1
|
Guldbrandsen H, Gopalasingam N, Christensen KH, Hørsdal OK, Nielsen R, Wiggers H, Berg-Hansen K. Cardiovascular and Metabolic Effects of Modulating Circulating Ketone Bodies With 1,3-Butanediol in Patients With Heart Failure With Reduced Ejection Fraction. J Am Heart Assoc 2025; 14:e038461. [PMID: 39719429 DOI: 10.1161/jaha.124.038461] [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: 08/29/2024] [Accepted: 11/21/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND Oral treatment with the exogenous ketone body 3-hydroxybutyrate improves cardiac function in patients with heart failure with reduced ejection fraction, but ketosis is limited to 3 to 4 hours. Treatment with (R)-1,3-butanediol (BD) provides prolonged ketosis in healthy controls, but the hemodynamic and metabolic profile is unexplored in patients with heart failure with reduced ejection fraction. METHODS AND RESULTS This was a randomized, single-blind, placebo-controlled, crossover study. Transthoracic echocardiography and venous blood samples were performed at baseline and hourly for 6 hours after an oral dose of BD (0.5 g/kg) or taste-matched placebo. The primary end point was the average between-treatment difference in cardiac output during the 6-hour period after intake. Secondary end points were stroke volume, heart rate, left ventricular ejection fraction, circulating 3-hydroxybutyrate, and free fatty acids. Twelve patients with heart failure with reduced ejection fraction were included. BD treatment provided significant increase in circulating 3-hydroxybutyrate by 1400 μmol/L (95% CI, 1262-1538 μmol/L, P<0.001) and increased cardiac output by 0.9 L/min (95% CI, 0.7-1.1 L/min, P<0.001) compared with placebo. Stroke volume increased by 15 mL (95% CI, 11-19 mL, P<0.001), and heart rate remained similar between treatments (P=0.150). Left ventricular ejection fraction increased by 3 percentage points (95% CI, 1-4 percentage points, P<0.001). Global longitudinal strain improved (P<0.001). Left ventricular contractility estimates increased after BD intake, and parameters of afterload were reduced. Finally, free fatty acids and glucose levels decreased. CONCLUSIONS Oral dosing of BD led to prolonged ketosis and cardiovascular and metabolic benefits in patients with heart failure with reduced ejection fraction. Treatment with BD is an attractive option to achieve beneficial effects from sustained therapeutic ketosis. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT05768100.
Collapse
Affiliation(s)
- Halvor Guldbrandsen
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
- Department of Cardiology Viborg Regional Hospital Viborg Denmark
- Department of Clinical Medicine, Faculty of Health Aarhus University Aarhus Denmark
| | - Nigopan Gopalasingam
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
- Department of Clinical Medicine, Faculty of Health Aarhus University Aarhus Denmark
- Department of Cardiology Gødstrup Hospital Herning Denmark
| | - Kristian Hylleberg Christensen
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
- Department of Clinical Medicine, Faculty of Health Aarhus University Aarhus Denmark
| | - Oskar Kjærgaard Hørsdal
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
- Department of Clinical Medicine, Faculty of Health Aarhus University Aarhus Denmark
| | - Roni Nielsen
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
- Department of Clinical Medicine, Faculty of Health Aarhus University Aarhus Denmark
| | - Henrik Wiggers
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
- Department of Clinical Medicine, Faculty of Health Aarhus University Aarhus Denmark
| | - Kristoffer Berg-Hansen
- Department of Cardiology Aarhus University Hospital Aarhus Denmark
- Department of Clinical Medicine, Faculty of Health Aarhus University Aarhus Denmark
| |
Collapse
|
2
|
Solis-Herrera C, Qin Y, Honka H, Cersosimo E, Triplitt C, Neppala S, Rajan J, Acosta FM, Moody AJ, Iozzo P, Fox P, Clarke G, DeFronzo RA. Effect of Hyperketonemia on Myocardial Function in Patients With Heart Failure and Type 2 Diabetes. Diabetes 2025; 74:43-52. [PMID: 39446133 DOI: 10.2337/db24-0406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024]
Abstract
We examined the effect of increased levels of plasma ketones on left ventricular (LV) function, myocardial glucose uptake (MGU), and myocardial blood flow (MBF) in patients with type 2 diabetes (T2DM) with heart failure. Three groups of patients with T2DM (n = 12 per group) with an LV ejection fraction (EF) ≤50% received incremental infusions of β-hydroxybutyrate (β-OH-B) for 3-6 h to increase the plasma β-OH-B concentration throughout the physiologic (groups I and II) and pharmacologic (group III) range. Cardiac MRI was performed at baseline and after each β-OH-B infusion to provide measures of cardiac function. On a separate day, group II also received a sodium bicarbonate (NaHCO3) infusion, thus serving as their own control for time, volume, and pH. Additionally, group II underwent positron emission tomography study with 18F-fluoro-2-deoxyglucose to examine effect of hyperketonemia on MGU. Groups I, II, and III achieved plasma β-OH-B levels (mean ± SEM) of 0.7 ± 0.3, 1.6 ± 0.2, 3.2 ± 0.2 mmol/L, respectively. Cardiac output (CO), LVEF, and stroke volume (SV) increased significantly during β-OH-B infusion in groups II (CO, from 4.54 to 5.30; EF, 39.9 to 43.8; SV, 70.3 to 80.0) and III (CO, from 5.93 to 7.16; EF, 41.1 to 47.5; SV, 89.0 to 108.4), and did not change with NaHCO3 infusion in group II. The increase in LVEF was greatest in group III (P < 0.001 vs. group II). MGU and MBF were not altered by β-OH-B. In patients with T2DM and LVEF ≤50%, increased plasma β-OH-B level significantly increased LV function dose dependently. Because MGU did not change, the myocardial benefit of β-OH-B resulted from providing an additional fuel for the heart without inhibiting MGU. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Carolina Solis-Herrera
- Division of Endocrinology, Department of Medicine, University of Texas Health Science Center, San Antonio, TX
| | - Yuejuan Qin
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center, San Antonio, TX
| | - Henri Honka
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center, San Antonio, TX
- Department of Internal Medicine, University of Turku, Turku, Finland
| | - Eugenio Cersosimo
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center, San Antonio, TX
- Texas Diabetes Institute, University Health System, San Antonio, TX
| | - Curtis Triplitt
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center, San Antonio, TX
- Texas Diabetes Institute, University Health System, San Antonio, TX
| | - Sivaram Neppala
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center, San Antonio, TX
| | - Jemena Rajan
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center, San Antonio, TX
| | - Francisca M Acosta
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX
| | - Alexander J Moody
- Department of Radiology and Research Imaging Institute, University of Texas Health Science Center, San Antonio, TX
| | - Patricio Iozzo
- Institute of Clinical Physiology, Consiglio Nazionale delle Ricerche, Pisa, Italy
| | - Peter Fox
- Department of Radiology and Research Imaging Institute, University of Texas Health Science Center, San Antonio, TX
| | - Geoffrey Clarke
- Department of Radiology and Research Imaging Institute, University of Texas Health Science Center, San Antonio, TX
| | - Ralph A DeFronzo
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center, San Antonio, TX
- Texas Diabetes Institute, University Health System, San Antonio, TX
| |
Collapse
|
3
|
Kanta JM, Lundsgaard AM, Havelund JF, Armour SL, Bæk O, Nguyen DN, Richter EA, Knudsen JG, Kleinert M, Færgeman NJ, Fritzen AM, Kiens B. Metabolic effects of medium-chain triacylglycerol consumption are preserved in obesity. Am J Physiol Endocrinol Metab 2025; 328:E1-E20. [PMID: 39441241 DOI: 10.1152/ajpendo.00234.2024] [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: 06/24/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Several health-beneficial effects are associated with intake of medium-chain triacylglycerol (MCT); however, the underlying mechanisms are unknown. Furthermore, it remains uncertain whether the acute metabolic effects of MCT differ between lean individuals and individuals with obesity-and whether these effects are sustained following chronic intake. This study aimed to elucidate the postprandial physiological and metabolic effects of MCT before and after 8 days intake compared with intake of energy-matched triacylglycerol consisting of long-chain fatty acids (long-chain triacylglycerols, LCT) using a randomized cross-over design in lean individuals (n = 8) and individuals with obesity (n = 8). The study revealed that consumption of MCT increased ketogenesis and metabolic rate while lowering blood glucose levels over 5 h. The hypoglycemic action of MCT intake was accompanied by a concomitant transient increase in plasma insulin and glucagon levels. Interestingly, the effects on ketogenesis, metabolic rate, and glycemia were preserved in individuals with obesity and sustained after 8 days of daily supplementation. Lipidomic plasma analysis in lean individuals (n = 4) showed that a part of the ingested MCT bypasses the liver and enters the systemic circulation as medium-chain fatty acids (MCFAs). The findings suggest that MCFAs, along with ketone bodies from the liver, may act as signaling molecules and/or substrates in the peripheral tissues, thereby contributing to the effects of MCT intake. In summary, these findings underscore the health benefits of MCT in metabolically compromised individuals after daily supplementation. Moreover, we uncover novel aspects of MCFA biology, providing insights into how these fatty acids orchestrate physiological effects in humans.NEW & NOTEWORTHY We reveal that medium-chain triacylglycerol (MCT) intake increases postprandial ketogenesis and metabolic rate and reduces plasma glucose levels in humans. Notably, these responses persist in individuals with obesity and are maintained following chronic MCT supplementation. Some medium-chain fatty acids entered the circulation, suggesting that these, together with ketone bodies, act as signaling molecules/substrates in peripheral tissues. The findings highlight health beneficial effects of dietary MCT in individuals with obesity and reveal new insights into lipid biology.
Collapse
Affiliation(s)
- Josephine M Kanta
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Anne-Marie Lundsgaard
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
- Novo Nordisk A/S, Søborg, Denmark
| | - Jesper F Havelund
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Sara L Armour
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Denmark
| | - Ole Bæk
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
- Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
| | - Erik A Richter
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Jakob G Knudsen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Denmark
| | - Maximilian Kleinert
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
- Muscle Physiology and Metabolism Group, German Institute of Human Nutrition (DIfE), Potsdam-Rehbruecke, Germany
| | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Andreas M Fritzen
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, University of Copenhagen, Denmark
| | - Bente Kiens
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| |
Collapse
|
4
|
Madhavan SS, Stubbs BJ. Beta-hydroxybutyrate. Trends Endocrinol Metab 2025; 36:96-97. [PMID: 39765208 PMCID: PMC11707391 DOI: 10.1016/j.tem.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 01/11/2025]
Affiliation(s)
- Sidharth S Madhavan
- Buck Institute for Research on Aging, Novato, CA, USA; Leonard Davis School of Gerontology at University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
5
|
Chen Y, You Y, Wang X, Jin Y, Zeng Y, Pan Z, Li D, Ling W. β-Hydroxybutyrate Alleviates Atherosclerotic Calcification by Inhibiting Endoplasmic Reticulum Stress-Mediated Apoptosis via AMPK/Nrf2 Pathway. Nutrients 2024; 17:111. [PMID: 39796543 DOI: 10.3390/nu17010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Atherosclerotic calcification (AC) is a common feature of atherosclerotic cardiovascular disease. β-Hydroxybutyrate (BHB) has been identified as a molecule that influences cardiovascular disease. However, whether BHB can influence AC is still unknown. METHODS AND RESULTS In this study, ApoE-/- mice, fed a Western diet, were used to examine the effects of BHB on AC. Rat vascular smooth muscle cells (VSMCs) were used to verify the impacts of BHB on AC and to explore the underlying mechanisms. The results show that Western diet-challenged ApoE-/- mice, supplemented with BHB for 24 weeks, exhibited reduced calcified areas, calcium content, and alkaline phosphatase (ALP) activity in the aortas, as well as ameliorated severity of AC. Furthermore, BHB downregulated the expression of glucose-regulated protein 78 (GRP78) and C/EBP homologous protein (CHOP), thereby reducing endoplasmic reticulum stress (ERS) and ERS-mediated apoptosis in the aortas of the mice. Consistently, in vitro studies showed that BHB reduced ALP activity and calcium content in VSMCs, and inhibited VSMC calcification. Additionally, BHB suppressed ERS-mediated apoptosis in VSMCs. CONCLUSIONS In summary, the present results demonstrate that BHB can alleviate atherosclerotic calcification by inhibiting ERS-mediated apoptosis. Therefore, BHB may serve as a viable therapeutic agent for AC.
Collapse
Affiliation(s)
- Yu Chen
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yiran You
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xin Wang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yufeng Jin
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yupeng Zeng
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zhijun Pan
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Dan Li
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750101, China
| |
Collapse
|
6
|
Ferrannini E, Baldi S, Scozzaro MT, Ferrannini G, Hansen MK. Fasting substrates predict chronic kidney disease progression in CREDENCE trial patients with type 2 diabetes. JCI Insight 2024; 9:e180637. [PMID: 39704168 DOI: 10.1172/jci.insight.180637] [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: 02/26/2024] [Accepted: 11/05/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUNDSodium-glucose cotransporter 2 inhibitors slow down progression of chronic kidney disease (CKD). We tested whether the circulating substrate mix is related to CKD progression and cardiovascular outcomes in patients with type 2 diabetes (T2D) and albuminuric CKD in the CREDENCE trial.METHODSWe measured fasting substrates in 2,543 plasma samples at baseline and 1 year after randomization to either 100 mg canagliflozin or placebo and used multivariate Cox models to explore their association with CKD progression, heart failure hospitalization/cardiovascular death (hHF/CVD), and mortality.RESULTSHigher baseline lactate and free fatty acids (FFAs) were independently associated with a lower risk of CKD progression (HR = 0.73 [95% CI: 0.54-0.98] and HR = 0.67 [95% CI: 0.48-0.95], respectively) and hHF/CVD HR = 0.70 [95% CI: 0.50-0.99] and HR = 0.63 [95% CI: 0.42-0.94]). Canagliflozin led to a rise in plasma FFAs, glycerol, β-hydroxybutyrate, and acetoacetate. Changes in substrate between baseline and year 1 predicted an approximately 30% reduction in relative risk of both CKD progression and hHF/CVD independently of treatment. More patients who did not respond to canagliflozin treatment in terms of CKD progression belonged to the bottom lactate and FFA distribution tertiles.CONCLUSIONIn T2D patients with albuminuric CKD, basic energy substrates selectively influenced major long-term endpoints; canagliflozin treatment amplified their effects by chronically raising their circulating levels.
Collapse
Affiliation(s)
- Ele Ferrannini
- CNR (National Research Council) Institute of Clinical Physiology, Pisa, Italy
| | - Simona Baldi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Giulia Ferrannini
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Internal Medicine Unit, Södertälje Hospital, Stockholm, Sweden
| | | |
Collapse
|
7
|
Berger JH, Finck BN. Beyond ketosis: the search for the mechanism underlying SGLT2-inhibitor benefit continues. J Clin Invest 2024; 134:e187097. [PMID: 39680453 DOI: 10.1172/jci187097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024] Open
Abstract
Despite the impressive clinical benefits and widespread adoption of sodium glucose cotransporter 2 inhibitors (SGLT2i) to treat all classes of heart failure, their cardiovascular mechanisms of action are poorly understood. Proposed mechanisms range broadly and include enhanced ketogenesis, where the mild ketosis associated with SGLT2i use is presumed to be beneficial. However, in this issue of the JCI, carefully conducted metabolic flux studies by Goedeke et al. comparing the effects of SGLT2i and exogenous ketones suggest differential effects. Thus, the mechanisms of action for SGLT2i are likely pleiotropic, and further work is needed to fully understand their beneficial effects.
