1
|
Panov AV, Mayorov VI, Dikalov SI. Role of Fatty Acids β-Oxidation in the Metabolic Interactions Between Organs. Int J Mol Sci 2024; 25:12740. [PMID: 39684455 DOI: 10.3390/ijms252312740] [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: 10/25/2024] [Revised: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
In recent decades, several discoveries have been made that force us to reconsider old ideas about mitochondria and energy metabolism in the light of these discoveries. In this review, we discuss metabolic interaction between various organs, the metabolic significance of the primary substrates and their metabolic pathways, namely aerobic glycolysis, lactate shuttling, and fatty acids β-oxidation. We rely on the new ideas about the supramolecular structure of the mitochondrial respiratory chain (respirasome), the necessity of supporting substrates for fatty acids β-oxidation, and the reverse electron transfer via succinate dehydrogenase during β-oxidation. We conclude that ATP production during fatty acid β-oxidation has its upper limits and thus cannot support high energy demands alone. Meanwhile, β-oxidation creates conditions that significantly accelerate the cycle: glucose-aerobic glycolysis-lactate-gluconeogenesis-glucose. Therefore, glycolytic ATP production becomes an important energy source in high energy demand. In addition, lactate serves as a mitochondrial substrate after converting to pyruvate + H+ by the mitochondrial lactate dehydrogenase. All coupled metabolic pathways are irreversible, and the enzymes are organized into multienzyme structures.
Collapse
Affiliation(s)
- Alexander V Panov
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31201, USA
| | - Vladimir I Mayorov
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31201, USA
| | | |
Collapse
|
2
|
Augusto SN, Suresh A, Tang WHW. Ceramides as Biomarkers of Cardiovascular Diseases and Heart Failure. Curr Heart Fail Rep 2024; 22:2. [PMID: 39560878 DOI: 10.1007/s11897-024-00689-3] [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] [Accepted: 09/17/2024] [Indexed: 11/20/2024]
Abstract
PURPOSE OF REVIEW Ceramides are lipid species that play several physiological roles in the body, including stress response, inflammation, and apoptosis, and their involvement in lipid metabolism and energy production makes them crucial in the pathophysiology of heart failure (HF). RECENT FINDINGS Several species of ceramides and ceramide signatures have recently been investigated as possible biomarkers of cardiovascular disease (CVD), and risk scores have demonstrated prognostic value in stratifying patients by risk and possibly predicting adverse cardiac events. With growing interest in targeting metabolic dysfunction, understanding the role of ceramides in CVD also opens the possibility of novel therapeutics that target ceramide metabolism to improve cardiac function and cardiac outcomes in patients. Understanding the role of ceramides in CVD opens the possibility of novel diagnostics and theragnostic targeting ceramide metabolism to improve cardiac function and cardiac outcomes in patients with heart failure.
Collapse
Affiliation(s)
- Silvio N Augusto
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA, 9500 Euclid Avenue, Desk J3-4, 44195
| | - Abhilash Suresh
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - W H Wilson Tang
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA, 9500 Euclid Avenue, Desk J3-4, 44195.
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
3
|
Panov AV. The Structure of the Cardiac Mitochondria Respirasome Is Adapted for the β-Oxidation of Fatty Acids. Int J Mol Sci 2024; 25:2410. [PMID: 38397087 PMCID: PMC10889813 DOI: 10.3390/ijms25042410] [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: 12/26/2023] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
It is well known that in the heart and kidney mitochondria, more than 95% of ATP production is supported by the β-oxidation of long-chain fatty acids. However, the β-oxidation of fatty acids by mitochondria has been studied much less than the substrates formed during the catabolism of carbohydrates and amino acids. In the last few decades, several discoveries have been made that are directly related to fatty acid oxidation. In this review, we made an attempt to re-evaluate the β-oxidation of long-chain fatty acids from the perspectives of new discoveries. The single set of electron transporters of the cardiac mitochondrial respiratory chain is organized into three supercomplexes. Two of them contain complex I, a dimer of complex III, and two dimers of complex IV. The third, smaller supercomplex contains a dimer of complex III and two dimers of complex IV. We also considered other important discoveries. First, the enzymes of the β-oxidation of fatty acids are physically associated with the respirasome. Second, the β-oxidation of fatty acids creates the highest level of QH2 and reverses the flow of electrons from QH2 through complex II, reducing fumarate to succinate. Third, β-oxidation is greatly stimulated in the presence of succinate. We argue that the respirasome is uniquely adapted for the β-oxidation of fatty acids. The acyl-CoA dehydrogenase complex reduces the membrane's pool of ubiquinone to QH2, which is instantly oxidized by the smaller supercomplex, generating a high energization of mitochondria and reversing the electron flow through complex II, which reverses the electron flow through complex I, increasing the NADH/NAD+ ratio in the matrix. The mitochondrial nicotinamide nucleotide transhydrogenase catalyzes a hydride (H-, a proton plus two electrons) transfer across the inner mitochondrial membrane, reducing the cytosolic pool of NADP(H), thus providing the heart with ATP for muscle contraction and energy and reducing equivalents for the housekeeping processes.
Collapse
Affiliation(s)
- Alexander V Panov
- Department of Biomedical Sciences, School of Medicine, Mercer University, Macon, GA 31201, USA
| |
Collapse
|
4
|
Ma F, Li P, Zhang S, Shi W, Wang J, Ma Q, Zhao M, Nie Z, Xiao H, Chen X, Xie X. Decreased lipid levels in adult with congenital heart disease: a systematic review and Meta-analysis. BMC Cardiovasc Disord 2023; 23:523. [PMID: 37891491 PMCID: PMC10612202 DOI: 10.1186/s12872-023-03455-w] [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: 03/06/2023] [Accepted: 08/17/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Metabolic disorders were a health problem for many adults with congenital heart disease, however, the differences in metabolic syndrome-related metabolite levels in adults with congenital heart disease compared to the healthy population were unknown. METHODS We collected 18 studies reporting metabolic syndrome-associated metabolite levels in patients with congenital heart disease. Data from different studies were combined under a random-effects model using Cohen's d values. RESULTS The results found that the levels of total cholesterol (Cohen's d -0.68, 95% CI: -0.91 to -0.45), high-density lipoprotein cholesterol (Cohen's d -0.63, 95% CI: -0.89 to -0.37), and low-density lipoprotein cholesterol (Cohen's d -0.32, 95% CI: -0.54 to -0.10) were significantly lower in congenital heart disease patients compared with controls. Congenital heart disease patients also had a lower body mass index (Cohen's d -0.27, 95% CI: -0.42 to -0.12) compared with controls. On the contrary, congenital heart disease patients had higher levels of hemoglobin A1c (Cohen's d 0.93, 95% CI: 0.17 to 1.70) than controls. Meanwhile, there were no significant differences in triglyceride (Cohen's d 0.07, 95% CI: -0.09 to 0.23), blood glucose (Cohen's d -0.12, 95% CI: -0.94 to 0.70) levels, systolic (Cohen's d 0.07, 95% CI: -0.30 to 0.45) and diastolic blood pressure (Cohen's d -0.10, 95% CI: -0.39 to 0.19) between congenital heart disease patients and controls. CONCLUSIONS The lipid levels in patients with congenital heart disease were significantly lower than those in the control group. These data will help in the health management of patients with congenital heart disease and guide clinicians. PROSPERO REGISTRATION NUMBER CRD42022228156.
Collapse
Affiliation(s)
- Fengdie Ma
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Peiqiang Li
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China.
| | - Shasha Zhang
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Wenjing Shi
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Jing Wang
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Qinglong Ma
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Meie Zhao
- The first people's hospital of Lanzhou city, Lanzhou, Gansu, China
| | - Ziyan Nie
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Handan Xiao
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Xinyi Chen
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaodong Xie
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
5
|
Silva AFR, Silva-Reis R, Ferreira R, Oliveira PA, Faustino-Rocha AI, Pinto MDL, Coimbra MA, Silva AMS, Cardoso SM. The Impact of Resveratrol-Enriched Bread on Cardiac Remodeling in a Preclinical Model of Diabetes. Antioxidants (Basel) 2023; 12:antiox12051066. [PMID: 37237932 DOI: 10.3390/antiox12051066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/27/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
The World Health Organization aims to stop the rise of diabetes by 2025, and diet is one of the most efficient non-pharmacological strategies used to prevent it. Resveratrol (RSV) is a natural compound with anti-diabetic properties, and incorporating it into bread is a suitable way to make it more accessible to consumers as it can be included as part of their daily diet. This study aimed to evaluate the effect of RSV-enriched bread in preventing early type 2 diabetes cardiomyopathy in vivo. Male Sprague Dawley rats (3 weeks old) were divided into four groups: controls with plain bread (CB) and RSV bread (CBR), and diabetics with plain bread (DB) and RSV bread (DBR). Type 2 diabetes was induced by adding fructose to the drinking water for two weeks followed by an injection of streptozotocin (STZ) (40 mg/kg). Then, plain bread and RSV bread (10 mg RSV/kg body weight) were included in the rats' diet for four weeks. Cardiac function, anthropometric, and systemic biochemical parameters were monitored, as well as the histology of the heart and molecular markers of regeneration, metabolism, and oxidative stress. Data showed that an RSV bread diet decreased the polydipsia and body weight loss observed in the early stages of the disease. At the cardiac level, an RSV bread diet diminished fibrosis but did not counteract the dysfunction and metabolic changes seen in fructose-fed STZ-injected rats.
Collapse
Affiliation(s)
- Andreia F R Silva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rita Silva-Reis
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Paula A Oliveira
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro UTAD, 5000-801 Vila Real, Portugal
| | - Ana I Faustino-Rocha
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Department of Zootechnics, Comprehensive Health Research Center, School of Sciences and Technology, University of Évora, 7004-516 Évora, Portugal
| | - Maria de Lurdes Pinto
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro UTAD, 5000-801 Vila Real, Portugal
| | - Manuel A Coimbra
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Artur M S Silva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Susana M Cardoso
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
6
|
Nordmeyer S, Kraus M, Ziehm M, Kirchner M, Schafstedde M, Kelm M, Niquet S, Stephen MM, Baczko I, Knosalla C, Schapranow MP, Dittmar G, Gotthardt M, Falcke M, Regitz-Zagrosek V, Kuehne T, Mertins P. Disease- and sex-specific differences in patients with heart valve disease: a proteome study. Life Sci Alliance 2023; 6:e202201411. [PMID: 36627164 PMCID: PMC9834574 DOI: 10.26508/lsa.202201411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Pressure overload in patients with aortic valve stenosis and volume overload in mitral valve regurgitation trigger specific forms of cardiac remodeling; however, little is known about similarities and differences in myocardial proteome regulation. We performed proteome profiling of 75 human left ventricular myocardial biopsies (aortic stenosis = 41, mitral regurgitation = 17, and controls = 17) using high-resolution tandem mass spectrometry next to clinical and hemodynamic parameter acquisition. In patients of both disease groups, proteins related to ECM and cytoskeleton were more abundant, whereas those related to energy metabolism and proteostasis were less abundant compared with controls. In addition, disease group-specific and sex-specific differences have been observed. Male patients with aortic stenosis showed more proteins related to fibrosis and less to energy metabolism, whereas female patients showed strong reduction in proteostasis-related proteins. Clinical imaging was in line with proteomic findings, showing elevation of fibrosis in both patient groups and sex differences. Disease- and sex-specific proteomic profiles provide insight into cardiac remodeling in patients with heart valve disease and might help improve the understanding of molecular mechanisms and the development of individualized treatment strategies.
Collapse
Affiliation(s)
- Sarah Nordmeyer
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Institute for Cardiovascular Computer-Assisted Medicine, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Congenital Heart Disease - Pediatric Cardiology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Milena Kraus
- Hasso Plattner Institute for Digital Engineering, Digital Health Center, University of Potsdam, Potsdam, Germany
| | - Matthias Ziehm
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Proteomics Platform, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marieluise Kirchner
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Proteomics Platform, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marie Schafstedde
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Institute for Cardiovascular Computer-Assisted Medicine, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Congenital Heart Disease - Pediatric Cardiology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Kelm
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Institute for Cardiovascular Computer-Assisted Medicine, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Congenital Heart Disease - Pediatric Cardiology, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sylvia Niquet
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Proteomics Platform, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mariet Mathew Stephen
- Hasso Plattner Institute for Digital Engineering, Digital Health Center, University of Potsdam, Potsdam, Germany
| | - Istvan Baczko
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| | - Christoph Knosalla
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart, Department of Cardiothoracic and Vascular Surgery, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Matthieu-P Schapranow
- Hasso Plattner Institute for Digital Engineering, Digital Health Center, University of Potsdam, Potsdam, Germany
| | - Gunnar Dittmar
- Proteomics of Cellular Signaling, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Michael Gotthardt
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Neuromuscular and Cardiovascular Cell Biology, Berlin, Germany
| | - Martin Falcke
- Max Delbrück Center for Molecular Medicine, Mathematical Cell Physiology, Berlin, Germany
| | - Vera Regitz-Zagrosek
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Cardiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Titus Kuehne
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Institute for Cardiovascular Computer-Assisted Medicine, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Congenital Heart Disease - Pediatric Cardiology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp Mertins
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Proteomics Platform, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
7
|
Sun G, Su W, Bao J, Teng T, Song X, Wang J, Shi B. Dietary full-fat rice bran prevents the risk of heart ferroptosis and imbalance of energy metabolism induced by prolonged cold stimulation. Food Funct 2023; 14:1530-1544. [PMID: 36655680 DOI: 10.1039/d2fo03673h] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The threat to human health from cold stimulation is increasing due to the frequent occurrence of temperature extremes. It is a challenge for people to resist the negative effects of prolonged cold stimulation on the heart. In this study, we created prolonged cold stimulation pig models to investigate the cardiac energy metabolism and injury during prolonged cold stimulation, and the molecular mechanisms by which dietary supplementation with full-fat rice bran reduces cardiac injury. The results showed that lesions in the morphological structure of the heart were detected under prolonged cold stimulation. At the same time, dystrophin was downregulated under the effect of prolonged cold stimulation. Cardiac fatty acid transport and utilization were promoted, and oxidative stress was increased under prolonged cold stimulation. It also increased MDA content and decreased T-AOC level in the heart, while promoting the mRNA expression of Nrf2 and NQO1, as well as the protein content of Nrf2 and HO-1. Prolonged cold stimulation induced mitochondrial lesions, mitochondrial fusion, and mitophagy in the heart. Prolonged cold stimulation promoted the mRNA expression of PTGS2, TLR4, MyD88, NLRP3, and IL-1β; and protein expression of PTGS2, NLRP3, and mature-IL-1β. GCH1 and FtH inhibited by prolonged cold stimulation caused the activation of heart ferroptosis. In addition, dietary supplementation with full-fat rice bran improved oxidative stress in the heart and inhibited mitophagy, ferroptosis, and pyroptosis. In conclusion, prolonged cold stimulation heightens the risk of cardiac ferroptosis and imbalance of energy metabolism, whereas dietary supplementation with full-fat rice bran mitigates the adverse effects of prolonged cold stimulation on the heart.
