Sie sind auf Seite 1von 16

Epilepsia, 48(1):43–58, 2007

Blackwell Publishing, Inc.



C 2007 International League Against Epilepsy

Anticonvulsant Mechanisms of the Ketogenic Diet

∗ Kristopher J. Bough and †Jong M. Rho

∗ Center for Drug Evaluation & Research, Food and Drug Administration, Rockville, Maryland;
†Barrow Neurological Institute, Phoenix, Arizona, U.S.A.

Summary: The ketogenic diet (KD) is a broadly effective treat- polyunsaturated fatty acids increased after KD induce the ex-
ment for medically refractory epilepsy. Despite nearly a century pression of neuronal uncoupling proteins (UCPs), a collective
of use, the mechanisms underlying its clinical efficacy remain up-regulation of numerous energy metabolism genes, and mito-
unknown. In this review, we present one intersecting view of how chondrial biogenesis. These effects further limit ROS generation
the KD may exert its anticonvulsant activity against the backdrop and increase energy production. As a result of limited glucose
of several seemingly disparate mechanistic theories. We summa- and enhanced oxidative phosphorylation, reduced glycolytic flux
rize key insights gleaned from experimental and clinical studies is hypothesized to activate metabolic KATP channels and hyper-
of the KD, and focus particular attention on the role that ketone polarize neurons and/or glia. Although it is unlikely that a single
bodies, fatty acids, and limited glucose may play in seizure con- mechanism, however well substantiated, will explain all of the
trol. Chronic ketosis is anticipated to modify the tricarboxcylic diet’s clinical benefits, these diverse, coordinated changes seem
acid cycle to increase GABA synthesis in brain, limit reactive poised to stabilize synaptic function and increase the resistance
oxygen species (ROS) generation, and boost energy production to seizures throughout the brain. Key Words: Ketogenic diet—
in brain tissue. Among several direct neuro-inhibitory actions, Epilepsy—Metabolism—Polyunsaturated fatty acids.

The ketogenic diet (KD) is a high-fat, low-protein, low- lying mechanisms of action. Although some studies sug-
carbohydrate diet that has been employed as a treatment gest that dietary constituents or metabolites have direct
for medically refractory epilepsy for 86 years. The “clas- anticonvulsant effects, emerging evidence indicates that
sic” KD is based upon consumption of long-chain satu- adaptations to chronic administration of the KD result in
rated triglycerides (LCTs) in a 3:1–4:1 ketogenic diet ratio improved seizure control. These data suggest that the KD
(KD ratio) of fats to carbohydrates + protein (by weight). activates several endogenous metabolic and genetic “pro-
The vast majority of calories (>90%) are derived from fat. grams” to stabilize and/or enhance cellular metabolism,
While clinical implementation of the KD has varied from and that these fundamental changes help counter neuronal
center to center (Kossoff and McGrogan, 2005), diet treat- dysfunction associated with seizure activity.
ment generally begins with a period of fasting followed by
gradual increase in calories to a target KD ratio of 3:1–4:1.
This is conducted in the inpatient setting over the course MECHANISTIC INSIGHTS FROM STUDIES
of several days, where blood glucose, urine ketones, and OF KD EFFICACY
several other metabolic variables are closely monitored. The anticonvulsant efficacy of the KD has been ex-
The hallmark feature of KD treatment is the production of amined in various acute and chronic animal models of
ketone bodies by the liver. Ketone bodies provide an al- epilepsy over the years (Stafstrom, 1999). Clinical and
ternative substrate to glucose for energy utilization, and in experimental studies have provided key insights into im-
developing brain, also constitute essential building blocks portant treatment-related variables and, when considered
for biosynthesis of cell membranes and lipids. together, these studies have helped direct mechanistic re-
While the clinical effectiveness of the KD is widely search. Commonalities between clinical and experimen-
accepted, surprisingly little is understood about its under- tal studies of efficacy are summarized in Table 1 (adapted
from Stafstrom, 2004).
Accepted October 24, 2006. Based on vast clinical experience, almost any diet that
Address correspondence and reprint requests to Kristopher J. Bough produces ketonemia and/or diminished blood glucose lev-
at FDA – Center for Drug Evaluation & Research, MPN 1 – Room els can induce an anticonvulsant effect. Ketogenic diets
1345, 7520 Standish Place, Rockville, MD 20855, U.S.A. E-mail:
Kristopher.Bough@fda.hhs.gov comprised of either LCTs (Freeman et al., 1998; Vin-
doi:10.1111/j.1528-1167.2007.00915.x ing et al., 1998) or medium-chain triglycerides (MCTs;

43
44 K. J. BOUGH AND J. M. RHO

TABLE 1. Translational correlations of ketogenic diet (KD) efficacy


Variable Experimental findings Clinical findings References

Age Young rats and mice <P40 at diet onset Infants, children and adolescents <19 Millichap (1964), Uhlemann and
Adult rats and mice >P40 at diet years of age Neims (1972), Appleton and DeVivo
onset Some evidence of efficacy in adults (1974), Nakazawa et al. (1983),
Kinsman et al. (1992), Hori et al.
(1997), Bough et al. (1999b),
Muller-Schwarze et al. (1999),
Sirven et al. (1999), Nordli et al.
(2001), Coppola et al. (2002),
Kossoff et al. (2002), Mady et al.
(2003)
Seizure type Effective in a wide variety of acute and Equally efficacious in a variety of Livingston (1972), Appleton and
chronic seizure models seizure types DeVivo (1974), Mahoney et al.
However, protection however can be (1983), Otani et al. (1984), Schwartz
transient; maximal seizure activity et al. (1989a), Hori et al. (1997),
can be prolonged Freeman et al. (1998), Vining et al.
(1998), Rho et al. (1999), Su et al.
(2000), Thavendiranathan et al.
(2000), Greene et al. (2001), Bough
et al. (2002)
Dietary composition Qualitative differences do not affect Qualitative differences do not affect Huttenlocher et al. (1971), Sills et al.
outcome outcome (1986), Schwartz et al. (1989b),
Classic LCT = MCT = PUFA fat diet Classic LCT, MCT, Atkins can all Freeman et al. (1998), Vining et al.
diminish seizure frequency (1998), Mak et al. (1999), Likhodii
et al. (2000), Dell et al. (2001),
Kossoff et al. (2003), Henderson
et al. (2006), Kossoff et al. (2006)
KD ratio Increase KD ratio = greater KD effect Increase KD ratio = greater KD effect Dekaban (1966), Livingston (1972),
Bough et al. (1999a), Bough et al.
(2000b), Freeman et al. (2000)
Calorie restriction CR is anticonvulsant CR is an integral part of KD regimen Bough et al.(2000a), Bough et al.
CR enhances KD anticonvulsant effect (CR associated with seizure control) (2000b), Freeman et al. (2000),
NAL < NCR < KAL < KCR Greene et al. (2001), Greene et al.
(2003)
Growth rate Initial transient decline in body weight Decline in weight/height over first few Rho et al. (1999), Freeman et al.
Followed by, resumption of normal or months (2000), Su et al. (2000), Vining et al.
near-normal growth rate Followed by resumption of normal or (2002), Peterson et al. (2005), Thio
near-normal height/weight gain et al. (2006)
Very young children do grow poorly
Latency to KD effect 1–2 weeks Seizures can be reduced within 1–2 Livingston (1972), Appleton and
days DeVivo (1974), Freeman and Vining
Fine-tuning is key to success of the (1999), Rho et al. (1999), Freeman
diet; at least 1–2 weeks are required et al. (2000), Bough et al. (2006)
to see if changes are maximally
effective
Reversal of KD effect Seizure are behaviorally more severe Breakthrough seizure activity can Appleton and DeVivo (1974),
within hours, but threshold returns to occur immediately with ingestion of Huttenlocher (1976), Freeman et al.
baseline over the course of 1–2 carbohydrates (2000), Mady et al. (2003), Bough
weeks et al. (2006)
Gender Males = females Males = females Millichap (1964), Nakazawa et al.
(1983), Schwartz et al. (1989b),
Freeman et al. (1998), Bough et al.
(2002), Mady et al. (2003)

Abbreviations: P40 age, postnatal day 40; LCT, long-chain triglycerides; MCT, medium-chain triglycerides; PUFA, polyunsaturated fatty acid; Atkins
diet, diet high in fats + protein, low in carbohydrates; CR, calorie-restriction; NAL, normal, ad libitum rodent diet; NCR, normal, calorie-restricted
(by ∼15%) rodent diet; KAL, ketogenic, ad libitum diet; KCR, ketogenic, calorie-restricted diet; KD ratio, ratio of [fats / (carbohydrates + proteins)];
KAP, ketogenic/antiketogenic potential. Table adapted from Stafstrom (2004).

Huttenlocher et al., 1971; Sills et al., 1986; Schwartz seizure control (Likhodii et al., 2000; Dell et al., 2001).
et al., 1989b; Mak et al., 1999) can control seizures with Thus, available evidence indicates that dietary composi-
similar efficacy. Even the high-fat, high-protein, and low- tion per se does not appear to affect the anticonvulsant
carbohydrate Atkins diet that produced a ketotic state, re- efficacy of the diet, as long as there is a degree of sus-
duced seizures in epileptic patients (Kossoff et al., 2003, tained ketosis.
2006). Similar effects have been observed experimentally. By comparison, KD ratios and calorie-restriction (CR)
Ketogenic diets containing myriad types of fats (i.e., with appear to be important variables in enabling seizure pro-
low carbohydrate content) all produced similar levels of tection. Seizure control is reportedly optimized when KDs

