You are on page 1of 13

Circulation

IN DEPTH
Implications of Altered Ketone Metabolism
and Therapeutic Ketosis in Heart Failure

ABSTRACT: Despite existing therapy, patients with heart failure Senthil Selvaraj, MD, MA
(HF) experience substantial morbidity and mortality, highlighting the Daniel P. Kelly, MD
urgent need to identify novel pathophysiological mechanisms and Kenneth B. Margulies, MD
therapies, as well. Traditional models for pharmacological intervention
have targeted neurohormonal axes and hemodynamic disturbances
in HF. However, several studies have now highlighted the potential
for ketone metabolic modulation as a promising treatment paradigm.
During the pathophysiological progression of HF, the failing heart
reduces fatty acid and glucose oxidation, with associated increases in
ketone metabolism. Recent studies indicate that enhanced myocardial
ketone use is adaptive in HF, and limited data demonstrate beneficial
effects of exogenous ketone therapy in studies of animal models
Downloaded from http://ahajournals.org by on June 2, 2020

and humans with HF. This review will summarize current evidence
supporting a salutary role for ketones in HF including (1) normal
myocardial ketone use, (2) alterations in ketone metabolism in the
failing heart, (3) effects of therapeutic ketosis in animals and humans
with HF, and (4) the potential significance of ketosis associated with
sodium-glucose cotransporter 2 inhibitors. Although a number of
important questions remain regarding the use of therapeutic ketosis
and mechanism of action in HF, current evidence suggests potential
benefit, in particular, in HF with reduced ejection fraction, with
theoretical rationale for its use in HF with preserved ejection fraction.
Although it is early in its study and development, therapeutic ketosis
across the spectrum of HF holds significant promise.

Key Words:  fatty acids ◼ glucose


◼ heart failure ◼ ketones ◼ metabolism
◼ sodium-glucose transporter 2

© 2020 American Heart Association, Inc.

https://www.ahajournals.org/journal/circ

1800 June 2, 2020 Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033


Selvaraj et al Therapeutic Ketosis in Heart Failure

H
eart failure (HF) affects 6.2 million people in the these tasks. FAs provide the bulk of energetic substrate
United States.1 Paradigm shifts in treatment ap- (60%–90%) under normal conditions. However, the

STATE OF THE ART


proaches may help reduce the residual morbidity heart demonstrates metabolic flexibility to adapt to
common to HF with reduced ejection fraction (HFrEF) changing substrate availability, using glucose (through
and preserved ejection fraction (HFpEF). At its most ba- glycolysis and glucose oxidation), ketone bodies, lac-
sic level, HF can be viewed as a state of metabolic insuf- tate, and branched-chain amino acids as necessary.
ficiency, along with the well-known functional impair- Mitochondrial β-oxidation supplies the majority of ATP
ment. Although original descriptions of HF focused on substrate.4 Comprehensive reviews of cardiovascular
hemodynamic maladaptations, and, in general, treat- metabolic substrates have been discussed elsewhere.4,11
ment has targeted the renin-angiotensin-aldosterone However, for the purposes of this article, we will re-
and sympathetic nervous systems, fuel metabolic de- view the role and physiology of ketone metabolism in
rangements are relevant to the pathogenesis of HF. The greater detail.
energy-starvation hypothesis is now well established
and has led some to characterize the failing heart as an
Hepatic Production and Myocardial
“engine out of fuel.”2 As a metabolically demanding
organ, the heart consumes >7 times its weight in ATP Metabolism of Ketone Bodies
daily, with limited reserves available.3 Therefore, aug- Normally, the myocardium minimally extracts ketone
menting cardiac ATP production in a bioenergetically bodies under nonfasting conditions12 because ketones
efficient manner is of significant interest to the HF field. are not normally present in great quantity. Teleologi-
Specific fuel metabolic derangements during the cally, ketosis has been hypothesized as an evolutionary
pathophysiological transition to HF include early impair- mechanism to preserve carbohydrate stores in periods
ments in fatty acid oxidation (FAO) followed by altera- of stress and starvation. Because the brain is largely
tions in glucose oxidation.4 Given that fatty acids (FAs) incapable of oxidizing FAs and predominantly relies
are the chief source of fuel for the normal heart, these on glucose, ketone bodies conserve glucose because
metabolic reprogramming events set the stage for re- ketones can be used in the periphery (and can them-
duced ability to regenerate ATP. In this setting, animal selves constitute neurological fuel). In addition, ketones
and human models have demonstrated increased ke- reduce proteolysis that would be required to support
tone body use in the failing heart.3,5 Very recently, pre- gluconeogenesis from amino acids. Ketogenesis is
Downloaded from http://ahajournals.org by on June 2, 2020

clinical and clinical studies have suggested that exog- therefore a critical part of maintaining metabolic ho-
enous delivery of ketones may improve cardiovascular meostasis during periods of fasting or stress.13
function and prevent the development of pathological Ketone bodies are produced mainly in the liver from
remodeling en route to HF.6,7 These small, early studies mobilized FAs, although other fuel sources (ketogenic
offered a glimpse into a promising new target for phar- amino acids leucine and lysine) may contribute to lim-
macological treatment of HF. The potential significance ited ketogenesis.14,15 During states of FA catabolism,
of ketosis associated with sodium-glucose cotransporter such as fasting (Figure 1), mobilized FAs enter hepato-
2 (SGLT2) inhibitors and reductions in HF morbidity and cytes and ultimately undergo complete mitochondrial
cardiovascular mortality observed in patients with HF β-oxidation to form several molecules of acetyl coen-
(irrespective of diabetes status) further motivates inves- zyme A (acetyl-CoA). Several enzymatic reactions cata-
tigation of the potential benefits of ketones in patients lyze the conversion of acetyl-CoA to acetoacetate, the
with HF.8,9 Accordingly, the goals of this review are to first ketone body able to be delivered in the circulation.
characterize normal myocardial ketone metabolism, de- Acetoacetate can also be reduced to β-hydroxybutyrate
scribe metabolic reprogramming and shifts in substrate (BHB) for systemic transport. Therefore, BHB, acetoace-
use in patients with HF, and review recent studies of tic acid, and the breakdown product acetone are the
therapeutic ketosis both in humans and animal models. 3 ketone bodies endogenously produced. Levels of ≈2
to 3 mmol/L BHB can be reached after a few days of
fasting.16 Ketogenesis is influenced by hormonal signal-
KETONE METABOLISM: A BRIEF ing, transcriptional regulation, and posttranscriptional
modification.13 Fates of ketone bodies include urinary
PRIMER or lung elimination, lipid synthesis, or transport to ex-
The energetic requirements of the human myocardium trahepatic tissue for use.
are robust because of the persistent pumping of ≈7200 Circulating ketone bodies are taken up by tissues
L of blood per day against systemic vascular resistance, through monocarboxylate transporters (MCTs) for use
using 35 L of oxygen to generate the requisite kilo- in organs capable of ketone oxidation. In myocardial
gram quantities of ATP daily.10 Given a limited capac- mitochondria, BHB dehydrogenase 1 (BDH1) oxidizes
ity to store energy, the heart turns over ATP stores at BHB to acetoacetate. Acetoacetate is then activated
the remarkable rate of every 10 seconds to accomplish to acetoacetyl-CoA by succinyl-CoA:3 ketoacid-CoA

Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033 June 2, 2020 1801


Selvaraj et al Therapeutic Ketosis in Heart Failure
STATE OF THE ART
Downloaded from http://ahajournals.org by on June 2, 2020

Figure 1. Pathways of hepatic ketogenesis and myocardial ketolysis.


Exogenous fats are absorbed through the gastrointestinal tract (specifically, medium-chain triglycerides can be hydrolyzed by lipases to generate medium-chain
fatty acids). In addition, free fatty acids generated by adipolysis are circulated to the liver, where they undergo fatty acid oxidation in the hepatic mitochon-
dria to produce acetyl coenzyme A (acetyl-CoA). Subsequent ketogenesis ultimately produces 2 mature ketone bodies: acetoacetate and β-hydroxybutyrate
(β-OHB). Export from the hepatocytes and import into the cardiomyocyte is accomplished through monocarboxylate transporters. In cardiomyocyte mitochon-
dria, ketolysis produces acetyl-CoA that can be used in the tricyclic acid cycle. Circulating ketones that are not oxidized by tissue can be (1) used for lipid or
cholesterol synthesis (acetoacetate only) or (2) eliminated through the urine or exhaled as acetone (derived from decarboxylation of acetoacetate). Therapies
used to achieve ketosis are noted in red. ACAT indicates acetyl-CoA acetyltransferase; BHB, β-hydroxybutyrate; BDH, BHB dehydrogenase; CoA-SH, coenzyme
A with sulfhydryl functional group; HMG-CoA, 3-hydroxy-3-methylglutaryl coenzyme A; HMGCL, 3-hydroxy-3-methylglutaryl coenzyme A lyase; HMGCS2,
3-hydroxy-3-methylglutaryl coenzyme A synthase; MCT, monocarboxylate transporter; SCOT, succinyl-CoA:3 ketoacid-CoA transferase; SGLT2, sodium-glucose
cotransporter 2; and TCA; tricyclic acid.

