Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Intermittent metabolic switching, neuroplasticity and brain health

A Publisher Correction to this article was published on 30 June 2020

Key Points

  • Brain evolution, including higher cortical functions of humans (imagination, creativity and language), was driven by the necessity of sustaining high levels of performance in a food-deprived (fasted) state

  • Intermittent metabolic switching (IMS) occurs when eating and exercise patterns result in periodic depletion of liver glycogen stores and the associated production of ketones from fatty acids. IMS occurs rarely or not at all in individuals who eat three or more meals per day and who are fairly sedentary

  • The ketone β-hydroxybutyrate (BHB) is transported into the brain and into neuronal mitochondria, where it is used to generate acetyl CoA and ATP. BHB also acts as a signalling molecule in neurons that can induce the expression of brain-derived neurotrophic factor and thereby promote synaptic plasticity and cellular stress resistance

  • During fasting and extended exercise, adaptive cellular stress-response signalling pathways are activated and autophagy is stimulated, whereas overall protein synthesis is reduced. Upon refeeding, rest and sleep, protein synthesis is upregulated and mitochondrial biogenesis occurs, enabling neurogenesis and synaptogenesis

  • IMS can enhance cognition and motor performance and protects neurons against dysfunction and degeneration in animal models of stroke, epilepsy, traumatic brain and spinal cord injury, Alzheimer disease and Parkinson disease

  • Intermittent fasting can improve indicators of metabolic and cardiovascular health in humans by mechanisms involving reductions in oxidative damage and inflammation. However, it remains to be determined whether and how intermittent fasting impacts the brains of healthy humans and those affected with a neurological disorder

Abstract

During evolution, individuals whose brains and bodies functioned well in a fasted state were successful in acquiring food, enabling their survival and reproduction. With fasting and extended exercise, liver glycogen stores are depleted and ketones are produced from adipose-cell-derived fatty acids. This metabolic switch in cellular fuel source is accompanied by cellular and molecular adaptations of neural networks in the brain that enhance their functionality and bolster their resistance to stress, injury and disease. Here, we consider how intermittent metabolic switching, repeating cycles of a metabolic challenge that induces ketosis (fasting and/or exercise) followed by a recovery period (eating, resting and sleeping), may optimize brain function and resilience throughout the lifespan, with a focus on the neuronal circuits involved in cognition and mood. Such metabolic switching impacts multiple signalling pathways that promote neuroplasticity and resistance of the brain to injury and disease.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Biochemical pathways involved in the metabolic switch.
Figure 2: Signalling pathways by which neurons respond to the metabolic switch during fasting and exercise.
Figure 3: Model for how intermittent metabolic switching may optimize brain performance and increase resistance to injury and disease.

Similar content being viewed by others

References

  1. Mattson, M. P. Lifelong brain health is a lifelong challenge: from evolutionary principles to empirical evidence. Ageing Res. Rev. 20, 37–45 (2015).

    PubMed  Google Scholar 

  2. Bramble, D. M. & Lieberman, D. E. Endurance running and the evolution of Homo. Nature 432, 345–352 (2004).

    CAS  PubMed  Google Scholar 

  3. Mattson, M. P. Superior pattern processing is the essence of the evolved human brain. Front. Neurosci. 8, 265 (2014).

    PubMed  PubMed Central  Google Scholar 

  4. Courchesne-Loyer, A. et al. Inverse relationship between brain glucose and ketone metabolism in adults during short-term moderate dietary ketosis: a dual tracer quantitative positron emission tomography study. J. Cereb. Blood Flow Metab. 37, 2485–2493 (2017).

    CAS  PubMed  Google Scholar 

  5. Camandola, S. & Mattson, M. P. Brain metabolism in health, aging, and neurodegeneration. EMBO J. 36, 1474–1492 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Mattson, M. P. Energy intake and exercise as determinants of brain health and vulnerability to injury and disease. Cell Metab. 16, 706–722 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Longo, V. D. & Mattson, M. P. Fasting: molecular mechanisms and clinical applications. Cell Metab. 19, 181–192 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Anson, R. M. et al. Intermittent fasting dissociates beneficial effects of dietary restriction on glucose metabo-lism and neuronal resistance to injury from calorie intake. Proc. Natl Acad. Sci. USA 100, 6216–6220 (2003). The data in this study provide the first evidence that IMS exerts beneficial effects on the brain that are independent of overall energy intake.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Hatori, M. et al. Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Cell Metab. 15, 848–860 (2012). This study finds that 16 hours of fasting each day can prevent obesity and related metabolic morbidities in mice fed a high-fat diet.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Harvie, M. N. et al. The effects of intermittent or continuous energy restriction on weight loss and metabolic disease risk markers: a randomized trial in young overweight women. Int. J. Obes. (Lond.). 35, 714–727 (2011).

    CAS  Google Scholar 

  11. Harvie, M. et al. The effect of intermittent energy and carbohydrate restriction v. daily energy restriction on weight loss and metabolic disease risk markers in overweight women. Br. J. Nutr. 110, 1534–1547 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Vanevski, F. & Xu, B. Molecular and neural bases underlying roles of BDNF in the control of body weight. Front. Neurosci. 7, 37 (2013).

    PubMed  PubMed Central  Google Scholar 

  13. Bechtold, D. A. & Loudon, A. S. Hypothalamic clocks and rhythms in feeding behaviour. Trends Neurosci. 36, 74–82 (2013).

    CAS  PubMed  Google Scholar 

  14. Ingram, D. K., Weindruch, R., Spangler, E. L., Freeman, J. R. & Walford, R. L. Dietary restriction benefits learning and motor performance of aged mice. J. Gerontol. 42, 78–81 (1987).

    CAS  PubMed  Google Scholar 

  15. Means, L. W., Higgins, J. L. & Fernandez, T. J. Mid-life onset of dietary restriction extends life and prolongs cognitive functioning. Physiol. Behav. 54, 503–508 (1993).

