1
|
Iveljic I, Young M, Corhodzic E, Cullen F, Prag HA, Murphy MP, Aksentijevic D. Type 2 diabetes worsens the outcome of ischemia/reperfusion in female STEMI patients and female db/db mice with HFpEF cardiometabolic phenotype. Cardiovasc Diabetol 2025; 24:243. [PMID: 40483445 PMCID: PMC12144705 DOI: 10.1186/s12933-025-02771-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 05/02/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) poses a significant global health challenge, disproportionately affecting women. Diabetic women with HFpEF represent a high-risk subgroup, particularly after experiencing ST-segment elevation myocardial infarction (STEMI), exhibiting increased mortality compared to men. While prolonged door-to-balloon (DTB) times, reflecting delayed reperfusion, are a critical factor in STEMI outcomes, they alone do not fully capture the observed outcome variability in diabetic women. Using an integrated clinical and pre-clinical approach this study aimed to investigate the relative contributions of metabolic dysfunction and coronary artery disease (CAD) in type 2 diabetes (T2D) to STEMI outcomes in women, beyond the impact of DTB time. METHODS A retrospective case-control study analysed female STEMI patients undergoing primary percutaneous coronary intervention (pPCI, n = 40 T2D, n = 40 non-diabetic controls), comparing clinical characteristics, treatment strategies, and early outcomes. A preclinical model (female db/db mice) assessed cardiac function via echocardiography, Langendorff perfusions, and ischemia-reperfusion protocols. Metabolome of heart, liver, and skeletal muscle was assessed by 1H NMR spectroscopy. RESULTS Our study reveals significantly higher mortality, impaired left ventricular function post-pPCI, and increased implantable cardioverter-defibrillator (ICD) implantation rates in diabetic STEMI patients, irrespective of DTB time, when compared to non-diabetic controls. Elevated inflammatory markers, acute hyperglycaemia and evidence of cardio-hepatic damage were identified in T2D patients. db/db mice exhibited analogous T2D-associated pathophysiology, including increased ischemia-reperfusion injury exacerbated by metabolic disturbances in the myocardium, liver, and skeletal muscle versus non-diabetic controls. CONCLUSIONS In diabetic women, multiple factors beyond reperfusion delays exacerbate acute myocardial injury. This necessitates the development of sex-specific strategies to manage the cardiovascular complications of diabetic HFpEF. The db/db mouse model provides a relevant preclinical tool for future research as it mimics human T2D-associated HFpEF and STEMI outcome.
Collapse
Affiliation(s)
- Ivana Iveljic
- Faculty of Medicine, University of Tuzla, Tuzla, Bosnia and Herzegovina
- University Clinical Center Tuzla, Tuzla, Bosnia and Herzegovina
| | - Megan Young
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Elvira Corhodzic
- Faculty of Medicine, University of Tuzla, Tuzla, Bosnia and Herzegovina
- University Clinical Center Tuzla, Tuzla, Bosnia and Herzegovina
| | - Fenn Cullen
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Hiran A Prag
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Dunja Aksentijevic
- William Harvey Research Institute, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
2
|
Huang Y, Li L, Hong Y, Cheng L, Gu Z. The effect of carbohydrates with different levels of digestibility on energy metabolism in vivo under hypobaric hypoxic conditions. Carbohydr Polym 2025; 351:123114. [PMID: 39779022 DOI: 10.1016/j.carbpol.2024.123114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025]
Abstract
Current strategies for improving energy supply in hypobaric hypoxic environments are limited. Therefore, this study investigates the effects of four carbohydrates with different levels of digestibility on energy metabolism in vivo in hypobaric hypoxic environments. First, we characterized the four types of carbohydrates. Subsequently, reverse transcription quantitative polymerase chain reaction (RT-qPCR) was used to characterize the expression of GLUT1, GLUT2, and SGLT1 in the glucose transport pathway in vivo. In addition, the effects of different levels of carbohydrate digestibility on energy expenditure were evaluated in vivo. The results showed that pre-gelatinized corn starch significantly increased GLUT1 gene expression in the hypobaric hypoxic conditions (1.58 times, compared to normobaric normoxic). In addition, pre-gelatinized corn starch increased energy expenditure in the hypobaric hypoxic conditions and performed better in terms of glycogen accumulation and glucose transport. Therefore, pre-gelatinized corn starch administration may be a promising strategy for long-term energy supplementation in hypobaric hypoxic.
Collapse
Affiliation(s)
- Yali Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China
| | - Lingjin Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China
| | - Yan Hong
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China.
| | - Li Cheng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China
| | - Zhengbiao Gu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
3
|
Zervou S, McAndrew DJ, Lake HA, Kuznecova E, Preece C, Davies B, Neubauer S, Lygate CA. Cardiac function and energetics in mice with combined genetic augmentation of creatine and creatine kinase activity. J Mol Cell Cardiol 2024; 196:105-114. [PMID: 39276853 DOI: 10.1016/j.yjmcc.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Improving energy provision in the failing heart by augmenting the creatine kinase (CK) system is a desirable therapeutic target. However, over-expression of the creatine transporter (CrT-OE) has shown that very high creatine levels result in cardiac hypertrophy and dysfunction. We hypothesise this is due to insufficient endogenous CK activity to maintain thermodynamically favourable metabolite ratios. If correct, then double transgenic mice (dTg) overexpressing both CrT and the muscle isoform of CK (CKM-OE) would rescue the adverse phenotype. In Study 1, overexpressing lines were crossed and cardiac function assessed by invasive haemodynamics and echocardiography. This demonstrated that CKM-OE was safe, but too few hearts had creatine in the toxic range. In Study 2, a novel CrT-OE line was generated with higher, homogeneous, creatine levels and phenotyped as before. Myocardial creatine was 4-fold higher in CrT-OE and dTg hearts compared to wildtype and was associated with hypertrophy and contractile dysfunction. The inability of dTg hearts to rescue this phenotype was attributed to downregulation of CK activity, as occurs in the failing heart. Nevertheless, combining both studies in a linear regression analysis suggests a modest positive effect of CKM over a range of creatine concentrations. In conclusion, we confirm that moderate elevation of creatine is well tolerated, but very high levels are detrimental. Correlation analysis lends support to the theory that this may be a consequence of limited CK activity. Future studies should focus on preventing CKM downregulation to unlock the potential synergy of augmenting both creatine and CK in the heart.
Collapse
Affiliation(s)
- Sevasti Zervou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK; Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Debra J McAndrew
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK; Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Hannah A Lake
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK; Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Elina Kuznecova
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK; Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | | | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK; Centre for Human Genetics, University of Oxford, Oxford, UK; School of Cardiovascular and Metabolic Health, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
4
|
Kacmaz M, Schlettert C, Kreimer F, Abumayyaleh M, Akin I, Mügge A, Aweimer A, Hamdani N, El-Battrawy I. Ejection Fraction-Related Differences of Baseline Characteristics and Outcomes in Troponin-Positive Patients without Obstructive Coronary Artery Disease. J Clin Med 2024; 13:2826. [PMID: 38792370 PMCID: PMC11121874 DOI: 10.3390/jcm13102826] [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: 02/16/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Background: The development and course of myocardial infarction with non-obstructive coronary artery (MINOCA) disease is still not fully understood. In this study, we aimed to examine the baseline characteristics of in-hospital outcomes and long-term outcomes of a cohort of troponin-positive patients without obstructive coronary artery disease based on different left ventricular ejection fractions (LVEFs). Methods and results: We included a cohort of 254 patients (mean age: 64 (50.8-75.3) years, 120 females) with suspected myocardial infarction and no obstructive coronary artery disease (MINOCA) in our institutional database between 2010 and 2021. Among these patients, 170 had LVEF ≥ 50% (84 females, 49.4%), 31 patients had LVEF 40-49% (15 females, 48.4%), and 53 patients had LVEF < 40% (20 females, 37.7%). The mean age in the LVEF ≥ 50% group was 61.5 (48-73) years, in the LVEF 40-49% group was 67 (57-78) years, and in the LVEF < 40% group was 68 (56-75.5) years (p = 0.05). The mean troponin value was highest in the LVEF < 40% group, at 3.8 (1.7-4.6) µg/L, and lowest in the LVEF ≥ 50% group, at 1.1 (0.5-2.1) µg/L (p = 0.05). Creatine Phosphokinase (CK) levels were highest in the LVEF ≥ 50% group (156 (89.5-256)) and lowest in the LVEF 40-49% group (127 (73-256)) (p < 0.05), while the mean BNP value was lowest in the LVEF ≥ 50% group (98 (48-278) pg/mL) and highest in the <40% group (793 (238.3-2247.5) pg/mL) (p = 0.001). Adverse in-hospital cardiovascular events were highest in the LVEF < 40% group compared to the LVEF 40-49% group and the LVEF ≥ 50% group (56% vs. 55% vs. 27%; p < 0.001). Over a follow-up period of 6.2 ± 3.1 years, the all-cause mortality was higher in the LVEF < 40% group compared to the LVEF 40-49% group and the LVEF ≥ 50% group. Among the different factors, LVEF < 40% and LVEF 40-49% were associated with an increased risk of in-hospital cardiovascular events in the multivariable Cox regression analysis. Conclusions: LVEF has different impacts on in-hospital cardiovascular events in this cohort. Furthermore, LVEF influences long-term all-cause mortality.
Collapse
Affiliation(s)
- Mustafa Kacmaz
- Institute of Physiology, Department of Cellular and Translational Physiology and Institute für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr-University Bochum, 44791 Bochum, Germany; (M.K.); (A.A.); (N.H.)
- HCEMM-SU Cardiovascular Comorbidities Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Clara Schlettert
- Department of Cardiology and Angiology, Bergmannsheil University Hospital, Ruhr University of Bochum, 44789 Bochum, Germany;
| | - Fabienne Kreimer
- Department of Cardiology and Rhythmology, University Hospital St. Josef Hospital Bochum, Ruhr University Bochum, 44791 Bochum, Germany; (F.K.); (A.M.)
| | - Mohammad Abumayyaleh
- First Department of Medicine, University Medical Centre Mannheim (UMM), 68167 Mannheim, Germany; (M.A.); (I.A.)
| | - Ibrahim Akin
- First Department of Medicine, University Medical Centre Mannheim (UMM), 68167 Mannheim, Germany; (M.A.); (I.A.)
| | - Andreas Mügge
- Department of Cardiology and Rhythmology, University Hospital St. Josef Hospital Bochum, Ruhr University Bochum, 44791 Bochum, Germany; (F.K.); (A.M.)
| | - Assem Aweimer
- Institute of Physiology, Department of Cellular and Translational Physiology and Institute für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr-University Bochum, 44791 Bochum, Germany; (M.K.); (A.A.); (N.H.)
| | - Nazha Hamdani
- Institute of Physiology, Department of Cellular and Translational Physiology and Institute für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr-University Bochum, 44791 Bochum, Germany; (M.K.); (A.A.); (N.H.)
- HCEMM-SU Cardiovascular Comorbidities Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
- Department of Physiology, Cardiovascular Research Institute Maastricht, University Maastricht, 6200 Maastricht, The Netherlands
| | - Ibrahim El-Battrawy
- Institute of Physiology, Department of Cellular and Translational Physiology and Institute für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr-University Bochum, 44791 Bochum, Germany; (M.K.); (A.A.); (N.H.)
