1
|
Corsetti G, Pasini E, Scarabelli TM, Romano C, Singh A, Scarabelli CC, Dioguardi FS. Importance of Energy, Dietary Protein Sources, and Amino Acid Composition in the Regulation of Metabolism: An Indissoluble Dynamic Combination for Life. Nutrients 2024; 16:2417. [PMID: 39125298 PMCID: PMC11313897 DOI: 10.3390/nu16152417] [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: 06/07/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
PURPOSE This paper aims to present a unique perspective that emphasizes the intricate interplay between energy, dietary proteins, and amino acid composition, underscoring their mutual dependence for health-related considerations. Energy and protein synthesis are fundamental to biological processes, crucial for the sustenance of life and the growth of organisms. METHODS AND RESULTS We explore the intricate relationship between energy metabolism, protein synthesis, regulatory mechanisms, protein sources, amino acid availability, and autophagy in order to elucidate how these elements collectively maintain cellular homeostasis. We underscore the vital role this dynamic interplay has in preserving cell life. CONCLUSIONS A deeper understanding of the link between energy and protein synthesis is essential to comprehend fundamental cellular processes. This insight could have a wide-ranging impact in several medical fields, such as nutrition, metabolism, and disease management.
Collapse
Affiliation(s)
- Giovanni Corsetti
- Division of Human Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25023 Brescia, Italy;
| | - Evasio Pasini
- Italian Association of Functional Medicine, 20855 Lesmo, Italy;
- Department of Clinical and Experimental Sciences, University of Brescia, 25023 Brescia, Italy
| | | | - Claudia Romano
- Division of Human Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25023 Brescia, Italy;
| | - Arashpreet Singh
- School of Osteopathic Medicine, Campbell University, Lillington, NC 27546, USA;
| | | | | |
Collapse
|
2
|
Li W, Hou Z, Li Y, Zhang X, Bao X, Hou X, Zhang H, Zhang S. Amelioration of metabolic disorders in H9C2 cardiomyocytes induced by PM 2.5 treated with vitamin C. Drug Chem Toxicol 2024; 47:347-355. [PMID: 36815321 DOI: 10.1080/01480545.2023.2181971] [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: 09/22/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023]
Abstract
OBJECTIVE Particulate matter with an aerodynamic diameter ≤2.5 μm (PM2.5) is a public health risk. We investigate PM2.5 on metabolites in cardiomyocytes and the influence of vitamin C on PM2.5 toxicity. MATERIALS AND METHODS For 24 hours, H9C2 were exposed to various concentrations of PM2.5 (0, 100, 200, 400, 800 μg/ml), after which the levels of reactive oxygen species (ROS) and cell viability were measured using the cell counting kit-8 (CCK-8) and 2',7'-dichlorofluoresceindiacetate (DCFH2-DA), respectively. H9C2 were treated with PM2.5 (200 μg/ml) in the presence or absence of vitamin C (40 μmol/L). mRNA levels of interleukin 6(IL-6), caspase-3, fatty acid-binding protein 3 (FABP3), and hemeoxygenase-1 (HO-1) were investigated by quantitative reverse-transcription polymerase chain reaction. Non-targeted metabolomics by LC-MS/MS was applied to evaluate the metabolic profile in the cell. RESULTS Results revealed a concentration-dependent reduction in cell viability, death, ROS, and increased expression of caspase-3, FABP3, and IL-6. In total, 15 metabolites exhibited significant differential expression (FC > 2, p < 0.05) between the control and PM2.5 group. In the PM2.5 group, lysophosphatidylcholines (LysoPC,3/3) were upregulated, whereas amino acids (5/5), amino acid analogues (3/3), and other acids and derivatives (4/4) were downregulated. PM2.5 toxicity was lessened by vitamin C. It reduced PM2.5-induced elevation of LysoPC (16:0), LysoPC (16:1), and LysoPC (18:1). DISCUSSION AND CONCLUSIONS PM2.5 induces metabolic disorders in H9C2 cardiomyocytes that can be ameliorated by treatment with vitamin C.
Collapse
Affiliation(s)
- Wenjie Li
- Department of Clinical Laboratory, Anyang Center for Disease Control and Prevention, Anyang, Henan, P.R. China
| | - Ziyuan Hou
- Department of Clinical Laboratory, Anyang Center for Disease Control and Prevention, Anyang, Henan, P.R. China
| | - Yang Li
- Department of Clinical Laboratory, Anyang Center for Disease Control and Prevention, Anyang, Henan, P.R. China
- The State Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, P.R. China
| | - Xiangping Zhang
- Department of Clinical Laboratory, Anyang Center for Disease Control and Prevention, Anyang, Henan, P.R. China
| | - Xiaobing Bao
- Department of Clinical Laboratory, Anyang Center for Disease Control and Prevention, Anyang, Henan, P.R. China
| | - Xiaoyan Hou
- Department of Clinical Laboratory, Anyang Center for Disease Control and Prevention, Anyang, Henan, P.R. China
| | - Hongjin Zhang
- Department of Clinical Laboratory, Anyang Center for Disease Control and Prevention, Anyang, Henan, P.R. China
| | - Shuanhu Zhang
- Department of Clinical Laboratory, Anyang Center for Disease Control and Prevention, Anyang, Henan, P.R. China
| |
Collapse
|
3
|
Lal H, Verma SK, Wang Y, Xie M, Young ME. Circadian Rhythms in Cardiovascular Metabolism. Circ Res 2024; 134:635-658. [PMID: 38484029 PMCID: PMC10947116 DOI: 10.1161/circresaha.123.323520] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/23/2024] [Indexed: 03/19/2024]
Abstract
Energetic demand and nutrient supply fluctuate as a function of time-of-day, in alignment with sleep-wake and fasting-feeding cycles. These daily rhythms are mirrored by 24-hour oscillations in numerous cardiovascular functional parameters, including blood pressure, heart rate, and myocardial contractility. It is, therefore, not surprising that metabolic processes also fluctuate over the course of the day, to ensure temporal needs for ATP, building blocks, and metabolism-based signaling molecules are met. What has become increasingly clear is that in addition to classic signal-response coupling (termed reactionary mechanisms), cardiovascular-relevant cells use autonomous circadian clocks to temporally orchestrate metabolic pathways in preparation for predicted stimuli/stresses (termed anticipatory mechanisms). Here, we review current knowledge regarding circadian regulation of metabolism, how metabolic rhythms are synchronized with cardiovascular function, and whether circadian misalignment/disruption of metabolic processes contribute toward the pathogenesis of cardiovascular disease.
Collapse
Affiliation(s)
- Hind Lal
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Suresh Kumar Verma
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yajing Wang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Min Xie
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Martin E. Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
4
|
Meng S, Yu Y, Yu S, Zhu S, Shi M, Xiang M, Ma H. Advances in Metabolic Remodeling and Intervention Strategies in Heart Failure. J Cardiovasc Transl Res 2024; 17:36-55. [PMID: 37843752 DOI: 10.1007/s12265-023-10443-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/27/2023] [Indexed: 10/17/2023]
Abstract
The heart is the most energy-demanding organ throughout the whole body. Perturbations or failure in energy metabolism contributes to heart failure (HF), which represents the advanced stage of various heart diseases. The poor prognosis and huge economic burden associated with HF underscore the high unmet need to explore novel therapies targeting metabolic modulators beyond conventional approaches focused on neurohormonal and hemodynamic regulators. Emerging evidence suggests that alterations in metabolic substrate reliance, metabolic pathways, metabolic by-products, and energy production collectively regulate the occurrence and progression of HF. In this review, we provide an overview of cardiac metabolic remodeling, encompassing the utilization of free fatty acids, glucose metabolism, ketone bodies, and branched-chain amino acids both in the physiological condition and heart failure. Most importantly, the latest advances in pharmacological interventions are discussed as a promising therapeutic approach to restore cardiac function, drawing insights from recent basic research, preclinical and clinical studies.
