1
|
Zhao Z, Sun Y, Jia M, Jiang M, Ruan X. Prophylactic administration of Kanli granule maintains fatty acid oxidation in the myocardium to prevent heart failure via activating AMPK/PPARα/CPT1A pathway: A network pharmacology-based study. Fitoterapia 2025; 184:106633. [PMID: 40398514 DOI: 10.1016/j.fitote.2025.106633] [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: 02/25/2025] [Revised: 04/18/2025] [Accepted: 05/17/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Abnormal energy metabolism plays a crucial role in the pathogenesis of heart failure (HF). Kanli granule (KLG), as an effective herbal medicine for treating HF, has been used in clinical practice for nearly 30 years. However, its underlying mechanisms have not been fully elucidated. METHODS This study combined network pharmacology, molecular docking, and in vivo experiments to explore KLG's effect on HF. Wistar rats with AAC-induced HF were orally administered KLG (0.675/1.35/2.7 g/kg) for 32 weeks. Assessments included heart weight index, echocardiography, histopathology, fatty acid metabolism (FAM) targets, and myocardial energy metrics. We focused on fatty acid oxidation (FAO) pathway, measuring AMPK, PPARα, and CPT1A at protein and gene levels. RESULTS KLG maintained cardiac function in AAC rats. Network pharmacology identified PPAR and AMPK pathways as key in FAM. Molecular docking showed strong affinity of KLG components to FAO targets PPARα and CPT1A. KLG significantly enhanced myocardial energy metabolism, reduced myocardial FFA levels, and increased ATP/ADP ratios. It activated AMPK and upregulated FAO-related genes, including PPARα and CPT1A. CONCLUSION KLG improves FAO in AAC-induced HF rats by activating the AMPK/PPARα/CPT1A pathway, reducing myocardial FFA levels, and improving myocardial microstructure and cardiac function.
Collapse
Affiliation(s)
- Zhejun Zhao
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuanlong Sun
- Cardiovascular Department, Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Meijun Jia
- Cardiovascular Department, Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Meixian Jiang
- Cardiovascular Department, Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xiaofen Ruan
- Cardiovascular Department, Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Chindamo M, Chehade H, Sordet A, Humbert-Droz G, Cachat F, Mauduit C, Benahmed M, Simeoni U, Siddeek B. Maternal exposure to high-fat diet induces long-term mitochondrial alterations in the offspring heart. Nutrition 2025; 137:112796. [PMID: 40408919 DOI: 10.1016/j.nut.2025.112796] [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: 08/12/2024] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 05/25/2025]
Abstract
OBJECTIVES Heart disease is a leading cause of death worldwide, with its prevalence exacerbated by inadequate nutritional intake. Particularly concerning is the elevated risk induced by imbalanced nutrition during development, which can impact lifelong heart health. Recent research has underscored mitochondrial dysregulation as a pivotal mechanism driving the enduring consequences of nutritional excess. Building upon previous findings wherein a maternal high-fat diet (HFD) led to cardiac hypertrophy and fibrosis, our current study aimed to evaluate the impact of such a challenge on myocardial mitochondrial function. METHODS Female rats were fed a chow diet or HFD during gestation and lactation. The hearts of male offspring were analyzed at adulthood. Mitochondrial DNA abundance was evaluated by quantitative polymerase chain reaction. Proteins involved in mitochondrial biogenesis, fusion, fission, damage to the electron transport chain, metabolism, cell death, proliferation, and inflammation were measured by western blot. Mitochondrial clearance was evaluated by the measurement of mitophagy markers on isolated mitochondria. Lipids were visualized by histologic approaches. RESULTS We detected decreased cardiac mitochondrial fission factor and mitochondrial adenosine triphosphate synthase beta subunit and increased Parkin, pro-tumor necrosis factor alpha, and pro-interleukin 1 beta protein levels associated with decreased microtubule-associated protein 1A/1B light chain 3B levels in cardiac mitochondrial fraction, with a tendency for increased Oil Red O staining in the adult hearts of male offspring exposed to HFD. CONCLUSIONS Maternal exposure to HFD enhanced mitochondrial damage and impaired fission and clearance in offspring hearts at adulthood. These alterations were associated with altered expression of proteins involved in the mitochondrial electron transport chain coupled with a propensity for increased fatty acid accumulation and elevated proinflammatory markers.
Collapse
Affiliation(s)
- Mariapia Chindamo
- Division of Pediatrics, Woman-Mother-Child Department, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Hassib Chehade
- Division of Pediatrics, Woman-Mother-Child Department, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Anthony Sordet
- Division of Pediatrics, Woman-Mother-Child Department, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Gaël Humbert-Droz
- Division of Pediatrics, Woman-Mother-Child Department, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - François Cachat
- Division of Pediatrics, Woman-Mother-Child Department, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Claire Mauduit
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Mohamed Benahmed
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | - Umberto Simeoni
- Division of Pediatrics, Woman-Mother-Child Department, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Benazir Siddeek
- Division of Pediatrics, Woman-Mother-Child Department, Developmental Origins of Health and Disease (DOHaD) Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
3
|
Das BB. Mechanism and Treatment of Right Ventricular Failure Due to Pulmonary Hypertension in Children. CHILDREN (BASEL, SWITZERLAND) 2025; 12:476. [PMID: 40310168 PMCID: PMC12025609 DOI: 10.3390/children12040476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025]
Abstract
Pulmonary hypertension (PH) is a progressive disorder characterized by obstructive changes in the pulmonary vasculature, leading to increased pulmonary vascular resistance (PVR), right ventricular (RV) strain, and eventual RV failure (RVF). Despite advancements in medical therapy, PH remains associated with significant morbidity and mortality, particularly in children. RVF is a clinical syndrome resulting from complex structural and functional remodeling of the right heart, leading to inadequate pulmonary circulation, reduced cardiac output, and elevated venous pressure. Management paradigms for pediatric PH diverge significantly from those in adults, particularly due to the predominance of congenital heart disease (CHD) and the dynamic nature of pediatric cardiovascular and pulmonary development. CHD remains a principal driver of PH in children, and its associated pathophysiology demands a nuanced approach. In patients with unrepaired left-to-right shunts, elevated pulmonary blood flow can lead to progressive pulmonary vascular remodeling and increased PVR. The postoperative persistence or progression of PH may occur if irreversible vascular changes have already developed. Current PH treatments primarily focus on reducing PVR, yet distinguishing between therapeutic approaches that target the pulmonary vasculature and those aimed at improving RV function remain challenging. In pediatric patients with progressive PH despite optimal therapy, additional targeted interventions may be necessary to mitigate RV dysfunction and disease progression. This review provides a comprehensive analysis of the mechanisms underlying RVF in PH, incorporating insights from clinical studies in adults and experimental models, while highlighting the unique considerations in children. Furthermore, it explores current pharmacological and interventional treatment strategies, emphasizing the need for novel therapeutic approaches aimed at directly reversing RV remodeling. Given the complexities of RV adaptation in pediatric PH, further research into disease-modifying treatments and innovative interventions is crucial to improving long-term outcomes in affected children.
Collapse
Affiliation(s)
- Bibhuti B Das
- Department of Pediatrics, Division of Pediatric Cardiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
4
|
Parker LE, Papanicolaou KN, Zalesak-Kravec S, Weinberger EM, Kane MA, Foster DB. Retinoic acid signaling and metabolism in heart failure. Am J Physiol Heart Circ Physiol 2025; 328:H792-H813. [PMID: 39933792 DOI: 10.1152/ajpheart.00871.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 12/24/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Nearly 70 years after studies first showed that the offspring of vitamin A (retinol, ROL)-deficient rats exhibit structural cardiac defects and over 20 years since the role of vitamin A's potent bioactive metabolite hormone, all-trans retinoic acid (ATRA), was elucidated in embryonic cardiac development, the role of the vitamin A metabolites, or retinoids, in adult heart physiology and heart and vascular disease, remains poorly understood. Studies have shown that low serum levels of retinoic acid correlate with higher all-cause and cardiovascular mortality, though the relationship between circulating retinol and ATRA levels, cardiac tissue ATRA levels, and intracellular cardiac ATRA signaling in the context of heart and vascular disease has only begun to be addressed. We have recently shown that patients with idiopathic dilated cardiomyopathy show a nearly 40% decline of in situ cardiac ATRA levels, despite adequate local stores of retinol. Moreover, we and others have shown that the administration of ATRA forestalls the development of heart failure (HF) in rodent models. In this review, we summarize key facets of retinoid metabolism and signaling and discuss mechanisms by which impaired ATRA signaling contributes to several HF hallmarks including hypertrophy, contractile dysfunction, poor calcium handling, redox imbalance, and fibrosis. We highlight unresolved issues in cardiac ATRA metabolism whose pursuit will help refine therapeutic strategies aimed at restoring ATRA homeostasis.
Collapse
Affiliation(s)
- Lauren E Parker
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Kyriakos N Papanicolaou
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | | | - Eva M Weinberger
- School of Medicine, Imperial College London, London, United Kingdom
| | - Maureen A Kane
- School of Pharmacy, University of Maryland, Baltimore, Maryland, United States
| | - D Brian Foster
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
5
|
Wang Z, Zhu H, Xiong W. Metabolism and metabolomics in senescence, aging, and age-related diseases: a multiscale perspective. Front Med 2025; 19:200-225. [PMID: 39821730 DOI: 10.1007/s11684-024-1116-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025]
Abstract
The pursuit of healthy aging has long rendered aging and senescence captivating. Age-related ailments, such as cardiovascular diseases, diabetes, and neurodegenerative disorders, pose significant threats to individuals. Recent studies have shed light on the intricate mechanisms encompassing genetics, epigenetics, transcriptomics, and metabolomics in the processes of senescence and aging, as well as the establishment of age-related pathologies. Amidst these underlying mechanisms governing aging and related pathology metabolism assumes a pivotal role that holds promise for intervention and therapeutics. The advancements in metabolomics techniques and analysis methods have significantly propelled the study of senescence and aging, particularly with the aid of multiscale metabolomics which has facilitated the discovery of metabolic markers and therapeutic potentials. This review provides an overview of senescence and aging, emphasizing the crucial role metabolism plays in the aging process as well as age-related diseases.
Collapse
Affiliation(s)
- Ziyi Wang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hongying Zhu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| | - Wei Xiong
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| |
Collapse
|
6
|
Dunzhu D, Han G, Shanshan Q, Li S, Yang J, He J, Gou S, Dong G, Jiang C, Hou J. The role of Perilipin 5 in pathological myocardial remodeling. Front Pharmacol 2025; 16:1526494. [PMID: 40166465 PMCID: PMC11955653 DOI: 10.3389/fphar.2025.1526494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Pathological cardiac remodeling (REM), caused by various pathological factors and characterized by changes in cardiac structure and geometry, is strongly associated with heart failure (HF). It damages cardiac tissue, alters energy metabolism, increases oxidative stress, and cause matrix metalloproteinase activation, cardiomyocyte hypertrophy, and interstitial fibrosis, leading to HF. REM determines the outcome of cardiovascular disease. Current treatments have limitations. REM is associated with cardiac energetic remodeling, and modulation of metabolic substrates may slow down the disease. Perilipin 5 (Plin5), positioned as a structural protein located on the surface of lipid droplets (LDs), is abundant in tissues and cells that rely on mitochondrial β-oxidation for energy production. It is the most recently identified member of the perilipin protein (PAT) family, with a notable enrichment in the cardiac muscle. Emerging evidence highlights the critical role of intracellular LD in the regulation of energy metabolism, with metabolic disruptions of LD being directly correlated with the incidence of metabolic disease. As a key barrier to LD, Plin5 is instrumental in controlling the catabolism of LD and regulating the metabolism and transport of fatty acids (FAs). As a protectant against excessive β-oxidation of free fatty acids (FFAs), Plin5 acts to isolate and neutralize overly oxidized fatty acids, thereby shielding the heart from myocardial remodeling instigated by a variety of etiological factors. This protective mechanism helps to ameliorate the progression of persistent and detrimental myocardial remodeling, which can otherwise lead to the development of severe heart failure. This systematic review attempts to delineate the metabolic disorders associated with pathological cardiac remodeling, focusing on the properties and regulatory mechanisms of Plin5. By synthesising current literature, it investigates the pivotal role of Plin5 in modulating the distinctive attributes, initiating factors, and molecular signaling networks underpinning pathological cardiac remodeling.
