Next Article in Journal
Correction: Chaby et al. Cross-Platform Evaluation of Commercially Targeted and Untargeted Metabolomics Approaches to Optimize the Investigation of Psychiatric Disease. Metabolites 2021, 11, 609
Previous Article in Journal
Evolution of Status of Trace Elements and Metallothioneins in Patients with COVID-19: Relationship with Clinical, Biochemical, and Inflammatory Parameters
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Comparative Metabolomics in Single Ventricle Patients after Fontan Palliation: A Strong Case for a Targeted Metabolic Therapy

1
Fundamental and Biomedical Sciences, Paris-Cité University, 75006 Paris, France
2
Health Sciences Faculty, Universidad Europea Miguel de Cervantes, 47012 Valladolid, Spain
3
Fundacja Recover, 05-124 Skrzeszew, Poland
4
Department of Child and Adolescent Health, Division of Pediatrics I—Inherited Metabolic Disorders, Medical University of Innsbruck, 6020 Innsbruck, Austria
5
Department of Child and Adolescent Health, Division of Pediatrics III—Cardiology, Pulmonology, Allergology and Cystic Fibrosis, Medical University of Innsbruck, 6020 Innsbruck, Austria
*
Author to whom correspondence should be addressed.
Metabolites 2023, 13(8), 932; https://doi.org/10.3390/metabo13080932
Submission received: 3 July 2023 / Revised: 28 July 2023 / Accepted: 3 August 2023 / Published: 9 August 2023
(This article belongs to the Section Endocrinology and Clinical Metabolic Research)

Abstract

:
Most studies on single ventricle (SV) circulation take a physiological or anatomical approach. Although there is a tight coupling between cardiac contractility and metabolism, the metabolic perspective on this patient population is very recent. Early findings point to major metabolic disturbances, with both impaired glucose and fatty acid oxidation in the cardiomyocytes. Additionally, Fontan patients have systemic metabolic derangements such as abnormal glucose metabolism and hypocholesterolemia. Our literature review compares the metabolism of patients with a SV circulation after Fontan palliation with that of patients with a healthy biventricular (BV) heart, or different subtypes of a failing BV heart, by Pubmed review of the literature on cardiac metabolism, Fontan failure, heart failure (HF), ketosis, metabolism published in English from 1939 to 2023. Early evidence demonstrates that SV circulation is not only a hemodynamic burden requiring staged palliation, but also a metabolic issue with alterations similar to what is known for HF in a BV circulation. Alterations of fatty acid and glucose oxidation were found, resulting in metabolic instability and impaired energy production. As reported for patients with BV HF, stimulating ketone oxidation may be an effective treatment strategy for HF in these patients. Few but promising clinical trials have been conducted thus far to evaluate therapeutic ketosis with HF using a variety of instruments, including ketogenic diet, ketone esters, and sodium-glucose co-transporter-2 (SGLT2) inhibitors. An initial trial on a small cohort demonstrated favorable outcomes for Fontan patients treated with SGLT2 inhibitors. Therapeutic ketosis is worth considering in the treatment of Fontan patients, as ketones positively affect not only the myocardial energy metabolism, but also the global Fontan physiopathology. Induced ketosis seems promising as a concerted therapeutic strategy.

1. Introduction

Children with complex congenital heart disease (CHD) and single ventricle (SV) physiology typically undergo several-step surgical palliation with the aim of a total cavopulmonary connection. In this so-called Fontan circulation, the subpulmonary pump is missing. Instead, the vena cavae are anastomosed directly to the pulmonary arteries, causing elevated systemic venous pressure and chronically decreased cardiac output [1,2,3,4].
Even if the outcome of SV patients is steadily improving, particularly their largest subgroup—i.e., SV patients with aortic atresia and a hypoplastic left heart (HLHS, 40% of cases [5]) and a morphologically right ventricle (RV) serving as the subsystemic ventricle—performs worse than patients with a morphologically left ventricle (LV) [5,6,7,8,9,10,11,12,13,14], particularly with respect to atrioventricular valve failure, impaired ventricular function and/or failure of the Fontan circulation with upstream issues such as liver cirrhosis or protein-losing enteropathy [15,16,17]. Ventricular dysfunction is currently considered to be inevitable for SV patients [18]. In addition to these complications, Fontan patients reveal alterations in their pulmonary, hematologic, immunologic, endocrinologic and metabolic systems [1,19].
Pharmacological interventions for both patients with a failing biventricular (BV) and patients with a failing SV heart currently target either hemodynamics or the neuro-hormonal axis, with conflicting evidence regarding the clinical benefit [20,21,22,23,24]. A more recent axis actively targeted in BV HF therapy is the metabolic axis [25,26,27,28,29,30,31,32,33,34,35,36]. Maintaining ketone metabolism is reported to have a protective effect in hypertrophic and failing BV hearts [37,38,39,40,41]. Keeping in mind the metabolic alterations in the Fontan patient [42,43], metabolism may be a promising therapeutic target also in Fontan patients with a failing SV or Fontan circulatory failure.
Reviewing the literature, we compare the energy metabolism in patients with healthy, or with a failing BV heart with that reported in SV patients after Fontan palliation. Focusing on lipid and ketone metabolism, and focusing on the role of therapeutic ketosis in BV HF, a potential role of metabolism-targeted therapy in Fontan patients will be discussed.

2. Cardiac Energy Metabolism

The heart is a biomechanical pump with complex hemodynamics. Ninety percent of the cellular adenosine triphosphate (ATP) is used to sustain the contraction-relaxation of the cardiac muscle [44]. Mitochondria make up one-third of the cardiac myocyte volume [45]. In the whole human body, cardiomyocytes exhibit the highest content in mitochondria, consistent with the fact that the heart is the organ with the highest energy consumption [46]. Thus, it is not surprising that an altered energy state could contribute to HF. The energy starvation model has been proposed as the basis of progressive HF [47,48,49,50], taking into account alterations of the three different stages of ATP production: perturbed substrate consumption; altered oxidative phosphorylation; and reduced energy transfer to ATP-consuming reactions. ATP production is controlled and regulated by a very complex set of transcriptions of metabolically relevant proteins (receptors, regulators, transporters, enzymes) that can be quantified through analysis of metabolites.

2.1. Energy Metabolism in the Healthy Heart

The “omnivore heart” and its diverse substrate consumption. Cardiomyocytes have the capacity to oxidize fatty acids, carbohydrates, amino acids, ketones, and lactate [51]. The substrate preferences are changing with development. The fetus lives in a hypoxic environment, what was called ‘Everest in utero’ [52], and depends on his mother‘s metabolism. Glucose is the dominant substrate used for ATP production, with a very low level of fatty acids and high level of lactate [53,54]. Change of available substrates, increasing oxygen level and improved cardiac workload drive metabolic maturation. Originally, it was thought that within the first week postnatally, glucose use in the heart drops significantly with fatty acid oxidation rising [55], reaching an adult metabolic pattern. Recent studies hypothesize that transition to fatty acid oxidation may start earlier, from the late gestational period [56]. Due to the surge of oxidative capacity—one third of cardiac myocytes’ volume being mitochondria—60–90% of the energy used for mechanical performance originates from mitochondrial fatty acid oxidation, with the remainder originating from glucose, lactate, and ketone bodies [57,58] (Table 1).
Biosynthesis of ketone bodies is connected to various metabolic pathways such as beta-oxidation, Krebs cycle, sterol biosynthesis, glucose metabolism, and mitochondrial electron transport chain [36,99,100]. Among all organs, the heart tissue exhibits the highest levels of ketolytic enzyme activity, reflecting its ability to use ketone bodies [101]. Nonetheless, under physiological conditions, ketone bodies are not contributing significantly to cardiac metabolism [102], glucose and fatty acids being the predominant substrates [61,62].
Substrate utilization is meticulously regulated, which is mandatory due to the relatively small amount of stored ATP relative to the rates of myocytal ATP consumption [49]. One of the control mechanisms is the glucose-fatty-acid-cycle. Numerous parallel mechanisms regulating the substrate utilization in cardiomyocytes were discovered, such as glucose transporter (Glut) 1, Glut 4, or peroxisome proliferator activated receptors (PPAR) [59,103].
From fuel to ATP: oxidative phosphorylation. Oxidative phosphorylation begins with the oxidation of substrates, such as fatty acids and glucose, which are converted into acetyl-coenzyme A (CoA) through pyruvate or beta-oxidation. Acetyl-CoA is fed into the Krebs cycle. Under normal conditions, mitochondrial oxidative phosphorylation matches 90–95% of the myocardial ATP demand with glycolysis filling the gap [61].
From ATP to contractile work: creatine phosphate reserve of ATP. The creatine kinase (CK) energy shuttle plays a crucial role in maintaining energy balance in cells with high and fluctuating energy demands, such as cardiomyocytes: It comprises the conversion of creatine to phosphocreatine (and back), which serves as a rapid and reversible energy storage system. This pathway enables the efficient transfer of energy from the mitochondria, where ATP is produced by oxidative phosphorylation, to the cytosol, where it is consumed during contraction.
Regulation of metabolism through gene expression. Control and regulation of cardiac metabolism are complex and include overexpression or deletion of metabolically relevant proteins, such as receptors, regulators, transporters, or enzymes.
Peroxisome proliferator activated receptors. PPAR are a family of nuclear receptor proteins that play a crucial role in regulating the expression of genes involved in lipid and glucose metabolism, inflammation, and cellular differentiation. PPAR is an abundantly expressed key regulator of cardiac substrate switching [104,105,106,107], including fatty acid oxidation, ketogenesis, and triglyceride synthesis, by upregulation of genes involved in fatty acid metabolism. Activation of PPAR upregulates genes of fatty acid oxidation (fatty acid transport protein/cluster of differentiation 36, malonyl-CoA decarboxylase, carnitine acyltransferase (CPT)-1, medium and long chain acyl-CoA dehydrogenases) [102]. PPAR was downregulated by hypoxia in rats’ hearts [108].
Adenosine monophosphate-activated protein kinase pathway, GLUT and CPT. The adenosine monophosphate-activated protein kinase (AMPK) pathway is a critical cellular signaling pathway that plays a crucial role in regulating energy homeostasis in cells. The activation of the AMPK pathway leads to the phosphorylation of numerous downstream targets involved in metabolism, transcription, and protein synthesis. Through this pathway, CPT1, medium-chain acyl-CoA dehydrogenase, cluster of differentiation 36, and fatty acid transport protein 1 are decreased in HF [61]. Inhibition of CPT1 directly inhibits fatty acid oxidation by malonyl-CoA—a phenomenon called reverse Randle effect [105].
Histone acetylation/deacetylation. Acetylation is a protein post-translational modification controlling expression and transcription of genes, regulating embryonic development, postnatal fatty acid oxidation maturation, and heart hypertrophy. It enables the cell to quickly and effectively react to cellular stress [109]. Histone acetylation regulates the electrostatic connections between DNA and histones as well as between adjacent nucleosomes within a nucleosomal fiber, which controls transcription [110]. Histone acetylationtransferases (HAT), also known as lysine acetyltransferases, which relax chromatin structure, and histone deacetylases (HDAC) which reverse the HAT process, reduce transcriptional activity and are the main regulators of HAT [111]. HDAC can be divided into two types based on their architectures and patterns of expression [112]. All tissues express class I HDAC. Class II histone deacetylases interact with the MEF2 transcription factor to control fetal cardiac and stress-responsive genes [113,114]. Activity of GLUT1, GLUT4, PDK2, muscle-glycogen synthase, mCPT-1, MCAD, and ACC is higher in the non-failing adult human heart than in the fetal heart. In the failing human heart, those metabolic genes‘ activities decrease to the same levels as in the fetal heart [60].

2.2. Energy Metabolism in Biventricular Patients with Congestive Heart Failure

Disturbed substrate consumption. Once the heart has reached its metabolic adulthood, the main substrates used for ATP production are fatty acids (60–70%), followed by pyruvate (glucose/lactate), ketone bodies, and amino acids. In early stages of HF, myocardial fatty acid utilization may be unchanged or augmented. In advanced stages, the myocytes switch from fatty acid to glucose oxidation, returning to a fetal pattern of energy substrate metabolism [48,59,60] (Table 1). Furthermore, myocytes may become insulin-resistant, leading to a decline in glucose/pyruvate utilization (metabolic inflexibility) [60,61,62,63,64,65,66]. Even though glucose uptake is increased, it does not always translate into increased glucose oxidation. Through overexpression of Glut1, the uptake increases glycolysis. Per glucose molecule, glycolysis produces two molecules of ATP compared to 31 molecules of ATP by oxidation. Thus, the energy deficit is not compensated for by substrate switch [115,116].
As described by Ritterhof et al. there is an upregulation of glucose uptake and glycolysis with either no change or even a decrease in glucose oxidation, resulting in uncoupling of substrate uptake and oxidation [59]. Ultimately, this uncoupling reduces cardiac energy availability, the affected heart exhibiting up to 30% less ATP than the healthy one [46,117].
Myocardial ketone body oxidation is increased in HF. Recent studies show this metabolic shift as a key metabolic adaptation in the failing human heart, indicating the potential of ketone bodies as an alternative fuel for HF with reduced and preserved ejection fraction (EF) [35,65,67,68,69,70,71,72,73,74,75]. An increased ketone body oxidation is also seen in RV failure like in pulmonary arterial hypertension (PAH) [118].
Reduced energy production. In the failing BV heart, electron transport chain activity is altered (Table 1). Alterations in mitochondrial number, structure and function, in part due to accumulation of reactive oxidative species (ROS) harming mitochondrial deoxyribonucleic acid, may be causes of altered electron transport chain activity [97,119].
Increase in reactive oxidative species. Oxidative stress is involved in the development and progression of cardiac remodeling in HF [76]. ROS impair the electrophysiology of the contractile function by denaturing proteins involved in contractility (including L-type calcium channels, sodium channels, potassium channels, and sodium-calcium exchangers [120]) and trigger hypertrophy through modifications in the extracellular matrix [121].
Insufficient energy transfer to ATP-consuming reactions. CK energy transfer shuttle works by transferring high-energy phosphate groups from creatine phosphate to ADP to produce ATP. It was the initial mechanism of energy starvation discovered in HF, with creatine deficiency [47]: In HF, the CK system is compromised due to a variety of factors, including decreased levels of CK enzymes and alterations in the composition of the mitochondrial membrane (Table 1). As a result, ATP levels in the heart decrease. Phosphocreatine and total creatine levels decrease by up to 30–70% in an early stage of HF [48,77,78,79]. Consequence is the inability to deliver ATP on increased workload [122]. Reduced CK flux is a significant predictor of HF outcome [117]. Acting on reduced CK flux and on its enzymes is considered a potential HF treatment target [117,123].
Epigenetic and transcriptional changes: reactivation of fetal gene expression in the failing BV heart. The activation of fetal cardiac genes which encode proteins involved in contraction, calcium management, and metabolism, is associated with pathological heart hypertrophy. A deterioration in heart function is associated with such reprogramming. Conversely, improvement in cardiac function is associated with normalization of cardiac gene expression in the failing heart [124]. The maturation of fatty acid oxidation was delayed according to a clinical investigation based on RV myocardial biopsies from patients with CHD. Key metabolic enzyme hyperacetylation was prevented by secondary hypertrophy [125]. Lysine acetylation is involved in regulating cardiometabolic diseases. Nicotinamide adenine dinucleotide-dependent deacetylase sirtuin-3 (SIRT3) expression was downregulated in failing hearts from patients with obesity and metabolic syndrome, which led to cyclophilin D hyperacetylation, hyperacetylation, mitochondrial permeability transition pore opening, and cardiac dysfunction [126]. In pressure overload-induced cardiac hypertrophy, histone acetylation is related to inflammation, collagen deposition, and cardiac contractile function [127]. Histone deacetylase inhibition was found to reduce cardiac hypertrophy and fibrosis in spontaneously hypertensive rats by increasing 3-acetylation on the promoters of MR target genes and suppressing gene expression [128]. The HDAC inhibitor valproic acid was shown to improve the development of atrial remodeling and postpone the onset of atrial fibrillation in mice by 4 to 8 weeks [129]. Another HDAC inhibitor, suberoylanilide hydroxamic acid, was discovered to prevent cardiac arrhythmias in dystrophic mice, including QT interval prolongation [130]. Both class I and class II HDAC are inhibited by the HDAC inhibitors that prevent LV hypertrophy.
Metabolomics as a bridge between gene and metabolism. Metabolomics is more and more used to analyze metabolites and intermediates in cardiology [131], as well as pediatric cardiology [132,133]. Interpretation can be complex.
Ketolytic metabolism was described as being increased in HF [134]. In atrial fibrillation, elevated blood levels of acylcarnitine were found and discussed as reflective of defective mitochondrial beta-oxidation [135,136].