    CAS  PubMed  Google Scholar 

  16. Parikh, I. et al. Caloric restriction preserves memory and reduces anxiety of aging mice with early enhancement of neurovascular functions. Aging 8, 2814–2826 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Brandhorst, S. et al. A periodic diet that mimics fasting promotes multi-system regeneration, enhanced cognitive performance, and healthspan. Cell Metab. 22, 86–99 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Stillman, C. M., Cohen, J., Lehman, M. E. & Erickson, K. I. Mediators of physical activity on neurocognitive function: a review at multiple levels of analysis. Front. Hum. Neurosci. 10, 626 (2016).

    PubMed  PubMed Central  Google Scholar 

  19. Cooper, C., Moon, H. Y. & van Praag, H. On the run for hippocampal plasticity. Cold Spring Harb. Perspect. Med. http://dx.doi.org/10.1101/cshperspect.a029736 (2017).

  20. Raichlen, D. A. & Alexander, G. E. Adaptive capacity: an evolutionary neuroscience model linking exercise, cognition, and brain health. Trends Neurosci. 40, 408–421 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Stranahan, A. M. et al. Voluntary exercise and caloric restriction enhance hippocampal dendritic spine density and BDNF levels in diabetic mice. Hippocampus 19, 951–961 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhu, X., Yin, S., Lang, M., He, R. & Li, J. The more the better? A meta-analysis on effects of combined cognitive and physical intervention on cognition in healthy older adults. Ageing Res. Rev. 31, 67–79 (2016).

    PubMed  Google Scholar 

  23. Hippocrates. On the Sacred Disease (400 bce). English translation by F. Adams: http://classics.mit.edu/Hippocrates/sacred.html

  24. Ruskin, D. N. & Masino, S. A. The nervous system and metabolic dysregulation: emerging evidence converges on ketogenic diet therapy. Front. Neurosci. 6, 33 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Bruce-Keller, A. J., Umberger, G., McFall, R. & Mattson, M. P. Food restriction reduces brain damage and improves behavioral outcome following excitotoxic and metabolic insults. Ann. Neurol. 45, 8–15 (1999).

    CAS  PubMed  Google Scholar 

  26. Hartman, A. L., Rubenstein, J. E. & Kossoff, E. H. Intermittent fasting: a “new” historical strategy for controlling seizures? Epilepsy Res. 104, 275–279 (2013).

    PubMed  Google Scholar 

  27. van Praag, H., Christie, B. R., Sejnowski, T. J. & Gage, F. H. Running enhances neurogenesis, learning, and long-term potentiation in mice. Proc. Natl Acad. Sci. USA 96, 13427–13431 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Farmer, J. et al. Effects of voluntary exercise on synaptic plasticity and gene expression in the dentate gyrus of adult male Sprague–Dawley rats in vivo. Neuroscience 124, 71–79 (2004).

    CAS  PubMed  Google Scholar 

  29. O'Callaghan, R. M., Ohle, R. & Kelly, A. M. The effects of forced exercise on hippocampal plasticity in the rat: a comparison of LTP, spatial- and non-spatial learning. Behav. Brain Res. 176, 362–366 (2007).

    PubMed  Google Scholar 

  30. Talani, G. et al. Enhanced glutamatergic synaptic plasticity in the hippocampal CA1 field of food-restricted rats: involvement of CB1 receptors. Neuropsychopharmacology 41, 1308–1318 (2016).

    CAS  PubMed  Google Scholar 

  31. Eckles-Smith, K., Clayton, D., Bickford, P. & Browning, M. D. Caloric restriction prevents age-related deficits in LTP and in NMDA receptor expression. Mol. Brain Res. 78, 154–162 (2000).

    CAS  PubMed  Google Scholar 

  32. Hori, N., Hirotsu, I., Davis, P. J. & Carpenter, D. O. Long-term potentiation is lost in aged rats but preserved by calorie restriction. Neuroreport 3, 1085–1088 (1992).

    CAS  PubMed  Google Scholar 

  33. Vivar, C. et al. Monosynaptic inputs to new neurons in the dentate gyrus. Nat. Commun. 3, 1107 (2012).

    PubMed  Google Scholar 

  34. Lee, J., Duan, W. & Mattson, M. P. Evidence that brain-derived neurotrophic factor is required for basal neurogenesis and mediates, in part, the enhancement of neurogenesis by dietary restriction in the hippocampus of adult mice. J. Neurochem. 82, 1367–1375 (2002).

    CAS  PubMed  Google Scholar 

  35. Vivar, C., Peterson, B. D. & van Praag, H. Running rewires the neuronal network of adult-born dentate granule cells. Neuroimage 131, 29–41 (2016).

    PubMed  Google Scholar 

  36. Cohen, S. M., Li, B., Tsien, R. W. & Ma, H. Evolutionary and functional perspectives on signaling from neuronal surface to nucleus. Biochem. Biophys. Res. Commun. 460, 88–99 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Mattson, M. P. & Meffert, M. K. Roles for NF-kappaB in nerve cell survival, plasticity, and disease. Cell Death Differ. 13, 852–860 (2006).

    CAS  PubMed  Google Scholar 

  38. Estrada, N. M. & Isokawa, M. Metabolic demand stimulates CREB signaling in the limbic cortex: implication for the induction of hippocampal synaptic plasticity by intrinsic stimulus for survival. Front. Syst. Neurosci. 3, 5 (2009).

    PubMed  PubMed Central  Google Scholar 

  39. Yang, J. L., Lin, Y. T., Chuang, P. C., Bohr, V. A. & Mattson, M. P. BDNF and exercise enhance neuronal DNA repair by stimulating CREB-mediated production of apurinic/apyrimidinic endonuclease 1. Neuromolecular Med. 16, 161–174 (2014).

    CAS  PubMed  Google Scholar 

  40. Hirano, Y. et al. Fasting launches CRTC to facilitate long-term memory formation in Drosophila. Science 339, 443–446 (2013).

    CAS  PubMed  Google Scholar 

  41. Rogawski, M. A., Löscher, W. & Rho, J. M. Mechanisms of action of antiseizure drugs and the ketogenic diet. Cold Spring Harb. Perspect. Med. 6, a022780 (2016).

    PubMed  PubMed Central  Google Scholar 

  42. Li, J., O'Leary, E. I. & Tanner, G. R. The ketogenic diet metabolite beta-hydroxybutyrate (β-HB) reduces incidence of seizure-like activity (SLA) in a Katp- and GABAb-dependent manner in a whole-animal Drosophila melanogaster model. Epilepsy Res. 133, 6–9 (2017).

