Keywords: β-hydroxybutyrate, cardiovascular disease, diabetes, heart failure, ketone bodies, skeletal muscle
Abstract
β-Hydroxybutyrate (βOHB) is the major ketone in the body, and it is recognized as a metabolic energy source and an important signaling molecule. While ketone oxidation is essential in the brain during prolonged fasting/starvation, other organs such as skeletal muscle and the heart also use ketones as metabolic substrates. Additionally, βOHB-mediated molecular signaling events occur in heart and skeletal muscle cells, and via metabolism and/or signaling, ketones may contribute to optimal skeletal muscle health and cardiac function. Of importance, when the use of ketones for ATP production and/or as signaling molecules becomes disturbed in the presence of underlying obesity, type 2 diabetes, and/or cardiovascular diseases, these changes may contribute to cardiometabolic disease. As a result of these disturbances in cardiometabolic disease, multiple approaches have been used to elevate circulating ketones with the goal of optimizing either ketone metabolism or ketone-mediated signaling. These approaches have produced significant improvements in heart and skeletal muscle during cardiometabolic disease with a wide range of benefits that include improved metabolism, weight loss, better glycemic control, improved cardiac and vascular function, as well as reduced inflammation and oxidative stress. Herein, we present the evidence that indicates that ketone therapy could be used as an approach to help treat cardiometabolic diseases by targeting cardiac and skeletal muscles.
INTRODUCTION
Ketone bodies (herein referred to as ketones) were first identified in the late 19th century in the urine of patients in diabetic coma, which led to the field viewing ketones as unwanted products of incomplete fat oxidation (1, 2). However, it was not until the 1960s that pivotal studies in starved human participants demonstrated that ketones become the predominant fuel supporting brain ATP production, illustrating that ketones have a major physiological purpose (3). Indeed, the use of ketones as an alternative fuel source for the brain during times of limited carbohydrate availability is essential given the fact that the brain cannot oxidize fatty acids.
While ketones can be synthesized by amino acids such as leucine, ketones are primarily synthesized in the liver as breakdown products of fatty acid oxidation-derived acetyl CoA and consist of β-hydroxybutyrate (βOHB), acetoacetate (AcAc), and acetone. The major regulatory enzyme of ketone synthesis is 3-hydroxymethyglutaryl-CoA (HMG-CoA) synthase (HMGCS2) and is thus necessary for ketogenesis (Fig. 1). Once synthesized, the major ketones βOHB and AcAc are either transported out of hepatocytes via monocarboxylic acid transporters, or passively diffuse through hepatocyte membranes into the circulation to be taken up by extrahepatic organs to serve as a fuel source for oxidative ATP production. When βOHB and AcAc are imported into extrahepatic cells of the body, βOHB dehydrogenase 1 (BDH1) converts d-βOHB to AcAc while reducing NAD+ to NADH, following which succinyl-CoA:3-ketoacid CoA transferase (SCOT) will transfer the CoA moiety from succinyl-CoA to AcAc to generate acetoacetyl-CoA (Fig. 1). Finally, acetyl-CoA acetyltransferase (ACAT) converts 1 mol of acetoacetyl-CoA into 2 mol of acetyl-CoA that can then enter the Krebs cycle to support ATP production via the generation of reducing equivalents (i.e., NADH) for the electron transport chain. More comprehensive and detailed discussions on ketone metabolism have been extensively reviewed elsewhere (1, 4–6).
Figure 1.
Overview of ketone metabolism. Hepatic fatty acid β-oxidation during fasting/starvation increases acetyl-CoA supply to support ketogenesis. The synthesized ketones, specifically AcAc and βOHB, are transported to the mitochondria of extrahepatic tissues (i.e. skeletal muscle, heart), where they are oxidized back into acetyl-CoA to fuel the Krebs cycle and subsequent oxidative ATP synthesis. AcAc, acetoacetate; Ac-CoA, acetyl CoA; ACAT, acetoacetyl CoA transferase; AcAc-CoA, acetoacetyl CoA; BDH1, β-hydroxybutyrate dehydrogenase 1; βOHB, β-hydroxybutyrate; FFA, free fatty acids; HMG-CoA, hydroxymethylglutaryl CoA; HMGCL, hydroxymethylglutaryl CoA lyase; HMGCS2, hydroxymethylglutaryl CoA synthase; SCOT, succinyl-CoA:3-ketoacid CoA transferase.
As mentioned, ketone oxidation occurs in extrahepatic organs, with the organs with high metabolic demands typically using more ketones as a source of ATP. These organs include brain, skeletal muscle, and the heart. Indeed, the skeletal muscle comprises nearly 40% of an individual’s total body mass (7) and is thus a major consumer of many metabolic substrates, including ketones. Additionally, the heart is the most metabolically demanding organ on a per gram basis (8), thus, can also become a major consumer of ketones particularly when ketone levels are elevated. Accordingly, the molecular and physiological control of ketone metabolism in skeletal muscle and the heart is a key determinant of optimal skeletal muscle health and cardiac function. Furthermore, if the complex regulation of ketone metabolism becomes disturbed, such as in the presence of underlying obesity, these changes may contribute to cardiometabolic diseases [i.e., type 2 diabetes (T2D) and heart failure (HF)] where metabolic disturbances are often hallmarks of these conditions (8, 9).
Although ketones are generally viewed as a fuel source, it is becoming increasingly recognized that ketones have metabolism-independent functions. Indeed, ketones influence several aspects of cellular signaling and gene expression, which subsequently regulate inflammation, oxidative stress, and even appetite (1, 5). Importantly, all of these biological actions are proposed to be key mediators of cardiometabolic disease. Thus, based on the crucial role that ketones play in contributing to overall health, herein we will review how cardiac and skeletal muscle may be therapeutically targeted by ketone supplementation to modify T2D and cardiovascular disease (CVD) outcomes. We will also discuss the differences between the metabolic effects of ketones and the signaling actions of ketones, while presenting evidence about the potential for ketone therapy to alleviate cardiometabolic disease. In most instances, this review will focus on the effects of βOHB and not AcAc as a therapeutic tool to alter signaling and metabolism.
THE SIGNALING EFFECTS OF KETONES
While ketones are an important fuel source, their numerous nonmetabolic signaling effects have been reported within the last 2 decades and are also now well recognized. For instance, βOHB can regulate signaling via serving as a ligand for G protein-coupled receptors (GPCRs), which includes activating GPR109a to both inhibit adipocyte lipolysis (10) and mitigate atherogenesis (11), and it also inhibits GPR41 to reduce sympathetic nervous system activity (12). AcAc can also act on GPCRs, and their distinct signaling differences have been reviewed elsewhere (4, 5).
In addition to GPCR signaling, ketones can also regulate epigenetic modifications. Specifically, βOHB directly inhibits class I histone deacetylases (HDACs), thereby increasing resistance to reactive oxygen species (ROS) via upregulating superoxide dismutase 2 (SOD2) and catalase (13). Furthermore, βOHB-mediated inhibition of class I HDACs also manifests in protective cardiac, spinal, and hepatic effects in healthy and disease models such as diabetes, spinal cord injury, and nonalcoholic (metabolic dysfunction-associated) fatty liver disease (14–17). In other cases, such as the retina and diabetic kidney, βOHB can also confer similar protection against oxidative stress by modulating upstream effectors, such as activating the transcription factor nuclear factor erythroid 2–related factor 2 (Nrf2), which results in HDAC inhibition and subsequently induces antioxidant enzymes such as NAD(P)H-quinone oxidoreductase 1, heme oxygenase 1, and glutathione S-transferases (18–20). While these findings connect βOHB-mediated HDAC inhibition to defense against oxidative stress, HDAC inhibition may also have other favorable effects in cardiometabolic disease, such as T2D and HF. Other epigenetic modifications via βOHB that are now recognized include β-hydroxybutyrylation, a primarily concentration-dependent posttranslational modification. For context, β-hydroxybutyrylation is 10- to 40-fold greater than the ∼2-fold increase in acetylation when assessed in vitro as well as in the livers of fasted or diabetic mice (21). Since β-hydroxybutyrylation competes with acetylation binding sites, the beneficial effects of ketone therapy may also arise from reducing the hyperacetylation observed in cardiometabolic disease (22). However, the impact of these signaling mechanisms has yet to be fully explored.
A rapidly growing area of interest in ketone signaling is the ability of ketones to modulate inflammation by regulating inflammatory cytokine production and subsequent signaling cascades. For instance, βOHB can potently inhibit the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome activation independent of effects on oxidative metabolism, energy status, glycolysis, GPR109a, HDAC inhibition, ROS, or autophagy (23). While these other mechanisms are not necessary for βOHB to inhibit NLRP3, these additional mechanisms can still modulate NLRP3 inhibition in different disease states and therefore should not be excluded when considering the mechanisms by which ketones exert benefit (11, 24–27). By inhibiting the NLRP3 inflammasome, βOHB decreases interleukin (IL)-1β production and secretion (23). This is particularly important as IL-1β plays a role in the pathogenesis and progression of cardiometabolic diseases (28–31). Indeed, reducing IL-1β production or inhibiting IL-1β and its downstream signals has been shown to improve outcomes in preclinical and clinical studies investigating cardiometabolic disease (28–31). Although ketones themselves can initiate signaling cascades, the effects of ketone signaling on inflammation are not necessarily independent of ketone metabolism, as evidenced by the finding that ketone metabolism is necessary for optimal CD8+ T-cell and macrophage function (32, 33). Therefore, it is likely that both ketone metabolism and ketone signaling are important for reducing inflammation and inflammatory cytokine production.
Taken together, ketones have a wide array of signaling effects that can act on several GPCRs, regulate epigenetic modifications, enhance the response to oxidative stress, and inhibit NLRP3-mediated inflammation (Fig. 2). While many of these effects can occur independent of ketone metabolism, ketone metabolism is important in various cell populations and is likely needed to sustain the beneficial effects of ketones in these specific cell types. Moreover, it should be noted that βOHB exists as either a d-βOHB or l-βOHB enantiomer, and while both share signaling actions in many cases, they may also have differing signaling effects as the target proteins may be stereoselective. The general differences between these enantiomers have already been described (34), and the differences in enantiomer-specific effects on cardiometabolic diseases are only beginning to be defined. Nonetheless, the majority of βOHB in mammalian systems exist as D-βOHB, whereas l-βOHB is not normally found in high levels in the systemic circulation (34).
Figure 2.
The cellular signaling effects of ketones. Ketones regulate an array of cellular signaling pathways. They suppress lipolysis, atherogenesis, and sympathetic activity through the activation of GPCRs, specifically GPR109a and GPR41. Additionally, ketones facilitate vasodilation via potassium channels, modulate chromatin architecture to enhance the expression of various antioxidant genes, promote autophagy, and reduce inflammation by inhibiting the NLRP3 inflammasome. AC, acetylation; BHB, β-hydroxybutyrylation; CAT, catalase; GPCRs, G protein-coupled receptors; HDAC, class I histone deacetylases; GST, glutathione S-transferases; HMOX-1, heme oxygenase 1; NLRP3, nucleotide-binding domain-like receptor protein 3; Nrf2, nuclear factor erythroid 2–related factor 2; ROS, reactive oxygen species; SOD2, superoxide dismutase 2.
ROLE OF KETONES IN OBESITY AND TYPE 2 DIABETES
Ketones and Ketogenic Diets
Because insulin inhibits both adipose tissue lipolysis and fatty acid oxidation rates, insulin is a key indirect regulator of hepatic ketogenesis (1, 2). Based on this, dysregulated ketone metabolism is often viewed as a readout of impaired insulin action and worsening diabetes. Indeed, a population-based study of 9,398 Finnish nondiabetic men and men with newly diagnosed T2D [metabolic syndrome in men (METSIM)] observed that increases in fasting blood glucose levels were positively associated with elevated levels of βOHB and AcAc (35). In addition, follow-up examinations over 5 yr in the nondiabetic men from METSIM revealed that elevations in βOHB and AcAc predicted worsening of oral glucose tolerance and a new diagnosis of T2D, further highlighting the important link between circulating ketone levels and T2D. More recently, observations from the Prevention of REnal and Vascular ENd-stage Disease (PREVEND) longitudinal study of 3,307 participants from northern Netherlands also reported that increases in fasting ketone levels were positively associated with incident T2D over 7.3 yr (36). Thus, because elevated ketone levels are correlated with T2D, the concept of elevating ketone levels as a therapeutic approach to treating T2D has not received much attention.