Collapse
Affiliation(s)
- Guodong Sun
- School of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| | - Wei Su
- School of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| | - Jiaxin Bao
- School of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| | - Teng Teng
- School of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| | - Xin Song
- School of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| | - Jiawei Wang
- School of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| | - Baoming Shi
- School of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
8
|
Long-Chain and Medium-Chain Fatty Acids in Energy Metabolism of Murine Kidney Mitochondria. Int J Mol Sci 2022; 24:ijms24010379. [PMID: 36613826 PMCID: PMC9820327 DOI: 10.3390/ijms24010379] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Scientists have long established that fatty acids are the primary substrates for kidney mitochondria. However, to date we still do not know how long-chain and middle-chain fatty acids are oxidized at the mitochondrial level. Our previous research has shown that mitochondria from the heart, brain, and kidney oxidize palmitoylcarnitine at a high rate only in the presence of succinate, glutamate, or pyruvate. In this paper, we report properties of the isolated kidney mitochondria and how malate and succinate affect the oxidation of C16 and C8 acylcarnitines. The isolated kidney mitochondria contain very few endogenous substrates and require malate to oxidize pyruvate, glutamate, and C16 or C8 acylcarnitines. We discovered that with 10 µM of C16 or C8 acylcarnitines, low concentrations of malate (0.2 mM) or succinate (0.5 mM) enhance the States 4 and 3 respiratory rates several times. The highest respiration rates were observed with C16 or C8 acylcarnitines and 5 mM succinate mixtures. Results show that kidney mitochondria, unlike the heart and brain mitochondria, lack the intrinsic inhibition of succinate dehydrogenase. Additionally, results show that the oxidation of fatty acid by the small respirasome's supercomplex generates a high level of CoQH2, and this makes SDH in the presence of succinate reverse the flow of electrons from CoQH2 to reduce fumarate to succinate. Finally, we report evidence that succinate dehydrogenase is a key mitochondrial enzyme that allows fast oxidation of fatty acids and turns the TCA cycle function from the catabolic to the anabolic and anaplerotic metabolic pathways.
Collapse
|
9
|
Oka SI, Sreedevi K, Shankar TS, Yedla S, Arowa S, James A, Stone KG, Olmos K, Sabry AD, Horiuchi A, Cawley KM, O’very SA, Tong M, Byun J, Xu X, Kashyap S, Mourad Y, Vehra O, Calder D, Lunde T, Liu T, Li H, Mashchek JA, Cox J, Saijoh Y, Drakos SG, Warren JS. PERM1 regulates energy metabolism in the heart via ERRα/PGC-1α axis. Front Cardiovasc Med 2022; 9:1033457. [PMID: 36419485 PMCID: PMC9676655 DOI: 10.3389/fcvm.2022.1033457] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022] Open
Abstract
Aims PERM1 is a striated muscle-specific regulator of mitochondrial bioenergetics. We previously demonstrated that PERM1 is downregulated in the failing heart and that PERM1 positively regulates metabolic genes known as targets of the transcription factor ERRα and its coactivator PGC-1α in cultured cardiomyocytes. The aims of this study were to determine the effect of loss of PERM1 on cardiac function and energetics using newly generated Perm1-knockout (Perm1 -/-) mice and to investigate the molecular mechanisms of its transcriptional control. Methods and results Echocardiography showed that ejection fraction and fractional shortening were lower in Perm1 -/- mice than in wild-type mice (both p < 0.05), and the phosphocreatine-to-ATP ratio was decreased in Perm1 -/- hearts (p < 0.05), indicating reduced contractile function and energy reserves of the heart. Integrated proteomic and metabolomic analyses revealed downregulation of oxidative phosphorylation and upregulation of glycolysis and polyol pathways in Perm1 -/- hearts. To examine whether PERM1 regulates energy metabolism through ERRα, we performed co-immunoprecipitation assays, which showed that PERM1 bound to ERRα in cardiomyocytes and the mouse heart. DNA binding and reporter gene assays showed that PERM1 was localized to and activated the ERR target promoters partially through ERRα. Mass spectrometry-based screening in cardiomyocytes identified BAG6 and KANK2 as potential PERM1's binding partners in transcriptional regulation. Mammalian one-hybrid assay, in which PERM1 was fused to Gal4 DNA binding domain, showed that the recruitment of PERM1 to a gene promoter was sufficient to activate transcription, which was blunted by silencing of either PGC-1α, BAG6, or KANK2. Conclusion This study demonstrates that PERM1 is an essential regulator of cardiac energetics and function and that PERM1 is a novel transcriptional coactivator in the ERRα/PGC-1α axis that functionally interacts with BAG6 and KANK2.
Collapse
Affiliation(s)
- Shin-ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Karthi Sreedevi
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Thirupura S. Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Shreya Yedla
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Sumaita Arowa
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Amina James
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Kathryn G. Stone
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Katia Olmos
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
| | - Amira D. Sabry
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Amanda Horiuchi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Keiko M. Cawley
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Sean A. O’very
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Mingming Tong
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Jaemin Byun
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Sanchita Kashyap
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Youssef Mourad
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Omair Vehra
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Dallen Calder
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Ty Lunde
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Tong Liu
- Department of Microbiology, Biochemistry, and Molecular Genetics, Center for Advanced Proteomics Research, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, NJ, United States
| | - Hong Li
- Department of Microbiology, Biochemistry, and Molecular Genetics, Center for Advanced Proteomics Research, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, NJ, United States
| | - J. Alan Mashchek
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, United States
| | - James Cox
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, United States
- Department of Biochemistry, University of Utah, Salt Lake City, UT, United States
| | - Yukio Saijoh
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Stavros G. Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Junco S. Warren
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, United States
- Department of Human Nutrition, Food and Exercise, Virginia Tech, Blacksburg, VA, United States
- Division of Developmental Genetics, Institute of Resource Developmental and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
10
|
Aggarwal R, Potel KN, McFalls EO, Butterick TA, Kelly RF. Novel Therapeutic Approaches Enhance PGC1-alpha to Reduce Oxidant Stress-Inflammatory Signaling and Improve Functional Recovery in Hibernating Myocardium. Antioxidants (Basel) 2022; 11:2155. [PMID: 36358527 PMCID: PMC9686496 DOI: 10.3390/antiox11112155] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/29/2022] [Accepted: 10/30/2022] [Indexed: 09/02/2023] Open
Abstract
Ischemic heart disease affects millions of people around the world. Current treatment options, including coronary artery bypass grafting, do not result in full functional recovery, highlighting the need for novel adjunctive therapeutic approaches. Hibernation describes the myocardial response to prolonged ischemia and involves a set of complex cytoprotective metabolic and functional adaptations. PGC1-alpha, a key regulator of mitochondrial energy metabolism and inhibitor of oxidant-stress-inflammatory signaling, is known to be downregulated in hibernating myocardium. PGC1-alpha is a critical component of cellular stress responses and links cellular metabolism with inflammation in the ischemic heart. While beneficial in the acute setting, a chronic state of hibernation can be associated with self-perpetuating oxidant stress-inflammatory signaling which leads to tissue injury. It is likely that incomplete functional recovery following revascularization of chronically ischemic myocardium is due to persistence of metabolic changes as well as prooxidant and proinflammatory signaling. Enhancement of PGC1-alpha signaling has been proposed as a possible way to improve functional recovery in patients with ischemic heart disease. Adjunctive mesenchymal stem cell therapy has been shown to induce PGC1-alpha signaling in hibernating myocardium and could help improve clinical outcomes for patients undergoing bypass surgery.
Collapse
Affiliation(s)
- Rishav Aggarwal
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Koray N. Potel
- School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK
| | - Edward O. McFalls
- Division of Cardiology, Richmond VA Medical Center, Richmond, VA 23249-4915, USA
| | - Tammy A. Butterick
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Research, Center for Veterans Research and Education, Minneapolis, MN 55417, USA
| | - Rosemary F. Kelly
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
11
|
Afolabi OA, Alabi BA, Oluranti O. Diet-induced insulin resistance altered cardiac GLUT4 and FATP/CD36 expression in rats. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2022. [DOI: 10.1186/s43088-022-00312-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Altered substrate transport protein expression is central to the effect of insulin resistance on cardiac metabolism. The present study was thus designed to investigate the comparative effects of high fat, high sucrose and salt-induced IR on cardiac expression of fatty acid transporter (FATP) and glucose transporter (GLUT4) in rats.
Results
Rats fed with high fat, high sucrose and salt diets developed impaired glucose tolerance (p > 0.05) and hyperinsulinemia (p < 0.05) compared with control group. Myocardial glucose transporter expression was significantly increased (p < 0.001 for salt-induced IR; p < 0.01 for sucrose-induced IR; p < 0.01 for fat-induced IR) across all IR groups compared with control. Fatty acid transporter expression was also increased (p < 0.001) in high salt diet-induced IR rats, and high fat diet-induced IR rats (p < 0.05).
Conclusions
Our results demonstrate that salt and not caloric excess has a potential role in IR alteration of myocardial substrate transport protein expression in the rat.
Collapse
|
12
|
Beneficial Effects of RNS60 in Cardiac Ischemic Injury. Curr Issues Mol Biol 2022; 44:4877-4887. [PMID: 36286046 PMCID: PMC9600597 DOI: 10.3390/cimb44100331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
RNS60 is a physically modified saline solution hypothesized to contain oxygen nanobubbles. It has been reported to reduce ischemia/reperfusion injury in a pig model of acute myocardial infarction. We investigated the effects of RNS60 during cardiac hypoxia in mice and as an additive to cardioplegic solution in rat hearts. ApoE−/−LDLr−/− mice were treated by intravenous injection of RNS60 or saline as a control while monitoring the ECG and post-hypoxic serum release of troponin T and creatine kinase activity. Hearts infused with Custodiol containing 10% RNS60 or saline as the control were subjected to 4 h of 4 °C preservation, followed by an assessment of myocardial metabolites, purine release, and mechanical function. RNS60 attenuated changes in the ECG STU area during hypoxia, while the troponin T concentration and creatine kinase activity were significantly higher in the serum of the controls. During reperfusion after 4 h of cold ischemia, the Custodiol/RNS60-treated hearts had about 30% lower LVEDP and better dp/dtmax and dp/dtmin together with a decreased release of purine catabolites vs. the controls. The myocardial ATP, total adenine nucleotides, and phosphocreatine concentrations were higher in the RNS60-treated hearts. This study indicates that RNS60 enhances cardioprotection in experimental myocardial hypoxia and under conditions of cardioplegic arrest. Improved cardiac energetics are involved in the protective effect, but complete elucidation of the mechanism requires further study.
Collapse
|
13
|
Scott SR, Singh K, Yu Q, Sen CK, Wang M. Sex as Biological Variable in Cardiac Mitochondrial Bioenergetic Responses to Acute Stress. Int J Mol Sci 2022; 23:9312. [PMID: 36012574 PMCID: PMC9409303 DOI: 10.3390/ijms23169312] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiac dysfunction/damage following trauma, shock, sepsis, and ischemia impacts clinical outcomes. Acute inflammation and oxidative stress triggered by these injuries impair mitochondria, which are critical to maintaining cardiac function. Despite sex dimorphisms in consequences of these injuries, it is unclear whether mitochondrial bioenergetic responses to inflammation/oxidative stress are sex-dependent. We hypothesized that sex disparity in mitochondrial bioenergetics following TNFα or H2O2 exposure is responsible for reported sex differences in cardiac damage/dysfunction. Methods and Results: Cardiomyocytes isolated from age-matched adult male and female mice were subjected to 1 h TNFα or H2O2 challenge, followed by detection of mitochondrial respiration capacity using the Seahorse XF96 Cell Mito Stress Test. Mitochondrial membrane potential (ΔΨm) was analyzed using JC-1 in TNFα-challenged cardiomyocytes. We found that cardiomyocytes isolated from female mice displayed a better mitochondrial bioenergetic response to TNFα or H2O2 than those isolated from male mice did. TNFα decreased ΔΨm in cardiomyocytes isolated from males but not from females. 17β-estradiol (E2) treatment improved mitochondrial metabolic function in cardiomyocytes from male mice subjected to TNFα or H2O2 treatment. Conclusions: Cardiomyocyte mitochondria from female mice were more resistant to acute stress than those from males. The female sex hormone E2 treatment protected cardiac mitochondria against acute inflammatory and oxidative stress.
Collapse
Affiliation(s)
- Susan R. Scott
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kanhaiya Singh
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Qing Yu
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chandan K. Sen
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Meijing Wang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
14
|
An Energy Metabolism Study on the Efficacy of Naoxintong Capsules against Myocardial Infarction in a Rat Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3712500. [PMID: 35915610 PMCID: PMC9338863 DOI: 10.1155/2022/3712500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/15/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022]
Abstract
Background In myocardial ischemia, optimizing the myocardial metabolic phenotype to improve cardiac function is critical. Naoxintong capsules (NXT) are widely prescribed in Chinese medicine for the treatment of cerebrovascular and cardiovascular diseases. Methods In this study, a rat model of myocardial infarction was established by ligation of the left anterior descending coronary artery. The structure and function of the heart were evaluated using echocardiography. The pathological changes of the rat myocardium and the myocardial volume collagen fraction (CVF) were examined using hematoxylin-eosin (HE) and Masson's trichrome staining (Masson). The expression of TNF-α and IL-6 were detected by immunohistochemistry. The level of cTnT was also measured to evaluate myocardial injury. In order to study the changes in energy metabolism in myocardial infarction and the effects of NXT, a targeted analysis method for detecting the 29 energy metabolites in cardiac muscle tissue was developed based on UPLC-QQQ-MS. Western blotting was used to detect the expression of proteins related to energy metabolism in myocardia. Results In the rat model of myocardial infarction, NXT showed obvious effects, such as improving heart function and increasing LVEF and LVFS. HE staining, Masson staining, and immunohistochemical results revealed that NXT decreased inflammatory infiltration, improved myocardial fibrosis, and reduced infarct size. In addition, NXT significantly reduced the level of serum cTnT. The levels of the 29 energy metabolites in cardiac muscle tissue were analyzed using a newly developed targeted analysis method. Compared to the sham group, the levels of 17 metabolites from different energy metabolic pathways, including four compounds in glycolysis metabolism, four compounds in TCA cycle, three compounds in oxidative phosphorylation, four compounds in purine metabolism, and two compounds in glutathione metabolism, displayed obvious changes induced by myocardial ischemia. Expressions of SIRT1, PGC-1α, and ATP5D proteins related to energy metabolism were decreased after myocardial infarction. These perturbations could all be reversed by NXT intervention, suggesting that the therapeutic effects of NXT were partially due to interferences with energy metabolisms. Conclusion This study provides a useful approach for investigating the mechanism of myocardial infarction and evaluating the efficacy of NXT from energy metabolism.