    CAS  PubMed  Google Scholar 

  43. Blier, P. & El Mansari, M. Serotonin and beyond: therapeutics for major depression. Phil. Trans. R. Soc. B Biol Sci. 368, 20120536 (2013).

    Google Scholar 

  44. Kondo, M., Nakamura, Y., Ishida, Y. & Shimada, S. The 5-HT3 receptor is essential for exercise-induced hippocampal neurogenesis and antidepressant effects. Mol. Psychiatry. 20, 1428–1437 (2015).

    CAS  PubMed  Google Scholar 

  45. Billman, G. E. et al. Exercise training-induced bradycardia: evidence for enhanced parasympathetic regulation without changes in intrinsic sinoatrial node function. J. Appl. Physiol. 118, 1344–1355 (2015).

    PubMed  PubMed Central  Google Scholar 

  46. Wan, R., Camandola, S. & Mattson, M. P. Intermittent food deprivation improves cardiovascular and neuroendocrine responses to stress in rats. J. Nutr. 133, 1921–1929 (2003).

    CAS  PubMed  Google Scholar 

  47. Mager, D. E. et al. Caloric restriction and intermittent fasting alter spectral measures of heart rate and blood pressure variability in rats. FASEB J. 20, 631–637 (2006). This study establishes that IF lowers blood pressure and heart rate and increases heart rate variability by a mechanism involving enhancement of parasympathetic tone.

    CAS  PubMed  Google Scholar 

  48. Wan, R. et al. Evidence that BDNF regulates heart rate by a mechanism involving increased brainstem parasympathetic neuron excitability. J. Neurochem. 129, 573–580 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Marosi, K. & Mattson, M. P. BDNF mediates adaptive brain and body responses to energetic challenges. Trends Endocrinol. Metab. 25, 89–98 (2014).

    CAS  PubMed  Google Scholar 

  50. Li, Y. et al. TrkB regulates hippocampal neurogenesis and governs sensitivity to antidepressive treatment. Neuron 59, 399–412 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Cheng, C. M. et al. A ketogenic diet increases brain insulin-like growth factor receptor and glucose transporter gene expression. Endocrinology 144, 2676–2682 (2003).

    CAS  PubMed  Google Scholar 

  52. Bohannon, N. J. et al. Characterization of insulin-like growth factor I receptors in the median eminence of the brain and their modulation by food restriction. Endocrinology 122, 1940–1947 (1988).

    CAS  PubMed  Google Scholar 

  53. Llorens-Martín, M., Torres-Alemán, I. & Trejo, J. L. Mechanisms mediating brain plasticity: IGF1 and adult hippocampal neurogenesis. Neuroscientist 15, 134–148 (2009).

    PubMed  Google Scholar 

  54. Marosi, K. et al. 3-Hydroxybutyrate regulates energy metabolism and induces BDNF expression in cerebral cortical neurons. J. Neurochem. 139, 769–781 (2016). This study shows that the ketone BHB can act directly on neurons to induce transcription of the gene encoding BDNF by a mechanism involving NF-κB.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Sleiman, S. F. et al. Exercise promotes the expression of brain derived neurotrophic factor (BDNF) through the action of the ketone body β-hydroxybutyrate. eLife 5, e1509 (2016). This study finds that the ketone BHB can induce Bdnf gene expression in the hippocampus in vivo.

    Google Scholar 

  56. Zechel, S., Werner, S., Unsicker, K. & von Bohlen und Halbach, O. Expression and functions of fibroblast growth factor 2 (FGF-2) in hippocampal formation. Neuroscientist 16, 357–373 (2010).

    CAS  PubMed  Google Scholar 

  57. Mattson, M. P., Murrain, M., Guthrie, P. B. & Kater, S. B. Fibroblast growth factor and glutamate: opposing roles in the generation and degeneration of hippocampal neuroarchitecture. J. Neurosci. 9, 3728–3740 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Mark, R. J., Keller, J. N., Kruman, I. & Mattson, M. P. Basic FGF attenuates amyloid beta-peptide-induced oxidative stress, mitochondrial dysfunction, and impairment of Na+/K+-ATPase activity in hippocampal neurons. Brain Res. 756, 205–214 (1997).

    CAS  PubMed  Google Scholar 

  59. Arumugam, T. V. et al. Age and energy intake interact to modify cell stress pathways and stroke outcome. Ann. Neurol. 67, 41–52 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Gómez-Pinilla, F., Dao, L. & So, V. Physical exercise induces FGF-2 and its mRNA in the hippocampus. Brain Res. 764, 1–8 (1997).

    PubMed  Google Scholar 

  61. Graham, B. M. & Richardson, R. Memory of fearful events; the role of fibroblast growth factor-2 in fear acquisition and extinction. Neuroscience 189, 156–169 (2011).

    CAS  PubMed  Google Scholar 

  62. Hood, D. A., Tryon, L. D., Carter, H. N., Kim, Y. & Chen, C. C. Unravelling the mechanisms regulating muscle mitochondrial biogenesis. Biochem. J. 473, 2295–2314 (2016).

    CAS  PubMed  Google Scholar 

  63. Kerr, J. S. et al. Mitophagy and Alzheimer's disease: cellular and molecular mechanisms. Trends Neurosci. 40, 151–166 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Hepple, R. T. Why eating less keeps mitochondria working in aged skeletal muscle. Exerc. Sport Sci. Rev. 37, 23–28 (2009).

    PubMed  Google Scholar 

  65. Cheng, A. et al. Involvement of PGC-1α in the formation and maintenance of neuronal dendritic spines. Nat. Commun. 3, 1250 (2012).

    PubMed  Google Scholar 

  66. Wrann, C. D. et al. Exercise induces hippocampal BDNF through a PGC-1α/FNDC5 pathway. Cell Metab. 18, 649–659 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Fusco, S. et al. A role for neuronal cAMP responsive-element binding (CREB)-1 in brain responses to calorie restriction. Proc. Natl Acad. Sci. USA 109, 621–626 (2012).

