101
|
Yoshii A, Tian R. Deciphering metabolic remodeling of the failing hearts. NATURE CARDIOVASCULAR RESEARCH 2022; 1:800-801. [PMID: 39196073 DOI: 10.1038/s44161-022-00118-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Akira Yoshii
- Mitochondria and Metabolism Center, University of Washington, Seattle, WA, USA
| | - Rong Tian
- Mitochondria and Metabolism Center, University of Washington, Seattle, WA, USA.
| |
Collapse
|
102
|
Jedrzejewska A, Braczko A, Kawecka A, Hellmann M, Siondalski P, Slominska E, Kutryb-Zajac B, Yacoub MH, Smolenski RT. Novel Targets for a Combination of Mechanical Unloading with Pharmacotherapy in Advanced Heart Failure. Int J Mol Sci 2022; 23:9886. [PMID: 36077285 PMCID: PMC9456495 DOI: 10.3390/ijms23179886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 12/19/2022] Open
Abstract
LVAD therapy is an effective rescue in acute and especially chronic cardiac failure. In several scenarios, it provides a platform for regeneration and sustained myocardial recovery. While unloading seems to be a key element, pharmacotherapy may provide powerful tools to enhance effective cardiac regeneration. The synergy between LVAD support and medical agents may ensure satisfying outcomes on cardiomyocyte recovery followed by improved quality and quantity of patient life. This review summarizes the previous and contemporary strategies for combining LVAD with pharmacotherapy and proposes new therapeutic targets. Regulation of metabolic pathways, enhancing mitochondrial biogenesis and function, immunomodulating treatment, and stem-cell therapies represent therapeutic areas that require further experimental and clinical studies on their effectiveness in combination with mechanical unloading.
Collapse
Affiliation(s)
- Agata Jedrzejewska
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| | - Alicja Braczko
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| | - Ada Kawecka
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| | - Marcin Hellmann
- Department of Cardiac Diagnostics, Medical University of Gdansk, Smoluchowskiego 17, 80-214 Gdansk, Poland
| | - Piotr Siondalski
- Department of Cardiac Surgery, Medical University of Gdansk, Debinki 7 Street, 80-211 Gdansk, Poland
| | - Ewa Slominska
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| | - Barbara Kutryb-Zajac
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| | - Magdi H. Yacoub
- Heart Science Centre, Imperial College of London at Harefield Hospital, Harefield UB9 6JH, UK
| | - Ryszard T. Smolenski
- Department of Biochemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| |
Collapse
|
103
|
Liu M, Lv J, Pan Z, Wang D, Zhao L, Guo X. Mitochondrial dysfunction in heart failure and its therapeutic implications. Front Cardiovasc Med 2022; 9:945142. [PMID: 36093152 PMCID: PMC9448986 DOI: 10.3389/fcvm.2022.945142] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/04/2022] [Indexed: 11/18/2022] Open
Abstract
The ATP consumption in heart is very intensive to support muscle contraction and relaxation. Mitochondrion is the power plant of the cell. Mitochondrial dysfunction has long been believed as the primary mechanism responsible for the inability of energy generation and utilization in heart failure. In addition, emerging evidence has demonstrated that mitochondrial dysfunction also contributes to calcium dysregulation, oxidative stress, proteotoxic insults and cardiomyocyte death. These elements interact with each other to form a vicious circle in failing heart. The role of mitochondrial dysfunction in the pathogenesis of heart failure has attracted increasing attention. The complex signaling of mitochondrial quality control provides multiple targets for maintaining mitochondrial function. Design of therapeutic strategies targeting mitochondrial dysfunction holds promise for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Miaosen Liu
- Clinical Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Jialan Lv
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhicheng Pan
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongfei Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liding Zhao
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Xiaogang Guo,
| |
Collapse
|
104
|
GDH promotes isoprenaline-induced cardiac hypertrophy by activating mTOR signaling via elevation of α-ketoglutarate level. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:1373-1385. [PMID: 35904584 DOI: 10.1007/s00210-022-02252-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/01/2022] [Indexed: 10/16/2022]
Abstract
Numerous studies reveal that metabolism dysfunction contributes to the development of pathological cardiac hypertrophy. While the abnormal lipid and glucose utilization in cardiomyocytes responding to hypertrophic stimuli have been extensively studied, the alteration and implication of glutaminolysis are rarely discussed. In the present work, we provide the first evidence that glutamate dehydrogenase (GDH), an enzyme that catalyzes conversion of glutamate into ɑ-ketoglutarate (AKG), participates in isoprenaline (ISO)-induced cardiac hypertrophy through activating mammalian target of rapamycin (mTOR) signaling. The expression and activity of GDH were enhanced in cultured cardiomyocytes and rat hearts following ISO treatment. Overexpression of GDH, but not its enzymatically inactive mutant, provoked cardiac hypertrophy. In contrast, GDH knockdown could relieve ISO-triggered hypertrophic responses. The intracellular AKG level was elevated by ISO or GDH overexpression, which led to increased phosphorylation of mTOR and downstream effector ribosomal protein S6 kinase (S6K). Exogenous supplement of AKG also resulted in mTOR activation and cardiomyocyte hypertrophy. However, incubation with rapamycin, an mTOR inhibitor, attenuated hypertrophic responses in cardiomyocytes. Furthermore, GDH silencing protected rats from ISO-induced cardiac hypertrophy. These findings give a further insight into the role of GDH in cardiac hypertrophy and suggest it as a potential target for hypertrophy-related cardiomyopathy.
Collapse
|
105
|
'Warburg effect' controls tumor growth, bacterial, viral infections and immunity - Genetic deconstruction and therapeutic perspectives. Semin Cancer Biol 2022; 86:334-346. [PMID: 35820598 DOI: 10.1016/j.semcancer.2022.07.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/16/2022]
Abstract
The evolutionary pressure for life transitioning from extended periods of hypoxia to an increasingly oxygenated atmosphere initiated drastic selections for a variety of biochemical pathways supporting the robust life currently present on the planet. First, we discuss how fermentative glycolysis, a primitive metabolic pathway present at the emergence of life, is instrumental for the rapid growth of cancer, regenerating tissues, immune cells but also bacteria and viruses during infections. The 'Warburg effect', activated via Myc and HIF-1 in response to growth factors and hypoxia, is an essential metabolic and energetic pathway which satisfies nutritional and energetic demands required for rapid genome replication. Second, we present the key role of lactic acid, the end-product of fermentative glycolysis able to move across cell membranes in both directions via monocarboxylate transporting proteins (i.e. MCT1/4) contributing to cell-pH homeostasis but also to the complex immune response via acidosis of the tumour microenvironment. Importantly lactate is recycled in multiple organs as a major metabolic precursor of gluconeogenesis and energy source protecting cells and animals from harsh nutritional or oxygen restrictions. Third, we revisit the Warburg effect via CRISPR-Cas9 disruption of glucose-6-phosphate isomerase (GPI-KO) or lactate dehydrogenases (LDHA/B-DKO) in two aggressive tumours (melanoma B16-F10, human adenocarcinoma LS174T). Full suppression of lactic acid production reduces but does not suppress tumour growth due to reactivation of OXPHOS. In contrast, disruption of the lactic acid transporters MCT1/4 suppressed glycolysis, mTORC1, and tumour growth as a result of intracellular acidosis. Finally, we briefly discuss the current clinical developments of an MCT1 specific drug AZ3965, and the recent progress for a specific in vivo MCT4 inhibitor, two drugs of very high potential for future cancer clinical applications.
Collapse
|
106
|
Gao ZW, Zhang X, Zhuo QY, Chen MX, Yang C, Chen ZJ, Chen Y, Liao YQ, Wang LL. Metabolomics and integrated network pharmacology analysis reveal attenuates cardiac hypertrophic mechanisms of HuoXin pill. JOURNAL OF ETHNOPHARMACOLOGY 2022; 292:115150. [PMID: 35304274 DOI: 10.1016/j.jep.2022.115150] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cardiac hypertrophy (CH) is maladaptive and contributes to the pathogenesis of heart failure. Huoxin pill (HXP), a Chinese herbal prescription, is widely applied in the treatment of cardiovascular disease (CAD). Its mechanism, however, is unclear. AIM OF THE STUDY This study investigated the mechanism of action for Huoxin pill in the treatment of CH, an important stage of CAD. MATERIALS AND METHODS A total of 60 rats were injected with isoprenaline (ISO) to establish a model of CH. Echocardiography and histopathologic evaluation were performed to evaluate the disease severity, whereas ELISAs were conducted to determine the expression of oxidative stress. Network pharmacology and metabolomic analyses were conducted to identify the key compounds, core targets and pathways that mediate the effects of HXP against CH. Western blotting and immunohistochemistry were used to test apoptosis protein levels. RESULTS HXP administration in ISO-treated rats decreased hypertrophy indices, alleviated cardiac pathological damage, and downregulated oxidative stress levels when compared to those of rats subjected to ISO treatment only. Moreover, network pharmacology results suggested that the PI3K-Akt pathway is a main mechanism by which HXP inhibits cardiac hypertrophy, and experimental verification showed that HXP inhibited cardiomyocyte apoptosis via activation of the PI3K-Akt pathway. The results of metabolomic analysis identified 21 differential metabolites between the HXPH group and ISO group, which were considered to be metabolic biomarkers of HXP in the treatment of CH. Among them, 6 differential metabolites were significantly upregulated, and 15 were significantly downregulated. CONCLUSIONS The present study presents an integrated strategy for investigating the mechanisms of HXP in the treatment of CH and sheds new light on the application of HXP as a traditional Chinese medicine.
