Ketogenic diet in neuromuscular and neurodegenerative diseases

Antonio Paoli, Antonino Bianco, Ernesto Damiani, Gerardo Bosco, Antonio Paoli, Antonino Bianco, Ernesto Damiani, Gerardo Bosco

Abstract

An increasing number of data demonstrate the utility of ketogenic diets in a variety of metabolic diseases as obesity, metabolic syndrome, and diabetes. In regard to neurological disorders, ketogenic diet is recognized as an effective treatment for pharmacoresistant epilepsy but emerging data suggests that ketogenic diet could be also useful in amyotrophic lateral sclerosis, Alzheimer, Parkinson's disease, and some mitochondriopathies. Although these diseases have different pathogenesis and features, there are some common mechanisms that could explain the effects of ketogenic diets. These mechanisms are to provide an efficient source of energy for the treatment of certain types of neurodegenerative diseases characterized by focal brain hypometabolism; to decrease the oxidative damage associated with various kinds of metabolic stress; to increase the mitochondrial biogenesis pathways; and to take advantage of the capacity of ketones to bypass the defect in complex I activity implicated in some neurological diseases. These mechanisms will be discussed in this review.

Figures

Figure 1
Figure 1
Glucose is necessary not only to supply energy for the central nervous system but also to produce pyruvate that can be transformed into oxaloacetate. Oxaloacetate must be maintained at a level sufficient to allow citric acid cycle function (i.e., the condensation between acetyl-CoA and oxaloacetate). Oxaloacetate is unstable and must be refurnished (this kind of reactions is called anaplerotic). The main way to produce oxaloacetate is from pyruvate that derives from glucose. In mammals pyruvate cannot be produced from acetyl-CoA as shown in the figure.
Figure 2
Figure 2
A reduced availability of dietary carbohydrates leads to an increased liver production of KBs. The liver cannot utilize KBs because it lacks the mitochondrial enzyme succinyl-CoA: 3-ketoacid (oxoacid) CoA transferase (SCOT) necessary for activation of acetoacetate to acetoacetyl-CoA. KBs are utilized by tissues, in particular by brain. KBs enter the citric acid cycle after being converted to acetyl-CoA by succinyl-CoA: 3-CoA transferase (SCOT) and methylacetoacetyl-CoA thiolase (MAT).

References

    1. Danial NN, Hartman AL, Stafstrom CE, Thio LL. How does the ketogenic diet work? Four potential mechanisms. Journal of Child Neurology. 2013;28(8):1027–1033.
    1. Kossoff E. The fat is in the fire: ketogenic diet for refractory status epilepticus. Epilepsy Currents. 2011;11(3):88–89.
    1. Levy RG, Cooper PN, Giri P. Ketogenic diet and other dietary treatments for epilepsy. Cochrane Database of Systematic Reviews. 2012;3
    1. Paoli A. Ketogenic diet for obesity: friend or foe? International Journal of Environmental Research and Public Health. 2014;11:2092–2107.
    1. Mavropoulos JC, Yancy WS, Hepburn J, Westman EC. The effects of a low-carbohydrate, ketogenic diet on the polycystic ovary syndrome: a pilot study. Nutrition and Metabolism. 2005;2, article 35
    1. Klement RJ, Kämmerer U. Is there a role for carbohydrate restriction in the treatment and prevention of cancer? Nutrition & Metabolism. 2011;8, article 75
    1. Seyfried TN, Marsh J, Shelton LM, Huysentruyt LC, Mukherjee P. Is the restricted ketogenic diet a viable alternative to the standard of care for managing malignant brain cancer? Epilepsy Research. 2012;100(3):310–326.
    1. Accurso A, Bernstein RK, Dahlqvist A, et al. Dietary carbohydrate restriction in type 2 diabetes mellitus and metabolic syndrome: time for a critical appraisal. Nutrition and Metabolism. 2008;5(1, article 9)
    1. Paoli A, Rubini A, Volek JS, Grimaldi KA. Beyond weight loss: a review of the therapeutic uses of very-low-carbohydrate (ketogenic) diets. European Journal of Clinical Nutrition. 2013;67(8):789–796.