Collapse
Affiliation(s)
- Justin H Berger
- Division of Pediatric Cardiology, Department of Pediatrics and
| | - Brian N Finck
- Division of Nutritional Science and Obesity Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
8
|
Goedeke L, Ma Y, Gaspar RC, Nasiri A, Lee J, Zhang D, Galsgaard KD, Hu X, Zhang J, Guerrera N, Li X, LaMoia T, Hubbard BT, Haedersdal S, Wu X, Stack J, Dufour S, Butrico GM, Kahn M, Perry RJ, Cline GW, Young LH, Shulman GI. SGLT2 inhibition alters substrate utilization and mitochondrial redox in healthy and failing rat hearts. J Clin Invest 2024; 134:e176708. [PMID: 39680452 DOI: 10.1172/jci176708] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 10/17/2024] [Indexed: 12/18/2024] Open
Abstract
Previous studies highlight the potential for sodium-glucose cotransporter type 2 (SGLT2) inhibitors (SGLT2i) to exert cardioprotective effects in heart failure by increasing plasma ketones and shifting myocardial fuel utilization toward ketone oxidation. However, SGLT2i have multiple in vivo effects and the differential impact of SGLT2i treatment and ketone supplementation on cardiac metabolism remains unclear. Here, using gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) methodology combined with infusions of [13C6]glucose or [13C4]βOHB, we demonstrate that acute SGLT2 inhibition with dapagliflozin shifts relative rates of myocardial mitochondrial metabolism toward ketone oxidation, decreasing pyruvate oxidation with little effect on fatty acid oxidation in awake rats. Shifts in myocardial ketone oxidation persisted when plasma glucose levels were maintained. In contrast, acute βOHB infusion similarly augmented ketone oxidation, but markedly reduced fatty acid oxidation and did not alter glucose uptake or pyruvate oxidation. After inducing heart failure, dapagliflozin increased relative rates of ketone and fatty acid oxidation, but decreased pyruvate oxidation. Dapagliflozin increased mitochondrial redox and reduced myocardial oxidative stress in heart failure, which was associated with improvements in left ventricular ejection fraction after 3 weeks of treatment. Thus, SGLT2i have pleiotropic effects on systemic and heart metabolism, which are distinct from ketone supplementation and may contribute to the long-term cardioprotective benefits of SGLT2i.
Collapse
Affiliation(s)
- Leigh Goedeke
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Medicine (Cardiology) and The Cardiovascular Research Institute and
- Department of Medicine (Endocrinology) and The Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yina Ma
- Department of Internal Medicine (Cardiovascular Medicine) and The Yale Cardiovascular Research Center, Yale School of Medicine, New Haven Connecticut, USA
| | - Rafael C Gaspar
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Ali Nasiri
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Jieun Lee
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Dongyan Zhang
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Katrine Douglas Galsgaard
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Xiaoyue Hu
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Jiasheng Zhang
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Nicole Guerrera
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Xiruo Li
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Traci LaMoia
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Brandon T Hubbard
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Sofie Haedersdal
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Clinical Research, Copenhagen University Hospital, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Xiaohong Wu
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - John Stack
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Sylvie Dufour
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Gina Marie Butrico
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Mario Kahn
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Rachel J Perry
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Gary W Cline
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
| | - Lawrence H Young
- Department of Internal Medicine (Cardiovascular Medicine) and The Yale Cardiovascular Research Center, Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
| | - Gerald I Shulman
- Department of Internal Medicine (Endocrinology), Yale School of Medicine, New Haven Connecticut, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven Connecticut, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
9
|
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
|
10
|
Kansakar U, Nieves Garcia C, Santulli G, Gambardella J, Mone P, Jankauskas SS, Lombardi A. Exogenous Ketones in Cardiovascular Disease and Diabetes: From Bench to Bedside. J Clin Med 2024; 13:7391. [PMID: 39685849 DOI: 10.3390/jcm13237391] [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: 10/17/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Ketone bodies are molecules produced from fatty acids in the liver that act as energy carriers to peripheral tissues when glucose levels are low. Carbohydrate- and calorie-restricted diets, known to increase the levels of circulating ketone bodies, have attracted significant attention in recent years due to their potential health benefits in several diseases. Specifically, increasing ketones through dietary modulation has been reported to be beneficial for cardiovascular health and to improve glucose homeostasis and insulin resistance. Interestingly, although excessive production of ketones may lead to life-threatening ketoacidosis in diabetic patients, mounting evidence suggests that modest levels of ketones play adaptive and beneficial roles in pancreatic beta cells, although the exact mechanisms are still unknown. Of note, Sodium-Glucose Transporter 2 (SGLT2) inhibitors have been shown to increase the levels of beta-hydroxybutyrate (BHB), the most abundant ketone circulating in the human body, which may play a pivotal role in mediating some of their protective effects in cardiovascular health and diabetes. This systematic review provides a comprehensive overview of the scientific literature and presents an analysis of the effects of ketone bodies on cardiovascular pathophysiology and pancreatic beta cell function. The evidence from both preclinical and clinical studies indicates that exogenous ketones may have significant beneficial effects on both cardiomyocytes and pancreatic beta cells, making them intriguing candidates for potential cardioprotective therapies and to preserve beta cell function in patients with diabetes.
Collapse
Affiliation(s)
- Urna Kansakar
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Crystal Nieves Garcia
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Gaetano Santulli
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Jessica Gambardella
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Pasquale Mone
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Medicine and Health Sciences, University of Molise, 86100 Campobasso, Italy
- Casa di Cura Montevergine, 83013 Mercogliano, Avellino, Italy
| | - Stanislovas S Jankauskas
- Department of Molecular Pharmacology, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Angela Lombardi
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
| |
Collapse
|
11
|
Ng YH, Koay YC, Marques FZ, Kaye DM, O’Sullivan JF. Leveraging metabolism for better outcomes in heart failure. Cardiovasc Res 2024; 120:1835-1850. [PMID: 39351766 PMCID: PMC11630082 DOI: 10.1093/cvr/cvae216] [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: 03/09/2024] [Revised: 06/26/2024] [Accepted: 08/07/2024] [Indexed: 12/11/2024] Open
Abstract
Whilst metabolic inflexibility and substrate constraint have been observed in heart failure for many years, their exact causal role remains controversial. In parallel, many of our fundamental assumptions about cardiac fuel use are now being challenged like never before. For example, the emergence of sodium-glucose cotransporter 2 inhibitor therapy as one of the four 'pillars' of heart failure therapy is causing a revisit of metabolism as a key mechanism and therapeutic target in heart failure. Improvements in the field of cardiac metabolomics will lead to a far more granular understanding of the mechanisms underpinning normal and abnormal human cardiac fuel use, an appreciation of drug action, and novel therapeutic strategies. Technological advances and expanding biorepositories offer exciting opportunities to elucidate the novel aspects of these metabolic mechanisms. Methodologic advances include comprehensive and accurate substrate quantitation such as metabolomics and stable-isotope fluxomics, improved access to arterio-venous blood samples across the heart to determine fuel consumption and energy conversion, high quality cardiac tissue biopsies, biochemical analytics, and informatics. Pairing these technologies with recent discoveries in epigenetic regulation, mitochondrial dynamics, and organ-microbiome metabolic crosstalk will garner critical mechanistic insights in heart failure. In this state-of-the-art review, we focus on new metabolic insights, with an eye on emerging metabolic strategies for heart failure. Our synthesis of the field will be valuable for a diverse audience with an interest in cardiac metabolism.
Collapse
Affiliation(s)
- Yann Huey Ng
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room 3E71 D17, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Office 3E72, Camperdown, NSW 2006, Australia
| | - Yen Chin Koay
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room 3E71 D17, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Office 3E72, Camperdown, NSW 2006, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, VIC 3800, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC 3800, Australia
- Victorian Heart Institute, Monash University, Melbourne, VIC 3800, Australia
| | - David M Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC 3800, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, VIC 3004, Australia
- Monash-Alfred-Baker Centre for Cardiovascular Research, Monash University, Melbourne, VIC 3800, Australia
| | - John F O’Sullivan
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Room 3E71 D17, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Office 3E72, Camperdown, NSW 2006, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- Department of Medicine, Technische Univeristat Dresden, 01062 Dresden, Germany
| |
Collapse
|
12
|
Abstract
OBJECTIVES A comprehensive review of the current literature was conducted to summarize the potential therapeutic and management roles of ketogenic diet (KD) for cardiovascular disease (CVD). BACKGROUND Consensus has not been reached on the optimal diet for individuals with cardiovascular risk factors. KDs are characterized by high-fat, low-carbohydrate, and appropriate protein content, and have gained popularity in recent years in the management of various conditions, including cardiovascular and metabolic diseases. METHODS Original research, systematic reviews, and meta-analyses available in the PubMed, Web of Science, and Google Scholar databases were reviewed. RESULTS The current body of preclinical and clinical evidence on the efficacy of KD in the management of CVD remains limited. Specific applications of KD seem to suggest a positive impact on management of CVD. However, conflicting results and a lack of precise molecular and biochemical mechanisms of action provide ample opportunity for future investigation. CONCLUSION More multidisciplinary studies are needed to determine the true clinical benefit of KD in the management of CVD and so justify its expanded clinical use.
Collapse
Affiliation(s)
- Mohamed S Zaghloul
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Santiago Elizondo-Benedetto
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Mohamed A Zayed
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Molecular Cell Biology, Washington University School of Medicine, St. Louis, MO, USA
- McKelvey School of Engineering, Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
- Department of Surgery, Veterans Affairs St. Louis Health Care System, St. Louis, MO, USA
| |
Collapse
|
13
|
Haghighat L, DeJong C, Teerlink JR. New and future heart failure drugs. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1389-1407. [PMID: 39632985 DOI: 10.1038/s44161-024-00576-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/30/2024] [Indexed: 12/07/2024]
Abstract
In the past decade, our understanding of heart failure pathophysiology has advanced significantly, resulting in the development of new medications such as angiotensin-neprilysin inhibitors, sodium-glucose cotransporter-2 inhibitors and oral soluble guanylate cyclase stimulators. Backed by positive findings from large randomized controlled trials, recommendations for their use were recently included in the 2022 AHA/ACC/HFSA guidelines and 2023 ESC guidelines for management of heart failure. Promising drugs for future heart failure treatment include agents that modulate the neurohormonal system, vasodilators, anti-inflammatory drugs, mitotropes, which improve deranged energy metabolism of the failing heart, and myotropes, which increase cardiac contractility by affecting cardiac sarcomere function. Here, we discuss these new and future heart failure drugs. We explain their mechanisms of action, critically evaluate their performance in clinical trials and summarize the clinical scenarios in which the latest guidelines recommend their use. This Review aims to offer clinicians and researchers a comprehensive overview of novel therapeutic classes in heart failure treatment.
Collapse
Affiliation(s)
- Leila Haghighat
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
- Sutter Health, Palo Alto Medical Foundation, Burlingame, CA, USA
| | - Colette DeJong
- Section of Cardiology, VA Palo Alto Health Care System and School of Medicine, Stanford University, Palo Alto, CA, USA
| | - John R Teerlink
- Section of Cardiology, San Francisco Veterans Affairs Medical Center and School of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
14
|
Maduray K, Zhong J. Emerging roles of ketone bodies in cardiac fibrosis. Am J Physiol Cell Physiol 2024; 327:C1416-C1432. [PMID: 39401423 DOI: 10.1152/ajpcell.00241.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 12/11/2024]
Abstract
Cardiac fibrosis, characterized by excessive extracellular matrix (ECM) deposition within the myocardium, poses a significant challenge in cardiovascular health, contributing to various cardiac pathologies. Ketone bodies (KBs), particularly β-hydroxybutyrate (β-OHB), have emerged as subjects of interest due to their potential cardioprotective effects. However, their specific influence on cardiac fibrosis remains underexplored. This literature review comprehensively examines the relationship between KBs and cardiac fibrosis, elucidating potential mechanisms through which KBs modulate fibrotic pathways. A multifaceted interplay exists between KBs and key mediators of cardiac fibrosis. While some studies indicate a profibrotic role for KBs, others highlight their potential to attenuate fibrosis and cardiac remodeling. Mechanistically, KBs may regulate fibrotic pathways through modulation of cellular components such as cardiac fibroblasts, macrophages, and lymphocytes, as well as extracellular matrix proteins. Furthermore, the impact of KBs on cellular processes implicated in fibrosis, including oxidative stress, chemokine and cytokine expression, caspase activation, and inflammasome signaling is explored. While conflicting findings exist regarding the effects of KBs on these processes, emerging evidence suggests a predominantly beneficial role in mitigating inflammation and oxidative stress associated with fibrotic remodeling. Overall, this review underscores the importance of elucidating the complex interplay between KB metabolism and cardiac fibrosis. The insights gained have the potential to inform novel therapeutic strategies for managing cardiac fibrosis and associated cardiovascular disorders, highlighting the need for further research in this area.
Collapse
Affiliation(s)
- Kellina Maduray
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Jingquan Zhong
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
- Department of Cardiology, Qilu Hospital of Shandong University (Qingdao), Shandong University, Qingdao, Shandong, China
| |
Collapse
|
15
|
Göpel SO, Adingupu D, Wang J, Semenova E, Behrendt M, Jansson-Löfmark R, Ahlström C, Jönsson-Rylander AC, Gopaul VS, Esterline R, Gan LM, Xiao RP. SGLT2 inhibition improves coronary flow velocity reserve and contractility: role of glucagon signaling. Cardiovasc Diabetol 2024; 23:408. [PMID: 39548491 PMCID: PMC11568596 DOI: 10.1186/s12933-024-02491-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/24/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND SGLT2 inhibitors, a T2DM medication to lower blood glucose, markedly improve cardiovascular outcomes but the underlying mechanism(s) are not fully understood. SGLT2i's produce a unique metabolic pattern by lowering blood glucose without increasing insulin while increasing ketone body and glucagon levels and reducing body weight. We tested if glucagon signaling contributes to SGLT2i induced improvement in CV function. METHODS Cardiac contractility and coronary flow velocity reserve (CFVR) were monitored in ob/ob mice and rhesus monkeys with metabolic syndrome using echocardiography. Metabolic status was characterized by measuring blood ketone levels, glucose tolerance during glucose challenge and Arg and ADMA levels were measured. Baysian models were developed to analyse the data. RESULTS Dapagliflozin improved CFVR and contractility, co-application of a glucagon receptor inhibitor (GcgRi) blunted the effect on CFVR but not contractility. Dapagliflozin increased the Arg/ADMA ratio and ketone levels and co-treatment with GcgRi blunted only the Dapagliflozin induced increase in Arg/ADMA ratio but not ketone levels. CONCLUSIONS Since GcgRi co-treatment only reduced the Arg/ADMA increase we hypothesize that dapagliflozin via a glucagon-signaling dependent pathway improves vascular function through the NO-signaling pathway leading to improved vascular function. Increase in ketone levels might be a contributing factor in SGLT2i induced contractility increase and does not require glucagon signaling.