Collapse
Affiliation(s)
- Zhan-Wang Gao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou Higher Education Mega Center, Guangzhou, 510006, PR China.
| | - Xin Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou Higher Education Mega Center, Guangzhou, 510006, PR China.
| | - Qing-Yuan Zhuo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou Higher Education Mega Center, Guangzhou, 510006, PR China.
| | - Mei-Xian Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou Higher Education Mega Center, Guangzhou, 510006, PR China.
| | - Chong Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou Higher Education Mega Center, Guangzhou, 510006, PR China.
| | - Zhao-Jie Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou Higher Education Mega Center, Guangzhou, 510006, PR China.
| | - Ying Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou Higher Education Mega Center, Guangzhou, 510006, PR China.
| | - Yi-Qiu Liao
- Baiyunshan Pharmaceutical General Factory, Guangzhou Baiyunshan Pharmaceutical Holdings Co., Ltd., Guangzhou, 510515, PR China; Key Laboratory of Key Technology Research on Chemical Raw Materials and Preparations of Guangdong Province, Guangzhou, 510515, PR China.
| | - Ling-Li Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, No. 232, Waihuandong Road, Guangzhou Higher Education Mega Center, Guangzhou, 510006, PR China.
| |
Collapse
|
107
|
Wang X, Zhang G, Dasgupta S, Niewold EL, Li C, Li Q, Luo X, Tan L, Ferdous A, Lorenzi PL, Rothermel BA, Gillette TG, Adams CM, Scherer PE, Hill JA, Wang ZV. ATF4 Protects the Heart From Failure by Antagonizing Oxidative Stress. Circ Res 2022; 131:91-105. [PMID: 35574856 PMCID: PMC9351829 DOI: 10.1161/circresaha.122.321050] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cellular redox control is maintained by generation of reactive oxygen/nitrogen species balanced by activation of antioxidative pathways. Disruption of redox balance leads to oxidative stress, a central causative event in numerous diseases including heart failure. Redox control in the heart exposed to hemodynamic stress, however, remains to be fully elucidated. METHODS Pressure overload was triggered by transverse aortic constriction in mice. Transcriptomic and metabolomic regulations were evaluated by RNA-sequencing and metabolomics, respectively. Stable isotope tracer labeling experiments were conducted to determine metabolic flux in vitro. Neonatal rat ventricular myocytes and H9c2 cells were used to examine molecular mechanisms. RESULTS We show that production of cardiomyocyte NADPH, a key factor in redox regulation, is decreased in pressure overload-induced heart failure. As a consequence, the level of reduced glutathione is downregulated, a change associated with fibrosis and cardiomyopathy. We report that the pentose phosphate pathway and mitochondrial serine/glycine/folate metabolic signaling, 2 NADPH-generating pathways in the cytosol and mitochondria, respectively, are induced by transverse aortic constriction. We identify ATF4 (activating transcription factor 4) as an upstream transcription factor controlling the expression of multiple enzymes in these 2 pathways. Consistently, joint pathway analysis of transcriptomic and metabolomic data reveal that ATF4 preferably controls oxidative stress and redox-related pathways. Overexpression of ATF4 in neonatal rat ventricular myocytes increases NADPH-producing enzymes' whereas silencing of ATF4 decreases their expression. Further, stable isotope tracer experiments reveal that ATF4 overexpression augments metabolic flux within these 2 pathways. In vivo, cardiomyocyte-specific deletion of ATF4 exacerbates cardiomyopathy in the setting of transverse aortic constriction and accelerates heart failure development, attributable, at least in part, to an inability to increase the expression of NADPH-generating enzymes. CONCLUSIONS Our findings reveal that ATF4 plays a critical role in the heart under conditions of hemodynamic stress by governing both cytosolic and mitochondrial production of NADPH.
Collapse
Affiliation(s)
- Xiaoding Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Guangyu Zhang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Subhajit Dasgupta
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Erica L. Niewold
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chao Li
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Qinfeng Li
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xiang Luo
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anwarul Ferdous
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Philip L. Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Beverly A. Rothermel
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas G. Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher M. Adams
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph A. Hill
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zhao V. Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
108
|
Zhang X, Wang Y, Hu S. Editorial: Novel Insights Into Obesity-Related Diseases. Front Physiol 2022; 13:952682. [PMID: 35812312 PMCID: PMC9261280 DOI: 10.3389/fphys.2022.952682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Xiang Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Xiang Zhang, ; Sanyuan Hu,
| | - Yanmin Wang
- California Medical Innovations Institute, San Diego, CA, United States
| | - Sanyuan Hu
- First Affiliated Hospital of Shandong First Medical University, Jinan, China
- Shandong University, Jinan, China
- *Correspondence: Xiang Zhang, ; Sanyuan Hu,
| |
Collapse
|
109
|
Kyriakopoulos CP, Kapelios CJ, Stauder EL, Taleb I, Hamouche R, Sideris K, Koliopoulou AG, Bonios MJ, Drakos SG. LVAD as a Bridge to Remission from Advanced Heart Failure: Current Data and Opportunities for Improvement. J Clin Med 2022; 11:3542. [PMID: 35743611 PMCID: PMC9225013 DOI: 10.3390/jcm11123542] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 02/04/2023] Open
Abstract
Left ventricular assist devices (LVADs) are an established treatment modality for advanced heart failure (HF). It has been shown that through volume and pressure unloading they can lead to significant functional and structural cardiac improvement, allowing LVAD support withdrawal in a subset of patients. In the first part of this review, we discuss the historical background, current evidence on the incidence and assessment of LVAD-mediated cardiac recovery, and out-comes including quality of life after LVAD support withdrawal. In the second part, we discuss current and future opportunities to promote LVAD-mediated reverse remodeling and improve our pathophysiological understanding of HF and recovery for the benefit of the greater HF population.
Collapse
Affiliation(s)
- Christos P. Kyriakopoulos
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health & School of Medicine, Salt Lake City, UT 84132, USA; (C.P.K.); (C.J.K.); (E.L.S.); (I.T.); (K.S.); (A.G.K.); (M.J.B.)
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA;
| | - Chris J. Kapelios
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health & School of Medicine, Salt Lake City, UT 84132, USA; (C.P.K.); (C.J.K.); (E.L.S.); (I.T.); (K.S.); (A.G.K.); (M.J.B.)
| | - Elizabeth L. Stauder
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health & School of Medicine, Salt Lake City, UT 84132, USA; (C.P.K.); (C.J.K.); (E.L.S.); (I.T.); (K.S.); (A.G.K.); (M.J.B.)
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA;
| | - Iosif Taleb
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health & School of Medicine, Salt Lake City, UT 84132, USA; (C.P.K.); (C.J.K.); (E.L.S.); (I.T.); (K.S.); (A.G.K.); (M.J.B.)
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA;
| | - Rana Hamouche
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA;
| | - Konstantinos Sideris
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health & School of Medicine, Salt Lake City, UT 84132, USA; (C.P.K.); (C.J.K.); (E.L.S.); (I.T.); (K.S.); (A.G.K.); (M.J.B.)
| | - Antigone G. Koliopoulou
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health & School of Medicine, Salt Lake City, UT 84132, USA; (C.P.K.); (C.J.K.); (E.L.S.); (I.T.); (K.S.); (A.G.K.); (M.J.B.)
- Divisions of Cardiology & Cardiothoracic Surgery, Onassis Cardiac Surgery Center, 17674 Athens, Greece
| | - Michael J. Bonios
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health & School of Medicine, Salt Lake City, UT 84132, USA; (C.P.K.); (C.J.K.); (E.L.S.); (I.T.); (K.S.); (A.G.K.); (M.J.B.)
- Divisions of Cardiology & Cardiothoracic Surgery, Onassis Cardiac Surgery Center, 17674 Athens, Greece
| | - Stavros G. Drakos
- Divisions of Cardiovascular Medicine and Cardiothoracic Surgery, University of Utah Health & School of Medicine, Salt Lake City, UT 84132, USA; (C.P.K.); (C.J.K.); (E.L.S.); (I.T.); (K.S.); (A.G.K.); (M.J.B.)
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA;
| |
Collapse
|
110
|
Xu B, Li F, Zhang W, Su Y, Tang L, Li P, Joshi J, Yang A, Li D, Wang Z, Wang S, Xie J, Gu H, Zhu W. Identification of metabolic pathways underlying FGF1 and CHIR99021-mediated cardioprotection. iScience 2022; 25:104447. [PMID: 35707727 PMCID: PMC9189130 DOI: 10.1016/j.isci.2022.104447] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/16/2022] [Accepted: 05/18/2022] [Indexed: 12/05/2022] Open
Abstract
Acute myocardial infarction is a leading cause of death worldwide. We have previously identified two cardioprotective molecules — FGF1 and CHIR99021— that confer cardioprotection in mouse and pig models of acute myocardial infarction. Here, we aimed to determine if improved myocardial metabolism contributes to this cardioprotection. Nanofibers loaded with FGF1 and CHIR99021 were intramyocardially injected to ischemic myocardium of adult mice immediately following surgically induced myocardial infarction. Animals were euthanized 3 and 7 days later. Our data suggested that FGF1/CHIR99021 nanofibers enhanced the heart’s capacity to utilize glycolysis as an energy source and reduced the accumulation of branched-chain amino acids in ischemic myocardium. The impact of FGF1/CHIR99021 on metabolism was more obvious in the first three days post myocardial infarction. Taken together, these findings suggest that FGF1/CHIR99021 protects the heart against ischemic injury via improving myocardial metabolism which may be exploited for treatment of acute myocardial infarction in humans. FGF1/CHIR confer cardioprotection in myocardial infarction animals FGF1/CHIR enhance the capability of ischemic hearts to produce energy via glycolysis FGF1/CHIR reduce the abundance of branched chain amino acids in ischemic hearts This study reveals a novel approach to correct metabolic disorders in ischemic hearts
Collapse
Affiliation(s)
- Bing Xu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259.,Department of Cardiology, Northern Jiangsu People's Hospital, Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Fan Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259.,Department of Kinesiology, South China Normal University, Guangzhou 510631, China
| | - Wenjing Zhang
- Center for Translational Science, Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Port St. Lucie, FL 34987, USA.,College of Health Solutions, Arizona State University, Phoenix, AZ 85287, USA
| | - Yajuan Su
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ling Tang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Pengsheng Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Jyotsna Joshi
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Aaron Yang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Dong Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| | - Zhao Wang
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Shu Wang
- College of Health Solutions, Arizona State University, Phoenix, AZ 85287, USA
| | - Jingwei Xie
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Haiwei Gu
- Center for Translational Science, Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Port St. Lucie, FL 34987, USA.,College of Health Solutions, Arizona State University, Phoenix, AZ 85287, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ, USA 85259
| |
Collapse
|
111
|
Abstract
As a muscular pump that contracts incessantly throughout life, the heart must constantly generate cellular energy to support contractile function and fuel ionic pumps to maintain electrical homeostasis. Thus, mitochondrial metabolism of multiple metabolic substrates such as fatty acids, glucose, ketones, and lactate is essential to ensuring an uninterrupted supply of ATP. Multiple metabolic pathways converge to maintain myocardial energy homeostasis. The regulation of these cardiac metabolic pathways has been intensely studied for many decades. Rapid adaptation of these pathways is essential for mediating the myocardial adaptation to stress, and dysregulation of these pathways contributes to myocardial pathophysiology as occurs in heart failure and in metabolic disorders such as diabetes. The regulation of these pathways reflects the complex interactions of cell-specific regulatory pathways, neurohumoral signals, and changes in substrate availability in the circulation. Significant advances have been made in the ability to study metabolic regulation in the heart, and animal models have played a central role in contributing to this knowledge. This review will summarize metabolic pathways in the heart and describe their contribution to maintaining myocardial contractile function in health and disease. The review will summarize lessons learned from animal models with altered systemic metabolism and those in which specific metabolic regulatory pathways have been genetically altered within the heart. The relationship between intrinsic and extrinsic regulators of cardiac metabolism and the pathophysiology of heart failure and how these have been informed by animal models will be discussed.