    1. Paoli A, Grimaldi K, Toniolo L, Canato M, Bianco A, Fratter A. Nutrition and acne: therapeutic potential of ketogenic diets. Skin Pharmacology and Physiology. 2012;25(3):111–117.
    1. Paoli A, Canato M, Toniolo L, et al. The ketogenic diet: an underappreciated therapeutic option? La Clinica Terapeutica. 2011;162(5):e145–e153.
    1. Stafstrom CE, Rho JM. The ketogenic diet as a treatment paradigm for diverse neurological disorders. Frontiers in Pharmacology. 2012;3(article 59)
    1. Felig P, Owen OE, Wahren J, Cahill GF., Jr. Amino acid metabolism during prolonged starvation. Journal of Clinical Investigation. 1969;48(3):584–594.
    1. Owen OE, Felig P, Morgan AP, Wahren J, Cahill GF., Jr. Liver and kidney metabolism during prolonged starvation. The Journal of Clinical Investigation. 1969;48(3):574–583.
    1. Jitrapakdee S, Vidal-Puig A, Wallace JC. Anaplerotic roles of pyruvate carboxylase in mammalian tissues. Cellular and Molecular Life Sciences. 2006;63(7-8):843–854.
    1. George FC. Fuel metabolism in starvation. Annual Review of Nutrition. 2006;26:1–22.
    1. Owen OE, Morgan AP, Kemp HG, Sullivan JM, Herrera MG, Cahill GF., Jr. Brain metabolism during fasting. Journal of Clinical Investigation. 1967;46(10):1589–1595.
    1. Fukao T, Mitchell G, Sass JO, Hori T, Orii K, Aoyama Y. Ketone body metabolism and its defects. Journal of Inherited Metabolic Disease. 2014
    1. Fukao T, Lopaschuk GD, Mitchell GA. Pathways and control of ketone body metabolism: on the fringe of lipid biochemistry. Prostaglandins Leukotrienes and Essential Fatty Acids. 2004;70(3):243–251.
    1. Paoli A, Cenci L, Fancelli M, et al. Ketogenic diet and phytoextracts comparison of the efficacy of mediterranean, zone and tisanoreica diet on some health risk factors. Agro Food Industry Hi-Tech. 2010;21(4):24–29.
    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 Leukotrienes and Essential Fatty Acids. 2004;70(3):309–319.
    1. Leino RL, Gerhart DZ, Duelli R, Enerson BE, Drewes LR. Diet-induced ketosis increases monocarboxylate transporter (MCT1) levels in rat brain. Neurochemistry International. 2001;38(6):519–527.
    1. McCue MD. Starvation physiology: reviewing the different strategies animals use to survive a common challenge. Comparative Biochemistry and Physiology A: Molecular & Integrative Physiology. 2010;156(1):1–18.
    1. Sato K, Kashiwaya Y, Keon CA, et al. Insulin, ketone bodies, and mitochondrial energy transduction. The FASEB Journal. 1995;9(8):651–658.
    1. Kashiwaya Y, Sato K, Tsuchiya N, et al. Control of glucose utilization in working perfused rat heart. The Journal of Biological Chemistry. 1994;269(41):25502–25514.
    1. Paoli A, Cenci L, Grimaldi KA. Effect of ketogenic mediterranean diet with phytoextracts and low carbohydrates/high-protein meals on weight, cardiovascular risk factors, body composition and diet compliance in Italian council employees. Nutrition Journal. 2011;10(1, article 112)
    1. Seyfried TN, Mukherjee P. Targeting energy metabolism in brain cancer: review and hypothesis. Nutrition and Metabolism. 2005;2, article 30
    1. Vazquez JA, Kazi U. Lipolysis and gluconeogenesis from glycerol during weight reduction with very-low-calorie diets. Metabolism: Clinical and Experimental. 1994;43(10):1293–1299.
    1. Veldhorst MAB, Westerterp-Plantenga MS, Westerterp KR. Gluconeogenesis and energy expenditure after a high-protein, carbohydrate-free diet. The American Journal of Clinical Nutrition. 2009;90(3):519–526.