Based in part on the studies described above, it is now becoming increasingly recognized that changes in ketone metabolism may directly contribute to the pathogenesis of obesity and/or T2D. However, several studies have investigated the impact of very high-fat and low-carbohydrate ketogenic diets on weight loss in overweight/obese individuals and the regulation of glycemia. A meta-analysis of individuals adhering to a ketogenic diet for 1–2 yr reported significant decreases in body weight, although the overall mean weight loss was 0.91 kg and was not significant in the studies that followed their participants over 2 yr (37). Thus it appears as though ketogenic diets are more effective in shorter timeframes (38, 39). Several hypotheses have been put forward to explain why such a dietary strategy may be beneficial. As the carbohydrate-insulin model of obesity argues that elevations in postprandial insulin promote body fat accumulation due to the anabolic actions of insulin (40), the low-carbohydrate nature of ketogenic diets would counteract this. Furthermore, the low basal circulating insulin levels associated with adherence to a ketogenic diet would result in elevated rates of adipose tissue lipolysis.
While ketogenic diet-induced ketosis has also been shown to promote satiety and decrease appetite (41), a recent study in normal-weight subjects demonstrated that consumption of exogenous ketone esters (KEs) to induce ketosis may suppress appetite by lowering circulating ghrelin levels (42). In addition to potential weight-loss actions, the low-carbohydrate nature of ketogenic diets is a major contributor to reduced glycemia (even in the absence of weight loss), which leads to less stimulation of islet β-cells to constantly produce and secrete insulin, thereby indirectly improving β-cell function (43). Notwithstanding this, in some preclinical and clinical studies, ketogenic diets induce variable responses. For instance, some studies demonstrated reductions in glycemia and body weight (38, 44–46), whereas others have reported no improvements in these parameters (47, 48). Nevertheless, despite the positive correlation between elevated circulating ketone concentrations and T2D, elevating ketones may benefit patients with T2D by stimulating weight loss and/or improving glycemic control.
Ketone Metabolism and Signaling in Skeletal Muscle
Although many beneficial effects of a ketogenic diet are mediated via weight loss and improvements in glucose handling, there is growing evidence that alterations in skeletal muscle ketone oxidation may also contribute to insulin resistance/T2D. For example, a study of 47 lean and 47 age-matched obese women reported that rectus abdominus muscle biopsy homogenates had decreased βOHB oxidation rates in obese women (49), demonstrating perturbations in muscle ketone metabolism in obesity. Although this may suggest that impaired muscle ketone oxidation contributes to obesity-related insulin resistance, this decrease was related to a marked reduction in mitochondrial content and/or function, as citrate synthase activity was reduced by ∼50%. In contrast, incubation of isolated mouse soleus muscles with 5 mM βOHB decreased insulin-stimulated Akt phosphorylation and 2-deoxyglucose uptake (50). Moreover, the mRNA/protein expression and enzyme activity of SCOT are increased in gastrocnemius muscles from male mice subjected to high-fat diet-induced obesity (51). This would suggest that elevations in muscle ketone oxidation may actually contribute to obesity-related insulin resistance/T2D. In support of this, pharmacological inhibition of SCOT improved glycemia in obese male mice (51, 52). Similarly, skeletal muscle-specific deletion of SCOT in mice also resulted in a robust improvement in glycemia and glucose tolerance in response to experimental obesity or T2D (51).
Mechanisms explaining why decreasing skeletal muscle ketone oxidation would protect against insulin resistance/T2D remain elusive but may simply arise from substrate competition, whereby restricting the use of ketones as fuel leads to a corresponding increase in carbohydrate utilization. In support of this, pharmacological inhibition of SCOT failed to improve glycemia in obese mice with a genetic modification selectively preventing carbohydrate oxidation in skeletal muscle (51). While this may explain why decreasing skeletal muscle βOHB oxidation can alleviate insulin resistance/T2D, contrasting findings also suggest that increases in ketones improve insulin sensitivity via other mechanisms. By interacting with GPR109A on adipocytes and inhibiting lipolysis, βOHB reduces the release of free fatty acids, thereby decreasing hepatic gluconeogenesis and ameliorating the fatty acid-induced antagonism of insulin-mediated glucose uptake. These effects ultimately result in enhanced insulin sensitivity (10). Concurrently, ketones exert an anorectic effect that can pivot the energy balance toward a deficit, causing weight loss and enhancing peripheral insulin sensitivity (42).
Because SCOT inhibition would also increase the intracellular accumulation of AcAc and βOHB, as well as circulating ketone levels, it is possible that the signaling aspects of ketones described in the signaling effects of ketones play a role in regulating glycemia. As previously stated, increases in βOHB have been linked to inhibition of class I HDACs (13), and pharmacological inhibition of class I HDACs has been reported to improve glucose homeostasis in genetically diabetic mice, which was associated with increases in skeletal muscle oxidative metabolism (53). However, it remains to be determined whether inhibiting ketone metabolism per se decreases HDAC signaling specifically in the skeletal muscle.
In contrast to the studies that show that restricting skeletal muscle ketone utilization improves glycemia in obesity/T2D, increases in circulating ketones secondary to treatment with KEs also appear to be beneficial. For example, KE treatment has been shown to improve glycemia in mice and humans with obesity/T2D (54–57). While KEs can acutely stimulate insulin secretion (57–59), it was observed in isolated perifused islets of lean and obese mice that the effect of ketones on insulin secretion was mild at best (60). Despite these mild actions on insulin secretion, acute elevations in circulating ketone levels following oral KE ingestion improved glucose tolerance in both obese mice and obese mice with skeletal muscle-specific deletion of SCOT (60). The latter illustrates that KE-mediated glucose lowering is independent of muscle ketone oxidation, suggesting that the signaling actions of ketones outweigh any potential benefit attributed to a reduction in their oxidation (51, 52).
Of relevance to skeletal muscle health, ketones also have an anticatabolic effect in muscle, which is mediated by signaling pathways such as Akt/FoxO3a and mammalian target of rapamycin/eIF4E-binding protein-1 (mTOR/4E-BP1) (61). The anabolic potential of βOHB was demonstrated in studies where its infusion in healthy men led to a decrease in leucine oxidation and an approximate 10% increase in muscle protein synthesis (62). The previously mentioned mild actions on insulin secretion would also indirectly increase muscle protein synthesis. In addition, βOHB significantly diminishes phenylalanine efflux from the muscle by up to 70% in the setting of lipopolysaccharide-induced muscle inflammation (63, 64). It is noteworthy that the anticatabolic effects of βOHB are minimally increased by hyperinsulinemia, suggesting that its anabolic role is independent of elevated insulin levels. In T2D where the anabolic efficacy of insulin on skeletal muscle is impaired (anabolic resistance) and there is an attenuated rate of muscle protein synthesis and elevated muscle protein breakdown, ketone therapy may represent a novel strategy to improve muscle mass/quality and insulin sensitivity.
Another important aspect of skeletal muscle ketone metabolism that needs to be considered relates to observations that increases in ketone oxidation secondary to acute elevations in circulating ketones have been linked to increases in exercise performance in humans (65). Mechanisms that may explain these improvements in performance may be linked to increases in muscle mitochondrial function and oxygen consumption (66), whereas other studies have reported increases in muscle glucose uptake and glycogen synthesis (67), both of which would benefit overall insulin sensitivity. Nonetheless, the impact of ketones on performance appears to be linked not only to the timing of the ketone intake but also to the specific nature and length of the physical activity [reviewed elsewhere (68)]. While the existing literature mostly focuses on healthy or physically trained individuals, there have been contrasting results, with some studies suggesting improvements in endurance and others showing limited or negative impacts on high-intensity exercise (69, 70, 111). Thus the effects of ketone supplementation on exercise performance in humans have yet to be fully elucidated.
Taken together, it appears that skeletal muscle ketone metabolism influences whole body glucose homeostasis in obesity/T2D (Fig. 3), while increases in muscle ketone metabolism may improve exercise performance and subsequent metabolic health. However, as ketone oxidation and signaling may have contrasting roles in the heart and other organs involved in CVD (as discussed below) versus the muscle, further work is necessary before strategies aimed at decreasing ketone oxidation can be considered as a glucose-lowering strategy.
Figure 3.
Both increasing and decreasing ketone availability to skeletal muscle paradoxically improves glycemia in T2D/obesity. Decreases in skeletal muscle ketone oxidation have glucose-lowering effects in T2D, yet providing exogenous ketones also lowers glucose in T2D, independent of skeletal muscle ketone metabolism. This may involve both an increase in glucose oxidation and/or the signaling effects of ketones. However, the signaling pathways by which ketones improve glycemia are incompletely understood but are suspected to outweigh the impacts of reducing ketone oxidation. T2D, type 2 diabetes.
ROLE OF KETONES IN CARDIAC AND VASCULAR INJURY
Myocardial Infarction and Ischemia
As NLRP3 inflammasome activation is a key mediator of myocardial ischemia/reperfusion (I/R) injury (71–73), the ability of βOHB to inhibit NLRP3 activation (23) suggests that it may have clinical utility in the treatment of myocardial infarction (MI) and/or acute coronary syndromes. While ketone-mediated inhibition of NLRP3 may be attributed to multiple systemic effects, βOHB still induces cardioprotection in ex vivo hearts, indicating a direct cardiac effect. For instance, the addition of βOHB to isolated Langendorff-perfused mouse hearts subjected to I/R results in reduced NLRP3 inflammasome activation and improved cardiac function (74). Similarly, 24 h of subcutaneous βOHB administration in mice post-I/R reduced infarct size and apoptosis while preserving cardiac function (75). These beneficial effects were also associated with improvements in autophagy, mitochondrial function, and oxidative and endoplasmic reticulum stress (75). While intravenous infusion of βOHB before an MI has been shown to reduce infarct size and decrease cardiomyocyte apoptosis in mice (76), as well as reduce oxidative stress and cardiac dysfunction in pigs (77), whether intravenous infusion of βOHB is also effective in a post-MI setting is unknown. Despite some reports identifying a positive correlation between circulating βOHB and MI (78, 79), these other data suggest that further augmenting circulating βOHB beyond the physiological response is cardioprotective.
In support of ketone therapy having beneficial effects following MI and ischemia, investigations using oral KEs in larger animal models and patients have been conducted. Yurista et al. (80) tested whether pigs subjected to I/R and then treated with KE for 3 days would benefit in a post-MI setting. In this study, the KE-treated pigs showed notably lower cardiac NLRP3-related and macrophage-mediated inflammation, oxidative stress, and apoptosis (80), consistent with a previous study that demonstrated that an elevated circulating βOHB reduced cardiac inflammation (74). In addition, in a small trial of patients with cardiogenic shock due to acute MI or decompensated HF, acute enteral KE administration improved cardiac output and peripheral perfusion beyond the effects of the existing vasoactive treatment for cardiogenic shock (81). Taken together, these studies suggest that elevating circulating levels of βOHB via KE supplementation can reduce cardiac injury following hypoxia, ischemia, and MI, and thus lay the foundations for further exploration of ketone therapy in acute coronary syndromes, MI, and decompensated HF.
Heart Failure
Current pharmacological therapies used to treat HF are vast and are continuing to expand (82). While these therapies [such as sodium glucose cotransporter 2 (SGLT2) inhibitors, glucagon-like peptide 1 receptor agonists, angiotensin-converting enzyme inhibitors, angiotensin receptor/neprilysin inhibitors, soluble guanylate cyclase stimulators, β-blockers, etc.] have helped improved cardiac mortality in people with HF, the overall prevalence of HF is still increasing (82). Thus new therapeutic approaches are needed to help treat this devastating condition. One such therapeutic option to help treat HF may be ketone therapy, where this approach may be most effective as an adjunct therapy. The rationale for this potential approach originated with the findings of two independent groups who reported increases in myocardial ketone utilization during HF with reduced ejection fraction (HFrEF) (83, 84). These reports coincided with data from SGLT2 inhibitor cardiovascular outcome trials, which suggested that the cardiovascular benefit of SGLT2 inhibition could be partly due to elevations in ketones (85, 86). As such, investigations into the therapeutic potential of ketones on the failing heart grew considerably.
βOHB is a more oxygen-efficient substrate than fatty acids and can theoretically improve energy production efficiency in the failing heart, which is known to have defects in metabolic processes that normally allow for proper ATP production necessary to maintain contractile function (4, 87). The importance of ketones for cardiac function is exemplified by the findings that a cardiac-specific SCOT-deficient mouse model accelerated the progression of pressure overload-induced cardiac dysfunction and remodeling (88), while another genetically modified mouse with increased circulating levels of βOHB prevented the development of HFrEF (74). Furthermore, various studies have linked the protective effects of ketones in HF with the increase in βOHB utilization/metabolism (89, 90). However, despite this earlier hypothesis, more recent work has challenged this concept and has shown that this protective effect does not appear to be due to an improved cardiac energy efficiency (91, 92). Thus it appears as though improvements in oxygen efficiency are likely not the primary factor driving improved cardiac function and remodeling in HF in the presence of elevated βOHB, and other mechanisms may play more important roles.