Collapse
Affiliation(s)
- Simin Meng
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Yi Yu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Shuo Yu
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Shiyu Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Mengjia Shi
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
5
|
Shu L, Wang Y, Huang W, Fan S, Pan J, Lv Q, Wang L, Wang Y, Xu J, Yan H, Bai Y, Wang Y, Li Y. Integrating Metabolomics and Network Pharmacology to Explore the Mechanism of Tongmai Yangxin Pills in Ameliorating Doxorubicin-Induced Cardiotoxicity. ACS OMEGA 2023; 8:18128-18139. [PMID: 37251132 PMCID: PMC10210219 DOI: 10.1021/acsomega.3c01441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023]
Abstract
Doxorubicin (DOX) is a broad-spectrum chemotherapeutic drug used in clinical treatment of malignant tumors. It has a high anticancer activity but also high cardiotoxicity. The aim of this study was to explore the mechanism of Tongmai Yangxin pills (TMYXPs) in ameliorating DOX-induced cardiotoxicity through integrated metabolomics and network pharmacology. In this study, first, an ultrahigh-performance liquid chromatography-quadrupole-time-of-flight/mass spectrometry (UPLC-Q-TOF/MS) metabonomics strategy was established to obtain metabolite information and potential biomarkers were determined after data processing. Second, network pharmacological analysis was used to evaluate the active components, drug-disease targets, and key pathways of TMYXPs to alleviate DOX-induced cardiotoxicity. Targets from the network pharmacology analysis and metabolites from plasma metabolomics were jointly analyzed to select crucial metabolic pathways. Finally, the related proteins were verified by integrating the above results and the possible mechanism of TMYXPs to alleviate DOX-induced cardiotoxicity was studied. After metabolomics data processing, 17 different metabolites were screened, and it was found that TMYXPs played a role in myocardial protection mainly by affecting the tricarboxylic acid (TCA) cycle of myocardial cells. A total of 71 targets and 20 related pathways were screened out with network pharmacological analysis. Based on the combined analysis of 71 targets and different metabolites, TMYXPs probably played a role in myocardial protection through regulating upstream proteins of the insulin signaling pathway, MAPK signaling pathway, and p53 signaling pathway, as well as the regulation of metabolites related to energy metabolism. They then further affected the downstream Bax/Bcl-2-Cyt c-caspase-9 axis, inhibiting the myocardial cell apoptosis signaling pathway. The results of this study may contribute to the clinical application of TMYXPs in DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Lexin Shu
- School
of Chinese Materia Medica, Tianjin University
of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuming Wang
- School
of Chinese Materia Medica, Tianjin University
of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wei Huang
- School
of Chinese Materia Medica, Tianjin University
of Traditional Chinese Medicine, Tianjin 301617, China
| | - Simiao Fan
- School
of Chinese Materia Medica, Tianjin University
of Traditional Chinese Medicine, Tianjin 301617, China
| | - Junhua Pan
- Hainan
Province Key Laboratory for Drug Preclinical Study of Pharmacology
and Toxicology Research, Hainan Medical
University, Haikou 571199, China
| | - Qingbo Lv
- Institute
of Traditional Chinese Medicine, Tianjin University of Traditional
Chinese Medicine, Tianjin 301617, China
| | - Lin Wang
- Tianjin
Zhongxin Pharmaceutical Group Co., Ltd., Le Ren Tang Pharmaceutical
Factory, Tianjin 301617, China
| | - Yujing Wang
- Tianjin
Zhongxin Pharmaceutical Group Co., Ltd., Le Ren Tang Pharmaceutical
Factory, Tianjin 301617, China
| | - Jinpeng Xu
- Tianjin
Zhongxin Pharmaceutical Group Co., Ltd., Tianjin 301617, China
| | - Haifeng Yan
- Institute
of Traditional Chinese Medicine, Tianjin University of Traditional
Chinese Medicine, Tianjin 301617, China
| | - Yuchao Bai
- School
of Chinese Materia Medica, Tianjin University
of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yi Wang
- Institute
of Traditional Chinese Medicine, Tianjin University of Traditional
Chinese Medicine, Tianjin 301617, China
| | - Yubo Li
- School
of Chinese Materia Medica, Tianjin University
of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
6
|
Qin X, Zhang Y, Zheng Q. Metabolic Inflexibility as a Pathogenic Basis for Atrial Fibrillation. Int J Mol Sci 2022; 23:ijms23158291. [PMID: 35955426 PMCID: PMC9368187 DOI: 10.3390/ijms23158291] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Atrial fibrillation (AF), the most common sustained arrhythmia, is closely intertwined with metabolic abnormalities. Recently, a metabolic paradox in AF pathogenesis has been suggested: under different forms of pathogenesis, the metabolic balance shifts either towards (e.g., obesity and diabetes) or away from (e.g., aging, heart failure, and hypertension) fatty acid oxidation, yet they all increase the risk of AF. This has raised the urgent need for a general consensus regarding the metabolic changes that predispose patients to AF. “Metabolic flexibility” aptly describes switches between substrates (fatty acids, glucose, amino acids, and ketones) in response to various energy stresses depending on availability and requirements. AF, characterized by irregular high-frequency excitation and the contraction of the atria, is an energy challenge and triggers a metabolic switch from preferential fatty acid utilization to glucose metabolism to increase the efficiency of ATP produced in relation to oxygen consumed. Therefore, the heart needs metabolic flexibility. In this review, we will briefly discuss (1) the current understanding of cardiac metabolic flexibility with an emphasis on the specificity of atrial metabolic characteristics; (2) metabolic heterogeneity among AF pathogenesis and metabolic inflexibility as a common pathological basis for AF; and (3) the substrate-metabolism mechanism underlying metabolic inflexibility in AF pathogenesis.
Collapse
Affiliation(s)
- Xinghua Qin
- Xi’an Key Laboratory of Special Medicine and Health Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China;
| | - Yudi Zhang
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China;
| | - Qiangsun Zheng
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China;
- Correspondence: or
| |
Collapse
|
7
|
Ouyang H, Gao X, Zhang J. Impaired expression of BCAT1 relates to muscle atrophy of mouse model of sarcopenia. BMC Musculoskelet Disord 2022; 23:450. [PMID: 35562710 PMCID: PMC9102634 DOI: 10.1186/s12891-022-05332-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/13/2022] [Indexed: 12/13/2022] Open
Abstract
Background The underlying mechanism of muscle atrophy in sarcopenia is still not fully understood; branched chain aminotransferase 1(BCAT1) isocitrate dehydrogenase-1 encodes an evolutionarily conserved cytoplasmic aminotransferase for glutamate and branched-chain amino acids (BCAAs), thus constituting a regulatory component of cytoplasmic amino and keto acid metabolism. In human gliomas carrying wild-type isocitrate dehydrogenase-1, BCAT1 promotes cell proliferation through amino acid catabolism. Hence, the goals of this study were to unravel the potential role of BCAT1 expression in muscle atrophy and to explore the mechanisms underlying this process. Methods We first measured Bcat1 expression by RT-qPCR and western blotting in murine and cellular models of muscle atrophy. To understand how the Bcat1-driven changes sustained muscle cell growth, we analyzed reactive oxygen species (ROS) levels and activation of the mTORC1/S6K1 pathway in muscle cells. Furthermore, we performed Cell Counting Kit-8(CCK8) assays and fluorescence staining to evaluate growth rate of cells and ROS levels. Finally, we verified that depletion of Bcat1 impairs the growth rate of muscle cells and increases ROS levels, indicating that muscle atrophy resulted from the downregulation of the mTORC1/S6K1 pathway. Data were analyzed by two-tailed unpaired Student’s t-test or Mann-Whitney U test for two groups to determine statistical significance. Statistical analyses were performed using GraphPad Prism version 6.0 and SPSS 16.0 software. Results Bcat1 expression level in skeletal muscles was lower in murine and cellular models of sarcopenia than in the control groups. Bcat1 knockdown not only suppressed the growth of muscle cells but also increased the production of ROS. Impaired cell growth and increased ROS production was rescued by co-introduction of an shRNA-resistant Bcat1 cDNA or addition of the mTORC1 stimulator MYH1485. Muscle cells with Bcat1 knockdown featured lower mTORC1 and S6K1 phosphorylation (pS6K1) than NT muscle cells. Addition of either shRNA-resistant Bcat1 cDNA or MYH1485 rescued the suppression of cell growth, increase in ROS production, and decrease in pS6K1. Conclusions The branched chain amino acids catabolic enzyme BCAT1 is essential for the growth of muscle cells. BCAT1 expression contributes to sustained growth of muscle cells by activating mTOR signaling and reducing ROS production. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-022-05332-7.
Collapse
Affiliation(s)
- Hui Ouyang
- Department of Neuromedicine, Peking University People's Hospital, 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Xuguang Gao
- Department of Neuromedicine, Peking University People's Hospital, 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Jun Zhang
- Department of Neuromedicine, Peking University People's Hospital, 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| |
Collapse
|
8
|
Abstract
The cardiomyocyte circadian clock temporally governs fundamental cellular processes, leading to 24-h rhythms in cardiac properties (such as electrophysiology and contractility). The importance of this cell-autonomous clock is underscored by reports that the disruption of the mechanism leads to adverse cardiac remodeling and heart failure. In healthy non-stressed mice, the cardiomyocyte circadian clock modestly augments both cardiac protein synthesis (~14%) and mass (~11%) at the awake-to-sleep transition (relative to their lowest values in the middle of the awake period). However, the increased capacity for cardiac growth at the awake-to-sleep transition exacerbates the responsiveness of the heart to pro-hypertrophic stimuli/stresses (e.g., adrenergic stimulation, nutrients) at this time. The cardiomyocyte circadian clock orchestrates time-of-day-dependent rhythms in cardiac growth through numerous mechanisms. Both ribosomal RNA (e.g., 28S) and the PI3K/AKT/mTOR/S6 signaling axis are circadian regulated, peaking at the awake-to-sleep transition in the heart. Conversely, the negative regulators of translation (including PER2, AMPK, and the integrated stress response) are elevated in the middle of the awake period in a coordinated fashion. We speculate that persistent circadian governance of cardiac growth during non-dipping/nocturnal hypertension, sleep apnea, and/or shift work may exacerbate left ventricular hypertrophy and cardiac disease development, highlighting a need for the advancement of chronotherapeutic interventions.
Collapse
Affiliation(s)
| | - Martin E. Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| |
Collapse
|
9
|
Boldt K, Joumaa V, Turnbull J, Fedak PW, Herzog W. A high-whey-protein diet does not enhance mechanical and structural remodeling of cardiac muscle in response to aerobic exercise in rats. Phys Act Nutr 2022; 26:28-38. [PMID: 35510443 PMCID: PMC9081358 DOI: 10.20463/pan.2022.0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/18/2022] [Accepted: 03/30/2022] [Indexed: 11/22/2022] Open
Abstract
PURPOSE Aerobic exercise training results in distinct structural and mechanical myocardial adaptations. In skeletal muscle, whey protein supplementation is effective in enhancing muscle adaptation following resistance exercise. However, it is unclear whether cardiac adaptation to aerobic exercise can be enhanced by systematic protein supplementation. METHODS Twelve-week-old rats were assigned to 12 weeks of either sedentary or aerobic exercise with either a standard (Sed+Standard, Ex+Standard) or high-protein (Sed+Pro, Ex+Pro) diet. Echocardiography was used to measure cardiac structural remodeling and performance. Skinned cardiac fiber bundles were used to determine the active and passive stress properties, maximum shortening velocity, and calcium sensitivity. RESULTS Aerobic training was characterized structurally by increases in ventricle volume (Ex+Standard, 19%; Ex+Pro, 29%) and myocardial thickness (Ex+Standard, 26%; Ex+- Pro, 12%) compared to that of baseline. Skinned trabecula r fiber bundles also had a greater unloaded shortening velocity (Sed+Standard, 1.04±0.05; Sed+Pro, 1.07±0.03; Ex- +Standard, 1.16±0.04; Ex+Pro, 1.18±0.05 FL/s) and calcium sensitivity (pCa50: Sed+Standard, 6.04±0.17; Sed+Pro, 6.08±0.19; Ex+Standard, 6.30±0.09; Ex+Pro, 6.36±0.12) in trained hearts compared to that of hearts from sedentary animals. However, the addition of a high-protein diet did not provide additional benefits to either the structural or mechanical adaptations of the myocardium. CONCLUSION Therefore, it seems that a high-whey-protein diet does not significantly enhance adaptations of the heart to aerobic exercise in comparison to that of a standard diet.
