Ketone Ester Treatment Improves Cardiac Function and Reduces Pathologic Remodeling in Preclinical Models of Heart Failure

Salva R Yurista, Timothy R Matsuura, Herman H W Silljé, Kirsten T Nijholt, Kendra S McDaid, Swapnil V Shewale, Teresa C Leone, John C Newman, Eric Verdin, Dirk J van Veldhuisen, Rudolf A de Boer, Daniel P Kelly, B Daan Westenbrink, Salva R Yurista, Timothy R Matsuura, Herman H W Silljé, Kirsten T Nijholt, Kendra S McDaid, Swapnil V Shewale, Teresa C Leone, John C Newman, Eric Verdin, Dirk J van Veldhuisen, Rudolf A de Boer, Daniel P Kelly, B Daan Westenbrink

Abstract

Background: Accumulating evidence suggests that the failing heart reprograms fuel metabolism toward increased utilization of ketone bodies and that increasing cardiac ketone delivery ameliorates cardiac dysfunction. As an initial step toward development of ketone therapies, we investigated the effect of chronic oral ketone ester (KE) supplementation as a prevention or treatment strategy in rodent heart failure models.

Methods: Two independent rodent heart failure models were used for the studies: transverse aortic constriction/myocardial infarction (MI) in mice and post-MI remodeling in rats. Seventy-five mice underwent a prevention treatment strategy with a KE comprised of hexanoyl-hexyl-3-hydroxybutyrate KE (KE-1) diet, and 77 rats were treated in either a prevention or treatment regimen using a commercially available β-hydroxybutyrate-(R)-1,3-butanediol monoester (DeltaG; KE-2) diet.

Results: The KE-1 diet in mice elevated β-hydroxybutyrate levels during nocturnal feeding, whereas the KE-2 diet in rats induced ketonemia throughout a 24-hour period. The KE-1 diet preventive strategy attenuated development of left ventricular dysfunction and remodeling post-transverse aortic constriction/MI (left ventricular ejection fraction±SD, 36±8 in vehicle versus 45±11 in KE-1; P=0.016). The KE-2 diet therapeutic approach also attenuated left ventricular dysfunction and remodeling post-MI (left ventricular ejection fraction, 41±11 in MI-vehicle versus 61±7 in MI-KE-2; P<0.001). In addition, ventricular weight, cardiomyocyte cross-sectional area, and the expression of ANP (atrial natriuretic peptide) were significantly attenuated in the KE-2-treated MI group. However, treatment with KE-2 did not influence cardiac fibrosis post-MI. The myocardial expression of the ketone transporter and 2 ketolytic enzymes was significantly increased in rats fed KE-2 diet along with normalization of myocardial ATP levels to sham values.

Conclusions: Chronic oral supplementation with KE was effective in both prevention and treatment of heart failure in 2 preclinical animal models. In addition, our results indicate that treatment with KE reprogrammed the expression of genes involved in ketone body utilization and normalized myocardial ATP production following MI, consistent with provision of an auxiliary fuel. These findings provide rationale for the assessment of KEs as a treatment for patients with heart failure.

Keywords: esters; heart failure; ketone bodies; myocardial infarction; myocardium.

Conflict of interest statement

The University Medical Center Groningen, which employs Dr Yurista, Dr Silljé, K.T. Nijholt, Dr van Veldhuisen, Dr de Boer, and Dr Westenbrink, has received research grants and fees from Abbott, AstraZeneca, Bristol-Myers Squibb, Novartis, Novo Nordisk, and Roche. Dr de Boer received personal fees from Abbott, AstraZeneca, and Novartis. Drs Newman and Verdin are cofounders with equity interest in BHB Therapeutics, Inc. Dr Verdin is an advisor to KetoFuel, Inc. Dr Newman serves on the scientific advisory board of Virta Health, Inc, and Roche.

Figures

Figure 1.
Figure 1.
Ketone ester diet increases nocturnal circulating β-hydroxybutyrate (βHB or BHB) in mice.A, Structure of the hexanoyl-hexyl-3-hydroxybutyrate ketone ester (KE-1) depicting a central βHB flanked by two 6-carbon moieties. B, Levels of blood βHB measured after 1 wk of feeding normal chow, 10% w/w KE-1 diet, or 15% w/w KE-1 diet (n=5–9). βHB measurements were taken during the day (09:00–13:00) and at night (00:00–02:00). Two-way ANOVA with Tukey multiple comparison test. C, Body weight normalized to starting weight measured after 1 wk of feeding normal chow, 10% w/w KE-1 diet, or 15% w/w KE-1 diet. Data are presented as mean±SEM. Comparisons between groups were performed using 1-way ANOVA with Tukey multiple comparison test.
Figure 2.
Figure 2.
Preventive treatment with ketone ester diet attenuates pathological cardiac remodeling and left ventricular dysfunction in a mouse model of heart failure.A, Schematic showing the experimental timeline. Male C57Bl/6NJ mice aged 7 wk were randomized to diet of either standard chow or 10% hexanoyl-hexyl-3-hydroxybutyrate ketone ester (KE-1) diet. One week later, mice received either sham surgery or transverse aortic constriction (TAC)/myocardial infarction (MI) surgery. B, Ratio of biventricular weight (BV) to tibia length (T). C, Left ventricular (LV) end-systolic volume (ESV) and end-diastolic volume (EDV) 4 wk after TAC/MI surgery as determined by echocardiography. D, LV ejection fraction (LVEF) 4 wk post-TAC/MI surgery (n=11–16). Data are presented as mean±SEM. Comparisons between groups were performed using 2-way ANOVA with Tukey multiple comparison test.
Figure 3.
Figure 3.
Ketone ester supplementation induces sustained ketonemia in rats.A, Experimental design of early and late β-hydroxybutyrate (βHB or BHB)–butanediol monoester (KE-2) diet supplementation regimens in rats with heart failure (HF) after myocardial infarction (MI). B, Plasma βHB concentrations measured after 10 d of KE-2 supplementation during the day (09:00–10:30) and at night (22:00–22:30; n=8–26). C, Body weight at the end of the study (n=8–20). Data are presented as mean±SEM. Comparisons between groups were performed using 2-way ANOVA with Tukey multiple comparison test. Echo indicates echocardiography.
Figure 4.
Figure 4.
Ketone ester treatment improves pathological cardiac remodeling and left ventricular (LV) dysfunction in a rat model of heart failure.A, Representative LV sections stained with Masson trichrome. B, Quantification of infarct size from Masson trichrome stained section (n=7–22). C, Ratio of biventricular weight (BV) to tibia length (T; n=7–21). D, LV internal dimensions in diastole (LVIDd) as determined by echocardiography (n=7–22). E, LV ejection fraction (LVEF; n=7–22). Comparisons between groups were performed using 1-way ANOVA with Tukey multiple comparison test. F, Longitudinal change in LVEF between 2 wk post-myocardial infarction (MI) and termination in the βHB-butanediol monoester (KE-2)-late (KE-2-L) compared with the chow control group, tested using Wilcoxon signed-rank test. Data are presented as mean±SEM. KE-2-E indicates KE-2-early.
Figure 5.
Figure 5.
Ketone ester treatment attenuates cardiac hypertrophy independent of fibrotic response.A, Representative left ventricular (LV) sections stained with wheat germ agglutinin (WGA) and Masson trichrome to assess cardiomyocyte hypertrophy and fibrosis, respectively. B, Quantification of cardiomyocyte cross-sectional area from WGA-stained section (n=7–22). C, Quantification of fibrosis in noninfarcted LV from Masson trichrome–stained section (n=7–22). The graph denotes fold change over sham-chow control group. D and E, Measurement of mRNA levels to assess molecular markers for remodeling and fibrosis, respectively, normalized to 36B4 (n=7–22). The graph denotes fold change over sham-chow group. Data are presented as mean±SEM. Comparisons between groups were performed using 1-way ANOVA with Tukey multiple comparison test. ANP indicates atrial natriuretic peptide; COL1A1, collagen type I alpha 1; KE-2-E, βHB-butanediol monoester-early; KE-2-L, βHB-butanediol monoester-late; and MI, myocardial infarction.
Figure 6.
Figure 6.
Ketone ester treatment enhances cardiac energy. Levels of mRNA encoding MCT1 (monocarboxylate transporter 1; n=9–21; A) and BDH1 (3-hydroxybutyrate dehydrogenase 1; n=9–21; B) in left ventricular (LV) free wall remote from the infarction (as determined by quantitative polymerase chain reaction) shown as arbitrary units normalized to the value of sham-chow control (+1.0). C, Top, Representative immunoblot analyses of succinyl CoA:3-oxo acid CoA transferase (SCOT) and GAPDH from protein extracts derived from the noninfarcted LV free wall. Bottom, Average SCOT protein expression normalized to GADPH and presented as fold change over the sham-chow group (n=5–6). D, ATP levels in the LV tissue (n=9–21). Data are presented as mean±SEM. Comparisons between groups were performed using Kruskal-Wallis test with Dunn multiple comparisons test. MI indicates myocardial infarction.

References

    1. Bishop SP, Altschuld RA. Increased glycolytic metabolism in cardiac hypertrophy and congestive failure. Am J Physiol. 1970;218:153–159. doi: 10.1152/ajplegacy.1970.218.1.153
    1. Taegtmeyer H, Overturf ML. Effects of moderate hypertension on cardiac function and metabolism in the rabbit. Hypertension. 1988;11:416–426. doi: 10.1161/01.hyp.11.5.416
    1. Allard MF, Schönekess BO, Henning SL, English DR, Lopaschuk GD. Contribution of oxidative metabolism and glycolysis to ATP production in hypertrophied hearts. Am J Physiol. 1994;2672 pt 2H742–H750. doi: 10.1152/ajpheart.1994.267.2.H742
    1. Christe ME, Rodgers RL. Altered glucose and fatty acid oxidation in hearts of the spontaneously hypertensive rat. J Mol Cell Cardiol. 1994;26:1371–1375. doi: 10.1006/jmcc.1994.1155
    1. Paolisso G, Gambardella A, Galzerano D, D’Amore A, Rubino P, Verza M, Teasuro P, Varricchio M, D’Onofrio F. Total-body and myocardial substrate oxidation in congestive heart failure. Metabolism. 1994;43:174–179. doi: 10.1016/0026-0495(94)90241-0
    1. Sambandam N, Lopaschuk GD, Brownsey RW, Allard MF. Energy metabolism in the hypertrophied heart. Heart Fail Rev. 2002;7:161–173. doi: 10.1023/a:1015380609464
    1. Chandler MP, Kerner J, Huang H, Vazquez E, Reszko A, Martini WZ, Hoppel CL, Imai M, Rastogi S, Sabbah HN, et al. Moderate severity heart failure does not involve a downregulation of myocardial fatty acid oxidation. Am J Physiol Heart Circ Physiol. 2004;287:H1538–H1543. doi: 10.1152/ajpheart.00281.2004
    1. Sack MN, Rader TA, Park S, Bastin J, McCune SA, Kelly DP. Fatty acid oxidation enzyme gene expression is downregulated in the failing heart. Circulation. 1996;94:2837–2842. doi: 10.1161/01.cir.94.11.2837
    1. Wallhaus TR, Taylor M, DeGrado TR, Russell DC, Stanko P, Nickles RJ, Stone CK. Myocardial free fatty acid and glucose use after carvedilol treatment in patients with congestive heart failure. Circulation. 2001;103:2441–2446. doi: 10.1161/01.cir.103.20.2441
    1. Dávila-Román VG, Vedala G, Herrero P, de las Fuentes L, Rogers JG, Kelly DP, Gropler RJ. Altered myocardial fatty acid and glucose metabolism in idiopathic dilated cardiomyopathy. J Am Coll Cardiol. 2002;40:271–277. doi: 10.1016/s0735-1097(02)01967-8
    1. de las Fuentes L, Herrero P, Peterson LR, Kelly DP, Gropler RJ, Dávila-Román VG. Myocardial fatty acid metabolism: independent predictor of left ventricular mass in hypertensive heart disease. Hypertension. 2003;41:83–87. doi: 10.1161/01.hyp.0000047668.48494.39
    1. Lai L, Leone TC, Keller MP, Martin OJ, Broman AT, Nigro J, Kapoor K, Koves TR, Stevens R, Ilkayeva OR, Vega RB, Attie AD, Muoio DM, Kelly DP. Energy metabolic reprogramming in the hypertrophied and early stage failing heart. Circ Hear Fail. 2014;7:1022–1031.
    1. Aubert G, Martin OJ, Horton JL, Lai L, Vega RB, Leone TC, Koves T, Gardell SJ, Krüger M, Hoppel CL, et al. The failing heart relies on ketone bodies as a fuel. Circulation. 2016;133:698–705. doi: 10.1161/CIRCULATIONAHA.115.017355
    1. 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 intramyocardial disruption of lipid metabolism and increased myocardial ketone utilization in advanced human heart failure. Circulation. 2016;133:706–716. doi: 10.1161/CIRCULATIONAHA.115.017545
    1. 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. 2019;4:e124079 doi: 10.1172/jci.insight.124079
    1. Nielsen R, Møller N, Gormsen LC, Tolbod LP, Hansson NH, Sorensen J, Harms HJ, Frøkiær J, Eiskjaer H, Jespersen NR, et al. Cardiovascular effects of treatment with the ketone body 3-hydroxybutyrate in chronic heart failure patients. Circulation. 2019;139:2129–2141. doi: 10.1161/CIRCULATIONAHA.118.036459
    1. Abdul Kadir A, Clarke K, Evans RD. Cardiac ketone body metabolism. Biochim Biophys Acta Mol Basis Dis. 2020;1866:165739 doi: 10.1016/j.bbadis.2020.165739
    1. Veech RL, Mehlman MA. Liver metabolite content, redox and phosphorylation states in rats fed diets containing 1,3-butanediol and ethanol. In: Energy Metabolism and the Regulation of Metabolic Processes in Mitochondria. 1972. Elsevier; 171–183.
    1. Kashiwaya Y, Pawlosky R, Markis W, King MT, Bergman C, Srivastava S, Murray A, Clarke K, Veech RL. A ketone ester diet increases brain malonyl-CoA and uncoupling proteins 4 and 5 while decreasing food intake in the normal Wistar Rat. J Biol Chem. 2010;285:25950–25956. doi: 10.1074/jbc.M110.138198
    1. Shivva V, Cox PJ, Clarke K, Veech RL, Tucker IG, Duffull SB. The population pharmacokinetics of D-β-hydroxybutyrate following administration of (R)-3-hydroxybutyl (R)-3-hydroxybutyrate. AAPS J. 2016;18:678–688. doi: 10.1208/s12248-016-9879-0
    1. 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. 2017;8:848 doi: 10.3389/fphys.2017.00848
    1. Kashiwaya Y, Bergman C, Lee JH, Wan R, King MT, Mughal MR, Okun E, Clarke K, Mattson MP, Veech RL. A ketone ester diet exhibits anxiolytic and cognition-sparing properties, and lessens amyloid and tau pathologies in a mouse model of Alzheimer’s disease. Neurobiol Aging. 2013;34:1530–1539. doi: 10.1016/j.neurobiolaging.2012.11.023
    1. Murray AJ, Knight NS, Cole MA, Cochlin LE, Carter E, Tchabanenko K, Pichulik T, Gulston MK, Atherton HJ, Schroeder MA, et al. Novel ketone diet enhances physical and cognitive performance. FASEB J. 2016;30:4021–4032. doi: 10.1096/fj.201600773R
    1. Pawlosky RJ, Kemper MF, Kashiwaya Y, King MT, Mattson MP, Veech RL. Effects of a dietary ketone ester on hippocampal glycolytic and tricarboxylic acid cycle intermediates and amino acids in a 3xTgAD mouse model of Alzheimer’s disease. J Neurochem. 2017;141:195–207. doi: 10.1111/jnc.13958
    1. Cox PJ, Kirk T, Ashmore T, Willerton K, Evans R, Smith A, Murray AJ, Stubbs B, West J, McLure SW, et al. Nutritional ketosis alters fuel preference and thereby endurance performance in athletes. Cell Metab. 2016;24:256–268. doi: 10.1016/j.cmet.2016.07.010
    1. Clarke K, Tchabanenko K, Pawlosky R, Carter E, Knight NS, Murray AJ, Cochlin LE, King MT, Wong AW, Roberts A, et al. Oral 28-day and developmental toxicity studies of (R)-3-hydroxybutyl (R)-3-hydroxybutyrate. Regul Toxicol Pharmacol. 2012;63:196–208. doi: 10.1016/j.yrtph.2012.04.001
    1. Weinheimer CJ, Lai L, Kelly DP, Kovacs A. Novel mouse model of left ventricular pressure overload and infarction causing predictable ventricular remodelling and progression to heart failure. Clin Exp Pharmacol Physiol. 2015;42:33–40. doi: 10.1111/1440-1681.12318
    1. Yurista SR, Silljé HHW, Oberdorf-Maass SU, Schouten EM, Pavez Giani MG, Hillebrands JL, van Goor H, van Veldhuisen DJ, de Boer RA, Westenbrink BD. Sodium-glucose co-transporter 2 inhibition with empagliflozin improves cardiac function in non-diabetic rats with left ventricular dysfunction after myocardial infarction. Eur J Heart Fail. 2019;21:862–873. doi: 10.1002/ejhf.1473
    1. Yurista SR, Silljé HHW, Nijholt KT, Dokter MM, van Veldhuisen DJ, de Boer RA, Westenbrink BD. Factor Xa inhibition with apixaban does not influence cardiac remodelling in rats with heart failure after myocardial infarction. Cardiovasc Drugs Ther. 2020. doi: 10.1007/s10557-020-06999-7
    1. Yurista SR, Silljé HHW, van Goor H, Hillebrands JL, Heerspink HJL, de Menezes Montenegro L, Oberdorf-Maass SU, de Boer RA, Westenbrink BD. Effects of sodium-glucose co-transporter 2 inhibition with empaglifozin on renal structure and function in non-diabetic rats with left ventricular dysfunction after myocardial infarction. Cardiovasc Drugs Ther. 2020;34:311–321. doi: 10.1007/s10557-020-06954-6
    1. Davis RAH, Deemer SE, Bergeron JM, Little JT, Warren JL, Fisher G, Smith DL, Jr, Fontaine KR, Dickinson SL, Allison DB, et al. Dietary R, S-1,3-butanediol diacetoacetate reduces body weight and adiposity in obese mice fed a high-fat diet. FASEB J. 2019;33:2409–2421. doi: 10.1096/fj.201800821RR
    1. Bleeker JC, Visser G, Clarke K, Ferdinandusse S, de Haan FH, Houtkooper RH, IJlst L, Kok IL, Langeveld M, van der Pol WL, et al. Nutritional ketosis improves exercise metabolism in patients with very long-chain acyl-CoA dehydrogenase deficiency. J Inherit Metab Dis. 2020;43:787–799. doi: 10.1002/jimd.12217
    1. Soto-Mota A, Vansant H, Evans RD, Clarke K. Safety and tolerability of sustained exogenous ketosis using ketone monoester drinks for 28 days in healthy adults. Regul Toxicol Pharmacol. 2019;109:104506 doi: 10.1016/j.yrtph.2019.104506
    1. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, D’Agostino D, Planavsky N, Lupfer C, Kanneganti TD, et al. The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015;21:263–269. doi: 10.1038/nm.3804
    1. Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, Grueter CA, Lim H, Saunders LR, Stevens RD, et al. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science. 2013;339:211–214. doi: 10.1126/science.1227166
    1. Xie Z, Zhang D, Chung D, Tang Z, Huang H, Dai L, Qi S, Li J, Colak G, Chen Y, et al. Metabolic regulation of gene expression by histone lysine β-hydroxybutyrylation. Mol Cell. 2016;62:194–206. doi: 10.1016/j.molcel.2016.03.036
    1. Gormsen LC, Svart M, Thomsen HH, Søndergaard E, Vendelbo MH, Christensen N, Tolbod LP, Harms HJ, Nielsen R, Wiggers H, et al. Ketone body infusion with 3-hydroxybutyrate reduces myocardial glucose uptake and increases blood flow in humans: a positron emission tomography study. J Am Heart Assoc. 2017;6:e005066 doi: 10.1161/JAHA.116.005066
    1. Epstein FH, Kelly DP, Strauss AW. Inherited cardiomyopathies. N Engl J Med. 1994;330:913–919. doi: 10.1056/NEJM199403313301308
    1. Ritterhoff J, Young S, Villet O, Shao D, Neto FC, Bettcher LF, Hsu YA, Kolwicz SC, Jr, Raftery D, Tian R. Metabolic remodeling promotes cardiac hypertrophy by directing glucose to aspartate biosynthesis. Circ Res. 2020;126:182–196. doi: 10.1161/CIRCRESAHA.119.315483
    1. Kolwicz SC, Airhart S, Tian R. Ketones step to the plate. Circulation. 2016;133:689–691. doi: 10.1161/CIRCULATIONAHA.116.021230
    1. Solomon SD, McMurray JJV, Anand IS, Ge J, Lam CSP, Maggioni AP, Martinez F, Packer M, Pfeffer MA, Pieske B, et al. ; PARAGON-HF Investigators and Committees. Angiotensin-neprilysin inhibition in heart failure with preserved ejection fraction. N Engl J Med. 2019;381:1609–1620. doi: 10.1056/NEJMoa1908655
    1. Armstrong PW, Pieske B, Anstrom KJ, Ezekowitz J, Hernandez AF, Butler J, Lam CSP, Ponikowski P, Voors AA, Jia G, et al. ; VICTORIA Study Group. Vericiguat in patients with heart failure and reduced ejection fraction. N Engl J Med. 2020;382:1883–1893. doi: 10.1056/NEJMoa1915928
    1. Santos-Gallego CG, Requena-Ibanez JA, San Antonio R, Ishikawa K, Watanabe S, Picatoste B, Flores E, Garcia-Ropero A, Sanz J, Hajjar RJ, et al. Empagliflozin ameliorates adverse left ventricular remodeling in nondiabetic heart failure by enhancing myocardial energetics. J Am Coll Cardiol. 2019;73:1931–1944. doi: 10.1016/j.jacc.2019.01.056
    1. Petrie MC, Verma S, Docherty KF, Inzucchi SE, Anand I, Belohlávek J, Böhm M, Chiang CE, Chopra VK, de Boer RA, et al. Effect of dapagliflozin on worsening heart failure and cardiovascular death in patients with heart failure with and without diabetes. JAMA. 2020;323:1353–1368. doi: 10.1001/jama.2020.1906
    1. McMurray JJV, Solomon SD, Inzucchi SE, Køber L, Kosiborod MN, Martinez FA, Ponikowski P, Sabatine MS, Anand IS, Bělohlávek J, et al. ; DAPA-HF Trial Committees and Investigators. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med. 2019;381:1995–2008. doi: 10.1056/NEJMoa1911303
    1. Zelniker TA, Braunwald E. Mechanisms of cardiorenal effects of sodium-glucose cotransporter 2 inhibitors: JACC state-of-the-art review. J Am Coll Cardiol. 2020;75:422–434. doi: 10.1016/j.jacc.2019.11.031
    1. Yu Y, Yu Y, Zhang Y, Zhang Z, An W, Zhao X. Treatment with D-β-hydroxybutyrate protects heart from ischemia/reperfusion injury in mice. Eur J Pharmacol. 2018;829:121–128. doi: 10.1016/j.ejphar.2018.04.019

Source: PubMed

3
購読する