β-hydroxybutyrate as an Anti-Aging Metabolite
Abstract
:1. Introduction
2. Overview of β-HB Metabolism
2.1. Ketone Body Production and Utilization
2.1.1. Ketogenesis in Liver
2.1.2. Ketolysis in Extra-Hepatic Tissues
2.2. β-HB as an Energy Substrate
2.3. β-HB as a Signaling Mediator
2.3.1. Cell Surface Receptor
2.3.2. Anti-Oxidative Stress
2.3.3. Neuroprotection
2.4. β-HB as an Epigenetic Regulator
2.4.1. Histone Deacetylases Inhibition
2.4.2. Histone Lysine β-hydroxybutyrylation
3. Role of β-HB in Age-Related Diseases
3.1. Relationship between β-HB and Aging
3.2. Age-Related Diseases
3.2.1. Cancers
3.2.2. Neurological Disorders
3.2.3. Cardiovascular Diseases
3.2.4. Muscle Dysfunction
3.2.5. Inflammation
3.2.6. Metabolic Syndrome
4. Therapeutic Application of β-HB in Aging
4.1. Endogenous Ketosis
4.1.1. Intermittent Fasting and Caloric Restriction
4.1.2. Ketogenic Diets
4.2. Exogenous Ketosis and Supplementation of β-HB
4.2.1. Ketone Salts
4.2.2. Ketone Esters
4.2.3. Medium Chain Triglycerides
4.2.4. R, S-1,3-Butanediol
4.2.5. β-HB Enantiomers
4.3. Comparisons between Endogenous and Exogenous Ketosis Induced by NK
5. Future Perspectives
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Wakeman, A.J.; Dakin, H.D. On the decomposition of β-oxybutyric acid and aceto-acetic acid by enzymes of the liver. J. Biol. Chem. 1909, 6, 373–389. [Google Scholar] [CrossRef]
- Poff, A.M.; Koutnik, A.P.; Egan, B. Nutritional Ketosis with Ketogenic Diets or Exogenous Ketones: Features, Convergence, and Divergence. Curr. Sports Med. Rep. 2020, 19, 251–259. [Google Scholar] [CrossRef]
- Kovács, Z.; Brunner, B.; Ari, C. Beneficial Effects of Exogenous Ketogenic Supplements on Aging Processes and Age-Related Neurodegenerative. Dis. Nutr. 2021, 13, 2197. [Google Scholar]
- Bhattacharya, K.; Matar, W.; Tolun, A.A.; Devanapalli, B.; Thompson, S.; Dalkeith, T.; Lichkus, K.; Tchan, M. The use of sodium DL-3-Hydroxybutyrate in severe acute neuro-metabolic compromise in patients with inherited ketone body synthetic disorders. Orphanet J. Rare Dis. 2020, 15, 53. [Google Scholar] [CrossRef] [Green Version]
- Sharma, R.; Ramanathan, A. The Aging Metabolome-Biomarkers to Hub Metabolites. Proteomics 2020, 20, e1800407. [Google Scholar] [CrossRef] [Green Version]
- Edwards, C.; Copes, N.; Bradshaw, P.C. D-ß-hydroxybutyrate: An anti-aging ketone body. Oncotarget 2015, 6, 3477–3478. [Google Scholar] [CrossRef]
- Thomas, L.K.; Ittmann, M.; Cooper, C. The role of leucine in ketogenesis in starved rats. Biochem. J. 1982, 204, 399–403. [Google Scholar] [CrossRef] [Green Version]
- Soto-Mota, A.; Norwitz, N.G.; Clarke, K. Why a d-β-hydroxybutyrate monoester? Biochem. Soc. Trans. 2020, 48, 51–59. [Google Scholar] [CrossRef] [Green Version]
- Cahill, G.F., Jr.; Herrera, M.G.; Morgan, A.P.; Soeldner, J.S.; Steinke, J.; Levy, P.L.; Reichard, G.A., Jr.; Kipnis, D.M. Hormone-fuel interrelationships during fasting. J. Clin. Investig. 1966, 45, 1751–1769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Flatt, J.P. On the maximal possible rate of ketogenesis. Diabetes 1972, 21, 50–53. [Google Scholar] [CrossRef]
- Garber, A.J.; Menzel, P.H.; Boden, G.; Owen, O.E. Hepatic ketogenesis and gluconeogenesis in humans. J. Clin. Investig. 1974, 54, 981–989. [Google Scholar] [CrossRef] [Green Version]
- Reichard, G.A., Jr.; Owen, O.E.; Haff, A.C.; Paul, P.; Bortz, W.M. Ketone-body production and oxidation in fasting obese humans. J. Clin. Investig. 1974, 53, 508–515. [Google Scholar] [CrossRef]
- Newman, J.C.; Verdin, E. β-hydroxybutyrate: Much more than a metabolite. Diabetes Res. Clin. Pr. 2014, 106, 173–181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bock, H.; Fleischer, S. Preparation of a homogeneous soluble D-beta-hydroxybutyrate apodehydrogenase from mitochondria. J. Biol. Chem. 1975, 250, 5774–5781. [Google Scholar] [CrossRef]
- Lehninger, A.L.; Sudduth, H.C.; Wise, J.B. D-beta-Hydroxybutyric dehydrogenase of muitochondria. J. Biol. Chem. 1960, 235, 2450–2455. [Google Scholar] [CrossRef]
- Halestrap, A.P.; Wilson, M.C. The monocarboxylate transporter family--role and regulation. IUBMB Life 2012, 64, 109–119. [Google Scholar] [CrossRef]
- Puchalska, P.; Crawford, P.A. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 2017, 25, 262–284. [Google Scholar] [CrossRef] [Green Version]
- Halestrap, A.P. The SLC16 gene family—Structure, role and regulation in health and disease. Mol. Asp. Med. 2013, 34, 337–349. [Google Scholar] [CrossRef]
- Fukao, T.; Lopaschuk, G.D.; Mitchell, G.A. Pathways and control of ketone body metabolism: On the fringe of lipid biochemistry. Prostaglandins Leukot. Essent. Fat. Acids 2004, 70, 243–251. [Google Scholar] [CrossRef]
