissera
- Graduate Program in Pharmacology, Universidade Federal de Santa Maria (UFSM), Av. Roraima, n 1000, Cidade Universitária, Camobi 97105-900, RS, Brazil
| | - Ricardo E. Mendes
- Laboratory of Veterinary Pathology, Instituto Federal Catarinense, Rodovia SC 283—km 17, Concordia 89703-720, SC, Brazil
| | - Marcel M. Boiago
- Animal Science Graduate Program, Universidade do Estado de Santa Catarina (UDESC), Beloni Trombeta Zanini, n 680-E, Santo Antonio 89815-630, SC, Brazil
- Department of Animal Science, Universidade do Estado de Santa Catarina (UDESC), Beloni Trombeta Zanini, n 680-E, Santo Antonio 89815-630, SC, Brazil
| | - Aleksandro S. da Silva
- Animal Science Graduate Program, Universidade do Estado de Santa Catarina (UDESC), Beloni Trombeta Zanini, n 680-E, Santo Antonio 89815-630, SC, Brazil
- Department of Animal Science, Universidade do Estado de Santa Catarina (UDESC), Beloni Trombeta Zanini, n 680-E, Santo Antonio 89815-630, SC, Brazil
- Correspondence: ; Tel.: +55-49-20499560
| |
Collapse
|
6
|
Sakurai K, Takeba Y, Osada Y, Mizuno M, Tsuzuki Y, Aso K, Kida K, Ohta Y, Ootaki M, Iiri T, Hokuto I, Shimizu N, Matsumoto N. Antenatal Glucocorticoid Administration Promotes Cardiac Structure and Energy Metabolism Maturation in Preterm Fetuses. Int J Mol Sci 2022; 23:10186. [PMID: 36077580 PMCID: PMC9456503 DOI: 10.3390/ijms231710186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Although the rate of preterm birth has increased in recent decades, a number of preterm infants have escaped death due to improvements in perinatal and neonatal care. Antenatal glucocorticoid (GC) therapy has significantly contributed to progression in lung maturation; however, its potential effects on other organs remain controversial. Furthermore, the effects of antenatal GC therapy on the fetal heart show both pros and cons. Translational research in animal models indicates that constant fetal exposure to antenatal GC administration is sufficient for lung maturation. We have established a premature fetal rat model to investigate immature cardiopulmonary functions in the lungs and heart, including the effects of antenatal GC administration. In this review, we explain the mechanisms of antenatal GC actions on the heart in the fetus compared to those in the neonate. Antenatal GCs may contribute to premature heart maturation by accelerating cardiomyocyte proliferation, angiogenesis, energy production, and sarcoplasmic reticulum function. Additionally, this review specifically focuses on fetal heart growth with antenatal GC administration in experimental animal models. Moreover, knowledge regarding antenatal GC administration in experimental animal models can be coupled with that from developmental biology, with the potential for the generation of functional cells and tissues that could be used for regenerative medical purposes in the future.
Collapse
Affiliation(s)
- Kenzo Sakurai
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| | - Yuko Takeba
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| | - Yosuke Osada
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| | - Masanori Mizuno
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| | - Yoshimitsu Tsuzuki
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| | - Kentaro Aso
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| | - Keisuke Kida
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| | - Yuki Ohta
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| | - Masanori Ootaki
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| | - Taroh Iiri
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| | - Isamu Hokuto
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| | - Naoki Shimizu
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| | - Naoki Matsumoto
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki 216-8511, Kanagawa, Japan
| |
Collapse
|
7
|
Abstract
The design of the energy metabolism system in striated muscle remains a major area of investigation. Here, we review our current understanding and emerging hypotheses regarding the metabolic support of muscle contraction. Maintenance of ATP free energy, so called energy homeostasis, via mitochondrial oxidative phosphorylation is critical to sustained contractile activity, and this major design criterion is the focus of this review. Cell volume invested in mitochondria reduces the space available for generating contractile force, and this spatial balance between mitochondria acontractile elements to meet the varying sustained power demands across muscle types is another important design criterion. This is accomplished with remarkably similar mass-specific mitochondrial protein composition across muscle types, implying that it is the organization of mitochondria within the muscle cell that is critical to supporting sustained muscle function. Beyond the production of ATP, ubiquitous distribution of ATPases throughout the muscle requires rapid distribution of potential energy across these large cells. Distribution of potential energy has long been thought to occur primarily through facilitated metabolite diffusion, but recent analysis has questioned the importance of this process under normal physiological conditions. Recent structural and functional studies have supported the hypothesis that the mitochondrial reticulum provides a rapid energy distribution system via the conduction of the mitochondrial membrane potential to maintain metabolic homeostasis during contractile activity. We extensively review this aspect of the energy metabolism design contrasting it with metabolite diffusion models and how mitochondrial structure can play a role in the delivery of energy in the striated muscle.
Collapse
Affiliation(s)
- Brian Glancy
- Muscle Energetics Laboratory, National Heart, Lung, and Blood Insititute and National Institute of Arthritis and Musculoskeletal and Skin Disease, Bethesda, Maryland
- Laboratory of Cardiac Energetics, National Heart, Lung, and Blood Insititute, Bethesda, Maryland
| | - Robert S Balaban
- Muscle Energetics Laboratory, National Heart, Lung, and Blood Insititute and National Institute of Arthritis and Musculoskeletal and Skin Disease, Bethesda, Maryland
- Laboratory of Cardiac Energetics, National Heart, Lung, and Blood Insititute, Bethesda, Maryland
| |
Collapse
|
8
|
Lygate CA. The Pitfalls of in vivo Cardiac Physiology in Genetically Modified Mice - Lessons Learnt the Hard Way in the Creatine Kinase System. Front Physiol 2021; 12:685064. [PMID: 34054587 PMCID: PMC8160301 DOI: 10.3389/fphys.2021.685064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/22/2021] [Indexed: 12/30/2022] Open
Abstract
In order to fully understand gene function, at some point, it is necessary to study the effects in an intact organism. The creation of the first knockout mouse in the late 1980's gave rise to a revolution in the field of integrative physiology that continues to this day. There are many complex choices when selecting a strategy for genetic modification, some of which will be touched on in this review, but the principal focus is to highlight the potential problems and pitfalls arising from the interpretation of in vivo cardiac phenotypes. As an exemplar, we will scrutinize the field of cardiac energetics and the attempts to understand the role of the creatine kinase (CK) energy buffering and transport system in the intact organism. This story highlights the confounding effects of genetic background, sex, and age, as well as the difficulties in interpreting knockout models in light of promiscuous proteins and metabolic redundancy. It will consider the dose-dependent effects and unintended consequences of transgene overexpression, and the need for experimental rigour in the context of in vivo phenotyping techniques. It is intended that this review will not only bring clarity to the field of cardiac energetics, but also aid the non-expert in evaluating and critically assessing data arising from in vivo genetic modification.
Collapse
Affiliation(s)
- Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
9
|
Physical performance level in sarcomeric mitochondria creatine kinase knockout mouse model throughout ageing. Exp Gerontol 2021; 146:111246. [PMID: 33515657 DOI: 10.1016/j.exger.2021.111246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/10/2021] [Accepted: 01/16/2021] [Indexed: 11/20/2022]
Abstract
PURPOSE The objective of the present study was to establish the role of sarcomeric mitochondrial creatine kinase (Mt-CK) in muscle energy output during exercise in a murine model of ageing (the Mt-CK knock-out mouse, Mt-CK-/-). METHODS Three age groups of Mt-CK-/- mice and control male mice (6, 9, and 18 months of age) underwent incremental treadmill running tests. The maximum speed (Vpeak) and maximal oxygen consumption (VO2peak) values were recorded. Urine samples were analyzed using metabolomic techniques. The skeletal muscle (quadriceps) expression of proteins involved in mitochondria biogenesis, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and dynamin-related GTPase mitofusin 2 (Mnf2) were quantified. RESULTS The VO2 peak (normalized to heart weight: HW) of 18-month-old (mo) Mt-CK-/- mice was 27% (p < 0.001) lower than in 18-mo control mice. The VO2peak/HW ratio was 29% (p < 0.001) lower in 18-mo Mt-CK-/- mice than in 6-mo (p < 0.001) and 32% (p < 0.001) than 9-mo Mt-CK-/- mice. With a 0° slope, Vpeak was 10% (p < 0.05) lower in 18-mo Mt-CK-/- mice than in 6-mo Mt-CK-/- mice but did not differ when comparing the 18-mo and 6-mo control groups. The skeletal muscles weight normalized on body weight in 6-mo Mt-CK-/- were 13 to 14% (p < 0.001, p < 0.05) lower versus the 6-mo control, in addition, the presence of branched-chain amino acids in the urine of 6-mo Mt-CK-/- mice suggests an imbalance in protein turnover (catabolism rather than anabolism) but we did not observe any age-related differences. The expression of PGC-1α and Mnf2 proteins in the quadriceps showed that age-related effects were more prominent than genotype effects. CONCLUSION The present study showed ageing is potentialized by Mt-CK deficiency with regard to VO2peak, Vpeak and mitochondrial protein expression. Our results support that Mt-CK-/- mice undergo physiological adaptations, enabling them to survive and to perform as well as wild-type mice. Furthermore, it is possible that these adaptations in Mt-CK-/- mice have a high energy cost and might trigger premature ageing.
Collapse
|
10
|
Shao D, Kolwicz SC, Wang P, Roe ND, Villet O, Nishi K, Hsu YWA, Flint GV, Caudal A, Wang W, Regnier M, Tian R. Increasing Fatty Acid Oxidation Prevents High-Fat Diet-Induced Cardiomyopathy Through Regulating Parkin-Mediated Mitophagy. Circulation 2020; 142:983-997. [PMID: 32597196 DOI: 10.1161/circulationaha.119.043319] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Increased fatty acid oxidation (FAO) has long been considered a culprit in the development of obesity/diabetes mellitus-induced cardiomyopathy. However, enhancing cardiac FAO by removing the inhibitory mechanism of long-chain fatty acid transport into mitochondria via deletion of acetyl coenzyme A carboxylase 2 (ACC2) does not cause cardiomyopathy in nonobese mice, suggesting that high FAO is distinct from cardiac lipotoxicity. We hypothesize that cardiac pathology-associated obesity is attributable to the imbalance of fatty acid supply and oxidation. Thus, we here seek to determine whether further increasing FAO by inducing ACC2 deletion prevents obesity-induced cardiomyopathy, and if so, to elucidate the underlying mechanisms. METHODS We induced high FAO in adult mouse hearts by cardiac-specific deletion of ACC2 using a tamoxifen-inducible model (ACC2 iKO). Control and ACC2 iKO mice were subjected to high-fat diet (HFD) feeding for 24 weeks to induce obesity. Cardiac function, mitochondria function, and mitophagy activity were examined. RESULTS Despite both control and ACC2 iKO mice exhibiting a similar obese phenotype, increasing FAO oxidation by deletion of ACC2 prevented HFD-induced cardiac dysfunction, pathological remodeling, and mitochondria dysfunction, as well. Similarly, increasing FAO by knockdown of ACC2 prevented palmitate-induced mitochondria dysfunction and cardiomyocyte death in vitro. Furthermore, HFD suppressed mitophagy activity and caused damaged mitochondria to accumulate in the heart, which was attenuated, in part, in the ACC2 iKO heart. Mechanistically, ACC2 iKO prevented HFD-induced downregulation of parkin. During stimulation for mitophagy, mitochondria-localized parkin was severely reduced in control HFD-fed mouse heart, which was restored, in part, in ACC2 iKO HFD-fed mice. CONCLUSIONS These data show that increasing cardiac FAO alone does not cause cardiac dysfunction, but protects against cardiomyopathy in chronically obese mice. The beneficial effect of enhancing cardiac FAO in HFD-induced obesity is mediated, in part, by the maintenance of mitochondria function through regulating parkin-mediated mitophagy. Our findings also suggest that targeting the parkin-dependent mitophagy pathway could be an effective strategy against the development of obesity-induced cardiomyopathy.