Collapse
Affiliation(s)
| | - Gao Han
- School of Stomatology, Qilu Medical University, Zibo, China
| | - Qin Shanshan
- School of Medicine, Tibet University, Lhasa, China
| | - Shangshi Li
- The Department of High Mountain Sickness, The General Hospital of Xizang Military Area Command, Xizang, China
| | - Jiali Yang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jian He
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Siyu Gou
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Gang Dong
- School of Stomatology, Qilu Medical University, Zibo, China
| | - Chunrong Jiang
- School of Stomatology, Qilu Medical University, Zibo, China
| | - Jun Hou
- The Third People’s Hospital of Chengdu, Chengdu, China
| |
Collapse
|
7
|
Williams T, Groß R, Arias-Loza AP, Nordbeck P, Noerpel M, Cirnu A, Kimmel L, Ashour D, Ramos G, Waschke J, Higuchi T, Gerull B. Illuminating Cardiac Remodeling: Insights From [ 18F]-Fluorodeoxyglucose Positron Emission Tomography Imaging in Plakoglobin-Associated Arrhythmogenic Cardiomyopathy. J Am Heart Assoc 2025; 14:e038331. [PMID: 40028850 DOI: 10.1161/jaha.124.038331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 01/07/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Arrhythmogenic cardiomyopathy (ACM) is a genetic heart muscle disease, which presents with arrhythmias and sudden cardiac death, along with progressive cardiac remodeling and myocardial inflammation. This study aims to elucidate the patterns of [18F]-fluorodeoxyglucose ([18F]-FDG) uptake in a mouse model of plakoglobin-associated cardiac disease to better understand its diagnostic potential. METHODS AND RESULTS Plakoglobin (Jup) knockout mice developed a cardiomyopathy that presented an ACM-like phenotype at 6 weeks of age. Flow cytometry experiments showed a significant increase of immune cells, for example, an expansion of proinflammatory and tissue-injury macrophages. In vivo positron emission tomography and ex vivo autoradiography showed increased [18F]-FDG uptake in genotype positive hearts. A correlative analysis between [18F]-FDG positivity and macrophage infiltration using CD68 and CD206 staining did not show colocalization. CD68 and CD206 positivity was primarily observed within the fibrotic scar, whereas [18F]-FDG uptake was predominantly identified in CD68 and CD206-negative tissue areas. Instead, [18F]-FDG signal seemed to originate from cardiomyocytes adjacent to areas of fibrotic remodeling. Morphometric analysis revealed hypertrophy of these cardiomyocytes, which may reflect metabolic remodeling as a compensatory response. CONCLUSIONS In our murine model of Jup-related ACM, strong cardiac [18F]-FDG uptake was detected, which colocalized with regional hypertrophic cardiomyocytes rather than inflammatory cells. These findings indicate that [18F]-FDG positron emission tomography is a valuable tool for identifying and localizing hypermetabolic areas associated with cardiac remodeling in ACM, providing insights into disease mechanisms and potential diagnostic strategies.
Collapse
Affiliation(s)
- Tatjana Williams
- Comprehensive Heart Failure Center, Department of Cardiovascular Genetics University Hospital Würzburg Würzburg Germany
| | - Regina Groß
- Comprehensive Heart Failure Center, Department of Cardiovascular Genetics University Hospital Würzburg Würzburg Germany
| | - Anahi-Paula Arias-Loza
- Comprehensive Heart Failure Center, Department of Nuclear Medicine University Hospital Würzburg Würzburg Germany
| | - Peter Nordbeck
- Department of Internal Medicine I University Hospital Würzburg Würzburg Germany
| | - Mike Noerpel
- Comprehensive Heart Failure Center, Department of Cardiovascular Genetics University Hospital Würzburg Würzburg Germany
| | - Alexandra Cirnu
- Comprehensive Heart Failure Center, Department of Cardiovascular Genetics University Hospital Würzburg Würzburg Germany
| | - Laura Kimmel
- Comprehensive Heart Failure Center, Department of Cardiovascular Genetics University Hospital Würzburg Würzburg Germany
| | - DiyaaEldin Ashour
- Comprehensive Heart Failure Center, Immunocardiology Lab, University Hospital Würzburg Würzburg Germany
| | - Gustavo Ramos
- Comprehensive Heart Failure Center, Immunocardiology Lab, University Hospital Würzburg Würzburg Germany
| | - Jens Waschke
- Faculty of Medicine, Ludwig-Maximilians-University Munich Munich Germany
| | - Takahiro Higuchi
- Comprehensive Heart Failure Center, Department of Nuclear Medicine University Hospital Würzburg Würzburg Germany
| | - Brenda Gerull
- Comprehensive Heart Failure Center, Department of Cardiovascular Genetics University Hospital Würzburg Würzburg Germany
- Department of Internal Medicine I University Hospital Würzburg Würzburg Germany
| |
Collapse
|
8
|
Selvaraj S, Kwee LC, Thompson EJ, He M, Hornik CP, Devore AD, Patel CB, Mentz RJ, Fudim M, Taylor L, Milosovic S, Hurdle M, Cade WT, Ilkayeva O, Muehlbauer MJ, Newgard CB, Kelly DP, Zamani P, Margulies KB, Shah SH. Metabolic and Pharmacokinetic Profiling of a Ketone Ester by Background SGLT2 Inhibitor Therapy in HFrEF. JACC Basic Transl Sci 2025; 10:290-303. [PMID: 40139871 PMCID: PMC12013845 DOI: 10.1016/j.jacbts.2024.10.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 03/29/2025]
Abstract
Growing evidence supports therapeutic ketosis in heart failure with reduced ejection fraction, though uncertainty exists regarding use with SGLT2i and dose-dependent effects. In a phase I trial of 2 ketone ester (KE) doses in 20 heart failure with reduced ejection fraction participants, stratified by background SGLT2i, the authors detailed pharmacokinetic parameters, noting rapid ketosis and short half-life. KE was associated with lower non-esterified fatty acid, branched-chain amino acids, and most acylcarnitines (except C2 and C4-OH, which increased); differences were observed by SGLT2i and KE dose. Increases in heart rate and decreases in systolic blood pressure, pH, and bicarbonate were generally transient. KE ingestion induces rapid changes in key metabolic pathways, differentially affected by SGLT2i (fatty acid metabolism) and KE dose (ketone metabolism). Hemodynamic effects were transient and irrespective of dose or SGLT2i. (Ketone Pharmacokinetic Study in HFrEF; NCT05757193).
Collapse
Affiliation(s)
- Senthil Selvaraj
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Duke Molecular Physiology Institute, Durham, North Carolina, USA; Division of Cardiovascular Medicine, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | | | - Elizabeth J Thompson
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA; Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Mengshu He
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | - Christoph P Hornik
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA; Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Adam D Devore
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Chetan B Patel
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Robert J Mentz
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Marat Fudim
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Lacey Taylor
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Stephanie Milosovic
- Duke Molecular Physiology Institute, Durham, North Carolina, USA; Physical Therapy Division, Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Melissa Hurdle
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | - William T Cade
- Duke Molecular Physiology Institute, Durham, North Carolina, USA; Physical Therapy Division, Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Olga Ilkayeva
- Duke Molecular Physiology Institute, Durham, North Carolina, USA; Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | | | | - Daniel P Kelly
- Division of Cardiovascular Medicine, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Payman Zamani
- Division of Cardiovascular Medicine, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kenneth B Margulies
- Division of Cardiovascular Medicine, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Svati H Shah
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA; Duke Molecular Physiology Institute, Durham, North Carolina, USA; Duke Clinical Research Institute, Durham, North Carolina, USA
| |
Collapse
|
9
|
Zakharyan R, Hakobyan S, Brojakowska A, Davitavyan S, Stepanyan A, Sirunyan T, Khachatryan G, Khlgatian MK, Bisserier M, Zhang S, Sahoo S, Hadri L, Garikipati VNS, Arakelyan A, Goukassian DA. Long-lasting sex-specific alteration in left ventricular cardiac transcriptome following gamma and simGCRsim radiation. Sci Rep 2025; 15:5963. [PMID: 39966642 PMCID: PMC11836050 DOI: 10.1038/s41598-025-89815-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Space irradiation (IR) is an important health risk for deep-space missions. We reported heart failure with preserved ejection fraction like cardiac phenotype 660-days following exposure to a single-dose of a simplified galactic cosmic ray simulation (simGCRsim) only in males with functional and structural impairment in left ventricular (LV) function. This sex-based dichotomy prompted us to investigate sex-specific changes in the LV transcriptome in three-month-old male and female mice exposed to 137Cs-γ- or simGCRsim-IR. Non-IR male and female (10 each) mice served as controls. LVs were collected at 440/660- and 440/550-days post-IR, male and female, respectively. RNA sequencing, differential gene expression, and functional annotation were performed on tissues from 5 mice/group. Sex and post-IR time points had the greatest influence on gene expression, surpassing the IR-type effects. SimGCRsim-IR showed more persistent transcriptome changes than γ-IR. We suggest that the single IR effects can persist up to 550-660 days, with overwhelmingly sex-biased responses at individual gene expression level.
Collapse
Affiliation(s)
- Roksana Zakharyan
- Institute of Molecular Biology, National Academy of Science of Republic of Armenia, Yerevan, Armenia.
- Russian-Armenian University, Yerevan, Armenia.
| | - Siras Hakobyan
- Institute of Molecular Biology, National Academy of Science of Republic of Armenia, Yerevan, Armenia
| | - Agnieszka Brojakowska
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
| | - Suren Davitavyan
- Institute of Molecular Biology, National Academy of Science of Republic of Armenia, Yerevan, Armenia
- Russian-Armenian University, Yerevan, Armenia
| | - Ani Stepanyan
- Institute of Molecular Biology, National Academy of Science of Republic of Armenia, Yerevan, Armenia
| | - Tamara Sirunyan
- Institute of Molecular Biology, National Academy of Science of Republic of Armenia, Yerevan, Armenia
- Russian-Armenian University, Yerevan, Armenia
| | - Gisane Khachatryan
- Institute of Molecular Biology, National Academy of Science of Republic of Armenia, Yerevan, Armenia
- Russian-Armenian University, Yerevan, Armenia
| | - Mary K Khlgatian
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Malik Bisserier
- Department of Cell Biology and Anatomy and Physiology, New York Medical College, Valhalla, NY, USA
| | - Shihong Zhang
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lahouaria Hadri
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Venkata Naga Srikanth Garikipati
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | - Arsen Arakelyan
- Institute of Molecular Biology, National Academy of Science of Republic of Armenia, Yerevan, Armenia
- Russian-Armenian University, Yerevan, Armenia
| | - David A Goukassian
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
10
|
Isaacs A, Zeemering S, Winters J, Batlle M, Bidar E, Boukens B, Casadei B, Chua W, Crijns HJGM, Fabritz L, Guasch E, Hatem SN, Hermans B, Kääb S, Kawczynski M, Maesen B, Maessen J, Mont L, Sinner MF, Wakili R, Verheule S, Kirchhof P, Schotten U, Stoll M. Lateral Atrial Expression Patterns Provide Insights into Local Transcription Disequilibrium Contributing to Disease Susceptibility. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025; 18:e004594. [PMID: 39846178 DOI: 10.1161/circgen.124.004594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 11/19/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Transcriptional dysregulation, possibly affected by genetic variation, contributes to disease etiology. Due to dissimilarities in development, function, and remodeling during disease progression, transcriptional differences between the left atrium (LA) and right atrium (RA) may provide insight into diseases such as atrial fibrillation. METHODS Lateral differences in atrial transcription were evaluated in CATCH ME (Characterizing Atrial fibrillation by Translating its Causes into Health Modifiers in the Elderly) using a 2-stage discovery and replication design. The design took advantage of the availability of 32 paired samples, for which both LA and RA tissue were obtained, as a discovery cohort, and 98 LA and 69 RA unpaired samples utilized as a replication cohort. RESULTS A total of 714 transcripts were identified and replicated as differentially expressed (DE) between LA and RA, as well as 98 exons in 55 genes. Approximately 50% of DE transcripts were colocated with another frequently correlated DE transcript (PFDR ≤0.05 for 579 regions). These "transcription disequilibrium" blocks contained examples including side-specific differential exon usage, such as the PITX2 locus, where ENPEP showed evidence of differential exon usage. Analysis of this region in conjunction with BMP10 identified rs9790621 as associated with ENPEP transcription in LA, while rs7687878 was associated with BMP10 expression in RA. In RA, BMP10 and ENPEP were strongly correlated in noncarriers, which was attenuated in risk-allele carriers, where BMP10 and PITX2 expression were strongly correlated. CONCLUSIONS These results significantly expand knowledge of the intricate, tissue-specific transcriptional landscape in human atria, including DE transcripts and side-specific isoform expression. Furthermore, they suggest the existence of blocks of transcription disequilibrium influenced by genetics.
Collapse
Affiliation(s)
- Aaron Isaacs
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Maastricht Center for Systems Biology (A.I.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Stef Zeemering
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Joris Winters
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Montserrat Batlle
- Institute of Biomedical Research August Pi Sunyer, Barcelona, Spain (M.B., E.G., L.M.)
- CIBERCV, Madrid, Spain (M.B., E.G., L.M.)
| | - Elham Bidar
- Departments of Cardiothoracic Surgery (E.B., M.K., B.M., J.M.), Maastricht University Medical Centre, the Netherlands
| | - Bas Boukens
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Barbara Casadei
- NIHR Oxford Biomedical Research Center, John Radcliffe Hospital, University of Oxford, United Kingdom (B.C.)
| | - Winnie Chua
- Institute of Cardiovascular Sciences, Birmingham, United Kingdom (W.C., L.F., P.K.)
| | - Harry J G M Crijns
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Cardiology (H.J.G.M.C.), Maastricht University Medical Centre, the Netherlands
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, Birmingham, United Kingdom (W.C., L.F., P.K.)
- University Center of Cardiovascular Sciences, University Medical Center Hamburg Eppendorf, Germany (L.F.)
- German Center for Cardiovascular Research (DZHK), Hamburg/Kiel/Lübeck, Germany (L.F., S.K., M.F.S., P.K.)
| | - Eduard Guasch
- Institute of Biomedical Research August Pi Sunyer, Barcelona, Spain (M.B., E.G., L.M.)
- CIBERCV, Madrid, Spain (M.B., E.G., L.M.)
- Clinic Barcelona, Universitat de Barcelona, Spain (E.G., L.M.)
| | - Stephane N Hatem
- INSERM UMRS 1166, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne University, Paris, France (S.N.H.)
| | - Ben Hermans
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Stefan Kääb
- German Center for Cardiovascular Research (DZHK), Hamburg/Kiel/Lübeck, Germany (L.F., S.K., M.F.S., P.K.)
- University Heart and Vascular Center, University Hospital Hamburg Eppendorf, Germany (S.K., M.F.S.)