- Fukao, T.; Song, X.Q.; Mitchell, G.A.; Yamaguchi, S.; Sukegawa, K.; Orii, T.; Kondo, N. Enzymes of ketone body utilization in human tissues: Protein and messenger RNA levels of succinyl-coenzyme A (CoA):3-ketoacid CoA transferase and mitochondrial and cytosolic acetoacetyl-CoA thiolases. Pediatr. Res. 1997, 42, 498–502. [Google Scholar] [CrossRef] [Green Version]
- Orii, K.E.; Fukao, T.; Song, X.Q.; Mitchell, G.A.; Kondo, N. Liver-specific silencing of the human gene encoding succinyl-CoA: 3-ketoacid CoA transferase. Tohoku J. Exp. Med. 2008, 215, 227–236. [Google Scholar] [CrossRef] [Green Version]
- Owen, O.E.; Morgan, A.P.; Kemp, H.G.; Sullivan, J.M.; Herrera, M.G.; Cahill, G.F., Jr. Brain metabolism during fasting. J. Clin. Investig. 1967, 46, 1589–1595. [Google Scholar] [CrossRef]
- Sultan, A.M. D-3-hydroxybutyrate metabolism in the perfused rat heart. Mol. Cell Biochem. 1988, 79, 113–118. [Google Scholar] [CrossRef]
- Abbasi, J. Ketone Body Supplementation-A Potential New Approach for Heart Disease. JAMA 2021, 326, 17–18. [Google Scholar] [CrossRef]
- Paoli, A.; Bosco, G.; Camporesi, E.M.; Mangar, D. Ketosis, ketogenic diet and food intake control: A complex relationship. Front. Psychol. 2015, 6, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Krebs, H. Biochemical aspects of ketosis. Proc. R. Soc. Med. 1960, 53, 71–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robinson, A.M.; Williamson, D.H. Physiological roles of ketone bodies as substrates and signals in mammalian tissues. Physiol. Rev. 1980, 60, 143–187. [Google Scholar] [CrossRef] [PubMed]
- Laffel, L. Ketone bodies: A review of physiology, pathophysiology and application of monitoring to diabetes. Diabetes Metab. Res. Rev. 1999, 15, 412–426. [Google Scholar] [CrossRef]
- Johnson, R.H.; Walton, J.L.; Krebs, H.A.; Williamson, D.H. Metabolic fuels during and after severe exercise in athletes and non-athletes. Lancet 1969, 2, 452–455. [Google Scholar] [CrossRef]
- Gershuni, V.M.; Yan, S.L.; Medici, V. Nutritional ketosis for weight management and reversal of metabolic syndrome. Curr. Nutr. Rep. 2018, 7, 97–106. [Google Scholar] [CrossRef]
- Cahill, G.F., Jr. Fuel metabolism in starvation. Annu. Rev. Nutr. 2006, 26, 1–22. [Google Scholar] [CrossRef] [Green Version]
- Balasse, E.O.; Féry, F. Ketone body production and disposal: Effects of fasting, diabetes, and exercise. Diabetes Metab. Rev. 1989, 5, 247–270. [Google Scholar] [CrossRef]
- Balasse, E.O.; Fery, F.; Neef, M.A. Changes induced by exercise in rates of turnover and oxidation of ketone bodies in fasting man. J. Appl. Physiol. Respir. Environ. Exerc. Physiol. 1978, 44, 5–11. [Google Scholar] [CrossRef]
- Rojas-Morales, P.; Tapia, E.; Pedraza-Chaverri, J. β-Hydroxybutyrate: A signaling metabolite in starvation response? Cell. Signal. 2016, 28, 917–923. [Google Scholar] [CrossRef] [PubMed]
- Cotter, D.G.; Schugar, R.C.; Crawford, P.A. Ketone body metabolism and cardiovascular disease. Am. J. Physiol. Heart Circ. Physiol. 2013, 304, H1060–H1076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taggart, A.K.; Kero, J.; Gan, X.; Cai, T.Q.; Cheng, K.; Ippolito, M.; Ren, N.; Kaplan, R.; Wu, K.; Wu, T.J.; et al. (D)-beta-Hydroxybutyrate inhibits adipocyte lipolysis via the nicotinic acid receptor PUMA-G. J. Biol. Chem. 2005, 280, 26649–26652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Offermanns, S. The nicotinic acid receptor GPR109A (HM74A or PUMA-G) as a new therapeutic target. Trends Pharm. Sci. 2006, 27, 384–390. [Google Scholar] [CrossRef] [PubMed]
- Stilling, R.M.; van de Wouw, M.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochem. Int. 2016, 99, 110–132. [Google Scholar] [CrossRef] [PubMed]
- Won, Y.J.; Lu, V.B.; Puhl, H.L., 3rd; Ikeda, S.R. β-Hydroxybutyrate modulates N-type calcium channels in rat sympathetic neurons by acting as an agonist for the G-protein-coupled receptor FFA3. J. Neurosci. 2013, 33, 19314–19325. [Google Scholar] [CrossRef]
- Tunaru, S.; Kero, J.; Schaub, A.; Wufka, C.; Blaukat, A.; Pfeffer, K.; Offermanns, S. PUMA-G and HM74 are receptors for nicotinic acid and mediate its anti-lipolytic effect. Nat. Med. 2003, 9, 352–355. [Google Scholar] [CrossRef] [PubMed]
- Lukasova, M.; Malaval, C.; Gille, A.; Kero, J.; Offermanns, S. Nicotinic acid inhibits progression of atherosclerosis in mice through its receptor GPR109A expressed by immune cells. J. Clin. Investig. 2011, 121, 1163–1173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kovács, Z.; D’Agostino, D.P.; Diamond, D.; Kindy, M.S.; Rogers, C.; Ari, C. Therapeutic Potential of Exogenous Ketone Supplement Induced Ketosis in the Treatment of Psychiatric Disorders: Review of Current Literature. Front. Psychiatry 2019, 10, 363. [Google Scholar] [CrossRef] [PubMed]