Collapse
Affiliation(s)
- Dan Shao
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine (D.S., S.C.K., P.W., N.D.R., O.V., K.N., Y.-W.A.H., A.C., W.W., R.T.), University of Washington, Seattle
| | - Stephen C Kolwicz
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine (D.S., S.C.K., P.W., N.D.R., O.V., K.N., Y.-W.A.H., A.C., W.W., R.T.), University of Washington, Seattle
| | - Pei Wang
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine (D.S., S.C.K., P.W., N.D.R., O.V., K.N., Y.-W.A.H., A.C., W.W., R.T.), University of Washington, Seattle
| | - Nathan D Roe
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine (D.S., S.C.K., P.W., N.D.R., O.V., K.N., Y.-W.A.H., A.C., W.W., R.T.), University of Washington, Seattle
| | - Outi Villet
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine (D.S., S.C.K., P.W., N.D.R., O.V., K.N., Y.-W.A.H., A.C., W.W., R.T.), University of Washington, Seattle
| | - Kiyoto Nishi
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine (D.S., S.C.K., P.W., N.D.R., O.V., K.N., Y.-W.A.H., A.C., W.W., R.T.), University of Washington, Seattle
| | - Yun-Wei A Hsu
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine (D.S., S.C.K., P.W., N.D.R., O.V., K.N., Y.-W.A.H., A.C., W.W., R.T.), University of Washington, Seattle
| | - Galina V Flint
- Department of Bioengineering (G.V.F., M.R.), University of Washington, Seattle
| | - Arianne Caudal
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine (D.S., S.C.K., P.W., N.D.R., O.V., K.N., Y.-W.A.H., A.C., W.W., R.T.), University of Washington, Seattle
| | - Wang Wang
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine (D.S., S.C.K., P.W., N.D.R., O.V., K.N., Y.-W.A.H., A.C., W.W., R.T.), University of Washington, Seattle
| | - Michael Regnier
- Department of Bioengineering (G.V.F., M.R.), University of Washington, Seattle
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine (D.S., S.C.K., P.W., N.D.R., O.V., K.N., Y.-W.A.H., A.C., W.W., R.T.), University of Washington, Seattle
| |
Collapse
|
11
|
Panagia M, He H, Baka T, Pimentel DR, Croteau D, Bachschmid MM, Balschi JA, Colucci WS, Luptak I. Increasing mitochondrial ATP synthesis with butyrate normalizes ADP and contractile function in metabolic heart disease. NMR IN BIOMEDICINE 2020; 33:e4258. [PMID: 32066202 PMCID: PMC7165026 DOI: 10.1002/nbm.4258] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 12/17/2019] [Accepted: 12/22/2019] [Indexed: 06/10/2023]
Abstract
Metabolic heart disease (MHD), which is strongly associated with heart failure with preserved ejection fraction, is characterized by reduced mitochondrial energy production and contractile performance. In this study, we tested the hypothesis that an acute increase in ATP synthesis, via short chain fatty acid (butyrate) perfusion, restores contractile function in MHD. Isolated hearts of mice with MHD due to consumption of a high fat high sucrose (HFHS) diet or on a control diet (CD) for 4 months were studied using 31 P NMR spectroscopy to measure high energy phosphates and ATP synthesis rates during increased work demand. At baseline, HFHS hearts had increased ADP and decreased free energy of ATP hydrolysis (ΔG~ATP ), although contractile function was similar between the two groups. At high work demand, the ATP synthesis rate in HFHS hearts was reduced by over 50%. Unlike CD hearts, HFHS hearts did not increase contractile function at high work demand, indicating a lack of contractile reserve. However, acutely supplementing HFHS hearts with 4mM butyrate normalized ATP synthesis, ADP, ΔG~ATP and contractile reserve. Thus, acute reversal of depressed mitochondrial ATP production improves contractile dysfunction in MHD. These findings suggest that energy starvation may be a reversible cause of myocardial dysfunction in MHD, and opens new therapeutic opportunities.
Collapse
Affiliation(s)
- Marcello Panagia
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA
| | - Huamei He
- Physiological NMR Core Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Tomas Baka
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - David R. Pimentel
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA
| | - Dominique Croteau
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA
| | | | - James A. Balschi
- Physiological NMR Core Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Wilson S. Colucci
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA
| | - Ivan Luptak
- Myocardial Biology Unit, Boston University School of Medicine, Boston, MA
| |
Collapse
|
12
|
Magadum A, Singh N, Kurian AA, Munir I, Mehmood T, Brown K, Sharkar MTK, Chepurko E, Sassi Y, Oh JG, Lee P, Santos CXC, Gaziel-Sovran A, Zhang G, Cai CL, Kho C, Mayr M, Shah AM, Hajjar RJ, Zangi L. Pkm2 Regulates Cardiomyocyte Cell Cycle and Promotes Cardiac Regeneration. Circulation 2020; 141:1249-1265. [PMID: 32078387 PMCID: PMC7241614 DOI: 10.1161/circulationaha.119.043067] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The adult mammalian heart has limited regenerative capacity, mostly attributable to postnatal cardiomyocyte cell cycle arrest. In the last 2 decades, numerous studies have explored cardiomyocyte cell cycle regulatory mechanisms to enhance myocardial regeneration after myocardial infarction. Pkm2 (Pyruvate kinase muscle isoenzyme 2) is an isoenzyme of the glycolytic enzyme pyruvate kinase. The role of Pkm2 in cardiomyocyte proliferation, heart development, and cardiac regeneration is unknown. METHODS We investigated the effect of Pkm2 in cardiomyocytes through models of loss (cardiomyocyte-specific Pkm2 deletion during cardiac development) or gain using cardiomyocyte-specific Pkm2 modified mRNA to evaluate Pkm2 function and regenerative affects after acute or chronic myocardial infarction in mice. RESULTS Here, we identify Pkm2 as an important regulator of the cardiomyocyte cell cycle. We show that Pkm2 is expressed in cardiomyocytes during development and immediately after birth but not during adulthood. Loss of function studies show that cardiomyocyte-specific Pkm2 deletion during cardiac development resulted in significantly reduced cardiomyocyte cell cycle, cardiomyocyte numbers, and myocardial size. In addition, using cardiomyocyte-specific Pkm2 modified RNA, our novel cardiomyocyte-targeted strategy, after acute or chronic myocardial infarction, resulted in increased cardiomyocyte cell division, enhanced cardiac function, and improved long-term survival. We mechanistically show that Pkm2 regulates the cardiomyocyte cell cycle and reduces oxidative stress damage through anabolic pathways and β-catenin. CONCLUSIONS We demonstrate that Pkm2 is an important intrinsic regulator of the cardiomyocyte cell cycle and oxidative stress, and highlight its therapeutic potential using cardiomyocyte-specific Pkm2 modified RNA as a gene delivery platform.
Collapse
Affiliation(s)
- Ajit Magadum
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Neha Singh
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ann Anu Kurian
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Irsa Munir
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Talha Mehmood
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kemar Brown
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mohammad Tofael Kabir Sharkar
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Elena Chepurko
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yassine Sassi
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jae Gyun Oh
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Philyoung Lee
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Celio XC Santos
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine & Sciences, London SE5 9NU, UK
| | - Avital Gaziel-Sovran
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Guoan Zhang
- Proteomics and Metabolomics Core Facility, Weill Cornell Medicine, New York, NY 10021, USA
| | - Chen-Leng Cai
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Changwon Kho
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Manuel Mayr
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine & Sciences, London SE5 9NU, UK
| | - Ajay M. Shah
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine & Sciences, London SE5 9NU, UK
| | - Roger J. Hajjar
- Phospholamban Foundation, Amsterdam,1775 ZH Middenmeer, Netherlands
| | - Lior Zangi
- Cardiovascular Research Center, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
13
|
Borlaug BA, Malloy CR. Energetic Adaptations and Stress Reserve in the Obese Heart. Circulation 2020; 141:1164-1167. [PMID: 32250704 DOI: 10.1161/circulationaha.120.045259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN (B.A.B.)
| | - Craig R Malloy
- Department of Internal Medicine and Advanced Imaging Research Center, University of Texas Southwestern, Dallas, TX (C.R.M.)
| |
Collapse
|
14
|
The creatine kinase system as a therapeutic target for myocardial ischaemia-reperfusion injury. Biochem Soc Trans 2018; 46:1119-1127. [PMID: 30242115 PMCID: PMC6195634 DOI: 10.1042/bst20170504] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/18/2018] [Accepted: 08/22/2018] [Indexed: 12/13/2022]
Abstract
Restoring blood flow following an acute myocardial infarction saves lives, but results in tissue damage due to ischaemia–reperfusion injury (I/R). Ameliorating this damage is a major research goal to improve recovery and reduce subsequent morbidity due to heart failure. Both the ischaemic and reperfusion phases represent crises of cellular energy provision in which the mitochondria play a central role. This mini-review will explore the rationale and therapeutic potential of augmenting the creatine kinase (CK) energy shuttle, which constitutes the primary short-term energy buffer and transport system in the cardiomyocyte. Proof-of-principle data from several transgenic mouse models have demonstrated robust cardioprotection by either raising myocardial creatine levels or by overexpressing specific CK isoforms. The effect on cardiac function, high-energy phosphates and myocardial injury will be discussed and possible directions for future research highlighted. We conclude that the CK system represents a viable target for therapeutic intervention in I/R injury; however, much needed translational studies will require the development of new pharmacological tools.