Collapse
Affiliation(s)
- Heiko Bugger
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - Nikole J Byrne
- University Heart Center Graz, Department of Cardiology, Medical University of Graz, Graz, Austria, Austria (H.B., N.J.B.)
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (E.D.A.)
| |
Collapse
|
112
|
Metabolic Determinants in Cardiomyocyte Function and Heart Regenerative Strategies. Metabolites 2022; 12:metabo12060500. [PMID: 35736435 PMCID: PMC9227827 DOI: 10.3390/metabo12060500] [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: 05/11/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/04/2023] Open
Abstract
Heart disease is the leading cause of mortality in developed countries. The associated pathology is characterized by a loss of cardiomyocytes that leads, eventually, to heart failure. In this context, several cardiac regenerative strategies have been developed, but they still lack clinical effectiveness. The mammalian neonatal heart is capable of substantial regeneration following injury, but this capacity is lost at postnatal stages when cardiomyocytes become terminally differentiated and transit to the fetal metabolic switch. Cardiomyocytes are metabolically versatile cells capable of using an array of fuel sources, and the metabolism of cardiomyocytes suffers extended reprogramming after injury. Apart from energetic sources, metabolites are emerging regulators of epigenetic programs driving cell pluripotency and differentiation. Thus, understanding the metabolic determinants that regulate cardiomyocyte maturation and function is key for unlocking future metabolic interventions for cardiac regeneration. In this review, we will discuss the emerging role of metabolism and nutrient signaling in cardiomyocyte function and repair, as well as whether exploiting this axis could potentiate current cellular regenerative strategies for the mammalian heart.
Collapse
|
113
|
Xiao D, Hu C, Xu X, Lü C, Wang Q, Zhang W, Gao C, Xu P, Wang X, Ma C. A d,l-lactate biosensor based on allosteric transcription factor LldR and amplified luminescent proximity homogeneous assay. Biosens Bioelectron 2022; 211:114378. [PMID: 35617798 DOI: 10.1016/j.bios.2022.114378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 11/02/2022]
Abstract
Lactate, a hydroxycarboxylic acid commercially produced by microbial fermentation, is widely applied in diverse industrial fields. Lactate exists in two stereoisomeric forms (d-lactate and l-lactate). d-Lactate and l-lactate are often simultaneously present in many biological samples. Therefore, a biosensor able to detect both d- and l-lactate is required but previously unavailable. Herein, an allosteric transcription factor LldR from Pseudomonas aeruginosa PAO1, which responds to both d-lactate and l-lactate, was combined with amplified luminescent proximity homogeneous assay technology to develop a d,l-lactate biosensor. The proposed biosensor was optimized by mutation of DNA sequence in binding site of LldR. The optimized biosensor BLac-6 can accurately detect the concentration of lactate independent on ratio of the two isomers in pending test samples. The biosensor was also tentatively used in quantitative analysis of d-lactate, l-lactate, or d,l-lactate in fermentation samples produced by three recombinant strains of Klebsiella oxytoca. With its desirable properties, the biosensor BLac-6 may be a potential choice for monitoring the concentration of lactate during industrial fermentation.
Collapse
Affiliation(s)
- Dan Xiao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, PR China
| | - Chunxia Hu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, PR China
| | - Xianzhi Xu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, PR China
| | - Chuanjuan Lü
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, PR China
| | - Qian Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, PR China
| | - Wen Zhang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, PR China
| | - Chao Gao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, PR China
| | - Ping Xu
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Xia Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, PR China
| | - Cuiqing Ma
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, PR China.
| |
Collapse
|
114
|
Zhou B, Caudal A, Tang X, Chavez JD, McMillen TS, Keller A, Villet O, Zhao M, Liu Y, Ritterhoff J, Wang P, Kolwicz SC, Wang W, Bruce JE, Tian R. Upregulation of mitochondrial ATPase inhibitory factor 1 (ATPIF1) mediates increased glycolysis in mouse hearts. J Clin Invest 2022; 132:e155333. [PMID: 35575090 PMCID: PMC9106352 DOI: 10.1172/jci155333] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 04/01/2022] [Indexed: 12/20/2022] Open
Abstract
In hypertrophied and failing hearts, fuel metabolism is reprogrammed to increase glucose metabolism, especially glycolysis. This metabolic shift favors biosynthetic function at the expense of ATP production. Mechanisms responsible for the switch are poorly understood. We found that inhibitory factor 1 of the mitochondrial FoF1-ATP synthase (ATPIF1), a protein known to inhibit ATP hydrolysis by the reverse function of ATP synthase during ischemia, was significantly upregulated in pathological cardiac hypertrophy induced by pressure overload, myocardial infarction, or α-adrenergic stimulation. Chemical cross-linking mass spectrometry analysis of hearts hypertrophied by pressure overload suggested that increased expression of ATPIF1 promoted the formation of FoF1-ATP synthase nonproductive tetramer. Using ATPIF1 gain- and loss-of-function cell models, we demonstrated that stalled electron flow due to impaired ATP synthase activity triggered mitochondrial ROS generation, which stabilized HIF1α, leading to transcriptional activation of glycolysis. Cardiac-specific deletion of ATPIF1 in mice prevented the metabolic switch and protected against the pathological remodeling during chronic stress. These results uncover a function of ATPIF1 in nonischemic hearts, which gives FoF1-ATP synthase a critical role in metabolic rewiring during the pathological remodeling of the heart.
Collapse
Affiliation(s)
- Bo Zhou
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, and
| | - Arianne Caudal
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, and
| | - Xiaoting Tang
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Juan D. Chavez
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Timothy S. McMillen
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, and
| | - Andrew Keller
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Outi Villet
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, and
| | - Mingyue Zhao
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, and
| | - Yaxin Liu
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, and
| | - Julia Ritterhoff
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, and
| | - Pei Wang
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, and
| | - Stephen C. Kolwicz
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, and
| | - Wang Wang
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, and
| | - James E. Bruce
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, and
| |
Collapse
|
115
|
Cheng X, Chu J, Zhang L, Suo Z, Tang W. Intracellular and extracellular untargeted metabolomics reveal the effect of acute uranium exposure in HK-2 cells. Toxicology 2022; 473:153196. [PMID: 35525329 DOI: 10.1016/j.tox.2022.153196] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/30/2022] [Accepted: 04/30/2022] [Indexed: 11/15/2022]
Abstract
Uranium exposure poses a serious threat to the health of occupational populations and the public. Although metabolomics is a promising research approach to study the toxicological mechanisms of uranium exposure, in vitro studies using human cells are scarce. Applying cultured cell metabolomics, we exhaustively analyzed the intracellular and extracellular differential metabolites upon uranium exposure and characterized the possible biological effects of uranium exposure on human kidney cells. Uranium exposure significantly induced disturbance in the amino acid biosynthesis and linoleic acid metabolism of the cells. Cells exposed to uranium produce excessive amounts of arachidonic acid, which has the potential to cause oxidative stress and damage cells. The results provide new evidence for an oxidative stress mechanism of uranium-induced renal cell injury. Cell metabolomics has proven to be a useful diagnostic tool to study the molecular mechanisms of uranium poisoning.
Collapse
Affiliation(s)
- Xuedan Cheng
- School of Materials Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China; Center for Medical Radiation Biology, 903 Hospital, Institute of Materials, China Academy of Engineering Physics, Mianyang, 621907, China
| | - Jian Chu
- Center for Medical Radiation Biology, 903 Hospital, Institute of Materials, China Academy of Engineering Physics, Mianyang, 621907, China
| | - Liandong Zhang
- Center for Medical Radiation Biology, 903 Hospital, Institute of Materials, China Academy of Engineering Physics, Mianyang, 621907, China
| | - Zhirong Suo
- School of Materials Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China.
| | - Wei Tang
- Center for Medical Radiation Biology, 903 Hospital, Institute of Materials, China Academy of Engineering Physics, Mianyang, 621907, China.
| |
Collapse
|
116
|
Heart failure with improved ejection fraction: The current and future in Asian populations. Int J Cardiol 2022; 358:72-73. [PMID: 35487319 DOI: 10.1016/j.ijcard.2022.04.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 11/20/2022]
|
117
|
Liu J, Li W, Deng KQ, Tian S, Liu H, Shi H, Fang Q, Liu Z, Chen Z, Tian T, Gan S, Hu F, Hu M, Cheng X, Ji YX, Zhang P, She ZG, Zhang XJ, Chen S, Cai J, Li H. The E3 Ligase TRIM16 Is a Key Suppressor of Pathological Cardiac Hypertrophy. Circ Res 2022; 130:1586-1600. [PMID: 35437018 DOI: 10.1161/circresaha.121.318866] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Pathological cardiac hypertrophy is one of the leading causes of heart failure with highly complicated pathogeneses. The E3 ligase TRIM16 (tripartite motif-containing protein 16) has been recognized as a pivotal regulator to control cell survival, immune response, and oxidative stress. However, the role of Trim16 in cardiac hypertrophy is unknown. METHODS We generated cardiac-specific knockout mice and adeno-associated virus serotype 9-Trim16 mice to evaluate the function of Trim16 in pathological myocardial hypertrophy. The direct effect of TRIM16 on cardiomyocyte enlargement was examined using an adenovirus system. Furthermore, we combined RNA-sequencing and interactome analysis that was followed by multiple molecular biological methodologies to identify the direct target and corresponding molecular events contributing to TRIM16 function. RESULTS We found an intimate correlation of Trim16 expression with hypertrophy-related heart failure in both human and mouse. Our functional investigations and unbiased transcriptomic analyses clearly demonstrated that Trim16 deficiency markedly exacerbated cardiomyocyte enlargement in vitro and in transverse aortic constriction-induced cardiac hypertrophy mouse model, whereas Trim16 overexpression attenuated cardiac hypertrophy and remodeling. Mechanistically, Prdx1 (peroxiredoxin 1) is an essential target of Trim16 in cardiac hypertrophy. We found that Trim16 interacts with Prdx1 and inhibits its phosphorylation, leading to a robust enhancement of its downstream Nrf2 (nuclear factor-erythroid 2-related factor 2) pathway to block cardiac hypertrophy. Trim16-blocked Prdx1 phosphorylation was largely dependent on a direct interaction between Trim16 and Src and the resultant Src ubiquitinational degradation. Notably, Prdx1 knockdown largely abolished the anti-hypertrophic effects of Trim16 overexpression. CONCLUSIONS Our findings provide the first evidence supporting Trim16 as a novel suppressor of pathological cardiac hypertrophy and indicate that targeting the Trim16-Prdx1 axis represents a promising therapeutic strategy for hypertrophy-related heart failure.