    CAS  PubMed  Google Scholar 

  68. van de Ven, R. A. H., Santos, D. & Haigis, M. C. Mitochondrial sirtuins and molecular mechanisms of aging. Trends Mol. Med. 23, 320–331 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Cheng, A. et al. Mitochondrial SIRT3 mediates adaptive responses of neurons to exercise and metabolic and excitatory challenges. Cell Metab. 23, 128–142 (2016). This study shows that the mitochondrial deacetylase SIRT3 is upregulated in response to exercise and excitatory synaptic activity and that SIRT3 protects neurons against excitotoxic and metabolic stress.

    CAS  PubMed  Google Scholar 

  70. Zhang, X. et al. PGC-1α/ERRα-Sirt3 pathway regulates DAergic neuronal death by directly deacetylating SOD2 and ATP synthase β. Antioxid. Redox Signal. 24, 312–328 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Johnson, S. C., Rabinovitch, P. S. & Kaeberlein, M. mTOR is a key modulator of ageing and age-related disease. Nature 493, 338–345 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Bockaert, J. & Marin, P. mTOR in brain physiology and pathologies. Physiol. Rev. 95, 1157–1187 (2015).

    CAS  PubMed  Google Scholar 

  73. Alirezaei, M. et al. Short-term fasting induces profound neuronal autophagy. Autophagy 6, 702–710 (2010).

    PubMed  PubMed Central  Google Scholar 

  74. He, C., Sumpter, R. Jr & Levine, B. Exercise induces autophagy in peripheral tissues and in the brain. Autophagy 8, 1548–1551 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Finn, P. F. & Dice, J. F. Ketone bodies stimulate chaperone-mediated autophagy. J. Biol. Chem. 280, 25864–25870 (2005).

    CAS  PubMed  Google Scholar 

  76. Saxton, R. A. & Sabatini, D. M. mTOR signaling in growth, metabolism, and disease. Cell 168, 960–976 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Nixon, R. A. The role of autophagy in neurodegenerative disease. Nat. Med. 19, 983–997 (2013).

    CAS  PubMed  Google Scholar 

  78. Harrison, D. E. et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460, 392–395 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Tang, S. J. et al. A rapamycin-sensitive signaling pathway contributes to long-term synaptic plasticity in the hippocampus. Proc. Natl Acad. Sci. USA 99, 467–472 (2002). This study demonstrates a role for the nutrient-sensing mTOR pathway in hippocampal synaptic plasticity.

    CAS  PubMed  Google Scholar 

  80. Cammalleri, M. et al. Time-restricted role for dendritic activation of the mTOR-p70S6K pathway in the induction of late-phase long-term potentiation in the CA1. Proc. Natl Acad. Sci. USA 100, 14368–14373 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Hoeffer, C. A. et al. Removal of FKBP12 enhances mTOR-Raptor interactions, LTP, memory, and perseverative/repetitive behavior. Neuron 60, 832–845 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Weston, M. C., Chen, H. & Swann, J. W. Multiple roles for mammalian target of rapamycin signaling in both glutamatergic and GABAergic synaptic transmission. J. Neurosci. 32, 11441–11452 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Shehata, M., Matsumura, H., Okubo-Suzuki, R., Ohkawa, N. & Inokuchi, K. Neuronal stimulation induces autophagy in hippocampal neurons that is involved in AMPA receptor degradation after chemical long-term depression. J. Neurosci. 32, 10413–10422 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Hernandez, D. et al. Regulation of presynaptic neurotransmission by macroautophagy. Neuron 74, 277–284 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Palacios, O. M. et al. Diet and exercise signals regulate SIRT3 and activate AMPK and PGC-1alpha in skeletal muscle. Aging 1, 771–783 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Mounier, R., Théret, M., Lantier, L., Foretz, M. & Viollet, B. Expanding roles for AMPK in skeletal muscle plasticity. Trends Endocrinol. Metab. 26, 275–286 (2015).

    CAS  PubMed  Google Scholar 

  87. Marosi, K. et al. Long-term exercise treatment reduces oxidative stress in the hippocampus of aging rats. Neuroscience 226, 21–28 (2012).

    CAS  PubMed  Google Scholar 

  88. Han, Y. et al. AMPK signaling in the dorsal hippocampus negatively regulates contextual fear memory formation. Neuropsychopharmacology 41, 1849–1864 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Kong, D. et al. A postsynaptic AMPK — p21-activated kinase pathway drives fasting-induced synaptic plasticity in AgRP neurons. Neuron 91, 25–33 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Kobilo, T. et al. AMPK agonist AICAR improves cognition and motor coordination in young and aged mice. Learn. Mem. 21, 119–126 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Ramamurthy, S., Chang, E., Cao, Y., Zhu, J. & Ronnett, G. V. AMPK activation regulates neuronal structure in developing hippocampal neurons. Neuroscience 259, 13–24 (2014).

    CAS  PubMed  Google Scholar 

  92. Ferrario, C. R. et al. Homeostasis meets motivation in the battle to control food intake. J. Neurosci. 36, 11469–11481 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Andrews, Z. B. The extra-hypothalamic actions of ghrelin on neuronal function. Trends Neurosci. 34, 31–40 (2011).

    CAS  PubMed  Google Scholar 

  94. Kim, Y. et al. Ghrelin is required for dietary restriction-induced enhancement of hippocampal neurogenesis: lessons from ghrelin knockout mice. Endocr. J. 62, 269–275 (2015).

    CAS  PubMed  Google Scholar 

  95. Jensen, M. et al. Anxiolytic-like effects of increased ghrelin receptor signaling in the amygdala. Int. J. Neuropsychopharmacol. 19, pyv123 (2016).

    PubMed  Google Scholar 

  96. Wallenius, V. et al. Interleukin-6-deficient mice develop mature-onset obesity. Nat. Med. 8, 75–79 (2002).

    CAS  PubMed  Google Scholar 

  97. Wueest, S. et al. Interleukin-6 contributes to early fasting-induced free fatty acid mobilization in mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 306, R861–R867 (2014).

    CAS  PubMed  Google Scholar 

  98. Baier, P. C., May, U., Scheller, J., Rose-John, S. & Schiffelholz, T. Impaired hippocampus-dependent and -independent learning in IL-6 deficient mice. Behav. Brain Res. 200, 192–196 (2009).