Based on the findings discussed above, several studies have investigated other potential mechanisms explaining the beneficial effects of ketones in HF. Indeed, many reports focusing on the pleiotropic signaling effects of ketones have shown that the benefits of ketones in HF could occur via reduced NLRP3 activation, inflammation, and oxidative stress, as well as alterations to mitochondrial function, mitophagy, and autophagy (56, 74, 75, 89, 90, 93, 94). For example, when humans or animals with HFrEF or HF with preserved ejection fraction (HFpEF) are supplemented with exogenous βOHB both acutely or chronically (either through KE, a ketogenic diet, or βOHB injection/infusion), most studies observe an alleviation of cardiac dysfunction and pathological remodeling (22, 80, 90, 91, 95–97). This cardioprotection also exists in mice with T2D, where KE treatment prevents the decline of both systolic and diastolic function (56), similar to the effects observed when on a ketogenic diet (98). Furthermore, treatments that elevate βOHB directly reduce systemic vascular resistance and improve myocardial contractility and blood flow (91, 93, 99, 100), indicating a vascular benefit to treatment with βOHB. While we discuss some of the mechanisms that have been shown to contribute to the beneficial effects of elevated ketones in HF (Fig. 4), it is quite likely that other mechanisms are also involved and have yet to be elucidated.
Figure 4.
Pleiotropic actions by which ketones attenuate cardiovascular dysfunction in cardiometabolic disease. The net therapeutic benefits of ketones are due to a complex interplay of both nonmetabolic cellular signaling and metabolic ATP-producing oxidation. ATP, adenosine triphosphate.
One important distinction of the beneficial effects of ketones in HF is that the salutary effects of βOHB do not appear to be consistent across all forms of HF. While the therapeutic effects of βOHB in HFrEF may occur due to a combination of augmenting both βOHB signaling and oxidation (80, 91, 95, 96), enhanced βOHB signaling and decreased βOHB oxidation appear to confer the beneficial effects observed in mice with HFpEF (22). Importantly, while KE treatment did not reduce body weight in mice with HFpEF, it did ameliorate their hypertension and improve their exercise tolerance (22), thereby demonstrating that ketones can improve HFpEF independent of weight loss. Furthermore, myocardial ketone uptake is decreased in HFpEF patients versus HFrEF patients (101), suggesting differential perturbations of ketone handling or metabolism depending on the type of HF. Although myocardial ketone uptake is generally concentration-dependent, this alone cannot explain the differences in relative extraction among various HF pathologies (101–105). In addition, even though the differences in ketone utilization, kinetics, signaling, and metabolism between different forms of HF remain incompletely understood, ketones still have therapeutic benefits in HFpEF (22, 97). This may be due to the fact that IL-1β is required for HFpEF pathogenesis (28, 106, 107), and since βOHB reduces IL-1β production via NLRP3 inhibition, the provision of KEs may attenuate the development and/or progression of HFpEF via this pathway. This is also consistent with Deng et al. (22) who reported that KE treatment reduced cardiac NLRP3 activation and subsequent IL-1β and IL-18 production. Additionally, ketone therapy may contribute to rescuing diastolic function in HFpEF by inhibiting HDAC activity, which has been shown to govern diastolic dysfunction (108). That said, despite the distinct differences in HFrEF and HFpEF (109), these syndromes still exist on a dynamic continuum (110) and more studies are needed to clarify what specific patient phenotypes may best benefit from ketone therapy. The current clinical trials using ketone therapies for HF and other cardiometabolic diseases are shown in Table 1, and these trials may help to address which diseases are most responsive to ketone therapy.
Table 1.
Clinical trials using exogenous ketones as a therapeutic approach for cardiometabolic disease
Patient Characteristics | Intervention | Aim of Investigation | Clinical Trial Identifier |
---|---|---|---|
20 HFrEF patients | Acute KE vs. placebo; crossover trial | Effect on exercise performance and skeletal muscle PCr (via 31 P-MRS) during exercise | NCT05348460 |
20 HFpEF patients | Acute KE vs. placebo; crossover trial | Effect on exercise performance and cardiovascular function | NCT04633460 |
10 HFpEF patients with T2D | 14 days of KE vs. placebo; crossover trial | Effects on lipolysis rate, and protein/glucose kinetics | NCT05159570 |
20 HFrEF patients | Acute ketone salt vs. carbohydrate supplement; crossover trial | Effect on cardiac function (via cardiac MRI) and blood biomarkers | NCT05651529 |
24 patients with T2D and HFpEF | 14 days of KE vs. placebo; crossover trial | Effect on cardiac function and exercise capacity | NCT05236335 |
10 HFrEF patients | 14 days of KE vs. placebo; crossover trial | Effects on lipolysis rate and protein and glucose kinetics | NCT05161676 |
18 nonischemic HFrEF patients | 10 days of KE vs. ketogenic diet vs. placebo | Effect on cardiac function (via MRI) and biomarkers | NCT04921293 |
8 HFrEF patients | Acute KE vs. ketone salt vs. placebo; crossover trial | Effect on cardiac function | NCT04443426 |
8 HFrEF patients | Acute weight-adujusted dose of KE vs. placebo; crossover trial | Effect on cardiac function | NCT04594265 |
50 HFrEF patients | 12 weeks of KE vs. placebo | Effect on cardiac function and creatine (via MRI) | NCT05924802 |
24 HFrEF patients | 14 days of KE vs. placebo; crossover trial | Effect on cardiac function and exercise capacity | NCT05161650 |
20 HFrEF patients | Acute KE (startified by SGLT2 inhibitor use) | Safety of KE and effect on blood pH, βOHB, glucose, and pressure | NCT05757193 |
12 patients hospitalized with HF | Acute KE vs. placebo (maltodextrin) | Effect on cardiac function | NCT04442555 |
16 HFrEF patients | 2 weeks of KE vs. placebo; crossover trial | Effect on cardiac function and exercise performance | NCT04370600 |
24 patients with acute HFrEF decompensation | Acute KE vs. placebo | Effect on cardiac and hemodynamic function and blood biochemistry | NCT04698005 |
10 comatose survivors of out-of-hospital cardiac arrest | 48 h of KE (nasogastric) | Effect on blood biochemistry, biomarkers, and acid-base status | NCT03226197 |
90 patients with T2D, HF (±T2D), and healthy controls | 14 days of KE | Effect on cardiac function and energetics (PCr via MRS) during rest and dobutamine stress test | ISRCTN24885065 |
12 HFrEF patients | Acute 1,3-butanediol vs. placebo; crossover trial | Effect on cardiac function | NCT05768100 |
15 healthy male adults | Acute KE vs. placebo; crossover trial | Effect on diurnal and nocturnal blood pressure and glucose homeostasis | NCT05794802 |
40 elder adults (>50 years) | Acute KE vs. placebo; crossover trial | Effect on nocturnal blood pressure in aging adults | NCT05888506 |
11 elder adults (50–75 years) | 2 weeks of KE vs. placebo; crossover trial | Effect on vascular function, blood metabolites, and cognition | NCT04236388 |
78 T2D patients with EF <50% | 6-hour dose-dependent βOHB infusion vs. time control | Effect on cardiac function and myocardial glucose uptake (via MRI) | NCT03560323 |
14 T2D patients | Acute pre-prandial KE vs. Na-βOHB (in water) vs. water; crossover trial | Effect on postprandial glucose and metabolites, lipid and glucose kinetics, and metabolic hormones | NCT05263401 |
20 T2D patients | 90 days of concentrated βOHB acid with 1,3-butanediol vs. placebo | Feasibility and safety of chronic ketone supplenmtation; glycemic control, weight, and blood biochemistry/biomarkers | NCT05983562 |
20 T2D patients | 90 days of βOHB acid with 1,3-butanediol vs. placebo | Feasibility and safety of chronic ketone supplenmtation; glycemic control, weight, and blood biochemistry/biomarkers | NCT05477368 |
12 T2D patients | Acute KE vs. placebo; crossover design | Effect on hepatic glucose production and postprandial glycemia | NCT05518448 |
30 T2D or obese patients and healthy controls | Acute KE vs. placebo; crossover trial | Effects on hemodynamic function, appetite, and cognition | NCT05651243 |
18 T2D patients | Acute KE vs. placebo; crossover trial | Effect on glycemia, lipids, inflammation, and blood pressure | NCT04194450 |
15 T2D patients | 14 days of pre-prandial KE vs. placebo; crossover trial | Effect on glycemia, vascular function, and cognition | NCT05155410 |
16 T2D patients | Acute KE vs. placebo; crossover design | Effect on cardiac function, exercise capacity, metabolic rate, and systemic inflammation | NCT04854330 |
T1D patients | Acute KE vs. palcebo; crossover trial | Effect on blood pH and metabolites | NCT04487678 |
31P, 31 phosphorus; EF, ejection fraction; HFpEF, heart failure with preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction; KE, ketone ester; MRI, magnetic resonance imaging; MRS, magnetic resonance spectroscopy; PCr, phosphocreatine; SGLT2, sodium-glucose cotransporter 2; T2D, type 2 diabetes.
As previously mentioned, studies on athletes and fit individuals have suggested that ketone supplementation can improve exercise performance (65, 111, 112). In HF, exercise intolerance is a particularly important symptom contributing to patient morbidity due to reduced exercise and functional capacity (113). Despite this, to date, there have not been any direct investigations to determine whether ketone therapy during HF can improve exercise capacity or symptoms of exercise intolerance, and thus is an area that remains to be explored.
Atrial Fibrillation
Atrial fibrillation (AF) is a common comorbidity in cardiometabolic disease (114, 115) and an area where ketone therapy remains understudied. Since SGLT2 inhibitors are becoming better recognized as an approach to improve AF and arrhythmias (116, 117), and because ketones are believed to mediate some of the cardioprotective effects of SGLT2 inhibitors, it is possible that ketones may also improve AF. Furthermore, since NLRP3 inflammasome activation (and downstream inflammation) is important in the development of AF (118, 119), and because βOHB inhibits NLRP3 activation (23), ketone therapy may be a clinically relevant adjunct for patients with AF. While some studies suggest no correlation between circulating βOHB and new-onset AF (120), other studies show that cardiac βOHB is elevated in AF patients (121) and that circulating βOHB levels are elevated in arrhythmogenic cardiomyopathy patients (105), suggesting the potential for high βOHB concentrations to contribute to AF. While it is unknown how this may occur, βOHB enantiomers themselves have different effects on cardiomyocyte action potential peak current and calcium transients alone or dependent on the presence of other metabolites (122). However, βOHB can augment cardiomyocyte contraction and may improve excitation-contraction coupling (123). Despite these positive correlations/indications, direct investigation of whether βOHB administration ameliorates or exacerbates AF should be explored not only to identify a potential adjunct therapy but also to determine whether ketone therapy may be contraindicated in patients with AF.
Hypertension and Vascular Function
Vascular dysfunction is a major contributor to CVD (110), and thus the effects of ketones on vessel function have been studied. Clinical and preclinical findings recently demonstrated that βOHB reduces vascular resistance and alleviates high-salt-induced hypertension (91, 93, 100, 124, 125), the latter of which was associated with reduced renal inflammation and NLRP3 cascade activation (125). In addition, 2 wk of KE treatment in obese adults also improves vascular function, as measured by flow-mediated dilation (54), further supporting a beneficial vascular effect as a result of elevated circulating ketone levels. While βOHB-mediated vasodilation likely occurs via numerous mechanisms, their direct effects involve small (SK) and intermediate (IK) conductance calcium-activated potassium channels as well as the sodium/potassium ATPase (93). In contrast, ATP-sensitive potassium channels were not involved in mesenteric artery βOHB-dependent vasodilation (93), although they are activated by βOHB and reduce firing in neuronal tissue (126–129). This latter finding may be important in reducing the excess sympathetic drive typically present in hypertension as well as HF. Furthermore, βOHB-dependent vasodilation is not mediated by nitric oxide or cyclooxygenase products in healthy rat mesenteric vessels and also not through GPR41 or GPR109a (93). This lack of involvement of GPR109a was also shown in HFrEF patients where activating GPR109a did not elicit a vasodilatory response like the one that was induced with βOHB (130), suggesting that the vasodilatory mechanisms of βOHB are not mediated by these GPCRs. Regardless of the mechanisms, the ability of βOHB to induce vasodilation may be important in numerous diseases involving restricted blood flow. Indeed, newer lines of investigation have suggested that βOHB may also be of therapeutic utility in pulmonary arterial hypertension and potentially other subtypes of pulmonary hypertension. For example, recent preclinical and clinical evidence demonstrates that increasing circulating βOHB in pulmonary arterial hypertension improves right ventricular function and reduces pulmonary vascular resistance, inflammation, fibrosis, and macrophage activation/infiltration (124, 131). Thus the beneficial effects of ketone therapy may be important for several CVDs.