transferase, which then undergoes a thiolysis reaction pathology.20–22 These studies highlight the importance
by acetyl-CoA acetyltransferase to produce 2 molecules of ensuring an anaplerotic input to the TCA cycle in the
of acetyl-CoA that can enter the tricyclic acid (TCA) context of increasing ketone body metabolism. Thus,
cycle. These enzymes are critical to ketolysis, consis- some degree of glucose metabolism is required to pro-
tent with observations that genetic defects in succinyl- vide functional benefits from ketone therapies.
CoA:3 ketoacid-CoA transferase, acetyl-CoA acetyl- Loss of ketone metabolic capability can be function-
transferase, and MCT1 cause recurrent ketoacidosis.17 ally significant in settings of myocardial stress. This is
The myocardium has among the highest levels of keto- illustrated in a cardiomyocyte-specific succinyl-CoA:3
lytic enzyme activity in the body.18 Indeed, states associ- ketoacid-CoA transferase knockout mice model, result-
ated with physiological increases in circulating ketones ing in impaired terminal ketone body oxidation. After 8
increase myocardial glycogen content in contrast with weeks of transaortic constriction (TAC), left ventricular
decreases observed in skeletal muscle.19 (LV) volume markedly increased and ejection fraction
In contrast with glucose, an anaplerotic fuel source (EF) decreased in succinyl-CoA:3 ketoacid-CoA transfer-
that replenishes TCA cycle intermediates, ketones are ase knockout mice in comparison with control mice.23
cataplerotic and deplete TCA intermediates. Conse- Similarly, mice with BDH1 gene knockout exhibit wors-
quently, in rat hearts, ketones as a sole source of fuel led ened pathological remodeling in response to combined
to impairments in contractile function, whereas delivery TAC and myocardial infarction.5 BDH1 knockout mice
of anaplerotic substrates such as pyruvate reversed the also exhibit cardiac dysfunction with prolonged fasting.7

1802 June 2, 2020 Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033


Selvaraj et al Therapeutic Ketosis in Heart Failure

Impact of Ketone Bodies on Cardiac of these shifts.33 In chronic pressure overload models
Energetic Properties and Efficiency of HF, FAO mRNA levels were markedly reduced in LV

STATE OF THE ART


hypertrophy rats in comparison with controls, whereas
Beyond their contribution to energy generation, ketone protein levels and enzyme activities were not signifi-
oxidation is relatively efficient in terms of oxygen use.4 cantly different until HF develops.30 Similar shifts away
FAO using a substrate such as palmitate renders 105 from FAO to glucose oxidation are observed early in a
ATP molecules at the expense of 23 molecules of oxy- canine chronic tachypacing model of HF.7 This impair-
gen per 2-carbon moiety, with a phosphate-to-oxygen ment in FAO combined with increased reliance on glu-
ratio of 2.3.4 Glucose oxidation generates only 31 ATP cose oxidation resembles the fetal metabolic program.7
molecules while consuming 6 molecules of oxygen; de- Overall, these shifts in metabolic programming dimin-
spite a lower ATP yield, glucose oxidation is more ef- ish the capacity of the mitochondria to generate high
ficient than FAO with a higher phosphate-to-oxygen ra- quantities of ATP through FAO.
tio (2.6).4 In this context, ketone bodies strike a balance
between energetic production and efficiency. Ketone
oxidation has a phosphate-to-oxygen ratio of 2.5, and, Alterations in Glucose Metabolism
thus, ketone bodies are energetically more efficient As an efficient metabolic substrate, glucose is theoreti-
than FAO and generate more ATP per 2-carbon moiety cally well positioned to support the high ATP require-
than glucose. Indeed, the addition of ketone bodies to ments of the failing heart and is generally consumed
working rat hearts perfused with glucose buffer signifi- to a greater degree in early stages of HF.35 However,
cantly increases cardiac output and efficiency in a man- progressive deficits in glucose oxidation lead to greater
ner similar to insulin.24 flux through the glycolytic pathway, a much less effi-
cient process for generation of ATP. In hypertrophied
hearts, the reduced contribution of FAO to ATP gen-
Preferential Use of Ketones in the
eration at low workloads is matched by an increase in
Normal Heart glycolysis.36–38 Moreover, the increase in glucose use in
Under normal conditions, oxygen consumption is not early-stage HF and LV hypertrophy may deteriorate by
limiting, and therefore the myocardium primarily uses later stages of HF,39,40 potentially mediated by the devel-
FAO and secondarily glucose oxidation. A formal rec- opment of insulin resistance or other unknown mecha-
ognition of the substrate hierarchy of mitochondrial nisms.41 Myocardial tissue obtained at the time of LV
Downloaded from http://ahajournals.org by on June 2, 2020

fuel selection (FAO at the expense of glucose use) has assist device implantation demonstrated decreased
been termed the “Randle Effect.”25 In a like manner, transcript levels encoding enzymes involved in glucose
several studies have demonstrated preferential use of and pyruvate oxidation.42 These metabolic deficits in
ketones in normal hearts, such that, in the presence both FAO and glucose metabolism in HF conspire to
of increased ketones, both FA and glucose metabolism produce an energy-starved heart that is primed to use
appear to be reduced.26–29 alternative ATP-generating fuels.

KETONE METABOLISM IN HF Switch to Ketone Fuel Use


As the failing heart progressively loses its capacity to
Impaired FAO oxidize FA and glucose, ketone bodies provide a po-
The transition from normal myocardium to pathological tential alternative substrate because ketone oxidation
remodeling and ultimately HF consists of well-described bypasses the complex dysregulation of the β-oxidation
fuel metabolic alterations, including a reduction in ca- pathway and pyruvate dehydrogenase complex in HF.
pacity for, and rates of, FAO.30,31 Indeed, FAO impair- In fact, one recent study of patients with end-stage
ment is observed in early compensated cardiac hyper- HF found an abundance of a ketogenic derivative, β-
trophy32 and with more severe cardiac dysfunction.30 hydroxybutyryl-CoA, along with decreased BHB in myo-
A case-control lipidomic analysis of myocardial tissues cardial tissue in association with increased expression
from patients with end-stage HF showed significantly of ketolytic enzymes such as BDH1.3 Extending these
decreased myocardial concentrations of the majority of findings, a mouse model of progressive HF, induced
lipids, including long-chain acylcarnitines.3 Analogous by TAC and distal coronary ligation (TAC/myocardial
substrate shifts can occur rapidly during the induction infarction), likewise demonstrated increased expres-
of HF in animal models. Acute pressure overload mod- sion of BDH1 along with increased expression of genes
els of HF (TAC) engender a prompt switch from FAO encoding ketone body transporters (MCT1 and MCT2)
to glucose use,33,34 and deactivation of the upstream and increased 13C-BHB use on the basis of nuclear
transcriptional regulator peroxisome proliferator-acti- magnetic resonance studies.5 Thus, despite well-known
vated receptor alpha has been implicated as a regulator mitochondrial dysfunction that occurs during the

Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033 June 2, 2020 1803


Selvaraj et al Therapeutic Ketosis in Heart Failure

progression to HF and in contrast with FAO, ketones diet, oral therapy, or intravenous infusion) and the tar-
can be completely oxidized given that the metabolic get population, as well, may account for distinctions
STATE OF THE ART

pathway is upregulated in the failing heart and use can observed among these studies.
occur with a shorter number of steps.3,5 Reports that
cardiac-specific BDH1 knockout mice exhibit more se-
vere ventricular modeling and dysfunction after TAC/ Endogenous Ketosis in Healthy Humans
myocardial infarction, whereas BDH1 overexpression In humans, the optimal mode to increase systemic
attenuates cardiac remodeling and DNA damage in a ketone levels is unclear. The ketogenic diet (KD) is in-
pressure-overloaded model43 suggest that increased ke- tended to mimic the biochemical effects of starvation
tone use is adaptive.7 and consists of high-fat with low-carbohydrate intake,
a regimen that can engender difficulty with long-term
adherence. The KD lowers blood glucose (through re-
Ketones as a Biomarker in HF duced glucose availability and lower gluconeogenesis)
Several studies have reported increases in peripheral and elevates FA concentrations. Endogenous ketosis
ketone levels in HF.44,45 Exhaled acetone, for example, occurs in states of low insulin and elevated glucagon
was able to identify patients who have HF with similar levels that, in turn, drive lipolysis, releasing FAs as a
predictive value to B-type natriuretic peptide, and ex- substrate for hepatic ketogenesis. The lipolytic compo-
haled acetone levels positively correlated to New York nent of the KD forms part of the basis of its popular-
Heart Association class.45 Among 45 patients with pre- ity as a weight loss regimen.58 KDs typically result in
dominantly systolic HF, higher circulating levels of ke- mild to moderate ketosis with BHB levels of ≈0.5 to 1.5
tones were associated with lower EF, worse New York mmol/L, which is currently considered an accepted defi-
Heart Association class, and elevated filling pressures.44 nition of ketosis (≥0.3 mmol/L).59 This level of ketosis
Increases in peripheral ketone levels do not them- may also reduce appetite.
selves demonstrate the mechanism of their presence, Classical KDs focus on augmenting intake of long-
because such an elevation could reflect a decrease in chain FAs (≈55%–75% of daily caloric intake) and sig-
use or an increase in production of ketones. In addi- nificantly reducing carbohydrate intake (5%–10% of
tion, increased ketone levels may be physiological and daily caloric intake, typically <30–50 g per day). How-
compensatory in HF. In a related example, natriuretic ever, an alternative diet consisting of medium-chain
peptides are elevated in HF and strongly prognostic
Downloaded from http://ahajournals.org by on June 2, 2020

