You are on page 1of 5

Seizure 52 (2017) 15–19

Contents lists available at ScienceDirect

Seizure
journal homepage: www.elsevier.com/locate/yseiz

Review

The mechanisms mediating the antiepileptic effects of the ketogenic


diet, and potential opportunities for improvement with
metabolism-altering drugs
Neil A. Youngsona,b,* , Margaret J. Morrisa , J. William O. Ballardb
a
Department of Pharmacology, School of Medical Sciences, UNSW Sydney, NSW, 2052, Australia
b
School of Biotechnology and Biomolecular Sciences, UNSW Sydney, NSW, 2052, Australia

A R T I C L E I N F O A B S T R A C T

Article history:
Received 2 August 2017 The ketogenic diet (KD) is increasingly being used to treat patients with intractable epilepsy. Despite
Received in revised form 9 September 2017 decades of research, the reason for its success, when anticonvulsants have failed, is still not completely
Accepted 11 September 2017 understood. There are, however, many candidate mechanisms which can be grouped into those that alter
neuronal excitability at the synapse, and those that confer neuroprotection. The molecular underpinning
Keywords: of these mechanisms centres on the shift from glucose- to lipid-based energy generation that
Ketogenic diet accompanies a high fat/low carbohydrate diet. Here we describe how changes in the relative abundances
Ketogenesis of energy substrates (ketone bodies), intermediates of glycolysis and fat metabolism, and metabolic end
Liver energy metabolism
products (ATP, reactive oxygen species) underlie many of the antiepileptic effects of the KD. We propose
that the success of the KD for treating epilepsy lies in the large variety of antiepileptic mechanisms that it
confers. Different subsets of the mechanisms may be clinically relevant in different patients. We extend
this to suggest that the broad benefits of the KD could therefore be achieved by pharmacologically
promoting the production of ketone bodies in the liver as they represent a key mediator that is common
to all of the proposed mechanisms.
© 2017 British Epilepsy Association. Published by Elsevier Ltd. All rights reserved.

1. Introduction the turn of the century, particularly for the 30% of patients with
intractable epilepsy. New variants of the KD have been developed
The use of the ketogenic diet (KD) as a therapy for epilepsy with slightly increased carbohydrate or protein content and/or
developed from observations that fasting could reduce seizures, a medium chain triglycerides rather than the longer chain trigly-
strategy that was in use as far back as ancient Greece [1]. Fasting cerides of the traditional KD. Nonetheless all the variants are
continued as a treatment up to the 1920s, but work around that essentially high fat/low carbohydrate diets that induce the
time also revealed that both starvation and high fat/low carbohy- production of ketone bodies (KBs) in the liver through fatty acid
drate diets increase plasma levels of the ketone bodies acetoace- catabolism. The KBs then enter the bloodstream and are taken up
tate and b-hydroxybutyrate [2]. It was also proposed that these by organs including the brain where they are further metabolised
metabolic products may benefit children with neurological in mitochondria to generate energy. Currently the KD is commonly
disorders [3]. In 1928 a study by Barborka [4] confirmed the used in children who have not improved after trialling two to three
effectiveness of the KD for treating epilepsy, as seizures in adults anti-epileptic drugs, although recent studies have confirmed its
receiving the diet were completely controlled or improved in 56 effectiveness in adults [5,6]. Patients typically undergo a three
out of 100 patients. From then on the KD was a common epilepsy month trial to see if the KD is effective, and if so they can remain on
treatment until it was gradually superseded by anticonvulsant it for several years until they are seizure-free, significantly
drugs, so that by the 1980s it was rarely used. However, there has improved or stop using it due to the difficulties in complying
been a resurgence of interest and usage of the KD for epilepsy since with such a restrictive regime. As for a reason why a high fat diet is
beneficial for a neuronal disorder, one possible explanation is that
KBs are a major fuel source for brain development in utero and in
infancy [1,7]. Perhaps the similarities between development and
* Corresponding author at: School of Biotechnology and Biomolecular Sciences, repair mean that the reinstatement of KB-based brain metabolism
UNSW Sydney, NSW, 2052, Australia
E-mail address: n.youngson@unsw.edu.au (N.A. Youngson).