    1. Robinson AM, Williamson DH. Physiological roles of ketone bodies as substrates and signals in mammalian tissues. Physiological Reviews. 1980;60(1):143–187.
    1. Krebs HA. The regulation of the release of ketone bodies by the liver. Advances in Enzyme Regulation. 1966;4:339–353.
    1. Amen-Ra N. Humans are evolutionarily adapted to caloric restriction resulting from ecologically dictated dietary deprivation imposed during the Plio-Pleistocene period. Medical Hypotheses. 2006;66(5):978–984.
    1. Newman JC, Verdin E. Ketone bodies as signaling metabolites. Trends in Endocrinology and Metabolism. 2014;25(1):42–52.
    1. Jäer S, Handschin C, St-Pierre J, Spiegelman BM. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1α . Proceedings of the National Academy of Sciences of the United States of America. 2007;104(29):12017–12022.
    1. Benton CR, Wright DC, Bonen A. PGC-1alpha-mediated regulation of gene expression and metabolism: implications for nutrition and exercise prescriptions. Applied Physiology, Nutrition and Metabolism. 2008;33(5):843–862.
    1. Yu J, Auwerx J. Protein deacetylation by SIRT1: an emerging key post-translational modification in metabolic regulation. Pharmacological Research. 2010;62(1):35–41.
    1. Williamson DL. Normalizing a hyperactive mTOR initiates muscle growth during obesity. Aging. 2011;3(2):83–84.
    1. Draznin B, Wang C, Adochio R, Leitner JW, Cornier M-A. Effect of dietary macronutrient composition on AMPK and SIRT1 expression and activity in human skeletal muscle. Hormone and Metabolic Research. 2012;44(9):650–655.
    1. Yoon JC, Puigserver P, Chen G, et al. Control of hepatic gluconeogenesis through the transcriptional coaotivator PGC-1. Nature. 2001;413(6852):131–138.
    1. Ruderman NB, Xu XJ, Nelson L, et al. AMPK and SIRT1: a long-standing partnership? The American Journal of Physiology: Endocrinology and Metabolism. 2010;298(4):E751–E760.
    1. Miller RG, Mitchell JD, Lyon M, Moore DH. Riluzole for amyotrophic lateral sclerosis (ALS)/motor neuron disease (MND) Cochrane Database of Systematic Reviews. 2002;(2)CD001447
    1. Bruijn LI, Miller TM, Cleveland DW. Unraveling the mechanisms involved in motor neuron degeneration in ALS. Annual Review of Neuroscience. 2004;27:723–749.
    1. Rowland LP, Shneider NA. Amyotrophic lateral sclerosis. The New England Journal of Medicine. 2001;344(22):1688–1700.
    1. Strong M, Rosenfeld J. Amyotrophic lateral sclerosis: a review of current concepts. Amyotrophic Lateral Sclerosis and Other Motor Neuron Disorders. 2003;4(3):136–143.
    1. Siva N. Can ketogenic diet slow progression of ALS? The Lancet Neurology. 2006;5(6, article 476)
    1. Rosen DR, Siddique T, Patterson D, et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;362(6415):59–62.
    1. Pasinelli P, Belford ME, Lennon N, et al. Amyotrophic lateral sclerosis-associated SOD1 mutant proteins bind and aggregate with Bcl-2 in spinal cord mitochondria. Neuron. 2004;43(1):19–30.
    1. Vielhaber S, Kunz D, Winkler K, et al. Mitochondrial DNA abnormalities in skeletal muscle of patients with sporadic amyotrophic lateral sclerosis. Brain. 2000;123(7):1339–1348.
    1. Tieu K, Perier C, Caspersen C, et al. D-β-Hydroxybutyrate rescues mitochondrial respiration and mitigates features of Parkinson disease. Journal of Clinical Investigation. 2003;112(6):892–901.
    1. Zhao Z, Lange DJ, Voustianiouk A, et al. A ketogenic diet as a potential novel therapeutic intervention in amyotrophic lateral sclerosis. BMC Neuroscience. 2006;7, article 29
    1. Zhao W, Varghese M, Vempati P, et al. Caprylic triglyceride as a novel therapeutic approach to effectively improve the performance and attenuate the symptoms due to the motor neuron loss in ALS disease. PLoS ONE. 2012;7(11)e49191
    1. Posse de Chaves EI, Rusinol AE, Vance DE, Campenot RB, Vance JE. Role of lipoproteins in the delivery of lipids to axons during axonal regeneration. Journal of Biological Chemistry. 1997;272(49):30766–30773.
    1. Dupuis L, Corcia P, Fergani A, et al. Dyslipidemia is a protective factor in amyotrophic lateral sclerosis. Neurology. 2008;70(13):1004–1009.
    1. Paganoni S, Deng J, Jaffa M, Cudkowicz ME, Wills A. Body mass index, not dyslipidemia, is an independent predictor of survival in amyotrophic lateral sclerosis. Muscle and Nerve. 2011;44(1):20–24.
    1. Wills AM, Hubbard J, Macklin EA, et al. Hypercaloric enteral nutrition in patients with amyotrophic lateral sclerosis: a randomised, double-blind, placebo-controlled phase 2 trial. The Lancet. 2014;383(9934):2065–2072.
    1. Fergani A, Oudart H, De Aguilar JG, et al. Increased peripheral lipid clearance in an animal model of amyotrophic lateral sclerosis. Journal of Lipid Research. 2007;48(7):1571–1580.
    1. Bough KJ, Wetherington J, Hassel B, et al. Mitochondrial biogenesis in the anticonvulsant mechanism of the ketogenic diet. Annals of Neurology. 2006;60(2):223–235.
    1. Kim DY, Vallejo J, Rho JM. Ketones prevent synaptic dysfunction induced by mitochondrial respiratory complex inhibitors. Journal of Neurochemistry. 2010;114(1):130–141.
    1. McDaniel SS, Rensing NR, Thio LL, Yamada KA, Wong M. The ketogenic diet inhibits the mammalian target of rapamycin (mTOR) pathway. Epilepsia. 2011;52(3):e7–e11.
    1. Srivastava S, Kashiwaya Y, King MT, et al. Mitochondrial biogenesis and increased uncoupling protein 1 in brown adipose tissue of mice fed a ketone ester diet. FASEB Journal. 2012;26(6):2351–2362.
    1. Wallace DC, Fan W, Procaccio V. Mitochondrial energetics and therapeutics. Annual Review of Pathology. 2010;5:297–348.
    1. Kang H, Lee Y, Kim HD. Mitochondrial disease and epilepsy. Brain and Development. 2013;35(8):757–761.
    1. Kang H, Lee Y, Kim HD, Lee JS, Slama A. Safe and effective use of the ketogenic diet in children with epilepsy and mitochondrial respiratory chain complex defects. Epilepsia. 2007;48(1):82–88.
    1. Ahola-Erkkilä S, Carroll CJ, Peltola-Mjösund K, et al. Ketogenic diet slows down mitochondrial myopathy progression in mice. Human Molecular Genetics. 2010;19(10):1974–1984.ddq076
    1. Kossoff EH, Zupec-Kania BA, Amark PE, et al. Optimal clinical management of children receiving the ketogenic diet: recommendations of the International Ketogenic Diet Study Group. Epilepsia. 2009;50(2):304–317.
    1. Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT. Neuropathological alterations in Alzheimer disease. Cold Spring Harbor perspectives in Medicine. 2011;1(1)a006189
    1. Ridge PG, Ebbert MTW, Kauwe JSK. Genetics of alzheimer's disease. BioMed Research International. 2013;2013:13 pages.254954
    1. Weuve J, Hebert LE, Scherr PA, Evans DA. Deaths in the United States among persons with Alzheimer's disease (2010–2050) Alzheimer's & Dementia. 2014;10(2):e40–e46.
    1. Balin BJ, Hudson AP. Etiology and pathogenesis of late-onset Alzheimer's disease. Current Allergy and Asthma Reports. 2014;14, article 417
    1. Holtzman DM, Mandelkow E, Selkoe DJ. Alzheimer disease in 2020. Cold Spring Harbor Perspectives in Medicine. 2012;2(11)
    1. Selkoe D, Mandelkow E, Holtzman D. Deciphering Alzheimer disease. Cold Spring Harbor perspectives in Medicine. 2012;2(1)a011460
    1. Yao J, Brinton RD. Targeting mitochondrial bioenergetics for Alzheimer's prevention and treatment. Current Pharmaceutical Design. 2011;17(31):3474–3479.
    1. Hashim SA, Vanitallie TB. Ketone Body Therapy: from ketogenic diet to oral administration of ketone ester. Journal of Lipid Research. 2014
    1. Kashiwaya Y, Takeshima T, Mori N, Nakashima K, Clarke K, Veech RL. D-beta-hydroxybutyrate protects neurons in models of Alzheimer's and Parkinson's disease. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(10):5440–5444.
    1. Kashiwaya Y, Bergman C, Lee J, et al. A ketone ester diet exhibits anxiolytic and cognition-sparing properties, and lessens amyloid and tau pathologies in a mouse model of Alzheimer's disease. Neurobiology of Aging. 2013;34(6):1530–1539.
    1. Van Der Auwera I, Wera S, Van Leuven F, Henderson ST. A ketogenic diet reduces amyloid beta 40 and 42 in a mouse model of Alzheimer's disease. Nutrition and Metabolism. 2005;2, article 28
    1. Studzinski CM, MacKay WA, Beckett TL, et al. Induction of ketosis may improve mitochondrial function and decrease steady-state amyloid-β precursor protein (APP) levels in the aged dog. Brain Research. 2008;1226:209–217.
    1. Beckett TL, Studzinski CM, Keller JN, Paul Murphy M, Niedowicz DM. A ketogenic diet improves motor performance but does not affect β-amyloid levels in a mouse model of Alzheimer's disease. Brain Research. 2013;1505:61–67.
    1. Akter K, Lanza EA, Martin SA, Myronyuk N, Rua M, Raffa RB. Diabetes mellitus and Alzheimer's disease: shared pathology and treatment? British Journal of Clinical Pharmacology. 2011;71(3):365–376.
    1. Dashti HM, Mathew TC, Khadada M, et al. Beneficial effects of ketogenic diet in obese diabetic subjects. Molecular and Cellular Biochemistry. 2007;302(1-2):249–256.
    1. Paoli A, Bianco A, Grimaldi KA, Lodi A, Bosco G. Long term successful weight loss with a combination biphasic ketogenic mediterranean diet and mediterranean diet maintenance protocol. Nutrients. 2013;5(12):5205–5217.
    1. Westman EC, Yancy WS, Jr., Mavropoulos JC, Marquart M, McDuffie JR. The effect of a low-carbohydrate, ketogenic diet versus a low-glycemic index diet on glycemic control in type 2 diabetes mellitus. Nutrition and Metabolism. 2008;5(1, article 36)
    1. Srikanth V, Maczurek A, Phan T, et al. Advanced glycation endproducts and their receptor RAGE in Alzheimer's disease. Neurobiology of Aging. 2011;32(5):763–777.
    1. Balietti M, Giorgetti B, Di Stefano G, et al. A ketogenic diet increases succinic dehydrogenase (SDH) activity and recovers age-related decrease in numeric density of SDH-positive mitochondria in cerebellar Purkinje cells of late-adult rats. Micron. 2010;41(2):143–148.
    1. Balietti M, Fattoretti P, Giorgetti B, et al. A ketogenic diet increases succinic dehydrogenase activity in aging cardiomyocytes. Annals of the New York Academy of Sciences. 2009;1171:377–384.
    1. Maalouf M, Rho JM, Mattson MP. The neuroprotective properties of calorie restriction, the ketogenic diet, and ketone bodies. Brain Research Reviews. 2009;59(2):293–315.
    1. Boumezbeur F, Mason GF, de Graaf RA, et al. Altered brain mitochondrial metabolism in healthy aging as assessed by in vivo magnetic resonance spectroscopy. Journal of Cerebral Blood Flow & Metabolism. 2010;30(1):211–221.
    1. Hoyer S. Causes and consequences of disturbances of cerebral glucose metabolism in sporadic alzheimer disease: therapeutic Implications. Advances in Experimental Medicine and Biology. 2003;541:135–152.
    1. Mosconi L, Mistur R, Switalski R, et al. Declining brain glucose metabolism in normal individuals with a maternal history of Alzheimer disease. Neurology. 2009;72(6):513–520.
    1. Vanitallie TB. Preclinical sporadic Alzheimer's disease: target for personalized diagnosis and preventive intervention. Metabolism: Clinical and Experimental. 2013;62(supplement 1):S30–S33.
    1. Liu Y, Liu F, Iqbal K, Grundke-Iqbal I, Gong C. Decreased glucose transporters correlate to abnormal hyperphosphorylation of tau in Alzheimer disease. FEBS Letters. 2008;582(2):359–364.
    1. Veggiotti P, Teutonico F, Alfei E, et al. Glucose transporter type 1 deficiency: Ketogenic diet in three patients with atypical phenotype. Brain and Development. 2010;32(5):404–408.
    1. Reger MA, Henderson ST, Hale C, et al. Effects of β-hydroxybutyrate on cognition in memory-impaired adults. Neurobiology of Aging. 2004;25(3):311–314.
    1. Camilleri A, Vassallo N. The centrality of mitochondria in the pathogenesis and treatment of Parkinson's disease. CNS Neuroscience & Therapeutics. 2014;20(7):591–602.
    1. Cheng B, Yang X, An L, Gao B, Liu X, Liu S. Ketogenic diet protects dopaminergic neurons against 6-OHDA neurotoxicity via up-regulating glutathione in a rat model of Parkinson's disease. Brain Research. 2009;1286:25–31.
    1. VanItallie TB, Nonas C, Di Rocco A, Boyar K, Hyams K, Heymsfield SB. Treatment of Parkinson disease with diet-induced hyperketonemia: a feasibility study. Neurology. 2005;64(4):728–730.
    1. Shin YS. Glycogen storage disease: clinical, biochemical, and molecular heterogeneity. Seminars in Pediatric Neurology. 2006;13(2):115–120.
    1. Gazzerro E, Andreu AL, Bruno C. Neuromuscular disorders of glycogen metabolism. Current Neurology and Neuroscience Reports. 2013;13, article 333
    1. Goldberg T, Slonim AE. Nutrition therapy for hepatic glycogen storage diseases. Journal of the American Dietetic Association. 1993;93(12):1423–1430.
    1. Heller S, Worona L, Consuelo A. Nutritional therapy for glycogen storage diseases. Journal of Pediatric Gastroenterology and Nutrition. 2008;47:S15–S21.
    1. Triomphe TJ. Glycogen storage disease: a basic understanding and guide to nursing care. Journal of pediatric nursing. 1997;12(4):238–249.
    1. Walter JB. General Pathology. 1987.
    1. Busch V, Gempel K, Hack A, et al. Treatment of glycogenosis type V with ketogenic diet. Annals of Neurology. 2005;58(2, article 341)
    1. Vorgerd M, Zange J. Treatment of glycogenosys type V (McArdle disease) with creatine and ketogenic diet with clinical scores and with 31P-MRS on working leg muscle. Acta Myologica. 2007;26(1):61–63.
    1. Rho JM, Sankar R. The ketogenic diet in a pill: is this possible? Epilepsia. 2008;49(supplement 8):127–133.
    1. Veggiotti P, De Giorgis V. Dietary treatments and new therapeutic perspective in GLUT1 deficiency syndrome. Current Treatment Options in Neurology. 2014;16(5):p. 291.
    1. Valayannopoulos V, Bajolle F, Arnoux J, et al. Successful treatment of severe cardiomyopathy in glycogen storage disease type III with d,l-3-hydroxybutyrate, ketogenic and high-protein diet. Pediatric Research. 2011;70(6):638–641.

Source: PubMed

Подписаться