Epilepsia, Vol. 48, No. 1, 2007


ANTICONVULSANT ACTIONS OF KETOGENIC DIET 45

are administered in ratios of ≥3:1 (Freeman et al., 2000), Muller-Schwarze et al., 1999). CR, too, is equally anti-
and higher KD ratios increased both clinical (Dekaban, convulsant in both juvenile and adult mice (Greene et al.,
1966; Livingston, 1972) and experimental anticonvulsant 2001). Thus, increasing evidence suggests the anticonvul-
efficacy (Bough et al., 2000b). Similar to the KD ratio, sant effects of KDs do not appear to be age-dependent.
increasing the extent of CR resulted in improved seizure Clinical reports indicate that outcome is unrelated to
control in epileptic mice (Greene et al., 2001), irrespec- seizure type or frequency (Freeman et al., 1998; Schwartz
tive of the type of diet that was restricted (Eagles et al., et al., 1989a; Vining et al., 1998). At least 50% of pa-
2003). In general, extra calories in the form of carbohy- tients treated with a classical KD will exhibit at least a
drates or proteins translate to additional metabolic sub- 50% reduction in seizures (Livingston, 1972; Freeman
strates for gluconeogenesis and diminished KD efficacy. et al., 1998). Comparatively, anticonvulsant effects of KD
Breakthrough seizures are believed to result from overes- in animals are more modest. Rats and mice only demon-
timation and administration of excess calories (Freeman strate a 15–20% increase in seizure threshold (Appleton
et al., 2000). As such, CR may share common anticonvul- and DeVivo, 1974; Bough and Eagles, 1999; Rho et al.,
sant mechanisms and adjunctively optimize KD efficacy. 1999; Bough et al., 2000b). Despite efficacy across a vari-
In rodents, maximal seizure control develops 1–2 weeks ety of acute and chronic seizure models (Hori et al., 1997;
after initiation of a KD (Appleton and DeVivo, 1974; Rho Muller-Schwarze et al., 1999; Su et al., 2000), KD-induced
et al., 1999; Bough et al., 2006). Similarly in humans, anticonvulsant effects have been incomplete (Bough et al.,
clinical efficacy does not reach its zenith in many patients 2002) or of limited duration (Hori et al., 1997). Further,
until after 2 weeks (Dekaban, 1966; Freeman et al., 2000). there is little evidence to indicate that KDs diminish sever-
One notable feature of the KD is the rapid occurrence of ity once a seizure begins. Indeed, many studies—including
breakthrough seizures and loss of ketosis when carbohy- our own—have shown that CR and/or KDs can even ex-
drates are introduced (e.g., after a child sneaks a cookie; acerbate (maximal) seizures (Mahoney et al., 1983; Otani
Huttenlocher, 1976). As a result, the KD must be strictly et al., 1984; Bough et al., 2000a; Thavendiranathan et al.,
enforced in order for efficacy to be maintained. However, a 2000; Bough et al., 2003). If one considers that seizure
breakthrough seizure may not necessarily translate to a to- activity requires large amounts of energy, seizure exacer-
tal loss of seizure control. Studies have shown that, despite bation may be a reflection of enhanced energy reserves
an abrupt discontinuation of the KD, the increased resis- after KD treatment, a situation that would allow for pro-
tance to seizures waned gradually when switched back to longed ictal activity once it begins (discussed below).
control (Bough et al., 2006) or even high-carbohydrate, In summary, the following generalizations can be made
antiketogenic chow (Appleton and DeVivo, 1974). This about the KD: (1) its anticonvulsant effects appear inde-
decline in seizure threshold generally occurred over 1–2 pendent of dietary formulation, but appear to be strongly
weeks, mirroring the onset of seizure protection (Apple- linked to the total quantity of calories consumed; (2) the
ton and DeVivo, 1974; Bough et al., 2006). This indicates KD must be strictly adhered to, if the anticonvulsant effect
that a critical, minimal level of sustained ketosis is neces- is to be maintained; (3) CR may work synergistically with
sary but not sufficient to maintain seizure control. Thus, KD to limit seizures and optimize treatment; (4) maximum
it would seem that metabolic adaptations to KDs underlie efficacy is not achieved for several days or weeks after ini-
their key anticonvulsant actions. tiation, suggesting that adaptive metabolic and/or genetic
Many studies and anecdotal observations have sug- “programs” underlie KD-induced seizure protection; (5)
gested that the KD is most effective in immature animals these adaptations are likely generalized throughout the
or infants and children (Livingston, 1972; Uhlemann and (epileptic) brain, irrespective of underlying pathology or
Neims, 1972; Otani et al., 1984; Bough et al., 1999b; Rho genetic predisposition to seizures since the KD is an ef-
et al., 1999). This is perhaps due to enhanced metabolic ca- fective treatment for diverse epileptic conditions; and (6)
pacity, more efficient extraction of ketone bodies from the efficacy is independent of gender and age, suggesting that
blood (Morris, 2005), and/or greater compliance of KDs KD treatment produces seizure control via a common set
in the pediatric population. However, a lack of efficacy of pathways in all clinical responders.
in older children or adults may simply reflect noncompli-
ance or dietary intolerance rather than an inadequate re-
sponse physiological (Livingston, 1972). The KD has been
ANTICONVULSANT MECHANISMS
demonstrated to be similarly effective in infants (Nordli
OF THE KETOGENIC DIET
et al., 2001; Kossoff et al., 2002), adolescents (Kinsman
et al., 1992; Mady et al., 2003), and adults (Sirven et al., Since the KD was originated over 85 years ago, several
1999; Coppola et al., 2002). Furthermore, experimental major hypotheses have been advanced, but none have been
KDs are effective in both young (i.e., <P40 days; Uhle- widely accepted. Several key aspects of the KD might ul-
mann and Neims, 1972; Otani et al., 1984; Bough et al., timately result in seizure protection. Ketone bodies, free
1999b) and adult rodents (Appleton and DeVivo, 1974; fatty acids (in particular, polyunsaturated fatty acids), or

Epilepsia, Vol. 48, No. 1, 2007


46 K. J. BOUGH AND J. M. RHO

glucose restriction might each lead directly or indirectly and Keith, 1930; Yamashita et al., 1976; Vodickova et al.,
to seizure control. While it is possible that any one of 1995). Clinically, acetone levels of up to 1 millimolar
these KD-induced changes is responsible for the anticon- (mM) were detected in the brains of five of seven well-
vulsant action of the KD, available evidence suggests that controlled epileptic patients following KD using mag-
improved seizure control, at a minimum, likely requires netic resonance spectroscopic techniques (Seymour et al.,
all three. 1999). Although acetone could not be detected in two other
seizure-free patients, the authors concluded that acetone
Role of ketone bodies contributes to the anticonvulsant effect of the KD. Inter-
Beta-hydroxybutyrate (BHB) is the predominant ketone estingly, the concept that a lipophilic solvent may potently
body measured in the blood, and as such, has been used as block seizure activity is not new. The classic example of
a clinical measure of KD implementation (Fig. 1). Accord- this is valproic acid, which was initially used as a solvent
ingly, nearly all KD studies have attempted to establish a to dissolve investigational anticonvulsant compounds, but
causative link between ketonemia and anticonvulsant ef- was serendipitously discovered to possess intrinsic anti-
ficacy. Although robust elevations in plasma BHB levels convulsant properties.
have been observed during KD treatment (Bough et al., Whereas in vivo pharmacodynamic studies have
1999b; Thavendiranathan et al., 2000), there is no signif- suggested that both ACA and acetone may act as
icant correlation between plasma BHB levels and seizure anticonvulsant agents, there is no evidence that ketone
protection. Optimal seizure protection generally lags days bodies can directly modulate synaptic transmission and/or
to weeks behind the development of ketonemia, which oc- neuronal excitability. In vitro cellular electrophysiologi-
curs within hours of KD onset. cal experiments have failed to demonstrate an effect on the
Nevertheless, there is some evidence that ketones other principal ion channels that mediate neuronal excitability
than BHB may possess anticonvulsant properties. When and inhibition. Specifically, neither L-BHB nor ACA were
injected into animals, acetone and its parent acetoacetate found to modulate GABAA receptors, AMPA receptors,
(ACA), prevent acutely provoked seizures. Seminal work or NMDA receptors in both hippocampus and neocortex
in the 1930s revealed that acute intraperitoneal admin- of rats (Thio et al., 2000; Donevan et al., 2003). Despite
istration of acetone or ethyl-acetoacetate protected rab- these negative observations, it remained possible that ke-
bits from thujone-induced seizures (Helmholz and Keith, tone bodies might affect network activity or synchrony.
1930; Keith, 1933). Thujone is the active constituent of However, in field potential recordings conducted in vitro,
wormword oil, and is an antagonist of GABAA receptors Thio et al. (2000) demonstrated clearly that neither ACA
(Hold et al., 2000). More recent experimental studies have nor BHB modified evoked excitatory postsynaptic poten-
shown similar results in rodents. Acetone (Likhodii et al., tials (EPSPs) or population spikes in the CA1 subfield of
2003) and ACA (Rho et al., 2002)—but not BHB—were the hippocampal tissue. In summary, there is no evidence
anticonvulsant in a variety of acute and chronic models of for direct anticonvulsant effects for either ACA or BHB,
epilepsy, consistent with earlier observations (Helmholz and acetone has yet to be studied in neuronal (CNS) tissue.

FIG. 1. Metabolic pathways highlighting the production of ketone bodies fatty acids during fasting or treatment with the ketogenic diet (KD).
Estimated fasting- or KD-induced concentrations of beta-hydroxybutyrate, acetoacetate, and acetone in blood are listed (large boxes).
Measures of beta-hydroxybutyrate levels in blood are most commonly used as the clinical indicator of successful KD treatment. From
Likhodii and Burnham (2004).

Epilepsia, Vol. 48, No. 1, 2007


ANTICONVULSANT ACTIONS OF KETOGENIC DIET 47

This may, in large part, reflect the technical difficulties in Others have hypothesized that glucose restriction dur-
investigating a compound that is highly volatile and can ing KD treatment activates ATP-sensitive potassium
react with perfusion systems ordinarily used in pharma- (KATP ) channels (Schwartzkroin, 1999; Vamecq et al.,
cological in vitro experiments. 2005). Interestingly, KATP channels are ligand-gated re-
Recently, it has been suggested that ACA and/or its ceptors broadly expressed throughout the central nervous
metabolic byproduct, acetone, may activate a novel class system, in both neurons and glia (Thomzig et al., 2005).
of potassium leak channels known as the two-pore do- These channels act as metabolic sensors, linking cellular
main or K2P channels (Vamecq et al., 2005). K2P channels membrane excitability to fluctuating levels of ADP and
represent a diverse superfamily of channels that generally ATP. Activation of this receptor by reduced ATP/ADP ra-
hyperpolarize cell membranes, and regulate membrane ex- tios opens the channel and leads to membrane hyperpo-
citability both pre- and postsynaptically (Lesage, 2003). larization. When glucose is limited (e.g., during adminis-
These channels can be modulated by changes in pH, os- tration of a classic KD, which is typically CR by 25%),
molality, temperature, mechanical pressure, and certain KATP channels might open to hyperpolarize the cell as the
fatty acids (Franks and Honore, 2004). Links between KD- intracellular ATP concentrations fall. Conversely, when
induced elevations in ketone bodies (and/or fatty acids, as glucose is present and ATP concentrations rise, KATP chan-
discussed below) and K2P channels, however, have yet to nels close. As such, KATP channels may provide a mea-
be explored. sure of protection against a variety of metabolic stressors
In conclusion, although ketone bodies have been shown such as hypoxia, ischemia, and hypoglycemia, and are
to possess anticonvulsant properties in vivo, there is no ev- believed to regulate seizure threshold (Seino and Miki,
idence to date that they mediate directly these effects. It 2003).
is clear that some degree of sustained ketosis is required KATP channels are particularly abundant in the substan-
for clinical efficacy and that efficacy is maximized over a tia nigra (Hicks et al., 1994), a region of the brain thought
period of weeks versus days, despite a rapid onset of keto- to act centrally in the propagation of seizure activity
sis within hours. Whereas it is plausible that some dietary, (Iadarola and Gale, 1982). KATP channels would therefore
pharmacokinetic factor(s) results in some level of seizure be ideally positioned to metabolically regulate the onset
protection, the approximate 2-week time course for opti- of several different seizure types, as does the KD. There is
mal seizure protection suggests a pharmacodynamic effect growing evidence that KATP channels may critically reg-
of the KD (e.g., parallel time course for changes in gene ulate seizure activity. Genetically engineered mice that
expression, mitochondrial proliferation, up-regulation of overexpress the sulfonylurea (SUR) subunit of KATP chan-
UCPs/transporters, etc) likely underlies the anticonvulsant nels were significantly more resistant to seizures induced
nature of the diet. Thus, available data suggest that adap- by kainate, and showed no marked cell loss in hippocam-
tations to, rather than a direct effect of, ketosis underlie pus (Hernandez-Sanchez et al., 2001). Studies of KATP
the anticonvulsant nature of the KD. channel (Kir6.2−/− ) knockout mice suggested that these
channels help determine seizure threshold (Yamada et al.,
2001). Following hypoxic challenge (∼5% O2 ), knock-
Role of glucose restriction out mice exhibited myoclonic–tonic seizure activity, and,
Whereas most studies have suggested that persistent ultimately, death compared to controls who all recovered
ketosis is essential to KD-induced seizure protection, oth- without sequelae.
ers have posited that glucose restriction is the key fea- Despite these observations, there is one important
ture (Greene et al., 2003). In addition to ketosis, it is caveat in implicating KATP channels as mediators of a KD-
clear that as ketonemia develops, another immediate con- induced anticonvulsant effect. Other studies have demon-
sequence of CR and/or KDs is a ‘moderate’ reduction strated an increase in energy reserves (specifically, ATP)
in blood glucose. Does caloric restriction simply act to after KD treatment (DeVivo et al., 1978; Pan et al., 1999).
limit gluconeogenic substrates that would otherwise re- These data predict that KATP channels would remain
duce KD ratio and counter efficacy? Or, might glucose re- closed, not open, during diet treatment, and would thus
striction result in another metabolic adaptation that helps contribute to neuronal/glial cell membrane depolarization.
quell aberrant hyperexcitability? Calorie restriction alone Nevertheless, several findings are consistent with the no-
was sufficient to retard seizure susceptibility in juvenile tion that KATP channels are selectively activated during
and adult epileptic EL mice; and, blood glucose lev- administration of a low-glucose, high-fat KD. First, KATP
els were inversely correlated with a decreased risk of channels are regulated preferentially via glycolytic energy
seizures (Greene et al., 2001). Greene et al. (2003) hypoth- sources (Dubinsky et al., 1998). It has recently been shown
esized that CR reduces energy production through glycol- that the glycolytic enzyme glyceraldehyde 3-phosphate
ysis, which limits a neuron’s ability to reach (and main- dehydrogenase (GAPDH) serves as an accessory pro-
tain) high levels of synaptic activity necessary for seizure tein to KATP channels and regulates directly their activity
genesis. (Dhar-Chowdhury et al., 2005; Jovanovic et al., 2005).