Collapse
|
15
|
Zhang Y, Zhang H, Yang Z, Zhang XH, Miao Q, Li M, Zhai TY, Zheng B, Wen JK. miR-155 down-regulation protects the heart from hypoxic damage by activating fructose metabolism in cardiac fibroblasts. J Adv Res 2022; 39:103-117. [PMID: 35777901 PMCID: PMC9263644 DOI: 10.1016/j.jare.2021.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/15/2021] [Accepted: 10/17/2021] [Indexed: 12/09/2022] Open
Abstract
INTRODUCTION Hypoxia-inducible factor (HIF)1α has been shown to be activated and induces a glycolytic shift under hypoxic condition, however, little attention was paid to the role of HIF1α-actuated fructolysis in hypoxia-induced heart injury. OBJECTIVES In this study, we aim to explore the molecular mechanisms of miR-155-mediated fructose metabolism in hypoxic cardiac fibroblasts (CFs). METHODS Immunostaining, western blot and quantitative real-time reverse transcription PCR (qRT-PCR) were performed to detect the expression of glucose transporter 5 (GLUT5), ketohexokinase (KHK)-A and KHK-C in miR-155-/- and miR-155wt CFs under normoxia or hypoxia. A microarray analysis of circRNAs was performed to identify circHIF1α. Then CoIP, RIP and mass spectrometry analysis were performed and identified SKIV2L2 (MTR4) and transformer 2 alpha (TRA2A), a member of the transformer 2 homolog family. pAd-SKIV2L2 was administrated after coronary artery ligation to investigate whether SKIV2L2 can provide a protective effect on the infarcted heart. RESULTS When both miR-155-/- and miR-155wt CFs were exposed to hypoxia for 24 h, these two cells exhibited an increased glycolysis and decreased glycogen synthesis, and the expression of KHK-A and KHK-C, the central fructose-metabolizing enzyme, was upregulated. Mechanistically, miR-155 deletion in CFs enhanced SKIV2L2 expression and its interaction with TRA2A, which suppresses the alternative splicing of HIF1α pre-mRNA to form circHIF1α, and then decreased circHIF1α contributed to the activation of fructose metabolism through increasing the production of the KHK-C isoform. Finally, exogenous delivery of SKIV2L2 reduced myocardial damage in the infarcted heart. CONCLUSION In this study, we demonstrated that miR-155 deletion facilitates the activation of fructose metabolism in hypoxic CFs through regulating alternative splicing of HIF1α pre-mRNA and thus circHIF1ɑ formation.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Hong Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Department of Urology, Second Hospital of Hebei Medical University 050000, China
| | - Zhan Yang
- Department of Urology, Second Hospital of Hebei Medical University 050000, China
| | - Xin-Hua Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Qing Miao
- Department of Cardiovascular Medicine, Second Hospital of Hebei Medical University, 050000, China
| | - Min Li
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Tian-Ying Zhai
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China.
| | - Jin-Kun Wen
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
16
|
Ness HO, Ljones K, Gjelsvik RH, Tjønna AE, Malmo V, Nilsen HO, Hollekim-Strand SM, Dalen H, Høydal MA. Acute effects of high intensity training on cardiac function: a pilot study comparing subjects with type 2 diabetes to healthy controls. Sci Rep 2022; 12:8239. [PMID: 35581305 PMCID: PMC9114004 DOI: 10.1038/s41598-022-12375-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/09/2022] [Indexed: 01/15/2023] Open
Abstract
This study evaluated acute cardiac stress after a high-intensity interval training session in patients with type 2 diabetes (T2D) versus healthy controls. High intensity aerobic exercise was performed by 4 × 4-min intervals (90-95% of maximal heart rate), followed by a ramp protocol to peak oxygen uptake. Echocardiography was performed before and 30 min after exercise. Holter electrocardiography monitored heart rhythms 24 h before, during, and 24 h after the exercise. Left atrial end-systolic volume, peak early diastolic mitral annular velocity, and the ratio of peak early to late diastolic mitral inflow velocity were reduced by approximately 18%, 15%, and 31%, respectively, after exercise across groups. Left ventricular end-diastolic wall thickness was the only echo parameter that significantly differed between groups in response to exercise. The T2D group had a rate of supraventricular extrasystoles per hour that was 265% greater than that of the controls before exercise, which remained higher after exercise. A single exhaustive exercise session impaired left ventricular diastolic function in both groups. The findings also indicated impaired right ventricular function in patients with T2D after exercise.ClinicalTrials.gov Identifier: NCT02998008.
Collapse
Affiliation(s)
- Henning O. Ness
- grid.5947.f0000 0001 1516 2393Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Prinsesse Kristinas gt. 3, Akutten og Hjerte-lunge-senteret, 3.etg, 7030 Trondheim, Norway
| | - Kristine Ljones
- grid.5947.f0000 0001 1516 2393Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Prinsesse Kristinas gt. 3, Akutten og Hjerte-lunge-senteret, 3.etg, 7030 Trondheim, Norway
| | - Randi H. Gjelsvik
- grid.5947.f0000 0001 1516 2393Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Prinsesse Kristinas gt. 3, Akutten og Hjerte-lunge-senteret, 3.etg, 7030 Trondheim, Norway
| | - Arnt Erik Tjønna
- grid.5947.f0000 0001 1516 2393Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Prinsesse Kristinas gt. 3, Akutten og Hjerte-lunge-senteret, 3.etg, 7030 Trondheim, Norway
| | - Vegard Malmo
- grid.5947.f0000 0001 1516 2393Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Prinsesse Kristinas gt. 3, Akutten og Hjerte-lunge-senteret, 3.etg, 7030 Trondheim, Norway ,grid.52522.320000 0004 0627 3560Clinic of Cardiology, St. Olavs University Hospital, Trondheim, Norway
| | - Hans Olav Nilsen
- grid.5947.f0000 0001 1516 2393Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Prinsesse Kristinas gt. 3, Akutten og Hjerte-lunge-senteret, 3.etg, 7030 Trondheim, Norway ,grid.52522.320000 0004 0627 3560Clinic of Cardiology, St. Olavs University Hospital, Trondheim, Norway
| | - Siri Marte Hollekim-Strand
- grid.5947.f0000 0001 1516 2393Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Håvard Dalen
- grid.5947.f0000 0001 1516 2393Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Prinsesse Kristinas gt. 3, Akutten og Hjerte-lunge-senteret, 3.etg, 7030 Trondheim, Norway ,grid.52522.320000 0004 0627 3560Clinic of Cardiology, St. Olavs University Hospital, Trondheim, Norway ,grid.414625.00000 0004 0627 3093Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Morten Andre Høydal
- grid.5947.f0000 0001 1516 2393Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Prinsesse Kristinas gt. 3, Akutten og Hjerte-lunge-senteret, 3.etg, 7030 Trondheim, Norway
| |
Collapse
|
17
|
Isei MO, Stevens D, Kamunde C. Copper modulates heart mitochondrial H 2O 2 emission differently during fatty acid and pyruvate oxidation. Comp Biochem Physiol C Toxicol Pharmacol 2022; 254:109267. [PMID: 35026399 DOI: 10.1016/j.cbpc.2022.109267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 12/28/2022]
Abstract
Although the preferred cardiac metabolic fuels are fatty acids, glucose metabolism also plays an important role. However, irrespective of substrate type, energy generation results in mitochondrial reactive oxygen species (ROS) formation. To determine if the preference of fat over carbohydrates predisposes cardiomyocytes to oxidant production, we measured total and site-specific H2O2 emission in heart mitochondria oxidizing palmitoylcarnitine or pyruvate during copper (Cu) exposure. H2O2 emission was higher during oxidation of palmitoylcarnitine compared with pyruvate. Moreover, the bulk of the H2O2 emitted during palmitoylcarnitine oxidation originated from the outer ubiquinone binding site of complex III (site IIIQo) and the flavin site of electron transfer flavoprotein (site EF). We found no evidence of ROS production from complex I ubiquinone-binding site (site IQ) by reverse electron transport during oxidation of palmitoylcarnitine. Pyruvate oxidation also drove H2O2 emission primarily from sites IIIQo; however, the flavin sites of pyruvate dehydrogenase (site PF) and complex II (site IIF) contributed substantially. The effect of Cu depended on substrate and redox site, with effects at sites OF and IIIQo being more pronounced in mitochondria oxidizing pyruvate compared with palmitoylcarnitine. Cu imposed a concentration-saturable effect at site PF but concentration-dependently stimulated H2O2 emission at site EF. The substrate-dependent differences in H2O2 emission and effects of Cu suggest that fuel type and points of entry of electrons into the mitochondrial electron transport system determine the mitochondrial ROS production rate. Importantly, knowledge of sites of mitochondrial ROS production is crucial to the understanding of cardiac dysfunction associated with impaired substrate metabolism.
Collapse
Affiliation(s)
- Michael O Isei
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Don Stevens
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Collins Kamunde
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada.
| |
Collapse
|
18
|
Radhakrishnan J, Baetiong A, Gazmuri RJ. Enhanced Oxygen Utilization Efficiency With Concomitant Activation of AMPK-TBC1D1 Signaling Nexus in Cyclophilin-D Conditional Knockout Mice. Front Physiol 2021; 12:756659. [PMID: 34955879 PMCID: PMC8692870 DOI: 10.3389/fphys.2021.756659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/12/2021] [Indexed: 12/02/2022] Open
Abstract
We have previously reported in HEK 293 T cells and in constitutive cyclophilin-D (Cyp-D) knockout (KO) mice that Cyp-D ablation downregulates oxygen consumption (VO2) and triggers an adaptive response that manifest in higher exercise endurance with less VO2. This adaptive response involves a metabolic switch toward preferential utilization of glucose via AMPK-TBC1D1 signaling nexus. We now investigated whether a similar response could be triggered in mice after acute ablation of Cyp-D using tamoxifen-induced ROSA26-Cre-mediated (i.e., conditional KO, CKO) by subjecting them to treadmill exercise involving five running sessions. At their first treadmill running session, CKO mice and controls had comparable VO2 (208.4 ± 17.9 vs. 209.1 ± 16.8 ml/kg min−1), VCO2 (183.6 ± 17.2 vs. 184.8 ± 16.9 ml/kg min−1), and RER (0.88 ± 0.043 vs. 0.88 ± 0.042). With subsequent sessions, CKO mice displayed more prominent reduction in VO2 (genotype & session interaction p = 0.000) with less prominent reduction in VCO2 resulting in significantly increased RER (genotype and session interaction p = 0.013). The increase in RER was consistent with preferential utilization of glucose as respiratory substrate (4.6 ± 0.8 vs. 4.0 ± 0.9 mg/min, p = 0.003). CKO mice also performed a significantly higher treadmill work for given VO2 expressed as a power/VO2 ratio (7.4 ± 0.2 × 10−3 vs. 6.7 ± 0.2 10−3 ratio, p = 0.025). Analysis of CKO skeletal muscle tissue after completion of five treadmill running sessions showed enhanced AMPK activation (0.669 ± 0.06 vs. 0.409 ± 0.11 pAMPK/β-tubulin ratio, p = 0.005) and TBC1D1 inactivation (0.877 ± 0.16 vs. 0.565 ± 0.09 pTBC1D1/β-tubulin ratio, p < 0.05) accompanied by increased glucose transporter-4 levels consistent with activation of the AMPK-TBC1D1 signaling nexus enabling increased glucose utilization. Taken together, our study demonstrates that like constitutive Cyp-D ablation, acute Cyp-D ablation also induces a state of increased O2 utilization efficiency, paving the way for exploring the use of pharmacological approach to elicit the same response, which could be beneficial under O2 limiting conditions.
Collapse
Affiliation(s)
- Jeejabai Radhakrishnan
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Department of Clinical Sciences, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Alvin Baetiong
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Raúl J Gazmuri
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Department of Clinical Sciences, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Captain James A. Lovell Federal Health Care Center, North Chicago, IL, United States
| |
Collapse
|
19
|
Al-Shamasi AA, Elkaffash R, Mohamed M, Rayan M, Al-Khater D, Gadeau AP, Ahmed R, Hasan A, Eldassouki H, Yalcin HC, Abdul-Ghani M, Mraiche F. Crosstalk between Sodium-Glucose Cotransporter Inhibitors and Sodium-Hydrogen Exchanger 1 and 3 in Cardiometabolic Diseases. Int J Mol Sci 2021; 22:12677. [PMID: 34884494 PMCID: PMC8657861 DOI: 10.3390/ijms222312677] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 12/14/2022] Open
Abstract
Abnormality in glucose homeostasis due to hyperglycemia or insulin resistance is the hallmark of type 2 diabetes mellitus (T2DM). These metabolic abnormalities in T2DM lead to cellular dysfunction and the development of diabetic cardiomyopathy leading to heart failure. New antihyperglycemic agents including glucagon-like peptide-1 receptor agonists and the sodium-glucose cotransporter-2 inhibitors (SGLT2i) have been shown to attenuate endothelial dysfunction at the cellular level. In addition, they improved cardiovascular safety by exhibiting cardioprotective effects. The mechanism by which these drugs exert their cardioprotective effects is unknown, although recent studies have shown that cardiovascular homeostasis occurs through the interplay of the sodium-hydrogen exchangers (NHE), specifically NHE1 and NHE3, with SGLT2i. Another theoretical explanation for the cardioprotective effects of SGLT2i is through natriuresis by the kidney. This theory highlights the possible involvement of renal NHE transporters in the management of heart failure. This review outlines the possible mechanisms responsible for causing diabetic cardiomyopathy and discusses the interaction between NHE and SGLT2i in cardiovascular diseases.