Collapse
Affiliation(s)
- Kevin Boldt
- Human Performance Laboratory, University of Calgary, Calgary, Canada
| | - Venus Joumaa
- Human Performance Laboratory, University of Calgary, Calgary, Canada
| | - Jeannine Turnbull
- Health Center, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Paul W.M. Fedak
- Health Center, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Walter Herzog
- Human Performance Laboratory, University of Calgary, Calgary, Canada
- Department of Cardiac Sciences, University of Calgary, Calgary, Canada
| |
Collapse
|
10
|
Amaral AG, da Silva CCC, Serna JDC, Honorato-Sampaio K, Freitas JA, Duarte-Neto AN, Bloise AC, Cassina L, Yoshinaga MY, Chaves-Filho AB, Qian F, Miyamoto S, Boletta A, Bordin S, Kowaltowski AJ, Onuchic LF. Disruption of polycystin-1 cleavage leads to cardiac metabolic rewiring in mice. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166371. [PMID: 35218894 DOI: 10.1016/j.bbadis.2022.166371] [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: 10/04/2021] [Revised: 02/15/2022] [Accepted: 02/20/2022] [Indexed: 11/18/2022]
Abstract
Cardiovascular manifestations account for marked morbi-mortality in autosomal dominant polycystic kidney disease (ADPKD). Pkd1- and Pkd2-deficient mice develop cardiac dysfunction, however the underlying mechanisms remain largely unclear. It is unknown whether impairment of polycystin-1 cleavage at the G-protein-coupled receptor proteolysis site, a significant ADPKD mutational mechanism, is involved in this process. We analyzed the impact of polycystin-1 cleavage on heart metabolism using Pkd1V/V mice, a model unable to cleave this protein and with early cardiac dysfunction. Pkd1V/V hearts showed lower levels of glucose and amino acids and higher lipid levels than wild-types, as well as downregulation of p-AMPK, p-ACCβ, CPT1B-Cpt1b, Ppara, Nppa and Acta1. These findings suggested decreased fatty acid β-oxidation, which was confirmed by lower oxygen consumption by Pkd1V/V isolated mitochondria using palmitoyl-CoA. Pkd1V/V hearts also presented increased oxygen consumption in response to glucose, suggesting that alternative substrates may be used to generate energy. Pkd1V/V hearts displayed a higher density of decreased-size mitochondria, a finding associated with lower MFN1, Parkin and BNIP3 expression. These derangements were correlated with increased apoptosis and inflammation but not hypertrophy. Notably, Pkd1V/V neonate cardiomyocytes also displayed shifts in oxygen consumption and p-AMPK downregulation, suggesting that, at least partially, the metabolic alterations are not induced by kidney dysfunction. Our findings reveal that disruption of polycystin-1 cleavage leads to cardiac metabolic rewiring in mice, expanding the understanding of heart dysfunction associated with Pkd1 deficiency and likely with human ADPKD.
Collapse
Affiliation(s)
- Andressa G Amaral
- Disciplinas de Nefrologia e Medicina Molecular, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 01246903, Brazil
| | - Camille C C da Silva
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Julian D C Serna
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Kinulpe Honorato-Sampaio
- Faculdade de Medicina, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG 31270901, Brazil
| | - Jéssica A Freitas
- Disciplinas de Nefrologia e Medicina Molecular, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 01246903, Brazil
| | - Amaro N Duarte-Neto
- Disciplina de Emergências Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 01246903, Brazil
| | - Antonio C Bloise
- Departamento de Física Aplicada, Instituto de Física, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Laura Cassina
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Marcos Y Yoshinaga
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Adriano B Chaves-Filho
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Feng Qian
- Division of Nephrology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sayuri Miyamoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Alessandra Boletta
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Silvana Bordin
- Departamento de Fisiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Luiz F Onuchic
- Disciplinas de Nefrologia e Medicina Molecular, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 01246903, Brazil.
| |
Collapse
|
11
|
Habaybeh D, de Moraes MB, Slee A, Avgerinou C. Nutritional interventions for heart failure patients who are malnourished or at risk of malnutrition or cachexia: a systematic review and meta-analysis. Heart Fail Rev 2021; 26:1103-1118. [PMID: 32124164 PMCID: PMC8310486 DOI: 10.1007/s10741-020-09937-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Malnutrition is common in heart failure (HF), and it is associated with higher hospital readmission and mortality rates. This review aims to answer the question whether nutritional interventions aiming to increase protein and energy intake are effective at improving outcomes for patients with HF who are malnourished or at risk of malnutrition or cachexia. Systematic searches of four databases (Medline, Embase, CINAHL and Cochrane Central Register of Controlled Trials (CENTRAL)) were conducted on 21 June 2019. Randomized controlled trials (RCTs) or other interventional studies using protein or energy supplementation for adult HF patients who are malnourished or at risk of malnutrition or cachexia were included. Two independent reviewers assessed study eligibility and risk of bias. Five studies (four RCTs and one pilot RCT) met the inclusion criteria. The majority of studies were small and of limited quality. The pooled weighted mean difference (WMD) for body weight showed a benefit from the nutritional intervention by 3.83 kg (95% confidence interval (CI) 0.17 to 7.50, P = 0.04) from three trials with no significant benefit for triceps skinfold thickness (WMD = - 2.14 mm, 95% CI - 9.07 to 4.79, P = 0.55) from two trials. The combination of personalized nutrition intervention with conventional treatment led to a decrease in all-cause mortality and hospital readmission in one study. Findings of this review suggest that nutritional interventions could potentially improve outcomes in HF patients who are malnourished or at risk of malnutrition. However, the strength of the evidence is poor, and more robust studies with a larger number of participants are needed.
Collapse
Affiliation(s)
- Dina Habaybeh
- Division of Medicine, University College London, London, UK
| | | | - Adrian Slee
- Division of Medicine, University College London, London, UK
| | - Christina Avgerinou
- Department of Primary Care and Population Health, University College London, Rowland Hill Street, London, NW3 2PF, UK.
| |
Collapse
|
12
|
Pasqua T, Rocca C, Giglio A, Angelone T. Cardiometabolism as an Interlocking Puzzle between the Healthy and Diseased Heart: New Frontiers in Therapeutic Applications. J Clin Med 2021; 10:721. [PMID: 33673114 PMCID: PMC7918460 DOI: 10.3390/jcm10040721] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiac metabolism represents a crucial and essential connecting bridge between the healthy and diseased heart. The cardiac muscle, which may be considered an omnivore organ with regard to the energy substrate utilization, under physiological conditions mainly draws energy by fatty acids oxidation. Within cardiomyocytes and their mitochondria, through well-concerted enzymatic reactions, substrates converge on the production of ATP, the basic chemical energy that cardiac muscle converts into mechanical energy, i.e., contraction. When a perturbation of homeostasis occurs, such as an ischemic event, the heart is forced to switch its fatty acid-based metabolism to the carbohydrate utilization as a protective mechanism that allows the maintenance of its key role within the whole organism. Consequently, the flexibility of the cardiac metabolic networks deeply influences the ability of the heart to respond, by adapting to pathophysiological changes. The aim of the present review is to summarize the main metabolic changes detectable in the heart under acute and chronic cardiac pathologies, analyzing possible therapeutic targets to be used. On this basis, cardiometabolism can be described as a crucial mechanism in keeping the physiological structure and function of the heart; furthermore, it can be considered a promising goal for future pharmacological agents able to appropriately modulate the rate-limiting steps of heart metabolic pathways.
Collapse
Affiliation(s)
- Teresa Pasqua
- Department of Health Science, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy
| | - Anita Giglio
- Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy;
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, E. and E.S. (Di.B.E.S.T.), University of Calabria, 87036 Rende (CS), Italy
- National Institute of Cardiovascular Research (I.N.R.C.), 40126 Bologna, Italy
| |
Collapse
|
13
|
Saleem TH, Algowhary M, Kamel FEM, El-Mahdy RI. Plasma amino acid metabolomic pattern in heart failure patients with either preserved or reduced ejection fraction: The relation to established risk variables and prognosis. Biomed Chromatogr 2020; 35:e5012. [PMID: 33119901 DOI: 10.1002/bmc.5012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/08/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022]
Abstract
Communication between amino acids (AAs) and heart failure (HF) is unclear. We evaluate the plasma metabolomic profile of AAs in HF and its subgroups and association with clinical features. This is a case-control study in which 90 patients with HF, 63 with reduced ejection fraction (HFrEF) and 27 with preserved ejection fraction (HFpEF), were compared with 60 controls. The quantitative measurement of plasma concentrations of AA metabolites was performed using an AA analyzer. Compared with controls, HF patients had significantly higher levels of nine AAs and significantly lower levels of seven AAs. Leu, phenylalanine (Phe), and methionine (Met) were the independent predictors of HF that remained significant after adjustment for confounding factors in multivariate analysis. There was a significant difference in 10 AAs and some clinical features between HFpEF and HFrEF. The plasma levels of six AAs were significantly increased across the different New York Heart Association (NYHA) classes, (class II, class III, class IV) but they were not predictor of reduced EF and NYHA in multivariate regression analysis. There were significant associations between Leu, Phe, and Met with cardiovascular risk variables and prognosis. In conclusion, plasma Leu, Phe, and Met provide early prediction and prognostic values of HF. The plasma AAs could have significant impact on the risk-stratifying HFrEF and HFpEF and NYHA functional class but do not predict them.