- Department of Cardiology, University Hospital of Munich, Germany (S.K., M.F.S.)
| | - Michal Kawczynski
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
- Departments of Cardiothoracic Surgery (E.B., M.K., B.M., J.M.), Maastricht University Medical Centre, the Netherlands
| | - Bart Maesen
- Departments of Cardiothoracic Surgery (E.B., M.K., B.M., J.M.), Maastricht University Medical Centre, the Netherlands
| | - Jos Maessen
- Departments of Cardiothoracic Surgery (E.B., M.K., B.M., J.M.), Maastricht University Medical Centre, the Netherlands
| | - Lluis Mont
- Institute of Biomedical Research August Pi Sunyer, Barcelona, Spain (M.B., E.G., L.M.)
- CIBERCV, Madrid, Spain (M.B., E.G., L.M.)
- Clinic Barcelona, Universitat de Barcelona, Spain (E.G., L.M.)
| | - Moritz F Sinner
- German Center for Cardiovascular Research (DZHK), Hamburg/Kiel/Lübeck, Germany (L.F., S.K., M.F.S., P.K.)
- University Heart and Vascular Center, University Hospital Hamburg Eppendorf, Germany (S.K., M.F.S.)
- Department of Cardiology, University Hospital of Munich, Germany (S.K., M.F.S.)
| | - Reza Wakili
- Department of Medicine and Cardiology, Goethe University, Frankfurt, Germany (R.W.)
- German Center for Cardiovascular Research DZHK, Rhine-Main (R.W.)
| | - Sander Verheule
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Paulus Kirchhof
- Institute of Cardiovascular Sciences, Birmingham, United Kingdom (W.C., L.F., P.K.)
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany (P.K.)
- German Center for Cardiovascular Research (DZHK), Hamburg/Kiel/Lübeck, Germany (L.F., S.K., M.F.S., P.K.)
| | - Ulrich Schotten
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Monika Stoll
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Biochemistry (M.S.), Maastricht University, the Netherlands
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Germany (M.S.)
| |
Collapse
|
11
|
Lv T, Liu C, Guo S, Wu M, Wang X, Zhang Z, Zhou J, Yao Y, Shen Z, Yang J, Sun S, Liu Z, Chi J. Targeting Ketone Body Metabolism Improves Cardiac Function and Hemodynamics in Patients With Heart Failure: A Systematic Review and Meta-Analysis. Nutr Rev 2025:nuae179. [PMID: 39873669 DOI: 10.1093/nutrit/nuae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
CONTEXT The impacts of elevated ketone body levels on cardiac function and hemodynamics in patients with heart failure (HF) remain unclear. OBJECTIVE The effects of ketone intervention on these parameters in patients with HF were evaluated quantitatively in this meta-analysis. DATA SOURCES We searched the PubMed, Cochrane Library, and Embase databases for relevant studies published from inception to April 13, 2024. Ketone therapy included ketone ester and β-hydroxybutyrate intervention. DATA EXTRACTION Seven human studies were included for the quantitative analysis. DATA ANALYSIS Our results showed that ketone therapy significantly improved left ventricular ejection fraction (standardized mean difference, 0.52 [95% CI, 0.25-0.80]; I2 = 0%), cardiac output (0.84 [95% CI, 0.36-1.32]; I2 = 68%) and stroke volume (0.47 [95% CI, 0.10-0.84]; I2 = 39%), and significantly reduced systemic vascular resistance (-0.92 [95% CI, -1.52 to -0.33]; I2 = 74%) without influencing mean arterial pressure (-0.09 [95% CI: -0.40 to 0.22]; I2 = 0%) in patients with HF. Subgroup analysis revealed that the enhanced cardiac function and favorable hemodynamic effects of ketone therapy were also applicable to individuals without HF. CONCLUSIONS Ketone therapy may significantly improve cardiac systolic function and hemodynamics in patients with HF and in patients without HF, suggesting it may be a promising treatment for patients with HF and also a beneficial medical strategy for patients without HF or healthy individuals.
Collapse
Affiliation(s)
- Tingting Lv
- Department of General Practice, Shaoxing People's Hospital, Shaoxing 312000, P. R. China
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Chunyan Liu
- Department of Infection Management, Shaoxing People's Hospital, Shaoxing 312000, P. R. China
| | - Shitian Guo
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P. R. China
| | - Menglu Wu
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Xiang Wang
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Ziyi Zhang
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Jiedong Zhou
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Yiying Yao
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Zeyu Shen
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Juntao Yang
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Shijia Sun
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Zheng Liu
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Jufang Chi
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Department of Cardiology, Zhuji People's Hospital (Zhuji Hospital, Wenzhou Medical University), Zhuji, Zhejiang 311800, P. R. China
| |
Collapse
|
12
|
Zou E, Xu X, Chen L. Potential of plasma biomarkers for heart failure prediction, management, and prognosis: A multiomics perspective. Heart Fail Rev 2025; 30:55-67. [PMID: 39377997 DOI: 10.1007/s10741-024-10443-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 12/15/2024]
Abstract
Heart failure (HF) remains a major global health challenge, and more effective and comprehensive plasma biomarkers are needed to effectively treat HF patients. Multiomics studies have shown that DNA fragments, noncoding RNAs, proteins, and metabolites may be potential plasma biomarkers for HF. However, comprehensive reviews that focus on research on plasma biomarkers for HF from an omics perspective are lacking. This review summarizes the applications of various omics approaches in the exploration of biomarkers related to the risk assessment, diagnosis, subtype classification, medical management, and prognosis prediction of HF. Moreover, as heart transplantation and left ventricular assistant device (LVAD) implantation are terminal therapies for end-stage HF patients, this review also discusses the role of cell-free DNA as a biomarker for cardiac transplant rejection and omics studies of plasma biomarkers in patients who respond to LVAD therapy. Our findings suggest that future omics research on HF biomarkers should employ integrated multiomics methods and expand the sample size to increase the robustness of the results and that the identified biomarkers should be further validated in large cohorts.
Collapse
Affiliation(s)
- Erhou Zou
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Chen
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
13
|
Sun Q, Karwi QG, Wong N, Lopaschuk GD. Advances in myocardial energy metabolism: metabolic remodelling in heart failure and beyond. Cardiovasc Res 2024; 120:1996-2016. [PMID: 39453987 PMCID: PMC11646102 DOI: 10.1093/cvr/cvae231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/28/2024] [Accepted: 07/03/2024] [Indexed: 10/27/2024] Open
Abstract
The very high energy demand of the heart is primarily met by adenosine triphosphate (ATP) production from mitochondrial oxidative phosphorylation, with glycolysis providing a smaller amount of ATP production. This ATP production is markedly altered in heart failure, primarily due to a decrease in mitochondrial oxidative metabolism. Although an increase in glycolytic ATP production partly compensates for the decrease in mitochondrial ATP production, the failing heart faces an energy deficit that contributes to the severity of contractile dysfunction. The relative contribution of the different fuels for mitochondrial ATP production dramatically changes in the failing heart, which depends to a large extent on the type of heart failure. A common metabolic defect in all forms of heart failure [including heart failure with reduced ejection fraction (HFrEF), heart failure with preserved EF (HFpEF), and diabetic cardiomyopathies] is a decrease in mitochondrial oxidation of pyruvate originating from glucose (i.e. glucose oxidation). This decrease in glucose oxidation occurs regardless of whether glycolysis is increased, resulting in an uncoupling of glycolysis from glucose oxidation that can decrease cardiac efficiency. The mitochondrial oxidation of fatty acids by the heart increases or decreases, depending on the type of heart failure. For instance, in HFpEF and diabetic cardiomyopathies myocardial fatty acid oxidation increases, while in HFrEF myocardial fatty acid oxidation either decreases or remains unchanged. The oxidation of ketones (which provides the failing heart with an important energy source) also differs depending on the type of heart failure, being increased in HFrEF, and decreased in HFpEF and diabetic cardiomyopathies. The alterations in mitochondrial oxidative metabolism and glycolysis in the failing heart are due to transcriptional changes in key enzymes involved in the metabolic pathways, as well as alterations in redox state, metabolic signalling and post-translational epigenetic changes in energy metabolic enzymes. Of importance, targeting the mitochondrial energy metabolic pathways has emerged as a novel therapeutic approach to improving cardiac function and cardiac efficiency in the failing heart.
Collapse
Affiliation(s)
- Qiuyu Sun
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Qutuba G Karwi
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John’s, NL A1B 3V6, Canada
| | - Nathan Wong
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Center, University of Alberta, Edmonton, AB T6G 2S2, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2S2, Canada
| |
Collapse
|
14
|
Dabbaghi MM, Soleimani Roudi H, Safaei R, Baradaran Rahimi V, Fadaei MR, Askari VR. Unveiling the Mechanism of Protective Effects of Tanshinone as a New Fighter Against Cardiovascular Diseases: A Systematic Review. Cardiovasc Toxicol 2024; 24:1467-1509. [PMID: 39306819 DOI: 10.1007/s12012-024-09921-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/08/2024] [Indexed: 11/15/2024]
Abstract
Tanshinone, a natural compound found in the roots of Salvia miltiorrhiza, has been shown to possess various pharmacological properties, including anti-inflammatory, antioxidant, and cardiovascular protective effects. This article aims to review the literature on the cardiovascular protective effects of tanshinone and its underlying mechanisms. Tanshinone has been demonstrated to improve cardiac function, reduce oxidative stress, and inhibit inflammation in various animal models of cardiovascular diseases. Additionally, it has been shown to regulate multiple signaling pathways involved in the pathogenesis of cardiovascular diseases, such as the PI3K/AKT, MAPK, and NF-κB pathways. Clinical studies have also suggested that tanshinone may have therapeutic potential for treating cardiovascular diseases. In conclusion, tanshinone has emerged as a promising natural compound with significant cardiovascular protective effects, and further research is warranted to explore its potential clinical applications.
Collapse
Affiliation(s)
- Mohammad Mahdi Dabbaghi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran
| | - Hesan Soleimani Roudi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran
| | - Rozhan Safaei
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Fadaei
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Azadi Sq, Vakil Abad Highway, Mashhad, 9177948564, Iran.
| |
Collapse
|
15
|
Zhu W, Guo S, Sun J, Zhao Y, Liu C. Lactate and lactylation in cardiovascular diseases: current progress and future perspectives. Metabolism 2024; 158:155957. [PMID: 38908508 DOI: 10.1016/j.metabol.2024.155957] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Cardiovascular diseases (CVDs) are often linked to structural and functional impairments, such as heart defects and circulatory dysfunction, leading to compromised peripheral perfusion and heightened morbidity risks. Metabolic remodeling, particularly in the context of cardiac fibrosis and inflammation, is increasingly recognized as a pivotal factor in the pathogenesis of CVDs. Metabolic syndromes further predispose individuals to these conditions, underscoring the need to elucidate the metabolic underpinnings of CVDs. Lactate, a byproduct of glycolysis, is now recognized as a key molecule that connects cellular metabolism with the regulation of cellular activity. The transport of lactate between different cells is essential for metabolic homeostasis and signal transduction. Disruptions to lactate dynamics are implicated in various CVDs. Furthermore, lactylation, a novel post-translational modification, has been identified in cardiac cells, where it influences protein function and gene expression, thereby playing a significant role in CVD pathogenesis. In this review, we summarized recent advancements in understanding the role of lactate and lactylation in CVDs, offering fresh insights that could guide future research directions and therapeutic interventions. The potential of lactate metabolism and lactylation as innovative therapeutic targets for CVD is a promising avenue for exploration.
Collapse
Affiliation(s)
- Wengen Zhu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China.
| | - Siyu Guo
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Junyi Sun
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Yudan Zhao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, PR China.
| | - Chen Liu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China.
| |
Collapse
|
16
|
Bornstein MR, Tian R, Arany Z. Human cardiac metabolism. Cell Metab 2024; 36:1456-1481. [PMID: 38959861 PMCID: PMC11290709 DOI: 10.1016/j.cmet.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/12/2024] [Accepted: 06/05/2024] [Indexed: 07/05/2024]
Abstract
The heart is the most metabolically active organ in the human body, and cardiac metabolism has been studied for decades. However, the bulk of studies have focused on animal models. The objective of this review is to summarize specifically what is known about cardiac metabolism in humans. Techniques available to study human cardiac metabolism are first discussed, followed by a review of human cardiac metabolism in health and in heart failure. Mechanistic insights, where available, are reviewed, and the evidence for the contribution of metabolic insufficiency to heart failure, as well as past and current attempts at metabolism-based therapies, is also discussed.
Collapse
Affiliation(s)
- Marc R Bornstein
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Zoltan Arany
- Cardiovascular Institute Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Challa AA, Vidal P, Maurya SK, Maurya CK, Baer LA, Wang Y, James NM, Pardeshi PJ, Fasano M, Carley AN, Stanford KI, Lewandowski ED. UCP1-dependent brown adipose activation accelerates cardiac metabolic remodeling and reduces initial hypertrophic and fibrotic responses to pathological stress. FASEB J 2024; 38:e23709. [PMID: 38809700 PMCID: PMC11163965 DOI: 10.1096/fj.202400922r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/06/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
Brown adipose tissue (BAT) is correlated to cardiovascular health in rodents and humans, but the physiological role of BAT in the initial cardiac remodeling at the onset of stress is unknown. Activation of BAT via 48 h cold (16°C) in mice following transverse aortic constriction (TAC) reduced cardiac gene expression for LCFA uptake and oxidation in male mice and accelerated the onset of cardiac metabolic remodeling, with an early isoform shift of carnitine palmitoyltransferase 1 (CPT1) toward increased CPT1a, reduced entry of long chain fatty acid (LCFA) into oxidative metabolism (0.59 ± 0.02 vs. 0.72 ± 0.02 in RT TAC hearts, p < .05) and increased carbohydrate oxidation with altered glucose transporter content. BAT activation with TAC reduced early hypertrophic expression of β-MHC by 61% versus RT-TAC and reduced pro-fibrotic TGF-β1 and COL3α1 expression. While cardiac natriuretic peptide expression was yet to increase at only 3 days TAC, Nppa and Nppb expression were elevated in Cold TAC versus RT TAC hearts 2.7- and 2.4-fold, respectively. Eliminating BAT thermogenic activation with UCP1 KO mice eliminated differences between Cold TAC and RT TAC hearts, confirming effects of BAT activation rather than autonomous cardiac responses to cold. Female responses to BAT activation were blunted, with limited UCP1 changes with cold, partly due to already activated BAT in females at RT compared to thermoneutrality. These data reveal a previously unknown physiological mechanism of UCP1-dependent BAT activation in attenuating early cardiac hypertrophic and profibrotic signaling and accelerating remodeled metabolic activity in the heart at the onset of cardiac stress.