Collapse
Affiliation(s)
- Al-Anood Al-Shamasi
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Rozina Elkaffash
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Meram Mohamed
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Menatallah Rayan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Dhabya Al-Khater
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Alain-Pierre Gadeau
- INSERM, Biology of Cardiovascular Disease, University of Bordeaux, U1034 Pessac, France;
| | - Rashid Ahmed
- Department of Mechanical and Chemical Engineering, College of Engineering, Qatar University, Doha P.O. Box 2713, Qatar; (R.A.); (A.H.)
- Biomedical Research Centre (BRC), Qatar University, Doha P.O. Box 2713, Qatar;
| | - Anwarul Hasan
- Department of Mechanical and Chemical Engineering, College of Engineering, Qatar University, Doha P.O. Box 2713, Qatar; (R.A.); (A.H.)
- Biomedical Research Centre (BRC), Qatar University, Doha P.O. Box 2713, Qatar;
| | - Hussein Eldassouki
- College of Kinesiology, University of Saskatchewan, Saskatoon, SK S7N 5B5, Canada;
| | | | - Muhammad Abdul-Ghani
- Division of Diabetes, University of Texas Health Science Center at San Antonio, Floyd Curl Drive, San Antonio, TX 7703, USA;
| | - Fatima Mraiche
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (A.-A.A.-S.); (R.E.); (M.M.); (M.R.); (D.A.-K.)
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
20
|
Costa Silva RCM, Correa LHT. Heme Oxygenase 1 in Vertebrates: Friend and Foe. Cell Biochem Biophys 2021; 80:97-113. [PMID: 34800278 DOI: 10.1007/s12013-021-01047-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/07/2021] [Indexed: 10/19/2022]
Abstract
HO-1 is the inducible form of the enzyme heme-oxygenase. HO-1 catalyzes heme breakdown, reducing the levels of this important oxidant molecule and generating antioxidant, anti-inflammatory, and anti-apoptotic byproducts. Thus, HO-1 has been described as an important stress response mechanism during both physiologic and pathological processes. Interestingly, some findings are demonstrating that uncontrolled levels of HO-1 byproducts can be associated with cell death and tissue destruction as well. Furthermore, HO-1 can be located in the nucleus, influencing gene transcription, cellular proliferation, and DNA repair. Here, we will discuss several studies that approach HO-1 effects as a protective or detrimental mechanism in different pathological conditions. In this sense, as the major organs of vertebrates will deal specifically with distinct types of stresses, we discuss the HO-1 role in each of them, exposing the contradictions associated with HO-1 expression after different insults and circumstances.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Leonardo Holanda Travassos Correa
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
21
|
Metformin Prevents Low-dose Isoproterenol-induced Cardiac Dilatation and Systolic Dysfunction in Male Sprague-Dawley Rats. J Cardiovasc Pharmacol 2021; 79:289-295. [PMID: 34775423 DOI: 10.1097/fjc.0000000000001172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 10/23/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Myocardial metabolic abnormalities are well recognized alterations in chronic heart failure, effects that may contribute to progressive cardiac dysfunction. However, whether metabolic alterations in-part mediate their deleterious effects by modifying the chronic impact of excess low dose sympathetic stimulation on cardiac chamber dilatation, is uncertain. We therefore aimed to determine the effect of metformin administration on cardiac function and mitochondrial architectural changes in a rat model of chronic sympathetic-induced left ventricular (LV) remodeling and systolic dysfunction (daily subcutaneous isoproterenol [ISO] injection at a low-dose of 0.02 mg/kg for 7 months). Echocardiography was used to assess in vivo LV dimensions and function, and mitochondrial and myofibril arrangement was assessed using transmission electron microscopy. 7 months of low-dose ISO administration increased left ventricular diastolic diameter (in mm) (CONT: 7.29±0.19 vs. ISO: 8.76±0.21; p=0.001), an effect that was attenuated by metformin (ISO+MET: 7.63±0.29 vs ISO: p=0.001) administration. Similarly, ISO increased LV end systolic diameter (CONT: 4.43±0.16 vs ISO: 5.49±0.16: p<0.0001) an effect prevented by metformin (ISO+MET: 4.04±0.25 vs. ISO: p<0.0001). Moreover, chronic ISO administration reduced LV endocardial fractional shortening (p=0.0001), midwall fractional shortening (p=0.0001) and ejection fraction (p=0.0001), effects similarly prevented by metformin administration. Furthermore, changes in mitochondrial arrangement and relative mitochondrial area (CONT: 37.7±2.2 vs. ISO: 28.1±2.9; p=0.05) were produced by ISO administration, effects prevented by metformin. In conclusion, metformin offers cardiac protection against chronic sympathetic-induced LV dilatation and systolic dysfunction. These data support a role for myocardial metabolic changes in mediating LV dilatation and LV dysfunction produced by chronic neurohumoral activation in cardiac disease.
Collapse
|
22
|
Cunha FM, Cidade-Rodrigues C, Elias C, Oliveira D, Bettencourt P, Lourenço P. Glucose variability predicts 6-month mortality in patients hospitalized with acute heart failure. Intern Emerg Med 2021; 16:2121-2128. [PMID: 33818704 DOI: 10.1007/s11739-021-02719-7] [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] [Received: 02/09/2021] [Accepted: 03/17/2021] [Indexed: 12/29/2022]
Abstract
In diabetes mellitus (DM), glycaemic fluctuations associate with higher oxidative stress than sustained chronic hyperglycaemia and glucose variability increases the risk of chronic diabetic complications. Our hypothesis was that higher glucose variability would associate with mortality after an acute heart failure (HF) episode. We retrospectively analysed patients with DM hospitalized with acute HF between 2009 and 2010. Patients with < 2 point-of-care glucose values/day were excluded. Glucose coefficient of variation (GCV) was defined as (glucose standard deviation/mean glucose) × 100. Patients were categorized according GCV ≤ 30.0 and > 30.0%. Follow-up: 6-months. Endpoint: all-cause mortality. A Cox-regression analysis was used to study the association of glucose variability with 6-month mortality. We studied 214 diabetic patients with acute HF, 49.1% male, mean age 76 years. Mean glycaemia during hospitalization was 187 ± 50 mg/dL, hypoglycaemia (< 70 mg/dL) was reported in 21 patients and mean GCV was 28.3 ± 7.6%. Patients with GCV > 30.0% had higher mean glycaemia, more hypoglycaemic episodes and higher HbA1c; they were also more often treated with insulin. Patients were similar concerning age, gender, comorbidities, left ventricular systolic dysfunction and ischemic heart disease. During the 6-month follow-up, 38 (17.8%) patients died. Patients with GCV > 30.0% had a HR of 6-month mortality of 2.21 (95% CI: 1.16-4.21), p = 0.02. This association with more than twofold higher short-term mortality was independent of main confounders. Elevated glycaemic variability in acute HF admissions of patients with DM predicts short-term mortality. Patients with GCV > 30.0% have an independent more than twofold higher risk of 6-month death after an acute HF hospitalization.
Collapse
Affiliation(s)
- Filipe M Cunha
- Endocrinology Department, Centro Hospitalar do Tâmega e Sousa, Avenida Do Hospital Padre Américo 210, Guilhufe, 4564-007, Penafiel, Portugal.
| | - Catarina Cidade-Rodrigues
- Endocrinology Department, Centro Hospitalar do Tâmega e Sousa, Avenida Do Hospital Padre Américo 210, Guilhufe, 4564-007, Penafiel, Portugal
| | - Catarina Elias
- Internal Medicine Department, Centro Hospitalar e Universitário de São João, Porto, Portugal
| | - Diana Oliveira
- Internal Medicine Department, Centro Hospitalar e Universitário de São João, Porto, Portugal
| | - Paulo Bettencourt
- Medicine Faculty, Porto University, Porto, Portugal
- Internal Medicine Department, Hospital CUF Porto, Porto, Portugal
| | - Patrícia Lourenço
- Internal Medicine Department, Centro Hospitalar e Universitário de São João, Porto, Portugal
- Medicine Faculty, Porto University, Porto, Portugal
- Heart Failure Clinic of the Internal Medicine Department, Centro Hospitalar e Universitário de São João, Porto, Portugal
| |
Collapse
|
23
|
Scheiber D, Zweck E, Albermann S, Jelenik T, Spieker M, Bönner F, Horn P, Schultheiss HP, Aleshcheva G, Escher F, Boeken U, Akhyari P, Roden M, Kelm M, Szendroedi J, Westenfeld R. Human myocardial mitochondrial oxidative capacity is impaired in mild acute heart transplant rejection. ESC Heart Fail 2021; 8:4674-4684. [PMID: 34490749 PMCID: PMC8712779 DOI: 10.1002/ehf2.13607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 12/28/2022] Open
Abstract
Aims Acute cellular rejection (ACR) following heart transplantation (HTX) is associated with long‐term graft loss and increased mortality. Disturbed mitochondrial bioenergetics have been identified as pathophysiological drivers in heart failure, but their role in ACR remains unclear. We aimed to prove functional disturbances of myocardial bioenergetics in human heart transplant recipients with mild ACR by assessing myocardial mitochondrial respiration using high‐resolution respirometry, digital image analysis of myocardial inflammatory cell infiltration, and clinical assessment of HTX patients. We hypothesized that (i) mild ACR is associated with impaired myocardial mitochondrial respiration and (ii) myocardial inflammation, systemic oxidative stress, and myocardial oedema relate to impaired mitochondrial respiration and myocardial dysfunction. Methods and results We classified 35 HTX recipients undergoing endomyocardial biopsy according International Society for Heart and Lung Transplantation criteria to have no (0R) or mild (1R) ACR. Additionally, we quantified immune cell infiltration by immunohistochemistry and digital image analysis. We analysed mitochondrial substrate utilization in myocardial fibres by high‐resolution respirometry and performed cardiovascular magnetic resonance (CMR). ACR (1R) was diagnosed in 12 patients (34%), while the remaining 23 patients revealed no signs of ACR (0R). Underlying cardiomyopathies (dilated cardiomyopathy 50% vs. 65%; P = 0.77), comorbidities (type 2 diabetes mellitus: 50% vs. 35%, P = 0.57; chronic kidney disease stage 5: 8% vs. 9%, P > 0.99; arterial hypertension: 59% vs. 30%, P = 0.35), medications (tacrolimus: 100% vs. 91%, P = 0.54; mycophenolate mofetil: 92% vs. 91%, P > 0.99; prednisolone: 92% vs. 96%, P > 0.99) and time post‐transplantation (21.5 ± 26.0 months vs. 29.4 ± 26.4 months, P = 0.40) were similar between groups. Mitochondrial respiration was reduced by 40% in ACR (1R) compared with ACR (0R) (77.8 ± 23.0 vs. 128.0 ± 33.0; P < 0.0001). Quantitative assessment of myocardial CD3+‐lymphocyte infiltration identified ACR (1R) with a cut‐off of >14 CD3+‐lymphocytes/mm2 (100% sensitivity, 82% specificity; P < 0.0001). Myocardial CD3+ infiltration (r = −0.41, P < 0.05), systemic oxidative stress (thiobarbituric acid reactive substances; r = −0.42, P < 0.01) and myocardial oedema depicted by global CMR derived T2 time (r = −0.62, P < 0.01) correlated with lower oxidative capacity and overt cardiac dysfunction (global longitudinal strain; r = −0.63, P < 0.01). Conclusions Mild ACR with inflammatory cell infiltration associates with impaired mitochondrial bioenergetics in cardiomyocytes. Our findings may help to identify novel checkpoints in cardiac immune metabolism as potential therapeutic targets in post‐transplant care.
Collapse
Affiliation(s)
- Daniel Scheiber
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstraße 5, Düsseldorf, 40225, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, München-Neuherberg, Germany
| | - Elric Zweck
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstraße 5, Düsseldorf, 40225, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, München-Neuherberg, Germany
| | - Sophie Albermann
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstraße 5, Düsseldorf, 40225, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, München-Neuherberg, Germany
| | - Tomas Jelenik
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, München-Neuherberg, Germany
| | - Maximilian Spieker
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstraße 5, Düsseldorf, 40225, Germany
| | - Florian Bönner
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstraße 5, Düsseldorf, 40225, Germany
| | - Patrick Horn
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstraße 5, Düsseldorf, 40225, Germany
| | | | - Ganna Aleshcheva
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany
| | - Felicitas Escher
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany
| | - Udo Boeken
- Department of Cardiac Surgery, Heinrich-Heine University, Düsseldorf, Germany
| | - Payam Akhyari
- Department of Cardiac Surgery, Heinrich-Heine University, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, München-Neuherberg, Germany.,Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstraße 5, Düsseldorf, 40225, Germany.,Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), Partner Düsseldorf, München-Neuherberg, Germany.,Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.,Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany.,Institute for Diabetes and Cancer (IDC) & Joint Heidelberg-IDC Translational Diabetes Program, Helmholtz Center Munich, Neuherberg, Germany
| | - Ralf Westenfeld
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Moorenstraße 5, Düsseldorf, 40225, Germany
| |
Collapse
|
24
|
Di Fonso A, Pietrangelo L, D’Onofrio L, Michelucci A, Boncompagni S, Protasi F. Ageing Causes Ultrastructural Modification to Calcium Release Units and Mitochondria in Cardiomyocytes. Int J Mol Sci 2021; 22:8364. [PMID: 34445071 PMCID: PMC8395047 DOI: 10.3390/ijms22168364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/20/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
Ageing is associated with an increase in the incidence of heart failure, even if the existence of a real age-related cardiomyopathy remains controversial. Effective contraction and relaxation of cardiomyocytes depend on efficient production of ATP (handled by mitochondria) and on proper Ca2+ supply to myofibrils during excitation-contraction (EC) coupling (handled by Ca2+ release units, CRUs). Here, we analyzed mitochondria and CRUs in hearts of adult (4 months old) and aged (≥24 months old) mice. Analysis by confocal and electron microscopy (CM and EM, respectively) revealed an age-related loss of proper organization and disposition of both mitochondria and EC coupling units: (a) mitochondria are improperly disposed and often damaged (percentage of severely damaged mitochondria: adults 3.5 ± 1.1%; aged 16.5 ± 3.5%); (b) CRUs that are often misoriented (longitudinal) and/or misplaced from the correct position at the Z line. Immunolabeling with antibodies that mark either the SR or T-tubules indicates that in aged cardiomyocytes the sarcotubular system displays an extensive disarray. This disarray could be in part caused by the decreased expression of Cav-3 and JP-2 detected by western blot (WB), two proteins involved in formation of T-tubules and in docking SR to T-tubules in dyads. By WB analysis, we also detected increased levels of 3-NT in whole hearts homogenates of aged mice, a product of nitration of protein tyrosine residues, recognized as marker of oxidative stress. Finally, a detailed EM analysis of CRUs (formed by association of SR with T-tubules) points to ultrastructural modifications, i.e., a decrease in their frequency (adult: 5.1 ± 0.5; aged: 3.9 ± 0.4 n./50 μm2) and size (adult: 362 ± 40 nm; aged: 254 ± 60 nm). The changes in morphology and disposition of mitochondria and CRUs highlighted by our results may underlie an inefficient supply of Ca2+ ions and ATP to the contractile elements, and possibly contribute to cardiac dysfunction in ageing.