Collapse
Affiliation(s)
- Tahia H Saleem
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Magdy Algowhary
- Department of Cardiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Fatma Elzahraa M Kamel
- Pharmacist at Cardiology Department, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Reham I El-Mahdy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
14
|
Yue S, Ding S, Zhou J, Yang C, Hu X, Zhao X, Wang Z, Wang L, Peng Q, Xue B. Metabolomics Approach Explore Diagnostic Biomarkers and Metabolic Changes in Heat-Stressed Dairy Cows. Animals (Basel) 2020; 10:E1741. [PMID: 32992834 PMCID: PMC7601318 DOI: 10.3390/ani10101741] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023] Open
Abstract
In the present experiment, we investigated the impact of heat stress (HS) on physiological parameters, dry matter intake, milk production, the metabolome of milk, and blood plasma in lactating Holstein dairy cows. For this purpose, 20 Holstein lactating cows were distributed in two groups in such a way that each group had 10 cows. A group of 10 cows was reared in HS conditions, while the other group of 10 cows was reared in the thermoneutral zone. The results of the experiment showed that cows subjected to HS had higher respiration rates (p < 0.01) and greater rectal temperature (p < 0.01). Results of milk production and composition explored that HS lowered milk production (p < 0.01) and milk protein percentage (p < 0.05) than cows raised in a thermoneutral place. Furthermore, HS increased the concentrations of N-acetyl glycoprotein, scyllo-inositol, choline, and pyridoxamine in milk, while HS decreased the concentrations of O-acetyl glycoprotein, glycerophosphorylcholine, citrate, and methyl phosphate in milk. Moreover, HS enhanced plasma concentrations of alanine, glucose, glutamate, urea, 1-methylhistidine, histidine, and formate in cows, while the plasma concentration of low-density lipoprotein, very-low-density lipoprotein, leucine, lipid, and 3-hydroxybutyrate decreased due to HS. Based on the findings of the current research, it is concluded that HS alters the milk and blood plasma metabolites of lactating Holstein dairy cows. Overall, in the current experiment, HS altered eight metabolites in milk and twelve metabolites in the plasma of lactating Holstein dairy cows. Furthermore, the current study explored that these metabolites were mainly involved in proteolysis, gluconeogenesis, and milk fatty acid synthesis and could be potential biomarkers for dairy cows undergoing HS.
Collapse
Affiliation(s)
- Shuangming Yue
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.Y.); (S.D.); (J.Z.); (X.H.); (X.Z.); (Z.W.); (L.W.); (Q.P.)
- Department of Bioengineering, Sichuan Water Conservancy Vocation College, Chengdu 611845, China;
| | - Siyan Ding
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.Y.); (S.D.); (J.Z.); (X.H.); (X.Z.); (Z.W.); (L.W.); (Q.P.)
| | - Jia Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.Y.); (S.D.); (J.Z.); (X.H.); (X.Z.); (Z.W.); (L.W.); (Q.P.)
| | - Chao Yang
- Department of Bioengineering, Sichuan Water Conservancy Vocation College, Chengdu 611845, China;
| | - Xiaofei Hu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.Y.); (S.D.); (J.Z.); (X.H.); (X.Z.); (Z.W.); (L.W.); (Q.P.)
| | - Xiaonan Zhao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.Y.); (S.D.); (J.Z.); (X.H.); (X.Z.); (Z.W.); (L.W.); (Q.P.)
| | - Zhisheng Wang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.Y.); (S.D.); (J.Z.); (X.H.); (X.Z.); (Z.W.); (L.W.); (Q.P.)
| | - Lizhi Wang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.Y.); (S.D.); (J.Z.); (X.H.); (X.Z.); (Z.W.); (L.W.); (Q.P.)
| | - Quanhui Peng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.Y.); (S.D.); (J.Z.); (X.H.); (X.Z.); (Z.W.); (L.W.); (Q.P.)
| | - Bai Xue
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.Y.); (S.D.); (J.Z.); (X.H.); (X.Z.); (Z.W.); (L.W.); (Q.P.)
| |
Collapse
|
15
|
Mehta A, Liu C, Nayak A, Tahhan AS, Ko YA, Dhindsa DS, Kim JH, Hayek SS, Sperling LS, Mehta PK, Sun YV, Uppal K, Jones DP, Quyyumi AA. Untargeted high-resolution plasma metabolomic profiling predicts outcomes in patients with coronary artery disease. PLoS One 2020; 15:e0237579. [PMID: 32810196 PMCID: PMC7444579 DOI: 10.1371/journal.pone.0237579] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/29/2020] [Indexed: 12/22/2022] Open
Abstract
Objective Patients with CAD have substantial residual risk of mortality, and whether hitherto unknown small-molecule metabolites and metabolic pathways contribute to this risk is unclear. We sought to determine the predictive value of plasma metabolomic profiling in patients with CAD. Approach and results Untargeted high-resolution plasma metabolomic profiling of subjects undergoing coronary angiography was performed using liquid chromatography/mass spectrometry. Metabolic features and pathways associated with mortality were identified in 454 subjects using metabolome-wide association studies and Mummichog, respectively, and validated in 322 subjects. A metabolomic risk score comprising of log-transformed HR estimates of metabolites that associated with mortality and passed LASSO regression was created and its performance validated. In 776 subjects (66.8 years, 64% male, 17% Black), 433 and 357 features associated with mortality (FDR-adjusted q<0.20); and clustered into 21 and 9 metabolic pathways in first and second cohorts, respectively. Six pathways (urea cycle/amino group, tryptophan, aspartate/asparagine, lysine, tyrosine, and carnitine shuttle) were common. A metabolomic risk score comprising of 7 metabolites independently predicted mortality in the second cohort (HR per 1-unit increase 2.14, 95%CI 1.62, 2.83). Adding the score to a model of clinical predictors improved risk discrimination (delta C-statistic 0.039, 95%CI -0.006, 0.086; and Integrated Discrimination Index 0.084, 95%CI 0.030, 0.151) and reclassification (continuous Net Reclassification Index 23.3%, 95%CI 7.9%, 38.2%). Conclusions Differential regulation of six metabolic pathways involved in myocardial energetics and systemic inflammation is independently associated with mortality in patients with CAD. A novel risk score consisting of representative metabolites is highly predictive of mortality.
Collapse
Affiliation(s)
- Anurag Mehta
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Chang Liu
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Aditi Nayak
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Ayman S. Tahhan
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Yi-An Ko
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Devinder S. Dhindsa
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Jeong Hwan Kim
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Salim S. Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Laurence S. Sperling
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Puja K. Mehta
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Yan V. Sun
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
- Atlanta VA Health Care System, Decatur, Georgia
| | - Karan Uppal
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Dean P. Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Arshed A. Quyyumi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
- * E-mail:
| |
Collapse
|
16
|
Designing Novel Therapies to Mend Broken Hearts: ATF6 and Cardiac Proteostasis. Cells 2020; 9:cells9030602. [PMID: 32138230 PMCID: PMC7140506 DOI: 10.3390/cells9030602] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/28/2020] [Indexed: 12/12/2022] Open
Abstract
The heart exhibits incredible plasticity in response to both environmental and genetic alterations that affect workload. Over the course of development, or in response to physiological or pathological stimuli, the heart responds to fluctuations in workload by hypertrophic growth primarily by individual cardiac myocytes growing in size. Cardiac hypertrophy is associated with an increase in protein synthesis, which must coordinate with protein folding and degradation to allow for homeostatic growth without affecting the functional integrity of cardiac myocytes (i.e., proteostasis). This increase in the protein folding demand in the growing cardiac myocyte activates the transcription factor, ATF6 (activating transcription factor 6α, an inducer of genes that restore proteostasis. Previously, ATF6 has been shown to induce ER-targeted proteins functioning primarily to enhance ER protein folding and degradation. More recent studies, however, have illuminated adaptive roles for ATF6 functioning outside of the ER by inducing non-canonical targets in a stimulus-specific manner. This unique ability of ATF6 to act as an initial adaptive responder has bolstered an enthusiasm for identifying small molecule activators of ATF6 and similar proteostasis-based therapeutics.
Collapse
|
17
|
Cardiac ketone body metabolism. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165739. [PMID: 32084511 DOI: 10.1016/j.bbadis.2020.165739] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/11/2020] [Accepted: 02/15/2020] [Indexed: 12/14/2022]
Abstract
The ketone bodies, d-β-hydroxybutyrate and acetoacetate, are soluble 4-carbon compounds derived principally from fatty acids, that can be metabolised by many oxidative tissues, including heart, in carbohydrate-depleted conditions as glucose-sparing energy substrates. They also have important signalling functions, acting through G-protein coupled receptors and histone deacetylases to regulate metabolism and gene expression including that associated with anti-oxidant activity. Their concentration, and hence availability, increases in diabetes mellitus and heart failure. Whilst known to be substrates for ATP production, especially in starvation, their role(s) in the heart, and in heart disease, is uncertain. Recent evidence, reviewed here, indicates that increased ketone body metabolism is a feature of heart failure, and is accompanied by other changes in substrate selection. Whether the change in myocardial ketone body metabolism is adaptive or maladaptive is unknown, but it offers the possibility of using exogenous ketones to treat the failing heart.
Collapse
|
18
|
Zhang Y, Ji X, Ku T, Li B, Li G, Sang N. Ambient fine particulate matter exposure induces cardiac functional injury and metabolite alterations in middle-aged female mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 248:121-132. [PMID: 30784831 DOI: 10.1016/j.envpol.2019.01.080] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 01/17/2019] [Accepted: 01/19/2019] [Indexed: 06/09/2023]
Abstract
Plenty of epidemiological studies have shown that exposure to ambient particulate matter (PM2.5) is linked to cardiovascular diseases (CVDs) in older even in middle-aged populations; however, experimental evidence through intuitive metabolic analysis to confirm the age susceptibility and explain the related molecular mechanism of PM2.5-induced cardiotoxicity is relatively rare. In the present study, C57BL/6 mice (adult (4-month) and middle-aged (10-month)) were given 3 mg/kg PM2.5 every other day by oropharyngeal aspiration for 4 weeks, and then, body and cardiac parameter, containing weight data, cardiac function, ultrastructure, metabolic analysis, and molecular detection were conducted to investigate the PM2.5-induced cardiotoxicity. The results indicated that middle-aged mice were more susceptible to PM2.5, displaying slow cardiac growth, cardiac dysfunction, abnormal mitochondrial structure and function, and cardiac metabolic disorders. The altered metabolites were enriched in carbohydrate metabolism, fatty acid metabolism, amino acid metabolism, nucleotide metabolism and nicotinate and nicotinamide metabolism. In conclusion, we speculated that the cardiac metabolic disorders may be important factors in PM2.5-induced cardiac dysfunction and mitochondrial structure destruction in middle-aged mice, providing a new direction for the study of the association between PM2.5 and CVDs.