http://dx.doi.org/10.1016/j.seizure.2017.09.005
1059-1311/© 2017 British Epilepsy Association. Published by Elsevier Ltd. All rights reserved.
16 N.A. Youngson et al. / Seizure 52 (2017) 15–19

creates an environment in which brain cells can repair the The antiepileptic effects of the KD are thought to be due to
structural damage associated with epileptogenesis. changes in the levels of the molecules or enzymes involved in
Despite the success of the KD in reducing seizures, and also the energy metabolism. It should be noted that it isn’t just the end-
considerable amount of research into the cellular and molecular products glucose- versus fat-based energy metabolism that are
changes that the diet induces, no single mechanism has been candidates for antiepileptic effects, as proposed mechanisms also
identified that explains its efficacy. The question, “How does the involve the KBs themselves, and the intermediates of their
ketogenic diet reduce seizures” is an unusually challenging one. catabolism. The links between several of these molecules and
Finding the answer is complicated by both the extreme variability neuronal excitability and/or neuroprotection form the basis of our
of the causes of epilepsy as well as the extensive changes in understanding of the antiepileptic mechanism of the KD and are
physiology induced by the diet. The shift in energy generation from described in the next section (Fig. 1).
carbohydrate sources to lipid essentially rewires many fundamen-
tal biological systems, and as with all therapies that alter 3. Summary of mechanisms that may explain the anti-seizure
metabolism, both the molecular end products and intermediates effects of the ketogenic diet
in a pathway can be potentially therapeutic. Furthermore, research
that goes deeper into the molecular changes induced by the KD, The cellular state associated with seizures is increased neuronal
rather than supporting or refuting existing hypotheses, often ends excitability which involves an increase in action potentials which
up providing yet more potentially seizure-reducing mechanisms are in turn induced by depolarization of the cell membrane at a
[1,8]. Finally, translation of discoveries in animal models into synapse. The polarity at a synapse is regulated by inhibitory and
clinical studies is complicated by the huge inter-individual excitatory neurotransmitters which control ion pumps and
variability in the underlying causes of epilepsy. This means that channels that facilitate the influx and efflux of ions such as Na2
+
rejection of a proposed mechanism would require its examination , K+, Cl and Ca2+. Accordingly potential anti-seizure mechanisms
in multiple groups that represent the various forms of epilepsy of the KD are linked to changes in the amounts of neuro-
(potentially at multiple ages, and/or severities), which is beyond transmitters as well changes that influence neural membrane
the scope of most research projects. polarity. The major excitatory neurotransmitter glutamate can be
However, despite these challenges, several KD-induced changes synthesised from the TCA cycle intermediate a-ketoglutarate.
have been identified that are beneficial to neuronal structure and/ Glutamate can then either be converted to the major inhibitory
or function. They can be broadly grouped into changes that alter neurotransmitter GABA, or be transaminated to aspartate in a
neurotransmission at synapses, and those that improve neuronal reaction that requires another TCA cycle compound, oxaloacetate.
structure and homeostasis, though there are many overlapping As oxaloacetate is required for energy generation, there is a relative
molecules and processes. Recent reviews have described these in increase in the amount of glutamate that is converted to GABA [14].
more detail [1,8,9]. Some human and rat studies support this anti-seizure mechanism
by showing that a KD leads to increased GABA and reduced
2. The ketogenic diet changes energy metabolism in the liver glutamate in the brain [15,16]. Another potential player is the
and the brain inhibitory neurotransmitter neuropeptide Y (NPY) which would be
increased by ketogenesis or starvation [17], and is known to be
In a carbohydrate rich diet, glucose is the preferred substrate for antiepileptic [18].
energy production in the brain. Glucose is initially catabolised in The increase in ATP generation by the KD provides fertile
the cytoplasm of a neuron or glial cell through the process of ground for theories to explain the anti-seizure effects as a
glycolysis which produces ATP and NADH. The latter can be used in multitude of cellular processes require ATP. The ATP requirement
the mitochondrial electron transport chain (ETC) to create the of several types of ion channels and transporters is a leading
proton gradient across the inner mitochondrial membrane which candidate. Perhaps the best example of the influence of ATP as an
is used in oxidative phosphorylation to produce more ATP. The end anti-seizure mechanism is the export of ATP from neurons which is
product of glycolysis is pyruvate which enters the mitochondria then converted to adenosine in the synapse which activates
and is converted to acetyl-CoA. This central molecule of energy adenosine A1 receptors (A1R) leading to activation of potassium
metabolism can then combine with oxaloacetate to enter the channels thereby leading to hyperpolarization of the cell
tricarboxylic acid (TCA) cycle and produce more NADH that can be membrane and a reduction in excitability (Fig. 1) [19].
used in the ETC. ATP and energy metabolism are intimately connected to
All of the variants of the KD instigate shifts in body-wide another process through which the KD is antiepileptic neuro-
energy metabolism away from glucose-based and towards fat- protection. Neuroprotective mechanisms are proposed to both
based energy generation. The ingested fatty acids are metabolised increase the seizure threshold and to reduce the damage to the
in liver mitochondria into KBs which are then released into the brain that is generated by seizures. As mentioned above the KD is
blood stream and are taken up by multiple organs including the known to increase the number of mitochondria in brain cells
brain. In the mitochondria of neurons and glial cells KBs are [20,21]. This is expected to be due to the requirement for
catabolised to acetyl-CoA, which can then enter the TCA cycle for mitochondria for energy generation from fats as well as the
energy generation (producing NADH and ultimately ATP), or it can reduction in glycolysis (which doesn’t require mitochondria). At
be used in lipogenesis to produce fatty acids. A consequence of the onset of the KD the existing mitochondria will experience a
the increased dependence on mitochondria for energy generation high workload which reduces the efficiency of ATP production
with the KD is that the numbers of that organelle increase in leading to the production of reactive oxygen species (ROS).
neurons and glia. This is one proposed contributor to the Increased ROS levels in seizures are actually a contributor to the
observation that ketones and the KD lead to high levels of ATP cell damage and death, so the KD-induced increase contradicts the
production [10,11]. observed benefits of the KD. However this state of increased
With the KD the reduction in the use of glucose as an energy oxidative stress is temporary as the increased ROS induces
source is compounded as not only is there simply less dietary mitochondrial biogenesis to deal with the increased workload,
carbohydrate, but glycolysis is additionally inhibited by shifts in and also induces anti-ROS mechanisms such as glutathione. The
the mitochondrial ratios of acetyl-CoA/CoA and NADH/NAD+ end result is neurons and glia that have an improved cellular
[12,13]. infrastructure for energy generation and reducing oxidative stress
N.A. Youngson et al. / Seizure 52 (2017) 15–19 17