    CAS  PubMed  Google Scholar 

  99. Mattson, M. P., Murrain, M., Guthrie, P. B. & Kater, S. B. Fibroblast growth factor and glutamate: opposing roles in the generation and degeneration of hippocampal neuroarchitecture. J. Neurosci. 9, 3728–3740 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Mattson, M. P., Maudsley, S. & Martin, B. A neural signaling triumvirate that influences ageing and age-related disease: insulin/IGF-1, BDNF and serotonin. Ageing Res. Rev. 3, 445–464 (2004).

    CAS  PubMed  Google Scholar 

  101. Vendelbo, M. H. et al. Exercise and fasting activate growth hormone-dependent myocellular signal transducer and activator of transcription-5b phosphorylation and insulin-like growth factor-I messenger ribonucleic acid expression in humans. J. Clin. Endocrinol. Metab. 95, E64–68 (2010).

    PubMed  Google Scholar 

  102. Pedersen, B. K. & Febbraio, M. A. Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nat. Rev. Endocrinol. 8, 457–465 (2012).

    CAS  PubMed  Google Scholar 

  103. Carro, E., Nuñez, A., Busiguina, S. & Torres-Aleman, I. Circulating insulin-like growth factor I mediates effects of exercise on the brain. J. Neurosci. 20, 2926–2933 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Carro, E., Trejo, J. L., Busiguina, S. & Torres-Aleman, I. Circulating insulin-like growth factor I mediates the protective effects of physical exercise against brain insults of different etiology and anatomy. J. Neurosci. 21, 5678–5684 (2001). The authors of this study use an IGF1-blocking antibody to demonstrate a role for circulating IGF1 in the neuroprotective actions of exercise in mouse models of excitotoxic neuronal degeneration.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Moon, H. Y. et al. Running-induced systemic cathepsin B secretion is associated with memory function. Cell Metab. 24, 332–340 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Newman, J. C. & Verdin, E. Ketone bodies as signaling metabolites. Trends Endocrinol. Metab. 25, 42–52 (2014).

    CAS  PubMed  Google Scholar 

  107. Shimazu, T. et al. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 339, 211–214 (2013).

    CAS  PubMed  Google Scholar 

  108. Murray, A. J. et al. Novel ketone diet enhances physical and cognitive performance. FASEB J. 30, 4021–4032 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Webber, R. J. & Edmond, J. The in vivo utilization of acetoacetate, D-(–)-3-hydroxybutyrate, and glucose for lipid synthesis in brain in the 18-day-old rat. Evidence for an acetyl-CoA bypass for sterol synthesis. J. Biol. Chem. 254, 3912–3920 (1979).

    CAS  PubMed  Google Scholar 

  110. Chamorro, Á., Dirnagl, U., Urra, X. & Planas, A. M. Neuroprotection in acute stroke: targeting excitotoxicity, oxidative and nitrosative stress, and inflammation. Lancet Neurol. 15, 869–881 (2016).

    PubMed  Google Scholar 

  111. Greene, A. E., Todorova, M. T., McGowan, R. & Seyfried, T. N. Caloric restriction inhibits seizure susceptibility in epileptic EL mice by reducing blood glucose. Epilepsia 42, 1371–1378 (2001).

    CAS  PubMed  Google Scholar 

  112. Yu, Z. F. & Mattson, M. P. Dietary restriction and 2-deoxyglucose administration reduce focal ischemic brain damage and improve behavioral outcome: evidence for a preconditioning mechanism. J. Neurosci. Res. 57, 830–839 (1999).

    CAS  PubMed  Google Scholar 

  113. Roberge, M. C., Messier, C., Staines, W. A. & Plamondon, H. Food restriction induces long-lasting recovery of spatial memory deficits following global ischemia in delayed matching and non-matching-to-sample radial arm maze tasks. Neuroscience 156, 11–29 (2008).

    CAS  PubMed  Google Scholar 

  114. Jeong, M. A. et al. Intermittent fasting improves functional recovery after rat thoracic contusion spinal cord injury. J. Neurotrauma. 28, 479–492 (2011).

    PubMed  PubMed Central  Google Scholar 

  115. Plunet, W. T. et al. Dietary restriction started after spinal cord injury improves functional recovery. Exp. Neurol. 213, 28–35 (2008).

    PubMed  Google Scholar 

  116. Davis, L. M., Pauly, J. R., Readnower, R. D., Rho, J. M. & Sullivan, P. G. Fasting is neuroprotective following traumatic brain injury. J. Neurosci. Res. 86, 1812–1822 (2008). This study shows that when initiated after traumatic brain injury, fasting and ketone administration reduce neuronal loss in rats.

    CAS  PubMed  Google Scholar 

  117. Lee, C. K., Weindruch, R. & Prolla, T. A. Gene-expression profile of the ageing brain in mice. Nat. Genet. 25, 294–297 (2000).

    CAS  PubMed  Google Scholar 

  118. Xu, X. et al. Gene expression atlas of the mouse central nervous system: impact and interactions of age, energy intake and gender. Genome Biol. 8, R234 (2007).

    PubMed  PubMed Central  Google Scholar 

  119. Schafer, M. J., Dolgalev, I., Alldred, M. J., Heguy, A. & Ginsberg, S. D. Calorie restriction suppresses age-dependent hippocampal transcriptional signatures. PLoS ONE 10, e0133923 (2015).

    PubMed  PubMed Central  Google Scholar 

  120. Di Benedetto, S., Müller, L., Wenger, E., Düzel, S. & Pawelec, G. Contribution of neuroinflammation and immunity to brain aging and the mitigating effects of physical and cognitive interventions. Neurosci. Biobehav Rev. 75, 114–128 (2017).

    PubMed  Google Scholar 

  121. Kiprianova, I. et al. Enlarged infarct volume and loss of BDNF mRNA induction following brain ischemia in mice lacking FGF-2. Exp. Neurol. 189, 252–260 (2004).

    CAS  PubMed  Google Scholar 

  122. Bo, H. et al. Exercise-induced neuroprotection of hippocampus in APP/PS1 transgenic mice via upregulation of mitochondrial 8-oxoguanine DNA glycosylase. Oxid. Med. Cell Longev. 2014, 834502 (2014).