Ketones also appear to have other favorable vascular effects. This includes a ketone-mediated regulation of cardiac endothelial cell proliferation, migration, and sprouting capacity, the combination of which contributes to preserving vascularization in the hypertrophied mouse heart (132). Likewise, ketones also promote proliferative effects in lymphatic endothelial cells to functionally enable lymphangiogenesis after MI in mice (133). When considering that KE supplementation in healthy males subjected to endurance training increases angiogenesis and skeletal muscle capillarization (112), these data suggest that ketones contribute to vascular remodeling in the organs that have increased energetic demand in both physiological and pathological conditions. Conversely, βOHB signaling also promotes quiescence of endothelial cells, vascular smooth muscle cells, and muscular stem cells (134, 135). However, the reasons for these contrasting proliferative and quiescent effects have yet to be understood. Collectively, ketones prevent vascular and endothelial aging and preserve vessel density in lymphatic and blood vessels, demonstrating the favorable effects of ketones in the vasculature and in vessel injury.
LIMITATIONS AND ADDITIONAL CONSIDERATIONS OF KETONE THERAPY
The growing body of preclinical and clinical evidence supports the expanding use of ketone therapy to treat cardiometabolic diseases. However, ketone therapy is not without its pitfalls and there may be concerns with its use in some clinical contexts. One of the most apparent safety concerns of ketone therapy is diabetic ketoacidosis (DKA), a condition that is predominantly a risk in T1D and rare in T2D patients (136). Although the body can adaptively respond to an elevation in blood ketones up until ∼10 mM βOHB, pathological levels may occur at 10–20 mM or higher and can result in ketoacidosis and detrimental organ effects (137, 138). In most studies though, KEs or other methods to augment circulating ketones result in nonpathological elevations of βOHB. Thus, while ketone therapy may be a potential concern in those with poorly controlled T2D, it does not appear to be of notable concern for those with well-controlled T2D. For example, T2D patients who received 4 wk of KEs did not have concerning changes in acid-base status or a risk of DKA and, instead, had an improvement in glycemic control (55). Furthermore, KE use in these patients also did not result in hypoglycemia, renal dysfunction, electrolyte imbalances, or hypertension (55). Similar to other healthy or diseased individuals using KEs, the potential adverse effects in T2D or obese patients are mild and transient, but further research on the long-term risks of KEs in patients with cardiometabolic disease is still needed. It is also unknown whether KE use may further predispose T2D patients to euglycemic DKA, a syndrome now associated with the use of SGLT2 inhibitors, although preliminary data suggest that this predisposition is unlikely (55). Furthermore, while T1D patients are at risk of DKA, or even euglycemic ketoacidosis, the use of ketone therapy in the context of T1D remains understudied. However, with the increasing popularity and therapeutic uses of exogenous ketones, it will be important to determine the safety of ketone therapy use in patients with well-controlled T1D (Table 1).
We have used ketone therapy to refer to ketogenic diets, KEs, and ketone salts, although there are still other methods of elevating circulating ketones (139). However, unlike KEs or ketone salts, ketogenic diets should not necessarily be considered an equivalent therapeutic option. Indeed, ketogenic diets stimulate endogenous ketosis by virtually eliminating dietary carbohydrate intake. This alone can result in significant changes to the body’s metabolic profile (140). Thus the resulting positive or negative effects cannot directly be attributed to the addition of ketones. Conversely, increases in circulating ketones by KEs or ketone salts do not deprive the body of carbohydrates nor affect the body’s metabolic profile to the same degree. Moreover, in the case of HF, many reports have indicated that the failing heart is metabolically inflexible and has an increased reliance on glucose to meet its energetic demands (9). Thus limiting glucose availability could impair the heart from producing adequate ATP levels, despite whatever beneficial effects may come from ketones. In addition, some reports have observed that ketogenic diets do not improve cardiac function in HF or I/R (84, 90, 141). One also needs to consider the very high-fat nature of ketogenic diets, which may lead to other adverse effects that are not likely to be observed with KEs or ketone salts. One potential adverse effect worth noting with ketogenic diets is that they are frequently associated with increases in circulating low-density lipoprotein (LDL) cholesterol levels (142, 143). However, T2D patients receiving a ketogenic diet compared to the standard of care for 1 yr had favorable improvements in parameters such as high-density lipoprotein (HDL) cholesterol and 10-yr risk of atherosclerotic CVD despite an increase in LDL cholesterol. While it is unknown if sustained elevations in LDL cholesterol levels may contribute to worsening cardiovascular outcomes with more chronic ketogenic diet adherence, this finding is consistent with the notion that the correlation of LDL cholesterol with CVD is oversimplified and incomplete (144). Thus, in general, while the long-term effects of ketogenic diets in T2D/obesity and HF are more ambiguous (37), the preliminary evidence with KEs seems to be more promising (54–56, 60).
With regards to the optimal doses for ketone therapy, elevating circulating βOHB can be therapeutic anywhere from ∼0.5 mM to 5 mM (139, 145), although it stands to reason that a modest, but prolonged, elevation would be preferred to achieve consistency of effects. However, it remains unknown whether pulsatile or consistently elevated levels of circulating βOHB yield greater beneficial effects, as endogenous ketone levels exhibit a diurnal fluctuation (146, 147). Moreover, different KEs can also affect whether or not circulating ketone levels are elevated during the day (95), suggesting that the modality of ketone supplementation should also be considered in determining the ideal dosing times and intervals.
With the various strategies to achieve ketosis (139), the metabolism and the kinetics of all these compounds are different. It also needs to be considered that KEs have a bitter taste and are not palatable. Because of this, and to prolong the duration of ketosis, there are continued efforts to create new KEs that may be more advantageous than the primary ketone monoester (R)-3-hydroxybutyl (R)-3-hydroxybutyrate and the ketone diester 1,3-butanediol acetoacetate diester (148–150). Before the widespread use of KEs and ketone salts, 1,3-butanediol (1,3-BD) was another commonly known ketone precursor used to induce ketosis. While a low dose of 1,3-BD has generally been reported to be safe, using a dose of 1,3-BD that elicits the same βOHB concentrations as with a 24-h fast is deleterious and toxic in rats (151). Interestingly, a low dose of 1,3-BD may improve cardiometabolic parameters independent of its conversion to βOHB (151), suggesting that the beneficial effects of 1,3-BD might occur via an alternate mechanism. While evidence demonstrates improvements in cardiometabolic parameters in up to 4 wk of KE use in humans, there are relatively limited data with longer periods of KE use, and this is needed to better understand the efficacy of chronic ketone therapy. In any case, it should be noted that the use of KEs is likely superior to using salt-based preparations (i.e., sodium-βOHB) as ketone salts have a shorter half-life and may cause potentially dangerous sodium loads (139), even though both are classified as Generally Recognized as Safe supplements by the Food and Drug Administration.
One particularly striking theme of almost all preclinical studies using ketone therapy is the lack of female animals. The lack of study in females is a major shortcoming in translating preclinical findings to use in humans. Interestingly, ketone metabolism appears to decline at a greater rate in male than female mice as they age (152), although more research on female animals and humans is needed to determine what effects this has in the context of cardiometabolic disease. Of relevance, HFpEF is more common in women (153), and thus further investigation of ketone therapy and its potential sexual dimorphic benefits in cardiometabolic disease is warranted.
CONCLUDING REMARKS
There is a growing body of evidence demonstrating the benefits of ketone therapy for cardiometabolic disease, particularly with regard to obesity, T2D, and CVD. The numerous beneficial effects of ketone metabolism and signaling in the heart also extend to the vasculature and skeletal muscle, although which component contributes the most to attenuating cardiometabolic diseases is still unclear. Furthermore, the goal(s) of ketone therapy may require contrasting approaches for improving functional outcomes in the skeletal muscle versus the heart, which will also need to be carefully considered. It is also imperative that future studies continue to explore the benefits of using ketones as a potential adjunct therapy, while also determining whether it may pose certain risks in patients with cardiometabolic disease. As it stands to date, it is likely that ketones may correct metabolic and signaling perturbations to improve inflammation, oxidative stress, and cardiac and vascular function. In addition, ketone therapy can potentially stimulate weight loss and improve glycemic control to improve metabolic outcomes. With further advancements in our understanding of the multifaceted actions of ketones as a fuel source and a signaling molecule, ketone therapy may become an important component of the approach to treating cardiometabolic diseases.
GRANTS
J.R.B.D. receives funding from the Canadian Institutes of Health Research (CIHR), the Heart and Stroke Foundation of Canada, and Diabetes Canada. J.R.U. is supported by CIHR and Diabetes Canada. SS is supported by the CIHR Canada Graduate Doctoral Scholarship and the Izaak Walton Killam Memorial Scholarship. S.A.T.D. is supported by a postdoctoral fellowship from the CIHR (MFE-186352). J.R.U. is a Canada Research Chair in Pharmacotherapy of Energy Metabolism in Obesity. J.R.B.D. is a Canada Research Chair in Molecular Medicine.
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
S.S., S.A.T.D., J.R.U., and J.R.B.D. prepared figures; S.S., S.A.T.D., J.R.U., and J.R.B.D. drafted manuscript; S.S., S.A.T.D., J.R.U., and J.R.B.D. edited and revised manuscript; S.S., S.A.T.D., J.R.U., and J. R.B.D. approved final version of manuscript.