Collapse
Affiliation(s)
- Yingying Zhang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Xiaotong Ji
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Tingting Ku
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Ben Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, PR China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, PR China.
| |
Collapse
|
19
|
Plasma Amino Acid Abnormalities in Chronic Heart Failure. Mechanisms, Potential Risks and Targets in Human Myocardium Metabolism. Nutrients 2017; 9:nu9111251. [PMID: 29140312 PMCID: PMC5707723 DOI: 10.3390/nu9111251] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 10/31/2017] [Accepted: 11/09/2017] [Indexed: 12/20/2022] Open
Abstract
The goal of this study was to measure arterial amino acid levels in patients with chronic heart failure (CHF), and relate them to left ventricular function and disease severity. Amino acids (AAs) play a crucial role for heart protein-energy metabolism. In heart failure, arterial AAs, which are the major determinant of AA uptake by the myocardium, are rarely measured. Forty-one subjects with clinically stable CHF (New York Heart Association (NYHA) class II to IV) were analyzed. After overnight fasting, blood samples from the radial artery were taken to measure AA concentrations. Calorie (KcalI), protein-, fat-, carbohydrate-intake, resting energy expenditure (REE), total daily energy expenditure (REE × 1.3), and cardiac right catheterization variables were all measured. Eight matched controls were compared for all measurements, with the exception of cardiac catheterization. Compared with controls, CHF patients had reduced arterial AA levels, of which both their number and reduced rates are related to Heart Failure (HF) severity. Arterial aspartic acid correlated with stroke volume index (r = 0.6263; p < 0.0001) and cardiac index (r = 0.4243; p = 0.0028). The value of arterial aspartic acid (µmol/L) multiplied by the cardiac index was associated with left ventricular ejection fraction (r = 0.3765; p = 0.0076). All NYHA groups had adequate protein intake (≥1.1 g/kg/day) and inadequate calorie intake (KcalI < REE × 1.3) was found only in class IV patients. This study showed that CHF patients had reduced arterial AA levels directly related to clinical disease severity and left ventricular dysfunction.
Collapse
|
20
|
Cheema BS, Sabbah HN, Greene SJ, Gheorghiade M. Protein turnover in the failing heart: an ever-changing landscape. Eur J Heart Fail 2017; 19:1218-1221. [PMID: 28805968 DOI: 10.1002/ejhf.905] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/01/2017] [Accepted: 05/10/2017] [Indexed: 01/09/2023] Open
Affiliation(s)
- Baljash S Cheema
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hani N Sabbah
- Division of Cardiovascular Medicine, Department of Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Stephen J Greene
- Duke Clinical Research Institute, Durham, NC, USA.,Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Mihai Gheorghiade
- Center for Cardiovascular Innovation, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
21
|
Guo X, Huang C, Lian K, Wang S, Zhao H, Yan F, Zhang X, Zhang J, Xie H, An R, Tao L. BCKA down-regulates mTORC2-Akt signal and enhances apoptosis susceptibility in cardiomyocytes. Biochem Biophys Res Commun 2016; 480:106-113. [PMID: 27697526 DOI: 10.1016/j.bbrc.2016.09.162] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 09/29/2016] [Indexed: 12/15/2022]
Abstract
Diabetic mellitus (DM) portends poor prognosis concerning pressure overloaded heart disease. Branched-chain amino acids (BCAAs), elements of essential amino acids, have been found altered in its catabolism in diabetes decades ago. However, the relationship between BCAAs and DM induced deterioration of pressure overloaded heart disease remains controversial. This study is aimed to investigate the particular effect of BCKA, a metabolite of BCAA, on myocardial injury induced by pressure overloaded. Primary cardiomyocytes were incubated with or without BCKA and followed by treatment with isoproterenol (ISO); then cell viability was detected by CCK8 and apoptosis was examined by TUNNEL stain and caspase-3 activity analysis. Compared to non-BCKA incubated group, BCKA incubation decreased cell survival and increased apoptosis concentration dependently. Furthermore, Western blot assay showed that mTORC2-Akt pathway was significantly inactivated by BCKA incubation. Moreover, overexpression of rictor, a vital component of mTORC2, significantly abolished the adverse effects of BCKA on apoptosis susceptibility of cardiomyocytes. These results indicate that BCKA contribute to vulnerability of cardiomyocytes in stimulated stress via inactivation of mTORC2-Akt pathway.
Collapse
Affiliation(s)
- Xiong Guo
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Chong Huang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Kun Lian
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shan Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Huishou Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Feng Yan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaomeng Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jinglong Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Huaning Xie
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Rui An
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
22
|
Abstract
The heart is a biological pump that converts chemical to mechanical energy. This process of energy conversion is highly regulated to the extent that energy substrate metabolism matches energy use for contraction on a beat-to-beat basis. The biochemistry of cardiac metabolism includes the biochemistry of energy transfer, metabolic regulation, and transcriptional, translational as well as posttranslational control of enzymatic activities. Pathways of energy substrate metabolism in the heart are complex and dynamic, but all of them conform to the First Law of Thermodynamics. The perspectives expand on the overall idea that cardiac metabolism is inextricably linked to both physiology and molecular biology of the heart. The article ends with an outlook on emerging concepts of cardiac metabolism based on new molecular models and new analytical tools. © 2016 American Physiological Society. Compr Physiol 6:1675-1699, 2016.
Collapse
Affiliation(s)
- Heinrich Taegtmeyer
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston
| | - Truong Lam
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston
| | - Giovanni Davogustto
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston
| |
Collapse
|
23
|
Sun H, Olson KC, Gao C, Prosdocimo DA, Zhou M, Wang Z, Jeyaraj D, Youn JY, Ren S, Liu Y, Rau CD, Shah S, Ilkayeva O, Gui WJ, William NS, Wynn RM, Newgard CB, Cai H, Xiao X, Chuang DT, Schulze PC, Lynch C, Jain MK, Wang Y. Catabolic Defect of Branched-Chain Amino Acids Promotes Heart Failure. Circulation 2016; 133:2038-49. [PMID: 27059949 DOI: 10.1161/circulationaha.115.020226] [Citation(s) in RCA: 384] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 03/28/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Although metabolic reprogramming is critical in the pathogenesis of heart failure, studies to date have focused principally on fatty acid and glucose metabolism. Contribution of amino acid metabolic regulation in the disease remains understudied. METHODS AND RESULTS Transcriptomic and metabolomic analyses were performed in mouse failing heart induced by pressure overload. Suppression of branched-chain amino acid (BCAA) catabolic gene expression along with concomitant tissue accumulation of branched-chain α-keto acids was identified as a significant signature of metabolic reprogramming in mouse failing hearts and validated to be shared in human cardiomyopathy hearts. Molecular and genetic evidence identified the transcription factor Krüppel-like factor 15 as a key upstream regulator of the BCAA catabolic regulation in the heart. Studies using a genetic mouse model revealed that BCAA catabolic defect promoted heart failure associated with induced oxidative stress and metabolic disturbance in response to mechanical overload. Mechanistically, elevated branched-chain α-keto acids directly suppressed respiration and induced superoxide production in isolated mitochondria. Finally, pharmacological enhancement of branched-chain α-keto acid dehydrogenase activity significantly blunted cardiac dysfunction after pressure overload. CONCLUSIONS BCAA catabolic defect is a metabolic hallmark of failing heart resulting from Krüppel-like factor 15-mediated transcriptional reprogramming. BCAA catabolic defect imposes a previously unappreciated significant contribution to heart failure.
Collapse
Affiliation(s)
- Haipeng Sun
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Kristine C Olson
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Chen Gao
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Domenick A Prosdocimo
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Meiyi Zhou
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Zhihua Wang
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Darwin Jeyaraj
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Ji-Youn Youn
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Shuxun Ren
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Yunxia Liu
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Christoph D Rau
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Svati Shah
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Olga Ilkayeva
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Wen-Jun Gui
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Noelle S William
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - R Max Wynn
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Christopher B Newgard
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Hua Cai
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Xinshu Xiao
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - David T Chuang
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Paul Christian Schulze
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Christopher Lynch
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Mukesh K Jain
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany
| | - Yibin Wang
- From Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (H.S., M.Z., Y.L., Y.W.); Division of Molecular Medicine, Departments of Anesthesiology, Medicine, and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles (H.S., C.G., Z.W., J.-Y.Y., S.R., C.D.R., H.C., X.X., Y.W.); Department of Cellular and Molecular Physiology, Penn State University College of Medicine, Hershey, PA (K.C.O., C.L.); Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH (D.A.P., D.J., M.K.J.); Division of Cardiology, Department of Medicine (S.S.) and Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology (O.I., C.B.N.), and Duke University School of Medicine, Durham, NC; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (W.-J.G., N.S.W., R.M.W., D.T.C.); and Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY (P.C.S.). Dr Schulze is now at the Department of Internal Medicine, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, Friedrich-Schiller-University, Jena, Germany.
| |
Collapse
|
24
|
Carubelli V, Castrini AI, Lazzarini V, Gheorghiade M, Metra M, Lombardi C. Amino acids and derivatives, a new treatment of chronic heart failure? Heart Fail Rev 2015; 20:39-51. [PMID: 24925377 DOI: 10.1007/s10741-014-9436-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Amino acids play a key role in multiple cellular processes. Amino acids availability is reduced in patients with heart failure (HF) with deleterious consequences on cardiac and whole-body metabolism. Several metabolic abnormalities have been identified in the failing heart, and many of them lead to an increased need of amino acids. Recently, several clinical trials have been conducted to demonstrate the benefits of amino acids supplementation in patients with HF. Although they have shown an improvement of exercise tolerance and, in some cases, of left ventricular function, they have many limitations, namely small sample size, differences in patients' characteristics and nutritional supplementations, and lack of data regarding outcomes. Moreover recent data suggest that a multi-nutritional approach, including also antioxidants, vitamins, and metals, may be more effective. Larger trials are needed to ascertain safety, efficacy, and impact on prognosis of such an approach in HF.
Collapse
Affiliation(s)
- Valentina Carubelli
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | | | | | | | | | | |
Collapse
|
25
|
Malik A, Sharma U, Lakshmy R, Narang R, Jagannathan NR. Biochemical characterization of blood plasma of coronary artery disease patients by in vitro high-resolution proton NMR spectroscopy. J Biosci 2015; 40:31-9. [DOI: 10.1007/s12038-014-9493-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
26
|
Visser M, Davids M, Verberne HJ, Kok WEM, Tepaske R, Cocchieri R, Kemper EM, Teerlink T, Jonker MA, Wisselink W, de Mol BAJM, van Leeuwen PAM. Nutrition before, during, and after surgery increases the arginine:asymmetric dimethylarginine ratio and relates to improved myocardial glucose metabolism: a randomized controlled trial. Am J Clin Nutr 2014; 99:1440-9. [PMID: 24695897 DOI: 10.3945/ajcn.113.075473] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Nitric oxide (NO) is essential for the optimal perfusion of the heart and its vasculature. NO may be insufficient in surgical patients because its precursor arginine is decreased, and the inhibitor of NO synthesis asymmetric dimethylarginine (ADMA) is increased. Besides arginine, the presence of other amino acids essential for the proper metabolism of cardiac cells may be decreased too. Supplementation of these amino acids with enteral and parenteral nutrition before, during, and after surgery may augment the myocardial and plasma arginine:ADMA ratio and availability of amino acids. Myocardial glucose metabolism and nutritional conditioning may result in a reduction of cardiac injury and support rapid recovery after major surgery. OBJECTIVE We investigated the effect of nutrition before, during, and after surgery on amino acids and the myocardial arginine:ADMA ratio and its relation to myocardial glucose metabolism. DESIGN In this trial, 33 patients who were undergoing off-pump coronary artery bypass grafting (CABG) were randomly assigned between enteral, parenteral, or no nutrition (control) from 2 d before, during, and until 2 d after surgery. Both enteral and parenteral solutions were prepared with commercially available products and included proteins or amino acids, glucose, vitamins, and minerals. Concentrations of amino acids including ADMA were analyzed in myocardial tissue and plasma samples. ¹⁸F-fluorodeoxyglucose positron emission tomography was performed before and after surgery to assess myocardial glucose metabolism. RESULTS The myocardial arginine:ADMA ratio increased during surgery and was significantly higher in the enteral and parenteral groups than in the control group [median (IQR): 115.0 (98.0-142.2) (P = 0.012), 116.9 (100.3-135.3) (P = 0.004), and 93.3 (82.7-101.1), respectively]. Furthermore, the change in the preoperative to postoperative plasma arginine:ADMA ratio correlated with the change in myocardial glucose metabolism in positron emission tomography (r = 0.427, P = 0.033). CONCLUSION Enteral or parenteral nutrition before, during, and after CABG may positively influence myocardial glucose metabolism by increasing the plasma and myocardial arginine:ADMA ratio.
Collapse
Affiliation(s)
- Marlieke Visser
- From the Departments of Surgery (MV, WW, and PAMvL), Clinical Chemistry (MD and TT), and Epidemiology and Biostatistics (MAJ), VU University Medical Center, Amsterdam, Netherlands, and the Departments of Cardio-thoracic Surgery (MV, RC, and BAJMdM), Nuclear Medicine (HJV), Cardiology (WEMK), and Pharmacy (EMK) and the Intensive Care Unit (RT), Academic Medical Center University of Amsterdam, Amsterdam, Netherlands
| | - Mariska Davids
- From the Departments of Surgery (MV, WW, and PAMvL), Clinical Chemistry (MD and TT), and Epidemiology and Biostatistics (MAJ), VU University Medical Center, Amsterdam, Netherlands, and the Departments of Cardio-thoracic Surgery (MV, RC, and BAJMdM), Nuclear Medicine (HJV), Cardiology (WEMK), and Pharmacy (EMK) and the Intensive Care Unit (RT), Academic Medical Center University of Amsterdam, Amsterdam, Netherlands
| | - Hein J Verberne
- From the Departments of Surgery (MV, WW, and PAMvL), Clinical Chemistry (MD and TT), and Epidemiology and Biostatistics (MAJ), VU University Medical Center, Amsterdam, Netherlands, and the Departments of Cardio-thoracic Surgery (MV, RC, and BAJMdM), Nuclear Medicine (HJV), Cardiology (WEMK), and Pharmacy (EMK) and the Intensive Care Unit (RT), Academic Medical Center University of Amsterdam, Amsterdam, Netherlands
| | - Wouter E M Kok
- From the Departments of Surgery (MV, WW, and PAMvL), Clinical Chemistry (MD and TT), and Epidemiology and Biostatistics (MAJ), VU University Medical Center, Amsterdam, Netherlands, and the Departments of Cardio-thoracic Surgery (MV, RC, and BAJMdM), Nuclear Medicine (HJV), Cardiology (WEMK), and Pharmacy (EMK) and the Intensive Care Unit (RT), Academic Medical Center University of Amsterdam, Amsterdam, Netherlands
| | - Robert Tepaske
- From the Departments of Surgery (MV, WW, and PAMvL), Clinical Chemistry (MD and TT), and Epidemiology and Biostatistics (MAJ), VU University Medical Center, Amsterdam, Netherlands, and the Departments of Cardio-thoracic Surgery (MV, RC, and BAJMdM), Nuclear Medicine (HJV), Cardiology (WEMK), and Pharmacy (EMK) and the Intensive Care Unit (RT), Academic Medical Center University of Amsterdam, Amsterdam, Netherlands
| | - Riccardo Cocchieri
- From the Departments of Surgery (MV, WW, and PAMvL), Clinical Chemistry (MD and TT), and Epidemiology and Biostatistics (MAJ), VU University Medical Center, Amsterdam, Netherlands, and the Departments of Cardio-thoracic Surgery (MV, RC, and BAJMdM), Nuclear Medicine (HJV), Cardiology (WEMK), and Pharmacy (EMK) and the Intensive Care Unit (RT), Academic Medical Center University of Amsterdam, Amsterdam, Netherlands
| | - Elles M Kemper
- From the Departments of Surgery (MV, WW, and PAMvL), Clinical Chemistry (MD and TT), and Epidemiology and Biostatistics (MAJ), VU University Medical Center, Amsterdam, Netherlands, and the Departments of Cardio-thoracic Surgery (MV, RC, and BAJMdM), Nuclear Medicine (HJV), Cardiology (WEMK), and Pharmacy (EMK) and the Intensive Care Unit (RT), Academic Medical Center University of Amsterdam, Amsterdam, Netherlands
| | - Tom Teerlink
- From the Departments of Surgery (MV, WW, and PAMvL), Clinical Chemistry (MD and TT), and Epidemiology and Biostatistics (MAJ), VU University Medical Center, Amsterdam, Netherlands, and the Departments of Cardio-thoracic Surgery (MV, RC, and BAJMdM), Nuclear Medicine (HJV), Cardiology (WEMK), and Pharmacy (EMK) and the Intensive Care Unit (RT), Academic Medical Center University of Amsterdam, Amsterdam, Netherlands
| | - Marianne A Jonker
- From the Departments of Surgery (MV, WW, and PAMvL), Clinical Chemistry (MD and TT), and Epidemiology and Biostatistics (MAJ), VU University Medical Center, Amsterdam, Netherlands, and the Departments of Cardio-thoracic Surgery (MV, RC, and BAJMdM), Nuclear Medicine (HJV), Cardiology (WEMK), and Pharmacy (EMK) and the Intensive Care Unit (RT), Academic Medical Center University of Amsterdam, Amsterdam, Netherlands
| | - Willem Wisselink
- From the Departments of Surgery (MV, WW, and PAMvL), Clinical Chemistry (MD and TT), and Epidemiology and Biostatistics (MAJ), VU University Medical Center, Amsterdam, Netherlands, and the Departments of Cardio-thoracic Surgery (MV, RC, and BAJMdM), Nuclear Medicine (HJV), Cardiology (WEMK), and Pharmacy (EMK) and the Intensive Care Unit (RT), Academic Medical Center University of Amsterdam, Amsterdam, Netherlands
| | - Bas A J M de Mol
- From the Departments of Surgery (MV, WW, and PAMvL), Clinical Chemistry (MD and TT), and Epidemiology and Biostatistics (MAJ), VU University Medical Center, Amsterdam, Netherlands, and the Departments of Cardio-thoracic Surgery (MV, RC, and BAJMdM), Nuclear Medicine (HJV), Cardiology (WEMK), and Pharmacy (EMK) and the Intensive Care Unit (RT), Academic Medical Center University of Amsterdam, Amsterdam, Netherlands
| | - Paul A M van Leeuwen
- From the Departments of Surgery (MV, WW, and PAMvL), Clinical Chemistry (MD and TT), and Epidemiology and Biostatistics (MAJ), VU University Medical Center, Amsterdam, Netherlands, and the Departments of Cardio-thoracic Surgery (MV, RC, and BAJMdM), Nuclear Medicine (HJV), Cardiology (WEMK), and Pharmacy (EMK) and the Intensive Care Unit (RT), Academic Medical Center University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
27
|
Lombardi C, Carubelli V, Lazzarini V, Vizzardi E, Quinzani F, Guidetti F, Rovetta R, Nodari S, Gheorghiade M, Metra M. Effects of oral amino Acid supplements on functional capacity in patients with chronic heart failure. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2014; 8:39-44. [PMID: 24899826 PMCID: PMC4039212 DOI: 10.4137/cmc.s14016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 02/11/2014] [Accepted: 02/15/2014] [Indexed: 11/05/2022]
Abstract
Amino acids (AAs) availability is reduced in patients with heart failure (HF) leading to abnormalities in cardiac and skeletal muscle metabolism, and eventually to a reduction in functional capacity and quality of life. In this study, we investigate the effects of oral supplementation with essential and semi-essential AAs for three months in patients with stable chronic HF. The primary endpoints were the effects of AA’s supplementation on exercise tolerance (evaluated by cardiopulmonary stress test and six minutes walking test (6MWT)), whether the secondary endpoints were change in quality of life (evaluated by Minnesota Living with Heart Failure Questionnaire—MLHFQ) and N-terminal pro-brain natriuretic peptide (NT-proBNP) levels. We enrolled 13 patients with chronic stable HF on optimal therapy, symptomatic in New York Heart Association (NYHA) class II/III, with an ejection fraction (EF) <45%. The mean age was 59 ± 14 years, and 11 (84.6%) patients were male. After three months, peak VO2 (baseline 14.8 ± 3.9 mL/minute/kg vs follow-up 16.8 ± 5.1 mL/minute/kg; P = 0.008) and VO2 at anaerobic threshold improved significantly (baseline 9.0 ± 3.8 mL/minute/kg vs follow-up 12.4 ± 3.9 mL/minute/kg; P = 0.002), as the 6MWT distance (baseline 439.1 ± 64.3 m vs follow-up 474.2 ± 89.0 m; P = 0.006). However, the quality of life did not change significantly (baseline 21 ± 14 vs follow-up 25 ± 13; P = 0.321). A non-significant trend in the reduction of NT-proBNP levels was observed (baseline 1502 ± 1900 ng/L vs follow-up 1040 ± 1345 ng/L; P = 0.052). AAs treatment resulted safe and was well tolerated by all patients. In our study, AAs supplementation in patients with chronic HF improved exercise tolerance but did not change quality of life.