Epilepsia, Vol. 48, No. 1, 2007


48 K. J. BOUGH AND J. M. RHO

The observed reduction in glycolytic processes af- C20:4ω6), or eicosapentanoic acid (EPA, C20:5ω3) are
ter KD treatment (specifically, the concentration of believed to affect profoundly cardiovascular function and
fructose-1,6-bisphosphate, the key regulatory enzyme health (Leaf and Kang, 1996; Nordoy, 1999; Leaf et al.,
of glycolysis) is consistent with this notion (DeVivo 2003). In cardiac myocytes, PUFAs inhibited fast, voltage-
et al., 1978; Puchowicz et al., 2005; Melo et al., 2006). gated sodium channels (Xiao et al., 1998) and L-type cal-
Glycolytic flux may be further limited as a consequence cium channels (Xiao et al., 1997). Similar findings have
of elevated ATP (DeVivo et al., 1978; Otani et al., 1984; been observed in neuronal tissue. For example, DHA and
Pan et al., 1999; Bough et al., 2006) and citrate (Yudkoff EPA diminished neuronal excitability and bursting in hip-
et al., 2001) levels on KD treatment; both ATP and citrate pocampus (Xiao and Li, 1999).
are feedback inhibitors of glycolysis. It is not surprising then that PUFAs are becoming an in-
Second, it is hypothesized that the accumulation of free creasingly popular focus of KD research. After KD treat-
fatty acids over the course of KD administration (Dekaban, ment, specific PUFAs (i.e., AA and DHA) were found to
1966) may boost KATP channel activation (Vamecq et al., be elevated in both serum (Cunnane et al., 2002; Fraser
2005). Whereas PUFAs freely cross the BBB, saturated et al., 2003) and brain (Taha et al., 2005) of patients and
free fatty acids are transported across the BBB via carrier- animals. Importantly, one report documented that the rise
mediated processes (Avellini et al., 1994). Fatty acids (or drop) in total fatty acids during KD treatment closely
that intercalate within neuronal cell membranes have been paralleled clinical improvement (or loss) of seizure control
shown to interact potently with KATP channels, specifically (Dekaban, 1966). An additional study found that dietary
reducing their affinity for (and inhibition by) ATP (Shyng supplementation with 5 g of (65%) n-3 PUFAs once per
and Nichols, 1998). Overall, these findings suggest that day produced a marked reduction in seizure frequency
the unique nature of low-glucose, high-fat KDs promotes and intensity in a few epileptic patients (Schlanger et al.,
KATP channel activation, despite observed enhancements 2002). These findings suggest that KD-induced elevations
in oxidative energy production. in PUFAs such as DHA and/or AA might act directly to
Recent experiments involving 2-deoxyglucose (2-DG) limit neuronal excitability and dampen seizure activity.
provide further support for a glucose-restriction hypothe- PUFAs could ultimately block seizure activity in a num-
sis of KD action. Two-deoxyglucose is a glucose analogue, ber of ways (Fig. 2). First, PUFAs may inhibit directly ion
which inhibits phosphoglucose isomerase and, hence, gly- channel activity. Omega-3 (ω-3) PUFAs have been shown
colysis. Stafstrom et al. (2005) reported that the addi- to: (1) inhibit both voltage-gated Na+ and Ca2+ channels,
tion of 1 mM 2-DG decreased epileptiform burst fre- (2) increase the resistance to bursting induced by bicu-
quency to 25–80% of baseline in rat hippocampal slices culline, zero Mg2+ , pentylenetetrazole or glutamate, and
exposed to elevated extracellular potassium. More signifi- (3) prolong the recovery time from inactivation in hip-
cantly, the same group also showed that 2-DG (250 mg/kg, pocampal neurons (Vreugdenhil et al., 1996; Xiao and Li,
i.p) elevated the after-discharge threshold in olfactory 1999; Young et al., 2000). Second, in conjunction with ke-
bulb of perforant-path kindled rats, markedly reduced the tone bodies, PUFAs may activate a lipid-sensitive class of
progression of kindling, and limited the expression of K2P potassium channels (Vamecq et al., 2005). And, third,
BDNF and its cognate receptor, trkB (Garriga-Canut et al., PUFAs may enhance the activity of the Na+ /K+ -ATPase
2006). (sodium pump). Elevated ω-3 and diminished ω-6 PU-
Interestingly, there are a number of anticonvulsant par- FAs levels in plasma membranes significantly increased
allels between 2-DG (Stafstrom et al., 2005; Garriga- sodium pump function (Wu et al., 2004). These findings
Canut et al., 2006) and KD treatment (Bough et al., 2003). indicate that elevations in brain levels of PUFAs after KD
First, both 2-DG and KD elevated electrographic seizure treatment (Taha et al., 2005) could help reduce neuronal
threshold in vivo; second, both 2-DG and KD potently re- hyperexcitability via a variety of direct mechanisms.
tarded the progression of epileptogenesis in kindling mod-
els of epilepsy in vivo; and, third, both 2-DG (in vitro)
and KD (in vivo) diminished measures of hippocampal Uncoupling proteins
hyperexcitability. These results collectively suggest that In addition to their direct actions on neuronal ex-
the anticonvulsant actions of KD may work, in large part, citability, PUFAs may also act indirectly to limit ex-
via an inhibition of glycolysis. Importantly, because 2-DG citotoxicity and neurodegeneration. PUFAs regulate the
is fairly well tolerated when administered orally (Pelicano expression of numerous genes in brain via transcription
et al., 2006), this compound may represent a novel treat- factors such as PPARα (peroxisome proliferator-activated
ment strategy for epilepsy. receptor-α; Sampath and Ntambi, 2004). Through in-
duction of PPARα and its coactivator PGC-1, PUFAs
Role of fatty acids induce the expression of mitochondrial uncoupling pro-
Polyunsaturated fatty acids (PUFAs) such as docosa- teins (UCPs) and activate these proteins directly as well
hexanoic acid (DHA, C22:6ω3), arachidonic acid (AA, (Jaburek et al., 1999; Diano et al., 2003). Recent evidence

Epilepsia, Vol. 48, No. 1, 2007


ANTICONVULSANT ACTIONS OF KETOGENIC DIET 49

FIG. 2. Potential pathways through which polyunsaturated fatty acids (PUFAs) may limit hyperexcitability in the brain. Acting directly,
PUFAs such as arachidonic acid (AA), docosahexanoic acid (DHA), and/or eicosapentanoic acid (EPA) might inhibit both voltage-gated
Na+ and Ca2+ channels, activate a lipid-sensitive class of K2P potassium channels, and enhance the activity of the Na+ /K+ -ATPase to
limit neuronal excitability and dampen seizure activity. Acting indirectly, PUFAs might induce the expression and activity of uncoupling
proteins (UCPs) to diminish reactive oxygen species (ROS), reduce neuronal dysfunction and induce a neuroprotective effect. Finally,
PUFAs are expected to activate PPARα and induce a coordinate up-regulation of energy transcripts leading to enhanced energy reserves,
stabilized synaptic function and limited hyperexcitability.

suggests that PUFAs are required for mitochondrial UCP et al., 2003). The protective role of UCPs was recently
activity (Garlid et al., 2001). highlighted by Sullivan et al. (2003) who demonstrated
Uncoupling proteins are homodimers that span the in- that dietary enhancement of UCP expression and function
ner mitochondrial membrane and allow a proton leak from in immature rats protected against kainate-induced exci-
the intermembrane space to the mitochondrial matrix. totoxicity, most likely by decreasing ROS generation (An-
There are three major isoforms that have been identified drews et al., 2005). Further work demonstrated that mice
in the brain, UCP2, UCP4 and UCP5 (a.k.a., BMCP-1 maintained on a high-fat KD demonstrated an increase in
or brain mitochondrial carrier protein-1). UCP proteins the hippocampal expression and activity of all three mi-
are increasingly implicated in the regulation of neuronal tochondrial UCPs and exhibited a significant reduction in
excitability and survival (Andrews et al., 2005). The un- ROS generation in mitochondria isolated from the same
coupling effect, albeit of small magnitude, reduces the brain region (Sullivan et al., 2004). In conjunction with
proton-motive force, disassociates or ‘uncouples’ elec- reports that ketone bodies potently decrease ROS gen-
tron transport from ATP production, and indirectly de- eration (Veech et al., 2001; Veech, 2004), these reports
creases the production of reactive oxygen species (ROS). suggest that the KD compensates for seizure-induced el-
Although it would seem that increased levels of UCP pro- evations in ROS generation and neuronal dysfunction to
teins would diminish cellular energy production, Diano provide a neuroprotective effect.
et al. (2003) showed that chronic overexpression of UCP2
in neuronal tissue increased cellular ATP and ADP levels Energy production
by triggering mitochondrial biogenesis. KD appears to do Polyunsaturated fatty acids additionally regulate the
the same; that is, studies show that the KD induces UCP transcription of numerous genes linked to energy
expression, stimulates mitochondrial biogenesis, and en- metabolism (Sampath and Ntambi, 2005) through activa-
hances energy production (see also below). Seizures, by tion of PPARα, a scenario in which the KD is thought to re-
comparison, increase ROS generation and/or mitochon- program cellular metabolism (Cullingford, 2004). Indeed,
drial dysfunction, which can lead to neuronal dysfunction numerous studies have described changes consistent with
and excitotoxicity (Layton and Pazdernik, 1999; Kovacs an enhancement in energy production following KD treat-
et al., 2001; Kovacs et al., 2002; Sullivan et al., 2003). ment. First, microarray expression studies demonstrated
Interestingly, UCP2 is up-regulated after seizures (Diano that KD induces a coordinated up-regulation of several