REFERENCES
- 1. Puchalska P, Crawford PA. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab 25: 262–284, 2017. doi: 10.1016/j.cmet.2016.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Robinson AM, Williamson DH. Physiological roles of ketone bodies as substrates and signals in mammalian tissues. Physiol Rev 60: 143–187, 1980. doi: 10.1152/physrev.1980.60.1.143. [DOI] [PubMed] [Google Scholar]
- 3. Owen OE, Morgan AP, Kemp HG, Sullivan JM, Herrera MG, Cahill GF Jr.. Brain metabolism during fasting. J Clin Invest 46: 1589–1595, 1967. doi: 10.1172/JCI105650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Lopaschuk GD, Dyck JR. Ketones and the cardiovascular system. Nat Cardiovasc Res 2: 425–437, 2023. doi: 10.1038/s44161-023-00259-1. [DOI] [Google Scholar]
- 5. Puchalska P, Crawford PA. Metabolic and signaling roles of ketone bodies in health and disease. Annu Rev Nutr 41: 49–77, 2021. doi: 10.1146/annurev-nutr-111120-111518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Matsuura TR, Puchalska P, Crawford PA, Kelly DP. Ketones and the heart: metabolic principles and therapeutic implications. Circ Res 132: 882–898, 2023. doi: 10.1161/CIRCRESAHA.123.321872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Frontera WR, Ochala J. Skeletal muscle: a brief review of structure and function. Calcif Tissue Int 96: 183–195, 2015. doi: 10.1007/s00223-014-9915-y. [DOI] [PubMed] [Google Scholar]
- 8. Taegtmeyer H, Young ME, Lopaschuk GD, Abel ED, Brunengraber H, Darley-Usmar V, Des Rosiers C, Gerszten R, Glatz JF, Griffin JL, Gropler RJ, Holzhuetter HG, Kizer JR, Lewandowski ED, Malloy CR, Neubauer S, Peterson LR, Portman MA, Recchia FA, Van Eyk JE, Wang TJ; American Heart Association Council on Basic Cardiovascular Sciences. Assessing cardiac metabolism: a scientific statement from the American Heart Association. Circ Res 118: 1659–1701, 2016. [Erratum in Circ Res 118: e35, 2016]. doi: 10.1161/RES.0000000000000097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Ussher JR, Elmariah S, Gerszten RE, Dyck JR. The emerging role of metabolomics in the diagnosis and prognosis of cardiovascular disease. J Am Coll Cardiol 68: 2850–2870, 2016. doi: 10.1016/j.jacc.2016.09.972. [DOI] [PubMed] [Google Scholar]
- 10. Taggart AK, Kero J, Gan X, Cai TQ, Cheng K, Ippolito M, Ren N, Kaplan R, Wu K, Wu TJ, Jin L, Liaw C, Chen R, Richman J, Connolly D, Offermanns S, Wright SD, Waters MG. (D)-beta-Hydroxybutyrate inhibits adipocyte lipolysis via the nicotinic acid receptor PUMA-G. J Biol Chem 280: 26649–26652, 2005. doi: 10.1074/jbc.C500213200. [DOI] [PubMed] [Google Scholar]
- 11. Zhang SJ, Li ZH, Zhang YD, Chen J, Li Y, Wu FQ, Wang W, Cui ZJ, Chen GQ. Ketone body 3-hydroxybutyrate ameliorates atherosclerosis via receptor Gpr109a-mediated calcium influx. Adv Sci (Weinh) 8: 2003410, 2021. doi: 10.1002/advs.202003410. . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S, Kobayashi M, Hirasawa A, Tsujimoto G. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc Natl Acad Sci U S A 108: 8030–8035, 2011. doi: 10.1073/pnas.1016088108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, Grueter CA, Lim H, Saunders LR, Stevens RD, Newgard CB, Farese RV Jr, de Cabo R, Ulrich S, Akassoglou K, Verdin E. Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339: 211–214, 2013. doi: 10.1126/science.1227166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Futatani T, Shimao A, Ina S, Higashiyama H, Fujita S, Ueno K, Igarashi N, Hatasaki K. Capillary blood ketone levels as an indicator of inadequate breast milk intake in the early neonatal period. J Pediatr 191: 76–81, 2017. doi: 10.1016/j.jpeds.2017.08.080. [DOI] [PubMed] [Google Scholar]
- 15. Li B, Yu Y, Liu K, Zhang Y, Geng Q, Zhang F, Li Y, Qi J. beta-Hydroxybutyrate inhibits histone deacetylase 3 to promote claudin-5 generation and attenuate cardiac microvascular hyperpermeability in diabetes. Diabetologia 64: 226–239, 2021. doi: 10.1007/s00125-020-05305-2. [DOI] [PubMed] [Google Scholar]
- 16. Kong G, Huang Z, Ji W, Wang X, Liu J, Wu X, Huang Z, Li R, Zhu Q. The ketone metabolite beta-hydroxybutyrate attenuates oxidative stress in spinal cord injury by suppression of class i histone deacetylases. J Neurotrauma 34: 2645–2655, 2017. doi: 10.1089/neu.2017.5192. [DOI] [PubMed] [Google Scholar]
- 17. Moore MP, Cunningham RP, Davis RA, Deemer SE, Roberts BM, Plaisance EP, Rector RS. 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 320: G564–G572, 2021. doi: 10.1152/ajpgi.00259.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Izuta Y, Imada T, Hisamura R, Oonishi E, Nakamura S, Inagaki E, Ito M, Soga T, Tsubota K. Ketone body 3-hydroxybutyrate mimics calorie restriction via the Nrf2 activator, fumarate, in the retina. Aging Cell 17: e12699, 2018. doi: 10.1111/acel.12699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Fang Y, Chen B, Gong AY, Malhotra DK, Gupta R, Dworkin LD, Gong R. The ketone body beta-hydroxybutyrate mitigates the senescence response of glomerular podocytes to diabetic insults. Kidney Int 100: 1037–1053, 2021. [Erratum in Kidney Int 101: 1301–1302, 2022]. doi: 10.1016/j.kint.2021.06.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Kim MN, Moon JH, Cho YM. Sodium-glucose cotransporter-2 inhibition reduces cellular senescence in the diabetic kidney by promoting ketone body-induced NRF2 activation. Diabetes Obes Metab 23: 2561–2571, 2021. doi: 10.1111/dom.14503. [DOI] [PubMed] [Google Scholar]
- 21. Xie Z, Zhang D, Chung D, Tang Z, Huang H, Dai L, Qi S, Li J, Colak G, Chen Y, Xia C, Peng C, Ruan H, Kirkey M, Wang D, Jensen LM, Kwon OK, Lee S, Pletcher SD, Tan M, Lombard DB, White KP, Zhao H, Li J, Roeder RG, Yang X, Zhao Y. Metabolic regulation of gene expression by histone lysine beta-hydroxybutyrylation. Mol Cell 62: 194–206, 2016. doi: 10.1016/j.molcel.2016.03.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Deng Y, Xie M, Li Q, Xu X, Ou W, Zhang Y, Xiao H, Yu H, Zheng Y, Liang Y, Jiang C, Chen G, Du D, Zheng W, Wang S, Gong M, Chen Y, Tian R, Li T. Targeting mitochondria-inflammation circuit by beta-hydroxybutyrate mitigates HFpEF. Circ Res 128: 232–245, 2021. [Erratum in Circ Res 130: e24, 2022]. doi: 10.1161/CIRCRESAHA.120.317933. [DOI] [PubMed] [Google Scholar]
- 23. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, D’Agostino D, Planavsky N, Lupfer C, Kanneganti TD, Kang S, Horvath TL, Fahmy TM, Crawford PA, Biragyn A, Alnemri E, Dixit VD. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med 21: 263–269, 2015. doi: 10.1038/nm.3804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Bae HR, Kim DH, Park MH, Lee B, Kim MJ, Lee EK, Chung KW, Kim SM, Im DS, Chung HY. beta-Hydroxybutyrate suppresses inflammasome formation by ameliorating endoplasmic reticulum stress via AMPK activation. Oncotarget 7: 66444–66454, 2016. doi: 10.18632/oncotarget.12119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Trotta MC, Maisto R, Guida F, Boccella S, Luongo L, Balta C, D’Amico G, Herman H, Hermenean A, Bucolo C, D’Amico M. The activation of retinal HCA2 receptors by systemic beta-hydroxybutyrate inhibits diabetic retinal damage through reduction of endoplasmic reticulum stress and the NLRP3 inflammasome. PloS one 14: e0211005, 2019. doi: 10.1371/journal.pone.0211005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Miyauchi T, Uchida Y, Kadono K, Hirao H, Kawasoe J, Watanabe T, Ueda S, Okajima H, Terajima H, Uemoto S. Up-regulation of FOXO1 and reduced inflammation by beta-hydroxybutyric acid are essential diet restriction benefits against liver injury. Proc Natl Acad Sci U S A 116: 13533–13542, 2019. doi: 10.1073/pnas.1820282116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Soni S, Martens MD, Takahara S, Silver HL, Maayah ZH, Ussher JR, Ferdaoussi M, Dyck JR. Exogenous ketone ester administration attenuates systemic inflammation and reduces organ damage in a lipopolysaccharide model of sepsis. Biochim Biophys Acta Mol Basis Dis 1868: 166507, 2022. doi: 10.1016/j.bbadis.2022.166507. [DOI] [PubMed] [Google Scholar]
- 28. Srinivas BK, Bourdi A, O’Regan JD, Malavalli KD, Rhaleb NE, Belmadani S, Matrougui K. Interleukin-1beta disruption protects male mice from heart failure with preserved ejection fraction pathogenesis. J Am Heart Assoc 12: e029668, 2023. [Erratum in J Am Heart Assoc 12: e027720, 2023]. doi: 10.1161/JAHA.122.029668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, Ravussin E, Stephens JM, Dixit VD. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med 17: 179–188, 2011. doi: 10.1038/nm.2279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, Kastelein JJ, Cornel JH, Pais P, Pella D, Genest J, Cifkova R, Lorenzatti A, Forster T, Kobalava Z, Vida-Simiti L, Flather M, Shimokawa H, Ogawa H, Dellborg M, Rossi PR, Troquay RP, Libby P, Glynn RJ, Group CT, CANTOS Trial Group. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med 377: 1119–1131, 2017. doi: 10.1056/NEJMoa1707914. [DOI] [PubMed] [Google Scholar]
- 31. Lei Y, Devarapu SK, Motrapu M, Cohen CD, Lindenmeyer MT, Moll S, Kumar SV, Anders HJ. Interleukin-1beta inhibition for chronic kidney disease in obese mice with type 2 diabetes. Front Immunol 10: 1223, 2019. doi: 10.3389/fimmu.2019.01223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Luda KM, Longo J, Kitchen-Goosen SM, Duimstra LR, Ma EH, Watson MJ, Oswald BM, Fu Z, Madaj Z, Kupai A, Dickson BM, DeCamp LM, Dahabieh MS, Compton SE, Teis R, Kaymak I, Lau KH, Kelly DP, Puchalska P, Williams KS, Krawczyk CM, Levesque D, Boisvert FM, Sheldon RD, Rothbart SB, Crawford PA, Jones RG. Ketolysis drives CD8(+) T cell effector function through effects on histone acetylation. Immunity 56: 2021–2035.e8, 2023. doi: 10.1016/j.immuni.2023.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Puchalska P, Martin SE, Huang X, Lengfeld JE, Daniel B, Graham MJ, Han X, Nagy L, Patti GJ, Crawford PA. Hepatocyte-macrophage acetoacetate shuttle protects against tissue fibrosis. Cell Metab 29: 383–398, 2019. doi: 10.1016/j.cmet.2018.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Newman JC, Verdin E. beta-Hydroxybutyrate: a signaling metabolite. Annu Rev Nutr 37: 51–76, 2017. doi: 10.1146/annurev-nutr-071816-064916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Mahendran Y, Vangipurapu J, Cederberg H, Stancakova A, Pihlajamaki J, Soininen P, Kangas AJ, Paananen J, Civelek M, Saleem NK, Pajukanta P, Lusis AJ, Bonnycastle LL, Morken MA, Collins FS, Mohlke KL, Boehnke M, Ala-Korpela M, Kuusisto J, Laakso M. Association of ketone body levels with hyperglycemia and type 2 diabetes in 9,398 Finnish men. Diabetes 62: 3618–3626, 2013. doi: 10.2337/db12-1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Szili-Torok T, de Borst MH, Garcia E, Gansevoort RT, Dullaart RP, Connelly MA, Bakker SJ, Tietge UJ. Fasting ketone bodies and incident type 2 diabetes in the general population. Diabetes 72: 1187–1192, 2023. doi: 10.2337/db22-0826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Bueno NB, de Melo IS, de Oliveira SL, da Rocha Ataide T. Very-low-carbohydrate ketogenic diet v. low-fat diet for long-term weight loss: a meta-analysis of randomised controlled trials. Br J Nutr 110: 1178–1187, 2013. doi: 10.1017/S0007114513000548. [DOI] [PubMed] [Google Scholar]