    PubMed  PubMed Central  Google Scholar 

  123. Yang, J. L., Lin, Y. T., Chuang, P. C., Bohr, V. A. & Mattson, M. P. BDNF and exercise enhance neuronal DNA repair by stimulating CREB-mediated production of apurinic/apyrimidinic endonuclease 1. Neuromolecular Med. 16, 161–174 (2014).

    CAS  PubMed  Google Scholar 

  124. Prins, M. L., Lee, S. M., Fujima, L. S. & Hovda, D. A. Increased cerebral uptake and oxidation of exogenous betaHB improves ATP following traumatic brain injury in adult rats. J. Neurochem. 90, 666–672 (2004).

    CAS  PubMed  Google Scholar 

  125. Rahman, M. et al. The β-hydroxybutyrate receptor HCA2 activates a neuroprotective subset of macrophages. Nat. Commun. 5, 3944 (2014).

    CAS  PubMed  Google Scholar 

  126. Yin, J., Han, P., Tang, Z., Liu, Q. & Shi, J. Sirtuin 3 mediates neuroprotection of ketones against ischemic stroke. J. Cereb. Blood Flow Metab. 35, 1783–1789 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Simeone, T. A., Simeone, K. A. & Rho, J. M. Ketone bodies as anti-seizure agents. Neurochem. Res. 42, 2011–2018 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Mattson, M. P. Pathways towards and away from Alzheimer's disease. Nature 430, 631–639 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Poewe, W. et al. Parkinson disease. Nat. Rev. Dis. Primers 3, 17013 (2017).

    PubMed  Google Scholar 

  130. Kontis, V. et al. Future life expectancy in 35 industrialised countries: projections with a Bayesian model ensemble. Lancet 389, 1323–1335 (2017).

    PubMed  PubMed Central  Google Scholar 

  131. Frisardi, V. et al. Metabolic-cognitive syndrome: a cross-talk between metabolic syndrome and Alzheimer's disease. Ageing Res. Rev. 9, 399–417 (2010).

    PubMed  Google Scholar 

  132. Lee, E. B. & Mattson, M. P. The neuropathology of obesity: insights from human disease. Acta Neuropathol. 127, 3–28 (2014).

    CAS  PubMed  Google Scholar 

  133. Baumgart, M. et al. Summary of the evidence on modifiable risk factors for cognitive decline and dementia: a population-based perspective. Alzheimers Dement. 11, 718–726 (2015).

    PubMed  Google Scholar 

  134. LaHue, S. C., Comella, C. L. & Tanner, C. M. The best medicine? The influence of physical activity and inactivity on Parkinson's disease. Mov Disord. 31, 1444–1454 (2016).

    PubMed  Google Scholar 

  135. Groot, C. et al. The effect of physical activity on cognitive function in patients with dementia: a meta-analysis of randomized control trials. Ageing Res. Rev. 25, 13–23 (2016).

    CAS  PubMed  Google Scholar 

  136. Uhrbrand, A., Stenager, E., Pedersen, M. S. & Dalgas, U. Parkinson's disease and intensive exercise therapy — a systematic review and meta-analysis of randomized controlled trials. J. Neurol. Sci. 353, 9–19 (2015).

    PubMed  Google Scholar 

  137. Wang, J. et al. Caloric restriction attenuates beta-amyloid neuropathology in a mouse model of Alzheimer's disease. FASEB J. 19, 659–661 (2005).

    PubMed  Google Scholar 

  138. Patel, N. V. et al. Caloric restriction attenuates a beta-deposition in Alzheimer transgenic models. Neurobiol. Aging. 26, 995–1000 (2005).

    CAS  PubMed  Google Scholar 

  139. Halagappa, V. K. et al. Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer's disease. Neurobiol. Dis. 26, 212–220 (2007).

    CAS  PubMed  Google Scholar 

  140. Ryan, S. M. & Kelly, Á. M. Exercise as a pro-cognitive, pro-neurogenic and anti inflammatory intervention in transgenic mouse models of Alzheimer's disease. Ageing Res. Rev. 27, 77–92 (2016).

    CAS  PubMed  Google Scholar 

  141. Qin, W. et al. Neuronal SIRT1 activation as a novel mechanism underlying the prevention of Alzheimer disease amyloid neuropathology by calorie restriction. J. Biol. Chem. 281, 21745–21754 (2006).

    CAS  PubMed  Google Scholar 

  142. Zilberter, M. et al. Dietary energy substrates reverse early neuronal hyperactivity in a mouse model of Alzheimer's disease. J. Neurochem. 125, 157–171 (2013).

    CAS  PubMed  Google Scholar 

  143. Kashiwaya, Y. 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. Neurobiol. Aging. 34, 1530–1539 (2013). This study shows that dietary supplementation with a ketone (BHB) ester ameliorates behavioural deficits and lessens amyloid and tau pathologies in a triple transgenic mouse model of AD.

    CAS  PubMed  Google Scholar 

  144. Castellano, C. A. et al. A 3-month aerobic training program improves brain energy metabolism in mild Alzheimer's disease: preliminary results from a neuroimaging study. J. Alzheimers Dis. 56, 1459–1468 (2017).

    CAS  PubMed  Google Scholar 

  145. Bentourkia, M. et al. PET study of 11C-acetoacetate kinetics in rat brain during dietary treatments affecting ketosis. Am. J. Physiol. Endocrinol. Metab. 296, E796–E801 (2009).

    CAS  PubMed  Google Scholar 

  146. Croteau, E. et al. A cross-sectional comparison of brain glucose and ketone metabolism in cognitively healthy older adults, mild cognitive impairment and early Alzheimer's disease. Exp. Gerontol. http://dx.doi.org/10.1016/j.exger.2017.07.004 (2017). Employing PET imaging with radiotracers for glucose and the ketone AcAc, the authors of this study provide evidence that although cerebral glucose utilization is impaired in patients with AD, their neurons remain capable of utilizing ketones.

  147. Duan, W. & Mattson, M. P. Dietary restriction and 2-deoxyglucose administration improve behavioral outcome and reduce degeneration of dopaminergic neurons in models of Parkinson's disease. J. Neurosci. Res. 57, 195–206 (1999).

