Metabolic and Signaling Roles of Ketone Bodies in Health and Disease

Patrycja Puchalska, Peter A Crawford, Patrycja Puchalska, Peter A Crawford

Abstract

Ketone bodies play significant roles in organismal energy homeostasis, serving as oxidative fuels, modulators of redox potential, lipogenic precursors, and signals, primarily during states of low carbohydrate availability. Efforts to enhance wellness and ameliorate disease via nutritional, chronobiological, and pharmacological interventions have markedly intensified interest in ketone body metabolism. The two ketone body redox partners, acetoacetate and D-β-hydroxybutyrate, serve distinct metabolic and signaling roles in biological systems. We discuss the pleiotropic roles played by both of these ketones in health and disease. While enthusiasm is warranted, prudent procession through therapeutic applications of ketogenic and ketone therapies is also advised, as a range of metabolic and signaling consequences continue to emerge. Organ-specific and cell-type-specific effects of ketone bodies are important to consider as prospective therapeutic and wellness applications increase.

Keywords: ketones and SGLT inhibitors; ketones and cancer; ketones and fatty liver disease; ketones and heart failure; ketones and neurodegenerative disease; ketones and the gut.

Figures

Figure 1
Figure 1
Overview of organismal physiology and turnover. Ketone bodies are primarily generated within hepatic mitochondria from fatty acid–derived acetyl-CoA by the sequence of metabolic reactions requiring the fate-committing enzyme HMGCS2. AcAc and βOHB are transported in circulation to extrahepatic tissue for terminal oxidation through reactions requiring the enzyme SCOT. Adipose tissue lipolysis is inhibited by βOHB, creating a negative feedback loop. Question marks represent the uncertain molecular identity of mitochondrial ketone transporter(s). Abbreviations: AcAc, acetoacetate; ATP, adenosine triphosphate; BDH1, D-β-hydroxybutyrate dehydrogenase 1; βOHB, β-hydroxybutyrate; βox, β-oxidation; CoA, coenzyme A; CoA-SH, CoA sodium salt hydrate; CPT, carnitine palmitoyltransferase; e−, electron; CS, citrate synthase; ETC, electron transport chain; HMGCL, 3-hydroxymethylglutaryl-CoA lyase; HMGCS2, 3-hydroxymethylglutaryl-CoA synthase 2; mThiolase, mitochondrial thiolase; SCOT, succinyl-CoA:3-oxoacid-CoA transferase; TCA, tricarboxylic acid. Figure adapted from images created with BioRender.com.
Figure 2
Figure 2
Ketone body metabolic fates beyond terminal oxidation. Ketone body metabolism in hepatic or extrahepatic cytosol is integrated with nonoxidative pathways such as cholesterogenesis and DNL. In extrahepatic cells, AcAc carbon converges with itaconate and HBPs in mitochondrial and cytosolic compartments, respectively. Question marks represent the uncertain molecular identity of mitochondrial ketone transporter(s). Abbreviations: AACS, AcAc-CoA synthetase; AcAc, acetoacetate; ACC, acetyl-CoA carboxylase; ACLY, adenosine triphosphate citrate lyase; ACSS2, acetyl-CoA synthetase; βox, β-oxidation; CIC, citrate transporter; CoA, coenzyme A; cThiolase, cytoplasmic thiolase; DNL, de novo lipogenesis; HBP, hexosamine biosynthetic pathway; TCA, tricarboxylic acid; UDP-GlcNAc, uridine diphospho-N-acetylglucosamine. Figure adapted from images created with BioRender.com.
Figure 3
Figure 3
Regulatory mechanisms for key mitochondrial drivers of ketone body metabolism. HMGCS2 is a fate-committing ketogenic enzyme abundant in hepatocytes. BDH1 catalyzes the reduction/oxidation between AcAc and D-βOHB in hepatic and extrahepatic cells. SCOT is required for extrahepatic mitochondrial ketolysis. Abbreviations: AcAc, acetoacetate; BDH1, D-β-hydroxybutyrate dehydrogenase 1; D-βOHB, D-β-hydroxybutyrate; FGF21, fibroblast growth factor 21; HMG-CoA, 3-hydroxymethylglutaryl-CoA; HMGCS2, 3-hydroxymethylglutaryl-CoA synthase 2; IL-6, interleukin 6; KAc, lysine-acetylation; KSuc, lysine-succinylation; MG-CoA, 3-methylglutaryl-CoA; mTORC1, mammalian target of rapamycin complex 1; PI3K, phosphatidylinositol-3-kinase; PPARα, peroxisome proliferator activated receptor alpha; PTM, posttranslational modification; SCOT, succinyl-CoA:3-oxoacid-CoA transferase; SIRT, sirtuin. Figure adapted from images created with BioRender.com.
Figure 4
Figure 4
Noncanonical cellular responses to AcAc and βOHB. Ketone bodies exert different cellular effects through changes transduced by epigenetic, posttranslational, and cell surface signaling mechanisms. Abbreviations: 3-HDMP, 3-hydroxypyrrole-derivative; 3-HHD, 3-hydroxyhexane-2,5-dione; 3-HTO, 3-hexane-2,3,4-trione; AcAc, acetoacetate; bhb, lysine β-hydroxybutyrylation; βOHB, β-hydroxybutyrate; ERK, extracellular signal–regulated kinase; FFA, free fatty acid; FGF, fibroblast growth factor; GPR, G-protein coupled receptor; HDAC, histone deacetylase; HSL, hormone-sensitive lipase; IL, interleukin; LPL, lipoprotein lipase; MAPK, mitogen-activated protein kinase; MCT, monocarboxylate transporter; MEK, MAPK/ERK kinase; MetG, methylglyoxal; NFκB, nuclear factor kappa B; PLCβ, phospholipase C-β; SCFA, short-chain fatty acid; SIRT, sirtuin; SNS, sympathetic nervous system; TAG, triacylglycerol; TNF, tumor necrosis factor. Figure adapted from images created with BioRender.com.
Figure 5
Figure 5
Overview of ketone body metabolism in small and large intestine. HMGCS2 distribution and the roles of βOHB in the small and large intestine vary. In small intestine, βOHB triggers Notch signaling and Th17 immunomodulatory changes. In large intestine, anaerobic bacterial fermentation yields butyrate, a source of βOHB that influences the gut community. Abbreviations: βOHB, β-hydroxybutyrate; HDAC, histone deacetylase; HMGCS2, 3-hydroxymethylglutaryl-CoA synthase. Figure adapted from images created with BioRender.com.