Collapse
|
15
|
Nabeebaccus AA, Zoccarato A, Hafstad AD, Santos CX, Aasum E, Brewer AC, Zhang M, Beretta M, Yin X, West JA, Schröder K, Griffin JL, Eykyn TR, Abel ED, Mayr M, Shah AM. Nox4 reprograms cardiac substrate metabolism via protein O-GlcNAcylation to enhance stress adaptation. JCI Insight 2017; 2:96184. [PMID: 29263294 PMCID: PMC5752273 DOI: 10.1172/jci.insight.96184] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 11/16/2017] [Indexed: 12/21/2022] Open
Abstract
Cardiac hypertrophic remodeling during chronic hemodynamic stress is associated with a switch in preferred energy substrate from fatty acids to glucose, usually considered to be energetically favorable. The mechanistic interrelationship between altered energy metabolism, remodeling, and function remains unclear. The ROS-generating NADPH oxidase-4 (Nox4) is upregulated in the overloaded heart, where it ameliorates adverse remodeling. Here, we show that Nox4 redirects glucose metabolism away from oxidation but increases fatty acid oxidation, thereby maintaining cardiac energetics during acute or chronic stresses. The changes in glucose and fatty acid metabolism are interlinked via a Nox4-ATF4–dependent increase in the hexosamine biosynthetic pathway, which mediates the attachment of O-linked N-acetylglucosamine (O-GlcNAcylation) to the fatty acid transporter CD36 and enhances fatty acid utilization. These data uncover a potentially novel redox pathway that regulates protein O-GlcNAcylation and reprograms cardiac substrate metabolism to favorably modify adaptation to chronic stress. Our results also suggest that increased fatty acid oxidation in the chronically stressed heart may be beneficial. Nox4 reprograms intermediary metabolism in the heart through an ATF4-mediated enhancement of protein O-GlcNAcylation, and the resulting switch to increased fatty acid oxidation protects the overloaded heart.
Collapse
Affiliation(s)
- Adam A Nabeebaccus
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Anna Zoccarato
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Anne D Hafstad
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom.,Cardiovascular Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Celio Xc Santos
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Ellen Aasum
- Cardiovascular Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Alison C Brewer
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Min Zhang
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Matteo Beretta
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Xiaoke Yin
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - James A West
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, Frankfurt am Main, Germany
| | - Julian L Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
| | - Thomas R Eykyn
- Division of Imaging Sciences & Biomedical Engineering, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - E Dale Abel
- Department of Medicine and Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Manuel Mayr
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Ajay M Shah
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| |
Collapse
|
16
|
Abdurrachim D, Prompers JJ. Evaluation of cardiac energetics by non-invasive 31P magnetic resonance spectroscopy. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1939-1948. [PMID: 29175056 DOI: 10.1016/j.bbadis.2017.11.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/17/2017] [Accepted: 11/18/2017] [Indexed: 01/10/2023]
Abstract
Alterations in myocardial energy metabolism have been implicated in the pathophysiology of cardiac diseases such as heart failure and diabetic cardiomyopathy. 31P magnetic resonance spectroscopy (MRS) is a powerful tool to investigate cardiac energetics non-invasively in vivo, by detecting phosphorus (31P)-containing metabolites involved in energy supply and buffering. In this article, we review the historical development of cardiac 31P MRS, the readouts used to assess cardiac energetics from 31P MRS, and how 31P MRS studies have contributed to the understanding of cardiac energy metabolism in heart failure and diabetes. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.
Collapse
Affiliation(s)
- Desiree Abdurrachim
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Functional Metabolism Group, Singapore Bioimaging Consortium, Agency for Science, Technology and Research, Singapore
| | - Jeanine J Prompers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
17
|
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: 65] [Impact Index Per Article: 8.1] [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
|
18
|
Abstract
Cardiac and skeletal striated muscles are intricately designed machines responsible for muscle contraction. Coordination of the basic contractile unit, the sarcomere, and the complex cytoskeletal networks are critical for contractile activity. The sarcomere is comprised of precisely organized individual filament systems that include thin (actin), thick (myosin), titin, and nebulin. Connecting the sarcomere to other organelles (e.g., mitochondria and nucleus) and serving as the scaffold to maintain cellular integrity are the intermediate filaments. The costamere, on the other hand, tethers the sarcomere to the cell membrane. Unique structures like the intercalated disc in cardiac muscle and the myotendinous junction in skeletal muscle help synchronize and transmit force. Intense investigation has been done on many of the proteins that make up these cytoskeletal assemblies. Yet the details of their function and how they interconnect have just started to be elucidated. A vast number of human myopathies are contributed to mutations in muscle proteins; thus understanding their basic function provides a mechanistic understanding of muscle disorders. In this review, we highlight the components of striated muscle with respect to their interactions, signaling pathways, functions, and connections to disease. © 2017 American Physiological Society. Compr Physiol 7:891-944, 2017.
Collapse
Affiliation(s)
- Christine A Henderson
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Christopher G Gomez
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Stefanie M Novak
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Lei Mi-Mi
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
19
|
Jang A, Xiong Q, Zhang P, Zhang J. Transmurally differentiated measurement of ATP hydrolysis rates in the in vivo porcine hearts. Magn Reson Med 2016; 75:1859-66. [PMID: 26892710 DOI: 10.1002/mrm.26162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/24/2015] [Accepted: 01/20/2016] [Indexed: 11/11/2022]
Abstract
PURPOSE Compare the transmural distribution of forward creatine kinase reaction (kf,CK ) and ATP hydrolysis rate (kr,ATPase ) in the myocardium of normal porcine heart. Rate constants were extracted from partially relaxed spectra by applying the T1nom method, effectively reducing data acquisition time by up to an order of magnitude. THEORY AND METHODS T1nom method for double saturation of PCr and Pi is introduced and validated through simulations. Bioenergetics was measured in vivo utilizing one-dimensional chemical shift imaging (1D-CSI) magnetic resonance (31) P spectroscopy. RESULTS At basal conditions, there was no significant difference between subepicardial layers (EPI) vs. the subendocardial layers (ENDO) for both fluxf,CK and fluxr,ATPase . At high cardiac workload (HWL), where the rate pressure product increased 2.6-fold, PCr/ATP ratio and fluxf,CK showed no significant change in both EPI and ENDO layers, while fluxr,ATPase increased significantly (baseline: 1.11 ± 0.12 and 1.12 ± 0.13 μmol/g/s, EPI and ENDO, respectively; to HWL: 2.35 ± 0.27 and 2.21 ± 0.08 μmol/g/s, EPI and ENDO, respectively, each P < 0.01 vs. baseline). CONCLUSION In the normal heart, increase of cardiac work state is accompanied by an increase in ATP hydrolysis rate with no changes in CK flux rate. There are no significant differences between EPI vs. ENDO concerning the ATP hydrolysis rate or CK flux rate in both baseline and high cardiac work states.
Collapse
Affiliation(s)
- Albert Jang
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota, USA.,Center for Magnetic Resonance Research and Department of Radiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Qiang Xiong
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, Minnesota, USA
| | - Pengyuan Zhang
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jianyi Zhang
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Biomedical Engineering, School of Medicine, School of Engineering, UAB
- The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
20
|
Bakermans AJ, Abdurrachim D, van Nierop BJ, Koeman A, van der Kroon I, Baartscheer A, Schumacher CA, Strijkers GJ, Houten SM, Zuurbier CJ, Nicolay K, Prompers JJ. In vivo mouse myocardial (31)P MRS using three-dimensional image-selected in vivo spectroscopy (3D ISIS): technical considerations and biochemical validations. NMR IN BIOMEDICINE 2015; 28:1218-1227. [PMID: 26269430 PMCID: PMC4573916 DOI: 10.1002/nbm.3371] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 06/24/2015] [Accepted: 07/09/2015] [Indexed: 05/31/2023]
Abstract
(31)P MRS provides a unique non-invasive window into myocardial energy homeostasis. Mouse models of cardiac disease are widely used in preclinical studies, but the application of (31)P MRS in the in vivo mouse heart has been limited. The small-sized, fast-beating mouse heart imposes challenges regarding localized signal acquisition devoid of contamination with signal originating from surrounding tissues. Here, we report the implementation and validation of three-dimensional image-selected in vivo spectroscopy (3D ISIS) for localized (31)P MRS of the in vivo mouse heart at 9.4 T. Cardiac (31)P MR spectra were acquired in vivo in healthy mice (n = 9) and in transverse aortic constricted (TAC) mice (n = 8) using respiratory-gated, cardiac-triggered 3D ISIS. Localization and potential signal contamination were assessed with (31)P MRS experiments in the anterior myocardial wall, liver, skeletal muscle and blood. For healthy hearts, results were validated against ex vivo biochemical assays. Effects of isoflurane anesthesia were assessed by measuring in vivo hemodynamics and blood gases. The myocardial energy status, assessed via the phosphocreatine (PCr) to adenosine 5'-triphosphate (ATP) ratio, was approximately 25% lower in TAC mice compared with controls (0.76 ± 0.13 versus 1.00 ± 0.15; P < 0.01). Localization with one-dimensional (1D) ISIS resulted in two-fold higher PCr/ATP ratios than measured with 3D ISIS, because of the high PCr levels of chest skeletal muscle that contaminate the 1D ISIS measurements. Ex vivo determinations of the myocardial PCr/ATP ratio (0.94 ± 0.24; n = 8) confirmed the in vivo observations in control mice. Heart rate (497 ± 76 beats/min), mean arterial pressure (90 ± 3.3 mmHg) and blood oxygen saturation (96.2 ± 0.6%) during the experimental conditions of in vivo (31)P MRS were within the normal physiological range. Our results show that respiratory-gated, cardiac-triggered 3D ISIS allows for non-invasive assessments of in vivo mouse myocardial energy homeostasis with (31)P MRS under physiological conditions.
Collapse
Affiliation(s)
- Adrianus J. Bakermans
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Desiree Abdurrachim
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Bastiaan J. van Nierop
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Anneke Koeman
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Inge van der Kroon
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Antonius Baartscheer
- Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Cees A. Schumacher
- Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Gustav J. Strijkers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sander M. Houten
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, and Department of Pediatrics, Emma Children’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Coert J. Zuurbier
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Klaas Nicolay
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Jeanine J. Prompers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
21
|
Schweinberger BM, Turcatel E, Rodrigues AF, Wyse ATS. Gestational hypermethioninaemia alters oxidative/nitrative status in skeletal muscle and biomarkers of muscular injury and inflammation in serum of rat offspring. Int J Exp Pathol 2015; 96:277-84. [PMID: 26303039 PMCID: PMC4693554 DOI: 10.1111/iep.12136] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/31/2015] [Indexed: 12/25/2022] Open
Abstract
In this study we evaluated oxidative/nitrative stress parameters (reactive oxygen species production, lipid peroxidation, sulfhydryl content, superoxide dismutase, catalase and nitrite levels), as well as total protein content in the gastrocnemius skeletal muscle of the offspring of rats that had been subjected to gestational hypermethioninaemia. The occurrence of muscular injury and inflammation was also measured by creatine kinase activity, levels of creatinine, urea and C-reactive protein and the presence of cardiac troponin I in serum. Wistar female rats (70-90 days of age) received methionine (2.68 μmol/g body weight) or saline (control) twice a day by subcutaneous injections during the gestational period (21 days). After the rats gave birth, pups were killed at the twenty-first day of life for removal of muscle and serum. Methionine treatment increased reactive oxygen species production and lipid peroxidation and decreased sulfhydryl content, antioxidant enzymes activities and nitrite levels, as well as total protein content in skeletal muscle of the offspring. Creatine kinase activity was reduced and urea and C-reactive protein levels were increased in serum of pups. These results were accompanied by reduced muscle mass. Our findings showed that maternal gestational hypermethioninaemia induced changes in oxidative/nitrative status in gastrocnemius skeletal muscle of the offspring. This may represent a mechanism which can contribute to the myopathies and loss of muscular mass that is found in some hypermethioninaemic patients. In addition, we believe that these results may be relevant as gestational hypermethioninaemia could cause damage to the skeletal muscle during intrauterine life.