Fig. 1. Multiple proposed anti-epileptogenic effects of the ketogenic diet involve the metabolism of ketone bodies in the mitochondria. A. products of mitochondrial
metabolism of ketone bodies in neurons (or in astrocytes with subsequent transfer of the products to neurons) that are proposed to confer anti-epileptic effects. B. The effects
of certain products from A on ion transfer at the synapse. A1R, adenosine A1 receptor; ACA, acetoacetate; b-HB, beta hydroxybutyrate; PUFA, polyunsaturated fatty acid; ROS,
reactive oxygen species.

[20]. The former change increases the ‘energy reserve’ of neurons, also directly modulate different types of ion pumps and channels
improving neuronal homeostasis before seizures and also reduces thereby reducing neuronal hyperexcitability [23].
the impacts of the high energy drain during seizures. The latter is There is evidence that the decrease in glycolysis in patients on
proposed to improve the resilience of brain cells to the damagingly the KD contributes to the improved seizure control and reduced
high levels of ROS that are generated in a seizure [22]. epileptogenesis. Indeed, there is a rapid resumption of seizures
Increases in the biosynthesis of certain fat species in patients on when patients on a KD ingest carbohydrates or glucose [24]. In
the KD provide other neuroprotective mechanisms. Hypomyeli- animal models treatment with inhibitors of glycolysis can have
nation is a feature of some epilepsies. The KD increases brain levels rapid anticonvulsant effects [25,26]. The effects may be linked to
of acetyl-CoA (from KBs) and aspartate (from TCA-cycle inter- reductions in lactate (which is produced by glycolysis) which can
mediates) which both contribute to the synthesis of myelin [8]. alter neuronal membrane polarity through ATP-dependent potas-
Another product of fat metabolism with multiple anti-seizure sium channels [27,28].
effects is poly-unsaturated fatty acids (PUFAs). These fat species Research has also been undertaken to determine whether
are increased in the brain of patients on the KD and they are ketone bodies themselves have direct antiepileptic actions in the
thought to protect neurons through simulating mitochondrial brain. b-hydroxybutyrate can slow spontaneous neuronal firing by
uncoupling proteins which reduce ROS production, and PUFAs can opening ATP-dependent potassium channels [29]. Furthermore,