Collapse
Affiliation(s)
- Carlo Lombardi
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| | - Valentina Carubelli
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| | - Valentina Lazzarini
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| | - Enrico Vizzardi
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| | - Filippo Quinzani
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| | - Federica Guidetti
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| | - Riccardo Rovetta
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| | - Savina Nodari
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| | - Mihai Gheorghiade
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| | - Marco Metra
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| |
Collapse
|
28
|
Lombardi C, Carubelli V, Lazzarini V, Vizzardi E, Bordonali T, Ciccarese C, Castrini AI, Dei Cas A, Nodari S, Metra M. Effects of oral administration of orodispersible levo-carnosine on quality of life and exercise performance in patients with chronic heart failure. Nutrition 2014; 31:72-8. [PMID: 25287762 DOI: 10.1016/j.nut.2014.04.021] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 03/01/2014] [Accepted: 04/24/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE Chronic heart failure (CHF) is characterized by several micronutrient deficits. Amino acid supplementation may have a positive effect on nutritional and metabolic status in patients with CHF. Levo-carnosine (β-alanyl-L-histidine) is expressed at a high concentration in myocardium and muscle. Preliminary studies with L-carnosine in healthy individuals have suggested a potential role in improving exercise performance. To our knowledge, no study has been conducted in patients with heart failure. The aim of this study was to test the oral supplementation of L-carnosine and its effects on quality of life and exercise performance in patients with stable CHF. METHODS Fifty patients with stable CHF and severe left-ventricular systolic dysfunction on optimal medical therapy were randomized 1:1 to receive oral orodispersible L-carnosine (500 mg OD) or standard treatment. Left-ventricular ejection fraction (LVEF) was measured by echocardiography. Cardiopulmonary stress test, 6-minute walking test (6 MWT) and quality-of-life (visual analog scale score and the EuroQOL five dimensions questionnaire [EQ-5D]) were performed at baseline and after 6 mo. RESULTS Patients receiving orodispersible L-carnosine had an improvement in 6 MWT distance (P = 0.014) and in quality-of-life (VAS score) (P = 0.039) between baseline and follow-up. Compared with controls, diet supplementation with orodispersible L-carnosine was associated with an improvement in peakVO2 (P < 0.0001), VO2 at anaerobic threshold, peak exercise workload, 6 MWT and quality-of-life assessed by the EQ-5D test and the VAS score. CONCLUSION This study suggests that L-carnosine, added to conventional therapy, has beneficial effects on exercise performance and quality of life in stable CHF. More data are necessary to evaluate its effects on left-ventricular ejection fraction and prognosis in CHF.
Collapse
Affiliation(s)
- Carlo Lombardi
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health University and Civil Hospital of Brescia, Brescia, Italy.
| | - Valentina Carubelli
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health University and Civil Hospital of Brescia, Brescia, Italy
| | - Valentina Lazzarini
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health University and Civil Hospital of Brescia, Brescia, Italy
| | - Enrico Vizzardi
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health University and Civil Hospital of Brescia, Brescia, Italy
| | - Tania Bordonali
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health University and Civil Hospital of Brescia, Brescia, Italy
| | - Camilla Ciccarese
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health University and Civil Hospital of Brescia, Brescia, Italy
| | - Anna Isotta Castrini
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health University and Civil Hospital of Brescia, Brescia, Italy
| | - Alessandra Dei Cas
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health University and Civil Hospital of Brescia, Brescia, Italy
| | - Savina Nodari
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health University and Civil Hospital of Brescia, Brescia, Italy
| | - Marco Metra
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health University and Civil Hospital of Brescia, Brescia, Italy
| |
Collapse
|
29
|
Andreadou I, Mikros E, Ioannidis K, Sigala F, Naka K, Kostidis S, Farmakis D, Tenta R, Kavantzas N, Bibli SI, Gikas E, Skaltsounis L, Kremastinos DT, Iliodromitis EK. Oleuropein prevents doxorubicin-induced cardiomyopathy interfering with signaling molecules and cardiomyocyte metabolism. J Mol Cell Cardiol 2014; 69:4-16. [DOI: 10.1016/j.yjmcc.2014.01.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 01/16/2014] [Accepted: 01/19/2014] [Indexed: 11/30/2022]
|
30
|
Heger Z, Cernei N, Kudr J, Gumulec J, Blazkova I, Zitka O, Eckschlager T, Stiborova M, Adam V, Kizek R. A novel insight into the cardiotoxicity of antineoplastic drug doxorubicin. Int J Mol Sci 2013; 14:21629-46. [PMID: 24185911 PMCID: PMC3856025 DOI: 10.3390/ijms141121629] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 09/26/2013] [Accepted: 10/09/2013] [Indexed: 01/04/2023] Open
Abstract
Doxorubicin is a commonly used antineoplastic agent in the treatment of many types of cancer. Little is known about the interactions of doxorubicin with cardiac biomolecules. Serious cardiotoxicity including dilated cardiomyopathy often resulting in a fatal congestive heart failure may occur as a consequence of chemotherapy with doxorubicin. The purpose of this study was to determine the effect of exposure to doxorubicin on the changes in major amino acids in tissue of cardiac muscle (proline, taurine, glutamic acid, arginine, aspartic acid, leucine, glycine, valine, alanine, isoleucine, threonine, lysine and serine). An in vitro interaction study was performed as a comparison of amino acid profiles in heart tissue before and after application of doxorubicin. We found that doxorubicin directly influences myocardial amino acid representation even at low concentrations. In addition, we performed an interaction study that resulted in the determination of breaking points for each of analyzed amino acids. Lysine, arginine, β-alanine, valine and serine were determined as the most sensitive amino acids. Additionally we compared amino acid profiles of myocardium before and after exposure to doxorubicin. The amount of amino acids after interaction with doxorubicin was significantly reduced (p = 0.05). This fact points at an ability of doxorubicin to induce changes in quantitative composition of amino acids in myocardium. Moreover, this confirms that the interactions between doxorubicin and amino acids may act as another factor most likely responsible for adverse effects of doxorubicin on myocardium.
Collapse
Affiliation(s)
- Zbynek Heger
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, Brno CZ-613 00, Czech Republic, E-Mails: (Z.H.); (N.C.); (J.K.); (J.G.); (I.B.); (O.Z.); (V.A.)
| | - Natalia Cernei
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, Brno CZ-613 00, Czech Republic, E-Mails: (Z.H.); (N.C.); (J.K.); (J.G.); (I.B.); (O.Z.); (V.A.)
- Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, Brno CZ-616 00, Czech Republic
| | - Jiri Kudr
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, Brno CZ-613 00, Czech Republic, E-Mails: (Z.H.); (N.C.); (J.K.); (J.G.); (I.B.); (O.Z.); (V.A.)
| | - Jaromir Gumulec
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, Brno CZ-613 00, Czech Republic, E-Mails: (Z.H.); (N.C.); (J.K.); (J.G.); (I.B.); (O.Z.); (V.A.)
- Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, Brno CZ-616 00, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Komenskeho namesti 2, Brno CZ-662 43, Czech Republic
| | - Iva Blazkova
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, Brno CZ-613 00, Czech Republic, E-Mails: (Z.H.); (N.C.); (J.K.); (J.G.); (I.B.); (O.Z.); (V.A.)
| | - Ondrej Zitka
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, Brno CZ-613 00, Czech Republic, E-Mails: (Z.H.); (N.C.); (J.K.); (J.G.); (I.B.); (O.Z.); (V.A.)
- Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, Brno CZ-616 00, Czech Republic
| | - Tomas Eckschlager
- Department of Paediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University, and University Hospital Motol, V Uvalu 84, Prague 5 CZ-15006, Czech Republic; E-Mail:
| | - Marie Stiborova
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, Prague 2 CZ-12840, Czech Republic; E-Mail:
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, Brno CZ-613 00, Czech Republic, E-Mails: (Z.H.); (N.C.); (J.K.); (J.G.); (I.B.); (O.Z.); (V.A.)
- Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, Brno CZ-616 00, Czech Republic
| | - Rene Kizek
- Department of Chemistry and Biochemistry, Faculty of Agronomy, Mendel University in Brno, Zemedelska 1, Brno CZ-613 00, Czech Republic, E-Mails: (Z.H.); (N.C.); (J.K.); (J.G.); (I.B.); (O.Z.); (V.A.)
- Central European Institute of Technology, Brno University of Technology, Technicka 3058/10, Brno CZ-616 00, Czech Republic
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +420-545-133-350; Fax: +420-545-212-044
| |
Collapse
|
31
|
Glembotski CC. Roles for ATF6 and the sarco/endoplasmic reticulum protein quality control system in the heart. J Mol Cell Cardiol 2013; 71:11-5. [PMID: 24140798 DOI: 10.1016/j.yjmcc.2013.09.018] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/26/2013] [Accepted: 09/27/2013] [Indexed: 11/30/2022]
Abstract
The hypertrophic growth of cardiac myocytes is a highly dynamic process that underlies physiological and pathological adaptation of the heart. Accordingly, a better understanding of the molecular underpinnings of cardiac myocyte hypertrophy is required in order to fully appreciate the causes and functional consequences of the changes in the size of the healthy and diseased heart. Hypertrophy is driven by increases in cardiac myocyte protein, which must be balanced by cellular ability to maintain protein quality in order to avoid maladaptive accumulation of toxic misfolded proteins. Recent studies have shown that the endoplasmic reticulum (ER), which, in cardiac myocytes, comprises the sarco/endoplasmic reticulum (SR/ER), is the site of most protein synthesis. Thus, the protein quality control machinery located at the SR/ER is likely to be an important determinant of whether the heart responds adaptively to hypertrophic growth stimuli. The SR/ER-transmembrane protein, ATF6, serves a critical protein quality control function as a first responder to the accumulation of potentially toxic, misfolded proteins. Misfolded proteins transform ATF6 into a transcription factor that regulates a gene program that is partly responsible for enhancing protein quality control. Two ATF6-inducible genes that have been studied in the heart and shown to be adaptive are RCAN1 and Derl3, which encode proteins that decrease protein-folding demand, and enhance degradation of misfolded proteins, respectively. Thus, the ATF6-regulated SR/ER protein quality control system is important for maintaining protein quality during growth, making ATF6, and other components of the system, potentially attractive targets for the therapeutic management pathological cardiac hypertrophy. This article is part of a Special Issue entitled "Protein Quality Control, the Ubiquitin Proteasome System, and Autophagy".
Collapse
Affiliation(s)
- Christopher C Glembotski
- San Diego State University Heart Institute, Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA.
| |
Collapse
|
32
|
Pasini E, Aquilani R, Dioguardi FS. "The enemy within". How to identify chronic diseases induced-protein metabolism impairment and its possible pharmacological treatment. Pharmacol Res 2013; 76:28-33. [PMID: 23827158 DOI: 10.1016/j.phrs.2013.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 06/24/2013] [Indexed: 12/17/2022]
Abstract
Recent clinical and experimental data show that considerable impairment of protein metabolism occurs in patients with chronic diseases such as heart failure. However, too often the extent of impairment is under-estimated or ignored by most clinicians and no therapy is considered leading to progressive loss of body proteins, increase morbidity, hospital stay and mortality. This paper illustrates the possible biological markers to evaluate general protein metabolism, including quantification of related damage and possible improvement of the metabolism using specific therapeutical metabolic strategies recently studied in a clinical setting.
Collapse
Affiliation(s)
- Evasio Pasini
- Foundation "Salvatore Maugeri", IRCCS, Scientific Institute of Lumezzane, via Mazzini 129, 25066 Lumezzane (BS), Italy.
| | | | | |
Collapse
|
33
|
Shimura D, Nakai G, Jiao Q, Osanai K, Kashikura K, Endo K, Soga T, Goda N, Minamisawa S. Metabolomic profiling analysis reveals chamber-dependent metabolite patterns in the mouse heart. Am J Physiol Heart Circ Physiol 2013; 305:H494-505. [PMID: 23792677 DOI: 10.1152/ajpheart.00867.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Energy of the cardiac muscle largely depends on fatty acid oxidation. It is known that the atrium and ventricle have chamber-specific functions, structures, gene expressions, and pathologies. The left ventricle works as a high-pressure chamber to pump blood toward the body, and its muscle wall is thicker than those of the other chambers, suggesting that energy utilization in each of the chambers should be different. However, a chamber-specific pattern of metabolism remains incompletely understood. Recently, innovative techniques have enabled the comprehensive analysis of metabolites. Therefore, we aimed to clarify differences in metabolic patterns among the chambers. Male C57BL6 mice at 6 wk old were subject to a comprehensive measurement of metabolites in the atria and ventricles by capillary electrophoresis and mass spectrometry. We found that overall metabolic profiles, including nucleotides and amino acids, were similar between the right and left ventricles. On the other hand, the atria exhibited a distinct metabolic pattern from those of the ventricles. Importantly, the high-energy phosphate pool (the total concentration of ATP, ADP, and AMP) was higher in both ventricles. In addition, the levels of lactate, acetyl CoA, and tricarboxylic acid cycle contents were higher in the ventricles. Accordingly, the activities and/or expression levels of key enzymes were higher in the ventricles to produce more energy. The present study provides a basis for understanding the chamber-specific metabolism underlining pathophysiology in the heart.
Collapse
Affiliation(s)
- Daisuke Shimura
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Drake KJ, Sidorov VY, McGuinness OP, Wasserman DH, Wikswo JP. Amino acids as metabolic substrates during cardiac ischemia. Exp Biol Med (Maywood) 2012; 237:1369-78. [PMID: 23354395 PMCID: PMC3816490 DOI: 10.1258/ebm.2012.012025] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The heart is well known as a metabolic omnivore in that it is capable of consuming fatty acids, glucose, ketone bodies, pyruvate, lactate, amino acids and even its own constituent proteins, in order of decreasing preference. The energy from these substrates supports not only mechanical contraction, but also the various transmembrane pumps and transporters required for ionic homeostasis, electrical activity, metabolism and catabolism. Cardiac ischemia - for example, due to compromise of the coronary vasculature or end-stage heart failure - will alter both electrical and metabolic activity. While the effects of myocardial ischemia on electrical propagation and stability have been studied in depth, the effects of ischemia on metabolic substrate preference has not been fully appreciated: oxygen deprivation during ischemia will significantly alter the relative ability of the heart to utilize each of these substrates. Although changes in cardiac metabolism are understood to be an underlying component in almost all cardiac myopathies, the potential contribution of amino acids in maintaining cardiac electrical conductance and stability during ischemia is underappreciated. Despite clear evidence that amino acids exert cardioprotective effects in ischemia and other cardiac disorders, their role in the metabolism of the ischemic heart has yet to be fully elucidated. This review synthesizes the current literature of the metabolic contribution of amino acids during ischemia by analyzing relevant historical and recent research.
Collapse
Affiliation(s)
- Kenneth J. Drake
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235
| | - Veniamin Y. Sidorov
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | - Owen P. McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235
| | - David H. Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
| | - John P. Wikswo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37235
| |
Collapse
|
35
|
Ardehali H, Sabbah HN, Burke MA, Sarma S, Liu PP, Cleland JGF, Maggioni A, Fonarow GC, Abel ED, Campia U, Gheorghiade M. Targeting myocardial substrate metabolism in heart failure: potential for new therapies. Eur J Heart Fail 2012; 14:120-9. [PMID: 22253453 DOI: 10.1093/eurjhf/hfr173] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The incidence and prevalence of heart failure have increased significantly over the past few decades. Available data suggest that patients with heart failure independent of the aetiology have viable but dysfunctional myocardium that is potentially salvageable. Although a great deal of research effort has focused on characterizing the molecular basis of heart failure, cardiac metabolism in this disorder remains an understudied discipline. It is known that many aspects of cardiomyocyte energetics are altered in heart failure. These include a shift from fatty acid to glucose as a preferred substrate and a decline in the levels of ATP. Despite these demonstrated changes, there are currently no approved drugs that target metabolic enzymes or proteins in heart failure. This is partly due to our limited knowledge of the mechanisms and pathways that regulate cardiac metabolism. Better characterization of these pathways may potentially lead to new therapies for heart failure. Targeting myocardial energetics in the viable and potentially salvageable tissue may be particularly effective in the treatment of heart failure. Here, we will review metabolic changes that occur in fatty acid and glucose metabolism and AMP-activated kinase in heart failure. We propose that cardiac energetics should be considered as a potential target for therapy in heart failure and more research should be done in this area.
Collapse
Affiliation(s)
- Hossein Ardehali
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
|
37
|
Visser M, Davids M, Verberne HJ, Kok WEM, Niessen HWM, van Venrooij LMW, Cocchieri R, Wisselink W, de Mol BAJM, van Leeuwen PAM. Rationale and design of a proof-of-concept trial investigating the effect of uninterrupted perioperative (par)enteral nutrition on amino acid profile, cardiomyocytes structure, and cardiac perfusion and metabolism of patients undergoing coronary artery bypass grafting. J Cardiothorac Surg 2011; 6:36. [PMID: 21439030 PMCID: PMC3072317 DOI: 10.1186/1749-8090-6-36] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 03/25/2011] [Indexed: 12/18/2022] Open
Abstract
Background Malnutrition is very common in patients undergoing cardiac surgery. Malnutrition can change myocardial substrate utilization which can induce adverse effects on myocardial metabolism and function. We aim to investigate the hypothesis that there is a disturbed amino acids profile in the cardiac surgical patient which can be normalized by (par)enteral nutrition before, during and after surgery, subsequently improving cardiomyocyte structure, cardiac perfusion and glucose metabolism. Methods/Design This randomized controlled intervention study investigates the effect of uninterrupted perioperative (par)enteral nutrition on cardiac function in 48 patients undergoing coronary artery bypass grafting. Patients are given enteral nutrition (n = 16) or parenteral nutrition (n = 16), at least two days before, during, and two days after coronary artery bypass grafting, or are treated according to the standard guidelines (control) (n = 16). We will illustrate the effect of (par)enteral nutrition on differences in concentrations of amino acids and asymmetric dimethylarginine and in activity of dimethylarginine dimethylaminohydrolase and arginase in cardiac tissue and blood plasma. In addition, cardiomyocyte structure by histological, immuno-histochemical and ultrastructural analysis will be compared between the (par)enteral and control group. Furthermore, differences in cardiac perfusion and global left ventricular function and glucose metabolism, and their changes after coronary artery bypass grafting are evaluated by electrocardiography-gated myocardial perfusion scintigraphy and 18F-fluorodeoxy-glucose positron emission tomography respectively. Finally, fat free mass is measured before and after intervention with bioelectrical impedance spectrometry in order to evaluate nutritional status. Trial registration Netherlands Trial Register (NTR): NTR2183
Collapse
Affiliation(s)
- Marlieke Visser
- Department of Cardiothoracic Surgery, Academic Medical Center University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Micronutrient Deficiencies. J Am Coll Cardiol 2009; 54:1660-73. [DOI: 10.1016/j.jacc.2009.08.012] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 08/24/2009] [Accepted: 08/25/2009] [Indexed: 01/08/2023]
|