Collapse
Affiliation(s)
- Sven O Göpel
- Global Patient Safety BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden.
| | - Damilola Adingupu
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jue Wang
- College of Future Technology, Peking University, Beijing, 100871, China
| | - Elizaveta Semenova
- Data Sciences and Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
- Imperial College London, School of Public Health, Department of Epidemiology and Biostatistics, London, United Kingdom
| | - Margareta Behrendt
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rasmus Jansson-Löfmark
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Christine Ahlström
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ann-Cathrine Jönsson-Rylander
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - V Sashi Gopaul
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Li-Ming Gan
- Ribocure Pharmaceuticals AB, Gothenburg, Sweden & SuZhou Ribo Life Science Co. Ltd., Gothenburg, Sweden
- Department of Cardiology, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Rui-Ping Xiao
- College of Future Technology, Peking University, Beijing, 100871, China
| |
Collapse
|
16
|
Gopalasingam N, Berg-Hansen K, Christensen KH, Ladefoged BT, Poulsen SH, Andersen MJ, Borlaug BA, Nielsen R, Møller N, Wiggers H. Randomized Crossover Trial of 2-Week Ketone Ester Treatment in Patients With Type 2 Diabetes and Heart Failure With Preserved Ejection Fraction. Circulation 2024; 150:1570-1583. [PMID: 39162035 DOI: 10.1161/circulationaha.124.069732] [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: 03/21/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a major cause of morbidity and mortality in patients with type 2 diabetes (T2D). Acute increases in circulating levels of ketone body 3-hydroxybutyrate have beneficial acute hemodynamic effects in patients without T2D with chronic heart failure with reduced ejection fraction. However, the cardiovascular effects of prolonged oral ketone ester (KE) treatment in patients with T2D and HFpEF remain unknown. METHODS A total of 24 patients with T2D and HFpEF completed a 6-week randomized, double-blind crossover study. All patients received 2 weeks of KE treatment (25 g D-ß-hydroxybutyrate-(R)-1,3-butanediol × 4 daily) and isocaloric and isovolumic placebo, separated by a 2-week washout period. At the end of each treatment period, patients underwent right heart catheterization, echocardiography, and blood samples at trough levels of intervention, and then during a 4-hour resting period after a single dose. A subsequent second dose was administered, followed by an exercise test. The primary end point was cardiac output during the 4-hour rest period. RESULTS During the 4-hour resting period, circulating 3-hydroxybutyrate levels were 10-fold higher after KE treatment (1010±56 µmol/L; P<0.001) compared with placebo (91±55 µmol/L). Compared with placebo, KE treatment increased cardiac output by 0.2 L/min (95% CI, 0.1 to 0.3) during the 4-hour period and decreased pulmonary capillary wedge pressure at rest by 1 mm Hg (95% CI, -2 to 0) and at peak exercise by 5 mm Hg (95% CI, -9 to -1). KE treatment decreased the pressure-flow relationship (∆ pulmonary capillary wedge pressure/∆ cardiac output) significantly during exercise (P<0.001) and increased stroke volume by 10 mL (95% CI, 0 to 20) at peak exercise. KE right-shifted the left ventricular end-diastolic pressure-volume relationship, suggestive of reduced left ventricular stiffness and improved compliance. Favorable hemodynamic responses of KE treatment were also observed in patients treated with sodium-glucose transporter-2 inhibitors and glucagon-like peptide-1 analogs. CONCLUSIONS In patients with T2D and HFpEF, a 2-week oral KE treatment increased cardiac output and reduced cardiac filling pressures and ventricular stiffness. At peak exercise, KE treatment markedly decreased pulmonary capillary wedge pressure and improved pressure-flow relationship. Modulation of circulating ketone levels is a potential new treatment modality for patients with T2D and HFpEF. REGISTRATION URL: https://www.clinicaltrials.gov; Unique Identifier: NCT05236335.
Collapse
Affiliation(s)
- Nigopan Gopalasingam
- Department of Cardiology, Aarhus University Hospital, Denmark (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., H.W.)
- Department of Clinical Medicine (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., N.M., H.W.), Aarhus University, Denmark
| | - Kristoffer Berg-Hansen
- Department of Cardiology, Aarhus University Hospital, Denmark (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., H.W.)
- Department of Clinical Medicine (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., N.M., H.W.), Aarhus University, Denmark
| | - Kristian Hylleberg Christensen
- Department of Cardiology, Aarhus University Hospital, Denmark (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., H.W.)
- Department of Clinical Medicine (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., N.M., H.W.), Aarhus University, Denmark
| | - Bertil T Ladefoged
- Department of Cardiology, Aarhus University Hospital, Denmark (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., H.W.)
- Department of Clinical Medicine (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., N.M., H.W.), Aarhus University, Denmark
| | - Steen Hvitfeldt Poulsen
- Department of Cardiology, Aarhus University Hospital, Denmark (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., H.W.)
- Department of Clinical Medicine (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., N.M., H.W.), Aarhus University, Denmark
| | - Mads Jønsson Andersen
- Department of Cardiology, Aarhus University Hospital, Denmark (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., H.W.)
- Department of Clinical Medicine (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., N.M., H.W.), Aarhus University, Denmark
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic Hospital, Rochester, MN (B.A.B.)
| | - Roni Nielsen
- Department of Cardiology, Aarhus University Hospital, Denmark (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., H.W.)
- Department of Clinical Medicine (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., N.M., H.W.), Aarhus University, Denmark
| | - Niels Møller
- Department of Clinical Medicine (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., N.M., H.W.), Aarhus University, Denmark
- Department of Endocrinology and Metabolism (N.M.), Aarhus University, Denmark
| | - Henrik Wiggers
- Department of Cardiology, Aarhus University Hospital, Denmark (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., H.W.)
- Department of Clinical Medicine (N.G., K.B.-H., K.H.C., B.T.L., S.H.P., M.J.A., R.N., N.M., H.W.), Aarhus University, Denmark
| |
Collapse
|
17
|
Voorrips SN, Westenbrink BD. Ketone Treatment in Heart Failure With Preserved Ejection Fraction: Recharging the Heart or Reducing Filling Pressures? Circulation 2024; 150:1584-1587. [PMID: 39527663 DOI: 10.1161/circulationaha.124.071608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Affiliation(s)
- Suzanne N Voorrips
- University Medical Center Groningen, University of Groningen, the Netherlands
| | - B Daan Westenbrink
- University Medical Center Groningen, University of Groningen, the Netherlands
| |
Collapse
|
18
|
Wiggers H. SGLT2 Inhibitors and Their Effect on Metabolism in Patients With Heart Failure. Circ Heart Fail 2024; 17:e012373. [PMID: 39421945 DOI: 10.1161/circheartfailure.124.012373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Affiliation(s)
- Henrik Wiggers
- Department of Cardiology, Aarhus University Hospital, Denmark
| |
Collapse
|
19
|
Kjærulff MLG, Luong TV, Richard G, St-Pierre V, Søndergaard E, Møller N, Gormsen LC, Tremblay S, Croteau E, Cunnane SC. Cerebral and myocardial kinetics of [ 11C]acetoacetate and [ 11C]β-hydroxybutyrate: A comparative crossover study in healthy rats. Nucl Med Biol 2024; 138-139:108967. [PMID: 39476467 DOI: 10.1016/j.nucmedbio.2024.108967] [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: 08/02/2024] [Revised: 10/03/2024] [Accepted: 10/23/2024] [Indexed: 12/11/2024]
Abstract
BACKGROUND Ketone metabolism has been studied using positron emission tomography (PET) with the radiotracers [11C]acetoacetate and [11C]β-hydroxybutyrate. However, whether these two radiotracers actually yield equivalent estimates of cerebral and myocardial ketone metabolism has not yet been investigated. This study aimed to investigate and compare the kinetics of both tracers in the brain and heart of healthy rats under varying levels of circulating ketones at baseline and after a single-dose exogenous ketone ester (KE) supplement. METHODS Six healthy Sprague-Dawley rats each underwent two scans with each tracer: one following oral KE administration and one with a placebo. Cerebral kinetic parameters (Ki, VT, and cerebral metabolic rate (CMR)) were obtained using the Patlak method, whereas myocardial kinetic parameters (K1, k2, and VT) were derived using a 1-tissue compartment model. Parameters were compared through mixed-effects, correlation, and Bland-Altman analyses. RESULTS Global CMR increased 3-4-fold in the KE group versus placebo, with strong positive correlations between CMR and plasma ketone levels for both tracers. Correlations between [11C]acetoacetate and [11C]β-hydroxybutyrate were moderate and non-significant for relative cerebral uptake expressed as Ki (ρ = 0.40) and for VT (ρ = 0.38) but strongly positive for absolute uptake, CMR (r = 0.84), with a non-significant mean bias of -0.03. In contrast, myocardial kinetics showed only non-significant weak to moderate correlations between the radiotracers (K1 (r = 0.04), k2 (r = -0.27), and VT (ρ = 0.43)), with no systematic biases. CONCLUSION [11C]acetoacetate and [11C]β-hydroxybutyrate can be used interchangeably for measuring global CMR in healthy rats but differ in certain cerebral and myocardial kinetics. Whether these findings are generalizable to pathological conditions warrants further studies to explore the kinetics of these tracers in disease models.
Collapse
Affiliation(s)
- Mette Louise Gram Kjærulff
- Department of Clinical Medicine - Nuclear Medicine & PET, Aarhus University, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Denmark; Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Denmark.
| | - Thien Vinh Luong
- Department of Clinical Medicine - Nuclear Medicine & PET, Aarhus University, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Denmark; Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Denmark
| | - Gabriel Richard
- Centre d'imagerie Moléculaire de Sherbrooke, Sherbrooke, Québec, Canada; Centre de Recherche du CHUS, Sherbrooke, Québec, Canada
| | - Valérie St-Pierre
- Department of Medicine and Research Center on Aging, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Esben Søndergaard
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Denmark; Department of Clinical Medicine - Medical/Steno Aarhus Research Laboratory, Aarhus University, Denmark
| | - Niels Møller
- Department of Clinical Medicine - Medical/Steno Aarhus Research Laboratory, Aarhus University, Denmark; Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark
| | - Lars Christian Gormsen
- Department of Clinical Medicine - Nuclear Medicine & PET, Aarhus University, Denmark; Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Denmark
| | - Sébastien Tremblay
- Centre d'imagerie Moléculaire de Sherbrooke, Sherbrooke, Québec, Canada; Centre de Recherche du CHUS, Sherbrooke, Québec, Canada
| | - Etienne Croteau
- Centre d'imagerie Moléculaire de Sherbrooke, Sherbrooke, Québec, Canada; Centre de Recherche du CHUS, Sherbrooke, Québec, Canada
| | - Stephen C Cunnane
- Department of Medicine and Research Center on Aging, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
20
|
Xin Q, Liang X, Yang J, Wang X, Hu F, Jiang M, Liu Y, Gong J, Pan Y, Liu L, Xu J, Cui Y, Qin H, Bai H, Li Y, Ma J, Zhang C, Shi B. Metabolomic alterations in the plasma of patients with various clinical manifestations of COVID-19. Virol J 2024; 21:266. [PMID: 39468659 PMCID: PMC11520427 DOI: 10.1186/s12985-024-02523-7] [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: 05/16/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND The metabolomic profiles of individuals with different clinical manifestations of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have not been clearly characterized. METHODS We performed metabolomics analysis of 166 individuals, including 62 healthy controls, 16 individuals with asymptomatic SARS-CoV-2 infection, and 88 patients with moderate (n = 42) and severe (n = 46) symptomatic 2019 coronavirus disease (COVID-19; 17 with short-term and 34 with long-term nucleic-acid test positivity). By examining differential expression, we identified candidate metabolites associated with different SARS-CoV-2 infection presentations. Functional and machine learning analyses were performed to explore the metabolites' functions and verify their candidacy as biomarkers. RESULTS A total of 417 metabolites were detected. We discovered 70 differentially expressed metabolites that may help differentiate asymptomatic infections from healthy controls and COVID-19 patients with different disease severity. Cyclamic acid and N-Acetylneuraminic Acid were identified to distinguish symptomatic infected patients and asymptomatic infected patients. Shikimic Acid, Glycyrrhetinic acid and 3-Hydroxybutyrate can supply significant insights for distinguishing short-term and long-term nucleic-acid test positivity. CONCLUSION Metabolomic profiling may highlight novel biomarkers for the identification of individuals with asymptomatic SARS-CoV-2 infection and further our understanding of the molecular pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Qi Xin
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xiao Liang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Jin Yang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaorui Wang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Fang Hu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Meng Jiang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yijia Liu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Jin Gong
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yiwen Pan
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Lijuan Liu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Jiao Xu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yuxin Cui
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Hongyu Qin
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Han Bai
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an, 710000, China
| | - Yixin Li
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an, 710000, China
| | - Junpeng Ma
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Chengsheng Zhang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China.
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an, 710000, China.
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China.
| |
Collapse
|
21
|
Liu K, Yang Y, Yang JH. Underlying mechanisms of ketotherapy in heart failure: current evidence for clinical implementations. Front Pharmacol 2024; 15:1463381. [PMID: 39512825 PMCID: PMC11540999 DOI: 10.3389/fphar.2024.1463381] [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: 07/11/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
Heart failure (HF) is a life-threatening cardiac syndrome characterized by high morbidity and mortality, but current anti-heart failure therapies have limited efficacy, necessitating the urgent development of new treatment drugs. Exogenous ketone supplementation helps prevent heart failure development in HF models, but therapeutic ketosis in failing hearts has not been systematically elucidated, limiting the use of ketones to treat HF. Here, we summarize current evidence supporting ketotherapy in HF, emphasizing ketone metabolism in the failing heart, metabolic and non-metabolic therapeutic effects, and mechanisms of ketotherapy in HF, involving the dynamics within the mitochondria. We also discuss clinical strategies for therapeutic ketosis, aiming to deepen the understanding of the characteristics of ketone metabolism, including mitochondrial involvement, and its clinical therapeutic potential in HF.
Collapse
Affiliation(s)
| | | | - Jing-Hua Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
22
|
Zhang N, Liu D, Zhao J, Tse G, Zhou J, Zhang Q, Lip GYH, Liu T. Circulating ketone bodies, genetic susceptibility, with left atrial remodeling and atrial fibrillation: A prospective study from the UK Biobank. Heart Rhythm 2024:S1547-5271(24)03454-4. [PMID: 39433077 DOI: 10.1016/j.hrthm.2024.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND Ketone bodies (KBs) are an important cardiac metabolic energy source. Metabolic remodeling has recently been found to play an important role in the pathological process of atrial fibrillation (AF). OBJECTIVE The purpose of this study was to evaluate the associations of circulating KB levels with incident AF risk in the general population. METHODS We studied 237,163 participants [mean age, 56.5 years; 129,472 women (55%)] from the UK Biobank who were free of AF at baseline and had data on circulating β-hydroxybutyrate (β-OHB), acetoacetate, and acetone. The associations of KBs with new-onset AF were evaluated using Cox regression in the general population and across the 3 genetic risk groups: low, moderate, and high polygenic risk score of AF. RESULTS During a median follow-up of 14.8 (13.8, 15.5) years, 16,638 participants (7.0%) developed AF. There was a U-shaped association of total KBs and β-OHB with incident AF, with nadirs at 60.6 and 40.8 μmol/L, respectively (Pnonlinear < .05), whereas there was a positive association of acetoacetate and acetone with AF (Poverall < .001; Pnonlinear > .05). Consistently, there was a U-shaped association of total KBs and β-OHB with left atrial (LA) volume parameters, including LA maximum volume, LA minimum volume, and their body surface area-indexed counterparts, and there was an inverted U-shaped association of total KBs and β-OHB with LA ejection fraction (Pnonlinear < .05 for all). The associations of KBs with AF were stronger in individuals with low genetic risk (Pinteraction < .05), while the highest AF risk was in those with high genetic risk with high KB levels. Significant mediation effects of inflammatory markers on the associations between KBs and AF were identified. CONCLUSION There was a U-shaped association of circulating total KBs and β-OHB with incident AF as well as a positive association of acetoacetate and acetone levels with AF risk in the general population.
Collapse
Affiliation(s)
- Nan Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Daiqi Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jinhua Zhao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China; School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China; Cardiovascular Analytics Group, PowerHealth Research Institute, Hong Kong, China
| | - Jiandong Zhou
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Qingpeng Zhang
- School of Data Science, City University of Hong Kong, Hong Kong, China
| | - Gregory Y H Lip
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China; Liverpool Centre for Cardiovascular Sciences, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom.
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
23
|
Lu P, Fan J, Li X, Liu Z, Qi Y, Shen Z, Wen Z, Yi C, Song M, Wang X. Serum Free Fatty Acid Concentration Predicts ARDS after Off-Pump CABG: A Prospective Observational Study. Lung 2024; 202:523-532. [PMID: 38753182 DOI: 10.1007/s00408-024-00704-3] [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: 02/09/2024] [Accepted: 04/30/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Free fatty acids (FFAs) are established risk factors for various cardiovascular and metabolic disorders. Elevated FFAs can trigger inflammatory response, which may be associated with the occurrence of acute respiratory distress syndrome (ARDS) in cardiac surgery. In this prospective study, we aimed to investigate the association between circulating FFA and the incidence of ARDS, as well as the length of ICU stay, in patients undergoing off-pump coronary artery bypass grafting (CABG). METHODS We conducted a single-center, prospective, observational study among patients undergoing off-pump CABG. The primary endpoint was the occurrence of ARDS within 6 days after off-pump CABG. Serum FFA were measured at baseline and 24 h post-procedure, and the difference (Δ-FFA) was calculated. RESULTS A total of 180 patients were included in the primary analysis. The median FFA was 2.3 mmol/L (quartile 1 [Q1]-Q3, 1.4-3.2) at baseline and 1.5 mmol/L (Q1-Q3, 0.9-2.3) 24 h after CABG, with a Δ-FFA of 0.6 mmol/L (Q1-Q3, -0.1 to 1.6). Patients with elevated Δ-FFA levels had a significantly higher ARDS occurrence (55.6% vs. 22.2%; P < 0.001). Elevated Δ-FFA after off-pump CABG correlated with a significantly lower PaO2/FiO2 ratio, prolonged mechanical ventilation, and extended length of ICU stay. The area under the curve (AUC) of Δ-FFA for predicting ARDS (AUC, 0.758; 95% confidence interval, 0.686-0.831) significantly exceeded the AUC of postoperative FFA (AUC, 0.708; 95% CI 0.628-0.788; P < 0.001). CONCLUSIONS Elevated Δ-FFA levels correlated with ARDS following off-pump CABG. Monitoring FFA may assist in identifying high-risk patients for ARDS, facilitating timely interventions to improve clinical outcomes.
Collapse
Affiliation(s)
- Peng Lu
- Department of Cardiovascular Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taihu Road 366, Taizhou, Jiangsu, 225300, China
- Department of Cardiovascular Surgery, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Jidan Fan
- Department of Cardiovascular Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taihu Road 366, Taizhou, Jiangsu, 225300, China
| | - Xiangyu Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Zhaoyang Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Yuanpu Qi
- Department of Cardiovascular Surgery, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Zihao Shen
- Department of Cardiovascular Surgery, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Ziang Wen
- Department of Cardiovascular Surgery, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Chenlong Yi
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Meijuan Song
- Department of Geriatrics, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China.
| | - Xiaowei Wang
- Department of Cardiovascular Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taihu Road 366, Taizhou, Jiangsu, 225300, China.
- Department of Cardiovascular Surgery, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China.
| |
Collapse
|
24
|
Kjaerulff MLG, Nielsen EN, Gormsen LC, Dias AH. First-in-human whole-body PET/CT imaging with the optimized ketone tracer [ 11C]β-hydroxybutyrate using a long axial field-of-view scanner. Eur J Nucl Med Mol Imaging 2024; 51:3802-3804. [PMID: 38867106 DOI: 10.1007/s00259-024-06787-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/01/2024] [Indexed: 06/14/2024]
Affiliation(s)
- Mette Louise Gram Kjaerulff
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, Aarhus, 8200, Denmark.
| | - Erik Nguyen Nielsen
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, Aarhus, 8200, Denmark
| | - Lars Christian Gormsen
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, Aarhus, 8200, Denmark
| | - André Henrique Dias
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, Aarhus, 8200, Denmark
| |
Collapse
|
25
|
Smeijer JD, Wasehuus VS, Dhaun N, Górriz JL, Soler MJ, Åstrand M, Mercier AK, Greasley PJ, Ambery P, Heerspink HJ. Effects of Zibotentan Alone and in Combination with Dapagliflozin on Fluid Retention in Patients with CKD. J Am Soc Nephrol 2024; 35:1381-1390. [PMID: 39352861 PMCID: PMC11452182 DOI: 10.1681/asn.0000000000000436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024] Open
Abstract
Key Points Increasing doses of the endothelin receptor antagonist zibotentan and lower eGFR were associated with a higher risk of fluid retention. The higher risk of fluid retention could be attenuated by the combination of zibotentan with the sodium-glucose cotransporter 2 inhibitor dapagliflozin. Background Endothelin receptor antagonists (ERAs) reduce albuminuria but are limited by fluid retention risk, particularly in patients with CKD. Combining ERAs with sodium-glucose cotransporter 2 (SGLT2) inhibitors, which have diuretic effects, offers a promising strategy to mitigate fluid retention. In this post hoc analysis of the Zibotentan and Dapagliflozin for the Treatment of CKD (ZENITH-CKD) trial, we assessed fluid dynamics in patients with CKD treated with the ERA zibotentan alone and in combination with the SGLT2 inhibitor dapagliflozin. Methods In the ZENITH-CKD trial, 508 patients with CKD (eGFR ≥20 ml/min per 1.73 m2 and a urinary albumin-creatinine ratio of 150–5000 mg/g) were randomized to treatment with placebo, dapagliflozin 10 mg plus placebo, zibotentan (0.25, 1.5, or 5 mg) plus dapagliflozin 10 mg, and zibotentan 5 mg plus placebo. We evaluated correlations between changes in fluid retention markers and bioimpedance-measured extracellular fluid in response to zibotentan treatment. We used Cox proportional hazards regression to assess the association between zibotentan/dapagliflozin treatment, baseline characteristics, and fluid retention and the relationship between zibotentan plasma exposure and fluid retention. Results After 3 weeks of treatment with zibotentan 0.25, 1.5, or 5 mg plus dapagliflozin 10 mg, changes in body weight (β =0.36 [95% confidence interval (CI), 0.26 to 0.45]) per kg, B-type natriuretic peptide (β =0.38 [95% CI, 0.22 to 0.54]) per doubling, and hemoglobin (β =−0.29 [95% CI, −0.48 to −0.10]) per g/dl were independently associated with changes in extracellular fluid. Higher doses of zibotentan were associated with significantly higher risk of fluid retention compared with dapagliflozin alone (zibotentan 5 mg hazard ratio (HR) 8.50 [95% CI, 3.40 to 21.30]). The HR attenuated when zibotentan was combined with dapagliflozin (zibotentan/dapagliflozin 5/10 mg HR 3.09 [95% CI, 1.08 to 8.80], zibotentan/dapagliflozin 1.5/10 mg 2.70 [95% CI, 1.44 to 5.07], and zibotentan/dapagliflozin 0.25/10 mg HR 1.21 [95% CI, 0.50 to 2.91]). The risk of fluid retention was higher with higher zibotentan exposure and lower eGFR. Conclusions High doses of zibotentan were associated with a higher risk of fluid retention, which was attenuated with lower doses and the addition of dapagliflozin. Clinical Trial registry name and registration number: ZENITH-CKD Trial, NCT04724837 .
Collapse
Affiliation(s)
- J. David Smeijer
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Neeraj Dhaun
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - José Luis Górriz
- Department of Nephrology, University Clinical Hospital, INCLIVA Research Institute, University of Valencia, Valencia, Spain
| | - Maria José Soler
- Nephrology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Research, Barcelona, Spain
| | - Magnus Åstrand
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Anne-Kristina Mercier
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Peter J. Greasley
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Phil Ambery
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Hiddo J.L. Heerspink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- George Institute for Global Health, Barangaroo, New South Wales, Australia
| |
Collapse
|
26
|
Shahpasand S, Khatami SH, Ehtiati S, Alehossein P, Salmani F, Toutounchi AH, Zarei T, Shahmohammadi MR, Khodarahmi R, Aghamollaii V, Tafakhori A, Karima S. Therapeutic potential of the ketogenic diet: A metabolic switch with implications for neurological disorders, the gut-brain axis, and cardiovascular diseases. J Nutr Biochem 2024; 132:109693. [PMID: 38880191 DOI: 10.1016/j.jnutbio.2024.109693] [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: 01/10/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/18/2024]
Abstract
The Ketogenic Diet (KD) is a dietary regimen that is low in carbohydrates, high in fats, and contains adequate protein. It is designed to mimic the metabolic state of fasting. This diet triggers the production of ketone bodies through a process known as ketosis. The primary objective of KD is to induce and sustain ketosis, which has been associated with numerous health benefits. Recent research has uncovered promising therapeutic potential for KD in the treatment of various diseases. This includes evidence of its effectiveness as a dietary strategy for managing intractable epilepsy, a form of epilepsy that is resistant to medication. We are currently assessing the efficacy and safety of KD through laboratory and clinical studies. This review focuses on the anti-inflammatory properties of the KD and its potential benefits for neurological disorders and the gut-brain axis. We also explore the existing literature on the potential effects of KD on cardiac health. Our aim is to provide a comprehensive overview of the current knowledge in these areas. Given the encouraging preliminary evidence of its therapeutic effects and the growing understanding of its mechanisms of action, randomized controlled trials are warranted to further explore the rationale behind the clinical use of KD. These trials will ultimately enhance our understanding of how KD functions and its potential benefits for various health conditions. We hope that our research will contribute to the body of knowledge in this field and provide valuable insights for future studies.
Collapse
Affiliation(s)
- Sheyda Shahpasand
- Department of Biology, Faculty of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Ehtiati
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parsa Alehossein
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Salmani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Alireza Haghbin Toutounchi
- Department of general surgery,Imam Hosein medical and educational center, Shahid Beheshti University of medical sciences, Tehran, Iran
| | - Tayebe Zarei
- Clinical Trial Department, Behbalin Co., Ltd., Tehran, Iran
| | - Mohammad Reza Shahmohammadi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Vajiheh Aghamollaii
- Neurology Department, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Tafakhori
- Department of Neurology, School of Medicine, Iranian Center of Neurological Research, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran.
| |
Collapse
|
27
|
Foster MW, Riley JM, Kaki PC, Al Soueidy A, Aligholiazadeh E, Rame JE. Metabolic Adaptation in Heart Failure and the Role of Ketone Bodies as Biomarkers. Curr Heart Fail Rep 2024; 21:498-503. [PMID: 39242479 DOI: 10.1007/s11897-024-00678-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/12/2024] [Indexed: 09/09/2024]
Abstract
PURPOSE OF REVIEW The development and progression of heart failure is characterized by metabolic and physiologic adaptations allowing patients to cope with cardiac insufficiency. This review explores the changes in metabolism in heart failure and the potential role of biomarkers, particularly ketone bodies, in staging and prognosticating heart failure progression. RECENT FINDINGS Recent insights into myocardial metabolism shed light on the heart's response to stress, highlighting the shift towards reliance on ketone bodies as an alternative fuel source. Elevated blood ketone levels have been shown to correlate with the severity of cardiac dysfunction, emphasizing their potential as prognostic indicators. Furthermore, studies exploring therapeutic interventions targeting specific metabolic pathways offer promise for improving outcomes in heart failure. Ketones have prognostic utility in heart failure, and potentially, an avenue for therapeutic intervention. Challenges remain in deciphering the optimal balance between metabolic support and exacerbating cardiac remodeling. Future research endeavors must address these complexities to advance personalized approaches in managing heart failure.
Collapse
Affiliation(s)
- Michael W Foster
- Department of Medicine, Division of Cardiology, Thomas Jefferson University Hospital, 833 Chestnut Street, Suite 600, Philadelphia, PA, 19107, USA
| | - Joshua M Riley
- Department of Medicine, Division of Cardiology, Thomas Jefferson University Hospital, 833 Chestnut Street, Suite 600, Philadelphia, PA, 19107, USA
| | - Praneet C Kaki
- Sidney Kimmel Medical College, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Amine Al Soueidy
- Department of Medicine, Copper University Hospital, Camden, NJ, USA
| | | | - J Eduardo Rame
- Department of Medicine, Division of Cardiology, Thomas Jefferson University Hospital, 833 Chestnut Street, Suite 600, Philadelphia, PA, 19107, USA.
| |
Collapse
|
28
|
Wang T, Ning M, Mo Y, Tian X, Fu Y, Laher I, Li S. Metabolomic Profiling Reveals That Exercise Lowers Biomarkers of Cardiac Dysfunction in Rats with Type 2 Diabetes. Antioxidants (Basel) 2024; 13:1167. [PMID: 39456421 PMCID: PMC11505272 DOI: 10.3390/antiox13101167] [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: 08/02/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 10/28/2024] Open
Abstract
The increasing prevalence of type 2 diabetes mellitus (T2DM) leads to significant global health challenges, including cardiac structural and functional deficits, which in severe cases can progress to heart failure that can further strain healthcare resources. Aerobic exercise can ameliorate cardiac dysfunction in individuals with diabetes, although a comprehensive understanding of its underlying mechanisms remains elusive. This study utilizes untargeted metabolomics to reveal aerobic-exercise-activated metabolic biomarkers in the cardiac tissues of Sprague Dawley rats with T2DM. Metabolomics analysis revealed that diabetes altered 1029 myocardial metabolites, while aerobic exercise reversed 208 of these metabolites, of which 112 were upregulated and 96 downregulated. Pathway topology analysis suggested that these metabolites predominantly contributed to purine metabolism and arginine biosynthesis. Furthermore, receiver operating characteristic curve analysis identified 10 potential biomarkers, including xanthine, hypoxanthine, inosine, dGMP, l-glutamic acid, l-arginine, l-tryptophan, (R)-3-hydroxybutyric acid, riboflavin, and glucolepidiin. Finally, data from Pearson correlation analysis indicated that some metabolic biomarkers strongly correlated with cardiac function. Our data suggest that certain metabolic biomarkers play an important role in ameliorating diabetes-related cardiac dysfunction by aerobic exercise.
