1
|
Bornstein MR, Tian R, Arany Z. Human cardiac metabolism. Cell Metab 2024; 36:1456-1481. [PMID: 38959861 PMCID: PMC11290709 DOI: 10.1016/j.cmet.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/12/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024]
Abstract
The heart is the most metabolically active organ in the human body, and cardiac metabolism has been studied for decades. However, the bulk of studies have focused on animal models. The objective of this review is to summarize specifically what is known about cardiac metabolism in humans. Techniques available to study human cardiac metabolism are first discussed, followed by a review of human cardiac metabolism in health and in heart failure. Mechanistic insights, where available, are reviewed, and the evidence for the contribution of metabolic insufficiency to heart failure, as well as past and current attempts at metabolism-based therapies, is also discussed.
Collapse
Affiliation(s)
- Marc R Bornstein
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Zoltan Arany
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Bohnert S, Reinert C, Trella S, Cattaneo A, Preiß U, Bohnert M, Zwirner J, Büttner A, Schmitz W, Ondruschka B. Neuroforensomics: metabolites as valuable biomarkers in cerebrospinal fluid of lethal traumatic brain injuries. Sci Rep 2024; 14:13651. [PMID: 38871842 DOI: 10.1038/s41598-024-64312-0] [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: 07/04/2023] [Accepted: 06/07/2024] [Indexed: 06/15/2024] Open
Abstract
Traumatic brain injury (TBI) is a ubiquitous, common sequela of accidents with an annual prevalence of several million cases worldwide. In forensic pathology, structural proteins of the cellular compartments of the CNS in serum and cerebrospinal fluid (CSF) have been predominantly used so far as markers of an acute trauma reaction for the biochemical assessment of neuropathological changes after TBI. The analysis of endogenous metabolites offers an innovative approach that has not yet been considered widely in the assessment of causes and circumstances of death, for example after TBI. The present study, therefore, addresses the question whether the detection of metabolites by liquid-chromatography-mass spectrometry (LC/MS) analysis in post mortem CSF is suitable to identify TBI and to distinguish it from acute cardiovascular control fatalities (CVF). Metabolite analysis of 60 CSF samples collected during autopsies was performed using high resolution (HR)-LC/MS. Subsequent statistical and graphical evaluation as well as the calculation of a TBI/CVF quotient yielded promising results: numerous metabolites were identified that showed significant concentration differences in the post mortem CSF for lethal acute TBI (survival times up to 90 min) compared to CVF. For the first time, this forensic study provides an evaluation of a new generation of biomarkers for diagnosing TBI in the differentiation to other causes of death, here CVF, as surrogate markers for the post mortem assessment of complex neuropathological processes in the CNS ("neuroforensomics").
Collapse
Affiliation(s)
- Simone Bohnert
- Institute of Forensic Medicine, University of Würzburg, Würzburg, Germany
| | - Christoph Reinert
- Institute of Forensic Medicine, University of Würzburg, Würzburg, Germany
| | - Stefanie Trella
- Institute of Forensic Medicine, University of Würzburg, Würzburg, Germany
| | - Andrea Cattaneo
- Department of Neurosurgery, University Hospital of Würzburg, Würzburg, Germany
| | - Ulrich Preiß
- Institute of Forensic Medicine, University of Würzburg, Würzburg, Germany
| | - Michael Bohnert
- Institute of Forensic Medicine, University of Würzburg, Würzburg, Germany
| | - Johann Zwirner
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Oral Sciences, University of Otago, Dunedin, New Zealand
| | - Andreas Büttner
- Institute of Forensic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Werner Schmitz
- Institute of Biochemistry and Molecular Biology, Biozentrum, University of Würzburg, Würzburg, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
3
|
Truby LK, Kwee LC, Bowles DE, Casalinova S, Ilkayeva O, Muehlbauer MJ, Huebner JL, Holley CL, DeVore AD, Patel CB, Kang L, Pla MM, Gross R, McGarrah RW, Schroder JN, Milano CA, Shah SH. Metabolomic profiling during ex situ normothermic perfusion before heart transplantation defines patterns of substrate utilization and correlates with markers of allograft injury. J Heart Lung Transplant 2024; 43:716-726. [PMID: 38065238 DOI: 10.1016/j.healun.2023.12.002] [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: 04/05/2023] [Revised: 11/21/2023] [Accepted: 12/02/2023] [Indexed: 12/22/2023] Open
Abstract
BACKGROUND Cardiac metabolism is altered in heart failure and ischemia-reperfusion injury states. We hypothesized that metabolomic profiling during ex situ normothermic perfusion before heart transplantation (HT) would lend insight into myocardial substrate utilization and report on subclinical and clinical allograft dysfunction risk. METHODS Metabolomic profiling was performed on serial samples of ex situ normothermic perfusate assaying biomarkers of myocardial injury in lactate and cardiac troponin I (TnI) as well as metabolites (66 acylcarnitines, 15 amino acids, nonesterified fatty acids [NEFA], ketones, and 3-hydroxybutyrate). We tested for change over time in injury biomarkers and metabolites, along with differential changes by recovery strategy (donation after circulatory death [DCD] vs donation after brain death [DBD]). We examined associations between metabolites, injury biomarkers, and primary graft dysfunction (PGD). Analyses were performed using linear mixed models adjusted for recovery strategy, assay batch, donor-predicted heart mass, and time. RESULTS A total of 176 samples from 92 ex situ perfusion runs were taken from donors with a mean age of 35 (standard deviation 11.3) years and a median total ex situ perfusion time of 234 (interquartile range 84) minutes. Lactate trends over time differed significantly by recovery strategy, while TnI increased during ex situ perfusion regardless of DCD vs DBD status. We found fuel substrates were rapidly depleted during ex situ perfusion, most notably the branched-chain amino acids leucine/isoleucine, as well as ketones, 3-hydroxybutyrate, and NEFA (least squares [LS] mean difference from the first to last time point -1.7 to -4.5, false discovery rate q < 0.001). Several long-chain acylcarnitines (LCAC), including C16, C18, C18:1, C18:2, C18:3, C20:3, and C20:4, increased during the perfusion run (LS mean difference 0.42-0.67, q < 0.001). Many LCACs were strongly associated with lactate and TnI. The change over time of many LCACs was significantly different for DCD vs DBD, suggesting differential trends in fuel substrate utilization by ischemic injury pattern. Changes in leucine/isoleucine, arginine, C12:1-OH/C10:1-DC, and C16-OH/C14-DC were associated with increased odds of moderate-severe PGD. Neither end-of-run nor change in lactate or TnI was associated with PGD. CONCLUSIONS Metabolomic profiling of ex situ normothermic perfusion solution reveals a pattern of fuel substrate utilization that correlates with subclinical and clinical allograft dysfunction. This study highlights a potential role for interventions focused on fuel substrate modification in allograft conditioning during ex situ perfusion to improve allograft outcomes.
Collapse
Affiliation(s)
- Lauren K Truby
- University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - Dawn E Bowles
- Duke University Medical Center, Durham, North Carolina
| | | | - Olga Ilkayeva
- Duke Molecular Physiology Institute, Durham, North Carolina
| | | | | | | | - Adam D DeVore
- Duke University Medical Center, Durham, North Carolina
| | | | - Lillian Kang
- Duke University Medical Center, Durham, North Carolina
| | | | - Ryan Gross
- Duke University Medical Center, Durham, North Carolina
| | | | | | | | - Svati H Shah
- Duke Molecular Physiology Institute, Durham, North Carolina.
| |
Collapse
|
4
|
Pal N, Acharjee A, Ament Z, Dent T, Yavari A, Mahmod M, Ariga R, West J, Steeples V, Cassar M, Howell NJ, Lockstone H, Elliott K, Yavari P, Briggs W, Frenneaux M, Prendergast B, Dwight JS, Kharbanda R, Watkins H, Ashrafian H, Griffin JL. Metabolic profiling of aortic stenosis and hypertrophic cardiomyopathy identifies mechanistic contrasts in substrate utilization. FASEB J 2024; 38:e23505. [PMID: 38507255 DOI: 10.1096/fj.202301710rr] [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: 08/24/2023] [Revised: 12/24/2023] [Accepted: 02/07/2024] [Indexed: 03/22/2024]
Abstract
Aortic stenosis (AS) and hypertrophic cardiomyopathy (HCM) are distinct disorders leading to left ventricular hypertrophy (LVH), but whether cardiac metabolism substantially differs between these in humans remains to be elucidated. We undertook an invasive (aortic root, coronary sinus) metabolic profiling in patients with severe AS and HCM in comparison with non-LVH controls to investigate cardiac fuel selection and metabolic remodeling. These patients were assessed under different physiological states (at rest, during stress induced by pacing). The identified changes in the metabolome were further validated by metabolomic and orthogonal transcriptomic analysis, in separately recruited patient cohorts. We identified a highly discriminant metabolomic signature in severe AS in all samples, regardless of sampling site, characterized by striking accumulation of long-chain acylcarnitines, intermediates of fatty acid transport across the inner mitochondrial membrane, and validated this in a separate cohort. Mechanistically, we identify a downregulation in the PPAR-α transcriptional network, including expression of genes regulating fatty acid oxidation (FAO). In silico modeling of β-oxidation demonstrated that flux could be inhibited by both the accumulation of fatty acids as a substrate for mitochondria and the accumulation of medium-chain carnitines which induce competitive inhibition of the acyl-CoA dehydrogenases. We present a comprehensive analysis of changes in the metabolic pathways (transcriptome to metabolome) in severe AS, and its comparison to HCM. Our results demonstrate a progressive impairment of β-oxidation from HCM to AS, particularly for FAO of long-chain fatty acids, and that the PPAR-α signaling network may be a specific metabolic therapeutic target in AS.
Collapse
Affiliation(s)
- Nikhil Pal
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Department of Experimental Therapeutics, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Animesh Acharjee
- Department of Biochemistry, Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- MRC-Human Nutrition Research Unit, University of Cambridge, Cambridge, UK
- Institute of Cancer and Genomic Sciences, Centre for Computational Biology, University of Birmingham, Birmingham, UK
| | - Zsuzsanna Ament
- Department of Biochemistry, Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- MRC-Human Nutrition Research Unit, University of Cambridge, Cambridge, UK
| | - Tim Dent
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Arash Yavari
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Department of Experimental Therapeutics, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Masliza Mahmod
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Rina Ariga
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - James West
- Department of Biochemistry, Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- MRC-Human Nutrition Research Unit, University of Cambridge, Cambridge, UK
| | - Violetta Steeples
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, UK
| | - Mark Cassar
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Neil J Howell
- Department of Cardiothoracic Surgery, University Hospital Birmingham, Birmingham, UK
| | - Helen Lockstone
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, UK
| | - Kate Elliott
- Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, UK
| | - Parisa Yavari
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - William Briggs
- Department of Biochemistry, Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Michael Frenneaux
- Norwich Medical School, University of East Anglia, Bob Champion Research and Educational Building, Norwich, UK
| | - Bernard Prendergast
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Jeremy S Dwight
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Rajesh Kharbanda
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Houman Ashrafian
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Department of Experimental Therapeutics, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Julian L Griffin
- Department of Biochemistry, Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- MRC-Human Nutrition Research Unit, University of Cambridge, Cambridge, UK
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
5
|
Chen DX, Wang TH, Xiong XL, Shi J, Zhou L. Incidence, factors, and prognostic analyses of challenging cardiopulmonary bypass separation in Chinese cardiac surgical populations. Minerva Anestesiol 2024; 90:144-153. [PMID: 38127467 DOI: 10.23736/s0375-9393.23.17727-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
BACKGROUND Challenging separation from cardiopulmonary bypass (CPB) has been associated with multiple medical adversities, while its incidence, associated factors, and prognosis among cardiac surgery populations are substantially understudied. METHODS Adult cardiac surgical patients in two medical centers were retrospectively analyzed. Separation from CPB was stratified as easy, difficult, or complex, based on the use of pharmacologic assistance agents and mechanical supports. The various in-hospital adverse outcomes (e.g., mortality, common complications) were assessed. RESULTS The incidence of difficult and complex separation from CPB was 21.9% (1159 cases, 95% CI 20.8% to 23.1%), and 6.1% (320 cases, 95% CI 5.4% to 6.7%), respectively. High age, the presence of pulmonary hypertension or unstable angina, decreased ejection fraction, and emergency surgery were more frequently associated with challenging separation from CPB. Patients who experienced challenging separation from CPB had an elevated risk of adverse outcomes, including in-hospital mortality (complex: odds ratio [OR] 2.85), composite infection events (difficult: OR=1.82; complex: OR=1.88), major adverse cardiac events (difficult: OR=1.40; complex: OR=1.57), pulmonary complications (difficult: OR=1.31; complex: OR=1.20), acute kidney injury (difficult: OR=1.75; complex: OR=2.64), and prolonged postoperative hospital stays. CONCLUSIONS We depicted the incidence of challenging separation from CPB among cardiac surgery population. Additionally, results of influential factors and various adverse outcome analyses emphasize the potential of interventions aimed at preventing difficult or complex separation from CPB and reducing associated adverse outcomes.
Collapse
Affiliation(s)
- Dong X Chen
- Department of Anesthesiology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Tian H Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xing L Xiong
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Jing Shi
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Leng Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China -
| |
Collapse
|
6
|
Zhang W, Chen R, Xu K, Guo H, Li C, Sun X. Protective effect of Xinmai'an tablets via mediation of the AMPK/SIRT1/PGC-1α signaling pathway on myocardial ischemia-reperfusion injury in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155034. [PMID: 37611465 DOI: 10.1016/j.phymed.2023.155034] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/17/2023] [Accepted: 08/15/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Xinmai'an tablets are a compound Chinese medicine comprising six traditional Chinese medicines that have been clinically applied to treat cardiovascular diseases such as premature ventricular contractions for many years. However, pharmacological effects and underlying mechanisms of Xinmai'an tablet in protecting against myocardial ischemia-reperfusion injury (MIRI) were barely ever studied. PURPOSE To investigate the cardioprotective properties of Xinmai'an tablet against MIRI and the underlying molecular mechanism in rats. METHODS We initially established the UHPLC-QTRAP-MS/MS analysis method to ensure the controllable quality of Xinmai'an tablet. We further identified the cardioprotective effects of Xinmai'an tablet against MIRI using TTC staining, hematoxylin and eosin, echocardiography, the transmission electron microscope analysis, biochemical analysis, and ELISA. We then investigated whether the safeguarding effect of Xinmai'an tablet on MIRI model rats was related to AMPK/SIRT1/PGC-1α pathway via western blotting. RESULTS Xinmai'an tablet decreased myocardial infarct size; ameliorated cardiac function; alleviated myocardial and mitochondrial damage; and suppressed oxidative stress injury, vascular endothelial damage, and apoptosis response in MIRI model rats. Mechanistically, our results showed that Xinmai'an tablet can dramatically activate the AMPK/SIRT1/PGC-1αpathway and subsequently diminish mitochondrial oxidative stress damage. This was evidenced by increased ATP, Na+-K+-ATPase, and Ca2+-Mg2+-ATPase levels, upregulation of GLUT4, p-AMPK, SIRT1, and PGC-1α protein levels; and reduced GLUT1 protein level. CONCLUSION To the knowledge of the author of this article, this study is the first report of Xinmai'an tablet attenuating MIRI, potentially associated with the activation of the AMPK/SIRT1/PGC-1α pathway and subsequent reduction of mitochondrial oxidative stress damage. These findings reveal a novel pharmacological effect and mechanism of action of Xinmai'an tablet and highlight a promising therapeutic drug for ischemic cardiovascular diseases.