- Department of Cardiology and Rhythmology, University Hospital St. Josef Hospital Bochum, Ruhr University Bochum, 44791 Bochum, Germany; (F.K.); (A.M.)
| |
Collapse
|
5
|
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
|
6
|
Lomivorotov V, Merekin D, Fominskiy E, Ponomarev D, Bogachev-Prokophiev A, Zalesov A, Cherniavsky A, Shilova A, Guvakov D, Lomivorotova L, Lembo R, Landoni G. Myocardial protection with phosphocreatine in high-risk cardiac surgery patients: a randomized trial. BMC Anesthesiol 2023; 23:389. [PMID: 38030971 PMCID: PMC10685505 DOI: 10.1186/s12871-023-02341-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND This study was conducted to test the hypothesis that phosphocreatine (PCr), administered intravenously and as cardioplegia adjuvant in patients undergoing cardiac surgery with prolonged aortic cross clamping and cardiopulmonary bypass (CPB) time, would decrease troponin I concentration after surgery. METHODS In this randomized, double-blind, placebo-controlled pilot study we included 120 patients undergoing double/triple valve repair/replacement under cardiopulmonary bypass in the cardiac surgery department of a tertiary hospital. The treatment group received: intravenous administration of 2 g of PCr after anesthesia induction; 2.5 g of PCr in every 1 L of cardioplegic solution (concentration = 10 mmol/L); intravenous administration of 2 g of PCr immediately after heart recovery following aorta declamping; 4 g of PCr at intensive care unit admission. The control group received an equivolume dose of normosaline. RESULTS The primary endpoint was peak concentration of troponin I after surgery. Secondary endpoints included peak concentration of serum creatinine, need for, and dosage of inotropic support, number of defibrillations after aortic declamping, incidence of arrhythmias, duration of Intensive Care Unit (ICU) stay, length of hospitalization. There was no difference in peak troponin I concentration after surgery (PCr, 10,508 pg/ml [IQR 6,838-19,034]; placebo, 11,328 pg/ml [IQR 7.660-22.894]; p = 0.24). There were also no differences in median peak serum creatinine (PCr, 100 µmol/L [IQR 85.0-117.0]; placebo, 99.5 µmol/L [IQR 90.0-117.0]; p = 0.87), the number of patients on vasopressor/inotropic agents (PCr, 49 [88%]; placebo, 57 [91%]; p = 0.60), the inotropic score on postoperative day 1 (PCr, 4.0 (0-7); placebo, 4.0 (0-10); p = 0.47), mean SOFA score on postoperative day 1 (PCr, 5.25 ± 2.33; placebo, 5,45 ± 2,65; p = 0.83), need for defibrillation after declamping of aorta (PCr, 22 [39%]; placebo, 25 [40%]; p = 0.9),, duration of ICU stay and length of hospitalization as well as 30-day mortality (PCr, 0 (0%); placebo,1 (4.3%); p = 0.4). CONCLUSION PCr administration to patients undergoing double/triple valve surgery under cardiopulmonary bypass is safe but is not associated with a decrease in troponin I concentration. Phosphocreatine had no beneficial effect on clinical outcomes after surgery. TRIAL REGISTRATION The study is registered at ClinicalTrials.gov with the Identifier: NCT02757443. First posted (published): 02/05/2016.
Collapse
Affiliation(s)
- Vladimir Lomivorotov
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Anesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Dmitry Merekin
- Department of Anesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Evgeny Fominskiy
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Dmitry Ponomarev
- Department of Anesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | | | - Anton Zalesov
- Department of Heart Valve Surgery, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Alexander Cherniavsky
- Department of Aortic and Coronary Artery Surgery, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Anna Shilova
- Department of Anesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Dmitry Guvakov
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Liudmila Lomivorotova
- Department of Anesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Rosalba Lembo
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
7
|
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
|
8
|
Maguire ML, McAndrew DJ, Lake HA, Ostrowski PJ, Zervou S, Neubauer S, Lygate CA, Schneider JE. Synergistic effect on cardiac energetics by targeting the creatine kinase system: in vivo application of high-resolution 31P-CMRS in the mouse. J Cardiovasc Magn Reson 2023; 25:6. [PMID: 36740688 PMCID: PMC9900916 DOI: 10.1186/s12968-023-00911-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 01/05/2023] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Phosphorus cardiovascular magnetic resonance spectroscopy (31P-CMRS) has emerged as an important tool for the preclinical assessment of myocardial energetics in vivo. However, the high rate and diminutive size of the mouse heart is a challenge, resulting in low resolution and poor signal-to-noise. Here we describe a refined high-resolution 31P-CMRS technique and apply it to a novel double transgenic mouse (dTg) with elevated myocardial creatine and creatine kinase (CK) activity. We hypothesised a synergistic effect to augment energetic status, evidenced by an increase in the ratio of phosphocreatine-to-adenosine-triphosphate (PCr/ATP). METHODS AND RESULTS Single transgenic Creatine Transporter overexpressing (CrT-OE, n = 7) and dTg mice (CrT-OE and CK, n = 6) mice were anaesthetised with isoflurane to acquire 31P-CMRS measurements of the left ventricle (LV) utilising a two-dimensional (2D), threefold under-sampled density-weighted chemical shift imaging (2D-CSI) sequence, which provided high-resolution data with nominal voxel size of 8.5 µl within 70 min. (1H-) cine-CMR data for cardiac function assessment were obtained in the same imaging session. Under a separate examination, mice received invasive haemodynamic assessment, after which tissue was collected for biochemical analysis. Myocardial creatine levels were elevated in all mouse hearts, but only dTg exhibited significantly elevated CK activity, resulting in a 51% higher PCr/ATP ratio in heart (3.01 ± 0.96 vs. 2.04 ± 0.57-mean ± SD; dTg vs. CrT-OE), that was absent from adjacent skeletal muscle. No significant differences were observed for any parameters of LV structure and function, confirming that augmentation of CK activity does not have unforeseen consequences for the heart. CONCLUSIONS We have developed an improved 31P-CMRS methodology for the in vivo assessment of energetics in the murine heart which enabled high-resolution imaging within acceptable scan times. Mice over-expressing both creatine and CK in the heart exhibited a synergistic elevation in PCr/ATP that can now be tested for therapeutic potential in models of chronic heart failure.
Collapse
Affiliation(s)
- Mahon L Maguire
- Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Debra J McAndrew
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Hannah A Lake
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Philip J Ostrowski
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Sevasti Zervou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, UK.
| | - Jurgen E Schneider
- Experimental and Preclinical Imaging Centre (ePIC), Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK.
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW Myocardial metabolism is intricately linked to cardiac function. Perturbations of cardiac energy metabolism result in an energy-starved heart and the development of contractile dysfunction. In this review, we discuss alterations in myocardial energy supply, transcriptional changes in response to different energy demands, and mitochondrial function in the development of heart failure. RECENT FINDINGS Recent studies on substrate modulation through modifying energy substrate supply have shown cardioprotective properties. In addition, large cardiovascular outcome trials of anti-diabetic agents have demonstrated prognostic benefit, suggesting the importance of myocardial metabolism in cardiac function. Understanding molecular and transcriptional controls of cardiac metabolism promises new research avenues for metabolic treatment targets. Future studies assessing the impact of substrate modulation on cardiac energetic status and function will better inform development of metabolic therapies.
Collapse
Affiliation(s)
- Sher May Ng
- Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
| | - Stefan Neubauer
- Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
- Department of Cardiology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Oliver J Rider
- Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK.
- Department of Cardiology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
10
|
McAndrew DJ, Lake HA, Zervou S, Schwedhelm E, Schneider JE, Neubauer S, Lygate CA. Homoarginine and creatine deficiency do not exacerbate murine ischaemic heart failure. ESC Heart Fail 2022; 10:189-199. [PMID: 36178450 PMCID: PMC9871656 DOI: 10.1002/ehf2.14183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/23/2022] [Accepted: 09/15/2022] [Indexed: 01/27/2023] Open
Abstract
AIMS Low levels of homoarginine and creatine are associated with heart failure severity in humans, but it is unclear to what extent they contribute to pathophysiology. Both are synthesized via L-arginine:glycine amidinotransferase (AGAT), such that AGAT-/- mice have a combined creatine and homoarginine deficiency. We hypothesized that this would be detrimental in the setting of chronic heart failure. METHODS AND RESULTS Study 1: homoarginine deficiency-female AGAT-/- and wild-type mice were given creatine-supplemented diet so that both had normal myocardial creatine levels, but only AGAT-/- had low plasma homoarginine. Myocardial infarction (MI) was surgically induced and left ventricular (LV) structure and function assessed at 6-7 weeks by in vivo imaging and haemodynamics. Study 2: homoarginine and creatine-deficiency-as before, but AGAT-/- mice were given creatine-supplemented diet until 1 week post-MI, when 50% were changed to a creatine-free diet. Both groups therefore had low homoarginine levels, but one group also developed lower myocardial creatine levels. In both studies, all groups had LV remodelling and dysfunction commensurate with the development of chronic heart failure, for example, LV dilatation and mean ejection fraction <20%. However, neither homoarginine deficiency alone or in combination with creatine deficiency had a significant effect on mortality, LV remodelling, or on any indices of contractile and lusitropic function. CONCLUSIONS Low levels of homoarginine and creatine do not worsen chronic heart failure arguing against a major causative role in disease progression. This suggests that it is unnecessary to correct hArg deficiency in patients with heart failure, although supra-physiological levels may still be beneficial.
Collapse
Affiliation(s)
- Debra J. McAndrew
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK,British Heart Foundation Centre for Research ExcellenceUniversity of OxfordOxfordUK,Wellcome Centre for Human GeneticsRoosevelt DriveOxfordOX3 7BNUK
| | - Hannah A. Lake
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK,British Heart Foundation Centre for Research ExcellenceUniversity of OxfordOxfordUK,Wellcome Centre for Human GeneticsRoosevelt DriveOxfordOX3 7BNUK
| | - Sevasti Zervou
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK,British Heart Foundation Centre for Research ExcellenceUniversity of OxfordOxfordUK,Wellcome Centre for Human GeneticsRoosevelt DriveOxfordOX3 7BNUK
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and ToxicologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Jurgen E. Schneider
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK,Experimental and Preclinical Imaging Centre (ePIC), Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK,British Heart Foundation Centre for Research ExcellenceUniversity of OxfordOxfordUK
| | - Craig A. Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK,British Heart Foundation Centre for Research ExcellenceUniversity of OxfordOxfordUK,Wellcome Centre for Human GeneticsRoosevelt DriveOxfordOX3 7BNUK
| |
Collapse
|
11
|
Prokic VZ, Rankovic MR, Draginic ND, Andjic MM, Sretenovic JZ, Zivkovic VI, Jeremic JN, Milinkovic MV, Bolevich S, Jakovljevic VLJ, Pantovic SB. Guanidinoacetic acid provides superior cardioprotection to its combined use with betaine and (or) creatine in HIIT-trained rats. Can J Physiol Pharmacol 2022; 100:772-786. [PMID: 35894232 DOI: 10.1139/cjpp-2021-0801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study aimed to determine how guanidinoacetic acid (GAA) or its combined administration with betaine (B) or creatine (C) influences the cardiac function, morphometric parameters, and redox status of rats subjected to high-intensity interval training (HIIT). This research was conducted on male Wistar albino rats exposed to HIIT for 4 weeks. The animals were randomly divided into five groups: HIIT, HIIT + GAA, HIIT + GAA + C, HIIT + GAA + B, and HIIT + GAA + C + B. After completing the training protocol, GAA (300 mg/kg), C (280 mg/kg), and B (300 mg/kg) were applied daily per os for 4 weeks. GAA supplementation in combination with HIIT significantly decreased the level of both systemic and cardiac prooxidants ( O 2 - , H2O2, NO 2 - , and thiobarbituric acid reactive substances) compared with nontreated HIIT (p < 0.05). Also, GAA treatment led to an increase in glutathione and superoxide dismutase levels. None of the treatment regimens altered cardiac function. A larger degree of cardiomyocyte hypertrophy was observed in the HIIT + GAA group, which was reflected through an increase of the cross-sectional area of 27% (p < 0.05) and that of the left ventricle wall thickness of 27% (p < 0.05). Since we showed that GAA in combination with HIIT may ameliorate oxidative stress and does not alter cardiac function, the present study is a basis for future research exploring the mechanisms of cardioprotection induced by this supplement in an HIIT scenario.