Figure 6
Figure 6
Integrative role of ketone bodies in the nervous system. Mechanisms supporting the pleiotropic effects of ketone bodies or a ketogenic diet in the nervous system still require elucidation. However, effects on feeding behavior, energy expenditure, mood and behavior, and neuroprotection have all been observed. Abbreviations: Glc, glucose; KB, ketone body; TCA, tricarboxylic acid. Figure adapted from images created with BioRender.com.

References

    1. Adijanto J, Du J, Moffat C, Seifert EL, Hurley JB, Philp NJ. 2014. The retinal pigment epithelium utilizes fatty acids for ketogenesis: implications for metabolic coupling with the outer retina. J. Biol. Chem 289:20570–82
    1. Al Batran R, Gopal K, Capozzi ME, Chahade JJ, Saleme B, et al. 2020. Pimozide alleviates hyperglycemia in diet-induced obesity by inhibiting skeletal muscle ketone oxidation. Cell Metab. 31:909–19. e8
    1. Ang QY, Alexander M, Newman JC, Tian Y, Cai J, et al. 2020. Ketogenic diets alter the gut microbiome resulting in decreased intestinal Th17 cells. Cell 181:1263–75. e16
    1. Aubert G, Martin OJ, Horton JL, Lai L, Vega RB, et al. 2016. The failing heart relies on ketone bodies as a fuel. Circulation 133:698–705
    1. Badman MK, Pissios P, Kennedy AR, Koukos G, Flier JS, Maratos-Flier E. 2007. Hepatic fibroblast growth factor 21 is regulated by PPARα and is a key mediator of hepatic lipid metabolism in ketotic states. Cell Metab. 5:426–37
    1. Balasse EO, Fery F. 1989. Ketone body production and disposal: effects of fasting, diabetes, and exercise. Diabetes Metab. Rev 5:247–70
    1. Bedi KC Jr., Snyder NW, Brandimarto J, Aziz M, Mesaros C, et al. 2016. Evidence for intramyocardial disruption of lipid metabolism and increased myocardial ketone utilization in advanced human heart failure. Circulation 133:706–16
    1. Beland-Millar A, Takimoto M, Hamada T, Messier C. 2020. Brain and muscle adaptation to high-fat diets and exercise: metabolic transporters, enzymes and substrates in the rat cortex and muscle. Brain Res. 1749:147126.
    1. Benyó Z, Gille A, Kero J, Csiky M, Suchánková MC, et al. 2005. GPR109A (PUMA-G/HM74A) mediates nicotinic acid-induced flushing. J. Biol. Chem 115:3634–40
    1. Bergstrom JD, Wong GA, Edwards PA, Edmond J. 1984. The regulation of acetoacetyl-CoA synthetase activity by modulators of cholesterol synthesis in vivo and the utilization of acetoacetate for cholesterogenesis. J. Biol. Chem 259:14548–53
    1. BonDurant LD, Potthoff MJ. 2018. Fibroblast growth factor 21: a versatile regulator of metabolic homeostasis. Annu. Rev. Nutr 38:173–96
    1. Bregere C, Rebrin I, Gallaher TK, Sohal RS. 2010. Effects of age and calorie restriction on tryptophan nitration, protein content, and activity of succinyl-CoA:3-ketoacid CoA transferase in rat kidney mitochondria. Free Radic. Biol. Med 48:609–18
    1. Bricker DK, Taylor EB, Schell JC, Orsak T, Boutron A, et al. 2012. A mitochondrial pyruvate carrier required for pyruvate uptake in yeast, Drosophila, and humans. Science 337:96–100
    1. Browning JD, Baxter J, Satapati S, Burgess SC. 2012. The effect of short-term fasting on liver and skeletal muscle lipid, glucose, and energy metabolism in healthy women and men. J. Lipid Res 53:577–86
    1. Byrne NJ, Matsumura N, Maayah ZH, Ferdaoussi M, Takahara S, et al. 2020. Empagliflozin blunts worsening cardiac dysfunction associated with reduced NLRP3 (nucleotide-binding domain-like receptor protein 3) inflammasome activation in heart failure. Circ. Heart Fail 13:e006277.
    1. Byrne NJ, Parajuli N, Levasseur JL, Boisvenue J, Beker DL, et al. 2017. Empagliflozin prevents worsening of cardiac function in an experimental model of pressure overload-induced heart failure. JACC Basic Transl. Sci 2:347–54
    1. Byrne NJ, Soni S, Takahara S, Ferdaoussi M, Al Batran R, et al. 2020. Chronically elevating circulating ketones can reduce cardiac inflammation and blunt the development of heart failure. Circ. Heart Fail 13:e006573.
    1. Caffa I, Spagnolo V, Vernieri C, Valdemarin F, Becherini P, et al. 2020. Fasting-mimicking diet and hormone therapy induce breast cancer regression. Nature 583:620–24
    1. Cahill GF Jr. 2006. Fuel metabolism in starvation. Annu. Rev. Nutr 26:1–22
    1. Camarero N, Mascaró C, Mayordomo C, Vilardell F, Haro D, Marrero PF. 2006. Ketogenic HMGCS2 is a c-Myc target gene expressed in differentiated cells of human colonic epithelium and down-regulated in colon cancer. Mol. Cancer Res 4:645–53
    1. Carley AN, Taegtmeyer H, Lewandowski ED. 2014. Mechanisms linking energy substrate metabolism to the function of the heart. Circ. Res 114:717–29
    1. Carneiro L, Geller S, Fioramonti X, Hébert A, Repond C, et al. 2016. Evidence for hypothalamic ketone body sensing: impact on food intake and peripheral metabolic responses in mice. Am. J. Physiol. Endocrinol. Metab 310:E103–15
    1. Chakraborty S, Galla S, Cheng X, Yeo JY, Mell B, et al. 2018. Salt-responsive metabolite, β-hydroxybutyrate, attenuates hypertension. Cell Rep. 25:677–89. e4
    1. Chavan R, Feillet C, Costa SS, Delorme JE, Okabe T, et al. 2016. Liver-derived ketone bodies are necessary for food anticipation. Nat. Commun 7:10580.