- Islam, M.T. Oxidative stress and mitochondrial dysfunction-linked neurodegenerative disorders. Neurol. Res. 2017, 39, 73–82. [Google Scholar] [CrossRef]
- Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef] [Green Version]
- Yang, H.; Shan, W.; Zhu, F.; Wu, J.; Wang, Q. Ketone bodies in neurological diseases: Focus on neuroprotection and underlying mechanisms. Front. Neurol. 2019, 10, 585. [Google Scholar] [CrossRef] [Green Version]
- Kashiwaya, Y.; Takeshima, T.; Mori, N.; Nakashima, K.; Clarke, K.; Veech, R.L. D-beta-hydroxybutyrate protects neurons in models of Alzheimer’s and Parkinson’s disease. Proc. Natl. Acad. Sci. USA 2000, 97, 5440–5444. [Google Scholar] [CrossRef] [Green Version]
- Fu, S.P.; Li, S.N.; Wang, J.F.; Li, Y.; Xie, S.S.; Xue, W.J.; Liu, H.M.; Huang, B.X.; Lv, Q.K.; Lei, L.C.; et al. BHBA suppresses LPS-induced inflammation in BV-2 cells by inhibiting NF-κB activation. Mediat. Inflamm. 2014, 2014, 983401. [Google Scholar] [CrossRef] [Green Version]
- Norwitz, N.G.; Hu, M.T.; Clarke, K. The mechanisms by which the ketone body D-β-Hydroxybutyrate may improve the multiple cellular pathologies of Parkinson’s disease. Front. Nutr. 2019, 6, 63. [Google Scholar] [CrossRef]
- Włodarek, D. Role of ketogenic diets in neurodegenerative diseases (Alzheimer’s disease and Parkinson’s disease). Nutrients 2019, 11, 169. [Google Scholar] [CrossRef] [Green Version]
- Shaafi, S.; Najmi, S.; Aliasgharpour, H.; Mahmoudi, J.; Sadigh-Etemad, S.; Farhoudi, M.; Baniasadi, N. The efficacy of the ketogenic diet on motor functions in Parkinson’s disease: A rat model. Iran. J. Neurol. 2016, 15, 63–69. [Google Scholar] [PubMed]
- Tieu, K.; Perier, C.; Caspersen, C.; Teismann, P.; Wu, D.C.; Yan, S.D.; Naini, A.; Vila, M.; Jackson-Lewis, V.; Ramasamy, R.; et al. D-beta-hydroxybutyrate rescues mitochondrial respiration and mitigates features of Parkinson disease. J. Clin. Investig. 2003, 112, 892–901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bird, A. Perceptions of epigenetics. Nature 2007, 447, 396–398. [Google Scholar] [CrossRef] [PubMed]
- Newman, J.C.; Verdin, E. Ketone bodies as signaling metabolites. Trends Endocrinol. Metab. 2014, 25, 42–52. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Kontopoulos, E.; Parvin, J.D.; Feany, M.B. Alpha-synuclein acts in the nucleus to inhibit histone acetylation and promote neurotoxicity. Hum. Mol. Genet. 2006, 15, 3012–3023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lang, C.; Campbell, K.R.; Ryan, B.J.; Carling, P.; Attar, M.; Vowles, J.; Perestenko, O.V.; Bowden, R.; Baig, F.; Kasten, M.; et al. Single-cell sequencing of iPSC-dopamine neurons reconstructs disease progression and identifies HDAC4 as a regulator of Parkinson cell phenotypes. Cell Stem Cell 2019, 24, 93–106. [Google Scholar] [CrossRef] [Green Version]
- Sleiman, S.F.; Henry, J.; Al-Haddad, R.; El Hayek, L.; Abou Haidar, E.; Stringer, T.; Ulja, D.; Karuppagounder, S.S.; Holson, E.B.; Ratan, R.R.; et al. Exercise promotes the expression of brain derived neurotrophic factor (BDNF) through the action of the ketone body β-hydroxybutyrate. eLife 2016, 5, e15092. [Google Scholar] [CrossRef]
- Marosi, K.; Kim, S.W.; Moehl, K.; Scheibye-Knudsen, M.; Cheng, A.; Cutler, R.; Camandola, S.; Mattson, M.P. 3-Hydroxybutyrate regulates energy metabolism and induces BDNF expression in cerebral cortical neurons. J. Neurochem. 2016, 139, 769–781. [Google Scholar] [CrossRef]
- Xu, D.; Lian, D.; Wu, J.; Liu, Y.; Zhu, M.; Sun, J.; He, D.; Li, L. Brain-derived neurotrophic factor reduces inflammation and hippocampal apoptosis in experimental Streptococcus pneumoniae meningitis. J. Neuroinflamm. 2017, 14, 156. [Google Scholar] [CrossRef] [Green Version]
- Makar, T.K.; Trisler, D.; Sura, K.T.; Sultana, S.; Patel, N.; Bever, C.T. Brain derived neurotrophic factor treatment reduces inflammation and apoptosis in experimental allergic encephalomyelitis. J. Neurol. Sci. 2008, 270, 70–76. [Google Scholar] [CrossRef]
- Mattson, M.P.; Lovell, M.A.; Furukawa, K.; Markesbery, W.R. Neurotrophic factors attenuate glutamate-induced accumulation of peroxides, elevation of intracellular Ca2+ concentration, and neurotoxicity and increase antioxidant enzyme activities in hippocampal neurons. J. Neurochem. 1995, 65, 1740–1751. [Google Scholar] [CrossRef]
- Kirschner, P.B.; Jenkins, B.G.; Schulz, J.B.; Finkelstein, S.P.; Matthews, R.T.; Rosen, B.R.; Beal, M.F. NGF, BDNF and NT-5, but not NT-3 protect against MPP+ toxicity and oxidative stress in neonatal animals. Brain Res. 1996, 713, 178–185. [Google Scholar] [CrossRef]