Collapse
Affiliation(s)
- Veljko Z Prokic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Marina R Rankovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nevena D Draginic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.,Department of Human Pathology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Marijana M Andjic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Jasmina Z Sretenovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Vladimir I Zivkovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Jovana N Jeremic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Milica V Milinkovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Sergey Bolevich
- Department of Human Pathology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vladimir L J Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.,Department of Human Pathology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Suzana B Pantovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
12
|
Peng JF, Salami OM, Lei C, Ni D, Habimana O, Yi GH. Targeted mitochondrial drugs for treatment of Myocardial ischemia-reperfusion injury. J Drug Target 2022; 30:833-844. [PMID: 35652502 DOI: 10.1080/1061186x.2022.2085728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Myocardial ischemia-reperfusion injury (MI/RI) refers to the further damage done to ischemic cardiomyocytes when restoring blood flow. A large body of evidence shows that MI/RI is closely associated with excessive production of mitochondrial reactive oxygen species, mitochondrial calcium overload, disordered mitochondrial energy metabolism, mitophagy, mitochondrial fission, and mitochondrial fusion. According to the way it affects mitochondria, it can be divided into mitochondrial quality abnormalities and mitochondrial quantity abnormalities. Abnormal mitochondrial quality refers to the dysfunction caused by the severe destruction of mitochondria, which then affects the balance of mitochondrial density and number, causing an abnormal mitochondrial quantity. In the past, most of the reports were limited to the study of the mechanism of myocardial ischemia-reperfusion injury, some of which involved mitochondria, but no specific countermeasures were proposed. In this review, we outline the mechanisms for treating myocardial ischemia-reperfusion injury from the direction of mitochondria and focus on targeted interventions and drugs to restore mitochondrial health during abnormal mitochondrial quality control and abnormal mitochondrial quantity control. This is an update in the field of myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Jin-Fu Peng
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China.,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | | | - Cai Lei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Dan Ni
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Olive Habimana
- International College, University of South China, 28 W Changsheng Road, Hengyang, Hunan 421001, China
| | - Guang-Hui Yi
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China.,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
13
|
Li Q. Metabolic Reprogramming, Gut Dysbiosis, and Nutrition Intervention in Canine Heart Disease. Front Vet Sci 2022; 9:791754. [PMID: 35242837 PMCID: PMC8886228 DOI: 10.3389/fvets.2022.791754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/05/2022] [Indexed: 12/15/2022] Open
Abstract
This review provides a state-of-the-art overview on recent advances in systems biology in canine cardiac disease, with a focus on our current understanding of bioenergetics and amino acid metabolism in myxomatous mitral valve disease (MMVD). Cross-species comparison is drawn to highlight the similarities between human and canine heart diseases. The adult mammalian heart exhibits a remarkable metabolic flexibility and shifts its energy substrate preference according to different physiological and pathological conditions. The failing heart suffers up to 40% ATP deficit and is compared to an engine running out of fuel. Bioenergetics and metabolic readaptations are among the major research topics in cardiac research today. Myocardial energy metabolism consists of three interconnected components: substrate utilization, oxidative phosphorylation, and ATP transport and utilization. Any disruption or uncoupling of these processes can result in deranged energy metabolism leading to heart failure (HF). The review describes the changes occurring in each of the three components of energy metabolism in MMVD and HF. It also provides an overview on the changes in circulating and myocardial glutathione, taurine, carnitines, branched-chain amino acid catabolism and tryptophan metabolic pathways. In addition, the review summarizes the potential role of the gut microbiome in MMVD and HF. As our knowledge and understanding in these molecular and metabolic processes increase, it becomes possible to use nutrition to address these changes and to slow the progression of the common heart diseases in dogs.
Collapse
|
14
|
Supplementing Soy-Based Diet with Creatine in Rats: Implications for Cardiac Cell Signaling and Response to Doxorubicin. Nutrients 2022; 14:nu14030583. [PMID: 35276943 PMCID: PMC8840593 DOI: 10.3390/nu14030583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/16/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
Nutritional habits can have a significant impact on cardiovascular health and disease. This may also apply to cardiotoxicity caused as a frequent side effect of chemotherapeutic drugs, such as doxorubicin (DXR). The aim of this work was to analyze if diet, in particular creatine (Cr) supplementation, can modulate cardiac biochemical (energy status, oxidative damage and antioxidant capacity, DNA integrity, cell signaling) and functional parameters at baseline and upon DXR treatment. Here, male Wistar rats were fed for 4 weeks with either standard rodent diet (NORMAL), soy-based diet (SOY), or Cr-supplemented soy-based diet (SOY + Cr). Hearts were either freeze-clamped in situ or following ex vivo Langendorff perfusion without or with 25 μM DXR and after recording cardiac function. The diets had distinct cardiac effects. Soy-based diet (SOY vs. NORMAL) did not alter cardiac performance but increased phosphorylation of acetyl-CoA carboxylase (ACC), indicating activation of rather pro-catabolic AMP-activated protein kinase (AMPK) signaling, consistent with increased ADP/ATP ratios and lower lipid peroxidation. Creatine addition to the soy-based diet (SOY + Cr vs. SOY) slightly increased left ventricular developed pressure (LVDP) and contractility dp/dt, as measured at baseline in perfused heart, and resulted in activation of the rather pro-anabolic protein kinases Akt and ERK. Challenging perfused heart with DXR, as analyzed across all nutritional regimens, deteriorated most cardiac functional parameters and also altered activation of the AMPK, ERK, and Akt signaling pathways. Despite partial reprogramming of cell signaling and metabolism in the rat heart, diet did not modify the functional response to supraclinical DXR concentrations in the used acute cardiotoxicity model. However, the long-term effect of these diets on cardiac sensitivity to chronic and clinically relevant DXR doses remains to be established.
Collapse
|
15
|
Koch V, Weber C, Riffel JH, Buchner K, Buss SJ, Hein S, Mereles D, Hagenmueller M, Erbel C, März W, Booz C, Albrecht MH, Vogl TJ, Frey N, Hardt SE, Ochs M. Impact of Homoarginine on Myocardial Function and Remodeling in a Rat Model of Chronic Renal Failure. J Cardiovasc Pharmacol Ther 2022; 27:10742484211054620. [PMID: 34994208 DOI: 10.1177/10742484211054620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE Low plasma concentrations of the amino acid homoarginine (HA) have been shown to correlate with adverse cardiovascular outcome, particularly in patients with chronic kidney disease. The present study sought to investigate the effect of HA treatment on cardiac remodeling in rats undergoing artificially induced renal insufficiency by 5/6 nephrectomy (5/6 Nx). METHODS A total of 33 male Wistar rats were randomly divided into sham and 5/6 Nx groups, receiving either placebo treatment or 400 mg·kg-1·day-1 HA over a 4-week period. RESULTS 5/6 Nx per se resulted in adverse myocardial remodeling with aggravated cardiac function and associated cardiac overload as the most obvious alteration (-23% ejection fraction, P < 0.0001), as well as increased myocardial fibrosis (+80%, P = 0.0005) compared to placebo treated sham animals. HA treatment of 5/6 Nx rats has led to an improvement of ejection fraction (+24%, P = 0.0003) and fractional shortening (+21%, P = 0.0126), as well as a decrease of collagen deposition (-32%, P = 0.0041), left ventricular weight (-14%, P = 0.0468), and myocyte cross-sectional area (-12%, P < 0.0001). These changes were accompanied by a downregulation of atrial natriuretic factor (-65% P < 0.0001) and collagen type V alpha 1 chain (-44%, P = 0.0006). Sham animals revealed no significant changes in cardiac function, myocardial fibrosis, or any of the aforementioned molecular changes after drug treatment. CONCLUSION Dietary HA supplementation appears to have the potential of preventing cardiac remodeling and improving heart function in the setting of chronic kidney disease. Our findings shed new light on HA as a possible new therapeutic agent for patients at high cardiovascular risk.
Collapse
Affiliation(s)
- Vitali Koch
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Christophe Weber
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Johannes H Riffel
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Kristina Buchner
- Institute of Human Genetics, Section for Developmental Genetics, 27178University of Heidelberg, Heidelberg, Germany
| | - Sebastian J Buss
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Selina Hein
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Derliz Mereles
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Marco Hagenmueller
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Erbel
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Winfried März
- Synlab Academy, Synlab Holding Deutschland GmbH, Augsburg, Germany
| | - Christian Booz
- 9173Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Thomas J Vogl
- 9173Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan E Hardt
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| | - Marco Ochs
- Department of Cardiology, Angiology and Pulmonology, 27178Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
16
|
The Influence of Mitochondrial-DNA-Driven Inflammation Pathways on Macrophage Polarization: A New Perspective for Targeted Immunometabolic Therapy in Cerebral Ischemia-Reperfusion Injury. Int J Mol Sci 2021; 23:ijms23010135. [PMID: 35008558 PMCID: PMC8745401 DOI: 10.3390/ijms23010135] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/14/2021] [Accepted: 12/19/2021] [Indexed: 12/18/2022] Open
Abstract
Cerebral ischemia-reperfusion injury is related to inflammation driven by free mitochondrial DNA. At the same time, the pro-inflammatory activation of macrophages, that is, polarization in the M1 direction, aggravates the cycle of inflammatory damage. They promote each other and eventually transform macrophages/microglia into neurotoxic macrophages by improving macrophage glycolysis, transforming arginine metabolism, and controlling fatty acid synthesis. Therefore, we propose targeting the mtDNA-driven inflammatory response while controlling the metabolic state of macrophages in brain tissue to reduce the possibility of cerebral ischemia-reperfusion injury.
Collapse
|
17
|
Greiner JV, Glonek T. Intracellular ATP Concentration and Implication for Cellular Evolution. BIOLOGY 2021; 10:1166. [PMID: 34827159 PMCID: PMC8615055 DOI: 10.3390/biology10111166] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022]
Abstract
Crystalline lens and striated muscle exist at opposite ends of the metabolic spectrum. Lens is a metabolically quiescent tissue, whereas striated muscle is a mechanically dynamic tissue with high-energy requirements, yet both tissues contain millimolar levels of ATP (>2.3 mM), far exceeding their underlying metabolic needs. We explored intracellular concentrations of ATP across multiple cells, tissues, species, and domains to provide context for interpreting lens/striated muscle data. Our database revealed that high intracellular ATP concentrations are ubiquitous across diverse life forms including species existing from the Precambrian Era, suggesting an ancient highly conserved role for ATP, independent of its widely accepted view as primarily "metabolic currency". Our findings reinforce suggestions that the primordial function of ATP was non-metabolic in nature, serving instead to prevent protein aggregation.
Collapse
Affiliation(s)
- Jack V. Greiner
- The Schepens Eye Research Institute of Massachusetts Eye & Ear Infirmary, Boston, MA 02114, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
- Clinical Eye Research of Boston, Boston, MA 02114, USA;
| | - Thomas Glonek
- Clinical Eye Research of Boston, Boston, MA 02114, USA;
| |
Collapse
|
18
|
Abstract
Impaired cardiac energy metabolism has been proposed as a mechanism common to different heart failure aetiologies. The energy-depletion hypothesis was pursued by several researchers, and is still a topic of considerable interest. Unlike most organs, in the heart, the creatine kinase system represents a major component of the metabolic machinery, as it functions as an energy shuttle between mitochondria and cytosol. In heart failure, the decrease in creatine level anticipates the reduction in adenosine triphosphate, and the degree of myocardial phosphocreatine/adenosine triphosphate ratio reduction correlates with disease severity, contractile dysfunction, and myocardial structural remodelling. However, it remains to be elucidated whether an impairment of phosphocreatine buffer activity contributes to the pathophysiology of heart failure and whether correcting this energy deficit might prove beneficial. The effects of creatine deficiency and the potential utility of creatine supplementation have been investigated in experimental and clinical models, showing controversial findings. The goal of this article is to provide a comprehensive overview on the role of creatine in cardiac energy metabolism, the assessment and clinical value of creatine deficiency in heart failure, and the possible options for the specific metabolic therapy.