Collapse
Affiliation(s)
- Alessia Di Fonso
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
- DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy
| | - Laura Pietrangelo
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
- DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy
| | - Laura D’Onofrio
- IZSAM, Istituto Zooprofilattico Sperimentale dell’Abruzzo e del Molise G. Caporale of Teramo, 64100 Teramo, Italy;
| | - Antonio Michelucci
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
| | - Simona Boncompagni
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
- DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy
| | - Feliciano Protasi
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
- DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
25
|
Bae J, Paltzer WG, Mahmoud AI. The Role of Metabolism in Heart Failure and Regeneration. Front Cardiovasc Med 2021; 8:702920. [PMID: 34336958 PMCID: PMC8322239 DOI: 10.3389/fcvm.2021.702920] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/23/2021] [Indexed: 12/25/2022] Open
Abstract
Heart failure is the leading cause of death worldwide. The inability of the adult mammalian heart to regenerate following injury results in the development of systolic heart failure. Thus, identifying novel approaches toward regenerating the adult heart has enormous therapeutic potential for adult heart failure. Mitochondrial metabolism is an essential homeostatic process for maintaining growth and survival. The emerging role of mitochondrial metabolism in controlling cell fate and function is beginning to be appreciated. Recent evidence suggests that metabolism controls biological processes including cell proliferation and differentiation, which has profound implications during development and regeneration. The regenerative potential of the mammalian heart is lost by the first week of postnatal development when cardiomyocytes exit the cell cycle and become terminally differentiated. This inability to regenerate following injury is correlated with the metabolic shift from glycolysis to fatty acid oxidation that occurs during heart maturation in the postnatal heart. Thus, understanding the mechanisms that regulate cardiac metabolism is key to unlocking metabolic interventions during development, disease, and regeneration. In this review, we will focus on the emerging role of metabolism in cardiac development and regeneration and discuss the potential of targeting metabolism for treatment of heart failure.
Collapse
Affiliation(s)
- Jiyoung Bae
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Wyatt G Paltzer
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Ahmed I Mahmoud
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
26
|
Vujic A, Koo ANM, Prag HA, Krieg T. Mitochondrial redox and TCA cycle metabolite signaling in the heart. Free Radic Biol Med 2021; 166:287-296. [PMID: 33675958 DOI: 10.1016/j.freeradbiomed.2021.02.041] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/18/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023]
Abstract
Mitochondria are essential signaling organelles that regulate a broad range of cellular processes and thereby heart function. Multiple mechanisms participate in the communication between mitochondria and the nucleus that maintain cardiomyocyte homeostasis, including mitochondrial reactive oxygen species (ROS) and metabolic shifts in TCA cycle metabolite availability. An increased rate of ROS generation can cause irreversible damage to the cell and proposed to be a leading cause of many pathologies, including accelerated aging and heart disease. Myocardial impairments are also characterised by specific coordinated metabolic changes and dysregulated inflammatory responses. Hence, the mitochondrial respiratory chain is an important mediator between health and disease in the heart. This review will first outline the sources of ROS in the heart, mitochondrial metabolite dynamics, and provide an overview of their implications for heart disease. In addition, we will concentrate our discussion around current cardioprotective strategies relevant to mitochondrial ROS. Thorough understanding of mitochondrial signaling and the complex interplay with vital signaling pathways in the heart might allow us to develop novel therapeutic approaches to cardiovascular disease.
Collapse
Affiliation(s)
- Ana Vujic
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Amy N M Koo
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Hiran A Prag
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK; MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, CB2 0XY, UK
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
27
|
Yuan Y, Yao S, Luo GH, Zhang XY. Impact of metabolism-related mutations on the heart rate of gastric cancer patients after peritoneal lavage. World J Clin Cases 2021; 9:1318-1328. [PMID: 33644198 PMCID: PMC7896693 DOI: 10.12998/wjcc.v9.i6.1318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/04/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND During surgery for gastric cancer, peritoneal lavage using warm distilled water can cause temporary hemodynamic changes.
AIM To examine the associations between changes in heart rate and single nucleotide polymorphisms (SNPs).
METHODS This was a prospective observational study of patients with gastric cancer who underwent gastrectomy and peritoneal hypotonic lavage at the Third Affiliated Hospital of Soochow University from March 2018 to March 2019. Related SNPs were selected, and the verified exons were analyzed. Heart rate and blood pressure (BP) were measured before and after lavage. The patients were grouped as heart rate change ≥ 30% vs < 30%. Comparison and regression analyses of the selected SNPs were performed between the two groups.
RESULTS According to the inclusion/exclusion criteria, 194 patients were included in the analysis. Of these patients, 138 were male, with a mean age of 65.9 ± 0.8 years, and 56 were female, with a mean age of 65.0 ± 1.3 years. Heart rate dropped by 0%-10% in 65 participants, by 10%-15% in 29, by 15%-20% in 23, by 20%-50% in 39, by 50%-100% in four, six had a cardiac arrest, and 28 had an increase in heart rate. Considering the possible impact of exonic SNPs on the phenotypes, TEP1 (rs938886), TEP1 (rs1713449), and RECQL5 (rs820196) were analyzed. The haplotype analysis suggested that the haplotypes CTT [odds ratio (OR) = 2.018, 95% confidence interval (CI): 1.012-4.025, P = 0.0430] and GCC (OR = 2.293, 95%CI: 1.174-4.477, P = 0.0131) of TEP1 (rs938886), TEP1 (rs1713449), and RECQL5 (rs820196) increased the risk of a drop in heart rate > 30%.
CONCLUSION The TEP1 (rs938886), TEP1 (rs1713449), and RECQL5 (rs820196) SNPs were associated with changes in heart rate ≥ 30% during intraperitoneal lavage using distilled water after gastrectomy for gastric cancer.
Collapse
Affiliation(s)
- Yan Yuan
- Department of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu Province, China
| | - Shuang Yao
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu Province, China
| | - Guang-Hua Luo
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu Province, China
| | - Xiao-Ying Zhang
- Department of Cardio-Thoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu Province, China
| |
Collapse
|
28
|
Ness H, Ljones K, Pinho M, Høydal M. Acute high-intensity aerobic exercise increases gene expression of calcium-related proteins and activates endoplasmic reticulum stress responses in diabetic hearts. COMPARATIVE EXERCISE PHYSIOLOGY 2021. [DOI: 10.3920/cep200022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Regular aerobic exercise training has a wide range of beneficial cardiac effects, but recent data also show that acute very strenuous aerobic exercise may impose a transient cardiac exhaustion. The aim of this study was to assess the response to acute high-intensity aerobic exercise on properties of mitochondrial respiration, cardiomyocyte contractile function, Ca2+ handling and transcriptional changes for key proteins facilitating Ca2+ handling and endoplasmic reticulum (ER) stress responses in type 2 diabetic mice. Diabetic mice were assigned to either sedentary control or an acute bout of exercise, consisting of a 10×4 minutes high-intensity interval treadmill run. Mitochondrial respiration, contractile and Ca2+ handling properties of cardiomyocytes were analysed 1 hour after completion of exercise. Gene expression levels of key Ca2+ handling and ER stress response proteins were measured in cardiac tissue samples harvested 1 hour and 24 hours after exercise. We found no significant changes in mitochondrial respiration, cardiomyocyte contractile function or Ca2+ handling 1 hour after the acute exercise. However, gene expression of Atp2a2, Slc8a1 and Ryr2, encoding proteins involved in cardiomyocyte Ca2+ handling, were all significantly upregulated 24 hours after the acute exercise bout. Acute exercise also altered gene expression of several key proteins in ER stress response and unfolded protein response, including Grp94, total Xbp1, Gadd34, and Atf6. The present results show that despite no significant alterations in functional properties of cardiomyocyte function, Ca2+ handling or mitochondrial respiration following one bout of high intensity aerobic exercise training, the expression of genes involved in Ca2+ handling and key components in ER stress and the unfolded protein response were changed. These transcriptional changes may constitute important steps in initiating adaptive remodelling to exercise training in type 2 diabetes.
Collapse
Affiliation(s)
- H.O. Ness
- NTNU, Norwegian University of Technology and Science (NTNU), Faculty of Medicine and Health, Department of Circulation and Medical Imaging, Group of Molecular and Cellular Cardiology, Prinsesse Kristinas gate 9, Trondheim, 7489, Norway
| | - K. Ljones
- NTNU, Norwegian University of Technology and Science (NTNU), Faculty of Medicine and Health, Department of Circulation and Medical Imaging, Group of Molecular and Cellular Cardiology, Prinsesse Kristinas gate 9, Trondheim, 7489, Norway
| | - M. Pinho
- NTNU, Norwegian University of Technology and Science (NTNU), Faculty of Medicine and Health, Department of Circulation and Medical Imaging, Group of Molecular and Cellular Cardiology, Prinsesse Kristinas gate 9, Trondheim, 7489, Norway
| | - M.A. Høydal
- NTNU, Norwegian University of Technology and Science (NTNU), Faculty of Medicine and Health, Department of Circulation and Medical Imaging, Group of Molecular and Cellular Cardiology, Prinsesse Kristinas gate 9, Trondheim, 7489, Norway
| |
Collapse
|
29
|
Huang Q, Su H, Qi B, Wang Y, Yan K, Wang X, Li X, Zhao D. A SIRT1 Activator, Ginsenoside Rc, Promotes Energy Metabolism in Cardiomyocytes and Neurons. J Am Chem Soc 2021; 143:1416-1427. [PMID: 33439015 DOI: 10.1021/jacs.0c10836] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Targeting SIRT1 signaling pathway could improve glucose aerobic metabolism and mitochondrial biosynthesis to resist cardiac and neurological injuries. Ginsenoside Rc has been identified for targeting mitochondrial function, but how ginsenoside Rc interacts with SIRT1 to regulate energy metabolism in cardiomyocytes and neurons under physiological or ischemia/reperfusion (I/R)-injured conditions has not been clearly investigated. Here, we confirm the interaction of Rc on the residue sites of SIRT1 in promoting its activity. Ginsenoside Rc significantly promotes mitochondrial biogenesis and increases the levels of electron-transport chain complex II-IV in cardiomyocytes and neurons. Meanwhile, ginsenoside Rc pretreatment increases ATP production, glucose uptake, and the levels of hexokinase I/II and mitochondrial pyruvate carrier I/II in both cell models. In addition, ginsenoside Rc activates the PGC1α pathway to induce mitochondrial biosynthesis. More importantly, ginsenoside Rc reduces mitochondrial damage and apoptosis through SIRT1 restoration-mediated reduction of PGC1α acetylation in the I/R-induced cardiac and neuronal models. Collectively, the in vitro and in vivo data indicate that ginsenoside Rc as a SIRT1 activator promotes energy metabolism to improve cardio- and neuroprotective functions under normal and I/R injury conditions, which provides new insights into the molecular mechanism of ginsenoside Rc as a protective agent.
Collapse
Affiliation(s)
| | | | | | | | | | - Xinglin Wang
- Guangdong Hanfang Health Research Institute, Guangzhou 510550, P. R. China
| | | | | |
Collapse
|
30
|
McNally LA, Altamimi TR, Fulghum K, Hill BG. Considerations for using isolated cell systems to understand cardiac metabolism and biology. J Mol Cell Cardiol 2020; 153:26-41. [PMID: 33359038 DOI: 10.1016/j.yjmcc.2020.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/13/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022]
Abstract
Changes in myocardial metabolic activity are fundamentally linked to cardiac health and remodeling. Primary cardiomyocytes, induced pluripotent stem cell-derived cardiomyocytes, and transformed cardiomyocyte cell lines are common models used to understand how (patho)physiological conditions or stimuli contribute to changes in cardiac metabolism. These cell models are helpful also for defining metabolic mechanisms of cardiac dysfunction and remodeling. Although technical advances have improved our capacity to measure cardiomyocyte metabolism, there is often heterogeneity in metabolic assay protocols and cell models, which could hinder data interpretation and discernment of the mechanisms of cardiac (patho)physiology. In this review, we discuss considerations for integrating cardiomyocyte cell models with techniques that have become relatively common in the field, such as respirometry and extracellular flux analysis. Furthermore, we provide overviews of metabolic assays that complement XF analyses and that provide information on not only catabolic pathway activity, but biosynthetic pathway activity and redox status as well. Cultivating a more widespread understanding of the advantages and limitations of metabolic measurements in cardiomyocyte cell models will continue to be essential for the development of coherent metabolic mechanisms of cardiac health and pathophysiology.
Collapse
Affiliation(s)
- Lindsey A McNally
- Department of Medicine, Division of Environmental Medicine, Christina Lee Brown Envirome Institute, Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA
| | - Tariq R Altamimi
- Department of Medicine, Division of Environmental Medicine, Christina Lee Brown Envirome Institute, Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA
| | - Kyle Fulghum
- Department of Medicine, Division of Environmental Medicine, Christina Lee Brown Envirome Institute, Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA
| | - Bradford G Hill
- Department of Medicine, Division of Environmental Medicine, Christina Lee Brown Envirome Institute, Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
31
|
Corrêa TD, Pereira AJ, Takala J, Jakob SM. Regional venous-arterial CO 2 to arterial-venous O 2 content difference ratio in experimental circulatory shock and hypoxia. Intensive Care Med Exp 2020; 8:64. [PMID: 33119834 PMCID: PMC7596113 DOI: 10.1186/s40635-020-00353-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/22/2020] [Indexed: 01/28/2023] Open
Abstract
Background Venous–arterial carbon dioxide (CO2) to arterial–venous oxygen (O2) content difference ratio (Cv-aCO2/Ca-vO2) > 1 is supposed to be both sensitive and specific for anaerobic metabolism. What regional hemodynamic and metabolic parameters determine the ratio has not been clarified. Objectives To address determinants of systemic and renal, spleen, gut and liver Cv-aCO2/Ca-vO2. Methods Post hoc analysis of original data from published experimental studies aimed to address effects of different fluid resuscitation strategies on oxygen transport, lactate metabolism and organ dysfunction in fecal peritonitis and endotoxin infusion, and from animals in cardiac tamponade or hypoxic hypoxia. Systemic and regional hemodynamics, blood flow, lactate uptake, carbon dioxide and oxygen-derived variables were determined. Generalized estimating equations (GEE) were fit to assess contributors to systemic and regional Cv-aCO2/Ca-vO2. Results Median (range) of pooled systemic Cv-aCO2/Ca-vO2 in 64 pigs was 1.02 (0.02 to 3.84). While parameters reflecting regional lactate exchange were variably associated with the respective regional Cv-aCO2/Ca-vO2 ratios, only regional ratios were independently correlated with systemic ratio: renal Cv-aCO2 /Ca-vO2 (β = 0.148, 95% CI 0.062 to 0.234; p = 0.001), spleen Cv-aCO2/Ca-vO2 (β = 0.065, 95% CI 0.002 to 0.127; p = 0.042), gut Cv-aCO2/Ca-vO2 (β = 0.117, 95% CI 0.025 to 0.209; p = 0.013), liver Cv-aCO2/Ca-vO2 (β = − 0.159, 95% CI − 0.297 to − 0.022; p = 0.023), hepatosplanchnic Cv-aCO2/Ca-vO2 (β = 0.495, 95% CI 0.205 to 0.786; p = 0.001). Conclusion In a mixed set of animals in different shock forms or during hypoxic injury, hepatosplanchnic Cv-aCO2/Ca-vO2 ratio had the strongest independent association with systemic Cv-aCO2/Ca-vO2, while no independent association was demonstrated for lactate or hemodynamic variables.
