You are on page 1of 17

Molecular Neurobiology

https://doi.org/10.1007/s12035-020-02065-3

Emerging Therapeutic Promise of Ketogenic Diet to Attenuate


Neuropathological Alterations in Alzheimer’s Disease
Md. Sahab Uddin 1,2 & Md. Tanvir Kabir 3 & Devesh Tewari 4 & Abdullah Al Mamun 1,2 & George E. Barreto 5,6 &
Simona G. Bungau 7 & May N. Bin-Jumah 8 & Mohamed M. Abdel-Daim 9,10 & Ghulam Md Ashraf 11,12

Received: 1 May 2020 / Accepted: 7 August 2020


# Springer Science+Business Media, LLC, part of Springer Nature 2020

Abstract
Alzheimer’s disease (AD) is a multifactorial and chronic neurodegenerative disorder that interferes with memory, thinking, and
behavior. The consumption of dietary fat has been considered a vital factor for AD as this disease is related to blood-brain barrier
function and cholesterol signaling. The ε4 allele of apolipoprotein E (APOE4) is a primary genetic risk factor that encodes one of
many proteins accountable for the transport of cholesterol and it is deemed as the leading cholesterol transport proteins in the
brain. In case of AD development, the causative factor is the high level of serum/plasma cholesterol. However, this statement is
arguable and, in the meantime, the levels of brain cholesterol in individuals with AD are extremely inconstant and levels of
cholesterol in the brain and serum/plasma of AD individuals do not reflect cholesterol as a risk factor. In fact, APOE4 is neither
fundamental nor sufficient for the advancement of AD; it just acts as a synergistic and increases the danger of AD. Another
noticeable characteristic of AD is area-specific decreases in the metabolism of brain glucose. It has been found that the brain cells
cannot efficiently metabolize fats; hence, they totally rely upon glucose as a vitality substrate. Thus, suppression of glucose
metabolism can possess an intense effect on brain actions. Hypometabolism is frequently found in AD and has quite recently
achieved impressive consideration as a plausible target for interfering in the progression of the disease. One promising approach
is to keep up the normal supply of glucose to the brain with ketone bodies from the ketogenic diet signifies a potential therapeutic
agent for AD. Therefore, this review represents the role of ketogenic diets to combat AD pathogenesis by considering the
influence of APOE.

Keywords Ketogenic diets . APOE4 . Alzheimer’s disease . Hypometabolism . Lipids

Abbreviations KBs Ketone bodies


AD Alzheimer’s disease BHB β-Hydroxybutyrate
KD Ketogenic diet ApoE Apolipoprotein E

* Md. Sahab Uddin 6


Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile,
msu-neuropharma@hotmail.com; msu_neuropharma@hotmail.com Santiago, Chile
* George E. Barreto 7
Department of Pharmacy, Faculty of Medicine and Pharmacy,
George.Barreto@ul.ie University of Oradea, Oradea, Romania
* Ghulam Md Ashraf 8
Department of Biology, College of Science, Princess Nourah bint
ashraf.gm@gmail.com; gashraf@kau.edu.sa Abdulrahman University, Riyadh 11474, Saudi Arabia
9
1 Department of Zoology, College of Science, King Saud University,
Department of Pharmacy, Southeast University, Dhaka, Bangladesh P.O. Box 2455, Riyadh 11451, Saudi Arabia
2
Pharmakon Neuroscience Research Network, Dhaka, Bangladesh 10
Pharmacology Department, Faculty of Veterinary Medicine, Suez
3
Department of Pharmacy, Brac University, Dhaka, Bangladesh Canal University, Ismailia 41522, Egypt
4 11
School of Pharmaceutical Sciences, Lovely Professional University, King Fahd Medical Research Center, King Abdulaziz University,
Phagwara, Punjab, India Jeddah, Saudi Arabia
5 12
Department of Biological Sciences, University of Limerick, Department of Medical Laboratory Technology, Faculty of Applied
Limerick, Ireland Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
Mol Neurobiol

Introduction related aspects. For the dysregulation of the mitochondrial


function, both amyloid beta (Aβ) [29, 30] and the ApoE4
Alzheimer’s disease (AD) is the most predominant cause of protein fragmentation have been implicated [11, 31].
dementia in people more than 60 years old, affecting about 44 Additionally, many studies have suggested alterations in insu-
million individuals globally [1–3]. The most well-known ge- lin signaling as an effective supporter of the progression of
netic risk factor for AD is the possession of the ε4 allele of hypometabolism [32–34]. Despite its cause, treatments that
apolipoprotein E (APOE4) [4–6]. Although about 70% of the focused on the correction of cellular metabolism may demon-
risk factor is supposed to be genetic [7], non-genetic risk fac- strate usefulness to individuals with AD [35].
tors have also been associated with AD, including lifestyle- Induction of ketosis is one of such auspicious therapies
related factors of exercise, eating routine, and weight, and [36]. Glucose is the principal energy substrate for the brain,
these factors may represent up to around 33% of AD cases under normal conditions [37]. However, in some cases, for
[8, 9]. In fact, with the concurrence of both the aging people example during prolonged fasting, the liver generates ketone
and increasingly obese and overweight society, comprehen- bodies (KBs) in order to be used by extrahepatic tissues, such
sion regarding the crossing point of eating regimen, genotype, as the brain. Acetone, acetoacetate (ACA), and β-
and cognitive impairment is very important. APOE4 acts as a hydroxybutyrate (BHB) are generally regarded as KBs; for
predominant, dose-dependent mode; one copy of APOE4 in- cells, they serve as an efficient fuel [38, 39] as shown in
creases the risk of developing AD approximately 3-fold, Fig. 1. Interestingly, through adherence to a high-fat, low-
whereas two copies increase the risk around 10-fold [10, carbohydrate ketogenic diet (KD), levels of circulating KBs
11]. AD is clinically featured by the chronic deterioration in can be elevated. AD patients are often found to undergo alter-
memory as well as language, and pathologically by aggrega- ations in food choice toward sweet and carbohydrate-rich
tion of neurofibrillary tangles and senile plaques [12–15]. foods [40, 41]. It is well-known that induction of the hepatic
Regional cerebral hypometabolism is another noticeable production of KBs can be instigated by the diets that contain a
characteristic of AD. On the other hand, low cerebral meta- great amount of fat content and very low carbohydrate [42]. A
bolic rates of glucose utilization in probable AD individuals number of studies have confirmed that KDs have neuropro-
were uncovered by early imaging investigations [16], and this tective effects and improve cognitive functions [42–45].
is currently accepted as a common characteristic of the disease Analysis to explore the therapeutic potentiality of KB and
[17]. It is known that the most generally affected regions in ketosis can be regarded as a new auspicious area of
Alzheimer’s are parietal, posterior cingulate, prefrontal, and Alzheimer’s research [36, 46]. In this review, we represent
temporal regions [18]. Indeed, these impairments can be iden- the therapeutic potential of KBs in AD.
tified in pre-symptomatic, in individuals who are at risk, for
example, E4 carriers, as young adults. Interestingly, such al-
terations have been identified in cognitively normal individ- Glucose Metabolism in Alzheimer’s Disease
uals well before there is extensive loss of neurons or antici-
pated deposition of plaque, which further recommends that A conspicuous characteristic of AD is chronic and area-spe-
low cerebral metabolic rates of glucose (CMRglc) use are an cific, and decreases in the CMRglc. On the other hand, in
early event in the disease [19]. CMRglc across the whole brain, previous investigations com-
Numerous studies have investigated the impact of alter- paring AD patients with normal controls observed a 17 to 24%
ation in lifestyle-related factors in improving AD and their decline. Furthermore, noticeable associations were found
interaction with APOE genotype [20–23]. For instance, taking among cognitive function and CMRglc, such that the low rate
part in regular exercise enhances cognitive functions and re- of CMRglc is linked with low cognitive scores [16].
lated cognitive impairments in AD patients and diminishes Subsequently, a number of imaging studies have validated
amyloid in the brain [24], and APOE4 carriers experience these justifications. In the AD brain, unusually low CMRglc
more noteworthy positive advantages of exercise engagement rates are observed in a characteristic arrangement, predomi-
[25]. Like exercise, a Western diet introduction and related nantly in the areas of posterior cingulate, parietal, temporal,
weight gain to a nation correspond with an increased occur- and prefrontal cortex. In addition, as a diagnostic tool for AD,
rence of AD in that country [26], while consumption of a this arrangement is found to be reproducible and has been
Mediterranean diet is found to be linked with lesser AD risk recommended [18, 47]. Fascinatingly, brain regions
and mild cognitive impairment (MCI) [27]. Nevertheless, exhibiting hypometabolism markedly overlap with areas de-
rather than exercise, dietary intervention seems to be less ef- tected in the default network. In addition, it has been recom-
fective in APOE4 carriers [28]. mended that the utilization of elevated concentrations of glu-
The reason for the hypometabolism remains obscure and cose in these areas might be beneficial to the forthcoming
might be owing to the loss of dendritic fields or neurons. development of cell atrophy, deposition of plaque, and
Hypometabolism has been ascribed to the activity of AD- hypometabolism that describes AD [48].
Mol Neurobiol

Fig. 1 The schematic representation of ketogenic metabolism and typical hydroxybutyrate are converted to acetyl-CoA. On the other hand, in typ-
oxidative glucose metabolism that lead to the formation of energy. During ical oxidative glucose metabolism, glucose is metabolized to acetyl-CoA
fasting, in ketogenic metabolism, lipids are metabolized by β-oxidation via glycolysis. The generated acetyl-CoA then entered into Krebs Cycle
to generated ketone bodies (i.e. acetoacetate, β-hydroxybutyrate, and and ETC to engender ATP. ETC, electron transport chain; ATP, adeno-
acetone). Later by multi-step pathways, acetoacetate and β- sine triphosphate