2.3. Right Ventricular Failure: The Case of Pulmonary Arterial Hypertension

In fetal circulation, the right ventricle (RV) is responsible for up to 60% of the cardiac output [137]. Thickness and contractility are likely to be similar between LV and RV at this stage [138]. However, there are deep metabolic and electrophysiological differences between LV and RV, such as differences in systolic Ca2+ [139]. The molecular structure of the RV is quite different from that of the LV [140,141]. Metabolic pathways and gene transcription mechanisms in the state of pressure-overload are different in LV and RV [142]. Dysfunction of the RV is accompanied by a release of ROS, combined with chronic nitric oxide deficiency [143]. Additionally, in RV hypertrophy specific mitochondrial remodeling was described [144].
RV failure shares with LV failure the perturbation in substrate consumption. Glucose homeostasis as well as fatty acid metabolism are impaired [82,83,84,145,146,147,148]. Acylcarnitine levels are disrupted in RV failure and have been associated with insulin resistance and adverse outcome [149,150,151,152].
Untypical substrate consumption. PAH is a disease resulting in RV failure. Initially regarded an isolated disease of the pulmonary circulation, evidence is accumulating that PAH is to be considered from the metabolic perspective [153,154].
Abnormal glucose oxidation. Abnormal glucose metabolism is observed in patients with PAH, even without manifest diabetes mellitus [82,83,84] (Table 1). In Pugh et al. [82], 56% of patients with PAH had an Hb1ac level of over 6.0%, and 15% of patients of over 6.5%—this being undiagnosed DM, reminding of findings in Fontan patients of Ohuchi et al. [155]. Insulin resistance worsened pulmonary phenotype in the West study, implying a possible causal role in PAH [83,85].
Altered fatty acid oxidation. Parallel to abnormal glucose metabolism and insulin resistance, alterations of fatty acid oxidation were found in PAH [81]. In plasma, circulating free fatty acids and acylcarnitines are significantly elevated in a similar pattern as found in Fontan patients [43]. In an animal model, altered carnitine function was tied to decreased mitochondrial function and altered nitric oxide signaling [156].
Increased ketogenesis. An increased conversion of fatty acids into ketones (ketogenesis) was found to correlate with better clinical health in PAH [118]. This is consistent with the adaptively increased uptake of ketones in HF [69]. Aarhus university is conducting a clinical trial on ketones for PAH patients [86]. There are some rationales behind such an approach. In a previous study, they found a 40% increase in cardiac output under treatment with beta-hydroxybutyrate (BHB) infusion, with an increase in RV function as well as a decrease in pulmonary vascular resistance by 20%. Their published results focus on the hemodynamic effects with an average increase in cardiac output by 27%, and a decrease in pulmonary vascular resistance by 18%, irrespective of the cause of right-sided HF (10 patients with PAH, 10 patients with chronic thromboembolic pulmonary hypertension), and at average a blood ketones level of 3.3 mmol/L [87].
While our article was submitted, a non-reviewed article from the Lillehei heart institute (Minnesota university) describes that compensatory ketosis is absent in RV failure, in contrast to LV failure. The therapeutic stimulation of ketolytic activity is improving RV function, suppresses NLRP3 inflammasome activation and blunts myocardial fibrosis [88]. Those authors hypothesized an RV-liver-axis behind this specific RV dysregulation.
Oxidative stress. The electron transport chain in PAH shows pronounced alterations [89,90]. Proteomics studies in PAH identified an increased ROS production that might be related to a loss of antioxidant response [91,92].
Decreased creatine kinase shuttle. Alterations in CK were found in diastolic dysfunction in an animal model with RV HF [93,157].
Alterations in signaling pathways. Histone deacetylase inhibitors are acting differently in LV, or RV hypertrophy [158,159]. Due to the complex pathogenesis of PAH, more than a single epigenetic modulation to reverse PAH might be required [160].
Metabolomics findings. In right-sided HF, metabolomics analysis found specifically elevated blood levels of L-carnitine, acetyl-L-carnitine and long-chain acylcarnitine. Alteration of beta-oxidation of fatty acids increases the concentration of acyl-CoA, thus increasing acylcarnitine levels. The conclusion is that an increased level of acylcarnitine may reflect significant inhibition of the mitochondrial fatty acid beta-oxidation in PAH [161].

2.4. Energy Metabolism and the Single Ventricle after Fontan Palliation

Altered substrate consumption—glucose oxidation. Pyruvate metabolism might be altered in SV patients, especially in those with HF. One metabolomics study found elevated levels of circulating pyruvate in this group of patients, which might indicate an alteration of pyruvate metabolism and glucose oxidation [95] (Table 1). Noteworthy is that the authors consider pyruvate therapy.
Altered substrate consumption—fatty acid oxidation. From biopsies collected during cardiac surgery, it was discovered that cardiac metabolic maturation happened in HLHS (dominant RV) through an increase in AMPK and PPAR-gamma coactivator 1 alpha, and that control of fatty acid oxidation is not impaired in the hearts of HLHS children [162]. This suggests beta-oxidation to work properly. Thus, SV HF might follow a metabolic pattern similar to that of BV HF, exhibiting the reverse Randle effect [105].
Surprisingly, in a very recent metabolomics study functional analysis of the mitochondrial CPT system demonstrated significantly decreased activity of the mitochondrial CPT transporters, suggesting that the diminished myocardial acylcarnitine is related to an overall decreased capacity of the failing SV to oxidize long-chain fatty acids, resulting in a diminished rate of cardiac ATP production [94]. Those findings suggest that due to the decreased enzymatic activity of CPT1/CPT2, beta-oxidation is altered in univentricular hearts which would explain the higher acylcarnitine levels found in Fontan patients compared to healthy controls [43].
Another recent metabolomics study suggests differences in 2-oxoglutarate, isocitric acid, malic acid, and cis-aconitic acid that could reflect alterations of the Krebs cycle [163]. The authors suggest Krebs cycle activation might be necessary to increase cardiac output to counteract hypoxia in Fontan patients. Nevertheless, this interesting finding would require further research, as the study compares metabolomics of various single ventricle architecture to different malformations with inhomogeneous presence or grade of pressure or volume burden such as tetralogy of Fallot or ventricular septum defects. Moreover, the number of patients included is low (n = 14), and the study includes patients with severe atrioventricular valve regurgitation or a history of protein-losing enteropathy.
Induced ketolysis in single ventricle physiology—early reports on the use of Sodium-glucose co-transporter-2 inhibitors. The decreased activity of CPT2 in SV patients without HF, and CPT1/CPT2 in SV patients with HF [94], as well as elevated circulating level of carnitine [95] might indicate metabolic perturbations. Pires da Silva et al. consider pyruvate therapy [95,164]. This is part of an approach to compensate for the decreased activity of CPT1/CPT2 by therapeutic use of cardiac anaplerosis [165]. An alternative approach is to make use of therapeutic ketosis or induced cardiac ketolysis. One means to stimulate ketone metabolism is the use of sodium-glucose co-transporter-2 (SGLT2) inhibitors as studied in BV HF [166,167,168,169,170,171], an approach supported by a case report (Fontan circulation, 5 patients), where rehospitalization rate was reduced without acute adverse effects [172]. Although based on a very limited patient number, objective data such as increases in systemic oxygen saturation, serum albumin level, and estimated glomerular filtration rate, as well as a decrease in plasma NT-proBNP-level are promising, as NT-proBNP > 100 pg/mL has a 91% sensitivity for significant ventricular dilation, and one of 82% for ejection fraction <50% [173], and is predictive of adverse outcome [174].
While SGLT2 inhibitors’ mechanism still is not completely understood, one of the main hypotheses is that SGLT2 inhibitors are increasing circulating ketone levels [168]. The improvement of parameters seen in the first use of SGLT2 inhibitors on Fontan patients match the effects of ketone bodies on hypoxia [175] and glomerular filtration rate [176], as reported in non-Fontan studies.
Inflammation and oxidative stress. In HLHS patients, disorders of cellular respiration are suspected to be present [96]. In an animal model, untypically elevated mitochondrial respiratory capacity was discovered, different from the reduced respiratory capacity typically seen in BV LV HF [96,177]. The cause of this elevation is controversial. Based on the Ohia mouse model, Xu et al., hypothesized a genetic mutation [96]. Other investigators suggested an alteration in the cardiac metabolic maturation [178]. We hypothesize that the unphysiological ventricle switch with its mechanical load changes may trigger metabolic alterations. Irrespective of the cause, such a mechanism is able to trigger an elevation of ROS, known to be one mechanism leading to cardiac hypertrophy [76,97,98]. A recent Proteomics study on young children with an SV lesion (prior to Fontan palliation) showed decreased inflammatory cytokines and increased vascular tone modulators compared to healthy controls before stage 2 palliation, and an increase in these analytes shortly after stage 2 palliation [179]. Interestingly, tissue inhibitor of metalloproteinases-1 or and matrix metalloproteinase 7 levels were associated with greater morbidity, suggesting an important role for regulation of extracellular matrix production.
Systemic metabolic changes. Abnormalities in Fontan patients’ glucose and lipid metabolism as well as in the neurohumoral axis have been reported, and this even in ‘stable’ Fontan patients with good exercise capacity and without signs of imminent Fontan failure [19,180,181,182,183,184,185], in part revealing similarities to findings in the BV patient with LV or RV failure. Interestingly, metabolomics studies found that even Fontan patients with good ventricular function and without signs of a Fontan failure exhibited similar lipid metabolic pattern as the BV patient with HF, particularly with respect to alterations in serum cholesterol, lipoprotein, phospholipid, and acylcarnitine concentrations [43]. Moreover, there are first reports on alterations in amino acid pathways, with some key analytes showing altered serum levels (asparagine, histidine, taurine, threonine; amino acid-derived analytes such as dimethylarginine, methionine-sulfoxide or glutamic acid), hinting at inflammation, oxidative stress and endothelial dysfunction, altered cell energy metabolism, and elevated myocardial turnover [42], similar to those found in BV patients with congestive HF.

3. Induced Ketosis in Patients with a Failing Biventricular Heart

Initially being an intuition in biochemistry [51], evidence accumulates on the therapeutic role of targeting mitochondrial oxidative metabolism [186], and especially ketone metabolism for the BV LV HF [32]. In the development of HF, cardiac alterations of metabolic processes contribute to a reduction of ATP availability, causing a decline in myocyte contractile function. In advanced HF, ketone metabolism is increased; its nature is discussed to be adaptive [29,102,187]. Nevertheless, therapeutic elevation of ketone bodies may have positive results, not only for cardiometabolic health in general but also systemic health in patients with HF in particular [188]. To date, research in this field is conducted on both LV and RV HF.
Ketones as a substrate for the failing heart. Early experimental studies showed that BHB is not only a fuel, but a super fuel for the heart [189,190,191,192,193,194]. Twenty-five percent increased contractility and decreased oxygen consumption were found at 5 mM blood level of ketone bodies [191]. It was discovered that in the healthy heart, ketones do not increase cardiac efficiency. However, ketone fuels are capable of increasing ATP production when the main cardiac fuels—fatty acids and glucose—are deficient [195]. Clinical applications are currently in development, for congestive and acute HF [188,196,197].
Ketones and oxidative stress. Oxidative stress is involved in the progression of congestive HF, showing a positive correlation between elevated oxidative stress and myocardial dysfunction [76,121,198,199]. BHB is an endogenous specific inhibitor of class I HDAC. In an experimental study, elevated levels of BHB inhibited HDAC, correlating with changes in transcription including those of the genes encoding oxidative stress resistance factors, conferring substantial protection against oxidative stress with decreased ROS production [200]. Oxidation of ketone bodies contributes to free radical homeostasis [40]. Reduced oxidative stress was also observed in mice using a ketogenic diet [201].
Ketones and inflammation. Inflammation is associated with cardiac remodeling and HF [202,203,204]. NLRP3 inflammasome is a new therapeutic target in the treatment of HF [205]. BHB is inhibiting NLRP3 inflammasome [206]. In ketogenic-diet-fed mice, BHB serum levels increased to 0.75–1 mM, and inhibited activation of NLRP3 inflammasome. Overexpression of D-beta-hydroxybutyrate dehydrogenase I enhanced antioxidant enzyme expression and attenuated peroxide-induced apoptosis [39]. In neurons, ketone bodies were shown to decrease mitochondrial production of ROS without affecting the endogenous antioxidant glutathione [207]. Consistently, a low-carbohydrate diet reduced inflammation [208]. High-fat diet elevating BHB was able to reduce inflammation and mitigate HF with preserved EF.
Ketones and mitochondrial respiratory complex activity. In an experimental model, ketogenic diet normalized complex I and improved complex II-III activities in rats [209]. This was discussed as originating from providing an alternative substrate as well as through the ketone-mediated downregulated oxidative stress.
Ketones, myocardial contractility, and ventricular ejection fraction. Infusion of 3-hydroxybutyrate to patients with HF with reduced EF increased the EF by 8% [80]. At the same time, cardiac output increased by 40%, with a concomitant increase in RV function and decrease of pulmonary vascular resistance by 20% each.
Ketones, myocardial remodeling, and prevention of cardiac hypertrophy. Inefficient myocardial fuel consumption can cause pathological hypertrophy [210,211,212,213]. Class I HDAC have been found responsible in the development of pathological cardiac hypertrophy and HF [124,214]. HDAC inhibition was found to be a therapeutic target for cardiac remodeling [215]. Overexpression of BBH dehydrogenase 1 has a protective role regarding resilience to pressure overload-induced cardiac remodeling [39]. In an experimental model, BHB infusion increased histone acetylation in the heart, inhibiting HDAC [200]. In preclinical model HF, elevation of BHB through ketone esters reduced pathologic remodeling [216]. Chronic elevation of BHB in dogs decreased adverse remodeling [38]. In a similar way, strict dietary carbohydrate restriction, causing elevation of ketone levels and decreasing mammalian target of rapamycin expression, suppressed hypertrophy in experimental studies [217,218]. This phenomenon was discussed to have clinical implications. In patients with HF with reduced EF, acute infusions of BHB improved contractility [80].
Ketones, endothelial function, and vascular resistance. Keeping pulmonary vascular resistance low is necessary in Fontan circulation [219,220]. In BV patients with HF, endothelial dysfunction, induced by oxidative stress, and elevated vascular resistance contribute to the development of HF and are associated with an increased mortality [221,222]. Mechanisms involved are complex, including oxidative stress, inflammation, and alteration of nitric oxide metabolism. The ketone body BHB presented as a potent vasodilator both in experimental models and human trials [223]. Under BHB infusion, myocardial blood flow increased and induced vasodilation [80,224]. BHB infusion increased blood flow in the renal system [170]. Ketone ester reduced risk of aortic dissection [225].
Antiarrhythmic potential of ketones. Ketone oxidation, membrane excitability, and arrhythmogenesis are interrelated. BHB contributed to a 24% improvement in cardiac efficiency, mitochondrial function and the stabilization of cellular membrane potential, enhancing the antiarrhythmic potential of the myocardial cell [36].
Ketones and oxygen consumption. Ketone oxidation spares oxygen consumption and is neuroprotective through two mechanisms, oxygenation improvement and decreased blood carbon dioxide [226,227]. Compared to fatty acid oxidation, ketones produce more ATP per molecule of oxygen [228,229].
Ketones in RV failure. Nutritional ketosis improved PAH through reversal of the metabolic syndrome [230]. In a similar way, use of the SGLT2 inhibitor empagliflozin prevented the progression of PAH [231]. A clinical trial is currently conducted on the use of an OHB infusion in patients with idiopathic PAH [86] (Table 1).