Collapse
Affiliation(s)
- Jiayi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.L.).,Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Wei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.L.).,Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Ke-Qiong Deng
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Department of Cardiology, Zhongnan Hospital of Wuhan University, China. (K.-Q.D., Z.C.)
| | - Song Tian
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Hui Liu
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China. (H. Liu, M.H., X.C.).,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China. (H. Liu, M.H., X.C.)
| | - Hongjie Shi
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Qian Fang
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Zhen Liu
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Ze Chen
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Department of Cardiology, Zhongnan Hospital of Wuhan University, China. (K.-Q.D., Z.C.)
| | - Tian Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.L.).,Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Shanyu Gan
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Fengjiao Hu
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Medical Science Research Center, Zhongnan Hospital of Wuhan University, China. (F.H., H. Li)
| | - Manli Hu
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China. (H. Liu, M.H., X.C.).,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China. (H. Liu, M.H., X.C.)
| | - Xu Cheng
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China. (H. Liu, M.H., X.C.).,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China. (H. Liu, M.H., X.C.)
| | - Yan-Xiao Ji
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Peng Zhang
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.L.).,Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| | - Xiao-Jing Zhang
- Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li)
| | - Shaoze Chen
- Department of Cardiology, Huanggang Central Hospital, China (S.C.).,Huanggang Institute of Translational Medicine, Huanggang, China (S.C.)
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China (J.C.)
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China (J.L., W.L., T.T., Z.-G.S., H.L.).,Institute of Model Animal of Wuhan University, China (J.L., W.L., K.-Q.D., S.T., H. Liu, H.S., Q.F., Z.L., Z.C., T.T., S.G., F.H., M.H., X.C., Y.-X.J., P.Z., Z.-G.S., X.-J.Z., H. Li).,Medical Science Research Center, Zhongnan Hospital of Wuhan University, China. (F.H., H. Li).,School of Basic Medical Sciences, Wuhan University, China (H.S., S.G., Y.-X.J., P.Z., X.-J.Z., H. Li)
| |
Collapse
|
118
|
Vandewalle J, Libert C. Sepsis: a failing starvation response. Trends Endocrinol Metab 2022; 33:292-304. [PMID: 35181202 DOI: 10.1016/j.tem.2022.01.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 12/22/2022]
Abstract
Sepsis is involved in ~ 20% of annual global deaths. Despite decades of research, the current management of sepsis remains supportive rather than curative. Clinical trials in sepsis have mainly been focused on targeting the inflammatory pathway, but without success. Recent data indicate that metabolic dysregulation takes place in sepsis, and targeting metabolic pathways might hold much promise for the management of sepsis. Sepsis yields a strong starvation response, including the release of high-energy metabolites such as lactate and free fatty acids. However, the activity of two major transcription factors, GR and PPARα, is downregulated in hepatocytes, leading to the accumulation and toxicity of metabolites that, moreover, fail to be transformed into useful molecules such as glucose and ketones. We review the literature and suggest mechanisms and potential therapeutic targets that might prevent or revert the fatal metabolic dysregulation in sepsis.
Collapse
Affiliation(s)
- Jolien Vandewalle
- Center for Inflammation Research, Vlaams Instituut voor Biotechnologie (VIB), Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Claude Libert
- Center for Inflammation Research, Vlaams Instituut voor Biotechnologie (VIB), Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
119
|
Chen W, Li Q, Hou R, Liang H, Zhang Y, Yang Y. An integrated metabonomics study to reveal the inhibitory effect and metabolism regulation of taurine on breast cancer. J Pharm Biomed Anal 2022; 214:114711. [PMID: 35306435 DOI: 10.1016/j.jpba.2022.114711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 12/23/2022]
|
120
|
Sommakia S, Almaw NH, Lee SH, Ramadurai DKA, Taleb I, Kyriakopoulos CP, Stubben CJ, Ling J, Campbell RA, Alharethi RA, Caine WT, Navankasattusas S, Hoareau GL, Abraham AE, Fang JC, Selzman CH, Drakos SG, Chaudhuri D. FGF21 (Fibroblast Growth Factor 21) Defines a Potential Cardiohepatic Signaling Circuit in End-Stage Heart Failure. Circ Heart Fail 2022; 15:e008910. [PMID: 34865514 PMCID: PMC8930477 DOI: 10.1161/circheartfailure.121.008910] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Extrinsic control of cardiomyocyte metabolism is poorly understood in heart failure (HF). FGF21 (Fibroblast growth factor 21), a hormonal regulator of metabolism produced mainly in the liver and adipose tissue, is a prime candidate for such signaling. METHODS To investigate this further, we examined blood and tissue obtained from human subjects with end-stage HF with reduced ejection fraction at the time of left ventricular assist device implantation and correlated serum FGF21 levels with cardiac gene expression, immunohistochemistry, and clinical parameters. RESULTS Circulating FGF21 levels were substantially elevated in HF with reduced ejection fraction, compared with healthy subjects (HF with reduced ejection fraction: 834.4 [95% CI, 628.4-1040.3] pg/mL, n=40; controls: 146.0 [86.3-205.7] pg/mL, n=20, P=1.9×10-5). There was clear FGF21 staining in diseased cardiomyocytes, and circulating FGF21 levels negatively correlated with the expression of cardiac genes involved in ketone metabolism, consistent with cardiac FGF21 signaling. FGF21 gene expression was very low in failing and nonfailing hearts, suggesting extracardiac production of the circulating hormone. Circulating FGF21 levels were correlated with BNP (B-type natriuretic peptide) and total bilirubin, markers of chronic cardiac and hepatic congestion. CONCLUSIONS Circulating FGF21 levels are elevated in HF with reduced ejection fraction and appear to bind to the heart. The liver is likely the main extracardiac source. This supports a model of hepatic FGF21 communication to diseased cardiomyocytes, defining a potential cardiohepatic signaling circuit in human HF.
Collapse
Affiliation(s)
- Salah Sommakia
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Naredos H. Almaw
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Sandra H. Lee
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Dinesh K. A. Ramadurai
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Iosif Taleb
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Christos P. Kyriakopoulos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Chris J. Stubben
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112
| | - Jing Ling
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Robert A. Campbell
- Department of Internal Medicine, Division of General Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Rami A. Alharethi
- U.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant Program: University of Utah Healthcare and School of Medicine, Intermountain Medical Center, Salt Lake Veterans Affairs Health Care System, Salt Lake City, UT
| | - William T. Caine
- U.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant Program: University of Utah Healthcare and School of Medicine, Intermountain Medical Center, Salt Lake Veterans Affairs Health Care System, Salt Lake City, UT
| | - Sutip Navankasattusas
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Guillaume L. Hoareau
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- Department of Surgery, Division of Emergency Medicine, University of Utah, Salt Lake City, UT, USA
| | - Anu E. Abraham
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT
| | - James C. Fang
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT
| | - Craig H. Selzman
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- U.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant Program: University of Utah Healthcare and School of Medicine, Intermountain Medical Center, Salt Lake Veterans Affairs Health Care System, Salt Lake City, UT
- Department of Surgery, Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, UT
| | - Stavros G. Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Utah, Salt Lake City, UT
| |
Collapse
|
121
|
Beuchel C, Dittrich J, Pott J, Henger S, Beutner F, Isermann B, Loeffler M, Thiery J, Ceglarek U, Scholz M. Whole Blood Metabolite Profiles Reflect Changes in Energy Metabolism in Heart Failure. Metabolites 2022; 12:metabo12030216. [PMID: 35323659 PMCID: PMC8949022 DOI: 10.3390/metabo12030216] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/15/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
A variety of atherosclerosis and cardiovascular disease (ASCVD) phenotypes are tightly linked to changes in the cardiac energy metabolism that can lead to a loss of metabolic flexibility and to unfavorable clinical outcomes. We conducted an association analysis of 31 ASCVD phenotypes and 97 whole blood amino acids, acylcarnitines and derived ratios in the LIFE-Adult (n = 9646) and LIFE-Heart (n = 5860) studies, respectively. In addition to hundreds of significant associations, a total of 62 associations of six phenotypes were found in both studies. Positive associations of various amino acids and a range of acylcarnitines with decreasing cardiovascular health indicate disruptions in mitochondrial, as well as peroxisomal fatty acid oxidation. We complemented our metabolite association analyses with whole blood and peripheral blood mononuclear cell (PBMC) gene-expression analyses of fatty acid oxidation and ketone-body metabolism related genes. This revealed several differential expressions for the heart failure biomarker N-terminal prohormone of brain natriuretic peptide (NT-proBNP) in peripheral blood mononuclear cell (PBMC) gene expression. Finally, we constructed and compared three prediction models of significant stenosis in the LIFE-Heart study using (1) traditional risk factors only, (2) the metabolite panel only and (3) a combined model. Area under the receiver operating characteristic curve (AUC) comparison of these three models shows an improved prediction accuracy for the combined metabolite and classical risk factor model (AUC = 0.78, 95%-CI: 0.76–0.80). In conclusion, we improved our understanding of metabolic implications of ASCVD phenotypes by observing associations with metabolite concentrations and gene expression of the mitochondrial and peroxisomal fatty acid oxidation. Additionally, we demonstrated the predictive potential of the metabolite profile to improve classification of patients with significant stenosis.
Collapse
Affiliation(s)
- Carl Beuchel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, 04107 Leipzig, Germany; (J.P.); (S.H.); (M.L.)
- Correspondence: (C.B.); (U.C.); (M.S.)
| | - Julia Dittrich
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany; (J.D.); (B.I.); (J.T.)
| | - Janne Pott
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, 04107 Leipzig, Germany; (J.P.); (S.H.); (M.L.)