Epilepsia, Vol. 48, No. 1, 2007


50 K. J. BOUGH AND J. M. RHO

dozen metabolic genes associated with oxidative phospho- originally hypothesized that KD-induced elevations in
rylation after KD (Noh et al., 2004; Bough et al., 2006). ATP concentrations might enhance and/or prolong the ac-
Second, KD treatment stimulated mitochondrial biogene- tivation of the Na+ /K+ -ATPase , perhaps via an increase in

sis, resulting in a striking 46% increase in the number of the delta-G of ATP hydrolysis (Veech et al., 2001; Veech,
mitochondria in the hilar/dentate gyrus region of rat hip- 2004). In neurons, increased sodium pump activity might
pocampus (Bough et al., 2006). And, third, levels of en- hyperpolarize the cell and/or reduce the resting mem-
ergy metabolites were increased after KD. Brain glycogen brane potential to diminish firing probability. Enhanced
and ATP concentrations were boosted throughout rodent Na+ /K+ -ATPase function in neurons might also preserve
brain (DeVivo et al., 1978; Otani et al., 1984) and there was normal neuronal functioning and/or delay a pathological
an elevation in the phosphocreatine-to-creatine (PCr:Cr) buildup of high external K+ (Xiong and Stringer, 2000).
energy-reserve ratio in both animals (Bough et al., 2006) In glia, increased activation of the Na+ /K+ -ATPase might
and humans (Pan et al., 1999). These findings are consis- slow glial depolarization and allow for prolonged uptake
tent with results that show ketones (4 mM BHB + 1 mM of extracellular K+ during periods of intense neuronal ac-
ACA) increased hydraulic work by 14% and improved tivity (e.g., high-frequency bursting). Increases in neu-
energy status in perfused heart tissue (Sato et al., 1995). ronal and/or glial action of the sodium pump would be
Further, there is an overall increased metabolic efficiency expected to limit hyperexcitability and increase the resis-
(DeVivo et al., 1978; Bough et al., 2006), decreased res- tance to seizures, as is noted after treatment with KD.
piratory quotient (Bough et al., 2000b), and maximal mi- Although no studies have tested this sodium-pump
tochondrial respiratory rate in rodents following the KD hypothesis directly, a recent report suggests that KD
(Sullivan et al., 2004). Collectively, these data provide tissue is more resistant to metabolic stress. When chal-
compelling evidence that the KD enhances oxidative en- lenged with mild hypoglycemia, synaptic transmission
ergy production by activating a variety of transcriptional, within the dentate gyrus was maintained for approxi-
translational, and biochemical mechanisms in a concerted mately 60% longer in tissue from KD-fed animals com-
fashion. pared to controls (Bough et al., 2006). These data suggest
Metabolic dysfunction has been identified in regions of that the KD stabilizes synaptic transmission (both excita-
hyperexcitability within the brain and is associated with tory and inhibitory) for prolonged periods of time during
several epileptic conditions. Impairment of mitochondrial metabolic stress such as during seizure activity. Hence,
function has been observed in the seizure foci of both hu- it seems likely that the KD induces seizure protection
man and experimental epilepsies (Kunz et al., 2000). Se- in part by preventing neuronal dysfunction (diminution
vere metabolic dysfunction occurred in both human and of ROS/enhancement of energy reserves) and stabilizing
rat hippocampal tissue during periods of heightened neu- synaptic transmission (enhancement in energy reserves).
ronal activity (Kann et al., 2005). Kudin et al. (2002)
demonstrated that seizure activity down-regulated mito-
A role for neurotransmitter systems
chondrial enzymes involved in oxidative phosphorylation.
In an earlier study, the same group demonstrated a specific The noradrenergic hypothesis
deficiency in complex I activity and mitochondrial ultra- One of the more intriguing observations regarding KD
structural abnormalities within the hippocampal CA3 re- action involves norepinephrine, its receptors and signaling
gion of epileptic tissue resected from 57 human patients cascades. In general, increases in noradrenergic tone re-
(Kunz et al., 2000). In view of previous studies demon- sult in an anticonvulsant effect. Several lines of evidence
strating impaired oxidative phosphorylation capacity in support this view. Norepinephrine (NE) re-uptake in-
pilocarpine-treated rats (Kudin et al., 2002) and in patients hibitors can prevent seizure activity in genetically epilepsy
with epilepsy (Antozzi et al., 1995; Kunz et al., 2000), a prone rats (GEPRs; Yan et al., 1993) and pharmacolog-
KD-induced augmentation in oxidative phosphorylation ical NE agonists are generally, though not always, an-
and energy reserves seems likely to counter energetic de- ticonvulsant (Weinshenker and Szot, 2002); damage to
ficiencies in epileptic tissue, making neuronal tissue more the locus coeruleus—the principal region of the brain
resilient to aberrant neuronal activity and, in this way, con- from which ascending and descending noradrenergic in-
tributing to the diet’s anticonvulsant actions. nervation originates—converts occasional seizures into
self-sustaining status epilepticus (SSSE) in rats (Giorgi
Stabilized synaptic function et al., 2004); animals are more prone to seizures when NE
Intriguing as this argument may be, how exactly would is chemically depleted with reserpine (Weinshenker and
enhanced energy reserves lead to stabilized synaptic func- Szot, 2002); and, interestingly, there are several reports of
tion and diminished seizures? One possibility is via the diminished brain levels of NE in several animal models
sodium pump. ATP is primarily used to maintain ionic gra- of epilepsy, including GEPRs, kindled animals, EL mice,
dients, especially through actions of the transmembrane seizure-sensitive Mongolian gerbils, and tottering mice
sodium pump (Hulbert and Else, 2000). Schwartzkroin (Weinshenker and Szot, 2002).

Epilepsia, Vol. 48, No. 1, 2007


ANTICONVULSANT ACTIONS OF KETOGENIC DIET 51

Of significant interest is the observation that mice lack- duced by pentylenetetrazole, bicuculline, picrotoxin, and
ing the ability to produce NE (Dbh−/− knockout mice) γ -butyrolactone. In contrast, the diet demonstrates lit-
do not exhibit an increased resistance to flurothyl seizures tle if any efficacy in acute seizure models involving ac-
when treated with a KD (Szot et al., 2001). These data tivation of ionotropic glutamate receptors (e.g., kainic
indicate that NE is required for the anticonvulsant effect acid), voltage-dependent sodium channels (e.g., maximal
of KD, at least in the flurothyl seizure threshold model. electroshock [MES]), or glycine receptor inhibition (e.g.,
Weinshenker and Szot (2002) additionally reported an ap- strychnine; Bough et al., 2002).
proximate twofold increase in NE levels in hippocampus Yudkoff et al. (2005) have proposed that ketosis in-
following a KD, suggesting that KD increases basal re- duces major shifts in brain amino acid handling favoring
lease of NE. These studies indicate the anticonvulsant ac- the production of GABA. This results from a reduction
tion of KD may result in part from an enhancement in of aspartate relative to glutamate, the precursor to GABA
noradrenergic signaling in the brain. synthesis, and a shift in the equilibrium of the aspartate
If the KD enhances NE release as described above, it aminotransferase reaction in the ketotic state. As a re-
may also promote the corelease of anticonvulsant orexi- sult, there is an increase in glutamic acid decarboxylase
genic peptides such as neuropeptide-Y (NPY) and galanin. (GAD) activity and GABA production (Fig. 3). Elevated
NPY has been shown to inhibit glutamatergic synaptic GABA levels would, in turn, be expected to dampen hyper-
transmission and epileptogenesis in vitro (Rhim et al., excitability throughout the brain. Several studies support
1997; Richichi et al., 2004; Vezzani and Sperk, 2004); this possibility. First, KD and CR diet treatments both
galanin has been shown to limit SSSE (Saar et al., 2002) increased GAD transcript and protein levels in inferior
and diminish both excitatory synaptic transmission and and superior colliculi, cerebellar and temporal cortex, and
ictal activity in vitro (Schlifke et al., 2006). Both neu- striatum (the latter, KD only; Cheng et al., 2004). Sec-
ropeptides are elevated after calorie restriction. However, ond, both BHB and ACA increased the rate and extent
there was no evidence for enhanced transcription of either of GABA formation in synaptosomes (Erecinska et al.,
of these peptides in the brain after KD treatment, suggest- 1996; Yudkoff et al., 1997). And, finally, KD treatment
ing that neither NPY nor galanin contribute significantly in vivo modified amino acid levels in a manner consistent
to the anticonvulsant actions of KD (Tabb et al., 2004). with enhanced GABA production (Yudkoff et al., 2001;
Melo et al., 2006). Although brain levels of glutamate
The GABAergic hypothesis and GABA have not been consistently elevated in rodents
One of the more popular hypotheses for KD action in- (DeVivo et al., 1978; Al-Mudallal et al., 1996; Yudkoff
volves γ -aminobutyric acid (GABA), the major inhibitory et al., 2001; Bough et al., 2006), two recent clinical stud-
neurotransmitter in the mammalian brain. In general, the ies report significant increases in GABA levels following
KD is most effective against seizures evoked by GABAer- KD treatment (Wang et al., 2003; Dahlin et al., 2005),
gic antagonists. The KD potently inhibits seizures in- further substantiating this view.

FIG. 3. Metabolic modifications of glutamate and GABA synthesis as a consequence of diminished glucose and ketosis. In ketosis, beta-
hydroxybutyrate and acetoacetate contribute heavily to brain energy needs. A variable fraction of pyruvate (1) is ordinarily converted to
acetyl-CoA via pyruvate dehydrogenase. In contrast, all ketone bodies generate acetyl-CoA, which enters the tricarboxcylic acid (TCA)
cycle via the citrate synthetase pathway (2). This involves the consumption of oxaloacetate, which is necessary for the transamination of
glutamate to aspartate. Oxaloacetate is then less available as a reactant of the aspartate aminotransferase pathway, which couples the
glutamate-aspartate interchange via transamination to the metabolism of glucose through the TCA cycle. Less glutamate is converted
to aspartate and thus, more glutamate is available for synthesis of GABA (3) through glutamic acid decarboxylase (GAD). Adapted from
Yudkoff et al. (2004).