triglycerides (with corresponding FAs of 6–12 carbon


for adverse cardiovascular events, although they serve atoms) has been used to allow greater carbohydrate
necessary physiological roles in natriuresis and sympa- intake. Shorter FAs can rapidly generate ketones and
thetic inhibition in HF. Accordingly, although elevated have been studied in epilepsy, inherited disorders of
levels of ketone track with HF diagnosis and severity, metabolism, and diabetes mellitus.60 Potential benefits
this observation does not necessarily indicate that el- of medium-chain triglycerides include modest weight
evated ketones are maladaptive. loss and favorable changes in body composition in
comparison with long-chain triglycerides, with similar
Ketone Metabolism in HFpEF versus effects on lipids.61
Limited data are available regarding the cardiovascu-
HFrEF lar effects of the KD. Short-term KD induces a decrease
In an investigation using arterial and coronary sinus in high-energy phosphate metabolism by cardiac mag-
sampling of patients with aortic stenosis with LV hy- netic resonance imaging spectroscopy, without echo-
pertrophy and preserved LVEF, both ketones and FA cardiographic changes in function.62 It has been shown
uptake increased in comparison with controls, where- that increases in serum free fatty acids (FFAs) favor mi-
as only ketone uptake increased in patients with tochondrial uncoupling, potentially accounting for the
HFrEF.46 Thus, ketone uptake appears to be used as spectroscopic findings.63 Observational data further
metabolic fuel in both HFpEF and HFrEF. Studies as- suggest that increasing FFA concentration is associated
sessing peripheral blood ketone levels showed that with diastolic dysfunction.64
patients with HFrEF had lower,47 or similar,48 levels in Chronic use of a KD raises several other long-term
comparison to HFpEF. concerns. First, the KD can induce dyslipidemia with
increased low-density lipoprotein cholesterol, which
may be problematic in patients with, or prone to, ath-
EFFECTS OF THERAPEUTIC KETOSIS erosclerotic cardiovascular disease.58 Second, it reduces
As highlighted in the Table, recent studies have investi- peripheral glycogen stores, a strong determinant of ex-
gated the role of therapeutic ketosis in animal and hu- ercise capacity.65 In addition, there are concerns regard-
man studies in both normal and HF states with promis- ing anaplerosis and depletion of TCA intermediates
ing results. The method of inducing ketosis (ketogenic with the KD (depending on the strictness of conformity

1804 June 2, 2020 Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033


Selvaraj et al Therapeutic Ketosis in Heart Failure

Table.  Studies Relevant to Augmenting Ketosis in Human Heart Failure

STATE OF THE ART


First Author and Year Population Intervention Main Findings
Human studies
 Cox, 201649 8 endurance athletes Crossover study of ketone monoester Athletes after ingesting drink containing
and dextrose vs carbohydrate drink ketone monoester cycled on average 411 m
further over 30 min.
 Verma, 201650 10 patients with diabetes mellitus Before-and-after study with Empagliflozin reduced left ventricle mass and
and established cardiovascular empagliflozin improved lateral E′ velocity.
disease
 Gormsen, 201751 8 healthy subjects Randomized, crossover trial of Ketone infusion decreased myocardial glucose
sodium-3-OHB infusion vs saline uptake and increased myocardial blood flow
by 75%.
 Nielsen, 20196 16 patients with heart failure with Randomized, crossover trial of 3-OHB 3-OHB infusion increased cardiac output,
reduced ejection fraction infusion vs isotonic saline reduced systemic and pulmonary vascular
resistance, and marginally decreased
biventricular filling pressures.
 McMurray, 20199 4744 patients with heart failure with Randomized, crossover trial of Dapagliflozin reduced risk of worsening heart
reduced ejection fraction dapagliflozin vs placebo failure or cardiovascular death, irrespective of
underlying diabetes mellitus.
Animal studies
 Joubert, 201752 Lipodystrophic (seipin knockout) Dapagliflozin only Dapagliflozin treatment significantly improved
mice diastolic function in the hypertrophic heart.
 Lee, 201753 Normoglycemic male Wistar rats Dapagliflozin therapy and control Dapagliflozin reduced cardiac fibrosis.
after coronary ligation arm
 Verma, 201854 Mice with diabetes mellitus Empagliflozin and control arms Empagliflozin increased cardiac ATP
production, but not through ketone oxidation.
 Abdurrachim, 201955 Rats with obesity, diabetes mellitus, Empagliflozin and control arms Empagliflozin lowered myocardial ketone
hypertension, and heart failure utilization; no effect observed on left
ventricular hypertrophy or fibrosis.
 Horton, 20197 Transaortic constriction/myocardial Ketogenic and normal chow for Ketogenic chow improved left ventricle
infarction mice and tachycardia- mice; 3-OHB infusion vs no infusion remodeling in mice; 3-OHB infusion
Downloaded from http://ahajournals.org by on June 2, 2020

induced cardiomyopathy dogs in dogs with heart failure significantly improved systolic dysfunction and
left ventricle remodeling in the canine model.
 Santos-Gallego, 201956 Heart failure after left anterior Empagliflozin vs control Empagliflozin improved systolic function and
descending artery ligation in cardiac remodeling. Empagliflozin-treated pigs
nondiabetic pigs switched substrate utilization to ketones, fatty
acids, and branched-chain amino acids.
 Yurista, 201957 Rat models without diabetes mellitus Empagliflozin and control chow arms Empagliflozin significantly improved ejection
after myocardial infarction fraction, attenuated hypertrophy, diminished
fibrosis, and reduced oxidative stress after
myocardial infarction. Empagliflozin increased
myocardial expression of the ketone body
transporters and ketogenic enzymes.

3-OHB indicates 3-hydroxybutyrate.

to the low-carbohydrate portion of the diet), and it Effects of Ketone Supplementation in


is important to investigate whether anaplerotic sub- Healthy Humans
strates should be incorporated into diets. Last, popula-
tion-level studies suggest that low-carbohydrate diets Exogenous delivery of ketones induces a unique
are associated with increased mortality, albeit with ef- state, because ketosis can be achieved in the pres-
fect modification by the source of fat or protein used ence of normal carbohydrate and FA levels. Ketone
to supplement the diet.66 Thus, endogenous ketosis supplementation demonstrates consistent improve-
by dietary modification may not be an ideal long-term ments in glucose control and reduced circulating
solution to maintain ketosis. Future studies of the KD FFAs, as well. Intravenous infusions of sodium BHB
should elucidate the long-term atherosclerotic cardio- (5 mmol·kg–1·h–1 for 1.5 hours67 and 4.7/9.4/18.8
vascular consequences, the impact of supplementation μmol·kg–1·min–1 each for 1 hour68) significantly reduc-
with medium-chain triglycerides and anaplerotic sub- es glucose and FFA concentration. The latter regimen
strates, and the effect on myocardial metabolism by has achieved BHB levels of ≈0.5 to 2 mmol/L.68 Glu-
using cardiac magnetic resonance imaging and nuclear cose reduction is probably related to limiting hepatic
metabolic tracers. gluconeogenesis and increasing peripheral glucose

Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033 June 2, 2020 1805


Selvaraj et al Therapeutic Ketosis in Heart Failure

uptake because it is not mediated by pancreatic Effects of Therapeutic Ketosis in Animals


stimulation.67 FFA reduction may be attributable to With Cardiovascular Disease
STATE OF THE ART

exogenous ketones negatively feeding back on their


own production through BHB-mediated agonism of Models of therapeutic ketosis in animals tend to support
the PUMA-G receptor in adipose tissue, ultimately a cardioprotective effect following ischemic-reperfusion
suppressing lipolysis.69 injury. Ketonemia induced by 3-day fasting in mice re-
The myocardial effects of relatively high doses of duced myocardial damage and arrhythmias.76 However,
exogenous therapy (to a BHB level of nearly 4 mmol/L), studies of KD after ischemia-reperfusion conflict as to
are substantial under carefully controlled circumstanc- whether the dietary intervention is protective77 or in-
es. One study demonstrated an increase in heart rate, creases myocardial injury.76 Furthermore, the possible
an increase in pulse pressure (mediated by lowering protective effect of KD may be enhanced by exogenous
of diastolic blood pressure), a 75% increase myocar- ketone therapy: fasting in addition to ketone infusion
dial blood flow, and near halving of glucose uptake.51 led to the least myocardial damage relative to fasting
There was little change in FFA, insulin, and glucose without ketone infusion or a fed state with ketone in-
levels, although these assessments were made in the fusion in rat hearts after coronary occlusion.78 In hyper-
presence of a hyperinsulinemic-euglycemic clamp, tensive heart disease models, low-carbohydrate, high-
which may complicate the assessment. The mecha- fat (ketogenic) diets in Dahl salt-sensitive hypertensive
nism of the increased heart rate is not clear but may rats and rats exposed to pressure-overload banding
be a compensatory response to vasodilation rather models results in attenuated ventricular remodeling
than a sympathetic response, which is typically down- and dysfunction in comparison with high-carbohydrate
regulated by ketones.70 and low-fat diets.79–81
Recognizing that intravenous infusion of ketones is Increased myocardial delivery of ketone bodies has
not practical for therapeutic ketosis among outpatients, been shown to ameliorate HF in mice and in large ani-
different formulations of oral ketone therapy have been mals. A KD that doubled levels of ketones led to signifi-
studied, including ketone salts and ketone esters. Un- cant reduction in ventricular remodeling caused by TAC/
fortunately, oral ketone salts result in a very high so- myocardial infarction in wild-type mice in comparison
dium intake and half the plasma concentration of the with mice fed normal chow.7 A complementary study
d-stereoisomer of BHB in comparison with a ketone in a canine tachypacing HF model demonstrated that a
ester. The differences in isomers are important to con- 2.5-fold increase in plasma ketones achieved through
Downloaded from http://ahajournals.org by on June 2, 2020