- LIFE—Leipzig Research Center for Civilization Diseases, Leipzig University, 04103 Leipzig, Germany
| | - Sylvia Henger
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, 04107 Leipzig, Germany; (J.P.); (S.H.); (M.L.)
- LIFE—Leipzig Research Center for Civilization Diseases, Leipzig University, 04103 Leipzig, Germany
| | | | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany; (J.D.); (B.I.); (J.T.)
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, 04107 Leipzig, Germany; (J.P.); (S.H.); (M.L.)
- LIFE—Leipzig Research Center for Civilization Diseases, Leipzig University, 04103 Leipzig, Germany
| | - Joachim Thiery
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany; (J.D.); (B.I.); (J.T.)
- Faculty of Medicine, Christian-Albrecht University of Kiel, 24118 Kiel, Germany
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, 04103 Leipzig, Germany; (J.D.); (B.I.); (J.T.)
- LIFE—Leipzig Research Center for Civilization Diseases, Leipzig University, 04103 Leipzig, Germany
- Correspondence: (C.B.); (U.C.); (M.S.)
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, 04107 Leipzig, Germany; (J.P.); (S.H.); (M.L.)
- LIFE—Leipzig Research Center for Civilization Diseases, Leipzig University, 04103 Leipzig, Germany
- IFB AdiposityDiseases, University Hospital Leipzig, 04103 Leipzig, Germany
- Correspondence: (C.B.); (U.C.); (M.S.)
| |
Collapse
|
122
|
Taleb I, Tseliou E, Fang JC, Drakos SG. A Mechanical Bridge to Recovery as a Bridge to Discovery: Learning From Few and Applying to Many. Circulation 2022; 145:562-564. [PMID: 35188797 PMCID: PMC8900596 DOI: 10.1161/circulationaha.120.052141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Iosif Taleb
- Division of Cardiovascular Medicine, University of Utah Health, Salt Lake City, UT,Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah Health, Salt Lake City, UT
| | - Eleni Tseliou
- Division of Cardiovascular Medicine, University of Utah Health, Salt Lake City, UT,Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah Health, Salt Lake City, UT
| | - James C. Fang
- Division of Cardiovascular Medicine, University of Utah Health, Salt Lake City, UT
| | - Stavros G. Drakos
- Division of Cardiovascular Medicine, University of Utah Health, Salt Lake City, UT,Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah Health, Salt Lake City, UT
| |
Collapse
|
123
|
Lazaropoulos MP, Elrod JW. Mitochondria in Pathological Cardiac Remodeling. CURRENT OPINION IN PHYSIOLOGY 2022; 25:100489. [PMID: 35274068 PMCID: PMC8903307 DOI: 10.1016/j.cophys.2022.100489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Adverse cardiac remodeling is often precipitated by chronic stress or injury inflicted upon the heart during the progression of cardiovascular diseases. Mitochondria play an important role in the cardiomyocyte response to stress by serving as a signaling hub for changes in cellular energetics, redox balance, contractile function, and cell death. Cardiac remodeling involves alterations to mitochondrial form and function that are either compensatory to maintain contractility or maladaptive, which promotes heart failure progression. In this mini-review, we focus on three mitochondrial processes that contribution to cardiac remodeling: Ca2+ signaling, mitochondrial dynamics, and mitochondrial metabolism.
Collapse
|
124
|
Yamamoto T, Sano M. Deranged Myocardial Fatty Acid Metabolism in Heart Failure. Int J Mol Sci 2022; 23:996. [PMID: 35055179 PMCID: PMC8779056 DOI: 10.3390/ijms23020996] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 01/27/2023] Open
Abstract
The heart requires fatty acids to maintain its activity. Various mechanisms regulate myocardial fatty acid metabolism, such as energy production using fatty acids as fuel, for which it is known that coordinated control of fatty acid uptake, β-oxidation, and mitochondrial oxidative phosphorylation steps are important for efficient adenosine triphosphate (ATP) production without unwanted side effects. The fatty acids taken up by cardiomyocytes are not only used as substrates for energy production but also for the synthesis of triglycerides and the replacement reaction of fatty acid chains in cell membrane phospholipids. Alterations in fatty acid metabolism affect the structure and function of the heart. Recently, breakthrough studies have focused on the key transcription factors that regulate fatty acid metabolism in cardiomyocytes and the signaling systems that modify their functions. In this article, we reviewed the latest research on the role of fatty acid metabolism in the pathogenesis of heart failure and provide an outlook on future challenges.
Collapse
Affiliation(s)
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| |
Collapse
|
125
|
Chatfield KC, Sparagna GC, Specht KS, Whitcomb LA, Omar AK, Miyamoto SD, Wolfe LM, Chicco AJ. Long-chain fatty acid oxidation and respiratory complex I deficiencies distinguish Barth Syndrome from idiopathic pediatric cardiomyopathy. J Inherit Metab Dis 2022; 45:111-124. [PMID: 34821394 DOI: 10.1002/jimd.12459] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/14/2021] [Accepted: 11/23/2021] [Indexed: 12/16/2022]
Abstract
Barth syndrome (BTHS) is an X-linked disorder that results from mutations in the TAFAZZIN gene, which encodes a phospholipid transacylase responsible for generating the mature form of cardiolipin in inner mitochondrial membranes. BTHS patients develop early onset cardiomyopathy and a derangement of intermediary metabolism consistent with mitochondrial disease, but the precise alterations in cardiac metabolism that distinguish BTHS from idiopathic forms of cardiomyopathy are unknown. We performed the first metabolic analysis of myocardial tissue from BTHS cardiomyopathy patients compared to age- and sex-matched patients with idiopathic dilated cardiomyopathy (DCM) and nonfailing controls. Results corroborate previous evidence for deficiencies in cardiolipin content and its linoleoyl enrichment as defining features of BTHS cardiomyopathy, and reveal a dramatic accumulation of hydrolyzed (monolyso-) cardiolipin molecular species. Respiratory chain protein deficiencies were observed in both BTHS and DCM, but a selective depletion of complex I was seen only in BTHS after controlling for an apparent loss of mitochondrial density in cardiomyopathic hearts. Distinct shifts in the expression of long-chain fatty acid oxidation enzymes and the tissue acyl-CoA profile of BTHS hearts suggest a specific block in mitochondrial fatty acid oxidation upstream of the conventional matrix beta-oxidation cycle, which may be compensated for by a greater reliance upon peroxisomal fatty acid oxidation and the catabolism of ketones, amino acids, and pyruvate to meet cardiac energy demands. These results provide a comprehensive foundation for exploring novel therapeutic strategies that target the adaptive and maladaptive metabolic features of BTHS cardiomyopathy.
Collapse
Affiliation(s)
- Kathryn C Chatfield
- Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital of Colorado, Aurora, Colorado, USA
| | - Genevieve C Sparagna
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kalyn S Specht
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Luke A Whitcomb
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Asma K Omar
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Shelley D Miyamoto
- Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital of Colorado, Aurora, Colorado, USA
| | - Lisa M Wolfe
- Proteomics and Metabolomics Facility, Colorado State University, Fort Collins, Colorado, USA
| | - Adam J Chicco
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
126
|
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
|
127
|
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in the general population. Energy metabolism disturbance is one of the early abnormalities in CVDs, such as coronary heart disease, diabetic cardiomyopathy, and heart failure. To explore the role of myocardial energy homeostasis disturbance in CVDs, it is important to understand myocardial metabolism in the normal heart and their function in the complex pathophysiology of CVDs. In this article, we summarized lipid metabolism/lipotoxicity and glucose metabolism/insulin resistance in the heart, focused on the metabolic regulation during neonatal and ageing heart, proposed potential metabolic mechanisms for cardiac regeneration and degeneration. We provided an overview of emerging molecular network among cardiac proliferation, regeneration, and metabolic disturbance. These novel targets promise a new era for the treatment of CVDs.
Collapse
Affiliation(s)
- Lu-Yun WANG
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| | - Chen CHEN
- Division of Cardiology, Tongji Hospital, Tongji Medical College and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
128
|
Montaigne D, Butruille L, Staels B. PPAR control of metabolism and cardiovascular functions. Nat Rev Cardiol 2021; 18:809-823. [PMID: 34127848 DOI: 10.1038/s41569-021-00569-6] [Citation(s) in RCA: 359] [Impact Index Per Article: 119.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 12/22/2022]
Abstract
Peroxisome proliferator-activated receptor-α (PPARα), PPARδ and PPARγ are transcription factors that regulate gene expression following ligand activation. PPARα increases cellular fatty acid uptake, esterification and trafficking, and regulates lipoprotein metabolism genes. PPARδ stimulates lipid and glucose utilization by increasing mitochondrial function and fatty acid desaturation pathways. By contrast, PPARγ promotes fatty acid uptake, triglyceride formation and storage in lipid droplets, thereby increasing insulin sensitivity and glucose metabolism. PPARs also exert antiatherogenic and anti-inflammatory effects on the vascular wall and immune cells. Clinically, PPARγ activation by glitazones and PPARα activation by fibrates reduce insulin resistance and dyslipidaemia, respectively. PPARs are also physiological master switches in the heart, steering cardiac energy metabolism in cardiomyocytes, thereby affecting pathological heart failure and diabetic cardiomyopathy. Novel PPAR agonists in clinical development are providing new opportunities in the management of metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- David Montaigne
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Laura Butruille
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Bart Staels
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| |
Collapse
|
129
|
Dong S, Qian L, Cheng Z, Chen C, Wang K, Hu S, Zhang X, Wu T. Lactate and Myocadiac Energy Metabolism. Front Physiol 2021; 12:715081. [PMID: 34483967 PMCID: PMC8415870 DOI: 10.3389/fphys.2021.715081] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/29/2021] [Indexed: 12/05/2022] Open
Abstract
The myocardium is capable of utilizing different energy substrates, which is referred to as “metabolic flexibility.” This process assures ATP production from fatty acids, glucose, lactate, amino acids, and ketones, in the face of varying metabolic contexts. In the normal physiological state, the oxidation of fatty acids contributes to approximately 60% of energy required, and the oxidation of other substrates provides the rest. The accumulation of lactate in ischemic and hypoxic tissues has traditionally be considered as a by-product, and of little utility. However, recent evidence suggests that lactate may represent an important fuel for the myocardium during exercise or myocadiac stress. This new paradigm drives increasing interest in understanding its role in cardiac metabolism under both physiological and pathological conditions. In recent years, blood lactate has been regarded as a signal of stress in cardiac disease, linking to prognosis in patients with myocardial ischemia or heart failure. In this review, we discuss the importance of lactate as an energy source and its relevance to the progression and management of heart diseases.