- Zhang, X.; Cao, R.; Niu, J.; Yang, S.; Ma, H.; Zhao, S.; Li, H. Molecular basis for hierarchical histone de-β-hydroxybutyrylation by SIRT3. Cell Discov. 2019, 5, 35. [Google Scholar] [CrossRef]
- Dąbek, A.; Wojtala, M.; Pirola, L.; Balcerczyk, A. Modulation of cellular biochemistry, epigenetics and metabolomics by ketone bodies. Implications of the ketogenic diet in the physiology of the organism and pathological states. Nutrients 2020, 12, 788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boison, D. New insights into the mechanisms of the ketogenic diet. Curr. Opin. Neurol. 2017, 30, 187–192. [Google Scholar] [CrossRef] [Green Version]
- Ruan, H.B.; Crawford, P.A. Ketone bodies as epigenetic modifiers. Curr. Opin. Clin. Nutr. Metab. Care 2018, 21, 260–266. [Google Scholar] [CrossRef] [PubMed]
- Liu, K.; Li, F.; Sun, Q.; Lin, N.; Han, H.; You, K.; Tian, F.; Mao, Z.; Li, T.; Tong, T.; et al. p53 β-hydroxybutyrylation attenuates p53 activity. Cell Death Dis. 2019, 10, 243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, H.; Zhang, D.; Weng, Y.; Delaney, K.; Tang, Z.; Yan, C.; Qi, S.; Peng, C.; Cole, P.A.; Roeder, R.G.; et al. The regulatory enzymes and protein substrates for the lysine β-hydroxybutyrylation pathway. Sci. Adv. 2021, 7, eabe2771. [Google Scholar] [CrossRef] [PubMed]
- Witte, A.V.; Fobker, M.; Gellner, R.; Knecht, S.; Flöel, A. Caloric restriction improves memory in elderly humans. Proc. Natl. Acad. Sci. USA 2009, 106, 1255–1260. [Google Scholar] [CrossRef] [Green Version]
- Roberts, M.N.; Wallace, M.A.; Tomilov, A.A.; Zhou, Z.; Marcotte, G.R.; Tran, D.; Perez, G.; Gutierrez-Casado, E.; Koike, S.; Knotts, T.A.; et al. A ketogenic diet extends longevity and healthspan in adult mice. Cell Metab. 2017, 26, 539–546. [Google Scholar] [CrossRef] [Green Version]
- Veech, R.L.; Bradshaw, P.C.; Clarke, K.; Curtis, W.; Pawlosky, R.; King, M.T. Ketone bodies mimic the life span extending properties of caloric restriction. IUBMB Life 2017, 69, 305–314. [Google Scholar] [CrossRef] [PubMed]
- Edwards, C.; Canfield, J.; Copes, N.; Rehan, M.; Lipps, D.; Bradshaw, P.C. D-beta-hydroxybutyrate extends lifespan in C. elegans. Aging 2014, 6, 621–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, Y.M.; Bedarida, T.; Ding, Y.; Somba, B.K.; Lu, Q.; Wang, Q.; Song, P.; Zou, M.H. β-Hydroxybutyrate prevents vascular senescence through hnRNP A1-mediated upregulation of Oct4. Mol. Cell 2018, 71, 1064–1078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, C.W.; Biton, M.; Haber, A.L.; Gunduz, N.; Eng, G.; Gaynor, L.T.; Tripathi, S.; Calibasi-Kocal, G.; Rickelt, S.; Butty, V.L.; et al. Ketone body signaling mediates intestinal stem cell homeostasis and adaptation to diet. Cell 2019, 178, 1115–1131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aunan, J.R.; Cho, W.C.; Søreide, K. The biology of aging and cancer: A brief overview of shared and divergent molecular hallmarks. Aging Dis. 2017, 8, 628–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumari, S.; Badana, A.K.; Malla, R. Reactive oxygen species: A key constituent in cancer survival. Biomark Insights 2018, 13, 1177271918755391. [Google Scholar] [CrossRef] [Green Version]
- Klement, R.J.; Champ, C.E.; Otto, C.; Kämmerer, U. Anti-tumor effects of ketogenic diets in mice: A meta-analysis. PLoS ONE 2016, 11, e0155050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Poff, A.M.; Ari, C.; Seyfried, T.N.; D’Agostino, D.P. The ketogenic diet and hyperbaric oxygen therapy prolong survival in mice with systemic metastatic cancer. PLoS ONE 2013, 8, e65522. [Google Scholar] [CrossRef] [Green Version]
- Otto, C.; Kaemmerer, U.; Illert, B.; Muehling, B.; Pfetzer, N.; Wittig, R.; Voelker, H.U.; Thiede, A.; Coy, J.F. Growth of human gastric cancer cells in nude mice is delayed by a ketogenic diet supplemented with omega-3 fatty acids and medium-chain triglycerides. BMC Cancer 2008, 8, 122. [Google Scholar] [CrossRef] [Green Version]
- Allen, B.G.; Bhatia, S.K.; Anderson, C.M.; Eichenberger-Gilmore, J.M.; Sibenaller, Z.A.; Mapuskar, K.A.; Schoenfeld, J.D.; Buatti, J.M.; Spitz, D.R.; Fath, M.A. Ketogenic diets as an adjuvant cancer therapy: History and potential mechanism. Redox Biol. 2014, 2, 963–970. [Google Scholar] [CrossRef] [Green Version]
- Klement, R.J.; Sweeney, R.A. Impact of a ketogenic diet intervention during radiotherapy on body composition: I. Initial clinical experience with six prospectively studied patients. BMC Res. Notes 2016, 9, 143. [Google Scholar] [CrossRef] [Green Version]