Collapse
Affiliation(s)
- Bruna M. Schweinberger
- Laboratório de Neuroproteção e Doenças NeurometabólicasUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Programa de Pós‐Graduação em Ciências Biológicas – Bioquímica. Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Elias Turcatel
- Laboratório de Neuroproteção e Doenças NeurometabólicasUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Programa de Pós‐Graduação em Ciências Biológicas – Bioquímica. Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - André F. Rodrigues
- Laboratório de Neuroproteção e Doenças NeurometabólicasUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Programa de Pós‐Graduação em Ciências Biológicas – Bioquímica. Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Angela T. S. Wyse
- Laboratório de Neuroproteção e Doenças NeurometabólicasUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Programa de Pós‐Graduação em Ciências Biológicas – Bioquímica. Departamento de BioquímicaInstituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
| |
Collapse
|
22
|
Fowler ED, Benoist D, Drinkhill MJ, Stones R, Helmes M, Wüst RCI, Stienen GJM, Steele DS, White E. Decreased creatine kinase is linked to diastolic dysfunction in rats with right heart failure induced by pulmonary artery hypertension. J Mol Cell Cardiol 2015; 86:1-8. [PMID: 26116865 PMCID: PMC4564291 DOI: 10.1016/j.yjmcc.2015.06.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 06/19/2015] [Accepted: 06/22/2015] [Indexed: 02/01/2023]
Abstract
Our objective was to investigate the role of creatine kinase in the contractile dysfunction of right ventricular failure caused by pulmonary artery hypertension. Pulmonary artery hypertension and right ventricular failure were induced in rats by monocrotaline and compared to saline-injected control animals. In vivo right ventricular diastolic pressure-volume relationships were measured in anesthetized animals; diastolic force-length relationships in single enzymatically dissociated myocytes and myocardial creatine kinase levels by Western blot. We observed diastolic dysfunction in right ventricular failure indicated by significantly steeper diastolic pressure-volume relationships in vivo and diastolic force-length relationships in single myocytes. There was a significant reduction in creatine kinase protein expression in failing right ventricle. Dysfunction also manifested as a shorter diastolic sarcomere length in failing myocytes. This was associated with a Ca(2+)-independent mechanism that was sensitive to cross-bridge cycling inhibition. In saponin-skinned failing myocytes, addition of exogenous creatine kinase significantly lengthened sarcomeres, while in intact healthy myocytes, inhibition of creatine kinase significantly shortened sarcomeres. Creatine kinase inhibition also changed the relatively flat contraction amplitude-stimulation frequency relationship of healthy myocytes into a steeply negative, failing phenotype. Decreased creatine kinase expression leads to diastolic dysfunction. We propose that this is via local reduction in ATP:ADP ratio and thus to Ca(2+)-independent force production and diastolic sarcomere shortening. Creatine kinase inhibition also mimics a definitive characteristic of heart failure, the inability to respond to increased demand. Novel therapies for pulmonary artery hypertension are needed. Our data suggest that cardiac energetics would be a potential ventricular therapeutic target.
Collapse
Affiliation(s)
- Ewan D Fowler
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, UK
| | - David Benoist
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, UK; L'Institut de Rythmologie et Modélisation Cardiaque, Inserm U-1045, Université de Bordeaux, France
| | - Mark J Drinkhill
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, UK
| | - Rachel Stones
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, UK
| | - Michiel Helmes
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Centre, Amsterdam, The Netherlands; IonOptix LLC, Milton, MA, USA
| | - Rob C I Wüst
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Centre, Amsterdam, The Netherlands
| | - Ger J M Stienen
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Centre, Amsterdam, The Netherlands; Department of Physics and Astronomy, Faculty of Science, VU University, Amsterdam, The Netherlands
| | - Derek S Steele
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, UK
| | - Ed White
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, UK.
| |
Collapse
|
23
|
Guzun R, Kaambre T, Bagur R, Grichine A, Usson Y, Varikmaa M, Anmann T, Tepp K, Timohhina N, Shevchuk I, Chekulayev V, Boucher F, Dos Santos P, Schlattner U, Wallimann T, Kuznetsov AV, Dzeja P, Aliev M, Saks V. Modular organization of cardiac energy metabolism: energy conversion, transfer and feedback regulation. Acta Physiol (Oxf) 2015; 213:84-106. [PMID: 24666671 DOI: 10.1111/apha.12287] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/23/2013] [Accepted: 03/16/2014] [Indexed: 12/19/2022]
Abstract
To meet high cellular demands, the energy metabolism of cardiac muscles is organized by precise and coordinated functioning of intracellular energetic units (ICEUs). ICEUs represent structural and functional modules integrating multiple fluxes at sites of ATP generation in mitochondria and ATP utilization by myofibrillar, sarcoplasmic reticulum and sarcolemma ion-pump ATPases. The role of ICEUs is to enhance the efficiency of vectorial intracellular energy transfer and fine tuning of oxidative ATP synthesis maintaining stable metabolite levels to adjust to intracellular energy needs through the dynamic system of compartmentalized phosphoryl transfer networks. One of the key elements in regulation of energy flux distribution and feedback communication is the selective permeability of mitochondrial outer membrane (MOM) which represents a bottleneck in adenine nucleotide and other energy metabolite transfer and microcompartmentalization. Based on the experimental and theoretical (mathematical modelling) arguments, we describe regulation of mitochondrial ATP synthesis within ICEUs allowing heart workload to be linearly correlated with oxygen consumption ensuring conditions of metabolic stability, signal communication and synchronization. Particular attention was paid to the structure-function relationship in the development of ICEU, and the role of mitochondria interaction with cytoskeletal proteins, like tubulin, in the regulation of MOM permeability in response to energy metabolic signals providing regulation of mitochondrial respiration. Emphasis was given to the importance of creatine metabolism for the cardiac energy homoeostasis.
Collapse
Affiliation(s)
- R. Guzun
- Laboratory of Fundamental and Applied Bioenergetics; INSERM U1055; Joseph Fourier University; Grenoble France
- Department of Rehabilitation and Physiology; University Hospital; Grenoble France
| | - T. Kaambre
- Laboratory of Bioenergetics; National Institute of Chemical Physics and Biophysics; Tallinn Estonia
| | - R. Bagur
- Laboratory of Fundamental and Applied Bioenergetics; INSERM U1055; Joseph Fourier University; Grenoble France
- Experimental, Theoretical and Applied Cardio-Respiratory Physiology; Laboratory TIMC-IMAG; UMR5525; Joseph Fourier University; Grenoble France
| | - A. Grichine
- Life Science Imaging - In Vitro Platform; IAB CRI INSERM U823; Joseph Fourier University; Grenoble France
| | - Y. Usson
- Experimental, Theoretical and Applied Cardio-Respiratory Physiology; Laboratory TIMC-IMAG; UMR5525; Joseph Fourier University; Grenoble France
| | - M. Varikmaa
- Laboratory of Bioenergetics; National Institute of Chemical Physics and Biophysics; Tallinn Estonia
| | - T. Anmann
- Laboratory of Bioenergetics; National Institute of Chemical Physics and Biophysics; Tallinn Estonia
| | - K. Tepp
- Laboratory of Bioenergetics; National Institute of Chemical Physics and Biophysics; Tallinn Estonia
| | - N. Timohhina
- Laboratory of Bioenergetics; National Institute of Chemical Physics and Biophysics; Tallinn Estonia
| | - I. Shevchuk
- Laboratory of Bioenergetics; National Institute of Chemical Physics and Biophysics; Tallinn Estonia
| | - V. Chekulayev
- Laboratory of Bioenergetics; National Institute of Chemical Physics and Biophysics; Tallinn Estonia
| | - F. Boucher
- Experimental, Theoretical and Applied Cardio-Respiratory Physiology; Laboratory TIMC-IMAG; UMR5525; Joseph Fourier University; Grenoble France
| | - P. Dos Santos
- University of Bordeaux Segalen; INSERM U1045; Bordeaux France
| | - U. Schlattner
- Laboratory of Fundamental and Applied Bioenergetics; INSERM U1055; Joseph Fourier University; Grenoble France
| | - T. Wallimann
- Emeritus; Biology Department; ETH; Zurich Switzerland
| | - A. V. Kuznetsov
- Cardiac Surgery Research Laboratory; Department of Heart Surgery; Innsbruck Medical University; Innsbruck Austria
| | - P. Dzeja
- Division of Cardiovascular Diseases; Department of Medicine; Mayo Clinic; Rochester MN USA
| | - M. Aliev
- Institute of Experimental Cardiology; Cardiology Research Center; Moscow Russia
| | - V. Saks
- Laboratory of Fundamental and Applied Bioenergetics; INSERM U1055; Joseph Fourier University; Grenoble France
| |
Collapse
|
24
|
Bottomley PA, Panjrath GS, Lai S, Hirsch GA, Wu K, Najjar SS, Steinberg A, Gerstenblith G, Weiss RG. Metabolic rates of ATP transfer through creatine kinase (CK Flux) predict clinical heart failure events and death. Sci Transl Med 2013; 5:215re3. [PMID: 24337482 PMCID: PMC4440545 DOI: 10.1126/scitranslmed.3007328] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Morbidity and mortality from heart failure (HF) are high, and current risk stratification approaches for predicting HF progression are imperfect. Adenosine triphosphate (ATP) is required for normal cardiac contraction, and abnormalities in creatine kinase (CK) energy metabolism, the primary myocardial energy reserve reaction, have been observed in experimental and clinical HF. However, the prognostic value of abnormalities in ATP production rates through CK in human HF has not been investigated. Fifty-eight HF patients with nonischemic cardiomyopathy underwent ³¹P magnetic resonance spectroscopy (MRS) to quantify cardiac high-energy phosphates and the rate of ATP synthesis through CK (CK flux) and were prospectively followed for a median of 4.7 years. Multiple-event analysis (MEA) was performed for HF-related events including all-cause and cardiac death, HF hospitalization, cardiac transplantation, and ventricular-assist device placement. Among baseline demographic, clinical, and metabolic parameters, MEA identified four independent predictors of HF events: New York Heart Association (NYHA) class, left ventricular ejection fraction (LVEF), African-American race, and CK flux. Reduced myocardial CK flux was a significant predictor of HF outcomes, even after correction for NYHA class, LVEF, and race. For each increase in CK flux of 1 μmol g⁻¹ s⁻¹, risk of HF-related composite outcomes decreased by 32 to 39%. These findings suggest that reduced CK flux may be a potential HF treatment target. Newer imaging strategies, including noninvasive ³¹P MRS that detect altered ATP kinetics, could thus complement risk stratification in HF and add value in conditions involving other tissues with high energy demands, including skeletal muscle and brain.