Collapse
Affiliation(s)
- Thiago Domingos Corrêa
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland. .,Intensive Care Unit, Hospital Israelita Albert Einstein, Av. Albert Einstein, 627/701, 5th floor, São Paulo, 05651-901, Brazil.
| | - Adriano José Pereira
- Intensive Care Unit, Hospital Israelita Albert Einstein, Av. Albert Einstein, 627/701, 5th floor, São Paulo, 05651-901, Brazil.,Research Group, Hospital Municipal da Vila Santa Catarina, São Paulo, Brazil.,Postgraduate Program of Health Sciences, Federal University of Lavras, Lavras, Brazil
| | - Jukka Takala
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Stephan Mathias Jakob
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
32
|
Dorji J, Vander Jagt CJ, Garner JB, Marett LC, Mason BA, Reich CM, Xiang R, Clark EL, Cocks BG, Chamberlain AJ, MacLeod IM, Daetwyler HD. Expression of mitochondrial protein genes encoded by nuclear and mitochondrial genomes correlate with energy metabolism in dairy cattle. BMC Genomics 2020; 21:720. [PMID: 33076826 PMCID: PMC7574280 DOI: 10.1186/s12864-020-07018-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022] Open
Abstract
Background Mutations in the mitochondrial genome have been implicated in mitochondrial disease, often characterized by impaired cellular energy metabolism. Cellular energy metabolism in mitochondria involves mitochondrial proteins (MP) from both the nuclear (NuMP) and mitochondrial (MtMP) genomes. The expression of MP genes in tissues may be tissue specific to meet varying specific energy demands across the tissues. Currently, the characteristics of MP gene expression in tissues of dairy cattle are not well understood. In this study, we profile the expression of MP genes in 29 adult and six foetal tissues in dairy cattle using RNA sequencing and gene expression analyses: particularly differential gene expression and co-expression network analyses. Results MP genes were differentially expressed (DE; over-expressed or under-expressed) across tissues in cattle. All 29 tissues showed DE NuMP genes in varying proportions of over-expression and under-expression. On the other hand, DE of MtMP genes was observed in < 50% of tissues and notably MtMP genes within a tissue was either all over-expressed or all under-expressed. A high proportion of NuMP (up to 60%) and MtMP (up to 100%) genes were over-expressed in tissues with expected high metabolic demand; heart, skeletal muscles and tongue, and under-expressed (up to 45% of NuMP, 77% of MtMP genes) in tissues with expected low metabolic rates; leukocytes, thymus, and lymph nodes. These tissues also invariably had the expression of all MtMP genes in the direction of dominant NuMP genes expression. The NuMP and MtMP genes were highly co-expressed across tissues and co-expression of genes in a cluster were non-random and functionally enriched for energy generation pathway. The differential gene expression and co-expression patterns were validated in independent cow and sheep datasets. Conclusions The results of this study support the concept that there are biological interaction of MP genes from the mitochondrial and nuclear genomes given their over-expression in tissues with high energy demand and co-expression in tissues. This highlights the importance of considering MP genes from both genomes in future studies related to mitochondrial functions and traits related to energy metabolism.
Collapse
Affiliation(s)
- Jigme Dorji
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC, 3083, Australia. .,Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia.
| | - Christy J Vander Jagt
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
| | - Josie B Garner
- Agriculture Victoria, Ellinbank Dairy Centre, Ellinbank, VIC, 3822, Australia
| | - Leah C Marett
- Agriculture Victoria, Ellinbank Dairy Centre, Ellinbank, VIC, 3822, Australia
| | - Brett A Mason
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
| | - Coralie M Reich
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
| | - Ruidong Xiang
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia.,Faculty of Veterinary & Agricultural Science, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Emily L Clark
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, UK
| | - Benjamin G Cocks
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC, 3083, Australia.,Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
| | - Amanda J Chamberlain
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
| | - Iona M MacLeod
- Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
| | - Hans D Daetwyler
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC, 3083, Australia.,Agriculture Victoria, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
| |
Collapse
|
33
|
de Lacerda Alexandre JV, Viana YIP, David CEB, Cunha PLO, Albuquerque AC, Varela ALN, Kowaltowski AJ, Facundo HT. Quercetin treatment increases H 2O 2 removal by restoration of endogenous antioxidant activity and blocks isoproterenol-induced cardiac hypertrophy. Naunyn Schmiedebergs Arch Pharmacol 2020; 394:217-226. [PMID: 32930861 DOI: 10.1007/s00210-020-01953-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/20/2020] [Indexed: 02/01/2023]
Abstract
Oxidative stress, characterized by the accumulation of reactive oxygen species (ROS), is implicated in the pathogenesis of several diseases, including cardiac hypertrophy. The flavonoid quercetin is a potent ROS scavenger, with several beneficial effects for the cardiovascular system, including antihypertrophic effects. Oxidative imbalance has been implicated in cardiac hypertrophy and heart failure. In this work, we tested whether quercetin could attenuate cardiac hypertrophy by improving redox balance and mitochondrial homeostasis. To test this hypothesis, we treated a group of mice with isoproterenol (30 mg/kg/day) for 4 or 8 consecutive days. Another group received quercetin (10 mg/kg/day) from day 5th of isoproterenol treatment. We carried out the following assays in cardiac tissue: measurement of cardiac hypertrophy, protein sulfhydryl, catalase, Cu/Zn and Mn-superoxide dismutase (SOD) activity, detection of H2O2, and opening of the mitochondrial permeability transition pore. The animals treated with isoproterenol for the initial 4 days showed increased cardiac weight/tibia length ratio, decreased protein sulfhydryl content, compromised SOD and catalase activity, and high H2O2 levels. Quercetin was able to attenuate cardiac hypertrophy, restore protein sulfhydryl, and antioxidant activity, in addition to efficiently blocking the H2O2. We also observed that isoproterenol decreases mitochondrial SOD activity, while quercetin reverses it. Strikingly, quercetin also protects mitochondria against the opening of mitochondrial permeability transition pore. Taken together, these results suggest that quercetin is capable of reversing established isoproterenol-induced cardiac hypertrophy through the restoration of cellular redox balance and protecting mitochondria.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
34
|
Mitochondria: Aging, Metabolic Syndrome and Cardiovascular Diseases. Formation of a New Paradigm. ACTA BIOMEDICA SCIENTIFICA 2020. [DOI: 10.29413/abs.2020-5.4.5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cardiovascular diseases are among the major causes of mortality among aged people in most developed countries. Oxidative stress, which causes mutations of mitochondrial DNA and mitochondrial dysfunctions, was considered as the main mechanism of heart failure and other pathologies of old age. However, in recent years the prior paradigm of mechanisms of aging, oxidative stress and antioxidative defense was questioned and in some aspects even turned out to be wrong. In this review, we discuss the new data that led to the need to reconsider paradigms. We show that although the mitochondrial free radical theory of aging remains valid, the radical responsible for the aging is the protonated form of the superoxide radical, namely perhydroxyl radical, which was largely ignored all previous years. Perhydroxyl radical initiates the isoprostane pathway of lipid peroxidation (IPLP) of polyunsaturated fatty acids, which are part of the phospholipid core of the mitochondrial inner membrane. IPLP was discovered 30 years ago by Roberts and Morrow at the Vanderbilt University, but the mechanism of its initiation remained unknown. The IPLP causes formation of the racemic mixture of hundreds of biologically active products, named isoprostanes, and highly toxic molecules, first of all isolevuglandins. We distinguish two types of damages caused by IPLP during aging. The first one is associated with oxidative damages to cardiolipin and phosphatidylethanolamine (PEA), which result in disruption of polyenzymatic complexes of the oxidative phosphorylation system. The second type of dysfunctions is caused by the direct actions of toxic products on the lysine-containing proteins and PEA. To this type of mitochondrial damages evidently belongs the oxidative damage of the mitochondrial DNA polymerase, which results in a 20-fold increase in mutations of mitochondrial mtDNA.
Collapse
|
35
|
Song Y, Qi Z, Zhang Y, Wei J, Liao X, Li R, Dong C, Zhu L, Yang Z, Cai Z. Effects of exposure to ambient fine particulate matter on the heart of diet-induced obesity mouse model. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 732:139304. [PMID: 32438171 DOI: 10.1016/j.scitotenv.2020.139304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 06/11/2023]
Abstract
Exposure to fine particulate matter (PM2.5) is associated with decreased cardiac function, especially in high risk populations such as obese ones. In this study, impacts of PM2.5 exposure on cardiac function were investigated by using the diet-induced obesity mice model. Mice were fed with normal diet or high-fat diet (HFD) for four weeks and then exposed to phosphate-buffered solution or Taiyuan winter PM2.5 (0.25 mg/kg body/day) through intratracheal instillation for another four weeks. Among physiological indices recorded, heart rate and blood pressure were increased after PM2.5 exposure in the heart of the obese mice. Metabolomics and lipidomics were applied to explore molecular alterations in response to the co-treatment of PM2.5 and HFD. Our results demonstrated both direct impacts on cardiac function and indirect effects resulted from the injury of other organs. Inflammation of lung and hypothalamus may be responsible for the elevation of phenylalanine metabolism in serum and its downstream products: epinephrine and norepinephrine, the catecholamines involves in regulating cardiac system. In intracardiac system, the co-treatment led to imbalance of energy metabolism, in addition to oxidative stress and inflammation. In contrast to the upregulation of glucose and fatty acids uptake and CoA synthesis, levels of ATP, acetyl-CoA and the intermediates in glycolysis pathway decreased in the heart. The results indicated that energy metabolism disorder was possibly one of the important contributing factors to the more severe adverse effects of the combined treatment of HFD and PM2.5.
Collapse
Affiliation(s)
- Yuanyuan Song
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Zenghua Qi
- School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, China
| | - Yanhao Zhang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Juntong Wei
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Xiaoliang Liao
- School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, China
| | - Ruijin Li
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| | - Chuan Dong
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| | - Lin Zhu
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Zhu Yang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
36
|
Xiang K, Qin Z, Zhang H, Liu X. Energy Metabolism in Exercise-Induced Physiologic Cardiac Hypertrophy. Front Pharmacol 2020; 11:1133. [PMID: 32848751 PMCID: PMC7403221 DOI: 10.3389/fphar.2020.01133] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022] Open
Abstract
Physiologic hypertrophy of the heart preserves or enhances systolic function without interstitial fibrosis or cell death. As a unique form of physiological stress, regular exercise training can trigger the adaptation of cardiac muscle to cause physiological hypertrophy, partly due to its ability to improve cardiac metabolism. In heart failure (HF), cardiac dysfunction is closely associated with early initiation of maladaptive metabolic remodeling. A large amount of clinical and experimental evidence shows that metabolic homeostasis plays an important role in exercise training, which is conducive to the treatment and recovery of cardiovascular diseases. Potential mechanistic targets for modulation of cardiac metabolism have become a hot topic at present. Thus, exploring the energy metabolism mechanism in exercise-induced physiologic cardiac hypertrophy may produce new therapeutic targets, which will be helpful to design novel effective strategies. In this review, we summarize the changes of myocardial metabolism (fatty acid metabolism, carbohydrate metabolism, and mitochondrial adaptation), metabolically-related signaling molecules, and probable regulatory mechanism of energy metabolism during exercise-induced physiological cardiac hypertrophy.
Collapse
Affiliation(s)
- Kefa Xiang
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Zhen Qin
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Huimin Zhang
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Xia Liu
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
37
|
Zhao J, Conklin DJ, Guo Y, Zhang X, Obal D, Guo L, Jagatheesan G, Katragadda K, He L, Yin X, Prodhan MAI, Shah J, Hoetker D, Kumar A, Kumar V, Wempe MF, Bhatnagar A, Baba SP. Cardiospecific Overexpression of ATPGD1 (Carnosine Synthase) Increases Histidine Dipeptide Levels and Prevents Myocardial Ischemia Reperfusion Injury. J Am Heart Assoc 2020; 9:e015222. [PMID: 32515247 PMCID: PMC7429021 DOI: 10.1161/jaha.119.015222] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Myocardial ischemia reperfusion (I/R) injury is associated with complex pathophysiological changes characterized by pH imbalance, the accumulation of lipid peroxidation products acrolein and 4-hydroxy trans-2-nonenal, and the depletion of ATP levels. Cardioprotective interventions, designed to address individual mediators of I/R injury, have shown limited efficacy. The recently identified enzyme ATPGD1 (Carnosine Synthase), which synthesizes histidyl dipeptides such as carnosine, has the potential to counteract multiple effectors of I/R injury by buffering intracellular pH and quenching lipid peroxidation products and may protect against I/R injury. METHODS AND RESULTS We report here that β-alanine and carnosine feeding enhanced myocardial carnosine levels and protected the heart against I/R injury. Cardiospecific overexpression of ATPGD1 increased myocardial histidyl dipeptides levels and protected the heart from I/R injury. Isolated cardiac myocytes from ATPGD1-transgenic hearts were protected against hypoxia reoxygenation injury. The overexpression of ATPGD1 prevented the accumulation of acrolein and 4-hydroxy trans-2-nonenal-protein adducts in ischemic hearts and delayed acrolein or 4-hydroxy trans-2-nonenal-induced hypercontracture in isolated cardiac myocytes. Changes in the levels of ATP, high-energy phosphates, intracellular pH, and glycolysis during low-flow ischemia in the wild-type mice hearts were attenuated in the ATPGD1-transgenic hearts. Two natural dipeptide analogs (anserine and balenine) that can either quench aldehydes or buffer intracellular pH, but not both, failed to protect against I/R injury. CONCLUSIONS Either exogenous administration or enhanced endogenous formation of histidyl dipeptides prevents I/R injury by attenuating changes in intracellular pH and preventing the accumulation of lipid peroxidation derived aldehydes.