In AD, a decrease in the regional CMRglc levels might be Like the aforementioned study, in spite of exhibiting no signs
owing to the decrease in the activity or density of glial cells or of weakened cognitive function, E4 carriers had low CMRglc
terminal neuronal fields, a metabolic defect observed within bilaterally in the posterior cingulate and prefrontal, temporal,
glia or neurons, or a mixture of these factors [19]. In a study, and parietal cortices. It was found that 30.7 years was the
Reiman et al. [19] observed the beginning of hypometabolism mean age of the E4 carriers [19]. Henceforth, in the disease
in non-demented subjects at risk for developing AD. The same process, a low level of regional CMRglc seems to be a very
research also observed, in an early experiment, screened indi- early event, well before any medical signs of dementia are
viduals within the ages of 50 and 65 years of age with a family obvious, and well before deposition of plaque or loss of cell
history of AD and who were homozygous for the E4 allele, is expected to have happened.
and thus at greater risk of AD development [18]. On the other The low level of regional CMRglc is not specific for E4
hand, 11 E4/E4 subjects (mean age was 55.4) were selected carriers. In another study, Corder et al. [49] studied 46 patients
and subsequently compared with 22 matched controls (mean in a memory disorder clinic. Among them, 31 subjects were
age was 56.3). Interestingly, in any cognitive test, the controls diagnosed with possible AD; in addition, 4 of them were E2/
and homozygotes did not vary; however, the E4/E4 homozy- E3, 11 were E3/E3, and 16 were E3/E4. In the frontal area,
gotes exhibited declines in the metabolism of glucose in the these 31 individuals showed a mean 11% decline in the level
same areas detected in AD individuals [18]. Then 12 E4 car- of CMRglc. In contrast, in the temporoparietal areas of the
riers (E4/E3) were compared with E4 non-carriers by Reiman cortex, 27 to 31% decrease was observed. Furthermore, each
et al. [19] in a follow-up study investigating young adults. genotype was studied to determine the variations in the level
Mol Neurobiol

of CMRglc, and none were observed. In a different study Another postulate is that the signs of a more general distur-
involving 83 individuals with probable AD, yet again, no bance are altered APP processing and low rate of CMRglc.
variations were observed based on the absence or presence One likely culprit has changed the metabolism of lipid/
of the E4 allele [50]. Nonetheless, some variations among glucose-stimulated either by several factors of environment,
E4(−) and E4(+) participants have been identified via studies for instance, genetic predisposition, or by diet, for example,
involving more advanced voxel-based methods. Since low APOE4 allele possession, or both. In addition, changes in the
rates were identified in other regions of the brain not observed level of lipid metabolism that cause low levels of circulating
in E4(−) individuals, E4(+) individuals might possess more docosahexaenoic acid (DHA) might elevate the risk of AD
global declines in the level of CMRglc [51]. Henceforth, cer- development [60]. Incorrect APP cleavage and weakened ac-
tain variations might be present among E4 carriers in other tions of other sensitive proteins (for example glucose trans-
areas of the brain or in the advancement of the CMRglc de- porters) may be caused by the disruptions in the metabolism of
cline [52]. lipid. The changes might be apparent in the production of
The exact molecular cause of hypometabolism is still reactive oxygen species, amyloid accumulation, and
obscure. The lower level of CMRglc might be owing to hypometabolism. In reality, possibly the changes in the me-
the changes in the processing of either the ApoE4 protein tabolism of lipids are due to the modern style of diet consump-
or amyloid precursor protein (APP). On the other hand, tion and evolutionarily acrimonious diets; all of these are cru-
ApoE4 protein fragments avoid the normal secretory cial to the AD etiology [34, 36].
mechanism and move in the mitochondria, suggested by
Mahley and Huang [31]. The E4 fragments might bind
with F1-ATPase subunits within the mitochondria; there- APOE in Lipid and Glucose Metabolism
fore, they can cause a reduction in the production of cellu-
lar energy [31]. It has been recommended by other re- APOE assumes a key part in the process of lipid metabolism in
searchers that the Aβ peptide produced from the APP pro- the brain through ApoE receptor-mediated uptake of the lipo-
tein cleavage [53, 54] binds heme and can lead to complex protein [61]. ApoE4 allies especially with triglyceride-rich
IV activity loss [55]. However, some other researchers still very-low-density lipoprotein (VLDL) particles whereas
suggest that Aβ peptide–stimulated activity of glycogen ApoE2 and ApoE3 have an inclination for small,
synthase kinase 3β (GSK3β) can lead to a decrease in phospholipid-rich high-density lipoprotein (HDL) in the plas-
energy metabolism and phosphorylation of pyruvate dehy- ma [184]. The only lipoprotein particle in the brain is HDL-
drogenase [56] as given in Fig. 2. In other researches, it has like size particles. Currently, it has been demonstrated that the
been suggested that the impairments in energy metabolism sizes of ApoE particles in the cerebrospinal fluid (CSF) of
might be owing to the low level of expression of energy moderately aged non-demented individuals follow the
metabolism genes in affected regions of the brain, for pattern of ApoE 2/3 > ApoE3/3 > ApoE3/4 > ApoE 4/4
example posterior cingulate neurons [57]. [62]. In aged adults, the sizes of the CSF ApoE complex
Low CMRglc rates are found in the early stage of the dis- follow the pattern of ApoE4-negative > ApoE4-positive.
ease, before greater amounts of Aβ are anticipated to exist, Though, there is no distinction among non-demented and per-
and in the AD individuals who are E4(−). Henceforth, several sons with the mildest dementia in the sizes of CSF ApoE
researchers studied whether disruptions in energy metabolism lipoprotein particles.
precede changes in the processing of APP. Interestingly, it has To examine the impacts of ApoE in the brain metabolome,
been found that a decrease in the production of energy via Lee et al. [63] examined metabolites of young low-density
treating cells with sodium azide can lead to an 80-times in- lipoprotein receptor (LDLR) knockout and ApoE knockout
crease in the production of C-terminal fragment of APP as mice. The serum cholesterol was dramatically increased in
well as aggregation of APP in the secretory pathway [58]. the absence of LDLR or ApoE while having extremely subtle
This increase in fragmentation of APP might be owing to consequences on the lipid metabolism or fatty acids in the
the increased activity of β-secretase (BACE), which is con- brain [63]. It was concluded by the researchers that either
sidered the rate-limiting step for Aβ production. On the other LDLR or ApoE alone may not play an essential role in these
hand, a long-term increase in the levels of BACE1 enzyme mouse models at an initial stage, however, it rather require the
was noticed when acute energy deprivation inducing agents combined impact from various pathways that prompt dysfunc-
(i.e. 2-deoxyglucose and insulin) were used to treat wild-type tion at a considerably later phase of life. In another study
mice. In addition, following treatment, it was observed that conducted by Zhu et al. [64], it was found that brain
increased BACE1 levels were long-lasting and lasted at least phosphoinositol biphosphate (PIP2) was regulated by ApoE3
1 week. Moreover, it was observed that such treatments through suppression of the levels of a PIP2-degrading enzyme,
caused a two-times increase in the levels of Aβ1–40, 1 week phosphoinositol phosphatase synaptojanin 1, whereas ApoE4
after treatment [59]. is non-functional during this process.
Mol Neurobiol

Fig. 2 The regulation of glucose metabolism by Aβ in nerve cells. Aβ As a result, the formation of ATP is reduced that activates AMPK
peptide by reducing the efficiency of oxidative phosphorylation in which stimulates β-oxidation of fatty acid and reduces protein,
mitochondria increases the production of ROS. Aβ causes glycogen synthesis as well as stimulate autophagy. ROS, reactive
phosphorylation of pyruvate dehydrogenase by stimulating GSK3β and oxygen species; GSK3β, glycogen synthase kinase 3β; ETC, electron
blocks the formation of acetyl-CoA from pyruvate as well as exert transport chain; AMPK, AMP-activated protein kinase; ATP, adenosine
deleterious effects on ETC by inhibiting mitochondrial redox carriers. triphosphate

Type 2 diabetes is also related to AD in spite of the fact that vulnerable to glycation and play a pivotal role in converting
it is as yet uncertain whether it is a cause or outcome of AD them into advanced glycation end products (AGEs). In the
[65–68]. The APOE genotype alters the response to insulin brains and cerebrospinal fluid of AD patients, AGEs are ob-
treatment in humans. The connection between insulin resis- served in large amounts [73]. This glycation damage is ac-
tance and Aβ is also controlled by APOE [69]. An investiga- countable for the impairment of ApoE that hampers its capa-
tion was done to show the interaction among insulin, Aβ, and bility to carry lipids to astrocytes, eventually contributing to
APOE isoforms through an APP mouse model that expressed inadequate levels of vital fats like cholesterol in neurons.
human APOE isoforms [70, 71]. The Aβ-induced disability of Regarding this, there is a cascade effect of neuronal dysfunc-
insulin signaling was upgraded by ApoE4 and quickened the tion, mitochondrial dysfunction, and oxidative stress, conse-
cognitive defects that were hippocampal-dependent as com- quently leading to neuronal cell death [72, 74, 75]. The eluci-
pared with ApoE3 [71]. It was also reported that insulin re- dation of the pathogenesis events of AD [72] could strengthen
ceptors that were co-immunoprecipitated with ApoE in the numerous research outcomes that were explained earlier. For
brain lysates of APP/ApoE4 mice were lesser with ApoE in example, the relationship between AD and diabetes can be
comparison with APP/ApoE3 mice which suggested that there owing to the resistance of insulin as well as the excess blood
was a feebler interaction between the insulin receptor and glucose level raising the glycation of ApoE proteins.
ApoE4 when compared with ApoE3 [70]. Since amyloid Moreover, ApoE proteins transcribed and translated by
plaque load is higher in the ApoE4 mice compared with the E4 isoform of the gene are 3 times more AGE-binding
ApoE3 mice, it is difficult to rule out if the higher amount of compared with those of other APOE isoforms [76]. This
Aβ in the APP/ApoE4 mice really advocates the insulin recommends ApoE4-positive people may be more vulner-
resistance. able to the cascade effect suggested by Seneff et al. [72],
Seneff et al. [72] suggested that modern diets, which are which might clarify why they are considered the most risk
low in fats and rich in carbohydrates, produce high blood factors for the development of AD. In addition, this finding
sugar levels after meals, which in turn impair vital proteins, might also elucidate why Henderson et al. [77] observed a
such as the ApoE protein. ApoE proteins are predominantly noteworthy enhancement in ApoE4-negative but not in
Mol Neurobiol

ApoE4-positive people, as the research participants were Hypometabolism as a Therapeutic Target


allowed to eat carbohydrates. Ultimately, lipidated ApoE
levels are responsible for the increase or decrease of the Irrespective of the cause, energy deprivation can be harmful to
levels of Aβ levels, which indicates that the deficiency of neurons. Furthermore, this deprivation may play a pivotal role
lipidated ApoE may contribute to the increase of Aβ, in progressing AD [81, 82]. Thus, enhancing the overall cere-
whereas copiousness of lipidated ApoE possibly improves bral metabolic rate might be beneficial for individuals with
the removal of Aβ [78, 79]. Furthermore, a study found AD. Indeed, one way to overcome this problem is to study
decreased Aβ levels in mice model that ate KDs, as the how the body typically survives with situations of low glucose
KDs potentially lessen glycation via dropping blood glu- availability and if such circumstances may be exogenously
cose levels, but also increase the availability of lipids, and used to AD. Extended starvation or fasting can be a simple
both effects may contribute to increased lipidated ApoE way to study the response toward low glucose availability. In
[80]. Additionally, this increase in existing lipidated classic studies investigating brain metabolism during fasting,
ApoE may supply astrocytes with vital facts that are sig- Owen et al. [83] studied the utilization of substrate when
nificant for the growth as well as the function of neurons, obese participants went through 5 to 6 weeks of complete
possibly stopping the cascade effect suggested by Seneff starvation. Participants of that study were limited to the intake
et al. [72] as shown in Fig. 3. These mounting proofs rec- of 1.5-l water, 17 mEq of sodium chloride, and one multivi-
ommend that glycation of the ApoE protein and the resul- tamin capsule per day during the period of starvation. In ad-
tant downstream effects may elucidate factors of the AD dition, glucose synthesis rates were also calculated during the
pathogenesis, as well as KDs, which may improve damage starvation period. Glucose is mainly generated under such
through the stabilization of blood glucose level and in- circumstances from the catabolism of proteins, lactate from
crease the availability of vital fats such as cholesterol. glycolytic tissues, or glycerol from fat stores. It was

Fig. 3 The role of high


carbohydrate diets in the
pathogenesis of Alzheimer’s
disease. ApoE protein modulates
the biosynthesis of lipoproteins.
Thus, generated ApoE-labeled li-
poproteins are transported intact
across the endothelium of the
blood-brain barrier and taken up
by astrocytes and finally trans-
ferred to neurons for neuronal
metabolism. On the other hand,
high carbohydrate diets cause
glycation of ApoE and block the
formation of lipoproteins which
in turn starves neuron and causes
neurodegeneration reported in
AD. ApoE, apolipoprotein E
Mol Neurobiol

determined based on rates of nitrogen excretion and other [89, 90]; nonetheless, in these prior investigations, fat was
factors that total glucose synthesis and availability to the body introduced to the eating regimen without a marked reduction
during starvation was around 33 g glucose/day, and this was in the content of carbohydrate. It is dubious these animals
below the brain’s normal oxidation of 110 g/day. The brain generated KB. In these study designs, the distinctions feature
will obtain most of its required energy from the oxidation of the remark that not all high-fat diets are high-fat diets [91].
KBs under such fasting conditions. On the other hand, during Animal models have been used by the researchers for a
the period of starvation, the concentration of the major better understanding of how ketosis may provide protection
KB, BHB was within the range of 4–8 mM, 10- to 20-times to the human brain against neurodegeneration. Yin et al. [92]
over normal fasting values. Furthermore, it is evaluated that elucidated the relationship between the metabolic benefits of
approximately 60% of the brain’s energy requirements are ketones, lower Aβ, and enhanced cognitive function. Indeed,
provided by the supply of KBs under such scenarios [83]. a novel neuroprotective pathway has been discovered by this
Amino acids are the main sources of de novo synthesized study in which they blocked the entry of Aβ1–42 into neurons.
glucose. A considerable tissue (mainly muscle) breakdown is It was found that inhibition of intracellular Aβ1–42 accumula-
required for the brain in order to provide high glucose levels. It tion ameliorated synaptic plasticity, decreased oxidative
has been observed that such lean body mass loss during pe- stress, and rescued mitochondrial complex I effects. In the
riods of food insufficiency would likely be maladaptive. As an symptomatic AD mouse model, it was also revealed by this
alternative, the use of KBs permits for the tapping of abundant study that peripheral administration of ketones caused a
fat stores [83]. marked reduction of Aβ burden and also significantly en-
hanced memory and learning ability [92]. Collectively, these
findings provide a better understanding and also can help to
Ketogenic Diet and Alzheimer’s Hallmarks establish a novel therapeutic use of ketones for AD treatment.

The capability of KBs to enhance the mitochondria efficacy


and glucose supplementation makes them alluring compounds Therapeutic Potential of Ketogenic Diet
for the treatment of AD [84]. A ketosis condition can be ac- to Combat Alzheimer’s Pathology
tuated via enhanced levels of free fatty acid (FFA) circulation
and encouraging oxidation of fatty acid. This can be accom- The capacity of the brain in glucose utilization seems to be
plished through adherence to KDs. The KDs gained attention pathologically decreased in AD [93, 94]. Neurobiological
when it developed to decrease seizure incidence in epileptics. proofs revealed that the KBs are successful elective energy
These diets are effectively utilized for a long time for the substrate for the brain [95, 96]. Increase in the levels of plasma
treatment of refractive childhood epilepsy. The KDs have KBs by medium chain triglycerides (MCTs) oral dose might
likewise been utilized effectively to moderate the manifesta- enhance cognitive function in adult people with memory dis-
tions of glucose deprivation, for example those that happen in orders. It has been confirmed by several studies that KDs are
glucose transporter type 1 deficiency syndrome [85]. effective in treating AD as given in Table 1 and Table 2.
Including AD, KDs have exhibited potential in the treat- Reduced levels of blood glucose and increased KB concentra-
ment of many neurological conditions like amyotrophic hori- tion are the key features of the neuroprotective effects of KD
zontal sclerosis [86], traumatic brain injury [87], and (Fig. 4). Additionally, KBs increased the action of brain-
Parkinson’s disease [194]. In a study, Van der Auwera et al. specific uncoupling proteins (UCP), which decrease the gen-
[80] directly assessed a KD in an AD animal model. In addi- eration of reactive oxygen species (ROS) through mitochon-
tion to this, this investigation utilized a transgenic line of mice drial complex I, moderate neuron dysfunction, and neurode-
that were expressing mutation of APP (V717I) that was driven generation [97, 98]. The expression of UCP is induced by
via THY1 promoter. It has been observed that these animals ketosis and it also upregulates many genes associated with
demonstrate noteworthy levels of soluble Aβ in as little as the energy metabolism of oxidation through the nuclear tran-
3 months, and widespread deposition of plaque by 12 to scription factor peroxisome proliferator-activated receptor-α
14 months of age [88]. In this study, either standard chow or (PPARα) and peroxisome proliferator-activated receptor γ
ketogenic chow (i.e. composed mainly of lard and butterfat, coactivator-1 (PGC-1α) [97, 98]. Ketosis is reported to stim-
mainly saturated fat and lesser carbohydrate) was feed for ulate the mitochondrial biogenesis, hence produces higher
43 days to 3-month-old, sixteen female mice. During the treat- adenosine triphosphate (ATP) and also increases energy re-
ment time, the levels of KBs were substantially enhanced in serves, which are recognized to stabilize the synaptic activity.
the range of 2–9 mM, and finally, it was found that there was a Perhaps, the ratio of increased phosphocreatine to creatinine
decrease of 25% in the levels of Aβ40 and Aβ42 [80]. (PCr:Cr) energy reserve might potentiate GABAergic output,
Initially, this outcome may appear inconsistent with prior ex- which is most probably linked with the enhanced γ-
aminations that ascribed a high-fat diet to enhance Aβ loads aminobutyric acid (GABA) synthesis triggered by ketosis
Mol Neurobiol

Table 1 Promising preclinical


studies regarding the Species/ Experimental model Outcomes References
neuroprotective effect of studied
ketogenic diet against material
neurodegeneration
Mice Alzheimer’s disease Reduced Aβ levels [80]
(AD)
Cell culture AD Increased cell survival [100]
Rats Hypoxia ischemia Neuroprotection [101]
Cell culture Glutamate toxicity Increased mitochondrial efficiency [102]
Cell culture Glutamate toxicity Neuroprotection [103]
Cell culture Hypoxia Increased cell survival [104]
Rats Glutamate toxicity Neuroprotection and reduced lipid peroxidation [105]
Cell culture Glutamate toxicity Increased cell survival [106]
Mice Hypoxia Maintained adenosine triphosphate ) and low lactate [107]
Mice Ischemia Decreased cerebral edema formation and infarct area [108]
Mice Ketogenic diet Enhanced mitochondrial function, reduced reactive [46]
treatment oxygen species generation, and elevated levels of
cerebral ATP
Rats Ketogenic diet Decreased levels of insulin, phosphorylation of S6 and [109]
treatment protein kinase B, and reduced activation of
mammalian target of rapamycin
Mice Hypercholesterolemia Elevated transcriptional activity of peroxisome [110]
proliferator-activated receptor γ and suppressed his-
tone deacetylases
Mice AD Decreased Aβ deposition, and enhanced energy [111–114]
metabolism and motor function
Mice AD Improved tau and Aβ pathology, and enhanced [115]
cognitive performance

Table 2 Promising clinical studies regarding the neuroprotective effect of ketogenic diet against Alzheimer’s disease

Type of study Participants Disease condition Duration Outcomes References


of the participants

Randomized, double-blind, 152 Mild to moderate 90 days AC-1202 rapidly elevated serum ketone bodies [77]
placebo-controlled, Alzheimer’s (β-hydroxybutyrate) that resulted in noticeably en-
parallel-group study disease (AD) hanced ADAS-Cog score as compared to placebo
Longitudinal, open-label study 20 Mild-to-moderate 12 weeks Medium-chain triglyceride (MCT)-based ketogenic diet [116]
AD improved cognitive performance, verbal memory,
and processing speed
Randomized study 23 Mild cognitive 6 weeks Very low-carbohydrate consumption, even in the short [117]
impairment term, can improve memory function
(MCI)
Single-arm, pilot clinical study 15 Mild-to-moderate 3 months MCT supplemented ketogenic diet enhanced the [118]
AD ADAS-Cog score
Randomized placebo-controlled 6 MCI 24 weeks MCT supplementation increased serum ketone bodies [119]
study and improved memory as compared to placebo
Double-blind, placebo-controlled 20 AD or MCI 90 min MCT-based ketogenic diet significantly increased level [120]
study of β-hydroxybutyrate (i.e. higher in case of ApoE4(+
) as compared to APOE4(−));
Enhanced cognitive function and memory in the
participants; APOE4(+) carriers were less responsive
to ketogenic diet as compared to APOE4(−)
Prospective, open-label, 22 Mild-to-moderate 90 days Axona did not improve cognitive function [121]
observational study AD
Singe-patient case study 1 Early-onset AD 20 months Ketone monoester noticeably enhanced the daily [122]
activities, self-care, and mood;
Enhanced cognitive function in APOE4(+) patient
Mol Neurobiol

[97–99]. Augmented acetone levels are linked with KB effect higher ketone was related to the more noteworthy change in
and this might activate K2P channels that limit neuronal excit- memory enhancements (passage recall) with MCTs treatment
ability and hyperpolarize neurons [97, 99]. The change in the with respect to placebo overall individuals. Further research is
metabolism of glutamate and GABA in the brain is also af- justified to decide the impact of MCTs for AD patients and
fected by KBs. how APOE4 might intervene in the BHB effect.
In a study conducted by Reger et al. [123], it was found that Henderson et al. [77] later on followed up these short-term,
on different days, 20 individuals with MCI or AD were ad- initial results through the use of AC-1202 (i.e. an oral keto-
ministered a drink comprising emulsified MCTs or placebo. genic compound) among 152 people (with possible AD) for
There was a significant increase in levels of KB, BHB after the duration of 90 days. It has been observed that as compared
90 min of treatment when administered the subjective tests, with placebo, AC-1202 markedly increased the level of a se-
and the moderation of BHB increases was directed through rum BHB following 2 h of its administration. It was observed
APOE. in each of the population groups that there was a noticeable
In the case of ε4+ individuals, levels of BHB kept on as- difference between AC-1202 and placebo in terms of mean
cending between the 90- and 120-min blood draws in the change from baseline in ADAS-Cog score on the 45th day. On
treatment condition, while the levels of BHB of ε4− individ- the other hand, it was found among the individuals who did
uals held consistently. On the other hand, in the case of cog- not contain the APOE4 allele (E4(−)) that there was a marked
nition test, MCTs treatment encouraged execution on the AD difference between AC-1202 and placebo in terms of mean
Assessment Scale-Cognitive Subscale (ADAS-Cog) for ε4− alteration from baseline in ADAS-Cog score on the 45th and
subjects, yet not for ε4+ individuals. It has been found that 90th day. In case of intention-to-treat population, it was

Fig. 4 Possible neuroprotective


effects of the ketogenic diet to
combat Alzheimer’s disease.
Ketogenic diet induces metabolic
shifts by increasing fatty acids
and reducing glucose levels.
Elevated fatty acids are suggested
to induce ketosis and finally
causes a reduction of neuronal
excitability and vesicular
glutamate transporters that lead to
the stabilization of synaptic
activity. On the other hand,
accompanying decreased glucose
availability increased oxidative
phosphorylation of ATP thus
enhanced energy reserves that
stabilize synaptic activity and
finally increased GABAergic
output. Ketogenic diets also
increase the activity of
uncoupling proteins that lead to
the reduction of ROS and
stimulate mitochondrial
biogenesis thus leading to
enhanced energy reserves that
stabilize synaptic activity and
finally potentiate GABAergic
output. ATP, adenosine
triphosphate; ROS, reactive
oxygen species
Mol Neurobiol

observed that when AC-1202 was administered in E4(−) par- manner, ketone levels were correlated with memory perfor-
ticipants (N = 55), a marked 4.77 point difference was ob- mance [117]. Collectively, these results denote that, even in
served in mean change from baseline in ADAS-Cog scores the short term, consumption of very low carbohydrate can lead
on the 45th day and a 3.36 point difference on the 90th day in to an improvement in the memory function in older people
comparison with placebo. Interestingly, it has been observed who are at increased risk for AD. This observation might be
in the case of per-protocol (PP) population that when AC- due to the ability to correct hyperinsulinemia and other pro-
1202 was administered to E4(−) subjects (N = 37), there was cesses linked with ketosis, for example increased energy me-
a 5.73 point difference on 45th day and a 4.39 point difference tabolism and decreased inflammation might also be able to
on the 90th day. On the other hand, in the case of dosage contribute to the improvement of neurocognitive function. In
compliant population, it was observed that when AC-1202 the context of early neurodegeneration, more studies are re-
was administered to E4(−) subjects (N = 38), there was a quired in order to assess its mechanisms of action and preven-
6.26 point difference on 45th day and a 5.33 point difference tive potential.
on the 90th day (N = 35). In addition to this, in E4(−) subjects, Established approaches in order to induce therapeutic
a noteworthy pharmacologic response was detected between hyperketonemia have entailed adherence to KDs [125] and/
change in ADAS-Cog scores and the levels of serum BHB on or repetitive medium-chain triglyceride (MCTG) administra-
the 90th day [77]. It was observed that adverse events were tion [77]. In caregivers and patients, the strong drive is re-
more prevalent among the participants who received AC- quired in order to successfully adhere to the classical KDs
1202. Nevertheless, those adverse events were limited to the containing very low carbohydrate and very high fat.
gastrointestinal system and were largely mild to moderate in However, liberalized versions might offer clinical benefits at
severity and transient in nature. lower concentrations of plasma ketone [125]. Chronic intake
As stated earlier, multiple AD risk factors include lifestyle of KDs might also elevate the atherogenic lipid levels and
and genetic. Lifestyle risk factors include exercise and diet, generate other adverse effects [77, 125]. It has been observed
while genetic risk factors include possession of APOE4 over that following the consumption of a very-low-carbohydrate
risk-neutral APOE3. However, the connection of these risk diet for 8 weeks, a group of MCI containing elderly people
factors with the disease is poorly understood. In a study, showed enhanced verbal memory performance [117].
Lane-Donovan et al. [124] demonstrated the effect of diet on MCTG-administering individuals can also increase con-
ApoE levels by feeding wild-type and ApoE3- and ApoE4- centrations of plasma ketone while they are on their usual diet.
targeted replacement mice with chow, ketogenic (i.e. very Individuals with AD and MCI were given 20 g of MCTG per
low-carbohydrate, high-fat), or high-fat diets. Moreover, they day in a cohort study; it was observed that concentrations of
also observed that the high-fat diet has an impact on both BHB increased from ~ 0.09 mM (pre-dose) to 0.3– 0.4 mM
hippocampal and plasma levels of ApoE in an isoform- (i.e. 2 h post-dose). However, only among the individuals who
dependent mode. In addition to this, a high-fat diet can cause are APOE4-negative did marked improvements in their cog-
a marked decrease in the hippocampal levels of ApoE in nitive performances during hyperketonemia exhibit [77].
ApoE3 targeted replacement mice. In contrast, it was observed Indeed, the highest levels of KBs were found to possess the
that KDs has no effect on the hippocampal levels of ApoE most benefit. Due to the minor gastrointestinal side effects,
[124]. Collectively, these results recommend that APOE ge- around 12.5% of the individuals discontinued their
notypes should be taken into account while using the dietary participation.
interventions in order to slow the AD progression. In fact, high concentrations of plasma KBs (6–8 mM) can
These promising findings of ketogenic compounds suggest be maintained for an indefinite period when ketone monoester
that dietary interventions might be beneficial for brain health. (KME) is consumed at 3–4-h intervals, which might increase
In a study, Krikorian et al. [117] randomly assigned MCI concerns regarding the long-term safety of this extent of
containing 23 older adults to either a high or very low hyperketonemia. Indeed, strict adherence to a very-low-
carbohydrate-containing diet. Interestingly, after 6-week inter- carbohydrate and very-high-fat KDs or prolonged fasting
vention period, it was found in the low-carbohydrate subjects was the only way to achieve comparable elevations of plasma
that there was an improvement in the verbal memory perfor- KBs before KME became available [83]. Nevertheless, these
mance along with a decrease in fasting insulin, fasting glu- approaches contain various metabolic side effects including
cose, waist circumference, and weight. However, symptoms amenorrhea, marked weight changes, urate nephrolithiasis,
of depression were found to be not affected in that study. and dehydration [126]. Furthermore, these methods cannot
Interestingly, for the entire sample, alterations in weight, in- play a key role as a satisfactory test in order to determine the
sulin level, and calorie intake were not linked with memory long-term safety of hyperketonemia induced by KME.
performance, even though it was observed within the low- Interestingly, while going through 5–6 weeks of starvation
carbohydrate group that there was a trend toward a moderate for weight loss, 3 severely obese participants (i.e. who
relationship between memory and insulin. In a positive sustained mean concentrations of KB at ~ 8 mM) did not show
Mol Neurobiol

any adverse clinical effects which can be attributed to the between KME and plasma BHB, concentrations of plasma
prolonged hyperketonemia [83]. BHB were estimated before and during treatment with
In rare cases, it has been observed that in hyperketonemia, KME. Interestingly, significant alterations were observed only
plasma concentrations of uric acid can get increased due to the in the case of certain plasma lipids. It was noticed that a total
decreased renal clearance of uric acid, which can further cause cholesterol level dropped from a pre-KME mean (n = 2) of
precipitation of a flare in gout-susceptible people [127]. This 244 to 163 mg/dl. In contrast, low-density lipoprotein (LDL)
complication can be prevented by the drugs which can inhibit cholesterol dropped from 145 to 81 mg/dl and HDL choles-
uric acid synthesis. Presently, there is no strong proof that terol from 85 to 68 mg/dl. It needs to be noted that the partic-
chronic hyperketonemia (i.e. at concentrations of ~ 8 mM) ipant was an APOE4 carrier; therefore, the researchers con-
might be clinically unsafe. Nevertheless, if valid safety con- cluded that ketosis can be substantially beneficial for the
cerns are increased, then they should be regarded in the light APOE4 carriers, although former studies revealed that ketosis
of a probable beneficial action of KME on devastating disor- is even more beneficial for the individuals with this risk factor
ders like AD, and on disabling convulsions in children and [122].
infants who are refractory to anti-seizure drugs [128]. In addition, MCT-supplemented ≥ 1:1 ratio KD was ad-
In a study, Henderson et al. [77] reported that APOE4- ministered for 3 months in 15 mild-to-moderate AD patients
positive participants failed to exhibit statistically significant in a single-arm pilot trial [118]. In that study, improvement in
and enhanced ADAS-Cog scores in response to the treatment cognitive functions was observed in 9 out of 10 individuals
with MCTG. Indeed, the patient (although APOE4-positive) who successfully finished the study and achieved ketosis
showed clear enhancement in their behavior and cognitive [118]. Furthermore, MCT-supplemented KD was given to
functions, while consuming larger or equivalent amounts of 15 AD individuals (around 70% of energy as fat) in a different
ketogenic, medium-chain fatty acids. However, the results of study named Ketogenic Diet Retention and Feasibility Trial.
Henderson et al. [77] do not exclude the chance that APOE4 ADAS-Cog test was considerably ameliorated during the KD,
allele carriers may exhibit improvements in their cognitive once the ketosis was attained [118]. In a study, MCT was
functions if higher MCTG doses are given and/or studied for provided to 20 Japanese mild-to-moderate AD individuals
longer periods of time. for more than 12 weeks [116]. Following 120 min of admin-
In formal clinical trials, the therapeutic potential of the istration, the level of KBs was elevated; then, following
ketosis has not been robustly studied. However, Newport 8 weeks, the individuals showed a marked enhancement in
et al. [122] in their recent study revealed a surprising thera- their delayed and immediate memory tests in comparison with
peutic target in order to control AD. Interestingly, in that their baseline score [116].
study, an elderly person (i.e. aged 63 years) with sporadic, Currently, several randomized clinical trials by the
younger onset AD was administered 35 ml of coconut oil University of British Columbia [129], Universite de
(CO) on a daily basis. It has been found that CO contains ~ Sherbrooke [130], Wake Forest University [131–133], and
15% ketogenic medium-chain fatty acids (MCFA). MCTG Johns Hopkins University [134] are conducted to explore
was introduced after several months and elevated progressive- more evidence about the effect of KD on AD [135].
ly to a 4:3 mixture with CO, ultimately reaching 165 ml/day Participants in these studies with mild AD, subjective memory
divided into 3 to 4 servings. After 2.5 months, rapid improve- impairment, and/or healthy controls were utilized to assess the
ment in the score of Mini-Mental State Examination (MMSE) influence of 6–18 weeks of the modified ketogenic-
from 12 to 20 was observed due to the use of CO/MCTG Mediterranean diet to a low-fat diet; 12 weeks of the modified
mixtures as dietary supplements. In addition to this, gradual Atkins diet in comparison with the recommended diet for
amelioration has also occurred in the recall of recent events, seniors to attain a healthy eating index; 1-month treatment
word-finding, social participation, and gait. Therefore, during with the two different MCTs oil emulsions (C8 oil or 40–60
treatment with MCFA for 20 months, it was observed that the oil), or 10 days, two times a day, and administration of a
patient’s activities of daily living increased to 14 points and lactose-free skim milk drink containing either 10–50 g/day
the ADAS-Cog score was increased to 6 points, followed by of placebo (high-oleic sunflower oil) or 10–50 g/day of
stabilization. No further brain atrophy was observed in the MCT oil.
patient’s magnetic resonance imaging. Subsequently, KME Another probable effect of KD on the progression of AD
was introduced in the diet of the patient. Henceforth, improve- has also been revealed. Studies have suggested that an elevat-
ments were observed in cognitive function, living functions, ed dietary supply of unsaturated fatty acids (particularly ome-
and behaviors of the patient. Following 6 to 8 weeks of con- ga 3 and polyunsaturated fatty acids) might lower the risk of
sumption of 28.7 g KME three times a day, the participant this condition [136–138]. Possibly, the useful effect of KD on
started to show amelioration in memory retrieval, spontane- the improvement of AD is linked with an increased supply of
ously discussing events that took place up to 7 days earlier. unsaturated fatty acids versus a high or normal-carbohydrate
Moreover, in order to estimate the dose-response relationships diet [42].
Mol Neurobiol

Adverse Effects of the Ketogenic Diet Furthermore, the identification of maximum and minimum
effective dose is imperative to settle the Alzheimer’s treatment
Limited data is available on adverse effects of KD in adults; regimen. Notwithstanding these questions, the therapeutic
however, some predictable effects are hypoglycemia and de- promise of KDs recommends a great weapon against AD
hydration. Other adverse effects are generally less common pathogenesis.
and present following long-term intake. Previously, KBs were
considered toxic which is an outcome of the association of Author Contributions MSU conceived the original idea and designed the
outlines of the study. MSU, MTK, DT, and AAM wrote the draft of the
therapeutic ketosis with diabetic ketoacidosis, which leads to
manuscript. MSU prepared the figures for the manuscript. GEB, SGB,
higher ketone concentrations over 20 mM, and can be re- MNB-J, MMA-D, and GMA revised and improved the manuscript. All
versed by administration of insulin [139]. It is notable that in authors have read and approved the final manuscript.
severe cases, hyperketonemia developed by insulin deficiency
may cause severe acidosis, sometimes death [139, 140]. Some Acknowledgments This work was funded by Deanship of Scientific
adverse effects on KD like weight loss, gastrointestinal ef- Research at Princess Nourah bint Abdulrahman University through the
Fast-Track Research Funding Program. This work was supported by
fects, and transient hyperlipidemia were reported frequently King Saud University, Deanship of Scientific Research, College of
by patients with epilepsy [141]. The side effects related to Science Research Center.
gastrointestinal are nausea, vomiting, constipation, and lower
appetite [141, 142]. Moreover, lipid profile change like tri- Compliance with Ethical Standards
glycerides, fasting total serum cholesterol, and LDL choles-
terol is initially increased during KD treatment and normalizes Competing Interests The authors declare that they have no competing
after ~ 1 year [143]. Apart from these, hepatitis, dehydration, interests.
hypoglycemia, hyperuricemia, pancreatitis, hypomagnesemia,
hypertransaminemia, and hyponatremia are some of the major
adverse effects of the KD [142, 144]. On the prolonged KD
References
treatment, it may increase cardiomyopathy, anemia,
nephrolithiasis, atherosclerosis, neuropathy of the optic nerve, 1. Uddin MS, Mamun AA, Takeda S et al (2019) Analyzing the
impaired hepatic functions, decrease of mineral bone density, chance of developing dementia among geriatric people: a cross-
and also the deficits of vitamins and minerals [99, 144]. sectional pilot study in Bangladesh. Psychogeriatrics 19:87–94.
https://doi.org/10.1111/psyg.12368
Chronic treatment with KD might lead to disturbances in
2. Kabir MT, Uddin MS, Begum MM, Thangapandiyan S, Rahman
catabolism and decreased production of functional proteins. MS, Aleya L, Mathew B, Ahmed M et al (2019) Cholinesterase
Bearing in mind the lower organoleptic attractiveness and loss inhibitors for Alzheimer’s disease: multitargeting strategy based
of appetite, it is likely to be problematic to attain a proper on anti-Alzheimer’s drugs repositioning. Curr Pharm Des 25:
supply of energy and protein in individuals on the KD, since 3519–3535. https://doi.org/10.2174/
1381612825666191008103141
an inadequate intake of protein or any energetic deficiency
3. Zaplatic E, Bule M, Shah SZA, Uddin MS, Niaz K (2019)
might result in severe consequences for health [118, 144]. Molecular mechanisms underlying protective role of quercetin in
Interestingly, when KD was provided for 24 weeks, no note- attenuating Alzheimer’s disease. Life Sci 224:109–119. https://
worthy adverse effects were seen in 83 obese individuals doi.org/10.1016/j.lfs.2019.03.055
[145]. Furthermore, in AD individuals, KD might consider- 4. Corder E, Saunders A, Strittmatter W et al (1993) Gene dose of
apolipoprotein E type 4 allele and the risk of Alzheimer’s disease
ably influence food intake through disturbances in the senses in late onset families. Science (80-) 261:921–923. https://doi.org/
of taste and smell; neurological symptoms, for instance, be- 10.1126/science.8346443
havioral disturbances; dysphagia; and apraxia disturbances 5. Uddin MS, Haque A, Mamun AA et al (2016) Searching the
during eating [144]. linkage between high fat diet and Alzheimer’s disease: a debatable
proof stand for Ketogenic diet to alleviate symptoms of
Alzheimer’s patient with APOE ε4 allele. J Neurol
Neurophysiol 07:1–9. https://doi.org/10.4172/2155-9562.
Conclusion 1000397
6. Hossain MF, Uddin MS, Uddin GMS, Sumsuzzman DM, Islam
The auspicious effect of KBs gives a new way to combat AD MS, Barreto GE, Mathew B, Ashraf GM (2019) Melatonin in
Alzheimer’s disease: a latent endogenous regulator of
pathogenesis by a dietary intervention. For the effective AD neurogenesis to mitigate Alzheimer’s neuropathology. Mol
treatment, if ketosis becomes avowed, further studies are vital Neurobiol 58:8255–8276. https://doi.org/10.1007/s12035-019-
to establishing the best possible mode of administration, since 01660-3
MCFA, diets containing high-fat, ketogenic compounds, or 7. Uddin MS, Al Mamun A, Kabir MT et al (2019) Nootropic and
anti-Alzheimer’s actions of medicinal plants: molecular insight
MCT supplements might be individually effective. into therapeutic potential to alleviate Alzheimer’s neuropathology.
Additionally, patient compliance could also indicate chal- Mol Neurobiol 56:4925–4944. https://doi.org/10.1007/s12035-
lenges due to the administration of a strict diet or supplements. 018-1420-2
Mol Neurobiol

8. Norton S, Matthews FE, Barnes DE, Yaffe K, Brayne C (2014) 24. Heyn P, Abreu BC, Ottenbacher KJ (2004) The effects of exercise
Potential for primary prevention of Alzheimer’s disease: an anal- training on elderly persons with cognitive impairment and demen-
ysis of population-based data. Lancet Neurol 13:788–794. https:// tia: a meta-analysis. Arch Phys Med Rehabil 85:1694–1704
doi.org/10.1016/S1474-4422(14)70136-X 25. Bugg JM, Bugg JM, Goate AM et al (2012) Exercise engagement
9. Kabir MT, Abu Sufian M, Uddin MS et al (2019) NMDA receptor as a moderator of the effects of APOE genotype on amyloid de-
antagonists: repositioning of memantine as multitargeting agent position. Arch Neurol 69:636–643. https://doi.org/10.1001/
for Alzheimer’s therapy. Curr Pharm Des 25:3506–3518. https:// archneurol.2011.845
doi.org/10.2174/1381612825666191011102444 26. Grant WB (2013) Trends in diet and Alzheimer’s disease during
10. Strittmatter WJ, Roses AD (1996) Apolipoprotein E and the nutrition transition in Japan and developing countries. J
Alzheimer’s disease. Annu Rev Neurosci 19:53–77. https://doi. Alzheimers Dis 38:611–620. https://doi.org/10.3233/JAD-
org/10.1146/annurev.ne.19.030196.000413 130719
11. Uddin MS, Kabir MT, Al Mamun A et al (2019) APOE and 27. Singh B, Parsaik AK, Mielke MM, Erwin PJ, Knopman DS,
Alzheimer’s disease: evidence mounts that targeting APOE4 Petersen RC, Roberts RO (2014) Association of Mediterranean
may combat Alzheimer’s pathogenesis. Mol Neurobiol 56: diet with mild cognitive impairment and Alzheimer’s disease: a
2450–2465. https://doi.org/10.1007/s12035-018-1237-z systematic review and meta-analysis. J Alzheimers Dis 39:271–
12. Sharma P, Sharma A, Fayaz F et al (2020) Biological signatures of 282. https://doi.org/10.3233/JAD-130830
Alzheimer’s disease. Curr Top Med Chem 20:770–781. https:// 28. Martínez-Lapiscina EH, Galbete C, Corella D, Toledo E, Buil-
doi.org/10.2174/1568026620666200228095553 Cosiales P, Salas-Salvado J, Ros E, Martinez-Gonzalez MÁ
(2014) Genotype patterns at CLU, CR1, PICALM and APOE,
13. Uddin MS, Kabir MT, Tewari D, Mathew B, Aleya L (2020)
cognition and Mediterranean diet: the PREDIMED-NAVARRA
Emerging signal regulating potential of small molecule
trial. Genes Nutr 9:393. https://doi.org/10.1007/s12263-014-
biflavonoids to combat neuropathological insults of Alzheimer’s
0393-7
disease. Sci Total Environ 700:134836. https://doi.org/10.1016/j.
29. Onyango IG, Dennis J, Khan SM (2016) Mitochondrial dysfunc-
scitotenv.2019.134836
tion in Alzheimer’s disease and the rationale for bioenergetics
14. Uddin MS, Al Mamun A, Asaduzzaman M et al (2018) Spectrum based therapies. Aging Dis 7:201. https://doi.org/10.14336/AD.
of disease and prescription pattern for outpatients with neurolog- 2015.1007
ical disorders: an empirical pilot study in Bangladesh. Ann 30. Uddin MS, Mamun AA, Labu ZK et al (2019) Autophagic dys-
Neurosci 25:25–37. https://doi.org/10.1159/000481812 function in Alzheimer’s disease: cellular and molecular mechanis-
15. Mamun AA, Uddin MS, Mathew B, Ashraf GM (2020) Toxic tau: tic approaches to halt Alzheimer’s pathogenesis. J Cell Physiol
Structural origins of tau aggregation in Alzheimer’s disease. 234:8094–8112. https://doi.org/10.1002/jcp.27588
Neural Regen Res 15:1417–1420. https://doi.org/10.4103/1673- 31. Mahley RW, Huang Y (2006) Apolipoprotein (apo) E4 and
5374.274329 Alzheimer’s disease: unique conformational and biophysical
16. de Leon MJ, Ferris SH, George AE et al (1983) Positron emission properties of apoE4 can modulate neuropathology. Acta Neurol
tomographic studies of aging and Alzheimer disease. AJNR Am J Scand 114:8–14. https://doi.org/10.1111/j.1600-0404.2006.
Neuroradiol 4:568–571 00679.x
17. Mosconi L, De Santi S, Rusinek H et al (2004) Magnetic reso- 32. Hoyer S (1992) Oxidative energy metabolism in Alzheimer brain.
nance and PET studies in the early diagnosis of Alzheimer’s dis- Studies in early-onset and late-onset cases. Mol Chem
ease. Expert Rev Neurother 4:831–849. https://doi.org/10.1586/ Neuropathol 16:207–224
14737175.4.5.831 33. Lannert H, Hoyer S (1998) Intracerebroventricular administration
18. Reiman EM, Caselli RJ, Yun LS, Chen K, Bandy D, Minoshima of streptozotocin causes long-term diminutions in learning and
S, Thibodeau SN, Osborne D (1996) Preclinical evidence of memory abilities and in cerebral energy metabolism in adult rats.
Alzheimer’s disease in persons homozygous for the ε4 allele for Behav Neurosci 112:1199–1208
apolipoprotein E. N Engl J Med 334:752–758. https://doi.org/10. 34. Henderson ST (2004) High carbohydrate diets and Alzheimer’s
1056/NEJM199603213341202 disease. Med Hypotheses 62:689–700. https://doi.org/10.1016/j.
19. Reiman EM, Chen K, Alexander GE, Caselli RJ, Bandy D, mehy.2003.11.028
Osborne D, Saunders AM, Hardy J (2004) Functional brain ab- 35. Costantini LC, Barr LJ, Vogel JL, Henderson ST (2008)
normalities in young adults at genetic risk for late-onset Hypometabolism as a therapeutic target in Alzheimer’s disease.
Alzheimer’s dementia. Proc Natl Acad Sci 101:284–289. https:// BMC Neurosci 9:S16. https://doi.org/10.1186/1471-2202-9-S2-
doi.org/10.1073/pnas.2635903100 S16
20. O’Donoghue MC, Murphy SE, Zamboni G et al (2018) APOE 36. Henderson ST (2008) Ketone bodies as a therapeutic for
genotype and cognition in healthy individuals at risk of Alzheimer’s disease. Neurotherapeutics 5:470–480. https://doi.
Alzheimer’s disease: a review. Cortex 104:103–123 org/10.1016/j.nurt.2008.05.004
21. Wirth M, Villeneuve S, La Joie R et al (2014) Gene-environment 37. Uddin MS, Upaganlawar AB (2019) Oxidative stress and antiox-
interactions: lifetime cognitive activity, APOE genotype, and beta- idant defense: biomedical value in health and diseases. Nova
amyloid burden. J Neurosci 34:8612–8617. https://doi.org/10. Science Publishers, New York
1523/JNEUROSCI.4612-13.2014 38. VanItallie TB, Nufert TH (2003) Ketones: metabolism’s ugly
22. Kivipelto M, Rovio S, Ngandu T, Kåreholt I, Eskelinen M, duckling. Nutr Rev 61:327–341
Winblad B, Hachinski V, Cedazo-Minguez A et al (2008) 39. Sato K, Kashiwaya Y, Keon CA, Tsuchiya N, King MT, Radda
Apolipoprotein e ε4 magnifies lifestyle risks for dementia: a GK, Chance B, Clarke K et al (1995) Insulin, ketone bodies, and
population-based study. J Cell Mol Med 12:2762–2771. https:// mitochondrial energy transduction. FASEB J 9:651–658
doi.org/10.1111/j.1582-4934.2008.00296.x 40. Wolf-Klein GP, Silverstone FA, Levy AP (1991) Sweet cravings
and Alzheimer’s disease. J Am Geriatr Soc 39:535–536
23. Mamun AA, Uddin MS, Bin Bashar MF et al (2020) Molecular
41. Keene JM, Hope T (1997) Hyperphagia in dementia: 2. food
insight into the therapeutic promise of targeting APOE4 for
choices and their macronutrient contents in hyperphagia, dementia
Alzheimer’s disease. Oxidative Med Cell Longev 2020:1–16.
and ageing. Appetite 28:167–175. https://doi.org/10.1006/APPE.
https://doi.org/10.1155/2020/5086250
1996.0068
Mol Neurobiol

42. Gasior M, Rogawski MA, Hartman AL (2006) Neuroprotective 58. Gabuzda D, Busciglio J, Chen LB, Matsudaira P, Yankner BA
and disease-modifying effects of the ketogenic diet. Behav (1994) Inhibition of energy metabolism alters the processing of
Pharmacol 17:431–439 amyloid precursor protein and induces a potentially
43. Appelberg KS, Hovda DA, Prins ML (2009) The effects of a amyloidogenic derivative. J Biol Chem 269:13623–13628
ketogenic diet on behavioral outcome after controlled cortical im- 59. Velliquette RA, O’Connor T, Vassar R (2005) Energy inhibition
pact injury in the juvenile and adult rat. J Neurotrauma 26:497– elevates -Secretase levels and activity and is potentially
506. https://doi.org/10.1089/neu.2008.0664 Amyloidogenic in APP transgenic mice: Possible early events in
44. Xu K, Sun X, Eroku BO et al (2010) Diet-induced ketosis im- Alzheimer’s disease pathogenesis. J Neurosci 25:10874–10883.
proves cognitive performance in aged rats. In: Advances in exper- https://doi.org/10.1523/JNEUROSCI.2350-05.2005
imental medicine and biology, pp. 71–75 60. Calon F, Cole G (2007) Neuroprotective action of omega-3 poly-
45. Choragiewicz TJ et al (2010) Neuroprotective effect of ketone unsaturated fatty acids against neurodegenerative diseases: evi-
bodies and ketogenic diet in Nmda-induced Rgc damage in rat. dence from animal studies. Prostaglandins Leukot Essent Fat
Possible Involvement of Kynurenic Acid? | IOVS | ARVO Acids 77:287–293. https://doi.org/10.1016/j.plefa.2007.10.019
Journals. Invest Ophthalmol Vis Sci 51:3456–3467 61. Huang Y, Mahley RW (2014) Apolipoprotein E: Structure and
46. Sullivan PG, Rippy NA, Dorenbos K et al (2004) The ketogenic function in lipid metabolism, neurobiology, and Alzheimer’s dis-
diet increases mitochondrial uncoupling protein levels and activi- eases. Neurobiol Dis 72:3–12. https://doi.org/10.1016/j.nbd.2014.
ty. Ann Neurol 55:576–580. https://doi.org/10.1002/ana.20062 08.025
47. Small GW, Ercoli LM, Silverman DHS, Huang SC, Komo S, 62. Heinsinger NM, Gachechiladze MA, Rebeck GW (2016)
Bookheimer SY, Lavretsky H, Miller K et al (2000) Cerebral Apolipoprotein E genotype affects size of ApoE complexes in
metabolic and cognitive decline in persons at genetic risk for cerebrospinal fluid. J Neuropathol Exp Neurol 75:918–924.
Alzheimer’s disease. Proc Natl Acad Sci 97:6037–6042. https:// https://doi.org/10.1093/jnen/nlw067
doi.org/10.1073/pnas.090106797 63. Lee J, Choi J, Wong GW, Wolfgang MJ (2016) Neurometabolic
48. Buckner RL, Snyder AZ, Shannon BJ, LaRossa G, Sachs R, roles of ApoE and Ldl-R in mouse brain. J Bioenerg Biomembr
Fotenos AF, Sheline YI, Klunk WE et al (2005) Molecular, struc- 48:13–21. https://doi.org/10.1007/s10863-015-9636-6
tural, and functional characterization of Alzheimer’s disease: evi- 64. Zhu L, Zhong M, Elder GA, Sano M, Holtzman DM, Gandy S,
dence for a relationship between default activity, amyloid, and Cardozo C, Haroutunian V et al (2015) Phospholipid dysregula-
memory. J Neurosci 25:7709–7717. https://doi.org/10.1523/ tion contributes to apoe4-associated cognitive deficits in
JNEUROSCI.2177-05.2005 Alzheimer’s disease pathogenesis. Proc Natl Acad Sci U S A
49. Corder EH, Jelic V, Basun H et al (1997) No difference in cerebral 112:11965–11970. https://doi.org/10.1073/pnas.1510011112
glucose metabolism in patients with Alzheimer disease and differ- 65. Li X, Song D, Leng SX (2015) Link between type 2 diabetes and
ing apolipoprotein E genotypes. Arch Neurol 54:273–277. https:// Alzheimer’s disease: from epidemiology to mechanism and treat-
doi.org/10.1001/archneur.1997.00550150035013 ment. Clin Interv Aging 10:549–560
50. Hirono N, Hashimoto M, Yasuda M, Ishii K, Sakamoto S, Kazui 66. Chatterjee S, Mudher A (2018) Alzheimer’s disease and type 2
H, Mori E (2002) The effect of APOE epsilon4 allele on cerebral diabetes: a critical assessment of the shared pathological traits.
glucose metabolism in AD is a function of age at onset. Neurology Front Neurosci 12:383. https://doi.org/10.3389/fnins.2018.00383
58:743–750 67. Tumminia A, Vinciguerra F, Parisi M, Frittitta L (2018) Type 2
51. Mosconi L, Nacmias B, Sorbi S, de Cristofaro MT, Fayazz M, diabetes mellitus and alzheimer’s disease: role of insulin signalling
Tedde A, Bracco L, Herholz K et al (2004) Brain metabolic de- and therapeutic implications. Int J Mol Sci 19:3306. https://doi.
creases related to the dose of the ApoE e4 allele in Alzheimer’s org/10.3390/ijms19113306
disease. J Neurol Neurosurg Psychiatry 75:370–376. https://doi. 68. Harilal S, Jose J, Parambi DGT, Kumar R, Mathew GE, Uddin
org/10.1136/JNNP.2003.014993 MS, Kim H, Mathew B (2019) Advancements in nanotherapeutics
52. Lehtovirta M, Kuikka J, Helisalmi S, Hartikainen P, Mannermaa for Alzheimer’s disease: current perspectives. J Pharm Pharmacol
A, Ryynanen M, Riekkinen PS, Soininen H (1998) Longitudinal 71:1370–1383. https://doi.org/10.1111/jphp.13132
SPECT study in Alzheimer’s disease: relation to apolipoprotein E 69. Salameh TS, Rhea EM, Banks WA, Hanson AJ (2016) Insulin
polymorphism. J Neurol Neurosurg Psychiatry 64:742–746 resistance, dyslipidemia, and apolipoprotein E interactions as
53. Uddin MS, Kabir MT (2019) Emerging signal regulating potential mechanisms in cognitive impairment and Alzheimer’s disease.
of Genistein against Alzheimer’s disease: a promising molecule of Exp Biol Med 241:1676–1683. https://doi.org/10.1177/
interest. Front Cell Dev Biol 7:197. https://doi.org/10.3389/fcell. 1535370216660770
2019.00197 70. Chan ES, Chen C, Cole GM, Wong B-S (2015) Differential inter-
54. Uddin MS, Kabir MT, Tewari D et al (2020) Revisiting the role of action of Apolipoprotein-E isoforms with insulin receptors mod-
brain and peripheral Aβ in the pathogenesis of Alzheimer’s dis- ulates brain insulin signaling in mutant human amyloid precursor
ease. J Neurol Sci 416:116974. https://doi.org/10.1016/j.jns.2020. protein transgenic mice. Sci Rep 5:13842. https://doi.org/10.1038/
116974 srep13842
55. Atamna H, Frey WH (2007) Mechanisms of mitochondrial dys- 71. Chan ES, Shetty MS, Sajikumar S, Chen C, Soong TW, Wong BS
function and energy deficiency in Alzheimer’s disease. (2016) ApoE4 expression accelerates hippocampus-dependent
Mitochondrion 7:297–310. https://doi.org/10.1016/j.mito.2007. cognitive deficits by enhancing Aβ impairment of insulin signal-
06.001 ing in an Alzheimer’s disease mouse model. Sci Rep 6:26119.
56. Veech L, Chance B, Yoshihiro R, Chance B, Kashiwaya Y et al https://doi.org/10.1038/srep26119
(2001) Ketone bodies, potential therapeutic uses. IUBMB Life 72. Seneff S, Wainwright G, Mascitelli L (2011) Nutrition and
(International Union Biochem Mol Biol Life) 51:241–247. Alzheimer’s disease: the detrimental role of a high carbohydrate
https://doi.org/10.1080/152165401753311780 diet. Eur J Intern Med 22:134–140
57. Liang WS, Reiman EM, Valla J, Dunckley T, Beach TG, Grover 73. Shuvaev VV, Laffont I, Serot JM et al Increased protein glycation
A, Niedzielko TL, Schneider LE et al (2008) Alzheimer’s disease in cerebrospinal fluid of Alzheimer’s disease. Neurobiol Aging
is associated with reduced expression of energy metabolism genes 22:397–402. https://doi.org/10.1016/s0197-4580(00)00253-0
in posterior cingulate neurons. Proc Natl Acad Sci 105:4441– 74. Uddin MS, Kabir MT (2019) Oxidative stress in Alzheimer’s dis-
4446. https://doi.org/10.1073/pnas.0709259105 ease: molecular hallmarks of underlying vulnerability. In:
Mol Neurobiol

Biological, diagnostic and therapeutic advances in Alzheimer’s 92. Yin JX, Maalouf M, Han P, Zhao M, Gao M, Dharshaun T, Ryan
disease. Springer Singapore, Singapore, pp. 91–115 C, Whitelegge J et al (2016) Ketones block amyloid entry and
75. Uddin MS, Kabir MT, Jakaria M, Mamun AA, Niaz K, Amran improve cognition in an Alzheimer’s model. Neurobiol Aging
MS, Barreto GE, Ashraf GM (2019) Endothelial PPARγ is crucial 39:25–37. https://doi.org/10.1016/j.neurobiolaging.2015.11.018
for averting age-related vascular dysfunction by stalling oxidative 93. Liu Y, Liu F, Iqbal K, Grundke-Iqbal I, Gong CX (2008)
stress and ROCK. Neurotox Res 36:583–601. https://doi.org/10. Decreased glucose transporters correlate to abnormal
1007/s12640-019-00047-5 hyperphosphorylation of tau in Alzheimer disease. FEBS Lett
76. Li YM, Dickson DW (1997) Enhanced binding of advanced 582:359–364. https://doi.org/10.1016/j.febslet.2007.12.035
glycation endproducts (AGE) by the ApoE4 isoform links the 94. Mosconi L (2013) Glucose metabolism in normal aging and
mechanism of plaque deposition in Alzheimer’s disease. Alzheimer’s disease: methodological and physiological consider-
Neurosci Lett 226:155–158. https://doi.org/10.1016/s0304- ations for PET studies. Clin Transl Imaging 1:217–233
3940(97)00266-8 95. Puchalska P, Crawford PA (2017) Multi-dimensional roles of ke-
77. Henderson ST, Vogel JL, Barr LJ, Garvin F, Jones JJ, Costantini tone bodies in fuel metabolism, signaling, and therapeutics. Cell
LC (2009) Study of the ketogenic agent AC-1202 in mild to mod- Metab 25:262–284
erate Alzheimer’s disease: a randomized, double-blind, placebo- 96. Cunnane SC (2018) Ketones, omega-3 fatty acids and the Yin-
controlled, multicenter trial. Nutr Metab (Lond) 6:31. https://doi. Yang balance in the brain: insights from infant development and
org/10.1186/1743-7075-6-31 Alzheimer’s disease, and implications for human brain evolution.
78. Hirsch-Reinshagen V, Burgess BL, Wellington CL (2009) Why OCL 25:D409. https://doi.org/10.1051/ocl/2018020
lipids are important for Alzheimer disease? In: Molecular and 97. Veyrat-Durebex C, Reynier P, Procaccio V, Hergesheimer R,
Cellular Biochemistry, pp. 121–129 Corcia P, Andres CR, Blasco H (2018) How can a ketogenic diet
79. Jiang Q, Lee CYD, Mandrekar S, Wilkinson B, Cramer P, Zelcer improve motor function? Front Mol Neurosci 11:15. https://doi.
N, Mann K, Lamb B et al (2008) ApoE promotes the proteolytic org/10.3389/fnmol.2018.00015
degradation of Aβ. Neuron 58:681–693. https://doi.org/10.1016/j. 98. Noebels J, Avoli M, Rogawski M et al (2012) Jasper’s basic
neuron.2008.04.010 mechanisms of the epilepsies. Oxford University Press, UK
80. Van der Auwera I, Wera S, Van Leuven F, Henderson ST (2005) 99. Rusek M, Pluta R, Ułamek-Kozioł M, Czuczwar SJ (2019)
A ketogenic diet reduces amyloid beta 40 and 42 in a mouse Ketogenic diet in Alzheimer’s disease. Int J Mol Sci 20:3892.
model of Alzheimer’s disease. Nutr Metab (Lond) 2:28. https:// https://doi.org/10.3390/ijms20163892
doi.org/10.1186/1743-7075-2-28 100. Kashiwaya Y, Takeshima T, Mori N, Nakashima K, Clarke K,
81. Saxena U (2011) Bioenergetics breakdown in Alzheimer’s dis- Veech RL (2000) D-beta-Hydroxybutyrate protects neurons in
ease: targets for new therapies. Int J Physiol Pathophysiol models of Alzheimer’s and Parkinson’s disease. Proc Natl Acad
Pharmacol 3:133–139 Sci U S A 97:5440–5444. https://doi.org/10.1073/PNAS.97.10.
82. Mazzali G, Bissoli L, Gambina S et al (2002) Energy balance in 5440
Alzheimer’s disease. J Nutr Health Aging 6:247–253 101. Dardzinski BJ, Smith SL, Towfighi J, Williams GD, Vannucci
83. Owen OE, Morgan AP, Kemp HG, Sullivan JM, Herrera MG, RC, Smith MB (2000) Increased plasma beta-hydroxybutyrate,
Cahill GF Jr (1967) Brain metabolism during fasting. J Clin preserved cerebral energy metabolism, and amelioration of brain
Invest 46:1589–1595. https://doi.org/10.1172/JCI105650 damage during neonatal hypoxia ischemia with dexamethasone
84. Evans M, Cogan KE, Egan B (2017) Metabolism of ketone bodies pretreatment. Pediatr Res 48:248–255. https://doi.org/10.1203/
during exercise and training: physiological basis for exogenous 00006450-200008000-00021
supplementation. J Physiol 595:2857–2871 102. Maalouf M, Sullivan PG, Davis L, Kim DY, Rho JM (2007)
85. Klepper J, Scheffer H, Leiendecker B et al (2005) Seizure control Ketones inhibit mitochondrial production of reactive oxygen spe-
and acceptance of the ketogenic diet in GLUT1 deficiency syn- cies production following glutamate excitotoxicity by increasing
drome: a 2- to 5-year follow-up of 15 children enrolled prospec- NADH oxidation. Neuroscience 145:256–264. https://doi.org/10.
tively. Neuropediatrics 36:302–308. https://doi.org/10.1055/s- 1016/J.NEUROSCIENCE.2006.11.065
2005-872843 103. Massieu L, Haces M, Montiel T, Hernández-Fonseca K (2003)
86. Zhao Z, Lange DJ, Voustianiouk A et al (2006) A ketogenic diet Acetoacetate protects hippocampal neurons against glutamate-
as a potential novel therapeutic intervention in amyotrophic lateral mediated neuronal damage during glycolysis inhibition.
sclerosis. BMC Neurosci 7:29. https://doi.org/10.1186/1471- Neuroscience 120:365–378. https://doi.org/10.1016/S0306-
2202-7-29 4522(03)00266-5
87. Prins ML, Fujima LS, Hovda DA (2005) Age-dependent reduc- 104. Masuda R, Monahan JW, Kashiwaya Y (2005) D-β-
tion of cortical contusion volume by ketones after traumatic brain Hydroxybutyrate is neuroprotective against hypoxia in serum-
injury. J Neurosci Res 82:413–420. https://doi.org/10.1002/jnr. free hippocampal primary cultures. J Neurosci Res 80:501–509.
20633 https://doi.org/10.1002/jnr.20464
88. Moechars D, Dewachter I, Lorent K, Reversé D, Baekelandt V, 105. Mejía-Toiber J, Montiel T, Massieu L (2006) d-β-
Naidu A, Tesseur I, Spittaels K et al (1999) Early phenotypic Hydroxybutyrate prevents glutamate-mediated lipoperoxidation
changes in transgenic mice that overexpress different mutants of and neuronal damage elicited during glycolysis inhibition
amyloid precursor protein in brain. J Biol Chem 274:6483–6492 in vivo. Neurochem Res 31:1399–1408. https://doi.org/10.
89. Ho L, Qin W, Pompl PN et al (2004) Diet-induced insulin resis- 1007/s11064-006-9189-5
tance promotes amyloidosis in a transgenic mouse model of 106. Noh HS, Hah Y-S, Nilufar R, Han J, Bong JH, Kang SS, Cho GJ,
Alzheimer’s disease. FASEB J 18:902–904. https://doi.org/10. Choi WS (2006) Acetoacetate protects neuronal cells from oxida-
1096/fj.03-0978fje tive glutamate toxicity. J Neurosci Res 83:702–709. https://doi.
90. Levin-Allerhand JA, Lominska CE, Smith JD (2002) Increased org/10.1002/jnr.20736
amyloid- levels in APPSWE transgenic mice treated chronically 107. Suzuki M, Suzuki M, Sato K, Dohi S, Sato T, Matsuura A, Hiraide
with a physiological high-fat high-cholesterol diet. J Nutr Health A (2001) Effect of beta-hydroxybutyrate, a cerebral function im-
Aging 6:315–319 proving agent, on cerebral hypoxia, anoxia and ischemia in mice
91. Feinman RD (2005) When is a high fat diet not a high fat diet? and rats. Jpn J Pharmacol 87:143–150. https://doi.org/10.1254/jjp.
Nutr Metab (Lond) 2:27. https://doi.org/10.1186/1743-7075-2-27 87.143
Mol Neurobiol

108. Suzuki M, Suzuki M, Kitamura Y et al (2002) β- 122. Newport MT, VanItallie TB, Kashiwaya Y et al (2015) A new
Hydroxybutyrate, a cerebral function improving agent, protects way to produce hyperketonemia: use of ketone ester in a case of
rat brain against ischemic damage caused by permanent and tran- Alzheimer’s disease. Alzheimers Dement 11:99–103. https://doi.
sient focal cerebral ischemia. Jpn J Pharmacol 89:36–43. https:// org/10.1016/j.jalz.2014.01.006
doi.org/10.1254/jjp.89.36 123. Reger MA, Henderson ST, Hale C et al (2004) Effects of β-
109. McDaniel SS, Rensing NR, Thio LL et al (2011) The ketogenic hydroxybutyrate on cognition in memory-impaired adults.
diet inhibits the mammalian target of rapamycin (mTOR) path- Neurobiol Aging 25:311–314. https://doi.org/10.1016/S0197-
way. Epilepsia 52:e7–e11. https://doi.org/10.1111/j.1528-1167. 4580(03)00087-3
2011.02981.x 124. Lane-Donovan C, Herz J (2016) High-fat diet changes hippocam-
110. Shie FS, Jin LW, Cook DG, Leverenz JB, LeBoeuf RC (2002) pal apolipoprotein E (ApoE) in a genotype- and carbohydrate-
Diet-induced hypercholesterolemia enhances brain Aβ accumula- dependent manner in mice. PLoS One 11:e0148099. https://doi.
tion in transgenic mice. Neuroreport 13:455–459. https://doi.org/ org/10.1371/journal.pone.0148099
10.1097/00001756-200203250-00019 125. Kossoff E, Freeman J, Turner Z, Rubenstein J (2011) Ketogenic
111. Beckett TL, Studzinski CM, Keller JN, Paul Murphy M, diets: treatments for epilepsy and other disorders. Demos Health
Niedowicz DM (2013) A ketogenic diet improves motor perfor- 126. Kerndt PR, Naughton JL, Driscoll CE, Loxterkamp DA (1982)
mance but does not affect β-amyloid levels in a mouse model of Fasting: the history, pathophysiology and complications. West J
Alzheimer’s disease. Brain Res 1505:61–67. https://doi.org/10. Med 137:379–399
1016/j.brainres.2013.01.046 127. Lecocq FR, McPhaul JJ (1965) The effects of starvation, high fat
112. Brownlow ML, Benner L, D’Agostino D, Gordon MN, Morgan D diets, and ketone infusions on uric acid balance. Metabolism 14:
(2013) Ketogenic diet improves motor performance but not cog- 186–197. https://doi.org/10.1016/S0026-0495(65)80039-7
nition in two mouse models of Alzheimer’s pathology. PLoS One 128. Vining EP (1999) Clinical efficacy of the ketogenic diet. Epilepsy
8. https://doi.org/10.1371/journal.pone.0075713 Res 37:181–190
113. Pawlosky RJ, Kemper MF, Kashiwaya Y et al (2017) Effects of a 129. ClinicalTrials.gov (2020) A medium chain triglyceride interven-
dietary ketone ester on hippocampal glycolytic and tricarboxylic tion for patients with Alzheimer disease. https://clinicaltrials.gov/
acid cycle intermediates and amino acids in a 3xTgAD mouse ct2/show/NCT02912936. Accessed 30 June 2020
model of Alzheimer’s disease. J Neurochem 141:195–207.
130. ClinicalTrials.gov (2020) Medium chain triglycerides and brain
https://doi.org/10.1111/jnc.13958
metabolism in Alzheimer’s disease. https://clinicaltrials.gov/ct2/
114. Zhang J, Cao Q, Li S, Lu X, Zhao Y, Guan JS, Chen JC, Wu Q
show/NCT02709356. Accessed 30 June 2020
et al (2013) 3-Hydroxybutyrate methyl ester as a potential drug
131. ClinicalTrials.gov (2020) Biodistribution of [11C]acetoacetate/
against Alzheimer’s disease via mitochondria protection mecha-
[18F]fluorodeoxyglucose in subjects with risk factors for
nism. Biomaterials 34:7552–7562. https://doi.org/10.1016/j.
Alzheimer’s disease. https://clinicaltrials.gov/ct2/show/
biomaterials.2013.06.043
NCT03130036. Accessed 30 June 2020
115. Kashiwaya Y, Bergman C, Lee J-H, Wan R, King MT, Mughal
132. ClinicalTrials.gov (2020) Brain energy for amyloid transformation
MR, Okun E, Clarke K et al (2013) A ketone ester diet exhibits
in Alzheimer’s disease study. https://clinicaltrials.gov/ct2/show/
anxiolytic and cognition-sparing properties, and lessens amyloid
NCT03472664. Accessed 30 June 2020
and tau pathologies in a mouse model of Alzheimer’s disease.
Neurobiol Aging 34:1530–1539. https://doi.org/10.1016/j. 133. ClinicalTrials.gov (2020) Effect of a modified ketogenic-
neurobiolaging.2012.11.023 Mediterranean diet on Alzheimer’s disease. https://clinicaltrials.
116. Ota M, Matsuo J, Ishida I, Takano H, Yokoi Y, Hori H, Yoshida S, gov/ct2/show/NCT02984540. Accessed 30 June 2020
Ashida K et al (2019) Effects of a medium-chain triglyceride- 134. ClinicalTrials.gov (2020) Dietary treatments for cognitive impair-
based ketogenic formula on cognitive function in patients with ment in older adults. https://clinicaltrials.gov/ct2/show/
mild-to-moderate Alzheimer’s disease. Neurosci Lett 690:232– NCT02521818. Accessed 30 June 2020
236. https://doi.org/10.1016/j.neulet.2018.10.048 135. McDonald TJW, Cervenka MC (2018) The expanding role of
117. Krikorian R, Shidler MD, Dangelo K et al (2012) Dietary ketosis ketogenic diets in adult neurological disorders. Brain Sci 8:148.
enhances memory in mild cognitive impairment. Neurobiol Aging https://doi.org/10.3390/brainsci8080148
33:425.e19–425.e27. https://doi.org/10.1016/j.neurobiolaging. 136. Solfrizzi V, Colacicco AM, D’Introno A, Capurso C, Torres F,
2010.10.006 Rizzo C, Capurso A, Panza F (2006) Dietary intake of unsaturated
118. Taylor MK, Sullivan DK, Mahnken JD et al (2018) Feasibility and fatty acids and age-related cognitive decline: a 8.5-year follow-up
efficacy data from a ketogenic diet intervention in Alzheimer’s of the Italian Longitudinal Study on Aging. Neurobiol Aging 27:
disease. Alzheimers Dement Transl Res Clin Interv 4:28–36. 1694–1704. https://doi.org/10.1016/j.neurobiolaging.2005.09.
https://doi.org/10.1016/j.trci.2017.11.002 026
119. Rebello CJ, Keller JN, Liu AG, Johnson WD, Greenway FL 137. van Gelder BM, Tijhuis M, Kalmijn S, Kromhout D (2007) Fish
(2015) Pilot feasibility and safety study examining the effect of consumption, n−3 fatty acids, and subsequent 5-y cognitive de-
medium chain triglyceride supplementation in subjects with mild cline in elderly men: the Zutphen Elderly Study. Am J Clin Nutr
cognitive impairment: a randomized controlled trial. BBA Clin 3: 85:1142–1147. https://doi.org/10.1093/ajcn/85.4.1142
123–125. https://doi.org/10.1016/j.bbacli.2015.01.001 138. Lopez LB, Kritz-Silverstein D, Barrett-Connor E (2011) High
120. Reger MA, Henderson ST, Hale C, Cholerton B, Baker LD, dietary and plasma levels of the omega-3 fatty acid
Watson GS, Hyde K, Chapman D et al (2004) Effects of β- docosahexaenoic acid are associated with decreased dementia
hydroxybutyrate on cognition in memory-impaired adults. risk: the Rancho Bernardo Study. J Nutr Health Aging 15:25–
Neurobiol Aging 25:311–314. https://doi.org/10.1016/S0197- 31. https://doi.org/10.1007/s12603-011-0009-5
4580(03)00087-3 139. Hashim SA, VanItallie TB (2014) Ketone body therapy: from the
121. Ohnuma T, Toda A, Kimoto A et al (2016) Benefits of use, and ketogenic diet to the oral administration of ketone ester. J Lipid
tolerance of, medium-chain triglyceride medical food in the man- Res 55:1818–1826. https://doi.org/10.1194/jlr.R046599
agement of Japanese patients with Alzheimer’s disease: a prospec- 140. Paoli A, Bianco A, Damiani E, Bosco G (2014) Ketogenic diet in
tive, open-label pilot study. Clin Interv Aging 11:29–36. https:// neuromuscular and neurodegenerative diseases. Biomed Res Int
doi.org/10.2147/CIA.S95362 2014:474296–474210. https://doi.org/10.1155/2014/474296
Mol Neurobiol

141. McDonald TJW, Cervenka MC (2018) Ketogenic diets for adult 144. Włodarek D (2019) Role of ketogenic diets in neurodegenerative
neurological disorders. Neurother J Am Soc Exp Neurother 15: diseases (Alzheimer’s disease and Parkinson’s disease). Nutrients
1018–1031. https://doi.org/10.1007/s13311-018-0666-8 11. https://doi.org/10.3390/nu11010169
142. Ułamek-Kozioł M, Pluta R, Bogucka-Kocka A, Czuczwar SJ 145. Dashti HM, Mathew TC, Hussein T, et al (2004) Long-term ef-
(2016) To treat or not to treat drug-refractory epilepsy by the fects of a ketogenic diet in obese patients. Exp Clin Cardiol 9:200–
ketogenic diet? That is the question. Ann Agric Environ Med 205
23:533–536. https://doi.org/10.5604/12321966.1226841
143. Klein P, Janousek J, Barber A, Weissberger R (2010) Ketogenic
Publisher’s Note Springer Nature remains neutral with regard to jurisdic-
diet treatment in adults with refractory epilepsy. Epilepsy Behav
tional claims in published maps and institutional affiliations.
19:575–579. https://doi.org/10.1016/j.yebeh.2010.09.016

You might also like