Table 1
Potential drugs to promote ketogenesis in liver.

Effect on liver metabolism Candidate drugs (those with evidence of having antiepileptic properties in bold) Reference
Activate hepatic b-oxidation Sirtuin activators e.g. Resveratrol, NMN Milne et al. [36]
PARP inhibitors Uddin et al. [37]
Thiazolidinediones Gariani et al. [38]
PPARa activators (synthetic ligands) Grabacka et al. [41]
Inhibit hepatic lipogenesis Acetyl-CoA Carboxylase inhibitors Corbett et al. [39]
Griffith et al. [47]
Inhibit hepatic glycolysis (Randle cycle manipulation) Lactate dehydrogenase inhibitor stiripentol Sada et al. [28]
Glucagon McGarry et al. [48]
Glycolysis inhibitors developed for cancer Pelicano et al. [40]
PPARa activators(synthetic ligands) Grabacka et al. [41]
Inhibit mitochondrial pyruvate carrier (MPC) Thiazolidinediones e.g. Rosiglitazone and Pioglitazone Simeone et al. [43]
UK5099 Mishra et al. [44]
Vacanti et al. [49]
Hildyard et al. [50]
Inhibit liver mitochondrial complex I to favour fat metabolism Metformin Yang et al. [46]
18 N.A. Youngson et al. / Seizure 52 (2017) 15–19