Collapse
Affiliation(s)
- Jingjing Zhao
- Diabetes and Obesity CenterUniversity of LouisvilleKY
- Christina Lee Brown Envirome InstituteUniversity of LouisvilleKY
| | - Daniel J. Conklin
- Diabetes and Obesity CenterUniversity of LouisvilleKY
- Christina Lee Brown Envirome InstituteUniversity of LouisvilleKY
| | - Yiru Guo
- Division of Cardiovascular MedicineDepartment of MedicineUniversity of LouisvilleKY
| | - Xiang Zhang
- Department of ChemistryUniversity of LouisvilleKY
| | - Detlef Obal
- Department of Anesthesiology and Perioperative and Pain MedicineStanford UniversityPalo AltoCA
| | - Luping Guo
- Diabetes and Obesity CenterUniversity of LouisvilleKY
- Christina Lee Brown Envirome InstituteUniversity of LouisvilleKY
| | - Ganapathy Jagatheesan
- Diabetes and Obesity CenterUniversity of LouisvilleKY
- Christina Lee Brown Envirome InstituteUniversity of LouisvilleKY
| | - Kartik Katragadda
- Diabetes and Obesity CenterUniversity of LouisvilleKY
- Christina Lee Brown Envirome InstituteUniversity of LouisvilleKY
| | - Liqing He
- Department of ChemistryUniversity of LouisvilleKY
| | - Xinmin Yin
- Department of ChemistryUniversity of LouisvilleKY
| | | | - Jasmit Shah
- Department of MedicineThe Aga Khan UniversityMedical CollegeNairobiKenya
| | - David Hoetker
- Diabetes and Obesity CenterUniversity of LouisvilleKY
- Christina Lee Brown Envirome InstituteUniversity of LouisvilleKY
| | - Amit Kumar
- Department of Pharmaceutical SciencesUniversity of ColoradoDenverCO
| | - Vijay Kumar
- Department of Pharmaceutical SciencesUniversity of ColoradoDenverCO
| | - Michael F. Wempe
- Department of Pharmaceutical SciencesUniversity of ColoradoDenverCO
| | - Aruni Bhatnagar
- Diabetes and Obesity CenterUniversity of LouisvilleKY
- Christina Lee Brown Envirome InstituteUniversity of LouisvilleKY
| | - Shahid P. Baba
- Diabetes and Obesity CenterUniversity of LouisvilleKY
- Christina Lee Brown Envirome InstituteUniversity of LouisvilleKY
| |
Collapse
|
38
|
Stotland AB, Spivia W, Orosco A, Andres AM, Gottlieb RA, Van Eyk JE, Parker SJ. MitoPlex: A targeted multiple reaction monitoring assay for quantification of a curated set of mitochondrial proteins. J Mol Cell Cardiol 2020; 142:1-13. [PMID: 32234390 PMCID: PMC7347090 DOI: 10.1016/j.yjmcc.2020.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 12/21/2022]
Abstract
Mitochondria are the major source of cellular energy (ATP), as well as critical mediators of widespread functions such as cellular redox balance, apoptosis, and metabolic flux. The organelles play an especially important role in the maintenance of cardiac homeostasis; their inability to generate ATP following impairment due to ischemic damage has been directly linked to organ failure. Methods to quantify mitochondrial content are limited to low throughput immunoassays, measurement of mitochondrial DNA, or relative quantification by untargeted mass spectrometry. Here, we present a high throughput, reproducible and quantitative mass spectrometry multiple reaction monitoring based assay of 37 proteins critical to central carbon chain metabolism and overall mitochondrial function termed 'MitoPlex'. We coupled this protein multiplex with a parallel analysis of the central carbon chain metabolites (219 metabolite assay) extracted in tandem from the same sample, be it cells or tissue. In tests of its biological applicability in cells and tissues, "MitoPlex plus metabolites" indicated profound effects of HMG-CoA Reductase inhibition (e.g., statin treatment) on mitochondria of i) differentiating C2C12 skeletal myoblasts, as well as a clear opposite trend of statins to promote mitochondrial protein expression and metabolism in heart and liver, while suppressing mitochondrial protein and ii) aspects of metabolism in the skeletal muscle obtained from C57Bl6 mice. Our results not only reveal new insights into the metabolic effect of statins in skeletal muscle, but present a new high throughput, reliable MS-based tool to study mitochondrial dynamics in both cell culture and in vivo models.
Collapse
Affiliation(s)
- Aleksandr B Stotland
- Molecular Cardiobiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Weston Spivia
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Amanda Orosco
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Allen M Andres
- Molecular Cardiobiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Roberta A Gottlieb
- Molecular Cardiobiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Sarah J Parker
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America.
| |
Collapse
|
39
|
Evans LW, Stratton MS, Ferguson BS. Dietary natural products as epigenetic modifiers in aging-associated inflammation and disease. Nat Prod Rep 2020; 37:653-676. [PMID: 31993614 PMCID: PMC7577396 DOI: 10.1039/c9np00057g] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Covering: up to 2020Chronic, low-grade inflammation is linked to aging and has been termed "inflammaging". Inflammaging is considered a key contributor to the development of metabolic dysfunction and a broad spectrum of diseases or disorders including declines in brain and heart function. Genome-wide association studies (GWAS) coupled with epigenome-wide association studies (EWAS) have shown the importance of diet in the development of chronic and age-related diseases. Moreover, dietary interventions e.g. caloric restriction can attenuate inflammation to delay and/or prevent these diseases. Common themes in these studies entail the use of phytochemicals (plant-derived compounds) or the production of short chain fatty acids (SCFAs) as epigenetic modifiers of DNA and histone proteins. Epigenetic modifications are dynamically regulated and as such, serve as potential therapeutic targets for the treatment or prevention of age-related disease. In this review, we will focus on the role for natural products that include phytochemicals and short chain fatty acids (SCFAs) as regulators of these epigenetic adaptations. Specifically, we discuss regulators of methylation, acetylation and acylation, in the protection from chronic inflammation driven metabolic dysfunction and deterioration of neurocognitive and cardiac function.
Collapse
Affiliation(s)
- Levi W Evans
- Department of Nutrition, University of Nevada, Reno, NV 89557, USA.
| | | | | |
Collapse
|
40
|
Tateishi E, Kiso K, Fukuda T. Assessing the Clinical Value of Myocardial Perfusion SPECT in Cardiac Sarcoidosis with Diffuse Myocardial 18F-FDG Uptake. ANNALS OF NUCLEAR CARDIOLOGY 2020; 6:39-45. [PMID: 37123497 PMCID: PMC10133934 DOI: 10.17996/anc.20-00125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/27/2020] [Accepted: 07/30/2020] [Indexed: 05/02/2023]
Abstract
Background: Myocardial 18F-fluorodeoxyglucose (18F-FDG) uptake is a sign of active inflammation in patients with cardiac sarcoidosis (CS) under the correct circumstance. However, even under the proper preparation, diffuse myocardial 18F-FDG uptake is frequently observed in the failing heart and misleads the CS disease activity. The aim of this study was to establish the diagnostic value of resting myocardial perfusion single photon emission computed tomography (SPECT) for assessing CS disease activity in patients with diffuse myocardial 18F-FDG uptake. Methods: We examined 39 patients with either histologically or clinically proven CS. All patients underwent 18F-FDG positron emission tomography (PET) and resting 99mTc-SPECT. The presence of perfusion-metabolic mismatch was evaluated with generating polar maps of 18F-FDG PET and 99mTc-SPECT images. Results: Increased myocardial 18F-FDG uptake was observed in 33 (85%) of 39 patients. Focal 18F-FDG uptake was detected in 16 patients and diffuse 18F-FDG uptake was seen in 17 patients. Brain natriuretic peptide (BNP) levels were significantly higher in patients with diffuse 18F-FDG uptake than those with focal 18F-FDG uptake (p=0.002). With comparing polar maps of 18F-FDG PET and 99mTc-SPECT images, 8 of 16 patients with diffuse 18F-FDG uptake and myocardial perfusion defects demonstrated perfusion-metabolic mismatch which represented active inflammatory lesions in CS. Conclusions: Simultaneous evaluation of myocardial 18F-FDG PET and 99mTc-SPECT by polar map analysis provides more relevant information for assessing disease activity in CS than 18F-FDG PET images alone. Perfusion-metabolic mismatch might indicate latent active inflammation in CS patients with diffuse myocardial 18F-FDG uptake, who had advanced heart failure.
Collapse
Affiliation(s)
- Emi Tateishi
- Department of Radiology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
- Reprint requests and correspondence: Emi Tateishi, MD, PhD, Department of Radiology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan / E-mail:
| | - Keisuke Kiso
- Department of Radiology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Tetsuya Fukuda
- Department of Radiology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| |
Collapse
|
41
|
Radhakrishnan J, Baetiong A, Kaufman H, Huynh M, Leschinsky A, Fresquez A, White C, DiMario JX, Gazmuri RJ. Improved exercise capacity in cyclophilin-D knockout mice associated with enhanced oxygen utilization efficiency and augmented glucose uptake via AMPK-TBC1D1 signaling nexus. FASEB J 2019; 33:11443-11457. [PMID: 31339770 DOI: 10.1096/fj.201802238r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We previously reported in HEK 293T cells that silencing the mitochondrial peptidyl prolyl isomerase cyclophilin-D (Cyp-D) reduces Vo2. We now report that in vivo Cyp-D ablation using constitutive Cyp-D knockout (KO) mice also reduces Vo2 both at rest (∼15%) and during treadmill exercise (∼12%). Yet, despite Vo2 reduction, these Cyp-D KO mice ran longer (1071 ± 77 vs. 785 ± 79 m; P = 0.002), for longer time (43 ± 3 vs. 34 ± 3 min; P = 0.004), and at higher speed (34 ± 1 vs. 29 ± 1 m/s; P ≤ 0.001), resulting in increased work (87 ± 6 vs. 58 ± 6 J; P ≤ 0.001). There were parallel reductions in carbon dioxide production, but of lesser magnitude, yielding a 2.3% increase in the respiratory exchange ratio consistent with increased glucose utilization as respiratory substrate. In addition, primary skeletal muscle cells of Cyp-D KO mice subjected to electrical stimulation exhibited higher glucose uptake (4.4 ± 0.55 vs. 2.6 ± 0.04 pmol/mg/min; P ≤ 0.001) with enhanced AMPK activation (0.58 ± 0.06 vs. 0.38 ± 0.03 pAMPK/β-tubulin ratio; P ≤ 0.01) and TBC1 (Tre-2/USP6, BUB2, Cdc16) domain family, member 1 (TBC1D1) inactivation. Likewise, pharmacological activation of AMPK also increased glucose uptake (3.2 ± 0.3 vs. 2.3 ± 0.2 pmol/mg/min; P ≤ 0.001). Moreover, lactate and ATP levels were increased in these cells. Taken together, Cyp-D ablation triggered an adaptive response resulting in increased exercise capacity despite less oxygen utilization associated with increased glucose uptake and utilization involving AMPK-TBC1D1 signaling nexus.-Radhakrishnan, J., Baetiong, A., Kaufman, H., Huynh, M., Leschinsky, A., Fresquez, A., White, C., DiMario, J. X., Gazmuri, R. J. Improved exercise capacity in cyclophilin-D knockout mice associated with enhanced oxygen utilization efficiency and augmented glucose uptake via AMPK-TBC1D1 signaling nexus.
Collapse
Affiliation(s)
- Jeejabai Radhakrishnan
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Department of Clinical Sciences, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Alvin Baetiong
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Harrison Kaufman
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Michelle Huynh
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Angela Leschinsky
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Adriana Fresquez
- Discipline of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Carl White
- Discipline of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Joseph X DiMario
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Department of Biomedical Research, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | - Raúl J Gazmuri
- Resuscitation Institute, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Department of Clinical Sciences, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
- Captain James A. Lovell Federal Health Care Center, North Chicago, Illinois, USA
| |
Collapse
|
42
|
Abstract
Metabolic pathways integrate to support tissue homeostasis and to prompt changes in cell phenotype. In particular, the heart consumes relatively large amounts of substrate not only to regenerate ATP for contraction but also to sustain biosynthetic reactions for replacement of cellular building blocks. Metabolic pathways also control intracellular redox state, and metabolic intermediates and end products provide signals that prompt changes in enzymatic activity and gene expression. Mounting evidence suggests that the changes in cardiac metabolism that occur during development, exercise, and pregnancy as well as with pathological stress (eg, myocardial infarction, pressure overload) are causative in cardiac remodeling. Metabolism-mediated changes in gene expression, metabolite signaling, and the channeling of glucose-derived carbon toward anabolic pathways seem critical for physiological growth of the heart, and metabolic inefficiency and loss of coordinated anabolic activity are emerging as proximal causes of pathological remodeling. This review integrates knowledge of different forms of cardiac remodeling to develop general models of how relationships between catabolic and anabolic glucose metabolism may fortify cardiac health or promote (mal)adaptive myocardial remodeling. Adoption of conceptual frameworks based in relational biology may enable further understanding of how metabolism regulates cardiac structure and function.
Collapse
Affiliation(s)
- Andrew A Gibb
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (A.A.G.)
| | - Bradford G Hill
- the Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville School of Medicine, KY (B.G.H.).
| |
Collapse
|
43
|
Kocabaş U, Yılmaz Ö, Kurtoğlu V. Diabetic cardiomyopathy: acute and reversible left ventricular systolic dysfunction due to cardiotoxicity of hyperglycaemic hyperosmolar state-a case report†. EUROPEAN HEART JOURNAL-CASE REPORTS 2019; 3:5479977. [PMID: 31449603 PMCID: PMC6601186 DOI: 10.1093/ehjcr/ytz049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 04/12/2019] [Indexed: 11/14/2022]
Abstract
Background Diabetic cardiomyopathy (DC) is defined as a ventricular diastolic and/or systolic dysfunction, which is directly related to diabetes mellitus (DM) in the absence of coronary artery disease, valvular, congenital or hypertensive heart disease, and alcoholism. In this report, we present an unusual case of a patient with DC and reversible, acute left ventricular systolic dysfunction due to cardiotoxicity of hyperosmolar hyperglycaemic state (HHS). Case summary A 20-year-old male patient presented with weakness and polyuria. Physical examination and electrocardiogram were normal. Laboratory results and arterial blood gas analysis were consistent with HHS. Baseline echocardiography showed global left ventricular hypokinesis with an ejection fraction (EF) of 36%. The patient’s clinical condition improved after blood glucose level normalization and echocardiography revealed progressive improvement in the left ventricular systolic function with an EF of 54% at the 5-day follow-up and an EF of 69% at the 15-day follow-up. Discussion Uncontrolled DM and hyperglycaemic crisis may result in cardiotoxicity, acute left ventricular systolic dysfunction, and DC. The pathophysiological mechanism of this phenomenon is still unclear. Blood glucose control is the most important strategy for the prevention of DC.