Collapse
|
19
|
Wang H, Wang X, Li P, Dong M, Yao SQ, Tang B. Fluorescent probes for visualizing ROS-associated proteins in disease. Chem Sci 2021; 12:11620-11646. [PMID: 34659698 PMCID: PMC8442704 DOI: 10.1039/d1sc02165f] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/06/2021] [Indexed: 12/14/2022] Open
Abstract
Abnormal expression of proteins, including catalytic and expression dysfunction, is directly related to the development of various diseases in living organisms. Reactive oxygen species (ROS) could regulate protein expression by redox modification or cellular signal pathway and thus influence the development of disease. Determining the expression level and activity of these ROS-associated proteins is of considerable importance in early-stage disease diagnosis and the identification of new drug targets. Fluorescence imaging technology has emerged as a powerful tool for specific in situ imaging of target proteins by virtue of its non-invasiveness, high sensitivity and good spatiotemporal resolution. In this review, we summarize advances made in the past decade for the design of fluorescent probes that have contributed to tracking ROS-associated proteins in disease. We envision that this review will attract significant attention from a wide range of researchers in their utilization of fluorescent probes for in situ investigation of pathological processes synergistically regulated by both ROS and proteins.
Collapse
Affiliation(s)
- Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University Jinan 250014 P. R. China
| | - Xin Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University Jinan 250014 P. R. China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University Jinan 250014 P. R. China
| | - Mingyan Dong
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University Jinan 250014 P. R. China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore Singapore 117543 Singapore
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University Jinan 250014 P. R. China
| |
Collapse
|
20
|
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
|
21
|
Ostojic SM. Modulation of CT1 Function: From Klotho Protein to Ammonia and Beyond. Front Nutr 2021; 8:660021. [PMID: 34041260 PMCID: PMC8143434 DOI: 10.3389/fnut.2021.660021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Sergej M Ostojic
- FSPE Applied Bioenergetics Lab, University of Novi Sad, Novi Sad, Serbia
| |
Collapse
|
22
|
Role of Creatine in the Heart: Health and Disease. Nutrients 2021; 13:nu13041215. [PMID: 33917009 PMCID: PMC8067763 DOI: 10.3390/nu13041215] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 12/26/2022] Open
Abstract
Creatine is a key player in heart contraction and energy metabolism. Creatine supplementation (throughout the paper, only supplementation with creatine monohydrate will be reviewed, as this is by far the most used and best-known way of supplementing creatine) increases creatine content even in the normal heart, and it is generally safe. In heart failure, creatine and phosphocreatine decrease because of decreased expression of the creatine transporter, and because phosphocreatine degrades to prevent adenosine triphosphate (ATP) exhaustion. This causes decreased contractility reserve of the myocardium and correlates with left ventricular ejection fraction, and it is a predictor of mortality. Thus, there is a strong rationale to supplement with creatine the failing heart. Pending additional trials, creatine supplementation in heart failure may be useful given data showing its effectiveness (1) against specific parameters of heart failure, and (2) against the decrease in muscle strength and endurance of heart failure patients. In heart ischemia, the majority of trials used phosphocreatine, whose mechanism of action is mostly unrelated to changes in the ergogenic creatine-phosphocreatine system. Nevertheless, preliminary data with creatine supplementation are encouraging, and warrant additional studies. Prevention of cardiac toxicity of the chemotherapy compounds anthracyclines is a novel field where creatine supplementation may also be useful. Creatine effectiveness in this case may be because anthracyclines reduce expression of the creatine transporter, and because of the pleiotropic antioxidant properties of creatine. Moreover, creatine may also reduce concomitant muscle damage by anthracyclines.
Collapse
|
23
|
Zervou S, McAndrew DJ, Whittington HJ, Lake HA, Park KC, Cha KM, Ostrowski PJ, Eykyn TR, Schneider JE, Neubauer S, Lygate CA. Subtle Role for Adenylate Kinase 1 in Maintaining Normal Basal Contractile Function and Metabolism in the Murine Heart. Front Physiol 2021; 12:623969. [PMID: 33867998 PMCID: PMC8044416 DOI: 10.3389/fphys.2021.623969] [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: 11/02/2020] [Accepted: 03/15/2021] [Indexed: 11/22/2022] Open
Abstract
Aims Adenylate kinase 1 (AK1) catalyses the reaction 2ADP ↔ ATP + AMP, extracting extra energy under metabolic stress and promoting energetic homeostasis. We hypothesised that increased AK1 activity would have negligible effects at rest, but protect against ischaemia/reperfusion (I/R) injury. Methods and Results Cardiac-specific AK1 overexpressing mice (AK1-OE) had 31% higher AK1 activity (P = 0.009), with unchanged total creatine kinase and citrate synthase activities. Male AK1-OE exhibited mild in vivo dysfunction at baseline with lower LV pressure, impaired relaxation, and contractile reserve. LV weight was 19% higher in AK1-OE males due to higher tissue water content in the absence of hypertrophy or fibrosis. AK1-OE hearts had significantly raised creatine, unaltered total adenine nucleotides, and 20% higher AMP levels (P = 0.05), but AMP-activated protein kinase was not activated (P = 0.85). 1H-NMR revealed significant differences in LV metabolite levels compared to wild-type, with aspartate, tyrosine, sphingomyelin, cholesterol all elevated, whereas taurine and triglycerides were significantly lower. Ex vivo global no-flow I/R, caused four-of-seven AK1-OE hearts to develop terminal arrhythmia (cf. zero WT), yet surviving AK1-OE hearts had improved functional recovery. However, AK1-OE did not influence infarct size in vivo and arrhythmias were only observed ex vivo, probably as an artefact of adenine nucleotide loss during cannulation. Conclusion Modest elevation of AK1 may improve functional recovery following I/R, but has unexpected impact on LV weight, function and metabolite levels under basal resting conditions, suggesting a more nuanced role for AK1 underpinning myocardial energy homeostasis and not just as a response to stress.
Collapse
Affiliation(s)
- Sevasti Zervou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Debra J McAndrew
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Hannah J Whittington
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Hannah A Lake
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Kyung Chan Park
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, United Kingdom.,Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Kuan Minn Cha
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Philip J Ostrowski
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Thomas R Eykyn
- British Heart Foundation Centre for Research Excellence, King's College London, St. Thomas Hospital, London, United Kingdom
| | - Jürgen E Schneider
- Experimental and Preclinical Imaging Centre (ePIC), Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
24
|
Piquereau J, Veksler V, Novotova M, Ventura-Clapier R. Energetic Interactions Between Subcellular Organelles in Striated Muscles. Front Cell Dev Biol 2020; 8:581045. [PMID: 33134298 PMCID: PMC7561670 DOI: 10.3389/fcell.2020.581045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/15/2020] [Indexed: 01/12/2023] Open
Abstract
Adult striated muscle cells present highly organized structure with densely packed intracellular organelles and a very sparse cytosol accounting for only few percent of cell volume. These cells have a high and fluctuating energy demand that, in continuously working oxidative muscles, is fulfilled mainly by oxidative metabolism. ATP produced by mitochondria should be directed to the main energy consumers, ATPases of the excitation-contraction system; at the same time, ADP near ATPases should rapidly be eliminated. This is achieved by phosphotransfer kinases, the most important being creatine kinase (CK). Specific CK isoenzymes are located in mitochondria and in close proximity to ATPases, forming efficient energy shuttle between these structures. In addition to phosphotransfer kinases, ATP/ADP can be directly channeled between mitochondria co-localized with ATPases in a process called “direct adenine nucleotide channeling, DANC.” This process is highly plastic so that inactivation of the CK system increases the participation of DANC to energy supply owing to the rearrangement of cell structure. The machinery for DANC is built during postnatal development in parallel with the increase in mitochondrial mass, organization, and complexification of the cell structure. Disorganization of cell architecture remodels the mitochondrial network and decreases the efficacy of DANC, showing that this process is intimately linked to cardiomyocyte structure. Accordingly, in heart failure, disorganization of the cell structure along with decrease in mitochondrial mass reduces the efficacy of DANC and together with alteration of the CK shuttle participates in energetic deficiency contributing to contractile failure.
Collapse
Affiliation(s)
- Jérôme Piquereau
- Université Paris-Saclay, Inserm, UMR-S 1180, Châtenay-Malabry, France
| | - Vladimir Veksler
- Université Paris-Saclay, Inserm, UMR-S 1180, Châtenay-Malabry, France
| | - Marta Novotova
- Department of Cellular Cardiology, Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | |
Collapse
|
25
|
Ramachandra CJA, Hernandez-Resendiz S, Crespo-Avilan GE, Lin YH, Hausenloy DJ. Mitochondria in acute myocardial infarction and cardioprotection. EBioMedicine 2020; 57:102884. [PMID: 32653860 PMCID: PMC7355051 DOI: 10.1016/j.ebiom.2020.102884] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022] Open
Abstract
Acute myocardial infarction (AMI) and the heart failure (HF) that often follows are among the leading causes of death and disability worldwide. As such, new treatments are needed to protect the myocardium against the damaging effects of the acute ischaemia and reperfusion injury (IRI) that occurs in AMI, in order to reduce myocardial infarct (MI) size, preserve cardiac function, and improve patient outcomes. In this regard, cardiac mitochondria play a dual role as arbiters of cell survival and death following AMI. Therefore, preventing mitochondrial dysfunction induced by acute myocardial IRI is an important therapeutic strategy for cardioprotection. In this article, we review the role of mitochondria as key determinants of acute myocardial IRI, and we highlight their roles as therapeutic targets for reducing MI size and preventing HF following AMI. In addition, we discuss the challenges in translating mitoprotective strategies into the clinical setting for improving outcomes in AMI patients.
Collapse
Affiliation(s)
- Chrishan J A Ramachandra
- National Heart Research Institute Singapore, National Heart Centre, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | - Sauri Hernandez-Resendiz
- National Heart Research Institute Singapore, National Heart Centre, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany
| | - Gustavo E Crespo-Avilan
- National Heart Research Institute Singapore, National Heart Centre, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany
| | - Ying-Hsi Lin
- National Heart Research Institute Singapore, National Heart Centre, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre, Singapore; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore; Department of Biochemistry, Medical Faculty, Justus Liebig-University, Giessen, Germany; Yong Loo Lin School of Medicine, National University Singapore, Singapore; The Hatter Cardiovascular Institute, University College London, London, UK; Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan.
| |
Collapse
|
26
|
The Effects of Creatine and Related Compounds on Cardiovascular System: From Basic to Applied Studies. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2020. [DOI: 10.2478/sjecr-2019-0066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Beneficial effects of creatine were firstly shown in sport, where itself has been recognized as an ergogenic substance, increasing exercise endurancе, muscle strength and lean body mass. Creatine supplementation is very interesting, due to the fact that creatine supplementation have been reported to be beneficial for wide spectrum of diseases and conditions referring neuro-degenerative, rheumatic diseases, myopathies, cancer, type 2 diabetes. Creatine is a principle component of the creatine kinase/phosphagen system. In cardiomyocytes, it plays an important role in the buffering and transport of chemical energy to ensure that supply meets the dynamic demands of the heart. Studies in mice proved that elevated creatine protects the heart from ischemia-reperfusion injury. A natural precursor of creatine, guanidinoacetic acid (GAA), plays an important role as an energy carrier/mediator in the cell. GAA is formed in the first step of creatine synthesis. Supplementation with GAA might be of great significance in some circumstances where biosynthesis of GAA is limited like deficient diet, kidney failure, renal insufficiency, exercise-related GAA depletion. Betaine is a neutral compound in the form of zwitterion. Betaine supplementation is associated with improved cognition, neuroprotection, cardioprotection and exercise physiology. Betaine insufficiency represents increased risk for secondary heart failure and acute myocardial infarction. This mini-review outlines the evidence in support of creatine and creatine related compounds (GAA and betaine) elevation and examines the pharmacological approaches that are currently available. Since data from the available studies, regarding cardioprotection are inconsistent, this review might help clarifying the benefits of creatine, GAA and betaine supplementation on cardiovascular system.