    1. Chen SW, Chou CT, Chang CC, Li YJ, Chen ST, et al. 2017. HMGCS2 enhances invasion and metastasis via direct interaction with PPARα to activate Src signaling in colorectal cancer and oral cancer. Oncotarget 8:22460–76
    1. Chen YJ, Mahieu NG, Huang X, Singh M, Crawford PA, et al. 2016. Lactate metabolism is associated with mammalian mitochondria. Nat. Chem. Biol 12:937–43
    1. Cheng A, Wang J, Ghena N, Zhao Q, Perone I, et al. 2020. SIRT3 haploinsufficiency aggravates loss of GABAergic interneurons and neuronal network hyperexcitability in an Alzheimer’s disease model. J. Neurosci 40:694–709
    1. Cheng CW, Biton M, Haber AL, Gunduz N, Eng G, et al. 2019. Ketone body signaling mediates intestinal stem cell homeostasis and adaptation to diet. Cell 178:1115–31. e15
    1. Cherbuy C, Andrieux C, Honvo-Houeto E, Thomas M, Ide C, et al. 2004. Expression of mitochondrial HMGCoA synthase and glutaminase in the colonic mucosa is modulated by bacterial species. Eur. J. Biochem 271:87–95
    1. Cherbuy C, Darcy-Vrillon B, Morel MT, Pegorier JP, Duee PH. 1995. Effect of germfree state on the capacities of isolated rat colonocytes to metabolize n-butyrate, glucose, and glutamine. Gastroenterology 109:1890–99
    1. Chriett S, Dabek A, Wojtala M, Vidal H, Balcerczyk A, Pirola L. 2019. Prominent action of butyrate over β-hydroxybutyrate as histone deacetylase inhibitor, transcriptional modulator and anti-inflammatory molecule. Sci. Rep 9:742.
    1. Comerford SA, Huang Z, Du X, Wang Y, Cai L, et al. 2014. Acetate dependence of tumors. Cell 159:1591–602
    1. Cotter DG, d’Avignon DA, Wentz AE, Weber ML, Crawford PA. 2011. Obligate role for ketone body oxidation in neonatal metabolic homeostasis. J. Biol. Chem 286:6902–10
    1. Cotter DG, Ercal B, Huang X, Leid JM, d’Avignon DA, et al. 2014. Ketogenesis prevents diet-induced fatty liver injury and hyperglycemia. J. Clin. Investig 124:5175–90
    1. Cotter DG, Schugar RC, Wentz AE, d’Avignon DA, Crawford PA. 2013. Successful adaptation to ketosis by mice with tissue-specific deficiency of ketone body oxidation. Am. J. Physiol. Endocrinol. Metab 304:E363–74
    1. Cox PJ, Kirk T, Ashmore T, Willerton K, Evans R, et al. 2016. Nutritional ketosis alters fuel preference and thereby endurance performance in athletes. Cell Metab. 24:256–68
    1. Crawford PA, Crowley JR, Sambandam N, Muegge BD, Costello EK, et al. 2009. Regulation of myocardial ketone body metabolism by the gut microbiota during nutrient deprivation. PNAS 106:11276–81
    1. Cui W, Luo W, Zhou X, Lu Y, Xu W, et al. 2019. Dysregulation of ketone body metabolism is associated with poor prognosis for clear cell renal cell carcinoma patients. Front. Oncol 9:1422.
    1. Cunnane SC, Trushina E, Morland C, Prigione A, Casadesus G, et al. 2020. Brain energy rescue: an emerging therapeutic concept for neurodegenerative disorders of ageing. Nat. Rev. Drug Discov 19:609–33
    1. d’Avignon DA, Puchalska P, Ercal B, Chang Y, Martin SE, et al. 2018. Hepatic ketogenic insufficiency reprograms hepatic glycogen metabolism and the lipidome. JCI Insight 3(12):e99762
    1. Davis JD, Wirtshafter D, Asin KE, Brief D. 1981. Sustained intracerebroventricular infusion of brain fuels reduces body weight and food intake in rats. Science 212:81–83
    1. Davuluri G, Song P, Liu Z, Wald D, Sakaguchi TF, et al. 2016. Inactivation of 3-hydroxybutyrate dehydrogenase 2 delays zebrafish erythroid maturation by conferring premature mitophagy. PNAS 113:E1460–69
    1. de Cabo R, Mattson MP. 2019. Effects of intermittent fasting on health, aging, and disease. N. Engl. J. Med 381:2541–51
    1. Dittenhafer-Reed KE, Richards AL, Fan J, Smallegan MJ, Fotuhi Siahpirani A, et al. 2015. SIRT3 mediates multi-tissue coupling for metabolic fuel switching. Cell Metab. 21:637–46
    1. Felig P, Wahren J, Hendler R, Brundin T. 1974. Splanchnic glucose and amino acid metabolism in obesity. J. Biol. Chem 53:582–90
    1. Ferrannini E, Baldi S, Frascerra S, Astiarraga B, Heise T, et al. 2016. Shift to fatty substrate utilization in response to sodium-glucose cotransporter 2 inhibition in subjects without diabetes and patients with type 2 diabetes. Diabetes 65:1190–95
    1. Fisher-Wellman KH, Draper JA, Davidson MT, Williams AS, Narowski TM, et al. 2019. Respiratory phenomics across multiple models of protein hyperacylation in cardiac mitochondria reveals a marginal impact on bioenergetics. Cell Rep. 26:1557–72. e8
    1. Fletcher JA, Deja S, Satapati S, Fu X, Burgess SC, Browning JD. 2019. Impaired ketogenesis and increased acetyl-CoA oxidation promote hyperglycemia in human fatty liver. JCI Insight 5:e127737
    1. Flint TR, Janowitz T, Connell CM, Roberts EW, Denton AE, et al. 2016. Tumor-induced IL-6 reprograms host metabolism to suppress anti-tumor immunity. Cell Metab. 24:672–84
    1. Fu SP, Liu BR, Wang JF, Xue WJ, Liu HM, et al. 2015. β-Hydroxybutyric acid inhibits growth hormone-releasing hormone synthesis and secretion through the GPR109A/extracellular signal-regulated 1/2 signalling pathway in the hypothalamus. J. Neuroendocrinol 27:212–22
    1. Fukao T, Lopaschuk GD, Mitchell GA. 2004. Pathways and control of ketone body metabolism: on the fringe of lipid biochemistry. Prostaglandins Leukot. Essent. Fatty Acids 70:243–51
    1. Gambhir D, Ananth S, Veeranan-Karmegam R, Elangovan S, Hester S, et al. 2012. GPR109A as an anti-inflammatory receptor in retinal pigment epithelial cells and its relevance to diabetic retinopathy. Investig. Ophthalmol. Vis. Sci 53:2208–17
    1. García-Caballero M, Zecchin A, Souffreau J, Truong ACK, Teuwen LA, et al. 2019. Role and therapeutic potential of dietary ketone bodies in lymph vessel growth. Nat. Metab 1:666–75
    1. Goedeke L, Peng L, Montalvo-Romeral V, Butrico GM, Dufour S, et al. 2019. Controlled-release mitochondrial protonophore (CRMP) reverses dyslipidemia and hepatic steatosis in dysmetabolic nonhuman primates. Sci. Transl. Med 11:eaay0284.
    1. Gormsen LC, Svart M, Thomsen HH, Sondergaard E, Vendelbo MH, et al. 2017. Ketone body infusion with 3-hydroxybutyrate reduces myocardial glucose uptake and increases blood flow in humans: a positron emission tomography study. J. Am. Heart Assoc 6:e005066.
    1. Green A, Bishop RE. 2019. Ketoacidosis—Where do the protons come from? Trends Biochem. Sci 44:484–89
    1. Grimsrud PA, Carson JJ, Hebert AS, Hubler SL, Niemi NM, et al. 2012. A quantitative map of the liver mitochondrial phosphoproteome reveals posttranslational control of ketogenesis. Cell Metab. 16:672–83
    1. Grinblat L, Pacheco Bolanos LF, Stoppani AO. 1986. Decreased rate of ketone-body oxidation and decreased activity of D-3-hydroxybutyrate dehydrogenase and succinyl-CoA:3-oxo-acid CoA-transferase in heart mitochondria of diabetic rats. Biochem. J 240:49–56
    1. Guzman M, Blazquez C. 2001. Is there an astrocyte-neuron ketone body shuttle? Trends Endocrinol. Metab 12:169–73
    1. Halestrap AP. 2012. The monocarboxylate transporter family—structure and functional characterization. IUBMB Life 64:1–9
    1. Halestrap AP, Wilson MC. 2012. The monocarboxylate transporter family—role and regulation. IUBMB Life 64:109–19
    1. Han YM, Bedarida T, Ding Y, Somba BK, Lu Q, et al. 2018. β-Hydroxybutyrate prevents vascular senescence through hnRNP A1-mediated upregulation of Oct4. Mol. Cell 71:1064–78. e5
    1. Harrison HC, Long CNH. 1940. The distribution of ketone bodies in tissues. J. Biol. Chem 133:209–18
    1. Hasegawa S, Noda K, Maeda A, Matsuoka M, Yamasaki M, Fukui T. 2012. Acetoacetyl-CoA synthetase, a ketone body-utilizing enzyme, is controlled by SREBP-2 and affects serum cholesterol levels. Mol. Genet. Metab 107:553–60
    1. Hashimoto T, Masuda S, Taguchi S, Brooks GA. 2005. Immunohistochemical analysis of MCT1, MCT2 and MCT4 expression in rat plantaris muscle. J. Physiol 567:121–29
    1. Hasselbalch SG, Madsen PL, Hageman LP, Olsen KS, Justesen N, et al. 1996. Changes in cerebral blood flow and carbohydrate metabolism during acute hyperketonemia. Am. J. Physiol. Endocrinol. Metab 270:E746–51
    1. Hegardt FG. 1999. Mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase: a control enzyme in ketogenesis. Biochem. J 338(Part 3):569–82
    1. Hennebelle M, Courchesne-Loyer A, St-Pierre V, Vandenberghe C, Castellano CA, et al. 2016. Preliminary evaluation of a differential effect of an α-linolenate-rich supplement on ketogenesis and plasma omega-3 fatty acids in young and older adults. Nutrition 32:1211–16
    1. Henning SJ, Hird FJ. 1972. Ketogenesis from butyrate and acetate by the caecum and the colon of rabbits. Biochem. J 130:785–90
    1. Ho KL, Karwi QG, Wagg C, Zhang L, Vo K, et al. 2020. Ketones can become the major fuel source for the heart but do not increase cardiac efficiency. Cardiovasc. Res 117(4):1178–87
    1. Ho KL, Zhang L, Wagg C, Al Batran R, Gopal K, et al. 2019. Increased ketone body oxidation provides additional energy for the failing heart without improving cardiac efficiency. Cardiovasc. Res 115:1606–16
    1. Hopkins BD, Pauli C, Du X, Wang DG, Li X, et al. 2018. Suppression of insulin feedback enhances the efficacy of PI3K inhibitors. Nature 560:499–503
    1. Horton JL, Davidson MT, Kurishima C, Vega RB, Powers JC, et al. 2019. The failing heart utilizes 3-hydroxybutyrate as a metabolic stress defense. JCI Insight 4:e124079
    1. Huang CK, Chang PH, Kuo WH, Chen CL, Jeng YM, et al. 2017. Adipocytes promote malignant growth of breast tumours with monocarboxylate transporter 2 expression via β-hydroxybutyrate. Nat. Commun 8:14706.
    1. Huang D, Li T, Wang L, Zhang L, Yan R, et al. 2016. Hepatocellular carcinoma redirects to ketolysis for progression under nutrition deprivation stress. Cell Res. 26:1112–30
    1. Huang H, Zhang D, Weng Y, Delaney K, Tang Z, et al. 2021. The regulatory enzymes and protein substrates for the lysine β-hydroxybutyrylation pathway. Sci. Adv 7:eabe2771.
    1. Hyde PN, Sapper TN, Crabtree CD, LaFountain RA, Bowling ML, et al. 2019. Dietary carbohydrate restriction improves metabolic syndrome independent of weight loss. JCI Insight 4(12):e128308
    1. Iozzo P, Bucci M, Roivainen A, Nagren K, Jarvisalo MJ, et al. 2010. Fatty acid metabolism in the liver, measured by positron emission tomography, is increased in obese individuals. Gastroenterology 139:846–56. e6
    1. Ishimwe JA, Garrett MR, Sasser JM. 2020. 1,3-Butanediol attenuates hypertension and suppresses kidney injury in female rats. Am. J. Physiol. Ren. Physiol 319:F106–14
    1. Jensen-Cody SO, Flippo KH, Claflin KE, Yavuz Y, Sapouckey SA, et al. 2020. FGF21 signals to glutamatergic neurons in the ventromedial hypothalamus to suppress carbohydrate intake. Cell Metab. 32:273–86
    1. Kajitani N, Iwata M, Miura A, Tsunetomi K, Yamanashi T, et al. 2020. Prefrontal cortex infusion of beta-hydroxybutyrate, an endogenous NLRP3 inflammasome inhibitor, produces antidepressant-like effects in a rodent model of depression. Neuropsychopharmacol. Rep 40:157–65
    1. Kakehashi A, Stefanov VE, Ishii N, Okuno T, Fujii H, et al. 2017. Proteome characteristics of non-alcoholic steatohepatitis liver tissue and associated hepatocellular carcinomas. Int. J. Mol. Sci 18(2):434
    1. Kang HB, Fan J, Lin R, Elf S, Ji Q, et al. 2015. Metabolic rewiring by oncogenic BRAF V600E links ketogenesis pathway to BRAF-MEK1 signaling. Mol. Cell 59:345–58
    1. Karwi QG, Biswas D, Pulinilkunnil T, Lopaschuk GD. 2020. Myocardial ketones metabolism in heart failure. J. Card. Fail 26:998–1005
    1. Kashiwaya Y, Sato K, Tsuchiya N, Thomas S, Fell DA, et al. 1994. Control of glucose utilization in working perfused rat heart. J. Biol. Chem 269:25502–14
    1. Katsu-Jimenez Y, Gimenez-Cassina A. 2019. Fibroblast growth factor-21 promotes ketone body utilization in neurons through activation of AMP-dependent kinase. Mol. Cell. Neurosci 101:103415.
    1. Kennedy AR, Pissios P, Otu H, Xue B, Asakura K, et al. 2007. A high-fat, ketogenic diet induces a unique metabolic state in mice. Am. J. Physiol. Endocrinol. Metab 292:E1724–39
    1. Kim CW, Addy C, Kusunoki J, Anderson NN, Deja S, et al. 2017. Acetyl CoA carboxylase inhibition reduces hepatic steatosis but elevates plasma triglycerides in mice and humans: a bedside to bench investigation. Cell Metab. 26:394–406. e6
    1. Kim SR, Lee SG, Kim SH, Kim JH, Choi E, et al. 2020. SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease. Nat. Commun 11:2127.
    1. Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, et al. 2011. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). PNAS 108:8030–35
    1. Koliaki C, Szendroedi J, Kaul K, Jelenik T, Nowotny P, et al. 2015. Adaptation of hepatic mitochondrial function in humans with non-alcoholic fatty liver is lost in steatohepatitis. Cell Metab. 21:739–46
    1. Koppel SJ, Swerdlow RH. 2018. Neuroketotherapeutics: a modern review of a century-old therapy. Neurochem. Int 117:114–25
    1. Koutnik AP, Poff AM, Ward NP, DeBlasi JM, Soliven MA, et al. 2020. Ketone bodies attenuate wasting in models of atrophy. J. Cachexia Sarcopenia Muscle 11:973–96
    1. Kuhla A, Hahn S, Butschkau A, Lange S, Wree A, Vollmar B. 2014. Lifelong caloric restriction reprograms hepatic fat metabolism in mice. J. Gerontol. A Biol. Sci. Med. Sci 69:915–22
    1. Laeger T, Metges CC, Kuhla B. 2010. Role of β-hydroxybutyric acid in the central regulation of energy balance. Appetite 54:450–55
    1. Lai L, Leone TC, Keller MP, Martin OJ, Broman AT, et al. 2014. Energy metabolic reprogramming in the hypertrophied and early stage failing heart: a multisystems approach. Circ. Heart Fail 7:1022–31
    1. Le Foll C, Levin BE. 2016. Fatty acid-induced astrocyte ketone production and the control of food intake. Am. J. Physiol. Regul. Integr. Comp. Physiol 310:R1186–92
    1. Leclercq S, Le Roy T, Furgiuele S, Coste V, Bindels LB, et al. 2020. Gut microbiota-induced changes in β-hydroxybutyrate metabolism are linked to altered sociability and depression in alcohol use disorder. Cell Rep. 33:108238.
    1. Lee J, Choi J, Selen Alpergin ES, Zhao L, Hartung T, et al. 2017. Loss of hepatic mitochondrial long-chain fatty acid oxidation confers resistance to diet-induced obesity and glucose intolerance. Cell Rep. 20:655–67
    1. Lehninger AL, Sudduth HC, Wise JB. 1960. D-β-Hydroxybutyric dehydrogenase of mitochondria. J. Biol. Chem 235:2450–55
    1. Li B, Yu Y, Liu K, Zhang Y, Geng Q, et al. 2021. β-Hydroxybutyrate inhibits histone deacetylase 3 to promote claudin-5 generation and attenuate cardiac microvascular hyperpermeability in diabetes. Diabetologia 64:226–39
    1. Lien EC, Vander Heiden MG. 2019. A framework for examining how diet impacts tumour metabolism. Nat. Rev. Cancer 19:651–61
    1. Lommi J, Kupari M, Koskinen P, Naveri H, Leinonen H, et al. 1996. Blood ketone bodies in congestive heart failure. J. Am. Coll. Cardiol 28:665–72
    1. London ED, Margolin RA, Duara R, Holloway HW, Robertson-Tchabo EA, et al. 1986. Effects of fasting on ketone body concentrations in healthy men of different ages. J. Gerontol 41:599–604
    1. Martinez-Outschoorn UE, Lin Z, Whitaker-Menezes D, Howell A, Lisanti MP, Sotgia F. 2012. Ketone bodies and two-compartment tumor metabolism: Stromal ketone production fuels mitochondrial biogenesis in epithelial cancer cells. Cell Cycle 11(21):3956–63
    1. McCommis KS, Chen Z, Fu X, McDonald WG, Colca JR, et al. 2015. Loss of mitochondrial pyruvate carrier 2 in the liver leads to defects in gluconeogenesis and compensation via pyruvate-alanine cycling. Cell Metab. 22:682–94
    1. McGarry JD, Foster DW. 1971. The regulation of ketogenesis from octanoic acid. The role of the tricarboxylic acid cycle and fatty acid synthesis. J. Biol. Chem 246:1149–59
    1. McGarry JD, Foster DW. 1980. Regulation of hepatic fatty acid oxidation and ketone body production. Annu. Rev. Biochem 49:395–420
    1. Miller VJ, LaFountain RA, Barnhart E, Sapper TS, Short J, et al. 2020. A ketogenic diet combined with exercise alters mitochondrial function in human skeletal muscle while improving metabolic health. Am.J. Physiol. Endocrinol. Metab 319:E995–1007
    1. Miyamoto J, Ohue-Kitano R, Mukouyama H, Nishida A, Watanabe K, et al. 2019. Ketone body receptor GPR43 regulates lipid metabolism under ketogenic conditions. PNAS 116:23813–21
    1. Mujica-Parodi LR, Amgalan A, Sultan SF, Antal B, Sun X, et al. 2020. Diet modulates brain network stability, a biomarker for brain aging, in young adults. PNAS 117:6170–77
    1. Murashige D, Jang C, Neinast M, Edwards JJ, Cowan A, et al. 2020. Comprehensive quantification of fuel use by the failing and nonfailing human heart. Science 370:364–68
    1. Neudorf H, Myette-Cote E, Little JP. 2020. The impact of acute ingestion of a ketone monoester drink on LPS-stimulated NLRP3 activation in humans with obesity. Nutrients 12(3):854
    1. Neufer PD. 2019. Cutting fuel offers new clues in diabetic mystery. J. Biol. Chem 294:12328–29
    1. Nielsen R, Moller N, Gormsen LC, Tolbod LP, Hansson NH, et al. 2019. Cardiovascular effects of treatment with the ketone body 3-hydroxybutyrate in chronic heart failure patients. Circulation 139:2129–41
    1. Orii KE, Fukao T, Song XQ, Mitchell GA, Kondo N. 2008. Liver-specific silencing of the human gene encoding succinyl-CoA: 3-ketoacid CoA transferase. Tohoku J. Exp. Med 215:227–36
    1. Otsuka H, Kimura T, Ago Y, Nakama M, Aoyama Y, et al. 2020. Deficiency of 3-hydroxybutyrate dehydrogenase (BDH1) in mice causes low ketone body levels and fatty liver during fasting. J. Inherit. Metab. Dis 43:960–68
    1. Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, et al. 2020. Cardiovascular and renal outcomes with empagliflozin in heart failure. N. Engl. J. Med 383:1413–24
    1. Paradies G, Papa S. 1976. Substrate regulation of the pyruvate-transporting system in rat liver mitochondria. FEBS Lett. 62:318–21
    1. Park S, Kim DS, Daily JW. 2011. Central infusion of ketone bodies modulates body weight and hepatic insulin sensitivity by modifying hypothalamic leptin and insulin signaling pathways in type 2 diabetic rats. Brain Res. 1401:95–103
    1. Pedersen KJ. 1929. The ketonic decomposition of beta-keto carboxylic acids. J. Am. Chem. Soc 51:2098–107
    1. Peng KY, Watt MJ, Rensen S, Greve JW, Huynh K, et al. 2018. Mitochondrial dysfunction-related lipid changes occur in nonalcoholic fatty liver disease progression. J. Lipid Res 59:1977–86
    1. Petersen KF, Befroy DE, Dufour S, Rothman DL, Shulman GI. 2016. Assessment of hepatic mitochondrial oxidation and pyruvate cycling in NAFLD by 13C magnetic resonance spectroscopy. Cell Metab. 24:167–71
    1. Pissios P, Hong S, Kennedy AR, Prasad D, Liu FF, Maratos-Flier E. 2013. Methionine and choline regulate the metabolic phenotype of a ketogenic diet. Mol. Metab 2:306–13
    1. Poff AM, Koutnik AP, Egan B. 2020. Nutritional ketosis with ketogenic diets or exogenous ketones: features, convergence, and divergence. Curr. Sports Med. Rep 19:251–59
    1. Pramfalk C, Pavlides M, Banerjee R, McNeil CA, Neubauer S, et al. 2015. Sex-specific differences in hepatic fat oxidation and synthesis may explain the higher propensity for NAFLD in men. J. Clin. Endocrinol. Metab 100:4425–33
    1. Puchalska P, Crawford PA. 2017. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 25:262–84
    1. Puchalska P, Huang X, Martin SE, Han X, Patti GJ, Crawford PA. 2018. Isotope tracing untargeted metabolomics reveals macrophage polarization-state-specific metabolic coordination across intracellular compartments. iScience 9:298–313
    1. Puchalska P, Martin SE, Huang X, Lengfeld JE, Daniel B, et al. 2019. Hepatocyte-macrophage acetoacetate shuttle protects against tissue fibrosis. Cell Metab. 29:383–98. e7
    1. Puchalska P, Nelson AB, Stagg DB, Crawford PA. 2021. Determination of ketone bodies in biological samples via rapid UPLC-MS/MS. Talanta 225:122048.
    1. Quant PA, Robin D, Robin P, Girard J, Brand MD. 1989. Control of acetoacetate production from exogenous palmitoyl-CoA in isolated rat liver mitochondria. Biochem. Soc. Trans 17:1089–90
    1. Rando G, Tan CK, Khaled N, Montagner A, Leuenberger N, et al. 2016. Glucocorticoid receptor-PPARα axis in fetal mouse liver prepares neonates for milk lipid catabolism. eLife 5:e11853.
    1. Rardin MJ, He W, Nishida Y, Newman JC, Carrico C, et al. 2013. SIRT5 regulates the mitochondrial lysine succinylome and metabolic networks. Cell Metab. 18:920–33
    1. Retterstol K, Svendsen M, Narverud I, Holven KB. 2018. Effect of low carbohydrate high fat diet on LDL cholesterol and gene expression in normal-weight, young adults: a randomized controlled study. Atherosclerosis 279:52–61
    1. Riehle C, Abel ED. 2016. Insulin signaling and heart failure. Circ. Res 118:1151–69
    1. Robinson AM, Williamson DH. 1980. Physiological roles of ketone bodies as substrates and signals in mammalian tissues. Physiol. Rev 60:143–87
    1. Rodrigues LM, Uribe-Lewis S, Madhu B, Honess DJ, Stubbs M, Griffiths JR. 2017. The action of β-hydroxybutyrate on the growth, metabolism and global histone H3 acetylation of spontaneous mouse mammary tumours: evidence of a β-hydroxybutyrate paradox. Cancer Metab. 5:4.
    1. Ruiz M, Labarthe F, Fortier A, Bouchard B, Thompson Legault J, et al. 2017. Circulating acylcarnitine profile in human heart failure: a surrogate of fatty acid metabolic dysregulation in mitochondria and beyond. Am. J. Physiol. Heart Circ. Physiol 313:H768–81
    1. Salomón T, Sibbersen C, Hansen J, Britz D, Svart MV, et al. 2017. Ketone body acetoacetate buffers methylglyoxal via a non-enzymatic conversion during diabetic and dietary ketosis. Cell Chem. Biol 24:935–43. e7
    1. Samuel VT, Shulman GI. 2018. Nonalcoholic fatty liver disease as a nexus of metabolic and hepatic diseases. Cell Metab. 27:22–41
    1. Santos-Gallego CG, Requena-Ibanez JA, San Antonio R, Ishikawa K, Watanabe S, et al. 2019. Empagliflozin ameliorates adverse left ventricular remodeling in nondiabetic heart failure by enhancing myocardial energetics. J. Am. Coll. Cardiol 73:1931–44
    1. Sanyal A, Charles ED, Neuschwander-Tetri BA, Loomba R, Harrison SA, et al. 2019. Pegbelfermin (BMS-986036), a PEGylated fibroblast growth factor 21 analogue, in patients with non-alcoholic steatohepatitis: a randomised, double-blind, placebo-controlled, phase 2a trial. Lancet 392:2705–17
    1. Sasaki K, Sasaki D, Hannya A, Tsubota J, Kondo A. 2020. In vitro human colonic microbiota utilises D-β-hydroxybutyrate to increase butyrogenesis. Sci. Rep 10:8516.
    1. Satapati S, Kucejova B, Duarte JA, Fletcher JA, Reynolds L, et al. 2015. Mitochondrial metabolism mediates oxidative stress and inflammation in fatty liver. J. Biol. Chem 125:4447–62
    1. Schugar RC, Huang X, Moll AR, Brunt EM, Crawford PA. 2013. Role of choline deficiency in the fatty liver phenotype of mice fed a low protein, very low carbohydrate ketogenic diet. PLOS ONE 8:e74806.
    1. Schugar RC, Moll AR, d’Avignon DA, Weinheimer CJ, Kovacs A, Crawford PA. 2014. Cardiomyocytespecific deficiency of ketone body metabolism promotes accelerated pathological remodeling. Mol. Metab 3:754–69
    1. Selvaraj S, Kelly DP, Margulies KB. 2020. Implications of altered ketone metabolism and therapeutic ketosis in heart failure. Circulation 141:1800–12
    1. Sengupta S, Peterson TR, Laplante M, Oh S, Sabatini D. 2010. mTORC1 controls fasting-induced ketogenesis and its modulation by ageing. Nature 468:1100–4
    1. Shi X, Li X, Li D, Li Y, Song Y, et al. 2014. β-Hydroxybutyrate activates the NF-κB signaling pathway to promote the expression of pro-inflammatory factors in calf hepatocytes. Cell. Physiol. Biochem 33:920–32
    1. Shimazu T, Hirschey MD, Hua L, Dittenhafer-Reed KE, Schwer B, et al. 2010. SIRT3 deacetylates mitochondrial 3-hydroxy-3-methylglutaryl CoA synthase 2 and regulates ketone body production. Cell Metab. 12:654–61
    1. Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, et al. 2013. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339:211–14
    1. Shippy DC, Wilhelm C, Viharkumar PA, Raife TJ, Ulland TK. 2020. β-Hydroxybutyrate inhibits inflammasome activation to attenuate Alzheimer’s disease pathology. J. Neuroinflamm 17:280
    1. Shukla SK, Gebregiworgis T, Purohit V, Chaika NV, Gunda V, Radhakrishnan P. 2014. Metabolic reprogramming induced by ketone bodies diminishes pancreatic cancer cachexia. Cancer Metab. 2:18.
    1. Simithy J, Sidoli S, Yuan ZF, Coradin M, Bhanu NV, et al. 2017. Characterization of histone acylations links chromatin modifications with metabolism. Nat. Commun 8:1141.
    1. Soeters MR, Sauerwein HP, Faas L, Smeenge M, Duran M, et al. 2009. Effects of insulin on ketogenesis following fasting in lean and obese men. Obesity 17:1326–31
    1. Sondhi V, Agarwala A, Pandey RM, Chakrabarty B, Jauhari P, et al. 2020. Efficacy of ketogenic diet, modified Atkins diet, and low glycemic index therapy diet among children with drug-resistant epilepsy: a randomized clinical trial. JAMA Pediatr. 174:944–51
    1. Song JP, Chen L, Chen X, Ren J, Zhang NN, et al. 2020. Elevated plasma β-hydroxybutyrate predicts adverse outcomes and disease progression in patients with arrhythmogenic cardiomyopathy. Sci. Transl. Med 12:eaay8329.
    1. Sperry J, Condro MC, Guo L, Braas D, Vanderveer-Harris N, et al. 2020. Glioblastoma utilizes fatty acids and ketone bodies for growth allowing progression during ketogenic diet therapy. iScience 23:101453.
    1. Stubbs BJ, Cox PJ, Evans RD, Santer P, Miller JJ, et al. 2017. On the metabolism of exogenous ketones in humans. Front. Physiol 8:848.
    1. Stubbs BJ, Koutnik AP, Goldberg EL, Upadhyay V, Turnbaugh PJ, et al. 2020. Investigating ketone bodies as immunometabolic countermeasures against respiratory viral infections. Med 1:43–65
    1. Sunny NE, Parks EJ, Browning JD, Burgess SC. 2011. Excessive hepatic mitochondrial TCA cycle and gluconeogenesis in humans with nonalcoholic fatty liver disease. Cell Metab. 14:804–10
    1. Taggart AK, Kero J, Gan X, Cai TQ, Cheng K, et al. 2005. (D)-β-Hydroxybutyrate inhibits adipocyte lipolysis via the nicotinic acid receptor PUMA-G. J. Biol. Chem 280:26649–52
    1. Thevenet J, De Marchi U, Domingo JS, Christinat N, Bultot L, et al. 2016. Medium-chain fatty acids inhibit mitochondrial metabolism in astrocytes promoting astrocyte-neuron lactate and ketone body shuttle systems. FASEB J. 30:1913–26
    1. Thomas LK, Ittmann M, Cooper C. 1982. The role of leucine in ketogenesis in starved rats. Biochem. J 204:399–403
    1. Thorrez L, Laudadio I, Van Deun K, Quintens R, Hendrickx N, et al. 2011. Tissue-specific disallowance of housekeeping genes: the other face of cell differentiation. Genome Res. 21:95–105
    1. Timper K, Del Rio-Martin A, Cremer AL, Bremser S, Alber J, et al. 2020. GLP-1 receptor signaling in astrocytes regulates fatty acid oxidation, mitochondrial integrity, and function. Cell Metab. 31:1189–205. e13
    1. Tomita I, Kume S, Sugahara S, Osawa N, Yamahara K, et al. 2020. SGLT2 inhibition mediates protection from diabetic kidney disease by promoting ketone body-induced mTORC1 inhibition. Cell Metab. 32:404–19. e6
    1. Tunaru S, Kero J, Schaub A, Wufka C, Blaukat A, et al. 2003. PUMA-G and HM74 are receptors for nicotinic acid and mediate its anti-lipolytic effect. Nat. Med 9:352–55
    1. Turko IV, Marcondes S, Murad F. 2001. Diabetes-associated nitration of tyrosine and inactivation of succinyl-CoA:3-oxoacid CoA-transferase. Am. J. Physiol. Heart Circ. Physiol 281:H2289–94
    1. van Hasselt PM, Ferdinandusse S, Monroe GR, Ruiter JP, Turkenburg M, et al. 2014. Monocarboxylate transporter 1 deficiency and ketone utilization. N. Engl. J. Med 371:1900–7
    1. Vanoverschelde JL, Wijns W, Kolanowski J, Bol A, Decoster PM, et al. 1993. Competition between palmitate and ketone bodies as fuels for the heart: study with positron emission tomography. Am. J. Physiol. Heart Circ. Physiol 264:H701–7
    1. Verma S, Rawat S, Ho KL, Wagg CS, Zhang L, et al. 2018. Empagliflozin increases cardiac energy production in diabetes: novel translational insights into the heart failure benefits of SGLT2 inhibitors. JACC Basic Transl. Sci 3:575–87
    1. Vila-Brau A, De Sousa-Coelho AL, Mayordomo C, Haro D, Marrero PF. 2011. Human HMGCS2 regulates mitochondrial fatty acid oxidation and FGF21 expression in HepG2 cell line. J. Biol. Chem 286:20423–30
    1. von Meyenn F, Porstmann T, Gasser E, Selevsek N, Schmidt A, et al. 2013. Glucagon-induced acetylation of Foxa2 regulates hepatic lipid metabolism. Cell Metab. 17:436–47
    1. Voros G, Ector J, Garweg C, Droogne W, Van Cleemput J, et al. 2018. Increased cardiac uptake of ketone bodies and free fatty acids in human heart failure and hypertrophic left ventricular remodeling. Circ. Heart Fail 11:e004953.
    1. Wagner GR, Bhatt DP, O’Connell TM, Thompson JW, Dubois LG, et al. 2017. A class of reactive acyl-CoA species reveals the non-enzymatic origins of protein acylation. Cell Metab. 25:823–37. e8
    1. Wang Q, Zhou Y, Rychahou P, Fan TWM, Lane AN, et al. 2017. Ketogenesis contributes to intestinal cell differentiation. Cell Death Differ. 24:458–68
    1. Wang W, Ishibashi J, Trefely S, Shao M, Cowan AJ, et al. 2019. A PRDM16-driven metabolic signal from adipocytes regulates precursor cell fate. Cell Metab. 30:174–89. e5
    1. Wang YH, Liu CL, Chiu WC, Twu YC, Liao YJ. 2019. HMGCS2 mediates ketone production and regulates the proliferation and metastasis of hepatocellular carcinoma. Cancers 11:1876
    1. Wang YH, Suk FM, Liao YJ. 2020. Loss of HMGCS2 enhances lipogenesis and attenuates the protective effect of the ketogenic diet in liver cancer. Cancers 12:1797
    1. Webber RJ, Edmond J. 1977. Utilization of L(+)-3-hydroxybutyrate, D(−)-3-hydroxybutyrate, acetoacetate, and glucose for respiration and lipid synthesis in the 18-day-old rat. J. Biol. Chem 252:5222–26
    1. Weidemann MJ, Krebs HA. 1969. The fuel of respiration of rat kidney cortex. Biochem. J 112:149–66
    1. Wentz AE, d’Avignon DA, Weber ML, Cotter DG, Doherty JM, et al. 2010. Adaptation of myocardial substrate metabolism to a ketogenic nutrient environment. J. Biol. Chem 285:24447–56
    1. Williamson DH, Bates MW, Page MA, Krebs HA. 1971. Activities of enzymes involved in acetoacetate utilization in adult mammalian tissues. Biochem. J 121:41–47
    1. Williamson DH, Lund P, Krebs HA. 1967. The redox state of free nicotinamide-adenine dinucleotide in the cytoplasm and mitochondria of rat liver. Biochem. J 103:514–27
    1. Williamson JR, Scholz R, Browning ET. 1969. Control mechanisms of gluconeogenesis and ketogenesis.II. Interactions between fatty acid oxidation and the citric acid cycle in perfused rat liver. J. Biol. Chem 244:4617–27
    1. Wolfrum C, Asilmaz E, Luca E, Friedman JM, Stoffel M. 2004. Foxa2 regulates lipid metabolism and ketogenesis in the liver during fasting and in diabetes. Nature 432:1027–32
    1. Xia S, Lin R, Jin L, Zhao L, Kang HB, et al. 2017. Prevention of dietary-fat-fueled ketogenesis attenuates BRAF V600E tumor growth. Cell Metab. 25:358–73
    1. Xie Z, Zhang D, Chung D, Tang Z, Huang H, et al. 2016. Metabolic regulation of gene expression by histone lysine β-hydroxybutyrylation. Mol. Cell 62:194–206
    1. Yang L, Achreja A, Yeung TL, Mangala LS, Jiang D, et al. 2016. Targeting stromal glutamine synthetase in tumors disrupts tumor microenvironment-regulated cancer cell growth. Cell Metab. 24:685–700
    1. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, et al. 2015. The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome–mediated inflammatory disease. Nat. Med 21:263–69
    1. Zelniker TA, Braunwald E. 2020. Mechanisms of cardiorenal effects of sodium-glucose cotransporter 2 inhibitors: JACC state-of-the-art review. J. Am. Coll. Cardiol 75:422–34
    1. Zhang D, Yang H, Kong X, Wang K, Mao X, et al. 2011. Proteomics analysis reveals diabetic kidney as a ketogenic organ in type 2 diabetes. Am. J. Physiol. Endocrinol. Metab 300:E287–95
    1. Zhang J, Jia PP, Liu QL, Cong MH, Gao Y, et al. 2018. Low ketolytic enzyme levels in tumors predict ketogenic diet responses in cancer cell lines in vitro and in vivo. J. Lipid Res 59:625–34
    1. Zhang S, Xie C. 2017. The role of OXCT1 in the pathogenesis of cancer as a rate-limiting enzyme of ketone body metabolism. Life Sci. 183:110–15
    1. Zhang WW, Churchill S, Lindahl R, Churchill P. 1989. Regulation of D-β-hydroxybutyrate dehydrogenase in rat hepatoma cell lines. Cancer Res. 49:2433–37
    1. Zou K, Hu Y, Li M, Wang H, Zhang Y, et al. 2019. Potential role of HMGCS2 in tumor angiogenesis in colorectal cancer and its potential use as a diagnostic marker. Can. J. Gastroenterol. Hepatol 2019:8348967.
    1. Zwiebel FM, Schwabe U, Olson MS, Scholz R. 1982. Role of pyruvate transporter in the regulation of the pyruvate dehydrogenase multienzyme complex in perfused rat liver. Biochemistry 21:346–53

Source: PubMed

3
Se inscrever