Collapse
Affiliation(s)
- Shuohui Dong
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Linhui Qian
- Department of Colorectal and Anal Surgery, Feicheng Hospital Affiliated to Shandong First Medical University, Feicheng, China
| | - Zhiqiang Cheng
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Chang Chen
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Kexin Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Sanyuan Hu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Xiang Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Tongzhi Wu
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, Australia.,Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
130
|
Fulghum KL, Audam TN, Lorkiewicz PK, Zheng Y, Merchant M, Cummins TD, Dean WL, Cassel TA, Fan TWM, Hill BG. In vivo deep network tracing reveals phosphofructokinase-mediated coordination of biosynthetic pathway activity in the myocardium. J Mol Cell Cardiol 2021; 162:32-42. [PMID: 34487754 PMCID: PMC8766935 DOI: 10.1016/j.yjmcc.2021.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/23/2021] [Accepted: 08/27/2021] [Indexed: 12/22/2022]
Abstract
Glucose metabolism comprises numerous amphibolic metabolites that provide precursors for not only the synthesis of cellular building blocks but also for ATP production. In this study, we tested how phosphofructokinase-1 (PFK1) activity controls the fate of glucose-derived carbon in murine hearts in vivo. PFK1 activity was regulated by cardiac-specific overexpression of kinase- or phosphatase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase transgenes in mice (termed GlycoLo or GlycoHi mice, respectively). Dietary delivery of 13C6-glucose to these mice, followed by deep network metabolic tracing, revealed that low rates of PFK1 activity promote selective routing of glucose-derived carbon to the purine synthesis pathway to form 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR). Consistent with a mechanism of physical channeling, we found multimeric protein complexes that contained phosphoribosylaminoimidazole carboxylase (PAICS)—an enzyme important for AICAR biosynthesis, as well as chaperone proteins such as Hsp90 and other metabolic enzymes. We also observed that PFK1 influenced glucose-derived carbon deposition in glycogen, but did not affect hexosamine biosynthetic pathway activity. These studies demonstrate the utility of deep network tracing to identify metabolic channeling and changes in biosynthetic pathway activity in the heart in vivo and present new potential mechanisms by which metabolic branchpoint reactions modulate biosynthetic pathways.
Collapse
Affiliation(s)
- Kyle L Fulghum
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, United States of America; Department of Physiology, University of Louisville, Louisville, KY, United States of America
| | - Timothy N Audam
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, United States of America; Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY, United States of America
| | - Pawel K Lorkiewicz
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, United States of America; Department of Chemistry, University of Louisville, Louisville, KY, United States of America
| | - Yuting Zheng
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Michael Merchant
- Division of Nephrology, Department of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Timothy D Cummins
- Division of Nephrology, Department of Medicine, University of Louisville, Louisville, KY, United States of America
| | - William L Dean
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, United States of America
| | - Teresa A Cassel
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States of America
| | - Teresa W M Fan
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States of America
| | - Bradford G Hill
- Diabetes and Obesity Center, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, United States of America.
| |
Collapse
|
131
|
Fernández-Ruiz I. Rebalancing the pyruvate-lactate axis to treat heart failure. Nat Rev Cardiol 2021; 18:150-151. [PMID: 33462418 DOI: 10.1038/s41569-021-00513-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
132
|
Li Q, Li C, Elnwasany A, Sharma G, An YA, Zhang G, Elhelaly WM, Lin J, Gong Y, Chen G, Wang M, Zhao S, Dai C, Smart CD, Liu J, Luo X, Deng Y, Tan L, Lv SJ, Davidson SM, Locasale JW, Lorenzi PL, Malloy CR, Gillette TG, Vander Heiden MG, Scherer PE, Szweda LI, Fu G, Wang ZV. PKM1 Exerts Critical Roles in Cardiac Remodeling Under Pressure Overload in the Heart. Circulation 2021; 144:712-727. [PMID: 34102853 PMCID: PMC8405569 DOI: 10.1161/circulationaha.121.054885] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Metabolic remodeling precedes most alterations during cardiac hypertrophic growth under hemodynamic stress. The elevation of glucose utilization has been recognized as a hallmark of metabolic remodeling. However, its role in cardiac hypertrophic growth and heart failure in response to pressure overload remains to be fully illustrated. Here, we aimed to dissect the role of cardiac PKM1 (pyruvate kinase muscle isozyme 1) in glucose metabolic regulation and cardiac response under pressure overload. METHODS Cardiac-specific deletion of PKM1 was achieved by crossing the floxed PKM1 mouse model with the cardiomyocyte-specific Cre transgenic mouse. PKM1 transgenic mice were generated under the control of tetracycline response elements, and cardiac-specific overexpression of PKM1 was induced by doxycycline administration in adult mice. Pressure overload was triggered by transverse aortic constriction. Primary neonatal rat ventricular myocytes were used to dissect molecular mechanisms. Moreover, metabolomics and nuclear magnetic resonance spectroscopy analyses were conducted to determine cardiac metabolic flux in response to pressure overload. RESULTS We found that PKM1 expression is reduced in failing human and mouse hearts. It is important to note that cardiomyocyte-specific deletion of PKM1 exacerbates cardiac dysfunction and fibrosis in response to pressure overload. Inducible overexpression of PKM1 in cardiomyocytes protects the heart against transverse aortic constriction-induced cardiomyopathy and heart failure. At the mechanistic level, PKM1 is required for the augmentation of glycolytic flux, mitochondrial respiration, and ATP production under pressure overload. Furthermore, deficiency of PKM1 causes a defect in cardiomyocyte growth and a decrease in pyruvate dehydrogenase complex activity at both in vitro and in vivo levels. CONCLUSIONS These findings suggest that PKM1 plays an essential role in maintaining a homeostatic response in the heart under hemodynamic stress.
Collapse
Affiliation(s)
- Qinfeng Li
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chao Li
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Abdallah Elnwasany
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Gaurav Sharma
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yu A. An
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Guangyu Zhang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Waleed M. Elhelaly
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jun Lin
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yingchao Gong
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Guihao Chen
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Meihui Wang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Shangang Zhao
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chongshan Dai
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Charles D. Smart
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Juan Liu
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Xiang Luo
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yingfeng Deng
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shuang-Jie Lv
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shawn M. Davidson
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jason W. Locasale
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Philip L. Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Craig R. Malloy
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas G. Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Matthew G. Vander Heiden
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Luke I. Szweda
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhao V. Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
133
|
Heinrich T, Sala-Hojman A, Ferretti R, Petersson C, Minguzzi S, Gondela A, Ramaswamy S, Bartosik A, Czauderna F, Crowley L, Wahra P, Schilke H, Böpple P, Dudek Ł, Leś M, Niedziejko P, Olech K, Pawlik H, Włoszczak Ł, Zuchowicz K, Suarez Alvarez JR, Martyka J, Sitek E, Mikulski M, Szczęśniak J, Jäckel S, Krier M, Król M, Wegener A, Gałęzowski M, Nowak M, Becker F, Herhaus C. Discovery of 5-{2-[5-Chloro-2-(5-ethoxyquinoline-8-sulfonamido)phenyl]ethynyl}-4-methoxypyridine-2-carboxylic Acid, a Highly Selective in Vivo Useable Chemical Probe to Dissect MCT4 Biology. J Med Chem 2021; 64:11904-11933. [PMID: 34382802 DOI: 10.1021/acs.jmedchem.1c00448] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Due to increased lactate production during glucose metabolism, tumor cells heavily rely on efficient lactate transport to avoid intracellular lactate accumulation and acidification. Monocarboxylate transporter 4 (MCT4/SLC16A3) is a lactate transporter that plays a central role in tumor pH modulation. The discovery and optimization of a novel class of MCT4 inhibitors (hit 9a), identified by a cellular screening in MDA-MB-231, is described. Direct target interaction of the optimized compound 18n with the cytosolic domain of MCT4 was shown after solubilization of the GFP-tagged transporter by fluorescence cross-correlation spectroscopy and microscopic studies. In vitro treatment with 18n resulted in lactate efflux inhibition and reduction of cellular viability in MCT4 high expressing cells. Moreover, pharmacokinetic properties of 18n allowed assessment of lactate modulation and antitumor activity in a mouse tumor model. Thus, 18n represents a valuable tool for investigating selective MCT4 inhibition and its effect on tumor biology.