Collapse
Affiliation(s)
- Tutu Wang
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (T.W.); (M.N.); (Y.M.); (X.T.); (Y.F.)
| | - Miaomiao Ning
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (T.W.); (M.N.); (Y.M.); (X.T.); (Y.F.)
| | - Yurou Mo
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (T.W.); (M.N.); (Y.M.); (X.T.); (Y.F.)
| | - Xinyu Tian
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (T.W.); (M.N.); (Y.M.); (X.T.); (Y.F.)
| | - Yu Fu
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (T.W.); (M.N.); (Y.M.); (X.T.); (Y.F.)
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
| | - Shunchang Li
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu 610041, China; (T.W.); (M.N.); (Y.M.); (X.T.); (Y.F.)
| |
Collapse
|
29
|
Annoni F, Su F, Peluso L, Lisi I, Caruso E, Pischiutta F, Gouvea Bogossian E, Garcia B, Njimi H, Vincent JL, Gaspard N, Ferlini L, Creteur J, Zanier ER, Taccone FS. Infusion of sodium DL-3-ß-hydroxybutyrate decreases cerebral injury biomarkers after resuscitation in experimental cardiac arrest. Crit Care 2024; 28:314. [PMID: 39304944 PMCID: PMC11414246 DOI: 10.1186/s13054-024-05106-8] [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: 07/12/2024] [Accepted: 09/18/2024] [Indexed: 09/22/2024] Open
Abstract
AIMS Cerebral complications after cardiac arrest (CA) remain a major problem worldwide. The aim was to test the effects of sodium-ß-hydroxybutyrate (SBHB) infusion on brain injury in a clinically relevant swine model of CA. RESULTS CA was electrically induced in 20 adult swine. After 10 min, cardiopulmonary resuscitation was performed for 5 min. After return of spontaneous circulation (ROSC), the animals were randomly assigned to receive an infusion of balanced crystalloid (controls, n = 11) or SBHB (theoretical osmolarity 1189 mOsm/l, n = 8) for 12 h. Multimodal neurological and cardiovascular monitoring were implemented in all animals. Nineteen of the 20 animals achieved ROSC. Blood sodium concentrations, osmolarity and circulating KBs were higher in the treated animals than in the controls. SBHB infusion was associated with significantly lower plasma biomarkers of brain injury at 6 (glial fibrillary acid protein, GFAP and neuron specific enolase, NSE) and 12 h (neurofilament light chain, NFL, GFAP and NSE) compared to controls. The amplitude of the stereoelectroencephalograph (sEEG) increased in treated animals after ROSC compared to controls. Cerebral glucose uptake was lower in treated animals. CONCLUSIONS In this experimental model, SBHB infusion after resuscitated CA was associated with reduced circulating markers of cerebral injury and increased sEEG amplitude.
Collapse
Affiliation(s)
- Filippo Annoni
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Lennik Road 808, 1070, Brussels, Belgium.
- Experimental Laboratory of Intensive Care, Free University of Brussels, Brussels, Belgium.
| | - Fuhong Su
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Lennik Road 808, 1070, Brussels, Belgium
- Experimental Laboratory of Intensive Care, Free University of Brussels, Brussels, Belgium
| | - Lorenzo Peluso
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Lennik Road 808, 1070, Brussels, Belgium
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Anesthesiology and Intensive Care, Humanitas Gavazzeni, Via M Gavazzeni 21, 24125, Bergamo, Italy
| | - Ilaria Lisi
- Laboratory of Traumatic Brain Injury and Neuroprotection, Department of Acute Brain and Cardiovascular Injury, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Enrico Caruso
- Laboratory of Traumatic Brain Injury and Neuroprotection, Department of Acute Brain and Cardiovascular Injury, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Francesca Pischiutta
- Laboratory of Traumatic Brain Injury and Neuroprotection, Department of Acute Brain and Cardiovascular Injury, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Elisa Gouvea Bogossian
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Lennik Road 808, 1070, Brussels, Belgium
| | - Bruno Garcia
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Lennik Road 808, 1070, Brussels, Belgium
- Experimental Laboratory of Intensive Care, Free University of Brussels, Brussels, Belgium
| | - Hassane Njimi
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Lennik Road 808, 1070, Brussels, Belgium
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Lennik Road 808, 1070, Brussels, Belgium
| | - Nicolas Gaspard
- Department of Neurology, Erasme Hospital, Lennik Road 808, 1070, Brussels, Belgium
- Neurology Department, School of Medicine, Yale University, New Haven, CT, USA
| | - Lorenzo Ferlini
- Department of Neurology, Erasme Hospital, Lennik Road 808, 1070, Brussels, Belgium
| | - Jacques Creteur
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Lennik Road 808, 1070, Brussels, Belgium
| | - Elisa R Zanier
- Laboratory of Traumatic Brain Injury and Neuroprotection, Department of Acute Brain and Cardiovascular Injury, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Fabio Silvio Taccone
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Lennik Road 808, 1070, Brussels, Belgium
- Experimental Laboratory of Intensive Care, Free University of Brussels, Brussels, Belgium
| |
Collapse
|
30
|
Ataran A, Pompian A, Hajirezaei H, Lodhi R, Javaheri A. Fueling the Heart: What Are the Optimal Dietary Strategies in Heart Failure? Nutrients 2024; 16:3157. [PMID: 39339757 PMCID: PMC11434961 DOI: 10.3390/nu16183157] [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: 08/01/2024] [Revised: 09/10/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
OBJECTIVES Heart failure (HF) is a global health concern with rising incidence and poor prognosis. While the essential role of nutritional and dietary strategies in HF patients is acknowledged in the existing scientific guidelines and clinical practice, there are no comprehensive nutritional recommendations for optimal dietary management of HF. METHODS In this review, we discuss results from recent studies on the obesity paradox and the effects of calorie restriction and weight loss, intermittent fasting, the Western diet, the Mediterranean diet, the ketogenic diet, and the DASH diet on HF progression. RESULTS Many of these strategies remain under clinical and basic investigation for their safety and efficacy, and there is considerable heterogeneity in the observed response, presumably because of heterogeneity in the pathogenesis of different types of HF. In addition, while specific aspects of cardiac metabolism, such as changes in ketone body utilization, might underlie the effects of certain dietary strategies on the heart, there is a critical divide between supplement strategies (i.e., with ketones) and dietary strategies that impact ketogenesis. CONCLUSION This review aims to highlight this gap by exploring emerging evidence supporting the importance of personalized dietary strategies in preventing progression and improving outcomes in the context of HF.
Collapse
Affiliation(s)
- Anahita Ataran
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63110, USA; (A.A.); (A.P.); (H.H.); (R.L.)
| | - Alexander Pompian
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63110, USA; (A.A.); (A.P.); (H.H.); (R.L.)
| | - Hamidreza Hajirezaei
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63110, USA; (A.A.); (A.P.); (H.H.); (R.L.)
| | - Rehman Lodhi
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63110, USA; (A.A.); (A.P.); (H.H.); (R.L.)
| | - Ali Javaheri
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO 63110, USA; (A.A.); (A.P.); (H.H.); (R.L.)
- John Cochran VA Hospital, St. Louis, MO 63110, USA
| |
Collapse
|
31
|
Stubbs BJ, Alvarez Azañedo G, Peralta S, Diaz SR, Gray W, Alexander L, Silverman-Martin W, Garcia TY, Blonquist TM, Upadhyay V, Turnbaugh PJ, Johnson JB, Newman JC. Rationale and protocol for a safety, tolerability and feasibility randomized, parallel arm, double-blind, placebo-controlled, pilot study of a novel ketone ester targeting frailty via immunometabolic geroscience mechanisms. PLoS One 2024; 19:e0307951. [PMID: 39292659 PMCID: PMC11410252 DOI: 10.1371/journal.pone.0307951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 07/14/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Frailty is a geriatric syndrome characterized by chronic inflammation and metabolic insufficiency that creates vulnerability to poor outcomes with aging. We hypothesize that interventions which target common underlying mechanism of aging could ameliorate frailty. Ketone bodies are metabolites produced during fasting or on a ketogenic diet that have pleiotropic effects on inflammatory and metabolic aging pathways in laboratory animal models. Ketone esters (KEs) are compounds that induce ketosis without dietary changes, but KEs have not been studied in an older adult population. Our long-term goal is to examine if KEs modulate aging biology mechanisms and clinical outcomes relevant to frailty in older adults. OBJECTIVES The primary objective of this randomized, placebo-controlled, double-blinded, parallel-group, pilot trial is to determine tolerability of 12-weeks of KE ingestion in a broad population of older adults (≥ 65 years). Secondary outcomes include safety and acute blood ketone kinetics. Exploratory outcomes include physical function, cognitive function, quality of life, aging biomarkers and inflammatory measures. METHODS Community-dwelling adults who are independent in activities of daily living, with no unstable acute medical conditions (n = 30) will be recruited. The study intervention is a KE or a taste, appearance, and calorie matched placebo beverage. Initially, acute 4-hour ketone kinetics after 12.5g or 25g of KE consumption will be assessed. After collection of baseline safety, functional, and biological measurements, subjects will randomly be allocated to consume KE 25g or placebo once daily for 12-weeks. Questionnaires will assess tolerability daily for 2-weeks, and then via phone interview at bi-monthly intervals. Safety assessments will be repeated at week 4. All measures will be repeated at week 12. CONCLUSION This study will evaluate feasibility, tolerability, and safety of KE consumption in older adults and provide exploratory data across a range of aging-related endpoints. This data will inform design of larger trials to rigorously test KE effects on aging mechanisms and clinical outcomes relevant to frailty.
Collapse
Affiliation(s)
- Brianna J. Stubbs
- Buck Institute for Research on Aging, Novato, California, United States of America
| | | | - Sawyer Peralta
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Stephanie Roa Diaz
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Wyatt Gray
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Laura Alexander
- Buck Institute for Research on Aging, Novato, California, United States of America
| | | | - Thelma Y. Garcia
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Traci M. Blonquist
- Biofortis, Mérieux NutriSciences, Addison, Illinois, United States of America
| | - Vaibhav Upadhyay
- Department of Microbiology & Immunology, UCSF, San Francisco, California, United States of America
- Independent Researcher, Greenbrae, California, United States of America
| | - Peter J. Turnbaugh
- Department of Microbiology & Immunology, UCSF, San Francisco, California, United States of America
- Division of Geriatrics, UCSF, San Francisco, California, United States of America
| | - James B. Johnson
- Department of Medicine, UCSF, San Francisco, California, United States of America
| | - John C. Newman
- Buck Institute for Research on Aging, Novato, California, United States of America
- Chan Zuckerberg Biohub-San Francisco, San Francisco, California, United States of America
| |
Collapse
|
32
|
Stubbs BJ, Stephens EB, Senadheera C, Diaz SR, Peralta S, Alexander L, Silverman-Martin W, Kurtzig J, Fernando BA, Yurkovich JT, Garcia TY, Yukawa M, Morris J, Johnson JB, Newman JC. Exploratory functional and quality of life outcomes with daily consumption of the ketone ester bis-octanoyl (R)-1,3-butanediol in healthy older adults: a randomized, parallel arm, double-blind, placebo-controlled study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.17.24313811. [PMID: 39371165 PMCID: PMC11451762 DOI: 10.1101/2024.09.17.24313811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Background Ketone bodies are metabolites produced during fasting or on a ketogenic diet that have pleiotropic effects on the inflammatory and metabolic aging pathways underpinning frailty in in vivo models. Ketone esters (KEs) are compounds that induce hyperketonemia without dietary changes and that may impact physical and cognitive function in young adults. The functional effects of KEs have not been studied in older adults. Objectives Our long-term goal is to examine if KEs modulate aging biology mechanisms and clinical outcomes relevant to frailty in older adults. Here, we report the exploratory functional and quality-of-life outcome measures collected during a 12-week safety and tolerability study of KE (NCT05585762). Design Randomized, placebo-controlled, double-blinded, parallel-group, pilot trial of 12-weeks of daily KE ingestion. Setting The Clinical Research Unit at the Buck Institute for Research on Aging, California. Participants Community-dwelling older adults (≥ 65 years), independent in activities of daily living, with no unstable acute medical conditions (n = 30). Intervention Subjects were randomly allocated (1:1) to consume 25 g daily of either KE (bis-octanoyl (R)-1,3-butanediol) or a taste, appearance, and calorie-matched placebo (PLA) containing canola oil. Measurements Longitudinal change in physical function, cognitive function and quality of life were assessed as exploratory outcomes in n = 23 completers (n = 11 PLA, n = 12 KE). A composite functional outcome to describe the vigor-frailty continuum was calculated. Heart rate and activity was measured throughout the study using digital wearables. Results There were no statistically significant longitudinal differences between groups in exploratory functional, activity-based or quality of life outcomes. Conclusion Daily ingestion of 25 g of KE did not affect exploratory functional or quality-of-life end points in this pilot cohort of healthy older adults. Future work will address these endpoints as primary and secondary outcomes in a larger trial of pre-frail older adults.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jamie Kurtzig
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - James T Yurkovich
- Buck Institute for Research on Aging, Novato, CA, USA
- Phenome Health, Seattle, WA, USA
| | | | - Michi Yukawa
- Veteran’s Affairs Medical Center, San Francisco, CA, USA
| | | | | | - John C Newman
- Buck Institute for Research on Aging, Novato, CA, USA
- Division of Geriatrics, University of California, San Francisco, CA, USA
| |
Collapse
|
33
|
Joergensen SH, Hansen ESS, Bøgh N, Bertelsen LB, Tougaard RS, Staehr PB, Laustsen C, Wiggers H. Hyperpolarized [1- 13C]pyruvate cardiovascular magnetic resonance imaging identifies metabolic phenotypes in patients with heart failure. J Cardiovasc Magn Reson 2024; 26:101095. [PMID: 39270801 PMCID: PMC11635003 DOI: 10.1016/j.jocmr.2024.101095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Hyperpolarized [1-13C]pyruvate cardiovascular magnetic resonance imaging (HP [1-13C]pyruvate CMR) visualizes key steps in myocardial metabolism. The present study aimed to examine patients with heart failure (HF) using HP [1-13C]pyruvate CMR. METHODS A cross-sectional study of patients with HF and healthy controls using HP [1-13C]pyruvate CMR. Metabolic imaging was obtained using a cardiac-gated spectral-spatial excitation with spiral read-out acquisition. The metabolite signal was analyzed for lactate, bicarbonate, and the alanine signal. Metabolite signal was normalized to the total carbon signal (TC). At the 1-year follow-up, echocardiography was performed in all patients and HP [1-13C]pyruvate MRI in two patients. RESULTS We included six patients with ischemic heart disease (IHD), six with dilated cardiomyopathy, and six healthy controls. In patients, left ventricular ejection fraction (LVEF) correlated with lactate/bicarbonate (r = -0.6, p = 0.03) and lactate/TC (r = -0.7, p = 0.01). In patients with LVEF <30%, lactate/TC was increased (p = 0.01) and bicarbonate/TC reduced (p = 0.03). Circumferential strain correlated with metabolite ratios: lactate/bicarbonate, r = 0.87 (p = 0.0002); lactate/TC, r = 0.85 (p = 0.0005); bicarbonate/TC, r = -0.82 (p = 0.001). In patients with IHD, a strong correlation was found between baseline metabolite ratios and the change in LVEF at follow-up: lactate/bicarbonate (p = 0.001), lactate/TC (p = 0.011), and bicarbonate/TC (p = 0.012). CONCLUSIONS This study highlighted the ability of HP [1-13C]pyruvate CMR to detect changes in metabolism in HF. HP [1-13C]pyruvate CMR has the potential for metabolic phenotyping of patients with HF and for predicting treatment response. TRIAL REGISTRATION EUDRACT, 2018-003533-15. Registered December 4, 2018, https://www.clinicaltrialsregister.eu/ctr-search/search?query=2018-003533-15.
Collapse
Affiliation(s)
- Steen Hylgaard Joergensen
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Esben Soevsoe S Hansen
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Nikolaj Bøgh
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lotte Bonde Bertelsen
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Rasmus Stilling Tougaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Christoffer Laustsen
- The MR Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Henrik Wiggers
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
34
|
Foulks Z, Weinheimer CJ, Kovacs A, Michael J, Pyles KD, Menezes TN, Cho K, Patti GJ, McCommis KS. Ketone Body Metabolism is Not Required for Improvement of Heart Failure by Ketogenic Diet in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610511. [PMID: 39282341 PMCID: PMC11398322 DOI: 10.1101/2024.08.30.610511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Failing hearts increasingly metabolize ketone bodies, and enhancing ketosis improves heart failure (HF) remodeling. Circulating ketones are elevated by fasting/starvation, which is mimicked with a high-fat, low-carbohydrate "ketogenic diet" (KD). While speculated that KD improves HF through increased ketone oxidation, some evidence suggests KD paradoxically downregulates cardiac ketone oxidation despite increased ketone delivery. We sought to clarify the significance of cardiac ketone metabolism during KD in HF. Mice were subjected to transverse aortic constriction with apical myocardial infarction (TAC-MI) and fed either low-fat (LF) control or KD. Cardiac-specific mitochondrial pyruvate carrier 2 (csMPC2-/-) mice were used as a second model of heart failure. In both mice, feeding a KD improved HF, determined by echocardiography, heart weights, and gene expression analyses. Although KD increases plasma ketone bodies, gene expression for ketone metabolic genes is decreased in the hearts of KD-fed mice. Cardiac-specific β-hydroxybutyrate dehydrogenase 1 (csBDH1-/-), the first enzyme in ketone catabolism, mice were also studied and crossed with the csMPC2-/- mice to create double knockout (DKO) mice. These mice were aged to 16 weeks and switched to LF or KD, and KD was able to completely normalize the hearts of both csMPC2-/- and DKO mice, suggesting that ketone metabolism is unnecessary for improving heart failure with ketogenic diet. These studies were then repeated, and mice injected with U-13C-β-hydroxybutyrate to evaluate ketone metabolism. KD feeding significantly decreased the enrichment of the TCA cycle from ketone body carbons, as did the BDH1-deletion in DKO mice. Gene expression and respirometry suggests that KD instead increases cardiac fat oxidation. In conclusion, these results suggest that ketogenic diet decreases cardiac ketone metabolism and does not require ketone metabolism to improve heart failure.
Collapse
Affiliation(s)
- Zachary Foulks
- Edward A. Doisy Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO
| | - Carla J. Weinheimer
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Attila Kovacs
- Center for Cardiovascular Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Jessica Michael
- Edward A. Doisy Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO
| | - Kelly D. Pyles
- Edward A. Doisy Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO
| | - Thiago N. Menezes
- Edward A. Doisy Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO
| | - Kevin Cho
- Departments of Chemistry, Medicine, and the Center for Mass Spectrometry & Metabolic Tracing, Washington University School of Medicine, St. Louis, MO
| | - Gary J. Patti
- Departments of Chemistry, Medicine, and the Center for Mass Spectrometry & Metabolic Tracing, Washington University School of Medicine, St. Louis, MO
| | - Kyle S. McCommis
- Edward A. Doisy Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, MO
| |
Collapse
|
35
|
Ho KL, Karwi QG, Wang F, Wagg C, Zhang L, Panidarapu S, Chen B, Pherwani S, Greenwell AA, Oudit GY, Ussher JR, Lopaschuk GD. The ketogenic diet does not improve cardiac function and blunts glucose oxidation in ischaemic heart failure. Cardiovasc Res 2024; 120:1126-1137. [PMID: 38691671 PMCID: PMC11368127 DOI: 10.1093/cvr/cvae092] [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: 12/06/2023] [Revised: 02/29/2024] [Accepted: 03/17/2024] [Indexed: 05/03/2024] Open
Abstract
AIMS Cardiac energy metabolism is perturbed in ischaemic heart failure and is characterized by a shift from mitochondrial oxidative metabolism to glycolysis. Notably, the failing heart relies more on ketones for energy than a healthy heart, an adaptive mechanism that improves the energy-starved status of the failing heart. However, whether this can be implemented therapeutically remains unknown. Therefore, our aim was to determine if increasing ketone delivery to the heart via a ketogenic diet can improve the outcomes of heart failure. METHODS AND RESULTS C57BL/6J male mice underwent either a sham surgery or permanent left anterior descending coronary artery ligation surgery to induce heart failure. After 2 weeks, mice were then treated with either a control diet or a ketogenic diet for 3 weeks. Transthoracic echocardiography was then carried out to assess in vivo cardiac function and structure. Finally, isolated working hearts from these mice were perfused with appropriately 3H or 14C labelled glucose (5 mM), palmitate (0.8 mM), and β-hydroxybutyrate (β-OHB) (0.6 mM) to assess mitochondrial oxidative metabolism and glycolysis. Mice with heart failure exhibited a 56% drop in ejection fraction, which was not improved with a ketogenic diet feeding. Interestingly, mice fed a ketogenic diet had marked decreases in cardiac glucose oxidation rates. Despite increasing blood ketone levels, cardiac ketone oxidation rates did not increase, probably due to a decreased expression of key ketone oxidation enzymes. Furthermore, in mice on the ketogenic diet, no increase in overall cardiac energy production was observed, and instead, there was a shift to an increased reliance on fatty acid oxidation as a source of cardiac energy production. This resulted in a decrease in cardiac efficiency in heart failure mice fed a ketogenic diet. CONCLUSION We conclude that the ketogenic diet does not improve heart function in failing hearts, due to ketogenic diet-induced excessive fatty acid oxidation in the ischaemic heart and a decrease in insulin-stimulated glucose oxidation.
Collapse
Affiliation(s)
- Kim L Ho
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Qutuba G Karwi
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Faqi Wang
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Cory Wagg
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Liyan Zhang
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Sai Panidarapu
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Brandon Chen
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Simran Pherwani
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Amanda A Greenwell
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
36
|
Trimigno A, Zhao J, Michaud WA, Paneitz DC, Chukwudi C, D’Alessandro DA, Lewis GD, Minie NF, Catricala JP, Vincent DE, Lopera Higuita M, Bolger-Chen M, Tessier SN, Li S, O’Day EM, Osho AA, Rabi SA. Metabolic Choreography of Energy Substrates During DCD Heart Perfusion. Transplant Direct 2024; 10:e1704. [PMID: 39220220 PMCID: PMC11365673 DOI: 10.1097/txd.0000000000001704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
Background The number of patients waiting for heart transplant far exceeds the number of hearts available. Donation after circulatory death (DCD) combined with machine perfusion can increase the number of transplantable hearts by as much as 48%. Emerging studies also suggest machine perfusion could enable allograft "reconditioning" to optimize outcomes. However, a detailed understanding of the energetic substrates and metabolic changes during perfusion is lacking. Methods Metabolites were analyzed using 1-dimensional 1H and 2-dimensional 13C-1H heteronuclear spectrum quantum correlation nuclear magnetic resonance spectroscopy on serial perfusate samples (N = 98) from 32 DCD hearts that were successfully transplanted. Wilcoxon signed-rank and Kruskal-Wallis tests were used to test for significant differences in metabolite resonances during perfusion and network analysis was used to uncover altered metabolic pathways. Results Metabolite differences were observed comparing baseline perfusate to samples from hearts at time points 1-2, 3-4, and 5-6 h of perfusion and all pairwise combinations. Among the most significant changes observed were a steady decrease in fatty acids and succinate and an increase in amino acids, especially alanine, glutamine, and glycine. This core set of metabolites was also altered in a DCD porcine model perfused with a nonblood-based perfusate. Conclusions Temporal metabolic changes were identified during ex vivo perfusion of DCD hearts. Fatty acids, which are normally the predominant myocardial energy source, are rapidly depleted, while amino acids such as alanine, glutamine, and glycine increase. We also noted depletion of ketone, β-hydroxybutyric acid, which is known to have cardioprotective properties. Collectively, these results suggest a shift in energy substrates and provide a basis to design optimal preservation techniques during perfusion.
Collapse
Affiliation(s)
| | | | - William A. Michaud
- Division of Cardiac Surgery, Corrigan Minehan Heart Center, Massachusetts General Hospital, Corrigan Minehan Heart Center, Boston, MA
| | - Dane C. Paneitz
- Division of Cardiac Surgery, Corrigan Minehan Heart Center, Massachusetts General Hospital, Corrigan Minehan Heart Center, Boston, MA
| | - Chijioke Chukwudi
- Division of Cardiac Surgery, Corrigan Minehan Heart Center, Massachusetts General Hospital, Corrigan Minehan Heart Center, Boston, MA
| | - David A. D’Alessandro
- Division of Cardiac Surgery, Corrigan Minehan Heart Center, Massachusetts General Hospital, Corrigan Minehan Heart Center, Boston, MA
| | - Greg D. Lewis
- Division of Cardiac Surgery, Corrigan Minehan Heart Center, Massachusetts General Hospital, Corrigan Minehan Heart Center, Boston, MA
| | - Nathan F. Minie
- Division of Cardiac Surgery, Corrigan Minehan Heart Center, Massachusetts General Hospital, Corrigan Minehan Heart Center, Boston, MA
| | - Joseph P. Catricala
- Division of Cardiac Surgery, Corrigan Minehan Heart Center, Massachusetts General Hospital, Corrigan Minehan Heart Center, Boston, MA
| | | | - Manuela Lopera Higuita
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Shriner Children’s Boston, Boston, MA
| | - Maya Bolger-Chen
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Shriner Children’s Boston, Boston, MA
| | - Shannon N. Tessier
- Center for Engineering in Medicine and Surgery, Harvard Medical School and Massachusetts General Hospital, Shriner Children’s Boston, Boston, MA
| | - Selena Li
- Division of Cardiac Surgery, Corrigan Minehan Heart Center, Massachusetts General Hospital, Corrigan Minehan Heart Center, Boston, MA
| | | | - Asishana A. Osho
- Division of Cardiac Surgery, Corrigan Minehan Heart Center, Massachusetts General Hospital, Corrigan Minehan Heart Center, Boston, MA
| | - S. Alireza Rabi
- Division of Cardiac Surgery, Corrigan Minehan Heart Center, Massachusetts General Hospital, Corrigan Minehan Heart Center, Boston, MA
| |
Collapse
|
37
|
Stubbs BJ, Stephens EB, Senadheera C, Peralta S, Roa-Diaz S, Alexander L, Silverman-Martin W, Garcia TY, Yukawa M, Morris J, Blonquist TM, Johnson JB, Newman JC. Daily consumption of ketone ester, bis-octanoyl (R)-1,3-butanediol, is safe and tolerable in healthy older adults in a randomized, parallel arm, double-blind, placebo-controlled, pilot study. J Nutr Health Aging 2024; 28:100329. [PMID: 39137624 PMCID: PMC11695072 DOI: 10.1016/j.jnha.2024.100329] [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: 05/03/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/15/2024]
Abstract
OBJECTIVES Ketone bodies are endogenous metabolites produced during fasting or a ketogenic diet that have pleiotropic effects on aging pathways. Ketone esters (KEs) are compounds that induce ketosis without dietary changes, but KEs have not been studied in an older adult population. The primary objective of this trial was to assess the tolerability and safety of KE ingestion in a cohort of older adults. DESIGN Randomized, placebo-controlled, double-blinded, parallel-arm trial (NCT05585762). SETTING General community, Northern California, USA. PARTICIPANTS Community-dwelling older adults, independent in activities of daily living, with no unstable acute medical conditions (n = 30; M = 15, F = 15; age = 76 y, range 65-90 y) were randomized and n = 23 (M = 14, F = 9) completed the protocol. INTERVENTION Participants were randomly allocated to consume either KE (25 g bis-octanoyl (R)-1,3-butanediol) or a taste, appearance, and calorie-matched placebo (PLA) containing canola oil daily for 12 weeks. MEASUREMENTS Tolerability was assessed using a composite score from a daily log for 2-weeks, and then via a bi-weekly phone interview. Safety was assessed by vital signs and lab tests at screening and weeks 0, 4 and 12, along with tabulation of adverse events. RESULTS There was no difference in the prespecified primary outcome of proportion of participants reporting moderate or severe nausea, headache, or dizziness on more than one day in a two-week reporting period (KE n = 2 (14.3% [90% CI = 2.6-38.5]); PLA n = 1 (7.1% [90% CI = 0.4-29.7]). Dropouts numbered four in the PLA group and two in the KE group. A greater number of symptoms were reported in both groups during the first two weeks; symptoms were reported less frequently between 2 and 12 weeks. There were no clinically relevant changes in safety labs or vital signs in either group. CONCLUSIONS This KE was safe and well-tolerated in this study of healthy older adults. These results provide an initial foundation for use of KEs in clinical research with older adults.
Collapse
Affiliation(s)
- Brianna J Stubbs
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945-1400, USA.
| | - Elizabeth B Stephens
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945-1400, USA
| | - Chatura Senadheera
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945-1400, USA
| | - Sawyer Peralta
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945-1400, USA
| | - Stephanie Roa-Diaz
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945-1400, USA
| | - Laura Alexander
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945-1400, USA
| | | | - Thelma Y Garcia
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945-1400, USA
| | - Michi Yukawa
- Division of Geriatrics, UCSF, 3575 Geary Blvd, Fl 1, San Francisco, CA 94118-3212, USA; Geriatrics, San Francisco VA Medical Center, 4150 Clement St, San Francisco, CA 94121-1563, USA
| | - Jenifer Morris
- Geriatrics, San Francisco VA Medical Center, 4150 Clement St, San Francisco, CA 94121-1563, USA
| | - Traci M Blonquist
- Biofortis, Mérieux NutriSciences, 800-A South Rohling Rd, Addison, IL 60101-4219, USA
| | | | - John C Newman
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA 94945-1400, USA; Division of Geriatrics, UCSF, 3575 Geary Blvd, Fl 1, San Francisco, CA 94118-3212, USA.
| |
Collapse
|
38
|
Hahn VS, Selvaraj S, Sharma K, Shah SH. Towards Metabolomic-Based Precision Approaches for Classifying and Treating Heart Failure. JACC Basic Transl Sci 2024; 9:1144-1158. [PMID: 39444924 PMCID: PMC11494393 DOI: 10.1016/j.jacbts.2024.04.008] [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: 11/08/2023] [Revised: 03/15/2024] [Accepted: 04/05/2024] [Indexed: 10/25/2024]
Abstract
Both heart failure and cardiometabolic disease are on the rise, and abnormal cardiac and peripheral metabolism are central to the syndrome of heart failure. Advances in metabolomic profiling have improved our understanding of the heart's metabolic flexibility in patients with and without heart failure. Prior studies have noted patients with heart failure display metabolomic profiles associated with marked abnormalities in the metabolism of fatty acids, branched-chain amino acids, ketones, and glucose compared with control subjects. Metabolomics can highlight specific pathways that are dysregulated; however, other metabolites beyond those related to fuel metabolism may also play a role in precision-medicine approaches. Novel approaches include metabolic flux studies, spatial and single-cell analysis, serial monitoring of treatment response, and integration with other -omics data. The goal of these innovative approaches should be to harness metabolomic technologies to affect precision care for patients with heart failure.