Collapse
|
27
|
Peterzan MA, Lewis AJM, Neubauer S, Rider OJ. Non-invasive investigation of myocardial energetics in cardiac disease using 31P magnetic resonance spectroscopy. Cardiovasc Diagn Ther 2020; 10:625-635. [PMID: 32695642 DOI: 10.21037/cdt-20-275] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cardiac metabolism and function are intrinsically linked. High-energy phosphates occupy a central and obligate position in cardiac metabolism, coupling oxygen and substrate fuel delivery to the myocardium with external work. This insight underlies the widespread clinical use of ischaemia testing. However, other deficits in high-energy phosphate metabolism (not secondary to supply-demand mismatch of oxygen and substrate fuels) may also be documented, and are of particular interest when found in the context of structural heart disease. This review introduces the scope of deficits in high-energy phosphate metabolism that may be observed in the myocardium, how to assess for them, and how they might be interpreted.
Collapse
Affiliation(s)
- Mark A Peterzan
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew J M Lewis
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Oliver J Rider
- University of Oxford Centre for Clinical Magnetic Resonance Research (OCMR), Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
28
|
Aksentijević D, Zervou S, Eykyn TR, McAndrew DJ, Wallis J, Schneider JE, Neubauer S, Lygate CA. Age-Dependent Decline in Cardiac Function in Guanidinoacetate- N-Methyltransferase Knockout Mice. Front Physiol 2020; 10:1535. [PMID: 32038270 PMCID: PMC6985570 DOI: 10.3389/fphys.2019.01535] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/05/2019] [Indexed: 01/15/2023] Open
Abstract
Aim Guanidinoacetate N-methyltransferase (GAMT) is the second essential enzyme in creatine (Cr) biosynthesis. Short-term Cr deficiency is metabolically well tolerated as GAMT–/– mice exhibit normal exercise capacity and response to ischemic heart failure. However, we hypothesized long-term consequences of Cr deficiency and/or accumulation of the Cr precursor guanidinoacetate (GA). Methods Cardiac function and metabolic profile were studied in GAMT–/– mice >1 year. Results In vivo LV catheterization revealed lower heart rate and developed pressure in aging GAMT–/– but normal lung weight and survival versus age-matched controls. Electron microscopy indicated reduced mitochondrial volume density in GAMT–/– hearts (P < 0.001), corroborated by lower mtDNA copy number (P < 0.004), and citrate synthase activity (P < 0.05), however, without impaired mitochondrial respiration. Furthermore, myocardial energy stores and key ATP homeostatic enzymes were barely altered, while pathology was unrelated to oxidative stress since superoxide production and protein carbonylation were unaffected. Gene expression of PGC-1α was 2.5-fold higher in GAMT–/– hearts while downstream genes were not activated, implicating a dysfunction in mitochondrial biogenesis signaling. This was normalized by 10 days of dietary Cr supplementation, as were all in vivo functional parameters, however, it was not possible to differentiate whether relief from Cr deficiency or GA toxicity was causative. Conclusion Long-term Cr deficiency in GAMT–/– mice reduces mitochondrial volume without affecting respiratory function, most likely due to impaired biogenesis. This is associated with hemodynamic changes without evidence of heart failure, which may represent an acceptable functional compromise in return for reduced energy demand in aging mice.
Collapse
Affiliation(s)
- Dunja Aksentijević
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine and Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Sevasti Zervou
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine and Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Thomas R Eykyn
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, United Kingdom
| | - Debra J McAndrew
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine and Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Julie Wallis
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine and Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Jurgen E Schneider
- Experimental and Preclinical Imaging Centre, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Stefan Neubauer
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine and Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Craig A Lygate
- Radcliffe Department of Medicine, Division of Cardiovascular Medicine and Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
Overexpression of mitochondrial creatine kinase preserves cardiac energetics without ameliorating murine chronic heart failure. Basic Res Cardiol 2020; 115:12. [PMID: 31925563 PMCID: PMC6954138 DOI: 10.1007/s00395-020-0777-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/02/2020] [Indexed: 01/24/2023]
Abstract
Mitochondrial creatine kinase (Mt-CK) is a major determinant of cardiac energetic status and is down-regulated in chronic heart failure, which may contribute to disease progression. We hypothesised that cardiomyocyte-specific overexpression of Mt-CK would mitigate against these changes and thereby preserve cardiac function. Male Mt-CK overexpressing mice (OE) and WT littermates were subjected to transverse aortic constriction (TAC) or sham surgery and assessed by echocardiography at 0, 3 and 6 weeks alongside a final LV haemodynamic assessment. Regardless of genotype, TAC mice developed progressive LV hypertrophy, dilatation and contractile dysfunction commensurate with pressure overload-induced chronic heart failure. There was a trend for improved survival in OE-TAC mice (90% vs 73%, P = 0.08), however, OE-TAC mice exhibited greater LV dilatation compared to WT and no functional parameters were significantly different under baseline conditions or during dobutamine stress test. CK activity was 37% higher in OE-sham versus WT-sham hearts and reduced in both TAC groups, but was maintained above normal values in the OE-TAC hearts. A separate cohort of mice received in vivo cardiac 31P-MRS to measure high-energy phosphates. There was no difference in the ratio of phosphocreatine-to-ATP in the sham mice, however, PCr/ATP was reduced in WT-TAC but preserved in OE-TAC (1.04 ± 0.10 vs 2.04 ± 0.22; P = 0.007). In conclusion, overexpression of Mt-CK activity prevented the changes in cardiac energetics that are considered hallmarks of a failing heart. This had a positive effect on early survival but was not associated with improved LV remodelling or function during the development of chronic heart failure.
Collapse
|
30
|
Wu GS, Li HK, Zhang WD. Metabolomics and its application in the treatment of coronary heart disease with traditional Chinese medicine. Chin J Nat Med 2020; 17:321-330. [PMID: 31171266 DOI: 10.1016/s1875-5364(19)30037-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Indexed: 12/19/2022]
Abstract
Traditional Chinese Medicine (TCM) is the treasure of Chinese Nation and gained the gradual acceptance of the international community. However, the methods and theories of TCM understanding of diseases are lack of appropriate modern scientific characterization systems. Moreover, traditional risk factors cannot promote to detection and prevent those patients with coronary artery disease (CAD) who have not developed acute myocardial infarction (MI) in time. To sum up, there is still no objective systematic evaluation system for the therapeutic mechanism of TCM in the prevention and cure of cardiovascular disease. Thus, new ideas and technologies are needed. The development of omics technology, especially metabolomics, can be used to predict the level of metabolites in vivo and diagnose the physiological state of the body in time to guide the corresponding intervention. In particular, metabolomics is also a very powerful tool to promote the modernization of TCM and the development of TCM in personalized medicine. This article summarized the application of metabolomics in the early diagnosis, the discovery of biomarkers and the treatment of TCM in CAD.
Collapse
Affiliation(s)
- Gao-Song Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hou-Kai Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Wei-Dong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
31
|
Zhang D, Ma M, Liu Y. Protective Effects of Incretin Against Age-Related Diseases. Curr Drug Deliv 2019; 16:793-806. [PMID: 31622202 DOI: 10.2174/1567201816666191010145029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/01/2019] [Accepted: 09/19/2019] [Indexed: 12/11/2022]
Abstract
Incretin contains two peptides named glucagon-like peptide-1(GLP-1) and glucose-dependent
insulinotropic polypeptide (GIP). Drug therapy using incretin has become a new strategy for diabetic
treatments due to its significant effects on improving insulin receptors and promoting insulinotropic
secretion. Considering the fact that diabetes millitus is a key risk factor for almost all age-related diseases,
the extensive protective roles of incretin in chronic diseases have received great attention. Based
on the evidence from animal experiments, where incretin can protect against the pathophysiological
processes of neurodegenerative diseases, clinical trials for the treatments of Alzheimer’s disease (AD)
and Parkinson’s disease (PD) patients are currently ongoing. Moreover, the protective effect of incretin
on heart has been observed in cardiac myocytes, smooth muscle cells and endothelial cells of vessels.
Meanwhile, incretin can also inhibit the proliferation of aortic vascular smooth muscle cells, which can
induce atherosclerogenesis. Incretin is also beneficial for diabetic microvascular complications, including
nephropathy, retinopathy and gastric ulcer, as well as the hepatic-related diseases such as NAFLD
and NASH. Besides, the anti-tumor properties of incretin have been proven in diverse cancers including
ovarian cancer, pancreas cancer, prostate cancer and breast cancer.
Collapse
Affiliation(s)
- Di Zhang
- Chemistry Department, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Mingzhu Ma
- Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yueze Liu
- Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
32
|
Whittington HJ, Ostrowski PJ, McAndrew DJ, Cao F, Shaw A, Eykyn TR, Lake HA, Tyler J, Schneider JE, Neubauer S, Zervou S, Lygate CA. Over-expression of mitochondrial creatine kinase in the murine heart improves functional recovery and protects against injury following ischaemia-reperfusion. Cardiovasc Res 2019; 114:858-869. [PMID: 29509881 PMCID: PMC5909653 DOI: 10.1093/cvr/cvy054] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 03/01/2018] [Indexed: 12/25/2022] Open
Abstract
Aims Mitochondrial creatine kinase (MtCK) couples ATP production via oxidative phosphorylation to phosphocreatine in the cytosol, which acts as a mobile energy store available for regeneration of ATP at times of high demand. We hypothesized that elevating MtCK would be beneficial in ischaemia-reperfusion (I/R) injury. Methods and results Mice were created over-expressing the sarcomeric MtCK gene with αMHC promoter at the Rosa26 locus (MtCK-OE) and compared with wild-type (WT) littermates. MtCK activity was 27% higher than WT, with no change in other CK isoenzymes or creatine levels. Electron microscopy confirmed normal mitochondrial cell density and mitochondrial localization of transgenic protein. Respiration in isolated mitochondria was unaltered and metabolomic analysis by 1 H-NMR suggests that cellular metabolism was not grossly affected by transgene expression. There were no significant differences in cardiac structure or function under baseline conditions by cine-MRI or LV haemodynamics. In Langendorff-perfused hearts subjected to 20 min ischaemia and 30 min reperfusion, MtCK-OE exhibited less ischaemic contracture, and improved functional recovery (Rate pressure product 58% above WT; P < 0.001). These hearts had reduced myocardial infarct size, which was confirmed in vivo: 55 ± 4% in WT vs. 29 ± 4% in MtCK-OE; P < 0.0001). Isolated cardiomyocytes from MtCK-OE hearts exhibited delayed opening of the mitochondrial permeability transition pore (mPTP) compared to WT, which was confirmed by reduced mitochondrial swelling in response to calcium. There was no detectable change in the structural integrity of the mitochondrial membrane. Conclusions Modest elevation of MtCK activity in the heart does not adversely affect cellular metabolism, mitochondrial or in vivo cardiac function, but modifies mPTP opening to protect against I/R injury and improve functional recovery. Our findings support MtCK as a prime therapeutic target in myocardial ischaemia.