Collapse
Affiliation(s)
- Paul A. Bottomley
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Division of Magnetic Resonance Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Gurusher S. Panjrath
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Shenghan Lai
- Division of Magnetic Resonance Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Glenn A. Hirsch
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Katherine Wu
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Samer S. Najjar
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- MedStar Health Research Institute, MedStar Washington Hospital Center, Washington, DC 20010, USA
| | - Angela Steinberg
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Gary Gerstenblith
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Division of Magnetic Resonance Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Robert G. Weiss
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Division of Magnetic Resonance Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
25
|
Affiliation(s)
- Heinrich Taegtmeyer
- The University of Texas Medical School at Houston, Department of Internal Medicine, Division of Cardiology, Houston, Texas
| | - Joanne S. Ingwall
- Professor of Medicine (Physiology), Emerita, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
26
|
Akki A, Gupta A, Weiss RG. Magnetic resonance imaging and spectroscopy of the murine cardiovascular system. Am J Physiol Heart Circ Physiol 2013; 304:H633-48. [PMID: 23292717 DOI: 10.1152/ajpheart.00771.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Magnetic resonance imaging (MRI) has emerged as a powerful and reliable tool to noninvasively study the cardiovascular system in clinical practice. Because transgenic mouse models have assumed a critical role in cardiovascular research, technological advances in MRI have been extended to mice over the last decade. These have provided critical insights into cardiac and vascular morphology, function, and physiology/pathophysiology in many murine models of heart disease. Furthermore, magnetic resonance spectroscopy (MRS) has allowed the nondestructive study of myocardial metabolism in both isolated hearts and in intact mice. This article reviews the current techniques and important pathophysiological insights from the application of MRI/MRS technology to murine models of cardiovascular disease.
Collapse
Affiliation(s)
- Ashwin Akki
- Division of Cardiology, Department of Medicine, and Division of Magnetic Resonance Research, Department of Radiology, The Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
27
|
Turer AT. Using metabolomics to assess myocardial metabolism and energetics in heart failure. J Mol Cell Cardiol 2012; 55:12-8. [PMID: 22982115 DOI: 10.1016/j.yjmcc.2012.08.025] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 08/28/2012] [Accepted: 08/29/2012] [Indexed: 12/22/2022]
Abstract
There is a long history of investigation into the metabolism of the failing heart. Congestive heart failure is marked both by severe disruptions in myocardial energy supply and an inability of the heart to efficiently uptake and oxidize fuels. Despite the many advancements in our understanding, there are still even more outstanding questions in the field. Metabolomics has the power to assist our understanding of the metabolic derangements which accompany myocardial dysfunction. Metabolomic investigations in animal models of heart failure have already highlighted several novel, potentially important pathways of substrate selection and toxicity. Metabolomic biomarker studies in humans, already successfully applied to other forms of cardiovascular disease, have the potential to improve diagnosis and patient care. This article is part of a Special Issue entitled "Focus on Cardiac Metabolism".
Collapse
Affiliation(s)
- Aslan T Turer
- Department of Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-8521, USA.
| |
Collapse
|
28
|
He H, Hoyer K, Tao H, Rice R, Jimenez J, Tardiff JC, Ingwall JS. Myosin-driven rescue of contractile reserve and energetics in mouse hearts bearing familial hypertrophic cardiomyopathy-associated mutant troponin T is mutation-specific. J Physiol 2012; 590:5371-88. [PMID: 22907055 DOI: 10.1113/jphysiol.2012.234252] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The thin filament protein troponin T (TnT) is a regulator of sarcomere function. Whole heart energetics and contractile reserve are compromised in transgenic mice bearing missense mutations at R92 within the tropomyosin-binding domain of cTnT, despite being distal to the ATP hydrolysis domain of myosin. These mutations are associated with familial hypertrophic cardiomyopathy (FHC). Here we test the hypothesis that genetically replacing murine αα-MyHC with murine ββ-MyHC in hearts bearing the R92Q cTnT mutation, a particularly lethal FHC-associated mutation, leads to sufficiently large perturbations in sarcomere function to rescue whole heart energetics and decrease the cost of contraction. By comparing R92Q cTnT and R92L cTnT mutant hearts, we also test whether any rescue is mutation-specific. We defined the energetic state of the isolated perfused heart using (31)P-NMR spectroscopy while simultaneously measuring contractile performance at four work states. We found that the cost of increasing contraction in intact mouse hearts with R92Q cTnT depends on the type of myosin present in the thick filament. We also found that the salutary effect of this manoeuvre is mutation-specific, demonstrating the major regulatory role of cTnT on sarcomere function at the whole heart level.
Collapse
Affiliation(s)
- Huamei He
- NMR Laboratory for Physiological Chemistry, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Lygate CA, Medway DJ, Ostrowski PJ, Aksentijevic D, Sebag-Montefiore L, Hunyor I, Zervou S, Schneider JE, Neubauer S. Chronic creatine kinase deficiency eventually leads to congestive heart failure, but severity is dependent on genetic background, gender and age. Basic Res Cardiol 2012; 107:276. [PMID: 22760499 PMCID: PMC3442167 DOI: 10.1007/s00395-012-0276-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 06/02/2012] [Accepted: 06/13/2012] [Indexed: 11/22/2022]
Abstract
The creatine kinase (CK) energy transport and buffering system supports cardiac function at times of high demand and is impaired in the failing heart. Mice deficient in muscle- and mitochondrial-CK (M/Mt-CK−/−) have previously been described, but exhibit an unexpectedly mild phenotype of compensated left ventricular (LV) hypertrophy. We hypothesised that heart failure would develop with age and performed echocardiography and LV haemodynamics at 1 year. Since all previous studies have utilised mice with a mixed genetic background, we backcrossed for >10 generations on to C57BL/6, and repeated the in vivo investigations. Male M/Mt-CK−/− mice on the mixed genetic background developed congestive heart failure as evidenced by significantly elevated end-diastolic pressure, impaired contractility, LV dilatation, hypertrophy and pulmonary congestion. Female mice were less severely affected, only showing trends for these parameters. After backcrossing, M/Mt-CK−/− mice had LV dysfunction consisting of impaired isovolumetric pressure changes and reduced contractile reserve, but did not develop congestive heart failure. Body weight was lower in knockout mice as a consequence of reduced total body fat. LV weight was not significantly elevated in relation to other internal organs and gene expression of LVH markers was normal, suggesting an absence of hypertrophy. In conclusion, the consequences of CK deficiency are highly dependent on genetic modifiers, gender and age. However, the observation that a primary defect in CK can, under the right conditions, result in heart failure suggests that impaired CK activity in the failing heart could contribute to disease progression.
Collapse
Affiliation(s)
- Craig A Lygate
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Nemutlu E, Zhang S, Gupta A, Juranic NO, Macura SI, Terzic A, Jahangir A, Dzeja P. Dynamic phosphometabolomic profiling of human tissues and transgenic models by 18O-assisted ³¹P NMR and mass spectrometry. Physiol Genomics 2012; 44:386-402. [PMID: 22234996 DOI: 10.1152/physiolgenomics.00152.2011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Next-generation screening of disease-related metabolomic phenotypes requires monitoring of both metabolite levels and turnover rates. Stable isotope (18)O-assisted (31)P nuclear magnetic resonance (NMR) and mass spectrometry uniquely allows simultaneous measurement of phosphometabolite levels and turnover rates in tissue and blood samples. The (18)O labeling procedure is based on the incorporation of one (18)O into P(i) from [(18)O]H(2)O with each act of ATP hydrolysis and the distribution of (18)O-labeled phosphoryls among phosphate-carrying molecules. This enables simultaneous recording of ATP synthesis and utilization, phosphotransfer fluxes through adenylate kinase, creatine kinase, and glycolytic pathways, as well as mitochondrial substrate shuttle, urea and Krebs cycle activity, glycogen turnover, and intracellular energetic communication. Application of expanded (18)O-labeling procedures has revealed significant differences in the dynamics of G-6-P[(18)O] (glycolysis), G-3-P[(18)O] (substrate shuttle), and G-1-P[(18)O] (glycogenolysis) between human and rat atrial myocardium. In human atria, the turnover of G-3-P[(18)O], which defects are associated with the sudden death syndrome, was significantly higher indicating a greater importance of substrate shuttling to mitochondria. Phosphometabolomic profiling of transgenic hearts deficient in adenylate kinase (AK1-/-), which altered levels and mutations are associated to human diseases, revealed a stress-induced shift in metabolomic profile with increased CrP[(18)O] and decreased G-1-P[(18)O] metabolic dynamics. The metabolomic profile of creatine kinase M-CK/ScCKmit-/--deficient hearts is characterized by a higher G-6-[(18)O]P turnover rate, G-6-P levels, glycolytic capacity, γ/β-phosphoryl of GTP[(18)O] turnover, as well as β-[(18)O]ATP and β-[(18)O]ADP turnover, indicating altered glycolytic, guanine nucleotide, and adenylate kinase metabolic flux. Thus, (18)O-assisted gas chromatography-mass spectrometry and (31)P NMR provide a suitable platform for dynamic phosphometabolomic profiling of the cellular energetic system enabling prediction and diagnosis of metabolic diseases states.
Collapse
Affiliation(s)
- Emirhan Nemutlu
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Gupta A, Akki A, Wang Y, Leppo MK, Chacko VP, Foster DB, Caceres V, Shi S, Kirk JA, Su J, Lai S, Paolocci N, Steenbergen C, Gerstenblith G, Weiss RG. Creatine kinase-mediated improvement of function in failing mouse hearts provides causal evidence the failing heart is energy starved. J Clin Invest 2011; 122:291-302. [PMID: 22201686 DOI: 10.1172/jci57426] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 11/02/2011] [Indexed: 01/06/2023] Open
Abstract
ATP is required for normal cardiac contractile function, and it has long been hypothesized that reduced energy delivery contributes to the contractile dysfunction of heart failure (HF). Despite experimental and clinical HF data showing reduced metabolism through cardiac creatine kinase (CK), the major myocardial energy reserve and temporal ATP buffer, a causal relationship between reduced ATP-CK metabolism and contractile dysfunction in HF has never been demonstrated. Here, we generated mice conditionally overexpressing the myofibrillar isoform of CK (CK-M) to test the hypothesis that augmenting impaired CK-related energy metabolism improves contractile function in HF. CK-M overexpression significantly increased ATP flux through CK ex vivo and in vivo but did not alter contractile function in normal mice. It also led to significantly increased contractile function at baseline and during adrenergic stimulation and increased survival after thoracic aortic constriction (TAC) surgery-induced HF. Withdrawal of CK-M overexpression after TAC resulted in a significant decline in contractile function as compared with animals in which CK-M overexpression was maintained. These observations provide direct evidence that the failing heart is "energy starved" as it relates to CK. In addition, these data identify CK as a promising therapeutic target for preventing and treating HF and possibly diseases involving energy-dependent dysfunction in other organs with temporally varying energy demands.