recent evidence has identified that KBs also function as extracel- regulatory pathways that activate energy storage often also repress
lular signalling ligands [30], that can activate stress-response and those of energy release, and vice versa [12]. Therefore pharmaco-
anti-inflammatory pathways [31]. Within the cell, KBs and logical strategies that either inhibit energy storage, or promote
intermediates of their catabolism can also have strong effects on energy release (often by mimicking starvation signals) in the liver
the epigenetic state of genes which facilitates the changes in will promote ketogenesis.
cellular metabolism and resistance to ROS [32,33]. Table 1 contains several drugs that in theory could promote
liver ketogenesis. Due to the multifunctional activities of metabo-
4. Future possibilities for the diet – pharmacological lism regulatory signals there is some overlap between the
enhancement mechanisms, but they can be separated into four main groups.
Ketogenesis could be enhanced by increasing the total acetyl-CoA
As mentioned in the introduction there are difficulties in through activating b-oxidation [36–38,41]. Similarly, the propor-
compliance with the KD. This is due to the restrictive and less tion of acetyl-CoA that is available for ketogenesis can be increased
palatable nature of the diet and side-effects such as gastrointesti- by repression of other processes that utilise it (e.g. lipogenesis)
nal problems. Consequently, one of the aims of research into anti- [39,47]. Several of the drugs in these groups have been developed
seizure mechanisms of KD has been to identify if it is possible to for tackling obesity and type 2 diabetes as they can increase the
achieve the same benefits with a drug or supplement. These efforts oxidation of fat stores in the liver and other organs [36–38] and/or
have included drugs that increase levels of GABA, PUFAs and prevent lipid storage [39]. The third group either inhibits glycolysis
antioxidants [34] or reducing glycolysis in the brain [28]. However, [28,40,41,50] or the transfer of the end product of glycolysis
none have yet demonstrated such consistent antiepileptic effects (pyruvate) into the mitochondria (MPC1 inhibition) [43,44,49,50].
as the KD. The challenge of replicating the benefits of the KD with a Inhibition of glycolysis pushes the hepatocyte away from glucose-
drug may be linked to the difficulty of identifying one main and towards fat-metabolism in a reciprocal process called the
mechanism that underpins its effectiveness. Perhaps a clue to this Randle cycle [12]. Several candidate drugs in this category for
puzzle lies in the fact that KD has been shown to improve seizures increasing ketogenesis have been developed as cancer therapies as
in patients with a variety of forms of epilepsy. It is possible that tumour cells often favour glycolysis for energy generation [40,41].
there is a different subset of the seizure-reducing mechanisms that The fourth group involves liver-specific inhibition of mitochondrial
are beneficial in each patient. Therefore one solution to the complex I (the main entry-point for electrons that are sourced from
challenge of developing a drug that mimics the KD may be, instead glucose metabolism) which can push mitochondrial metabolism to
of pharmacologically manipulating the terminal processes (e.g. favour fat metabolism (which has a reduced requirement for
neurotransmitter actions), and thereby limiting the treatment to complex I) and thereby increase acetyl-CoA levels [42,46]. This
altering a subset of the anti-seizure mechanisms, why not focus on inhibition may need to be liver-specific as complex I inhibition or
manipulating the upstream steps of the KD which are common to dysfunction in the brain is associated with increased seizure risk.
all of the mechanisms? This strategy would theoretically maintain Interestingly, many of the drugs in Table 1 have also been found to
the multi-mechanism aspect of the KD which may allow for the have anticonvulsant properties [28,43–46].
utility of the drug across a broad range of epilepsies. Therefore In summary we propose that stimulation of ketogenesis in the
drugs that promote ketogenesis in the liver may be candidates for liver could be a useful alternative or adjunct to the KD in
future research (Table 1/Fig. 2). management of epilepsy. Application of these drugs in combina-
The liver is an extremely important metabolic organ. Depend- tion with the KD, or a less restrictive KD (i.e. that allows increased
ing on the hormonal signals it receives and its existing energy dietary carbohydrates) could improve the compliance to the diet,
stores (glycogen or lipids) it will switch between multiple or possibly be used to perpetuate ketogenesis at a clinically-
processes of energy storage (gluconeogenesis, glycogenesis, effective level after the patient ends the diet.
lipogenesis) and mobilisation of energy supplies for itself and
the rest of the body (glycolysis, glycogenolysis, b-oxidation,
Conflicts of interest
ketogenesis). Ketogenesis is primarily a response to starvation
when carbohydrate stores are depleted and the lipid stores in the
None.
liver or adipose tissue are mobilised, converted to KBs in the liver
and released into the blood [35]. The signalling- and gene-
Funding sources

This research was funded by an Australian Research Council


(ARC) Project Grant to Prof. Ballard (DP160102575). Work in the
Morris lab is funded by the ARC and Australian National Health and
Medical Research Council (NHMRC).

References

[1] Koppel SJ, Swerdlow RH. Neuroketotherapeutics: a modern review of a


century-old therapy. Neurochem Int 2017, doi:http://dx.doi.org/10.1016/j.
neuint.2017.05.019.
[2] Woodyatt RT. Objects and method of diet adjustment in diabetes. Arch Intern
Med 1921;28:125–41.
[3] Wilder RM. The effect of ketonemia on the course of epilepsy. Mayo Clin Bull
19212:.
[4] Barborka CJ. Ketogenic diet treatment of epilepsy in adults. J Am Med Assoc
1928;91:73–8.
[5] Kverneland M, Selmer KK, Nakken KO, Iversen PO, Tauboll E. A prospective
study of the modified Atkins diet for adults with idiopathic generalized
epilepsy. Epilepsy Behav 2015;53:197–201, doi:http://dx.doi.org/10.1016/j.
yebeh.2015.10.021.
Fig. 2. Promotion of liver ketogenesis as a pharmacological replacement for the [6] Neal EG, Chaffe H, Schwartz RH, Lawson MS, Edwards N, Fitzsimmons G, et al.
ketogenic diet. ACA, acetoacetate; b-HB, beta hydroxybutyrate. The ketogenic diet for the treatment of childhood epilepsy: a randomised
N.A. Youngson et al. / Seizure 52 (2017) 15–19 19