Collapse
Affiliation(s)
- Hannah J Whittington
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, The Wellcome Centre for Human Genetics, and the BHF Centre of Research Excellence, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Philip J Ostrowski
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, The Wellcome Centre for Human Genetics, and the BHF Centre of Research Excellence, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Debra J McAndrew
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, The Wellcome Centre for Human Genetics, and the BHF Centre of Research Excellence, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Fang Cao
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, The Wellcome Centre for Human Genetics, and the BHF Centre of Research Excellence, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Andrew Shaw
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, The Wellcome Centre for Human Genetics, and the BHF Centre of Research Excellence, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Thomas R Eykyn
- School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, St Thomas' Hospital, London, UK
| | - Hannah A Lake
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, The Wellcome Centre for Human Genetics, and the BHF Centre of Research Excellence, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Jack Tyler
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, The Wellcome Centre for Human Genetics, and the BHF Centre of Research Excellence, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Jurgen E Schneider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, The Wellcome Centre for Human Genetics, and the BHF Centre of Research Excellence, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK.,Experimental and Preclinical Imaging Centre (ePIC), Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, The Wellcome Centre for Human Genetics, and the BHF Centre of Research Excellence, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Sevasti Zervou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, The Wellcome Centre for Human Genetics, and the BHF Centre of Research Excellence, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, The Wellcome Centre for Human Genetics, and the BHF Centre of Research Excellence, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
33
|
Takahashi Y, Kioka H, Shintani Y, Ohki A, Takashima S, Sakata Y, Higuchi T, Saito S. Detection of increased intracerebral lactate in a mouse model of Leigh syndrome using proton MR spectroscopy. Magn Reson Imaging 2019; 58:38-43. [DOI: 10.1016/j.mri.2019.01.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/12/2019] [Accepted: 01/12/2019] [Indexed: 12/16/2022]
|
34
|
Fowler ED, Hauton D, Boyle J, Egginton S, Steele DS, White E. Energy Metabolism in the Failing Right Ventricle: Limitations of Oxygen Delivery and the Creatine Kinase System. Int J Mol Sci 2019; 20:E1805. [PMID: 31013688 PMCID: PMC6514649 DOI: 10.3390/ijms20081805] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) results in hypertrophic remodeling of the right ventricle (RV) to overcome increased pulmonary pressure. This increases the O2 consumption of the myocardium, and without a concomitant increase in energy generation, a mismatch with demand may occur. Eventually, RV function can no longer be sustained, and RV failure occurs. Beta-adrenergic blockers (BB) are thought to improve survival in left heart failure, in part by reducing energy expenditure and hypertrophy, however they are not currently a therapy for PAH. The monocrotaline (MCT) rat model of PAH was used to investigate the consequence of RV failure on myocardial oxygenation and mitochondrial function. A second group of MCT rats was treated daily with the beta-1 blocker metoprolol (MCT + BB). Histology confirmed reduced capillary density and increased capillary supply area without indications of capillary rarefaction in MCT rats. A computer model of O2 flux was applied to the experimentally recorded capillary locations and predicted a reduction in mean tissue PO2 in MCT rats. The fraction of hypoxic tissue (defined as PO2 < 0.5 mmHg) was reduced following beta-1 blocker (BB) treatment. The functionality of the creatine kinase (CK) energy shuttle was measured in permeabilized RV myocytes by sequential ADP titrations in the presence and absence of creatine. Creatine significantly decreased the KmADP in cells from saline-injected control (CON) rats, but not MCT rats. The difference in KmADP with or without creatine was not different in MCT + BB cells compared to CON or MCT cells. Improved myocardial energetics could contribute to improved survival of PAH with chronic BB treatment.
Collapse
Affiliation(s)
- Ewan D Fowler
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK.
- Cardiac Research Laboratories, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK.
| | - David Hauton
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK.
- Metabolomics Research Group, Chemistry Research Laboratory, University of Oxford, Oxford OX1 3TA, UK.
| | - John Boyle
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK.
| | - Stuart Egginton
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK.
| | - Derek S Steele
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK.
| | - Ed White
- Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
35
|
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
|
36
|
Abstract
Isoforms of creatine kinase (CK) generate and use phosphocreatine, a concentrated and highly diffusible cellular "high energy" intermediate, for the main purpose of energy buffering and transfer in order to maintain cellular energy homeostasis. The mitochondrial CK isoform (mtCK) localizes to the mitochondrial intermembrane and cristae space, where it assembles into peripherally membrane-bound, large cuboidal homooctamers. These are part of proteolipid complexes wherein mtCK directly interacts with cardiolipin and other anionic phospholipids, as well as with the VDAC channel in the outer membrane. This leads to a stabilization and cross-linking of inner and outer mitochondrial membrane, forming so-called contact sites. Also the adenine nucleotide translocator of the inner membrane can be recruited into these proteolipid complexes, probably mediated by cardiolipin. The complexes have functions mainly in energy transfer to the cytosol and stimulation of oxidative phosphorylation, but also in restraining formation of reactive oxygen species and apoptosis. In vitro evidence indicates a putative role of mtCK in mitochondrial phospholipid distribution, and most recently a role in thermogenesis has been proposed. This review summarizes the essential structural and functional data of these mtCK complexes and describes in more detail the more recent advances in phospholipid interaction, thermogenesis, cancer and evolution of mtCK.
Collapse
|
37
|
Omori K, Katakami N, Yamamoto Y, Ninomiya H, Takahara M, Matsuoka TA, Bamba T, Fukusaki E, Shimomura I. Identification of Metabolites Associated with Onset of CAD in Diabetic Patients Using CE-MS Analysis: A Pilot Study. J Atheroscler Thromb 2018; 26:233-245. [PMID: 30068816 PMCID: PMC6402886 DOI: 10.5551/jat.42945] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aim: Coronary artery disease (CAD) is the result of a complex metabolic disorder caused by various environmental and genetic factors. Metabolomics is a potential tool for identifying biomarkers for better risk classification and for understanding the pathophysiological mechanisms of CAD. With this background, we performed a pilot study to identify metabolites associated with the future onset of CAD in patients with type 2 diabetes. Methods: Sixteen subjects who suffered from CAD event during the observation period and 39 non-CAD subjects who were matched to the CAD subjects for Framingham Coronary Heart Disease Risk Score, diabetes duration, and HbA1c were selected. Capillary electrophoresis time-of-flight mass spectrometry (CE-TOFMS) was used to perform non-targeted metabolome analysis of serum samples collected in 2005. Results: A total of 104 metabolites were identified. Unsupervised principal component analysis (PCA) did not to reveal two distinct clusters of individuals. However, a significant association with CAD was found for 7 metabolites (pelargonic acid, glucosamine:galactosamine, thymine, 3-hydroxybutyric acid, creatine, 2-aminoisobutyric acid, hypoxanthine) and the levels of all these metabolites were significantly lower in the CAD group compared with the non-CAD group. Conclusions: We identified 7 metabolites related to long-term future onset of CAD in Japanese patients with diabetes. Further studies with large sample size would be necessary to confirm our findings, and future studies using in vivo or in vitro models would be necessary to elucidate whether direct relationships exist between the detected metabolites and CAD pathophysiology.
Collapse
Affiliation(s)
- Kazuo Omori
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine
| | - Naoto Katakami
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine.,Department of Metabolism and Atherosclerosis, Osaka University Graduate School of Medicine
| | - Yuichi Yamamoto
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine
| | - Hiroyo Ninomiya
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine
| | - Mitsuyoshi Takahara
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine.,Department of Diabetes Care Medicine, Graduate, School of Medicine, Osaka University
| | - Taka-Aki Matsuoka
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine
| | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University
| | - Eiichiro Fukusaki
- Laboratory of Bioresource Engineering, Department of Biotechnology, Graduate School of Engineering, Osaka University
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine
| |
Collapse
|
38
|
Santacruz L, Arciniegas AJL, Darrabie M, Mantilla JG, Baron RM, Bowles DE, Mishra R, Jacobs DO. Hypoxia decreases creatine uptake in cardiomyocytes, while creatine supplementation enhances HIF activation. Physiol Rep 2018; 5:5/16/e13382. [PMID: 28821596 PMCID: PMC5582266 DOI: 10.14814/phy2.13382] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 07/18/2017] [Indexed: 12/23/2022] Open
Abstract
Creatine (Cr), phosphocreatine (PCr), and creatine kinases (CK) comprise an energy shuttle linking ATP production in mitochondria with cellular consumption sites. Myocytes cannot synthesize Cr: these cells depend on uptake across the cell membrane by a specialized creatine transporter (CrT) to maintain intracellular Cr levels. Hypoxia interferes with energy metabolism, including the activity of the creatine energy shuttle, and therefore affects intracellular ATP and PCr levels. Here, we report that exposing cultured cardiomyocytes to low oxygen levels rapidly diminishes Cr transport by decreasing Vmax and Km. Pharmacological activation of AMP‐activated kinase (AMPK) abrogated the reduction in Cr transport caused by hypoxia. Cr supplementation increases ATP and PCr content in cardiomyocytes subjected to hypoxia, while also significantly augmenting the cellular adaptive response to hypoxia mediated by HIF‐1 activation. Our results indicate that: (1) hypoxia reduces Cr transport in cardiomyocytes in culture, (2) the cytoprotective effects of Cr supplementation are related to enhanced adaptive physiological responses to hypoxia mediated by HIF‐1, and (3) Cr supplementation increases the cellular ATP and PCr content in RNCMs exposed to hypoxia.
Collapse
Affiliation(s)
- Lucia Santacruz
- Department of Molecular Biology and Biochemistry, The University of Texas Medical Branch, Galveston, Texas .,Department of Natural Sciences, Bowie State University, Bowie, Maryland
| | - Antonio Jose Luis Arciniegas
- Department of Medicine, Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | - Rebecca M Baron
- Department of Medicine, Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dawn E Bowles
- Duke University Medical Center, Durham, North Carolina
| | | | - Danny O Jacobs
- Department of Surgery, The University of Texas Medical Branch, Galveston, Texas.,Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
39
|
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
|
40
|
Increasing creatine kinase activity protects against hypoxia / reoxygenation injury but not against anthracycline toxicity in vitro. PLoS One 2017; 12:e0182994. [PMID: 28806770 PMCID: PMC5555628 DOI: 10.1371/journal.pone.0182994] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 07/27/2017] [Indexed: 02/03/2023] Open
Abstract
The creatine kinase (CK) phosphagen system is fundamental to cellular energy homeostasis. Cardiomyocytes express three CK isoforms, namely the mitochondrial sarcomeric CKMT2 and the cytoplasmic CKM and CKB. We hypothesized that augmenting CK in vitro would preserve cell viability and function and sought to determine efficacy of the various isoforms. The open reading frame of each isoform was cloned into pcDNA3.1, followed by transfection and stable selection in human embryonic kidney cells (HEK293). CKMT2- CKM- and CKB-HEK293 cells had increased protein and total CK activity compared to non-transfected cells. Overexpressing any of the three CK isoforms reduced cell death in response to 18h hypoxia at 1% O2 followed by 2h re-oxygenation as assayed using propidium iodide: by 33% in CKMT2, 47% in CKM and 58% in CKB compared to non-transfected cells (P<0.05). Loading cells with creatine did not modify cell survival. Transient expression of CK isoforms in HL-1 cardiac cells elevated isoenzyme activity, but only CKMT2 over-expression protected against hypoxia (0.1% for 24h) and reoxygenation demonstrating 25% less cell death compared to non-transfected control (P<0.01). The same cells were not protected from doxorubicin toxicity (250nM for 48h), in contrast to the positive control. These findings support increased CK activity as protection against ischaemia-reperfusion injury, in particular, protection via CKMT2 in a cardiac-relevant cell line, which merits further investigation in vivo.