sider: the l-isoform is much less readily oxidized, is more continuous delivery of ketones to the right ventricle
likely to accumulate, and is less well studied regarding markedly reduces cardiac dilatation and reduced cardi-
its signaling effects.71 One ketone monoester, (R)-3-hy- ac output along with blunting decreases in LVEF in com-
droxybutyl (R)-3-hydroxybutyrate, has been studied for parison with tachypacing and vehicle infusion.7 BHB de-
the purposes of therapeutic exogenous ketosis.65,71,72 livery also decreases total peripheral resistance, raising
This compound produces a rapid rise of blood levels of the question of whether ketones are direct vasodilators
BHB in humans to nearly 3 mmol/L within 60 minutes of or act indirectly such as through lowering sympathetic
consumption with 282 mg/kg ketone ester.71 No signifi- tone.70 In canines, BHB infusion reduces glucose oxida-
cant cardiovascular effects have been reported at rest in tion and lactate production, without altering FFA oxida-
healthy volunteers.72 tion, consistent with a Randle effect (increased acetyl-
In contrast with ketone salts,73 benefits of ketone CoA levels decrease pyruvate dehydrogenase activity).25
ester therapy among athletes have been demon- The corresponding decrease in glucose oxidation could
strated during exercise. Early studies indicate that be beneficial in cardiometabolic disease states to limit
athletes, in particular, appear to undergo meta- glucotoxicity. Although ketone infusion augmented
bolic reprogramming that results in favorable and myocardial oxygen consumption, myocardial mechanic
matched ketone use during exercise.74,75 In a recent energetic efficiency actually improved, implying that the
blinded, crossover study of endurance athletes, ke- increased oxygen consumption was not pathological.
tone ester therapy increased exercise distance by The direct impact of ketone bodies on cardiac ener-
an average of 411 m over 30 minutes cycling du- getics (phosphocreatine/ATP ratio) in the failing heart
ration, demonstrating that ketone therapy improves is not established. With that said, data suggest that
exercise capacity among highly trained athletes, al- infused ketones are oxidized, along with improvement
though a ceiling effect may be reached in that popu- in cardiac function, in the canine tachypacing model
lation.49 However, pathological conditions that cause of HF.7 Moreover, infusion of ketones competes with
metabolic dysregulation, and where incremental im- glucose oxidation in this setting. Last, mitochondria iso-
provements in energy transduction may translate to lated from the normal mouse heart exhibit increased
increases in exercise capacity, such as HF, may afford NADH/NAD levels and enhanced defense of the mi-
the greatest clinical benefit.49 tochondrial membrane potential when supplemented

1806 June 2, 2020 Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033


Selvaraj et al Therapeutic Ketosis in Heart Failure

with ketones in the setting of limited supply of FAs to long-term benefits because of concerns related to the
mimic the substrate use of the failing heart.7 high sodium load, increased heart rate (which may be

STATE OF THE ART


Although the energetic properties of ketones are detrimental in HFrEF), and depletion of TCA intermedi-
important, it is not clear that efficiency as defined by ates. Furthemore, the small sample size precludes fur-
phosphate/oxygen ratio accounts for their beneficial ef- ther assessment of subgroups that might differentially
fect in HF. In mice, ketone infusion significantly increas- respond to ketone infusions (eg, patients with diabetes
es the failing heart’s use of ketones (to >20% from mellitus). Last, understanding how the degree of mi-
<10%), but cardiac efficiency fails to improve and is tochondrial dysfunction affects the response to ketone
similar to FA metabolic efficiency.12 Nevertheless, stud- infusion is an important clinical consideration.
ies using isolated mitochondria in the context of limit- The effects of ketone supplementation in HFpEF
ing levels of FA input into the TCA cycle report that BHB have not yet been assessed. However, because many of
maintains the mitochondrial membrane gradient to a the control patients in the aforementioned study had
greater extent than other substrates.7 Thus, the respira- hypertension (a common comorbidity in those with HF-
tory efficiency may be improved by ketone bodies. Fu- pEF) and achieved similar hemodynamic findings with
ture animal studies should investigate the relevance of ketone infusion to HFrEF, the same benefit may be seen
exogenous ketosis in acute versus chronic models of HF, in HFpEF. It is also tempting to speculate that improve-
and in HFpEF, as well, given that these studies largely ment in systolic strain82 or vasodilatory effect of ketone
investigate models of HFrEF. therapy83 could be beneficial in patients with HFpEF.
Thus, ketone supplementation is therapeutically
promising in human HF. However, questions about the
Effects of Ketone Supplementation in mechanism of benefit, impact on peripheral muscles
Human HF and glycogen content, relevance in end-stage HF, and
Limited data in humans suggest that ketone supple- the effect of anaplerotic substrate supplementation
mentation has therapeutic potential in HF. In a random- are important to investigate further.
ized, crossover study in patients with compensated
HFrEF, a 3-hour infusion of a racemic mixture of 7.5%
3-hydroxybutyrate (at 0.18 g·kg–1·h–1) achieved serum
Nonmetabolic Benefits of Ketone Bodies
levels of 3.3 mmol/L and increased cardiac output by The benefits observed with ketone therapy in the
aforementioned studies might also relate to the pleio-
Downloaded from http://ahajournals.org by on June 2, 2020

40% in association with decreases in systemic and pul-


monary vascular resistances, a modest increase in heart tropic roles of ketone bodies in cellular signaling and
rate and stroke volume, and mildly reduced cardiac modulating oxidative stress and inflammation, which
filling pressures, which were not observed with saline recently have been comprehensively reviewed.13 In
infusion.6 These hemodynamic changes were dose de- particular, ketone bodies regulate both lipolysis and
pendent, and cardiac effects were seen with even mild sympathetic activity through their interactions with
ketosis. Laboratory analysis revealed little difference in specific G-protein–coupled receptors. For example,
insulin levels, B-type natriuretic peptide, or norepineph- BHB binds G-protein–coupled receptor GPR109A and
rine levels. Myocardial external efficiency (defined as provides negative feedback to lipolysis in adipocytes
the ratio between stroke work and myocardial oxygen through hormone-sensitive triglyceride lipase.69 In ad-
consumption) was unchanged with ketone infusion in dition, ketones interact with GPR41 to suppress sym-
comparison with isotonic saline. Similar hemodynamic pathetic nervous system activity in the sympathetic
effects of the ketone infusion were observed in an age- ganglion.70 This latter effect may be particularly rel-
matched control group who were largely hypertensive. evant in HF, given the benefits of sympathetic antago-
Although important, this study generates several nism. In addition, BHB has anti-inflammatory activity
additional questions and considerations. A key un- by suppressing activation of the NLRP3 (Nod-like re-
answered question with this study is whether the in- ceptor protein 3) inflammasome.84 However, the in-
creases in cardiac output were solely attributable to flammatory response may not be uniform among ke-
peripheral vasodilation or to the use of ketones as a tones, with studies supporting both proinflammatory
metabolic substrate. Given that vasodilators are already and anti-inflammatory effects of acetoacetate.85,86 Fur-
a mainstay of HFrEF treatment, clarifying this distinc- thermore, ketone bodies regulate FGF21 (fibroblastic
tion is key to understanding whether exogenous ke- growth factor 21), which plays an important paracrine
tones add unique benefits in HF. The comparison with role in the heart and favorably influences myocar-
normal saline, as opposed to an alternative energetic dial remodeling, because Fgf21−/− mice demonstrate
fuel, likewise leaves unanswered questions about how greater cardiac hypertrophy and more deleterious re-
the ketone infusion affected hemodynamics. In ad- sponses to isoproterenol infusion.87,88 Last, the role of
dition, favorable short-term effects (because the ke- ketones in regulating appetite and promoting satiety
tone washes out very quickly)6 may not translate to are complex and in need of further study. Although

Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033 June 2, 2020 1807


Selvaraj et al Therapeutic Ketosis in Heart Failure

speculative, the differential milieu of endogenous ver- neurohormonal pathways, is critical. Further defining
sus exogenous ketosis may be important in evoking a these pathways in HFpEF versus HFrEF and method of
STATE OF THE ART

specific response. Because endogenous ketosis often ketosis (exogenous versus endogenous) will be neces-
occurs during sympathetic states, the antilipolytic, an- sary in advancing the treatment paradigm.
tihyperlipidemic, antisympathetic, and anti-inflamma-
tory effects may be muted. Exogenous ketosis, how-
ever, might uniquely elicit these responses without
Relationship Between SGLT2 Inhibitors,
requiring the sympathetic state to promote ketosis. Systemic Ketosis, and HF
Epigenetic effects of BHB have also been observed. Very recently, administration of SGLT2 inhibitors as a
Ketone bodies inhibit class I histone deacetylases primary HF therapeutic demonstrated a striking re-
and increase the activity of 2 promoters involved duction in HF hospitalization and all-cause mortality.9
in protecting against oxidative stress (FOXO3A and The mechanisms responsible for the salutary impact
MT2).89,90 Increased histone acetylation was observed of SGLT2 inhibitors in HF are unknown. SGLT2 inhibi-
both with endogenous and exogenous means of tors induce systemic ketosis, but it is currently unclear
inducing ketosis.90 These globally beneficial ketotic whether this is a mechanism of benefit.8 Because
effects may be relevant to reversing the pathophysi- SGLT2 receptors are not located in cardiomyocytes, in-
ological remodeling induced by reactive oxygen spe- direct mechanisms, including ketosis, are likely to ac-
cies that contribute directly and indirectly to wors- count for the benefit.93 SGLT2 inhibitors decrease the
ening HF.91 A ketone-derived histone modification, insulin-to-glucagon ratio and increase lipolysis, both
β-hydroxybutyrylation, is another recently discovered of which favor the formation of ketones (Figure  1).94
epigenetic regulatory mark, although its significance In fact, empagliflozin treatment mildly increases BHB
needs further elucidation.92 levels (0.56 mmol/L in patients with diabetes mellitus,
Thus, the relevance of therapeutic ketosis in HF goes 0.27 mmol/L in patients without diabetes mellitus)
significantly beyond ATP generation. Understanding with chronic dosing (28 days).95 Because SGLT2 in-
the nonmetabolic benefits of ketone bodies in HF, such hibitor therapy increases ketone metabolism in animal
as the effects on inflammatory, oxidative stress, and models of HF,56,57 increased circulating levels of ketones
Downloaded from http://ahajournals.org by on June 2, 2020

Figure 2. Methods of achieving ketosis and related potential cardiovascular benefits.


Popular, nondietary ways of achieving ketosis include oral ketone ester drinks, intravenous ketone salt infusions, and SGLT2 inhibitors. Reported cardiovascular
benefits related to the ketosis achieved are listed underneath with the corresponding patient population studied. Note that the role of ketosis as a mediator of the
cardiovascular benefits observed with SGLT-2 inhibitors is speculative at this point. CV indicates cardiovascular; FA, fatty acid; HF, heart failure; LV, left ventricular;
PVR, pulmonary vascular resistance; SGLT2, sodium-glucose cotransporter 2; and SVR, systemic vascular resistance.

1808 June 2, 2020 Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033


Selvaraj et al Therapeutic Ketosis in Heart Failure

may plausibly increase myocardial use of ketones. In- ARTICLE INFORMATION


deed, the systemic ketosis achieved with SGLT2 inhibi-

STATE OF THE ART


Correspondence
tors that decrease HF hospitalization is similar to levels Kenneth B. Margulies, MD, Perelman School of Medicine, University of Pennsyl-
shown to be beneficial in HF.6 vania, Smilow Center for Translational Research, 3400 Civic Center Boulevard,
11-101, Philadelphia, PA 19104. Email kenb@pennmedicine.upenn.edu
Apart from ketosis, SGLT2 therapy may have other
cardioprotective effects in HF. SGLT2 inhibitors may en-
Affiliations
gender favorable changes in blood pressure, weight
Division of Cardiovascular Medicine, Department of Medicine (S.S., K.B.M.),
loss, uricosuria, volume control through osmotic diure- Cardiovascular Institute and Department of Medicine (D.P.K., K.B.M.), Heart
sis, natriuresis, and oxygen-carrying capacity with he- Failure and Transplant Program, Smilow Center for Translational Research
moconcentration.93 In animal and human hearts with (K.B.M.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.

pathological hypertrophy or diabetes mellitus, treat-


ment with SGLT2 inhibitors improved diastolic function Acknowledgments
Images were created with the support of Biorender.
and reduced cardiac fibrosis, but whether these are
ketone-dependent benefits is unknown.50,52,53 Because
Sources of Funding
SGLT2 inhibitors are used to treat patients with HF, bet-
Dr Selvaraj is supported by the National Institutes of Health (Training Grant
ter understanding of whether ketosis is central to their 5-T32HL007843-23). Dr Kelly is supported by the NIH (NHLBI) R01 HL058493 and
mechanism of action will have implications for their tar- R01 HL128349. Dr Margulies is supported by the NIH (NHLBI) U10 HL110338.
get population, potential risks, use of dietary adjuvants,
and safety monitoring. Disclosures
Dr Kelly reports a significant consulting relationship with Pfizer and a modest
consulting relationship with Amgen. Dr Margulies serves as a consultant for

CONCLUSIONS MyoKardia and Pfizer and receives research funding from Sanofi-Aventis and
GlaxoSmithKline. Dr Selvaraj receives research funding from the American So-
Emerging evidence suggests that therapeutic ketosis ciety of Nuclear Cardiology related to ketone esters and the ketogenic diet. No
other relevant disclosures are reported.
may be a viable treatment target in patients with HF
(Figure 2). Striking short-term cardiovascular benefits
observed with exogenous ketosis highlight the ke- REFERENCES
tone metabolic axis as a promising frontier in novel 1. Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW,
HF treatment. Interest in capitalizing on the unique Carson AP, Chamberlain AM, Chang AR, Cheng S, Das SR, et al; Ameri-
Downloaded from http://ahajournals.org by on June 2, 2020

can Heart Association Council on Epidemiology and Prevention Statistics


metabolic effects of ketone use is growing but begs Committee and Stroke Statistics Subcommittee. Heart disease and stroke
for greater understanding of the mechanisms through statistics-2019 update: a report from the American Heart Association. Cir-
culation. 2019;139:e56–e528. doi: 10.1161/CIR.0000000000000659
which ketones alter cardiovascular function. For ex-
2. Neubauer S. The failing heart–an engine out of fuel. N Engl J Med.
ample, do ketone bodies work as alternate cardiac fu- 2007;356:1140–1151. doi: 10.1056/NEJMra063052
els, vasodilators, or via other signaling pathways? Will 3. Bedi KC Jr, Snyder NW, Brandimarto J, Aziz M, Mesaros C, Worth AJ,
Wang LL, Javaheri A, Blair IA, Margulies KB, et al. Evidence for intramyo-
they prove useful for both chronic and acute (boosting cardial disruption of lipid metabolism and increased myocardial ketone
short-term activity tolerance or in decompensated HF) utilization in advanced human heart failure. Circulation. 2016;133:706–
indications? Could peripheral effects of ketosis con- 716. doi: 10.1161/CIRCULATIONAHA.115.017545
4. De Jong KA, Lopaschuk GD. Complex energy metabolic changes in
tribute to improved outcomes in cardiomyopathies? heart failure with preserved ejection fraction and heart failure with
How does ketone body supplementation interact with reduced ejection fraction. Can J Cardiol. 2017;33:860–871. doi:
10.1016/j.cjca.2017.03.009
myocardial branched-chain amino acid metabolism (a
5. Aubert G, Martin OJ, Horton JL, Lai L, Vega RB, Leone TC,
recently discovered pathway in HF genesis)? Several Koves T, Gardell SJ, Krüger M, Hoppel CL, et al. The failing heart re-
other questions remain to be answered. Future stud- lies on ketone bodies as a fuel. Circulation. 2016;133:698–705. doi:
10.1161/CIRCULATIONAHA.115.017355
ies should seek to clarify efficacy and safety (particu- 6. Nielsen R, Møller N, Gormsen LC, Tolbod LP, Hansson NH,
larly hepatic effects and acid-based disturbances) of Sorensen J, Harms HJ, Frøkiær J, Eiskjaer H, Jespersen NR, et al. Cardio-
long-term ketone administration, optimal methods of vascular effects of treatment with the ketone body 3-hydroxybutyrate
in chronic heart failure patients. Circulation. 2019;139:2129–2141. doi:
achieving ketosis in HF (oral versus intravenous ad- 10.1161/CIRCULATIONAHA.118.036459
ministration), effect on patient-centered outcomes, 7. Horton JL, Davidson MT, Kurishima C, Vega RB, Powers JC, Matsuura TR,
Petucci C, Lewandowski ED, Crawford PA, Muoio DM, et al. The failing
and differential effects in HFrEF and HFpEF. For ex-
heart utilizes 3-hydroxybutyrate as a metabolic stress defense. JCI Insight.
ample, peripheral glycogen-preserving effects may 2019;4: e124079.
be important to both subsets of HF, although they 8. Ferrannini E, Mark M, Mayoux E. CV protection in the EMPA-REG
OUTCOME trial: a “thrifty substrate” hypothesis. Diabetes Care.
may be particularly relevant to patients with HFpEF 2016;39:1108–1114. doi: 10.2337/dc16-0330
in whom improvements in systolic function might be 9. McMurray JJV, Solomon SD, Inzucchi SE, Køber L, Kosiborod MN,
less relevant. It is an exciting time to investigate the Martinez FA, Ponikowski P, Sabatine MS, Anand IS, Bělohlávek J, et
al; DAPA-HF Trial Committees and Investigators. Dapagliflozin in pa-
potential of ketone metabolic modulation as a thera- tients with heart failure and reduced ejection fraction. N Engl J Med.
peutic strategy for HF. 2019;381:1995–2008. doi: 10.1056/NEJMoa1911303

Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033 June 2, 2020 1809


Selvaraj et al Therapeutic Ketosis in Heart Failure

10. Taegtmeyer H, Young ME, Lopaschuk GD, Abel ED, 31. Aubert G, Vega RB, Kelly DP. Perturbations in the gene regulatory pathways
Brunengraber H, Darley-Usmar V, Des Rosiers C, Gerszten R, Glatz JF, controlling mitochondrial energy production in the failing heart. Biochim
STATE OF THE ART

Griffin JL, et al; American Heart Association Council on Basic Cardiovas- Biophys Acta. 2013;1833:840–847. doi: 10.1016/j.bbamcr.2012.08.015
cular Sciences. Assessing cardiac metabolism: a scientific statement from 32. Akki A, Smith K, Seymour AM. Compensated cardiac hypertrophy is
the American Heart Association. Circ Res. 2016;118:1659–1701. doi: characterised by a decline in palmitate oxidation. Mol Cell Biochem.
10.1161/RES.0000000000000097 2008;311:215–224. doi: 10.1007/s11010-008-9711-y
11. McGarrah RW, Crown SB, Zhang GF, Shah SH, Newgard CB. Cardio- 33. Barger PM, Brandt JM, Leone TC, Weinheimer CJ, Kelly DP. Deactivation of
vascular metabolomics. Circ Res. 2018;122:1238–1258. doi: 10.1161/ peroxisome proliferator-activated receptor-alpha during cardiac hypertro-
CIRCRESAHA.117.311002 phic growth. J Clin Invest. 2000;105:1723–1730. doi: 10.1172/JCI9056
12. Ho KL, Zhang L, Wagg C, Al Batran R, Gopal K, Levasseur J, Leone T, 34. Sack MN, Disch DL, Rockman HA, Kelly DP. A role for Sp and nuclear recep-
Dyck JRB, Ussher JR, Muoio DM, et al. Increased ketone body oxidation tor transcription factors in a cardiac hypertrophic growth program. Proc
provides additional energy for the failing heart without improving cardiac Natl Acad Sci U S A. 1997;94:6438–6443. doi: 10.1073/pnas.94.12.6438
efficiency. Cardiovasc Res. 2019;115:1606–1616. doi: 10.1093/cvr/cvz045 35. Dávila-Román VG, Vedala G, Herrero P, de las Fuentes L, Rogers JG,
13. Puchalska P, Crawford PA. Multi-dimensional roles of ketone bodies in fuel Kelly DP, Gropler RJ. Altered myocardial fatty acid and glucose metabolism
metabolism, signaling, and therapeutics. Cell Metab. 2017;25:262–284. in idiopathic dilated cardiomyopathy. J Am Coll Cardiol. 2002;40:271–
doi: 10.1016/j.cmet.2016.12.022 277. doi: 10.1016/s0735-1097(02)01967-8
14. Thomas LK, Ittmann M, Cooper C. The role of leucine in ketogenesis in 36. Allard MF, Schönekess BO, Henning SL, English DR, Lopaschuk GD. Con-
starved rats. Biochem J. 1982;204:399–403. doi: 10.1042/bj2040399 tribution of oxidative metabolism and glycolysis to ATP production in
15. Zhang D, Yang H, Kong X, Wang K, Mao X, Yan X, Wang Y, Liu S, Zhang X, hypertrophied hearts. Am J Physiol. 1994;267(2 pt 2):H742–H750. doi:
Li J, et al. Proteomics analysis reveals diabetic kidney as a ketogenic organ 10.1152/ajpheart.1994.267.2.H742
in type 2 diabetes. Am J Physiol Endocrinol Metab. 2011;300:E287–E295. 37. Leong HS, Grist M, Parsons H, Wambolt RB, Lopaschuk GD,
doi: 10.1152/ajpendo.00308.2010 Brownsey R, Allard MF. Accelerated rates of glycolysis in the hypertrophied
16. Cahill GF Jr, Herrera MG, Morgan AP, Soeldner JS, Steinke J, Levy PL, heart: are they a methodological artifact? Am J Physiol Endocrinol Metab.
Reichard GA Jr, Kipnis DM. Hormone-fuel interrelationships during fast- 2002;282:E1039–E1045. doi: 10.1152/ajpendo.00507.2001
ing. J Clin Invest. 1966;45:1751–1769. doi: 10.1172/JCI105481 38. Nascimben L, Ingwall JS, Lorell BH, Pinz I, Schultz V, Tornheim K, Tian R.
17. van Hasselt PM, Ferdinandusse S, Monroe GR, Ruiter JP, Turkenburg M, Mechanisms for increased glycolysis in the hypertrophied rat heart. Hyper-
Geerlings MJ, Duran K, Harakalova M, van der Zwaag B, Monavari AA, tension. 2004;44:662–667. doi: 10.1161/01.HYP.0000144292.69599.0c
et al. Monocarboxylate transporter 1 deficiency and ketone utilization. N 39. Zhabyeyev P, Gandhi M, Mori J, Basu R, Kassiri Z, Clanachan A,
Engl J Med. 2014;371:1900–1907. doi: 10.1056/NEJMoa1407778 Lopaschuk GD, Oudit GY. Pressure-overload-induced heart failure induces
18. Robinson AM, Williamson DH. Physiological roles of ketone bodies as sub- a selective reduction in glucose oxidation at physiological afterload. Car-
strates and signals in mammalian tissues. Physiol Rev. 1980;60:143–187. diovasc Res. 2013;97:676–685. doi: 10.1093/cvr/cvs424
doi: 10.1152/physrev.1980.60.1.143 40. Schroeder MA, Lau AZ, Chen AP, Gu Y, Nagendran J, Barry J, Hu X, Dyck JR,
19. Zorzano A, Balon TW, Brady LJ, Rivera P, Garetto LP, Young JC, Tyler DJ, Clarke K, et al. Hyperpolarized 13C magnetic resonance reveals
Goodman MN, Ruderman NB. Effects of starvation and exercise on con- early- and late-onset changes to in vivo pyruvate metabolism in the failing
centrations of citrate, hexose phosphates and glycogen in skeletal muscle heart. Eur J Heart Fail. 2013;15:130–140. doi: 10.1093/eurjhf/hfs192
and heart. Evidence for selective operation of the glucose-fatty acid cycle. 41. Riehle C, Abel ED. Insulin signaling and heart failure. Circ Res.
Biochem J. 1985;232:585–591. doi: 10.1042/bj2320585 2016;118:1151–1169. doi: 10.1161/CIRCRESAHA.116.306206
20. Russell RR 3rd, Taegtmeyer H. Changes in citric acid cycle flux and anaple- 42. Gupte AA, Hamilton DJ, Cordero-Reyes AM, Youker KA, Yin Z, Estep JD,
Downloaded from http://ahajournals.org by on June 2, 2020

rosis antedate the functional decline in isolated rat hearts utilizing aceto- Stevens RD, Wenner B, Ilkayeva O, Loebe M, et al. Mechanical unloading
acetate. J Clin Invest. 1991;87:384–390. doi: 10.1172/JCI115008 promotes myocardial energy recovery in human heart failure. Circ Cardio-
21. Taegtmeyer H. On the inability of ketone bodies to serve as the only en- vasc Genet. 2014;7:266–276. doi: 10.1161/CIRCGENETICS.113.000404
ergy providing substrate for rat heart at physiological work load. Basic Res 43. Uchihashi M, Hoshino A, Okawa Y, Ariyoshi M, Kaimoto S, Tateishi S,
Cardiol. 1983;78:435–450. doi: 10.1007/bf02070167 Ono K, Yamanaka R, Hato D, Fushimura Y, et al. Cardiac-specific bdh1
22. Russell RR 3rd, Taegtmeyer H. Pyruvate carboxylation prevents the de- overexpression ameliorates oxidative stress and cardiac remodeling in pres-
cline in contractile function of rat hearts oxidizing acetoacetate. Am sure overload-induced heart failure. Circ Heart Fail. 2017;10: e004417.
J Physiol. 1991;261(6 pt 2):H1756–H1762. doi: 10.1152/ajpheart. 44. Lommi J, Kupari M, Koskinen P, Näveri H, Leinonen H, Pulkki K,
1991.261.6.H1756 Härkönen M. Blood ketone bodies in congestive heart failure. J Am Coll
23. Schugar RC, Moll AR, André d’Avignon D, Weinheimer CJ, Cardiol. 1996;28:665–672. doi: 10.1016/0735-1097(96)00214-8
Kovacs A, Crawford PA. Cardiomyocyte-specific deficiency of ketone body 45. Marcondes-Braga FG, Gutz IGR, Batista GL, Saldiva PHN, Ayub-
metabolism promotes accelerated pathological remodeling. Mol Metab. Ferreira SM, Issa VS, Mangini S, Bocchi EA, Bacal F. Exhaled acetone as a
2014;3:754–769. doi: 10.1016/j.molmet.2014.07.010 new biomarker of heart failure severity. Chest. 2012;142:457–466. doi:
24. Keon CA, Tuschiya N, Kashiwaya Y, Sato K, Clarke K, Radda GK, 10.1378/chest.11-2892
Veech RL. Substrate dependence of the mitochondrial energy status in 46. Voros G, Ector J, Garweg C, Droogne W, Van Cleemput J,
the isolated working rat heart. Biochem Soc Trans. 1995;23:307S. doi: Peersman N, Vermeersch P, Janssens S. Increased cardiac uptake of ke-
10.1042/bst023307s tone bodies and free fatty acids in human heart failure and hypertro-
25. Randle PJ, Garland PB, Hales CN, Newsholme EA. The glucose fatty-acid cy- phic left ventricular remodeling. Circ Heart Fail. 2018;11:e004953. doi:
cle. Its role in insulin sensitivity and the metabolic disturbances of diabetes 10.1161/CIRCHEARTFAILURE.118.004953
mellitus. Lancet. 1963;1:785–789. doi: 10.1016/s0140-6736(63)91500-9 47. Zordoky BN, Sung MM, Ezekowitz J, Mandal R, Han B, Bjorndahl TC,
26. Bassenge E, Wendt VE, Schollmeyer P, Bluemchen G, Gudbjarnason S, Bouatra S, Anderson T, Oudit GY, Wishart DS, et al; Alberta HEART. Me-
Bing RJ. Effect of ketone bodies on cardiac metabolism. Am J Physiol. tabolomic fingerprint of heart failure with preserved ejection fraction.
1965;208:162–168. doi: 10.1152/ajplegacy.1965.208.1.162 PLoS One. 2015;10:e0124844. doi: 10.1371/journal.pone.0124844
27. Jeffrey FM, Diczku V, Sherry AD, Malloy CR. Substrate selection in the 48. Hunter WG, Kelly JP, McGarrah RW 3rd, Khouri MG, Craig D, Haynes C,
isolated working rat heart: effects of reperfusion, afterload, and concen- Ilkayeva O, Stevens RD, Bain JR, Muehlbauer MJ, et al. Metabolomic pro-
tration. Basic Res Cardiol. 1995;90:388–396. doi: 10.1007/bf00788500 filing identifies novel circulating biomarkers of mitochondrial dysfunction
28. Stanley WC, Meadows SR, Kivilo KM, Roth BA, Lopaschuk GD. β- differentially elevated in heart failure with preserved versus reduced ejec-
Hydroxybutyrate inhibits myocardial fatty acid oxidation in vivo indepen- tion fraction: evidence for shared metabolic impairments in clinical heart
dent of changes in malonyl-CoA content. Am J Physiol Heart Circ Physiol. failure. J Am Heart Assoc. 2016;5:e003190.
2003;285:H1626–H1631. doi: 10.1152/ajpheart.00332.2003 49. Cox PJ, Kirk T, Ashmore T, Willerton K, Evans R, Smith A, Murray AJ,
29. Ziegler A, Zaugg CE, Buser PT, Seelig J, Künnecke B. Non-invasive mea- Stubbs B, West J, McLure SW, et al. Nutritional ketosis alters fuel pref-
surements of myocardial carbon metabolism using in vivo 13C NMR spec- erence and thereby endurance performance in athletes. Cell Metab.
troscopy. NMR Biomed. 2002;15:222–234. doi: 10.1002/nbm.764 2016;24:256–268. doi: 10.1016/j.cmet.2016.07.010
30. Sack MN, Rader TA, Park S, Bastin J, McCune SA, Kelly DP. Fatty acid 50. Verma S, Garg A, Yan AT, Gupta AK, Al-Omran M, Sabongui A, Teoh H,
oxidation enzyme gene expression is downregulated in the failing heart. Mazer CD, Connelly KA. Effect of empagliflozin on left ventricular mass
Circulation. 1996;94:2837–2842. doi: 10.1161/01.cir.94.11.2837 and diastolic function in individuals with diabetes: an important clue to

1810 June 2, 2020 Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033


Selvaraj et al Therapeutic Ketosis in Heart Failure

the EMPA-REG OUTCOME trial? Diabetes Care. 2016;39:e212–e213. doi: beta-hydroxybutyrate infusions in man. Diabetologia. 1968;4:133–135.
10.2337/dc16-1312 doi: 10.1007/bf01219433

STATE OF THE ART


51. Gormsen LC, Svart M, Thomsen HH, Sondergaard E, Vendelbo MH, 68. Mikkelsen KH, Seifert T, Secher NH, Grøndal T, van Hall G. Systemic, ce-
Christensen N, Tolbod LP, Harms HJ, Nielsen R, Wiggers H, et al. Ketone rebral and skeletal muscle ketone body and energy metabolism during
body infusion with 3-hydroxybutyrate reduces myocardial glucose uptake acute hyper-D-β-hydroxybutyratemia in post-absorptive healthy males. J
and increases blood flow in humans: a positron emission tomography Clin Endocrinol Metab. 2015;100:636–643. doi: 10.1210/jc.2014-2608
study. J Am Heart Assoc. 2017;6:e005066. 69. Taggart AK, Kero J, Gan X, Cai TQ, Cheng K, Ippolito M, Ren N, Kaplan R,
52. Joubert M, Jagu B, Montaigne D, Marechal X, Tesse A, Ayer A, Dollet L, Wu K, Wu TJ, et al. (D)-β-Hydroxybutyrate inhibits adipocyte lipolysis
Le May C, Toumaniantz G, Manrique A, et al. The sodium-glucose co- via the nicotinic acid receptor PUMA-G. J Biol Chem. 2005;280:26649–
transporter 2 inhibitor dapagliflozin prevents cardiomyopathy in a dia- 26652. doi: 10.1074/jbc.C500213200
betic lipodystrophic mouse model. Diabetes. 2017;66:1030–1040. doi: 70. Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S, Kobayashi M,
10.2337/db16-0733 Hirasawa A, Tsujimoto G. Short-chain fatty acids and ketones directly
53. Lee TM, Chang NC, Lin SZ. Dapagliflozin, a selective SGLT2 Inhibi- regulate sympathetic nervous system via G protein-coupled recep-
tor, attenuated cardiac fibrosis by regulating the macrophage polariza- tor 41 (GPR41). Proc Natl Acad Sci U S A. 2011;108:8030–8035. doi:
tion via STAT3 signaling in infarcted rat hearts. Free Radic Biol Med. 10.1073/pnas.1016088108
2017;104:298–310. doi: 10.1016/j.freeradbiomed.2017.01.035 71. Stubbs BJ, Cox PJ, Evans RD, Santer P, Miller JJ, Faull OK, Magor-Elliott S,
54. Verma S, Rawat S, Ho KL, Wagg CS, Zhang L, Teoh H, Dyck JE, Uddin GM, Hiyama S, Stirling M, Clarke K. On the metabolism of exogenous ketones
Oudit GY, Mayoux E, et al. Empagliflozin increases cardiac energy  pro- in humans. Front Physiol. 2017;8:848. doi: 10.3389/fphys.2017.00848
duction  in diabetes: novel translational insights into the heart failure 72. Clarke K, Tchabanenko K, Pawlosky R, Carter E, Todd King M,
benefits of SGLT2 inhibitors. JACC Basic Transl Sci. 2018;3:575–587. doi: Musa-Veloso K, Ho M, Roberts A, Robertson J, Vanitallie TB, et al. Kinet-
10.1016/j.jacbts.2018.07.006 ics, safety and tolerability of (R)-3-hydroxybutyl (R)-3-hydroxybutyrate in
55. Abdurrachim D, Teo XQ, Woo CC, Chan WX, Lalic J, Lam CSP, Lee PTH. healthy adult subjects. Regul Toxicol Pharmacol. 2012;63:401–408. doi:
Empagliflozin reduces myocardial ketone utilization while preserving 10.1016/j.yrtph.2012.04.008
glucose utilization in diabetic hypertensive heart disease: a hyperpolar- 73. O’Malley T, Myette-Cote E, Durrer C, Little JP. Nutritional ketone salts
ized 13C magnetic resonance spectroscopy study. Diabetes Obes Metab. increase fat oxidation but impair high-intensity exercise performance in
2019;21:357–365. doi: 10.1111/dom.13536 healthy adult males. Appl Physiol Nutr Metab. 2017;42:1031–1035. doi:
56. Santos-Gallego CG, Requena-Ibanez JA, San Antonio R, Ishikawa K, 10.1139/apnm-2016-0641
Watanabe S, Picatoste B, Flores E, Garcia-Ropero A, Sanz J, Hajjar RJ, et al. 74. Johnson RH, Walton JL, Krebs HA, Williamson DH. Metabolic fuels
Empagliflozin ameliorates adverse left ventricular remodeling in nondia- during and after severe exercise in athletes and non-athletes. Lancet.
betic heart failure by enhancing myocardial energetics. J Am Coll Cardiol. 1969;2:452–455. doi: 10.1016/s0140-6736(69)90164-0
2019;73:1931–1944. doi: 10.1016/j.jacc.2019.01.056 75. Johnson RH, Walton JL. The effect of exercise upon acetoacetate me-
57. Yurista SR, Silljé HHW, Oberdorf-Maass SU, Schouten EM, Pavez Giani MG, tabolism in athletes and non-athletes. Q J Exp Physiol Cogn Med Sci.
Hillebrands JL, van Goor H, van Veldhuisen DJ, de Boer RA, Westenbrink BD. 1972;57:73–79. doi: 10.1113/expphysiol.1972.sp002139
Sodium-glucose co-transporter 2 inhibition with empagliflozin improves 76. Wang P, Tate JM, Lloyd SG. Low carbohydrate diet decreases myocardial
cardiac function in non-diabetic rats with left ventricular dysfunction insulin signaling and increases susceptibility to myocardial ischemia. Life
after myocardial infarction. Eur J Heart Fail. 2019;21:862–873. doi: Sci. 2008;83:836–844. doi: 10.1016/j.lfs.2008.09.024
10.1002/ejhf.1473 77. Al-Zaid NS, Dashti HM, Mathew TC, Juggi JS. Low carbohydrate keto-
58. Nordmann AJ, Nordmann A, Briel M, Keller U, Yancy WS Jr, Brehm BJ, genic diet enhances cardiac tolerance to global ischaemia. Acta Cardiol.
Downloaded from http://ahajournals.org by on June 2, 2020

Bucher HC. Effects of low-carbohydrate vs low-fat diets on weight loss and 2007;62:381–389. doi: 10.2143/AC.62.4.2022282
cardiovascular risk factors: a meta-analysis of randomized controlled trials. 78. Zou Z, Sasaguri S, Rajesh KG, Suzuki R. dl-3-Hydroxybutyrate admin-
Arch Intern Med. 2006;166:285–293. doi: 10.1001/archinte.166.3.285 istration prevents myocardial damage after coronary occlusion in rat
59. Gibson AA, Seimon RV, Lee CM, Ayre J, Franklin J, Markovic TP, hearts. Am J Physiol Heart Circ Physiol. 2002;283:H1968–H1974. doi:
Caterson ID, Sainsbury A. Do ketogenic diets really suppress appetite? 10.1152/ajpheart.00250.2002
A systematic review and meta-analysis. Obes Rev. 2015;16:64–76. doi: 79. Sharma N, Okere IC, Barrows BR, Lei B, Duda MK, Yuan CL, Previs SF,
10.1111/obr.12230 Sharov VG, Azimzadeh AM, Ernsberger P, et al. High-sugar diets increase
60. Augustin K, Khabbush A, Williams S, Eaton S, Orford M, Cross JH, Heales SJR, cardiac dysfunction and mortality in hypertension compared to low-car-
Walker MC, Williams RSB. Mechanisms of action for the medium-chain bohydrate or high-starch diets. J Hypertens. 2008;26:1402–1410. doi:
triglyceride ketogenic diet in neurological and metabolic disorders. Lancet 10.1097/HJH.0b013e3283007dda
Neurol. 2018;17:84–93. doi: 10.1016/S1474-4422(17)30408-8 80. Duda MK, O’Shea KM, Lei B, Barrows BR, Azimzadeh AM, McElfresh TE,
61. Mumme K, Stonehouse W. Effects of medium-chain triglycerides Hoit BD, Kop WJ, Stanley WC. Low-carbohydrate/high-fat diet attenuates
on weight loss and body composition: a meta-analysis of random- pressure overload-induced ventricular remodeling and dysfunction. J Card
ized controlled trials. J Acad Nutr Diet. 2015;115:249–263. doi: Fail. 2008;14:327–335. doi: 10.1016/j.cardfail.2007.11.003
10.1016/j.jand.2014.10.022 81. Okere IC, Young ME, McElfresh TA, Chess DJ, Sharov VG, Sabbah HN,
62. Holloway CJ, Cochlin LE, Emmanuel Y, Murray A, Codreanu I, Hoit BD, Ernsberger P, Chandler MP, Stanley WC. Low carbohydrate/
Edwards LM, Szmigielski C, Tyler DJ, Knight NS, Saxby BK, et al. A high- high-fat diet attenuates cardiac hypertrophy, remodeling, and altered
fat diet impairs cardiac high-energy phosphate metabolism and cognitive gene expression in hypertension. Hypertension. 2006;48:1116–1123. doi:
function in healthy human subjects. Am J Clin Nutr. 2011;93:748–755. 10.1161/01.HYP.0000248430.26229.0f
doi: 10.3945/ajcn.110.002758 82. Shah AM, Claggett B, Sweitzer NK, Shah SJ, Anand IS, Liu L,
63. Murray AJ, Anderson RE, Watson GC, Radda GK, Clarke K. Uncou- Pitt B, Pfeffer MA, Solomon SD. Prognostic importance of impaired sys-
pling proteins in human heart. Lancet. 2004;364:1786–1788. doi: tolic function in heart failure with preserved ejection fraction and
10.1016/S0140-6736(04)17402-3 the impact of spironolactone. Circulation. 2015;132:402–414. doi:
64. Leichman JG, Aguilar D, King TM, Vlada A, Reyes M, Taegtmeyer H. As- 10.1161/CIRCULATIONAHA.115.015884
sociation of plasma free fatty acids and left ventricular diastolic function 83. Weber T, Chirinos JA. Pulsatile arterial haemodynamics in heart failure. Eur
in patients with clinically severe obesity. Am J Clin Nutr. 2006;84:336–341. Heart J. 2018;39:3847–3854. doi: 10.1093/eurheartj/ehy346
doi: 10.1093/ajcn/84.1.336 84. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D,
65. Cox PJ, Clarke K. Acute nutritional ketosis: implications for exercise D’Agostino D, Planavsky N, Lupfer C, Kanneganti TD, et al. The ketone
performance and metabolism. Extrem Physiol Med. 2014;3:17. doi: metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated in-
10.1186/2046-7648-3-17 flammatory disease. Nat Med. 2015;21:263–269. doi: 10.1038/nm.3804
66. Seidelmann SB, Claggett B, Cheng S, Henglin M, Shah A, Steffen LM, 85. Kanikarla-Marie P, Jain SK. Hyperketonemia (acetoacetate) upregulates
Folsom AR, Rimm EB, Willett WC, Solomon SD. Dietary carbohydrate in- NADPH oxidase 4 and elevates oxidative stress, ICAM-1, and monocyte
take and mortality: a prospective cohort study and meta-analysis. Lancet adhesivity in endothelial cells. Cell Physiol Biochem. 2015;35:364–373.
Public Health. 2018;3:e419–e428. doi: 10.1016/S2468-2667(18)30135-X doi: 10.1159/000369702
67. Balasse E, Ooms HA. Changes in the concentrations of glucose, free 86. Jain SK, Kannan K, Lim G, McVie R, Bocchini JA Jr. Hyperketone-
fatty acids, insulin and ketone bodies in the blood during sodium mia increases tumor necrosis factor-alpha secretion in cultured U937

Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033 June 2, 2020 1811


Selvaraj et al Therapeutic Ketosis in Heart Failure

monocytes and type 1 diabetic patients and is apparently mediated by 91. Tsutsui H, Kinugawa S, Matsushima S. Oxidative stress and heart fail-
oxidative stress and cAMP deficiency. Diabetes. 2002;51:2287–2293. doi: ure. Am J Physiol Heart Circ Physiol. 2011;301:H2181–H2190. doi:
STATE OF THE ART

10.2337/diabetes.51.7.2287 10.1152/ajpheart.00554.2011
87. Fisher FM, Maratos-Flier E. Understanding the physiology of FGF21. 92. Xie Z, Zhang D, Chung D, Tang Z, Huang H, Dai L, Qi S, Li J,
Annu Rev Physiol. 2016;78:223–241. doi: 10.1146/annurev- Colak G, Chen Y, et al. Metabolic regulation of gene expression by his-
physiol-021115-105339 tone lysine β-hydroxybutyrylation. Mol Cell. 2016;62:194–206. doi:
88. Planavila A, Redondo I, Hondares E, Vinciguerra M, Munts C, Iglesias R, 10.1016/j.molcel.2016.03.036
Gabrielli LA, Sitges M, Giralt M, van Bilsen M, et al. Fibroblast growth 93. Lytvyn Y, Bjornstad P, Udell JA, Lovshin JA, Cherney DZI. Sodium glucose
factor 21 protects against cardiac hypertrophy in mice. Nat Commun. cotransporter-2 inhibition in heart failure: potential mechanisms, clinical
2013;4:2019. doi: 10.1038/ncomms3019 applications, and summary of clinical trials. Circulation. 2017;136:1643–
89. Nagao M, Toh R, Irino Y, Mori T, Nakajima H, Hara T, Honjo T, 1658. doi: 10.1161/CIRCULATIONAHA.117.030012
Satomi-Kobayashi S, Shinke T, Tanaka H, et al. β-Hydroxybutyrate eleva- 94. Bonner C, Kerr-Conte J, Gmyr V, Queniat G, Moerman E, Thévenet J,
tion as a compensatory response against oxidative stress in cardiomyo- Beaucamps C, Delalleau N, Popescu I, Malaisse WJ, et al. Inhibition of the
cytes. Biochem Biophys Res Commun. 2016;475:322–328. doi: 10.1016/j. glucose transporter SGLT2 with dapagliflozin in pancreatic alpha cells triggers
bbrc.2016.05.097 glucagon secretion. Nat Med. 2015;21:512–517. doi: 10.1038/nm.3828
90. Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, 95. Ferrannini E, Baldi S, Frascerra S, Astiarraga B, Heise T, Bizzotto R, Mari A,
Grueter CA, Lim H, Saunders LR, Stevens RD, et al. Suppression of oxi- Pieber TR, Muscelli E. Shift to fatty substrate utilization in response to sodium-
dative stress by β-hydroxybutyrate, an endogenous histone deacetylase glucose cotransporter 2 inhibition in subjects without diabetes and patients
inhibitor. Science. 2013;339:211–214. doi: 10.1126/science.1227166 with type 2 diabetes. Diabetes. 2016;65:1190–1195. doi: 10.2337/db15-1356
Downloaded from http://ahajournals.org by on June 2, 2020

1812 June 2, 2020 Circulation. 2020;141:1800–1812. DOI: 10.1161/CIRCULATIONAHA.119.045033

You might also like