- 38. Yuan X, Wang J, Yang S, Gao M, Cao L, Li X, Hong D, Tian S, Sun C. Effect of the ketogenic diet on glycemic control, insulin resistance, and lipid metabolism in patients with T2DM: a systematic review and meta-analysis. Nutr Diabetes 10: 38, 2020. doi: 10.1038/s41387-020-00142-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Goday A, Bellido D, Sajoux I, Crujeiras AB, Burguera B, Garcia-Luna PP, Oleaga A, Moreno B, Casanueva FF. Short-term safety, tolerability and efficacy of a very low-calorie-ketogenic diet interventional weight loss program versus hypocaloric diet in patients with type 2 diabetes mellitus. Nutr Diabetes 6: e230, 2016. doi: 10.1038/nutd.2016.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Ludwig DS, Ebbeling CB. The carbohydrate-insulin model of obesity: beyond “calories in, calories out”. JAMA Intern Med 178: 1098–1103, 2018. doi: 10.1001/jamainternmed.2018.2933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Gibson AA, Seimon RV, Lee CM, Ayre J, Franklin J, Markovic TP, Caterson ID, Sainsbury A. Do ketogenic diets really suppress appetite? A systematic review and meta-analysis. Obes Rev 16: 64–76, 2015. doi: 10.1111/obr.12230. [DOI] [PubMed] [Google Scholar]
- 42. Stubbs BJ, Cox PJ, Evans RD, Cyranka M, Clarke K, de Wet H. A ketone ester drink lowers human ghrelin and appetite. Obesity (Silver Spring) 26: 269–273, 2018. doi: 10.1002/oby.22051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Sakr HF, Sirasanagandla SR, Das S, Bima AI, Elsamanoudy AZ. Low-carbohydrate ketogenic diet for improvement of glycemic control: mechanism of action of ketosis and beneficial effects. Curr Diabetes Rev 19: e110522204580, 2023. doi: 10.2174/1573399818666220511121629. . [DOI] [PubMed] [Google Scholar]
- 44. Hallberg SJ, McKenzie AL, Williams PT, Bhanpuri NH, Peters AL, Campbell WW, Hazbun TL, Volk BM, McCarter JP, Phinney SD, Volek JS. Effectiveness and safety of a novel care model for the management of type 2 diabetes at 1 year: an open-label, non-randomized, controlled study. Diabetes Ther 9: 583–612, 2018. [Erratum in Diabetes Ther 9: 613–621, 2018]. doi: 10.1007/s13300-018-0373-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Durrer C, McKelvey S, Singer J, Batterham AM, Johnson JD, Gudmundson K, Wortman J, Little JP. A randomized controlled trial of pharmacist-led therapeutic carbohydrate and energy restriction in type 2 diabetes. Nat Commun 12: 5367, 2021. [Erratum in Nat Commun 13: 2590, 2022]. doi: 10.1038/s41467-021-25667-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Zhang Q, Xu L, Xia J, Wang D, Qian M, Ding S. Treatment of diabetic mice with a combination of ketogenic diet and aerobic exercise via modulations of PPARs gene programs. PPAR Res 2018: 4827643, 2018. doi: 10.1155/2018/4827643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Hall KD, Chen KY, Guo J, Lam YY, Leibel RL, Mayer LE, Reitman ML, Rosenbaum M, Smith SR, Walsh BT, Ravussin E. Energy expenditure and body composition changes after an isocaloric ketogenic diet in overweight and obese men. Am J Clin Nutr 104: 324–333, 2016. doi: 10.3945/ajcn.116.133561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Greenwell AA, Saed CT, Tabatabaei Dakhili SA, Ho KL, Gopal K, Chan JS, Kaczmar OO, Dyer SA, Eaton F, Lopaschuk GD, Al Batran R, Ussher JR. An isoproteic cocoa butter-based ketogenic diet fails to improve glucose homeostasis and promote weight loss in obese mice. Am J Physiol Endocrinol Metab 323: E8–E20, 2022. doi: 10.1152/ajpendo.00435.2021. [DOI] [PubMed] [Google Scholar]
- 49. Vice E, Privette JD, Hickner RC, Barakat HA. Ketone body metabolism in lean and obese women. Metabolism 54: 1542–1545, 2005. doi: 10.1016/j.metabol.2005.05.023. [DOI] [PubMed] [Google Scholar]
- 50. Ivarsson N, Zhang SJ, Katz A. AICAR reverses ketone body mediated insulin resistance in isolated oxidative muscle. Biochem Biophys Res Commun 414: 670–674, 2011. doi: 10.1016/j.bbrc.2011.09.122. [DOI] [PubMed] [Google Scholar]
- 51. Al Batran R, Gopal K, Capozzi ME, Chahade JJ, Saleme B, Tabatabaei-Dakhili SA, Greenwell AA, Niu J, Almutairi M, Byrne NJ, Masson G, Kim R, Eaton F, Mulvihill EE, Garneau L, Masters AR, Desta Z, Velazquez-Martinez CA, Aguer C, Crawford PA, Sutendra G, Campbell JE, Dyck JR, Ussher JR. Pimozide alleviates hyperglycemia in diet-induced obesity by inhibiting skeletal muscle ketone oxidation. Cell Metab 31: 909–919, 2020. doi: 10.1016/j.cmet.2020.03.017. [DOI] [PubMed] [Google Scholar]
- 52. Tabatabaei Dakhili SA, Greenwell AA, Yang K, Abou Farraj R, Saed CT, Gopal K, Chan JS, Chahade JJ, Eaton F, Lee C, Velazquez-Martinez CA, Crawford PA, Glover JN, Al Batran R, Ussher JR. The antipsychotic dopamine 2 receptor antagonist diphenylbutylpiperidines improve glycemia in experimental obesity by inhibiting succinyl-CoA:3-ketoacid CoA transferase. Diabetes 72: 126–134, 2023. doi: 10.2337/db22-0221. [DOI] [PubMed] [Google Scholar]
- 53. Galmozzi A, Mitro N, Ferrari A, Gers E, Gilardi F, Godio C, Cermenati G, Gualerzi A, Donetti E, Rotili D, Valente S, Guerrini U, Caruso D, Mai A, Saez E, De Fabiani E, Crestani M. Inhibition of class I histone deacetylases unveils a mitochondrial signature and enhances oxidative metabolism in skeletal muscle and adipose tissue. Diabetes 62: 732–742, 2013. doi: 10.2337/db12-0548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Walsh JJ, Neudorf H, Little JP. 14-Day ketone supplementation lowers glucose and improves vascular function in obesity: a randomized crossover trial. J Clin Endocrinol Metab 106: e1738–e1754, 2021. doi: 10.1210/clinem/dgaa925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Soto-Mota A, Norwitz NG, Evans R, Clarke K, Barber TM. Exogenous ketosis in patients with type 2 diabetes: Safety, tolerability and effect on glycaemic control. Endocrinol Diabetes Metab 4: e00264, 2021. doi: 10.1002/edm2.264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Thai PN, Miller CV, King MT, Schaefer S, Veech RL, Chiamvimonvat N, Bers DM, Dedkova EN. Ketone ester D-beta-hydroxybutyrate-(R)-1,3-butanediol prevents decline in cardiac function in type 2 diabetic mice. J Am Heart Assoc 10: e020729, 2021. doi: 10.1161/JAHA.120.020729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Falkenhain K, Daraei A, Forbes SC, Little JP. Effects of exogenous ketone supplementation on blood glucose: a systematic review and meta-analysis. Adv Nutr 13: 1697–1714, 2022. doi: 10.1093/advances/nmac036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Stubbs BJ, Cox PJ, Evans RD, Santer P, Miller JJ, Faull OK, Magor-Elliott S, Hiyama S, Stirling M, Clarke K. On the metabolism of exogenous ketones in humans. Front Physiol 8: 848, 2017. doi: 10.3389/fphys.2017.00848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Neudorf H, Durrer C, Myette-Cote E, Makins C, O’Malley T, Little JP. Oral ketone supplementation acutely increases markers of NLRP3 inflammasome activation in human monocytes. Mol Nutr Food Res 63: e1801171, 2019. doi: 10.1002/mnfr.201801171. [DOI] [PubMed] [Google Scholar]
- 60. Tabatabaei Dakhili SA, Yang K, Locatelli CA, Saed CT, Greenwell AA, Chan JS, Chahade JJ, Scharff J, Al-Imarah S, Eaton F, Crawford PA, Gopal K, Mulvihill EE, Ussher JR. Ketone ester administration improves glycemia in obese mice. Am J Physiol Cell Physiol 325: C750–C757, 2023. doi: 10.1152/ajpcell.00300.2023. [DOI] [PubMed] [Google Scholar]
- 61. Chen J, Li Z, Zhang Y, Zhang X, Zhang S, Liu Z, Yuan H, Pang X, Liu Y, Tao W, Chen X, Zhang P, Chen GQ. Mechanism of reduced muscle atrophy via ketone body (D)-3-hydroxybutyrate. Cell Biosci 12: 94, 2022. doi: 10.1186/s13578-022-00826-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Nair KS, Welle SL, Halliday D, Campbell RG. Effect of beta-hydroxybutyrate on whole-body leucine kinetics and fractional mixed skeletal muscle protein synthesis in humans. J Clin Invest 82: 198–205, 1988. doi: 10.1172/JCI113570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Mose M, Brodersen K, Rittig N, Schmidt J, Jessen N, Mikkelsen UR, Jorgensen JO, Moller N. Anabolic effects of oral leucine-rich protein with and without beta-hydroxybutyrate on muscle protein metabolism in a novel clinical model of systemic inflammation-a randomized crossover trial. Am J Clin Nutr 114: 1159–1172, 2021. doi: 10.1093/ajcn/nqab148. [DOI] [PubMed] [Google Scholar]
- 64. Thomsen HH, Rittig N, Johannsen M, Moller AB, Jorgensen JO, Jessen N, Moller N. Effects of 3-hydroxybutyrate and free fatty acids on muscle protein kinetics and signaling during LPS-induced inflammation in humans: anticatabolic impact of ketone bodies. Am J Clin Nutr 108: 857–867, 2018. doi: 10.1093/ajcn/nqy170. [DOI] [PubMed] [Google Scholar]
- 65. Cox PJ, Kirk T, Ashmore T, Willerton K, Evans R, Smith A, Murray AJ, Stubbs B, West J, McLure SW, King MT, Dodd MS, Holloway C, Neubauer S, Drawer S, Veech RL, Griffin JL, Clarke K. Nutritional ketosis alters fuel preference and thereby endurance performance in athletes. Cell Metab 24: 256–268, 2016. doi: 10.1016/j.cmet.2016.07.010. [DOI] [PubMed] [Google Scholar]
- 66. Da Costa D, Banagalee V, Naidoo R. Exogenous ketone supplement ingestion and submaximal exercise response in trained, male cyclists: a randomised, double-blind study. Annals Appl Sport Sci 8: S1, 2020. doi: 10.29252/aassjournal.890. [DOI] [Google Scholar]
- 67. Holdsworth DA, Cox PJ, Kirk T, Stradling H, Impey SG, Clarke K. A Ketone ester drink increases postexercise muscle glycogen synthesis in humans. Med Sci Sports Exerc 49: 1789–1795, 2017. doi: 10.1249/MSS.0000000000001292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Pinckaers PJ, Churchward-Venne TA, Bailey D, van Loon LJ. Ketone bodies and exercise performance: the next magic bullet or merely hype? Sports Med 47: 383–391, 2017. doi: 10.1007/s40279-016-0577-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Clark D, Munten S, Herzig KH, Gagnon DD. Exogenous ketone salt supplementation and whole-body cooling do not improve short-term physical performance. Front Nutr 8: 663206, 2021. doi: 10.3389/fnut.2021.663206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. O’Malley T, Myette-Cote E, Durrer C, Little JP. Nutritional ketone salts increase fat oxidation but impair high-intensity exercise performance in healthy adult males. Appl Physiol Nutr Metab 42: 1031–1035, 2017. doi: 10.1139/apnm-2016-0641. [DOI] [PubMed] [Google Scholar]
- 71. Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, Izawa A, Takahashi Y, Masumoto J, Koyama J, Hongo M, Noda T, Nakayama J, Sagara J, Taniguchi S, Ikeda U. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation 123: 594–604, 2011. doi: 10.1161/CIRCULATIONAHA.110.982777. [DOI] [PubMed] [Google Scholar]
- 72. Sandanger O, Ranheim T, Vinge LE, Bliksoen M, Alfsnes K, Finsen AV, Dahl CP, Askevold ET, Florholmen G, Christensen G, Fitzgerald KA, Lien E, Valen G, Espevik T, Aukrust P, Yndestad A. The NLRP3 inflammasome is up-regulated in cardiac fibroblasts and mediates myocardial ischaemia-reperfusion injury. Cardiovasc Res 99: 164–174, 2013. doi: 10.1093/cvr/cvt091. [DOI] [PubMed] [Google Scholar]
- 73. Toldo S, Abbate A. The NLRP3 inflammasome in acute myocardial infarction. Nat Rev Cardiol 15: 203–214, 2018. doi: 10.1038/nrcardio.2017.161. [DOI] [PubMed] [Google Scholar]
- 74. Byrne NJ, Soni S, Takahara S, Ferdaoussi M, Al Batran R, Darwesh AM, Levasseur JL, Beker D, Vos DY, Schmidt MA, Alam AS, Maayah ZH, Schertzer JD, Seubert JM, Ussher JR, Dyck JR. Chronically elevating circulating ketones can reduce cardiac inflammation and blunt the development of heart failure. Circ Heart Fail 13: e006573, 2020. doi: 10.1161/CIRCHEARTFAILURE.119.006573. [DOI] [PubMed] [Google Scholar]
- 75. Yu Y, Yu Y, Zhang Y, Zhang Z, An W, Zhao X. Treatment with D-beta-hydroxybutyrate protects heart from ischemia/reperfusion injury in mice. Eur J Pharmacol 829: 121–128, 2018. doi: 10.1016/j.ejphar.2018.04.019. [DOI] [PubMed] [Google Scholar]