Collapse
|
41
|
Suda M, Shimizu I, Yoshida Y, Hayashi Y, Ikegami R, Katsuumi G, Wakasugi T, Yoshida Y, Okuda S, Soga T, Minamino T. Inhibition of dipeptidyl peptidase-4 ameliorates cardiac ischemia and systolic dysfunction by up-regulating the FGF-2/EGR-1 pathway. PLoS One 2017; 12:e0182422. [PMID: 28771625 PMCID: PMC5542565 DOI: 10.1371/journal.pone.0182422] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 07/18/2017] [Indexed: 01/21/2023] Open
Abstract
Dipeptidyl peptidase 4 inhibitors are used worldwide in the management of diabetes, but their role in the prevention or treatment of cardiovascular disorders has yet to be defined. We found that linagliptin, a DPP-4 inhibitor, suppressed capillary rarefaction in the hearts of mice with dietary obesity. Metabolomic analysis performed with capillary electrophoresis/mass spectrometry (LC-MS/MS) showed that linagliptin promoted favorable metabolic remodeling in cardiac tissue, which was characterized by high levels of citrulline and creatine. DNA microarray analysis revealed that the cardiac tissue level of early growth response protein 1 (EGR-1), which activates angiogenesis, was significantly reduced in untreated mice with dietary obesity, while this decrease was inhibited by administration of linagliptin. Mature fibroblast growth factor 2 (FGF-2) has a putative truncation site for DPP-4 at the NH2-terminal, and LC-MS/MS showed that recombinant DPP-4 protein cleaved the NH2-terminal dipeptides of mature FGF-2. Incubation of cultured neonatal rat cardiomyocytes with FGF-2 increased Egr1 expression, while it was suppressed by recombinant DPP-4 protein. Furthermore, vascular endothelial growth factor-A had a critical role in mediating FGF-2/EGR-1 signaling. In conclusion, pharmacological inhibition of DPP-4 suppressed capillary rarefaction and contributed to favorable remodeling of cardiac metabolism in mice with dietary obesity.
Collapse
Affiliation(s)
- Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yuka Hayashi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ryutaro Ikegami
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Goro Katsuumi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takayuki Wakasugi
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yutaka Yoshida
- Department of Structural Pathology, Institute of Nephrology, Graduate School of Medical and Dental Sciences, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- * E-mail:
| |
Collapse
|
42
|
Mingxing F, Landoni G, Zangrillo A, Monaco F, Lomivorotov VV, Hui C, Novikov M, Nepomniashchikh V, Fominskiy E. Phosphocreatine in Cardiac Surgery Patients: A Meta-Analysis of Randomized Controlled Trials. J Cardiothorac Vasc Anesth 2017; 32:762-770. [PMID: 29409711 DOI: 10.1053/j.jvca.2017.07.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Indexed: 11/11/2022]
Abstract
OBJECTIVE There is experimental evidence that phosphocreatine (PCr) can decrease ischemia/reperfusion injury of the heart. The authors investigated if PCr would improve heart performance as compared with standard treatment in cardiac surgery. DESIGN Meta-analysis of randomized controlled trials. SETTING Hospitals. PARTICIPANTS Adult and pediatric patients undergoing cardiac surgery. INTERVENTIONS The ability of PCr to improve cardiac outcomes as compared with standard treatment was investigated. MEASUREMENTS AND MAIN RESULTS PubMed/Medline, Embase, Scopus, Cochrane Library, China National Knowledge Infrastructure, WANGFANG DATA, and VIP Paper Check System were searched to March 1 2017. The authors included 26 randomized controlled trials comprising 1,948 patients. Random and fixed-effects models were used to estimate odds ratio (OR) and mean difference (MD) with 95% confidence interval (CI). PCr use was associated with reduced rates of intraoperative inotropic support (27% v 44%; OR 0.47, 95% CI 0.35-0.61; p < 0.001), major arrhythmias (16% v 28%; OR 0.44, 95% CI 0.27-0.69; p < 0.001), as well as increased spontaneous recovery of the cardiac rhythm immediately after aortic declamping (50% v 34%; OR 2.45, 95% CI 1.82-3.30; p < 0.001) as compared with standard treatment. The use of PCr decreased myocardial damage and augmented left ventricular ejection fraction in the postoperative period; however, MD for these outcomes were small and do not seem to be clinically significant. CONCLUSIONS In randomized trials, PCr administration was associated with reduced rates of intraoperative inotropic support and major arrhythmias, and increased spontaneous recovery of the cardiac rhythm after aortic declamping. Large multicenter evidence is needed to validate these findings.
Collapse
Affiliation(s)
- Fang Mingxing
- Department of Intensive Care, The Third Hospital, Hebei Medical University, Shijiazhuang, Hebei Province, P.R. China
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University of Milan, Milan, Italy
| | - Alberto Zangrillo
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University of Milan, Milan, Italy
| | - Fabrizio Monaco
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vladimir V Lomivorotov
- Department of Anesthesia and Intensive Care, Siberian Biomedical Research Center of the Ministry of Health, Novosibirsk, Russia
| | - Cao Hui
- Department of Intensive Care, The Third Hospital, Hebei Medical University, Shijiazhuang, Hebei Province, P.R. China
| | - Maxim Novikov
- Department of Anesthesia and Intensive Care, Medical Center of Saint-Petersburg State University, Saint-Petersburg, Russia
| | - Valery Nepomniashchikh
- Department of Anesthesia and Intensive Care, Siberian Biomedical Research Center of the Ministry of Health, Novosibirsk, Russia
| | - Evgeny Fominskiy
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy; Department of Anesthesia and Intensive Care, Siberian Biomedical Research Center of the Ministry of Health, Novosibirsk, Russia.
| |
Collapse
|
43
|
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
|
44
|
Atzler D, McAndrew DJ, Cordts K, Schneider JE, Zervou S, Schwedhelm E, Neubauer S, Lygate CA. Dietary Supplementation with Homoarginine Preserves Cardiac Function in a Murine Model of Post-Myocardial Infarction Heart Failure. Circulation 2017; 135:400-402. [DOI: 10.1161/circulationaha.116.025673] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Dorothee Atzler
- From Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (D.A., D.J.M., J.E.S., S.Z., S.N., C.A.L.); Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilians-University Munich, Germany (D.A.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Deutsches Zentrum füz-Kreislauf-Forschung e.V., partner site Munich Heart Alliance, Germany (D.A.); Department of Clinical Pharmacology and Toxicology,
| | - Debra J. McAndrew
- From Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (D.A., D.J.M., J.E.S., S.Z., S.N., C.A.L.); Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilians-University Munich, Germany (D.A.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Deutsches Zentrum füz-Kreislauf-Forschung e.V., partner site Munich Heart Alliance, Germany (D.A.); Department of Clinical Pharmacology and Toxicology,
| | - Kathrin Cordts
- From Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (D.A., D.J.M., J.E.S., S.Z., S.N., C.A.L.); Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilians-University Munich, Germany (D.A.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Deutsches Zentrum füz-Kreislauf-Forschung e.V., partner site Munich Heart Alliance, Germany (D.A.); Department of Clinical Pharmacology and Toxicology,
| | - Jürgen E. Schneider
- From Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (D.A., D.J.M., J.E.S., S.Z., S.N., C.A.L.); Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilians-University Munich, Germany (D.A.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Deutsches Zentrum füz-Kreislauf-Forschung e.V., partner site Munich Heart Alliance, Germany (D.A.); Department of Clinical Pharmacology and Toxicology,
| | - Sevasti Zervou
- From Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (D.A., D.J.M., J.E.S., S.Z., S.N., C.A.L.); Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilians-University Munich, Germany (D.A.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Deutsches Zentrum füz-Kreislauf-Forschung e.V., partner site Munich Heart Alliance, Germany (D.A.); Department of Clinical Pharmacology and Toxicology,
| | - Edzard Schwedhelm
- From Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (D.A., D.J.M., J.E.S., S.Z., S.N., C.A.L.); Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilians-University Munich, Germany (D.A.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Deutsches Zentrum füz-Kreislauf-Forschung e.V., partner site Munich Heart Alliance, Germany (D.A.); Department of Clinical Pharmacology and Toxicology,
| | - Stefan Neubauer
- From Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (D.A., D.J.M., J.E.S., S.Z., S.N., C.A.L.); Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilians-University Munich, Germany (D.A.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Deutsches Zentrum füz-Kreislauf-Forschung e.V., partner site Munich Heart Alliance, Germany (D.A.); Department of Clinical Pharmacology and Toxicology,
| | - Craig A. Lygate
- From Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (D.A., D.J.M., J.E.S., S.Z., S.N., C.A.L.); Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilians-University Munich, Germany (D.A.); Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Deutsches Zentrum füz-Kreislauf-Forschung e.V., partner site Munich Heart Alliance, Germany (D.A.); Department of Clinical Pharmacology and Toxicology,
| |
Collapse
|
45
|
Abstract
Creatine is a principle component of the creatine kinase (CK) phosphagen system common to all vertebrates. It is found in excitable cells, such as cardiomyocytes, where it plays an important role in the buffering and transport of chemical energy to ensure that supply meets the dynamic demands of the heart. Multiple components of the CK system, including intracellular creatine levels, are reduced in heart failure, while ischaemia and hypoxia represent acute crises of energy provision. Elevation of myocardial creatine levels has therefore been suggested as potentially beneficial, however, achieving this goal is not trivial. This mini-review outlines the evidence in support of creatine elevation and critically examines the pharmacological approaches that are currently available. In particular, dietary creatine-supplementation does not sufficiently elevate creatine levels in the heart due to subsequent down-regulation of the plasma membrane creatine transporter (CrT). Attempts to increase passive diffusion and bypass the CrT, e.g. via creatine esters, have yet to be tested in the heart. However, studies in mice with genetic overexpression of the CrT demonstrate proof-of-principle that elevated creatine protects the heart from ischaemia-reperfusion injury. This suggests activation of the CrT as a major unmet pharmacological target. However, translation of this finding to the clinic will require a greater understanding of CrT regulation in health and disease and the development of small molecule activators.
Collapse
Affiliation(s)
| | | | | | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Headington OX3 7BN, UK.
| |
Collapse
|
46
|
Kitzenberg D, Colgan SP, Glover LE. Creatine kinase in ischemic and inflammatory disorders. Clin Transl Med 2016; 5:31. [PMID: 27527620 PMCID: PMC4987751 DOI: 10.1186/s40169-016-0114-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/02/2016] [Indexed: 12/20/2022] Open
Abstract
The creatine/phosphocreatine pathway plays a conserved and central role in energy metabolism. Compartmentalization of specific creatine kinase enzymes permits buffering of local high energy phosphates in a thermodynamically favorable manner, enabling both rapid energy storage and energy transfer within the cell. Augmentation of this metabolic pathway by nutritional creatine supplementation has been shown to elicit beneficial effects in a number of diverse pathologies, particularly those that incur tissue ischemia, hypoxia or oxidative stress. In these settings, creatine and phosphocreatine prevent depletion of intracellular ATP and internal acidification, enhance post-ischemic recovery of protein synthesis and promote free radical scavenging and stabilization of cellular membranes. The creatine kinase energy system is itself further regulated by hypoxic signaling, highlighting the existence of endogenous mechanisms in mammals that can enhance creatine metabolism during oxygen deprivation to promote tissue resolution and homeostasis. Here, we review recent insights into the creatine kinase pathway, and provide rationale for dietary creatine supplementation in human ischemic and inflammatory pathologies.
Collapse
Affiliation(s)
- David Kitzenberg
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, 12700 East 19th Ave. MS B-146, Aurora, CO, 80045, USA.,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Sean P Colgan
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, 12700 East 19th Ave. MS B-146, Aurora, CO, 80045, USA.,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Louise E Glover
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, 12700 East 19th Ave. MS B-146, Aurora, CO, 80045, USA. .,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
47
|
Landoni G, Zangrillo A, Lomivorotov VV, Likhvantsev V, Ma J, De Simone F, Fominskiy E. Cardiac protection with phosphocreatine: a meta-analysis. Interact Cardiovasc Thorac Surg 2016; 23:637-46. [PMID: 27318357 DOI: 10.1093/icvts/ivw171] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/09/2016] [Indexed: 11/12/2022] Open
Abstract
Phosphocreatine (PCr) plays an important role in the energy metabolism of the heart and a decrease in its intracellular concentration results in alteration of myocardium energetics and work. We conducted a meta-analysis of all randomized and matched trials that compared PCr with placebo or standard treatment in patients with coronary artery disease or chronic heart failure or in those undergoing cardiac surgery. We systematically searched PubMed/Medline, Embase, Cochrane Central Register of Controlled Trials and Google Scholar up to 1 November 2015, for pertinent trials. The primary outcome was all-cause mortality. Secondary outcomes included inotrope use, ejection fraction (EF), peak creatinine kinase-myocardial band (CK-MB) release and the incidence of major arrhythmias, as well as spontaneous recovery of the heart performance in the subgroup of patients undergoing cardiac surgery with cardiopulmonary bypass. We pooled odds ratio (OR) and mean difference (MD) using fixed- and random effects models. We identified 41 controlled trials, of them 32 were randomized. Patients receiving PCr had lower all-cause mortality when compared with the control group [61/1731 (3.5%) vs 177/1667 (10.6%); OR: 0.71, 95% CI: 0.51-0.99; P = 0.04; I(2) = 0%; with 3400 patients and 22 trials included]. Phosphocreatine administration was associated with higher LVEF (MD: 3.82, 95% CI: 1.18-6.46; P = 0.005; I(2) = 98%), lower peak CK-MB release (MD: -6.08, 95% CI: -8.01, -4.15; P < 0.001; I(2) = 97%), lower rate of major arrhythmias (OR: 0.42; 95% CI: 0.27-0.66; P < 0.001; I(2) = 0%), lower incidence of inotropic support (OR: 0.39, 95% CI: 0.25-0.61; P < 0.001; I(2) = 56%) and a higher level of spontaneous recovery of the heart performance after cardiopulmonary bypass (OR: 3.49, 95% CI: 2.28-5.35; P < 0.001; I(2) = 49%) when compared with the control group. In a mixed population of patients with coronary artery disease, chronic heart failure or in those undergoing cardiac surgery, PCr may reduce all-cause short-term mortality. In addition, PCr administration was associated with improved cardiac outcomes. Owing to the pharmacological plausibility of this effect and to the concordance of the beneficial effects of PCr on several secondary but important outcomes and survival, there is urgent need for a large multicentre randomized trial to confirm these findings.
Collapse
Affiliation(s)
- Giovanni Landoni
- Department of Anaesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy Vita-Salute San Raffaele University of Milan, Italy
| | - Alberto Zangrillo
- Department of Anaesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy Vita-Salute San Raffaele University of Milan, Italy
| | - Vladimir V Lomivorotov
- Department of Anaesthesia and Intensive Care, Academician EN Meshalkin Novosibirsk State Budget Research Institute of Circulation Pathology, Novosibirsk, Russia
| | | | - Jun Ma
- Centre for Anaesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Francesco De Simone
- Department of Anaesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Evgeny Fominskiy
- Department of Anaesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy Department of Anaesthesia and Intensive Care, Academician EN Meshalkin Novosibirsk State Budget Research Institute of Circulation Pathology, Novosibirsk, Russia
| |
Collapse
|
48
|
Zervou S, Yin X, Nabeebaccus AA, O’Brien BA, Cross RL, McAndrew DJ, Atkinson RA, Eykyn TR, Mayr M, Neubauer S, Lygate CA. Proteomic and metabolomic changes driven by elevating myocardial creatine suggest novel metabolic feedback mechanisms. Amino Acids 2016; 48:1969-81. [PMID: 27143170 PMCID: PMC4974297 DOI: 10.1007/s00726-016-2236-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/11/2016] [Indexed: 01/04/2023]
Abstract
Mice over-expressing the creatine transporter have elevated myocardial creatine levels [Cr] and are protected against ischaemia/reperfusion injury via improved energy reserve. However, mice with very high [Cr] develop cardiac hypertrophy and dysfunction. To investigate these contrasting effects, we applied a non-biased hypothesis-generating approach to quantify global protein and metabolite changes in the LV of mice stratified for [Cr] levels: wildtype, moderately elevated, and high [Cr] (65-85; 100-135; 160-250 nmol/mg protein, respectively). Male mice received an echocardiogram at 7 weeks of age with tissue harvested at 8 weeks. RV was used for [Cr] quantification by HPLC to select LV tissue for subsequent analysis. Two-dimensional difference in-gel electrophoresis identified differentially expressed proteins, which were manually picked and trypsin digested for nano-LC-MS/MS. Principal component analysis (PCA) showed efficient group separation (ANOVA P ≤ 0.05) and peptide sequences were identified by mouse database (UniProt 201203) using Mascot. A total of 27 unique proteins were found to be differentially expressed between normal and high [Cr], with proteins showing [Cr]-dependent differential expression, chosen for confirmation, e.g. α-crystallin B, a heat shock protein implicated in cardio-protection and myozenin-2, which could contribute to the hypertrophic phenotype. Nuclear magnetic resonance (¹H-NMR at 700 MHz) identified multiple strong correlations between [Cr] and key cardiac metabolites. For example, positive correlations with α-glucose (r² = 0.45; P = 0.002), acetyl-carnitine (r² = 0.50; P = 0.001), glutamine (r² = 0.59; P = 0.0002); and negative correlations with taurine (r² = 0.74; P < 0.0001), fumarate (r² = 0.45; P = 0.003), aspartate (r² = 0.59; P = 0.0002), alanine (r² = 0.66; P < 0.0001) and phosphocholine (r² = 0.60; P = 0.0002). These findings suggest wide-ranging and hitherto unexpected adaptations in substrate utilisation and energy metabolism with a general pattern of impaired energy generating pathways in mice with very high creatine levels.
Collapse
Affiliation(s)
- Sevasti Zervou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and the BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Xiaoke Yin
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | | | - Brett A. O’Brien
- Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, UK
| | - Rebecca L. Cross
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and the BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Debra J. McAndrew
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and the BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - R. Andrew Atkinson
- Randall Division of Cell and Molecular Biophysics, and the BHF Centre of Research Excellence, Centre for Biomolecular Spectroscopy, King’s College London, London, UK
| | - Thomas R. Eykyn
- Division of Imaging Sciences and Biomedical Engineering, King’s College London, London, UK
| | - Manuel Mayr
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and the BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Craig A. Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and the BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| |
Collapse
|
49
|
Santacruz L, Jacobs DO. Structural correlates of the creatine transporter function regulation: the undiscovered country. Amino Acids 2016; 48:2049-55. [PMID: 26951207 DOI: 10.1007/s00726-016-2206-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 02/23/2016] [Indexed: 10/22/2022]
Abstract
Creatine (Cr) and phosphocreatine constitute an energy shuttle that links ATP production in mitochondria to subcellular locations of ATP consumption. Cells in tissues that are reliant on this energy shuttle, such as myocytes and neurons, appear to have very limited ability to synthesize creatine. Therefore, these cells depend on Cr uptake across the cell membrane by a specialized creatine transporter (CrT solute carrier SLC6A8) in order to maintain intracellular creatine levels. Cr supplementation has been shown to have a beneficial effect in numerous in vitro and in vivo models, particularly in cases of oxidative stress, and is also widely used by athletes as a performance enhancement nutraceutical. Intracellular creatine content is maintained within narrow limits. However, the physiological and cellular mechanisms that mediate Cr transport during health and disease (such as cardiac failure) are not understood. In this narrative mini-review, we summarize the last three decades of research on CrT structure, function and regulation.
Collapse
Affiliation(s)
- Lucia Santacruz
- Department of Biochemistry and Molecular Biology, University of Texas, Medical Branch, 301 University Boulevard, Galveston, TX, USA.
| | - Danny O Jacobs
- Department of Biochemistry and Molecular Biology, University of Texas, Medical Branch, 301 University Boulevard, Galveston, TX, USA.,Departament of Surgery and the institute for Translational Sciences, University of Texas, Medical Branch, Galveston, USA
| |
Collapse
|
50
|
Almeida FM, Oliveira-Junior MC, Souza RA, Petroni RC, Soto SF, Soriano FG, Carvalho PTCD, Albertini R, Damaceno-Rodrigues NR, Lopes FDTQS, Castro-Faria-Neto HC, Martins MA, Dolhnikoff M, Pazetti R, Vieira RP. Creatine supplementation attenuates pulmonary and systemic effects of lung ischemia and reperfusion injury. J Heart Lung Transplant 2016; 35:242-250. [PMID: 26215332 DOI: 10.1016/j.healun.2015.06.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 06/16/2015] [Accepted: 06/24/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Creatine (Cr) is a dietary supplement that presents beneficial effects in experimental models of heart and brain ischemia and reperfusion (I/R) injury. It can improve adenosine 5'-triphosphate generation and reduce cell damage. This study evaluated the effects of Cr supplementation in a model of lung I/R. METHODS Forty male Wistar rats were divided into 4 groups: sham operated, Cr+sham, I/R, and Cr+I/R. We investigated the effects of 5 days of Cr supplementation (0.5 g/kg/day by gavage) before left pulmonary artery ischemia (90 minutes) and reperfusion (120 minutes) on pulmonary and systemic response. RESULTS Cr inhibited the I/R-induced increase in exhaled nitric oxide (p < 0.05), total cells (p < 0.01), and neutrophils (p < 0.001) in bronchoalveolar lavage fluid and in the systemic circulation (p < 0.001). The levels of interleukin-1β (p < 0.05), tissue damping, and tissue elastance (p < 0.05) were also minimized. Cr also inhibited pulmonary edema formation (total proteins in bronchoalveolar lavage fluid, p < 0.001; histologic edema index, p < 0.001) and neutrophils accumulation in lung tissue (p < 0.001). As possible mechanisms underlying Cr effects, we observed a reduced expression of caspase 3 (p < 0.05), reduced expression of Toll-like receptor (TLR) 4, and increased expression of TLR7 in lung tissue (p < 0.001). CONCLUSIONS Cr supplementation presents pulmonary and systemic protective effects in acute lung injury induced by I/R in rats. These beneficial effects seem to be related to the anti-inflammatory and anti-oxidant properties of Cr and modulation of TLRs.
Collapse
Affiliation(s)
- Francine Maria Almeida
- Department of Clinical Medicine (LIM 20), School of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | | | - Renato Aparecido Souza
- Laboratory of Pulmonary and Exercise Immunology (LABPEI), Nove de Julho University (UNINOVE), São Paulo, São Paulo, Brazil; Grupo de Estudos e Pesquisa em Ciência da Saúde (GEP-CS), Instituto Federal de Educação, Ciência e Tecnologia do Sul de Minas Gerais (IFSULDEMINAS), Campus Muzambinho, Muzambinho, Minas Gerais, Brazil
| | - Ricardo Costa Petroni
- Department of Clinical Medicine (LIM 51), School of Medicine University of São Paulo, São Paulo, São Paulo, Brazil
| | - Sonia Fatima Soto
- Department of Clinical Medicine (LIM 20), School of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Francisco Garcia Soriano
- Department of Clinical Medicine (LIM 51), School of Medicine University of São Paulo, São Paulo, São Paulo, Brazil
| | | | - Regiane Albertini
- Laboratory of Pulmonary and Exercise Immunology (LABPEI), Nove de Julho University (UNINOVE), São Paulo, São Paulo, Brazil
| | | | | | - Hugo Caire Castro-Faria-Neto
- Laboratory of Immunopharmacology, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Milton Arruda Martins
- Department of Clinical Medicine (LIM 20), School of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Marisa Dolhnikoff
- Department of Pathology (LIM 05), School of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Rogerio Pazetti
- Department of Cardiopneumology (LIM 61), School of Medicine, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Rodolfo Paula Vieira
- Laboratory of Pulmonary and Exercise Immunology (LABPEI), Nove de Julho University (UNINOVE), São Paulo, São Paulo, Brazil.
| |
Collapse
|