Collapse
Affiliation(s)
- Ashish Gupta
- Department of Medicine, Cardiology Division, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Isolated adult mouse cardiomyocytes are an important tool in cardiovascular research, but are challenging to prepare. Because the energy supply determines cell function and viability, we compared total creatine ([Cr]) and [ATP] in isolated cardiomyocytes with the intact mouse heart. Isolated myocytes suffered severe losses of Cr (-70%) and ATP (-53%). Myocytes were not able to replete [Cr] during a 5 h incubation period in medium supplemented with 1 mM Cr. In contrast, adding 20 mM Cr to the digestion buffers was sufficient to maintain normal [Cr]. Supplementing buffers with 5 mM of inosine (Ino) and adenosine (Ado) to prevent loss of cellular nucleosides partially protected against loss of ATP. To test whether maintaining [ATP] and [Cr] improves contractile function, myocytes were challenged by varying pacing rate from 0.5 to 10 Hz and by adding isoproterenol (Iso) at 5 and 10 Hz. All groups performed well up to 5 Hz, showing a positive cell shortening-frequency relationship; however, only 16% of myocytes isolated under standard conditions were able to sustain pacing with Iso challenge at 10 Hz. In contrast, 30-50% of the myocytes with normal Cr levels were able to contract and maintain low diastolic [Ca(2+)]. Cell yield also improved in Cr and the Cr/Ino/Ado-treated groups (85-90% vs. 70-75% rod shaped in untreated myocytes). These data suggest that viability and performance of isolated myocytes are improved when they are protected from the severe loss of Cr and ATP during the isolation, making them an even better research tool.
Collapse
Affiliation(s)
- Ilka Pinz
- NMR Laboratory for Physiological Chemistry, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | | | | | | |
Collapse
|
33
|
Dzeja PP, Hoyer K, Tian R, Zhang S, Nemutlu E, Spindler M, Ingwall JS. Rearrangement of energetic and substrate utilization networks compensate for chronic myocardial creatine kinase deficiency. J Physiol 2011; 589:5193-211. [PMID: 21878522 DOI: 10.1113/jphysiol.2011.212829] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Plasticity of the cellular bioenergetic system is fundamental to every organ function, stress adaptation and disease tolerance. Here, remodelling of phosphotransfer and substrate utilization networks in response to chronic creatine kinase (CK) deficiency, a hallmark of cardiovascular disease, has been revealed in transgenic mouse models lacking either cytosolic M-CK (M-CK(-/-)) or both M-CK and sarcomeric mitochondrial CK (M-CK/ScCKmit(-/-)) isoforms. The dynamic metabolomic signatures of these adaptations have also been defined. Tracking perturbations in metabolic dynamics with (18)O and (13)C isotopes and (31)P NMR and mass spectrometry demonstrate that hearts lacking M-CK have lower phosphocreatine (PCr) turnover but increased glucose-6-phosphate (G-6-P) turnover, glucose utilization and inorganic phosphate compartmentation with normal ATP γ-phosphoryl dynamics. Hearts lacking both M-CK and sarcomeric mitochondrial CK have diminished PCr turnover, total phosphotransfer capacity and intracellular energetic communication but increased dynamics of β-phosphoryls of ADP/ATP, G-6-P and γ-/β-phosphoryls of GTP, indicating redistribution of flux through adenylate kinase (AK), glycolytic and guanine nucleotide phosphotransfer circuits. Higher glycolytic and mitochondrial capacities and increased glucose tolerance contributed to metabolic resilience of M-CK/ScCKmit(-/-) mice. Multivariate analysis revealed unique metabolomic signatures for M-CK(-/-) and M-CK/ScCKmit(-/-) hearts suggesting that rearrangements in phosphotransfer and substrate utilization networks provide compensation for genetic CK deficiency. This new information highlights the significance of integrated CK-, AK-, guanine nucleotide- and glycolytic enzyme-catalysed phosphotransfer networks in supporting the adaptivity and robustness of the cellular energetic system.
Collapse
Affiliation(s)
- Petras P Dzeja
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
The most notable change in the metabolic profile of hypertrophied hearts is an increased reliance on glucose with an overall reduced oxidative metabolism, i.e. a reappearance of the foetal metabolic pattern. In animal models, this change is attributed to the down-regulation of the transcriptional cascades promoting gene expression for fatty acid oxidation and mitochondrial oxidative phosphorylation in adult hearts. Impaired myocardial energetics in cardiac hypertrophy also triggers AMP-activated protein kinase (AMPK), leading to increased glucose uptake and glycolysis. Aside from increased reliance on glucose as an energy source, changes in other glucose metabolism pathways, e.g. the pentose phosphate pathway, the glucosamine biosynthesis pathway, and anaplerosis, are also noted in the hypertrophied hearts. Studies using transgenic mouse models and pharmacological compounds to mimic or counter the switch of substrate preference in cardiac hypertrophy have demonstrated that increased glucose metabolism in adult heart is not harmful and can be beneficial when it provides sufficient fuel for oxidative metabolism. However, improvement in the oxidative capacity and efficiency rather than the selection of the substrate is likely the ultimate goal for metabolic therapies.
Collapse
Affiliation(s)
- Stephen C Kolwicz
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, 815 Mercer Street, Seattle, WA 98109, USA
| | | |
Collapse
|
35
|
Hettling H, van Beek JHGM. Analyzing the functional properties of the creatine kinase system with multiscale 'sloppy' modeling. PLoS Comput Biol 2011; 7:e1002130. [PMID: 21912519 PMCID: PMC3166207 DOI: 10.1371/journal.pcbi.1002130] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 06/08/2011] [Indexed: 11/18/2022] Open
Abstract
In this study the function of the two isoforms of creatine kinase (CK; EC 2.7.3.2) in myocardium is investigated. The 'phosphocreatine shuttle' hypothesis states that mitochondrial and cytosolic CK plays a pivotal role in the transport of high-energy phosphate (HEP) groups from mitochondria to myofibrils in contracting muscle. Temporal buffering of changes in ATP and ADP is another potential role of CK. With a mathematical model, we analyzed energy transport and damping of high peaks of ATP hydrolysis during the cardiac cycle. The analysis was based on multiscale data measured at the level of isolated enzymes, isolated mitochondria and on dynamic response times of oxidative phosphorylation measured at the whole heart level. Using 'sloppy modeling' ensemble simulations, we derived confidence intervals for predictions of the contributions by phosphocreatine (PCr) and ATP to the transfer of HEP from mitochondria to sites of ATP hydrolysis. Our calculations indicate that only 15±8% (mean±SD) of transcytosolic energy transport is carried by PCr, contradicting the PCr shuttle hypothesis. We also predicted temporal buffering capabilities of the CK isoforms protecting against high peaks of ATP hydrolysis (3750 µM*s(-1)) in myofibrils. CK inhibition by 98% in silico leads to an increase in amplitude of mitochondrial ATP synthesis pulsation from 215±23 to 566±31 µM*s(-1), while amplitudes of oscillations in cytosolic ADP concentration double from 77±11 to 146±1 µM. Our findings indicate that CK acts as a large bandwidth high-capacity temporal energy buffer maintaining cellular ATP homeostasis and reducing oscillations in mitochondrial metabolism. However, the contribution of CK to the transport of high-energy phosphate groups appears limited. Mitochondrial CK activity lowers cytosolic inorganic phosphate levels while cytosolic CK has the opposite effect.
Collapse
Affiliation(s)
- Hannes Hettling
- Centre for Integrative Bioinformatics VU, VU University Amsterdam, Amsterdam, The Netherlands.
| | | |
Collapse
|
36
|
Diguet N, Mallat Y, Ladouce R, Clodic G, Prola A, Tritsch E, Blanc J, Larcher JC, Delcayre C, Samuel JL, Friguet B, Bolbach G, Li Z, Mericskay M. Muscle creatine kinase deficiency triggers both actin depolymerization and desmin disorganization by advanced glycation end products in dilated cardiomyopathy. J Biol Chem 2011; 286:35007-19. [PMID: 21768101 DOI: 10.1074/jbc.m111.252395] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Alterations in the balance of cytoskeleton as well as energetic proteins are involved in the cardiac remodeling occurring in dilated cardiomyopathy (DCM). We used two-dimensional DIGE proteomics as a discovery approach to identify key molecular changes taking place in a temporally controlled model of DCM triggered by cardiomyocyte-specific serum response factor (SRF) knock-out in mice. We identified muscle creatine kinase (MCK) as the primary down-regulated protein followed by α-actin and α-tropomyosin down-regulation leading to a decrease of polymerized F-actin. The early response to these defects was an increase in the amount of desmin intermediate filaments and phosphorylation of the αB-crystallin chaperone. We found that αB-crystallin and desmin progressively lose their striated pattern and accumulate at the intercalated disk and the sarcolemma, respectively. We further show that desmin is a preferential target of advanced glycation end products (AGE) in mouse and human DCM. Inhibition of CK in cultured cardiomyocytes is sufficient to recapitulate both the actin depolymerization defect and the modification of desmin by AGE. Treatment with either cytochalasin D or glyoxal, a cellular AGE, indicated that both actin depolymerization and AGE contribute to desmin disorganization. Heat shock-induced phosphorylation of αB-crystallin provides a transient protection of desmin against glyoxal in a p38 MAPK-dependent manner. Our results show that the strong down-regulation of MCK activity contributes to F-actin instability and induces post-translational modification of αB-crystallin and desmin. Our results suggest that AGE may play an important role in DCM because they alter the organization of desmin filaments that normally support stress response and mitochondrial functions in cardiomyocytes.
Collapse
Affiliation(s)
- Nicolas Diguet
- Department of Aging, UPMC University Paris 6, 75005 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Li HH, Du J, Fan YN, Zhang ML, Liu DP, Li L, Lockyer P, Kang EY, Patterson C, Willis MS. The ubiquitin ligase MuRF1 protects against cardiac ischemia/reperfusion injury by its proteasome-dependent degradation of phospho-c-Jun. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1043-58. [PMID: 21356357 DOI: 10.1016/j.ajpath.2010.11.049] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Revised: 10/19/2010] [Accepted: 11/16/2010] [Indexed: 12/11/2022]
Abstract
Despite improvements in interventions of acute coronary syndromes, primary reperfusion therapies restoring blood flow to ischemic myocardium leads to the activation of signaling cascades that induce cardiomyocyte cell death. These signaling cascades, including the mitogen-activated protein kinase signaling pathways, activate cardiomyocyte death in response to both ischemia and reperfusion. We have previously identified muscle ring finger-1 (MuRF1) as a cardiac-specific protein that regulates cardiomyocyte mass through its ubiquitin ligase activity, acting to degrade sarcomeric proteins and inhibit transcription factors involved in cardiac hypertrophy signaling. To determine MuRF1's role in cardiac ischemia/reperfusion (I/R) injury, cardiomyocytes in culture and intact hearts were challenged with I/R injury in the presence and absence of MuRF1. We found that MuRF1 is cardioprotective, in part, by its ability to prevent cell death by inhibiting Jun N-terminal kinase (JNK) signaling. MuRF1 specifically targets JNK's proximal downstream target, activated phospho-c-Jun, for degradation by the proteasome, effectively inhibiting downstream signaling and the induction of cell death. MuRF1's inhibitory affects on JNK signaling through its ubiquitin proteasome-dependent degradation of activated c-Jun is the first description of a cardiac ubiquitin ligase inhibiting mitogen-activated protein kinase signaling. MuRF1's cardioprotection in I/R injury is attenuated in the presence of pharmacologic JNK inhibition in vivo, suggesting a prominent role of MuRF1's regulation of c-Jun in the intact heart.
Collapse
Affiliation(s)
- Hui-Hua Li
- Department of Pathology, School of Basic Medical Sciences, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Xiong Q, Du F, Zhu X, Zhang P, Suntharalingam P, Ippolito J, Kamdar FD, Chen W, Zhang J. ATP production rate via creatine kinase or ATP synthase in vivo: a novel superfast magnetization saturation transfer method. Circ Res 2011; 108:653-63. [PMID: 21293002 PMCID: PMC3090083 DOI: 10.1161/circresaha.110.231456] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 01/21/2010] [Indexed: 11/16/2022]
Abstract
RATIONALE ³¹P magnetization saturation transfer (MST) experiment is the most widely used method to study ATP metabolism kinetics. However, its lengthy data acquisition time greatly limits the wide biomedical applications in vivo, especially for studies requiring high spatial and temporal resolutions. OBJECTIVE We aimed to develop a novel superfast MST method that can accurately quantify ATP production rate constants (k(f)) through creatine kinase (CK) or ATP synthase (ATPase) with 2 spectra. METHODS AND RESULTS The T₁(nom) (T₁ nominal) method uses a correction factor to compensate the partially relaxed MST experiments, thus allowing measurement of enzyme kinetics with an arbitrary repetition time and flip angle, which consequently reduces the data acquisition time of a transmurally differentiated CK k(f) measurement by 91% as compared with the conventional method with spatial localization. The novel T₁(nom) method is validated theoretically with numeric simulation, and further verified with in vivo swine hearts, as well as CK and ATPase activities in rat brain at 9.4 Tesla. Importantly, the in vivo data from swine hearts demonstrate, for the first time, that within an observation window of 30 minutes, the inhibition of CK activity by iodoacetamide does not limit left ventricular chamber contractile function. CONCLUSIONS A novel MST method for superfast examination of enzyme kinetics in vivo has been developed and verified theoretically and experimentally. In the in vivo normal heart, redundant multiple supporting systems of myocardial ATP production, transportation, and utilization exist, such that inhibition of one mechanism does not impair the normal left ventricular contractile performance.
Collapse
Affiliation(s)
- Qiang Xiong
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
- Center for Magnetic Resonance Research, Department of Radiology, Harvard Medical School, Belmont, Massachusetts
| | - Fei Du
- Center for Magnetic Resonance Research, Department of Radiology, Harvard Medical School, Belmont, Massachusetts
- McLean Hospital, Department of Psychiatry, Harvard Medical School, Belmont, Massachusetts
| | - Xiaohong Zhu
- Center for Magnetic Resonance Research, Department of Radiology, Harvard Medical School, Belmont, Massachusetts
| | - Pengyuan Zhang
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Piradeep Suntharalingam
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Joseph Ippolito
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
- Center for Magnetic Resonance Research, Department of Radiology, Harvard Medical School, Belmont, Massachusetts
| | - Forum D. Kamdar
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Wei Chen
- Center for Magnetic Resonance Research, Department of Radiology, Harvard Medical School, Belmont, Massachusetts
| | - Jianyi Zhang
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
- Center for Magnetic Resonance Research, Department of Radiology, Harvard Medical School, Belmont, Massachusetts
| |
Collapse
|
39
|
Gupta A, Chacko VP, Schär M, Akki A, Weiss RG. Impaired ATP kinetics in failing in vivo mouse heart. Circ Cardiovasc Imaging 2011; 4:42-50. [PMID: 20926788 PMCID: PMC3066090 DOI: 10.1161/circimaging.110.959320] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 09/28/2010] [Indexed: 11/16/2022]
Abstract
BACKGROUND The hypothesis that the failing heart may be energy-starved is supported in part by observations of reduced rates of adenosine 5'-triphosphate (ATP) synthesis through the creatine kinase (CK) reaction, the primary myocardial energy reservoir, in patients with heart failure (HF). Although murine models have been used to probe HF pathophysiology, it has not been possible to noninvasively measure the rate of ATP synthesis through CK in the in vivo mouse heart. The purpose of this work was to exploit noninvasive spatially localized magnetic resonance spectroscopy techniques to measure ATP flux through CK in in vivo mouse hearts and determine the extent of any reductions in murine HF. METHODS AND RESULTS The Triple Repetition Time Saturation Transfer (TRiST) magnetic resonance spectroscopy method of measuring ATP kinetics was first validated in skeletal muscle, rendering similar results to conventional saturation transfer magnetic resonance spectroscopy. In normal mouse hearts, the in vivo CK pseudo-first-order-rate constant, k(F), was 0.32±0.03 s(-1) (mean±SD) and the rate of ATP synthesis through CK was 3.16±0.47 μmol/g/s. Thoracic aortic constriction reduced k(F) by 31% (0.23±0.03 s(-1), P<0.0001) and ATP synthesis through CK by 51% (1.54±0.25 μmol/g/s, P<0.0001), values analogous to those in failing human hearts. CONCLUSIONS Despite the small size and high murine heart rate, the ATP synthesis rate through CK is similar in vivo in murine and human hearts and comparably reduced in HF. Because murine thoracic aortic constriction shares fundamental energetic similarities with human HF, this model and new magnetic resonance spectroscopy approach promise a powerful means to noninvasively probe altered energetics in HF.
Collapse
Affiliation(s)
- Ashish Gupta
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
40
|
Aksentijević D, Lygate CA, Makinen K, Zervou S, Sebag-Montefiore L, Medway D, Barnes H, Schneider JE, Neubauer S. High-energy phosphotransfer in the failing mouse heart: role of adenylate kinase and glycolytic enzymes. Eur J Heart Fail 2010; 12:1282-9. [PMID: 20940173 PMCID: PMC2990411 DOI: 10.1093/eurjhf/hfq174] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 07/14/2010] [Accepted: 07/30/2010] [Indexed: 11/13/2022] Open
Abstract
AIMS To measure the activity of the key phosphotransfer enzymes creatine kinase (CK), adenylate kinase (AK), and glycolytic enzymes in two common mouse models of chronic heart failure. METHODS AND RESULTS C57BL/6 mice were subjected to transverse aortic constriction (TAC), myocardial infarction induced by coronary artery ligation (CAL), or sham operation. Activities of phosphotransfer enzymes CK, AK, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), 3-phosphoglycerate kinase (PGK), and pyruvate kinase were assessed spectrophotometrically. Mice were characterized by echocardiography or magnetic resonance imaging 5- to 8-week post-surgery and selected for the presence of congestive heart failure. All mice had severe left ventricular hypertrophy, impaired systolic function and pulmonary congestion compared with sham controls. A significant decrease in myocardial CK and maximal CK reaction velocity was observed in both experimental models of heart failure. However, the activity of AK and its isoforms remained unchanged, despite a reduction in its protein expression. In contrast, the activities of glycolytic phosphotransfer mediators GAPDH and PGK were 19 and 12% higher in TAC, and 31 and 23% higher in CAL models, respectively. CONCLUSION Chronic heart failure in the mouse is characterized by impaired CK function, unaltered AK, and increased activity of glycolytic phosphotransfer enzymes. This pattern of altered phosphotransfer activity was observed independent of the heart failure aetiology.
Collapse
Affiliation(s)
- Dunja Aksentijević
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Craig A. Lygate
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Kimmo Makinen
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Sevasti Zervou
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Liam Sebag-Montefiore
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Debra Medway
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Hannah Barnes
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
- British Heart Foundation Experimental Magnetic Resonance Unit, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Jurgen E. Schneider
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
- British Heart Foundation Experimental Magnetic Resonance Unit, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Stefan Neubauer
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
41
|
Efficiency of cross-bridges and mitochondria in mouse cardiac muscle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010. [PMID: 20824531 DOI: 10.1007/978-1-4419-6366-6_15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2025]
Abstract
The aim of this study was to make cellular-level measurements of the mechanical efficiency of mouse cardiac muscle and to use these measurements to determine (1) the work performed by a cross-bridge in one ATP-splitting cycle and (2) the fraction of the free energy available in metabolic substrates that is transferred by oxidative phosphorylation to free energy in ATP (i.e. mitochondrial thermodynamic efficiency). Experiments were performed using isolated left ventricular mouse papillary muscles (n = 9; studied at 27°C) and the myothermic technique. The production of work and heat was measured during and after 40 contractions at a contraction frequency of 2 Hz. Each contraction consisted of a brief isometric period followed by isovelocity shortening. Work output, heat output and enthalpy output were all independent of shortening velocity. Maximum initial mechanical efficiency (mean ± SEM) was 31.1 ± 1.3% and maximum net mechanical efficiency 16.9 ± 1.5%. It was calculated that the maximum work per cross-bridge cycle was 20 zJ, comparable to values for mouse skeletal muscle, and that mitochondrial thermodynamic efficiency was 72%. Analysis of data in the literature suggests that mitochondrial efficiency of cardiac muscle from other species is also likely to be between 70 and 80%.
Collapse
|
42
|
Gao J, Shen M, Guo X, Li X, Li J. Proteomic Mechanism of Myocardial Angiogenesis Augmented by Remote Ischemic Training of Skeletal Muscle in Rabbit. Cardiovasc Ther 2010; 29:199-210. [DOI: 10.1111/j.1755-5922.2009.00097.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
43
|
Cui Y, Piao CS, Ha KC, Kim DS, Lee GH, Kim HK, Chae SW, Lee YC, Park SJ, Yoo WH, Kim HR, Chae HJ. Measuring adriamycin-induced cardiac hemodynamic dysfunction with a proteomics approach. Immunopharmacol Immunotoxicol 2010; 32:376-86. [DOI: 10.3109/08923970903440168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
44
|
Rice R, Guinto P, Dowell-Martino C, He H, Hoyer K, Krenz M, Robbins J, Ingwall JS, Tardiff JC. Cardiac myosin heavy chain isoform exchange alters the phenotype of cTnT-related cardiomyopathies in mouse hearts. J Mol Cell Cardiol 2009; 48:979-88. [PMID: 20004663 DOI: 10.1016/j.yjmcc.2009.11.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2009] [Revised: 11/12/2009] [Accepted: 11/28/2009] [Indexed: 11/28/2022]
Abstract
Familial hypertrophic cardiomyopathy, FHC, is a clinically heterogeneous, autosomal-dominant disease of the cardiac sarcomere leading to extensive remodeling at both the whole heart and molecular levels. The remodeling patterns are mutation-specific, a finding that extends to the level of single amino acid substitutions at the same peptide residue. Here we utilize two well-characterized transgenic FHC mouse models carrying independent amino acid substitutions in the TM-binding region of cardiac troponin T (cTnT) at residue 92. R92Q and R92L cTnT domains have mutation-specific average peptide conformation and dynamics sufficient to alter thin filament flexibility and cross-bridge formation and R92 mutant myocytes demonstrate mutation-specific temporal molecular remodeling of Ca(2+) kinetics and impaired cardiac contractility and relaxation. To determine if a greater economy of contraction at the crossbridge level would rescue the mechanical defects caused by the R92 cTnT mutations, we replaced the endogenous murine alpha-myosin heavy chain (MyHC) with the beta-MyHC isoform. While beta-MyHC replacement rescued the systolic dysfunction in R92Q mice, it failed to rescue the defects in diastolic function common to FHC-associated R92 mutations. Surprisingly, a significant component of the whole heart and molecular contractile improvement in the R92Q mice was due to improvements in Ca(2+) homeostasis including SR uptake, [Ca2+](i) amplitude and phospholamban phosphorylation. Our data demonstrate that while genetically altering the myosin composition of the heart bearing a thin filament FHC mutation is sufficient to improve contractility, diastolic performance is refractory despite improved Ca(2+) kinetics. These data reveal a previously unrecognized role for MyHC isoforms with respect to Ca(2+) homeostasis in the setting of cardiomyopathic remodeling and demonstrate the overall dominance of the thin filament mutation in determining the degree of diastolic impairment at the myofilament level.
Collapse
Affiliation(s)
- Ron Rice
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Yeshiva University, 1300 Morris Park Avenue, Ullmann, Room 316, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mizuno M, Takeba Y, Matsumoto N, Tsuzuki Y, Asoh K, Takagi M, Kobayashi S, Yamamoto H. Antenatal glucocorticoid therapy accelerates ATP production with creatine kinase increase in the growth-enhanced fetal rat heart. Circ J 2009; 74:171-80. [PMID: 19952437 DOI: 10.1253/circj.cj-09-0311] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Previous study has demonstrated the increase of several cardiac function-related proteins, including creatine kinase (CK) as an important enzyme in the process of ATP synthesis in the fetal heart of rats administered glucocorticoid (GC) antenatally. In the present study the effect of antenatal GC administration on the CK expression in fetal and neonatal hearts was demonstrated. METHODS AND RESULTS Dexamethasone was administered to pregnant rats on days 19 and 20 of gestation. The mRNA levels of the CK isoforms, CK-M and Mi-CK, in 21-day-old fetal and 1-day-old neonatal hearts were significantly increased after antenatal GC administration. CK protein levels were also increased in both cultured cardiomyocytes and the mitochondria of the hearts. Uptake of 5, 5', 6, 6'-tetrachloro-1, 1', 3, 3'-tetraethyl-benzimidazolocarbocyanine iodide by mitochondria was significantly increased. An increased ATP level accompanied the CK increase in the neonatal hearts. Furthermore, in vitro these effects were mediated though the GC receptor of cardiomyocytes. Peroxisome proliferator-activated receptor gamma as the upstream transcription factor of CK was significantly increased in fetal hearts. CONCLUSIONS These results suggest that antenatal GC administration accelerates ATP synthesis through increased CK and may contribute to maturation of the premature heart so that it is ready for preterm delivery. (Circ J 2010; 74: 171 - 180).
Collapse
Affiliation(s)
- Masanori Mizuno
- Department of Pediatrics, St Marianna University School of Medicine, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
There has been growing interest in targeting myocardial substrate metabolism for the therapy of cardiovascular and metabolic diseases. This is largely based on the observation that cardiac metabolism undergoes significant changes during both physiologic and pathologic stresses. In search for an effective therapeutic strategy, recent studies have focused on the functional significance of the substrate switch in the heart during stress conditions, such as cardiac hypertrophy and failure, using both pharmacologic and genetic approaches. The results of these studies indicate that both the capacity and the flexibility of the cardiac metabolic network are essential for normal function; thus, their maintenance should be the primary goal for future metabolic therapy.
Collapse
Affiliation(s)
- Stephen C Kolwicz
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | | |
Collapse
|
47
|
Bovill E, Westaby S, Crisp A, Jacobs S, Shaw T. Reduction of four-and-a-half LIM-protein 2 expression occurs in human left ventricular failure and leads to altered localization and reduced activity of metabolic enzymes. J Thorac Cardiovasc Surg 2009; 137:853-61. [PMID: 19327508 DOI: 10.1016/j.jtcvs.2008.09.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 08/13/2008] [Accepted: 09/01/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVE We sought to identify changes in four-and-a-half LIM-protein 2 levels and location in human cardiomyocytes during the transition from compensated aortic stenosis to left ventricular failure. We also sought to characterize four-and-a-half LIM-protein 2 binding with the metabolic enzymes phosphofructokinase 2, adenylate kinase, and creatine kinase M isoform during this transition and their consequential subcellular localization in failing human ventricles. METHODS Left ventricular biopsy specimens from selected patients undergoing aortic valve replacement for aortic stenosis were allocated to one of 2 groups: (1) nondilated with preserved left ventricular function (nonfailing group, n = 16) and (2) grossly dilated with poor left ventricular function (failing group, n = 15). These were compared with a control group of unused donor hearts (n = 6). Protein levels and subcellular localization were determined by means of Western blotting and immunofluorescence. Four-and-a-half LIM-protein 2 binding to adenylate kinase, creatine kinase M isoform, or phosphofructokinase 2 was studied by means of coimmunoprecipitation. Phosphofructokinase 2, adenylate kinase, and creatine kinase M isoform activities were assayed in protein extractions. RESULTS Four-and-a-half LIM-protein 2 levels were preserved in nonfailing hypertrophied hearts but reduced by 53% in failing hearts. The pattern of four-and-a-half LIM-protein 2 staining was disrupted in failing hearts: four-and-a-half LIM-protein 2 was lost from the sarcomere but present in the perinuclear Golgi apparatus complex. Phosphofructokinase 2, adenylate kinase, and creatine kinase M isoform coimmunoprecipitated in vitro and colocalized with four-and-a-half LIM-protein 2 in both hypertrophied and failing hearts. Phosphofructokinase 2 and adenylate kinase activities were reduced to 77% and 58% of normal values in compensated aortic stenosis, with phosphofructokinase 2 activity decreased further to 56% of normal value in failing hearts, but creatine kinase activity remained unchanged. CONCLUSIONS Altered four-and-a-half LIM-protein 2 expression in heart failure is associated with disruption of the normal subcellular localization of phosphofructokinase 2, adenylate kinase, and creatine kinase M isoform and reduced activity of phosphofructokinase 2 and adenylate kinase, which might have important consequences for myocardial energy metabolism in heart failure.
Collapse
Affiliation(s)
- Esta Bovill
- Department of Medicine, University College London, London, United Kingdom.
| | | | | | | | | |
Collapse
|
48
|
Gnecchi M, He H, Melo LG, Noiseaux N, Morello F, de Boer RA, Zhang L, Pratt RE, Dzau VJ, Ingwall JS. Early beneficial effects of bone marrow-derived mesenchymal stem cells overexpressing Akt on cardiac metabolism after myocardial infarction. Stem Cells 2009; 27:971-9. [PMID: 19353525 PMCID: PMC2873075 DOI: 10.1002/stem.12] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Administration of mesenchymal stem cells (MSCs) is an effective therapy to repair cardiac damage after myocardial infarction (MI) in experimental models. However, the mechanisms of action still need to be elucidated. Our group has recently suggested that MSCs mediate their therapeutic effects primarily via paracrine cytoprotective action. Furthermore, we have shown that MSCs overexpressing Akt1 (Akt-MSCs) exert even greater cytoprotection than unmodified MSCs. So far, little has been reported on the metabolic characteristics of infarcted hearts treated with stem cells. Here, we hypothesize that Akt-MSC administration may influence the metabolic processes involved in cardiac adaptation and repair after MI. MI was performed in rats randomized in four groups: sham group and animals treated with control MSCs, Akt-MSCs, or phosphate-buffered saline (PBS). High energy metabolism and basal 2-deoxy-glucose (2-DG) uptake were evaluated on isolated hearts using phosphorus-31 nuclear magnetic resonance spectroscopy at 72 hours and 2 weeks after MI. Treatment with Akt-MSCs spared phosphocreatine stores and significantly limited the increase in 2-DG uptake in the residual intact myocardium compared with the PBS- or the MSC-treated animals. Furthermore, Akt-MSC-treated hearts had normal pH, whereas low pH was measured in the PBS and MSC groups. Correlative analysis indicated that functional recovery after MI was inversely related to the rate of 2-DG uptake. We conclude that administration of MSCs overexpressing Akt at the time of infarction results in preservation of normal metabolism and pH in the surviving myocardium.
Collapse
|
49
|
Wu F, Beard DA. Roles of the creatine kinase system and myoglobin in maintaining energetic state in the working heart. BMC SYSTEMS BIOLOGY 2009; 3:22. [PMID: 19228404 PMCID: PMC2667476 DOI: 10.1186/1752-0509-3-22] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Accepted: 02/19/2009] [Indexed: 11/10/2022]
Abstract
BACKGROUND The heart is capable of maintaining contractile function despite a transient decrease in blood flow and increase in cardiac ATP demand during systole. This study analyzes a previously developed model of cardiac energetics and oxygen transport to understand the roles of the creatine kinase system and myoglobin in maintaining the ATP hydrolysis potential during beat-to-beat transient changes in blood flow and ATP hydrolysis rate. RESULTS The theoretical investigation demonstrates that elimination of myoglobin only slightly increases the predicted range of oscillation of cardiac oxygenation level during beat-to-beat transients in blood flow and ATP utilization. In silico elimination of myoglobin has almost no impact on the cytoplasmic ATP hydrolysis potential (DeltaGATPase). In contrast, disabling the creatine kinase system results in considerable oscillations of cytoplasmic ADP and ATP levels and seriously deteriorates the stability of DeltaGATPase in the beating heart. CONCLUSION The CK system stabilizes DeltaGATPase by both buffering ATP and ADP concentrations and enhancing the feedback signal of inorganic phosphate in regulating mitochondrial oxidative phosphorylation.
Collapse
Affiliation(s)
- Fan Wu
- Biotechnology and Bioengineering Center, Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | - Daniel A Beard
- Biotechnology and Bioengineering Center, Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| |
Collapse
|
50
|
Abstract
Myocytes of the failing heart undergo impressive metabolic remodelling. The time line for changes in the pathways for ATP synthesis in compensated hypertrophy is: flux through the creatine kinase (CK) reaction falls as both creatine concentration ([Cr]) and CK activity fall; increases in [ADP] and [AMP] lead to increases in glucose uptake and utilization; fatty acid oxidation either remains the same or decreases. In uncompensated hypertrophy and in other forms of heart failure, CK flux and fatty acid oxidation are both lower; any increases in glucose uptake and utilization are not sufficient to compensate for overall decreases in the capacity for ATP supply and [ATP] falls. Metabolic remodelling is under transcriptional and post-transcriptional control. The lower metabolic reserve of the failing heart contributes to impaired contractile reserve.
Collapse
Affiliation(s)
- Joanne S Ingwall
- NMR Laboratory for Physiological Chemistry, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 221 Longwood Avenue, Room 247, Boston, MA 02115, USA.
| |
Collapse
|