Collapse
Affiliation(s)
- Azariyas A. Challa
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Pablo Vidal
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - Santosh K. Maurya
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Chandan K. Maurya
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Lisa A. Baer
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - Yang Wang
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Natasha Maria James
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - Parth J. Pardeshi
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - Matthew Fasano
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Andrew N. Carley
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| | - Kristin I. Stanford
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Department of Physiology and Cell Biology, College of Medicine, Ohio State University. Columbus, OH., 43210, USA
- Department of Surgery, General and Gastrointestinal Surgery, College of Medicine, The Ohio State University. Columbus, OH., 43210, USA
| | - E. Douglas Lewandowski
- Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
- Davis Heart and Lung Research Institute and Department of Internal Medicine, College of Medicine, Ohio State University. Columbus, OH, 43210, USA
| |
Collapse
|
18
|
Leow JWH, Chan ECY. CYP2J2-mediated metabolism of arachidonic acid in heart: A review of its kinetics, inhibition and role in heart rhythm control. Pharmacol Ther 2024; 258:108637. [PMID: 38521247 DOI: 10.1016/j.pharmthera.2024.108637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 02/06/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
Cytochrome P450 2 J2 (CYP2J2) is primarily expressed extrahepatically and is the predominant epoxygenase in human cardiac tissues. This highlights its key role in the metabolism of endogenous substrates. Significant scientific interest lies in cardiac CYP2J2 metabolism of arachidonic acid (AA), an omega-6 polyunsaturated fatty acid, to regioisomeric bioactive epoxyeicosatrienoic acid (EET) metabolites that show cardioprotective effects including regulation of cardiac electrophysiology. From an in vitro perspective, the accurate characterization of the kinetics of CYP2J2 metabolism of AA including its inhibition and inactivation by drugs could be useful in facilitating in vitro-in vivo extrapolations to predict drug-AA interactions in drug discovery and development. In this review, background information on the structure, regulation and expression of CYP2J2 in human heart is presented alongside AA and EETs as its endogenous substrate and metabolites. The in vitro and in vivo implications of the kinetics of this endogenous metabolic pathway as well as its perturbation via inhibition and inactivation by drugs are elaborated. Additionally, the role of CYP2J2-mediated metabolism of AA to EETs in cardiac electrophysiology will be expounded.
Collapse
Affiliation(s)
- Jacqueline Wen Hui Leow
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore.
| |
Collapse
|
19
|
Li Y, Lou N, Liu X, Zhuang X, Chen S. Exploring new mechanisms of Imeglimin in diabetes treatment: Amelioration of mitochondrial dysfunction. Biomed Pharmacother 2024; 175:116755. [PMID: 38772155 DOI: 10.1016/j.biopha.2024.116755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 04/26/2024] [Accepted: 05/13/2024] [Indexed: 05/23/2024] Open
Abstract
With the increasing prevalence of type 2 diabetes mellitus (T2DM), it has become critical to identify effective treatment strategies. In recent years, the novel oral hypoglycaemic drug Imeglimin has attracted much attention in the field of diabetes treatment. The mechanisms of its therapeutic action are complex and are not yet fully understood by current research. Current evidence suggests that pancreatic β-cells, liver, and skeletal muscle are the main organs in which Imeglimin lowers blood glucose levels and that it acts mainly by targeting mitochondrial function, thereby inhibiting hepatic gluconeogenesis, enhancing insulin sensitivity, promoting pancreatic β-cell function, and regulating energy metabolism. There is growing evidence that the drug also has a potentially volatile role in the treatment of diabetic complications, including metabolic cardiomyopathy, diabetic vasculopathy, and diabetic neuroinflammation. According to available clinical studies, its efficacy and safety profile are more evident than other hypoglycaemic agents, and it has synergistic effects when combined with other antidiabetic drugs, and also has potential in the treatment of T2DM-related complications. This review aims to shed light on the latest research progress in the treatment of T2DM with Imeglimin, thereby providing clinicians and researchers with the latest insights into Imeglimin as a viable option for the treatment of T2DM.
Collapse
Affiliation(s)
- Yilin Li
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China
| | - Nenngjun Lou
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China
| | - Xiaojing Liu
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China
| | - Xianghua Zhuang
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China; Multidisciplinary Innovation Center for Nephrology of the Second Hospital of Shandong University, Jinan 250033, China.
| | - Shihong Chen
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan 250033, China; Multidisciplinary Innovation Center for Nephrology of the Second Hospital of Shandong University, Jinan 250033, China.
| |
Collapse
|
20
|
Pherwani S, Connolly D, Sun Q, Karwi QG, Carr M, Ho KL, Wagg CS, Zhang L, Levasseur J, Silver H, Dyck JRB, Lopaschuk GD. Ketones provide an extra source of fuel for the failing heart without impairing glucose oxidation. Metabolism 2024; 154:155818. [PMID: 38369056 DOI: 10.1016/j.metabol.2024.155818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Cardiac glucose oxidation is decreased in heart failure with reduced ejection fraction (HFrEF), contributing to a decrease in myocardial ATP production. In contrast, circulating ketones and cardiac ketone oxidation are increased in HFrEF. Since ketones compete with glucose as a fuel source, we aimed to determine whether increasing ketone concentration both chronically with the SGLT2 inhibitor, dapagliflozin, or acutely in the perfusate has detrimental effects on cardiac glucose oxidation in HFrEF, and what effect this has on cardiac ATP production. METHODS 8-week-old male C57BL6/N mice underwent sham or transverse aortic constriction (TAC) surgery to induce HFrEF over 3 weeks, after which TAC mice were randomized to treatment with either vehicle or the SGLT2 inhibitor, dapagliflozin (DAPA), for 4 weeks (raises blood ketones). Cardiac function was assessed by echocardiography. Cardiac energy metabolism was measured in isolated working hearts perfused with 5 mM glucose, 0.8 mM palmitate, and either 0.2 mM or 0.6 mM β-hydroxybutyrate (βOHB). RESULTS TAC hearts had significantly decreased %EF compared to sham hearts, with no effect of DAPA. Glucose oxidation was significantly decreased in TAC hearts compared to sham hearts and did not decrease further in TAC hearts treated with high βOHB or in TAC DAPA hearts, despite βOHB oxidation rates increasing in both TAC vehicle and TAC DAPA hearts at high βOHB concentrations. Rather, increasing βOHB supply to the heart selectively decreased fatty acid oxidation rates. DAPA significantly increased ATP production at both βOHB concentrations by increasing the contribution of glucose oxidation to ATP production. CONCLUSION Therefore, increasing ketone concentration increases energy supply and ATP production in HFrEF without further impairing glucose oxidation.
Collapse
Affiliation(s)
- Simran Pherwani
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - David Connolly
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Qiuyu Sun
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Qutuba G Karwi
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2S2, Canada; Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Newfoundland and Labrador, St. John's A1B 3V6, Canada
| | - Michael Carr
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Kim L Ho
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Cory S Wagg
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Liyan Zhang
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Jody Levasseur
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Heidi Silver
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Jason R B Dyck
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2S2, Canada.
| |
Collapse
|
21
|
Sun Q, Güven B, Wagg CS, Almeida de Oliveira A, Silver H, Zhang L, Chen B, Wei K, Ketema EB, Karwi QG, Persad KL, Vu J, Wang F, Dyck JRB, Oudit GY, Lopaschuk GD. Mitochondrial fatty acid oxidation is the major source of cardiac adenosine triphosphate production in heart failure with preserved ejection fraction. Cardiovasc Res 2024; 120:360-371. [PMID: 38193548 DOI: 10.1093/cvr/cvae006] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 01/10/2024] Open
Abstract
AIMS Heart failure with preserved ejection fraction (HFpEF) is a prevalent disease worldwide. While it is well established that alterations of cardiac energy metabolism contribute to cardiovascular pathology, the precise source of fuel used by the heart in HFpEF remains unclear. The objective of this study was to define the energy metabolic profile of the heart in HFpEF. METHODS AND RESULTS Eight-week-old C57BL/6 male mice were subjected to a '2-Hit' HFpEF protocol [60% high-fat diet (HFD) + 0.5 g/L of Nω-nitro-L-arginine methyl ester]. Echocardiography and pressure-volume loop analysis were used for assessing cardiac function and cardiac haemodynamics, respectively. Isolated working hearts were perfused with radiolabelled energy substrates to directly measure rates of fatty acid oxidation, glucose oxidation, ketone oxidation, and glycolysis. HFpEF mice exhibited increased body weight, glucose intolerance, elevated blood pressure, diastolic dysfunction, and cardiac hypertrophy. In HFpEF hearts, insulin stimulation of glucose oxidation was significantly suppressed. This was paralleled by an increase in fatty acid oxidation rates, while cardiac ketone oxidation and glycolysis rates were comparable with healthy control hearts. The balance between glucose and fatty acid oxidation contributing to overall adenosine triphosphate (ATP) production was disrupted, where HFpEF hearts were more reliant on fatty acid as the major source of fuel for ATP production, compensating for the decrease of ATP originating from glucose oxidation. Additionally, phosphorylated pyruvate dehydrogenase levels decreased in both HFpEF mice and human patient's heart samples. CONCLUSION In HFpEF, fatty acid oxidation dominates as the major source of cardiac ATP production at the expense of insulin-stimulated glucose oxidation.
Collapse
Affiliation(s)
- Qiuyu Sun
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Berna Güven
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Canada
- Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey
| | - Cory S Wagg
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Amanda Almeida de Oliveira
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Heidi Silver
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Liyan Zhang
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Brandon Chen
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
| | - Kaleigh Wei
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
| | - Ezra B Ketema
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Qutuba G Karwi
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Saint John's, Canada
| | - Kaya L Persad
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Jennie Vu
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Faqi Wang
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Jason R B Dyck
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Gavin Y Oudit
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
22
|
Plessner M, Thiele L, Hofhuis J, Thoms S. Tissue-specific roles of peroxisomes revealed by expression meta-analysis. Biol Direct 2024; 19:14. [PMID: 38365851 PMCID: PMC10873952 DOI: 10.1186/s13062-024-00458-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/30/2024] [Indexed: 02/18/2024] Open
Abstract
Peroxisomes are primarily studied in the brain, kidney, and liver due to the conspicuous tissue-specific pathology of peroxisomal biogenesis disorders. In contrast, little is known about the role of peroxisomes in other tissues such as the heart. In this meta-analysis, we explore mitochondrial and peroxisomal gene expression on RNA and protein levels in the brain, heart, kidney, and liver, focusing on lipid metabolism. Further, we evaluate a potential developmental and heart region-dependent specificity of our gene set. We find marginal expression of the enzymes for peroxisomal fatty acid oxidation in cardiac tissue in comparison to the liver or cardiac mitochondrial β-oxidation. However, the expression of peroxisome biogenesis proteins in the heart is similar to other tissues despite low levels of peroxisomal fatty acid oxidation. Strikingly, peroxisomal targeting signal type 2-containing factors and plasmalogen biosynthesis appear to play a fundamental role in explaining the essential protective and supporting functions of cardiac peroxisomes.
Collapse
Affiliation(s)
- Matthias Plessner
- Department of Biochemistry and Molecular Medicine, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Leonie Thiele
- Department of Biochemistry and Molecular Medicine, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Julia Hofhuis
- Department of Biochemistry and Molecular Medicine, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Sven Thoms
- Department of Biochemistry and Molecular Medicine, Medical School OWL, Bielefeld University, Bielefeld, Germany.
- Department of Child and Adolescent Health, University Medical Center, Göttingen, Germany.
| |
Collapse
|
23
|
Choudhury C, Gill MK, McAleese CE, Butcher NJ, Ngo ST, Steyn FJ, Minchin RF. The Arylamine N-Acetyltransferases as Therapeutic Targets in Metabolic Diseases Associated with Mitochondrial Dysfunction. Pharmacol Rev 2024; 76:300-320. [PMID: 38351074 DOI: 10.1124/pharmrev.123.000835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 02/16/2024] Open
Abstract
In humans, there are two arylamine N-acetyltransferase genes that encode functional enzymes (NAT1 and NAT2) as well as one pseudogene, all of which are located together on chromosome 8. Although they were first identified by their role in the acetylation of drugs and other xenobiotics, recent studies have shown strong associations for both enzymes in a variety of diseases, including cancer, cardiovascular disease, and diabetes. There is growing evidence that this association may be causal. Consistently, NAT1 and NAT2 are shown to be required for healthy mitochondria. This review discusses the current literature on the role of both NAT1 and NAT2 in mitochondrial bioenergetics. It will attempt to relate our understanding of the evolution of the two genes with biologic function and then present evidence that several major metabolic diseases are influenced by NAT1 and NAT2. Finally, it will discuss current and future approaches to inhibit or enhance NAT1 and NAT2 activity/expression using small-molecule drugs. SIGNIFICANCE STATEMENT: The arylamine N-acetyltransferases (NATs) NAT1 and NAT2 share common features in their associations with mitochondrial bioenergetics. This review discusses mitochondrial function as it relates to health and disease, and the importance of NAT in mitochondrial function and dysfunction. It also compares NAT1 and NAT2 to highlight their functional similarities and differences. Both NAT1 and NAT2 are potential drug targets for diseases where mitochondrial dysfunction is a hallmark of onset and progression.