- 76. Zou Z, Sasaguri S, Rajesh KG, Suzuki R. dl-3-Hydroxybutyrate administration prevents myocardial damage after coronary occlusion in rat hearts. Am J Physiol Heart Circ Physiol 283: H1968–1974, 2002. doi: 10.1152/ajpheart.00250.2002. [DOI] [PubMed] [Google Scholar]
- 77. Santos-Gallego CG, Requena-Ibanez JA, Picatoste B, Fardman B, Ishikawa K, Mazurek R, Pieper M, Sartori S, Rodriguez-Capitan J, Fuster V, Badimon JJ. Cardioprotective effect of empagliflozin and circulating ketone bodies during acute myocardial infarction. Circ Cardiovasc Imaging 16: e015298, 2023. doi: 10.1161/CIRCIMAGING.123.015298. [DOI] [PubMed] [Google Scholar]
- 78. de Koning ML, Westenbrink BD, Assa S, Garcia E, Connelly MA, van Veldhuisen DJ, Dullaart RP, Lipsic E, van der Harst P. Association of circulating ketone bodies with functional outcomes after st-segment elevation myocardial infarction. J Am Coll Cardiol 78: 1421–1432, 2021. doi: 10.1016/j.jacc.2021.07.054. [DOI] [PubMed] [Google Scholar]
- 79. Holmes MV, Millwood IY, Kartsonaki C, Hill MR, Bennett DA, Boxall R, Guo Y, Xu X, Bian Z, Hu R, Walters RG, Chen J, Ala-Korpela M, Parish S, Clarke RJ, Peto R, Collins R, Li L, Chen Z, China Kadoorie Biobank Collaborative Group. Lipids, lipoproteins, and metabolites and risk of myocardial infarction and stroke. J Am Coll Cardiol 71: 620–632, 2018. doi: 10.1016/j.jacc.2017.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Yurista SR, Eder RA, Welsh A, Jiang W, Chen S, Foster AN, Mauskapf A, Tang WH, Hucker WJ, Coll-Font J, Rosenzweig A, Nguyen CT. Ketone ester supplementation suppresses cardiac inflammation and improves cardiac energetics in a swine model of acute myocardial infarction. Metabolism 145: 155608, 2023. doi: 10.1016/j.metabol.2023.155608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Berg-Hansen K, Christensen KH, Gopalasingam N, Nielsen R, Eiskjaer H, Moller 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 11: 1337–1347, 2023. doi: 10.1016/j.jchf.2023.05.029. [DOI] [PubMed] [Google Scholar]
- 82. Lam CSP, Docherty KF, Ho JE, McMurray JJ, Myhre PL, Omland T. Recent successes in heart failure treatment. Nat Med 29: 2424–2437, 2023. doi: 10.1038/s41591-023-02567-2. [DOI] [PubMed] [Google Scholar]
- 83. Bedi KC Jr, Snyder NW, Brandimarto J, Aziz M, Mesaros C, Worth AJ, Wang LL, Javaheri A, Blair IA, Margulies KB, Rame JE. Evidence for intramyocardial disruption of lipid metabolism and increased myocardial ketone utilization in advanced human heart failure. Circulation 133: 706–716, 2016. doi: 10.1161/CIRCULATIONAHA.115.017545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Aubert G, Martin OJ, Horton JL, Lai L, Vega RB, Leone TC, Koves T, Gardell SJ, Kruger M, Hoppel CL, Lewandowski ED, Crawford PA, Muoio DM, Kelly DP. The failing heart relies on ketone bodies as a fuel. Circulation 133: 698–705, 2016. doi: 10.1161/CIRCULATIONAHA.115.017355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Ferrannini E, Baldi S, Frascerra S, Astiarraga B, Heise T, Bizzotto R, Mari A, Pieber TR, Muscelli E. Shift to fatty substrate utilization in response to sodium-glucose cotransporter 2 inhibition in subjects without diabetes and patients with type 2 diabetes. Diabetes 65: 1190–1195, 2016. doi: 10.2337/db15-1356. [DOI] [PubMed] [Google Scholar]
- 86. Al Jobori H, Daniele G, Adams J, Cersosimo E, Triplitt C, DeFronzo RA, Abdul-Ghani M. Determinants of the increase in ketone concentration during SGLT2 inhibition in NGT, IFG and T2DM patients. Diabetes Obes Metab 19: 809–813, 2017. doi: 10.1111/dom.12881. [DOI] [PubMed] [Google Scholar]
- 87. Lopaschuk GD, Ussher JR, Folmes CD, Jaswal JS, Stanley WC. Myocardial fatty acid metabolism in health and disease. Physiol Rev 90: 207–258, 2010. doi: 10.1152/physrev.00015.2009. [DOI] [PubMed] [Google Scholar]
- 88. Schugar RC, Moll AR, Andre d’Avignon D, Weinheimer CJ, Kovacs A, Crawford PA. Cardiomyocyte-specific deficiency of ketone body metabolism promotes accelerated pathological remodeling. Mol Metab 3: 754–769, 2014. doi: 10.1016/j.molmet.2014.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Uchihashi M, Hoshino A, Okawa Y, Ariyoshi M, Kaimoto S, Tateishi S, Ono K, Yamanaka R, Hato D, Fushimura Y, Honda S, Fukai K, Higuchi Y, Ogata T, Iwai-Kanai E, Matoba S. Cardiac-specific Bdh1 overexpression ameliorates oxidative stress and cardiac remodeling in pressure overload-induced heart failure. Circ Heart Fail 10: e004417, 2017. doi: 10.1161/CIRCHEARTFAILURE.117.004417. [DOI] [PubMed] [Google Scholar]
- 90. Horton JL, Davidson MT, Kurishima C, Vega RB, Powers JC, Matsuura TR, Petucci C, Lewandowski ED, Crawford PA, Muoio DM, Recchia FA, Kelly DP. The failing heart utilizes 3-hydroxybutyrate as a metabolic stress defense. JCI Insight 4: e124079, 2019. doi: 10.1172/jci.insight.124079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Nielsen R, Moller N, Gormsen LC, Tolbod LP, Hansson NH, Sorensen J, Harms HJ, Frokiaer J, Eiskjaer H, Jespersen NR, Mellemkjaer S, Lassen TR, Pryds K, Botker HE, Wiggers H. Cardiovascular effects of treatment with the ketone body 3-hydroxybutyrate in chronic heart failure patients. Circulation 139: 2129–2141, 2019. doi: 10.1161/CIRCULATIONAHA.118.036459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Ho KL, Zhang L, Wagg C, Al Batran R, Gopal K, Levasseur J, Leone T, Dyck JR, Ussher JR, Muoio DM, Kelly DP, Lopaschuk GD. Increased ketone body oxidation provides additional energy for the failing heart without improving cardiac efficiency. Cardiovasc Res 115: 1606–1616, 2019. doi: 10.1093/cvr/cvz045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. McCarthy CG, Chakraborty S, Singh G, Yeoh BS, Schreckenberger ZJ, Singh A, Mell B, Bearss NR, Yang T, Cheng X, Vijay-Kumar M, Wenceslau CF, Joe B. Ketone body beta-hydroxybutyrate is an autophagy-dependent vasodilator. JCI Insight 6: e149037, 2021. doi: 10.1172/jci.insight.149037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Gambardella J, Jankauskas SS, Kansakar U, Varzideh F, Avvisato R, Prevete N, Sidoli S, Mone P, Wang X, Lombardi A, Santulli G. Ketone bodies rescue mitochondrial dysfunction via epigenetic remodeling. JACC Basic Transl Sci 8: 1123–1137, 2023. doi: 10.1016/j.jacbts.2023.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Yurista SR, Matsuura TR, Sillje HH, Nijholt KT, McDaid KS, Shewale SV, Leone TC, Newman JC, Verdin E, van Veldhuisen DJ, de Boer RA, Kelly DP, Westenbrink BD. Ketone ester treatment improves cardiac function and reduces pathologic remodeling in preclinical models of heart failure. Circ Heart Fail 14: e007684, 2021. doi: 10.1161/CIRCHEARTFAILURE.120.007684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Takahara S, Soni S, Phaterpekar K, Kim TT, Maayah ZH, Levasseur JL, Silver HL, Freed DH, Ferdaoussi M, Dyck JR. Chronic exogenous ketone supplementation blunts the decline of cardiac function in the failing heart. ESC Heart Fail 8: 5606–5612, 2021. doi: 10.1002/ehf2.13634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Liao S, Tang Y, Yue X, Gao R, Yao W, Zhou Y, Zhang H. beta-Hydroxybutyrate mitigated heart failure with preserved ejection fraction by increasing treg cells via Nox2/GSK-3beta. J Inflamm Res 14: 4697–4706, 2021. doi: 10.2147/JIR.S331320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Guo Y, Zhang C, Shang FF, 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 11: 229–240, 2020. doi: 10.14336/AD.2019.0510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Gormsen LC, Svart M, Thomsen HH, Sondergaard E, Vendelbo MH, Christensen N, Tolbod LP, Harms HJ, Nielsen R, Wiggers H, Jessen N, Hansen J, Botker HE, Moller N. 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 6: e005066, 2017. doi: 10.1161/JAHA.116.005066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Homilius C, Seefeldt JM, Axelsen JS, Pedersen TM, Sorensen TM, Nielsen R, Wiggers H, Hansen J, Matchkov VV, Botker HE, Boedtkjer E. Ketone body 3-hydroxybutyrate elevates cardiac output through peripheral vasorelaxation and enhanced cardiac contractility. Basic Res Cardiol 118: 37, 2023. doi: 10.1007/s00395-023-01008-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Murashige D, Jang C, Neinast M, Edwards JJ, Cowan A, Hyman MC, Rabinowitz JD, Frankel DS, Arany Z. Comprehensive quantification of fuel use by the failing and nonfailing human heart. Science 370: 364–368, 2020. doi: 10.1126/science.abc8861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. 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 11: e004953, 2018. doi: 10.1161/CIRCHEARTFAILURE.118.004953. [DOI] [PubMed] [Google Scholar]
- 103. Monzo L, Sedlacek K, Hromanikova K, Tomanova L, Borlaug BA, Jabor A, Kautzner J, Melenovsky V. Myocardial ketone body utilization in patients with heart failure: the impact of oral ketone ester. Metabolism 115: 154452, 2021. doi: 10.1016/j.metabol.2020.154452. [DOI] [PubMed] [Google Scholar]
- 104. Mizuno Y, Harada E, Nakagawa H, Morikawa Y, Shono M, Kugimiya F, Yoshimura M, Yasue H. The diabetic heart utilizes ketone bodies as an energy source. Metabolism 77: 65–72, 2017. doi: 10.1016/j.metabol.2017.08.005. [DOI] [PubMed] [Google Scholar]
- 105. Song JP, Chen L, Chen X, Ren J, Zhang NN, Tirasawasdichai T, Hu ZL, Hua W, Hu YR, Tang HR, Chen HV, Hu SS. Elevated plasma beta-hydroxybutyrate predicts adverse outcomes and disease progression in patients with arrhythmogenic cardiomyopathy. Sci Transl Med 12: eaay8329, 2020. doi: 10.1126/scitranslmed.aay8329. [DOI] [PubMed] [Google Scholar]
- 106. Mishra S, Kass DA. Cellular and molecular pathobiology of heart failure with preserved ejection fraction. Nat Rev Cardiol 18: 400–423, 2021. [Erratum in Nat Rev Cardiol 18: 735, 2021]. doi: 10.1038/s41569-020-00480-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Abbate A, Toldo S, Marchetti C, Kron J, Van Tassell BW, Dinarello CA. Interleukin-1 and the inflammasome as therapeutic targets in cardiovascular disease. Circ Res 126: 1260–1280, 2020. doi: 10.1161/CIRCRESAHA.120.315937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Jeong MY, Lin YH, Wennersten SA, Demos-Davies KM, Cavasin MA, Mahaffey JH, Monzani V, Saripalli C, Mascagni P, Reece TB, Ambardekar AV, Granzier HL, Dinarello CA, McKinsey TA. Histone deacetylase activity governs diastolic dysfunction through a nongenomic mechanism. Sci Transl Med 10: eaao0144, 2018. doi: 10.1126/scitranslmed.aao0144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Zordoky BN, Sung MM, Ezekowitz J, Mandal R, Han B, Bjorndahl TC, Bouatra S, Anderson T, Oudit GY, Wishart DS, Dyck JR, Alberta H, Alberta HEART. Metabolomic fingerprint of heart failure with preserved ejection fraction. PLoS One 10: e0124844, 2015. doi: 10.1371/journal.pone.0124844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Triposkiadis F, Butler J, Abboud FM, Armstrong PW, Adamopoulos S, Atherton JJ, et al. The continuous heart failure spectrum: moving beyond an ejection fraction classification. Eur Heart J 40: 2155–2163, 2019. doi: 10.1093/eurheartj/ehz158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Poffe C, Ramaekers M, Van Thienen R, Hespel P. Ketone ester supplementation blunts overreaching symptoms during endurance training overload. J Physiol 597: 3009–3027, 2019. doi: 10.1113/JP277831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Poffe C, Robberechts R, Van Thienen R, Hespel P. Exogenous ketosis elevates circulating erythropoietin and stimulates muscular angiogenesis during endurance training overload. J Physiol 601: 2345–2358, 2023. doi: 10.1113/JP284346. [DOI] [PubMed] [Google Scholar]