- Schmidt, M.; Pfetzer, N.; Schwab, M.; Strauss, I.; Kämmerer, U. Effects of a ketogenic diet on the quality of life in 16 patients with advanced cancer: A pilot trial. Nutr. Metab. 2011, 8, 54. [Google Scholar] [CrossRef] [Green Version]
- Zhang, C.; Rissman, R.A.; Feng, J. Characterization of ATP alternations in an Alzheimer’s disease transgenic mouse model. J. Alzheimers Dis. 2015, 44, 375–378. [Google Scholar] [CrossRef] [Green Version]
- Greenamyre, J.T.; Sherer, T.B.; Betarbet, R.; Panov, A.V. Complex I and Parkinson’s disease. IUBMB Life 2001, 52, 135–141. [Google Scholar] [CrossRef]
- Mosconi, L.; de Leon, M.; Murray, J.; E., L.; Lu, J.; Javier, E.; McHugh, P.; Swerdlow, R.H. Reduced mitochondria cytochrome oxidase activity in adult children of mothers with Alzheimer’s disease. J. Alzheimers Dis. 2011, 27, 483–490. [Google Scholar] [CrossRef]
- Fortier, M.; Castellano, C.A.; Croteau, E.; Langlois, F.; Bocti, C.; St-Pierre, V.; Vandenberghe, C.; Bernier, M.; Roy, M.; Descoteaux, M.; et al. A ketogenic drink improves brain energy and some measures of cognition in mild cognitive impairment. Alzheimers Dement. 2019, 15, 625–634. [Google Scholar] [CrossRef]
- Srivastava, S.; Baxa, U.; Niu, G.; Chen, X.; Veech, R.L. A ketogenic diet increases brown adipose tissue mitochondrial proteins and UCP1 levels in mice. IUBMB Life 2013, 65, 58–66. [Google Scholar] [CrossRef] [PubMed]
- Xu, X.; Zhang, Q.; Tu, J.; Ren, Z. D-β-hydroxybutyrate inhibits microglial activation in a cell activation model in vitro. J. Med. Coll. PLA 2011, 26, 117–127. [Google Scholar] [CrossRef]
- Youm, Y.H.; Nguyen, K.Y.; Grant, R.W.; Goldberg, E.L.; Bodogai, M.; Kim, D.; D’Agostino, D.; Planavsky, N.; Lupfer, C.; Kanneganti, T.D.; et al. The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat. Med. 2015, 21, 263–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, Y.M.; Ramprasath, T.; Zou, M.H. β-hydroxybutyrate and its metabolic effects on age-associated pathology. Exp. Mol. Med. 2020, 52, 548–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campisi, J.; Kapahi, P.; Lithgow, G.J.; Melov, S.; Newman, J.C.; Verdin, E. From discoveries in ageing research to therapeutics for healthy ageing. Nature 2019, 571, 183–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abiri, B.; Vafa, M. Dietary Restriction, Cardiovascular Aging and Age-Related Cardiovascular Diseases: A Review of the Evidence. Adv. Exp. Med. Biol. 2019, 1178, 113–127. [Google Scholar] [PubMed]
- Newman, J.C.; Covarrubias, A.J.; Zhao, M.; Yu, X.; Gut, P.; Ng, C.P.; Huang, Y.; Haldar, S.; Verdin, E. Ketogenic Diet Reduces Midlife Mortality and Improves Memory in Aging Mice. Cell Metab. 2017, 26, 547–557.e548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shou, J.; Chen, P.J.; Xiao, W.H. Mechanism of increased risk of insulin resistance in aging skeletal muscle. Diabetol. Metab. Syndr. 2020, 12, 14. [Google Scholar] [CrossRef] [Green Version]
- Bonaldo, P.; Sandri, M. Cellular and molecular mechanisms of muscle atrophy. Dis. Model Mech. 2013, 6, 25–39. [Google Scholar] [CrossRef] [Green Version]
- Stubbs, B.J.; Koutnik, A.P.; Volek, J.S.; Newman, J.C. From bedside to battlefield: Intersection of ketone body mechanisms in geroscience with military resilience. GeroScience 2021, 43, 1071–1081. [Google Scholar] [CrossRef] [PubMed]
- Koutnik, A.P.; D’Agostino, D.P.; Egan, B. Anticatabolic Effects of Ketone Bodies in Skeletal Muscle. Trends Endocrinol. Metab. 2019, 30, 227–229. [Google Scholar] [CrossRef] [PubMed]
- Ahola-Erkkilä, S.; Carroll, C.J.; Peltola-Mjösund, K.; Tulkki, V.; Mattila, I.; Seppänen-Laakso, T.; Oresic, M.; Tyynismaa, H.; Suomalainen, A. Ketogenic diet slows down mitochondrial myopathy progression in mice. Hum. Mol. Genet. 2010, 19, 1974–1984. [Google Scholar] [CrossRef] [Green Version]
- Kwak, S.E.; Bae, J.H.; Lee, J.H.; Shin, H.E.; Zhang, D.; Cho, S.C.; Song, W. Effects of exercise-induced beta-hydroxybutyrate on muscle function and cognitive function. Physiol. Rep. 2021, 9, e14497. [Google Scholar] [CrossRef]
- Munroe, M.; Pincu, Y.; Merritt, J.; Cobert, A.; Brander, R.; Jensen, T.; Rhodes, J.; Boppart, M.D. Impact of β-hydroxy β-methylbutyrate (HMB) on age-related functional deficits in mice. Exp. Gerontol. 2017, 87, 57–66. [Google Scholar] [CrossRef]
- Walsh, M.E.; Bhattacharya, A.; Sataranatarajan, K.; Qaisar, R.; Sloane, L.; Rahman, M.M.; Kinter, M.; Van Remmen, H. The histone deacetylase inhibitor butyrate improves metabolism and reduces muscle atrophy during aging. Aging Cell 2015, 14, 957–970. [Google Scholar] [CrossRef]
- Angiolilli, C.; Baeten, D.L.; Radstake, T.R.; Reedquist, K.A. The acetyl code in rheumatoid arthritis and other rheumatic diseases. Epigenomics 2017, 9, 447–461. [Google Scholar] [CrossRef]