Epilepsia, Vol. 48, No. 1, 2007


52 K. J. BOUGH AND J. M. RHO

In addition to biochemical measures of KD-enhanced table absence of Class I and II clinical studies. Today, few
GABAergic inhibition, there is functional evidence as question the clinical efficacy of the KD in both young and
well. Electrophysiological recordings conducted in vivo older patients (Vining, 1999; Coppola et al., 2002; Mady
demonstrated that network excitability was diminished in et al., 2003), and many successful international centers
both KD- and calorie-restricted rats (Bough et al., 2003); have evolved (Kossoff and McGrogan, 2005; Freeman
greater stimulus intensities were required to evoke popu- et al., 2006). However, since we do not fundamentally
lation spikes in both CR- and KD-fed animals compared to know how the KD prevents seizures, there exists as yet no
ad libitum controls. Paired-pulse inhibition was increased. rational basis for optimizing the efficacy of the diet, other
Both CR and KD dietary treatments resulted in greater than through trial and error.
paired-pulse inhibition compared to controls at the 30- When examining the accumulated clinical data, it ap-
ms interpulse interval (Bough et al., 2003), a result con- pears seizure control can be achieved in the majority
sistent with an enhancement in fast, GABAA -mediated of epileptic patients as long as there is a shift from
inhibition. Additionally, KD-fed animals exhibited an el- glycolytic flux to intermediary metabolism (resulting in
evated electrographic seizure threshold and an increased measurable ketosis), irrespective of the precise dietary
resistance to a modified, 1-day kindling protocol (max- formulation (Henderson et al., 2006). On the other hand,
imal dentate activation). These data suggested that both the experimental literature suggests that different treat-
KD and calorie-restricted diets limited network excitabil- ment protocols may result in differential efficacy or even
ity and elevated seizure threshold via an enhancement of lack of efficacy, despite significant ketosis (Bough et al.,
GABAergic inhibition. 2000a; Thavendiranathan et al., 2000; Bough et al., 2002;
GABAergic interneurons, which at baseline have more Thavendiranathan et al., 2003). Most of the published
depolarized resting membrane potentials, endure non- studies have been based on acute seizure models, and not
accommodating bursts of neuronal firing and must on developmental epilepsy models. Hence, of course, one
metabolically persist (Attwell and Laughlin, 2001), lest must bear in mind that, despite dozens of animal models
network inhibition becomes compromised. Previous stud- of the KD, none recapitulate all of the essential features
ies have shown that a KD increases total brain [ATP] in the human epileptic condition (Stafstrom, 1999).
(DeVivo et al., 1978) and PCr/Cr or PCr/ATP energy re- So how can we reach the goal of developing a safer and
serve (Pan et al., 1999; Bough et al., 2006). Accordingly, more effective KD? The reductionist approach posits that
KD-induced elevations in PCr are likely to play a pivotal were we to identify the critical mediator of the diet’s an-
role in maintaining the activity of the Na+ /K+ -ATPase ticonvulsant effect, administration of this substrate alone
during periods of intense seizure activity, in both gluta- would likely yield a similar clinical effect as the tradi-
matergic and GABAergic neurons. In a recent study of tional KD, and importantly, spare the patient significant
human temporal lobe epilepsy (Williamson et al., 2005), side-effects that may preclude its use—even in the face of
the PCr/ATP ratio correlated with the recovery of the clear clinical efficacy. The closest we have come to this
membrane potential following a stimulus train, which situation is the recent use of BHB as an oral neuroprotec-
was inversely correlated with granule cell bursting. Be- tant. Promising results have already been demonstrated
cause creatine kinase is predominantly localized within in Phase I clinical trials (Smith et al., 2005). Neverthe-
GABAergic interneurons (Boero et al., 2003), Boero et al. less, despite increasing experimental evidence that BHB
concluded that PCr and energy levels are especially crit- and ACA both possess neuroprotective properties (Kashi-
ical to the maintenance of GABAergic inhibitory output. waya et al., 2000; Noh et al., 2006), a direct anticonvulsant
In this manner, a KD-induced increase in energy reserves effect of ketone bodies has not yet been demonstrated in
might enhance GABAergic function in particular and im- epileptic brain, either animal or human. Intriguing, how-
prove seizure control. ever, are animal studies indicating that ACA and acetone
are anticonvulsant in acute seizure models. Yet, there re-
mains a perplexing lack of an acute anticonvulsant effect
HOW CAN THE KETOGENIC
of the principal ketone body, BHB.
DIET BE OPTIMIZED?
Conversely, if we believe that certain PUFAs, in lieu of
Historically, few guidelines have emerged regarding the ketone bodies, are direct mediators of an anticonvulsant ef-
clinical implementation of the KD and its variants – in- fect (Cunnane et al., 2002; Cunnane, 2004), as suggested
cluding the medium-chain triglyceride (MCT) formula- by clinical studies (Schlanger et al., 2002; Fraser et al.,
tion (Huttenlocher et al., 1971) and more recent options 2003; Fuehrlein et al., 2004; Yuen et al., 2005), we may be
such as the Atkins diet (Kossoff et al., 2003; Kossoff closer to distilling the essence of the KD. However, there
et al., 2006). This is largely a result of the fact that, until re- is likely no single fatty acid that is necessary and sufficient
cently, few KD centers existed throughout the world. Even for an anticonvulsant effect. And experimentally, while it
with a resurgence of interest in dietary approaches toward has been straightforward to demonstrate the inhibitory ef-
epilepsy treatment in the past decade, there remains a no- fects of PUFAs on specific voltage-gated ion channels and

Epilepsia, Vol. 48, No. 1, 2007


ANTICONVULSANT ACTIONS OF KETOGENIC DIET 53

FIG. 4. Hypothetical pathways leading to the anticonvulsant effects of the ketogenic diet (KD). Elevated free fatty acids (FFA) lead to
chronic ketosis and increased concentrations of polyunsaturated fatty acids (PUFAs) in the brain. Chronic ketosis is predicted to lead
to increased levels of acetone; this might activate K2P channels to hyperpolarize neurons and limit neuronal excitability. Chronic ketosis
is also anticipated to modify the tricarboxcylic acid (TCA) cycle. This would increase glutamate and, subsequently, GABA synthesis in
brain. Among several direct inhibitory actions (see also Fig. 2), PUFAs boost the activity of brain-specific uncoupling proteins (UCPs).
This is expected to limit ROS generation, neuronal dysfunction, and resultant neurodegeneration. Acting via the nuclear transcription
factor peroxisome proliferator-activated receptor-α (PPARα), PUFAs would induce the expression of UCPs and coordinately up-regulate
several dozen genes related to oxidative energy metabolism. PPARα expression is inversely correlated with IL-1β cytokine expression;
given the role of IL-1β in hyperexcitability and seizure generation (Vezzani et al., 2000), diminished expression of IL-β cytokines during KD
treatment could lead to improved seizure control. Ultimately, PUFAs would stimulate mitochondrial biogenesis. Mitochondrial biogenesis is
predicted to increase ATP production capacity and enhance energy reserves, leading to stabilized synaptic function and improved seizure
control. In particular, an elevated phosphocreatine:creatine (PCr:Cr) energy-reserve ratio is predicted to enhance GABAergic output,
perhaps in conjunction with the ketosis-induced elevated GABA production, leading to diminished hyperexcitability. Reduced glucose
coupled with elevated free fatty acids are proposed to reduce glycolytic flux during KD, which would further be feedback inhibited by high
concentrations of citrate and ATP produced during KD treatment. This would activate metabolic KATP channels. Opening of KATP channels
would hyperpolarize neurons and diminish neuronal excitability to contribute to the anticonvulsant (and perhaps neuroprotective) action
of the KD. Reduced glucose is also expected to downregulate brain-derived neurotrophic factor (BDNF) and trkB signaling in brain. As
activation of TrkB pathways by BDNF have been shown to promote hyperexcitability and kindling, these potential KD-induced effects would
be expected to limit the symptom (seizures) as well as the progression of epilepsy. Boxed variables depict findings taken from KD studies;
up (↑) or down (↓) arrows indicate the direction of the relationship between variables as a result of KD treatment.

the resultant diminution of cellular excitability in vitro, beta-oxidation. The collective data, from both animals and
it is not an easy task to demonstrate that ingestion of a humans, indicate that the critical condition necessary for
specific fatty acid or fatty acid cocktail, acts directly on achieving seizure control is a metabolic shift toward fatty
relevant brain receptor targets without first undergoing acid oxidation from glycolysis, reflected in the variable

Epilepsia, Vol. 48, No. 1, 2007


54 K. J. BOUGH AND J. M. RHO

rise in blood/brain ketone levels and a concomitant (mod- mechanistically to this broadly efficacious treatment for
erate) reduction in blood/brain glucose. Fatty acid compo- epilepsy. The challenge of finding key variables is made
sition may not ultimately matter, as long as this important ever more difficult by the intrinsic complexity of metabolic
metabolic shift occurs. And, interestingly, calorie restric- effects and their resultant actions on neurons, glia and on
tion (Greene et al., 2001; Bough et al., 2003; Eagles et al., the epileptic condition itself. We have reviewed here a
2003; Greene et al., 2003) or intake of 2-DG (Stafstrom number of seemingly disparate variables that must be sus-
et al., 2005), both of which result in mild hypoglycemia, tained for a meaningful anticonvulsant effect to be ren-
may be the only requirement for seizure protection, re- dered. These interrelationships are summarized in Fig. 4.
gardless of whether fats are consumed or not. The fact that a fundamental modification in diet can have
As we continue to explore putative anticonvulsant such profound, therapeutic effects on neurological disease
mechanisms of KD action, we are left with many out- underscores the importance of elucidating mechanisms of
standing clinical questions regarding dietary treatments KD action. Future studies will no doubt provide unique
for epilepsy. Well-designed, multicenter prospective- and insights into how diet can affect the brain, both in health
controlled clinical trials are essential toward developing and disease, and likely provide the scientific basis for the
the optimum KD. If woven together with pharmacokinetic development of potent new treatment strategies for the
and pharmacogenetic investigations, these clinical stud- epilepsies.
ies will not only provide further insights into mechanistic
underpinnings, but will also help differentiate responders REFERENCES
from non-responders and identify patients in whom the
diet is definitively contraindicated. Clinicians would be Al-Mudallal AS, LaManna JC, Lust WD, Harik SI. (1996) Diet-induced
ketosis does not cause cerebral acidosis. Epilepsia 37:258–261.
given the tools to make evidence-based decisions rather Andrews ZB, Diano S, Horvath TL. (2005) Mitochondrial uncoupling
than rely upon a few case–controlled studies, anecdotal re- proteins in the cns: in support of function and survival. Nature Re-
ports of efficacy, or clinical folklore as has been the prac- views Neuroscience 6:829–840.
Antozzi C, Franceschetti S, Filippini G, Barbiroli B, Savoiardo M, Fi-
tice in the past. Toward this end, information regarding the acchino F, Rimoldi M, Lodi R, Zaniol P, Zeviani M. (1995) Epilepsia
impact of pharmacogenetics on epilepsy treatment is now partialis continua associated with nadh-coenzyme q reductase defi-
beginning to emerge (Depondt and Shorvon, 2006; Spurr, ciency. Journal of the Neurological Sciences 129:152–161.
Appleton DB, DeVivo DC. (1974) An animal model for the ketogenic
2006), although much less is known regarding the genet- diet. Epilepsia 15:211–227.
ically determined variables influencing dietary impact on Attwell D, Laughlin SB. (2001) An energy budget for signaling in
brain function, particularly as it relates to the epileptic the grey matter of the brain. Journal of Cerebral Blood Flow and
Metabolism 21:1133–1145.
brain. Avellini L, Terracina L, Gaiti A. (1994) Linoleic acid passage through
the blood-brain barrier and a possible effect of age. Neurochemical
Research 19:129–133.
CONCLUSIONS Boero J, Qin W, Cheng J, Woolsey TA, Strauss AW, Khuchua Z. (2003)
Restricted neuronal expression of ubiquitous mitochondrial creatine
After nearly a century of clinical use, we still do not kinase: changing patterns in development and with increased activity.
know how the KD works. However, much progress in KD Molecular and Cellular Biochemistry 244:69–76.
Bough KJ, Chen RS, Eagles DA. (1999a) Path analysis shows that
research has been made in the past decade. Among other increasing ketogenic ratio, but not beta-hydroxybutarate, elevates
factors, current evidence indicates KD optimizes cellular seizure threshold in the rat. Developmental Neuroscience 21:400–
metabolism. Endogenous biochemical and genetic ‘pro- 406.
Bough KJ, Eagles DA. (1999) A ketogenic diet increases the resistance
grams’ are switched on in the brain in response to keto- to pentylenetetrazole-induced seizures in the rat. Epilepsia 40:138–
sis, glucose restriction, and elevated free fatty acids. This 143.
unique metabolic state, if maintained, induces a shift away Bough KJ, Valiyil R, Han FT, Eagles DA. (1999b) Seizure resistance
is dependent upon age and calorie restriction in rats fed a ketogenic
from glycolytic energy production (glucose restriction) diet. Epilepsy Research 35:21–28.
toward the production of energy via oxidative phospho- Bough KJ, Matthews PJ, Eagles DA. (2000a) A ketogenic diet has
rylation (beta-oxidation of fatty acids and production of different effects upon seizures induced by maximal electroshock
and by pentylenetetrazole infusion. Epilepsy Research 38:105–
ketone bodies). The reduction in glycolytic energy sup- 114.
ply may activate selectively KATP channels to increase the Bough KJ, Yao SG, Eagles DA. (2000b) Higher ketogenic diet ratios
resistance to onset of ictal activity. An increase in oxida- confer protection from seizures without neurotoxicity. Epilepsy Re-
search 38:15–25.
tive phosphorylation coupled with an induction of UCPs Bough KJ, Gudi K, Han FT, Rathod AH, Eagles DA. (2002) An anti-
and mitochondrial biogenesis can diminish ROS genera- convulsant profile of the ketogenic diet in the rat. Epilepsy Research
tion and increase energy reserves, both of which would 50:313–325.
Bough KJ, Schwartzkroin PA, Rho JM. (2003) Calorie restriction and
be expected to prevent neuronal dysfunction, seizures and ketogenic diet diminish neuronal excitability in rat dentate gyrus in
even neurodegeneration. vivo. Epilepsia 44:752–760.
It is improbable that one mechanistic target or medi- Bough KJ, Wetherington J, Hassel B, Pare JF, Gawryluk JW, Greene JG,
Shaw R, Smith Y, Geiger JD, Dingledine RJ. (2006) Mitochondrial
ator will produce entirely the seizure protection associ- biogenesis in the anticonvulsant mechanism of the ketogenic diet.
ated with the KD. Rather, several factors likely contribute Annals of Neurology 60:223–235.