Collapse
Affiliation(s)
- Virginia S. Hahn
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Senthil Selvaraj
- Division of Cardiology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Kavita Sharma
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Svati H. Shah
- Division of Cardiology, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
39
|
Volek JS, Kackley ML, Buga A. Nutritional Considerations During Major Weight Loss Therapy: Focus on Optimal Protein and a Low-Carbohydrate Dietary Pattern. Curr Nutr Rep 2024; 13:422-443. [PMID: 38814519 PMCID: PMC11327213 DOI: 10.1007/s13668-024-00548-6] [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] [Accepted: 05/01/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE OF REVIEW Considering the high prevalence of obesity and related metabolic impairments in the population, the unique role nutrition has in weight loss, reversing metabolic disorders, and maintaining health cannot be overstated. Normal weight and well-being are compatible with varying dietary patterns, but for the last half century there has been a strong emphasis on low-fat, low-saturated fat, high-carbohydrate based approaches. Whereas low-fat dietary patterns can be effective for a subset of individuals, we now have a population where the vast majority of adults have excess adiposity and some degree of metabolic impairment. We are also entering a new era with greater access to bariatric surgery and approval of anti-obesity medications (glucagon-like peptide-1 analogues) that produce substantial weight loss for many people, but there are concerns about disproportionate loss of lean mass and nutritional deficiencies. RECENT FINDINGS No matter the approach used to achieve major weight loss, careful attention to nutritional considerations is necessary. Here, we examine the recent findings regarding the importance of adequate protein to maintain lean mass, the rationale and evidence supporting low-carbohydrate and ketogenic dietary patterns, and the potential benefits of including exercise training in the context of major weight loss. While losing and sustaining weight loss has proven challenging, we are optimistic that application of emerging nutrition science, particularly personalized well-formulated low-carbohydrate dietary patterns that contain adequate protein (1.2 to 2.0 g per kilogram reference weight) and achieve the beneficial metabolic state of euketonemia (circulating ketones 0.5 to 5 mM), is a promising path for many individuals with excess adiposity.
Collapse
Affiliation(s)
- Jeff S Volek
- Department of Human Sciences, The Ohio State University, 305 Annie & John Glenn Ave, Columbus, OH, 43210, USA.
| | - Madison L Kackley
- Department of Human Sciences, The Ohio State University, 305 Annie & John Glenn Ave, Columbus, OH, 43210, USA
| | - Alex Buga
- Department of Human Sciences, The Ohio State University, 305 Annie & John Glenn Ave, Columbus, OH, 43210, USA
| |
Collapse
|
40
|
Liang X, Chen J, An X, Ren Y, Liu Q, Huang L, Zhang P, Qu P, Li J. The optimal time restricted eating interventions for blood pressure, weight, fat mass, glucose, and lipids: A meta-analysis and systematic review. Trends Cardiovasc Med 2024; 34:389-401. [PMID: 37838299 DOI: 10.1016/j.tcm.2023.10.002] [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: 08/02/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND No previous systematic review or meta-analysis has evaluated the effect of optimal time-restricted eating (TRE) interventions on cardiovascular (CVD) risk factors. This meta-analysis aimed to illustrate the effect of a suitable TRE on CVD risk factors. METHODS A systematic review was performed to identify trials reporting the effects of TRE, relative to non-diet controls, on CVD risk factors in humans. A random-effects model was used to evaluate the effect sizes, and the results are expressed as the mean difference (MD) and 95% confidence intervals (CIs). Subgroup analyses were performed to examine the influence of the study population, age, duration of intervention, and baseline mean BMI on the CVD indexes. RESULTS TRE intervention significantly reduced systolic pressure (SBP) (MD: -3.45 mmHg; 95%CI:(-6.20,-0.71) mmHg; P = 0.01), body weight (MD: -1.63 Kg; 95%CI:(-2.09,-1.17) Kg; P<0.001), body mass index (BMI) (MD: -0.47 Kg/m2; 95% CI: (-0.72, -0.22) Kg/m2; P<0.001), and fat mass (MD: -0.98 Kg; 95% CI: (-1.51,-0.44) Kg; P<0.001), and reduced blood glucose levels. Based on the results of subgroup analysis, this meta-analysis identified the optimal TRE for BP (with a 6 h feeding window, last eating time point at 6-8 PM, and male participants with obesity and aged ≥ 45 years), obesity (with a 6 h feeding window, last eating time point at 6-8 PM, and female participants aged ≥ 45 years), lipids (with an 8 h feeding window, last eating time point at 6-8 PM, and male participants aged < 45 years), and glucose (with a 10-12 h feeding window, last eating time point before 6 PM, and female participants aged < 45years). CONCLUSIONS Relative to a non-diet control, TRE is effective for the improvement of CVD risks. Moreover, individual TRE interventions should be developed for different populations to achieve the most effective health improvement for CVD risk factors.
Collapse
Affiliation(s)
- Xiaohua Liang
- Department of Clinical Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400016, China.
| | - Jingyu Chen
- Department of Clinical Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400016, China
| | - Xizou An
- Department of Clinical Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400016, China
| | - Yanling Ren
- Department of Clinical Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400016, China
| | - Qin Liu
- Department of Clinical Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400016, China
| | - Lan Huang
- Department of Clinical Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400016, China
| | - Ping Zhang
- Department of Clinical Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400016, China
| | - Ping Qu
- Department of Clinical Epidemiology and Biostatistics, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400016, China
| | - Jianxin Li
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
41
|
Samsø Mathiasen S, Kanta JM, Frydenberg RP, Lundsgaard A, Guo Z, Fritzen AM, Kiens B, Wiking L, Kleinert M. Novel methodology to enrich medium- and short-chain fatty acids in milk fat to improve metabolic health. Food Funct 2024; 15:7951-7960. [PMID: 38980698 DOI: 10.1039/d4fo00267a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Dietary short- and medium-chain fatty acids have been shown to elevate circulating ketone bodies and confer metabolic health benefits. Cow milk fat contains these lipids in a balanced mix but in relatively low concentrations. Enriching them could amplify health benefits of dairy products. Here, we used a volatility-based workflow to produce milk fat with a 2-fold enrichment of medium- and short-chain fatty acids (referred to as MSFAT). Our proof-of-concept studies in mice demonstrated that intake of MSFAT increased circulating ketone bodies, reduced blood glucose levels, and suppressed food intake. In humans, ingestion of MSFAT resulted in increased circulating ketone bodies, trended to attenuate (p = 0.07) postprandial glucose excursion, and acutely elevated energy expenditure. Our findings show that milk products enriched with MSFAT may hold significant metabolic advantages.
Collapse
Affiliation(s)
- Sally Samsø Mathiasen
- Department of Food Science, Aarhus University, Aarhus, Denmark.
- CiFood Multidisciplinary Center for Innovative Foods, Aarhus, Denmark
| | - Josephine M Kanta
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
| | - Rikke P Frydenberg
- Department of Food Science, Aarhus University, Aarhus, Denmark.
- CiFood Multidisciplinary Center for Innovative Foods, Aarhus, Denmark
| | - Annemarie Lundsgaard
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
| | - Zheng Guo
- CiFood Multidisciplinary Center for Innovative Foods, Aarhus, Denmark
- Department of Biological and Chemical Engineering, Aarhus University, Aarhus, Denmark
| | - Andreas M Fritzen
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bente Kiens
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
| | - Lars Wiking
- Department of Food Science, Aarhus University, Aarhus, Denmark.
- CiFood Multidisciplinary Center for Innovative Foods, Aarhus, Denmark
| | - Maximilian Kleinert
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
- Department of Molecular Physiology of Exercise and Nutrition, German Institute of Human Nutrition (DIfE), Potsdam-Rehbruecke, Nuthetal, Germany.
- Institute of Nutritional Sciences, University of Potsdam, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
| |
Collapse
|
42
|
Huang S, Huang X, Liu Z, Yao C, Liu J, He M, Xu X, Zhang T, Wang J, Jiang L, Chen HJ, Xie X. Advances in Multifunctional Electronic Catheters for Precise and Intelligent Diagnosis and Therapy in Minimally Invasive Surgery. ACS NANO 2024; 18:18129-18150. [PMID: 38954632 DOI: 10.1021/acsnano.4c03871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The advent of catheter-based minimally invasive surgical instruments has provided an effective means of diagnosing and treating human disease. However, conventional medical catheter devices are limited in functionalities, hindering their ability to gather tissue information or perform precise treatment during surgery. Recently, electronic catheters have integrated various sensing and therapeutic technologies through micro/nanoelectronics, expanding their capabilities. As micro/nanoelectronic devices become more miniaturized, flexible, and stable, electronic surgical catheters are evolving from simple tools to multiplexed sensing and theranostics for surgical applications. The review on multifunctional electronic surgical catheters is lacking and thus is not conducive to the reader's comprehensive understanding of the development trend in this field. This review covers the advances in multifunctional electronic catheters for precise and intelligent diagnosis and therapy in minimally invasive surgery. It starts with the summary of clinical minimally invasive surgical instruments, followed by the background of current clinical catheter devices for sensing and therapeutic applications. Next, intelligent electronic catheters with integrated electronic components are reviewed in terms of electronic catheters for diagnosis, therapy, and multifunctional applications. It highlights the present status and development potential of catheter-based minimally invasive surgical devices, while also illustrating several significant challenges that remain to be overcome.
Collapse
Affiliation(s)
- Shuang Huang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen 518107, China
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Xinshuo Huang
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhengjie Liu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Chuanjie Yao
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Jing Liu
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Mengyi He
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Xingyuan Xu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Tao Zhang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen 518107, China
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Ji Wang
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Lelun Jiang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen 518107, China
| | - Hui-Jiuan Chen
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Xi Xie
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen 518107, China
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
43
|
Queathem ED, Moazzami Z, Stagg DB, Nelson AB, Fulghum K, Hayir A, Seay A, Gillingham JR, d'Avignon DA, Han X, Ruan HB, Crawford PA, Puchalska P. Ketogenesis supports hepatic polyunsaturated fatty acid homeostasis via fatty acid elongation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602593. [PMID: 39026753 PMCID: PMC11257565 DOI: 10.1101/2024.07.09.602593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Therapeutic interventions targeting hepatic lipid metabolism in metabolic dysfunction-associated steatotic liver disease (MASLD) and steatohepatitis (MASH) remain elusive. Using mass spectrometry-based stable isotope tracing and shotgun lipidomics, we established a novel link between ketogenesis and MASLD pathophysiology. Our findings show that mouse liver and primary hepatocytes consume ketone bodies to support fatty acid (FA) biosynthesis via both de novo lipogenesis (DNL) and FA elongation. Analysis of 13 C-labeled FAs in hepatocytes lacking mitochondrial D-β-hydroxybutyrate dehydrogenase (BDH1) revealed a partial reliance on mitochondrial conversion of D-βOHB to acetoacetate (AcAc) for cytoplasmic DNL contribution, whereas FA elongation from ketone bodies was fully dependent on cytosolic acetoacetyl-CoA synthetase (AACS). Ketone bodies were essential for polyunsaturated FA (PUFA) homeostasis in hepatocytes, as loss of AACS diminished both free and esterified PUFAs. Ketogenic insufficiency depleted liver PUFAs and increased triacylglycerols, mimicking human MASLD, suggesting that ketogenesis supports PUFA homeostasis, and may mitigate MASLD-MASH progression in humans.
Collapse
|
44
|
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
|
45
|
Umemura A, Sasaki A, Kumagai H, Tanahashi Y, Iwasaki T, Nitta H. Relationships Between Changes in Serum Ketone Body Levels and Metabolic Effects in Patients with Severe Obesity Who Underwent Laparoscopic Sleeve Gastrectomy. Obes Surg 2024; 34:2607-2616. [PMID: 38842760 PMCID: PMC11217106 DOI: 10.1007/s11695-024-07337-8] [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: 04/04/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Serum ketone bodies increase due to dynamic changes in the lipid metabolisms of patients undergoing bariatric surgery. However, there have been few studies on the role of ketone bodies after bariatric surgery. We aimed to clarify the role of and relationship between the changes in serum ketone bodies and weight loss, as well as between those changes and the metabolic effects after laparoscopic sleeve gastrectomy (LSG). METHODS We recruited 52 patients with severe obesity who underwent LSG. We measured acetoacetic acid (AcAc) and β-hydroxybutyric acid (β-OHB) at the baseline, 1 month, and 6 months after LSG. Subsequently, we compared the changes in the serum ketone bodies with weight-loss effects and various metabolic parameters. RESULTS At 1 month after LSG, β-OHB significantly increased (p = 0.009), then significantly decreased 6 months after LSG (p = 0.002). In addition, β-OHB in patients without Type 2 diabetes (T2D) and metabolic dysfunction-associated steatohepatitis (MASH) was notably higher than in patients with T2D at 1 month after LSG (p < 0.001). In the early phase, both AcAc and β-OHB mainly had strong positive correlations with changes in T2D- and MASH-related parameters. In the middle term after LSG, changes in both AcAc and β-OHB were positively correlated with changes in lipid parameters and chronic kidney disease-related parameters. CONCLUSION We demonstrated that the postoperative surge of ketone bodies plays a crucial function in controlling metabolic effects after LSG. These findings suggest the cause- and consequence-related roles of ketone bodies in the metabolic benefits of bariatric surgery.
Collapse
Affiliation(s)
- Akira Umemura
- Department of Surgery, Iwate Medical University School of Medicine, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan.
| | - Akira Sasaki
- Department of Surgery, Iwate Medical University School of Medicine, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Hideki Kumagai
- Department of Surgery, Iwate Medical University School of Medicine, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Yota Tanahashi
- Department of Surgery, Iwate Medical University School of Medicine, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Takafumi Iwasaki
- Department of Surgery, Iwate Medical University School of Medicine, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Hiroyuki Nitta
- Department of Surgery, Iwate Medical University School of Medicine, 2-1-1 Idaidori, Yahaba, Iwate, 028-3695, Japan
| |
Collapse
|
46
|
Mansoor T, Khalid SN, Bilal MI, Ijaz SH, Fudim M, Greene SJ, Warraich HJ, Nambi V, Virani SS, Fonarow GC, Abramov D, Minhas AMK. Ongoing and Future Clinical Trials of Pharmacotherapy for Heart Failure. Am J Cardiovasc Drugs 2024; 24:481-504. [PMID: 38907865 DOI: 10.1007/s40256-024-00658-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/30/2024] [Indexed: 06/24/2024]
Abstract
Increasing knowledge of the processes leading to heart failure (HF) has allowed significant developments in therapies for HF over the past few decades. Despite the evolution of HF treatment, it still places a large burden on patients and health care systems across the world.We used clinicaltrials.gov to gather information about clinical trials as of August 2023 studying pharmacotherapy for HF. We included interventional trials that were "active, not recruiting", "recruiting", or looking for participants but "not yet recruiting". In total, 119 studies met our criteria of ongoing clinical trials studying novel as well as currently approved HF pharmacotherapies. The major interventions were novel medications/already approved medications for other diseases 29 % (34 trials), sodium-glucose co-transporter inhibitors 21 % (25 trials), angiotensin receptor blocker-neprilysin inhibitors 10 % (12 trials), diuretics 14 % (17 trials) and mineralocorticoid receptor antagonists 5 % (6 trials). Ongoing research will aid in reducing the impact of HF and we summarize clinical trials leading the way to better HF treatment in this review.
Collapse
Affiliation(s)
- Taha Mansoor
- Department of Internal Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, 1000 Oakland Drive, Kalamazoo, MI, 49008, USA.
| | - Subaina N Khalid
- Department of Internal Medicine, SUNY Upstate Medical University, Syracruse, NY, USA
| | | | | | - Marat Fudim
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Stephen J Greene
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Haider J Warraich
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Vijay Nambi
- Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
- Michael E. DeBakey, Veterans Affair Medical Center, Houston, TX, USA
| | - Salim S Virani
- Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Gregg C Fonarow
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Dmitry Abramov
- Division of Cardiology, Department of Medicine, Loma Linda University Health, Loma Linda, CA, USA
| | | |
Collapse
|
47
|
Horne BD, Anderson JL, Haddad F, May HT, Le VT, Knight S, Bair TL, Knowlton KU. Periodic Fasting and Acute Cardiac Events in Patients Evaluated for COVID-19: An Observational Prospective Cohort Study. Nutrients 2024; 16:2075. [PMID: 38999823 PMCID: PMC11243697 DOI: 10.3390/nu16132075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Periodic fasting was previously associated with greater longevity and a lower incidence of heart failure (HF) in a pre-pandemic population. In patients with coronavirus disease 2019 (COVID-19), periodic fasting was associated with a lower risk of death or hospitalization. This study evaluated the association between periodic fasting and HF hospitalization and major adverse cardiovascular events (MACEs). METHODS Patients enrolled in the INSPIRE registry from February 2013 to March 2020 provided periodic fasting information and were followed into the pandemic (n = 5227). Between March 2020 and February 2023, N = 2373 patients were studied, with n = 601 COVID-positive patients being the primary study population (2836 had no COVID-19 test; 18 were excluded due to fasting <5 years). A Cox regression was used to evaluate HF admissions, MACEs, and other endpoints through March 2023, adjusting for covariables, including time-varying COVID-19 vaccination. RESULTS In patients positive for COVID-19, periodic fasting was reported by 180 (30.0% of 601), who periodically fasted over 43.1 ± 19.2 years (min: 7, max: 83). HF hospitalization (n = 117, 19.5%) occurred in 13.3% of fasters and 22.1% of non-fasters [adjusted hazard ratio (aHR) = 0.63, CI = 0.40, 0.99; p = 0.044]. Most HF admissions were exacerbations, with a prior HF diagnosis in 111 (94.9%) patients hospitalized for HF. Fasting was also associated with a lower MACE risk (aHR = 0.64, CI = 0.43, 0.96; p = 0.030). In n = 1772 COVID-negative patients (29.7% fasters), fasting was not associated with HF hospitalization (aHR = 0.82, CI = 0.64, 1.05; p = 0.12). In COVID-positive and negative patients combined, periodic fasting was associated with lower mortality (aHR = 0.60, CI = 0.39, 0.93; p = 0.021). CONCLUSIONS Routine periodic fasting was associated with less HF hospitalization in patients positive for COVID-19.
Collapse
Affiliation(s)
- Benjamin D. Horne
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (J.L.A.); (H.T.M.); (V.T.L.); (S.K.); (T.L.B.); (K.U.K.)
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA;
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jeffrey L. Anderson
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (J.L.A.); (H.T.M.); (V.T.L.); (S.K.); (T.L.B.); (K.U.K.)
- Cardiology Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Francois Haddad
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA;
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Heidi T. May
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (J.L.A.); (H.T.M.); (V.T.L.); (S.K.); (T.L.B.); (K.U.K.)
| | - Viet T. Le
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (J.L.A.); (H.T.M.); (V.T.L.); (S.K.); (T.L.B.); (K.U.K.)
- Department of Physician Assistant Studies, Rocky Mountain University of Health Professions, Provo 84606, UT, USA
| | - Stacey Knight
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (J.L.A.); (H.T.M.); (V.T.L.); (S.K.); (T.L.B.); (K.U.K.)
- Genetic Epidemiology Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Tami L. Bair
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (J.L.A.); (H.T.M.); (V.T.L.); (S.K.); (T.L.B.); (K.U.K.)
| | - Kirk U. Knowlton
- Intermountain Medical Center Heart Institute, Salt Lake City, UT 84107, USA; (J.L.A.); (H.T.M.); (V.T.L.); (S.K.); (T.L.B.); (K.U.K.)
- Cardiology Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| |
Collapse
|
48
|
Hassan A, Samaan K, Asfour A, Baghdady Y, Samaan AA. Ventricular remodeling and hemodynamic changes in heart failure patients with non-ischemic dilated cardiomyopathy following dapagliflozin initiation. Egypt Heart J 2024; 76:76. [PMID: 38888761 PMCID: PMC11189362 DOI: 10.1186/s43044-024-00508-z] [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: 04/04/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND In heart failure with reduced ejection fraction (HFrEF), sodium-glucose co-transporter inhibitors (SGLT-2i) have persistently shown cardiovascular benefits through different trials. However, their impact on ventricular remodeling and cardiac hemodynamics has not been sufficiently studied. This study aimed to study how SGLT-2i initiation affects invasive hemodynamics and cardiac magnetic resonance imaging (CMR)-derived ventricular volumes, function, and fraction of the extracellular volume (ECV) in HFrEF patients with non-ischemic dilated cardiomyopathy (NIDCM). RESULTS In this study, 23 patients with HFrEF and a mean age of 42, including 82.6% males, all have NIDCM and underwent right heart catheterization and CMR at the initiation of dapagliflozin and at 6-month follow-up. The addition of dapagliflozin resulted in significant reductions in the following invasive hemodynamic parameters compared to baseline: left ventricular end-diastolic pressure (23.4 vs 19.7 mmHg, p = 0.003), mean pulmonary artery pressure (31.3 vs 27.7 mmHg, p = 0.03), and systemic vascular resistance (18 vs 15 Wood units, p = 0.047). Among the studied CMR-derived measurements, only the percentage of extracellular volume fraction was significantly less at follow-up (33.7 vs 32.16%, p = 0.001). Additionally, functional class showed significant improvement with a notable reduction of the NT-proBNP level and a considerable decrease in diuretic dose (median: 40 vs 80 mg, p = 0.01). CONCLUSION Adding dapagliflozin to patients with HFrEF due to NIDCM improved invasively measured hemodynamics and significantly reduced left ventricular extracellular volume fraction measured by CMR, with no significant change in ventricular volumes or ejection fraction.
Collapse
Affiliation(s)
- Ahmed Hassan
- Department of Cardiovascular Medicine, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Kerollos Samaan
- Department of Cardiovascular Medicine, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ahmed Asfour
- Department of Cardiovascular Medicine, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Yasser Baghdady
- Department of Cardiovascular Medicine, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Amir Anwar Samaan
- Department of Cardiovascular Medicine, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
49
|
Bruns B, Joos M, Elsous N, Katus HA, Schultz J, Frey N, Backs J, Meder B. Insulin resistance in Takotsubo syndrome. ESC Heart Fail 2024; 11:1515-1524. [PMID: 38123355 PMCID: PMC11098631 DOI: 10.1002/ehf2.14623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 11/07/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
AIMS Takotsubo syndrome (TTS) is an acute heart failure (AHF) syndrome mimicking the symptoms of acute myocardial infarction. Impaired outcome has been shown, making risk stratification and novel therapeutic concepts a necessity. We hypothesized insulin resistance with elevated plasma glucose and potentially myocardial glucose deprivation to contribute to the pathogenesis of TTS and investigated the therapeutic benefit of insulin in vivo. METHODS AND RESULTS First, we retrospectively analysed patient data of n = 265 TTS cases (85.7% female, mean age 71.1 ± 14.1 years) with documented initial plasma glucose from the Department of Cardiology of the University Hospital Heidelberg in Germany (May 2011 to May 2021). Median split of the study population according to glucose levels (≤123 mg/dL vs. >123 mg/dL) yielded significantly elevated mean heart rate (80.75 ± 18.96 vs. 90.01 ± 22.19 b.p.m., P < 0.001), left ventricular end-diastolic pressure (LVEDP, 18.51 ± 8.35 vs. 23.09 ± 7.97 mmHg, P < 0.001), C-reactive protein (26.14 ± 43.30 vs. 46.4 ± 68.6 mg/L, P = 0.006), leukocyte count (10.12 ± 4.29 vs. 15.05 ± 9.83/nL, P < 0.001), peak high-sensitive Troponin T (hs-TnT, 515.44 ± 672.15 vs. 711.40 ± 736.37 pg/mL, P = 0.005), reduced left ventricular ejection fraction (EF, 34.92 ± 8.94 vs. 31.35 ± 8.06%, P < 0.001), and elevated intrahospital mortality (2.3% vs. 12.1%, P = 0.002) in the high-glucose group (Student's t-test, Mann-Whitney U test, or chi-squared test). Linear regression indicated a significant association of glucose with HR (P < 0.001), LVEDP (P = 0.014), hs-TnT kinetics from admission to the next day (P < 0.001), hs-TnT the day after admission (P < 0.001), as well as peak hsTnT (P < 0.001). Logistic regression revealed significant association of glucose with a composite intrahospital outcome including catecholamine use, respiratory support, and resuscitation [OR 1.010 (1.004-1.015), P = 0.001]. To further investigate the potential role of glucose in TTS pathophysiology experimentally, we utilized an in vivo murine model of epinephrine (EPI)-driven reversible AHF. For this, male mice underwent therapeutic injection of insulin (INS, 1 IU/kg) or/and glucose (GLU, 0.5 g/kg) after EPI (2.5 mg/kg), both of which markedly improved mean EF (EPI 34.3% vs. EPI + INS + GLU 43.7%, P = 0.025) and significantly blunted mean hs-TnT (EPI 14 393 pg/mL vs. EPI + INS 6864 pg/mL at 24 h, P = 0.039). Particularly, insulin additionally ameliorated myocardial pro-inflammatory gene expression, suggesting an anti-inflammatory effect of acute insulin therapy. CONCLUSIONS Elevated initial plasma glucose was associated with adverse outcome-relevant parameters in TTS and may present a surrogate parameter of heightened catecholaminergic drive. In mice, insulin- and glucose injection both improved EPI-induced AHF and myocardial damage, indicating insulin resistance rather than detrimental effects of hyperglycaemia itself as the underlying cause. Future studies will investigate the role of HbA1c as a risk stratifier and of insulin-based therapy in TTS.
Collapse
Affiliation(s)
- Bastian Bruns
- Department of Cardiology, Angiology and PneumologyHeidelberg University HospitalHeidelbergGermany
- Institute of Experimental CardiologyHeidelberg University HospitalHeidelbergGermany
- Department of General Internal Medicine and PsychosomaticsHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Maximilian Joos
- Institute of Experimental CardiologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Nesrin Elsous
- Department of Cardiology, Angiology and PneumologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Hugo A. Katus
- Department of Cardiology, Angiology and PneumologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Jobst‐Hendrik Schultz
- Department of General Internal Medicine and PsychosomaticsHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Norbert Frey
- Department of Cardiology, Angiology and PneumologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Johannes Backs
- Institute of Experimental CardiologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| | - Benjamin Meder
- Department of Cardiology, Angiology and PneumologyHeidelberg University HospitalHeidelbergGermany
- DZHK (German Centre for Cardiovascular Research), Partner SiteHeidelberg/MannheimGermany
| |
Collapse
|
50
|
Genserová L, Duška F, Krajčová A. β-hydroxybutyrate exposure restores mitochondrial function in skeletal muscle satellite cells of critically ill patients. Clin Nutr 2024; 43:1250-1260. [PMID: 38653008 DOI: 10.1016/j.clnu.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 03/17/2024] [Accepted: 04/05/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND & AIM Dysfunction of skeletal muscle satellite cells might impair muscle regeneration and prolong ICU-acquired weakness, a condition associated with disability and delayed death. This study aimed to elucidate the distinct metabolic effects of critical illness and β-OH-butyrate on satellite cells isolated from these patients. METHODS Satellite cells were extracted from vastus lateralis muscle biopsies of patients with ICU-acquired weakness (n = 10) and control group of healthy volunteers or patients undergoing elective hip replacement surgery (n = 10). The cells were exposed to standard culture media supplemented with β-OH-butyrate to assess its influence on cell proliferation by ELISA, mitochondrial functions by extracellular flux analysis, electron transport chain complexes by high resolution respirometry, and ROS production by confocal microscopy. RESULTS Critical illness led to a decline in maximal respiratory capacity, ATP production and glycolytic capacity and increased ROS production in ICU patients' cells. Notably, the function of complex II was impaired due to critical illness but restored to normal levels upon exposure to β-OH-butyrate. While β-OH-butyrate significantly reduced ROS production in both control and ICU groups, it had no significant impact on global mitochondrial functions. CONCLUSION Critical illness induces measurable bioenergetic dysfunction of skeletal muscle satellite cells. β-OH-butyrate displayed a potential in rectifying complex II dysfunction caused by critical illness and this warrants further exploration.
Collapse
Affiliation(s)
- Lucie Genserová
- Department of Internal Medicine of the Third Faculty of Medicine, Královské Vinohrady University Hospital, Charles University, Prague, Czech Republic; Department of Anaesthesia and Intensive Care of the Third Faculty of Medicine, Královské Vinohrady University Hospital, OXYLAB-Laboratory for Mitochondrial Physiology, Charles University, Prague, Czech Republic
| | - František Duška
- Department of Anaesthesia and Intensive Care of the Third Faculty of Medicine, Královské Vinohrady University Hospital, OXYLAB-Laboratory for Mitochondrial Physiology, Charles University, Prague, Czech Republic
| | - Adéla Krajčová
- Department of Anaesthesia and Intensive Care of the Third Faculty of Medicine, Královské Vinohrady University Hospital, OXYLAB-Laboratory for Mitochondrial Physiology, Charles University, Prague, Czech Republic.
| |
Collapse
|