4. Impact of Ketones Apart from That on the Cardiovascular System Relevant to Fontan Circulation

The Fontan circulation with its elevated central venous pressure and limited cardiac output has consequences on all organ systems [19,232]. Ketone bodies are organ-protective. In the following, we aim to estimate to which extent Fontan circulation pathophysiology benefits from this protection (Figure 1).
Hepatoprotection. Deficiencies in hepatic ketogenesis are associated with non-alcoholic fatty liver disease and fibrosis [233]. Therapeutic ketosis was explored in the treatment of non-alcoholic fatty liver disease, with effective improvement and reduction of fibrosis [234,235,236,237].
Renoprotection. Numerous studies show detrimental effects of the Fontan circulation on the kidneys [238,239,240,241,242]. Renal ketogenesis requires further exploration. Nevertheless, renal ketogenesis is reported to be a mechanism protecting against renal ischemia-reperfusion injury [243,244].
Neuroprotection. Mild hypoxia is a common feature of the Fontan circulation [245], and ketones could be a therapeutic strategy to counteract the effects of this hypoxia. For certain anatomical variants of a SV, optimizing oxygen consumption might be critical considering altered coronary perfusion. A ketogenic diet improved the cerebral oxygen level in hypoxia after an epileptic event [175]—the assumed mechanism being altered substrate consumption, reduced glycolysis, and accumulation of lactate [246]. Exogenous ketones increased blood and muscle oxygenation in hypoxia [247].
Lymphangiogenesis. Evidence is appearing on the role of ketogenesis on the lymphangiogenesis after corneal injury and myocardial infarction in the BV patient [248]. The mechanism is also on trial for alleviating lymphedema [249,250]. Whether ketone bodies might play a therapeutic role in abnormal lymphatic flow in Fontan circulation requires further research.
Abnormal glucose patterns. Abnormal glucose metabolism is one of the underdiagnosed complications of Fontan circulation [155,180]. Alteration of insulin sensitivity is hypothesized to be a factor of Fontan-associated liver disease, which affects a high percentage of Fontan patients [251,252]. Having in mind that ketogenic diet and carbohydrate restriction are established therapies to treat insulin resistance and to improve glycemic control [253], facing the abnormal glucose metabolism inherent to Fontan, the therapeutic potential of ketogenic diet and carbohydrate restriction might be worth considering.

5. Rationale of a Targeted Metabolic Therapy in Fontan Patients

Current approaches to HF in Fontan patients are widely based on treatment regimens for BV HF. Their application varies among centers, and polymedication is common, often with the potential of complex drug interaction [22,23]. Apart from the therapeutic potential of ketone bodies in BV HF that should apply to SV patients, there are features unique to Fontan circulation that—based on the effects on similar complications—might benefit from therapeutic ketosis.
Therapeutic modulation of the ketone pathway. SGLT2 inhibitors are considered to positively address HF through an increase in cardiac ketone oxidation [168]. The first trial of SGLT2 inhibitors on Fontan patients is promising [172]. Though the trial size was limited, the benefits seen in the study support therapeutic ketosis in HF. SGLT2 inhibitor use is currently supported by FDA only for patients over 18 years old [254]. Other means to achieve therapeutic ketosis including ketogenic diet, medium chain triglycerides supplementation, or application of ketone esters (Figure 2).
A strong case for therapeutic ketosis in Fontan patients. Therapeutic ketosis has been in use already for 100 years in pediatric neurology [255]. Numerous reviews judge it a safe dietary therapy with minor adverse effects to be monitored [256,257]. It is a sustainable therapy [258], and it is used from toddler age to adult age [259]. The therapy has spread from pediatric neurology to other fields, such as certain inherited metabolic disorders [260], adults’ BV congestive LV HF [216,217,218], intensive care management [261], or oncology [262]. Due to the unique nature of SV patients and the potential of interactions with their polymedication commonly present, induction of ketosis should only be undertaken under close medical control.
Additional candidate mechanisms for metabolism-targeted therapy for Fontan patients. Cardiac anaplerosis might be able to replete tricarboxylic acid intermediates and alleviate the substrate consumption alterations [165]. Among other regimens, there is pyruvate therapy [164], application of glutamine [263] (PAH patients showed an increased anaplerosis under glutamine [264]), or branched-chain amino acids [265] and odd-chain fatty acids [266,267]. Octanoate is modulating metabolic acetyl-CoA histone acetylation, promoting cardiac repair after myocardial infarction [268]. It is currently applied to Fontan patients with lymphatic complications such as protein-losing enteropathy (supplementation of the low-fat diet with medium chain triglycerides) [269]. It is unclear how octanoate could bypass CPTI/CPTII transporter and act as a fuel in the mitochondria in the cardiomyocyte, the results being controversial [270,271].

6. Limitations

SV malformation is a rare disease. Most studies on cardiac and systemic metabolism of Fontan patients were conducted on a limited number of patients. The etiology of all SV malformations might not be identical, and metabolism as well as gene transcription might vary. Furthermore, metabolomics results should be treated with caution as interpretation can be complex. Large-scale (multicenter) studies are necessary to further explore the metabolic impairment of the SV heart and the ketones‘ therapeutic potential in the respective patients.

7. Methods

Our literature review compares the metabolism of patients with an SV circulation after Fontan palliation with that of patients with a healthy BV heart, or different subtypes of a failing BV heart, by Pubmed review of the literature on cardiac metabolism, Fontan failure, heart failure, ketosis, metabolism, published in English from 1939 to 2023.

8. Conclusions

Evidence is growing that SV circulation after Fontan palliation not only is a hemodynamically challenging state, but also raises a metabolic issue with its alterations of fatty acid oxidation as well as glucose oxidation, similar to those reported in the failing BV heart, generating metabolic instability and disturbed energy production that per se may become a cause of circulatory failure. Evidence accumulates that stimulating ketone oxidation as a targeted metabolic therapy might be a therapeutic strategy to address HF in BV patients. Therapeutic ketosis may be worth considering also in the treatment of Fontan patients, as ketones positively affect not only the myocardial energy metabolism, but also the global Fontan pathophysiology. Induced ketosis seems promising as a therapeutic strategy for chronic ventricular failure and low-grade inflammation, as well as diseased liver, kidney, and intestines. Ketone esters, 1,3 butadeniol, or octanoate might provide a third fuel to the SV heart, and a well-formulated ketogenic diet taking into account the micronutritional status inherent to Fontan patients may have an integrative, concerted effect on the several complications of Fontan circulation.

Author Contributions

Conceptualization, D.R. and M.M.; investigation, D.R. and M.M.; resources, M.M.; writing—original draft preparation, D.R.; writing—review and editing, M.M., S.S.-B. and D.K.; supervision, M.M.; project administration, D.R. and M.M.; funding acquisition, M.M. All authors have read and agreed to the published version of the manuscript.

Funding

Open Access Funding by the Austrian Science Fund (FWF) (grant no. KLI 1036-B) given to Miriam Michel.

Acknowledgments

This work is dedicated to the lovely Bernadette-Marie, born with HLHS. Open Access Funding by the Austrian Science Fund (FWF).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Rychik, J.; Atz, A.M.; Celermajer, D.S.; Deal, B.J.; Gatzoulis, M.A.; Gewillig, M.H.; Hsia, T.-Y.; Hsu, D.T.; Kovacs, A.H.; McCrindle, B.W.; et al. Evaluation and Management of the Child and Adult with Fontan Circulation: A Scientific Statement from the American Heart Association. Circulation 2019, 140, e234–e284. [Google Scholar] [CrossRef]
  2. Fontan, F.; Baudet, E. Surgical Repair of Tricuspid Atresia. Thorax 1971, 26, 240–248. [Google Scholar] [CrossRef] [Green Version]
  3. Kreutzer, G.; Galíndez, E.; Bono, H.; De Palma, C.; Laura, J.P. An Operation for the Correction of Tricuspid Atresia. J. Thorac. Cardiovasc. Surg. 1973, 66, 613–621. [Google Scholar] [CrossRef] [PubMed]
  4. Gewillig, M.; Brown, S.C. The Fontan Circulation after 45 Years: Update in Physiology. Heart 2016, 102, 1081–1086. [Google Scholar] [CrossRef] [PubMed]
  5. Alsoufi, B.; Gillespie, S.; Kim, D.; Shashidharan, S.; Kanter, K.; Maher, K.; Kogon, B. The Impact of Dominant Ventricle Morphology on Palliation Outcomes of Single Ventricle Anomalies. Ann. Thorac. Surg. 2016, 102, 593–601. [Google Scholar] [CrossRef] [Green Version]
  6. Kutty, S.; Jacobs, M.L.; Thompson, W.R.; Danford, D.A. Fontan Circulation of the Next Generation: Why It’s Necessary, What It Might Look Like. J. Am. Heart Assoc. 2020, 9, e013691. [Google Scholar] [CrossRef] [PubMed]
  7. Rychik, J. Path Taken in a Fontan Circulation: Room for Optimism in the Face of Uncertainty. Heart 2021, 107, 521–522. [Google Scholar] [CrossRef]
  8. Zhu, A.; Meza, J.M.; Prabhu, N.K.; McCrary, A.W.; Allareddy, V.; Turek, J.W.; Andersen, N.D. Survival After Intervention for Single-Ventricle Heart Disease Over 15 Years at a Single Institution. Ann. Thorac. Surg. 2022, 114, 2303–2312. [Google Scholar] [CrossRef]
  9. Lewis, M.; Rosenbaum, M. The Miracle Baby Grows Up: Hypoplastic Left Heart Syndrome in the Adult. Curr. Cardiol. Rep. 2017, 19, 74. [Google Scholar] [CrossRef]
  10. Julsrud, P.R.; Weigel, T.J.; Van Son, J.A.; Edwards, W.D.; Mair, D.D.; Driscoll, D.J.; Danielson, G.K.; Puga, F.J.; Offord, K.P. Influence of Ventricular Morphology on Outcome after the Fontan Procedure. Am. J. Cardiol. 2000, 86, 319–323. [Google Scholar] [CrossRef]
  11. McGuirk, S. The Impact of Ventricular Morphology on Midterm Outcome Following Completion Total Cavopulmonary Connection. Eur. J. Cardiothorac. Surg. 2003, 24, 37–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Anderson, P.A.W.; Sleeper, L.A.; Mahony, L.; Colan, S.D.; Atz, A.M.; Breitbart, R.E.; Gersony, W.M.; Gallagher, D.; Geva, T.; Margossian, R.; et al. Contemporary Outcomes after the Fontan Procedure. J. Am. Coll. Cardiol. 2008, 52, 85–98. [Google Scholar] [CrossRef] [Green Version]
  13. Backer, C.L. The Functionally Univentricular Heart. J. Am. Coll. Cardiol. 2012, 59, 1186–1187. [Google Scholar] [CrossRef] [PubMed]
  14. d’Udekem, Y.; Xu, M.Y.; Galati, J.C.; Lu, S.; Iyengar, A.J.; Konstantinov, I.E.; Wheaton, G.R.; Ramsay, J.M.; Grigg, L.E.; Millar, J.; et al. Predictors of Survival After Single-Ventricle Palliation. J. Am. Coll. Cardiol. 2012, 59, 1178–1185. [Google Scholar] [CrossRef] [Green Version]
  15. King, G.; Buratto, E.; Daley, M.; Iyengar, A.; Alphonso, N.; Grigg, L.; Cordina, R.; d’Udekem, Y.; Konstantinov, I.E. Impact of Aortic Atresia After Fontan Operation in Patients With Hypoplastic Left Heart Syndrome. Ann. Thorac. Surg. 2022, 116, 95–102. [Google Scholar] [CrossRef]
  16. Iyengar, A.J.; Winlaw, D.S.; Galati, J.C.; Wheaton, G.R.; Gentles, T.L.; Grigg, L.E.; Justo, R.N.; Radford, D.J.; Weintraub, R.G.; Bullock, A.; et al. The Extracardiac Conduit Fontan Procedure in Australia and New Zealand: Hypoplastic Left Heart Syndrome Predicts Worse Early and Late Outcomes. Eur. J. Cardiothorac. Surg. 2014, 46, 465–473. [Google Scholar] [CrossRef]
  17. Book, W.M.; Gerardin, J.; Saraf, A.; Marie Valente, A.; Rodriguez, F. Clinical Phenotypes of Fontan Failure: Implications for Management: Fontan Phenotypes. Congenit. Heart Dis. 2016, 11, 296–308. [Google Scholar] [CrossRef]
  18. Sable, C.; Foster, E.; Uzark, K.; Bjornsen, K.; Canobbio, M.M.; Connolly, H.M.; Graham, T.P.; Gurvitz, M.Z.; Kovacs, A.; Meadows, A.K.; et al. Best Practices in Managing Transition to Adulthood for Adolescents with Congenital Heart Disease: The Transition Process and Medical and Psychosocial Issues: A Scientific Statement from the American Heart Association. Circulation 2011, 123, 1454–1485. [Google Scholar] [CrossRef] [Green Version]
  19. Michel, M.; Zlamy, M.; Entenmann, A.; Pichler, K.; Scholl-Bürgi, S.; Karall, D.; Geiger, R.; Salvador, C.; Niederwanger, C.; Ohuchi, H. Impact of the Fontan Operation on Organ Systems. Cardiovasc. Hematol. Disord. Drug Targets 2019, 19, 205–214. [Google Scholar] [CrossRef]
  20. Harteveld, L.M.; Blom, N.A.; Terol Espinosa de Los Monteros, C.; Kuipers, I.M.; Rammeloo, L.A.J.; Hazekamp, M.G.; van Dijk, J.G.; ten Harkel, A.D.J. 3-Month Enalapril Treatment in Pediatric Fontan Patients with Moderate to Good Systolic Ventricular Function. Am. J. Cardiol. 2022, 163, 98–103. [Google Scholar] [CrossRef] [PubMed]
  21. Shaddy, R.E.; Boucek, M.M.; Hsu, D.T.; Boucek, R.J.; Canter, C.E.; Mahony, L.; Ross, R.D.; Pahl, E.; Blume, E.D.; Dodd, D.A.; et al. Carvedilol for Children and Adolescents With Heart Failure: A Randomized Controlled Trial. JAMA 2007, 298, 1171. [Google Scholar] [CrossRef] [PubMed]
  22. Schranz, D.; Voelkel, N.F. “Nihilism” of Chronic Heart Failure Therapy in Children and Why Effective Therapy Is Withheld. Eur. J. Pediatr. 2016, 175, 445–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Anderson, P.A.W.; Breitbart, R.E.; McCrindle, B.W.; Sleeper, L.A.; Atz, A.M.; Hsu, D.T.; Lu, M.; Margossian, R.; Williams, R.V. The Fontan Patient: Inconsistencies in Medication Therapy Across Seven Pediatric Heart Network Centers. Pediatr. Cardiol. 2010, 31, 1219–1228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ghanayem, N.S.; Berger, S.; Tweddell, J.S. Medical Management of the Failing Fontan. Pediatr. Cardiol. 2007, 28, 465–471. [Google Scholar] [CrossRef] [PubMed]
  25. Taegtmeyer, H. Metabolism—The Lost Child of Cardiology. J. Am. Coll. Cardiol. 2000, 36, 1386–1388. [Google Scholar] [CrossRef] [Green Version]
  26. Taegtmeyer, H. Cardiac Metabolism as a Target for the Treatment of Heart Failure. Circulation 2004, 110, 894–896. [Google Scholar] [CrossRef] [Green Version]
  27. Ashrafian, H.; Frenneaux, M.P.; Opie, L.H. Metabolic Mechanisms in Heart Failure. Circulation 2007, 116, 434–448. [Google Scholar] [CrossRef]
  28. Kimball, T.H.; Vondriska, T.M. Metabolism, Epigenetics, and Causal Inference in Heart Failure. Trends Endocrinol. Metab. 2020, 31, 181–191. [Google Scholar] [CrossRef]
  29. Selvaraj, S.; Kelly, D.P.; Margulies, K.B. Implications of Altered Ketone Metabolism and Therapeutic Ketosis in Heart Failure. Circulation 2020, 141, 1800–1812. [Google Scholar] [CrossRef]
  30. Taegtmeyer, H. Energy Metabolism of the Heart: From Basic Concepts to Clinical Applications Applications. Curr. Probl. Cardiol. 1994, 19, 61–86. [Google Scholar] [CrossRef]
  31. Christensen, K.H. Treatment with the Ketone Body 3-Hydroxybutyrate in Patients with Acute Heart Failure. 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT04442555 (accessed on 20 July 2022).
  32. Yurista, S.R.; Chong, C.-R.; Badimon, J.J.; Kelly, D.P.; de Boer, R.A.; Westenbrink, B.D. Therapeutic Potential of Ketone Bodies for Patients with Cardiovascular Disease. J. Am. Coll. Cardiol. 2021, 77, 1660–1669. [Google Scholar] [CrossRef] [PubMed]
  33. Takahara, S.; Soni, S.; Phaterpekar, K.; Kim, T.T.; Maayah, Z.H.; Levasseur, J.L.; Silver, H.L.; Freed, D.H.; Ferdaoussi, M.; Dyck, J.R.B. Chronic Exogenous Ketone Supplementation Blunts the Decline of Cardiac Function in the Failing Heart. ESC Heart Fail. 2021, 8, 5606–5612. [Google Scholar] [CrossRef] [PubMed]
  34. Takahara, S.; Soni, S.; Maayah, Z.H.; Ferdaoussi, M.; Dyck, J.R.B. Ketone Therapy for Heart Failure: Current Evidence for Clinical Use. Cardiovasc. Res. 2022, 118, 977–987. [Google Scholar] [CrossRef]
  35. Monzo, L.; Sedlacek, K.; Hromanikova, K.; Tomanova, L.; Borlaug, B.A.; Jabor, A.; Kautzner, J.; Melenovsky, V. Myocardial Ketone Body Utilization in Patients with Heart Failure: The Impact of Oral Ketone Ester. Metabolism 2021, 115, 154452. [Google Scholar] [CrossRef] [PubMed]
  36. Papazafiropoulou, A.; Georgopoulos, M.; Katsilambros, N. Ketone Bodies and the Heart. Arch. Med. Sci. Atheroscler. Dis. 2021, 6, 209–214. [Google Scholar] [CrossRef]
  37. Schulze, P.C.; Wu, J.M.F. Ketone Bodies for the Starving Heart. Nat. Metab. 2020, 2, 1183–1185. [Google Scholar] [CrossRef]
  38. Horton, J.L.; Davidson, M.T.; Kurishima, C.; Vega, R.B.; Powers, J.C.; Matsuura, T.R.; Petucci, C.; Lewandowski, E.D.; Crawford, P.A.; Muoio, D.M.; et al. The Failing Heart Utilizes 3-Hydroxybutyrate as a Metabolic Stress Defense. JCI Insight 2019, 4, e124079. [Google Scholar] [CrossRef] [Green Version]
  39. Uchihashi, M.; Hoshino, A.; Okawa, Y.; Ariyoshi, M.; Kaimoto, S.; Tateishi, S.; Ono, K.; Yamanaka, R.; Hato, D.; Fushimura, Y.; et al. Cardiac-Specific Bdh1 Overexpression Ameliorates Oxidative Stress and Cardiac Remodeling in Pressure Overload–Induced Heart Failure. Circ. Heart Fail. 2017, 10, e004417. [Google Scholar] [CrossRef]
  40. Schugar, R.C.; Moll, A.R.; André d’Avignon, D.; Weinheimer, C.J.; Kovacs, A.; Crawford, P.A. Cardiomyocyte-Specific Deficiency of Ketone Body Metabolism Promotes Accelerated Pathological Remodeling. Mol. Metab. 2014, 3, 754–769. [Google Scholar] [CrossRef]
  41. Kolb, H.; Kempf, K.; Röhling, M.; Lenzen-Schulte, M.; Schloot, N.C.; Martin, S. Ketone Bodies: From Enemy to Friend and Guardian Angel. BMC Med. 2021, 19, 313. [Google Scholar] [CrossRef]
  42. Michel, M.; Dubowy, K.-O.; Entenmann, A.; Karall, D.; Adam, M.G.; Zlamy, M.; Odri Komazec, I.; Geiger, R.; Niederwanger, C.; Salvador, C.; et al. Targeted Metabolomic Analysis of Serum Amino Acids in the Adult Fontan Patient with a Dominant Left Ventricle. Sci. Rep. 2020, 10, 8930. [Google Scholar] [CrossRef]
  43. Michel, M.; Dubowy, K.-O.; Zlamy, M.; Karall, D.; Adam, M.G.; Entenmann, A.; Keller, M.A.; Koch, J.; Odri Komazec, I.; Geiger, R.; et al. Targeted Metabolomic Analysis of Serum Phospholipid and Acylcarnitine in the Adult Fontan Patient with a Dominant Left Ventricle. Ther. Adv. Chronic Dis. 2020, 11, 204062232091603. [Google Scholar] [CrossRef] [PubMed]
  44. Opie, L.H. Heart Physiology: From Cell to Circulation, 4th ed.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2004. [Google Scholar]
  45. Barth, E. Ultrastructural Quantitation of Mitochondria and Myofilaments in Cardiac Muscle from 10 Different Animal Species Including Man. J. Mol. Cell. Cardiol. 1992, 24, 669–681. [Google Scholar] [CrossRef] [PubMed]
  46. Ingwall, J.S.; Weiss, R.G. Is the Failing Heart Energy Starved?: On Using Chemical Energy to Support Cardiac Function. Circ. Res. 2004, 95, 135–145. [Google Scholar] [CrossRef]
  47. Herrmann, G.; Decherd, G. The chemical nature of heart failure. Ann. Intern. Med. 1939, 12, 1233. [Google Scholar] [CrossRef]
  48. Neubauer, S. The Failing Heart—An Engine Out of Fuel. N. Engl. J. Med. 2007, 356, 1140–1151. [Google Scholar] [CrossRef] [Green Version]
  49. Sack, M.N.; Rader, T.A.; Park, S.; Bastin, J.; McCune, S.A.; Kelly, D.P. Fatty Acid Oxidation Enzyme Gene Expression Is Downregulated in the Failing Heart. Circulation 1996, 94, 2837–2842. [Google Scholar] [CrossRef]
  50. Katz, A.M. Energetics and the Failing Heart. Hosp. Pract. 1991, 26, 78–90. [Google Scholar] [CrossRef]
  51. Taegtmeyer, H. Failing Heart and Starving Brain. Circulation 2016, 134, 265–266. [Google Scholar] [CrossRef] [PubMed]
  52. Giussani, D.A.; Bennet, L.; Sferruzzi-Perri, A.N.; Vaughan, O.R.; Fowden, A.L. Hypoxia, fetal and neonatal physiology: 100 years on from Sir Joseph Barcroft. J. Physiol. 2016, 594, 1105–1111. [Google Scholar] [CrossRef] [Green Version]
  53. Girard, J.; Ferre, P.; Pegorier, J.P.; Duee, P.H. Adaptations of Glucose and Fatty Acid Metabolism during Perinatal Period and Suckling-Weaning Transition. Physiol. Rev. 1992, 72, 507–562. [Google Scholar] [CrossRef]
  54. Ascuitto, R.J.; Ross-Ascuitto, N.T. Substrate Metabolism in the Developing Heart. Semin. Perinatol. 1996, 20, 542–563. [Google Scholar] [CrossRef]
  55. Itoi, T.; Lopaschuk, G.D. The Contribution of Glycolysis, Glucose Oxidation, Lactate Oxidation, and Fatty Acid Oxidation to ATP Production in Isolated Biventricular Working Hearts from 2-Week-Old Rabbits. Pediatr. Res. 1993, 34, 735–741. [Google Scholar] [CrossRef] [Green Version]
  56. Dimasi, C.G.; Darby, J.R.T.; Morrison, J.L. A change of heart: Understanding the mechanisms regulating cardiac proliferation and metabolism before and after birth. J. Physiol. 2023, 601, 1319–1341. [Google Scholar] [CrossRef] [PubMed]
  57. Stanley, W.C.; Recchia, F.A.; Lopaschuk, G.D. Myocardial Substrate Metabolism in the Normal and Failing Heart. Physiol. Rev. 2005, 85, 1093–1129. [Google Scholar] [CrossRef] [Green Version]
  58. Lopaschuk, G.D.; Jaswal, J.S. Energy Metabolic Phenotype of the Cardiomyocyte During Development, Differentiation, and Postnatal Maturation. J. Cardiovasc. Pharmacol. 2010, 56, 130–140. [Google Scholar] [CrossRef] [PubMed]
  59. Ritterhoff, J.; Tian, R. Metabolism in Cardiomyopathy: Every Substrate Matters. Cardiovasc. Res. 2017, 113, 411–421. [Google Scholar] [CrossRef] [Green Version]
  60. Taegtmeyer, H.; Sen, S.; Vela, D. Return to the Fetal Gene Program: A Suggested Metabolic Link to Gene Expression in the Heart. Ann. N. Y. Acad. Sci. 2010, 1188, 191–198. [Google Scholar] [CrossRef] [Green Version]
  61. Lopaschuk, G.D.; Karwi, Q.G.; Tian, R.; Wende, A.R.; Abel, E.D. Cardiac Energy Metabolism in Heart Failure. Circ. Res. 2021, 128, 1487–1513. [Google Scholar] [CrossRef]
  62. Karwi, Q.G.; Uddin, G.M.; Ho, K.L.; Lopaschuk, G.D. Loss of Metabolic Flexibility in the Failing Heart. Front. Cardiovasc. Med. 2018, 5, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Shah, A.; Shannon, R.P. Insulin Resistance in Dilated Cardiomyopathy. Rev. Cardiovasc. Med. 2003, 4, S50–S57. [Google Scholar] [PubMed]
  64. Nikolaidis, L. The Development of Myocardial Insulin Resistance in Conscious Dogs with Advanced Dilated Cardiomyopathy. Cardiovasc. Res. 2004, 61, 297–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Taegtmeyer, H.; Golfman, L.; Sharma, S.; Razeghi, P.; Arsdall, M. Linking Gene Expression to Function: Metabolic Flexibility in the Normal and Diseased Heart. Ann. N. Y. Acad. Sci. 2004, 1015, 202–213. [Google Scholar] [CrossRef]
  66. Schulze, P.C.; Drosatos, K.; Goldberg, I.J. Lipid Use and Misuse by the Heart. Circ. Res. 2016, 118, 1736–1751. [Google Scholar] [CrossRef]
  67. Kolwicz, S.C.; Airhart, S.; Tian, R. Ketones Step to the Plate: A Game Changer for Metabolic Remodeling in Heart Failure? Circulation 2016, 133, 689–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Bedi, K.C.; Snyder, N.W.; Brandimarto, J.; Aziz, M.; Mesaros, C.; Worth, A.J.; Wang, L.L.; Javaheri, A.; Blair, I.A.; Margulies, K.B.; et al. Evidence for Intramyocardial Disruption of Lipid Metabolism and Increased Myocardial Ketone Utilization in Advanced Human Heart Failure. Circulation 2016, 133, 706–716. [Google Scholar] [CrossRef] [Green Version]
  69. Aubert, G.; Martin, O.J.; Horton, J.L.; Lai, L.; Vega, R.B.; Leone, T.C.; Koves, T.; Gardell, S.J.; Krüger, M.; Hoppel, C.L.; et al. The Failing Heart Relies on Ketone Bodies as a Fuel. Circulation 2016, 133, 698–705. [Google Scholar] [CrossRef] [Green Version]
  70. Huynh, K. Ketone Bodies as Fuel in Heart Failure. Nat. Rev. Cardiol. 2016, 13, 123. [Google Scholar] [CrossRef]
  71. Manolis, A.S.; Manolis, T.A.; Manolis, A.A. Ketone Bodies and Cardiovascular Disease: An Alternate Fuel Source to the Rescue. Int. J. Mol. Sci. 2023, 24, 3534. [Google Scholar] [CrossRef]
  72. Voros, G.; Ector, J.; Garweg, C.; Droogne, W.; Van Cleemput, J.; Peersman, N.; Vermeersch, P.; Janssens, S. Increased Cardiac Uptake of Ketone Bodies and Free Fatty Acids in Human Heart Failure and Hypertrophic Left Ventricular Remodeling. Circ. Heart Fail. 2018, 11, e004953. [Google Scholar] [CrossRef]
  73. Nakamura, M.; Sadoshima, J. Ketone Body Can Be a Fuel Substrate for Failing Heart. Cardiovasc. Res. 2019, 115, 1567–1569. [Google Scholar] [CrossRef]
  74. Liao, S.; Tang, Y.; Yue, X.; Gao, R.; Yao, W.; Zhou, Y.; Zhang, H. β-Hydroxybutyrate Mitigated Heart Failure with Preserved Ejection Fraction by Increasing Treg Cells via Nox2/GSK-3β. J. Inflamm. Res. 2021, 14, 4697–4706. [Google Scholar] [CrossRef]
  75. Deng, Y.; Xie, M.; Li, Q.; Xu, X.; Ou, W.; Zhang, Y.; Xiao, H.; Yu, H.; Zheng, Y.; Liang, Y.; et al. Targeting Mitochondria-Inflammation Circuit by β-Hydroxybutyrate Mitigates HFpEF. Circ. Res. 2021, 128, 232–245. [Google Scholar] [CrossRef] [PubMed]
  76. Tsutsui, H.; Kinugawa, S.; Matsushima, S. Oxidative Stress and Heart Failure. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H2181–H2190. [Google Scholar] [CrossRef] [Green Version]
  77. Beer, M.; Seyfarth, T.; Sandstede, J.; Landschütz, W.; Lipke, C.; Köstler, H.; von Kienlin, M.; Harre, K.; Hahn, D.; Neubauer, S. Absolute Concentrations of High-Energy Phosphate Metabolites in Normal, Hypertrophied, and Failing Human Myocardium Measured Noninvasively with 31P-SLOOP Magnetic Resonance Spectroscopy. J. Am. Coll. Cardiol. 2002, 40, 1267–1274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Weiss, R.G.; Gerstenblith, G.; Bottomley, P.A. ATP Flux through Creatine Kinase in the Normal, Stressed, and Failing Human Heart. Proc. Natl. Acad. Sci. USA 2005, 102, 808–813. [Google Scholar] [CrossRef]
  79. Smith, C.S.; Bottomley, P.A.; Schulman, S.P.; Gerstenblith, G.; Weiss, R.G. Altered Creatine Kinase Adenosine Triphosphate Kinetics in Failing Hypertrophied Human Myocardium. Circulation 2006, 114, 1151–1158. [Google Scholar] [CrossRef] [Green Version]
  80. Nielsen, R.; Møller, N.; Gormsen, L.C.; Tolbod, L.P.; Hansson, N.H.; Sorensen, J.; Harms, H.J.; Frøkiær, J.; Eiskjaer, H.; Jespersen, N.R.; et al. Cardiovascular Effects of Treatment with the Ketone Body 3-Hydroxybutyrate in Chronic Heart Failure Patients. Circulation 2019, 139, 2129–2141. [Google Scholar] [CrossRef]
  81. Brittain, E.L.; Talati, M.; Fessel, J.P.; Zhu, H.; Penner, N.; Calcutt, M.W.; West, J.D.; Funke, M.; Lewis, G.D.; Gerszten, R.E.; et al. Fatty Acid Metabolic Defects and Right Ventricular Lipotoxicity in Human Pulmonary Arterial Hypertension. Circulation 2016, 133, 1936–1944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Pugh, M.E.; Robbins, I.M.; Rice, T.W.; West, J.; Newman, J.H.; Hemnes, A.R. Unrecognized Glucose Intolerance Is Common in Pulmonary Arterial Hypertension. J. Heart Lung Transplant. 2011, 30, 904–911. [Google Scholar] [CrossRef]
  83. West, J.; Niswender, K.D.; Johnson, J.A.; Pugh, M.E.; Gleaves, L.; Fessel, J.P.; Hemnes, A.R. A Potential Role for Insulin Resistance in Experimental Pulmonary Hypertension. Eur. Respir. J. 2013, 41, 861–871. [Google Scholar] [CrossRef] [Green Version]
  84. Zamanian, R.T.; Hansmann, G.; Snook, S.; Lilienfeld, D.; Rappaport, K.M.; Reaven, G.M.; Rabinovitch, M.; Doyle, R.L. Insulin Resistance in Pulmonary Arterial Hypertension. Eur. Respir. J. 2008, 33, 318–324. [Google Scholar] [CrossRef]
  85. Zare, E.; Kafshbani, P.; Chenaghlou, M.; Noori, M.; Ghaemmaghami, Z.; Amin, A.; Taghavi, S.; Naderi, N. Prognostic Significance of Insulin Resistance in Pulmonary Hypertension. ESC Heart Fail. 2022, 9, 318–326. [Google Scholar] [CrossRef]
  86. University of Aarhus. Ketones for Pulmonary Hypertension—Effects on Hemodynamics (KEPAH). 2021. Available online: https://clinicaltrials.gov/ct2/show/NCT04615754 (accessed on 22 March 2023).
  87. Nielsen, R.; Christensen, K.H.; Gopalasingam, N.; Berg-Hansen, K.; Seefeldt, J.; Homilius, C.; Boedtkjer, E.; Andersen, M.J.; Wiggers, H.; Møller, N.; et al. Hemodynamic Effects of Ketone Bodies in Patients with Pulmonary Hypertension. J. Am. Heart Assoc. 2023, 12, e028232. [Google Scholar] [CrossRef]
  88. Blake, M.; Puchalska, P.; Kazmirczak, F.; Thenappan, T.; Crawford, P.A.; Prins, K.W. Ketone Bodies in Right Ventricular Failure: A Unique Therapeutic Opportunity. bioRxiv 2023. [Google Scholar] [CrossRef]
  89. McCullough, D.J.; Kue, N.; Mancini, T.; Vang, A.; Clements, R.T.; Choudhary, G. Endurance Exercise Training in Pulmonary Hypertension Increases Skeletal Muscle Electron Transport Chain Supercomplex Assembly. Pulm. Circ. 2020, 10, 1–11. [Google Scholar] [CrossRef] [Green Version]
  90. Cawthon, D.; Beers, K.; Bottje, W.G. Electron Transport Chain Defect and Inefficient Respiration May Underlie Pulmonary Hypertension Syndrome (Ascites)-Associated Mitochondrial Dysfunction in Broilers. Poult. Sci. 2001, 80, 474–484. [Google Scholar] [CrossRef]
  91. Xu, W.; Comhair, S.A.A.; Chen, R.; Hu, B.; Hou, Y.; Zhou, Y.; Mavrakis, L.A.; Janocha, A.J.; Li, L.; Zhang, D.; et al. Integrative Proteomics and Phosphoproteomics in Pulmonary Arterial Hypertension. Sci. Rep. 2019, 9, 18623. [Google Scholar] [CrossRef] [Green Version]
  92. Huertas, A.; Tu, L.; Humbert, M.; Guignabert, C. Chronic Inflammation within the Vascular Wall in Pulmonary Arterial Hypertension: More than a Spectator. Cardiovasc. Res. 2020, 116, 885–893. [Google Scholar] [CrossRef]
  93. Fowler, E.D.; Hauton, D.; Boyle, J.; Egginton, S.; Steele, D.S.; White, E. Energy Metabolism in the Failing Right Ventricle: Limitations of Oxygen Delivery and the Creatine Kinase System. Int. J. Mol. Sci. 2019, 20, 1805. [Google Scholar] [CrossRef] [Green Version]
  94. Garcia, A.M.; Toni, L.S.; Miyano, C.A.; Sparagna, G.C.; Jonscher, R.; Phillips, E.K.; Karimpour-Fard, A.; Chapman, H.L.; Baybayon-Grandgeorge, A.N.; Pietra, A.E.; et al. Cardiac Transcriptome Remodeling and Impaired Bioenergetics in Single-Ventricle Congenital Heart Disease. JACC Basic Transl. Sci. 2023, 8, 258–279. [Google Scholar] [CrossRef]
  95. Pires da Silva, J.; Pietra, A.E.; Baybayon-Grandgeorge, A.N.; Garcia, A.M. Serum Metabolic Profiling Identifies Key Differences between Patients with Single-Ventricle Heart Disease and Healthy Controls. Int. J. Transl. Med. 2022, 2, 78–96. [Google Scholar] [CrossRef]
  96. Xu, X.; Lin, J.-H.I.; Bais, A.S.; Reynolds, M.J.; Tan, T.; Gabriel, G.C.; Kondos, Z.; Liu, X.; Shiva, S.S.; Lo, C.W. Mitochondrial Respiration Defects in Single-Ventricle Congenital Heart Disease. Front. Cardiovasc. Med. 2021, 8, 734388. [Google Scholar] [CrossRef]
  97. Ide, T.; Tsutsui, H.; Hayashidani, S.; Kang, D.; Suematsu, N.; Nakamura, K.; Utsumi, H.; Hamasaki, N.; Takeshita, A. Mitochondrial DNA Damage and Dysfunction Associated with Oxidative Stress in Failing Hearts After Myocardial Infarction. Circ. Res. 2001, 88, 529–535. [Google Scholar] [CrossRef]
  98. Tsutsui, H. Mitochondrial Oxidative Stress and Heart Failure. Intern. Med. 2006, 45, 809–813. [Google Scholar] [CrossRef] [Green Version]
  99. Randle, P.J.; Garland, P.B.; Hales, C.N.; Newsholme, E.A. The glucose fatty-acid cycle its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1963, 281, 785–789. [Google Scholar] [CrossRef]
  100. Koeslag, J.H.; Noakes, T.D.; Sloan, A.W. Post-Exercise Ketosis. J. Physiol. 1980, 301, 79–90. [Google Scholar] [CrossRef]
  101. Robinson, A.M.; Williamson, D.H. Physiological Roles of Ketone Bodies as Substrates and Signals in Mammalian Tissues. Physiol. Rev. 1980, 60, 143–187. [Google Scholar] [CrossRef]
  102. Lopaschuk, G.D.; Karwi, Q.G.; Ho, K.L.; Pherwani, S.; Ketema, E.B. Ketone Metabolism in the Failing Heart. Biochim. Biophys. Acta BBA Mol. Cell Biol. Lipids 2020, 1865, 158813. [Google Scholar] [CrossRef]
  103. Hue, L.; Taegtmeyer, H. The Randle Cycle Revisited: A New Head for an Old Hat. Am. J. Physiol. Endocrinol. Metab. 2009, 297, E578–E591. [Google Scholar] [CrossRef] [Green Version]
  104. Barger, P.M.; Kelly, D.P. PPAR Signaling in the Control of Cardiac Energy Metabolism. Trends Cardiovasc. Med. 2000, 10, 238–245. [Google Scholar] [CrossRef]
  105. Taegtmeyer, H.; Wilson, C.R.; Razeghi, P.; Sharma, S. Metabolic Energetics and Genetics in the Heart. Ann. N. Y. Acad. Sci. 2005, 1047, 208–218. [Google Scholar] [CrossRef]
  106. Kelly, D.P. PPARs of the Heart: Three Is a Crowd. Circ. Res. 2003, 92, 482–484. [Google Scholar] [CrossRef]
  107. Lehman, J.J.; Kelly, D.P. Gene Regulatory Mechanisms Governing Energy Metabolism during Cardiac Hypertrophic Growth. Heart Fail. Rev. 2002, 7, 175–185. [Google Scholar] [CrossRef]
  108. Razeghi, P.; Young, M.E.; Abbasi, S.; Taegtmeyer, H. Hypoxia in Vivo Decreases Peroxisome Proliferator-Activated Receptor α-Regulated Gene Expression in Rat Heart. Biochem. Biophys. Res. Commun. 2001, 287, 5–10. [Google Scholar] [CrossRef]
  109. Yang, M.; Zhang, Y.; Ren, J. Acetylation in Cardiovascular Diseases: Molecular Mechanisms and Clinical Implications. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2020, 1866, 165836. [Google Scholar] [CrossRef]
  110. Shogren-Knaak, M.; Ishii, H.; Sun, J.-M.; Pazin, M.J.; Davie, J.R.; Peterson, C.L. Histone H4-K16 Acetylation Controls Chromatin Structure and Protein Interactions. Science 2006, 311, 844–847. [Google Scholar] [CrossRef] [Green Version]
  111. Peleg, S.; Feller, C.; Ladurner, A.G.; Imhof, A. The Metabolic Impact on Histone Acetylation and Transcription in Ageing. Trends Biochem. Sci. 2016, 41, 700–711. [Google Scholar] [CrossRef] [Green Version]
  112. Verdin, E.; Dequiedt, F.; Kasler, H.G. Class II Histone Deacetylases: Versatile Regulators. Trends Genet. 2003, 19, 286–293. [Google Scholar] [CrossRef] [Green Version]
  113. McKinsey, T.A.; Olson, E.N. Cardiac Histone Acetylation—Therapeutic Opportunities Abound. Trends Genet. 2004, 20, 206–213. [Google Scholar] [CrossRef]
  114. Backs, J.; Olson, E.N. Control of Cardiac Growth by Histone Acetylation/Deacetylation. Circ. Res. 2006, 98, 15–24. [Google Scholar] [CrossRef]
  115. Allard, M.F.; Schonekess, B.O.; Henning, S.L.; English, D.R.; Lopaschuk, G.D. Contribution of Oxidative Metabolism and Glycolysis to ATP Production in Hypertrophied Hearts. Am. J. Physiol. Heart Circ. Physiol. 1994, 267, H742–H750. [Google Scholar] [CrossRef]
  116. Diakos, N.A.; Navankasattusas, S.; Abel, E.D.; Rutter, J.; McCreath, L.; Ferrin, P.; McKellar, S.H.; Miller, D.V.; Park, S.Y.; Richardson, R.S.; et al. Evidence of Glycolysis Up-Regulation and Pyruvate Mitochondrial Oxidation Mismatch During Mechanical Unloading of the Failing Human Heart. JACC Basic Transl. Sci. 2016, 1, 432–444. [Google Scholar] [CrossRef] [Green Version]
  117. Bottomley, P.A.; Panjrath, G.S.; Lai, S.; Hirsch, G.A.; Wu, K.; Najjar, S.S.; Steinberg, A.; Gerstenblith, G.; Weiss, R.G. Metabolic Rates of ATP Transfer Through Creatine Kinase (CK Flux) Predict Clinical Heart Failure Events and Death. Sci. Transl. Med. 2013, 5, 215re3. [Google Scholar] [CrossRef] [Green Version]
  118. Mey, J.T.; Hari, A.; Axelrod, C.L.; Fealy, C.E.; Erickson, M.L.; Kirwan, J.P.; Dweik, R.A.; Heresi, G.A. Lipids and Ketones Dominate Metabolism at the Expense of Glucose Control in Pulmonary Arterial Hypertension: A Hyperglycaemic Clamp and Metabolomics Study. Eur. Respir. J. 2020, 55, 1901700. [Google Scholar] [CrossRef]
  119. Zhou, B.; Tian, R. Mitochondrial Dysfunction in Pathophysiology of Heart Failure. J. Clin. Investig. 2018, 128, 3716–3726. [Google Scholar] [CrossRef] [Green Version]
  120. Takimoto, E.; Kass, D.A. Role of Oxidative Stress in Cardiac Hypertrophy and Remodeling. Hypertension 2007, 49, 241–248. [Google Scholar] [CrossRef]
  121. van der Pol, A.; van Gilst, W.H.; Voors, A.A.; van der Meer, P. Treating Oxidative Stress in Heart Failure: Past, Present and Future. Eur. J. Heart Fail. 2019, 21, 425–435. [Google Scholar] [CrossRef] [Green Version]
  122. Nascimben, L.; Ingwall, J.S.; Pauletto, P.; Friedrich, J.; Gwathmey, J.K.; Saks, V.; Pessina, A.C.; Allen, P.D. Creatine Kinase System in Failing and Nonfailing Human Myocardium. Circulation 1996, 94, 1894–1901. [Google Scholar] [CrossRef]
  123. Keceli, G.; Gupta, A.; Sourdon, J.; Gabr, R.; Schär, M.; Dey, S.; Tocchetti, C.G.; Stuber, A.; Agrimi, J.; Zhang, Y.; et al. Mitochondrial Creatine Kinase Attenuates Pathologic Remodeling in Heart Failure. Circ. Res. 2022, 130, 741–759. [Google Scholar] [CrossRef]
  124. Olson, E.N.; Backs, J.; McKinsey, T.A. Control of Cardiac Hypertrophy and Heart Failure by Histone Acetylation/Deacetylation. In Novartis Foundation Symposia; Bock, G., Goode, J., Eds.; John Wiley & Sons, Ltd.: Chichester, UK, 2008; pp. 3–19. [Google Scholar] [CrossRef]
  125. Fukushima, A.; Zhang, L.; Huqi, A.; Lam, V.H.; Rawat, S.; Altamimi, T.; Wagg, C.S.; Dhaliwal, K.K.; Hornberger, L.K.; Kantor, P.F.; et al. Acetylation Contributes to Hypertrophy-Caused Maturational Delay of Cardiac Energy Metabolism. JCI Insight 2018, 3, e99239. [Google Scholar] [CrossRef] [Green Version]
  126. Castillo, E.C.; Morales, J.A.; Chapoy-Villanueva, H.; Silva-Platas, C.; Treviño-Saldaña, N.; Guerrero-Beltrán, C.E.; Bernal-Ramírez, J.; Torres-Quintanilla, A.; García, N.; Youker, K.; et al. Mitochondrial Hyperacetylation in the Failing Hearts of Obese Patients Mediated Partly by a Reduction in SIRT3: The Involvement of the Mitochondrial Permeability Transition Pore. Cell Physiol. Biochem. 2019, 53, 465–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Ooi, J.Y.Y.; Tuano, N.K.; Rafehi, H.; Gao, X.-M.; Ziemann, M.; Du, X.-J.; El-Osta, A. HDAC Inhibition Attenuates Cardiac Hypertrophy by Acetylation and Deacetylation of Target Genes. Epigenetics 2015, 10, 418–430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Kang, S.-H.; Seok, Y.M.; Song, M.; Lee, H.-A.; Kurz, T.; Kim, I. Histone Deacetylase Inhibition Attenuates Cardiac Hypertrophy and Fibrosis through Acetylation of Mineralocorticoid Receptor in Spontaneously Hypertensive Rats. Mol. Pharmacol. 2015, 87, 782–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Scholz, B.; Schulte, J.S.; Hamer, S.; Himmler, K.; Pluteanu, F.; Seidl, M.D.; Stein, J.; Wardelmann, E.; Hammer, E.; Völker, U.; et al. HDAC (Histone Deacetylase) Inhibitor Valproic Acid Attenuates Atrial Remodeling and Delays the Onset of Atrial Fibrillation in Mice. Circ. Arrhythm. Electrophysiol. 2019, 12, e007071. [Google Scholar] [CrossRef]
  130. Colussi, C.; Berni, R.; Rosati, J.; Straino, S.; Vitale, S.; Spallotta, F.; Baruffi, S.; Bocchi, L.; Delucchi, F.; Rossi, S.; et al. The Histone Deacetylase Inhibitor Suberoylanilide Hydroxamic Acid Reduces Cardiac Arrhythmias in Dystrophic Mice. Cardiovasc. Res. 2010, 87, 73–82. [Google Scholar] [CrossRef] [Green Version]
  131. Müller, J.; Bertsch, T.; Volke, J.; Schmid, A.; Klingbeil, R.; Metodiev, Y.; Karaca, B.; Kim, S.-H.; Lindner, S.; Schupp, T.; et al. Narrative Review of Metabolomics in Cardiovascular Disease. J. Thorac. Dis. 2021, 13, 2532–2550. [Google Scholar] [CrossRef]
  132. Bassareo, P.P.; McMahon, C.J. Metabolomics: A New Tool in Our Understanding of Congenital Heart Disease. Children 2022, 9, 1803. [Google Scholar] [CrossRef]
  133. Michel, M.; Laser, K.T.; Dubowy, K.-O.; Scholl-Bürgi, S.; Michel, E. Metabolomics and Random Forests in Patients with Complex Congenital Heart Disease. Front. Cardiovasc. Med. 2022, 9, 994068. [Google Scholar] [CrossRef]
  134. Murashige, D.; Jang, C.; Neinast, M.; Edwards, J.J.; Cowan, A.; Hyman, M.C.; Rabinowitz, J.D.; Frankel, D.S.; Arany, Z. Comprehensive Quantification of Fuel Use by the Failing and Nonfailing Human Heart. Science 2020, 370, 364–368. [Google Scholar] [CrossRef]
  135. Smith, E.; Fernandez, C.; Melander, O.; Ottosson, F. Altered Acylcarnitine Metabolism Is Associated With an Increased Risk of Atrial Fibrillation. J. Am. Heart Assoc. 2020, 9, e016737. [Google Scholar] [CrossRef]
  136. Ruiz, M.; Labarthe, F.; Fortier, A.; Bouchard, B.; Thompson Legault, J.; Bolduc, V.; Rigal, O.; Chen, J.; Ducharme, A.; Crawford, P.A.; et al. Circulating Acylcarnitine Profile in Human Heart Failure: A Surrogate of Fatty Acid Metabolic Dysregulation in Mitochondria and Beyond. Am. J. Physiol. Heart Circ. Physiol. 2017, 313, H768–H781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Penny, D.J.; Redington, A.N. Function of the Left and Right Ventricles and the Interactions Between Them. Pediatr. Crit. Care Med. 2016, 17, S112–S118. [Google Scholar] [CrossRef] [PubMed]
  138. Sanz, J.; Sánchez-Quintana, D.; Bossone, E.; Bogaard, H.J.; Naeije, R. Anatomy, Function, and Dysfunction of the Right Ventricle. J. Am. Coll. Cardiol. 2019, 73, 1463–1482. [Google Scholar] [CrossRef] [PubMed]
  139. Kondo, R.P.; Dederko, D.A.; Teutsch, C.; Chrast, J.; Catalucci, D.; Chien, K.R.; Giles, W.R. Comparison of Contraction and Calcium Handling between Right and Left Ventricular Myocytes from Adult Mouse Heart: A Role for Repolarization Waveform: Interventricular Heterogeneity of Cardiac Myocyte Contractions. J. Physiol. 2006, 571, 131–146. [Google Scholar] [CrossRef] [Green Version]
  140. Sedmera, D. Form Follows Function: Developmental and Physiological View on Ventricular Myocardial Architecture. Eur. J. Cardiothorac. Surg. 2005, 28, 526–528. [Google Scholar] [CrossRef] [Green Version]
  141. Garcia, A.M.; Beatty, J.-T.; Nakano, S.J. Heart Failure in Single Right Ventricle Congenital Heart Disease: Physiological and Molecular Considerations. Am. J. Physiol. Heart Circ. Physiol. 2020, 318, H947–H965. [Google Scholar] [CrossRef] [PubMed]
  142. Friehs, I.; Cowan, D.B.; Choi, Y.-H.; Black, K.M.; Barnett, R.; Bhasin, M.K.; Daly, C.; Dillon, S.J.; Libermann, T.A.; McGowan, F.X.; et al. Pressure-Overload Hypertrophy of the Developing Heart Reveals Activation of Divergent Gene and Protein Pathways in the Left and Right Ventricular Myocardium. Am. J. Physiol. Heart Circ. Physiol. 2013, 304, H697–H708. [Google Scholar] [CrossRef] [Green Version]
  143. Schreckenberg, R.; Rebelo, M.; Deten, A.; Weber, M.; Rohrbach, S.; Pipicz, M.; Csonka, C.; Ferdinandy, P.; Schulz, R.; Schlüter, K.-D. Specific Mechanisms Underlying Right Heart Failure: The Missing Upregulation of Superoxide Dismutase-2 and Its Decisive Role in Antioxidative Defense. Antioxid. Redox Signal. 2015, 23, 1220–1232. [Google Scholar] [CrossRef]
  144. Nagendran, J.; Gurtu, V.; Fu, D.Z.; Dyck, J.R.B.; Haromy, A.; Ross, D.B.; Rebeyka, I.M.; Michelakis, E.D. A Dynamic and Chamber-Specific Mitochondrial Remodeling in Right Ventricular Hypertrophy Can Be Therapeutically Targeted. J. Thorac. Cardiovasc. Surg. 2008, 136, 168–178.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Bokhari, S.; Raina, A.; Berman Rosenweig, E.; Schulze, P.C.; Bokhari, J.; Einstein, A.J.; Barst, R.J.; Johnson, L.L. PET Imaging May Provide a Novel Biomarker and Understanding of Right Ventricular Dysfunction in Patients With Idiopathic Pulmonary Arterial Hypertension. Circ. Cardiovasc. Imaging 2011, 4, 641–647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Gomez-Arroyo, J.; Mizuno, S.; Szczepanek, K.; Van Tassell, B.; Natarajan, R.; dos Remedios, C.G.; Drake, J.I.; Farkas, L.; Kraskauskas, D.; Wijesinghe, D.S.; et al. Metabolic Gene Remodeling and Mitochondrial Dysfunction in Failing Right Ventricular Hypertrophy Secondary to Pulmonary Arterial Hypertension. Circ. Heart Fail. 2013, 6, 136–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Piao, L.; Marsboom, G.; Archer, S.L. Mitochondrial Metabolic Adaptation in Right Ventricular Hypertrophy and Failure. J. Mol. Med. 2010, 88, 1011–1020. [Google Scholar] [CrossRef] [Green Version]
  148. Fessel, J.P.; Hamid, R.; Wittmann, B.M.; Robinson, L.J.; Blackwell, T.; Tada, Y.; Tanabe, N.; Tatsumi, K.; Hemnes, A.R.; West, J.D. Metabolomic Analysis of Bone Morphogenetic Protein Receptor Type 2 Mutations in Human Pulmonary Endothelium Reveals Widespread Metabolic Reprogramming. Pulm. Circ. 2012, 2, 201–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Schooneman, M.G.; Vaz, F.M.; Houten, S.M.; Soeters, M.R. Acylcarnitines. Diabetes 2013, 62, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Kalim, S.; Clish, C.B.; Wenger, J.; Elmariah, S.; Yeh, R.W.; Deferio, J.J.; Pierce, K.; Deik, A.; Gerszten, R.E.; Thadhani, R.; et al. A Plasma Long-Chain Acylcarnitine Predicts Cardiovascular Mortality in Incident Dialysis Patients. J. Am. Heart Assoc. 2013, 2, e000542. [Google Scholar] [CrossRef] [Green Version]
  151. Aitken-Buck, H.M.; Krause, J.; Zeller, T.; Jones, P.P.; Lamberts, R.R. Long-Chain Acylcarnitines and Cardiac Excitation-Contraction Coupling: Links to Arrhythmias. Front. Physiol. 2020, 11, 577856. [Google Scholar] [CrossRef]
  152. Brunner, N.W.; Skhiri, M.; Fortenko, O.; Hsi, A.; Haddad, F.; Khazeni, N.; Zamanian, R.T. Impact of Insulin Resistance on Ventricular Function in Pulmonary Arterial Hypertension. J. Heart Lung Transplant. 2014, 33, 721–726. [Google Scholar] [CrossRef]
  153. Assad, T.R.; Hemnes, A.R. Metabolic Dysfunction in Pulmonary Arterial Hypertension. Curr. Hypertens. Rep. 2015, 17, 20. [Google Scholar] [CrossRef] [Green Version]
  154. Graham, B.B.; Kumar, R.; Mickael, C.; Sanders, L.; Gebreab, L.; Huber, K.M.; Perez, M.; Smith-Jones, P.; Serkova, N.J.; Tuder, R.M. Severe Pulmonary Hypertension Is Associated with Altered Right Ventricle Metabolic Substrate Uptake. Am. J. Physiol. Lung Cell. Mol. Physiol. 2015, 309, L435–L440. [Google Scholar] [CrossRef] [Green Version]
  155. Ohuchi, H.; Negishi, J.; Hayama, Y.; Miike, H.; Suzuki, D.; Nakajima, K.; Konagai, N.; Iwasa, T.; Sakaguchi, H.; Kurosaki, K.; et al. Abnormal Glucose Metabolism in Patients with Fontan Circulation: Unique Characteristics and Associations with Fontan Pathophysiology. Am. Heart J. 2019, 216, 125–135. [Google Scholar] [CrossRef]
  156. Sharma, S.; Sud, N.; Wiseman, D.A.; Carter, A.L.; Kumar, S.; Hou, Y.; Rau, T.; Wilham, J.; Harmon, C.; Oishi, P.; et al. Altered Carnitine Homeostasis Is Associated with Decreased Mitochondrial Function and Altered Nitric Oxide Signaling in Lambs with Pulmonary Hypertension. Am. J. Physiol. Lung Cell. Mol. Physiol. 2008, 294, L46–L56. [Google Scholar] [CrossRef]
  157. Fowler, E.D.; Benoist, D.; Drinkhill, M.J.; Stones, R.; Helmes, M.; Wüst, R.C.I.; Stienen, G.J.M.; Steele, D.S.; White, E. Decreased Creatine Kinase Is Linked to Diastolic Dysfunction in Rats with Right Heart Failure Induced by Pulmonary Artery Hypertension. J. Mol. Cell. Cardiol. 2015, 86, 1–8. [Google Scholar] [CrossRef] [Green Version]
  158. Bernal-Ramirez, J.; Díaz-Vesga, M.C.; Talamilla, M.; Méndez, A.; Quiroga, C.; Garza-Cervantes, J.A.; Lázaro-Alfaro, A.; Jerjes-Sanchez, C.; Henríquez, M.; García-Rivas, G.; et al. Exploring Functional Differences between the Right and Left Ventricles to Better Understand Right Ventricular Dysfunction. Oxidative Med. Cell. Longev. 2021, 2021, 9993060. [Google Scholar] [CrossRef]
  159. Bogaard, H.J.; Mizuno, S.; Hussaini, A.A.A.; Toldo, S.; Abbate, A.; Kraskauskas, D.; Kasper, M.; Natarajan, R.; Voelkel, N.F. Suppression of Histone Deacetylases Worsens Right Ventricular Dysfunction after Pulmonary Artery Banding in Rats. Am. J. Respir. Crit. Care Med. 2011, 183, 1402–1410. [Google Scholar] [CrossRef]
  160. Chelladurai, P.; Boucherat, O.; Stenmark, K.; Kracht, M.; Seeger, W.; Bauer, U.; Bonnet, S.; Pullamsetti, S.S. Targeting Histone Acetylation in Pulmonary Hypertension and Right Ventricular Hypertrophy. Br. J. Pharmacol. 2021, 178, 54–71. [Google Scholar] [CrossRef] [Green Version]
  161. Chen, C.; Luo, F.; Wu, P.; Huang, Y.; Das, A.; Chen, S.; Chen, J.; Hu, X.; Li, F.; Fang, Z.; et al. Metabolomics Reveals Metabolite Changes of Patients with Pulmonary Arterial Hypertension in China. J. Cell. Mol. Med. 2020, 24, 2484–2496. [Google Scholar] [CrossRef]
  162. Rawat, S.; Fukushima, A.; Zhang, L.; Hugi, A.; Lam, V.; Altamimi, T.; Wagg, C.; Petinelli, R.; Dhaliwal, K.; Hornberger, L.; et al. Control of cardiac fatty acid metabolism in infants with hypoplastic left heart syndrome. J. Mol. Cell. Cardiol. 2018, 124, 91–92. [Google Scholar] [CrossRef]
  163. Motoki, N.; Motoki, H.; Utsumi, M.; Yamazaki, S.; Obinata, H.; Takei, K.; Yasukochi, S. Identification of metabolomic profile related to adult Fontan pathophysiology. Int. J. Cardiol. Heart Vasc. 2021, 37, 100921. [Google Scholar] [CrossRef]
  164. Li, M.; Zhou, S.; Chen, C.; Ma, L.; Luo, D.; Tian, X.; Dong, X.; Zhou, Y.; Yang, Y.; Cui, Y. Therapeutic Potential of Pyruvate Therapy for Patients with Mitochondrial Diseases: A Systematic Review. Ther. Adv. Endocrinol. Metab. 2020, 11, 204201882093824. [Google Scholar] [CrossRef]
  165. Des Rosiers, C.; Labarthe, F.; Lloyd, S.G.; Chatham, J.C. Cardiac Anaplerosis in Health and Disease: Food for Thought. Cardiovasc. Res. 2011, 90, 210–219. [Google Scholar] [CrossRef] [Green Version]
  166. McMurray, J.J.V.; Solomon, S.D.; Inzucchi, S.E.; Køber, L.; Kosiborod, M.N.; Martinez, F.A.; Ponikowski, P.; Sabatine, M.S.; Anand, I.S.; Bělohlávek, J.; et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N. Engl. J. Med. 2019, 381, 1995–2008. [Google Scholar] [CrossRef] [Green Version]
  167. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Pocock, S.J.; Carson, P.; Januzzi, J.; Verma, S.; Tsutsui, H.; Brueckmann, M.; et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N. Engl. J. Med. 2020, 383, 1413–1424. [Google Scholar] [CrossRef]
  168. Lopaschuk, G.D.; Verma, S. Mechanisms of Cardiovascular Benefits of Sodium Glucose Co-Transporter 2 (SGLT2) Inhibitors. JACC Basic Transl. Sci. 2020, 5, 632–644. [Google Scholar] [CrossRef]
  169. Maejima, Y. SGLT2 Inhibitors Play a Salutary Role in Heart Failure via Modulation of the Mitochondrial Function. Front. Cardiovasc. Med. 2020, 6, 186. [Google Scholar] [CrossRef]
  170. Pietschner, R.; Kolwelter, J.; Bosch, A.; Striepe, K.; Jung, S.; Kannenkeril, D.; Ott, C.; Schiffer, M.; Achenbach, S.; Schmieder, R.E. Effect of Empagliflozin on Ketone Bodies in Patients with Stable Chronic Heart Failure. Cardiovasc. Diabetol. 2021, 20, 219. [Google Scholar] [CrossRef]
  171. Takada, S.; Sabe, H.; Kinugawa, S. Treatments for Skeletal Muscle Abnormalities in Heart Failure: Sodium-Glucose Transporter 2 and Ketone Bodies. Am. J. Physiol. Heart Circ. Physiol. 2022, 322, H117–H128. [Google Scholar] [CrossRef]
  172. Muneuchi, J.; Sugitani, Y.; Kobayashi, M.; Ezaki, H.; Yamada, H.; Watanabe, M. Feasibility and Safety of Sodium Glucose Cotransporter-2 Inhibitors in Adults with Heart Failure after the Fontan Procedure. Case Rep. Cardiol. 2022, 2022, 5243594. [Google Scholar] [CrossRef]
  173. Ghelani, S.J.; Opotowsky, A.R.; Harrild, D.M.; Powell, A.J.; Azcue, N.; Ahmad, S.; Clair, N.S.; Bradwin, G.; Rathod, R.H. Characterization of Circulating and Urinary Biomarkers in the Fontan Circulation and Their Correlation with Cardiac Imaging. Am. J. Cardiol. 2022, 162, 177–183. [Google Scholar] [CrossRef]
  174. van den Bosch, E.; Bossers, S.S.M.; Kamphuis, V.P.; Boersma, E.; Roos-Hesselink, J.W.; Breur, J.M.P.J.; Ten Harkel, A.D.J.; Kapusta, L.; Bartelds, B.; Roest, A.A.W.; et al. Associations Between Blood Biomarkers, Cardiac Function, and Adverse Outcome in a Young Fontan Cohort. J. Am. Heart Assoc. 2021, 10, e015022. [Google Scholar] [CrossRef]
  175. Gom, R.C.; Bhatt, D.; Villa, B.R.; George, A.G.; Lohman, A.W.; Mychasiuk, R.; Rho, J.M.; Teskey, G.C. The Ketogenic Diet Raises Brain Oxygen Levels, Attenuates Postictal Hypoxia, and Protects against Learning Impairments. Neurobiol. Dis. 2021, 154, 105335. [Google Scholar] [CrossRef]
  176. Trevisan, R.; Nosadini, R.; Fioretto, P.; Avogaro, A.; Duner, E.; Jori, E.; Valerio, A.; Doria, A.; Crepaldi, G. Ketone Bodies Increase Glomerular Filtration Rate in Normal Man and in Patients with Type 1 (Insulin-Dependent) Diabetes Mellitus. Diabetologia 1987, 30, 214–221. [Google Scholar] [CrossRef] [Green Version]
  177. Zhou, B.; Wang, D.D.-H.; Qiu, Y.; Airhart, S.; Liu, Y.; Stempien-Otero, A.; O’Brien, K.D.; Tian, R. Boosting NAD Level Suppresses Inflammatory Activation of PBMCs in Heart Failure. J. Clin. Investig. 2020, 130, 6054–6063. [Google Scholar] [CrossRef]
  178. Lopaschuk, G.D.; Persad, K.L. Failure to Launch. JACC Basic Transl. Sci. 2023, 8, 280–282. [Google Scholar] [CrossRef]
  179. Frank, B.S.; Khailova, L.; Silveira, L.; Mitchell, M.B.; Morgan, G.J.; Hsieh, E.W.Y.; DiMaria, M.V.; Twite, M.; Klawitter, J.; Davidson, J.A. Proteomic profiling identifies key differences between inter-stage infants with single ventricle heart disease and healthy controls. Transl. Res. 2021, 229, 24–37. [Google Scholar] [CrossRef]
  180. Ohuchi, H.; Miyamoto, Y.; Yamamoto, M.; Ishihara, H.; Takata, H.; Miyazaki, A.; Yamada, O.; Yagihara, T. High Prevalence of Abnormal Glucose Metabolism in Young Adult Patients with Complex Congenital Heart Disease. Am. Heart J. 2009, 158, 30–39. [Google Scholar] [CrossRef]
  181. Whiteside, W.; Tan, M.; Ostlund, R.E.; Yu, S.; Ma, L.; Rocchini, A. Altered Cholesterol Metabolism and Hypocholesterolemia in Patients with Single Ventricle Following Fontan Palliation. J. Pediatr. 2016, 171, 73–77. [Google Scholar] [CrossRef] [Green Version]
  182. Whiteside, W.; Tan, M.; Yu, S.; Rocchini, A. Low Total, Low-Density Lipoprotein, High-Density Lipoprotein, and Non–High-Density Lipoprotein Cholesterol Levels in Patients with Complex Congenital Heart Disease after Fontan Palliation. J. Pediatr. 2013, 162, 1199–1204. [Google Scholar] [CrossRef]
  183. Lubert, A.M.; Alsaied, T.; Palermo, J.J.; Anwar, N.; Urbina, E.M.; Brown, N.M.; Alexander, C.; Almeneisi, H.; Wu, F.; Leventhal, A.R.; et al. Fontan-Associated Dyslipidemia. J. Am. Heart Assoc. 2021, 10, e019578. [Google Scholar] [CrossRef]
  184. Zyblewski, S.C.; Argraves, W.S.; Graham, E.M.; Slate, E.H.; Atz, A.M.; Bradley, S.M.; McQuinn, T.C.; Wilkerson, B.A.; Wing, S.B.; Argraves, K.M. Reduction in postoperative high-density lipoprotein cholesterol levels in children undergoing the Fontan operation. Pediatr. Cardiol. 2012, 33, 1154–1159. [Google Scholar] [CrossRef] [Green Version]
  185. Saraf, A.; De Staercke, C.; Everitt, I.; Haouzi, A.; Ko, Y.A.; Jennings, S.; Kim, J.H.; Rodriguez, F.H.; Kalogeropoulos, A.P.; Quyyumi, A.; et al. Biomarker profile in stable Fontan patients. Int. J. Cardiol. 2020, 305, 56–62. [Google Scholar] [CrossRef]
  186. Fillmore, N.; Lopaschuk, G.D. Targeting Mitochondrial Oxidative Metabolism as an Approach to Treat Heart Failure. Biochim. Biophys. Acta BBA Mol. Cell Res. 2013, 1833, 857–865. [Google Scholar] [CrossRef] [Green Version]
  187. Karwi, Q.G.; Biswas, D.; Pulinilkunnil, T.; Lopaschuk, G.D. Myocardial Ketones Metabolism in Heart Failure. J. Card. Fail. 2020, 26, 998–1005. [Google Scholar] [CrossRef]
  188. Lopaschuk, G.D.; Dyck, J.R.B. Ketones and the Cardiovascular System. Nat. Cardiovasc. Res. 2023, 2, 425–437. [Google Scholar] [CrossRef]
  189. Kashiwaya, Y.; Sato, K.; Tsuchiya, N.; Thomas, S.; Fell, D.A.; Veech, R.L.; Passonneau, J.V. Control of Glucose Utilization in Working Perfused Rat Heart. J. Biol. Chem. 1994, 269, 25502–25514. [Google Scholar] [CrossRef]
  190. Cahill, G.F.; Veech, R.L. Ketoacids? Good Medicine? Trans. Am. Clin. Climatol. Assoc. 2003, 114, 149–163. [Google Scholar]
  191. Veech, R.L.; Chance, B.; Kashiwaya, Y.; Lardy, H.A.; Cahill, G.F., Jr. Ketone Bodies, Potential Therapeutic Uses. IUBMB Life Int. Union Biochem. Mol. Biol. Life 2001, 51, 241–247. [Google Scholar] [CrossRef]
  192. Veech, R.L. The Therapeutic Implications of Ketone Bodies: The Effects of Ketone Bodies in Pathological Conditions: Ketosis, Ketogenic Diet, Redox States, Insulin Resistance, and Mitochondrial Metabolism. Prostaglandins Leukot. Essent. Fatty Acids 2004, 70, 309–319. [Google Scholar] [CrossRef]
  193. Sato, K.; Kashiwaya, Y.; Keon, C.A.; Tsuchiya, N.; King, M.T.; Radda, G.K.; Chance, B.; Clarke, K.; Veech, R.L. Insulin, Ketone Bodies, and Mitochondrial Energy Transduction. FASEB J. 1995, 9, 651–658. [Google Scholar] [CrossRef]
  194. Mudaliar, S.; Alloju, S.; Henry, R.R. Can a Shift in Fuel Energetics Explain the Beneficial Cardiorenal Outcomes in the EMPA-REG OUTCOME Study? A Unifying Hypothesis. Diabetes Care 2016, 39, 1115–1122. [Google Scholar] [CrossRef] [Green Version]
  195. Ho, K.L.; Karwi, Q.G.; Wagg, C.; Zhang, L.; Vo, K.; Altamimi, T.; Uddin, G.M.; Ussher, J.R.; Lopaschuk, G.D. Ketones Can Become the Major Fuel Source for the Heart but Do Not Increase Cardiac Efficiency. Cardiovasc. Res. 2021, 117, 1178–1187. [Google Scholar] [CrossRef]
  196. Berg-Hansen, K.; Christensen, K.H.; Gopalasingam, N.; Nielsen, R.; Eiskjær, H.; Møller, N.; Birkelund, T.; Christensen, S.; Wiggers, H. Beneficial Effects of Ketone Ester in Patients with Cardiogenic Shock: A Randomized, Controlled, Double-Blind Trial. JACC Heart Fail. 2023; ahead of print. [Google Scholar] [CrossRef]
  197. Lopaschuk, G.D.; Karwi, Q.G. Jump Starting the Heart: Ketone Esters Improve Cardiac Function in Patients with Cardiogenic Shock. JACC Heart Fail. 2023, in press. [CrossRef]
  198. Dubois-Deruy, E.; Peugnet, V.; Turkieh, A.; Pinet, F. Oxidative Stress in Cardiovascular Diseases. Antioxidants 2020, 9, 864. [Google Scholar] [CrossRef]
  199. Grieve, D. Oxidative Stress in Heart Failure More than Just Damage. Eur. Heart J. 2003, 24, 2161–2163. [Google Scholar] [CrossRef] [Green Version]
  200. Shimazu, T.; Hirschey, M.D.; Newman, J.; He, W.; Shirakawa, K.; Le Moan, N.; Grueter, C.A.; Lim, H.; Saunders, L.R.; Stevens, R.D.; et al. Suppression of Oxidative Stress by β-Hydroxybutyrate, an Endogenous Histone Deacetylase Inhibitor. Science 2013, 339, 211–214. [Google Scholar] [CrossRef] [Green Version]
  201. Guo, Y.; Zhang, C.; Shang, F.-F.; Luo, M.; You, Y.; Zhai, Q.; Xia, Y.; Suxin, L. Ketogenic Diet Ameliorates Cardiac Dysfunction via Balancing Mitochondrial Dynamics and Inhibiting Apoptosis in Type 2 Diabetic Mice. Aging Dis. 2020, 11, 229. [Google Scholar] [CrossRef] [Green Version]
  202. Murphy, S.P.; Kakkar, R.; McCarthy, C.P.; Januzzi, J.L. Inflammation in Heart Failure. J. Am. Coll. Cardiol. 2020, 75, 1324–1340. [Google Scholar] [CrossRef]
  203. Shirazi, L.F.; Bissett, J.; Romeo, F.; Mehta, J.L. Role of Inflammation in Heart Failure. Curr. Atheroscler. Rep. 2017, 19, 27. [Google Scholar] [CrossRef]
  204. Adamo, L.; Rocha-Resende, C.; Prabhu, S.D.; Mann, D.L. Reappraising the Role of Inflammation in Heart Failure. Nat. Rev. Cardiol. 2020, 17, 269–285. [Google Scholar] [CrossRef]
  205. Suetomi, T.; Willeford, A.; Brand, C.S.; Cho, Y.; Ross, R.S.; Miyamoto, S.; Brown, J.H. Inflammation and NLRP3 Inflammasome Activation Initiated in Response to Pressure Overload by Ca2+/Calmodulin-Dependent Protein Kinase II δ Signaling in Cardiomyocytes Are Essential for Adverse Cardiac Remodeling. Circulation 2018, 138, 2530–2544. [Google Scholar] [CrossRef] [Green Version]
  206. Youm, Y.-H.; Nguyen, K.Y.; Grant, R.W.; Goldberg, E.L.; Bodogai, M.; Kim, D.; D’Agostino, D.; Planavsky, N.; Lupfer, C.; Kanneganti, T.D.; et al. The Ketone Metabolite β-Hydroxybutyrate Blocks NLRP3 Inflammasome–Mediated Inflammatory Disease. Nat. Med. 2015, 21, 263–269. [Google Scholar] [CrossRef] [Green Version]
  207. Maalouf, M.; Sullivan, P.G.; Davis, L.; Kim, D.Y.; Rho, J.M. Ketones Inhibit Mitochondrial Production of Reactive Oxygen Species Production Following Glutamate Excitotoxicity by Increasing NADH Oxidation. Neuroscience 2007, 145, 256–264. [Google Scholar] [CrossRef] [Green Version]
  208. Forsythe, C.E.; Phinney, S.D.; Fernandez, M.L.; Quann, E.E.; Wood, R.J.; Bibus, D.M.; Kraemer, W.J.; Feinman, R.D.; Volek, J.S. Comparison of Low Fat and Low Carbohydrate Diets on Circulating Fatty Acid Composition and Markers of Inflammation. Lipids 2008, 43, 65–77. [Google Scholar] [CrossRef]
  209. Greco, T.; Glenn, T.C.; Hovda, D.A.; Prins, M.L. Ketogenic Diet Decreases Oxidative Stress and Improves Mitochondrial Respiratory Complex Activity. J. Cereb. Blood Flow Metab. 2016, 36, 1603–1613. [Google Scholar] [CrossRef]
  210. Yang, D.; Liu, H.-Q.; Liu, F.-Y.; Guo, Z.; An, P.; Wang, M.-Y.; Yang, Z.; Fan, D.; Tang, Q.-Z. Mitochondria in Pathological Cardiac Hypertrophy Research and Therapy. Front. Cardiovasc. Med. 2022, 8, 822969. [Google Scholar] [CrossRef]
  211. Abel, E.D.; Doenst, T. Mitochondrial Adaptations to Physiological vs. Pathological Cardiac Hypertrophy. Cardiovasc. Res. 2011, 90, 234–242. [Google Scholar] [CrossRef] [Green Version]
  212. Kolwicz, S.C.; Tian, R. Glucose Metabolism and Cardiac Hypertrophy. Cardiovasc. Res. 2011, 90, 194–201. [Google Scholar] [CrossRef] [Green Version]
  213. Lopaschuk, G.D.; Ussher, J.R.; Folmes, C.D.L.; Jaswal, J.S.; Stanley, W.C. Myocardial Fatty Acid Metabolism in Health and Disease. Physiol. Rev. 2010, 90, 207–258. [Google Scholar] [CrossRef]
  214. Kee, H.J.; Kook, H. Roles and Targets of Class I and IIa Histone Deacetylases in Cardiac Hypertrophy. J. Biomed. Biotechnol. 2011, 2011, 928326. [Google Scholar] [CrossRef] [Green Version]
  215. Hewitson, R.; Dargan, J.; Collis, D.; Green, A.; Moorjani, N.; Ohri, S.; Townsend, P.A. Heart Failure: The Pivotal Role of Histone Deacetylases. Int. J. Biochem. Cell Biol. 2013, 45, 448–453. [Google Scholar] [CrossRef]
  216. Yurista, S.R.; Matsuura, T.R.; Silljé, H.H.W.; Nijholt, K.T.; McDaid, K.S.; Shewale, S.V.; Leone, T.C.; Newman, J.C.; Verdin, E.; van Veldhuisen, D.J.; et al. Ketone Ester Treatment Improves Cardiac Function and Reduces Pathologic Remodeling in Preclinical Models of Heart Failure. Circ. Heart Fail. 2021, 14, e007684. [Google Scholar] [CrossRef]
  217. Nakamura, M.; Odanovic, N.; Nakada, Y.; Dohi, S.; Zhai, P.; Ivessa, A.; Yang, Z.; Abdellatif, M.; Sadoshima, J. Dietary Carbohydrates Restriction Inhibits the Development of Cardiac Hypertrophy and Heart Failure. Cardiovasc. Res. 2021, 117, 2365–2376. [Google Scholar] [CrossRef]
  218. Okere, I.C.; Young, M.E.; McElfresh, T.A.; Chess, D.J.; Sharov, V.G.; Sabbah, H.N.; Hoit, B.D.; Ernsberger, P.; Chandler, M.P.; Stanley, W.C. Low Carbohydrate/High-Fat Diet Attenuates Cardiac Hypertrophy, Remodeling, and Altered Gene Expression in Hypertension. Hypertension 2006, 48, 1116–1123. [Google Scholar] [CrossRef] [Green Version]
  219. Egbe, A.C.; Connolly, H.M.; Miranda, W.R.; Ammash, N.M.; Hagler, D.J.; Veldtman, G.R.; Borlaug, B.A. Hemodynamics of Fontan Failure: The Role of Pulmonary Vascular Disease. Circ. Heart Fail. 2017, 10, e004515. [Google Scholar] [CrossRef]
  220. Castaldi, B.; Bordin, G.; Padalino, M.; Cuppini, E.; Vida, V.; Milanesi, O. Hemodynamic Impact of Pulmonary Vasodilators on Single Ventricle Physiology. Cardiovasc. Ther. 2018, 36, e12314. [Google Scholar] [CrossRef] [Green Version]
  221. Zuchi, C.; Tritto, I.; Carluccio, E.; Mattei, C.; Cattadori, G.; Ambrosio, G. Role of Endothelial Dysfunction in Heart Failure. Heart Fail. Rev. 2020, 25, 21–30. [Google Scholar] [CrossRef]
  222. Giannitsi, S.; Maria, B.; Bechlioulis, A.; Naka, K. Endothelial Dysfunction and Heart Failure: A Review of the Existing Bibliography with Emphasis on Flow Mediated Dilation. JRSM Cardiovasc. Dis. 2019, 8, 204800401984304. [Google Scholar] [CrossRef]
  223. McCarthy, C.G.; Chakraborty, S.; Singh, G.; Yeoh, B.S.; Schreckenberger, Z.J.; Singh, A.; Mell, B.; Bearss, N.R.; Yang, T.; Cheng, X.; et al. Ketone Body β-Hydroxybutyrate Is an Autophagy-Dependent Vasodilator. JCI Insight 2021, 6, e149037. [Google Scholar] [CrossRef]
  224. Gormsen, L.C.; Svart, M.; Thomsen, H.H.; Søndergaard, E.; Vendelbo, M.H.; Christensen, N.; Tolbod, L.P.; Harms, H.J.; Nielsen, R.; Wiggers, H.; et al. Ketone Body Infusion With 3-Hydroxybutyrate Reduces Myocardial Glucose Uptake and Increases Blood Flow in Humans: A Positron Emission Tomography Study. J. Am. Heart Assoc. 2017, 6, e005066. [Google Scholar] [CrossRef]
  225. Ibrahim, A. The Effect of Ketone on β-Aminopropionitrile-Induced Vascular Remodeling. Master’s Thesis, Georgia State University, Atlanta, GA, USA, 2022. [Google Scholar] [CrossRef]
  226. Coleman, K.; Phillips, J.; Sciarini, M.; Stubbs, B.; Jackson, O.; Kernagis, D. A Metabolic Intervention for Improving Human Cognitive Performance During Hypoxia. Aerosp. Med. Hum. Perform. 2021, 92, 556–562. [Google Scholar] [CrossRef]
  227. Prins, P.J.; Buxton, J.D.; McClure, T.S.; D’Agostino, D.P.; Ault, D.L.; Welton, G.L.; Jones, D.W.; Atwell, A.D.; Slack, M.A.; Slack, M.L.; et al. Ketone Bodies Impact on Hypoxic CO2 Retention Protocol During Exercise. Front. Physiol. 2021, 12, 780755. [Google Scholar] [CrossRef]
  228. Kashiwaya, Y.; Pawlosky, R.; Markis, W.; King, M.T.; Bergman, C.; Srivastava, S.; Murray, A.; Clarke, K.; Veech, R.L. A Ketone Ester Diet Increases Brain Malonyl-CoA and Uncoupling Proteins 4 and 5 While Decreasing Food Intake in the Normal Wistar Rat. J. Biol. Chem. 2010, 285, 25950–25956. [Google Scholar] [CrossRef] [Green Version]
  229. Puchalska, P.; Crawford, P.A. Multi-Dimensional Roles of Ketone Bodies in Fuel Metabolism, Signaling, and Therapeutics. Cell Metab. 2017, 25, 262–284. [Google Scholar] [CrossRef] [Green Version]
  230. Kim, D.; Roberts, C.; McKenzie, A.; George, M.P. Nutritional Ketosis to Treat Pulmonary Hypertension Associated with Obesity and Metabolic Syndrome: A Case Report. Pulm. Circ. 2021, 11, 2045894021991426. [Google Scholar] [CrossRef]
  231. Chowdhury, B.; Luu, A.Z.; Luu, V.Z.; Kabir, M.G.; Pan, Y.; Teoh, H.; Quan, A.; Sabongui, S.; Al-Omran, M.; Bhatt, D.L.; et al. The SGLT2 Inhibitor Empagliflozin Reduces Mortality and Prevents Progression in Experimental Pulmonary Hypertension. Biochem. Biophys. Res. Commun. 2020, 524, 50–56. [Google Scholar] [CrossRef]
  232. Rychik, J. The Relentless Effects of the Fontan Paradox. Semin. Thorac. Cardiovasc. Surg. Pediatr. Card. Surg. Annu. 2016, 19, 37–43. [Google Scholar] [CrossRef] [Green Version]
  233. Mooli, R.G.R.; Ramakrishnan, S.K. Emerging Role of Hepatic Ketogenesis in Fatty Liver Disease. Front. Physiol. 2022, 13, 946474. [Google Scholar] [CrossRef]
  234. Liao, Y.-J.; Wang, Y.-H.; Wu, C.-Y.; Hsu, F.-Y.; Chien, C.-Y.; Lee, Y.-C. Ketogenic Diet Enhances the Cholesterol Accumulation in Liver and Augments the Severity of CCl4 and TAA-Induced Liver Fibrosis in Mice. Int. J. Mol. Sci. 2021, 22, 2934. [Google Scholar] [CrossRef]
  235. Moore, M.P.; Cunningham, R.P.; Davis, R.A.H.; Deemer, S.E.; Roberts, B.M.; Plaisance, E.P.; Rector, R.S. A Dietary Ketone Ester Mitigates Histological Outcomes of NAFLD and Markers of Fibrosis in High-Fat Diet Fed Mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2021, 320, G564–G572. [Google Scholar] [CrossRef]
  236. Luukkonen, P.K.; Dufour, S.; Lyu, K.; Zhang, X.-M.; Hakkarainen, A.; Lehtimäki, T.E.; Cline, G.W.; Petersen, K.F.; Shulman, G.I.; Yki-Järvinen, H. Effect of a Ketogenic Diet on Hepatic Steatosis and Hepatic Mitochondrial Metabolism in Nonalcoholic Fatty Liver Disease. Proc. Natl. Acad. Sci. USA 2020, 117, 7347–7354. [Google Scholar] [CrossRef] [Green Version]
  237. Sripongpun, P.; Churuangsuk, C.; Bunchorntavakul, C. Current Evidence Concerning Effects of Ketogenic Diet and Intermittent Fasting in Patients with Nonalcoholic Fatty Liver. J. Clin. Transl. Hepatol. 2022, 10, 730–739. [Google Scholar] [CrossRef] [PubMed]
  238. Khuong, J.N.; Wilson, T.G.; Grigg, L.E.; Bullock, A.; Celermajer, D.; Disney, P.; Wijesekera, V.A.; Hornung, T.; Zannino, D.; Iyengar, A.J.; et al. Fontan-Associated Nephropathy: Predictors and Outcomes. Int. J. Cardiol. 2020, 306, 73–77. [Google Scholar] [CrossRef] [PubMed]
  239. Lee, D.; Levin, A.; Kiess, M.; Sexsmith, G.; Chakrabarti, S.; Barlow, A.; Human, D.; Grewal, J. Chronic Kidney Damage in the Adult Fontan Population. Int. J. Cardiol. 2018, 257, 62–66. [Google Scholar] [CrossRef] [PubMed]
  240. Binotto, M.A. Renal Function and Fontan Patients: What Is the Real Impact in the Long-Term Outcomes? Int. J. Cardiol. 2020, 306, 86–87. [Google Scholar] [CrossRef]
  241. Niaz, T.; Stephens, E.H.; Gleich, S.J.; Dearani, J.A.; Johnson, J.N.; Sas, D.J.; Bly, S.; Driscoll, D.J.; Cetta, F. Acute Kidney Injury and Renal Replacement Therapy After Fontan Operation. Am. J. Cardiol. 2021, 161, 84–94. [Google Scholar] [CrossRef]
  242. Zafar, F.; Lubert, A.M.; Katz, D.A.; Hill, G.D.; Opotowsky, A.R.; Alten, J.A.; Goldstein, S.L.; Alsaied, T. Long-Term Kidney Function After the Fontan Operation. J. Am. Coll. Cardiol. 2020, 76, 334–341. [Google Scholar] [CrossRef]
  243. Hems, D.A.; Brosnan, J.T. Effects of Ischaemia on Content of Metabolites in Rat Liver and Kidney in Vivo. Biochem. J. 1970, 120, 105–111. [Google Scholar] [CrossRef] [Green Version]
  244. Tran, M.T.; Zsengeller, Z.K.; Berg, A.H.; Khankin, E.V.; Bhasin, M.K.; Kim, W.; Clish, C.B.; Stillman, I.E.; Karumanchi, S.A.; Rhee, E.P.; et al. PGC1α Drives NAD Biosynthesis Linking Oxidative Metabolism to Renal Protection. Nature 2016, 531, 528–532. [Google Scholar] [CrossRef] [Green Version]
  245. Ritmeester, E.; Veger, V.A.; van der Ven, J.P.G.; van Tussenbroek, G.M.J.W.; van Capelle, C.I.; Udink ten Cate, F.E.A.; Helbing, W.A. Fontan Circulation Associated Organ Abnormalities Beyond the Heart, Lungs, Liver, and Gut: A Systematic Review. Front. Cardiovasc. Med. 2022, 9, 826096. [Google Scholar] [CrossRef]
  246. Puchowicz, M.A.; Emancipator, D.S.; Xu, K.; Magness, D.L.; Ndubuizu, O.I.; Lust, W.D.; LaManna, J.C. Adaptation to Chronic Hypoxia During Diet-Induced Ketosis. In Oxygen Transport to Tissue XXVI; Okunieff, P., Williams, J., Chen, Y., Eds.; Advances in Experimental Medicine and Biology; Springer: New York, NY, USA, 2005; Volume 566, pp. 51–57. [Google Scholar] [CrossRef]
  247. Poffé, C.; Robberechts, R.; Podlogar, T.; Kusters, M.; Debevec, T.; Hespel, P. Exogenous Ketosis Increases Blood and Muscle Oxygenation but Not Performance during Exercise in Hypoxia. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2021, 321, R844–R857. [Google Scholar] [CrossRef]
  248. García-Caballero, M.; Zecchin, A.; Souffreau, J.; Truong, A.-C.K.; Teuwen, L.-A.; Vermaelen, W.; Martín-Pérez, R.; de Zeeuw, P.; Bouché, A.; Vinckier, S.; et al. Role and Therapeutic Potential of Dietary Ketone Bodies in Lymph Vessel Growth. Nat. Metab. 2019, 1, 666–675. [Google Scholar] [CrossRef]
  249. Universitaire Ziekenhuizen KU Leuven. Ketogenic Diet: A Novel Metabolic Strategy to Treat Lymphedema Patients? 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT03991897 (accessed on 4 April 2023).
  250. Puchalska, P.; Crawford, P.A. Ketogenic Therapies for Lymphedema? Nat. Metab. 2019, 1, 656–657. [Google Scholar] [CrossRef]
  251. Dodeja, A.; Urbina, F.; Moore-Padilla, M.; Mah, M.L.; Bradley, D.; Bradley, E. Fontan-Associated Liver Disease: Is Insulin Sensitivity Important? J. Am. Coll. Cardiol. 2020, 75, 549. [Google Scholar] [CrossRef]
  252. Emamaullee, J.; Zaidi, A.N.; Schiano, T.; Kahn, J.; Valentino, P.L.; Hofer, R.E.; Taner, T.; Wald, J.W.; Olthoff, K.M.; Bucuvalas, J.; et al. Fontan-Associated Liver Disease: Screening, Management, and Transplant Considerations. Circulation 2020, 142, 591–604. [Google Scholar] [CrossRef] [PubMed]
  253. Goldenberg, J.Z.; Day, A.; Brinkworth, G.D.; Sato, J.; Yamada, S.; Jönsson, T.; Beardsley, J.; Johnson, J.A.; Thabane, L.; Johnston, B.C. Efficacy and Safety of Low and Very Low Carbohydrate Diets for Type 2 Diabetes Remission: Systematic Review and Meta-Analysis of Published and Unpublished Randomized Trial Data. BMJ 2021, 372, m4743. [Google Scholar] [CrossRef]
  254. Tommerdahl, K.L.; Nelson, R.G.; Bjornstad, P. Dapagliflozin in young people with type 2 diabetes. Lancet Diabetes Endocrinol. 2022, 10, 303–304. [Google Scholar] [CrossRef] [PubMed]
  255. Jiang, W.; Liao, J.; Zhou, D.; Mu, J. Ketogenic Diet Therapy for Epilepsy: Past 100 Years of Practice. Acta Epileptol. 2022, 4, 15. [Google Scholar] [CrossRef]
  256. Suo, C.; Liao, J.; Lu, X.; Fang, K.; Hu, Y.; Chen, L.; Cao, D.; Huang, T.; Li, B.; Li, C. Efficacy and Safety of the Ketogenic Diet in Chinese Children. Seizure 2013, 22, 174–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Wells, J.; Swaminathan, A.; Paseka, J.; Hanson, C. Efficacy and Safety of a Ketogenic Diet in Children and Adolescents with Refractory Epilepsy—A Review. Nutrients 2020, 12, 1809. [Google Scholar] [CrossRef]
  258. Dressler, A.; Trimmel-Schwahofer, P. The Ketogenic Diet for Infants: How Long Can You Go? Epilepsy Res. 2020, 164, 106339. [Google Scholar] [CrossRef]
  259. Mady, M.A.; Kossoff, E.H.; McGregor, A.L.; Wheless, J.W.; Pyzik, P.L.; Freeman, J.M. The Ketogenic Diet: Adolescents Can Do It, Too. Epilepsia 2003, 44, 847–851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  260. Scholl-Bürgi, S.; Höller, A.; Pichler, K.; Michel, M.; Haberlandt, E.; Karall, D. Ketogenic Diets in Patients with Inherited Metabolic Disorders. J. Inherit. Metab. Dis. 2015, 38, 765–773. [Google Scholar] [CrossRef]
  261. Lin, K.-L.; Lin, J.-J.; Wang, H.-S. Application of Ketogenic Diets for Pediatric Neurocritical Care. Biomed. J. 2020, 43, 218–225. [Google Scholar] [CrossRef]
  262. Li, J.; Zhang, H.; Dai, Z. Cancer Treatment with the Ketogenic Diet: A Systematic Review and Meta-Analysis of Animal Studies. Front. Nutr. 2021, 8, 594408. [Google Scholar] [CrossRef] [PubMed]
  263. Lauzier, B.; Vaillant, F.; Merlen, C.; Gélinas, R.; Bouchard, B.; Rivard, M.-E.; Labarthe, F.; Dolinsky, V.W.; Dyck, J.R.B.; Allen, B.G.; et al. Metabolic Effects of Glutamine on the Heart: Anaplerosis versus the Hexosamine Biosynthetic Pathway. J. Mol. Cell. Cardiol. 2013, 55, 92–100. [Google Scholar] [CrossRef] [Green Version]
  264. Hernandez-Saavedra, D.; Sanders, L.; Freeman, S.; Reisz, J.A.; Lee, M.H.; Mickael, C.; Kumar, R.; Kassa, B.; Gu, S.; D’ Alessandro, A.; et al. Stable Isotope Metabolomics of Pulmonary Artery Smooth Muscle and Endothelial Cells in Pulmonary Hypertension and with TGF-Beta Treatment. Sci. Rep. 2020, 10, 413. [Google Scholar] [CrossRef] [Green Version]
  265. Sun, H.; Olson, K.C.; Gao, C.; Prosdocimo, D.A.; Zhou, M.; Wang, Z.; Jeyaraj, D.; Youn, J.-Y.; Ren, S.; Liu, Y.; et al. Catabolic Defect of Branched-Chain Amino Acids Promotes Heart Failure. Circulation 2016, 133, 2038–2049. [Google Scholar] [CrossRef]
  266. Vockley, J.; Charrow, J.; Ganesh, J.; Eswara, M.; Diaz, G.A.; McCracken, E.; Conway, R.; Enns, G.M.; Starr, J.; Wang, R.; et al. Triheptanoin Treatment in Patients with Pediatric Cardiomyopathy Associated with Long Chain-Fatty Acid Oxidation Disorders. Mol. Genet. Metab. 2016, 119, 223–231. [Google Scholar] [CrossRef]
  267. Zöggeler, T.; Stock, K.; Jörg-Streller, M.; Spenger, J.; Konstantopoulou, V.; Hufgard-Leitner, M.; Scholl-Bürgi, S.; Karall, D. Long-Term Experience with Triheptanoin in 12 Austrian Patients with Long-Chain Fatty Acid Oxidation Disorders. Orphanet J. Rare Dis. 2021, 16, 28. [Google Scholar] [CrossRef]
  268. Lei, I.; Tian, S.; Gao, W.; Liu, L.; Guo, Y.; Tang, P.; Chen, E.; Wang, Z. Acetyl-CoA Production by Specific Metabolites Promotes Cardiac Repair after Myocardial Infarction via Histone Acetylation. eLife 2021, 10, e60311. [Google Scholar] [CrossRef] [PubMed]
  269. Johnson, J.N.; Driscoll, D.J.; O’Leary, P.W. Protein-Losing Enteropathy and the Fontan Operation. Nutr. Clin. Pract. 2012, 27, 375–384. [Google Scholar] [CrossRef] [PubMed]
  270. Violante, S.; IJlst, L.; te Brinke, H.; Almeida, I.T.; Wanders, R.J.A.; Ventura, F.V.; Houten, S.M. Carnitine Palmitoyltransferase 2 and Carnitine/Acylcarnitine Translocase Are Involved in the Mitochondrial Synthesis and Export of Acylcarnitines. FASEB J. 2013, 27, 2039–2044. [Google Scholar] [CrossRef] [PubMed]
  271. Pereyra, A.S.; Harris, K.L.; Soepriatna, A.H.; Waterbury, Q.A.; Bharathi, S.S.; Zhang, Y.; Fisher-Wellman, K.H.; Goergen, C.J.; Goetzman, E.S.; Ellis, J.M. Octanoate Is Differentially Metabolized in Liver and Muscle and Fails to Rescue Cardiomyopathy in CPT2 Deficiency. J. Lipid Res. 2021, 62, 100069. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Proven or assumed benefit of ketone bodies on various organ systems. NT-proBNP, N-terminal pro-B-type natriuretic peptide. ↑, increase; ↓, decrease. Created with BioRender.com.
Figure 1. Proven or assumed benefit of ketone bodies on various organ systems. NT-proBNP, N-terminal pro-B-type natriuretic peptide. ↑, increase; ↓, decrease. Created with BioRender.com.
Metabolites 13 00932 g001
Figure 2. Means to achieve therapeutic ketosis. MCT, medium chain triglycerides; BHB, beta-hydroxybutyrate; SGLT2, sodium-glucose co-transporter 2. Created with BioRender.com.
Figure 2. Means to achieve therapeutic ketosis. MCT, medium chain triglycerides; BHB, beta-hydroxybutyrate; SGLT2, sodium-glucose co-transporter 2. Created with BioRender.com.
Metabolites 13 00932 g002
Table 1. State of the heart and metabolic state.
Table 1. State of the heart and metabolic state.
Metabolic State
Substrate ConsumptionETCCKTherapeutic Ketosis
Fatty acidsGlucoseKetone bodies
Healthy heart60–70% [57,58]Remaining [57,58]Remaining [57,58]balanced
HfrEFEarly HF: no change [48,59,60]
Late HF: ↓ [48,59,60]
Early HF: ↑ [48,59,60]
Late HF: ↓ [60,61,62,63,64,65,66]
Progressing HF: ↑ [35,65,67,68,69,70,71,72,73,74,75]Loss of electrons [76]
Accumulation of ROS [76]
early HF: ↓ [48,77,78,79]CO +40% [80]
EF +8% [80]
BV-HF↓ [81]↑ [82,83,84,85]Progressing HF: ↑ [69,86,87,88]Loss of electrons [89,90]
Accumulation of ROS [91,92]
↓ [89,93]CO +27% [87]
PVR—18% [87]
SV-HF↓ [94]↓ [95]?Loss of electrons [96]
Accumulation of ROS [76,97,98]
??
BV, biventricular; CK, creatine kinase energy shuttle; CO, cardiac output; EF, ejection fraction; ETC, electron transport chain; HF, heart failure; PVR, pulmonary vascular resistance; ROS, reactive oxygen species; SV, single ventricle; ↑, increase; ↓, decrease. Note the similar findings for BV HF and SV HF with regards to substrate consumption (fatty acids, glucose, ketone bodies). Thus, therapeutic ketosis is worth considering in the treatment of Fontan patients.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Renaud, D.; Scholl-Bürgi, S.; Karall, D.; Michel, M. Comparative Metabolomics in Single Ventricle Patients after Fontan Palliation: A Strong Case for a Targeted Metabolic Therapy. Metabolites 2023, 13, 932. https://doi.org/10.3390/metabo13080932

AMA Style

Renaud D, Scholl-Bürgi S, Karall D, Michel M. Comparative Metabolomics in Single Ventricle Patients after Fontan Palliation: A Strong Case for a Targeted Metabolic Therapy. Metabolites. 2023; 13(8):932. https://doi.org/10.3390/metabo13080932

Chicago/Turabian Style

Renaud, David, Sabine Scholl-Bürgi, Daniela Karall, and Miriam Michel. 2023. "Comparative Metabolomics in Single Ventricle Patients after Fontan Palliation: A Strong Case for a Targeted Metabolic Therapy" Metabolites 13, no. 8: 932. https://doi.org/10.3390/metabo13080932

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop