Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

  • Upload
    l10nass

  • View
    221

  • Download
    0

Embed Size (px)

Citation preview

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    1/16

     Epilepsia, 48(1):43–58, 2007Blackwell 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-ment for medically refractory epilepsy. Despite nearly a centuryof use, the mechanisms underlying its clinical efficacy remainunknown. In this review, we present one intersecting view of howthe KD may exertits anticonvulsantactivityagainst the backdropof several seemingly disparate mechanistic theories. We summa-rize key insights gleaned from experimental and clinical studiesof the KD, and focus particular attention on the role that ketonebodies, fatty acids, and limited glucose may play in seizure con-trol. Chronic ketosis is anticipated to modify the tricarboxcylicacid cycle to increase GABA synthesis in brain, limit reactiveoxygen species (ROS) generation, and boost energy productionin brain tissue. Among several direct neuro-inhibitory actions,

    polyunsaturated fatty acids increased after KD induce the ex-pression of neuronal uncoupling proteins (UCPs), a collectiveup-regulation of numerous energy metabolism genes, and mito-chondrial biogenesis. These effects further limit ROS generationand increase energy production. As a result of limited glucoseand enhanced oxidativephosphorylation,reduced glycolytic fluxis hypothesized to activate metabolic KATP  channels and hyper-polarize neurons and/or glia. Although it is unlikely that a singlemechanism, however well substantiated, will explain all of thediet’s clinical benefits, these diverse, coordinated changes seempoised to stabilize synaptic function and increase the resistanceto seizures throughout the brain.   Key Words:   Ketogenic diet— Epilepsy—Metabolism—Polyunsaturated fatty acids.

    The ketogenic diet (KD) is a high-fat, low-protein, low-

    carbohydrate diet that has been employed as a treatment

    for medically refractory epilepsy for 86 years. The “clas-

    sic” KD is based upon consumption of long-chain satu-rated triglycerides (LCTs) in a 3:1–4:1 ketogenic diet ratio

    (KD ratio) of fats to carbohydrates + protein (by weight).

    The vast majority of calories (>90%) are derived from fat.

    While clinical implementation of the KD has varied from

    center to center (Kossoff and McGrogan, 2005), diet treat-

    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

    of several days, where blood glucose, urine ketones, and

    several other metabolic variables are closely monitored.

    The hallmark feature of KD treatment is the production of 

    ketone bodies by the liver. Ketone bodies provide an al-

    ternative substrate to glucose for energy utilization, and in

    developing brain, also constitute essential building blocks

    for biosynthesis of cell membranes and lipids.

    While the clinical effectiveness of the KD is widely

    accepted, surprisingly little is understood about its under-

    Accepted October 24, 2006.

    Address correspondence and reprint requests to Kristopher J. Boughat FDA – Center for Drug Evaluation & Research, MPN 1 – Room1345, 7520 Standish Place, Rockville, MD 20855, U.S.A. E-mail:[email protected]

    doi:10.1111/j.1528-1167.2007.00915.x

    lying mechanisms of action. Although some studies sug-

    gest that dietary constituents or metabolites have direct

    anticonvulsant effects, emerging evidence indicates that

    adaptations to chronic administration of the KD result inimproved seizure control. These data suggest that the KD

    activates several endogenous metabolic and genetic “pro-

    grams” to stabilize and/or enhance cellular metabolism,

    and that these fundamental changes help counter neuronal

    dysfunction associated with seizure activity.

    MECHANISTIC INSIGHTS FROM STUDIES

    OF KD EFFICACY

    The anticonvulsant efficacy of the KD has been ex-

    amined in various acute and chronic animal models of 

    epilepsy over the years (Stafstrom, 1999). Clinical andexperimental studies have provided key insights into im-

    portant treatment-related variables and, when considered

    together, these studies have helped direct mechanistic re-

    search. Commonalities between clinical and experimen-

    tal studies of efficacy are summarized in Table 1 (adapted

    from Stafstrom, 2004).

    Based on vast clinical experience, almost any diet that

    produces ketonemia and/or diminished blood glucose lev-

    els can induce an anticonvulsant effect. Ketogenic diets

    comprised of either LCTs (Freeman et al., 1998; Vin-

    ing et al., 1998) or medium-chain triglycerides (MCTs;

     43

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    2/16

     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 dietonset

    Infants, children and adolescents 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    3/16

     ANTICONVULSANT ACTIONS OF KETOGENIC DIET 45

    are administered in ratios of ≥3:1 (Freeman et al., 2000),

    and higher KD ratios increased both clinical (Dekaban,

    1966; Livingston, 1972) and experimental anticonvulsant

    efficacy (Bough et al., 2000b). Similar to the KD ratio,

    increasing the extent of CR resulted in improved seizure

    control in epileptic mice (Greene et al., 2001), irrespec-

    tive of the type of diet that was restricted (Eagles et al.,2003). In general, extra calories in the form of carbohy-

    drates or proteins translate to additional metabolic sub-

    strates for gluconeogenesis and diminished KD efficacy.

    Breakthrough seizures are believed to result from overes-

    timation and administration of excess calories (Freeman

    et al., 2000). As such, CR may share common anticonvul-

    sant mechanisms and adjunctively optimize KD efficacy.

    In rodents, maximal seizure control develops 1 – 2 weeks

    after initiation of a KD (Appleton and DeVivo, 1974; Rho

    et al., 1999; Bough et al., 2006). Similarly in humans,

    clinical efficacy does not reach its zenith in many patients

    until after 2 weeks (Dekaban, 1966; Freeman et al., 2000).

    One notable feature of the KD is the rapid occurrence of 

    breakthrough seizures and loss of ketosis when carbohy-

    drates are introduced (e.g., after a child sneaks a cookie;

    Huttenlocher, 1976). As a result, the KD must be strictly

    enforced in order for efficacy to be maintained. However,a

    breakthrough seizure may not necessarily translate to a to-

    tal loss of seizure control. Studies have shown that, despite

    an abrupt discontinuation of the KD, the increased resis-

    tance to seizures waned gradually when switched back to

    control (Bough et al., 2006) or even high-carbohydrate,

    antiketogenic chow (Appleton and DeVivo, 1974). This

    decline in seizure threshold generally occurred over 1 – 2

    weeks, mirroring the onset of seizure protection (Apple-ton and DeVivo, 1974; Bough et al., 2006). This indicates

    that a critical, minimal level of sustained ketosis is neces-

    sary but not sufficient to maintain seizure control. Thus,

    it would seem that metabolic adaptations to KDs underlie

    their key anticonvulsant actions.

    Many studies and anecdotal observations have sug-

    gested that the KD is most effective in immature animals

    or infants and children (Livingston, 1972; Uhlemann and

    Neims, 1972; Otani et al., 1984; Bough et al., 1999b; Rho

    et al., 1999). This is perhaps dueto enhanced metabolicca-

    pacity, more efficient extraction of ketone bodies from the

    blood (Morris, 2005), and/or greater compliance of KDsin the pediatric population. However, a lack of efficacy

    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

    demonstrated to be similarly effective in infants (Nordli

    et al., 2001; Kossoff et al., 2002), adolescents (Kinsman

    et al., 1992; Mady et al., 2003), and adults (Sirven et al.,

    1999; Coppola et al., 2002). Furthermore, experimental

    KDs are effective in both young (i.e.,  

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    4/16

     46 K. J. BOUGH AND J. M. RHO

    glucose restriction might each lead directly or indirectly

    to seizure control. While it is possible that any one of 

    these KD-induced changes is responsible for the anticon-

    vulsant action of the KD, available evidence suggests that

    improved seizure control, at a minimum, likely requires

    all three.

    Role of ketone bodies

    Beta-hydroxybutyrate (BHB)is the predominant ketone

    body measured in the blood, and as such, has been used as

    a clinical measure of KD implementation (Fig. 1). Accord-

    ingly, nearly all KD studies have attempted to establish a

    causative link between ketonemia and anticonvulsant ef-

    ficacy. Although robust elevations in plasma BHB levels

    have been observed during KD treatment (Bough et al.,

    1999b; Thavendiranathan et al., 2000), there is no signif-

    icant correlation between plasma BHB levels and seizure

    protection. Optimal seizure protection generally lags days

    to weeks behind the development of ketonemia, which oc-

    curs within hours of KD onset.

    Nevertheless, there is some evidence that ketones other 

    than BHB may possess anticonvulsant properties. When

    injected into animals, acetone and its parent acetoacetate

    (ACA), prevent acutely provoked seizures. Seminal work

    in the 1930s revealed that acute intraperitoneal admin-

    istration of acetone or ethyl-acetoacetate protected rab-

    bits from thujone-induced seizures (Helmholz and Keith,

    1930; Keith, 1933). Thujone is the active constituent of 

    wormword oil, and is an antagonist of GABAA receptors

    (Hold et al., 2000). More recent experimental studies have

    shown similar results in rodents. Acetone (Likhodii et al.,

    2003) and ACA (Rho et al., 2002) — but not BHB — wereanticonvulsant in a variety of acute and chronic models of 

    epilepsy, consistent with earlier observations (Helmholz

    FIG. 1.   Metabolic pathwayshighlighting 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. FromLikhodii and Burnham (2004).

    and Keith, 1930; Yamashita et al., 1976; Vodickova et al.,

    1995). Clinically, acetone levels of up to 1 millimolar 

    (mM) were detected in the brains of five of seven well-

    controlled epileptic patients following KD using mag-

    netic resonance spectroscopic techniques (Seymour et al.,

    1999).Althoughacetone could notbe detected in twoother 

    seizure-free patients, the authors concluded that acetonecontributes to the anticonvulsant effect of the KD. Inter-

    estingly, the concept that a lipophilic solvent may potently

    block seizure activity is not new. The classic example of 

    this is valproic acid, which was initially used as a solvent

    to dissolve investigational anticonvulsant compounds, but

    was serendipitously discovered to possess intrinsic anti-

    convulsant properties.

    Whereas in vivo pharmacodynamic studies have

    suggested that both ACA and acetone may act as

    anticonvulsant agents, there is no evidence that ketone

    bodies can directly modulate synaptic transmission and/or 

    neuronal excitability. In vitro cellular electrophysiologi-

    cal experiments have failed to demonstrate an effect on the

    principal ion channels that mediate neuronal excitability

    and inhibition. Specifically, neither L-BHB nor ACA were

    found to modulate GABAA  receptors, AMPA receptors,

    or NMDA receptors in both hippocampus and neocortex

    of rats (Thio et al., 2000; Donevan et al., 2003). Despite

    these negative observations, it remained possible that ke-

    tone bodies might affect network activity or synchrony.

    However, in field potential recordings conducted in vitro,

    Thio et al. (2000) demonstrated clearly that neither ACA

    nor BHB modified evoked excitatory postsynaptic poten-

    tials (EPSPs) or population spikes in the CA1 subfield of 

    the hippocampal tissue. In summary, there is no evidencefor direct anticonvulsant effects for either ACA or BHB,

    and acetone has yet to be studied in neuronal (CNS) tissue.

     Epilepsia, Vol. 48, No. 1, 2007 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    5/16

     ANTICONVULSANT ACTIONS OF KETOGENIC DIET 47 

    This may, in large part, reflect the technical difficulties in

    investigating a compound that is highly volatile and can

    react with perfusion systems ordinarily used in pharma-

    cological in vitro experiments.

    Recently, it has been suggested that ACA and/or its

    metabolic byproduct, acetone, may activate a novel class

    of potassium leak channels known as the two-pore do-main or K2P channels (Vamecq et al., 2005). K2P channels

    represent a diverse superfamily of channels that generally

    hyperpolarize cell membranes,and regulate membrane ex-

    citability both pre- and postsynaptically (Lesage, 2003).

    These channels can be modulated by changes in pH, os-

    molality, temperature, mechanical pressure, and certain

    fatty acids (Franksand Honore, 2004).Links between KD-

    induced elevations in ketone bodies (and/or fatty acids, as

    discussed below) and K2P channels, however, have yet to

    be explored.

    In conclusion, although ketone bodies have been shown

    to possess anticonvulsant properties in vivo, there is no ev-

    idence to date that they mediate directly these effects. It

    is clear that some degree of sustained ketosis is required

    for clinical efficacy and that efficacy is maximized over a

    period of weeks versus days, despite a rapid onset of keto-

    sis within hours. Whereas it is plausible that some dietary,

    pharmacokinetic factor(s) results in some level of seizure

    protection, the approximate 2-week time course for opti-

    mal seizure protection suggests a pharmacodynamiceffect

    of the KD (e.g., parallel time course for changes in gene

    expression, mitochondrial proliferation, up-regulation of 

    UCPs/transporters, etc) likely underlies the anticonvulsant

    nature of the diet. Thus, available data suggest that adap-

    tations to, rather than a direct effect of, ketosis underliethe anticonvulsant nature of the KD.

    Role of glucose restriction

    Whereas most studies have suggested that persistent

    ketosis is essential to KD-induced seizure protection, oth-

    ers have posited that glucose restriction is the key fea-

    ture (Greene et al., 2003). In addition to ketosis, it is

    clear that as ketonemia develops, another immediate con-

    sequence of CR and/or KDs is a   ‘moderate’   reduction

    in blood glucose. Does caloric restriction simply act to

    limit gluconeogenic substrates that would otherwise re-duce KD ratio and counter efficacy? Or, might glucose re-

    striction result in another metabolic adaptation that helps

    quell aberrant hyperexcitability? Calorie restriction alone

    was sufficient to retard seizure susceptibility in juvenile

    and adult epileptic   EL   mice; and, blood glucose lev-

    els were inversely correlated with a decreased risk of 

    seizures (Greene et al., 2001). Greeneet al.(2003) hypoth-

    esized that CR reduces energy production through glycol-

    ysis, which limits a neuron’s ability to reach (and main-

    tain) high levels of synaptic activity necessary for seizure

    genesis.

    Others have hypothesized that glucose restriction dur-

    ing KD treatment activates ATP-sensitive potassium

    (KATP) channels (Schwartzkroin, 1999; Vamecq et al.,

    2005). Interestingly, KATP  channels are ligand-gated re-

    ceptors broadly expressed throughout the central nervous

    system, in both neurons and glia (Thomzig et al., 2005).

    These channels act as metabolic sensors, linking cellular membrane excitability to fluctuating levels of ADP and

    ATP. Activation of this receptor by reduced ATP/ADP ra-

    tios opens the channel and leads to membrane hyperpo-

    larization. When glucose is limited (e.g., during adminis-

    tration of a classic KD, which is typically CR by 25%),

    KATP channels might open to hyperpolarize the cell as the

    intracellular ATP concentrations fall. Conversely, when

    glucose is present andATP concentrations rise, KATP chan-

    nels close. As such, KATP  channels may provide a mea-

    sure of protection against a variety of metabolic stressors

    such as hypoxia, ischemia, and hypoglycemia, and are

    believed to regulate seizure threshold (Seino and Miki,

    2003).

    KATP channels are particularly abundant in the substan-

    tia nigra (Hicks et al., 1994), a region of the brain thought

    to act centrally in the propagation of seizure activity

    (Iadarola and Gale, 1982). KATP channels would therefore

    be ideally positioned to metabolically regulate the onset

    of several different seizure types, as does the KD. There is

    growing evidence that KATP  channels may critically reg-

    ulate seizure activity. Genetically engineered mice that

    overexpress the sulfonylurea (SUR) subunit of KATP chan-

    nels were significantly more resistant to seizures induced

    by kainate, and showed no marked cell loss in hippocam-

    pus (Hernandez-Sanchez et al., 2001). Studies of KATPchannel ( Kir6.2−/−) knockout mice suggested that these

    channels help determine seizure threshold (Yamada et al.,

    2001). Following hypoxic challenge (∼5% O2), knock-

    out mice exhibited myoclonic – tonic seizure activity, and,

    ultimately, death compared to controls who all recovered

    without sequelae.

    Despite these observations, there is one important

    caveat in implicating KATP channels as mediators of a KD-

    induced anticonvulsant effect. Other studies have demon-

    strated an increase in energy reserves (specifically, ATP)

    after KD treatment (DeVivo et al., 1978; Pan et al., 1999).

    These data predict that KATP   channels would remainclosed, not open, during diet treatment, and would thus

    contribute to neuronal/glial cell membrane depolarization.

    Nevertheless, several findings are consistent with the no-

    tion that KATP   channels are selectively activated during

    administration of a low-glucose, high-fat KD. First, K ATPchannels are regulated preferentially via glycolytic energy

    sources (Dubinskyet al., 1998). It hasrecentlybeen shown

    that the glycolytic enzyme glyceraldehyde 3-phosphate

    dehydrogenase (GAPDH) serves as an accessory pro-

    tein to KATP channels and regulates directly their activity

    (Dhar-Chowdhury et al., 2005; Jovanovic et al., 2005).

     Epilepsia, Vol. 48, No. 1, 2007 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    6/16

     48 K. J. BOUGH AND J. M. RHO

    The observed reduction in glycolytic processes af-

    ter KD treatment (specifically, the concentration of 

    fructose-1,6-bisphosphate, the key regulatory enzyme

    of glycolysis) is consistent with this notion (DeVivo

    et al., 1978; Puchowicz et al., 2005; Melo et al., 2006).

    Glycolytic flux may be further limited as a consequence

    of elevated ATP (DeVivo et al., 1978; Otani et al., 1984;Pan et al., 1999; Bough et al., 2006) and citrate (Yudkoff 

    et al., 2001) levels on KD treatment; both ATP and citrate

    are feedback inhibitors of glycolysis.

    Second, it is hypothesized that the accumulation of free

    fatty acids over thecourse of KD administration (Dekaban,

    1966) may boost KATP channel activation (Vamecq et al.,

    2005). Whereas PUFAs freely cross the BBB, saturated

    free fatty acids are transported across the BBB via carrier-

    mediated processes (Avellini et al., 1994). Fatty acids

    that intercalate within neuronal cell membranes have been

    shown to interact potently with KATP channels, specifically

    reducing their affinity for (and inhibition by) ATP (Shyng

    and Nichols, 1998). Overall, these findings suggest that

    the unique nature of low-glucose, high-fat KDs promotes

    KATP  channel activation, despite observed enhancements

    in oxidative energy production.

    Recent experiments involving 2-deoxyglucose (2-DG)

    provide further support for a glucose-restriction hypothe-

    sis of KD action. Two-deoxyglucoseis a glucose analogue,

    which inhibits phosphoglucose isomerase and, hence, gly-

    colysis. Stafstrom et al. (2005) reported that the addi-

    tion of 1 mM 2-DG decreased epileptiform burst fre-

    quency to 25 – 80% of baseline in rat hippocampal slices

    exposed to elevated extracellular potassium. More signifi-

    cantly, the same group also showedthat 2-DG (250 mg/kg,i.p) elevated the after-discharge threshold in olfactory

    bulb of perforant-path kindled rats, markedly reduced the

    progression of kindling, and limited the expression of 

    BDNF and its cognate receptor, trkB (Garriga-Canut et al.,

    2006).

    Interestingly, there are a number of anticonvulsant par-

    allels between 2-DG (Stafstrom et al., 2005; Garriga-

    Canut et al., 2006) and KD treatment (Bough et al., 2003).

    First, both 2-DG and KD elevated electrographic seizure

    threshold in vivo; second, both 2-DG and KD potently re-

    tarded the progression of epileptogenesisin kindling mod-

    els of epilepsy in vivo; and, third, both 2-DG (in vitro)and KD (in vivo) diminished measures of hippocampal

    hyperexcitability. These results collectively suggest that

    the anticonvulsant actions of KD may work, in large part,

    via an inhibition of glycolysis. Importantly, because 2-DG

    is fairly well tolerated when administered orally (Pelicano

    et al., 2006), this compound may represent a novel treat-

    ment strategy for epilepsy.

    Role of fatty acids

    Polyunsaturated fatty acids (PUFAs) such as docosa-

    hexanoic acid (DHA, C22:6ω3), arachidonic acid (AA,

    C20:4ω6), or eicosapentanoic acid (EPA, C20:5ω3) are

    believed to affect profoundly cardiovascular function and

    health (Leaf and Kang, 1996; Nordoy, 1999; Leaf et al.,

    2003).In cardiac myocytes, PUFAs inhibited fast, voltage-

    gated sodium channels (Xiao et al., 1998) and L-type cal-

    cium channels (Xiao et al., 1997). Similar findings have

    been observed in neuronal tissue. For example, DHA andEPA diminished neuronal excitability and bursting in hip-

    pocampus (Xiao and Li, 1999).

    It is not surprising then that PUFAs are becoming an in-

    creasingly popular focus of KD research. After KD treat-

    ment, specific PUFAs (i.e., AA and DHA) were found to

    be elevated in both serum (Cunnane et al., 2002; Fraser 

    et al., 2003) and brain (Taha et al., 2005) of patients and

    animals. Importantly, one report documented that the rise

    (or drop) in total fatty acids during KD treatment closely

    paralleled clinical improvement (or loss)of seizure control

    (Dekaban, 1966). An additional study found that dietary

    supplementation with 5 g of (65%) n-3 PUFAs once per 

    day produced a marked reduction in seizure frequency

    and intensity in a few epileptic patients (Schlanger et al.,

    2002). These findings suggest that KD-induced elevations

    in PUFAs such as DHA and/or AA might act directly to

    limit neuronal excitability and dampen seizure activity.

    PUFAs could ultimately block seizure activity in a num-

    ber of ways (Fig. 2). First, PUFAs may inhibit directly ion

    channel activity. Omega-3 (ω-3) PUFAs have been shown

    to: (1) inhibit both voltage-gated Na+ and Ca2+ channels,

    (2) increase the resistance to bursting induced by bicu-

    culline, zero Mg2+, pentylenetetrazole or glutamate, and

    (3) prolong the recovery time from inactivation in hip-

    pocampal neurons (Vreugdenhil et al., 1996; Xiao and Li,1999; Young et al., 2000). Second, in conjunction with ke-

    tone bodies, PUFAs may activate a lipid-sensitive class of 

    K2P potassium channels (Vamecq et al., 2005). And, third,

    PUFAs may enhance the activity of the Na+ /K+-ATPase

    (sodium pump). Elevated  ω-3 and diminished  ω-6 PU-

    FAs levels in plasma membranes significantly increased

    sodium pump function (Wu et al., 2004). These findings

    indicate that elevations in brain levels of PUFAs after KD

    treatment (Taha et al., 2005) could help reduce neuronal

    hyperexcitability via a variety of direct mechanisms.

    Uncoupling proteins

    In addition to their direct actions on neuronal ex-

    citability, PUFAs may also act indirectly to limit ex-

    citotoxicity and neurodegeneration. PUFAs regulate the

    expression of numerous genes in brain via transcription

    factors such as PPARα  (peroxisome proliferator-activated

    receptor-α; Sampath and Ntambi, 2004). Through in-

    duction of PPARα   and its coactivator PGC-1, PUFAs

    induce the expression of mitochondrial uncoupling pro-

    teins (UCPs) and activate these proteins directly as well

    (Jaburek et al., 1999; Diano et al., 2003). Recent evidence

     Epilepsia, Vol. 48, No. 1, 2007 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    7/16

     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-gatedNa+ and Ca2+ channels, activate a lipid-sensitive class of K2P   potassium channels, and enhance the activity of the Na

    + /K+-ATPase tolimit neuronal excitability and dampen seizure activity. Acting indirectly, PUFAs might induce the expression and activity of uncouplingproteins (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

    activity (Garlid et al., 2001).

    Uncoupling proteins are homodimers that span the in-

    ner mitochondrial membrane and allow a proton leak fromthe intermembrane space to the mitochondrial matrix.

    There are three major isoforms that have been identified

    in the brain, UCP2, UCP4 and UCP5 (a.k.a., BMCP-1

    or brain mitochondrial carrier protein-1). UCP proteins

    are increasingly implicated in the regulation of neuronal

    excitability and survival (Andrews et al., 2005). The un-

    coupling effect, albeit of small magnitude, reduces the

    proton-motive force, disassociates or   ‘uncouples’   elec-

    tron transport from ATP production, and indirectly de-

    creases the production of reactive oxygen species (ROS).

    Although it would seem that increased levels of UCP pro-

    teins would diminish cellular energy production, Diano

    et al. (2003) showed that chronic overexpression of UCP2

    in neuronal tissue increased cellular ATP and ADP levels

    by triggering mitochondrial biogenesis. KD appears to do

    the same; that is, studies show that the KD induces UCP

    expression, stimulates mitochondrial biogenesis, and en-

    hances energy production (see also below). Seizures, by

    comparison, increase ROS generation and/or mitochon-

    drial dysfunction, which can lead to neuronal dysfunction

    and excitotoxicity (Layton and Pazdernik, 1999; Kovacs

    et al., 2001; Kovacs et al., 2002; Sullivan et al., 2003).

    Interestingly, UCP2 is up-regulated after seizures (Diano

    et al., 2003). The protective role of UCPs was recently

    highlighted by Sullivan et al. (2003) who demonstrated

    that dietary enhancement of UCP expression and function

    in immature rats protected against kainate-induced exci-totoxicity, most likely by decreasing ROS generation (An-

    drews et al., 2005). Further work demonstrated that mice

    maintained on a high-fat KD demonstrated an increase in

    the hippocampal expression and activity of all three mi-

    tochondrial UCPs and exhibited a significant reduction in

    ROS generation in mitochondria isolated from the same

    brain region (Sullivan et al., 2004). In conjunction with

    reports that ketone bodies potently decrease ROS gen-

    eration (Veech et al., 2001; Veech, 2004), these reports

    suggest that the KD compensates for seizure-induced el-

    evations in ROS generation and neuronal dysfunction to

    provide a neuroprotective effect.

     Energy production

    Polyunsaturated fatty acids additionally regulate the

    transcription of numerous genes linked to energy

    metabolism (Sampath and Ntambi, 2005) through activa-

    tion of PPARα, a scenario inwhich the KDis thought to re-

    program cellular metabolism (Cullingford, 2004). Indeed,

    numerous studies have described changes consistent with

    an enhancement in energy production following KD treat-

    ment. First, microarray expression studies demonstrated

    that KD induces a coordinated up-regulation of several

     Epilepsia, Vol. 48, No. 1, 2007 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    8/16

     50 K. J. BOUGH AND J. M. RHO

    dozenmetabolic genesassociated with oxidative phospho-

    rylation after KD (Noh et al., 2004; Bough et al., 2006).

    Second, KD treatment stimulated mitochondrial biogene-

    sis, resulting in a striking 46% increase in the number of 

    mitochondria in the hilar/dentate gyrus region of rat hip-

    pocampus (Bough et al., 2006). And, third, levels of en-

    ergy metabolites were increased after KD. Brain glycogenand ATP concentrations were boosted throughout rodent

    brain (DeVivo et al., 1978; Otani et al., 1984) andtherewas

    an elevation in the phosphocreatine-to-creatine (PCr:Cr)

    energy-reserve ratio in both animals (Bough et al., 2006)

    and humans (Pan et al., 1999). These findings are consis-

    tent with results that show ketones (4 mM BHB + 1 mM

    ACA) increased hydraulic work by 14% and improved

    energy status in perfused heart tissue (Sato et al., 1995).

    Further, there is an overall increased metabolic efficiency

    (DeVivo et al., 1978; Bough et al., 2006), decreased res-

    piratory quotient (Bough et al., 2000b), and maximal mi-

    tochondrial respiratory rate in rodents following the KD

    (Sullivan et al., 2004). Collectively, these data provide

    compelling evidence that the KD enhances oxidative en-

    ergy production by activating a variety of transcriptional,

    translational, and biochemical mechanisms in a concerted

    fashion.

    Metabolic dysfunction has been identified in regions of 

    hyperexcitability within the brain and is associated with

    several epileptic conditions. Impairment of mitochondrial

    function has been observed in the seizure foci of both hu-

    man and experimental epilepsies (Kunz et al., 2000). Se-

    vere metabolic dysfunction occurred in both human and

    rat hippocampal tissue during periods of heightened neu-

    ronal activity (Kann et al., 2005). Kudin et al. (2002)demonstrated that seizure activity down-regulated mito-

    chondrial enzymes involved in oxidative phosphorylation.

    In an earlier study, the same group demonstrated a specific

    deficiency in complex I activity and mitochondrial ultra-

    structural abnormalities within the hippocampal CA3 re-

    gion of epileptic tissue resected from 57 human patients

    (Kunz et al., 2000). In view of previous studies demon-

    strating impaired oxidative phosphorylation capacity in

    pilocarpine-treated rats (Kudin et al., 2002) and in patients

    with epilepsy (Antozzi et al., 1995; Kunz et al., 2000), a

    KD-induced augmentation in oxidative phosphorylation

    and energy reserves seems likely to counter energetic de-ficiencies in epileptic tissue, making neuronal tissue more

    resilient to aberrant neuronal activity and, in this way, con-

    tributing to the diet’s anticonvulsant actions.

    Stabilized synaptic function

    Intriguing as this argument may be, how exactly would

    enhanced energy reserves lead to stabilized synaptic func-

    tion and diminished seizures? One possibility is via the

    sodium pump. ATP is primarily used to maintain ionic gra-

    dients, especially through actions of the transmembrane

    sodium pump (Hulbert and Else, 2000). Schwartzkroin

    originally hypothesized that KD-induced elevations in

    ATP concentrations might enhance and/or prolong the ac-

    tivation of theNa+ /K+-ATPase , perhaps via an increase in

    the delta-G

    of ATP hydrolysis (Veech et al., 2001; Veech,

    2004). In neurons, increased sodium pump activity might

    hyperpolarize the cell and/or reduce the resting mem-

    brane potential to diminish firing probability. EnhancedNa+ /K+-ATPase function in neurons might also preserve

    normal neuronal functioning and/or delay a pathological

    buildup of high external K+ (Xiong and Stringer, 2000).

    In glia, increased activation of the Na+ /K+-ATPase might

    slow glial depolarization and allow for prolonged uptake

    of extracellular K+ during periods of intense neuronal ac-

    tivity (e.g., high-frequency bursting). Increases in neu-

    ronal and/or glial action of the sodium pump would be

    expected to limit hyperexcitability and increase the resis-

    tance to seizures, as is noted after treatment with KD.

    Although no studies have tested this sodium-pump

    hypothesis directly, a recent report suggests that KD

    tissue is more resistant to metabolic stress. When chal-

    lenged with mild hypoglycemia, synaptic transmission

    within the dentate gyrus was maintained for approxi-

    mately 60% longer in tissue from KD-fed animals com-

    pared to controls (Bough et al., 2006). These data suggest

    that the KD stabilizes synaptic transmission (both excita-

    tory and inhibitory) for prolonged periods of time during

    metabolic stress such as during seizure activity. Hence,

    it seems likely that the KD induces seizure protection

    in part by preventing neuronal dysfunction (diminution

    of ROS/enhancement of energy reserves) and stabilizing

    synaptic transmission (enhancement in energy reserves).

    A role for neurotransmitter systems

    The noradrenergic hypothesis

    One of the more intriguing observations regarding KD

    action involves norepinephrine, its receptors and signaling

    cascades. In general, increases in noradrenergic tone re-

    sult in an anticonvulsant effect. Several lines of evidence

    support this view. Norepinephrine (NE) re-uptake in-

    hibitors can prevent seizure activity in genetically epilepsy

    prone rats (GEPRs; Yan et al., 1993) and pharmacolog-

    ical NE agonists are generally, though not always, an-

    ticonvulsant (Weinshenker and Szot, 2002); damage tothe locus coeruleus — the principal region of the brain

    from which ascending and descending noradrenergic in-

    nervation originates — converts occasional seizures into

    self-sustaining status epilepticus (SSSE) in rats (Giorgi

    et al., 2004); animals are more prone to seizures when NE

    is chemically depleted with reserpine (Weinshenker and

    Szot, 2002); and, interestingly, there are several reports of 

    diminished brain levels of NE in several animal models

    of epilepsy, including GEPRs, kindled animals,  EL  mice,

    seizure-sensitive Mongolian gerbils, and tottering mice

    (Weinshenker and Szot, 2002).

     Epilepsia, Vol. 48, No. 1, 2007 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    9/16

     ANTICONVULSANT ACTIONS OF KETOGENIC DIET 51

    Of significant interest is the observation that mice lack-

    ing the ability to produce NE ( Dbh−/− knockout mice)

    do not exhibit an increased resistance to flurothyl seizures

    when treated with a KD (Szot et al., 2001). These data

    indicate that NE is required for the anticonvulsant effect

    of KD, at least in the flurothyl seizure threshold model.

    Weinshenker and Szot (2002) additionally reported an ap-proximate twofold increase in NE levels in hippocampus

    following a KD, suggesting that KD increases basal re-

    lease of NE. These studies indicate the anticonvulsant ac-

    tion of KD may result in part from an enhancement in

    noradrenergic signaling in the brain.

    If the KD enhances NE release as described above, it

    may also promote the corelease of anticonvulsant orexi-

    genicpeptides such as neuropeptide-Y (NPY)and galanin.

    NPY has been shown to inhibit glutamatergic synaptic

    transmission and epileptogenesis in vitro (Rhim et al.,

    1997; Richichi et al., 2004; Vezzani and Sperk, 2004);

    galanin has been shown to limit SSSE (Saar et al., 2002)

    and diminish both excitatory synaptic transmission and

    ictal activity in vitro (Schlifke et al., 2006). Both neu-

    ropeptides are elevated after calorie restriction. However,

    there was no evidence for enhanced transcription of either 

    of these peptides in the brain after KD treatment, suggest-

    ing that neither NPY nor galanin contribute significantly

    to the anticonvulsant actions of KD (Tabb et al., 2004).

    The GABAergic hypothesis

    One of the more popular hypotheses for KD action in-

    volves γ -aminobutyric acid (GABA), the major inhibitory

    neurotransmitter in the mammalian brain. In general, the

    KD is most effective against seizures evoked by GABAer-gic antagonists. The KD potently inhibits seizures in-

    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 toacetyl-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 ofglutamate to aspartate. Oxaloacetate is then less available as a reactant of the aspartate aminotransferase pathway, which couples theglutamate-aspartate interchange via transamination to the metabolism of glucose through the TCA cycle. Less glutamate is convertedto aspartate and thus, more glutamate is available for synthesis of GABA (3) through glutamic acid decarboxylase (GAD). Adapted fromYudkoff et al. (2004).

    duced by pentylenetetrazole, bicuculline, picrotoxin, and

    γ -butyrolactone. In contrast, the diet demonstrates lit-

    tle if any efficacy in acute seizure models involving ac-

    tivation of ionotropic glutamate receptors (e.g., kainic

    acid), voltage-dependent sodium channels (e.g., maximal

    electroshock [MES]), or glycine receptor inhibition (e.g.,

    strychnine; Bough et al., 2002).Yudkoff et al. (2005) have proposed that ketosis in-

    duces major shifts in brain amino acid handling favoring

    the production of GABA. This results from a reduction

    of aspartate relative to glutamate, the precursor to GABA

    synthesis, and a shift in the equilibrium of the aspartate

    aminotransferase reaction in the ketotic state. As a re-

    sult, there is an increase in glutamic acid decarboxylase

    (GAD) activity and GABA production (Fig. 3). Elevated

    GABA levels would,in turn, be expectedto dampenhyper-

    excitability throughout the brain. Several studies support

    this possibility. First, KD and CR diet treatments both

    increased GAD transcript and protein levels in inferior 

    and superior colliculi, cerebellar and temporal cortex, and

    striatum (the latter, KD only; Cheng et al., 2004). Sec-

    ond, both BHB and ACA increased the rate and extent

    of GABA formation in synaptosomes (Erecinska et al.,

    1996; Yudkoff et al., 1997). And, finally, KD treatment

    in vivo modified amino acid levels in a manner consistent

    with enhanced GABA production (Yudkoff et al., 2001;

    Melo et al., 2006). Although brain levels of glutamate

    and GABA have not been consistently elevated in rodents

    (DeVivo et al., 1978; Al-Mudallal et al., 1996; Yudkoff 

    et al., 2001; Bough et al., 2006), two recent clinical stud-

    ies report significant increases in GABA levels following

    KD treatment (Wang et al., 2003; Dahlin et al., 2005),further substantiating this view.

     Epilepsia, Vol. 48, No. 1, 2007 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    10/16

     52 K. J. BOUGH AND J. M. RHO

    In addition to biochemical measures of KD-enhanced

    GABAergic inhibition, there is functional evidence as

    well. Electrophysiological recordings conducted in vivo

    demonstrated that network excitability was diminished in

    both KD- and calorie-restricted rats (Bough et al., 2003);

    greater stimulus intensities were required to evoke popu-

    lation spikesin both CR-and KD-fed animals compared toad libitum controls. Paired-pulse inhibition was increased.

    Both CR and KD dietary treatments resulted in greater 

    paired-pulse inhibition compared to controls at the 30-

    ms interpulse interval (Bough et al., 2003), a result con-

    sistent with an enhancement in fast, GABAA-mediated

    inhibition. Additionally, KD-fed animals exhibited an el-

    evated electrographic seizure threshold and an increased

    resistance to a modified, 1-day kindling protocol (max-

    imal dentate activation). These data suggested that both

    KD and calorie-restricted diets limited network excitabil-

    ity and elevated seizure threshold via an enhancement of 

    GABAergic inhibition.

    GABAergic interneurons, which at baseline have more

    depolarized resting membrane potentials, endure non-

    accommodating bursts of neuronal firing and must

    metabolically persist (Attwell and Laughlin, 2001), lest

    network inhibition becomes compromised. Previous stud-

    ies have shown that a KD increases total brain [ATP]

    (DeVivo et al., 1978) and PCr/Cr or PCr/ATP energy re-

    serve (Pan et al., 1999; Bough et al., 2006). Accordingly,

    KD-induced elevations in PCr are likely to play a pivotal

    role in maintaining the activity of the Na+ /K+-ATPase

    during periods of intense seizure activity, in both gluta-

    matergic and GABAergic neurons. In a recent study of 

    human temporal lobe epilepsy (Williamson et al., 2005),the PCr/ATP ratio correlated with the recovery of the

    membrane potential following a stimulus train, which

    was inversely correlated with granule cell bursting. Be-

    cause creatine kinase is predominantly localized within

    GABAergic interneurons (Boero et al., 2003), Boero et al.

    concluded that PCr and energy levels are especially crit-

    ical to the maintenance of GABAergic inhibitory output.

    In this manner, a KD-induced increase in energy reserves

    might enhance GABAergic function in particular and im-

    prove seizure control.

    HOW CAN THE KETOGENICDIET BE OPTIMIZED?

    Historically, few guidelines have emerged regarding the

    clinical implementation of the KD and its variants  –   in-

    cluding the medium-chain triglyceride (MCT) formula-

    tion (Huttenlocher et al., 1971) and more recent options

    such as the Atkins diet (Kossoff et al., 2003; Kossoff 

    et al., 2006). This is largely a result of the fact that, until re-

    cently, few KD centers existed throughout the world. Even

    with a resurgence of interest in dietary approaches toward

    epilepsy treatment in the past decade, there remains a no-

    table absence of Class I and II clinical studies. Today, few

    question the clinical efficacy of the KD in both young and

    older patients (Vining, 1999; Coppola et al., 2002; Mady

    et al., 2003), and many successful international centers

    have evolved (Kossoff and McGrogan, 2005; Freeman

    et al., 2006). However, since we do not fundamentally

    know how the KD prevents seizures, there exists as yet norational basis for optimizing the efficacy of the diet, other 

    than through trial and error.

    When examining the accumulated clinical data, it ap-

    pears seizure control can be achieved in the majority

    of epileptic patients as long as there is a shift from

    glycolytic flux to intermediary metabolism (resulting in

    measurable ketosis), irrespective of the precise dietary

    formulation (Henderson et al., 2006). On the other hand,

    the experimental literature suggests that different treat-

    ment protocols may result in differential efficacy or even

    lack of efficacy, despite significant ketosis (Bough et al.,

    2000a; Thavendiranathan et al., 2000; Bough et al., 2002;

    Thavendiranathan et al., 2003). Most of the published

    studies have been based on acute seizure models, and not

    on developmental epilepsy models. Hence, of course, one

    must bear in mind that, despite dozens of animal models

    of the KD, none recapitulate all of the essential features

    in the human epileptic condition (Stafstrom, 1999).

    So how can we reach the goal of developing a safer and

    more effective KD? The reductionist approach posits that

    were we to identify the critical mediator of the diet’s an-

    ticonvulsant effect, administration of this substrate alone

    would likely yield a similar clinical effect as the tradi-

    tional KD, and importantly, spare the patient significant

    side-effects that may preclude its use — even in the face of clear clinical efficacy. The closest we have come to this

    situation is the recent use of BHB as an oral neuroprotec-

    tant. Promising results have already been demonstrated

    in Phase I clinical trials (Smith et al., 2005). Neverthe-

    less, despite increasing experimental evidence that BHB

    and ACA both possess neuroprotective properties (Kashi-

    waya et al., 2000; Noh et al., 2006), a direct anticonvulsant

    effect of ketone bodies has not yet been demonstrated in

    epileptic brain, either animal or human. Intriguing, how-

    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 effectof the principal ketone body, BHB.

    Conversely, if we believe that certain PUFAs, in lieu of 

    ketone bodies, are direct mediators of an anticonvulsant ef-

    fect (Cunnane et al., 2002; Cunnane, 2004), as suggested

    by clinical studies (Schlanger et al., 2002; Fraser et al.,

    2003; Fuehrlein et al., 2004; Yuen et al., 2005), we may be

    closer to distilling the essence of the KD. However, there

    is likely no single fatty acid that is necessary and sufficient

    for an anticonvulsant effect. And experimentally, while it

    has been straightforward to demonstrate the inhibitory ef-

    fects of PUFAs on specific voltage-gated ion channels and

     Epilepsia, Vol. 48, No. 1, 2007 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    11/16

     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 tochronic ketosis and increased concentrations of polyunsaturated fatty acids (PUFAs) in the brain. Chronic ketosis is predicted to leadto increased levels of acetone; this might activate K2P  channels to hyperpolarize neurons and limit neuronal excitability. Chronic ketosisis also anticipated to modify the tricarboxcylic acid (TCA) cycle. This would increase glutamate and, subsequently, GABA synthesis inbrain. Among several direct inhibitory actions (see also Fig. 2), PUFAs boost the activity of brain-speci fic uncoupling proteins (UCPs).This is expected to limit ROS generation, neuronal dysfunction, and resultant neurodegeneration. Acting via the nuclear transcriptionfactor peroxisome proliferator-activated receptor-α  (PPARα), PUFAs would induce the expression of UCPs and coordinately up-regulateseveral 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 KDtreatment could lead to improved seizure control. Ultimately, PUFAs would stimulate mitochondrial biogenesis. Mitochondrial biogenesis ispredicted to increase ATP production capacity and enhance energy reserves, leading to stabilized synaptic function and improved seizurecontrol. 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 glucosecoupled with elevated free fatty acids are proposed to reduce glycolytic  flux during KD, which would further be feedback inhibited by highconcentrations of citrate and ATP produced during KD treatment. This would activate metabolic KATP channels. Opening of KATP channelswould hyperpolarize neurons and diminish neuronal excitability to contribute to the anticonvulsant (and perhaps neuroprotective) actionof the KD. Reduced glucose is also expected to downregulate brain-derived neurotrophic factor (BDNF) and trkB signaling in brain. Asactivation of TrkB pathways by BDNF have been shown to promote hyperexcitability and kindling, these potential KD-induced effects wouldbe 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,

    it is not an easy task to demonstrate that ingestion of a

    specific fatty acid or fatty acid cocktail, acts directly on

    relevant brain receptor targets without first undergoing

    beta-oxidation. The collective data, from both animals and

    humans, indicate that the critical condition necessary for 

    achieving seizure control is a metabolic shift toward fatty

    acid oxidation from glycolysis, reflected in the variable

     Epilepsia, Vol. 48, No. 1, 2007 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    12/16

     54 K. J. BOUGH AND J. M. RHO

    rise in blood/brain ketone levels and a concomitant (mod-

    erate) reduction in blood/brain glucose. Fatty acid compo-

    sition may not ultimately matter, as long as this important

    metabolic shift occurs. And, interestingly, calorie restric-

    tion (Greene et al., 2001; Bough et al., 2003; Eagles et al.,

    2003; Greene et al., 2003) or intake of 2-DG (Stafstrom

    et al., 2005), both of which result in mild hypoglycemia,may be the only requirement for seizure protection, re-

    gardless of whether fats are consumed or not.

    As we continue to explore putative anticonvulsant

    mechanisms of KD action, we are left with many out-

    standing clinical questions regarding dietary treatments

    for epilepsy. Well-designed, multicenter prospective- and

    controlled clinical trials are essential toward developing

    the optimum KD. If woven together with pharmacokinetic

    and pharmacogenetic investigations, these clinical stud-

    ies will not only provide further insights into mechanistic

    underpinnings, but will also help differentiate responders

    from non-responders and identify patients in whom the

    diet is definitively contraindicated. Clinicians would be

    given the tools to make evidence-based decisions rather 

    than rely upon a few case – controlled studies, anecdotal re-

    ports of efficacy, or clinical folklore as has been the prac-

    tice in the past. Toward this end, information regarding the

    impact of pharmacogenetics on epilepsy treatment is now

    beginning to emerge (Depondt and Shorvon, 2006; Spurr,

    2006), although much less is known regarding the genet-

    ically determined variables influencing dietary impact on

    brain function, particularly as it relates to the epileptic

    brain.

    CONCLUSIONS

    After nearly a century of clinical use, we still do not

    know how the KD works. However, much progress in KD

    research has been made in the past decade. Among other 

    factors, current evidence indicates KD optimizes cellular 

    metabolism. Endogenous biochemical and genetic   ‘pro-

    grams’  are switched on in the brain in response to keto-

    sis, glucose restriction, and elevated free fatty acids. This

    unique metabolic state, if maintained, induces a shift away

    from glycolytic energy production (glucose restriction)

    toward the production of energy via oxidative phospho-

    rylation (beta-oxidation of fatty acids and production of ketone bodies). The reduction in glycolytic energy sup-

    ply may activate selectively KATP channels to increase the

    resistance to onset of ictal activity. An increase in oxida-

    tive phosphorylation coupled with an induction of UCPs

    and mitochondrial biogenesis can diminish ROS genera-

    tion and increase energy reserves, both of which would

    be expected to prevent neuronal dysfunction, seizures and

    even neurodegeneration.

    It is improbable that one mechanistic target or medi-

    ator will produce entirely the seizure protection associ-

    ated with the KD. Rather, several factors likely contribute

    mechanistically to this broadly efficacious treatment for 

    epilepsy. The challenge of finding key variables is made

    ever more difficultby the intrinsic complexity of metabolic

    effects and their resultant actions on neurons, glia and on

    the epileptic condition itself. We have reviewed here a

    number of seemingly disparate variables that must be sus-

    tained for a meaningful anticonvulsant effect to be ren-dered. These interrelationships are summarized in Fig. 4.

    The fact that a fundamental modification in diet can have

    such profound, therapeutic effects on neurological disease

    underscores the importance of elucidating mechanisms of 

    KD action. Future studies will no doubt provide unique

    insights into how diet can affect the brain, both in health

    and disease, and likely provide the scientific basis for the

    development of potent new treatment strategies for the

    epilepsies.

    REFERENCES

    Al-Mudallal AS, LaManna JC, Lust WD, Harik SI. (1996) Diet-inducedketosis does not cause cerebral acidosis.  Epilepsia 37:258 – 261.

    Andrews ZB, Diano S, Horvath TL. (2005) Mitochondrial uncouplingproteins in the cns: in support of function and survival.   Nature Re-views Neuroscience 6:829 – 840.

    Antozzi C, Franceschetti S, Filippini G, Barbiroli B, Savoiardo M, Fi-acchino F, Rimoldi M, Lodi R, Zaniol P, Zeviani M. (1995) Epilepsiapartialis continua associated with nadh-coenzyme q reductase defi-ciency. Journal of the Neurological Sciences 129:152 – 161.

    Appleton DB, DeVivo DC. (1974) An animal model for the ketogenicdiet. Epilepsia 15:211 – 227.

    Attwell D, Laughlin SB. (2001) An energy budget for signaling inthe grey matter of the brain.  Journal of Cerebral Blood Flow and  Metabolism 21:1133 – 1145.

    Avellini L, Terracina L, Gaiti A. (1994) Linoleic acid passage throughthe blood-brain barrier and a possible effect of age.  Neurochemical

     Research 19:129 – 133.Boero J, Qin W, Cheng J, Woolsey TA, Strauss AW, Khuchua Z. (2003)

    Restricted neuronal expression of ubiquitous mitochondrial creatinekinase: changing patterns in developmentand withincreasedactivity. Molecular and Cellular Biochemistry  244:69 – 76.

    Bough KJ, Chen RS, Eagles DA. (1999a) Path analysis shows thatincreasing ketogenic ratio, but not beta-hydroxybutarate, elevatesseizure threshold in the rat.  Developmental Neuroscience  21:400 – 406.

    Bough KJ, Eagles DA. (1999) A ketogenic diet increases the resistanceto pentylenetetrazole-induced seizures in the rat.  Epilepsia 40:138 – 143.

    Bough KJ, Valiyil R, Han FT, Eagles DA. (1999b) Seizure resistance

    is dependent upon age and calorie restriction in rats fed a ketogenicdiet. Epilepsy Research  35:21 – 28.

    Bough KJ, Matthews PJ, Eagles DA. (2000a) A ketogenic diet hasdifferent effects upon seizures induced by maximal electroshock

    and by pentylenetetrazole infusion.   Epilepsy Research   38:105 – 114.

    Bough KJ, Yao SG, Eagles DA. (2000b) Higher ketogenic diet ratiosconfer protection from seizures without neurotoxicity. Epilepsy Re-search 38:15 – 25.

    Bough KJ, Gudi K, Han FT, Rathod AH, Eagles DA. (2002) An anti-convulsant profile of the ketogenic diet in the rat. Epilepsy Research50:313 – 325.

    Bough KJ, Schwartzkroin PA, Rho JM. (2003) Calorie restriction and

    ketogenic diet diminish neuronal excitability in rat dentate gyrus invivo. Epilepsia 44:752 – 760.

    Bough KJ,Wetherington J, HasselB, Pare JF,GawrylukJW, Greene JG,Shaw R, Smith Y, Geiger JD, Dingledine RJ. (2006) Mitochondrialbiogenesis in the anticonvulsant mechanism of the ketogenic diet. Annals of Neurology 60:223 – 235.

     Epilepsia, Vol. 48, No. 1, 2007 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    13/16

     ANTICONVULSANT ACTIONS OF KETOGENIC DIET 55

    Cheng CM, Hicks K, Wang J, Eagles DA, Bondy CA. (2004) Caloricrestriction augments brain glutamic acid decarboxylase-65 and -67expression. Journal of Neuroscience Research  77:270 – 276.

    Coppola G, Veggiotti P, Cusmai R, Bertoli S, Cardinali S, Dionisi-ViciC, Elia M, Lispi ML, Sarnelli C, Tagliabue A, Toraldo C, Pascotto A.(2002) The ketogenic diet in children, adolescents and young adultswith refractory epilepsy: an italian multicentric experience. Epilepsy Research 48:221 – 227.

    Cullingford TE. (2004) The ketogenic diet; fatty acids, fatty acid-activated receptors and neurological disorders.   Prostaglandins Leukot Essent Fatty Acids 70:253 – 264.

    Cunnane SC, Musa K, Ryan MA, Whiting S, Fraser DD. (2002) Poten-tial role of polyunsaturates in seizure protection achieved with theketogenic diet.   Prostaglandins Leukot Essent Fatty Acids   67:131 – 135.

    Cunnane SC. (2004) Metabolism of polyunsaturated fatty acids and ke-togenesis: an emerging connection.  Prostaglandins Leukot Essent  Fatty Acids 70:237 – 241.

    Dahlin M, Elfving A, Ungerstedt U, Amark P. (2005) The ketogenicdiet influences the levels of excitatory and inhibitory amino acids

    in the csf in children with refractory epilepsy.   Epilepsy Research64:115 – 125.

    Dekaban AS. (1966) Plasma lipids in epileptic children treated with thehigh fat diet. Archives of Neurology 15:177 – 184.

    Dell CA, Likhodii SS, Musa K, Ryan MA, Burnham WM, CunnaneSC. (2001) Lipid and fatty acid profiles in rats consuming differenthigh-fat ketogenic diets. Lipids 36:373 – 378.

    Depondt C, Shorvon SD. (2006) Genetic association studies in epilepsypharmacogenomics: lessons learnt and potential applications. Phar-macogenomics 7:731 – 745.

    DeVivo DC, Leckie MP, Ferrendelli JS, McDougal DB Jr. (1978)Chronic ketosis and cerebral metabolism.   Annals of Neurology3:331 – 337.

    Dhar-Chowdhury P, Harrell MD, Han SY, Jankowska D, Parachuru L,Morrissey A, Srivastava S, Liu W, Malester B, Yoshida H, CoetzeeWA. (2005) The glycolytic enzymes, glyceraldehyde-3-phosphatedehydrogenase, triose-phosphate isomerase, and pyruvate kinase arecomponents of the k(atp) channel macromolecular complex and reg-ulate its function.  The Journal of Biological Chemistry 280:38464 – 38470.

    Diano S, Matthews RT, Patrylo P, Yang L, Beal MF, Barnstable CJ,

    Horvath TL. (2003) Uncoupling protein 2 prevents neuronal deathincluding that occurring during seizures: a mechanism for precondi-tioning. Endocrinology 144:5014 – 5021.

    Donevan SD, White HS, Anderson GD, Rho JM. (2003) Voltage-dependent block of N-methyl-D-aspartate receptors by the novel an-

    ticonvulsant dibenzylamine, a bioactive constituent of l-(+)-beta-hydroxybutyrate. Epilepsia 44:1274 – 1279.

    Dubinsky WP, Mayorga-Wark O, Schultz SG. (1998) Colocalization of glycolytic enzyme activity and katp channels in basolateral mem-brane of necturus enterocytes.  The American Journal of Physiology275:C1653 – 1659.

    Eagles DA, Boyd SJ, Kotak A, Allan F. (2003) Calorie restriction of ahigh-carbohydrate diet elevates the threshold of ptz-induced seizuresto valuesequalto those seen with a ketogenicdiet. Epilepsy Research54:41 – 52.

    Erecinska M, Nelson D, Daikhin Y, Yudkoff M. (1996) Regulation of gaba level in rat brain synaptosomes: fluxes through enzymes of the

    gaba shunt and effects of glutamate, calcium, and ketone bodies. Journal of Neurochemistry 67:2325 – 2334.

    Franks NP, Honore E. (2004) The trek k2p channels and their role ingeneral anaesthesiaand neuroprotection. Trends in PharmacologicalSciences 25:601 – 608.

    Fraser DD, Whiting S, Andrew RD, Macdonald EA, Musa-Veloso K,Cunnane SC. (2003) Elevated polyunsaturated fatty acids in bloodserum obtained from children on the ketogenic diet.   Neurology60:1026 – 1029.

    Freeman J, Veggiotti P, Lanzi G, Tagliabue A, Perucca E. (2006) Theke-togenic diet:from molecular mechanismsto clinical effects. Epilepsy Research 68:145 – 180.

    Freeman JM,ViningEP,Pillas DJ,Pyzik PL,Casey JC,KellyLM. (1998)The efficacy of the ketogenic diet-1998: a prospective evaluation of 

    intervention in 150 children.  Pediatrics 102:1358 – 1363.

    Freeman JM, Vining EP. (1999) Seizures decrease rapidly after fasting:preliminary studies of the ketogenic diet.  Archives of Pediatrics & Adolescent Medicine  153:946 – 949.

    Freeman JM, Kelley MT, Freeman JB. (2000)   The ketogenic diet: atreatment for epilepsy, 3rd ed. Demos, New York.

    Fuehrlein BS, Rutenberg MS, Silver JN, Warren MW, TheriaqueDW, Duncan GE, Stacpoole PW, Brantly ML. (2004) Differentialmetabolic effects of saturated versus polyunsaturated fats in keto-

    genic diets. The Journal of Clinical Endocrinology and Metabolism89:1641 – 1645.

    GarlidKD, Jaburek M,JezekP. (2001)Mechanism ofuncoupling proteinaction. Biochemical Society Transactions  29:803 – 806.

    Garriga-CanutM, SchoenikeB, QaziR, BergendahlK, DaleyTJ, Pfender RM, Morrison JF, Ockuly J, Stafstrom CE, Sutula T, Roopra A.(2006)2-deoxy-d-glucosereducesepilepsyprogressionby nrsf-ctbp-dependent metabaolic regulationof chromatin structure. Nature Neu-roscience 9:1382 – 1387.

    Giorgi FS, Pizzanelli C, Biagioni F, Murri L, Fornai F. (2004) The roleof norepinephrine in epilepsy: from the bench to the bedside.  Neu-roscience and Biobehavioral Reviews 28:507 – 524.

    Greene AE, Todorova MT, McGowan R, Seyfried TN. (2001) Caloricrestriction inhibits seizure susceptibility in epileptic el mice by re-ducing blood glucose. Epilepsia 42:1371 – 1378.

    Greene AE, Todorova MT, Seyfried TN. (2003) Perspectives on themetabolic management of epilepsy through dietary reduction of glu-cose and elevation of ketone bodies.  Journal of Neurochemistry86:529 – 537.

    Helmholz HF, Keith HM. (1930) Eight years’  experience with the ke-togenic diet in the treatment of epilepsy.  Journal of the American Medical Association  95:707 – 709.

    Henderson CB, Filloux FM, Alder SC, Lyon JL, Caplin DA. (2006)Efficacy of the ketogenic diet as a treatment option for epilepsy:meta-analysis. Journal of Child Neurology  21:193 – 198.

    Hernandez-Sanchez C, Basile AS, Fedorova I, Arima H, StannardB, Fernandez AM, Ito Y, LeRoith D. (2001) Mice transgenicallyoverexpressing sulfonylurea receptor 1 in forebrain resist seizureinduction and excitotoxic neuron death.   Proceedings of the Na-tional Academy of Sciences of the United States of America   98:3549 –  3554.

    Hicks GA, Hudson AL, Henderson G. (1994) Localization of high affin-ity [3H]glibenclamide binding sites within the substantia nigra zona

    reticulata of the rat brain. Neuroscience 61:285 – 292.Hold KM, Sirisoma NS,Ikeda T, Narahashi T, Casida JE. (2000) Alpha-

    thujone (the active component of absinthe): gamma-aminobutyricacid type a receptor modulation and metabolic detoxification. Pro-ceedings of the National Academy of Sciences of the United Statesof America 97:3826 – 3831.

    Hori A, Tandon P, Holmes GL, Stafstrom CE. (1997) Ketogenic diet:effects on expression of kindled seizures and behavior in adult rats. Epilepsia 38:750 – 758.

    Hulbert AJ, Else PL. (2000) Mechanisms underlying the cost of livingin animals. Annual Review of Physiology 62:207 – 235.

    Huttenlocher PR, Wilbourn AJ, Signore JM. (1971) Medium-chaintriglycerides as a therapy for intractable childhood epilepsy.   Neu-rology  21:1097 – 1103.

    Huttenlocher PR. (1976) Ketonemia and seizures: metabolic and an-ticonvulsant effects of two ketogenic diets in childhood epilepsy. Pediatric Research 10:536 – 540.

    Iadarola MJ, Gale K. (1982) Substantia nigra: site of anticonvulsantactivity mediated by gamma-aminobutyric acid.  Science 218:1237 – 1240.

    Jaburek M, Varecha M, Gimeno RE, Dembski M, Jezek P, Zhang M,Burn P, Tartaglia LA, Garlid KD. (1999) Transport function andregulation of mitochondrialuncoupling proteins 2 and3. The Journalof Biological Chemistry 274:26003 – 26007.

    Jovanovic S, Du Q, Crawford RM, Budas GR, Stagljar I, JovanovicA. (2005) Glyceraldehyde 3-phosphate dehydrogenase serves as anaccessory protein of the cardiac sarcolemmal k(atp) channel.  EMBO Reports 6:848 – 852.

    Kann O, Kovacs R, Njunting M, Behrens CJ, Otahal J, Lehmann TN,Gabriel S, Heinemann U. (2005) Metabolic dysfunction during neu-ronal activation in the ex vivo hippocampus from chronic epileptic

    rats and humans. Brain 128:2396 – 2407.

     Epilepsia, Vol. 48, No. 1, 2007 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    14/16

     56 K. J. BOUGH AND J. M. RHO

    Kashiwaya Y, Takeshima T, Mori N, Nakashima K, Clarke K, VeechRL. (2000) D-beta-hydroxybutyrate protects neurons in models of alzheimer ’s and parkinson’s disease.  Proceedings of the National Academy of Sciences of the United States of America   97:5440 – 5444.

    Keith HM. (1933) Factors influencing experimentally produced convul-sions. Archives of Neurology Psychiatry  29:148 – 154.

    Kinsman SL, Vining EP, Quaskey SA, Mellits D, Freeman JM. (1992)

    Efficacyof the ketogenic dietfor intractableseizure disorders:reviewof 58 cases. Epilepsia 33:1132 – 1136.

    Kossoff EH, Pyzik PL, McGrogan JR, Vining EP, Freeman JM. (2002)Efficacy of the ketogenic diet for infantile spasms.   Pediatrics109:780 – 783.

    Kossoff EH, Krauss GL, McGrogan JR, Freeman JM. (2003) Efficacyof the atkins diet as therapy for intractable epilepsy.   Neurology61:1789 – 1791.

    Kossoff EH, McGrogan JR. (2005) Worldwide use of the ketogenic diet. Epilepsia 46:280 – 289.

    Kossoff EH,McGrogan JR,Bluml RM, PillasDJ, RubensteinJE, ViningEP. (2006) A modified atkins diet is effective for the treatment of 

    intractable pediatric epilepsy. Epilepsia 47:421 – 424.Kovacs R, Schuchmann S, Gabriel S, Kardos J, Heinemann U. (2001)

    Ca2+ signalling and changes of mitochondrial function during low-mg2+-induced epileptiform activity in organotypic hippocampalslicecultures.The EuropeanJournal of Neuroscience 13:1311 – 1319.

    Kovacs R, Schuchmann S, Gabriel S, Kann O, Kardos J, Heinemann U.(2002) Free radical-mediated cell damage after experimental statusepilepticus in hippocampal slice cultures.  Journal of Neurophysiol-ogy 88:2909 – 2918.

    Kudin AP, Kudina TA, Seyfried J, Vielhaber S, Beck H, Elger CE, KunzWS. (2002) Seizure-dependent modulation of mitochondrial oxida-tive phosphorylation in rat hippocampus.  The European Journal of  Neuroscience 15:1105 – 1114.

    Kunz WS, Kudin AP, Vielhaber S, Blumcke I, Zuschratter W, SchrammJ, Beck H, Elger CE. (2000) Mitochondrial complex i deficiency inthe epileptic focus of patients with temporal lobe epilepsy.  Annalsof Neurology 48:766 – 773.

    Layton ME, Pazdernik TL. (1999) Reactive oxidant species in piriformcortex extracellular fluid during seizures induced by systemic kainicacid in rats.  Journal of Molecular Neuroscience 13:63 – 68.

    Leaf A, Kang JX. (1996) Prevention of cardiac sudden death by n-3

    fatty acids: a review of the evidence.   Journal of Internal Medicine240:5 – 12.

    Leaf A, Xiao YF, Kang JX, Billman GE. (2003) Prevention of suddencardiac death by n-3 polyunsaturated fatty acids.  Pharmacology &Therapeutics 98:355 – 377.

    Lesage F. (2003) Pharmacology of neuronal background potassiumchannels. Neuropharmacology 44:1 – 7.

    Likhodii S, Burnham WM. (2004) Epilepsy and the ketogenic diet. In-StafstromCE, RhoJM (Eds).The effectsof ketonebodieson neuronalexcitability. Humana Press, Inc., Totowa, NJ, pp. 217 – 228.

    Likhodii SS, Musa K, Mendonca A, Dell C, Burnham WM, CunnaneSC. (2000) Dietary fat, ketosis, and seizure resistance in rats on theketogenic diet. Epilepsia 41:1400 – 1410.

    Likhodii SS, Serbanescu I, Cortez MA, Murphy P, Snead OC, III, Burn-ham WM. (2003) Anticonvulsant properties of acetone, a brain ke-tone elevated by the ketogenic diet.   Annals of Neurology   54:219 – 226.

    LivingstonS. (1972)Comprehensive management of epilepsy in infancy,childhood and adolescence. In Livingston S (Ed)  Dietary treatment of epilepsy. Charles C. Thomas, Springfield, IL, pp. 378 – 405.

    Mady MA,KossoffEH, McGregor AL,WhelessJW, Pyzik PL,FreemanJM. (2003) The ketogenic diet: adolescents can do it, too. Epilepsia44:847 – 851.

    Mahoney AW, Hendricks DG, Bernhard N, Sisson DV. (1983) Fastingand ketogenic diet effects on audiogenic seizures susceptibility of magnesium deficient rats. Pharmacology, Biochemistry, and Behav-ior  18:683 – 687.

    Mak SC, Chi CS, Wan CJ. (1999) Clinical experience of ketogenic dieton children with refractory epilepsy.   Acta Paediatrica Taiwanica40:97 – 100.

    Melo TM,Nehlig A, SonnewaldU. (2006) Neuronal-glial interactionsin

    rats fed a ketogenic diet. Neurochemistry International 48:498 – 507.

    Millichap JG, Jones JD, Rudis BP. (1964) Mechanism of anticonvulsantaction of ketogenic diet.  American Journal of Diseases of Children107:593 – 604.

    Morris AA. (2005) Cerebral ketone body metabolism. Journal of Inher-ited Metabolic Disease 28:109 – 121.

    Muller-Schwarze AB, Tandon P, Liu Z, Yang Y, Holmes GL, StafstromCE. (1999) Ketogenic diet reduces spontaneous seizures and mossyfiber sprouting in thekainicacid model. Neuroreport  10:1517 – 1522.

    Nakazawa M, Kodama S, Matsuo T. (1983) Effects of ketogenic dieton electroconvulsive threshold and brain contents of adenosine nu-cleotides. Brain & Development  5:375 – 380.

    Noh HS, Lee HP, Kim DW, Kang SS, Cho GJ, Rho JM, Choi WS.(2004) A cdna microarray analysis of gene expression profiles in rathippocampus following a ketogenic diet. Brain Research. Molecular  Brain Research 129:80 – 87.

    Noh HS, Hah YS, Nilufar R,HanJ, BongJH,Kang SS, Cho GJ, ChoiWS.(2006)Acetoacetate protects neuronal cellsfrom oxidativeglutamatetoxicity. Journal of Neuroscience Research  83:702 – 709.

    Nordli DR, Jr., Kuroda MM, Carroll J, Koenigsberger DY, Hirsch LJ,Bruner HJ, Seidel WT, De Vivo DC. (2001) Experience with theketogenic diet in infants.   Pediatrics 108:129 – 133.

    Nordoy A. (1999) Dietary fatty acids and coronary heart disease. Lipids34(suppl):S19 – 22.

    Otani K, Yamatodani A, Wada H, Mimaki T, Yabuuchi H. (1984) [Effectof ketogenic diet on the convulsive threshold and brain amino acidand monoamine levels in young mice]. No To Hattatsu 16:196 – 204.

    Pan JW, Bebin EM, Chu WJ, Hetherington HP. (1999) Ketosis andepilepsy: 31p spectroscopic imaging at 4.1 t. Epilepsia 40:703 – 707.

    Pelicano H, Martin DS, Xu RH, Huang P. (2006) Glycolysis inhibitionfor anticancer treatment. Oncogene 25:4633 – 4646.

    Peterson SJ, Tangney CC, Pimentel-Zablah EM, Hjelmgren B, Booth G,Berry-Kravis E. (2005) Changes in growth and seizure reduction inchildren on the ketogenic diet as a treatment for intractable epilepsy. Journal of the American Dietetic Association  105:718 – 725.

    Puchowicz MA, Emancipator DS, Xu K, Magness DL, Ndubuizu OI,Lust WD, LaManna JC. (2005) Adaptation to chronic hypoxia dur-ing diet-induced ketosis.  Advances in Experimental Medicine and  Biology 566:51 – 57.

    Rhim H, Kinney GA, Emmerson PJ, Miller RJ. (1997) Regulation of neurotransmission in the arcuate nucleus of the rat by different neu-ropeptide y receptors. The Journal of Neuroscience  17:2980 – 2989.

    Rho JM, Kim DW, Robbins CA, Anderson GD, Schwartzkroin PA.(1999) Age-dependent differences in flurothyl seizure sensitivityin mice treated with a ketogenic diet.   Epilepsy Research   37:233 – 240.

    Rho JM, Anderson GD, Donevan SD, White HS. (2002) Acetoac-

    etate, acetone, and dibenzylamine (a contaminant in l-(+)-beta-hydroxybutyrate) exhibit direct anticonvulsant actions in vivo. Epilepsia 43:358 – 361.

    Richichi C, LinEJ, Stefanin D, ColellaD, Ravizza T, Grignaschi G, Veg-lianese P, SperkG, DuringMJ, VezzaniA. (2004)Anticonvulsant andantiepileptogenic effects mediated by adeno-associated virus vector neuropeptide y expression in the rat hippocampus.  The Journal of  Neuroscience 24:3051 – 3059.

    Saar K, Mazarati AM, Mahlapuu R, Hallnemo G, Soomets U, Kilk K,Hellberg S, Pooga M, Tolf BR, Shi TS, Hokfelt T, Wasterlain C,Bartfai T, Langel U. (2002) Anticonvulsant activity of a nonpeptidegalanin receptor agonist.  Proceedings of the National Academy of 

    Sciences of the United States of America  99:7136 – 7141.Sampath H, Ntambi JM. (2004) Polyunsaturated fatty acid regulation of 

    gene expression. Nutrition Reviews 62:333 – 339.Sampath H, Ntambi JM. (2005) Polyunsaturated fatty acid regulation of 

    genes of lipid metabolism. Annual Review of Nutrition  25:317 – 340.Sato K, Kashiwaya Y, Keon CA, Tsuchiya N, King MT, Radda GK,

    Chance B, Clarke K, Veech RL. (1995) Insulin, ketone bodies, andmitochondrial energy transduction. The FASEB Journal  9:651 – 658.

    Schlanger S, Shinitzky M, Yam D. (2002) Diet enriched with omega-3 fatty acids alleviates convulsion symptoms in epilepsy patients. Epilepsia 43:103 – 104.

    Schlifke I, Kuteeva E, Hokfelt T, Kokaia M. (2006) Galanin expressedin the excitatory fibers attenuates synaptic strength and generalizedseizures in the piriform cortex of mice.   Experimental Neurology200:398 – 406.

     Epilepsia, Vol. 48, No. 1, 2007 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    15/16

     ANTICONVULSANT ACTIONS OF KETOGENIC DIET 57 

    Schwartz RH, Eaton J, Bower BD,Aynsley-Green A. (1989a)Ketogenicdiets in the treatment of epilepsy: short-term clinical effects.  Devel-opmental Medicine and Child Neurology  31:145 – 151.

    Schwartz RM, Boyes S, Aynsley-Green A. (1989b) Metabolic effects of three ketogenic diets in the treatment of severe epilepsy. Develop-mental Medicine and Child Neurology  31:152 – 160.

    Schwartzkroin PA. (1999) Mechanisms underlying the anti-epileptic ef-ficacy of the ketogenic diet.  Epilepsy Research  37:171 – 180.

    Seino S, Miki T. (2003) Physiological and pathophysiological roles of atp-sensitive k+   channels.  Progress in Biophysics and Molecular  Biology 81:133 – 176.

    Seymour KJ, Bluml S, Sutherling J, Sutherling W, Ross BD. (1999)Identification of cerebral acetone by 1h-mrs in patients with epilepsycontrolled by ketogenic diet. Magma 8:33 – 42.

    Shyng SL, Nichols CG. (1998) Membrane phospholipid control of nu-cleotide sensitivity of katp channels.  Science 282:1138 – 1141.

    Sills MA, Forsythe WI, Haidukewych D, MacDonald A, Robinson M.(1986) The medium chain triglyceride diet and intractable epilepsy. Archives of Disease in Childho od  61:1168 – 1172.

    Sirven J, Whedon B, Caplan D, Liporace J, Glosser D, O’Dwyer J,Sperling MR. (1999) The ketogenic diet for intractable epilepsy in

    adults: preliminary results. Epilepsia 40:1721 – 1726.Smith SL, Heal DJ, Martin KF. (2005) Ktx 0101: a potential metabolic

    approach to cytoprotection in major surgery and neurological disor-ders. CNS Drug Reviews 11:113 – 140.

    Spurr NK. (2006) Pharmacogenetic studies of epilepsy drugs: are wethere yet? Trends in Genetics 22:250 – 252.

    Stafstrom CE. (1999) Animal models of the ketogenic diet: whathave we learned, what can we learn?   Epilepsy Research   37:241 – 259.

    Stafstrom CE. (2004) Dietary approaches to epilepsy treatment: old andnew options on the menu. Epilepsy Currents  4:215 – 222.

    Stafstrom CE, Kriegler SM, Valley MT, Ockuly JC, Roopra AS, Sutula

    TP. (2005) 2-deoxyglucose exerts anticonvulsant and antiepilepticactions in experimental epilepsy models.  Epilepsia 46:268 – 269.

    Su SW, Cilio MR, Sogawa Y, Silveira DC, Holmes GL, Stafstrom CE.(2000)Timingof ketogenic dietinitiationin an experimental epilepsymodel.   Brain Research Developmental Brain Research   125:131 – 138.

    Sullivan PG, Dube C, Dorenbos K, Steward O, Baram TZ. (2003) Mi-tochondrial uncoupling protein-2 protects the immature brain from

    excitotoxic neuronal death. Annals of Neurology  53:711 – 717.SullivanPG,RippyNA, Dorenbos K, Concepcion RC,AgarwalAK, Rho

    JM. (2004) The ketogenic diet increases mitochondrial uncouplingprotein levels and activity.  Annals of Neurology  55:576 – 580.

    Szot P, Weinshenker D, Rho JM, Storey TW, Schwartzkroin PA. (2001)

    Norepinephrine is required for the anticonvulsant effect of the keto-genic diet. Brain Research Developmental Brain Research 129:211 – 214.

    Tabb K, Szot P, White SS, Liles LC, Weinshenker D. (2004) The ke-togenic diet does not alter brain expression of orexigenic neuropep-tides. Epilepsy Research 62:35 – 39.

    Taha AY, Ryan MA, Cunnane SC. (2005) Despite transient ketosis, theclassic high-fat ketogenic diet induces marked changes in fatty acidmetabolism in rats. Metabolism 54:1127 – 1132.

    Thavendiranathan P, Mendonca A, Dell C, Likhodii SS, Musa K, Ira-cleous C, Cunnane SC, Burnham WM. (2000) The mct ketogenicdiet: effects on animal seizure models.   Experimental Neurology

    161:696 – 703.Thavendiranathan P, Chow C, Cunnane S, McIntyre BW. (2003) The

    effect of the ‘classic’ ketogenic diet on animal seizure models.  Brain Research 959:206 – 213.

    Thio LL,WongM, Yamada KA.(2000)Ketone bodies do notdirectly al-ter excitatory or inhibitory hippocampal synaptic transmission. Neu-rology  54:325 – 331.

    Thio LL, Erbayat-Altay E, Rensing N, Yamada KA. (2006) Leptin con-tributes to slower weight gain in juvenile rodentson a ketogenic diet. Pediatric Research.

    Thomzig A, Laube G, Pruss H, Veh RW. (2005) Pore-forming subunitsof k-atp channels, kir6.1 and kir6.2, display prominent differencesin regional and cellular distribution in the rat brain. The Journal of Comparative Neurology 484:313 – 330.

    Uhlemann ER, Neims AH.(1972)Anticonvulsantproperties of theketo-

    genic diet in mice. The Journal of Pharmacology and ExperimentalTherapeutics 180:231 – 238.

    Vamecq J, Vallee L, LesageF, Gressens P, Stables JP.(2005)Antiepilep-tic popular ketogenic diet: emerging twists in an ancient story. Progress in Neurobiology 75:1 – 28.

    Veech RL, Chance B, Kashiwaya Y, Lardy HA, Cahill GF Jr. (2001)Ketone bodies, potential therapeutic uses.  IUBMB Life 51:241 – 247.

    Veech RL. (2004) The therapeutic implications of ketone bodies: the

    effects of ketonebodies in pathologicalconditions: ketosis,ketogenicdiet, redox states, insulin resistance, and mitochondrial metabolism. Prostaglandins Leukot Essent Fatty Acids 70:309 – 319.

    Vezzani A, Moneta D, Conti M, Richichi C, Ravizza T, De Luigi A,De Simoni MG, Sperk G, Andell-Jonsson S, Lundkvist J, IverfeldtK, Bartfai T. (2000) Powerful anticonvulsant action of il-1 receptor antagonist on intracerebral injection and astrocytic overexpressionin mice.  Proceedings of the National Academy of Sciences of theUnited States of America  97:11534 – 11539.

    Vezzani A, Sperk G. (2004) Overexpression of npy and y2 receptors inepileptic brain tissue: an endogenous neuroprotective mechanism intemporal lobe epilepsy? Neuropeptides 38:245 – 252.

    Vining EP, Freeman JM, Ballaban-Gil K, Camfield CS, Camfield PR,

    HolmesGL, Shinnar S, ShumanR, Trevathan E, Wheless JW.(1998)A multicenter study of the efficacy of the ketogenic diet.  Archives of  Neurology 55:1433 – 1437.

    Vining EP. (1999) Clinical efficacy of the ketogenic diet.  Epilepsy Re-search 37:181 – 190.

    Vining EP, Pyzik P, McGrogan J, Hladky H, Anand A, Kriegler S, Free-man JM. (2002) Growth of children on the ketogenic diet.  Develop-mental Medicine and Child Neurology  44:796 – 802.

    Vodickova L, Frantik E, Vodickova A. (1995) Neutrotropic effects andblood levels of solvents at combined exposures: binary mixtures of toluene, o-xylene and acetone in rats and mice.  Central European Journal of Public Health  3:57 – 64.

    Vreugdenhil M, Bruehl C, Voskuyl RA, Kang JX, Leaf A, Wadman WJ.

    (1996) Polyunsaturated fatty acids modulate sodium and calciumcurrents in CA1 neurons.  Proceedings of the National Academy of Sciences of the United States of America  93:12559 – 12563.

    WangZJ,BergqvistC, HunterJV,Jin D,Wang DJ,Wehrli S,ZimmermanRA. (2003) In vivo measurement of brain metabolites using two-dimensional double-quantum mr spectroscopy – exploration of gabalevels in a ketogenic diet. Magnetic Resonance in Medicine 49:615 – 

    619.Weinshenker D, Szot P. (2002) The role of catecholamines in seizure

    susceptibility: new results using genetically engineered mice.  Phar-macology & Therapeutics 94:213 – 233.

    Williamson A, Patrylo PR, Pan J, Spencer DD, Hetherington H. (2005)

    Correlations between granule cell physiology and bioenergetics inhuman temporal lobe epilepsy. Brain 128:1199 – 1208.

    Wu BJ, Hulbert AJ, Storlien LH, Else PL. (2004) Membrane lipidsand sodium pumps of cattle and crocodiles: an experimental test of the membrane pacemaker theory of metabolism. American Journalof Physiology. Regulatory, Integrative and Comparative Physiology287:R633 – 641.

    Xiao Y, Li X. (1999) Polyunsaturated fatty acids modify mouse hip-pocampal neuronal excitability during excitotoxic or convulsantstimulation. Brain Research 846:112 – 121.

    Xiao YF, Gomez AM, Morgan JP, Lederer WJ, Leaf A. (1997) Suppres-sion of voltage-gated l-type ca2+ currents by polyunsaturated fatty

    acids in adult and neonatal rat ventricular myocytes.  Proceedings of the National Academy of Sciences of the United States of America94:4182 – 4187.

    Xiao YF, Wright SN, Wang GK, Morgan JP, Leaf A. (1998) Fatty acidssuppress voltage-gated Na+  currents in hek293t cells transfectedwith the alpha-subunit of the human cardiac Na+ channel. Proceed-ings of the National Academy of Sciences of the United States of  America 95:2680 – 2685.

    Xiong ZQ, Stringer JL. (2000) Sodium pump activity, not glial spatialbuffering, clears potassium after epileptiform activity induced in thedentate gyrus. Journal of Neurophysiology 83:1443 – 1451.

    Yamada K, Ji JJ, Yuan H, Miki T, Sato S, Horimoto N, Shimizu T,Seino S, Inagaki N. (2001) Protective role of atp-sensitive potassiumchannels in hypoxia-induced generalizedseizure. Science 292:1543 – 1546.

     Epilepsia, Vol. 48, No. 1, 2007 

  • 8/18/2019 Bough2007 Anticonvulsant Mechanisms of the Ketogenic Diet

    16/16

     58 K. J. BOUGH AND J. M. RHO

    Yamashita M, Matsuki A, Oyama T. (1976) General anaesthesia for apatient with progressive muscular dystrophy. Anaesthesist 25:76 – 79.

    Yan QS, Jobe PC, Dailey JW. (1993) Noradrenergic mechanisms for the anticonvulsant effects of desipramine and yohimbine in geneti-callyepilepsy-prone rats:studies with microdialysis. Brain Research610:24 – 31.

    Young C, Gean PW, Chiou LC, Shen YZ. (2000) Docosahexaenoic acidinhibits synaptic transmission and epileptiform activity in the rat

    hippocampus. Synapse 37:90 – 94.Yudkoff M, Daikhin Y, Nissim I, Grunstein R. (1997) Effects of ketone

    bodies on astrocyte amino acid metabolism.  Journal of Neurochem-istry 69:682 – 692.

    Yudkoff M, Daikhin Y, Nissim I, Lazarow A. (2001) Brain amino acid

    metabolism and ketosis.   The Journal of Neuroscience Journal of  Neuroscience Research 66:272 – 281.

    Yudkoff M, Daikhin Y, Nissim I, Nissim I. (2004) Epilepsy and theketogenic diet. In Stafstrom CE, Rho JM (Eds) The ketogenic diet:interactions with brain amino acid handling. Humana Press, Inc.,Totowa, NJ, pp. 185 – 215.

    Yudkoff M, Daikhin Y, Nissim I, Horyn O, Lazarow A, Luhovyy B,WehrliS. (2005)Responseof brainamino acidmetabolismto ketosis.

     Neurochemistry International 47:119 – 128.Yuen AW, Sander JW, Fluegel D, Patsalos PN, Bell GS, Johnson T,

    Koepp MJ. (2005) Omega-3 fatty acid supplementation in patientswith chronic epilepsy: a randomized trial.   Epilepsy & Behavior 7:253 – 258.

    E il i V l 48 N 1 2007