- Pawelec, G.; Goldeck, D.; Derhovanessian, E. Inflammation, ageing and chronic disease. Curr. Opin. Immunol. 2014, 29, 23–28. [Google Scholar] [CrossRef]
- Chamorro, A.; Hallenbeck, J. The harms and benefits of inflammatory and immune responses in vascular disease. Stroke 2006, 37, 291–293. [Google Scholar] [CrossRef] [PubMed]
- Praticò, D.; Trojanowski, J.Q. Inflammatory hypotheses: Novel mechanisms of Alzheimer’s neurodegeneration and new therapeutic targets? Neurobiol. Aging 2000, 21, 441–453. [Google Scholar] [CrossRef]
- Pasyukova, E.G.; Vaiserman, A.M. HDAC inhibitors: A new promising drug class in anti-aging research. Mech. Ageing Dev. 2017, 166, 6–15. [Google Scholar] [CrossRef] [PubMed]
- Camberos-Luna, L.; Massieu, L. Therapeutic strategies for ketosis induction and their potential efficacy for the treatment of acute brain injury and neurodegenerative diseases. Neurochem. Int. 2020, 133, 104614. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.Y.; Hao, J.; Liu, R.; Turner, G.; Shi, F.D.; Rho, J.M. Inflammation-mediated memory dysfunction and effects of a ketogenic diet in a murine model of multiple sclerosis. PLoS ONE 2012, 7, e35476. [Google Scholar] [CrossRef] [Green Version]
- Alberti, K.G.; Zimmet, P.; Shaw, J. The metabolic syndrome--a new worldwide definition. Lancet 2005, 366, 1059–1062. [Google Scholar] [CrossRef]
- Ron, D.; Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat. Rev. Mol. Cell Biol. 2007, 8, 519–529. [Google Scholar] [CrossRef]
- Nunes-Souza, V.; César-Gomes, C.J.; Da Fonseca, L.J.; Guedes Gda, S.; Smaniotto, S.; Rabelo, L.A. Aging Increases Susceptibility to High Fat Diet-Induced Metabolic Syndrome in C57BL/6 Mice: Improvement in Glycemic and Lipid Profile after Antioxidant Therapy. Oxid. Med. Cell Longev. 2016, 2016, 1987960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mey, J.T.; Erickson, M.L.; Axelrod, C.L.; King, W.T.; Flask, C.A.; McCullough, A.J.; Kirwan, J.P. β-Hydroxybutyrate is reduced in humans with obesity-related NAFLD and displays a dose-dependent effect on skeletal muscle mitochondrial respiration in vitro. Am. J. Physiol. Endocrinol. Metab. 2020, 319, e187–e195. [Google Scholar] [CrossRef] [PubMed]
- Cavaleri, F.; Bashar, E. Potential Synergies of β-Hydroxybutyrate and Butyrate on the Modulation of Metabolism, Inflammation, Cognition, and General Health. J. Nutr. Metab. 2018, 2018, 7195760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, A.K.; Kim, D.H.; Bang, E.; Choi, Y.J.; Chung, H.Y. β-Hydroxybutyrate Suppresses Lipid Accumulation in Aged Liver through GPR109A-mediated Signaling. Aging Dis. 2020, 11, 777–790. [Google Scholar] [CrossRef]
- Bae, H.R.; Kim, D.H.; Park, M.H.; Lee, B.; Kim, M.J.; Lee, E.K.; Chung, K.W.; Kim, S.M.; Im, D.S.; Chung, H.Y. β-Hydroxybutyrate suppresses inflammasome formation by ameliorating endoplasmic reticulum stress via AMPK activation. Oncotarget 2016, 7, 66444–66454. [Google Scholar] [CrossRef] [Green Version]
- Møller, N. Ketone Body, 3-Hydroxybutyrate: Minor Metabolite—Major Medical Manifestations. J. Clin. Endocrinol. Metab. 2020, 105, dgaa370. [Google Scholar] [CrossRef]
- Paoli, A.; Rubini, A.; Volek, J.S.; Grimaldi, K.A. Beyond weight loss: A review of the therapeutic uses of very-low-carbohydrate (ketogenic) diets. Eur. J. Clin. Nutr. 2013, 67, 789–796. [Google Scholar] [CrossRef] [Green Version]
- Fontana, L. Excessive adiposity, calorie restriction, and aging. JAMA 2006, 295, 1577–1578. [Google Scholar] [CrossRef]
- Kim, B.H.; Joo, Y.; Kim, M.-S.; Choe, H.K.; Tong, Q.; Kwon, O. Effects of Intermittent Fasting on the Circulating Levels and Circadian Rhythms of Hormones. Endocrinol. Metab. 2021, 36, 745–756. [Google Scholar] [CrossRef]
- Hutchison, A.T.; Liu, B.; Wood, R.E.; Vincent, A.D.; Thompson, C.H.; O’Callaghan, N.J.; Wittert, G.A.; Heilbronn, L.K. Effects of Intermittent Versus Continuous Energy Intakes on Insulin Sensitivity and Metabolic Risk in Women with Overweight. Obesity 2019, 27, 50–58. [Google Scholar] [CrossRef] [Green Version]
- Gomez-Arbelaez, D.; Bellido, D.; Castro, A.I.; Ordoñez-Mayan, L.; Carreira, J.; Galban, C.; Martinez-Olmos, M.A.; Crujeiras, A.B.; Sajoux, I.; Casanueva, F.F. Body composition changes after very-low-calorie ketogenic diet in obesity evaluated by 3 standardized methods. J. Clin. Endocrinol. Metab. 2017, 102, 488–498. [Google Scholar] [CrossRef]
- Stewart, W.K.; Fleming, L.W. Features of a successful therapeutic fast of 382 days’ duration. Postgrad. Med. J. 1973, 49, 203–209. [Google Scholar] [CrossRef] [Green Version]
- Brandhorst, S.; Choi, I.Y.; Wei, M.; Cheng, C.W.; Sedrakyan, S.; Navarrete, G.; Dubeau, L.; Yap, L.P.; Park, R.; Vinciguerra, M.; et al. A Periodic Diet that Mimics Fasting Promotes Multi-System Regeneration, Enhanced Cognitive Performance, and Healthspan. Cell Metab. 2015, 22, 86–99. [Google Scholar] [CrossRef] [Green Version]
- Bordone, L.; Guarente, L. Calorie restriction, SIRT1 and metabolism: Understanding longevity. Nat. Rev. Mol. Cell Biol. 2005, 6, 298–305. [Google Scholar] [CrossRef]
- Pignatti, C.; D’Adamo, S.; Stefanelli, C.; Flamigni, F.; Cetrullo, S. Nutrients and Pathways that Regulate Health Span and Life Span. Geriatrics 2020, 5, 95. [Google Scholar] [CrossRef]
- Fontana, L.; Partridge, L.; Longo, V.D. Extending healthy life span--from yeast to humans. Science 2010, 328, 321–326. [Google Scholar] [CrossRef] [Green Version]
- Abdul Kadir, A.; Clarke, K.; Evans, R.D. Cardiac ketone body metabolism. Biochim. Biophys. Acta. Mol. Basis Dis. 2020, 1866, 165739. [Google Scholar] [CrossRef]
- Wallace, M.A.; Aguirre, N.W.; Marcotte, G.R.; Marshall, A.G.; Baehr, L.M.; Hughes, D.C.; Hamilton, K.L.; Roberts, M.N.; Lopez-Dominguez, J.A.; Miller, B.F.; et al. The ketogenic diet preserves skeletal muscle with aging in mice. Aging Cell 2021, 20, e13322. [Google Scholar] [CrossRef]
- O’Neill, B.; Raggi, P. The ketogenic diet: Pros and cons. Atherosclerosis 2020, 292, 119–126. [Google Scholar] [CrossRef] [Green Version]
- Ari, C.; Kovács, Z.; Juhasz, G.; Murdun, C.; Goldhagen, C.R.; Koutnik, A.P.; Poff, A.M.; Kesl, S.L.; D’Agostino, D.P. Exogenous Ketone Supplements Reduce Anxiety-Related Behavior in Sprague-Dawley and Wistar Albino Glaxo/Rijswijk Rats. Front. Mol. Neurosci. 2016, 9, 137. [Google Scholar] [CrossRef] [Green Version]
- CJ, D.C.H.; Schofield, G.M.; Williden, M.; McQuillan, J.A. The effect of medium chain triglycerides on time to nutritional ketosis and symptoms of keto-induction in healthy adults: A randomised controlled clinical trial. J. Nutr. Metab. 2018, 2018, 2630565. [Google Scholar]
- Kesl, S.L.; Poff, A.M.; Ward, N.P.; Fiorelli, T.N.; Ari, C.; Van Putten, A.J.; Sherwood, J.W.; Arnold, P.; D’Agostino, D.P. Effects of exogenous ketone supplementation on blood ketone, glucose, triglyceride, and lipoprotein levels in Sprague-Dawley rats. Nutr. Metab. 2016, 13, 9. [Google Scholar] [CrossRef] [Green Version]
- Brunengraber, H. Potential of ketone body esters for parenteral and oral nutrition. Nutrition 1997, 13, 233–235. [Google Scholar] [CrossRef]
- Ari, C.; Murdun, C.; Koutnik, A.P.; Goldhagen, C.R.; Rogers, C.; Park, C.; Bharwani, S.; Diamond, D.M.; Kindy, M.S.; D’Agostino, D.P.; et al. Exogenous Ketones Lower Blood Glucose Level in Rested and Exercised Rodent Models. Nutrients 2019, 11, 2330. [Google Scholar] [CrossRef] [Green Version]
- Veech, R.L. 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. Fat. Acids 2004, 70, 309–319. [Google Scholar] [CrossRef]
- Stubbs, B.J.; Blade, T.; Mills, S.; Thomas, J.; Yufei, X.; Nelson, F.R.; Higley, N.; Nikiforov, A.I.; Rhiner, M.O.; Verdin, E.; et al. In vitro stability and in vivo pharmacokinetics of the novel ketogenic ester, bis hexanoyl (R)-1,3-butanediol. Food Chem. Toxicol. 2021, 147, 111859. [Google Scholar] [CrossRef]
- Clarke, K.; Tchabanenko, K.; Pawlosky, R.; Carter, E.; Todd King, M.; Musa-Veloso, K.; Ho, M.; Roberts, A.; Robertson, J.; Vanitallie, T.B.; et al. Kinetics, safety and tolerability of (R)-3-hydroxybutyl (R)-3-hydroxybutyrate in healthy adult subjects. Regul. Toxicol. Pharm. 2012, 63, 401–408. [Google Scholar] [CrossRef] [Green Version]
- Stubbs, B.J.; Cox, P.J.; Evans, R.D.; Santer, P.; Miller, J.J.; Faull, O.K.; Magor-Elliott, S.; Hiyama, S.; Stirling, M.; Clarke, K. On the metabolism of exogenous ketones in humans. Front. Physiol. 2017, 8, 848. [Google Scholar] [CrossRef]
- Stubbs, B.J.; Cox, P.J.; Evans, R.D.; Cyranka, M.; Clarke, K.; de Wet, H. A ketone ester drink lowers human ghrelin and appetite. Obesity 2018, 26, 269–273. [Google Scholar] [CrossRef]
- Myette-Côté, É.; Neudorf, H.; Rafiei, H.; Clarke, K.; Little, J.P. Prior ingestion of exogenous ketone monoester attenuates the glycaemic response to an oral glucose tolerance test in healthy young individuals. J. Physiol. 2018, 596, 1385–1395. [Google Scholar] [CrossRef]
- Soto-Mota, A.; Vansant, H.; Evans, R.D.; Clarke, K. Safety and tolerability of sustained exogenous ketosis using ketone monoester drinks for 28 days in healthy adults. Regul. Toxicol. Pharm. 2019, 109, 104506. [Google Scholar] [CrossRef]
- Schönfeld, P.; Wojtczak, L. Short- and medium-chain fatty acids in energy metabolism: The cellular perspective. J. Lipid Res. 2016, 57, 943–954. [Google Scholar] [CrossRef] [Green Version]
- Huttenlocher, P.R.; Wilbourn, A.J.; Signore, J.M. Medium-chain triglycerides as a therapy for intractable childhood epilepsy. Neurology 1971, 21, 1097–1103. [Google Scholar] [CrossRef]
- Thorburn, M.S.; Vistisen, B.; Thorp, R.M.; Rockell, M.J.; Jeukendrup, A.E.; Xu, X.; Rowlands, D.S. Attenuated gastric distress but no benefit to performance with adaptation to octanoate-rich esterified oils in well-trained male cyclists. J. Appl. Physiol. (1985) 2006, 101, 1733–1743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Henderson, S.T. Ketone bodies as a therapeutic for Alzheimer’s disease. Neurotherapeutics 2008, 5, 470–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dymsza, H.A. Nutritional application and implication of 1,3-butanediol. Fed. Proc. 1975, 34, 2167–2170. [Google Scholar] [PubMed]
- Puchowicz, M.A.; Smith, C.L.; Bomont, C.; Koshy, J.; David, F.; Brunengraber, H. Dog model of therapeutic ketosis Induced by oral administration of R,S-1,3-butanediol diacetoacetate. J. Nutr. Biochem. 2000, 11, 281–287. [Google Scholar] [CrossRef]
- D’Agostino, D.P.; Pilla, R.; Held, H.E.; Landon, C.S.; Puchowicz, M.; Brunengraber, H.; Ari, C.; Arnold, P.; Dean, J.B. Therapeutic ketosis with ketone ester delays central nervous system oxygen toxicity seizures in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2013, 304, R829–R836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hashim, S.A.; VanItallie, T.B. Ketone body therapy: From the ketogenic diet to the oral administration of ketone ester. J. Lipid Res. 2014, 55, 1818–1826. [Google Scholar] [CrossRef] [Green Version]
- Scala, R.A.; Paynter, O.E. Chronic oral toxicity of 1,3-butanediol. Toxicol. Appl. Pharmacol. 1967, 10, 160–164. [Google Scholar] [CrossRef]
- Desrochers, S.; David, F.; Garneau, M.; Jetté, M.; Brunengraber, H. Metabolism of R- and S-1,3-butanediol in perfused livers from meal-fed and starved rats. Biochem. J. 1992, 285 Pt 2, 647–653. [Google Scholar] [CrossRef] [Green Version]
- Newman, J.C.; Verdin, E. β-Hydroxybutyrate: A signaling metabolite. Annu. Rev. Nutr. 2017, 37, 51–76. [Google Scholar] [CrossRef]
- Desrochers, S.; Dubreuil, P.; Brunet, J.; Jetté, M.; David, F.; Landau, B.R.; Brunengraber, H. Metabolism of (R,S)-1,3-butanediol acetoacetate esters, potential parenteral and enteral nutrients in conscious pigs. Am. J. Physiol. 1995, 268, E660–E667. [Google Scholar] [CrossRef]
- Webber, R.J.; Edmond, J. 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. 1977, 252, 5222–5226. [Google Scholar] [CrossRef]
- Valenzuela, P.L.; Morales, J.S.; Castillo-García, A.; Lucia, A. Acute Ketone Supplementation and Exercise Performance: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Int. J. Sports Physiol. Perform. 2020, 15, 298–308. [Google Scholar] [CrossRef]
- Thomsen, H.H.; Rittig, N.; Johannsen, M.; Møller, A.B.; Jørgensen, J.O.; Jessen, N.; Møller, N. Effects of 3-hydroxybutyrate and free fatty acids on muscle protein kinetics and signaling during LPS-induced inflammation in humans: Anticatabolic impact of ketone bodies. Am. J. Clin. Nutr. 2018, 108, 857–867. [Google Scholar] [CrossRef] [Green Version]
- Phinney, S.D.; Bistrian, B.R.; Wolfe, R.R.; Blackburn, G.L. The human metabolic response to chronic ketosis without caloric restriction: Physical and biochemical adaptation. Metabolism 1983, 32, 757–768. [Google Scholar] [CrossRef]
- Burke, L.M.; Ross, M.L.; Garvican-Lewis, L.A.; Welvaert, M.; Heikura, I.A.; Forbes, S.G.; Mirtschin, J.G.; Cato, L.E.; Strobel, N.; Sharma, A.P.; et al. Low carbohydrate, high fat diet impairs exercise economy and negates the performance benefit from intensified training in elite race walkers. J. Physiol. 2017, 595, 2785–2807. [Google Scholar] [CrossRef] [Green Version]
- Cox, P.J.; Kirk, T.; Ashmore, T.; Willerton, K.; Evans, R.; Smith, A.; Murray, A.J.; Stubbs, B.; West, J.; McLure, S.W.; et al. Nutritional Ketosis Alters Fuel Preference and Thereby Endurance Performance in Athletes. Cell Metab. 2016, 24, 256–268. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Wang, L.; Chen, P.; Xiao, W. β-hydroxybutyrate as an Anti-Aging Metabolite. Nutrients 2021, 13, 3420. https://doi.org/10.3390/nu13103420
Wang L, Chen P, Xiao W. β-hydroxybutyrate as an Anti-Aging Metabolite. Nutrients. 2021; 13(10):3420. https://doi.org/10.3390/nu13103420
Chicago/Turabian StyleWang, Lian, Peijie Chen, and Weihua Xiao. 2021. "β-hydroxybutyrate as an Anti-Aging Metabolite" Nutrients 13, no. 10: 3420. https://doi.org/10.3390/nu13103420
APA StyleWang, L., Chen, P., & Xiao, W. (2021). β-hydroxybutyrate as an Anti-Aging Metabolite. Nutrients, 13(10), 3420. https://doi.org/10.3390/nu13103420