Epilepsia, Vol. 48, No. 1, 2007


ANTICONVULSANT ACTIONS OF KETOGENIC DIET 55

Cheng CM, Hicks K, Wang J, Eagles DA, Bondy CA. (2004) Caloric Freeman JM, Vining EP. (1999) Seizures decrease rapidly after fasting:
restriction augments brain glutamic acid decarboxylase-65 and -67 preliminary studies of the ketogenic diet. Archives of Pediatrics &
expression. Journal of Neuroscience Research 77:270–276. Adolescent Medicine 153:946–949.
Coppola G, Veggiotti P, Cusmai R, Bertoli S, Cardinali S, Dionisi-Vici Freeman JM, Kelley MT, Freeman JB. (2000) The ketogenic diet: a
C, Elia M, Lispi ML, Sarnelli C, Tagliabue A, Toraldo C, Pascotto A. treatment for epilepsy, 3rd ed. Demos, New York.
(2002) The ketogenic diet in children, adolescents and young adults Fuehrlein BS, Rutenberg MS, Silver JN, Warren MW, Theriaque
with refractory epilepsy: an italian multicentric experience. Epilepsy DW, Duncan GE, Stacpoole PW, Brantly ML. (2004) Differential
Research 48:221–227. metabolic effects of saturated versus polyunsaturated fats in keto-
Cullingford TE. (2004) The ketogenic diet; fatty acids, fatty acid- genic diets. The Journal of Clinical Endocrinology and Metabolism
activated receptors and neurological disorders. Prostaglandins 89:1641–1645.
Leukot Essent Fatty Acids 70:253–264. Garlid KD, Jaburek M, Jezek P. (2001) Mechanism of uncoupling protein
Cunnane SC, Musa K, Ryan MA, Whiting S, Fraser DD. (2002) Poten- action. Biochemical Society Transactions 29:803–806.
tial role of polyunsaturates in seizure protection achieved with the Garriga-Canut M, Schoenike B, Qazi R, Bergendahl K, Daley TJ, Pfender
ketogenic diet. Prostaglandins Leukot Essent Fatty Acids 67:131– RM, Morrison JF, Ockuly J, Stafstrom CE, Sutula T, Roopra A.
135. (2006) 2-deoxy-d-glucose reduces epilepsy progression by nrsf-ctbp-
Cunnane SC. (2004) Metabolism of polyunsaturated fatty acids and ke- dependent metabaolic regulation of chromatin structure. Nature Neu-
togenesis: an emerging connection. Prostaglandins Leukot Essent roscience 9:1382–1387.
Fatty Acids 70:237–241. Giorgi FS, Pizzanelli C, Biagioni F, Murri L, Fornai F. (2004) The role
Dahlin M, Elfving A, Ungerstedt U, Amark P. (2005) The ketogenic of norepinephrine in epilepsy: from the bench to the bedside. Neu-
diet influences the levels of excitatory and inhibitory amino acids roscience and Biobehavioral Reviews 28:507–524.
in the csf in children with refractory epilepsy. Epilepsy Research Greene AE, Todorova MT, McGowan R, Seyfried TN. (2001) Caloric
64:115–125. restriction inhibits seizure susceptibility in epileptic el mice by re-
Dekaban AS. (1966) Plasma lipids in epileptic children treated with the ducing blood glucose. Epilepsia 42:1371–1378.
high fat diet. Archives of Neurology 15:177–184. Greene AE, Todorova MT, Seyfried TN. (2003) Perspectives on the
Dell CA, Likhodii SS, Musa K, Ryan MA, Burnham WM, Cunnane metabolic management of epilepsy through dietary reduction of glu-
SC. (2001) Lipid and fatty acid profiles in rats consuming different cose and elevation of ketone bodies. Journal of Neurochemistry
high-fat ketogenic diets. Lipids 36:373–378. 86:529–537.
Depondt C, Shorvon SD. (2006) Genetic association studies in epilepsy Helmholz HF, Keith HM. (1930) Eight years’ experience with the ke-
pharmacogenomics: lessons learnt and potential applications. Phar- togenic diet in the treatment of epilepsy. Journal of the American
macogenomics 7:731–745. Medical Association 95:707–709.
DeVivo DC, Leckie MP, Ferrendelli JS, McDougal DB Jr. (1978) Henderson CB, Filloux FM, Alder SC, Lyon JL, Caplin DA. (2006)
Chronic ketosis and cerebral metabolism. Annals of Neurology Efficacy of the ketogenic diet as a treatment option for epilepsy:
3:331–337. meta-analysis. Journal of Child Neurology 21:193–198.
Dhar-Chowdhury P, Harrell MD, Han SY, Jankowska D, Parachuru L, Hernandez-Sanchez C, Basile AS, Fedorova I, Arima H, Stannard
Morrissey A, Srivastava S, Liu W, Malester B, Yoshida H, Coetzee B, Fernandez AM, Ito Y, LeRoith D. (2001) Mice transgenically
WA. (2005) The glycolytic enzymes, glyceraldehyde-3-phosphate overexpressing sulfonylurea receptor 1 in forebrain resist seizure
dehydrogenase, triose-phosphate isomerase, and pyruvate kinase are induction and excitotoxic neuron death. Proceedings of the Na-
components of the k(atp) channel macromolecular complex and reg- tional Academy of Sciences of the United States of America 98:
ulate its function. The Journal of Biological Chemistry 280:38464– 3549– 3554.
38470. Hicks GA, Hudson AL, Henderson G. (1994) Localization of high affin-
Diano S, Matthews RT, Patrylo P, Yang L, Beal MF, Barnstable CJ, ity [3 H]glibenclamide binding sites within the substantia nigra zona
Horvath TL. (2003) Uncoupling protein 2 prevents neuronal death reticulata of the rat brain. Neuroscience 61:285–292.
including that occurring during seizures: a mechanism for precondi- Hold KM, Sirisoma NS, Ikeda T, Narahashi T, Casida JE. (2000) Alpha-
tioning. Endocrinology 144:5014–5021. thujone (the active component of absinthe): gamma-aminobutyric
Donevan SD, White HS, Anderson GD, Rho JM. (2003) Voltage- acid type a receptor modulation and metabolic detoxification. Pro-
dependent block of N-methyl-D-aspartate receptors by the novel an- ceedings of the National Academy of Sciences of the United States
ticonvulsant dibenzylamine, a bioactive constituent of l-(+)-beta- of America 97:3826–3831.
hydroxybutyrate. Epilepsia 44:1274–1279. Hori A, Tandon P, Holmes GL, Stafstrom CE. (1997) Ketogenic diet:
Dubinsky WP, Mayorga-Wark O, Schultz SG. (1998) Colocalization of effects on expression of kindled seizures and behavior in adult rats.
glycolytic enzyme activity and katp channels in basolateral mem- Epilepsia 38:750–758.
brane of necturus enterocytes. The American Journal of Physiology Hulbert AJ, Else PL. (2000) Mechanisms underlying the cost of living
275:C1653–1659. in animals. Annual Review of Physiology 62:207–235.
Eagles DA, Boyd SJ, Kotak A, Allan F. (2003) Calorie restriction of a Huttenlocher PR, Wilbourn AJ, Signore JM. (1971) Medium-chain
high-carbohydrate diet elevates the threshold of ptz-induced seizures triglycerides as a therapy for intractable childhood epilepsy. Neu-
to values equal to those seen with a ketogenic diet. Epilepsy Research rology 21:1097–1103.
54:41–52. Huttenlocher PR. (1976) Ketonemia and seizures: metabolic and an-
Erecinska M, Nelson D, Daikhin Y, Yudkoff M. (1996) Regulation of ticonvulsant effects of two ketogenic diets in childhood epilepsy.
gaba level in rat brain synaptosomes: fluxes through enzymes of the Pediatric Research 10:536–540.
gaba shunt and effects of glutamate, calcium, and ketone bodies. Iadarola MJ, Gale K. (1982) Substantia nigra: site of anticonvulsant
Journal of Neurochemistry 67:2325–2334. activity mediated by gamma-aminobutyric acid. Science 218:1237–
Franks NP, Honore E. (2004) The trek k2p channels and their role in 1240.
general anaesthesia and neuroprotection. Trends in Pharmacological Jaburek M, Varecha M, Gimeno RE, Dembski M, Jezek P, Zhang M,
Sciences 25:601–608. Burn P, Tartaglia LA, Garlid KD. (1999) Transport function and
Fraser DD, Whiting S, Andrew RD, Macdonald EA, Musa-Veloso K, regulation of mitochondrial uncoupling proteins 2 and 3. The Journal
Cunnane SC. (2003) Elevated polyunsaturated fatty acids in blood of Biological Chemistry 274:26003–26007.
serum obtained from children on the ketogenic diet. Neurology Jovanovic S, Du Q, Crawford RM, Budas GR, Stagljar I, Jovanovic
60:1026–1029. A. (2005) Glyceraldehyde 3-phosphate dehydrogenase serves as an
Freeman J, Veggiotti P, Lanzi G, Tagliabue A, Perucca E. (2006) The ke- accessory protein of the cardiac sarcolemmal k(atp) channel. EMBO
togenic diet: from molecular mechanisms to clinical effects. Epilepsy Reports 6:848–852.
Research 68:145–180. Kann O, Kovacs R, Njunting M, Behrens CJ, Otahal J, Lehmann TN,
Freeman JM, Vining EP, Pillas DJ, Pyzik PL, Casey JC, Kelly LM. (1998) Gabriel S, Heinemann U. (2005) Metabolic dysfunction during neu-
The efficacy of the ketogenic diet-1998: a prospective evaluation of ronal activation in the ex vivo hippocampus from chronic epileptic
intervention in 150 children. Pediatrics 102:1358–1363. rats and humans. Brain 128:2396–2407.