controlled trial. Lancet Neurol 2008;7:500–6, doi:http://dx.doi.org/10.1016/ neurons. J Neurosci 2011;31:8689–96, doi:http://dx.doi.org/10.1523/JNEUR-
S1474-4422(08)70092-9. OSCI.5951-10.2011.
[7] Cotter DG, d'Avignon DA, Wentz AE, Weber ML, Crawford PA. Obligate role for [30] Newman JC, Verdin E. Ketone bodies as signaling metabolites. Trends
ketone body oxidation in neonatal metabolic homeostasis. J Biol Chem Endocrinol Metab 2014;25:42–52, doi:http://dx.doi.org/10.1016/j.
2011;286:6902–10, doi:http://dx.doi.org/10.1074/jbc.M110.192369. tem.2013.09.002.
[8] Clanton RM, Wu G, Akabani G, Aramayo R. Control of seizures by ketogenic [31] Selfridge JE, EL, Carl SM, Funk E, Fields T, et al. Effect of one month duration
diet-induced modulation of metabolic pathways. Amino Acids 2017;49:1–20, ketogenic and non-ketogenic high fat diets on mouse brain bioenergetic
doi:http://dx.doi.org/10.1007/s00726-016-2336-7. infrastructure. J Bioenerg Biomembr 2015;47:1–11, doi:http://dx.doi.org/
[9] Rho JM. How does the ketogenic diet induce anti-seizure effects? Neurosci Lett 10.1007/s10863-014-9570-z.
2017;637:4–10, doi:http://dx.doi.org/10.1016/j.neulet.2015.07.034. [32] Xie Z, Zhang D, Chung D, Tang Z, Huang H, Dai L, et al. Metabolic regulation of
[10] DeVivo DC, Leckie MP, Ferrendelli JS, McDougal Jr. DB. Chronic ketosis and gene expression by histone lysine beta-hydroxybutyrylation. Mol Cell
cerebral metabolism. Ann Neurol 1978;3:331–7, doi:http://dx.doi.org/ 2016;62:194–206, doi:http://dx.doi.org/10.1016/j.molcel.2016.03.036.
10.1002/ana.410030410. [33] Achanta LB, Rae C. D. beta-Hydroxybutyrate in the brain: one molecule,
[11] Kim DY, Vallejo J, Rho JM. Ketones prevent synaptic dysfunction induced by multiple mechanisms. Neurochem Res 2017;42:35–49, doi:http://dx.doi.org/
mitochondrial respiratory complex inhibitors. J Neurochem 2010;114:130–41, 10.1007/s11064-016-2099-2.
doi:http://dx.doi.org/10.1111/j.1471-4159.2010.06728.x. [34] Rho JM, Sankar R. The ketogenic diet in a pill: is this possible? Epilepsia
[12] Hue L, Taegtmeyer H. The Randle cycle revisited: a new head for an old hat. Am 2008;49(Suppl. 8):127–33, doi:http://dx.doi.org/10.1111/j.1528-
J Physiol Endocrinol Metab 2009;297:E578–591, doi:http://dx.doi.org/10.1152/ 1167.2008.01857.x.
ajpendo.00093.2009. [35] Cotter DG, Schugar RC, Crawford PA. Ketone body metabolism and
[13] LaManna JC, Salem N, Puchowicz M, Erokwu B, Koppaka S, Flask C, et al. cardiovascular disease. Am J Physiol Heart Circ Physiol 2013;304:H1060–76,
Ketones suppress brain glucose consumption. Adv Exp Med Biol doi:http://dx.doi.org/10.1152/ajpheart.00646.2012.
2009;645:301–6, doi:http://dx.doi.org/10.1007/978-0-387-85998-9_45. [36] Milne JC, Lambert PD, Schenk S, Carney DP, Smith JJ, Gagne DJ, et al. Small
[14] Yudkoff M, Daikhin Y, Nissim I, Horyn O, Lazarow A, Luhovyy B, et al. Response molecule activators of SIRT1 as therapeutics for the treatment of type 2
of brain amino acid metabolism to ketosis. Neurochem Int 2005;47:119–28, diabetes. Nature 2007;450:712–6, doi:http://dx.doi.org/10.1038/nature06261.
doi:http://dx.doi.org/10.1016/j.neuint.2005.04.014. [37] Uddin GM, Youngson NA, Sinclair DA, Morris MJ. Head to head comparison of