    CAS  PubMed  Google Scholar 

  148. Maswood, N. et al. Caloric restriction increases neurotrophic factor levels and attenuates neurochemical and behavioral deficits in a primate model of Parkinson's disease. Proc. Natl Acad. Sci. USA 101, 18171–18176 (2004). This study demonstrates that daily CR lessens motor deficits and dopamine depletion and elevates striatal levels of glial-cell-line-derived neurotrophic factor in a nonhuman primate model of PD.

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Petzinger, G. M. et al. Effects of treadmill exercise on dopaminergic transmission in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned mouse model of basal ganglia injury. J. Neurosci. 27, 5291–5300 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Griffioen, K. J. et al. Dietary energy intake modifies brainstem autonomic dysfunction caused by mutant α-synuclein. Neurobiol. Aging. 34, 928–935 (2013).

    CAS  PubMed  Google Scholar 

  151. Gerecke, K. M., Jiao, Y., Pagala, V. & Smeyne, R. J. Exercise does not protect against MPTP-induced neurotoxicity in BDNF haploinsufficient mice. PLoS ONE 7, e43250 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Bayliss, J. A. et al. Ghrelin-AMPK signaling mediates the neuroprotective effects of calorie restriction in Parkinson's disease. J. Neurosci. 36, 3049–3063 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Tieu, K. et al. D-Beta-hydroxybutyrate rescues mitochondrial respiration and mitigates features of Parkinson disease. J. Clin. Invest. 112, 892–901 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Ferrari, A. J. Burden of depressive disorders by country, sex, age, and year: findings from the global burden of disease study 2010. PLoS Med. 10, e1001547 (2013).

    PubMed  PubMed Central  Google Scholar 

  155. Hryhorczuk, C., Sharma, S. & Fulton, S. E. Metabolic disturbances connecting obesity and depression. Front. Neurosci. 7, 177 (2013).

    PubMed  PubMed Central  Google Scholar 

  156. Hallgren, M. et al. Exercise, physical activity, and sedentary behavior in the treatment of depression: broadening the scientific perspectives and clinical opportunities. Front. Psychiatry 7, 36 (2016).

    PubMed  PubMed Central  Google Scholar 

  157. Johnson, J. B. et al. Alternate day calorie restriction improves clinical findings and reduces markers of oxidative stress and inflammation in overweight adults with moderate asthma. Free Radic. Biol. Med. 42, 665–674 (2007).

    CAS  PubMed  Google Scholar 

  158. Archer, T., Josefsson, T. & Lindwall, M. Effects of physical exercise on depressive symptoms and biomarkers in depression. CNS Neurol. Disord. Drug Targets. 13, 1640–1653 (2014).

    PubMed  Google Scholar 

  159. Patki, G. et al. Novel mechanistic insights into treadmill exercise based rescue of social defeat-induced anxiety-like behavior and memory impairment in rats. Physiol. Behav. 130, 135–144 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Adlard, P. A. & Cotman, C. W. Voluntary exercise protects against stress-induced decreases in brain-derived neurotrophic factor protein expression. Neuroscience 124, 985–992 (2004).

    CAS  PubMed  Google Scholar 

  161. Siuciak, J. A., Lewis, D. R., Wiegand, S. J. & Lindsay, R. M. Antidepressant-like effect of brain-derived neurotrophic factor (BDNF). Pharmacol. Biochem. Behav. 56, 131–137 (1997).

    CAS  PubMed  Google Scholar 

  162. Taliaz, D. et al. Resilience to chronic stress is mediated by hippocampal brain-derived neurotrophic factor. J. Neurosci. 31, 4475–4483 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Garcia, C., Chen, M. J., Garza, A. A., Cotman, C. W. & Russo-Neustadt, A. The influence of specific noradrenergic and serotonergic lesions on the expression of hippocampal brain-derived neurotrophic factortranscripts following voluntary physical activity. Neuroscience 119, 721–732 (2003).

    CAS  PubMed  Google Scholar 

  164. Aguiar, A. S. et al. Effects of exercise on mitochondrial function, neuroplasticity and anxio-depressive behavior of mice. Neuroscience 271, 56–63 (2014).

    CAS  PubMed  Google Scholar 

  165. Li, Y. et al. TrkB regulates hippocampal neurogenesis and governs sensitivity to antidepressive treatment. Neuron 59, 399–412 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Glasper, E. R., Llorens-Martin, M. V., Leuner, B., Gould, E. & Trejo, J. L. Blockade of insulin-like growth factor-I has complex effects on structural plasticity in the hippocampus. Hippocampus 20, 706–712 (2010).

    CAS  PubMed  Google Scholar 

  167. Duman, C. H. et al. Peripheral insulin-like growth factor-I produces antidepressant-like behavior and contributes to the effect of exercise. Behav. Brain Res. 198, 366–371 (2009).

    CAS  PubMed  Google Scholar 

  168. Zipfel, S., Giel, K. E., Bulik, C. M., Hay, P. & Schmidt, U. Anorexia nervosa: aetiology, assessment, and treatment. Lancet Psychiatry 2, 1099–1111 (2015).

    PubMed  Google Scholar 

  169. Devlin, B. & Scherer, S. W. Genetic architecture in autism spectrum disorder. Curr. Opin. Genet. Dev. 22, 229–237 (2012).

    CAS  PubMed  Google Scholar 

  170. Criado, K. K. et al. Overweight and obese status in children with autism spectrum disorder and disruptive behavior. Autism http://dx.doi.org/10.1177/1362361316683888 (2017).

  171. Han, J. C. et al. Association of brain-derived neurotrophic factor (BDNF) haploinsufficiency with lower adaptive behaviour and reduced cognitive functioning in WAGR/11p13 deletion syndrome. Cortex 49, 2700–2710 (2013).

    PubMed  Google Scholar 

  172. Huber, K. M., Klann, E., Costa-Mattioli, M. & Zukin, R. S. Dysregulation of mammalian target of rapamycin signaling in mouse models of autism. J. Neurosci. 35, 13836–13842 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Bremer, E., Crozier, M. & Lloyd, M. A systematic review of the behavioral outcomes following exercise interventions for children and youth with autism spectrum disorder. Autism 20, 899–915 (2016).

    PubMed  Google Scholar 

  174. Ruskin, D. N. et al. Ketogenic diet improves core symptoms of autism in BTBR mice. PLoS ONE 8, e65021 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. El-Rashidy, O. et al. Ketogenic diet versus gluten free casein free diet in autistic children: a case-control study. Metab. Brain Dis. 32, 1935–1941 (2017).

    CAS  PubMed  Google Scholar 

  176. Stranahan, A. M. Models and mechanisms for hippocampal dysfunction in obesity and diabetes. Neuroscience 309, 125–139 (2015).

    CAS  PubMed  Google Scholar 

  177. O'Brien, P. D., Hinder, L. M., Callaghan, B. C. & Feldman, E. L. Neurological consequences of obesity. Lancet Neurol. 16, 465–477 (2017).

    PubMed  PubMed Central  Google Scholar 

  178. Lindqvist, A. et al. High-fat diet impairs hippocampal neurogenesis in male rats. Eur. J. Neurol. 13, 1385–1388 (2006).

    CAS  PubMed  Google Scholar 

  179. Stranahan, A. M. et al. Diabetes impairs hippocampal function through glucocorticoid-mediated effects on new and mature neurons. Nat. Neurosci. 11, 309–317 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Stranahan, A. M. et al. Diet-induced insulin resistance impairs hippocampal synaptic plasticity and cognition in middle-aged rats. Hippocampus 18, 1085–1088 (2008).

    PubMed  PubMed Central  Google Scholar 

  181. MacInnis, M. J. & Gibala, M. J. Physiological adaptations to interval training and the role of exercise intensity. J. Physiol. 595, 2915–2930 (2017).

    CAS  PubMed  Google Scholar 

  182. Mattson, M. P., Longo, V. D. & Harvie, M. Impact of intermittent fasting on health and disease processes. Ageing Res. Rev. 39, 46–58 (2017).

    PubMed  Google Scholar 

  183. Ngandu, T. et al. A 2 year multidomain intervention of diet, exercise, cognitive training, and vascular risk monitoring versus control to prevent cognitive decline in at-risk elderly people (FINGER): a randomised controlled trial. Lancet 385, 2255–2263 (2015).

    PubMed  Google Scholar 

  184. Duffy, P. H., Feuers, R., Nakamura, K. D., Leakey, J. & Hart, J. Effect of chronic caloric restriction on the synchronization of various physiological measures in old female Fischer 344 rats. Chronobiol. Int. 7, 113–124 (1990).

    CAS  PubMed  Google Scholar 

  185. Duffy, P. H., Feuers, R. J. & Hart, R. W. Effect of chronic caloric restriction on the circadian regulation of physiological and behavioral variables in old male B6C3F1 mice. Chronobiol. Int. 7, 291–303 (1990).

    CAS  PubMed  Google Scholar 

  186. Acosta-Rodríguez, V. A., de Groot, M. H. M., Rijo-Ferreira, F., Green, C. B. & Takahashi, J. S. Mice under caloric restriction self-impose a temporal restriction of food intake as revealed by an automated feeder system. Cell Metab. 26, 267–277 (2017).

    PubMed  PubMed Central  Google Scholar 

  187. Schupp, M. et al. Metabolite and transcriptome analysis during fasting suggest a role for the p53-Ddit4 axis in major metabolic tissues. BMC Genomics 14, 758 (2013).

    PubMed  PubMed Central  Google Scholar 

  188. Askew, E. W., Dohm, G. L. & Huston, R. L. Fatty acid and ketone body metabolism in the rat: response to diet and exercise. J. Nutr. 105, 1422–1432 (1975).

    CAS  PubMed  Google Scholar 

  189. Raja, G., Bräu, L., Palmer, T. N. & Fournier, P. A. Repeated bouts of high-intensity exercise and muscle glycogen sparing in the rat. J. Exp. Biol. 206, 2159–2166 (2003).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the Intramural Research Program of the US National Institute on Ageing.

Author information

Authors and Affiliations

Authors

Contributions

M.P.M., K.M., N.G., M.S. and A.C. researched data for the article, made a substantial contribution to the discussion of content and contributed to the writing, review and editing of the manuscript before submission'

Corresponding author

Correspondence to Mark P. Mattson.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Glossary

β-Hydroxybutyrate

(BHB). A ketone, generated from fatty acids during fasting and extended exercise, that functions as a cellular energy source and as a signalling molecule that induces the expression of brain-derived neurotrophic factor.

Intermittent metabolic switching

(IMS). Repeating cycles of a metabolic challenge (fasting and/or exercise) sufficient to deplete liver glycogen stores and elevate circulating ketone levels, followed by a recovery period (eating, resting and sleeping).

Brain-derived neurotrophic factor

(BDNF). A protein produced and released from neurons in response to synaptic activity, exercise and fasting that acts to enhance synaptic plasticity and cellular stress resistance.

Mitochondrial biogenesis

The proliferation of mitochondria in neurons in response to metabolic challenges and neurotrophic factors to produce new mitochondria that promote synaptic plasticity and cellular stress resistance.

Deacetylase

An enzyme that removes an acetyl group from lysine residues of substrate proteins; the sirtuins SIRT1 and SIRT3 are deacetylases that play particularly important roles in adaptive responses of neurons to metabolic challenges.

Mechanistic target of rapamycin

(mTOR; also known as serine/threonine-protein kinase mTOR (MTOR) and mammalian target of rapamycin). A kinase that plays a pivotal role in stimulating cellular protein synthesis and suppressing autophagy when nutrients (glucose and amino acids) are plentiful.

Autophagy

A complex process by which cells recognize damaged dysfunctional proteins and organelles, engulf them in a membrane and target them for enzymatic degradation in lysosomes to generate recyclable undamaged components (for example, amino acids and lipids).

Ketogenesis

The process by which spillover of acetyl CoA results from β-oxidation of fatty acids in the liver during fasting and extended exercise.

Myokines

Proteins and peptides released from muscle cells during exercise that can enter the brain and affect neuroplasticity; examples include interleukin-6, cathepsin B and irisin.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mattson, M., Moehl, K., Ghena, N. et al. Intermittent metabolic switching, neuroplasticity and brain health. Nat Rev Neurosci 19, 81–94 (2018). https://doi.org/10.1038/nrn.2017.156

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn.2017.156

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing