You are on page 1of 15

Neurochem Res (2017) 42:35–49

DOI 10.1007/s11064-016-2099-2

ORIGINAL PAPER

β-Hydroxybutyrate in the Brain: One Molecule, Multiple


Mechanisms
Lavanya B. Achanta1,2 · Caroline D. Rae1,2  

Received: 29 September 2016 / Revised: 31 October 2016 / Accepted: 2 November 2016 / Published online: 8 November 2016
© Springer Science+Business Media New York 2016

Abstract β-Hydroxybutyrate (βOHB), a ketone body, the approach elevating plasma βOHB while allowing con-
is oxidised as a brain fuel. Although its contribution to sumption of normal diet. An improved understanding of the
energy metabolism in the healthy brain is minimal, it is mechanisms by which βOHB acts will allow better design
an interesting metabolite which is not only oxidised but of both diet and supplemental interventions.
also has other direct and collateral effects which make it
a molecule of interest for therapeutic purposes. In brain Keywords Ketone body metabolism · Histone
βOHB can be produced in astrocytes from oxidation of deacetylase inhibition: K+ channels · Brain energy
fatty acids or catabolism of amino acids and is metabo- metabolism
lised in the mitochondria of all brain cell types although
uptake across the blood brain barrier is a metabolic con-
trol point. βOHB possesses an intrinsic high heat of com- Introduction
bustion, making it an efficient mitochondrial fuel, where it
can alter the NAD+/NADH and Q/QH2 couples and reduce βHydroxybutyrate, (3-hydroxybutanoic acid; C4H8O3;
production of mitochondrial reactive oxygen species. It βOHB) is a water soluble principal ketone body formed
can directly interact as a signalling molecule influencing reversibly from acetoacetate [1]. It constitutes up to 70%
opening of K+ channels and regulation of Ca2+ channels. of the ketone bodies (βOHB, acetoacetate and their break-
βOHB is an inhibitor of histone deacetylases resulting in down product acetone) synthesized in liver mitochondria
upregulation of genes involved in protection against oxida- from the oxidation of fatty acids derived from adipose
tive stress and regulation of metabolism. It interacts with tissues [2] or human milk [3] and from the conversion of
an inflammasome in immune cells to reduce production of ketogenic amino acids [4]. Unlike acetone, βOHB is a non-
inflammatory cytokines and reduce inflammation. Use of volatile and stable compound that, once synthesized, may
βOHB as an efficient neurotherapeutic relies on increasing be released into the blood stream.
blood βOHB levels so as to encourage entry of βOHB to It is well-accepted that the preferred substrate to be oxi-
the brain. While use of βOHB as a sole therapeutic is cur- dised for brain energy supply is glucose. However, even
rently limited, with employment of a ketogenic diet a more under normal circumstances, brain will metabolise alter-
widely used approach, recent development and testing of native substrates such as monocarboxylates like pyruvate
esterified forms of βOHB have shown great promise, with [5], lactate [6], acetate [7] and βOHB [8]. Most of βOHB
oxidised by the brain is supplied by the liver via the blood-
stream. βOHB is also synthesised endogenously in astro-
* Caroline D. Rae cytes which are the only cell type in the brain capable of
c.rae@unsw.edu.au
oxidising fatty acids [9] and this βOHB can be released and
1
Neuroscience Research Australia, Barker St, Randwick, supplied to other brain cell types as fuel.
NSW 2031, Australia There are several intriguing aspects of βOHB metabo-
2
School of Medical Sciences, The University of New South lism and chemistry that potentially make it a more useful
Wales, Kensington, NSW 2052, Australia substrate than other alternative substrates; indeed some

13
36 Neurochem Res (2017) 42:35–49

have argued that there is likely a benefit to the brain in the very little affinity at all [23]. βOHB is also transported by a
body being mildly ketotic [10]. Here, we review what is sodium-dependent monocarboxylate transporter (SMCT1,
currently known about the biochemistry of βOHB and how SLC5A8) which is located in neurons [24]. Another
it could potentially benefit brain function. sodium-dependent transporter (SMCT2, SLC5A12) has
been identified in astrocytes but its affinity for βOHB has
not yet been established [25]. Taken together, it would seem
Brain Transport of βOHB that neuronal cells are well equipped to take up βOHB, but
that glial cells also have capacity to take up and release
The concentration of βOHB in plasma under healthy, βOHB. Uptake of βOHB has been measured in cultured rat
fasted conditions is relatively low with reference values astrocytes with a reported KM of 6.03 mM [26].
reported ~0.04  mmol/L ranging as high as 0.08  mmol/L Uptake of βOHB is generally a metabolic control point
[11] although this may increase in circumstances such as in the metabolism of βOHB with limited capacity and
fasting or starvation (5–6  mM; [12]), dietary interven- expression of MCT1 under normal dietary circumstances
tion or diabetic ketosis (~25  mM; [13]). In diabetic keto- and low plasma βOHB concentrations. Expression of
sis, the amount of plasma βOHB may increase more than MCT1 and hence uptake of βOHB by the brain is increased
one hundred fold with values in excess of 10  mmol/L significantly by ingestion of a high-fat (ketogenic) diet [27,
reported in the literature [14]. In non-diabetic subjects, a 28] or with fasting. In rats, a ketogenic diet has been shown
three-day fast has been reported to increase plasma levels to increase βOHB influx forty-fold due to increases in cap-
from 0.03 ± 0.04 to 3.15 ± 0.67. The brain concentration of illary density and threefold increase in MCT1 staining with
βOHB in these same subjects increased from 0.05 ± 0.05 a doubling of the number of capillaries staining positive for
to 0.98 ± 0.16  mmol/L across the same time span [15]. In MCT1 and no measurable increase in blood flow [29]. In
adolescent mice a ketogenic diet has been found to increase humans, increasing the plasma concentration of βOHB up
plasma βOHB levels to an average of 1.1 ± 0.2  mM [16]. to 12 times normal levels has been shown to have a linear
The same mice showed a concentration of βOHB in hip- relationship with brain oxidation of D-βOHB up to concen-
pocampal extracellular fluid of only 50.7 ± 5.5 μM indicat- trations of ~2  mmol/L [30]. Similarly, in adult humans, a
ing a βOHB plasma/hippocampal extracellular fluid ratio linear relationship has been demonstrated between plasma
of about 1:25, with the authors calculating an intracellu- acetoacetate concentration and the rate of brain acetoace-
lar βOHB concentration around threefold higher than the tate consumption using positron emission tomography [31].
extracellular concentration [16]. These authors also showed an inverse relationship between
Penetration of βOHB into the brain is carrier dependent. the rate of glucose consumption and the rate of ketone body
As a monocarboxylate, βOHB is a substrate for monocar- (acetoacetate) consumption after induction of mild ketosis
boxylate transporters, members of the SLC16 family which via 4 days of ketogenic diet. Interestingly, there seems to be
are proton symporters found widely throughout the brain no obvious relationship in rat brain between the amount of
[17]. MCT1 (SLC16A1) is a relatively low affinity trans- energy able to be extracted from ketones following starva-
porter located mainly in the blood brain barrier with lower tion and pre-starvation glucose utilisation rates [32].
levels in astrocytes [18]. MCT2 (SLC16A7) is a higher MCT1 blood brain barrier expression is also age-
affinity transporter mostly located in neurons in the post dependent, being high in neonates and decreasing with
synaptic density [19]. MCT4 (SLC16A3) is a low affinity age [33, 34]. A “developmental switch” has been identi-
transporter localised exclusively to astrocytes [20]. Each fied, where the brain shifts from metabolising fatty acids,
of these transporters display different affinities for βOHB ketones and monocarboxylates, to metabolising glucose
(Table  1) with the neuronally located MCT2 having the [35], In neonatal rats this is accompanied by a decrease in
highest affinity for βOHB [21] and displaying little if any the vascular expression of MCT1 in the brain, an increase
stereospecificity for D-βOHB over L-βOHB [18]. MCT1 in the blood brain barrier expression of the glucose trans-
has much less affinity for βOHB [22] while MCT4 has porter GLUT1 and an increase in the non-vascular brain

Table 1 Monocarboxylate Transporter D-βOHB (mM) L-βOHB (mM) Acetoacetate (mM)


transporter (MCT) affinities for
βOHB and acetoacetate MCT1 [22] 10.1 ± 0.53 11.4 ± 0.82 5.5 [140]
MCT2 [21] 1.2 ± 0.2 1.2 ± 0.2a 0.8 ± 0.1
MCT4 [23] 130 ± 96 824 ± 64 216 ± 27
SMCT1(SLC5A8) [24] 1.442 ± 0.124 2.33 ± 0.17 0.213 ± 0.039
a
DL-βOHB used in this work

13
Neurochem Res (2017) 42:35–49 37

expression of GLUT3. Expression of non-vascular brain mitochondrial NAD+/NADH ratio such that introduction
monocarboxylate transporters MCT1 and MCT2 is not of βOHB has the potential to shift this ratio. The intercon-
altered by development [35]. version of βOHB and acetoacetate is rapid [43] making it
Expression of MCT1 in the blood brain barrier shows difficult to study the effects of one without considering the
significant species difference, being much lower in adult other.
monkey (0.834 ± 0.368 fmol/μg protein) than in adult BDH is subject to lysine acetylation, a process whereby
mouse (23.7 ± 0.87 fmol/μg protein) [33] and similarly proteins can be modified post-translation and their activity
lower in human than in mouse (2.27 ± 0.85 vs 23.7 ± 1.6 may possibly be altered as a result. It is known that BDH
fmol/μg protein, respectively); [36]. acetylation can be altered by SIRT3 (silent information reg-
The question of how βOHB enters the mitochondrion, ulator 3) as SIRT3 knockout mice have altered BHD acety-
the site at which it is most likely metabolised, is less set- lation [44, 45] but it is not known what the effect of acety-
tled. βOHB is poor substrate for the mitochondrial pyru- lation or de-acetylation may have on the enzyme as this has
vate carrier (KM = 5.6  mM; [37]) although its metabolite not yet been examined.
acetoacetate is carried with reasonable affinity (0.56  mM; A cytosolic form of BDH has been reported in humans;
[37]). Under concentrations found in  vivo it is unlikely DHRS6, a short form of dehydrogenase with structural
that this carrier contributes significantly to βOHB uptake. homologies to bacterial hydroxybutyrate dehydrogenases.
βOHB is taken up into mitochondria, by a mechanism dis- DHRS6 substrate screening studies showed sole NAD+-
tinct from that of pyruvate, with reasonably high affinity. dependent conversion of (R)-β-hydroxybutyrate to ace-
Uptake is reported to be 50% saturated at 0.9 mM and capa- toacetate with a low affinity KM around 10 mM [46]. This
ble of accumulating high concentrations of βOHB inside enzyme is sometimes designated BHB2 and has been
the mitochondrion, suggestive of active uptake [38, 39]. reported as elevated in malignant gliomas [47] but little
is known yet about its expression or role in healthy brain
tissue.
The Chemistry of β-Hydroxybutyrate
Pathways for Conversion of Acetoacetate to Acetyl-CoA
βOHB has an inherently high heat of combustion compared
with the end product of glycolysis, pyruvate, for exam- There are several enzymes in mammalian systems catalys-
ple, producing 243.6  kcal/mol of C2 units compared with ing the interconversion of acetoacetate and acetyl-CoA. In
185.7  kcal/mol in the case of pyruvate; this derives from liver (Fig.  1), acetoacetate can be converted from acetyl-
the fact that βOHB is more reduced than pyruvate [40]. In CoA through the combined activity of hydroxymethylglu-
rat heart, addition of a physiological ratio of ketone bod- taryl-CoA synthase (E.C. 2.3.3.10) and 3-hydroxy-3-meth-
ies resulted in an improvement in the mitochondrial redox ylglutaryl-CoA lyase (HMG-CoA lyase; E.C. 4.3.1.4).
potential of the half reaction (Eh) NAD+/NADH couple These enzymes are key in the production of ketone bodies
from −280 to −300 mV and an increase in the Coenzyme from fatty acids. HMG-CoA is also an intermediate in the
Q couple EhQ/QH2 from −4 to +12 mV indicating that the breakdown pathway of branched chain amino acids such as
chemistry of ketone bodies themselves is capable of driving leucine. Ketone bodies can be produced from leucine by
mitochondrial efficiency ([41]). glial cells, showing an overlap between ketone body metab-
olism and metabolism of branched chain amino acids [48].
In tissues that produce no or little fatty acids, these
Oxidation of βOHB to Acetyl-CoA enzymes may be silenced by alternative slicing mechanisms
that produce inactive variants of both enzymes [49]. In
β-Hydroybutyrate Dehydrogenase Catalyses the First brain and other tissue which do not produce ketone bodies
Step in βOHB Oxidation in any appreciable amounts under normal circumstances,
the conversion of acetoacetate to acetyl-CoA is catalysed
Once βOHB has entered the brain, it is first oxidised by the mitochondrially located succinyl-CoA: 3-oxoacid-
by the mitochondrial enzyme β-hydroxybutyrate dehy- CoA transferase (SCOT; E.C. 2.3.8.5) and acetylacetylCoA
drogenase (BDH, E.C. 1.1.1.30) in a reaction requir- thiolase (E.C. 2.3.1.9) which has both mitochondrial (T2)
ing NAD+, producing acetoacetate and NADH (Fig.  1). and cytosolic (CT) forms [50] (Fig. 1).
D-β-Hydroxybutyrate is the naturally occurring isoform A cytosolic pathway for the direct activation of ace-
and the substrate for BDH in the brain; the inactive L-β- toacetate exists in brain [51], in the reaction catalysed by
hydroxybutyrate isomer does not support mitochondrial acetoacetylCo-A synthetase (AACS; E.C. 6.2.1.16). This
respiration [42]. This means that the two ketone bodies, reaction requires ATP and produces AMP and PPi [52].
βOHB and acetoacetate are in near equilibrium with the The enzyme is highly expressed in the brain of humans

13
38 Neurochem Res (2017) 42:35–49

Fig. 1 Pathways for synthesis and catabolism of ketone bodies in ketogenic and ketone body metabolising tissues

and rats and enriched in neuronal-like cells in the cortical Similar issues are present in 3-hydroxy-3-methylglutaryl
and hippocampal regions [53] where its expression patterns coenzyme A (HMG CoA) lyase deficiency where leucine
are different to that of SCOT [51], suggesting that the two metabolism and ketone body production are deranged.
pathways play different but complementary roles. Life-threatening episodes include hypoglycaemia, acidosis
and hyperammonemia but not ketosis [58]. The occurrence
of this disorder is similarly rare to that of SCOT deficiency
βOHB Metabolising Enzyme Deficiencies have Similar with around 18 patients identified in 1988 [58]. Brain
Outcomes and Therapies imaging studies of these patients show diffuse white mat-
ter abnormalities with more focal lesions reported related
There have been case reports of patients with hereditary to the degree of metabolic derangement experienced by the
succinyl-CoA: 3-oxoacid-CoA transferase (SCOT) defi- patient [59, 60].
ciency, first described in 1972 [54], which is characterised Mitochondrial acetoacetyl-CoA thiolase (T2) deficiency
by intermittent ketoacidotic attacks and permanent hyperke- has also been reported in a similar number of patients and
tonaemia [55]. The gene encoding SCOT occupies a large again, the outcomes are good if the metabolic derange-
100 kb region on chromosome 5p13 [56] and deficiencies ment is detected early and managed appropriately [61].
are relatively rare: fewer than 30 affected individuals were T2 is generally co-expressed in tissues along with SCOT
reported in 2011 [55]. Despite the ketotic episodes, the out- although the SCOT/T2 ratio is higher in brain than any
come is generally good. Hyperketonaemia is eliminated by other tissue [50]. A case of deficiency in the cytosolic form
restricting leucine intake and providing intravenous glu- of acetoacetyl-CoA thiolase (CT) has also been reported
cose. Less than optimal outcomes can be eliminated by fast associated with mental retardation, which was subsequently
diagnosis and prevention of hypoglycaemia [57]. stabilised with diet [62].

13
Neurochem Res (2017) 42:35–49 39

Metabolism and Metabolic Effects of βOHB Neurons, Astrocytes and Oligodendrocytes all


Metabolise Ketone Bodies
Compared to other alternative substrates for brain, such
as lactate, pyruvate and acetate, less is known about Edmond et al. studied the capacity of brain cells in culture
metabolism of βOHB. It was established in the 1960s that to use glucose or ketone bodies for respiration [9]. They
brain switches to metabolism of ketone bodies, includ- found that all cell types use βOHB in respiration but that
ing βOHB, during fasting, when glucose is in short sup- neurons and oligodendrocytes use βOHB about threefold
ply and the body resorts to metabolism of fat and pro- more efficiently than astrocytes, as do synaptic terminals
tein [12]. It was also noted that ketone body oxidation [73]. Perhaps surprisingly, investigation of the activities of
was greater in suckling than in adult rats [63] and Louis the three major ketone body metabolising enzymes, BDH,
Sokoloff noted in 1973 that the previous idea of the brain SCOT and T2 (Fig.  1) have been found to be highest in
as being exclusively restricted to metabolism of glucose astrocytes although each of the cell types is quite capable
must be reconsidered, in view of the fact that “under of using ketone bodies as substrate [74].
certain conditions and during certain periods of life the In contrast to acetate, where metabolism gives rise to a
brain can and does rely, at least in part, on other sub- higher specific activity in glutamine compared to glutamate
strates, particularly ketone bodies” [64]. Indeed ketones due to metabolic compartmentation of its metabolism [75,
can comprise as much as 60–70% of brain energy needs 76], metabolism of βOHB has been shown to give higher
during prolonged fasting and starvation [65]. specific activity in glutamate compared to glutamine [77].
Ketones are preferred over glucose in the developing Modelling of the relative distribution of label from gluta-
brain. Newborn rats incorporated significantly more radi- mate and glutamine in human brain has given rise to the
oactivity from [3-14C]βOHB into glutamate and aspartate suggestion that metabolism of βOHB is predominantly neu-
than from [2-14C]glucose [66]. It has been suggested [67] ronal [78]. This contrasts with the determination by others
that developing brain preferentially uses ketones to spare in rat brain that βOHB is more preferentially used in glial
glucose for metabolism in the pentose phosphate path- cells than neurons [8].
way, for example. The pentose phosphate pathway leads It is not immediately clear why these differences in cel-
to the biosynthesis of ribose for DNA and to the produc- lular preference may have been found. The infusion time
tion of NADPH for lipid biosynthesis, both of which are (2  h) was identical between the work of Pan and that of
crucial in brain development. The activities of BDH and Künnecke, with Pan et  al. infusing a higher concentration
glucose-6-phosphate dehydrogenase, the enzyme which for the first 20  min then a steady, lower level for the rest
generates NADPH [68], peak in rats around postnatal of the time, while Künnecke et al. used a single, constantly
day 18 and 21, respectively [69]. The changes in ketone infused concentration. The two papers also used different
body metabolism during development in are detailed in metabolic models and made different assumptions about
the excellent review from the laboratory of Astrid Nehlig pool sizes.
[70].
The switch to metabolism of ketones over glucose Enzyme Activities Vary with Age and Across Different
under conditions of starvation, or in switching to a Species
ketogenic diet, requires modification of transporters and
enzyme activities. Ketogenic diets and their effects on In the rat, there are quite large developmental changes in
brain metabolism and brain disorders have been reviewed enzyme activity, with this effect being largest in astrocytes,
extensively by others (e.g. [71, 72]) and are not the sub- which lose SCOT and T2 activity appreciably across the
ject of this review. 15  days since birth. BDH activity remains relatively con-
Examination of metabolism of βOHB under “normal” stant from birth to adulthood [74] but has been reported
conditions in brain can be a very informative and useful to decrease in adulthood with aging [79]. The activities
tool for researchers interested in metabolic compartmen- of these enzymes also varies appreciably between spe-
tation and metabolite interaction within the brain. βOHB, cies, with quite high levels of activity reported in rodents
along with competing substrates, or analysis of metabolism such as mice and rats, with appreciably (ten-fold) lower
of labelled βOHB (either radio-labelled or stable isotope) SCOT activity in guinea pig and sheep. These authors
has been employed successfully to give us unique insights also reported that the activities of these enzymes were not
into mitochondrial and cellular processes. From the first altered in rats by 48  h of starvation or alloxan diabetes
observation that respiration rates were altered in brain [80]. Levels of the three enzymes are lower in human brain
when metabolising βOHB it was apparent that metabolism than in rat and there is little reported change with devel-
of this ketone body was sufficiently different to that of glu- opment between 32 week gestation foetus and 70 year old
cose to warrant further attention. adults [81], although regional differences in the ability to

13
40 Neurochem Res (2017) 42:35–49

metabolise acetoacetate have been demonstrated to vary D-βOHB. L-βOHB competes with D-βOHB for uptake via
with age in humans [82]. Page and Williamson [81] con- the various MCTs (Table 1).
clude that there is sufficient enzyme activity in human As detailed above, βOHB is directly metabolised in
brain to account for consumption of ketone bodies, what- mitochondria, being used to produce acetyl-CoA for oxi-
ever their concentration in the blood. This is also sup- dation in the Krebs cycle for the ultimate production of
ported in humans by tracer studies showing that transport ATP. Glucose, on the other hand, generates cytosolic ATP
of βOHB across the blood brain barrier is the limiting fac- via substrate level phosphorylation, before also providing
tor, with βOHB utilisation found to increase almost linearly carbon units for mitochondrial oxidative phosphorylation.
with concentration, with the rate of use higher in gray than It has been suggested that there is compartmentalisation of
in white matter [83]. ATP in cells, with the cytosolic component being used to
power ion channels, such as the Na+/K+-ATPase or KATP
βOHB Metabolism Varies with Brain Region channel activity [88–90]. Neither glycolysis nor oxidative
phosphorylation alone are adequate to power ion gradients
A study injecting 3-14C βOHB intravenously into male in the brain [90].
Sprague–Dawley rats, followed by freezing and sectioning In keeping with this, the contribution of βOHB to basal
of the brain after 10 min, showed regional variation in the (housekeeping) needs in the brain vs fuelling neuronal
uptake of βOHB with the lower layers of the cortex show- activity has been estimated after 36  h of fasting in rats.
ing greater uptake than the upper layers. The striatum and Provided the blood concentration of βOHB is high enough,
globus pallidus showed low uptake while the area of the βOHB is capable of supplying all basal requirements and
hypothalamic arcuate nucleus-median eminence took up around half of the energy needs of neuronal activity [91].
large amounts of label, with the pituitary and pineal gland
taking up the most [84]. βOHB Metabolism Alters Glycolytic Activity,
BDH expression has been shown to vary across the Mitochondrial NAD+/NADH and Malate Aspartate
brain. Its expression in cerebellum has been reported to Shuttle Activity
be patchy with intense staining in Purkinje cells and den-
drites postnatally, with less prominent staining here and in βOHB has been shown to alter the activity of pyruvate
the surrounding molecular layer later in life, while the extra dehydrogenase through conversion of the enzyme to the
germinal layer of the cerebellum did not show appreciable inactive form [92] and to inhibit glycolysis through an
staining [85]. indirect mechanism at the level of phosphofructokinase
The cerebral metabolic rate of acetoacetate (CMRa) has [93]. When βOHB was added to synaptosomes metabo-
recently been measured using positron emission tomogra- lising [6-14C]-glucose the rate of 14CO2 production was
phy (PET) in humans and found to be reasonably constant reduced by ~35% but when βOHB was added to mitochon-
across brain regions, with the lowest rate in hippocam- dria this rate increased slightly by 12% [94]. In the reverse
pus (CMRa = 0.4 ± 0.0  μmol/100  g/min) and the high- case, when [3-14C]-3-hydroxybutyrate was incubated with
est rate in regions of the parietal and occipital cortices mitochondria and synaptosomes and glucose was added
(CMRa = 0.9 ± 0.1  μmol/100  g/min). These rates increase (5 mM), production of 14CO2 was reduced by 65 and 70%,
appreciably after four days on a ketogenic diet, rising to respectively. One possible explanation for this may lie in
3.3 ± 0.3  μmol/100  g/min in hippocampus and as high as the alteration of mitochondrial NAD+/NADH ratio due
6.5 ± 0.7 in frontal pole and to 6.2–6.4 μmol/100 g/min in to the 2  h of pre-incubation with βOHB. Recently, it has
parietal and occipital regions [31]. been demonstrated that the NAD+/NADH ratio is shifted
in cortical neurons preincubated for two hours with 8 mM
βOHB can Supply Baseline Energy Needs but may not βOHB [95]. The increased mitochondrial respiration drives
Support Synaptic Activity changes in BDNF expression through increased genera-
tion of reactive oxygen species, activation of NF-ĸB tran-
Early experiments suggested that, in Guinea pig hippocam- scription factor and activity of the histone acetyltransferase
pal slices, βOHB was able to support maintenance of high p300/EP300 [95].
energy phosphate levels, but unable to support synaptic Effects of βOHB on metabolism are seen through the
potentials [86]. This was also shown in the rat, although increase in acetyl-CoA levels which rise by 15- to 18-fold
there was an age-dependent effect, with βOHB able to sup- as a result of addition of ketone bodies such as βOHB or
port ~90% maintenance of synaptic potential compared acetoacetate to cells [41]. The direct consequence of this
to initial values in the p4 rat and ~20% in the p7 rat [87]. is a reduction in levels of “free” Coenzyme A and an effect
The interpretation of this work is complicated by the use of on the steady-state equilibrium of 2-oxoglutarate dehy-
DL-βOHB rather than the stereoscopically preferred isomer drogenase, which requires unesterified CoA as a cofactor

13
Neurochem Res (2017) 42:35–49 41

(Fig.  2); levels of 2-oxoglutarate rise while those of suc- cycle via succinic semialdehyde dehydrogenase, thus main-
cinate and oxaloacetate fall. The changes in 2-oxoglutarate taining levels of oxaloacetate and hence aspartate [97].
and oxaloacetate then result in concomitant increased glu- Ketone bodies such as βOHB have been reported to not
tamate and reduced aspartate levels. In compartments with be able to be used to synthesise pyruvate [99] but authors
glutamate decarboxylase this also results in a rise in GABA have reported pyruvate recycling from βOHB [8] with
levels due to stimulation of GAD by increased glutamate pyruvate recycling capability increasing after hypoglycae-
[96] (Fig. 2). mia [100]. This has important implications for anaplerosis
In astrocytes, the presence of βOHB (5  mM) in the in the Krebs cycle and for the ability to synthesise Krebs
absence of glucose with 15N-glutamate as substrate has cycle by-products, such as the neurotransmitters glutamate
been shown to result in decreased transamination of gluta- and GABA, as well as aspartate.
mate to aspartate, as well as a reduction in aspartate levels A reduction in the synthesis of neurotransmitter gluta-
[97]. By contrast, this reduction in aspartate levels is not mate has been reported in cultured mouse glutamatergic
seen when 15N-GABA is used as substrate. βOHB has no neurons, with significantly lower levels of glutamate and
measureable direct effect on aspartate aminotransferase so correspondingly higher levels of aspartate present when
the authors suggested that the effect was due to diversion superfused in the presence of 1  mM βOHB compared to
of oxaloacetate away from transamination and towards syn- 1 mM glucose [101]. Adding 1 mM βOHB to the neurons
thesis of citrate by combination with acetyl-CoA (which along with 1  mM glucose somewhat counterintuitively
would be elevated in the presence of βOHB). The authors resulted in further increases in glutamate and further reduc-
noted elevation in astrocytic citrate levels when βOHB was tions in aspartate. Depolarising the neurons with 300  μM
supplied. Secondarily, they noted that glutamine synthetase NMDA, 10  μM glycine and 15  mM K+ had no effect on
is inhibited by citrate [97] as are both the cytosolic and glutamate or aspartate levels in the presence of 1  mM
mitochondrial forms of malic enzyme [98]. GABA could βOHB while significant reductions in glutamate and total
reasonably be expected to supply the section of the Krebs glutamate + aspartate were seen under depolarisation in the

Fig. 2 βOHB metabolism in brain. Metabolism of βOHB by BDH glycolysis is decreased. This, in turn, reduces the exchange of malate
(βOHB dehydrogenase) produces NADH inside the mitochondria and α-ketoglutarate across the mitochondrial membrane, result-
while the second step of its metabolism catalysed by SCOT (succinyl- ing potentially in lower cytosolic α-ketoglutarate levels and reduced
CoA: 3-oxoacid-CoA transferase) produces succinate. Together, these ability to synthesise glutamate and aspartate via cytosolic aspartate
have the potential to alter mitochondrial redox potentials through aminotransferase. AcAc acetoacetate, CoASH Coenzyme-A, OAA
shifting the NAD+/NADH and Q/QH2 couples. βOHB metabolism oxaloacetate, AcAcCoA acetylacetoCo-A, MPC mitochondrial pyru-
also reduces glycolysis, thereby reducing the flux through cytosolic vate carrier, MCT1 monocarboxylate transporter 1
malate dehydrogenase as the drive to regenerate NAD+ is less when

13
42 Neurochem Res (2017) 42:35–49

presence of 1  mM glucose. The failure to increase βOHB also supported in purified mitochondria from G93A-SOD1
metabolism under depolarising conditions was attributed transgenic mice, a model of amyotrophic lateral sclerosis,
to increased mitochondrial Ca2+ under depolarising con- where βOHB was protective in maintaining ATP produc-
ditions with concomitant effects on NAD+/NADH and tion against a rotenone (Complex I inhibitor) challenge but
therefore on BDH activity [101]. Failure to increase βOHB not against a complex II inhibitor, malonate [106].
metabolism under acoustic activation has also been demon- Neurons treated with MPP+ (1-methyl-4-phenylpyridin-
strated using βOHB tracer [102]. ium) which produces a Parkinson’s disease like syndrome,
There has been some speculation as to the role of the or with Aβ1-42, a fragment of amyloid protein which is
malate aspartate shuttle when glycolysis is reduced during lethal to hippocampal cells and produces an Alzheimer’s
ketone metabolism. The role of this shuttle is the transfer like condition, have been shown to be protected from
of reducing equivalents between the cytosol and mitochon- these toxic effects by pre-treatment with βOHB [107]. The
drion and it is required because the inner mitochondrial authors suggest that this effect is due to alteration in the
membrane is impermeable to NADH and NAD+. In the Q/QH2 couple, with resultant reduction in the capacity of
presence of βOHB, glycolysis is reduced resulting in less Complex II to produce reactive oxygen species while at the
requirement to transfer NADH to the mitochondrion, and same time relieving the product inhibition of Complex I.
mitochondria are able to regenerate NADH via the mito- The capability of βOHB to reduce production of reactive
chondrial BDH-catalysed reaction. This may be expected to oxygen species and prevent neuronal death was also dem-
reduce the activity of both the cytosolic and mitochondrial onstrated in rats in a model of hypoglycaemia induced by
malate dehydrogenases (Fig. 2), with associated effects on glucose deprivation [108].
the movement of 2-oxoglutarate (α-ketoglutarate), a key
intermediate in the Krebs cycle but also a key metabolite βOHB Metabolism Alters Activity of K+-ATP Channels
in cytosolic transaminase reactions. Further complicat-
ing matters, the metabolism of βOHB via SCOT generates Supply of βOHB along with glucose has been shown to
succinate from succinyl-CoA, which are also Krebs cycle reduce the use of glucose in brain cellular preparations
intermediates, but bypassing the succinylCo-A synthetase [101, 109] and this is likely due to a reduction in glycolytic
step (Fig. 2). activity, with addition of 1  mM βOHB to neurons in the
presence of glucose reducing glycolysis by around a third
[110]. The effect of 1 mM βOHB was further examined and
βOHB Reduces Production of Reactive Oxygen Species found to alter the response of NMDA receptors which dis-
via Action at Complex II played both a higher EC50 to NMDA and a lower maximal
calcium response in the presence vs the absence of βOHB,
The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) but no effect on the release of neurotransmitter. By study-
treated mouse is a model of Parkinson’s disease. MPTP ing responses induced by the KATP channel blocker gliben-
exerts its effects through blocking complex I (NADH clamide and the KATP channel opener diazoxide the authors
dehydrogenase) in mitochondria [103]. Mice, treated with concluded that these effects of βOHB were via indirect
MPTP, were infused with βOHB which conferred partial effects on KATP channels. The presence of βOHB and the
protection against the dopaminergic neurodegeneration and resultant reduction in glycolytic activity and reduced avail-
motor deficits induced by MPTP. The authors suggested ability of ATP would result in either a higher opening prob-
that this effect was due to effects of βOHB on complex II ability for KATP channels under these circumstances, or a
(succinate dehydrogenase), thus increasing mitochondrial longer opening time [110]. Application of ketone bodies
respiration and bypassing the complex I deficits [104]. at supra-physiological concentrations (0.4 mM βOHB) has
This is because increased SCOT activity in metabolising been shown to reduce the spontaneous firing rates of sub-
the βOHB drives increased production of succinate; the stantia nigra pars reticulate neurons, and to reduce the vul-
authors showed that succinate was protective against MPTP nerability of these neurons to a challenge that caused them
toxicity, although to a higher degree than βOHB, and that to fire faster. This effect was mediated by D-βOHB but not
this effect was not driven by increased NADH, since added by the inactive L-βOHB. Further, this protective effect on
NADH had little effect. Investigation of this hypothesis the neurons was abolished when the KATP channels were
in C. elegans using worms with complex I or II defective ablated, either through pharmacological approaches or
mutants however, suggested that βOHB required com- gene knockout [111]. The effect of ketone bodies on KATP
plex I to exert its effects and that these were independent channels may be mediated via Bcl-2 associated agonist
of complex II [105] although they were also able to show of cell death (BAD) protein, which acts as a regulator of
succinate and other TCA cycle intermediate dependent glucose metabolism, including in brain [112]. Ablation of
effects. The finding of βOHB protection via Complex I is BAD through knockout results in significantly increased

13
Neurochem Res (2017) 42:35–49 43

mitochondrial use of βOHB and reduction in use of glu- activity of Akt/protein kinase B downstream of the insulin
cose. Knockout mice were also significantly more resistant receptor, decreasing insulin signalling [118].
to kainite-induced seizures. This effect is likely mediated Extension of life span in glial cells has also been demon-
through phosphorylation of the BH3 domain [112]. strated following addition of 5 or 10 mM βOHB with sig-
nificant reductions in apoptosis in the presence of βOHB.
βOHB Metabolism Alters Activity of Ca2+ Channels This effect was greater using methyl-βOHB, which has
improved membrane permeability compared to βOHB
There is considerable evidence that βOHB causes elevation [113].
in Ca2+. A study adding βOHB to glial cells showed dra-
matic and immediate elevations in cytosolic Ca2+ through a βOHB Reduces Neuroinflammation Generated
mechanism involving L-type Ca2+ channels [113]. In sym- by Immune Cells
pathetic neurons, βOHB has been shown to directly inter-
act with a G-protein coupled fatty acid receptor (GPR41; βOHB has been reported to reduce inflammation gener-
FFA3) and to modulate sympathetic activity, although ated by macrophages via a mechanism independent of
some have reported it to be an antagonist at this receptor any of those reported above [119]. The NLR family, pyrin
[114] and others an agonist [115] with EC50 2.0 ± 1.2 mM. domain-containing 3 (NLRP3) inflammasome is a multi-
This receptor (GPR41) is coupled to N-type (Cav2.2) Ca2+ protein complex that controls the activation of caspase-1
channels and βOHB is capable of modulating these cur- and the release of the pro-inflammatory cytokines IL-1β
rents at concentrations which are achievable in vivo [115]. and IL-18 in macrophages [120]. This inflammasome,
Acetoacetate, by contrast, has no effect. which is expressed mainly in immune cells [121], is acti-
vated by a shift to low cytoplasmic K+ levels. Youm et al.
βOHB is an Inhibitor of Histone Deacetylases HDAC 1, noted that βOHB prevented the decline in K+ levels and
3 and 4 at Concentrations Achievable In Vivo prevented activation of NLRP3. They worked thoroughly
through a list of known βOHB mechanisms to see if these
βOHB has been shown to improve defence against oxida- were responsible for preventing inflammasome activation.
tive stress through its direct inhibition of histone deacety- They were able to rule them all out but did not identify
lase (HDAC) activity. βOHB inhibits HDAC1, HDAC3 and what mechanism actually was responsible [119]. Ca2+ sig-
HDAC4 with IC50s of 5.3, 2.4 and 5.4  mM, respectively. nalling was not tested as a possible cause although the Ca2+
This inhibition results in direct upregulation of genes in sensing receptor in mice has been shown to be involved in
the FOXO3A network, including catalase, mitochondrial NLRP3 activation [122].
superoxide dismutase (Mn-SOD) and metallothionein 2.
The authors also treated mice with βOHB infusion for 24 h
with and challenged them with paraquat. They found sig- Summary of Metabolic Effects of βOHB
nificantly reduced evidence of oxidative stress, measured
by lipid peroxidation and protein carbonylation, in kidney • βOHB is taken up in to the brain via the monocarboxy-
from βOHB-treated compared to control mice [116]. βOHB late transporter. Brain uptake can be a metabolic con-
has also been shown to reduce lipoperoxidation caused by trol point. βOHB can also be produced endogenously in
three days of intrastriatal injections of glutamate in mice brain by astrocytes which have the capacity to metabo-
[117]. lise free fatty acids.
• βOHB is a high energy substrate and its addition to
βOHB Increases Lifespan in Worms and in Cultured mitochondria will alter the NAD+/NADH and Q/QH2
Cells couples. Glycolysis is reduced in the presence of βOHB
and mitochondria have less capacity to generate reactive
Supplementation with βOHB has been shown to extend the oxygen species from Complex II.
lifespan of C. elegans by 20%. This was suggested to be • βOHB can bind to BAD protein and influence the open-
via two possible mechanisms. Firstly, via HDAC inhibition ing of K+ channels. This effect may also be exacerbated
through an AMPK and Sir2-dependent effect (Sir2 is the C. under metabolically active conditions by reduced glyco-
elegans equivalent of the mammalian SIRT1), or through lysis, leading to reduced cytosolic ATP to power the K+
increasing the concentration of acetyl-CoA, again resulting channels.
in increased histone acetylation [105]. Secondly the effects • βOHB is an agonist at free fatty acid receptor GPR41,
of βOHB may be mediated through inhibition of the insulin directly modulating the activity of N-type Ca2+ chan-
signalling pathway. In mice, administration of βOHB has nels.
been shown to yield a 50% reduced phosphorylation and

13
44 Neurochem Res (2017) 42:35–49

• βOHB is an inhibitor of histone deacetylases HDAC short-echo (TE = 20 ms) STEAM in infants supplemented
1, 3 and 4 at concentrations achievable in  vivo. This with DL-βOHB [125].
inhibition results in direct upregulation of genes in the In 1998, an editing method was published to allow
FOXO3A network, including catalase, mitochondrial resolution of the methyl doublets from lactate and βOHB
superoxide dismutase (Mn-SOD) and metallothionein 2. from background lipid signal, and used to measure brain
• βOHB has specific anti-inflammatory effects on NLRP3 levels of these compounds in epilepsy both before and
inflammasome via a direct but as yet unidentified mech- after treatment with a ketogenic diet. The authors suc-
anism. cessfully detected both lactate and βOHB at 2.1  T; no
βOHB was detected in the patient with epilepsy at base-
line but a significant elevation in this compound was
Measurement of βOHB in Brain Using Magnetic noted after implementation of a ketogenic diet [126].
Resonance Spectroscopy This method, which used ISIS for localisation [127],
was further modified by this group and used to detect
βOHB is detectable in human and rat brain in vivo. Under βOHB in healthy humans and following fasting-induced
normal circumstances, the concentrations of βOHB in ketosis [15]. The authors were able to quantify the βOHB
brain are very low and difficult to detect reliably using with a reasonable reliability.
standard, or even edited, magnetic resonance spectros- In the absence of access to editing methods, others
copy. The main reported confound is in confusing the have detected βOHB in  vivo in diabetic ketosis using
methyl resonance from βOHB with that from lactate. single voxel PRESS (Point RESolved Spectroscopy)
Lactate is chemically similar with a methyl resonance with a long (144  ms) echo-time; this allows full inver-
linked to an hydroxymethyl group, but the two can be sion of the βOHB doublet and removes any contaminat-
resolved at field strengths as low as 1.5 T (Fig. 3). βOHB ing lipid signal due to relaxation [128]. A short echo
has been measured in human brain using a range of dif- (TE = 35  ms, plus a further TE = 144  ms acquisition)
ferent sequences. Early attempts to measure brain βOHB PRESS method has also been used to detect βOHB fol-
in diabetes mellitus used short-echo STEAM (STimu- lowing a ketogenic diet, recovering the βOHB signal by
lated Echo, Acquisition Mode; [123]) spectroscopy fitting the spectrum with a basis set of known priors,
(TE = 30  ms) at 1.5  T and detected peaks arising from including βOHB [129].
acetoacetate and acetone but not βOHB [124]. At 1.5  T The editing sequence MEGA-PRESS [130] has been
the methyl peaks from βOHB and lactate overlap con- adapted for measurement of lactate in  vivo [131] and
siderably and the presence of significant lipid or macro- could also be used successfully to measure βOHB with
molecule resonances at short echo times can also make improved signal to noise over ISIS localisation methods.
it difficult to resolve the βOHB doublet from the back- In summary, βOHB is difficult to detect reliably in
ground resonance; this is a similar issue to that found healthy human brain and is best measured using an edit-
when trying to measure lactate levels using short-echo ing sequence to resolve the βOHB resonances from back-
spectroscopy. Others have detected βOHB at 3  T using ground signals.

Fig. 3 High field section of


1
H magnetic resonance spectra
of βOHB and lactate at 3 T.
Spectra were generated using
NMRScope in jMRUI (v 4,
build 162) using published
coupling constants and chemi-
cal shifts and a PRESS (Point
RESolved Spectroscopy)
sequence (TE = 144 ms)

13
Neurochem Res (2017) 42:35–49 45

Therapeutic Effects of βOHB promise in a case of Alzheimer disease [135] and it is plain
that the use of ketone bodies such as (R)-3-hydroxybutyl
Much of the literature about the effects of ketone bodies (R)-3-hydroxybutyrate deserves further investigation [136].
on disease outcomes is focused on the ketogenic diet, mak- βOHB has been used experimentally in rats to try and
ing it difficult to divorce the effects of βOHB alone from prevent blood brain barrier disruption due to traumatic
the more complicated systems effects induced by this diet. brain injury. βOHB, administered by infusion prior to
Here, we examine the literature where βOHB alone has injury, not only failed to provide protection against blood
been used as a potential therapeutic. brain barrier disruption in brain injured rats but it caused
The main problem with using βOHB as a therapeutic lies blood brain barrier disruption in healthy animals [137]
in being able to deliver sufficient βOHB to the brain, due to resulting in decreased GSH and malondialdehyde levels
the limited uptake of βOHB across the blood brain barrier and reduced glucose transporter (GLUT1) expression. GSH
and the difficulty in sustaining high enough levels of βOHB levels are maintained by reduction with NADPH, which is
in the blood. As detailed above, uptake can be increased by mostly produced via the pentose phosphate pathway [68].
instigation of dietary changes, such as a high fat diet or reg- Although it might be expected that NADPH levels might be
ular ingestion of medium chain fatty acids. Ketogenic diets, altered if glycolytic activity is reduced, with concomitant
which can raise plasma βOHB to higher levels, are difficult reduction in GSH, the ketogenic diet has been shown to
to adhere to, both for those consuming them and for their increase GSH levels through increased GSH synthesis and
care-givers. Medium chain triacylglycerols, on the other reduction in production of reactive oxygen species [138].
hand, can be consumed along with the usual diet, making In this regard, it should be borne in mind that acute and
them more attractive. Consumption of medium chain tria- chronic exposure to βOHB have different outcomes. Indeed
cylglycerols produces a mild ketosis which has been esti- exposure to a ketogenic diet after contusion injury in rats
mated to contribute up to 8–9% to brain energy metabolism has been shown to reduce contusion volume and improve
and it is well tolerated [132]. neuronal survival, with better responses in younger rats
The effect was tested of a one-off drink of medium chain [139].
triglycerides on the cognition of 20 adults with impaired
cognition (diagnosed with Alzheimer’s disease or mild
cognitive impairment) compared to consumption of a pla- Conclusion
cebo on a separate day. βOHB levels were measured in
blood subsequent to consumption. The authors reported βOHB is a high energy metabolite that has significant met-
a different response in cognitive performance and blood abolic impact and, if well understood, could be used as a
βOHB levels depending on the subjects ApoE status; those well-tolerated therapeutic agent in a range of neurodegen-
with the APOE-ε4 allele responded to MCT consump- erative, inflammatory and traumatic brain disorders.
tion with continually increasing plasma βOHB levels, no
improvement on the Alzheimer disease assessment scale Acknowledgements The authors would like to thank Ben Rowlands
cognitive subscale (ADAS-cog) compared to plateauing of of Neuroscience Research Australia and Don Thomas of the UNSW
plasma βOHB levels and improvement on the ADAS-cog Mark Wainwright Analytical Centre for their assistance in the prepa-
ration of this manuscript. The authors would like to acknowledge the
in those without APOE-ε4 allele. All subjects performance lifetime commitment of Professor Mary McKenna to the pursuit of
on paragraph recall improved with MCT consumption but scientific excellence and education in neurochemistry and to dedicate
performance on the Stroop colour word test, which meas- this article to her contributions to this field. We have greatly enjoyed
ures selective attention, was unchanged [133]. This study and also benefitted from our interactions over the years.
had only 20 participants, meaning that statistical power was
limited, especially after subdividing the group by APOE
status. References
Recently, attention has been given to esterified versions
of βOHB, such as (R)-3-hydroxybutyl (R)-3-hydroxybu- 1. Wakeman A, Dakin H (1909) On the decomposition of
tyrate. This compound can be consumed with the normal β-oxybutyric acid and acetoacetic acid by enzymes of the liver.
J Biol Chem 6:373–389
diet and is hydrolysed completely in the small intestine 2. Dedkova EN, Blatter LA (2014) Role of β-hydroxybutyrate, its
producing βOHB and 1,3-butanediol which is converted polymer poly-β-hydroxybutyrate and inorganic polyphosphate
to βOHB and acetoacetate in the liver, producing a “thera- in mammalian health and disease. Front Physiol 5:260
peutic ketosis”. This method results in blood βOHB levels 3. Krebs H, Williamson D, Bates MW, Page MA, Hawkins R
(1971) The role of ketone bodies in caloric homeostasis. Adv
higher than those achieved after consuming medium chain Enzyme Regul 9:387–409
fatty acids and is well tolerated [134]. Supplementation 4. Longo VD, Mattson MP (2014) Fasting: molecular mechanisms
with (R)-3-hydroxybutyl (R)-3-hydroxybutyrate has shown and clinical applications. Cell Metab 19:181–192

13
46 Neurochem Res (2017) 42:35–49

5. Gonzalez SV, Nguyen NHT, Rise F, Hassel B (2005) Brain transporter for active uptake of L-lactate and ketone bodies in
metabolism of exogenous pyruvate. J Neurochem 95:284–293 the brain. J Neurochem 98:279–288
6. Dienel GA (2012) Brain lactate metabolism: the discov- 25. Gopal E, Umapathy NS, Martin PM, Ananth S, Gnana-
eries and the controversies. J Cereb Blood Flow Metab Prakasam JP, Becker H, Wagner CA, Ganapathy V, Prasad
32:1107–1138 PD (2007) Cloning and functional characterization of human
7. Rae C, Fekete AD, Kashem MA, Nasrallah FA, Bröer S (2012) SMCT2 (SLC5A12) and expression pattern of the transporter in
Metabolism, compartmentation, transport and production kidney. Biochim Et Biophys Acta Biomembr 1768:2690–2697
of acetate in the cortical brain tissue slice. Neurochem Res 26. Tildon JT, McKenna MC, Stevenson JH Jr (1994) Transport of
37:2541–2553 3-hydroxybutyrate by cultured rat brain astrocytes. Neurochem
8. Künnecke B, Cerdan S, Seelig J (1993) Cerebral metabolism Res 19:1237–1242
of [1, 2-13C2] glucose and [U-13C4] 3-hydroxybutyrate in rat 27. Leino RL, Gerhart DZ, Duelli R, Enerson BE, Drewes LR
brain as detected by 13C NMR spectroscopy. NMR Biomed (2001) Diet-induced ketosis increases monocarboxylate trans-
6:264–277 porter (MCT1) levels in rat brain. Neurochem Int 38:519–527
9. Edmond J, Robbins R, Bergstrom J, Cole R, De Vellis J (1987) 28. Regen DM, Callis JT, Sugden MC (1983) Studies of cerebral
Capacity for substrate utilization in oxidative metabolism by β-hydroxybutyrate transport by carotid injection; effects of age,
neurons, astrocytes, and oligodendrocytes from developing diet and injectant composition. Brain Res 271:289–299
brain in primary culture. J Neurosci Res 18:551–561 29. Puchowicz MA, Xu K, Sun X, Ivy A, Emancipator D, LaManna
10. Cahill GF, Veech RL (2003) Ketoacids? Good medicine? Trans JC (2007) Diet-induced ketosis increases capillary density with-
Am Clin Climatol Assoc 114:149 out altered blood flow in rat brain. Am J Physiol Endocrinol
11. Hansen JL, Freier EF (1978) Direct assays of lactate, pyruvate, Metab 292:E1607
beta-hydroxybutyrate, and acetoacetate with a centrifugal ana- 30. Mikkelsen KH, Seifert T, Secher NH, Grøndal T, van Hall G
lyzer. Clin Chem 24:475–479 (2014) Systemic, cerebral and skeletal muscle ketone body and
12. Owen O, Morgan A, Kemp H, Sullivan J, Herrera M, Cahill energy metabolism during acute hyper-D-β-hydroxybutyratemia
G Jr (1967) Brain metabolism during fasting. J Clin Invest in post-absorptive healthy males. J Clin Endocrinol Metab
46:1589 100:636–643
13. Mitchell GA, Kassovska-Bratinova S, Boukaftane Y, Robert 31. Courchesne-Loyer A, Croteau E, Castellano C-A, St-Pierre
MF, Wang SP, Ashmarina L, Lambert M, Lapierre P, Potier E V, Hennebelle M, Cunnane SC (2016) Inverse relationship
(1995) Medical aspects of ketone body metabolism. Clin Invest between brain glucose and ketone metabolism in adults dur-
Med 18:193–216 ing short-term moderate dietary ketosis: a dual tracer quantita-
14. Laffel L (1999) Ketone bodies: a review of physiology, patho- tive positron emission tomography study. J Cereb Blood Flow
physiology and application of monitoring to diabetes. Diabetes Metab. doi:10.1177/0271678X16669366
Metab Res Rev 15:412–426 32. Hawkins RA, Mans AM, Davis DW (1986) Regional ketone
15. Pan JW, Rothman DL, Behar KL, Stein DT, Hethering- body utilization by rat brain in starvation and diabetes. Am J
ton HP (2000) Human brain β-hydroxybutyrate and lactate Physiol Endocrinol Metab 250:E169
increase in fasting-induced ketosis. J Cereb Blood Flow Metab 33. Ito K, Uchida Y, Ohtsuki S, Aizawa S, Kawakami H, Katsukura
20:1502–1507 Y, Kamiie J, Terasaki T (2011) Quantitative membrane protein
16. Samala R, Klein J, Borges K (2011) The ketogenic diet changes expression at the blood–brain barrier of adult and younger cyn-
metabolite levels in hippocampal extracellular fluid. Neurochem omolgus monkeys. J Pharm Sci 100:3939–3950
Int 58:5–8 34. Gerhart DZ, Enerson BE, Zhdankina OY, Leino RL, Drewes
17. Halestrap AP, Meredith D (2004) The SLC16 gene family— LR (1997) Expression of monocarboxylate transporter MCT1
from monocarboxylate transporters (MCTs) to aromatic amino by brain endothelium and glia in adult and suckling rats. Am J
acid transporters and beyond. Pflugers Arch 447:619–628 Physiol Endocrinol Metab 273:E207–E213
18. Halestrap AP (2013) Monocarboxylic acid transport. Compr 35. Vannucci SJ, Simpson IA (2003) Developmental switch in brain
Physiol nutrient transporter expression in the rat. Am J Physiol Endo-
19. Bergersen LH, Magistretti PJ, Pellerin L (2005) Selective crinol Metab 285:E1127–E1134
postsynaptic co-localisation of MCT2 with AMPA recep- 36. Uchida Y, Ohtsuki S, Katsukura Y, Ikeda C, Suzuki T, Kamiie
tor GluR2/3 subunits at excitatory synapses exhibiting AMPA J, Terasaki T (2011) Quantitative targeted absolute proteomics
receptor trafficking. Cereb Cortex 15:361–370 of human blood–brain barrier transporters and receptors. J Neu-
20. Rafiki A, Boulland JL, Halestrap AP, Ottersen OP, Bergersen rochem 117:333–345
LH (2003) Highly differential expression of the monocarboxy- 37. Halestrap AP (1978) Pyruvate and ketone-body transport across
late transporters MCT2 and MCT4 in the developing rat brain. the mitochondrial membrane. Exchange properties, pH-depend-
Neurosci 122:677–688 ence and mechanism of the carrier. Biochem J 172:377–387
21. Broer S, Broer A, Schneider HP, Stegen C, Halestrap AP, Deit- 38. Paradies G, Papa S (1997) On the kinetics and substrate speci-
mer JW (1999) Characterization of the high-affinity monocar- ficity of the pyruvate translocator in rat liver mitochondria. Bio-
boxylate transporter MCT2 in Xenopus laevis oocytes. Bio- chim Et Biophys Acta Bioenerg 462:333–346
chem J 341:529–535 39. Booth RF, Clark JB (1981) Energy metabolism in rat brain:
22. Carpenter L, Halestrap AP (1994) The kinetics, substrate and inhibition of pyruvate decarboxylation by 3-hydroxybutyrate in
inhibitor specificity of the lactate transporter of ehrlich-lettre neonatal mitochondria. J Neurochem 37:179–185
tumor cells studied with the intracellular pH indicator BCECF. 40. Veech RL (2004) The therapeutic implications of ketone bodies:
Biochem J 304:751–760 the effects of ketone bodies in pathological conditions: ketosis,
23. Fox JEM, Meredith D, Halestrap AP (2000) Characterisation of ketogenic diet, redox states, insulin resistance, and mitochon-
human monocarboxylate transporter 4 substantiates its role in drial metabolism. Prostaglandin Leukotriene Essent Fatty Acid
lactic acid efflux from skeletal muscle. J Physiol 529:285–293 70:309–319
24. Martin PM, Gopal E, Ananth S, Zhuang L, Itagaki S, Prasad 41. Sato K, Kashiwaya Y, Keon C, Tsuchiya N, King M, Radda G,
BM, Smith SB, Prasad PD, Ganapathy V (2006) Identify Chance B, Clarke K, Veech R (1995) Insulin ketone bodies, and
of SMCT1(SLC5A8) as a neuron-specific Na+-coupled mitochondrial energy transduction. FASEB J 9:651–658

13
Neurochem Res (2017) 42:35–49 47

42. Klee CB, Sokoloff L (1967) Changes in d(−)-β-hydroxybutyric Succinyl-CoA:3-ketoacid CoA transferase (SCOT): cloning
dehydrogenase activity during brain maturation in the rat. J Biol of the human SCOT gene, tertiary structural modeling of the
Chem 242:3880–3883 human SCOT monomer, and characterization of three patho-
43. Latruffe N, Gaudemer Y (1974) Propriétés et mécanisme ciné- genic mutations. Genomics 68:144–151
tique de la D(−)β-hydroxybutyrique déshydrogénase de parti- 57. Berry G, Fukao T, Mitchell G, Mazur A, Ciafre M, Gibson J,
cules sous-mitochondriales de foie de rat; Effets comparés de Kondo N, Palmieri M (2001) Neonatal hypoglycaemia in severe
différents agents thiols. Biochimie 56:435–444 succinyl-CoA: 3-oxoacid CoA-transferase deficiency. J Inherit
44. Rardin MJ, Newman JC, Held JM, Cusack MP, Sorensen DJ, Li Metab Dis 24:587–595
B, Schilling B, Mooney SD, Kahn CR, Verdin E (2013) Label- 58. Gibson K, Breuer J, Nyhan W (1988) 3-Hydroxy-3-methyl-
free quantitative proteomics of the lysine acetylome in mito- glutaryl-coenzyme A lyase deficiency: review of 18 reported
chondria identifies substrates of SIRT3 in metabolic pathways. patients. Eur J Pediatr 148:180–186
Proc Natl Acad Sci USA 110:6601–6606 59. Van der Knaap M, Bakker H, Valk J (1998) MR imaging and
45. Hebert AS, Dittenhafer-Reed KE, Yu W, Bailey DJ, Selen ES, proton spectroscopy in 3-hydroxy-3-methylglutaryl coenzyme
Boersma MD, Carson JJ, Tonelli M, Balloon AJ, Higbee AJ, A lyase deficiency. Am J Neuroradiol 19:378–382
Westphall MS, Pagliarini DJ, Prolla TA, Assadi-Porter F, Roy 60. Gordon K, Riding M, Camfield P, Bawden H, Ludman M, Bag-
S, Denu JM Coon JJ (2013) Calorie restriction and SIRT3 trig- nell P (1994) CT and MR of 3-hydroxy-3-methylglutaryl-coen-
ger global reprogramming of the mitochondrial protein acety- zyme A lyase deficiency. Am J Neuroradiol 15:1474–1476
lome. Mol Cell 49:186–199 61. Fukao T, Scriver CR, Kondo N; Group TCW (2001) The clini-
46. Guo K, Lukacik P, Papagrigoriou E, Meier M, Lee WH, Adam- cal phenotype and outcome of mitochondrial acetoacetyl-CoA
ski J, Oppermann U (2006) Characterization of human DHRS6, thiolase deficiency (β-ketothiolase or T2 deficiency) in 26 enzy-
an orphan short chain dehydrogenase/reductase enzyme: a matically proved and mutation-defined patients. Mol Genet
novel, cytosolic type 2 R-beta-hydroxybutyrate dehydrogenase. Metab 72:109–114
J Biol Chem 281:10291–10297 62. Bennett M, Hosking G, Smith M, Gray R, Middleton B (1984)
47. Chang HT, Olson LK, Schwartz KA (2013) Ketolytic and gly- Biochemical investigations on a patient with a defect in cyto-
colytic enzymatic expression profiles in malignant gliomas: solic acetoacetyl-CoA thiolase, associated with mental retarda-
implication for ketogenic diet therapy. Nutr Metab 10:1 tion. J Inherit Metab Dis 7:125–128
48. Bixel MG, Hamprecht B (1995) Generation of ketone bod- 63. Hawkins R, Williamson D, Krebs H (1971) Ketone-body uti-
ies from leucine by cultured astroglial cells. J Neurochem lization by adult and suckling rat brain in  vivo. Biochem J
65:2450–2461 122:13–18
49. Puisac B, Ramos M, Arnedo M, Menao S, Gil-Rodríguez MC, 64. Sokoloff L (1973) Metabolism of ketone bodies by the brain.
Teresa-Rodrigo ME, Pié A, de Karam JC, Wesselink J-J, Gimé- Annu Rev Med 24:271–280
nez I, Ramos FJ, Casals N, Gómez-Puertas P, Hegardt FG, Pié 65. Cahill GFJ (2006) Fuel metabolism in starvation. Annu Rev
J (2012) Characterization of splice variants of the genes encod- Nutr 26:1–22
ing human mitochondrial HMG-CoA lyase and HMG-CoA syn- 66. DeVivo DC, Leckie MP, Agrawal HC (1973) The differential
thase, the main enzymes of the ketogenesis pathway. Mol Biol incorporation of β-hydroxybutyrate and glucose into brain glu-
Rep 39:4777–4785 tamate in the newborn rat. Brain Res 55:485–490
50. Fukao T, Song X-Q, Mitchell GA, Yamaguchi S, Sukegawa K, 67. Nehlig A (2004) Brain uptake and metabolism of ketone bodies
Or T, Kondo N (1997) Enzymes of ketone body utilization in in animal models. Prostaglandin Leukotriene Essent Fatty Acid
human tissues: protein and messenger RNA levels of succinyl- 70:265–275
coenzyme A (CoA):3-ketoacid CoA transferase and mitochon- 68. Hotta SS (1962) Glucose metabolism in brain tissue: the hex-
drial and cytosolic acetoacetyl-CoA thiolases. Pediatr Res ose monophosphate shunt and its role in glutathione reduction. J
42:498–502 Neurochem 9:43–51
51. Ohnuki M, Takahashi N, Yamasaki M, Fukui T (2005) Differ- 69. Bilger A, Nehlig A (1992) Quantitative histochemical changes
ent localization in rat brain of the novel cytosolic ketone body- in enzymes involved in energy metabolism in the rat brain dur-
utilizing enzyme, acetoacetyl-CoA synthetase, as compared to ing postnatal development 2. Glucose-6-phosphate dehydroge-
succinyl-CoA:3-oxoacid CoA-transferase. Biochim Et Biophys nase and beta-hydroxybutyrate dehydrogenase. Int J Dev Neuro-
Acta 1729:147–153 sci 10:143–152
52. Endemann G, Goetz PG, Edmond J, Brunengraber H (1982) 70. Nehlig A, de Vasconcelos AP (1993) Glucose and ketone
Lipogenesis from ketone bodies in the isolated perfused rat body utilization by the brain of neonatal rats. Prog Neurobiol
liver. Evidence for the cytosolic activation of acetoacetate. J 40:163–220
Biol Chem 257:3434–3440 71. Rho JM, Stafstrom CE (2012) The ketogenic diet as a treatment
53. Hasegawa S, Kume H, Iinuma S, Yamasaki M, Takahashi N, paradigm for diverse neurological disorders. Front Pharmacol
Fukui T (2012) Acetoacetyl-CoA synthetase is essential for 3:59
normal neuronal development. Biochem Biophys Res Commun 72. Maalouf M, Rho JM, Mattson MP (2009) The neuroprotective
427:398–403 properties of calorie restriction, the ketogenic diet, and ketone
54. Tildon JT, Cornblath M (1972) Succinyl-CoA: 3-ketoacid CoA- bodies. Brain Res Rev 59:293–315
transferase deficiency. A cause for ketoacidosis in infancy. J 73. McKenna MC, Tildon JT, Stevenson JH, Boatright R, Huang S
Clin Invest 51:493–498 (1993) Regulation of energy metabolism in synaptic terminals
55. Fukao T, Sass JO, Kursula P, Thimm E, Wendel U, Ficicioglu and cultured rat brain astrocytes: differences revealed using
C, Monastiri K, Guffon N, Barić I, Zabot MT, Kondo N (2011) aminooxyacetate. Dev Neurosci 15:320–329
Clinical and molecular characterization of five patients with 74. Chechik T, Roeder L, Tildon J, Poduslo S (1987) Ketone body
succinyl-CoA:3-ketoacid CoA transferase (SCOT) deficiency. enzyme activities in purified neurons, astrocytes and oligoden-
Biochim Et Biophys Acta 1812:619–624 droglia. Neurochem Int 10:95–99
56. Fukao T, Mitchell GA, Song X-Q, Nakamura H, Kassovska- 75. Berl S, Frigyesi TL (1969) Turnover of glutamate glutamine
Bratinova S, Orii KE, Wraith JE, Besley G, Wanders RJA, aspartate and GABA labeled with 1-14C acetate in caudate
Niezen-Koning KE, Berry GT, Palmieri M, Kondo N (2000) nucleus thalamus and motor cortex (cat). Brain Res 12:444–455

13
48 Neurochem Res (2017) 42:35–49

76. Berl S, Nicklas WJ, Clarke DD (1970) Compartmentation of 94. Tildon JT, Roeder LM, Stevenson JH (1985) Substrate oxida-
citric acid cycle metabolism in brain-labeilling of glutamate, tion by isolated rat brain mitochondria and synaptosomes. J
glutamine, aspartate and GABA by several radioactive tracer Neurosci Res 14:207–215
metabolites. J Neurochem 17:1009–1015 95. Marosi K, Kim SW, Moehl K, Scheibye-Knudsen M, Cheng A,
77. Cremer JE (1971) Incorporation of label from d-β-hydroxy- Cutler R, Camandola S, Mattson MP (2016) 3-Hydroxybutyrate
[14C] butyrate and [3-14C] acetoacetate into amino acids in rat regulates energy metabolism and induces BDNF expression in
brain in vivo. Biochem J 122:135–138 cerebral cortical neurons. J Neurochem. doi:10.1111/jnc.13868
78. Pan JW, de Graaf RA, Petersen KF, Shulman GI, Hetherington 96. Erecinska M, Nelson D, Daikhin Y, Yudkoff M (1996) Regula-
HP, Rothman DL (2002) [2,4-13C2]-hydroxybutyrate metabo- tion of GABA level in rat brain synaptosomes: fluxes through
lism in human brain. J Cereb Blood Flow Metab 22:890–898 enzymes of the GABA shunt and effects of glutamate, calcium
79. Leong SF, Lai JCK, Lim L, Clark JB (1981) Energy-metabolis- and ketone bodies. J Neurochem 67:2325–2334
ing enzymes in brain regions of adult and aging rats. J Neuro- 97. Yudkoff M, Daikhin Y, Nissim I, Grunstein R, Nissim I (1997)
chem 37:1548–1556 Effects of ketone bodies on astrocyte amino acid metabolism. J
80. Williamson D, Bates MW, Page MA, Krebs H (1971) Activities Neurochem 69:682–692
of enzymes involved in acetoacetate utilization in adult mam- 98. McKenna MC, Tildon JT, Stevenson J, Huang X, Kingwell
malian tissues. Biochem J 121:41–47 KG (1995) Regulation of mitochondrial and cytosolic malic
81. Page MA, Williamson D (1971) Enzymes of ketone-body utili- enzymes from cultured rat brain astrocytes. Neurochem Res
sation in human brain. Lancet 298:66–68 20:1491–1501
82. Nugent S, Tremblay S, Chen KW, Ayutyanont N, Roontiva A, 99. Lajtha A, Gibson G, Dienel G (2007) Handbook of neurochem-
Castellano C-A, Fortier M, Roy M, Courchesne-Loyer A, Bocti istry and molecular neurobiology. Neuroactive proteins and
C (2014) Brain glucose and acetoacetate metabolism: a compar- peptides. Springer, Springer-Verlag
ison of young and older adults. Neurobiol Aging 35:1386–1395 100. Amaral AI, Teixeira AP, Sonnewald U, Alves PM (2011) Esti-
83. Blomqvist G, Thorell J, Ingvar M, Grill V, Widen L, Stone- mation of intracellular fluxes in cerebellar neurons after hypo-
Elander S (1995) Use of R-beta-[1-11C] hydroxybutyrate in PET glycemia: importance of the pyruvate recycling pathway and
studies of regional cerebral uptake of ketone bodies in humans. glutamine oxidation. J Neurosci Res 89:700–710
Am J Physiol Endocrinol Metab 269:E948–E959 101. Lund TM, Risa O, Sonnewald U, Schousboe A, Waagepetersen
84. Hawkins RA, Biebuyck JF (1979) Ketone bodies are selectively HS (2009) Availability of neurotransmitter glutamate is dimin-
used by individual brain regions. Science 205:325–327 ished when beta-hydroxybutyrate replaces glucose in cultured. J
85. Gesink DS, Wilson JE A (1985) Histochemical study of the dis- Neurochem 110:80–91
tribution of β-hydroxybutyrate dehydrogenase in developing rat 102. Cruz NF, Lasater A, Zielke HR, Dienel GA (2005) Activa-
cerebellum. J Neurochem 44:1308–1311 tion of astrocytes in brain of conscious rats during acoustic
86. Arakawa T, Goto T, Okada Y (1991) Effect of ketone body stimulation: acetate utilization in working brain. J Neurochem
(D-3-hydroxybutyrate) on neural activity and energy metabo- 92:934–947
lism in hippocampal slices of the adult guinea pig. Neurosci 103. Nicklas WJ, Youngster SK, Kindt MV, Heikkila RE IV (1987)
Lett 130:53–56 MPTP, MPP+ and mitochondrial function. Life Sci 40:721–729
87. Wada H, Okada Y, Nabetani M, Nakamura H (1997) The effects 104. Tieu K, Perier C, Caspersen C, Teismann P, Wu D-C, Yan S-D,
of lactate and beta-hydroxybutyrate on the energy metabolism Naini A, Vila M, Jackson-Lewis V, Ramasamy R (2003) D-β-
and neural activity of hippocampal slices from adult and imma- Hydroxybutyrate rescues mitochondrial respiration and miti-
ture rat. Dev Brain Res 101:1–7 gates features of Parkinson disease. J Clin Invest 112:892–901
88. Weiss JN, Lamp ST (1989) Cardiac ATP-sensitive K+ channels. 105. Edwards C, Canfield J, Copes N, Rehan M, Lipps D, Bradshaw
Evidence for preferential regulation by glycolysis. J Gen Physiol PC (2014) D-beta-hydroxybutyrate extends lifespan in C. ele-
94:911–935 gans. Aging 6:621
89. Dhar-Chowdhury P, Harrell MD, Han SY, Jankowska D, 106. Zhao Z, Lange DJ, Voustianiouk A, MacGrogan D, Ho L, Suh
Parachuru L, Morrissey A, Srivastava S, Liu W, Malester B, J, Humala N, Thiyagarajan M, Wang J, Pasinetti GM (2006) A
Yoshida H (2005) The glycolytic enzymes, glyceraldehyde- ketogenic diet as a potential novel therapeutic intervention in
3-phosphate dehydrogenase, triose-phosphate isomerase, and amyotrophic lateral sclerosis. BMC Neurosci 7:1
pyruvate kinase are components of the KATP channel mac- 107. Kashiwaya Y, Takeshima T, Mori N, Nakashima K, Clarke
romolecular complex and regulate its function. J Biol Chem K, Veech RL (2000) d-β-Hydroxybutyrate protects neurons in
280:38464–38470 models of Alzheimer’s and Parkinson’s disease. Proc Natl Acad
90. Silver IA, Deas J, Erecinska M (1997) Ion homeostasis in brain Sci USA 97:5440–5444
cells: differences in intracellular ion reponses to energy limita- 108. Julio-Amilpas A, Montiel T, Soto-Tinoco E, Gerónimo-Olvera
tion between cultured neurons and glial cells. Neuroscience C, Massieu L (2015) Protection of hypoglycemia-induced neu-
78:598–601 ronal death by β-hydroxybutyrate involves the preservation of
91. Chowdhury GM, Jiang L, Rothman DL, Behar KL (2014) The energy levels and decreased production of reactive oxygen spe-
contribution of ketone bodies to basal and activity-depend- cies. J Cereb Blood Flow Metab 35:851–860
ent neuronal oxidation in  vivo. J Cereb Blood Flow Metab 109. McKenna M, Tildon J, Stevenson J, Hopkins I (1994) Energy
34:1233–1242 metabolism in cortical synaptic terminals from weanling and
92. Wieland O, Funcke Hv, Löffler G (1971) Interconversion of mature rat brain: evidence for multiple compartments of tricar-
pyruvate dehydrogenase in rat heart muscle upon perfusion boxylic acid cycle activity. Dev Neurosci 16:291–300
with fatty acids or ketone bodies. FEBS Lett 15:295–298 110. Lund TM, Ploug KB, Iversen A, Jensen AA, Jansen-Olesen I
93. Newsholme EA, Randle PJ, Manchester KL (1962) Inhibition (2015) The metabolic impact of β-hydroxybutyrate on neuro-
of the phosphofructokinase reaction in perfused rat heart by transmission: reduced glycolysis mediates changes in calcium
respiration of ketone bodies, fatty acids and pyruvate. Nature responses and KATP channel receptor sensitivity. J Neurochem
193:270–271 132:520–531

13
Neurochem Res (2017) 42:35–49 49

111. Ma W, Berg J, Yellen G (2007) Ketogenic diet metabolites 126. Shen J, Novotny EJ, Rothman DL (1998) In  vivo lactate and
reduce firing in central neurons by opening KATP channels. J β-hydroxybutyrate editing using a pure-phase refocusing pulse
Neurosci 27:3618–3625 train. Magn Reson Med 40:783–788
112. Giménez-Cassina A, Martínez-François Juan  R, Fisher Jill  K, 127. Ordidge R, Connelly A, Lohman J (1969) Image-selected
Szlyk B, Polak K, Wiwczar J, Tanner Geoffrey  R, Lutas A, in  vivo spectroscopy (ISIS). A new technique for spatially
Yellen G (2012) Danial Nika N BAD-dependent regulation of selective NMR spectroscopy. J Magn Reson 66:283–294
fuel metabolism and KATP channel activity confers resistance 128. Wootton-Gorges SL, Buonocore MH, Kuppermann N, Marcin
to epileptic seizures. Neuron 74:719–730 J, DiCarlo J, Neely EK, Barnes PD, Glaser N (2005) Detection
113. Xiao X-Q, Zhao Y, Chen G-Q (2007) The effect of 3-hydroxy- of cerebral β-hydroxy butyrate, acetoacetate, and lactate on pro-
butyrate and its derivatives on the growth of glial cells. Bioma- ton MR spectroscopy in children with diabetic ketoacidosis. Am
terials 28:3608–3616 J Neuroradiol 26:1286–1291
114. Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi 129. Cecil KM, Mulkey SB, Ou X, Glasier CM (2015) Brain ketones
S, Kobayashi M, Hirasawa A, Tsujimoto G (2011) Short-chain detected by proton magnetic resonance spectroscopy in an
fatty acids and ketones directly regulate sympathetic nervous infant with Ohtahara syndrome treated with ketogenic diet.
system via G protein-coupled receptor 41 (GPR41). Proc Natl Pediatr Radiol 45:133–137
Acad Sci USA 108:8030–8035 130. Mescher M, Merkle H, Kirsch J, Garwood M, Gruetter R (1998)
115. Won Y-J, Lu VB, Puhl HL, Ikeda SR (2013) β-Hydroxybutyrate Simultaneous in  vivo spectral editing and water suppression.
modulates N-type calcium channels in rat sympathetic neu- NMR Biomed 11:266–272
rons by acting as an agonist for the G-protein-coupled receptor 131. Edden RAE, Harris AD, Murphy K, Evans CJ, Saxena N, Hall
FFA3. J Neurosci 33:19314–19325 JE, Bailey DM, Wise RG (2010) Edited MRS is sensitive to
116. Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le changes in lactate concentration during inspiratory hypoxia. J
Moan N, Grueter CA, Lim H, Saunders LR, Stevens RD (2013) Magn Reson Imag 32:320–325
Suppression of oxidative stress by β-hydroxybutyrate, an endog- 132. Courchesne-Loyer A, Fortier M, Tremblay-Mercier J, Choui-
enous histone deacetylase inhibitor. Science 339:211–214 nard-Watkins R, Roy M, Nugent S, Castellano C-A, Cun-
117. Mejía-Toiber J, Montiel T, Massieu L (2006) D-β- nane SC (2013) Stimulation of mild, sustained ketonemia by
hydroxybutyrate prevents glutamate-mediated lipoperoxida- medium-chain triacylglycerols in healthy humans: estimated
tion and neuronal damage elicited during glycolysis inhibition potential contribution to brain energy metabolism. Nutrition
in vivo. Neurochem Res 31:1399–1408 29:635–640
118. Yamada T, Zhang S-J, Westerblad H, Katz A (2010) 133. Reger MA, Henderson ST, Hale C, Cholerton B, Baker LD,
β-Hydroxybutyrate inhibits insulin-mediated glucose transport Watson G, Hyde K, Chapman D, Craft S (2004) Effects of
in mouse oxidative muscle. Am J Physiol Endocrinol Metab β-hydroxybutyrate on cognition in memory-impaired adults.
299:E364–E373 Neurobiol Aging 25:311–314
119. Youm Y-H, Nguyen KY, Grant RW, Goldberg EL, Bodogai 134. Clarke K, Tchabanenko K, Pawlosky R, Carter E, King MT,
M, Kim D, D’Agostino D, Planavsky N, Lupfer C, Kanneganti Musa-Veloso K, Ho M, Roberts A, Robertson J, VanItallie TB
TD, Kang S, Horvath TL, Fahmy TM, Crawford PA, Biragyn (2012) Kinetics, safety and tolerability of (R)-3-hydroxybutyl
A, Alnemri E, Dixit VD (2015) Ketone body β-hydroxybutyrate (R)-3-hydroxybutyrate in healthy adult subjects. Regul Toxicol
blocks the NLRP3 inflammasome-mediated inflammatory dis- Pharmacol 63:401–408
ease. Nat Med 21:263–269 135. Newport MT, VanItallie TB, Kashiwaya Y, King MT, Veech RL
120. Tschopp J, Schroder K (2010) NLRP3 inflammasome activa- (2015) A new way to produce hyperketonemia: use of ketone
tion: the convergence of multiple signalling pathways on ROS ester in a case of Alzheimer’s disease. Alzheimers Dement
production? Nat Rev Immunol 10:210–215 11:99–103
121. Shao B-Z, Xu Z-Q, Han B-Z, Su D-F, Liu C (2015) NLRP3 136. Hertz L, Chen Y, Waagepetersen HS (2015) Effects of ketone
inflammasome and its inhibitors: a review. Front Pharmacol bodies in Alzheimer’s disease in relation to neural hypometabo-
6:262 lism, β-amyloid toxicity, and astrocyte function. J Neurochem
122. Lee G-S, Subramanian N, Kim AI, Aksentijevich I, Gold- 134:7–20
bach-Mansky R, Sacks DB, Germain RN, Kastner DL, Chae 137. Orhan N, Ugur Yilmaz C, Ekizoglu O, Ahishali B, Kucuk M,
JJ (2012) The calcium-sensing receptor regulates the NLRP3 Arican N, Elmas I, Gürses C, Kaya M (2016) Effects of beta-
inflammasome through Ca2+ and cAMP. Nature 492:123–127 hydroxybutyrate on brain vascular permeability in rats with
123. Frahm J, Merboldt KD, Hanicke W (1987) Localized pro- traumatic brain injury. Brain Res 1631:113–126
ton spectroscopy using stimulated echoes. J Magn Reson 138. Jarrett SG, Milder JB, Liang LP, Patel M (2008) The ketogenic
72:502–508 diet increases mitochondrial glutathione levels. J Neurochem
124. Kreis R, Ross B (1992) Cerebral metabolic disturbances in 106:1044–1051
patients with subacute and chronic diabetes mellitus: detection 139. Prins M, Fujima L, Hovda D (2005) Age-dependent reduction
with proton MR spectroscopy. Radiology 184:123–130 of cortical contusion volume by ketones after traumatic brain
125. Plecko B, Stoeckler-Ipsiroglu S, Schober E, Harrer G, Mlynarik injury. J Neurosci Res 82:413–420
V, Gruber S, Moser E, Moeslinger D, Silgoner H, Ipsiroglu O 140. Bröer S, Schneider HP, Bröer A, Rahman B, Hamprecht B,
(2002) Oral &bgr;-hydroxybutyrate supplementation in two Deitmer JW (1998) Characterization of the monocarboxylate
patients with hyperinsulinemic hypoglycemia: monitoring of transporter 1 expressed in Xenopus laevis oocytes by changes in
&bgr;-hydroxybutyrate levels in blood and cerebrospinal fluid, cytosolic pH. Biochem J 333:167–174
and in the brain by in  vivo magnetic resonance spectroscopy.
Pediatric Res 52:301–306

13

You might also like