Ketone Body Infusion With 3-Hydroxybutyrate Reduces Myocardial Glucose Uptake and Increases Blood Flow in Humans: A Positron Emission Tomography Study

Lars C Gormsen, Mads Svart, Henrik Holm Thomsen, Esben Søndergaard, Mikkel H Vendelbo, Nana Christensen, Lars Poulsen Tolbod, Hendrik Johannes Harms, Roni Nielsen, Henrik Wiggers, Niels Jessen, Jakob Hansen, Hans Erik Bøtker, Niels Møller, Lars C Gormsen, Mads Svart, Henrik Holm Thomsen, Esben Søndergaard, Mikkel H Vendelbo, Nana Christensen, Lars Poulsen Tolbod, Hendrik Johannes Harms, Roni Nielsen, Henrik Wiggers, Niels Jessen, Jakob Hansen, Hans Erik Bøtker, Niels Møller

Abstract

Background: High levels of ketone bodies are associated with improved survival as observed with regular exercise, caloric restriction, and-most recently-treatment with sodium-glucose linked transporter 2 inhibitor antidiabetic drugs. In heart failure, indices of ketone body metabolism are upregulated, which may improve energy efficiency and increase blood flow in skeletal muscle and the kidneys. Nevertheless, it is uncertain how ketone bodies affect myocardial glucose uptake and blood flow in humans. Our study was therefore designed to test whether ketone body administration in humans reduces myocardial glucose uptake (MGU) and increases myocardial blood flow.

Methods and results: Eight healthy subjects, median aged 60 were randomly studied twice: (1) During 390 minutes infusion of Na-3-hydroxybutyrate (KETONE) or (2) during 390 minutes infusion of saline (SALINE), together with a concomitant low-dose hyperinsulinemic-euglycemic clamp to inhibit endogenous ketogenesis. Myocardial blood flow was measured by 15O-H2O positron emission tomography/computed tomography, myocardial fatty acid metabolism by 11C-palmitate positron emission tomography/computed tomography and MGU by 18F-fluorodeoxyglucose positron emission tomography/computed tomography. Similar euglycemia, hyperinsulinemia, and suppressed free fatty acids levels were recorded on both study days; Na-3-hydroxybutyrate infusion increased circulating Na-3-hydroxybutyrate levels from zero to 3.8±0.5 mmol/L. MGU was halved by hyperketonemia (MGU [nmol/g per minute]: 304±97 [SALINE] versus 156±62 [KETONE], P<0.01), whereas no effects were observed on palmitate uptake oxidation or esterification. Hyperketonemia increased heart rate by ≈25% and myocardial blood flow by 75%.

Conclusions: Ketone bodies displace MGU and increase myocardial blood flow in healthy humans; these novel observations suggest that ketone bodies are important cardiac fuels and vasodilators, which may have therapeutic potentials.

Keywords: 11C‐palmitate; 18F‐fluorodeoxyglucose; cardiac metabolism; hyperketonemia; myocardial glucose uptake; myocardial perfusion; nuclear medicine; positron emission tomography/computed tomography.

© 2017 The Authors. Published on behalf of the American Heart Association, Inc., by Wiley Blackwell.

Figures

Figure 1
Figure 1
Study protocol. Study days (SALINE and KETONES) were identical except for the intervention, which was either infusion of Na‐betahydroxybutyrate (OHB) or NaCl (0.9%). PET/CT scans were initiated at t=180 minutes at which point quasi‐stable concentrations of metabolites were expected. CT indicates computed tomography; FDG, fluorodeoxyglucose; FFA, free fatty acids.
Figure 2
Figure 2
Hemodynamics. Systolic (treatment×time, P=0.54) blood pressure was comparable on the 2 study days (A), whereas heart diastolic blood pressure (treatment×time, P<0.001) decreased and heart rate (treatment×time, P<0.001) increased significantly during the KETONES study day (B). BPM indicates beats per minute.
Figure 3
Figure 3
Hormones and metabolites. A, Circulating 3‐OHB concentrations were below detection threshold (50 μmol/L) during the SALINE study day and increased to between 3 and 4 mmol/L between t=120 and 390 during the KETONE study day. By design, FFA concentrations (B) were suppressed to ≈50 μmol/L during both study days by the HE‐clamp, which increased insulin levels (C) to between 160 and 180 pmol/L. Glucose levels (D) were clamped ≈5 mmol/L through the study days, although a tendency to increasing glucose concentrations was observed from t=300 minutes and onwards. P‐values refer to the mixed model treatment vs time interaction. N=8, error bars are ±SE. FFA indicates free fatty acids; HE, hyperinsulinemic–euglycemic; 3‐OHB, 3‐hydroxybutyrate.
Figure 4
Figure 4
Glucose infusion rates reached plateau levels at t=180 minutes and started decreasing from t=300 onwards. No difference in insulin sensitivity was observed. N=8, error bars are ±SE for the GIR curves. Open boxes = KETONES, closed circles = SALINE. GIR indicates glucose infusion rates.
Figure 5
Figure 5
Myocardial 18F‐FDG uptake. Summed images (t=10–50 minutes) from a representative subject are presented in the left panel. Infusion of OHB significantly decreased 18F‐FDG uptake in all subjects, resulting in significantly lower relative uptake rates (Ki) when compared with the SALINE study day (A). Since blood glucose levels were clamped at ≈5 mmol/L, the resulting absolute MGU was roughly halved (B). N=8, error bars are SD. FDG indicates fluorodeoxyglucose; MGU, myocardial glucose uptake; OHB, hydroxybutyrate; SUV, standardized uptake value.
Figure 6
Figure 6
Myocardial fatty acid metabolism. A, Summed images (t=10–50 minutes) from a representative subject are presented in the left panel. There was no visible difference in accumulated 11C‐palmitate in most subjects. B, Myocardial 11C‐palmitate metabolism. The 3‐tissue compartment model has 3 free parameters (black arrows) and 3 fixed parameters (dotted arrows). C, No differences were observed in myocardial fatty acid uptake, esterification, or oxidation (black bar: Saline, white bar: Ketone). N=7, error bars are SD. MFAE indicates myocardial fatty acid esterification; MFAO, myocardial fatty acid oxidation; MFAU, myocardial fatty acid uptake; SUV, standardized uptake value.

References

    1. Taegtmeyer H. Failing heart and starving brain: ketone bodies to the rescue. Circulation. 2016;134:265–266.
    1. Ferrannini E, Mark M, Mayoux E. CV protection in the EMPA‐REG Outcome Trial: a “thrifty substrate” hypothesis. Diabetes Care. 2016;39:1108–1114.
    1. Cahill GF Jr. Fuel metabolism in starvation. Annu Rev Nutr. 2006;26:1–22.
    1. Kashiwaya Y, Sato K, Tsuchiya N, Thomas S, Fell DA, Veech RL, Passonneau JV. Control of glucose utilization in working perfused rat heart. J Biol Chem. 1994;269:25502–25514.
    1. Veech RL. The therapeutic implications of ketone bodies: the effects of ketone bodies in pathological conditions: ketosis, ketogenic diet, redox states, insulin resistance, and mitochondrial metabolism. Prostaglandins Leukot Essent Fatty Acids. 2004;70:309–319.
    1. Boehm EA, Jones BE, Radda GK, Veech RL, Clarke K. Increased uncoupling proteins and decreased efficiency in palmitate‐perfused hyperthyroid rat heart. Am J Physiol Heart Circ Physiol. 2001;280:H977–H983.
    1. Maalouf M, Rho JM, Mattson MP. The neuroprotective properties of calorie restriction, the ketogenic diet, and ketone bodies. Brain Res Rev. 2009;59:293–315.
    1. Reger MA, Henderson ST, Hale C, Cholerton B, Baker LD, Watson GS, Hyde K, Chapman D, Craft S. Effects of beta‐hydroxybutyrate on cognition in memory‐impaired adults. Neurobiol Aging. 2004;25:311–314.
    1. White H, Venkatesh B. Clinical review: ketones and brain injury. Crit Care. 2011;15:219.
    1. Lopaschuk GD, Ussher JR, Folmes CD, Jaswal JS, Stanley WC. Myocardial fatty acid metabolism in health and disease. Physiol Rev. 2010;90:207–258.
    1. Stanley WC, Meadows SR, Kivilo KM, Roth BA, Lopaschuk GD. Beta‐hydroxybutyrate inhibits myocardial fatty acid oxidation in vivo independent of changes in malonyl‐CoA content. Am J Physiol Heart Circ Physiol. 2003;285:H1626–H1631.
    1. Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, Grueter CA, Lim H, Saunders LR, Stevens RD, Newgard CB, Farese RV Jr, de Cabo R, Ulrich S, Akassoglou K, Verdin E. Suppression of oxidative stress by beta‐hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science. 2013;339:211–214.
    1. Aubert G, Martin OJ, Horton JL, Lai L, Vega RB, Leone TC, Koves T, Gardell SJ, Kruger M, Hoppel CL, Lewandowski ED, Crawford PA, Muoio DM, Kelly DP. The failing heart relies on ketone bodies as a fuel. Circulation. 2016;133:698–705.
    1. Bedi KC Jr, Snyder NW, Brandimarto J, Aziz M, Mesaros C, Worth AJ, Wang LL, Javaheri A, Blair IA, Margulies KB, Rame JE. Evidence for intramyocardial disruption of lipid metabolism and increased myocardial ketone utilization in advanced human heart failure. Circulation. 2016;133:706–716.
    1. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE; Investigators E‐RO . Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117–2128.
    1. Mudaliar S, Alloju S, Henry RR. Can a shift in fuel energetics explain the beneficial cardiorenal outcomes in the EMPA‐REG OUTCOME study? A unifying hypothesis. Diabetes Care. 2016;39:1115–1122.
    1. Mather KJ, Steinberg HO, Baron AD. Insulin resistance in the vasculature. J Clin Invest. 2013;123:1003–1004.
    1. Crea F, Camici PG, Bairey Merz CN. Coronary microvascular dysfunction: an update. Eur Heart J. 2014;35:1101–1111.
    1. Shah MS, Brownlee M. Molecular and cellular mechanisms of cardiovascular disorders in diabetes. Circ Res. 2016;118:1808–1829.
    1. Hasselbalch SG, Madsen PL, Hageman LP, Olsen KS, Justesen N, Holm S, Paulson OB. Changes in cerebral blood flow and carbohydrate metabolism during acute hyperketonemia. Am J Physiol. 1996;270:E746–E751.
    1. Fioretto P, Trevisan R, Velussi M, Cernigoi A, De Riva C, Bressan M, Doria A, Pauletto N, Angeli P, De Dona C, Nosadini R. Glomerular filtration rate is increased in man by the infusion of both D, L‐3‐hydroxybutyric acid and sodium D, L‐3‐hydroxybutyrate. J Clin Endocrinol Metab. 1987;65:331–338.
    1. Bing RJ, Siegel A, Ungar I, Gilbert M. Metabolism of the human heart. II. Studies on fat, ketone and amino acid metabolism. Am J Med. 1954;16:504–515.
    1. Landau BR, Wahren J. Nonproductive exchanges: the use of isotopes gone astray. Metabolism. 1992;41:457–459.
    1. Sorensen LK, Rittig NF, Holmquist EF, Jorgensen KA, Jorgensen JO, Moller N, Johannsen M. Simultaneous determination of beta‐hydroxybutyrate and beta‐hydroxy‐beta‐methylbutyrate in human whole blood using hydrophilic interaction liquid chromatography electrospray tandem mass spectrometry. Clin Biochem. 2013;46:1877–1883.
    1. Harms HJ, Knaapen P, de Haan S, Halbmeijer R, Lammertsma AA, Lubberink M. Automatic generation of absolute myocardial blood flow images using [15O]H2O and a clinical PET/CT scanner. Eur J Nucl Med Mol Imaging. 2011;38:930–939.
    1. Bergmann SR, Weinheimer CJ, Markham J, Herrero P. Quantitation of myocardial fatty acid metabolism using PET. J Nucl Med. 1996;37:1723–1730.
    1. Guiducci L, Jarvisalo M, Kiss J, Nagren K, Viljanen A, Naum AG, Gastaldelli A, Savunen T, Knuuti J, Salvadori PA, Ferrannini E, Nuutila P, Iozzo P. [11C]palmitate kinetics across the splanchnic bed in arterial, portal and hepatic venous plasma during fasting and euglycemic hyperinsulinemia. Nucl Med Biol. 2006;33:521–528.
    1. Patlak CS, Blasberg RG. Graphical evaluation of blood‐to‐brain transfer constants from multiple‐time uptake data. Generalizations. J Cereb Blood Flow Metab. 1985;5:584–590.
    1. Botker HE, Goodwin GW, Holden JE, Doenst T, Gjedde A, Taegtmeyer H. Myocardial glucose uptake measured with fluorodeoxyglucose: a proposed method to account for variable lumped constants. J Nucl Med. 1999;40:1186–1196.
    1. Crawford PA, Crowley JR, Sambandam N, Muegge BD, Costello EK, Hamady M, Knight R, Gordon JI. Regulation of myocardial ketone body metabolism by the gut microbiota during nutrient deprivation. Proc Natl Acad Sci USA. 2009;106:11276–11281.
    1. Pelletier A, Tardif A, Gingras MH, Chiasson JL, Coderre L. Chronic exposure to ketone bodies impairs glucose uptake in adult cardiomyocytes in response to insulin but not vanadate: the role of PI3‐K. Mol Cell Biochem. 2007;296:97–108.
    1. Garland PB, Newsholme EA, Randle PJ. Regulation of glucose uptake by muscle. 9. Effects of fatty acids and ketone bodies, and of alloxan‐diabetes and starvation, on pyruvate metabolism and on lactate‐pyruvate and L‐glycerol 3‐phosphate‐dihydroxyacetone phosphate concentration ratios in rat heart and rat diaphragm muscles. Biochem J. 1964;93:665–678.
    1. Russell RR III, Taegtmeyer H. Coenzyme A sequestration in rat hearts oxidizing ketone bodies. J Clin Invest. 1992;89:968–973.
    1. Ferrannini E, Santoro D, Bonadonna R, Natali A, Parodi O, Camici PG. Metabolic and hemodynamic effects of insulin on human hearts. Am J Physiol. 1993;264:E308–E315.
    1. Baron AD. Vascular reactivity. Am J Cardiol. 1999;84:25J–27J.
    1. Depre C, Vanoverschelde JL, Taegtmeyer H. Glucose for the heart. Circulation. 1999;99:578–588.
    1. Taylor M, Wallhaus TR, Degrado TR, Russell DC, Stanko P, Nickles RJ, Stone CK. An evaluation of myocardial fatty acid and glucose uptake using PET with [18F]fluoro‐6‐thia‐heptadecanoic acid and [18F]FDG in patients with congestive heart failure. J Nucl Med. 2001;42:55–62.
    1. O'Donnell JM, Kudej RK, LaNoue KF, Vatner SF, Lewandowski ED. Limited transfer of cytosolic NADH into mitochondria at high cardiac workload. Am J Physiol Heart Circ Physiol. 2004;286:H2237–H2242.
    1. Kobayashi K, Neely JR. Control of maximum rates of glycolysis in rat cardiac muscle. Circ Res. 1979;44:166–175.
    1. Kuang M, Febbraio M, Wagg C, Lopaschuk GD, Dyck JR. Fatty acid translocase/CD36 deficiency does not energetically or functionally compromise hearts before or after ischemia. Circulation. 2004;109:1550–1557.
    1. Luiken JJ, Coort SL, Koonen DP, van der Horst DJ, Bonen A, Zorzano A, Glatz JF. Regulation of cardiac long‐chain fatty acid and glucose uptake by translocation of substrate transporters. Pflugers Arch. 2004;448:1–15.
    1. van der Vusse GJ. The fascinating and elusive life of cardiac fatty acids. Cardiovasc Res. 2011;92:363–364.
    1. Kudej RK, Fasano M, Zhao X, Lopaschuk GD, Fischer SK, Vatner DE, Vatner SF, Lewandowski ED. Second window of preconditioning normalizes palmitate use for oxidation and improves function during low‐flow ischaemia. Cardiovasc Res. 2011;92:394–400.
    1. Vanoverschelde JL, Wijns W, Kolanowski J, Bol A, Decoster PM, Michel C, Cogneau M, Heyndrickx GR, Essamri B, Melin JA. Competition between palmitate and ketone bodies as fuels for the heart: study with positron emission tomography. Am J Physiol. 1993;264:H701–H707.
    1. Duncker DJ, Bache RJ. Regulation of coronary blood flow during exercise. Physiol Rev. 2008;88:1009–1086.
    1. Heusch G. Heart rate in the pathophysiology of coronary blood flow and myocardial ischaemia: benefit from selective bradycardic agents. Br J Pharmacol. 2008;153:1589–1601.
    1. Wang HH, Katz RL. Effects of changes in coronary blood pH on the heart. Circ Res. 1965;17:114–122.
    1. Walker M, Fulcher GR, Marsiaj H, Orskov H, Alberti KG. The independent effect of ketone bodies on forearm glucose metabolism in normal man. Scand J Clin Lab Invest. 1991;51:605–613.
    1. Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S, Kobayashi M, Hirasawa A, Tsujimoto G. Short‐chain fatty acids and ketones directly regulate sympathetic nervous system via G protein‐coupled receptor 41 (GPR41). Proc Natl Acad Sci USA. 2011;108:8030–8035.
    1. McNeely JD, Windham BG, Anderson DE. Dietary sodium effects on heart rate variability in salt sensitivity of blood pressure. Psychophysiology. 2008;45:405–411.
    1. Zajac A, Poprzecki S, Maszczyk A, Czuba M, Michalczyk M, Zydek G. The effects of a ketogenic diet on exercise metabolism and physical performance in off‐road cyclists. Nutrients. 2014;6:2493–2508.
    1. Miles JM, Nissen SL, Rizza RA, Gerich JE, Haymond MW. Failure of infused beta‐hydroxybutyrate to decrease proteolysis in man. Diabetes. 1983;32:197–205.
    1. Blomqvist G, Alvarsson M, Grill V, Von Heijne G, Ingvar M, Thorell JO, Stone‐Elander S, Widen L, Ekberg K. Effect of acute hyperketonemia on the cerebral uptake of ketone bodies in nondiabetic subjects and IDDM patients. Am J Physiol Endocrinol Metab. 2002;283:E20–E28.
    1. Muniyappa R, Montagnani M, Koh KK, Quon MJ. Cardiovascular actions of insulin. Endocr Rev. 2007;28:463–491.
    1. Taggart AK, Kero J, Gan X, Cai TQ, Cheng K, Ippolito M, Ren N, Kaplan R, Wu K, Wu TJ, Jin L, Liaw C, Chen R, Richman J, Connolly D, Offermanns S, Wright SD, Waters MG. (D)‐beta‐hydroxybutyrate inhibits adipocyte lipolysis via the nicotinic acid receptor PUMA‐G. J Biol Chem. 2005;280:26649–26652.
    1. Gormsen LC, Jessen N, Gjedsted J, Gjedde S, Norrelund H, Lund S, Christiansen JS, Nielsen S, Schmitz O, Moller N. Dose‐response effects of free fatty acids on glucose and lipid metabolism during somatostatin blockade of growth hormone and insulin in humans. J Clin Endocrinol Metab. 2007;92:1834–1842.
    1. Du Z, Shen A, Huang Y, Su L, Lai W, Wang P, Xie Z, Xie Z, Zeng Q, Ren H, Xu D. 1H‐NMR‐based metabolic analysis of human serum reveals novel markers of myocardial energy expenditure in heart failure patients. PLoS One. 2014;9:e88102.
    1. Cox PJ, Kirk T, Ashmore T, Willerton K, Evans R, Smith A, Murray AJ, Stubbs B, West J, McLure SW, King MT, Dodd MS, Holloway C, Neubauer S, Drawer S, Veech RL, Griffin JL, Clarke K. Nutritional ketosis alters fuel preference and thereby endurance performance in athletes. Cell Metab. 2016;24:256–268.
    1. Wentz AE, d'Avignon DA, Weber ML, Cotter DG, Doherty JM, Kerns R, Nagarajan R, Reddy N, Sambandam N, Crawford PA. Adaptation of myocardial substrate metabolism to a ketogenic nutrient environment. J Biol Chem. 2010;285:24447–24456.

Source: PubMed

3
Subscribe