Collapse
Affiliation(s)
- Umut Kocabaş
- Department of Cardiology, Başkent University Faculty of Medicine, Istanbul, Turkey
| | - Özgür Yılmaz
- Department of Internal Medicine, Soma State Hospital, Manisa, Turkey
| | - Volkan Kurtoğlu
- Department of Internal Medicine, Soma State Hospital, Manisa, Turkey
| |
Collapse
|
44
|
Polak-Iwaniuk A, Harasim-Symbor E, Gołaszewska K, Chabowski A. How Hypertension Affects Heart Metabolism. Front Physiol 2019; 10:435. [PMID: 31040794 PMCID: PMC6476990 DOI: 10.3389/fphys.2019.00435] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/29/2019] [Indexed: 01/15/2023] Open
Abstract
Hypertension is one of the most frequently observed cardiovascular diseases, which precedes heart failure in 75% of its cases. It is well-established that hypertensive patients have whole body metabolic complications such as hyperlipidemia, hyperglycemia, decreased insulin sensitivity or diabetes mellitus. Since myocardial metabolism is strictly dependent on hormonal status as well as substrate milieu, the above mentioned disturbances may affect energy generation status in the heart. Interestingly, it was found that hypertension induces a shift in substrate preference toward increased glucose utilization in cardiac muscle, prior to structural changes development. The present work reports advances in the aspect of heart metabolism under high blood pressure conditions, including human and the most common animal models of hypertension.
Collapse
Affiliation(s)
| | - Ewa Harasim-Symbor
- Department of Physiology, Medical University of Białystok, Białystok, Poland
| | | | - Adrian Chabowski
- Department of Physiology, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
45
|
Dragan S, Buleu F, Christodorescu R, Cobzariu F, Iurciuc S, Velimirovici D, Xiao J, Luca CT. Benefits of multiple micronutrient supplementation in heart failure: A comprehensive review. Crit Rev Food Sci Nutr 2018; 59:965-981. [PMID: 30507249 DOI: 10.1080/10408398.2018.1540398] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Multiple micronutrient supplementation has been suggested to have a role on health outcomes in patients with heart failure (HF), but the evidence is inconclusive. OBJECTIVE To elucidate the role of multiple micronutrient supplementation in heart failure we performed a comprehensive review of the literature. METHODS AND RESULTS The search in databases included PUBMED (until June 2018) to detect randomized controlled trials (RCTs) and meta-analyzes that investigated the impact of micronutrient supplementation in HF. RESULTS With more than 2357 titles and abstracts reviewed, we included only the studies suitable for the final review. Whether alone or in combination, micronutrients have been found to improve the health outcomes of patients with HF by improving symptoms, work capacity and left ventricular ejection fraction (LVEF), thus increasing the quality of life in these patients. CONCLUSION Future studies are needed to document the effects of multiple micronutrient associations in order to include them in nutritional guidelines to increase survival and to improve quality of life in patients with heart failure.
Collapse
Affiliation(s)
- Simona Dragan
- a Department of Cardiology, Discipline of Preventive Cardiology , "Victor Babes" University of Medicine and Pharmacy , Timisoara , Romania
| | - Florina Buleu
- a Department of Cardiology, Discipline of Preventive Cardiology , "Victor Babes" University of Medicine and Pharmacy , Timisoara , Romania
| | - Ruxandra Christodorescu
- b Department of Internal Medicine, Discipline of Medical Semiology II , "Victor Babes" University of Medicine and Pharmacy , Timisoara , Romania
| | - Florin Cobzariu
- a Department of Cardiology, Discipline of Preventive Cardiology , "Victor Babes" University of Medicine and Pharmacy , Timisoara , Romania
| | - Stela Iurciuc
- a Department of Cardiology, Discipline of Preventive Cardiology , "Victor Babes" University of Medicine and Pharmacy , Timisoara , Romania
| | - Dana Velimirovici
- a Department of Cardiology, Discipline of Preventive Cardiology , "Victor Babes" University of Medicine and Pharmacy , Timisoara , Romania
| | - Jianbo Xiao
- c Institute of Chinese Medical Sciences , University of Macau , Taipa , China
| | - Constantin Tudor Luca
- d Department of Cardiology, Discipline of Cardiology II , "Victor Babes" University of Medicine and Pharmacy , Timisoara , Romania
| |
Collapse
|
46
|
Cardiac mitochondrial respiration following a low-carbohydrate, high-fat diet in apolipoprotein E-deficient mice. J Physiol Biochem 2018; 75:65-72. [PMID: 30362048 DOI: 10.1007/s13105-018-0653-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 10/10/2018] [Indexed: 01/13/2023]
Abstract
Low-carbohydrate diets are considered to be an effective approach to weight loss and have, subsequently, grown in popularity. Despite the apparent health benefits that these diets may provide for insulin resistance, hypertension, and dyslipidemia, their implications on cardiomyocyte oxidative capacity have yet to be investigated. To evaluate the adaptations induced by a 6-week low-carbohydrate, high-fat (LCHF) diet on mitochondrial respiration, two groups of male mice were investigated: Apolipoprotein E-deficient mice on a LCHF diet (L-DIET) and apolipoprotein E-deficient mice on a regular rodent diet (CON). Heart tissue was extracted and used for high-resolution respirometry (HRR), while immunoblotting was performed to quantify mitochondrial density and complexes. The results demonstrate increased expression of all five mitochondrial subunits in the L-DIET group compared to control condition. Furthermore, HRR revealed increased efficiency of substrate consumption, implying augmented oxidative capacity in the L-DIET group. These findings further support the notion that cardiomyocytes prefer lipids as a primary fuel source, by demonstrating that the shift in metabolism caused by a LCHF diet facilitates such an environment. This provides important information regarding the effects of a LCHF on cardiomyocytes, especially when considering free radical production and heart dysfunction.
Collapse
|
47
|
Sözmen M, Devrim AK, Kabak YB, Devrim T. Periostin alters transcriptional profile in a rat model of isoproterenol-induced cardiotoxicity. Hum Exp Toxicol 2018; 38:255-266. [DOI: 10.1177/0960327118802617] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Periostin is an extracellular matrix protein from the fasciclin family that guides cellular trafficking and extracellular matrix organization. Periostin stimulates mature cardiomyocytes to reenter the cell cycle. The molecular mechanism behind such stimulation remains to be explored. A DNA microarray technology constituting 30,429 gene-level probe sets was utilized to investigate effects of recombinant murine periostin peptide on the gene expression pattern in a rat model of isoproterenol (ISO)-induced myocardial injury. The experiment was performed on 84 adult male Sprague-Dawley rats in four groups ( n = 21): (1) control group, (2) only periostin applied group, (3) ISO cardiotoxicity group, and (4) ISO + periostin group. The experiment was continued for 28 days, and rats were killed on days 1, 7, and 28 ( n = 7). Microarray analyses revealed that periostin significantly altered the expression of at least ±2-fold of 2474 genes in the ISO + periostin group compared to the ISO cardiotoxicity group of which 521 genes altered out of 30,429 gene-level probe sets. Ingenuity pathway analysis indicated that multiple pathway networks were affected by periostin, with predominant changes occurring in the expression of genes involved in oxidative phosphorylation, oxidative stress, fatty acid metabolism, and TNF-α NF-κB signaling pathways. These findings indicate that periostin alters gene expression profile in the ISO-induced myocardial injury and modulates local myocardial inflammation, possibly mitigating inflammation through TNF-α NF-κB signaling pathway along with a decreased Casp7 activity and apoptotic cell death.
Collapse
Affiliation(s)
- M Sözmen
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - AK Devrim
- Department of Biochemistry, Faculty of Veterinary Medicine, Kırıkkale University, Kırıkkale, Turkey
| | - YB Kabak
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - T Devrim
- Department of Pathology, Faculty of Medicine, Kırıkkale University, Kırıkkale, Turkey
| |
Collapse
|
48
|
Bernardo BC, Ooi JYY, Weeks KL, Patterson NL, McMullen JR. Understanding Key Mechanisms of Exercise-Induced Cardiac Protection to Mitigate Disease: Current Knowledge and Emerging Concepts. Physiol Rev 2018; 98:419-475. [PMID: 29351515 DOI: 10.1152/physrev.00043.2016] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The benefits of exercise on the heart are well recognized, and clinical studies have demonstrated that exercise is an intervention that can improve cardiac function in heart failure patients. This has led to significant research into understanding the key mechanisms responsible for exercise-induced cardiac protection. Here, we summarize molecular mechanisms that regulate exercise-induced cardiac myocyte growth and proliferation. We discuss in detail the effects of exercise on other cardiac cells, organelles, and systems that have received less or little attention and require further investigation. This includes cardiac excitation and contraction, mitochondrial adaptations, cellular stress responses to promote survival (heat shock response, ubiquitin-proteasome system, autophagy-lysosomal system, endoplasmic reticulum unfolded protein response, DNA damage response), extracellular matrix, inflammatory response, and organ-to-organ crosstalk. We summarize therapeutic strategies targeting known regulators of exercise-induced protection and the challenges translating findings from bench to bedside. We conclude that technological advancements that allow for in-depth profiling of the genome, transcriptome, proteome and metabolome, combined with animal and human studies, provide new opportunities for comprehensively defining the signaling and regulatory aspects of cell/organelle functions that underpin the protective properties of exercise. This is likely to lead to the identification of novel biomarkers and therapeutic targets for heart disease.
Collapse
Affiliation(s)
- Bianca C Bernardo
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Jenny Y Y Ooi
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Kate L Weeks
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Natalie L Patterson
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute , Melbourne , Australia ; Department of Paediatrics, University of Melbourne , Victoria , Australia ; Department of Diabetes, Central Clinical School, Monash University , Victoria , Australia ; Department of Medicine, Central Clinical School, Monash University , Victoria , Australia ; and Department of Physiology, School of Biomedical Sciences , Victoria , Australia
| |
Collapse
|
49
|
Ariyasinghe NR, Lyra-Leite DM, McCain ML. Engineering cardiac microphysiological systems to model pathological extracellular matrix remodeling. Am J Physiol Heart Circ Physiol 2018; 315:H771-H789. [PMID: 29906229 PMCID: PMC6230901 DOI: 10.1152/ajpheart.00110.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/27/2018] [Accepted: 06/08/2018] [Indexed: 12/11/2022]
Abstract
Many cardiovascular diseases are associated with pathological remodeling of the extracellular matrix (ECM) in the myocardium. ECM remodeling is a complex, multifactorial process that often contributes to declines in myocardial function and progression toward heart failure. However, the direct effects of the many forms of ECM remodeling on myocardial cell and tissue function remain elusive, in part because conventional model systems used to investigate these relationships lack robust experimental control over the ECM. To address these shortcomings, microphysiological systems are now being developed and implemented to establish direct relationships between distinct features in the ECM and myocardial function with unprecedented control and resolution in vitro. In this review, we will first highlight the most prominent characteristics of ECM remodeling in cardiovascular disease and describe how these features can be mimicked with synthetic and natural biomaterials that offer independent control over multiple ECM-related parameters, such as rigidity and composition. We will then detail innovative microfabrication techniques that enable precise regulation of cellular architecture in two and three dimensions. We will also describe new approaches for quantifying multiple aspects of myocardial function in vitro, such as contractility, action potential propagation, and metabolism. Together, these collective technologies implemented as cardiac microphysiological systems will continue to uncover important relationships between pathological ECM remodeling and myocardial cell and tissue function, leading to new fundamental insights into cardiovascular disease, improved human disease models, and novel therapeutic approaches.
Collapse
Affiliation(s)
- Nethika R Ariyasinghe
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California , Los Angeles, California
| | - Davi M Lyra-Leite
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California , Los Angeles, California
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California , Los Angeles, California
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California , Los Angeles, California
| |
Collapse
|
50
|
Fulghum K, Hill BG. Metabolic Mechanisms of Exercise-Induced Cardiac Remodeling. Front Cardiovasc Med 2018; 5:127. [PMID: 30255026 PMCID: PMC6141631 DOI: 10.3389/fcvm.2018.00127] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/23/2018] [Indexed: 12/13/2022] Open
Abstract
Exercise has a myriad of physiological benefits that derive in part from its ability to improve cardiometabolic health. The periodic metabolic stress imposed by regular exercise appears fundamental in driving cardiovascular tissue adaptation. However, different types, intensities, or durations of exercise elicit different levels of metabolic stress and may promote distinct types of tissue remodeling. In this review, we discuss how exercise affects cardiac structure and function and how exercise-induced changes in metabolism regulate cardiac adaptation. Current evidence suggests that exercise typically elicits an adaptive, beneficial form of cardiac remodeling that involves cardiomyocyte growth and proliferation; however, chronic levels of extreme exercise may increase the risk for pathological cardiac remodeling or sudden cardiac death. An emerging theme underpinning acute as well as chronic cardiac adaptations to exercise is metabolic periodicity, which appears important for regulating mitochondrial quality and function, for stimulating metabolism-mediated exercise gene programs and hypertrophic kinase activity, and for coordinating biosynthetic pathway activity. In addition, circulating metabolites liberated during exercise trigger physiological cardiac growth. Further understanding of how exercise-mediated changes in metabolism orchestrate cell signaling and gene expression could facilitate therapeutic strategies to maximize the benefits of exercise and improve cardiac health.
Collapse
Affiliation(s)
- Kyle Fulghum
- Department of Medicine, Envirome Institute, Institute of Molecular Cardiology, Diabetes and Obesity Center, Louisville, KY, United States
- Department of Physiology, University of Louisville, Louisville, KY, United States
| | - Bradford G. Hill
- Department of Medicine, Envirome Institute, Institute of Molecular Cardiology, Diabetes and Obesity Center, Louisville, KY, United States
| |
Collapse
|