Collapse
Affiliation(s)
- Chandra Choudhury
- School of Biomedical Sciences (C.C., M.K.G., C.E.M., N.J.B., F.J.S., R.F.M.) and Australian Institute for Bioengineering and Nanotechnology (S.T.N.), University of Queensland, Brisbane, Australia
| | - Melinder K Gill
- School of Biomedical Sciences (C.C., M.K.G., C.E.M., N.J.B., F.J.S., R.F.M.) and Australian Institute for Bioengineering and Nanotechnology (S.T.N.), University of Queensland, Brisbane, Australia
| | - Courtney E McAleese
- School of Biomedical Sciences (C.C., M.K.G., C.E.M., N.J.B., F.J.S., R.F.M.) and Australian Institute for Bioengineering and Nanotechnology (S.T.N.), University of Queensland, Brisbane, Australia
| | - Neville J Butcher
- School of Biomedical Sciences (C.C., M.K.G., C.E.M., N.J.B., F.J.S., R.F.M.) and Australian Institute for Bioengineering and Nanotechnology (S.T.N.), University of Queensland, Brisbane, Australia
| | - Shyuan T Ngo
- School of Biomedical Sciences (C.C., M.K.G., C.E.M., N.J.B., F.J.S., R.F.M.) and Australian Institute for Bioengineering and Nanotechnology (S.T.N.), University of Queensland, Brisbane, Australia
| | - Frederik J Steyn
- School of Biomedical Sciences (C.C., M.K.G., C.E.M., N.J.B., F.J.S., R.F.M.) and Australian Institute for Bioengineering and Nanotechnology (S.T.N.), University of Queensland, Brisbane, Australia
| | - Rodney F Minchin
- School of Biomedical Sciences (C.C., M.K.G., C.E.M., N.J.B., F.J.S., R.F.M.) and Australian Institute for Bioengineering and Nanotechnology (S.T.N.), University of Queensland, Brisbane, Australia
| |
Collapse
|
24
|
Meng S, Yu Y, Yu S, Zhu S, Shi M, Xiang M, Ma H. Advances in Metabolic Remodeling and Intervention Strategies in Heart Failure. J Cardiovasc Transl Res 2024; 17:36-55. [PMID: 37843752 DOI: 10.1007/s12265-023-10443-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/27/2023] [Indexed: 10/17/2023]
Abstract
The heart is the most energy-demanding organ throughout the whole body. Perturbations or failure in energy metabolism contributes to heart failure (HF), which represents the advanced stage of various heart diseases. The poor prognosis and huge economic burden associated with HF underscore the high unmet need to explore novel therapies targeting metabolic modulators beyond conventional approaches focused on neurohormonal and hemodynamic regulators. Emerging evidence suggests that alterations in metabolic substrate reliance, metabolic pathways, metabolic by-products, and energy production collectively regulate the occurrence and progression of HF. In this review, we provide an overview of cardiac metabolic remodeling, encompassing the utilization of free fatty acids, glucose metabolism, ketone bodies, and branched-chain amino acids both in the physiological condition and heart failure. Most importantly, the latest advances in pharmacological interventions are discussed as a promising therapeutic approach to restore cardiac function, drawing insights from recent basic research, preclinical and clinical studies.
Collapse
Affiliation(s)
- Simin Meng
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Yi Yu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Shuo Yu
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Shiyu Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Mengjia Shi
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University; State Key Laboratory of Transvascular Implantation Devices; Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
25
|
Zhang T, Xu L, Guo X, Tao H, Liu Y, Liu X, Zhang Y, Meng X. The potential of herbal drugs to treat heart failure: The roles of Sirt1/AMPK. J Pharm Anal 2024; 14:157-176. [PMID: 38464786 PMCID: PMC10921247 DOI: 10.1016/j.jpha.2023.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/09/2023] [Accepted: 09/05/2023] [Indexed: 03/12/2024] Open
Abstract
Heart failure (HF) is a highly morbid syndrome that seriously affects the physical and mental health of patients and generates an enormous socio-economic burden. In addition to cardiac myocyte oxidative stress and apoptosis, which are considered mechanisms for the development of HF, alterations in cardiac energy metabolism and pathological autophagy also contribute to cardiac abnormalities and ultimately HF. Silent information regulator 1 (Sirt1) and adenosine monophosphate-activated protein kinase (AMPK) are nicotinamide adenine dinucleotide (NAD+)-dependent deacetylases and phosphorylated kinases, respectively. They play similar roles in regulating some pathological processes of the heart through regulating targets such as peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), protein 38 mitogen-activated protein kinase (p38 MAPK), peroxisome proliferator-activated receptors (PPARs), and mammalian target of rapamycin (mTOR). We summarized the synergistic effects of Sirt1 and AMPK in the heart, and listed the traditional Chinese medicine (TCM) that exhibit cardioprotective properties by modulating the Sirt1/AMPK pathway, to provide a basis for the development of Sirt1/AMPK activators or inhibitors for the treatment of HF and other cardiovascular diseases (CVDs).
Collapse
Affiliation(s)
- Tao Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lei Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiaowei Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Honglin Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yue Liu
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianfeng Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yi Zhang
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, Sichuan, 620032, China
| |
Collapse
|
26
|
Vite A, Matsuura TR, Bedi KC, Flam EL, Arany Z, Kelly DP, Margulies KB. Functional Impact of Alternative Metabolic Substrates in Failing Human Cardiomyocytes. JACC Basic Transl Sci 2024; 9:1-15. [PMID: 38362346 PMCID: PMC10864907 DOI: 10.1016/j.jacbts.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 02/17/2024]
Abstract
Recent studies suggest that metabolic dysregulation in patients with heart failure might contribute to myocardial contractile dysfunction. To understand the correlation between function and energy metabolism, we studied the impact of different fuel substrates on human nonfailing or failing cardiomyocytes. Consistent with the concept of metabolic flexibility, nonfailing myocytes exhibited excellent contractility in all fuels provided. However, impaired contractility was observed in failing myocytes when carbohydrates alone were used but was improved when additional substrates were added. This study demonstrates the functional significance of fuel utilization shifts in failing human cardiomyocytes.
Collapse
Affiliation(s)
- Alexia Vite
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Timothy R. Matsuura
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kenneth C. Bedi
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Emily L. Flam
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zoltan Arany
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel P. Kelly
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kenneth B. Margulies
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
27
|
Carvalho RA. The glycolytic pathway to heart failure. GLYCOLYSIS 2024:235-266. [DOI: 10.1016/b978-0-323-91704-9.00010-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
28
|
Walker MA, Chen H, Yadav A, Ritterhoff J, Villet O, McMillen T, Wang Y, Purcell H, Djukovic D, Raftery D, Isoherranen N, Tian R. Raising NAD + Level Stimulates Short-Chain Dehydrogenase/Reductase Proteins to Alleviate Heart Failure Independent of Mitochondrial Protein Deacetylation. Circulation 2023; 148:2038-2057. [PMID: 37965787 PMCID: PMC10842390 DOI: 10.1161/circulationaha.123.066039] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/04/2023] [Indexed: 11/16/2023]
Abstract
BACKGROUND Strategies to increase cellular NAD+ (oxidized nicotinamide adenine dinucleotide) level have prevented cardiac dysfunction in multiple models of heart failure, but molecular mechanisms remain unclear. Little is known about the benefits of NAD+-based therapies in failing hearts after the symptoms of heart failure have appeared. Most pretreatment regimens suggested mechanisms involving activation of sirtuin, especially Sirt3 (sirtuin 3), and mitochondrial protein acetylation. METHODS We induced cardiac dysfunction by pressure overload in SIRT3-deficient (knockout) mice and compared their response with nicotinamide riboside chloride treatment with wild-type mice. To model a therapeutic approach, we initiated the treatment in mice with established cardiac dysfunction. RESULTS We found nicotinamide riboside chloride improved mitochondrial function and blunted heart failure progression. Similar benefits were observed in wild-type and knockout mice. Boosting NAD+ level improved the function of NAD(H) redox-sensitive SDR (short-chain dehydrogenase/reductase) family proteins. Upregulation of Mrpp2 (mitochondrial ribonuclease P protein 2), a multifunctional SDR protein and a subunit of mitochondrial ribonuclease P, improves mitochondrial DNA transcripts processing and electron transport chain function. Activation of SDRs in the retinol metabolism pathway stimulates RXRα (retinoid X receptor α)/PPARα (proliferator-activated receptor α) signaling and restores mitochondrial oxidative metabolism. Downregulation of Mrpp2 and impaired mitochondrial ribonuclease P were found in human failing hearts, suggesting a shared mechanism of defective mitochondrial biogenesis in mouse and human heart failure. CONCLUSIONS These findings identify SDR proteins as important regulators of mitochondrial function and molecular targets of NAD+-based therapy. Furthermore, the benefit is observed regardless of Sirt3-mediated mitochondrial protein deacetylation, a widely held mechanism for NAD+-based therapy for heart failure. The data also show that NAD+-based therapy can be useful in pre-existing heart failure.
Collapse
Affiliation(s)
- Matthew A. Walker
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Hongye Chen
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Aprajita Yadav
- Department of Pharmaceutics, School of Pharmacy, University
of Washington, Seattle, WA 98195
| | - Julia Ritterhoff
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Outi Villet
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Tim McMillen
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Yuliang Wang
- Department of Computer Science & Engineering,
University of Washington, Seattle, WA 98195
| | - Hayley Purcell
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Danijel Djukovic
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Daniel Raftery
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University
of Washington, Seattle, WA 98195
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of
Anesthesiology & Pain Medicine, University of Washington, Seattle, WA
98109
| |
Collapse
|
29
|
Su S, Ji X, Li T, Teng Y, Wang B, Han X, Zhao M. The changes of cardiac energy metabolism with sodium-glucose transporter 2 inhibitor therapy. Front Cardiovasc Med 2023; 10:1291450. [PMID: 38124893 PMCID: PMC10731052 DOI: 10.3389/fcvm.2023.1291450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023] Open
Abstract
Background/aims To investigate the specific effects of s odium-glucose transporter 2 inhibitor (SGLT2i) on cardiac energy metabolism. Methods A systematic literature search was conducted in eight databases. The retrieved studies were screened according to the inclusion and exclusion criteria, and relevant information was extracted according to the purpose of the study. Two researchers independently screened the studies, extracted information, and assessed article quality. Results The results of the 34 included studies (including 10 clinical and 24 animal studies) showed that SGLT2i inhibited cardiac glucose uptake and glycolysis, but promoted fatty acid (FA) metabolism in most disease states. SGLT2i upregulated ketone metabolism, improved the structure and functions of myocardial mitochondria, alleviated oxidative stress of cardiomyocytes in all literatures. SGLT2i increased cardiac glucose oxidation in diabetes mellitus (DM) and cardiac FA metabolism in heart failure (HF). However, the regulatory effects of SGLT2i on cardiac FA metabolism in DM and cardiac glucose oxidation in HF varied with disease types, stages, and intervention duration of SGLT2i. Conclusion SGLT2i improved the efficiency of cardiac energy production by regulating FA, glucose and ketone metabolism, improving mitochondria structure and functions, and decreasing oxidative stress of cardiomyocytes under pathological conditions. Thus, SGLT2i is deemed to exert a benign regulatory effect on cardiac metabolic disorders in various diseases. Systematic review registration https://www.crd.york.ac.uk/, PROSPERO (CRD42023484295).
Collapse
Affiliation(s)
- Sha Su
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xiang Ji
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Tong Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yu Teng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Baofu Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowan Han
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
30
|
Ritterhoff J, Tian R. Metabolic mechanisms in physiological and pathological cardiac hypertrophy: new paradigms and challenges. Nat Rev Cardiol 2023; 20:812-829. [PMID: 37237146 DOI: 10.1038/s41569-023-00887-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 05/28/2023]
Abstract
Cardiac metabolism is vital for heart function. Given that cardiac contraction requires a continuous supply of ATP in large quantities, the role of fuel metabolism in the heart has been mostly considered from the perspective of energy production. However, the consequence of metabolic remodelling in the failing heart is not limited to a compromised energy supply. The rewired metabolic network generates metabolites that can directly regulate signalling cascades, protein function, gene transcription and epigenetic modifications, thereby affecting the overall stress response of the heart. In addition, metabolic changes in both cardiomyocytes and non-cardiomyocytes contribute to the development of cardiac pathologies. In this Review, we first summarize how energy metabolism is altered in cardiac hypertrophy and heart failure of different aetiologies, followed by a discussion of emerging concepts in cardiac metabolic remodelling, that is, the non-energy-generating function of metabolism. We highlight challenges and open questions in these areas and finish with a brief perspective on how mechanistic research can be translated into therapies for heart failure.
Collapse
Affiliation(s)
- Julia Ritterhoff
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany.
- Mitochondria and Metabolism Center, Department of Anaesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA.
| | - Rong Tian
- Mitochondria and Metabolism Center, Department of Anaesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
31
|
Bhullar SK, Dhalla NS. Status of Mitochondrial Oxidative Phosphorylation during the Development of Heart Failure. Antioxidants (Basel) 2023; 12:1941. [PMID: 38001794 PMCID: PMC10669359 DOI: 10.3390/antiox12111941] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Mitochondria are specialized organelles, which serve as the "Power House" to generate energy for maintaining heart function. These organelles contain various enzymes for the oxidation of different substrates as well as the electron transport chain in the form of Complexes I to V for producing ATP through the process of oxidative phosphorylation (OXPHOS). Several studies have shown depressed OXPHOS activity due to defects in one or more components of the substrate oxidation and electron transport systems which leads to the depletion of myocardial high-energy phosphates (both creatine phosphate and ATP). Such changes in the mitochondria appear to be due to the development of oxidative stress, inflammation, and Ca2+-handling abnormalities in the failing heart. Although some investigations have failed to detect any changes in the OXPHOS activity in the failing heart, such results appear to be due to a loss of Ca2+ during the mitochondrial isolation procedure. There is ample evidence to suggest that mitochondrial Ca2+-overload occurs, which is associated with impaired mitochondrial OXPHOS activity in the failing heart. The depression in mitochondrial OXPHOS activity may also be due to the increased level of reactive oxygen species, which are formed as a consequence of defects in the electron transport complexes in the failing heart. Various metabolic interventions which promote the generation of ATP have been reported to be beneficial for the therapy of heart failure. Accordingly, it is suggested that depression in mitochondrial OXPHOS activity plays an important role in the development of heart failure.
Collapse
Affiliation(s)
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada;
| |
Collapse
|
32
|
Szrok-Jurga S, Czumaj A, Turyn J, Hebanowska A, Swierczynski J, Sledzinski T, Stelmanska E. The Physiological and Pathological Role of Acyl-CoA Oxidation. Int J Mol Sci 2023; 24:14857. [PMID: 37834305 PMCID: PMC10573383 DOI: 10.3390/ijms241914857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023] Open
Abstract
Fatty acid metabolism, including β-oxidation (βOX), plays an important role in human physiology and pathology. βOX is an essential process in the energy metabolism of most human cells. Moreover, βOX is also the source of acetyl-CoA, the substrate for (a) ketone bodies synthesis, (b) cholesterol synthesis, (c) phase II detoxication, (d) protein acetylation, and (d) the synthesis of many other compounds, including N-acetylglutamate-an important regulator of urea synthesis. This review describes the current knowledge on the importance of the mitochondrial and peroxisomal βOX in various organs, including the liver, heart, kidney, lung, gastrointestinal tract, peripheral white blood cells, and other cells. In addition, the diseases associated with a disturbance of fatty acid oxidation (FAO) in the liver, heart, kidney, lung, alimentary tract, and other organs or cells are presented. Special attention was paid to abnormalities of FAO in cancer cells and the diseases caused by mutations in gene-encoding enzymes involved in FAO. Finally, issues related to α- and ω- fatty acid oxidation are discussed.
Collapse
Affiliation(s)
- Sylwia Szrok-Jurga
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| | - Aleksandra Czumaj
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Jacek Turyn
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| | - Areta Hebanowska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| | - Julian Swierczynski
- Institue of Nursing and Medical Rescue, State University of Applied Sciences in Koszalin, 75-582 Koszalin, Poland;
| | - Tomasz Sledzinski
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Ewa Stelmanska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (J.T.); (A.H.)
| |
Collapse
|
33
|
Issa J, Lodewyckx P, Blasco H, Benz‐de‐Bretagne I, Labarthe F, Lefort B. Increased acylcarnitines in infant heart failure indicate fatty acid oxidation inhibition: towards therapeutic options? ESC Heart Fail 2023; 10:3114-3122. [PMID: 37614055 PMCID: PMC10567663 DOI: 10.1002/ehf2.14449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 08/25/2023] Open
Abstract
AIMS Heart failure in adults is characterized by reduction of long-chain fatty acid oxidation in favour of carbohydrate metabolism. This adaptive phenomenon becomes maladaptive because energy conversion decreases and lipid toxic derivatives known to impair cardiac function are accumulating. No data are available concerning metabolic modification in heart failure in children. METHODS AND RESULTS In order to evaluate the fatty acid oxidation in children suffering from heart failure, acylcarnitine profiles on dried blood spots were obtained from children under 16 years old with dilated cardiomyopathy and clinical heart failure (DCM-HF) and control children. Nine children were included in the DCM-HF group and eight in the control group. Acylcarnitine profiles revealed a significant 3.1-fold increase of total acylcarnitines (sum of C3 to C18 acylcarnitine species) in DCM-HF children compared with controls. This result persisted considering the sum of long-chain acylcarnitines (sum of C14 to C18 species), medium-chain acylcarnitines (sum of C8 to C12 species), and short-chain acylcarnitines (sum of C3 to C6 species), respectively, 2.0-, 2.6-, and 1.9-fold increase compared with the control group. A significant linear correlation was found between left ventricular dilatation or ejection fraction and acylcarnitines accumulation. Finally, acylcarnitine ratio C16OH/C16 and C18OH/C18 enhanced in the DCM-HF group, suggesting a diminution of the long-chain hydroxyl acyl-CoA dehydrogenase activity. CONCLUSIONS Our results suggest down-regulation of fatty acid oxidation in children with heart failure. Such lipidomic alteration could worsen heart function and may suggest considering a metabolic treatment of heart failure in children.
Collapse
Affiliation(s)
- Jean Issa
- Institut des Cardiopathies Congénitales de Tours, Hôpital Gatien de ClochevilleCHU Tours49 Boulevard BérangerTours37000France
- Université François RabelaisToursFrance
| | - Pierre Lodewyckx
- Institut des Cardiopathies Congénitales de Tours, Hôpital Gatien de ClochevilleCHU Tours49 Boulevard BérangerTours37000France
- Université François RabelaisToursFrance
| | - Hélène Blasco
- Université François RabelaisToursFrance
- Service de Biochimie et Biologie MoléculaireCHU ToursToursFrance
| | | | - François Labarthe
- Université François RabelaisToursFrance
- Département de PédiatrieCHU de ToursToursFrance
- INSERM UMR 1069ToursFrance
| | - Bruno Lefort
- Institut des Cardiopathies Congénitales de Tours, Hôpital Gatien de ClochevilleCHU Tours49 Boulevard BérangerTours37000France
- Université François RabelaisToursFrance
- INSERM UMR 1069ToursFrance
- FHU PreciCareToursFrance
| |
Collapse
|
34
|
Regan JA, Mentz RJ, Nguyen M, Green JB, Truby LK, Ilkayeva O, Newgard CB, Buse JB, Sourij H, Sjöström CD, Sattar N, McGarrah RW, Zheng Y, McGuire DK, Standl E, Armstrong P, Peterson ED, Hernandez AF, Holman RR, Shah SH. Mitochondrial metabolites predict adverse cardiovascular events in individuals with diabetes. JCI Insight 2023; 8:e168563. [PMID: 37552540 PMCID: PMC10544215 DOI: 10.1172/jci.insight.168563] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023] Open
Abstract
Metabolic mechanisms underlying the heterogeneity of major adverse cardiovascular (CV) event (MACE) risk in individuals with type 2 diabetes mellitus (T2D) remain unclear. We hypothesized that circulating metabolites reflecting mitochondrial dysfunction predict incident MACE in T2D. Targeted mass-spectrometry profiling of 60 metabolites was performed on baseline plasma samples from the Trial Evaluating Cardiovascular Outcomes with Sitagliptin (TECOS; discovery cohort) and Exenatide Study of Cardiovascular Event Lowering (EXSCEL; validation cohort) biomarker substudy cohorts. A principal components analysis metabolite factor comprising medium-chain acylcarnitines (MCACs) was associated with MACE in TECOS and validated in EXSCEL, with higher levels associated with higher MACE risk. Meta-analysis showed that long-chain acylcarnitines (LCACs) and dicarboxylacylcarnitines were also associated with MACE. Metabolites remained associated with MACE in multivariate models and favorably changed with exenatide therapy. A third cohort (Cardiac Catheterization Genetics [CATHGEN]) with T2D was assessed to determine whether these metabolites improved discriminative capability of multivariate models for MACE. Nine metabolites (MCACs and LCACs and 1 dicarboxylacylcarnitine) were associated with time to MACE in the CATHGEN cohort. Addition of these metabolites to clinical models minimally improved the discriminative capability for MACE but did significantly down reclassify risk. Thus, metabolites reporting on dysregulated mitochondrial fatty acid oxidation are present in higher levels in individuals with T2D who experience subsequent MACE. These biomarkers may improve CV risk prediction models, be therapy responsive, and highlight emerging risk mechanisms.
Collapse
Affiliation(s)
- Jessica A. Regan
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
- Duke University Department of Medicine, Durham, North Carolina, USA
| | - Robert J. Mentz
- Duke University Department of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Maggie Nguyen
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | - Jennifer B. Green
- Duke University Department of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Lauren K. Truby
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
- Duke University Department of Medicine, Durham, North Carolina, USA
| | - Olga Ilkayeva
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
- Duke University Department of Medicine, Durham, North Carolina, USA
| | | | - John B. Buse
- University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina, USA
| | - Harald Sourij
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - C. David Sjöström
- Late-stage Development, Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Naveed Sattar
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Robert W. McGarrah
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
- Duke University Department of Medicine, Durham, North Carolina, USA
| | - Yinggan Zheng
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Darren K. McGuire
- University of Texas Southwestern Medical Center and Parkland Health and Hospital System, Dallas, Texas, USA
| | - Eberhard Standl
- Diabetes Research Group at Munich Helmholtz Center, Munich, Germany
| | - Paul Armstrong
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Eric D. Peterson
- University of Texas Southwestern Medical Center and Parkland Health and Hospital System, Dallas, Texas, USA
| | - Adrian F. Hernandez
- Duke University Department of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Rury R. Holman
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Svati H. Shah
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
- Duke University Department of Medicine, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| |
Collapse
|
35
|
Tatman PD, Kao DP, Chatfield KC, Carroll IA, Wagner JA, Jonas ER, Sucharov CC, Port JD, Lowes BD, Minobe WA, Huebler SP, Karimpour-Fard A, Rodriguez EM, Liggett SB, Bristow MR. An extensive β1-adrenergic receptor gene signaling network regulates molecular remodeling in dilated cardiomyopathies. JCI Insight 2023; 8:e169720. [PMID: 37606047 PMCID: PMC10543724 DOI: 10.1172/jci.insight.169720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/11/2023] [Indexed: 08/23/2023] Open
Abstract
We investigated the extent, biologic characterization, phenotypic specificity, and possible regulation of a β1-adrenergic receptor-linked (β1-AR-linked) gene signaling network (β1-GSN) involved in left ventricular (LV) eccentric pathologic remodeling. A 430-member β1-GSN was identified by mRNA expression in transgenic mice overexpressing human β1-ARs or from literature curation, which exhibited opposite directional behavior in interventricular septum endomyocardial biopsies taken from patients with beta-blocker-treated, reverse remodeled dilated cardiomyopathies. With reverse remodeling, the major biologic categories and percentage of the dominant directional change were as follows: metabolic (19.3%, 81% upregulated); gene regulation (14.9%, 78% upregulated); extracellular matrix/fibrosis (9.1%, 92% downregulated); and cell homeostasis (13.3%, 60% upregulated). Regarding the comparison of β1-GSN categories with expression from 19,243 nonnetwork genes, phenotypic selection for major β1-GSN categories was exhibited for LV end systolic volume (contractility measure), ejection fraction (remodeling index), and pulmonary wedge pressure (wall tension surrogate), beginning at 3 months and persisting to study completion at 12 months. In addition, 121 lncRNAs were identified as possibly involved in cis-acting regulation of β1-GSN members. We conclude that an extensive 430-member gene network downstream from the β1-AR is involved in pathologic ventricular remodeling, with metabolic genes as the most prevalent category.
Collapse
Affiliation(s)
| | - David P. Kao
- Division of Cardiology, Department of Medicine, and
- Colorado Center for Personalized Medicine University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kathryn C. Chatfield
- Division of Cardiology, Department of Medicine, and
- Department of Pediatric Cardiology, Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Ian A. Carroll
- Division of Cardiology, Department of Medicine, and
- ARCA biopharma, Westminster, Colorado, USA
| | | | | | | | | | - Brian D. Lowes
- Division of Cardiovascular Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | - Anis Karimpour-Fard
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Stephen B. Liggett
- Departments of Medicine and Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Michael R. Bristow
- Division of Cardiology, Department of Medicine, and
- ARCA biopharma, Westminster, Colorado, USA
| |
Collapse
|
36
|
Renaud D, Scholl-Bürgi S, Karall D, Michel M. Comparative Metabolomics in Single Ventricle Patients after Fontan Palliation: A Strong Case for a Targeted Metabolic Therapy. Metabolites 2023; 13:932. [PMID: 37623876 PMCID: PMC10456471 DOI: 10.3390/metabo13080932] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023] Open
Abstract
Most studies on single ventricle (SV) circulation take a physiological or anatomical approach. Although there is a tight coupling between cardiac contractility and metabolism, the metabolic perspective on this patient population is very recent. Early findings point to major metabolic disturbances, with both impaired glucose and fatty acid oxidation in the cardiomyocytes. Additionally, Fontan patients have systemic metabolic derangements such as abnormal glucose metabolism and hypocholesterolemia. Our literature review compares the metabolism of patients with a SV circulation after Fontan palliation with that of patients with a healthy biventricular (BV) heart, or different subtypes of a failing BV heart, by Pubmed review of the literature on cardiac metabolism, Fontan failure, heart failure (HF), ketosis, metabolism published in English from 1939 to 2023. Early evidence demonstrates that SV circulation is not only a hemodynamic burden requiring staged palliation, but also a metabolic issue with alterations similar to what is known for HF in a BV circulation. Alterations of fatty acid and glucose oxidation were found, resulting in metabolic instability and impaired energy production. As reported for patients with BV HF, stimulating ketone oxidation may be an effective treatment strategy for HF in these patients. Few but promising clinical trials have been conducted thus far to evaluate therapeutic ketosis with HF using a variety of instruments, including ketogenic diet, ketone esters, and sodium-glucose co-transporter-2 (SGLT2) inhibitors. An initial trial on a small cohort demonstrated favorable outcomes for Fontan patients treated with SGLT2 inhibitors. Therapeutic ketosis is worth considering in the treatment of Fontan patients, as ketones positively affect not only the myocardial energy metabolism, but also the global Fontan physiopathology. Induced ketosis seems promising as a concerted therapeutic strategy.
Collapse
Affiliation(s)
- David Renaud
- Fundamental and Biomedical Sciences, Paris-Cité University, 75006 Paris, France
- Health Sciences Faculty, Universidad Europea Miguel de Cervantes, 47012 Valladolid, Spain
- Fundacja Recover, 05-124 Skrzeszew, Poland
| | - Sabine Scholl-Bürgi
- Department of Child and Adolescent Health, Division of Pediatrics I—Inherited Metabolic Disorders, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Daniela Karall
- Department of Child and Adolescent Health, Division of Pediatrics I—Inherited Metabolic Disorders, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Miriam Michel
- Department of Child and Adolescent Health, Division of Pediatrics III—Cardiology, Pulmonology, Allergology and Cystic Fibrosis, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
37
|
Dyńka D, Kowalcze K, Charuta A, Paziewska A. The Ketogenic Diet and Cardiovascular Diseases. Nutrients 2023; 15:3368. [PMID: 37571305 PMCID: PMC10421332 DOI: 10.3390/nu15153368] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
The most common and increasing causes of death worldwide are cardiovascular diseases (CVD). Taking into account the fact that diet is a key factor, it is worth exploring this aspect of CVD prevention and therapy. The aim of this article is to assess the potential of the ketogenic diet in the prevention and treatment of CVD. The article is a comprehensive, meticulous analysis of the literature in this area, taking into account the most recent studies currently available. The ketogenic diet has been shown to have a multifaceted effect on the prevention and treatment of CVD. Among other aspects, it has a beneficial effect on the blood lipid profile, even compared to other diets. It shows strong anti-inflammatory and cardioprotective potential, which is due, among other factors, to the anti-inflammatory properties of the state of ketosis, the elimination of simple sugars, the restriction of total carbohydrates and the supply of omega-3 fatty acids. In addition, ketone bodies provide "rescue fuel" for the diseased heart by affecting its metabolism. They also have a beneficial effect on the function of the vascular endothelium, including improving its function and inhibiting premature ageing. The ketogenic diet has a beneficial effect on blood pressure and other CVD risk factors through, among other aspects, weight loss. The evidence cited is often superior to that for standard diets, making it likely that the ketogenic diet shows advantages over other dietary models in the prevention and treatment of cardiovascular diseases. There is a legitimate need for further research in this area.
Collapse
Affiliation(s)
| | | | | | - Agnieszka Paziewska
- Institute of Health Sciences, Faculty of Medical and Health Sciences, Siedlce University of Natural Sciences and Humanities, 08-110 Siedlce, Poland; (D.D.); (K.K.); (A.C.)
| |
Collapse
|
38
|
Guo Y, Cen XF, Li D, Qiu HL, Chen YJ, Zhang M, Huang SH, Xia H, Xu M. Identify Tcea3 as a novel anti-cardiomyocyte hypertrophy gene involved in fatty acid oxidation and oxidative stress. Front Cardiovasc Med 2023; 10:1137429. [PMID: 37404738 PMCID: PMC10315901 DOI: 10.3389/fcvm.2023.1137429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/07/2023] [Indexed: 07/06/2023] Open
Abstract
Background Chronic pressure overload triggers pathological cardiac hypertrophy that eventually leads to heart failure. Effective biomarkers and therapeutic targets for heart failure remain to be defined. The aim of this study is to identify key genes associated with pathological cardiac hypertrophy by combining bioinformatics analyses with molecular biology experiments. Methods Comprehensive bioinformatics tools were used to screen genes related to pressure overload-induced cardiac hypertrophy. We identified differentially expressed genes (DEGs) by overlapping three Gene Expression Omnibus (GEO) datasets (GSE5500, GSE1621, and GSE36074). Correlation analysis and BioGPS online tool were used to detect the genes of interest. A mouse model of cardiac remodeling induced by transverse aortic constriction (TAC) was established to verify the expression of the interest gene during cardiac remodeling by RT-PCR and western blot. By using RNA interference technology, the effect of transcription elongation factor A3 (Tcea3) silencing on PE-induced hypertrophy of neonatal rat ventricular myocytes (NRVMs) was detected. Next, gene set enrichment analysis (GSEA) and the online tool ARCHS4 were used to predict the possible signaling pathways, and the fatty acid oxidation relevant pathways were enriched and then verified in NRVMs. Furthermore, the changes of long-chain fatty acid respiration in NRVMs were detected using the Seahorse XFe24 Analyzer. Finally, MitoSOX staining was used to detect the effect of Tcea3 on mitochondrial oxidative stress, and the contents of NADP(H) and GSH/GSSG were detected by relevant kits. Results A total of 95 DEGs were identified and Tcea3 was negatively correlated with Nppa, Nppb and Myh7. The expression level of Tcea3 was downregulated during cardiac remodeling both in vivo and in vitro. Knockdown of Tcea3 aggravated cardiomyocyte hypertrophy induced by PE in NRVMs. GSEA and online tool ARCHS4 predict Tcea3 involved in fatty acid oxidation (FAO). Subsequently, RT-PCR results showed that knockdown of Tcea3 up-regulated Ces1d and Pla2g5 mRNA expression levels. In PE induced cardiomyocyte hypertrophy, Tcea3 silencing results in decreased fatty acid utilization, decreased ATP synthesis and increased mitochondrial oxidative stress. Conclusion Our study identifies Tcea3 as a novel anti-cardiac remodeling target by regulating FAO and governing mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Yingying Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xian-feng Cen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hong-liang Qiu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ya-jie Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Meng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Si-hui Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Man Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
39
|
Li X, Bi X. Integrated Control of Fatty Acid Metabolism in Heart Failure. Metabolites 2023; 13:615. [PMID: 37233656 PMCID: PMC10220550 DOI: 10.3390/metabo13050615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/18/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
Disrupted fatty acid metabolism is one of the most important metabolic features in heart failure. The heart obtains energy from fatty acids via oxidation. However, heart failure results in markedly decreased fatty acid oxidation and is accompanied by the accumulation of excess lipid moieties that lead to cardiac lipotoxicity. Herein, we summarized and discussed the current understanding of the integrated regulation of fatty acid metabolism (including fatty acid uptake, lipogenesis, lipolysis, and fatty acid oxidation) in the pathogenesis of heart failure. The functions of many enzymes and regulatory factors in fatty acid homeostasis were characterized. We reviewed their contributions to the development of heart failure and highlighted potential targets that may serve as promising new therapeutic strategies.
Collapse
Affiliation(s)
| | - Xukun Bi
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China;
| |
Collapse
|
40
|
Baloglu E. Hypoxic Stress-Dependent Regulation of Na,K-ATPase in Ischemic Heart Disease. Int J Mol Sci 2023; 24:ijms24097855. [PMID: 37175562 PMCID: PMC10177966 DOI: 10.3390/ijms24097855] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
In cardiomyocytes, regular activity of the Na,K-ATPase (NKA) and its Na/K pump activity is essential for maintaining ion gradients, excitability, propagation of action potentials, electro-mechanical coupling, trans-membrane Na+ and Ca2+ gradients and, thus, contractility. The activity of NKA is impaired in ischemic heart disease and heart failure, which has been attributed to decreased expression of the NKA subunits. Decreased NKA activity leads to intracellular Na+ and Ca2+ overload, diastolic dysfunction and arrhythmias. One signal likely related to these events is hypoxia, where hypoxia-inducible factors (HIF) play a critical role in the adaptation of cells to low oxygen tension. HIF activity increases in ischemic heart, hypertension, heart failure and cardiac fibrosis; thus, it might contribute to the impaired function of NKA. This review will mainly focus on the regulation of NKA in ischemic heart disease in the context of stressed myocardium and the hypoxia-HIF axis and argue on possible consequences of treatment.
Collapse
Affiliation(s)
- Emel Baloglu
- Department of Medical Pharmacology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey
| |
Collapse
|
41
|
Schenkl C, Heyne E, Doenst T, Schulze PC, Nguyen TD. Targeting Mitochondrial Metabolism to Save the Failing Heart. Life (Basel) 2023; 13:life13041027. [PMID: 37109556 PMCID: PMC10143865 DOI: 10.3390/life13041027] [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: 02/17/2023] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Despite considerable progress in treating cardiac disorders, the prevalence of heart failure (HF) keeps growing, making it a global medical and economic burden. HF is characterized by profound metabolic remodeling, which mostly occurs in the mitochondria. Although it is well established that the failing heart is energy-deficient, the role of mitochondria in the pathophysiology of HF extends beyond the energetic aspects. Changes in substrate oxidation, tricarboxylic acid cycle and the respiratory chain have emerged as key players in regulating myocardial energy homeostasis, Ca2+ handling, oxidative stress and inflammation. This work aims to highlight metabolic alterations in the mitochondria and their far-reaching effects on the pathophysiology of HF. Based on this knowledge, we will also discuss potential metabolic approaches to improve cardiac function.
Collapse
Affiliation(s)
- Christina Schenkl
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Paul Christian Schulze
- Department of Medicine I (Cardiology, Angiology, Critical Care Medicine), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Tien Dung Nguyen
- Department of Medicine I (Cardiology, Angiology, Critical Care Medicine), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
42
|
Abstract
The ketone bodies beta-hydroxybutyrate and acetoacetate are hepatically produced metabolites catabolized in extrahepatic organs. Ketone bodies are a critical cardiac fuel and have diverse roles in the regulation of cellular processes such as metabolism, inflammation, and cellular crosstalk in multiple organs that mediate disease. This review focuses on the role of cardiac ketone metabolism in health and disease with an emphasis on the therapeutic potential of ketosis as a treatment for heart failure (HF). Cardiac metabolic reprogramming, characterized by diminished mitochondrial oxidative metabolism, contributes to cardiac dysfunction and pathologic remodeling during the development of HF. Growing evidence supports an adaptive role for ketone metabolism in HF to promote normal cardiac function and attenuate disease progression. Enhanced cardiac ketone utilization during HF is mediated by increased availability due to systemic ketosis and a cardiac autonomous upregulation of ketolytic enzymes. Therapeutic strategies designed to restore high-capacity fuel metabolism in the heart show promise to address fuel metabolic deficits that underpin the progression of HF. However, the mechanisms involved in the beneficial effects of ketone bodies in HF have yet to be defined and represent important future lines of inquiry. In addition to use as an energy substrate for cardiac mitochondrial oxidation, ketone bodies modulate myocardial utilization of glucose and fatty acids, two vital energy substrates that regulate cardiac function and hypertrophy. The salutary effects of ketone bodies during HF may also include extra-cardiac roles in modulating immune responses, reducing fibrosis, and promoting angiogenesis and vasodilation. Additional pleotropic signaling properties of beta-hydroxybutyrate and AcAc are discussed including epigenetic regulation and protection against oxidative stress. Evidence for the benefit and feasibility of therapeutic ketosis is examined in preclinical and clinical studies. Finally, ongoing clinical trials are reviewed for perspective on translation of ketone therapeutics for the treatment of HF.
Collapse
Affiliation(s)
- Timothy R. Matsuura
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Patrycja Puchalska
- Department of Medicine, Division of Molecular Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Peter A. Crawford
- Department of Medicine, Division of Molecular Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Daniel P. Kelly
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
43
|
Metabolic landscape in cardiac aging: insights into molecular biology and therapeutic implications. Signal Transduct Target Ther 2023; 8:114. [PMID: 36918543 PMCID: PMC10015017 DOI: 10.1038/s41392-023-01378-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/06/2023] [Accepted: 02/20/2023] [Indexed: 03/16/2023] Open
Abstract
Cardiac aging is evident by a reduction in function which subsequently contributes to heart failure. The metabolic microenvironment has been identified as a hallmark of malignancy, but recent studies have shed light on its role in cardiovascular diseases (CVDs). Various metabolic pathways in cardiomyocytes and noncardiomyocytes determine cellular senescence in the aging heart. Metabolic alteration is a common process throughout cardiac degeneration. Importantly, the involvement of cellular senescence in cardiac injuries, including heart failure and myocardial ischemia and infarction, has been reported. However, metabolic complexity among human aging hearts hinders the development of strategies that targets metabolic susceptibility. Advances over the past decade have linked cellular senescence and function with their metabolic reprogramming pathway in cardiac aging, including autophagy, oxidative stress, epigenetic modifications, chronic inflammation, and myocyte systolic phenotype regulation. In addition, metabolic status is involved in crucial aspects of myocardial biology, from fibrosis to hypertrophy and chronic inflammation. However, further elucidation of the metabolism involvement in cardiac degeneration is still needed. Thus, deciphering the mechanisms underlying how metabolic reprogramming impacts cardiac aging is thought to contribute to the novel interventions to protect or even restore cardiac function in aging hearts. Here, we summarize emerging concepts about metabolic landscapes of cardiac aging, with specific focuses on why metabolic profile alters during cardiac degeneration and how we could utilize the current knowledge to improve the management of cardiac aging.
Collapse
|
44
|
Farooq M, Jorde UP. Impact of Sodium-Glucose Cotransporter-2 Inhibitors on Cardiac Bioenergetic Properties and Cardiorespiratory Fitness: A Special Effect of SGLT2i In Heart Failure? Cardiol Rev 2023; 31:65-69. [PMID: 35191660 DOI: 10.1097/crd.0000000000000424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recent clinical trials have highlighted the profound benefits of sodium-glucose linked transporter 2 inhibitors (SGLT2i) on cardiovascular mortality and hospitalization for heart failure patients. Modest improvements in glycemic, lipid, or blood pressure control are unlikely to contribute to these significant beneficial outcomes, generating much interest in the relevant mechanisms leading to outcome improvements. In this review, we discuss the current evidence supporting a shift in myocardial substrate utilization from carbohydrates and fat oxidation toward energy efficient ketone bodies in the failing heart and the role of SGLT2i in this key metabolic adaptation to optimize myocardial fuel energetics. Further insights into the effect of SGLT2i on the indices of cardiorespiratory fitness are outlined and provide important clues into their mechanism of benefit. This mechanistic discussion in the context of recent trials of SGLT2i denotes a promising treatment paradigm of heart failure in individuals with and without diabetes.
Collapse
Affiliation(s)
- Muhammad Farooq
- From the Division of Cardiology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY
| | | |
Collapse
|
45
|
Li C, Zhang X, Li J, Liang L, Zeng J, Wen M, Pan L, Lv D, Liu M, Cheng Y, Huang H. Ginsenoside Rb1 promotes the activation of PPARα pathway via inhibiting FADD to ameliorate heart failure. Eur J Pharmacol 2023; 947:175676. [PMID: 37001580 DOI: 10.1016/j.ejphar.2023.175676] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/07/2023] [Accepted: 03/22/2023] [Indexed: 04/01/2023]
Abstract
PURPOSE Ginsenoside Rb1 (GRb1), a dammarane-type triterpene saponin compound mainly distributed in ginseng (Panax ginseng), has been demonstrated to ameliorate cardiovascular diseases. However, it remains unclear whether GRb1 alleviates heart failure (HF) by maintaining cardiac energy metabolism balance. Therefore, this work aimed to investigate the cardiac benefits of GRb1 against cardiac energy deficit and explore its mechanism of action. METHODS AND RESULTS Isoproterenol (ISO) induced HF Sprague-Dawley rats were administrated with GRb1 or fenofibrate for 6 weeks. ISO-induced primary neonatal rat cardiomyocytes (NRCMs) were used as the in vitro model. In vivo, GRb1 significantly improved the structural and metabolic disorder, as demonstrated by the restoration of cardiac function, inhibition of cardiac hypertrophy and fibrosis, and increased adenosine triphosphate (ATP) generation. In vitro, GRb1 effectively protected mitochondrial function and scavenged excessive reactive oxygen species. Moreover, in ISO-induced NRCMs, GRb1 significantly inhibited the abnormal upregulation of Fas-associated death domain (FADD), promoted transcriptional activation of peroxisome proliferator-activated receptor-alpha (PPARα), improved the aberrant expression of cardiac energy metabolism-related enzymes and cardiac fatty acid oxidation, and subsequently increased the synthesis of ATP. Noticeably, GRb1 could inhibit the increased binding between FADD and PPARα, which contributed to the activation of PPARα. Furthermore, GRb1 strengthened the thermal stabilization of FADD and might bind to FADD directly. CONCLUSIONS Collectively, it's part of the in-depth mechanism of GRb1's cardio-protection that GRb1 could directly bind to FADD and counteract its negative role in the transcription of PPARα thus ameliorating cardiac energy derangement and HF.
Collapse
Affiliation(s)
- Chuting Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xuting Zhang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jie Li
- Medical Research Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Liyin Liang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jingran Zeng
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Min Wen
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Linjie Pan
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Dongxin Lv
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Min Liu
- Guangzhou University of Traditional Chinese Medicine First Affiliated Hospital, Guangzhou, 510405, China.
| | - Yuanyuan Cheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Heqing Huang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
46
|
The KLF7/PFKL/ACADL axis modulates cardiac metabolic remodelling during cardiac hypertrophy in male mice. Nat Commun 2023; 14:959. [PMID: 36810848 PMCID: PMC9944323 DOI: 10.1038/s41467-023-36712-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Abstract
The main hallmark of myocardial substrate metabolism in cardiac hypertrophy or heart failure is a shift from fatty acid oxidation to greater reliance on glycolysis. However, the close correlation between glycolysis and fatty acid oxidation and underlying mechanism by which causes cardiac pathological remodelling remain unclear. We confirm that KLF7 simultaneously targets the rate-limiting enzyme of glycolysis, phosphofructokinase-1, liver, and long-chain acyl-CoA dehydrogenase, a key enzyme for fatty acid oxidation. Cardiac-specific knockout and overexpression KLF7 induce adult concentric hypertrophy and infant eccentric hypertrophy by regulating glycolysis and fatty acid oxidation fluxes in male mice, respectively. Furthermore, cardiac-specific knockdown phosphofructokinase-1, liver or overexpression long-chain acyl-CoA dehydrogenase partially rescues the cardiac hypertrophy in adult male KLF7 deficient mice. Here we show that the KLF7/PFKL/ACADL axis is a critical regulatory mechanism and may provide insight into viable therapeutic concepts aimed at the modulation of cardiac metabolic balance in hypertrophied and failing heart.
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW Myocardial metabolism is intricately linked to cardiac function. Perturbations of cardiac energy metabolism result in an energy-starved heart and the development of contractile dysfunction. In this review, we discuss alterations in myocardial energy supply, transcriptional changes in response to different energy demands, and mitochondrial function in the development of heart failure. RECENT FINDINGS Recent studies on substrate modulation through modifying energy substrate supply have shown cardioprotective properties. In addition, large cardiovascular outcome trials of anti-diabetic agents have demonstrated prognostic benefit, suggesting the importance of myocardial metabolism in cardiac function. Understanding molecular and transcriptional controls of cardiac metabolism promises new research avenues for metabolic treatment targets. Future studies assessing the impact of substrate modulation on cardiac energetic status and function will better inform development of metabolic therapies.
Collapse
Affiliation(s)
- Sher May Ng
- Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
| | - Stefan Neubauer
- Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK
- Department of Cardiology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Oliver J Rider
- Department of Medicine, University of Oxford Centre for Clinical Magnetic Resonance Research, Oxford, UK.
- Department of Cardiology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
48
|
Targeting mitochondrial impairment for the treatment of cardiovascular diseases: From hypertension to ischemia-reperfusion injury, searching for new pharmacological targets. Biochem Pharmacol 2023; 208:115405. [PMID: 36603686 DOI: 10.1016/j.bcp.2022.115405] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023]
Abstract
Mitochondria and mitochondrial proteins represent a group of promising pharmacological target candidates in the search of new molecular targets and drugs to counteract the onset of hypertension and more in general cardiovascular diseases (CVDs). Indeed, several mitochondrial pathways result impaired in CVDs, showing ATP depletion and ROS production as common traits of cardiac tissue degeneration. Thus, targeting mitochondrial dysfunction in cardiomyocytes can represent a successful strategy to prevent heart failure. In this context, the identification of new pharmacological targets among mitochondrial proteins paves the way for the design of new selective drugs. Thanks to the advances in omics approaches, to a greater availability of mitochondrial crystallized protein structures and to the development of new computational approaches for protein 3D-modelling and drug design, it is now possible to investigate in detail impaired mitochondrial pathways in CVDs. Furthermore, it is possible to design new powerful drugs able to hit the selected pharmacological targets in a highly selective way to rescue mitochondrial dysfunction and prevent cardiac tissue degeneration. The role of mitochondrial dysfunction in the onset of CVDs appears increasingly evident, as reflected by the impairment of proteins involved in lipid peroxidation, mitochondrial dynamics, respiratory chain complexes, and membrane polarization maintenance in CVD patients. Conversely, little is known about proteins responsible for the cross-talk between mitochondria and cytoplasm in cardiomyocytes. Mitochondrial transporters of the SLC25A family, in particular, are responsible for the translocation of nucleotides (e.g., ATP), amino acids (e.g., aspartate, glutamate, ornithine), organic acids (e.g. malate and 2-oxoglutarate), and other cofactors (e.g., inorganic phosphate, NAD+, FAD, carnitine, CoA derivatives) between the mitochondrial and cytosolic compartments. Thus, mitochondrial transporters play a key role in the mitochondria-cytosol cross-talk by leading metabolic pathways such as the malate/aspartate shuttle, the carnitine shuttle, the ATP export from mitochondria, and the regulation of permeability transition pore opening. Since all these pathways are crucial for maintaining healthy cardiomyocytes, mitochondrial carriers emerge as an interesting class of new possible pharmacological targets for CVD treatments.
Collapse
|
49
|
Ranea-Robles P, Pavlova NN, Bender A, Pereyra AS, Ellis JM, Stauffer B, Yu C, Thompson CB, Argmann C, Puchowicz M, Houten SM. A mitochondrial long-chain fatty acid oxidation defect leads to transfer RNA uncharging and activation of the integrated stress response in the mouse heart. Cardiovasc Res 2022; 118:3198-3210. [PMID: 35388887 PMCID: PMC9799058 DOI: 10.1093/cvr/cvac050] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 03/08/2022] [Accepted: 03/23/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS Cardiomyopathy and arrhythmias can be severe presentations in patients with inherited defects of mitochondrial long-chain fatty acid β-oxidation (FAO). The pathophysiological mechanisms that underlie these cardiac abnormalities remain largely unknown. We investigated the molecular adaptations to a FAO deficiency in the heart using the long-chain acyl-CoA dehydrogenase (LCAD) knockout (KO) mouse model. METHODS AND RESULTS We observed enrichment of amino acid metabolic pathways and of ATF4 target genes among the upregulated genes in the LCAD KO heart transcriptome. We also found a prominent activation of the eIF2α/ATF4 axis at the protein level that was independent of the feeding status, in addition to a reduction of cardiac protein synthesis during a short period of food withdrawal. These findings are consistent with an activation of the integrated stress response (ISR) in the LCAD KO mouse heart. Notably, charging of several transfer RNAs (tRNAs), such as tRNAGln was decreased in LCAD KO hearts, reflecting a reduced availability of cardiac amino acids, in particular, glutamine. We replicated the activation of the ISR in the hearts of mice with muscle-specific deletion of carnitine palmitoyltransferase 2. CONCLUSIONS Our results show that perturbations in amino acid metabolism caused by long-chain FAO deficiency impact cardiac metabolic signalling, in particular the ISR. These results may serve as a foundation for investigating the role of the ISR in the cardiac pathology associated with long-chain FAO defects.Translational Perspective: The heart relies mainly on mitochondrial fatty acid β-oxidation (FAO) for its high energy requirements. The heart disease observed in patients with a genetic defect in this pathway highlights the importance of FAO for cardiac health. We show that the consequences of a FAO defect extend beyond cardiac energy homeostasis and include amino acid metabolism and associated signalling pathways such as the integrated stress response.
Collapse
Affiliation(s)
- Pablo Ranea-Robles
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1498, New York, NY 10029, USA
| | - Natalya N Pavlova
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Aaron Bender
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1498, New York, NY 10029, USA
| | - Andrea S Pereyra
- Brody School of Medicine at East Carolina University, Department of Physiology, and East Carolina Diabetes and Obesity Institute, Greenville, NC 27858, USA
| | - Jessica M Ellis
- Brody School of Medicine at East Carolina University, Department of Physiology, and East Carolina Diabetes and Obesity Institute, Greenville, NC 27858, USA
| | - Brandon Stauffer
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1498, New York, NY 10029, USA
- Mount Sinai Genomics, Inc, Stamford, CT 06902, USA
| | - Chunli Yu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1498, New York, NY 10029, USA
- Mount Sinai Genomics, Inc, Stamford, CT 06902, USA
| | - Craig B Thompson
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carmen Argmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1498, New York, NY 10029, USA
| | - Michelle Puchowicz
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sander M Houten
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Box 1498, New York, NY 10029, USA
| |
Collapse
|
50
|
Maciejewska-Skrendo A, Massidda M, Tocco F, Leźnicka K. The Influence of the Differentiation of Genes Encoding Peroxisome Proliferator-Activated Receptors and Their Coactivators on Nutrient and Energy Metabolism. Nutrients 2022; 14:nu14245378. [PMID: 36558537 PMCID: PMC9782515 DOI: 10.3390/nu14245378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/27/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Genetic components may play an important role in the regulation of nutrient and energy metabolism. In the presence of specific genetic variants, metabolic dysregulation may occur, especially in relation to the processes of digestion, assimilation, and the physiological utilization of nutrients supplied to the body, as well as the regulation of various metabolic pathways and the balance of metabolic changes, which may consequently affect the effectiveness of applied reduction diets and weight loss after training. There are many well-documented studies showing that the presence of certain polymorphic variants in some genes can be associated with specific changes in nutrient and energy metabolism, and consequently, with more or less desirable effects of applied caloric reduction and/or exercise intervention. This systematic review focused on the role of genes encoding peroxisome proliferator-activated receptors (PPARs) and their coactivators in nutrient and energy metabolism. The literature review prepared showed that there is a link between the presence of specific alleles described at different polymorphic points in PPAR genes and various human body characteristics that are crucial for the efficacy of nutritional and/or exercise interventions. Genetic analysis can be a valuable element that complements the work of a dietitian or trainer, allowing for the planning of a personalized diet or training that makes the best use of the innate metabolic characteristics of the person who is the subject of their interventions.
Collapse
Affiliation(s)
- Agnieszka Maciejewska-Skrendo
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland
- Institute of Physical Culture Sciences, University of Szczecin, 71-065 Szczecin, Poland
- Correspondence:
| | - Myosotis Massidda
- Department of Medical Sciences and Public Health, Faculty of Medicine and Surgery, Sport and Exercise Sciences Degree Courses, University of Cagliari, 72-09124 Cagliari, Italy
| | - Filippo Tocco
- Department of Medical Sciences and Public Health, Faculty of Medicine and Surgery, Sport and Exercise Sciences Degree Courses, University of Cagliari, 72-09124 Cagliari, Italy
| | - Katarzyna Leźnicka
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland
| |
Collapse
|