Epilepsia, Vol. 48, No. 1, 2007


56 K. J. BOUGH AND J. M. RHO

Kashiwaya Y, Takeshima T, Mori N, Nakashima K, Clarke K, Veech Millichap JG, Jones JD, Rudis BP. (1964) Mechanism of anticonvulsant
RL. (2000) D-beta-hydroxybutyrate protects neurons in models of action of ketogenic diet. American Journal of Diseases of Children
alzheimer’s and parkinson’s disease. Proceedings of the National 107:593–604.
Academy of Sciences of the United States of America 97:5440– Morris AA. (2005) Cerebral ketone body metabolism. Journal of Inher-
5444. ited Metabolic Disease 28:109–121.
Keith HM. (1933) Factors influencing experimentally produced convul- Muller-Schwarze AB, Tandon P, Liu Z, Yang Y, Holmes GL, Stafstrom
sions. Archives of Neurology Psychiatry 29:148–154. CE. (1999) Ketogenic diet reduces spontaneous seizures and mossy
Kinsman SL, Vining EP, Quaskey SA, Mellits D, Freeman JM. (1992) fiber sprouting in the kainic acid model. Neuroreport 10:1517–1522.
Efficacy of the ketogenic diet for intractable seizure disorders: review Nakazawa M, Kodama S, Matsuo T. (1983) Effects of ketogenic diet
of 58 cases. Epilepsia 33:1132–1136. on electroconvulsive threshold and brain contents of adenosine nu-
Kossoff EH, Pyzik PL, McGrogan JR, Vining EP, Freeman JM. (2002) cleotides. Brain & Development 5:375–380.
Efficacy of the ketogenic diet for infantile spasms. Pediatrics Noh HS, Lee HP, Kim DW, Kang SS, Cho GJ, Rho JM, Choi WS.
109:780–783. (2004) A cdna microarray analysis of gene expression profiles in rat
Kossoff EH, Krauss GL, McGrogan JR, Freeman JM. (2003) Efficacy hippocampus following a ketogenic diet. Brain Research. Molecular
of the atkins diet as therapy for intractable epilepsy. Neurology Brain Research 129:80–87.
61:1789–1791. Noh HS, Hah YS, Nilufar R, Han J, Bong JH, Kang SS, Cho GJ, Choi WS.
Kossoff EH, McGrogan JR. (2005) Worldwide use of the ketogenic diet. (2006) Acetoacetate protects neuronal cells from oxidative glutamate
Epilepsia 46:280–289. toxicity. Journal of Neuroscience Research 83:702–709.
Kossoff EH, McGrogan JR, Bluml RM, Pillas DJ, Rubenstein JE, Vining Nordli DR, Jr., Kuroda MM, Carroll J, Koenigsberger DY, Hirsch LJ,
EP. (2006) A modified atkins diet is effective for the treatment of Bruner HJ, Seidel WT, De Vivo DC. (2001) Experience with the
intractable pediatric epilepsy. Epilepsia 47:421–424. ketogenic diet in infants. Pediatrics 108:129–133.
Kovacs R, Schuchmann S, Gabriel S, Kardos J, Heinemann U. (2001) Nordoy A. (1999) Dietary fatty acids and coronary heart disease. Lipids
Ca2+ signalling and changes of mitochondrial function during low- 34(suppl):S19–22.
mg2+-induced epileptiform activity in organotypic hippocampal Otani K, Yamatodani A, Wada H, Mimaki T, Yabuuchi H. (1984) [Effect
slice cultures. The European Journal of Neuroscience 13:1311–1319. of ketogenic diet on the convulsive threshold and brain amino acid
Kovacs R, Schuchmann S, Gabriel S, Kann O, Kardos J, Heinemann U. and monoamine levels in young mice]. No To Hattatsu 16:196–204.
(2002) Free radical-mediated cell damage after experimental status Pan JW, Bebin EM, Chu WJ, Hetherington HP. (1999) Ketosis and
epilepticus in hippocampal slice cultures. Journal of Neurophysiol- epilepsy: 31p spectroscopic imaging at 4.1 t. Epilepsia 40:703–707.
ogy 88:2909–2918. Pelicano H, Martin DS, Xu RH, Huang P. (2006) Glycolysis inhibition
Kudin AP, Kudina TA, Seyfried J, Vielhaber S, Beck H, Elger CE, Kunz for anticancer treatment. Oncogene 25:4633–4646.
WS. (2002) Seizure-dependent modulation of mitochondrial oxida- Peterson SJ, Tangney CC, Pimentel-Zablah EM, Hjelmgren B, Booth G,
tive phosphorylation in rat hippocampus. The European Journal of Berry-Kravis E. (2005) Changes in growth and seizure reduction in
Neuroscience 15:1105–1114. children on the ketogenic diet as a treatment for intractable epilepsy.
Kunz WS, Kudin AP, Vielhaber S, Blumcke I, Zuschratter W, Schramm Journal of the American Dietetic Association 105:718–725.
J, Beck H, Elger CE. (2000) Mitochondrial complex i deficiency in Puchowicz MA, Emancipator DS, Xu K, Magness DL, Ndubuizu OI,
the epileptic focus of patients with temporal lobe epilepsy. Annals Lust WD, LaManna JC. (2005) Adaptation to chronic hypoxia dur-
of Neurology 48:766–773. ing diet-induced ketosis. Advances in Experimental Medicine and
Layton ME, Pazdernik TL. (1999) Reactive oxidant species in piriform Biology 566:51–57.
cortex extracellular fluid during seizures induced by systemic kainic Rhim H, Kinney GA, Emmerson PJ, Miller RJ. (1997) Regulation of
acid in rats. Journal of Molecular Neuroscience 13:63–68. neurotransmission in the arcuate nucleus of the rat by different neu-
Leaf A, Kang JX. (1996) Prevention of cardiac sudden death by n-3 ropeptide y receptors. The Journal of Neuroscience 17:2980–2989.
fatty acids: a review of the evidence. Journal of Internal Medicine Rho JM, Kim DW, Robbins CA, Anderson GD, Schwartzkroin PA.
240:5–12. (1999) Age-dependent differences in flurothyl seizure sensitivity
Leaf A, Xiao YF, Kang JX, Billman GE. (2003) Prevention of sudden in mice treated with a ketogenic diet. Epilepsy Research 37:233–
cardiac death by n-3 polyunsaturated fatty acids. Pharmacology & 240.
Therapeutics 98:355–377. Rho JM, Anderson GD, Donevan SD, White HS. (2002) Acetoac-
Lesage F. (2003) Pharmacology of neuronal background potassium etate, acetone, and dibenzylamine (a contaminant in l-(+)-beta-
channels. Neuropharmacology 44:1–7. hydroxybutyrate) exhibit direct anticonvulsant actions in vivo.
Likhodii S, Burnham WM. (2004) Epilepsy and the ketogenic diet. In- Epilepsia 43:358–361.
Stafstrom CE, Rho JM (Eds). The effects of ketone bodies on neuronal Richichi C, Lin EJ, Stefanin D, Colella D, Ravizza T, Grignaschi G, Veg-
excitability. Humana Press, Inc., Totowa, NJ, pp. 217–228. lianese P, Sperk G, During MJ, Vezzani A. (2004) Anticonvulsant and
Likhodii SS, Musa K, Mendonca A, Dell C, Burnham WM, Cunnane antiepileptogenic effects mediated by adeno-associated virus vector
SC. (2000) Dietary fat, ketosis, and seizure resistance in rats on the neuropeptide y expression in the rat hippocampus. The Journal of
ketogenic diet. Epilepsia 41:1400–1410. Neuroscience 24:3051–3059.
Likhodii SS, Serbanescu I, Cortez MA, Murphy P, Snead OC, III, Burn- Saar K, Mazarati AM, Mahlapuu R, Hallnemo G, Soomets U, Kilk K,
ham WM. (2003) Anticonvulsant properties of acetone, a brain ke- Hellberg S, Pooga M, Tolf BR, Shi TS, Hokfelt T, Wasterlain C,
tone elevated by the ketogenic diet. Annals of Neurology 54:219– Bartfai T, Langel U. (2002) Anticonvulsant activity of a nonpeptide
226. galanin receptor agonist. Proceedings of the National Academy of
Livingston S. (1972) Comprehensive management of epilepsy in infancy, Sciences of the United States of America 99:7136–7141.
childhood and adolescence. In Livingston S (Ed) Dietary treatment Sampath H, Ntambi JM. (2004) Polyunsaturated fatty acid regulation of
of epilepsy. Charles C. Thomas, Springfield, IL, pp. 378–405. gene expression. Nutrition Reviews 62:333–339.
Mady MA, Kossoff EH, McGregor AL, Wheless JW, Pyzik PL, Freeman Sampath H, Ntambi JM. (2005) Polyunsaturated fatty acid regulation of
JM. (2003) The ketogenic diet: adolescents can do it, too. Epilepsia genes of lipid metabolism. Annual Review of Nutrition 25:317–340.
44:847–851. Sato K, Kashiwaya Y, Keon CA, Tsuchiya N, King MT, Radda GK,
Mahoney AW, Hendricks DG, Bernhard N, Sisson DV. (1983) Fasting Chance B, Clarke K, Veech RL. (1995) Insulin, ketone bodies, and
and ketogenic diet effects on audiogenic seizures susceptibility of mitochondrial energy transduction. The FASEB Journal 9:651–658.
magnesium deficient rats. Pharmacology, Biochemistry, and Behav- Schlanger S, Shinitzky M, Yam D. (2002) Diet enriched with omega-
ior 18:683–687. 3 fatty acids alleviates convulsion symptoms in epilepsy patients.
Mak SC, Chi CS, Wan CJ. (1999) Clinical experience of ketogenic diet Epilepsia 43:103–104.
on children with refractory epilepsy. Acta Paediatrica Taiwanica Schlifke I, Kuteeva E, Hokfelt T, Kokaia M. (2006) Galanin expressed
40:97–100. in the excitatory fibers attenuates synaptic strength and generalized
Melo TM, Nehlig A, Sonnewald U. (2006) Neuronal-glial interactions in seizures in the piriform cortex of mice. Experimental Neurology
rats fed a ketogenic diet. Neurochemistry International 48:498–507. 200:398–406.