[15] Wang ZJ, Bergqvist C, Hunter JV, Jin D, Wang DJ, Wehrli S, et al. In vivo short-term treatment with the NAD(+) precursor nicotinamide mononucleo-
measurement of brain metabolites using two-dimensional double-quantum tide (NMN) and 6 weeks of exercise in obese female mice. Front Pharmacol
MR spectroscopy–exploration of GABA levels in a ketogenic diet. Magn Reson 2016;7:258, doi:http://dx.doi.org/10.3389/fphar.2016.00258.
Med 2003;49:615–9, doi:http://dx.doi.org/10.1002/mrm.10429. [38] Gariani K, Ryu D, Menzies KJ, Yi HS, Stein S, Zhang H, et al. Inhibiting poly ADP-
[16] Melo TM, Nehlig A, Sonnewald U. Neuronal-glial interactions in rats fed a ribosylation increases fatty acid oxidation and protects against fatty liver
ketogenic diet. Neurochem Int 2006;48:498–507, doi:http://dx.doi.org/ disease. J Hepatol 2017;66:132–41, doi:http://dx.doi.org/10.1016/j.
10.1016/j.neuint.2005.12.037. jhep.2016.08.024.
[17] Mainardi P, Albano C. Is the antiepileptic effect of the ketogenic diet due to [39] Corbett JW. Review of recent acetyl-CoA carboxylase inhibitor patents: mid-
ketones? Med Hypotheses 2008;70:536–9, doi:http://dx.doi.org/10.1016/j. 2007-2008. Expert Opin Ther Pat 2009;19:943–56, doi:http://dx.doi.org/
mehy.2007.05.054. 10.1517/13543770902862180.
[18] Morris MJ, Gannan E, Stroud LM, Beck-Sickinger AG, O'Brien TJ. Neuropeptide Y [40] Pelicano H, Martin DS, Xu RH, Huang P. Glycolysis inhibition for anticancer
suppresses absence seizures in a genetic rat model primarily through effects treatment. Oncogene 2006;25:4633–46, doi:http://dx.doi.org/10.1038/sj.
on Y receptors. Eur J Neurosci 2007;25:1136–43, doi:http://dx.doi.org/10.1111/ onc.1209597.
j.1460-9568.2007.05348.x. [41] Grabacka M, Pierzchalska M, Reiss K. Peroxisome proliferator activated
[19] Masino SA, Kawamura Jr. M, Ruskin DN. Adenosine receptors and epilepsy: receptor alpha ligands as anticancer drugs targeting mitochondrial metabo-
current evidence and future potential. Int Rev Neurobiol 2014;119:233–55, lism. Curr Pharm Biotechnol 2013;14:342–56.
doi:http://dx.doi.org/10.1016/B978-0-12-801022-8.00011-8. [42] Ballard JWO, Youngson NA. Review: can diet influence the selective advantage
[20] Milder JB, Liang LP, Patel M. Acute oxidative stress and systemic Nrf2 activation of mitochondrial DNA haplotypes? Bioscience Rep 201535:e00277, doi:http://
by the ketogenic diet. Neurobiol Dis 2010;40:238–44, doi:http://dx.doi.org/ dx.doi.org/10.1042/BSR20150232.
10.1016/j.nbd.2010.05.030. [43] Simeone TA, Matthews SA, Samson KK, Simeone KA. Regulation of brain
[21] Bough KJ, Wetherington J, Hassel B, Pare JF, Gawryluk JW, Greene JG, et al. PPARgamma2 contributes to ketogenic diet anti-seizure efficacy. Exp Neurol
Mitochondrial biogenesis in the anticonvulsant mechanism of the ketogenic 2017;287:54–64, doi:http://dx.doi.org/10.1016/j.expneurol.2016.08.006.
diet. Ann Neurol 2006;60:223–35, doi:http://dx.doi.org/10.1002/ana.20899. [44] Mishra J, Chaudhary T, Kumar A. Rosiglitazone synergizes the neuroprotective
[22] Shin EJ, Jeong JH, Chung YH, Kim WK, Ko KH, Bach JH, et al. Role of oxidative effects of valproic acid against quinolinic acid-induced neurotoxicity in rats:
stress in epileptic seizures. Neurochem Int 2011;59:122–37, doi:http://dx.doi. targeting PPARgamma and HDAC pathways. Neurotox Res 2014;26:130–51,
org/10.1016/j.neuint.2011.03.025. doi:http://dx.doi.org/10.1007/s12640-014-9458-z.
[23] Bough KJ, Rho JM. Anticonvulsant mechanisms of the ketogenic diet. Epilepsia [45] Li Z, You ZY, Li M, Pang L, Cheng J, Wang LC. Protective effect of resveratrol on
2007;48:43–58, doi:http://dx.doi.org/10.1111/j.1528-1167.2007.00915.x. the brain in a rat model of epilepsy. Neurosci Bull 2017;33:273–80, doi:http://
[24] Huttenlocher PR. Ketonemia and seizures: metabolic and anticonvulsant dx.doi.org/10.1007/s12264-017-0097-2.
effects of two ketogenic diets in childhood epilepsy. Pediatr Res 1976;10:536– [46] Yang Y, Zhu B, Zheng F, Li Y, Zhang Y, Hu Y, et al. Chronic metformin treatment
40, doi:http://dx.doi.org/10.1203/00006450-197605000-00006. facilitates seizure termination. Biochem Biophys Res Commun 2017;484:450–
[25] Stafstrom CE, Ockuly JC, Murphree L, Valley MT, Roopra A, Sutula TP. 5, doi:http://dx.doi.org/10.1016/j.bbrc.2017.01.157.
Anticonvulsant and antiepileptic actions of 2-deoxy-D-glucose in epilepsy [47] Griffith DA, Kung DW, Esler WP, Amor PA, Bagley SW, Beysen C, et al.
models. Ann Neurol 2009;65:435–47, doi:http://dx.doi.org/10.1002/ Decreasing the rate of metabolic ketone reduction in the discovery of a clinical
ana.21603. acetyl-CoA carboxylase inhibitor for the treatment of diabetes. J Med Chem
[26] Lian XY, Khan FA, Stringer JL. Fructose-1,6-bisphosphate has anticonvulsant 2014;57:10512–26, doi:http://dx.doi.org/10.1021/jm5016022.
activity in models of acute seizures in adult rats. J Neurosci 2007;27:12007–11, [48] McGarry J, Wright PH, Foster DW. Hormonal control of ketogenesis. Rapid
doi:http://dx.doi.org/10.1523/JNEUROSCI.3163-07.2007. activation of hepatic ketogenic capacity in fed rats by anti-insulin serum and
[27] Parsons MP, Hirasawa M. ATP-sensitive potassium channel-mediated lactate glucagon. J Clin Invest 1975;55:1202–9, doi:http://dx.doi.org/10.1172/
effect on orexin neurons: implications for brain energetics during arousal. J JCI108038.
Neurosci 2010;30:8061–70, doi:http://dx.doi.org/10.1523/JNEUROSCI.5741- [49] Vacanti NM, Divakaruni AS, Green CR, Parker SJ, Henry RR, Ciaraldi TP, Murphy
09.2010. AN, Metallo CM. Regulation of substrate utilization by the mitochondrial
[28] Sada N, Lee S, Katsu T, Otsuki T, Inoue T. Epilepsy treatment. Targeting LDH pyruvate carrier. Mol Cell 2014;56:425–35, doi:http://dx.doi.org/10.1016/j.
enzymes with a stiripentol analog to treat epilepsy. Science 2015;347:1362–7, molcel.2014.09.024.
doi:http://dx.doi.org/10.1126/science.aaa1299. [50] Hildyard JC, Ammala C, Dukes ID, Thomson SA, Halestrap AP. Identification and
[29] Tanner GR, Lutas A, Martinez-Francois JR, Yellen G. Single K ATP channel characterisation of a new class of highly specific and potent inhibitors of the
opening in response to action potential firing in mouse dentate granule mitochondrial pyruvate carrier. Biochim Biophys Acta 2005;1707:221–30, doi:
http://dx.doi.org/10.1016/j.bbabio.2004.12.005.

You might also like