Collapse
Affiliation(s)
- Wei Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; Hutchison Whampoa Guangzhou Bai Yunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China
| | - Rongchang Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Keyi Xu
- Hutchison Whampoa Guangzhou Bai Yunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China
| | - Haibiao Guo
- Hutchison Whampoa Guangzhou Bai Yunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China
| | - Chuyuan Li
- Hutchison Whampoa Guangzhou Bai Yunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China.
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China.
| |
Collapse
|
7
|
Regan JA, Mentz RJ, Nguyen M, Green JB, Truby LK, Ilkayeva O, Newgard CB, Buse JB, Sourij H, Sjöström CD, Sattar N, McGarrah RW, Zheng Y, McGuire DK, Standl E, Armstrong P, Peterson ED, Hernandez AF, Holman RR, Shah SH. Mitochondrial metabolites predict adverse cardiovascular events in individuals with diabetes. JCI Insight 2023; 8:e168563. [PMID: 37552540 PMCID: PMC10544215 DOI: 10.1172/jci.insight.168563] [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: 01/06/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023] Open
Abstract
Metabolic mechanisms underlying the heterogeneity of major adverse cardiovascular (CV) event (MACE) risk in individuals with type 2 diabetes mellitus (T2D) remain unclear. We hypothesized that circulating metabolites reflecting mitochondrial dysfunction predict incident MACE in T2D. Targeted mass-spectrometry profiling of 60 metabolites was performed on baseline plasma samples from the Trial Evaluating Cardiovascular Outcomes with Sitagliptin (TECOS; discovery cohort) and Exenatide Study of Cardiovascular Event Lowering (EXSCEL; validation cohort) biomarker substudy cohorts. A principal components analysis metabolite factor comprising medium-chain acylcarnitines (MCACs) was associated with MACE in TECOS and validated in EXSCEL, with higher levels associated with higher MACE risk. Meta-analysis showed that long-chain acylcarnitines (LCACs) and dicarboxylacylcarnitines were also associated with MACE. Metabolites remained associated with MACE in multivariate models and favorably changed with exenatide therapy. A third cohort (Cardiac Catheterization Genetics [CATHGEN]) with T2D was assessed to determine whether these metabolites improved discriminative capability of multivariate models for MACE. Nine metabolites (MCACs and LCACs and 1 dicarboxylacylcarnitine) were associated with time to MACE in the CATHGEN cohort. Addition of these metabolites to clinical models minimally improved the discriminative capability for MACE but did significantly down reclassify risk. Thus, metabolites reporting on dysregulated mitochondrial fatty acid oxidation are present in higher levels in individuals with T2D who experience subsequent MACE. These biomarkers may improve CV risk prediction models, be therapy responsive, and highlight emerging risk mechanisms.
Collapse
Affiliation(s)
- Jessica A. Regan
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
- Duke University Department of Medicine, Durham, North Carolina, USA
| | - Robert J. Mentz
- Duke University Department of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Maggie Nguyen
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | - Jennifer B. Green
- Duke University Department of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Lauren K. Truby
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
- Duke University Department of Medicine, Durham, North Carolina, USA
| | - Olga Ilkayeva
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
- Duke University Department of Medicine, Durham, North Carolina, USA
| | | | - John B. Buse
- University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina, USA
| | - Harald Sourij
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - C. David Sjöström
- Late-stage Development, Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Naveed Sattar
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Robert W. McGarrah
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
- Duke University Department of Medicine, Durham, North Carolina, USA
| | - Yinggan Zheng
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Darren K. McGuire
- University of Texas Southwestern Medical Center and Parkland Health and Hospital System, Dallas, Texas, USA
| | - Eberhard Standl
- Diabetes Research Group at Munich Helmholtz Center, Munich, Germany
| | - Paul Armstrong
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Eric D. Peterson
- University of Texas Southwestern Medical Center and Parkland Health and Hospital System, Dallas, Texas, USA
| | - Adrian F. Hernandez
- Duke University Department of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Rury R. Holman
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Svati H. Shah
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
- Duke University Department of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| |
Collapse
|
8
|
Ullah A, Sajid S, Qureshi M, Kamran M, Anwaar MA, Naseem MA, Zaman MU, Mahmood F, Rehman A, Shehryar A, Nadeem MA. Novel Biomarkers and the Multiple-Marker Approach in Early Detection, Prognosis, and Risk Stratification of Cardiac Diseases: A Narrative Review. Cureus 2023; 15:e42081. [PMID: 37602073 PMCID: PMC10434821 DOI: 10.7759/cureus.42081] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Cardiac diseases are a primary cause of mortality worldwide, underscoring the importance of early identification and risk stratification to enhance patient outcomes. Biomarkers have become important tools for the risk assessment of cardiovascular disease and monitoring disease progression. This narrative review focuses on the multiple-marker approach, which involves simultaneously evaluating several biomarkers for the early detection and risk stratification of heart diseases. The review covers the clinical applications of novel biomarkers, such as high-sensitivity troponin, galectin-3, source of tumorigenicity 2, B-type natriuretic peptide and N-terminal pro-B-type natriuretic peptide, growth differentiation factor 15, myeloperoxidase, fatty acid-binding protein, C-reactive protein, lipoprotein-associated phospholipase A2, microRNAs, circulating endothelial cells, and ischemia-modified albumin. These biomarkers have demonstrated potential in identifying people who are at high risk for developing heart disease and in providing prognostic data. Given the complexity of cardiac illnesses, the multiple-marker approach to risk assessment is extremely beneficial. Implementing the multiple-marker strategy can improve risk stratification, diagnostic accuracy, and patient care in heart disease patients.
Collapse
Affiliation(s)
| | - Samar Sajid
- Medicine, Dow University of Health Sciences, Karachi, PAK
| | - Maria Qureshi
- Family Medicine, Ayub Medical College, Abbottabad, PAK
| | | | - Mohammad Ahsan Anwaar
- Internal Medicine, CMH Lahore Medical College and Institute of Dentistry, Lahore, PAK
| | | | | | - Fizza Mahmood
- Cardiology/Cardiac Surgery, Shifa International Hospital Islamabad, Islamabad, PAK
| | | | | | - Muhammad A Nadeem
- Medicine and Surgery, Shifa International Hospital Islamabad, Islamabad, PAK
| |
Collapse
|
9
|
Guo Z, Wang M, Ying X, Yuan J, Wang C, Zhang W, Tian S, Yan X. Caloric restriction increases the resistance of aged heart to myocardial ischemia/reperfusion injury via modulating AMPK-SIRT 1-PGC 1a energy metabolism pathway. Sci Rep 2023; 13:2045. [PMID: 36739302 PMCID: PMC9899227 DOI: 10.1038/s41598-023-27611-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 01/04/2023] [Indexed: 02/06/2023] Open
Abstract
A large number of data suggest that caloric restriction (CR) has a protective effect on myocardial ischemia/reperfusion injury (I/R) in the elderly. However, the mechanism is still unclear. In this study, we created the I/R model in vivo by ligating the mice left coronary artery for 45 min followed by reperfusion. C57BL/6J wild-type mice were randomly divided into a young group fed ad libitum (y-AL), aged fed ad libitum (a-AL) and aged calorie restriction group (a-CR, 70% diet restriction), and fed for 6 weeks. The area of myocardial infarction was measured by Evan's blue-TTC staining, plasma cholesterol content quantified by ELISA, fatty acids and glucose measured by Langendorff working system, as well as protein expression of AMPK/SIRT1/PGC1a signaling pathway related factors in myocardial tissue detected by immunoblotting. Our results showed that CR significantly reduced infarct size in elderly mice after I/R injury, promoted glycolysis regardless of I/R injury, and restored myocardial glucose uptake in elderly mice. Compared with a-AL group, CR significantly promoted the expression of p-AMPK, SIRT1, p-PGC1a, and SOD2, but decreased PPARγ expression in aged mice. In conclusion, our results suggest that CR protects elderly mice from I/R injury by altering myocardial substrate energy metabolism via the AMPK/SIRT1/PGC1a pathway.
Collapse
Affiliation(s)
- Zhijia Guo
- 1st Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Meng Wang
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaodong Ying
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiyu Yuan
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chenggang Wang
- Shanxi Traditional Chinese Medicine Hospital, Taiyuan, Shanxi, China
| | - Wenjie Zhang
- 1st Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shouyuan Tian
- 1st Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoyan Yan
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
10
|
Ge X, Meng Q, Liu X, Liu J, Ma X, Shi S, Li M, Lin F, Liang X, Gong X, Liu Z, Han W, Zhou X. Alterations of long noncoding RNAs and mRNAs in extracellular vesicles derived from the murine heart post-ischemia-reperfusion injury. J Cell Mol Med 2022; 26:6006-6018. [PMID: 36444487 PMCID: PMC9753460 DOI: 10.1111/jcmm.17617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/30/2022] Open
Abstract
Extracellular vesicles (EVs) play important roles in cardiovascular diseases by delivering their RNA cargos. However, the features and possible role of the lncRNAs and mRNAs in cardiac EVs during ischemia-reperfusion (IR) remain unclear. Therefore, we performed RNA sequencing analysis to profile the features of lncRNAs and mRNAs and predicted their potential functions. Here, we demonstrated that the severity of IR injury was significantly correlated with cardiac EV production. RNA sequencing identified 73 significantly differentially expressed (DE) lncRNAs (39 upregulated and 34 downregulated) and 720 DE-mRNAs (317 upregulated and 403 downregulated). Gene Ontology (GO) and pathway analysis were performed to predict the potential functions of the DE-lncRNAs and mRNAs. The lncRNA-miRNA-mRNA ceRNA network showed the possible functions of DE-lncRNAs with DE-mRNAs which are enriched in the pathways of T cell receptor signalling pathway and cell adhesion molecules. Moreover, the expressions of ENSMUST00000146010 and ENSMUST00000180630 were negatively correlated with the severity of IR injury. A significant positive correlation was revealed between TCONS_00010866 expression and the severity of the cardiac injury. These findings revealed the lncRNA and mRNA profiles in the heart derived EVs and provided potential targets and pathways involved in cardiac IR injury.
Collapse
Affiliation(s)
- Xinyu Ge
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Department of Cardiothoracic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Qingshu Meng
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Xuan Liu
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Department of Cardiothoracic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Jing Liu
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Department of Cardiothoracic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Xiaoxue Ma
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Shanshan Shi
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Mimi Li
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Fang Lin
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Department of Cardiothoracic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Xiaoting Liang
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji UniversityShanghaiChina
| | - Xin Gong
- Department of Heart FailureShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Zhongmin Liu
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Department of Cardiothoracic SurgeryShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Institute of Stem Cell Research and Clinical TranslationShanghaiChina
| | - Wei Han
- Department of Heart FailureShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Xiaohui Zhou
- Research Center for Translational MedicineShanghai East Hospital, Tongji University School of MedicineShanghaiChina,Shanghai Heart Failure Research CenterShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| |
Collapse
|
11
|
Schwartz B, Gjini P, Gopal DM, Fetterman JL. Inefficient Batteries in Heart Failure: Metabolic Bottlenecks Disrupting the Mitochondrial Ecosystem. JACC Basic Transl Sci 2022; 7:1161-1179. [PMID: 36687274 PMCID: PMC9849281 DOI: 10.1016/j.jacbts.2022.03.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 02/01/2023]
Abstract
Mitochondrial abnormalities have long been described in the setting of cardiomyopathies and heart failure (HF), yet the mechanisms of mitochondrial dysfunction in cardiac pathophysiology remain poorly understood. Many studies have described HF as an energy-deprived state characterized by a decline in adenosine triphosphate production, largely driven by impaired oxidative phosphorylation. However, impairments in oxidative phosphorylation extend beyond a simple decline in adenosine triphosphate production and, in fact, reflect pervasive metabolic aberrations that cannot be fully appreciated from the isolated, often siloed, interrogation of individual aspects of mitochondrial function. With the application of broader and deeper examinations into mitochondrial and metabolic systems, recent data suggest that HF with preserved ejection fraction is likely metabolically disparate from HF with reduced ejection fraction. In our review, we introduce the concept of the mitochondrial ecosystem, comprising intricate systems of metabolic pathways and dynamic changes in mitochondrial networks and subcellular locations. The mitochondrial ecosystem exists in a delicate balance, and perturbations in one component often have a ripple effect, influencing both upstream and downstream cellular pathways with effects enhanced by mitochondrial genetic variation. Expanding and deepening our vantage of the mitochondrial ecosystem in HF is critical to identifying consistent metabolic perturbations to develop therapeutics aimed at preventing and improving outcomes in HF.
Collapse
Key Words
- ADP, adenosine diphosphate
- ANT1, adenine translocator 1
- ATP, adenosine triphosphate
- CVD, cardiovascular disease
- DCM, dilated cardiomyopathy
- DRP-1, dynamin-related protein 1
- EET, epoxyeicosatrienoic acid
- FADH2/FAD, flavin adenine dinucleotide
- HETE, hydroxyeicosatetraenoic acid
- HF, heart failure
- HFpEF, heart failure with preserved ejection fraction
- HFrEF, heart failure with reduced ejection fraction
- HIF1α, hypoxia-inducible factor 1α
- LV, left ventricle
- LVAD, left ventricular assist device
- LVEF, left ventricular ejection fraction
- NADH/NAD+, nicotinamide adenine dinucleotide
- OPA1, optic atrophy protein 1
- OXPHOS, oxidative phosphorylation
- PGC1-α, peroxisome proliferator-activated receptor gamma coactivator 1 alpha
- SIRT1-7, sirtuins 1-7
- cardiomyopathy
- heart failure
- iPLA2γ, Ca2+-independent mitochondrial phospholipase
- mPTP, mitochondrial permeability transition pore
- metabolism
- mitochondria
- mitochondrial ecosystem
- mtDNA, mitochondrial DNA
Collapse
Affiliation(s)
- Brian Schwartz
- Evans Department of Medicine, Section of Internal Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Petro Gjini
- Evans Department of Medicine, Section of Internal Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Deepa M Gopal
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jessica L Fetterman
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Xiong Y, Jiang L, Li T. Aberrant branched-chain amino acid catabolism in cardiovascular diseases. Front Cardiovasc Med 2022; 9:965899. [PMID: 35911554 PMCID: PMC9334649 DOI: 10.3389/fcvm.2022.965899] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/29/2022] [Indexed: 01/04/2023] Open
Abstract
Globally, cardiovascular diseases are the leading cause of death. Research has focused on the metabolism of carbohydrates, fatty acids, and amino acids to improve the prognosis of cardiovascular diseases. There are three types of branched-chain amino acids (BCAAs; valine, leucine, and isoleucine) required for protein homeostasis, energy balance, and signaling pathways. Increasing evidence has implicated BCAAs in the pathogenesis of multiple cardiovascular diseases. This review summarizes the biological origin, signal transduction pathways and function of BCAAs as well as their significance in cardiovascular diseases, including myocardial hypertrophy, heart failure, coronary artery disease, diabetic cardiomyopathy, dilated cardiomyopathy, arrhythmia and hypertension.
Collapse
Affiliation(s)
- Yixiao Xiong
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Ling Jiang
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Tao Li
- Department of Anesthesiology, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Mitochondria and Metabolism, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Tao Li,
| |
Collapse
|
13
|
Mu H, Yang R, Wang S, Zhang W, Wang X, Li H, Dong J, Chen W, Yu X, Ji F. Association of Serum β-Hydroxybutyrate and Coronary Artery Disease in an Urban Chinese Population. Front Nutr 2022; 9:828824. [PMID: 35252305 PMCID: PMC8893320 DOI: 10.3389/fnut.2022.828824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Ketone bodies, including β-hydroxybutyrate (BHB), acetoacetate (AA), and acetone, can substitute and alternate with glucose under conditions of fuel/food deficiency. Ketone-body metabolism is increased in a myriad of tissue-metabolism disorders. Perturbations in metabolism are major contributors to coronary artery disease (CAD). We investigated the association of BHB with CAD. A total of 2,970 people of Chinese Han ethnicity were enrolled. The Gensini score was calculated for all patients who had positive findings. The serum level of BHB and other laboratory parameters were measured. The association of serum levels of metabolites with traditionally risk factors and CAD severity was analyzed. The BHB was found to be associated with some traditional risk factors of CAD and CAD severity, as determined by the Gensini score or the number of diseased regions. Moreover, BHB was associated with the T3/T1 tertiles of the Gensini score after the adjustment for traditional risk factors by multivariable logistic regression analysis. The association of BHB with CAD severity was more obvious in women. Taken together, these data suggest that the circulating BHB level is independently associated with CAD severity, and that this association is more pronounced in women.
Collapse
Affiliation(s)
- Hongna Mu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Ruiyue Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Siming Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Wenduo Zhang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinyue Wang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Hongxia Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Jun Dong
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Wenxiang Chen
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
| | - Xue Yu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Xue Yu
| | - Fusui Ji
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Fusui Ji
| |
Collapse
|
14
|
Jiang M, Xie X, Cao F, Wang Y. Mitochondrial Metabolism in Myocardial Remodeling and Mechanical Unloading: Implications for Ischemic Heart Disease. Front Cardiovasc Med 2021; 8:789267. [PMID: 34957264 PMCID: PMC8695728 DOI: 10.3389/fcvm.2021.789267] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022] Open
Abstract
Ischemic heart disease refers to myocardial degeneration, necrosis, and fibrosis caused by coronary artery disease. It can lead to severe left ventricular dysfunction (LVEF ≤ 35–40%) and is a major cause of heart failure (HF). In each contraction, myocardium is subjected to a variety of mechanical forces, such as stretch, afterload, and shear stress, and these mechanical stresses are clinically associated with myocardial remodeling and, eventually, cardiac outcomes. Mitochondria produce 90% of ATP in the heart and participate in metabolic pathways that regulate the balance of glucose and fatty acid oxidative phosphorylation. However, altered energetics and metabolic reprogramming are proved to aggravate HF development and progression by disturbing substrate utilization. This review briefly summarizes the current insights into the adaptations of cardiomyocytes to mechanical stimuli and underlying mechanisms in ischemic heart disease, with focusing on mitochondrial metabolism. We also discuss how mechanical circulatory support (MCS) alters myocardial energy metabolism and affects the detrimental metabolic adaptations of the dysfunctional myocardium.
Collapse
Affiliation(s)
- Min Jiang
- Department of Cardiology, National Clinical Research Center for Geriatric Disease, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,College of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army General Hospital, Beijing, China.,Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaoye Xie
- Department of Cardiology, National Clinical Research Center for Geriatric Disease, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China.,Department of Cadre Ward, The 960 Hospital of Chinese People's Liberation Army, Jinan, China
| | - Feng Cao
- Department of Cardiology, National Clinical Research Center for Geriatric Disease, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yabin Wang
- Department of Cardiology, National Clinical Research Center for Geriatric Disease, The Second Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China.,Medical School of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
15
|
Liu X, Jin F, Wang C, Zhao S, Han S, Jiang P, Cui C. Targeted metabolomics analysis of serum amino acid profiles in patients with Moyamoya disease. Amino Acids 2021; 54:137-146. [PMID: 34800175 DOI: 10.1007/s00726-021-03100-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/31/2021] [Indexed: 11/30/2022]
Abstract
Amino acids are one of the main metabolites in the body, and provide energy for the body and brain. The purpose of this study is to provide a profile of amino acid changes in the serum of patients with Moyamoya disease (MMD) and identify potential disease biomarkers. In this paper, we quantitatively determined the serum amino acid metabolic profiles of 43 MMD patients and 42 healthy controls (HCs). T test, multivariate statistical analysis, and receiver operating characteristic (ROC) curve analysis were used to identify candidate markers. Thirty-nine amino acids were quantified, and 12 amino acid levels differed significantly between the MMD patients and HCs. Moreover, based on ROC curve analysis, four amino acid (L-methionine, L-glutamic acid, β-alanine and o-phosphoserine) biomarkers showed high sensitivity and specificity (AUC > 0.90), and showed the best sensitivity and specificity in MetaboAnalyst 5.0 using binary logistic regression analysis. We have provided serum amino acid metabolic profiles of MMD patients, and identified four potential biomarkers which may both provide clinicians with an objective diagnostic method for early detection of MMD and further our understanding of MMD pathogenesis.
Collapse
Affiliation(s)
- Xi Liu
- Department of Pharmacy, Linfen People's Hospital, Linfen, China
| | - Feng Jin
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Changshui Wang
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Shiyuan Zhao
- Jining First People's Hospital, Jining Medical University, Jining, China
| | - Shasha Han
- Jining Life Science Center, Jining, China
| | - Pei Jiang
- Jining First People's Hospital, Jining Medical University, Jining, China.
| | - Changmeng Cui
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.
| |
Collapse
|
16
|
Wang L, Goldberg EM, Taylor CG, Zahradka P, Aliani M. Analyses of serum and urinary metabolites in individuals with peripheral artery disease (PAD) consuming a bean-rich diet: Relationships with drug metabolites. Appl Physiol Nutr Metab 2021; 47:243-252. [PMID: 34699735 DOI: 10.1139/apnm-2021-0495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Peripheral artery disease (PAD) has high morbidity and mortality rates. A metabolomics approach was employed to determine whether consumption of bean-rich diets for 8 weeks would impact the metabolomic profile of PAD individuals. Serum and urine, collected from 54 participants with clinical PAD at baseline and after 8 weeks on 0.3 cups beans/d (n=19), 0.6 cups beans/d (n= 20), or control (n=23) diet, and the beans were extracted and analyzed using LC-QTOF-MS. As a result, PGE2 p-acetamidophenyl ester, PGF2α diethyl amide and 5-L-glutamyl-L-alanine were significantly changed in the serum or urine of bean groups compared to control. Significant changes (P<0.05) in the profile and/or levels of 22 flavonoids present in bean extracts showed the potential importance of the mixture of beans used in this study. In a subset of participants taking metoprolol, after 8 weeks the bean-rich diets significantly elevated metoprolol in the serum while reducing it in urine compared to baseline. In addition, the diets significantly enhanced the urinary excretion of metformin. In conclusion, several biochemical pathways including prostaglandins and glutathione were affected by bean consumption. Significant changes in the metabolism of metoprolol and metformin with bean consumption suggested the presence of diet-drug interactions that may require adjustment of the prescribed dose. ClinicalTrials.gov Identifier: NCT01382056 Novelty: • Bean consumption by people with PAD alters the levels of certain metabolites in serum and urine • Different bean types (black, red kidney, pinto, navy) have unique flavonoid profiles • Metabolomics revealed potential diet-dug interactions as serum and/or urinary levels of metoprolol and metformin are modified by bean consumption.
Collapse
Affiliation(s)
- Le Wang
- University of Manitoba, 8664, Winnipeg, Manitoba, Canada;
| | | | - Carla G Taylor
- St. Boniface Hospital Research Centre, Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, Manitoba, Canada.,University of Manitoba, Physiology, Winnipeg, Manitoba, Canada;
| | - Peter Zahradka
- St. Boniface Hospital Research Centre, Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, Manitoba, Canada.,University of Manitoba, Physiology, Winnipeg, Manitoba, Canada;
| | - Michel Aliani
- University of Manitoba, 8664, Winnipeg, Manitoba, Canada, R3T 2N2;
| |
Collapse
|
17
|
Chacko S, Mamas MA, El-Omar M, Simon D, Haseeb S, Fath-Ordoubadi F, Clarke B, Neyses L, Dunn WB. Perturbations in cardiac metabolism in a human model of acute myocardial ischaemia. Metabolomics 2021; 17:76. [PMID: 34424431 PMCID: PMC8382649 DOI: 10.1007/s11306-021-01827-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 07/29/2021] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Acute myocardial ischaemia and the transition from reversible to irreversible myocardial injury are associated with abnormal metabolic patterns. Advances in metabolomics have extended our capabilities to define these metabolic perturbations on a metabolome-wide scale. OBJECTIVES This study was designed to identify cardiac metabolic changes in serum during the first 5 min following early myocardial ischaemia in humans, applying an untargeted metabolomics approach. METHODS Peripheral venous samples were collected from 46 patients in a discovery study (DS) and a validation study (VS) (25 for DS, 21 for VS). Coronary sinus venous samples were collected from 7 patients (4 for DS, 3 for VS). Acute myocardial ischaemia was induced by transient coronary occlusion during percutaneous coronary intervention (PCI). Plasma samples were collected at baseline (prior to PCI) and at 1 and 5 min post-coronary occlusion. Samples were analyzed by Ultra Performance Liquid Chromatography-Mass Spectrometry in an untargeted metabolomics approach. RESULTS The study observed changes in the circulating levels of metabolites at 1 and 5 min following transient coronary ischaemia. Both DS and VS identified 54 and 55 metabolites as significant (P < 0.05) when compared to baseline levels, respectively. Fatty acid beta-oxidation and anaerobic respiration, lysoglycerophospholipids, arachidonic acid, docosahexaenoic acid, tryptophan metabolism and sphingosine-1-phosphate were identified as mechanistically important. CONCLUSION Using an untargeted metabolomics approach, the study identified important cardiac metabolic changes in peripheral and coronary sinus plasma, in a human model of controlled acute myocardial ischaemia. Distinct classes of metabolites were shown to be involved in the rapid cardiac response to ischemia and provide insights into diagnostic and interventional targets.
Collapse
Affiliation(s)
- Sanoj Chacko
- Division of Cardiology, Queen's University, Kingston, ON, Canada.
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK.
- Keele Cardiovascular Research Group, Keele University, Stoke-on-Trent, UK.
- Manchester Heart Centre, Manchester Royal Infirmary, Central Manchester University Hospitals NHS Trust, Manchester, UK.
- Kingston Health Sciences Centre, Queen's University, 76 Stuart St, Kingston, ON, Canada.
| | - Mamas A Mamas
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
- Keele Cardiovascular Research Group, Keele University, Stoke-on-Trent, UK
| | - Magdi El-Omar
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
- Manchester Heart Centre, Manchester Royal Infirmary, Central Manchester University Hospitals NHS Trust, Manchester, UK
| | - David Simon
- Department of Chemistry, Queen's University, Kingston, ON, Canada
| | - Sohaib Haseeb
- Division of Cardiology, Queen's University, Kingston, ON, Canada
| | - Farzin Fath-Ordoubadi
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
- Manchester Heart Centre, Manchester Royal Infirmary, Central Manchester University Hospitals NHS Trust, Manchester, UK
| | - Bernard Clarke
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
- School of Chemistry and Manchester Centre for Integrative Systems Biology, Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Ludwig Neyses
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
- University of Luxembourg, 4365, Esch-sur-Alzette, Luxembourg
| | - Warwick B Dunn
- School of Chemistry and Manchester Centre for Integrative Systems Biology, Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
- School of Biosciences and Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
18
|
Cedars A, Manlhiot C, Ko JM, Bottiglieri T, Arning E, Weingarten A, Opotowsky A, Kutty S. Metabolomic Profiling of Adults with Congenital Heart Disease. Metabolites 2021; 11:metabo11080525. [PMID: 34436466 PMCID: PMC8398700 DOI: 10.3390/metabo11080525] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/29/2021] [Accepted: 07/31/2021] [Indexed: 11/16/2022] Open
Abstract
Metabolomic analysis may provide an integrated assessment in genetically and pathologically heterogeneous populations. We used metabolomic analysis to gain mechanistic insight into the small and diverse population of adults with congenital heart disease (ACHD). Consecutive ACHD patients seen at a single institution were enrolled. Clinical variables and whole blood were collected at regular clinical visits. Stored plasma samples were analyzed for the concentrations of 674 metabolites and metabolic markers using mass spectrometry with internal standards. These samples were compared to 28 simultaneously assessed healthy non-ACHD controls. Principal component analysis and multivariable regression modeling were used to identify metabolites associated with clinical outcomes in ACHD. Plasma from ACHD and healthy control patients differed in the concentrations of multiple metabolites. Differences between control and ACHD were greater in number and in degree than those between ACHD anatomic groups. A metabolite cluster containing amino acids and metabolites of amino acids correlated with negative clinical outcomes across all anatomic groups. Metabolites in the arginine metabolic pathway, betaine, dehydroepiandrosterone, cystine, 1-methylhistidine, serotonin and bile acids were associated with specific clinical outcomes. Metabolic markers of disease may both be useful as biomarkers for disease activity and suggest etiologically related pathways as possible targets for disease-modifying intervention.
Collapse
Affiliation(s)
- Ari Cedars
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21218, USA; (C.M.); (J.-M.K.); (S.K.)
- Correspondence:
| | - Cedric Manlhiot
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21218, USA; (C.M.); (J.-M.K.); (S.K.)
| | - Jong-Mi Ko
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21218, USA; (C.M.); (J.-M.K.); (S.K.)
| | - Teodoro Bottiglieri
- Center of Metabolomics, Baylor Scott & White Research Institute, Dallas, TX 75204, USA; (T.B.); (E.A.)
| | - Erland Arning
- Center of Metabolomics, Baylor Scott & White Research Institute, Dallas, TX 75204, USA; (T.B.); (E.A.)
| | - Angela Weingarten
- Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA;
| | - Alexander Opotowsky
- Department of Cardiology, Cincinnati Children’s Hospital, Cincinnati, OH 45229, USA;
| | - Shelby Kutty
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21218, USA; (C.M.); (J.-M.K.); (S.K.)
| |
Collapse
|
19
|
Nuñez-Gil IJ, Andrés M, Benito B, Bernardo E, Vedia O, Ferreira-Gonzalez I, Barba I. Serum Metabolomic Analysis Suggests Impairment of Myocardial Energy Production in Takotsubo Syndrome. Metabolites 2021; 11:metabo11070439. [PMID: 34357333 PMCID: PMC8303832 DOI: 10.3390/metabo11070439] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Takotsubo syndrome is a complex entity that, although it usually has a good prognosis, can be life threatening. While recent advances have improved the knowledge of takotsubo syndrome, many aspects of its etiology still remain uncertain. Metabolomics, a hypothesis generating approach, could provide novel pathophysiology information about this disease. METHODS AND RESULTS Serum samples were obtained from takotsubo (n = 19) and acute myocardial infarction patients (n = 8) at the cath lab and, in the case of takotsubo, again once the patient had recovered, 3 months after the main event. 1H NMR spectra of the serum were acquired at 9.4T using a CPMG pulse sequence (32 ms effective delay). Supervised and unsupervised pattern recognition approaches where applied to the data. Pattern recognition was able to differentiate between takotsubo and acute myocardial infarction during the acute phase with 95% accuracy. Myocardial infarction patients showed an increase in lipid signals, a known risk factor for the disease while takotsubo patients showed a relative increase in acetate that could suggest a reduced turnover of the Krebs cycle. When comparing acute and recovered phases, we could detect an increase in alanine and creatine once patients recovered. CONCLUSIONS Our results demonstrate that takotsubo syndrome is metabolically different than AMI, showing limited myocardial energy production capacity during the acute phase. We achieved high classification success against AMI; however, this study should be considered as a proof of concept regarding clinical application of metabolic profiling in takotsubo cardiomyopathy.
Collapse
Affiliation(s)
- Iván J. Nuñez-Gil
- Interventional Cardiology Unit, Cardiovascular Institute, Hospital Clínico San Carlos, Calle del Prof Martín Lagos, s/n, 28040 Madrid, Spain; (I.J.N.-G.); (E.B.); (O.V.)
| | - Mireia Andrés
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (M.A.); (B.B.)
| | - Begoña Benito
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (M.A.); (B.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Esther Bernardo
- Interventional Cardiology Unit, Cardiovascular Institute, Hospital Clínico San Carlos, Calle del Prof Martín Lagos, s/n, 28040 Madrid, Spain; (I.J.N.-G.); (E.B.); (O.V.)
| | - Oscar Vedia
- Interventional Cardiology Unit, Cardiovascular Institute, Hospital Clínico San Carlos, Calle del Prof Martín Lagos, s/n, 28040 Madrid, Spain; (I.J.N.-G.); (E.B.); (O.V.)
| | - Ignacio Ferreira-Gonzalez
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (M.A.); (B.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (I.F.-G.); (I.B.)
| | - Ignasi Barba
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (M.A.); (B.B.)
- Faculty of Medicine, University of Vic-Central University of Catalonia (UVicUCC), Can Baumann, Ctra, de Roda, 70, 08500 Vic, Spain
- Vall d’Hebron Institut d’Oncologia (VHIO), CELLEX CENTER C/ Natzaret 115-117, 08035 Barcelona, Spain
- Correspondence: (I.F.-G.); (I.B.)
| |
Collapse
|
20
|
Nodari A, Scambi I, Peroni D, Calabria E, Benati D, Mannucci S, Manfredi M, Frontini A, Visonà S, Bozzato A, Sbarbati A, Schena F, Marengo E, Krampera M, Galiè M. Interferon regulatory factor 7 impairs cellular metabolism in aging adipose-derived stromal cells. J Cell Sci 2021; 134:jcs256230. [PMID: 34096605 DOI: 10.1242/jcs.256230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 04/26/2021] [Indexed: 11/20/2022] Open
Abstract
Dysregulated immunity and widespread metabolic dysfunctions are the most relevant hallmarks of the passing of time over the course of adult life, and their combination at midlife is strongly related to increased vulnerability to diseases; however, the causal connection between them remains largely unclear. By combining multi-omics and functional analyses of adipose-derived stromal cells established from young (1 month) and midlife (12 months) mice, we show that an increase in expression of interferon regulatory factor 7 (IRF7) during adult life drives major metabolic changes, which include impaired mitochondrial function, altered amino acid biogenesis and reduced expression of genes involved in branched-chain amino acid (BCAA) degradation. Our results draw a new paradigm of aging as the 'sterile' activation of a cell-autonomous pathway of self-defense and identify a crucial mediator of this pathway, IRF7, as driver of metabolic dysfunction with age.
Collapse
Affiliation(s)
- Alice Nodari
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Ilaria Scambi
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Daniele Peroni
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Elisa Calabria
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Donatella Benati
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Silvia Mannucci
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Marcello Manfredi
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Alessandria, Italy
- Center for Translational Research on Autoimmune and Allergic Disease - CAAD, University of Piemonte Orientale, 28100 Novara, Italy
| | - Andrea Frontini
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 20121 Ancona, Italy
| | - Silvia Visonà
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
| | - Andrea Bozzato
- Department of Biomedical Sciences and Biotechnology, University of Brescia, 25123 Brescia, Italy
| | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Federico Schena
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| | - Emilio Marengo
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Alessandria, Italy
- Center for Translational Research on Autoimmune and Allergic Disease - CAAD, University of Piemonte Orientale, 28100 Novara, Italy
| | - Mauro Krampera
- Department of Medicine, Section of Hematology, Stem Cell Research Laboratory, University of Verona, 37134 Verona, Italy
| | - Mirco Galiè
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy
| |
Collapse
|
21
|
Abstract
Nitric oxide, studied to evaluate its role in cardiovascular physiology, has cardioprotective and therapeutic effects in cellular signaling, mitochondrial function, and in regulating inflammatory processes. Heme oxygenase (major role in catabolism of heme into biliverdin, carbon monoxide (CO), and iron) has similar effects as well. CO has been suggested as the molecule that is responsible for many of the above mentioned cytoprotective and therapeutic pathways as CO is a signaling molecule in the control of physiological functions. This is counterintuitive as toxic effects are related to its binding to hemoglobin. However, CO is normally produced in the body. Experimental evidence indicates that this toxic gas, CO, exerts cytoprotective properties related to cellular stress including the heart and is being assessed for its cytoprotective and cytotherapeutic properties. While survival of adult cardiomyocytes depends on oxidative phosphorylation (survival and resulting cardiac function is impaired by mitochondrial damage), mitochondrial biogenesis is modified by the heme oxygenase-1/CO system and can result in promotion of mitochondrial biogenesis by associating mitochondrial redox status to the redox-active transcription factors. It has been suggested that the heme oxygenase-1/CO system is important in differentiation of embryonic stem cells and maturation of cardiomyocytes which is thought to mitigate progression of degenerative cardiovascular diseases. Effects on other cardiac cells are being studied. Acute exposure to air pollution (and, therefore, CO) is associated with cardiovascular mortality, myocardial infarction, and heart failure, but changes in the endogenous heme oxygenase-1 system (and, thereby, CO) positively affect cardiovascular health. We will review the effect of CO on heart health and function in this article.
Collapse
Affiliation(s)
- Vicki L Mahan
- Department of Surgery and Pediatrics, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
22
|
Müller J, Bertsch T, Volke J, Schmid A, Klingbeil R, Metodiev Y, Karaca B, Kim SH, Lindner S, Schupp T, Kittel M, Poschet G, Akin I, Behnes M. Narrative review of metabolomics in cardiovascular disease. J Thorac Dis 2021; 13:2532-2550. [PMID: 34012599 PMCID: PMC8107570 DOI: 10.21037/jtd-21-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases are accompanied by disorders in the cardiac metabolism. Furthermore, comorbidities often associated with cardiovascular disease can alter systemic and myocardial metabolism contributing to worsening of cardiac performance and health status. Biomarkers such as natriuretic peptides or troponins already support diagnosis, prognosis and treatment of patients with cardiovascular diseases and are represented in international guidelines. However, as cardiovascular diseases affect various pathophysiological pathways, a single biomarker approach cannot be regarded as ideal to reveal optimal clinical application. Emerging metabolomics technology allows the measurement of hundreds of metabolites in biological fluids or biopsies and thus to characterize each patient by its own metabolic fingerprint, improving our understanding of complex diseases, significantly altering the management of cardiovascular diseases and possibly personalizing medicine. This review outlines current knowledge, perspectives as well as limitations of metabolomics for diagnosis, prognosis and treatment of cardiovascular diseases such as heart failure, atherosclerosis, ischemic and non-ischemic cardiomyopathy. Furthermore, an ongoing research project tackling current inconsistencies as well as clinical applications of metabolomics will be discussed. Taken together, the application of metabolomics will enable us to gain more insights into pathophysiological interactions of metabolites and disease states as well as improving therapies of patients with cardiovascular diseases in the future.
Collapse
Affiliation(s)
- Julian Müller
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Thomas Bertsch
- Institute of Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremburg General Hospital, Paracelsus Medical University, Nuremberg, Germany
| | - Justus Volke
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Alexander Schmid
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rebecca Klingbeil
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Yulian Metodiev
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Bican Karaca
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Seung-Hyun Kim
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Simon Lindner
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Tobias Schupp
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Maximilian Kittel
- Institute for Clinical Chemistry, Faculty of Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Gernot Poschet
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Ibrahim Akin
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael Behnes
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
23
|
Zhou H, Ren J, Toan S, Mui D. Role of mitochondrial quality surveillance in myocardial infarction: From bench to bedside. Ageing Res Rev 2021; 66:101250. [PMID: 33388396 DOI: 10.1016/j.arr.2020.101250] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Myocardial infarction (MI) is the irreversible death of cardiomyocyte secondary to prolonged lack of oxygen or fresh blood supply. Historically considered as merely cardiomyocyte powerhouse that manufactures ATP and other metabolites, mitochondrion is recently being identified as a signal regulator that is implicated in the crosstalk and signal integration of cardiomyocyte contraction, metabolism, inflammation, and death. Mitochondria quality surveillance is an integrated network system modifying mitochondrial structure and function through the coordination of various processes including mitochondrial fission, fusion, biogenesis, bioenergetics, proteostasis, and degradation via mitophagy. Mitochondrial fission favors the elimination of depolarized mitochondria through mitophagy, whereas mitochondrial fusion preserves the mitochondrial network upon stress through integration of two or more small mitochondria into an interconnected phenotype. Mitochondrial biogenesis represents a regenerative program to replace old and damaged mitochondria with new and healthy ones. Mitochondrial bioenergetics is regulated by a metabolic switch between glucose and fatty acid usage, depending on oxygen availability. To maintain the diversity and function of mitochondrial proteins, a specialized protein quality control machinery regulates protein dynamics and function through the activity of chaperones and proteases, and induction of the mitochondrial unfolded protein response. In this review, we provide an overview of the molecular mechanisms governing mitochondrial quality surveillance and highlight the most recent preclinical and clinical therapeutic approaches to restore mitochondrial fitness during both MI and post-MI heart failure.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China.
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Sam Toan
- Department of Chemical Engineering, University of Minnesota-Duluth, Duluth, MN 55812, USA
| | - David Mui
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
24
|
Easton ZJW, Regnault TRH. The Impact of Maternal Body Composition and Dietary Fat Consumption upon Placental Lipid Processing and Offspring Metabolic Health. Nutrients 2020; 12:nu12103031. [PMID: 33022934 PMCID: PMC7601624 DOI: 10.3390/nu12103031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/20/2022] Open
Abstract
The proportion of women of reproductive age who are overweight or obese is increasing globally. Gestational obesity is strongly associated in both human studies and animal models with early-onset development of adult-associated metabolic diseases including metabolic syndrome in the exposed offspring. However, animal model studies have suggested that gestational diet in obese pregnancies is an independent but underappreciated mediator of offspring risk for later life metabolic disease, and human diet consumption data have highlighted that many women do not follow nutritional guidelines prior to and during pregnancy. Thus, this review will highlight how maternal diet independent from maternal body composition impacts the risk for later-life metabolic disease in obesity-exposed offspring. A poor maternal diet, in combination with the obese metabolic state, are understood to facilitate pathological in utero programming, specifically through changes in lipid handling processes in the villous trophoblast layer of the placenta that promote an environment associated with the development of metabolic disease in the offspring. This review will additionally highlight how maternal obesity modulates villous trophoblast lipid processing functions including fatty acid transport, esterification and beta-oxidation. Further, this review will discuss how altering maternal gestational diet may ameliorate these functional changes in lipid metabolic processes in the obese placenta.
Collapse
Affiliation(s)
- Zachary J. W. Easton
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building Room 216, London, ON N6A 5C1, Canada;
- Correspondence: ; Tel.: +1-(519)-661-2111 (ext. 82869)
| | - Timothy R. H. Regnault
- Department of Physiology and Pharmacology, Western University, Medical Sciences Building Room 216, London, ON N6A 5C1, Canada;
- Department of Obstetrics and Gynaecology, London Health Science Centre-Victoria Hospital, B2-401, London, ON N6H 5W9, Canada
- Children’s Health Research Institute, 800 Commissioners Road East, London, ON N6C 2V5, Canada
- Lawson Health Research Institute, 750 Base Line Rd E, London, ON N6C 2R5, Canada
| |
Collapse
|
25
|
Li Y, Xiong Z, Yan W, Gao E, Cheng H, Wu G, Liu Y, Zhang L, Li C, Wang S, Fan M, Zhao H, Zhang F, Tao L. Branched chain amino acids exacerbate myocardial ischemia/reperfusion vulnerability via enhancing GCN2/ATF6/PPAR-α pathway-dependent fatty acid oxidation. Theranostics 2020; 10:5623-5640. [PMID: 32373236 PMCID: PMC7196282 DOI: 10.7150/thno.44836] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/08/2020] [Indexed: 12/15/2022] Open
Abstract
Rationale: Myocardial vulnerability to ischemia/reperfusion (I/R) injury is strictly regulated by energy substrate metabolism. Branched chain amino acids (BCAA), consisting of valine, leucine and isoleucine, are a group of essential amino acids that are highly oxidized in the heart. Elevated levels of BCAA have been implicated in the development of cardiovascular diseases; however, the role of BCAA in I/R process is not fully understood. The present study aims to determine how BCAA influence myocardial energy substrate metabolism and to further clarify the pathophysiological significance during cardiac I/R injury. Methods: Parameters of glucose and fatty acid metabolism were measured by seahorse metabolic flux analyzer in adult mouse cardiac myocytes with or without BCAA incubation. Chronic accumulation of BCAA was induced in mice receiving oral BCAA administration. A genetic mouse model with defective BCAA catabolism was also utilized. Mice were subjected to MI/R and the injury was assessed extensively at the whole-heart, cardiomyocyte, and molecular levels. Results: We confirmed that chronic accumulation of BCAA enhanced glycolysis and fatty acid oxidation (FAO) but suppressed glucose oxidation in adult mouse ventricular cardiomyocytes. Oral gavage of BCAA enhanced FAO in cardiac tissues, exacerbated lipid peroxidation toxicity and worsened myocardial vulnerability to I/R injury. Etomoxir, a specific inhibitor of FAO, rescued the deleterious effects of BCAA on I/R injury. Mechanistically, valine, leucine and their corresponding branched chain α-keto acid (BCKA) derivatives, but not isoleucine and its BCKA derivative, transcriptionally upregulated peroxisome proliferation-activated receptor alpha (PPAR-α). BCAA/BCKA induced PPAR-α upregulation through the general control nonderepresible-2 (GCN2)/ activating transcription factor-6 (ATF6) pathway. Finally, in a genetic mouse model with BCAA catabolic defects, chronic accumulation of BCAA increased FAO in myocardial tissues and sensitized the heart to I/R injury, which could be reversed by adenovirus-mediated PPAR-α silencing. Conclusions: We identify BCAA as an important nutrition regulator of myocardial fatty acid metabolism through transcriptional upregulation of PPAR-α. Chronic accumulation of BCAA, caused by either dietary or genetic factors, renders the heart vulnerable to I/R injury via exacerbating lipid peroxidation toxicity. These data support the notion that BCAA lowering methods might be potentially effective cardioprotective strategies, especially among patients with diseases characterized by elevated levels of BCAA, such as obesity and diabetes.
Collapse
|
26
|
Geng C, Cui C, Guo Y, Wang C, Zhang J, Han W, Jin F, Chen D, Jiang P. Metabolomic Profiling Revealed Potential Biomarkers in Patients With Moyamoya Disease. Front Neurosci 2020; 14:308. [PMID: 32372905 PMCID: PMC7186471 DOI: 10.3389/fnins.2020.00308] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/16/2020] [Indexed: 11/13/2022] Open
Abstract
Metabolomics is increasingly used to observe metabolic patterns and disease-specific metabolic biomarkers. However, serum metabolite analysis of moyamoya disease (MMD) is rarely reported. We investigated serum metabolites in MMD and compared them with those of healthy controls (HCs) using a non-targeted gas chromatography-mass spectrometry (GC-MS) approach to identify metabolic biomarkers associated with MMD. Forty-one patients with MMD diagnosed by cerebral angiography and 58 HCs were recruited for our study. Comparative analyses (univariate, multivariate, correlation, heatmaps, receiver operating characteristi curves) were performed between MMD patients and HCs. Twenty-five discriminating serum metabolic biomarkers between MMD patients and HCs were identified. Compared with HCs, MMD patients had higher levels of phenol, 2-hydroxybutyric acid, L-isoleucine, L-serine, glycerol, pelargonic acid, L-methionine, myristic acid, pyroglutamic acid, palmitic acid, palmitoleic acid, stearic acid, octadecanamide, monoglyceride (MG) (16:0/0:0/0:0), and MG (0:0/18:0/0:0), and lower levels of L-alanine, L-valine, urea, succinic acid, L-phenylalanine, L-threonine, L-tyrosine, edetic acid, and oleamide. These metabolic biomarkers are involved in several pathways and are closely associated with the metabolism of amino acids, lipids, carbohydrates, and carbohydrate translation. A GC-MS-based metabolomics approach could be useful in the clinical diagnosis of MMD. The identified biomarkers may be helpful to develop an objective diagnostic method for MMD and improve our understanding of MMD pathogenesis.
Collapse
Affiliation(s)
- Chunmei Geng
- Jining First People's Hospital, Jining Medical University, Jining, China
| | - Changmeng Cui
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Yujin Guo
- Jining First People's Hospital, Jining Medical University, Jining, China
| | - Changshui Wang
- Department of Clinical Translational Medicine, Jining Life Science Center, Jining, China
| | - Jun Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenxiu Han
- Jining First People's Hospital, Jining Medical University, Jining, China
| | - Feng Jin
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Dan Chen
- Jining First People's Hospital, Jining Medical University, Jining, China
| | - Pei Jiang
- Jining First People's Hospital, Jining Medical University, Jining, China
| |
Collapse
|
27
|
Liu T, Wen H, Li H, Xu H, Xiao N, Liu R, Chen L, Sun Y, Song L, Bai C, Ge J, Zhang Y, Chen J. Oleic Acid Attenuates Ang II (Angiotensin II)-Induced Cardiac Remodeling by Inhibiting FGF23 (Fibroblast Growth Factor 23) Expression in Mice. Hypertension 2020; 75:680-692. [DOI: 10.1161/hypertensionaha.119.14167] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Plasma metabolic profiles were compared between patients with hypertension with and without left ventricular hypertrophy and significantly decreased oleic acid (OA) levels were observed in the peripheral blood of patients with hypertension with left ventricular hypertrophy. We sought to determine the effect and underlying mechanisms of OA on cardiac remodeling. In vitro studies with isolated neonatal mouse cardiomyocytes and cardiac fibroblasts revealed that OA significantly attenuated Ang II (angiotensin II)-induced cardiomyocyte growth and cardiac fibroblast collagen expression. In vivo, cardiac function, hypertrophic growth of cardiomyocytes, and fibrosis were analyzed after an Ang II (1000 ng/kg/minute) pump was implanted for 14 days. We found that OA could significantly prevent Ang II-induced cardiac remodeling in mice. RNA sequencing served as a gene expression roadmap highlighting gene expression changes in the hearts of Ang II-induced mice and OA-treated mice. The results revealed that FGF23 (fibroblast growth factor 23) expression was significantly upregulated in mouse hearts in response to Ang II infusion, which was significantly suppressed in the hearts of OA-treated mice. Furthermore, overexpression of FGF23 in the heart by injection of an AAV-9 vector aggravated Ang II-induced cardiac remodeling and impaired the protective effect of OA on cardiac remodeling. Further study found that OA could suppress Ang II-induced FGF23 expression by inhibiting the translocation of Nurr1 (nuclear receptor–related 1 protein) from the cytoplasm to the nucleus. Our findings suggest a novel role of OA in preventing Ang II-induced cardiac remodeling via suppression of FGF23 expression.
Collapse
Affiliation(s)
- Tianlong Liu
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (T.L., H.W., H.L., N.X., Y.S., L.S., C.B., J.G., Y.Z.)
| | - Hongyan Wen
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (T.L., H.W., H.L., N.X., Y.S., L.S., C.B., J.G., Y.Z.)
| | - Hao Li
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (T.L., H.W., H.L., N.X., Y.S., L.S., C.B., J.G., Y.Z.)
| | | | - Ning Xiao
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (T.L., H.W., H.L., N.X., Y.S., L.S., C.B., J.G., Y.Z.)
| | | | - Luonan Chen
- Key Laboratory of Systems Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China (L.C.)
| | - Yingying Sun
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (T.L., H.W., H.L., N.X., Y.S., L.S., C.B., J.G., Y.Z.)
| | - Li Song
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (T.L., H.W., H.L., N.X., Y.S., L.S., C.B., J.G., Y.Z.)
| | - Congxia Bai
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (T.L., H.W., H.L., N.X., Y.S., L.S., C.B., J.G., Y.Z.)
| | - Jing Ge
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (T.L., H.W., H.L., N.X., Y.S., L.S., C.B., J.G., Y.Z.)
| | - Yinhui Zhang
- From the State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (T.L., H.W., H.L., N.X., Y.S., L.S., C.B., J.G., Y.Z.)
| | | |
Collapse
|
28
|
Wu G, Zhang W, Li H. Application of metabolomics for unveiling the therapeutic role of traditional Chinese medicine in metabolic diseases. JOURNAL OF ETHNOPHARMACOLOGY 2019; 242:112057. [PMID: 31279867 DOI: 10.1016/j.jep.2019.112057] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/12/2019] [Accepted: 07/03/2019] [Indexed: 05/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional medicine has been practiced for thousands of years in China and some Asian countries. Traditional Chinese Medicine (TCM) is characterized as multi-component and multiple targets in disease therapy, and it is a great challenge for elucidating the mechanisms of TCM. AIM OF THE REVIEW Comprehensively summarize the application of metabolomics in biomarker discovery, stratification of TCM syndromes, and mechanism underlying TCM therapy on metabolic diseases. METHODS This review systemically searched the publications with key words such as metabolomics, traditional Chinese medicine, metabolic diseases, obesity, cardiovascular disease, diabetes mellitus in "Title OR Abstract" in major databases including PubMed, the Web of Science, Google Scholar, Science Direct, CNKI from 2010 to 2019. RESULTS A total of 135 papers was searched and included in this review. An overview of articles indicated that metabolic characteristics may be a hallmark of different syndromes/models of metabolic diseases, which provides a new perspective for disease diagnosis and therapeutic optimization. Moreover, TCM treatment has significantly altered the metabolic perturbations associated with metabolic diseases, which may be an important mechanism for the therapeutic effect of TCM. CONCLUSIONS Until now, many metabolites and differential biomarkers related to the pathogenesis of metabolic diseases and TCM therapy have been discovered through metabolomics research. Unfortunately, the biological role and mechanism of disease-related metabolites were largely unclarified so far, which warrants further investigation.
Collapse
Affiliation(s)
- Gaosong Wu
- Interdisciplinary Science Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weidong Zhang
- Interdisciplinary Science Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Department of Phytochemistry, School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.
| | - Houkai Li
- Interdisciplinary Science Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
29
|
Lee TH, Cheng ML, Shiao MS, Lin CN. Metabolomics study in severe extracranial carotid artery stenosis. BMC Neurol 2019; 19:138. [PMID: 31234801 PMCID: PMC6589885 DOI: 10.1186/s12883-019-1371-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 06/18/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Significant genetic association has been found in patients with severe carotid artery stenosis (CAS). The present study wished to investigate if metabolites may also act as biomarkers for CAS. METHODS Consecutive patients with at least one carotid artery stenosis > = 60% on cerebral angiography were prospectively recruited from May 2007 to January 2016. Normal controls were recruited from outpatient clinic who had no stroke and coronary artery disease (CAD) history, and the brain magnetic resonance or computed tomographic angiography showed bilateral CAS < 30%. Risk factor profile, clinical characteristics, age, and clinical features were recorded. All subjects were male, and none had diabetes. 1H-NMR spectroscopy-based metabolomics analysis was carried out for plasma samples. RESULTS Totally, 130 male subjects were recruited. Age had no significant difference between the controls and CAS group (60.2 ± 5.9 vs. 63.3 ± 6.0, p = 0.050). The CAS group had significantly higher frequency of CAD, hypertension, smoking and alcohol but lower body mass index than the controls (p < 0.05). The laboratory tests showed CAS group had significantly higher level of homocysteine but lower levels of cholesterol, high-density lipoprotein and hemoglobin than the controls (p < 0.05). The 1H-NMR based plasma metabolomics analysis indicated that choline was significantly lower in CAS patients. The VIP values of lipids were greater than 1.0, which were considered significantly different. CONCLUSIONS Our results suggest homocysteine, choline and lipids in association with traditional risk factors may be involved in the pathogenesis of CAS. Diet adjustment to control homocysteine, choline and lipids may be helpful for the prevention of CAS.
Collapse
Affiliation(s)
- Tsong-Hai Lee
- Stroke Center and Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Mei-Ling Cheng
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Shi Shiao
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Ni Lin
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
30
|
Chen M, Gao C, Yu J, Ren S, Wang M, Wynn RM, Chuang DT, Wang Y, Sun H. Therapeutic Effect of Targeting Branched-Chain Amino Acid Catabolic Flux in Pressure-Overload Induced Heart Failure. J Am Heart Assoc 2019; 8:e011625. [PMID: 31433721 PMCID: PMC6585363 DOI: 10.1161/jaha.118.011625] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/18/2019] [Indexed: 12/14/2022]
Abstract
Background Branched-chain amino acid (BCAA) catabolic defect is an emerging metabolic hallmark in failing hearts in human and animal models. The therapeutic impact of targeting BCAA catabolic flux under pathological conditions remains understudied. Methods and Results BT2 (3,6-dichlorobenzo[b]thiophene-2-carboxylic acid), a small-molecule inhibitor of branched-chain ketoacid dehydrogenase kinase, was used to enhance BCAA catabolism. After 2 weeks of transaortic constriction, mice with significant cardiac dysfunctions were treated with vehicle or BT2. Serial echocardiograms showed continuing pathological deterioration in left ventricle of the vehicle-treated mice, whereas the BT2-treated mice showed significantly preserved cardiac function and structure. Moreover, BT2 treatment improved systolic contractility and diastolic mechanics. These therapeutic benefits appeared to be independent of impacts on left ventricle hypertrophy but associated with increased gene expression involved in fatty acid utilization. The BT2 administration showed no signs of apparent toxicity. Conclusions Our data provide the first proof-of-concept evidence for the therapeutic efficacy of restoring BCAA catabolic flux in hearts with preexisting dysfunctions. The BCAA catabolic pathway represents a novel and potentially efficacious target for treatment of heart failure.
Collapse
Affiliation(s)
- Mengping Chen
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of EducationDepartment of PathophysiologyShanghai Jiao Tong University School of MedicineShanghaiChina
- Departments of Anesthesiology, Medicine and PhysiologyDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA
| | - Chen Gao
- Departments of Anesthesiology, Medicine and PhysiologyDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA
| | - Jiayu Yu
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of EducationDepartment of PathophysiologyShanghai Jiao Tong University School of MedicineShanghaiChina
- Departments of Anesthesiology, Medicine and PhysiologyDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA
| | - Shuxun Ren
- Departments of Anesthesiology, Medicine and PhysiologyDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA
| | - Menglong Wang
- Department of CardiologyRenmin Hospital of Wuhan UniversityCardiovascular Research InstituteWuhan UniversityHubei Key Laboratory of CardiologyWuhanChina
| | - R. Max Wynn
- Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTX
| | - David T. Chuang
- Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTX
| | - Yibin Wang
- Departments of Anesthesiology, Medicine and PhysiologyDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA
| | - Haipeng Sun
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of EducationDepartment of PathophysiologyShanghai Jiao Tong University School of MedicineShanghaiChina
- Departments of Anesthesiology, Medicine and PhysiologyDavid Geffen School of Medicine at University of CaliforniaLos AngelesCA
| |
Collapse
|
31
|
Barba I, Andrés M, Garcia-Dorado D. Metabolomics and Heart Diseases: From Basic to Clinical Approach. Curr Med Chem 2019; 26:46-59. [PMID: 28990507 DOI: 10.2174/0929867324666171006151408] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 03/15/2017] [Accepted: 04/03/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND The field of metabolomics has been steadily increasing in size for the last 15 years. Advances in analytical and statistical methods have allowed metabolomics to flourish in various areas of medicine. Cardiovascular diseases are some of the main research targets in metabolomics, due to their social and medical relevance, and also to the important role metabolic alterations play in their pathogenesis and evolution. Metabolomics has been applied to the full spectrum of cardiovascular diseases: from patient risk stratification to myocardial infarction and heart failure. However - despite the many proof-ofconcept studies describing the applicability of metabolomics in the diagnosis, prognosis and treatment evaluation in cardiovascular diseases - it is not yet used in routine clinical practice. Recently, large phenome centers have been established in clinical environments, and it is expected that they will provide definitive proof of the applicability of metabolomics in clinical practice. But there is also room for small and medium size centers to work on uncommon pathologies or to resolve specific but relevant clinical questions. OBJECTIVES In this review, we will introduce metabolomics, cover the metabolomic work done so far in the area of cardiovascular diseases. CONCLUSION The cardiovascular field has been at the forefront of metabolomics application and it should lead the transfer to the clinic in the not so distant future.
Collapse
Affiliation(s)
- Ignasi Barba
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centro de Investigacion Biomedica en Red sobre Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - Mireia Andrés
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - David Garcia-Dorado
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall d'Hebron University Hospital and Research Institute, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centro de Investigacion Biomedica en Red sobre Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| |
Collapse
|
32
|
Misra BB, Puppala SR, Comuzzie AG, Mahaney MC, VandeBerg JL, Olivier M, Cox LA. Analysis of serum changes in response to a high fat high cholesterol diet challenge reveals metabolic biomarkers of atherosclerosis. PLoS One 2019; 14:e0214487. [PMID: 30951537 PMCID: PMC6450610 DOI: 10.1371/journal.pone.0214487] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/13/2019] [Indexed: 01/19/2023] Open
Abstract
Atherosclerotic plaques are characterized by an accumulation of macrophages, lipids, smooth muscle cells, and fibroblasts, and, in advanced stages, necrotic debris within the arterial walls. Dietary habits such as high fat and high cholesterol (HFHC) consumption are known risk factors for atherosclerosis. However, the key metabolic contributors to diet-induced atherosclerosis are far from established. Herein, we investigate the role of a 2-year HFHC diet challenge in the metabolic changes of development and progression of atherosclerosis. We used a non-human primate (NHP) model (baboons, n = 60) fed a HFHC diet for two years and compared metabolomic profiles in serum from animals on baseline chow with serum collected after the challenge diet using two-dimensional gas chromatography time-of-flight mass-spectrometry (2D GC-ToF-MS) for untargeted metabolomic analysis, to quantify metabolites that contribute to atherosclerotic lesion formation. Further, clinical biomarkers associated with atherosclerosis, lipoprotein measures, fat indices, and arterial plaque formation (lesions) were quantified. Using two chemical derivatization (i.e., silylation) approaches, we quantified 321 metabolites belonging to 66 different metabolic pathways, which revealed significantly different metabolic profiles of HFHC diet and chow diet fed baboon sera. We found heritability of two important metabolites, lactic acid and asparagine, in the context of diet-induced metabolic changes. In addition, abundance of cholesterol, lactic acid, and asparagine were sex-dependent. Finally, 35 metabolites correlated (R2, 0.068-0.271, P < 0.05) with total lesion burden assessed in three arteries (aortic arch, common iliac artery, and descending aorta) which could serve as potential biomarkers pending further validation. This study demonstrates the feasibility of detecting sex-specific and heritable metabolites in NHPs with diet-induced atherosclerosis using untargeted metabolomics allowing understanding of atherosclerotic disease progression in humans.
Collapse
Affiliation(s)
- Biswapriya B. Misra
- Center for Precision Medicine, Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina United States of America
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Sobha R. Puppala
- Center for Precision Medicine, Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina United States of America
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | | | - Michael C. Mahaney
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, The University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas, United States of America
| | - John L. VandeBerg
- South Texas Diabetes and Obesity Institute and Department of Human Genetics, The University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas, United States of America
| | - Michael Olivier
- Center for Precision Medicine, Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina United States of America
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Laura A. Cox
- Center for Precision Medicine, Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina United States of America
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| |
Collapse
|
33
|
Abstract
Branched chain amino acids (BCAAs) are building blocks for all life-forms. We review here the fundamentals of BCAA metabolism in mammalian physiology. Decades of studies have elicited a deep understanding of biochemical reactions involved in BCAA catabolism. In addition, BCAAs and various catabolic products act as signaling molecules, activating programs ranging from protein synthesis to insulin secretion. How these processes are integrated at an organismal level is less clear. Inborn errors of metabolism highlight the importance of organismal regulation of BCAA physiology. More recently, subtle alterations of BCAA metabolism have been suggested to contribute to numerous prevalent diseases, including diabetes, cancer, and heart failure. Understanding the mechanisms underlying altered BCAA metabolism and how they contribute to disease pathophysiology will keep researchers busy for the foreseeable future.
Collapse
Affiliation(s)
- Michael Neinast
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| | - Danielle Murashige
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| | - Zoltan Arany
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|
34
|
Limkakeng AT, Henao R, Voora D, O’Connell T, Griffin M, Tsalik EL, Shah S, Woods CW, Ginsburg GS. Pilot study of myocardial ischemia-induced metabolomic changes in emergency department patients undergoing stress testing. PLoS One 2019; 14:e0211762. [PMID: 30707740 PMCID: PMC6358091 DOI: 10.1371/journal.pone.0211762] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/21/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The heart is a metabolically active organ, and plasma acylcarnitines are associated with long-term risk for myocardial infarction. We hypothesized that myocardial ischemia from cardiac stress testing will produce dynamic changes in acylcarnitine and amino acid levels compared to levels seen in matched control patients with normal stress tests. METHODS We analyzed targeted metabolomic profiles in a pilot study of 20 case patients with inducible ischemia on stress testing from an existing prospectively collected repository of 357 consecutive patients presenting with symptoms of Acute Coronary Syndrome (ACS) in an Emergency Department (ED) observation unit between November 2012 and September 2014. We selected 20 controls matched on age, sex, and body-mass index (BMI). A peripheral blood sample was drawn <1 hour before stress testing and 2 hours after stress testing on each patient. We assayed 60 select acylcarnitines and amino acids by tandem mass spectrometry (MS/MS) using a Quattro Micro instrument (Waters Corporation, Milford, MA). Metabolite values were log transformed for skew. We then performed bivariable analysis for stress test outcome and both individual timepoint metabolite concentrations and stress-delta metabolite ratios (T2/T0). False discovery rates (FDR) were calculated for 60 metabolites while controlling for age, sex, and BMI. We built multivariable regularized linear models to predict stress test outcome from metabolomics data at times 0, 2 hours, and log ratio between these two. We used leave-one-out cross-validation to estimate the performance characteristics of the model. RESULTS Nine of our 20 case subjects were male. Cases' average age was 55.8, with an average BMI 29.5. Bivariable analysis identified 5 metabolites associated with positive stress tests (FDR < 0.2): alanine, C14:1-OH, C16:1, C18:2, C20:4. The multivariable regularized linear models built on T0 and T2 had Area Under the ROC Curve (AUC-ROC) between 0.5 and 0.55, however, the log(T2/T0) model yielded 0.625 AUC, with 65% sensitivity and 60% specificity. The top metabolites selected by the model were: Ala, Arg, C12-OH/C10-DC, C14:1-OH, C16:1, C18:2, C18:1, C20:4 and C18:1-DC. CONCLUSIONS Stress-delta metabolite analysis of patients undergoing stress testing is feasible. Future studies with a larger sample size are warranted.
Collapse
Affiliation(s)
- Alexander T. Limkakeng
- Division of Emergency Medicine, Department of Surgery, Duke University, Durham, North Carolina, United States of America
| | - Ricardo Henao
- Center for Applied Genomics & Precision Medicine, Duke University, Durham, North Carolina, United States of America
| | - Deepak Voora
- Center for Applied Genomics & Precision Medicine, Duke University, Durham, North Carolina, United States of America
- Division of Cardiology, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Thomas O’Connell
- Indiana University, Indianapolis, Indiana, United States of America
| | - Michelle Griffin
- Division of Emergency Medicine, Department of Surgery, Duke University, Durham, North Carolina, United States of America
| | - Ephraim L. Tsalik
- Center for Applied Genomics & Precision Medicine, Duke University, Durham, North Carolina, United States of America
- Emergency Medicine Service, Durham Veteran’s Affairs Medical Center, Durham, North Carolina, United States of America
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Svati Shah
- Center for Applied Genomics & Precision Medicine, Duke University, Durham, North Carolina, United States of America
- Division of Cardiology, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Christopher W. Woods
- Center for Applied Genomics & Precision Medicine, Duke University, Durham, North Carolina, United States of America
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Geoffrey S. Ginsburg
- Center for Applied Genomics & Precision Medicine, Duke University, Durham, North Carolina, United States of America
- Division of Cardiology, Department of Medicine, Duke University, Durham, North Carolina, United States of America
| |
Collapse
|
35
|
Lee CC, Hsieh YJ, Chen SW, Fu SH, Hsu CW, Wu CC, Han W, Li Y, Huan T, Chang YS, Yu JS, Li L, Chang CH, Chen YT. Bretschneider solution-induced alterations in the urine metabolome in cardiac surgery patients. Sci Rep 2018; 8:17774. [PMID: 30538262 PMCID: PMC6290005 DOI: 10.1038/s41598-018-35631-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/01/2018] [Indexed: 01/01/2023] Open
Abstract
The development of Bretschneider’s histidine-tryptophan-ketoglutarate (HTK) cardioplegia solution represented a major advancement in cardiac surgery, offering significant myocardial protection. However, metabolic changes induced by this additive in the whole body have not been systematically investigated. Using an untargeted mass spectrometry-based method to deeply explore the urine metabolome, we sought to provide a holistic and systematic view of metabolic perturbations occurred in patients receiving HTK. Prospective urine samples were collected from 100 patients who had undergone cardiac surgery, and metabolomic changes were profiled using a high-performance chemical isotope labeling liquid chromatography-mass spectrometry (LC-MS) method. A total of 14,642 peak pairs or metabolites were quantified using differential 13C-/12C-dansyl labeling LC-MS, which targets the amine/phenol submetabolome from urine specimens. We identified 223 metabolites that showed significant concentration change (fold change > 5) and assembled several potential metabolic pathway maps derived from these dysregulated metabolites. Our data indicated upregulated histidine metabolism with subsequently increased glutamine/glutamate metabolism, altered purine and pyrimidine metabolism, and enhanced vitamin B6 metabolism in patients receiving HTK. Our findings provide solid evidence that HTK solution causes significant perturbations in several metabolic pathways and establish a basis for further study of key mechanisms underlying its organ-protective or potential harmful effects.
Collapse
Affiliation(s)
- Cheng-Chia Lee
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Linkou branch, College of Medicine, Chang Gung University, Guishan, Taoyuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, College of medicine, Chang Gung University, Guishan, Taoyuan, Taiwan
| | - Ya-Ju Hsieh
- Molecular and Medicine Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan
| | - Shao-Wei Chen
- Graduate Institute of Clinical Medical Sciences, College of medicine, Chang Gung University, Guishan, Taoyuan, Taiwan.,Department of cardiothoracic and vascular surgery, Chang Gung Memorial Hospital, Linkou branch, College of Medicine, Chang Gung University, Guishan, Taoyuan, Taiwan
| | - Shu-Hsuan Fu
- Molecular and Medicine Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan
| | - Chia-Wei Hsu
- Molecular and Medicine Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan
| | - Chih-Ching Wu
- Molecular and Medicine Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan.,Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Guishan, Taoyuan, 33302, Taiwan.,Department of Otolaryngology-Head & Neck Surgery, Chang Gung Memorial Hospital at Linkou, Guishan, Taoyuan, 33305, Taiwan
| | - Wei Han
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G2G2, Canada
| | - Yunong Li
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G2G2, Canada
| | - Tao Huan
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G2G2, Canada
| | - Yu-Sun Chang
- Molecular and Medicine Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan.,Department of Otolaryngology-Head & Neck Surgery, Chang Gung Memorial Hospital at Linkou, Guishan, Taoyuan, 33305, Taiwan.,Graduate Institute of Biomedical Sciences, Chang Gung University, Guishan, Taoyuan, 33302, Taiwan
| | - Jau-Song Yu
- Molecular and Medicine Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan.,Liver Research Center, Chang Gung Memorial Hospital at Linkou, Guishan, Taoyuan, 33305, Taiwan.,Department of Cell and Molecular Biology, Chang Gung University, Guishan, Taoyuan, 33302, Taiwan
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G2G2, Canada.
| | - Chih-Hsiang Chang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Linkou branch, College of Medicine, Chang Gung University, Guishan, Taoyuan, Taiwan. .,Graduate Institute of Clinical Medical Sciences, College of medicine, Chang Gung University, Guishan, Taoyuan, Taiwan.
| | - Yi-Ting Chen
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Linkou branch, College of Medicine, Chang Gung University, Guishan, Taoyuan, Taiwan. .,Molecular and Medicine Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan. .,Department of Biomedical Sciences, College of Medicine, Chang Gung University, Guishan, Taoyuan, Taiwan. .,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Guishan, Taoyuan, Taiwan.
| |
Collapse
|
36
|
Evaluation of preoperative oral carbohydrate administration on insulin resistance in off-pump coronary artery bypass patients: A randomised trial. Eur J Anaesthesiol 2018; 34:740-747. [PMID: 28437263 DOI: 10.1097/eja.0000000000000637] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND In fasting cardiac surgery patients, preoperative carbohydrate (CHO) drink intake attenuated insulin resistance and improved cardiac metabolism, although its beneficial effects were not evident after cardiac surgery possibly due to cardiopulmonary bypass-related extreme systemic inflammation. OBJECTIVE We aimed to evaluate whether preoperative CHO intake affected insulin resistance and free-fatty acid (FFA) concentrations in off-pump coronary revascularisation. DESIGN A randomised controlled trial. SETTING Primary care in a university hospital in Korea from January 2015 to July 2016. PATIENTS Sixty patients who underwent elective multi-vessel off-pump coronary revascularisation were randomised into two groups. Three patients were excluded from analysis and 57 patients completed study. INTERVENTION The CHO group received oral CHO (400 ml) the prior evening and 2 to 3 h before surgery, and the control group was fasted from food and water according to standard protocol. MAIN OUTCOME MEASURES Insulin resistance was assessed twice, after anaesthetic induction and after surgery via short insulin tolerance test. FFA, C-reactive protein and creatine kinase-myocardial band concentrations were determined serially for 48 h after surgery. RESULTS Insulin sensitivity was greater (P = 0.002) and plasma FFA concentrations were lower (P = 0.001) after anaesthetic induction in the CHO group compared with the Control group, although there were no intergroup differences after surgery. The postoperative peak creatine kinase-myocardial band concentration was significantly lower in the CHO group compared with the Control group [8.8 (5.4 to 18.2) vs. 6.4 (3.5 to 9.7) ng ml, P = 0.031]. CONCLUSION A preoperative CHO supplement significantly reduced insulin resistance and FFA concentrations compared with fasting at the beginning of the surgery, but these benefits were lost after off-pump coronary revascularisation. Despite their transient nature, these beneficial effects resulted in less myocardial injury, mandating further studies focused on the impact of preoperative CHO on myocardial ischaemia and cardiac function after coronary revascularisation. TRIAL REGISTRATION Clinicaltrials.gov identifier: NCT 02330263.
Collapse
|
37
|
Targeted Metabolomic Profiling of Plasma and Survival in Heart Failure Patients. JACC-HEART FAILURE 2018; 5:823-832. [PMID: 29096792 DOI: 10.1016/j.jchf.2017.07.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/24/2017] [Accepted: 07/26/2017] [Indexed: 01/13/2023]
Abstract
OBJECTIVES This study sought to derive and validate plasma metabolite associations with survival in heart failure (HF) patients. BACKGROUND Profiling of plasma metabolites to predict the course of HF appears promising, but validation and incremental value of these profiles are less established. METHODS Patients (n = 1,032) who met Framingham HF criteria with a history of reduced ejection fraction were randomly divided into derivation and validation cohorts (n = 516 each). Amino acids, organic acids, and acylcarnitines were quantified using mass spectrometry in fasting plasma samples. We derived a prognostic metabolite profile (PMP) in the derivation cohort using Lasso-penalized Cox regression. Validity was assessed by 10-fold cross validation in the derivation cohort and by standard testing in the validation cohort. The PMP was analyzed as both a continuous variable (PMPscore) and dichotomized at the median (PMPcat), in univariate and multivariate models adjusted for clinical risk score and N-terminal pro-B-type natriuretic peptide. RESULTS Overall, 48% of patients were African American, 35% were women, and the average age was 69 years. After a median follow-up of 34 months, there were 256 deaths (127 and 129 in derivation and validation cohorts, respectively). Optimized modeling defined the 13 metabolite PMPs, which was cross validated as both the PMPscore (hazard ratio [HR]: 3.27; p < 2 × 10-16) and PMPcat (HR: 3.04; p = 2.93 × 10-8). The validation cohort showed similar results (PMPscore HR: 3.9; p < 2 × 10-16 and PMPcat HR: 3.99; p = 3.47 × 10-9). In adjusted models, PMP remained associated with mortality in the cross-validated derivation cohort (PMPscore HR: 1.63; p = 0.0029; PMPcat HR: 1.47; p = 0.081) and the validation cohort (PMPscore HR: 1.54; p = 0.037; PMPcat HR: 1.69; p = 0.043). CONCLUSIONS Plasma metabolite profiles varied across HF subgroups and were associated with survival incremental to conventional predictors. Additional investigation is warranted to define mechanisms and clinical applications.
Collapse
|
38
|
Dissecting Clinical and Metabolomics Associations of Left Atrial Phasic Function by Cardiac Magnetic Resonance Feature Tracking. Sci Rep 2018; 8:8138. [PMID: 29802321 PMCID: PMC5970174 DOI: 10.1038/s41598-018-26456-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 05/10/2018] [Indexed: 01/09/2023] Open
Abstract
Among community cohorts, associations between clinical and metabolite factors and complex left atrial (LA) phasic function assessed by cardiac magnetic resonance (CMR) feature tracking (FT) are unknown. Longitudinal LA strain comprising reservoir strain (εs), conduit strain (εe) and booster strain (εa) and their corresponding peak strain rates (SRs, SRe, SRa) were measured using CMR FT. Targeted mass spectrometry measured 83 circulating metabolites in serum. Sparse Principal Component Analysis was used for data reduction. Among community adults (n = 128, 41% female) (mean age: 70.5 ± 11.6 years), age was significantly associated with εs (β = -0.30, p < 0.0001), εe (β = -0.3, p < 0.0001), SRs (β = -0.02, p < 0.0001), SRe (β = 0.04, p < 0.0001) and SRe/SRa (β = -0.01, p = 0.012). In contrast, heart rate was significantly associated with εa (β = 0.1, p = 0.001) and SRa (β = -0.02, p < 0.0001). Serine was significantly associated with εs (β = 10.1, p = 0.015), SRs (β = 0.5, p = 0.033) and SRa (β = -0.9, p = 0.016). Citrulline was associated with εs (β = -4.0, p = 0.016), εa (β = -3.4, p = 0.002) and SRa (β = 0.4, p = 0.019). Valine was associated with ratio of SRe:SRa (β = -0.4, p = 0.039). Medium and long chain dicarboxyl carnitines were associated with εs (β = -0.6, p = 0.038). Phases of LA function were differentially associated with clinical and metabolite factors. Metabolite signals may be used to advance mechanistic understanding of LA disease in future studies.
Collapse
|
39
|
Ruoppolo M, Caterino M, Albano L, Pecce R, Di Girolamo MG, Crisci D, Costanzo M, Milella L, Franconi F, Campesi I. Targeted metabolomic profiling in rat tissues reveals sex differences. Sci Rep 2018; 8:4663. [PMID: 29549307 PMCID: PMC5856765 DOI: 10.1038/s41598-018-22869-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 03/02/2018] [Indexed: 12/15/2022] Open
Abstract
Sex differences affect several diseases and are organ-and parameter-specific. In humans and animals, sex differences also influence the metabolism and homeostasis of amino acids and fatty acids, which are linked to the onset of diseases. Thus, the use of targeted metabolite profiles in tissues represents a powerful approach to examine the intermediary metabolism and evidence for any sex differences. To clarify the sex-specific activities of liver, heart and kidney tissues, we used targeted metabolomics, linear discriminant analysis (LDA), principal component analysis (PCA), cluster analysis and linear correlation models to evaluate sex and organ-specific differences in amino acids, free carnitine and acylcarnitine levels in male and female Sprague-Dawley rats. Several intra-sex differences affect tissues, indicating that metabolite profiles in rat hearts, livers and kidneys are organ-dependent. Amino acids and carnitine levels in rat hearts, livers and kidneys are affected by sex: male and female hearts show the greatest sexual dimorphism, both qualitatively and quantitatively. Finally, multivariate analysis confirmed the influence of sex on the metabolomics profiling. Our data demonstrate that the metabolomics approach together with a multivariate approach can capture the dynamics of physiological and pathological states, which are essential for explaining the basis of the sex differences observed in physiological and pathological conditions.
Collapse
Affiliation(s)
- Margherita Ruoppolo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli, "Federico II", Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy.,Associazione Culturale DiSciMuS, RFC 80026, Casoria, Napoli, Italy
| | - Marianna Caterino
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli, "Federico II", Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy.,Associazione Culturale DiSciMuS, RFC 80026, Casoria, Napoli, Italy
| | - Lucia Albano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli, "Federico II", Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Rita Pecce
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli, "Federico II", Napoli, Italy
| | - Maria Grazia Di Girolamo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli, "Federico II", Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Daniela Crisci
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli, "Federico II", Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Michele Costanzo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli, "Federico II", Napoli, Italy
| | - Luigi Milella
- Dipartimento di Scienze, Università degli Studi della Basilicata, Potenza, Italy
| | - Flavia Franconi
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy.,Assessorato alle Politiche della Persone della Regione Basilicata, Potenza, Italy
| | - Ilaria Campesi
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy.
| |
Collapse
|
40
|
Hausenloy DJ, Garcia-Dorado D, Bøtker HE, Davidson SM, Downey J, Engel FB, Jennings R, Lecour S, Leor J, Madonna R, Ovize M, Perrino C, Prunier F, Schulz R, Sluijter JPG, Van Laake LW, Vinten-Johansen J, Yellon DM, Ytrehus K, Heusch G, Ferdinandy P. Novel targets and future strategies for acute cardioprotection: Position Paper of the European Society of Cardiology Working Group on Cellular Biology of the Heart. Cardiovasc Res 2018; 113:564-585. [PMID: 28453734 DOI: 10.1093/cvr/cvx049] [Citation(s) in RCA: 243] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 03/15/2017] [Indexed: 02/06/2023] Open
Abstract
Ischaemic heart disease and the heart failure that often results, remain the leading causes of death and disability in Europe and worldwide. As such, in order to prevent heart failure and improve clinical outcomes in patients presenting with an acute ST-segment elevation myocardial infarction and patients undergoing coronary artery bypass graft surgery, novel therapies are required to protect the heart against the detrimental effects of acute ischaemia/reperfusion injury (IRI). During the last three decades, a wide variety of ischaemic conditioning strategies and pharmacological treatments have been tested in the clinic-however, their translation from experimental to clinical studies for improving patient outcomes has been both challenging and disappointing. Therefore, in this Position Paper of the European Society of Cardiology Working Group on Cellular Biology of the Heart, we critically analyse the current state of ischaemic conditioning in both the experimental and clinical settings, provide recommendations for improving its translation into the clinical setting, and highlight novel therapeutic targets and new treatment strategies for reducing acute myocardial IRI.
Collapse
Affiliation(s)
- Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK; The National Institute of Health Research University College London Hospitals Biomedical Research Centre, 149 Tottenham Court Road London, W1T 7DN, UK; Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore 169857; National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Dr, Singapore 169609, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore; Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - David Garcia-Dorado
- Department of Cardiology, Vall d Hebron University Hospital and Research Institute. Universitat Autònoma, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital Skejby, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - James Downey
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, 5851 USA Dr. N., MSB 3074, Mobile, AL 36688, USA
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nßrnberg, Schloßplatz 4, 91054 Erlangen, Germany
| | - Robert Jennings
- Department of Cardiology, Duke University, Durham, NC 27708, USA
| | - Sandrine Lecour
- Department of Medicine, Hatter Institute for Cardiovascular Research in Africa and South African Medical Research Council Inter-University Cape Heart Group, Faculty of Health Sciences, University of Cape Town, Chris Barnard Building, Anzio Road, Observatory, 7925, Cape Town, Western Cape, South Africa
| | - Jonathan Leor
- Tamman Cardiovascular Research Institute, Sheba Medical Center, Tel Hashomer, Israel; Neufeld Cardiac Research Institute, Tel-Aviv University, Sheba Medical Center, Tel Hashomer, 5265601, Israel; Sheba Center for Regenerative Medicine, Stem Cell, and Tissue Engineering, Tel Hashomer, 5265601, Israel
| | - Rosalinda Madonna
- Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy; Institute of Cardiology, Department of Neurosciences, Imaging, and Clinical Sciences, "G. d'Annunzio University, Chieti, Italy; Texas Heart Institute and University of Texas Medical School in Houston, Department of Internal Medicine, 6770 Bertner Avenue, Houston, Texas 77030 USA
| | - Michel Ovize
- Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, 28 Avenue du Doyen Jean Lépine, 69500 Bron, France; UMR 1060 (CarMeN), Université Claude Bernard Lyon, 43 Boulevard du 11 Novembre 1918, 69100 Villeurbanne, France
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Division of Cardiology, Federico II University Corso Umberto I, 40, 80138 Napoli, Italy
| | - Fabrice Prunier
- Department of Cardiology, University of Angers, University Hospital of Angers, 4 Rue Larrey, 49100 Angers, France
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig, University of Giessen, Ludwigstraße 23, 35390 Gießen, Germany
| | - Joost P G Sluijter
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| | - Linda W Van Laake
- Division Heart and Lungs, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| | - Jakob Vinten-Johansen
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University, 201 Dowman Dr, Atlanta, GA 30322, USA
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK; The National Institute of Health Research University College London Hospitals Biomedical Research Centre, 149 Tottenham Court Road London, W1T 7DN, UK
| | - Kirsti Ytrehus
- Cardiovascular Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Hansine Hansens veg 18, 9019 Tromsø, Norway
| | - Gerd Heusch
- Institute for Pathophysiology, West-German Heart and Vascular Center, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Nagyvárad tér 4, 1089 Hungary; Pharmahungary Group, Graphisoft Park, 7 Záhony street, Budapest, H-1031, Hungary
| |
Collapse
|
41
|
Hunter WG, Kelly JP, McGarrah RW, Kraus WE, Shah SH. Metabolic Dysfunction in Heart Failure: Diagnostic, Prognostic, and Pathophysiologic Insights From Metabolomic Profiling. Curr Heart Fail Rep 2017; 13:119-31. [PMID: 27216948 DOI: 10.1007/s11897-016-0289-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Metabolic impairment is an intrinsic component of heart failure (HF) pathophysiology. Although initially conceived as a myocardial defect, metabolic dysfunction is now recognized as a systemic process with complex interplay between the myocardium and peripheral tissues and organs. Specifically, HF-associated metabolic dysfunction includes alterations in substrate utilization, insulin resistance, defects in energy production, and imbalanced anabolic-catabolic signaling leading to cachexia. Each of these metabolic abnormalities is associated with significant morbidity and mortality in patients with HF; however, their detection and therapeutic management remains challenging. Given the difficulty in obtaining human cardiac tissue for research purposes, peripheral blood metabolomic profiling, a well-established approach for characterizing small-molecule metabolite intermediates from canonical biochemical pathways, may be a useful technology for dissecting biomarkers and mechanisms of metabolic impairment in HF. In this review, metabolic abnormalities in HF will be discussed with particular emphasis on the application of metabolomic profiling to detecting, risk stratifying, and identifying novel targets for metabolic therapy in this heterogeneous population.
Collapse
Affiliation(s)
- Wynn G Hunter
- Duke University School of Medicine, 300 North Duke Street, Durham, NC, 27701, USA
| | - Jacob P Kelly
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Robert W McGarrah
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
| | - William E Kraus
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
| | - Svati H Shah
- Duke University School of Medicine, 300 North Duke Street, Durham, NC, 27701, USA.
- Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Duke Clinical Research Institute, Durham, NC, USA.
- Duke Molecular Physiology Institute, Durham, NC, USA.
| |
Collapse
|
42
|
Tsai CL, Tsai MS, Kuo CH, Chou TH, Chen WJ, Huang CH. Metabolomic profiling for outcome prediction in emergency department patients with out-of-hospital cardiac arrest. Resuscitation 2017; 123:e1-e2. [PMID: 29253647 DOI: 10.1016/j.resuscitation.2017.12.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/09/2017] [Accepted: 12/14/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Chu-Lin Tsai
- Department of Emergency Medicine (CLT, MST, THC, WJC, CHH), National Taiwan University Hospital, and School of Pharmacy (CHK), College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Min-Shan Tsai
- Department of Emergency Medicine (CLT, MST, THC, WJC, CHH), National Taiwan University Hospital, and School of Pharmacy (CHK), College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ching-Hwa Kuo
- Department of Emergency Medicine (CLT, MST, THC, WJC, CHH), National Taiwan University Hospital, and School of Pharmacy (CHK), College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Tzung-Hsin Chou
- Department of Emergency Medicine (CLT, MST, THC, WJC, CHH), National Taiwan University Hospital, and School of Pharmacy (CHK), College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Wen-Jone Chen
- Department of Emergency Medicine (CLT, MST, THC, WJC, CHH), National Taiwan University Hospital, and School of Pharmacy (CHK), College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chien-Hua Huang
- Department of Emergency Medicine (CLT, MST, THC, WJC, CHH), National Taiwan University Hospital, and School of Pharmacy (CHK), College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| |
Collapse
|
43
|
Angelini A, Pi X, Xie L. Dioxygen and Metabolism; Dangerous Liaisons in Cardiac Function and Disease. Front Physiol 2017; 8:1044. [PMID: 29311974 PMCID: PMC5732914 DOI: 10.3389/fphys.2017.01044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/29/2017] [Indexed: 12/19/2022] Open
Abstract
The heart must consume a significant amount of energy to sustain its contractile activity. Although the fuel demands are huge, the stock remains very low. Thus, in order to supply its daily needs, the heart must have amazing adaptive abilities, which are dependent on dioxygen availability. However, in myriad cardiovascular diseases, “fuel” depletion and hypoxia are common features, leading cardiomyocytes to favor low-dioxygen-consuming glycolysis rather than oxidation of fatty acids. This metabolic switch makes it challenging to distinguish causes from consequences in cardiac pathologies. Finally, despite the progress achieved in the past few decades, medical treatments have not improved substantially, either. In such a situation, it seems clear that much remains to be learned about cardiac diseases. Therefore, in this review, we will discuss how reconciling dioxygen availability and cardiac metabolic adaptations may contribute to develop full and innovative strategies from bench to bedside.
Collapse
Affiliation(s)
- Aude Angelini
- Department of Medicine-Athero and Lipo, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - Xinchun Pi
- Department of Medicine-Athero and Lipo, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - Liang Xie
- Department of Medicine-Athero and Lipo, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
44
|
Andersen LW. Lactate Elevation During and After Major Cardiac Surgery in Adults: A Review of Etiology, Prognostic Value, and Management. Anesth Analg 2017; 125:743-752. [PMID: 28277327 DOI: 10.1213/ane.0000000000001928] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Elevated lactate is a common occurrence after cardiac surgery. This review summarizes the literature on the complex etiology of lactate elevation during and after cardiac surgery, including considerations of oxygen delivery, oxygen utilization, increased metabolism, lactate clearance, medications and fluids, and postoperative complications. Second, the association between lactate and a variety of outcomes are described, and the prognostic role of lactate is critically assessed. Despite the fact that elevated lactate is strongly associated with many important outcomes, including postoperative complications, length of stay, and mortality, little is known about the optimal management of postoperative patients with lactate elevations. This review ends with an assessment of the limited literature on this subject.
Collapse
Affiliation(s)
- Lars W Andersen
- From the *Research Center for Emergency Medicine, Aarhus University Hospital, Aarhus, Denmark; †Center for Resuscitation Science, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; ‡Department of Anesthesiology, Aarhus University Hospital, Aarhus, Denmark; and §Department of Medicine, Regional Hospital Holstebro, Aarhus University, Holstebro, Denmark
| |
Collapse
|
45
|
Ruiz M, Labarthe F, Fortier A, Bouchard B, Thompson Legault J, Bolduc V, Rigal O, Chen J, Ducharme A, Crawford PA, Tardif JC, Des Rosiers C. Circulating acylcarnitine profile in human heart failure: a surrogate of fatty acid metabolic dysregulation in mitochondria and beyond. Am J Physiol Heart Circ Physiol 2017; 313:H768-H781. [PMID: 28710072 DOI: 10.1152/ajpheart.00820.2016] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 07/07/2017] [Accepted: 07/07/2017] [Indexed: 12/19/2022]
Abstract
Heart failure (HF) is associated with metabolic perturbations, particularly of fatty acids (FAs), which remain to be better understood in humans. This study aimed at testing the hypothesis that HF patients with reduced ejection fraction display systemic perturbations in levels of energy-related metabolites, especially those reflecting dysregulation of FA metabolism, namely, acylcarnitines (ACs). Circulating metabolites were assessed using mass spectrometry (MS)-based methods in two cohorts. The main cohort consisted of 72 control subjects and 68 HF patients exhibiting depressed left ventricular ejection fraction (25.9 ± 6.9%) and mostly of ischemic etiology with ≥2 comorbidities. HF patients displayed marginal changes in plasma levels of tricarboxylic acid cycle-related metabolites or indexes of mitochondrial or cytosolic redox status. They had, however, 22-79% higher circulating ACs, irrespective of chain length (P < 0.0001, adjusted for sex, age, renal function, and insulin resistance, determined by shotgun MS/MS), which reflects defective mitochondrial β-oxidation, and were significantly associated with levels of NH2-terminal pro-B-type natriuretic peptide levels, a disease severity marker. Subsequent extended liquid chromatography-tandem MS analysis of 53 plasma ACs in a subset group from the primary cohort confirmed and further substantiated with a comprehensive lipidomic analysis in a validation cohort revealed in HF patients a more complex circulating AC profile. The latter included dicarboxylic-ACs and dihydroxy-ACs as well as very long chain (VLC) ACs or sphingolipids with VLCFAs (>20 carbons), which are proxies of dysregulated FA metabolism in peroxisomes. Our study identified alterations in circulating ACs in HF patients that are independent of biological traits and associated with disease severity markers. These alterations reflect dysfunctional FA metabolism in mitochondria but also beyond, namely, in peroxisomes, suggesting a novel mechanism contributing to global lipid perturbations in human HF.NEW & NOTEWORTHY Mass spectrometry-based profiling of circulating energy metabolites, including acylcarnitines, in two cohorts of heart failure versus control subjects revealed multiple alterations in fatty acid metabolism in peroxisomes in addition to mitochondria, thereby highlighting a novel mechanism contributing to global lipid perturbations in heart failure.Listen to this article's corresponding podcast at http://ajpheart.podbean.com/e/acylcarnitines-in-human-heart-failure/.
Collapse
Affiliation(s)
- Matthieu Ruiz
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada.,Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - François Labarthe
- CHRU de Tours, Université François Rabelais, Institut National de la Santé et de la Recherche Médicale U1069, Nutrition, Croissance et Cancer, Tours, France
| | - Annik Fortier
- Montreal Health Innovations Coordinating Center, Montreal, Quebec, Canada
| | - Bertrand Bouchard
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada.,Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Julie Thompson Legault
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada.,Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Virginie Bolduc
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Odile Rigal
- Laboratoire de Biochimie, Hôpital R. Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jane Chen
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | - Anique Ducharme
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Peter A Crawford
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; and
| | | | - Christine Des Rosiers
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada; .,Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| |
Collapse
|
46
|
The Emerging Role of Metabolomics in the Diagnosis and Prognosis of Cardiovascular Disease. J Am Coll Cardiol 2017; 68:2850-2870. [PMID: 28007146 DOI: 10.1016/j.jacc.2016.09.972] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/09/2016] [Indexed: 12/12/2022]
Abstract
Perturbations in cardiac energy metabolism are major contributors to a number of cardiovascular pathologies. In addition, comorbidities associated with cardiovascular disease (CVD) can alter systemic and myocardial metabolism, often contributing to the worsening of cardiac function and health outcomes. State-of-the-art metabolomic technologies give us the ability to measure thousands of metabolites in biological fluids or biopsies, providing us with a metabolic fingerprint of individual patients. These metabolic profiles may serve as diagnostic and/or prognostic tools that have the potential to significantly alter the management of CVD. Herein, the authors review how metabolomics can assist in the interpretation of perturbed metabolic processes, and how this has improved our ability to understand the pathology of ischemic heart disease, atherosclerosis, and heart failure. Taken together, the integration of metabolomics with other "omics" platforms will allow us to gain insight into pathophysiological interactions of metabolites, proteins, genes, and disease states, while advancing personalized medicine.
Collapse
|
47
|
Stram AR, Wagner GR, Fogler BD, Pride PM, Hirschey MD, Payne RM. Progressive mitochondrial protein lysine acetylation and heart failure in a model of Friedreich's ataxia cardiomyopathy. PLoS One 2017; 12:e0178354. [PMID: 28542596 PMCID: PMC5444842 DOI: 10.1371/journal.pone.0178354] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/11/2017] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION The childhood heart disease of Friedreich's Ataxia (FRDA) is characterized by hypertrophy and failure. It is caused by loss of frataxin (FXN), a mitochondrial protein involved in energy homeostasis. FRDA model hearts have increased mitochondrial protein acetylation and impaired sirtuin 3 (SIRT3) deacetylase activity. Protein acetylation is an important regulator of cardiac metabolism and loss of SIRT3 increases susceptibility of the heart to stress-induced cardiac hypertrophy and ischemic injury. The underlying pathophysiology of heart failure in FRDA is unclear. The purpose of this study was to examine in detail the physiologic and acetylation changes of the heart that occur over time in a model of FRDA heart failure. We predicted that increased mitochondrial protein acetylation would be associated with a decrease in heart function in a model of FRDA. METHODS A conditional mouse model of FRDA cardiomyopathy with ablation of FXN (FXN KO) in the heart was compared to healthy controls at postnatal days 30, 45 and 65. We evaluated hearts using echocardiography, cardiac catheterization, histology, protein acetylation and expression. RESULTS Acetylation was temporally progressive and paralleled evolution of heart failure in the FXN KO model. Increased acetylation preceded detectable abnormalities in cardiac function and progressed rapidly with age in the FXN KO mouse. Acetylation was also associated with cardiac fibrosis, mitochondrial damage, impaired fat metabolism, and diastolic and systolic dysfunction leading to heart failure. There was a strong inverse correlation between level of protein acetylation and heart function. CONCLUSION These results demonstrate a close relationship between mitochondrial protein acetylation, physiologic dysfunction and metabolic disruption in FRDA hypertrophic cardiomyopathy and suggest that abnormal acetylation contributes to the pathophysiology of heart disease in FRDA. Mitochondrial protein acetylation may represent a therapeutic target for early intervention.
Collapse
Affiliation(s)
- Amanda R. Stram
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Gregory R. Wagner
- Division of Endocrinology, Metabolism, and Nutrition, Duke University, Durham, North Carolina, United States of America
| | - Brian D. Fogler
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - P. Melanie Pride
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Matthew D. Hirschey
- Division of Endocrinology, Metabolism, and Nutrition, Duke University, Durham, North Carolina, United States of America
| | - R. Mark Payne
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| |
Collapse
|
48
|
Abstract
Although statistical evidence is clear regarding the dangerousness of unstable angina (UA), a form of coronary heart disease (CHD) characterised by high mortality and morbidity globally, it is important to recognise that diagnostic precision for the condition is unfavourable. In the present research, to gain insight into candidate biomarkers, the author draws on 1H NMR-based serum metabolic profiling to analyze the unstable angina pectoris (UAP) metabolic signatures; this constitutes an effective way to produce medical diagnosis. 101 unstable angina pectoris patients and 132 healthy controls were enrolled and 22 serum samples from each group were analyzed. Effective separation was noted regarding the UAP and control groups, and, for the former group considered in relation to their counterpart, the serum concentrations of Lac, m-I, lipid, VLDL, 3-HB, and LDL were higher whereas the concentrations of Thr, Cr, Cho, PC/GPC, Glu, Gln, Lys, HDL, Ile, Leu, and Val were lower. The conclusion drawn in view of the results is that the plasma metabolomics examined by 1H NMR displayed promise for biomarker identification for UA. In addition to this, the analysis illuminated the metabolic processes of UA.
Collapse
|
49
|
Cheng S, Shah SH, Corwin EJ, Fiehn O, Fitzgerald RL, Gerszten RE, Illig T, Rhee EP, Srinivas PR, Wang TJ, Jain M. Potential Impact and Study Considerations of Metabolomics in Cardiovascular Health and Disease: A Scientific Statement From the American Heart Association. ACTA ACUST UNITED AC 2017; 10:HCG.0000000000000032. [PMID: 28360086 DOI: 10.1161/hcg.0000000000000032] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Through the measure of thousands of small-molecule metabolites in diverse biological systems, metabolomics now offers the potential for new insights into the factors that contribute to complex human diseases such as cardiovascular disease. Targeted metabolomics methods have already identified new molecular markers and metabolomic signatures of cardiovascular disease risk (including branched-chain amino acids, select unsaturated lipid species, and trimethylamine-N-oxide), thus in effect linking diverse exposures such as those from dietary intake and the microbiota with cardiometabolic traits. As technologies for metabolomics continue to evolve, the depth and breadth of small-molecule metabolite profiling in complex systems continue to advance rapidly, along with prospects for ongoing discovery. Current challenges facing the field of metabolomics include scaling throughput and technical capacity for metabolomics approaches, bioinformatic and chemoinformatic tools for handling large-scale metabolomics data, methods for elucidating the biochemical structure and function of novel metabolites, and strategies for determining the true clinical relevance of metabolites observed in association with cardiovascular disease outcomes. Progress made in addressing these challenges will allow metabolomics the potential to substantially affect diagnostics and therapeutics in cardiovascular medicine.
Collapse
|
50
|
Mueller-Hennessen M, Sigl J, Fuhrmann JC, Witt H, Reszka R, Schmitz O, Kastler J, Fischer JJ, Müller OJ, Giannitsis E, Weis T, Frey N, Katus HA. Metabolic profiles in heart failure due to non-ischemic cardiomyopathy at rest and under exercise. ESC Heart Fail 2017; 4:178-189. [PMID: 28451455 PMCID: PMC5396036 DOI: 10.1002/ehf2.12133] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 11/02/2016] [Accepted: 12/08/2016] [Indexed: 11/05/2022] Open
Abstract
AIMS Identification of metabolic signatures in heart failure (HF) patients and evaluation of their diagnostic potential to discriminate HF patients from healthy controls during baseline and exercise conditions. METHODS Plasma samples were collected from 22 male HF patients with non-ischemic idiopathic cardiomyopathy and left ventricular systolic dysfunction and 19 healthy controls before (t0), at peak (t1) and 1 h after (t2) symptom-limited cardiopulmonary exercise testing. Two hundred fifty-two metabolites were quantified by gas chromatography-mass spectrometry (GC-MS) and liquid chromatography (LC)-MS/MS-based metabolite profiling. RESULTS Plasma metabolite profiles clearly differed between HF patients and controls at t0 (P < 0.05). The metabolic signature of HF was characterized by decreased levels of complex lipids and fatty acids, notably phosphatidylcholines, cholesterol, and sphingolipids. Moreover, reduced glutamine and increased glutamate plasma levels, significantly increased purine degradation products, as well as signs of impaired glucose metabolism were observed. The metabolic differences increased strongly according to New York Heart Association functional class and the addition of three metabolites further improved prediction of exercise capacity (Q2 = 0.24 to 0.35). Despite a high number of metabolites changing significantly with exercise (30.2% at t1/t0), the number of significant alterations between HF and controls was almost unchanged at t1 and t2 (30.7 and 29.0% vs. 31.3% at t0) with a similar predictive group separation (Q2 = 0.50 for t0, 0.52 for t1, and 0.56 for t2, respectively). CONCLUSIONS Our study identified a metabolic signature of non-ischemic HF with prominent changes in complex lipids including phosphatidylcholines, cholesterol, and sphingolipids. The metabolic changes were already evident at rest and largely preserved under exercise.
Collapse
Affiliation(s)
- Matthias Mueller-Hennessen
- Department of Internal Medicine III, Cardiology, Angiology & PneumologyUniversity of HeidelbergHeidelbergGermany.,DZHK (German Centre for Cardiovascular Research)BerlinGermany
| | - Johanna Sigl
- Department of Internal Medicine III, Cardiology, Angiology & PneumologyUniversity of HeidelbergHeidelbergGermany
| | | | | | | | | | | | | | - Oliver J Müller
- Department of Internal Medicine III, Cardiology, Angiology & PneumologyUniversity of HeidelbergHeidelbergGermany.,DZHK (German Centre for Cardiovascular Research)BerlinGermany
| | - Evangelos Giannitsis
- Department of Internal Medicine III, Cardiology, Angiology & PneumologyUniversity of HeidelbergHeidelbergGermany
| | - Tanja Weis
- Department of Internal Medicine III, Cardiology, Angiology & PneumologyUniversity of HeidelbergHeidelbergGermany.,DZHK (German Centre for Cardiovascular Research)BerlinGermany
| | - Norbert Frey
- Department of Cardiology and AngiologyUniversity of KielKielGermany.,DZHK (German Centre for Cardiovascular Research)BerlinGermany
| | - Hugo A Katus
- Department of Internal Medicine III, Cardiology, Angiology & PneumologyUniversity of HeidelbergHeidelbergGermany.,DZHK (German Centre for Cardiovascular Research)BerlinGermany
| |
Collapse
|