Epilepsia, Vol. 48, No. 1, 2007


ANTICONVULSANT ACTIONS OF KETOGENIC DIET 57

Schwartz RH, Eaton J, Bower BD, Aynsley-Green A. (1989a) Ketogenic genic diet in mice. The Journal of Pharmacology and Experimental
diets in the treatment of epilepsy: short-term clinical effects. Devel- Therapeutics 180:231–238.
opmental Medicine and Child Neurology 31:145–151. Vamecq J, Vallee L, Lesage F, Gressens P, Stables JP. (2005) Antiepilep-
Schwartz RM, Boyes S, Aynsley-Green A. (1989b) Metabolic effects of tic popular ketogenic diet: emerging twists in an ancient story.
three ketogenic diets in the treatment of severe epilepsy. Develop- Progress in Neurobiology 75:1–28.
mental Medicine and Child Neurology 31:152–160. Veech RL, Chance B, Kashiwaya Y, Lardy HA, Cahill GF Jr. (2001)
Schwartzkroin PA. (1999) Mechanisms underlying the anti-epileptic ef- Ketone bodies, potential therapeutic uses. IUBMB Life 51:241–247.
ficacy of the ketogenic diet. Epilepsy Research 37:171–180. Veech RL. (2004) The therapeutic implications of ketone bodies: the
Seino S, Miki T. (2003) Physiological and pathophysiological roles of effects of ketone bodies in pathological conditions: ketosis, ketogenic
atp-sensitive k+ channels. Progress in Biophysics and Molecular diet, redox states, insulin resistance, and mitochondrial metabolism.
Biology 81:133–176. Prostaglandins Leukot Essent Fatty Acids 70:309–319.
Seymour KJ, Bluml S, Sutherling J, Sutherling W, Ross BD. (1999) Vezzani A, Moneta D, Conti M, Richichi C, Ravizza T, De Luigi A,
Identification of cerebral acetone by 1h-mrs in patients with epilepsy De Simoni MG, Sperk G, Andell-Jonsson S, Lundkvist J, Iverfeldt
controlled by ketogenic diet. Magma 8:33–42. K, Bartfai T. (2000) Powerful anticonvulsant action of il-1 receptor
Shyng SL, Nichols CG. (1998) Membrane phospholipid control of nu- antagonist on intracerebral injection and astrocytic overexpression
cleotide sensitivity of katp channels. Science 282:1138–1141. in mice. Proceedings of the National Academy of Sciences of the
Sills MA, Forsythe WI, Haidukewych D, MacDonald A, Robinson M. United States of America 97:11534–11539.
(1986) The medium chain triglyceride diet and intractable epilepsy. Vezzani A, Sperk G. (2004) Overexpression of npy and y2 receptors in
Archives of Disease in Childhood 61:1168–1172. epileptic brain tissue: an endogenous neuroprotective mechanism in
Sirven J, Whedon B, Caplan D, Liporace J, Glosser D, O’Dwyer J, temporal lobe epilepsy? Neuropeptides 38:245–252.
Sperling MR. (1999) The ketogenic diet for intractable epilepsy in Vining EP, Freeman JM, Ballaban-Gil K, Camfield CS, Camfield PR,
adults: preliminary results. Epilepsia 40:1721–1726. Holmes GL, Shinnar S, Shuman R, Trevathan E, Wheless JW. (1998)
Smith SL, Heal DJ, Martin KF. (2005) Ktx 0101: a potential metabolic A multicenter study of the efficacy of the ketogenic diet. Archives of
approach to cytoprotection in major surgery and neurological disor- Neurology 55:1433–1437.
ders. CNS Drug Reviews 11:113–140. Vining EP. (1999) Clinical efficacy of the ketogenic diet. Epilepsy Re-
Spurr NK. (2006) Pharmacogenetic studies of epilepsy drugs: are we search 37:181–190.
there yet? Trends in Genetics 22:250–252. Vining EP, Pyzik P, McGrogan J, Hladky H, Anand A, Kriegler S, Free-
Stafstrom CE. (1999) Animal models of the ketogenic diet: what man JM. (2002) Growth of children on the ketogenic diet. Develop-
have we learned, what can we learn? Epilepsy Research 37:241– mental Medicine and Child Neurology 44:796–802.
259. Vodickova L, Frantik E, Vodickova A. (1995) Neutrotropic effects and
Stafstrom CE. (2004) Dietary approaches to epilepsy treatment: old and blood levels of solvents at combined exposures: binary mixtures of
new options on the menu. Epilepsy Currents 4:215–222. toluene, o-xylene and acetone in rats and mice. Central European
Stafstrom CE, Kriegler SM, Valley MT, Ockuly JC, Roopra AS, Sutula Journal of Public Health 3:57–64.
TP. (2005) 2-deoxyglucose exerts anticonvulsant and antiepileptic Vreugdenhil M, Bruehl C, Voskuyl RA, Kang JX, Leaf A, Wadman WJ.
actions in experimental epilepsy models. Epilepsia 46:268–269. (1996) Polyunsaturated fatty acids modulate sodium and calcium
Su SW, Cilio MR, Sogawa Y, Silveira DC, Holmes GL, Stafstrom CE. currents in CA1 neurons. Proceedings of the National Academy of
(2000) Timing of ketogenic diet initiation in an experimental epilepsy Sciences of the United States of America 93:12559–12563.
model. Brain Research Developmental Brain Research 125:131– Wang ZJ, Bergqvist C, Hunter JV, Jin D, Wang DJ, Wehrli S, Zimmerman
138. RA. (2003) In vivo measurement of brain metabolites using two-
Sullivan PG, Dube C, Dorenbos K, Steward O, Baram TZ. (2003) Mi- dimensional double-quantum mr spectroscopy–exploration of gaba
tochondrial uncoupling protein-2 protects the immature brain from levels in a ketogenic diet. Magnetic Resonance in Medicine 49:615–
excitotoxic neuronal death. Annals of Neurology 53:711–717. 619.
Sullivan PG, Rippy NA, Dorenbos K, Concepcion RC, Agarwal AK, Rho Weinshenker D, Szot P. (2002) The role of catecholamines in seizure
JM. (2004) The ketogenic diet increases mitochondrial uncoupling susceptibility: new results using genetically engineered mice. Phar-
protein levels and activity. Annals of Neurology 55:576–580. macology & Therapeutics 94:213–233.
Szot P, Weinshenker D, Rho JM, Storey TW, Schwartzkroin PA. (2001) Williamson A, Patrylo PR, Pan J, Spencer DD, Hetherington H. (2005)
Norepinephrine is required for the anticonvulsant effect of the keto- Correlations between granule cell physiology and bioenergetics in
genic diet. Brain Research Developmental Brain Research 129:211– human temporal lobe epilepsy. Brain 128:1199–1208.
214. Wu BJ, Hulbert AJ, Storlien LH, Else PL. (2004) Membrane lipids
Tabb K, Szot P, White SS, Liles LC, Weinshenker D. (2004) The ke- and sodium pumps of cattle and crocodiles: an experimental test of
togenic diet does not alter brain expression of orexigenic neuropep- the membrane pacemaker theory of metabolism. American Journal
tides. Epilepsy Research 62:35–39. of Physiology. Regulatory, Integrative and Comparative Physiology
Taha AY, Ryan MA, Cunnane SC. (2005) Despite transient ketosis, the 287:R633–641.
classic high-fat ketogenic diet induces marked changes in fatty acid Xiao Y, Li X. (1999) Polyunsaturated fatty acids modify mouse hip-
metabolism in rats. Metabolism 54:1127–1132. pocampal neuronal excitability during excitotoxic or convulsant
Thavendiranathan P, Mendonca A, Dell C, Likhodii SS, Musa K, Ira- stimulation. Brain Research 846:112–121.
cleous C, Cunnane SC, Burnham WM. (2000) The mct ketogenic Xiao YF, Gomez AM, Morgan JP, Lederer WJ, Leaf A. (1997) Suppres-
diet: effects on animal seizure models. Experimental Neurology sion of voltage-gated l-type ca2+ currents by polyunsaturated fatty
161:696–703. acids in adult and neonatal rat ventricular myocytes. Proceedings of
Thavendiranathan P, Chow C, Cunnane S, McIntyre BW. (2003) The the National Academy of Sciences of the United States of America
effect of the ‘classic’ ketogenic diet on animal seizure models. Brain 94:4182–4187.
Research 959:206–213. Xiao YF, Wright SN, Wang GK, Morgan JP, Leaf A. (1998) Fatty acids
Thio LL, Wong M, Yamada KA. (2000) Ketone bodies do not directly al- suppress voltage-gated Na+ currents in hek293t cells transfected
ter excitatory or inhibitory hippocampal synaptic transmission. Neu- with the alpha-subunit of the human cardiac Na+ channel. Proceed-
rology 54:325–331. ings of the National Academy of Sciences of the United States of
Thio LL, Erbayat-Altay E, Rensing N, Yamada KA. (2006) Leptin con- America 95:2680–2685.
tributes to slower weight gain in juvenile rodents on a ketogenic diet. Xiong ZQ, Stringer JL. (2000) Sodium pump activity, not glial spatial
Pediatric Research. buffering, clears potassium after epileptiform activity induced in the
Thomzig A, Laube G, Pruss H, Veh RW. (2005) Pore-forming subunits dentate gyrus. Journal of Neurophysiology 83:1443–1451.
of k-atp channels, kir6.1 and kir6.2, display prominent differences Yamada K, Ji JJ, Yuan H, Miki T, Sato S, Horimoto N, Shimizu T,
in regional and cellular distribution in the rat brain. The Journal of Seino S, Inagaki N. (2001) Protective role of atp-sensitive potassium
Comparative Neurology 484:313–330. channels in hypoxia-induced generalized seizure. Science 292:1543–
Uhlemann ER, Neims AH. (1972) Anticonvulsant properties of the keto- 1546.

Epilepsia, Vol. 48, No. 1, 2007


58 K. J. BOUGH AND J. M. RHO

Yamashita M, Matsuki A, Oyama T. (1976) General anaesthesia for a metabolism and ketosis. The Journal of Neuroscience Journal of
patient with progressive muscular dystrophy. Anaesthesist 25:76–79. Neuroscience Research 66:272–281.
Yan QS, Jobe PC, Dailey JW. (1993) Noradrenergic mechanisms for Yudkoff M, Daikhin Y, Nissim I, Nissim I. (2004) Epilepsy and the
the anticonvulsant effects of desipramine and yohimbine in geneti- ketogenic diet. In Stafstrom CE, Rho JM (Eds) The ketogenic diet:
cally epilepsy-prone rats: studies with microdialysis. Brain Research interactions with brain amino acid handling. Humana Press, Inc.,
610:24–31. Totowa, NJ, pp. 185–215.
Young C, Gean PW, Chiou LC, Shen YZ. (2000) Docosahexaenoic acid Yudkoff M, Daikhin Y, Nissim I, Horyn O, Lazarow A, Luhovyy B,
inhibits synaptic transmission and epileptiform activity in the rat Wehrli S. (2005) Response of brain amino acid metabolism to ketosis.
hippocampus. Synapse 37:90–94. Neurochemistry International 47:119–128.
Yudkoff M, Daikhin Y, Nissim I, Grunstein R. (1997) Effects of ketone Yuen AW, Sander JW, Fluegel D, Patsalos PN, Bell GS, Johnson T,
bodies on astrocyte amino acid metabolism. Journal of Neurochem- Koepp MJ. (2005) Omega-3 fatty acid supplementation in patients
istry 69:682–692. with chronic epilepsy: a randomized trial. Epilepsy & Behavior
Yudkoff M, Daikhin Y, Nissim I, Lazarow A. (2001) Brain amino acid 7:253–258.

Epilepsia, Vol. 48, No. 1, 2007

Das könnte Ihnen auch gefallen