- 113. Del Buono MG, Arena R, Borlaug BA, Carbone S, Canada JM, Kirkman DL, Garten R, Rodriguez-Miguelez P, Guazzi M, Lavie CJ, Abbate A. Exercise intolerance in patients with heart failure: jacc state-of-the-art review. J Am Coll Cardiol 73: 2209–2225, 2019. doi: 10.1016/j.jacc.2019.01.072. [DOI] [PubMed] [Google Scholar]
- 114. Menezes AR, Lavie CJ, Dinicolantonio JJ, O’Keefe J, Morin DP, Khatib S, Abi-Samra FM, Messerli FH, Milani RV. Cardiometabolic risk factors and atrial fibrillation. Rev Cardiovasc Med 14: e73–e81, 2013. [Erratum in Rev Cardiovasc Med 15: 74, 2014]. doi: 10.3909/ricm0693. [DOI] [PubMed] [Google Scholar]
- 115. Odutayo A, Wong CX, Hsiao AJ, Hopewell S, Altman DG, Emdin CA. Atrial fibrillation and risks of cardiovascular disease, renal disease, and death: systematic review and meta-analysis. BMJ 354: i4482, 2016. doi: 10.1136/bmj.i4482. [DOI] [PubMed] [Google Scholar]
- 116. Philippaert K, Kalyaanamoorthy S, Fatehi M, Long W, Soni S, Byrne NJ, Barr A, Singh J, Wong J, Palechuk T, Schneider C, Darwesh AM, Maayah ZH, Seubert JM, Barakat K, Dyck JR, Light PE. Cardiac late sodium channel current is a molecular target for the sodium/glucose cotransporter 2 inhibitor empagliflozin. Circulation 143: 2188–2204, 2021. doi: 10.1161/CIRCULATIONAHA.121.053350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Light PE. Decoding the effects of SGLT2 inhibitors on cardiac arrhythmias in heart failure. Eur Heart J 42: 3739–3740, 2021. doi: 10.1093/eurheartj/ehab563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Yao C, Veleva T, Scott L Jr, Cao S, Li L, Chen G, Jeyabal P, Pan X, Alsina KM, Abu-Taha ID, Ghezelbash S, Reynolds CL, Shen YH, LeMaire SA, Schmitz W, Muller FU, El-Armouche A, Tony Eissa N, Beeton C, Nattel S, Wehrens XH, Dobrev D, Li N. Enhanced cardiomyocyte NLRP3 inflammasome signaling promotes atrial fibrillation. Circulation 138: 2227–2242, 2018. doi: 10.1161/CIRCULATIONAHA.118.035202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Dobrev D, Heijman J, Hiram R, Li N, Nattel S. Inflammatory signalling in atrial cardiomyocytes: a novel unifying principle in atrial fibrillation pathophysiology. Nat Rev Cardiol 20: 145–167, 2023. doi: 10.1038/s41569-022-00759-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Harskamp RE, Granger TM, Clare RM, White KR, Lopes RD, Pieper KS, Granger CB, Newgard CB, Shah SH, Newby LK. Peripheral blood metabolite profiles associated with new onset atrial fibrillation. Am Heart J 211: 54–59, 2019. doi: 10.1016/j.ahj.2019.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Mayr M, Yusuf S, Weir G, Chung YL, Mayr U, Yin X, Ladroue C, Madhu B, Roberts N, De Souza A, Fredericks S, Stubbs M, Griffiths JR, Jahangiri M, Xu Q, Camm AJ. Combined metabolomic and proteomic analysis of human atrial fibrillation. J Am Coll Cardiol 51: 585–594, 2008. doi: 10.1016/j.jacc.2007.09.055. [DOI] [PubMed] [Google Scholar]
- 122. Klos ML, Hou W, Nsengimana B, Weng S, Yan C, Xu S, Devaney E, Han S. Differential effects of beta-hydroxybutyrate enantiomers on induced pluripotent stem derived cardiac myocyte electrophysiology. Biomolecules 12: 1500, 2022. doi: 10.3390/biom12101500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Klos M, Morgenstern S, Hicks K, Suresh S, Devaney EJ. The effects of the ketone body beta-hydroxybutyrate on isolated rat ventricular myocyte excitation-contraction coupling. Arch Biochem Biophys 662: 143–150, 2019. doi: 10.1016/j.abb.2018.11.027. [DOI] [PubMed] [Google Scholar]
- 124. Nielsen R, Christensen KH, Gopalasingam N, Berg-Hansen K, Seefeldt J, Homilius C, Boedtkjer E, Andersen MJ, Wiggers H, Moller N, Botker HE, Mellemkjaer S. Hemodynamic effects of ketone bodies in patients with pulmonary hypertension. J Am Heart Assoc 12: e028232, 2023. doi: 10.1161/JAHA.122.028232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Chakraborty S, Galla S, Cheng X, Yeo JY, Mell B, Singh V, Yeoh B, Saha P, Mathew AV, Vijay-Kumar M, Joe B. Salt-responsive metabolite, beta-hydroxybutyrate, attenuates hypertension. Cell Rep 25: 677–689, 2018. doi: 10.1016/j.celrep.2018.09.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Ma W, Berg J, Yellen G. Ketogenic diet metabolites reduce firing in central neurons by opening K(ATP) channels. J Neurosci 27: 3618–3625, 2007. doi: 10.1523/JNEUROSCI.0132-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Kim DY, Abdelwahab MG, Lee SH, O’Neill D, Thompson RJ, Duff HJ, Sullivan PG, Rho JM. Ketones prevent oxidative impairment of hippocampal synaptic integrity through KATP channels. PLoS One 10: e0119316, 2015. doi: 10.1371/journal.pone.0119316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Tanner GR, Lutas A, Martinez-Francois JR, Yellen G. Single K ATP channel opening in response to action potential firing in mouse dentate granule neurons. J Neurosci 31: 8689–8696, 2011. doi: 10.1523/JNEUROSCI.5951-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Enders JD, Thomas S, Lynch P, Jack J, Ryals JM, Puchalska P, Crawford P, Wright DE. ATP-gated potassium channels contribute to ketogenic diet-mediated analgesia in mice (Preprint). bioRxiv 2023.05.22.541799, 2023. doi: 10.1101/2023.05.22.541799. [DOI] [PMC free article] [PubMed]
- 130. Gopalasingam N, Christensen KH, Berg Hansen K, Nielsen R, Johannsen M, Gormsen LC, Boedtkjer E, Norregaard R, Moller N, Wiggers H. Stimulation of the hydroxycarboxylic acid receptor 2 with the ketone body 3-hydroxybutyrate and niacin in patients with chronic heart failure: hemodynamic and metabolic effects. j Am Heart Assoc, 12: e029849, 2023. doi: 10.1161/JAHA.123.029849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Blake M, Puchalska P, Kazmirczak F, Blake J, Moon R, Thenappan T, Crawford PA, Prins KW. Ketone bodies in right ventricular failure: a unique therapeutic opportunity. Heliyon 9: e22227, 2023. doi: 10.1016/j.heliyon.2023.e22227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Weis EM, Puchalska P, Nelson AB, Taylor J, Moll I, Hasan SS, Dewenter M, Hagenmuller M, Fleming T, Poschet G, Hotz-Wagenblatt A, Backs J, Crawford PA, Fischer A. Ketone body oxidation increases cardiac endothelial cell proliferation. EMBO Mol Med 14: e14753, 2022. doi: 10.15252/emmm.202114753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. García-Caballero M, Zecchin A, Souffreau J, Truong AC, Teuwen LA, Vermaelen W, Martín-Pérez R, de Zeeuw P, Bouché A, Vinckier S, Cornelissen I, Eelen G, Ghesquière B, Mazzone M, Dewerchin M, Carmeliet P. Role and therapeutic potential of dietary ketone bodies in lymph vessel growth. Nat Metab 1: 666–675, 2019. doi: 10.1038/s42255-019-0087-y. [DOI] [PubMed] [Google Scholar]
- 134. Han YM, Bedarida T, Ding Y, Somba BK, Lu Q, Wang Q, Song P, Zou MH. beta-Hydroxybutyrate prevents vascular senescence through hnRNP A1-mediated upregulation of Oct4. Mol Cell 71: 1064–1078.e5, 2018. doi: 10.1016/j.molcel.2018.07.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Benjamin DI, Both P, Benjamin JS, Nutter CW, Tan JH, Kang J, Machado LA, Klein JD, de Morree A, Kim S, Liu L, Dulay H, Feraboli L, Louie SM, Nomura DK, Rando TA. Fasting induces a highly resilient deep quiescent state in muscle stem cells via ketone body signaling. Cell Metab 34: 902–918.e9, 2022. doi: 10.1016/j.cmet.2022.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Lizzo JM, Goyal A, Gupta V. Adult diabetic ketoacidosis. In: StatPearls. Treasure Island, FL: StatPearls Publishing, 2023. [PubMed] [Google Scholar]
- 137. Cahill GF Jr. Fuel metabolism in starvation. Annu Rev Nutr 26: 1–22, 2006. doi: 10.1146/annurev.nutr.26.061505.111258. [DOI] [PubMed] [Google Scholar]
- 138. Cahill GF Jr, Veech RL. Ketoacids? Good medicine? Trans Am Clin Climatol Assoc 114: 149–161, 2003. [PMC free article] [PubMed] [Google Scholar]
- 139. Takahara S, Soni S, Maayah ZH, Ferdaoussi M, Dyck JR. Ketone therapy for heart failure: current evidence for clinical use. Cardiovasc Res 118: 977–987, 2022. doi: 10.1093/cvr/cvab068. [DOI] [PubMed] [Google Scholar]
- 140. Effinger D, Hirschberger S, Yoncheva P, Schmid A, Heine T, Newels P, Schutz B, Meng C, Gigl M, Kleigrewe K, Holdt LM, Teupser D, Kreth S. A ketogenic diet substantially reshapes the human metabolome. Clin Nutr 42: 1202–1212, 2023. doi: 10.1016/j.clnu.2023.04.027. [DOI] [PubMed] [Google Scholar]
- 141. Wang P, Tate JM, Lloyd SG. Low carbohydrate diet decreases myocardial insulin signaling and increases susceptibility to myocardial ischemia. Life Sci 83: 836–844, 2008. doi: 10.1016/j.lfs.2008.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Crosier R, McPherson R. Profound elevation in LDL cholesterol level following a ketogenic diet: a case series. CJC Open 4: 732–734, 2022. doi: 10.1016/j.cjco.2022.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Schmidt T, Harmon DM, Kludtke E, Mickow A, Simha V, Kopecky S. Dramatic elevation of LDL cholesterol from ketogenic-dieting: a case series. Am J Prev Cardiol 14: 100495, 2023. doi: 10.1016/j.ajpc.2023.100495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. O’Neill B, Raggi P. The ketogenic diet: pros and cons. Atherosclerosis 292: 119–126, 2020. doi: 10.1016/j.atherosclerosis.2019.11.021. [DOI] [PubMed] [Google Scholar]
- 145. Yurista SR, Chong CR, Badimon JJ, Kelly DP, de Boer RA, Westenbrink BD. Therapeutic potential of ketone bodies for patients with cardiovascular disease: JACC state-of-the-art review. J Am Coll Cardiol 77: 1660–1669, 2021. doi: 10.1016/j.jacc.2020.12.065. [DOI] [PubMed] [Google Scholar]
- 146. Iwata S, Ozawa K, Shimahara Y, Mori K, Kobayashi N, Kumada K, Yamaoka Y. Diurnal fluctuations of arterial ketone body ratio in normal subjects and patients with liver dysfunction. Gastroenterology 100: 1371–1378, 1991. [PubMed] [Google Scholar]
- 147. Sato S, Parr EB, Devlin BL, Hawley JA, Sassone-Corsi P. Human metabolomics reveal daily variations under nutritional challenges specific to serum and skeletal muscle. Mol Metab 16: 1–11, 2018. doi: 10.1016/j.molmet.2018.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Chen O, Blonquist TM, Mah E, Sanoshy K, Beckman D, Nieman KM, Winters BL, Anthony JC, Verdin E, Newman JC, Stubbs BJ. Tolerability and safety of a novel ketogenic ester, bis-hexanoyl (R)-1,3-butanediol: a randomized controlled trial in healthy adults. Nutrients 13: 2066, 2021. doi: 10.3390/nu13062066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Stubbs BJ, Cook C, Blonquist TM, Taggart K, Beckman D, Kruger C, Conze D, Boileau AC. A randomized, open-label, cross-over pilot study investigating metabolic product kinetics of the palatable novel ketone ester, bis-octanoyl (R)-1,3-butanediol, and bis-hexanoyl (R)-1,3-butanediol ingestion in healthy adults. Toxicol Res Appl 7: 23978473231197835, 2023. doi: 10.1177/23978473231197835. [DOI] [Google Scholar]
- 150. King MT. Ketone ester-what’s in a name? Ambiguity begets uncertainty. Front Physiol 14: 1197768, 2023. doi: 10.3389/fphys.2023.1197768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. McCarthy CG, Waigi EW, Singh G, Castaneda TR, Mell B, Chakraborty S, Wenceslau CF, Joe B. Physiologic, metabolic, and toxicologic profile of 1,3-butanediol. J Pharmacol Exp Ther 379: 245–252, 2021. doi: 10.1124/jpet.121.000796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Eap B, Nomura M, Panda O, Garcia TY, King CD, Rose JP, Leone TC, Kelly DP, Schilling B, Newman JC. Ketone body metabolism declines with age in mice in a sex-dependent manner (Preprint). bioRxiv 10.05.511032, 2022. doi: 10.1101/2022.10.05.511032. [DOI]
- 153. Dunlay SM, Roger VL, Redfield MM. Epidemiology of heart failure with preserved ejection fraction. Nat Rev Cardiol 14: 591–602, 2017. doi: 10.1038/nrcardio.2017.65. [DOI] [PubMed] [Google Scholar]