Collapse
Affiliation(s)
- Timo Heinrich
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Ada Sala-Hojman
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Roberta Ferretti
- EMD Serono Research & Development Institute, Inc., 45A Middlesex Turnpike, Billerica, Massachusetts 01821, United States
| | - Carl Petersson
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Stefano Minguzzi
- Intana, Bioscience GmbH, Lochhamer Str. 29a, 82152 Planegg, Martinsried, Germany
| | | | - Shivapriya Ramaswamy
- EMD Serono Research & Development Institute, Inc., 45A Middlesex Turnpike, Billerica, Massachusetts 01821, United States
| | - Anna Bartosik
- Ryvu Therapeutics, Sternbacha 2, 30-394 Kraków, Poland
| | - Frank Czauderna
- EMD Serono Research & Development Institute, Inc., 45A Middlesex Turnpike, Billerica, Massachusetts 01821, United States
| | - Lindsey Crowley
- EMD Serono Research & Development Institute, Inc., 45A Middlesex Turnpike, Billerica, Massachusetts 01821, United States
| | - Pamela Wahra
- EMD Serono Research & Development Institute, Inc., 45A Middlesex Turnpike, Billerica, Massachusetts 01821, United States
| | - Heike Schilke
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Pia Böpple
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Łukasz Dudek
- Ryvu Therapeutics, Sternbacha 2, 30-394 Kraków, Poland
| | - Marcin Leś
- Ryvu Therapeutics, Sternbacha 2, 30-394 Kraków, Poland
| | | | - Kamila Olech
- Ryvu Therapeutics, Sternbacha 2, 30-394 Kraków, Poland
| | - Henryk Pawlik
- Ryvu Therapeutics, Sternbacha 2, 30-394 Kraków, Poland
| | | | | | | | | | - Ewa Sitek
- Ryvu Therapeutics, Sternbacha 2, 30-394 Kraków, Poland
| | | | | | - Sven Jäckel
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Mireille Krier
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Marcin Król
- Ryvu Therapeutics, Sternbacha 2, 30-394 Kraków, Poland
| | - Ansgar Wegener
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | | | - Mateusz Nowak
- Ryvu Therapeutics, Sternbacha 2, 30-394 Kraków, Poland
| | - Frank Becker
- Intana, Bioscience GmbH, Lochhamer Str. 29a, 82152 Planegg, Martinsried, Germany
| | - Christian Herhaus
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| |
Collapse
|
134
|
Cable J, Pourquié O, Wellen KE, Finley LWS, Aulehla A, Gould AP, Teleman A, Tu WB, Garrett WS, Miguel-Aliaga I, Perrimon N, Hooper LV, Walhout AJM, Wei W, Alexandrov T, Erez A, Ralser M, Rabinowitz JD, Hemalatha A, Gutiérrez-Pérez P, Chandel NS, Rutter J, Locasale JW, Landoni JC, Christofk H. Metabolic decisions in development and disease-a Keystone Symposia report. Ann N Y Acad Sci 2021; 1506:55-73. [PMID: 34414571 DOI: 10.1111/nyas.14678] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 07/31/2021] [Indexed: 12/11/2022]
Abstract
There is an increasing appreciation for the role of metabolism in cell signaling and cell decision making. Precise metabolic control is essential in development, as evident by the disorders caused by mutations in metabolic enzymes. The metabolic profile of cells is often cell-type specific, changing as cells differentiate or during tumorigenesis. Recent evidence has shown that changes in metabolism are not merely a consequence of changes in cell state but that metabolites can serve to promote and/or inhibit these changes. Metabolites can link metabolic pathways with cell signaling pathways via several mechanisms, for example, by serving as substrates for protein post-translational modifications, by affecting enzyme activity via allosteric mechanisms, or by altering epigenetic markers. Unraveling the complex interactions governing metabolism, gene expression, and protein activity that ultimately govern a cell's fate will require new tools and interactions across disciplines. On March 24 and 25, 2021, experts in cell metabolism, developmental biology, and human disease met virtually for the Keystone eSymposium, "Metabolic Decisions in Development and Disease." The discussions explored how metabolites impact cellular and developmental decisions in a diverse range of model systems used to investigate normal development, developmental disorders, dietary effects, and cancer-mediated changes in metabolism.
Collapse
Affiliation(s)
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, Massachusetts.,Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Stem Cell Institute, Boston, Massachusetts
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Lydia W S Finley
- Cell Biology Program and Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander Aulehla
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Aurelio Teleman
- German Cancer Research Center (DKFZ) and Heidelberg University, Heidelberg, Germany
| | - William B Tu
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Wendy Sarah Garrett
- Harvard T. H. Chan School of Public Health and Dana-Farber Cancer, Boston, Massachusetts
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences and Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Howard Hughes Institute, Boston, Massachusetts
| | - Lora V Hooper
- Department of Immunology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - A J Marian Walhout
- Program in Systems Biology and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Wei Wei
- Department of Pathology, Stanford University School of Medicine, Stanford, California.,Department of Biology and Stanford ChEM-H, Stanford University, Stanford, California
| | - Theodore Alexandrov
- Structural and Computational Biology Unit and Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Markus Ralser
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK.,Department of Biochemistry, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Joshua D Rabinowitz
- Department of Chemistry and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey
| | - Anupama Hemalatha
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
| | - Paula Gutiérrez-Pérez
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Navdeep S Chandel
- Department of Medicine, Robert H. Lurie Cancer Center, Chicago, Illinois.,Department of Biochemistry and Molecular Genetics, Robert H. Lurie Cancer Center Metabolomics Core, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jared Rutter
- Department of Biochemistry and Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Juan C Landoni
- Research Program in Stem Cells and Metabolism, University of Helsinki, Helsinki, Finland
| | - Heather Christofk
- Departments of Biological Chemistry and Molecular & Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
135
|
With a grain of salt: Sodium elevation and metabolic remodelling in heart failure. J Mol Cell Cardiol 2021; 161:106-115. [PMID: 34371034 PMCID: PMC7611640 DOI: 10.1016/j.yjmcc.2021.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/09/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022]
Abstract
Elevated intracellular Na (Nai) and metabolic impairment are interrelated pathophysiological features of the failing heart (HF). There have been a number of studies showing that myocardial sodium elevation subtly affects mitochondrial function. During contraction, mitochondrial calcium (Camito) stimulates a variety of TCA cycle enzymes, thereby providing reducing equivalents to maintain ATP supply. Nai elevation has been shown to impact Camito; however, whether metabolic remodelling in HF is caused by increased Nai has only been recently demonstrated. This novel insight may help to elucidate the contribution of metabolic remodelling in the pathophysiology of HF, the lack of efficacy of current HF therapies and a rationale for the development of future metabolism-targeting treatments. Here we review the relationship between Na pump inhibition, elevated Nai, and altered metabolic profile in the context of HF and their link to metabolic (in)flexibility and mitochondrial reprogramming.
Collapse
|
136
|
Wang C, Wang Y, Shen L. Mitochondrial proteins in heart failure: The role of deacetylation by SIRT3. Pharmacol Res 2021; 172:105802. [PMID: 34363948 DOI: 10.1016/j.phrs.2021.105802] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/28/2022]
Abstract
Heart failure (HF) is still the leading cause of death worldwide, occurring with a variety of complex mechanisms. However, most intervention for HF do not directly target the pathological mechanisms underlying cell damage in failing cardiomyocytes. Mitochondria are involved in many physiological processes, which is an important guarantee for normal heart function. Mitochondrial dysfunction is considered to be the critical node of the development of HF. Strict modulation of the mitochondrial function can ameliorate the myocardial injury and protect cardiac function. Acetylation plays an important role in mitochondrial protein homeostasis, and SIRT3, the most important deacetylation protein in mitochondria, is involved in the maintenance of mitochondrial function. SIRT3 can delay the progression of HF by improving mitochondrial function. Herein we summarize the interaction between SIRT3 and proteins related to mitochondrial function including oxidative phosphorylation (OXPHOS), fatty acid oxidation (FAO), mitochondrial biosynthesis, mitochondrial quality control. In addition, we also sum up the effects of this interaction on HF and the research progress of treatments targeting SIRT3, so as to find potential HF therapeutic for clinical use in the future.
Collapse
Affiliation(s)
- Chunfang Wang
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renming Road, Changsha, Hunan 410011, PR China.
| | - Yating Wang
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renming Road, Changsha, Hunan 410011, PR China.
| | - Li Shen
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renming Road, Changsha, Hunan 410011, PR China.
| |
Collapse
|
137
|
Karlstaedt A, Barrett M, Hu R, Gammons ST, Ky B. Cardio-Oncology: Understanding the Intersections Between Cardiac Metabolism and Cancer Biology. JACC Basic Transl Sci 2021; 6:705-718. [PMID: 34466757 PMCID: PMC8385559 DOI: 10.1016/j.jacbts.2021.05.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/21/2021] [Accepted: 05/23/2021] [Indexed: 12/24/2022]
Abstract
An important priority in the cardiovascular care of oncology patients is to reduce morbidity and mortality, and improve the quality of life in cancer survivors through cross-disciplinary efforts. The rate of survival in cancer patients has improved dramatically over the past decades. Nonetheless, survivors may be more likely to die from cardiovascular disease in the long term, secondary, not only to the potential toxicity of cancer therapeutics, but also to the biology of cancer. In this context, efforts from basic and translational studies are crucial to understanding the molecular mechanisms causal to cardiovascular disease in cancer patients and survivors, and identifying new therapeutic targets that may prevent and treat both diseases. This review aims to highlight our current understanding of the metabolic interaction between cancer and the heart, including potential therapeutic targets. An overview of imaging techniques that can support both research studies and clinical management is also provided. Finally, this review highlights opportunities and challenges that are necessary to advance our understanding of metabolism in the context of cardio-oncology.
Collapse
Key Words
- 99mTc-MIBI, 99mtechnetium-sestamibi
- CVD, cardiovascular disease
- D2-HG, D-2-hydroxyglutarate
- FAO, fatty acid oxidation
- FASN, fatty acid synthase
- GLS, glutaminase
- HF, heart failure
- IDH, isocitrate dehydrogenase
- IGF, insulin-like growth factor
- MCT1, monocarboxylate transporter 1
- MRS, magnetic resonance spectroscopy
- PDH, pyruvate dehydrogenase
- PET, positron emission tomography
- PI3K, insulin-activated phosphoinositide-3-kinase
- PTM, post-translational modification
- SGLT2, sodium glucose co-transporter 2
- TRF, time-restricted feeding
- [18F]FDG, 2-deoxy-2-[fluorine-18]fluoro-D-glucose
- cancer
- cardio-oncology
- heart failure
- metabolism
- oncometabolism
- α-KG, α-ketoglutarate
Collapse
Affiliation(s)
- Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Matthew Barrett
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ray Hu
- Departments of Medicine and Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Seth Thomas Gammons
- Department of Cancer Systems Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Bonnie Ky
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Departments of Medicine and Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
138
|
Shankar TS, Ramadurai DKA, Steinhorst K, Sommakia S, Badolia R, Thodou Krokidi A, Calder D, Navankasattusas S, Sander P, Kwon OS, Aravamudhan A, Ling J, Dendorfer A, Xie C, Kwon O, Cheng EHY, Whitehead KJ, Gudermann T, Richardson RS, Sachse FB, Schredelseker J, Spitzer KW, Chaudhuri D, Drakos SG. Cardiac-specific deletion of voltage dependent anion channel 2 leads to dilated cardiomyopathy by altering calcium homeostasis. Nat Commun 2021; 12:4583. [PMID: 34321484 PMCID: PMC8319341 DOI: 10.1038/s41467-021-24869-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/06/2021] [Indexed: 12/13/2022] Open
Abstract
Voltage dependent anion channel 2 (VDAC2) is an outer mitochondrial membrane porin known to play a significant role in apoptosis and calcium signaling. Abnormalities in calcium homeostasis often leads to electrical and contractile dysfunction and can cause dilated cardiomyopathy and heart failure. However, the specific role of VDAC2 in intracellular calcium dynamics and cardiac function is not well understood. To elucidate the role of VDAC2 in calcium homeostasis, we generated a cardiac ventricular myocyte-specific developmental deletion of Vdac2 in mice. Our results indicate that loss of VDAC2 in the myocardium causes severe impairment in excitation-contraction coupling by altering both intracellular and mitochondrial calcium signaling. We also observed adverse cardiac remodeling which progressed to severe cardiomyopathy and death. Reintroduction of VDAC2 in 6-week-old knock-out mice partially rescued the cardiomyopathy phenotype. Activation of VDAC2 by efsevin increased cardiac contractile force in a mouse model of pressure-overload induced heart failure. In conclusion, our findings demonstrate that VDAC2 plays a crucial role in cardiac function by influencing cellular calcium signaling. Through this unique role in cellular calcium dynamics and excitation-contraction coupling VDAC2 emerges as a plausible therapeutic target for heart failure.
Collapse
Affiliation(s)
- Thirupura S Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Dinesh K A Ramadurai
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Kira Steinhorst
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Salah Sommakia
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Rachit Badolia
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Aspasia Thodou Krokidi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Dallen Calder
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Sutip Navankasattusas
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Paulina Sander
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Oh Sung Kwon
- Department of Kinesiology, University of Connecticut, Storrs, CT, USA
- Geriatric Research, Education, and Clinical Center, Salt Lake City VA Medical Center, Salt Lake City, UT, USA
| | - Aishwarya Aravamudhan
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Jing Ling
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Andreas Dendorfer
- Walter-Brendel-Center of Experimental Medicine, Ludwig-Maximilians Universität Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Changmin Xie
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Ohyun Kwon
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | | | - Kevin J Whitehead
- Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Russel S Richardson
- Geriatric Research, Education, and Clinical Center, Salt Lake City VA Medical Center, Salt Lake City, UT, USA
| | - Frank B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Johann Schredelseker
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Kenneth W Spitzer
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA
- Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Stavros G Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, USA.
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
- Division of Cardiovascular Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA.
| |
Collapse
|
139
|
Abstract
Insulin receptors are highly expressed in the heart and vasculature. Insulin signaling regulates cardiac growth, survival, substrate uptake, utilization, and mitochondrial metabolism. Insulin signaling modulates the cardiac responses to physiological and pathological stressors. Altered insulin signaling in the heart may contribute to the pathophysiology of ventricular remodeling and heart failure progression. Myocardial insulin signaling adapts rapidly to changes in the systemic metabolic milieu. What may initially represent an adaptation to protect the heart from carbotoxicity may contribute to amplifying the risk of heart failure in obesity and diabetes. This review article presents the multiple roles of insulin signaling in cardiac physiology and pathology and discusses the potential therapeutic consequences of modulating myocardial insulin signaling.
Collapse
Affiliation(s)
- E Dale Abel
- Division of Endocrinology, Metabolism and Diabetes and Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
140
|
Monocarboxylate transporter antagonism reveals metabolic vulnerabilities of viral-driven lymphomas. Proc Natl Acad Sci U S A 2021; 118:2022495118. [PMID: 34161263 PMCID: PMC8237662 DOI: 10.1073/pnas.2022495118] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Epstein-Barr virus (EBV) is a ubiquitous herpesvirus that typically causes asymptomatic infection but can promote B lymphoid tumors in the immune suppressed. In vitro, EBV infection of primary B cells stimulates glycolysis during immortalization into lymphoblastoid cell lines (LCLs). Lactate export during glycolysis is crucial for continued proliferation of many cancer cells-part of a phenomenon known as the "Warburg effect"- and is mediated by monocarboxylate transporters (MCTs). However, the role of MCTs has yet to be studied in EBV-associated malignancies, which display Warburg-like metabolism in vitro. Here, we show that EBV infection of B lymphocytes directly promotes temporal induction of MCT1 and MCT4 through the viral proteins EBNA2 and LMP1, respectively. Functionally, MCT1 was required for early B cell proliferation, and MCT4 up-regulation promoted acquired resistance to MCT1 antagonism in LCLs. However, dual MCT1/4 inhibition led to LCL growth arrest and lactate buildup. Metabolic profiling in LCLs revealed significantly reduced oxygen consumption rates (OCRs) and NAD+/NADH ratios, contrary to previous observations of increased OCR and unaltered NAD+/NADH ratios in MCT1/4-inhibited cancer cells. Furthermore, U-13C6-glucose labeling of MCT1/4-inhibited LCLs revealed depleted glutathione pools that correlated with elevated reactive oxygen species. Finally, we found that dual MCT1/4 inhibition also sensitized LCLs to killing by the electron transport chain complex I inhibitors phenformin and metformin. These findings were extended to viral lymphomas associated with EBV and the related gammaherpesvirus KSHV, pointing at a therapeutic approach for targeting both viral lymphomas.
Collapse
|
141
|
Garde A, Sherwood DR. Fueling Cell Invasion through Extracellular Matrix. Trends Cell Biol 2021; 31:445-456. [PMID: 33549396 PMCID: PMC8122022 DOI: 10.1016/j.tcb.2021.01.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/07/2021] [Accepted: 01/14/2021] [Indexed: 01/10/2023]
Abstract
Cell invasion through extracellular matrix (ECM) has pivotal roles in cell dispersal during development, immune cell trafficking, and cancer metastasis. Many elegant studies have revealed the specialized cellular protrusions, proteases, and distinct modes of migration invasive cells use to overcome ECM barriers. Less clear, however, is how invasive cells provide energy, specifically ATP, to power the energetically demanding membrane trafficking, F-actin polymerization, and actomyosin machinery that mediate break down, remodeling, and movement through ECMs. Here, we provide an overview of the challenges of examining ATP generation and delivery within invading cells and how recent studies using diverse invasion models, experimental approaches, and energy biosensors are revealing that energy metabolism is an integral component of cell invasive behavior that is dynamically tuned to overcome the ECM environment.
Collapse
Affiliation(s)
- Aastha Garde
- Department of Cell Biology, Duke University, Box 3709, Durham, NC 27710, USA
| | - David R Sherwood
- Department of Biology, Duke University, Box 90338, Durham, NC 27708, USA; Regeneration Next, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
142
|
Abstract
Alterations in cardiac energy metabolism contribute to the severity of heart failure. However, the energy metabolic changes that occur in heart failure are complex and are dependent not only on the severity and type of heart failure present but also on the co-existence of common comorbidities such as obesity and type 2 diabetes. The failing heart faces an energy deficit, primarily because of a decrease in mitochondrial oxidative capacity. This is partly compensated for by an increase in ATP production from glycolysis. The relative contribution of the different fuels for mitochondrial ATP production also changes, including a decrease in glucose and amino acid oxidation, and an increase in ketone oxidation. The oxidation of fatty acids by the heart increases or decreases, depending on the type of heart failure. For instance, in heart failure associated with diabetes and obesity, myocardial fatty acid oxidation increases, while in heart failure associated with hypertension or ischemia, myocardial fatty acid oxidation decreases. Combined, these energy metabolic changes result in the failing heart becoming less efficient (ie, a decrease in cardiac work/O2 consumed). The alterations in both glycolysis and mitochondrial oxidative metabolism in the failing heart are due to both transcriptional changes in key enzymes involved in these metabolic pathways, as well as alterations in NAD redox state (NAD+ and nicotinamide adenine dinucleotide levels) and metabolite signaling that contribute to posttranslational epigenetic changes in the control of expression of genes encoding energy metabolic enzymes. Alterations in the fate of glucose, beyond flux through glycolysis or glucose oxidation, also contribute to the pathology of heart failure. Of importance, pharmacological targeting of the energy metabolic pathways has emerged as a novel therapeutic approach to improving cardiac efficiency, decreasing the energy deficit and improving cardiac function in the failing heart.
Collapse
Affiliation(s)
- Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada (G.D.L., Q.G.K.)
| | - Qutuba G Karwi
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada (G.D.L., Q.G.K.)
| | - Rong Tian
- Mitochondria and Metabolism Center, University of Washington, Seattle (R.T.)
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.)
| | - E Dale Abel
- Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City (E.D.A.).,Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City (E.D.A.)
| |
Collapse
|
143
|
Ruiz-Iglesias A, Mañes S. The Importance of Mitochondrial Pyruvate Carrier in Cancer Cell Metabolism and Tumorigenesis. Cancers (Basel) 2021; 13:cancers13071488. [PMID: 33804985 PMCID: PMC8037430 DOI: 10.3390/cancers13071488] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The characteristic metabolic hallmark of cancer cells is the massive catabolism of glucose by glycolysis, even under aerobic conditions—the so-called Warburg effect. Although energetically unfavorable, glycolysis provides “building blocks” to sustain the unlimited growth of malignant cells. Aberrant glycolysis is also responsible for lactate accumulation and acidosis in the tumor milieu, which fosters hypoxia and immunosuppression. One of the mechanisms used by cancer cells to increase glycolytic flow is the negative regulation of the proteins that conform the mitochondrial pyruvate carrier (MPC) complex, which transports pyruvate into the mitochondrial matrix to be metabolized in the tricarboxylic acid (TCA) cycle. Evidence suggests that MPC downregulation in tumor cells impacts many aspects of tumorigenesis, including cancer cell-intrinsic (proliferation, invasiveness, stemness, resistance to therapy) and -extrinsic (angiogenesis, anti-tumor immune activity) properties. In many cancers, but not in all, MPC downregulation is associated with poor survival. MPC regulation is therefore central to tackling glycolysis in tumors. Abstract Pyruvate is a key molecule in the metabolic fate of mammalian cells; it is the crossroads from where metabolism proceeds either oxidatively or ends with the production of lactic acid. Pyruvate metabolism is regulated by many enzymes that together control carbon flux. Mitochondrial pyruvate carrier (MPC) is responsible for importing pyruvate from the cytosol to the mitochondrial matrix, where it is oxidatively phosphorylated to produce adenosine triphosphate (ATP) and to generate intermediates used in multiple biosynthetic pathways. MPC activity has an important role in glucose homeostasis, and its alteration is associated with diabetes, heart failure, and neurodegeneration. In cancer, however, controversy surrounds MPC function. In some cancers, MPC upregulation appears to be associated with a poor prognosis. However, most transformed cells undergo a switch from oxidative to glycolytic metabolism, the so-called Warburg effect, which, amongst other possibilities, is induced by MPC malfunction or downregulation. Consequently, impaired MPC function might induce tumors with strong proliferative, migratory, and invasive capabilities. Moreover, glycolytic cancer cells secrete lactate, acidifying the microenvironment, which in turn induces angiogenesis, immunosuppression, and the expansion of stromal cell populations supporting tumor growth. This review examines the latest findings regarding the tumorigenic processes affected by MPC.
Collapse
|