You are on page 1of 14

Physiology & Behavior 240 (2021) 113528

Contents lists available at ScienceDirect

Physiology & Behavior


journal homepage: www.elsevier.com/locate/physbeh

High fat diet and its effects on cognitive health: alterations of neuronal and
vascular components of brain
Sorabh Sharma *
Division of Medical Sciences, University of Victoria, PO Box 1700 STN CSC, Victoria, BC, V8W2Y2, Canada

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

Keywords: It has been well recognized that intake of diets rich in saturated fats could result in development of metabolic
Blood brain barrier disorders such as type 2 diabetes mellitus, obesity and cardiovascular diseases. Recent studies have suggested
Cerebral blood flow that intake of high fat diet (HFD) is also associated with cognitive dysfunction. Various preclinical studies have
Cognitive decline
demonstrated the impact of short and long term HFD feeding on the biochemical and behavioural alterations.
Insulin resistance
Neurotrophic factor
This review summarizes studies and the protocols used to assess the impacts of HFD feeding on cognitive per­
formance in rodents. Further, it discuss the key mechanisms that are altered by HFD feeding, such as, insulin
resistance, oxidative stress, neuro-inflammation, transcriptional dysregulation and loss of synaptic plasticity.
Along with these, HFD feeding also alters the vascular components of brain such as loss of BBB integrity and
reduced cerebral blood flow. It is highly possible that these factors are responsible for the development of
cognitive deficits as a result of HFD feeding.

1. Introduction cognitive health [57, 105, 127, 223]. Thus, it is conceivable that the
composition, abundance or the lack of specific nutrients in the diet can
It is well known that the human brain requires a high amount of differentially affect the functions of the brain. In the present review, we
energy to perform a diverse array of actions. However, the brain pos­ will focus on this vital issue of fat composition in diet and how it ma­
sesses minimal energy reserves that can satisfy only a small portion of its nipulates the functioning of the brain with respect to cognitive perfor­
energy demand. Thus, the brain heavily relies on the continuous supply mance in both humans and rodents.
of nutrients from the blood via the blood-brain barrier (BBB) to meet its Traditionally, consumption of saturated fat rich diet is associated
required energy goals. Under physiological conditions, the brain uses with the development of metabolic and cardiovascular diseases [39,
glucose as a main fuel [49]. However, the brain can also utilize some 103, 182]. A growing body of research indicate that patients with
alternative energy substrates such as ketone bodies, triglycerides and metabolic syndrome (including diabetes, obesity, hypertension) have an
lactate during development and times when the glucose availability is increased relative risk for development of diverse cognitive impairments
scarce [21]. Recent research has provided exciting evidence that and AD [17, 30, 38, 46, 57, 68, 96, 112, 143, 193, 200]. Thus, consid­
adequate dietary intake and nutritional status can have a profound ering the progressive increase in global burden of both metabolic dis­
impact on cognitive capabilities, neuronal function, neuronal signaling, orders and neurological diseases, it is necessary to dig into the possible
and synaptic plasticity [70, 126]. For example, both clinical and pre­ mechanisms of association between these diseases. Availability of a
clinical studies have consistently shown that intake of diets rich in suitable animal model is very critical to study these mechanisms.
saturated fats can cause impaired cognitive performance [72, 132]. Considering the polygenic nature of metabolic diseases such as diabetes
Interestingly, studies in the adult human population suggested that even and obesity, it is not wise to use animal models that are monogenic in
short-term dietary intake of a high fat diet (HFD) is associated with nature for example, ob/ob mouse or the Zucker-(fa/fa) fatty rat for
impaired attention and visual memory [13, 52]. Notably, intake of studying all aspects of these polygenic diseases. Importantly, the HFD
saturated fat rich diet is widely considered as a driving factor for feeding model seems best to study these conditions as it is associated
cognitive impairment and AD development [57, 105]. In contrast, diets with a wide range of metabolic disturbances related to body weight,
rich in polyunsaturated fatty acids have been reported to promote blood glucose, insulin levels, lipid profile, cholesterol level etc. Thus,

* Corresponding author.
E-mail address: sharmasorabh88@gmail.com.

https://doi.org/10.1016/j.physbeh.2021.113528
Received 3 April 2021; Received in revised form 3 July 2021; Accepted 6 July 2021
Available online 11 July 2021
0031-9384/© 2021 Elsevier Inc. All rights reserved.
S. Sharma Physiology & Behavior 240 (2021) 113528

researchers have been using this model to generate the metabolic syn­ reports demonstrate that lard-based saturated fat-rich diet feeding for 4
drome in rodents. weeks results in mild hyperglycemia, hyperinsulinemia, glucose intol­
erance, hypertriglyceridemia, hypercholesterolemia, and hypertension
2. Historical aspects of HFD feeding animal model and its [148, 208]. Importantly, excessive intake of these saturated fatty
association with cognitive decline in rodents acids-rich diets is strongly associated as a risk factor for the development
of cognitive decline in humans [27, 95, 211]. Although most researchers
Although initially described as an animal model to study the impact use only a HFD, which is rich in saturated fatty acids to induce metabolic
of dietary fats on metabolic health, HFD feeding has now emerged as an alterations and study the effects on learning and memory tasks [128,
indispensable tool to study the effects of fat-rich diets on neuronal 242]. However, others have opted some additional stressors along with
health. During the 1940s, Samuels and colleagues demonstrated that HFD to better mimic the human disease conditions in rodents.
feeding rats with HFD (containing 70% energy as fat) result in an
elevation of basal and postprandial blood glucose along with the (ii) Additional Stressors: Streptozotocin with HFD
development of obesity [171]. However, it was not until the 1990′ s
when the initial connections of HFD feeding in rodents were found to One of the prime examples of an added stressor to HFD feeding is the
affect their cognitive health by Greenwood and Winocur [73]. They used use of pancreatic β-cell toxin, streptozotocin (STZ) at a low dose [120,
Long Evans rats (1-month-old) and fed them with different dietary 121, 238]. Administering high doses of STZ alone can severely impair
regimens including high saturated fatty acid (SFA) diet, a high poly­ insulin secretion and mimic the T1DM condition in rodents. However,
unsaturated fatty acid diet, or a normal rat chow diet, for 3 months. The when combined with HFD feeding protocol, a low-dose STZ has been
rats were then evaluated for the impact of these different dietary regi­ shown to induce a mild impairment of insulin secretion which is a
mens on their learning and memory performance using a battery of hallmark feature of later stages of T2DM [165, 186]. Reed and col­
behavioral tasks. Interestingly, they found that rats fed with a diet rich leagues first described this animal model and demonstrated that the
in saturated fat, performed poorly in all behavioral tasks as compared to animals fed with HFD alone exhibited high blood insulin levels but
other diets, highlighting the detrimental effects of a saturated fat-rich normal blood glucose levels [165]. They suggested that administration
diet on cognitive performance and brain health [73]. Later, seminal of STZ after HFD feeding can reduce the functional capacity of β-cells of
work by these researchers described a link between HFD feeding and the pancreas without completely compromising insulin secretion. They
cognitive dysfunction by highlighting insulin resistance as an important fed the male Sprague-Dawley rats with either normal chow (12% fat), or
mechanism [72, 220, 221, 219]. Since then, numerous preclinical a HFD (40% fat) for 2 weeks followed by STZ injections (50 mg/kg
studies showed that feeding the rodents with HFD results in significant intravenously) and found that the rats fed with HFD and injected with
impairment in their cognitive function ([33, 75, 128, 169, 177, 178, STZ have hyperglycemia, elevated insulin, free fatty acids (FFAs), and
210]). triglycerides (TGs) level as compared with chow-fed STZ-injected rats
[165]. Later, this model was further modified by Zhang and colleagues,
3. Variants of high fat diet and their effects on metabolic and who used low dose STZ (15 mg/kg) after HFD feeding (30% of calories as
cognitive health fat) for two months [237]. Further, Srinivasan and colleagues modified
this protocol by feeding male Sprague–Dawley rats with either a normal
Different compositions of HFDs have been used to induce obesity and pellet diet (NPD) (12% fat) or HFD (58% fat), respectively for 2 weeks.
metabolic disorders in rodents. Conventionally, for making these diets, Following 2 weeks of dietary regimen, they injected the rats with a lower
the researchers replace the carbohydrate-derived calories with fat- dose of STZ (35 mg/kg, i.p.) [186]. They reported that following STZ
derived calories and then compared this diet with a standard chow injection, the HFD fed rats developed frank hyperglycemia, however, the
diet as a control [106]. However, recently, the researchers have been NPD-fed rats showed just mild elevation in glucose levels. They also
modifying the diet composition based on their specific experimental showed that the HFD fed STZ injected rats had an insulin deficit as
objectives by altering the fat content or by adding additional stressors compared to normal rats [186]. These studies highlight the importance
such as streptozotocin or higher cholesterol concentration. In this sec­ of adding STZ as an additional stressor to develop a model which better
tion, we will review some of the most commonly used variations in the mimics the human T2DM condition instead of just using HFD alone.
diet composition for inducing metabolic and neurological dysfunctions Since these interesting studies have published, many research groups
in rodents. have taken advantage of this excellent model and screened a number of
potential drug candidates.
(i) Type of fat: Saturated vs Unsaturated
(iii) Addition of higher concentration of carbohydrates
Numerous factors such as carbon chain length, degree of unsatura­
tion, the position and configuration of double bonds can affect the In addition to using STZ as an additional stressor along with HFD,
metabolic fate of dietary fatty acids [48]. Classically, the HFDs fed to researchers have also utilized other approaches to modify the HFD
rodents contains a high fraction of saturated fatty acids and a very low model. Some researchers even replace the fat content with higher
fraction of monounsaturated and polyunsaturated fatty acids. In addi­ fructose concentrations in their diet composition. However, the effec­
tion, recent studies also showed that diets rich in saturated fats are more tiveness of such diets in inducing metabolic syndrome is still question­
obesogenic compared to diets containing unsaturated fatty acids [39, able. Zaman and colleagues compared the effects of a HFD and a high-
77]. A growing body of evidence suggests that excessive intake of a fructose diet on body weight, plasma lipid profiles, and insulin sensi­
saturated fat rich diet can result in an increased body weight gain, hy­ tivity in rats in their longitudinal study [236]. They fed the rats with
perglycemia, impaired glucose tolerance, and obesity [3, 153]. Alter­ either a HFD (65% calories from fat) or a high-fructose diet (65% cal­
natively, a recent prospective study demonstrates that increasing the ories from fructose) for a period of 10-weeks and demonstrated that rats
intake of unsaturated fatty acids at the expense of saturated fatty acids fed with HFD showed the development of obesity along with low insulin
may exert beneficial effects on body weight and obesity [15]. Even sensitivity. On the contrary, rats fed with a high-fructose diet showed no
short-term feeding of a saturated fat-rich diet is associated with signif­ change in insulin sensitivity and lipid profile. This study suggests that
icant effects on the metabolic profile. For example, oral ingestion of a HFD feeding has more deleterious effects on lipid profile and insulin
high-fat emulsion in Wistar rats for 10 days has been reported to induce sensitivity as compared to a high fructose diet [236].
insulin resistance, along with larger adipocyte and pancreatic islets, Recent studies suggest that feeding a diet that is composed of a
increased GLUT2 and α-glucosidase mRNA expression [1]. Similar combination of high fat and high sugar content possesses a higher risk

2
S. Sharma Physiology & Behavior 240 (2021) 113528

Table. 1
Preclinical studies evaluating the effects of HFD feeding on brain health and cognitive functioning of rodents. (Arranged in ascending order of the duration of
diet feeding).
Diet composition and duration of Compound tested Behavioral task performed References Animals (sex, strain, Effect of High fat diet feeding on brain health
feeding species, age) and behavioural tasks

HFD containing 45% fat by kcal – – [66] Male Long-Evans rats HFD fed rats showed increase in BDNF and
for 72 h (8–10 week-old) HDAC2 in the dorsal hippocampus and
significant increase in HDAC4 in the ventral
hippocampus.
High fat high fructose diet – – [26] Male Sprague Dawley HFD leads to decreased insulin signaling;
(HFFD) containing 10% lard in Rats weighing 250 to decreased hippocampal weight; reduction in
chow and 60% fructose in 270 g dendritic arborization in CA1 and MAP-2 levels;
drinking water for 7 days decreased dendritic spine number in CA1 and
synaptophysin content, an increase in tau
phosphorylation; and an increase in reactive
astrocyte associated with microglial changes.
Low-fat diet (10% energy from – Object-place-context [128] Male C57Bl/6 J mice Performance deficits were seen after only one
fat) and HFD (60% energy from (OPC) memory task; Object (12 week old) day of HFD. Switching from a high-fat diet back
fat) for 1 or 2 weeks exploration; Novel object to a low-fat diet reverses cognitive deficits
recognition (NOR) task
HFD 60% of kilocalories for 7 Tyr-Leu-Gly (YLG), Object recognition test [136] Eleven-week-old HFD intake reduced cognitive function in
days a tripeptide derived (ORT) and object location male ddY mice behavioural tasks; decreased the number of
from αS1-casein test (OLT). 5‑bromo-2′ -deoxyuridine (BrdU)–positive cells;
decreased nerve growth factor (NGF) and glial
cell line–derived neurotrophic factor (GDNF)
HFD (Coconut oil 5%; Atorvastatin Y maze; Hebb-William’s [18] Swiss albino mice HFD fed animals showed impaired performance
Cholesterol 2%; Cholic acid Maze; elevated plus; either sex (22–28 g) in all behavioural tasks
1%) for 28 days Passive Avoidance
HFD containing 60% calories Glyburide NOR and Object Location [65] C57BL/6 J male mice HFD fed mice showed impaired performance in
from fat for 1, 3 or 6 weeks Recognition (OLR) (3–4 weeks old) NOR task after 1 week. After 3 weeks, HFD-mice
had impaired NOR and OLR. These mice also
displayed anxiety-like behavior. After 6 weeks,
HFD-mice were comparable to LFD-mice in
NOR, open-field and elevated zero maze
performance, but they remained impaired
during OLR testing
HFD 58% fat kcal for 5 weeks Rosiglitazone MWM [148] Adult male SD rats HFD feeding results in severe deficit in learning
weighing 150–190 g and memory
High-fat high-carbohydrate diet Vitamin E RAM [4] Adult male Wistar HFD feeding results in impaired memory;
containing mainly 25% total rats weighing Elevated oxidative stress; Reduced antioxidant
fat; 44% carbohydrates for 6 200–250 g defense.
weeks
HFD containing 0.236 g fat/g of – – [114] Male and female After 4 weeks, HFD fed male rats showed
diet for 4days or 4 weeks Long-Evans rats reduction in BDNF expression in the VMN of the
hypothalamus, but no change in females.
HFD (21.2% fat) for 4 or 7 weeks – Object-place recognition; [216] Male C57BL/6 J mice HFD animals had increased oxidative stress and
Open-field test; fear (6–8 weeks old, biochemical alterations in the hippocampus
conditioning task 18–22 g) after 4 weeks and were aggravated by a longer
exposure time; HFD-fed mice displayed long-
term memory impairments, abnormal
expression of proteins associated with synaptic
function.
HFD (45% energy from fat) for 7 Salvianolic acid B MWM [228] Male C57BL/6 mice HFD feeding resulted in higher food intake,
weeks (4–6 weeks old dyslipidemia, cognitive impairment and
increasing hippocampal and frontal cortex
oxidative stress.
HFD (45% energy from fat) for 7 – – [146] Male C57BL/6 mice HFD feeding decreased the numbers of newly
weeks (5 week-old) generated cells in the dentate gyrus; increased
MDA and decreased the level of BDNF in the
hippocampus
HFD containing 60 kcal% fat for Pyridoxine – [233] Male C57BL/6 J mice In the HFD-fed group, Ki67-positive nuclei and
8 weeks DCX-ir neuroblasts were significantly decreased
in the DG compared with those in the LFD-fed
mice. HFD also reduced the levels of GAD67,
pCREB, BDNF.
HFD containing 60 kcal% fat for Metformin and – [232] Male C57BL/6 mice Ki67-immunoreactive (+) nuclei, DCX+
8 weeks Glimepiride (6 weeks age) neuroblasts and BDNF protein levels were
markedly decreased in the DG of the HFD mice
HFD (primarily from lard plus a Exercise MWM [133] Female Fisher 344 HFD feeding results in an increased reactive
small amount of corn oil, rats; 2 months old oxygen species; reduced BDNF and altered CREB
approximately 39% energy) for activity; impaired spatial learning in MWM
2 months
HFD (containing 60% kcal fat for – T-maze [7] C57BL/6 J male mice HFD mouse showed insulin resistance in
17days and 45% kcal fat for 8 (9 week old) cerebral cortex; their hippocampus exhibited
weeks) increased expression (IRS1-pS616), as well as
decreased expression of PSD-95, and
(continued on next page)

3
S. Sharma Physiology & Behavior 240 (2021) 113528

Table. 1 (continued )
Diet composition and duration of Compound tested Behavioral task performed References Animals (sex, strain, Effect of High fat diet feeding on brain health
feeding species, age) and behavioural tasks

synaptopodin; Spatial working memory was


impaired.
Diet high in monounsaturated fat – NOR; MWM; Operant [63] Male Sprague Dawley Rats fed with high fat and refined sugar diet
(≈ 38% energy) and refined Lever Press (OLP) and rats showed impaired performance in NOR and
sugar (≈ 38% energy) for 8 Extinction Task MWM tasks.
weeks
HFD lard based 39% fat for 2 Vitamin E MWM [224] Male Sprague Dawley Reduced CREB; Synapsin 1; BDNF; increased
months rats 200–240 g oxidative stress; impaired learning and memory
performance
HFD containing 32% fat for 8 β-Alanyl-L-Histidine Fear conditioning [80] B6C3-Tg (APP swe/ Significant decline in the contextual memory in
weeks PSEN1dE9)85Dbo/J transgenic mice fed with HFD; an increase e in
AD mice both sexes (4 the expression of RAGE in blood vessels as well
month old) as increased microglial activation in the
hippocampus of animals fed with HFD
HFD containing 60% of energy as Lipoic acid NOR [166] Male Wistar rats (6 HFD rats showed alterations in central insulin
lipids for 8 weeks week old) signaling; Significantly decreased VGlut1
expression, and memory impairments.
HFFD containing 45% fructose, Indirubin-3- – [173] Male Swiss albino HFFD fed-mice had significant amyloid deposits
20% fat for 60 days monoxime mice (7 weeks old) in cerebral cortex and hippocampus, and protein
expression analyses showed activation of GSK-
3β, nuclear translocation of NF-κB p65 and
upregulation of inflammatory (TNF-α, IL-6,
COX-2), astrocytic (GFAP), glial surface (CD-68)
and pro-apoptotic markers (Bax and caspase-3).
HFHC (21% fat and 1.25% – – [16] 4 month old C57BL/6 Presence of hyperphosphorylated tau correlated
cholesterol) for 2 months mice with activated GSK3
HFD containing 45% of fat for 11 BDNF – [69] Male SD rats Chronic VMN administration of BDNF reduced
weeks (225–250 g) 10-week insulin elevation and/or reversed
old hyperleptinemia
HFD containing 20 g lard/100 g – RAM; Variable-interval [73] Male Long-Evans Among different diets, the rats fed with lard-rich
diet for 3 months delayed alternation task, hooded rats; 1 month diet showed impaired performance in all tests.
Hebb-Williams maze of age and weighing
60–80 g
HFD containing 59.28% fat Vildagliptin MWM and Open field test [157] Male Wistar rats HFD results in neuronal insulin resistance and
mostly from lard for 12 weeks 180–200 g brain mitochondrial dysfunction with impaired
learning and memory.
HFD (60% of calorie from fat) for – [116] 3 month old C57BL/ Decreased IRS-1/ ERK/CREB and GLUT4 in the
12 weeks 6 J mice brain of HFD fed mice.
HFD 59.3% fat for 12 weeks Metformin MWM [156] male Wistar rats 180 Increased peripheral and brain oxidative stress
to 200 g (5 weeks old) levels, brain mitochondrial dysfunction;
impaired learning behavior
HFD 59.3% fat for 12 weeks Vildagliptin and MWM and Open field task [155] male Wistar rats HFD feeding results in increased circulating and
Sitagliptin 180–200 g (6–7 brain oxidative stress levels; brain and
weeks old) hippocampal mitochondrial dysfunction;
impaired learning behavior.
HFD (containing 45% of calories – Open field; MWM; Operant [129] Male Wistar rats HFD animals were consistently poorer in all
as crude fat) for 12 weeks task (150–175 g) aspects of an operant based delayed matching to
position task, no significant difference in open
field and MWM.
HFHF (46% fructose and 20% Cinnamon Y maze; Elevated plus [5] Male Wistar rats 5 Higher anxiety in an elevated plus maze test;
lard) for 12 weeks maze weeks down regulation of the mRNA coding for GLUT1
and GLUT3; increases in mRNA associated with
AD including PTEN, Tau and amyloid precursor
protein (App)
HFD containing 42% of calories – [99] Tg2576 female mice Elevated level of Aβ40 and Aβ42 in HFD-fed Tg
from fat for 12 weeks (4-week old) brain; decrease in the expression of the
anorexigenic neuropeptide, brain-derived
neurotrophic factor,
HFD contains Lard 310 g/1000 g Liver X receptor MWM [183] Swiss albino mice HFD produced a significant decline in MWM
for 90 days agonist T0901317 (20–25 g) of either performance of the animals, reflecting
sex impairment of learning and memory; Increased
oxidative stress; reduced anti-oxidant defense
HFD 310 g/1000 g for 90 days Silymarin MWM [139] Swiss albino mice HFD significantly impaired the cognitive
(20–25 g) of either abilities, along with increasing brain AchE,
sex TBARS, MPO, nitrate/nitrite, and serum
cholesterol levels. Marked reduction of brain
GSH levels was observed.
HFD containing 21.2% fat and Quercetin MWM [226] Male Chinese Escape latency was increased and percent time
1% cholesterol) for 13 weeks Kunming (KM) mice spent in the target quadrant was decreased, with
increased reactive carbonyls, malondialdehyde
(MDA) and declined expression of pi3k, akt,
nrf2, creb and bdnf in the hippocampus of HFD
HFD 32% kcal from fat for 13 calorie restriction MWM [94] Male Sprague- HFD results in reduced BDNF expression in the
weeks Dawley rats (350 to hippocampus and reduced cognitive
425 g) performance in MWM
(continued on next page)

4
S. Sharma Physiology & Behavior 240 (2021) 113528

Table. 1 (continued )
Diet composition and duration of Compound tested Behavioral task performed References Animals (sex, strain, Effect of High fat diet feeding on brain health
feeding species, age) and behavioural tasks

HFD containing 42% kcal from – T-water maze; New object [201] APPswe/PS1 mice (3 HFD accelerates age-associated cognitive
fat, for 4 months recognition and 12 month old) decline without affecting parenchymal Aβ; HFD
decreases MMP-9 enzymatic activity and brain-
derived neurotrophic factor mRNA and protein
levels; altering BBB functionality
HFD of 2 types: Western diet – 14-Unit T-maze [135] Male C57Bl/6 mice Only the HF Lard diet increased age-related
(41% fat), very high fat lard (20-month old) oxidative damage (protein carbonyls); impaired
diet (60% fat for 16 weeks) retention in the behavioral test; a decline in Nrf2
levels and Nrf2 activity
HFD containing 60 kcal% fat; for – MWM; contextual fear [213] Female C57BL6/J HFD fed mice displayed severe deficits in
4 months then switched to conditioning test mice 2 months of age learning and long-term memory consolidation.
normal diet till 22 months of HFD mice also showed increased expression of
age HDAC 5, accompanied by reduced expression of
BDNF in the brains 61 weeks after the mice had
been off the high-fat diet.
HFD 45% fat for 20 weeks Liraglutide ORT [159] Young Male Swiss TO LTP was completely abolished in high fat saline-
mice (6–8 weeks old) treated mice. HFD fed mice also performed
poorly in object recognition task.
HFD (60%) fat for 20 weeks. Resveratrol MWM [89] Male C57BL/6 J mice HFD induces hepatic steatosis, macrophage
(3 weeks old) infiltration, and insulin resistance. HFD mice
also showed increased expression of TNF-α and
Iba-1, tau phosphorylation in hippocampus and
memory deficits.
High cholesterol 5% for 20 weeks Troxerutin, a MWM; step-through [119] Male C57BL/6 mice Cholesterol rich diet feeding results in an
naturally occurring passive avoidance task (4-week-old) increased ROS, AGEs; endoplasmic reticulum
flavonoid stress and impaired PI3K/Akt/CREB pathway.
These mice also showed impaired performance
in behavioural tasks.
HFD 60% fat for 20 week Treadmill Exercise MWM; step-down [147] Four-week-old HFD fed mice showed a decrease in insulin
C57BL/6 male mice signaling, neuroplasticity in the hippocampus
and the dentate gyrus along with cognitive
impairment.
HFD containing 60% fat for 5 – MWM; Locomotor activity [82] Female 3xTg-AD HFD feeding result in an elevation in
months mice amyloidogenic Aβ1–42 and Aβ1–42 peptide;
altered insulin signaling and Impaired learning
in behavioural tasks.
HFD containing 60 kcal% fat; for HDAC II inhibitor – [214] Female C57BL6/J T2DM mice showed increased expression of
6 Months MC1568 mice 2 months of age HDAC5 and 9 in the brain, and increased
susceptibility to oligomeric Aβ-induced synaptic
impairments in the hippocampal formation and
synaptic dysfunction.
High-fat, high-glucose – MWM [191] Rats HFD fed rats exhibited impaired spatial learning
supplemented with 20% high- ability, reduced hippocampal dendritic spine
fructose corn syrup for 8 density, and reduced long-term potentiation at
months Schaffer collateral—CA1 synapses; Reduced
BDNF in hippocampus.
HFD containing 60% kcal fat for Insulin object-recognition task [206] Female 3xTg-AD HFD enhanced glucose intolerance, brain
9 months mice (at 6months of soluble Aβ, and memory impairment in 3xTg-AD
age) mice
HFD for 9–12 months – Contextual fear [85] C57BL/6 J mice both The obese male, but not female, mice showed
conditioning and step- sexes poorer learning performance than their normal
down passive avoidance counterpart in behavioural tasks.
tasks
HFD (45% kcal fat) for 10 months – Morris water maze; [130] Male C57BL/6 mice No effect in MWM; Impaired performance in an
Operant-learning task (17–20 g) operant bar-pressing task.
HFD containing 45 kcal% fat for Melatonin MWM [229] Female Sprague- HFD feeding increases Aβ accumulation, Tau
approximately 10–11 months Dawley rats (8- phosphorylation in the hippocampus along with
month-old) activated microglia and astrocytes. Cognitive
decline was observed in these rats in MWM task
along with reduced CREB activity and BDNF
levels in the hippocampus.
HFD containing 60% energy from – Y-maze spontaneous [98] Male 3xTgAD mice HFD impairs learning and memory in different
fat, for varying intervals and alternation, smell (8 week old) tasks.
age upto 3–16months recognition, NOR; MWM

AD: Alzheimer’s disease; HFD: High fat diet; HFHF: High fat-high fructose; HFHC: high fat high cholesterol; Aβ: Amyloid-β; MWM: Morris Water Maze; GLUT: Glucose
transporter; NOR: Novel Object Recognition task; TNF-α: Tumour Necrosis factor-α; CREB: cAMP Responsive element binding protein; GSK-3β: Glycogen synthase
kinase-3β; BDNF: Brain-derived neurotrophic factor; T2DM: Type 2 diabetes mellitus; PKB: Protein Kinase B; ROS: Reactive Oxygen Species; MDA: Malondialdehyde;
AGEs: Advanced glycation end products; LTP: Long term potentiation; HDAC: Histone deacetylases;MMP-9: matrix metalloproteinase-9: MPO: Myeloperoxidase;
TBARS: Thiobarbituric acid reactive substances; GSH: Reduced glutathione; AchE: Acetyl cholinesterase; ERK: Extracellular signal–regulated kinases; IRS: Insulin
Receptor Substrate; VMN: Ventromedial nucleus of the hypothalamus; RAGE: Receptor for Advanced Glycation End Products; OLP: Operant Lever Press; OLR: Object
Location Recognition; GDNF: glial cell line–derived neurotrophic factor; NGF: nerve growth factor.

5
S. Sharma Physiology & Behavior 240 (2021) 113528

Fig. 1. Pathways leading to hippocampal neuronal damage and cognitive impairment as a result of HFD feeding Note: DAG: Diacyl glycerol; IL-1β: Inter­
leukin-1β; PKC: Protein Kinase C; ROS: Reactive oxygen species; TNF- α: Tumour Necrosis factor- α.

for the development of metabolic syndrome as compared to diets rich in detrimental to brain health and could even account for mild cognitive
either fat or sugar [11, 117]. Lozano and colleagues showed that 2 impairment and dementia [62, 184, 185, 218]. Thus, not surprisingly,
months of feeding of either a high-fat high fructose diet (HFHF) or HFD researchers have used this idea and studied how the addition of
results in an increased body weight, leptin, and HOMA-IR in rats. cholesterol to HFD can affect the metabolic and neurological functions
However, the rats fed with alone high fructose diet had only a transient in rodents. Recently, a comparative study demonstrated that
increase of leptin and c-peptide [117]. Some studies even reported cholesterol-rich HFD induces more severe liver damage, inflammation,
better outcomes of this combination of a high-fat, high- carbohydrate fibrosis, and insulin resistance than HFD without cholesterol [79].
diet compared to HFD alone. For example, in their interesting study, Moreover, there is increasing evidence that a high-cholesterol diet can
Kohli and colleagues randomly assign C57BL/6 mice to different dietary induce neuroinflammation and neuronal apoptosis, ultimately resulting
regimens, including, a chow diet, a HFD (58 kcal% fat), or a high-fat, in neurodegeneration and cognitive deficits [28, 161]. Interesting
high-carbohydrate (HFHC) diet and drinking water enriched with studies by Thirumangalakudi and colleagues demonstrated that feeding
high-fructose corn syrup equivalent. They demonstrated similar effects a high fat/high cholesterol diet to C57BL/6 for 8 weeks results in a loss
of HFD and HFHC on weight gain, insulin resistance, body fat, and liver of working memory in mice, which was correlated with
steatosis. Interestingly, the mice fed with the HFHC diet had increased neuro-inflammatory changes and increased AβPP processing [202].
hepatic oxidative stress, fibrogenesis and collagen deposition as Thus, these authors suggested that cognitive deficit associated with
compared to the HFD fed group. Thus, it is proposed that the addition of hypercholesterolemia is mediated by increased neuro-inflammation and
fructose to HFD promotes liver fibrogenesis [100]. Based upon these APP processing in a non-transgenic mouse model. Moreover, recently
findings, several preclinical studies have used this diet composition to these authors also demonstrated that feeding the cholesterol-rich diet
reveal its effect on learning and memory in rodents [5, 26, 173]. can also result in hyperphosphorylated tau and increased GSK-3 activity
in the hippocampus of mice. These changes were attributed to insulin
(iv) Addition of higher cholesterol in HFD resistance conditions in the hippocampus [16]. Some other groups have
also observed significant deficits in working memory of mice fed with a
Cholesterol is essential for the normal functioning of the brain and high fat/high cholesterol diet [71]. Alternatively, some studies have also
plays an important role in signaling pathways, synaptic plasticity, shown that decreasing cholesterol levels in the brain could improve
learning, and memory. Previous research has suggested that the brain learning and memory performance in different behavioral tasks [93,
contains the remarkably highest level of cholesterol in the body, which 231].
is approximately 20% of whole-body cholesterol [19]. A growing body In summary, it is plausible that the composition of the diet is a vital
of evidence suggests that higher than normal cholesterol levels can be factor that affects the development of cognitive and metabolic

6
S. Sharma Physiology & Behavior 240 (2021) 113528

alterations. It is advisable that along with the composition of the diet, complex I of the mitochondrial respiratory chain, where most of these
many other important factors should also be kept in the mind while electrons safely combine with oxygen and protons to form water.
studying the effects of HFD on cognitive functions such as duration of However, a portion of these electrons reacts with oxygen to form su­
the HFD feeding, age, sex, and strain of the animal. Perhaps all these peroxide radicals. In contrast, during HFD feeding, elevated mitochon­
factors make it difficult to compare studies among each other. For drial β-oxidation of FFAs leads to an excess electron flow and increased
example, most of the studies have shown that cognitive deficits can be production of ROS, and eventually, elevated superoxide anion level
observed in HFD fed rodents within 4 to 12 weeks period. However, a [125, 152]. Importantly, the ROS can also consume the endogenous
recent study by McLean and colleagues exhibited that HFD-mediated antioxidant enzymes and put the brain at higher risk from oxidative
cognitive deficits can be observed within a week of diet feeding [128]. damage [109, 152]. A growing body of evidence suggests a possible
This study provided evidence that HFD mediated cognitive decline starts relationship between increased oxidative stress and the development of
much earlier than anticipated based on previous literature [37, 90]. cognitive decline [78, 134, 149, 192]. Considering these evidences, it is
Another study reported that the biochemical alterations in the brain highly likely that HFD mediated cognitive deficits are mediated by
associated with HFD feeding could be observed as early as 72 h after the increased oxidative stress.
initiation of diet feeding [66]. These researchers observed that levels of
enzyme histone deacetylases (HDACs) were significantly higher in the 4.2. HFD and neuro-inflammation
hippocampus of HFD-fed rats after 72 h of feeding [66]. Moreover,
recent studies have demonstrated that the underlying insulin resistance Chronic unchecked inflammation has been found to be deleterious
in the brain can occur after only 3 days of a HFD feeding initiation [35, that could result in the pathology of metabolic and neurodegenerative
142]. diseases [60, 164]. Literature has suggested that HFD feeding is not only
Some other factors such as the strain of rats/mice can affect the associated with the development of inflammation in the peripheral or­
development of metabolic and neurological alterations. For example, gans such as the liver, adipose tissue, skeletal muscle, but it can also lead
some strains such as C57BL/6 J mice or Wistar rats develop obesity and to the development of neuro-inflammation. Several possible mecha­
insulin resistance as a result of HFD feeding but 129S6 mice [2] or A/J nisms by which this happens include, generation of peripherally derived
mice do not [194]. Thus, in this review, we have distilled hundreds of pro-inflammatory cytokines that cross BBB and could eventually lead to
papers and provided a summary of various factors such as diet compo­ damage of neuronal function in the brain [9, 42]. Several studies have
sition (% of fat), diet duration, animal variables (strain, age, and sex), shown that HFD feeding increased the expression of various cytokines
behavioral task performed that can affect the metabolic and neurolog­ including TNF-α, interleukin-6 (IL-6), IL-1β, interferon-γ in the hippo­
ical functioning (Table. 1). As discussing each of this factor is beyond the campus, neocortex, and hypothalamus [6, 20, 29, 55, 89, 115, 131, 173,
scope of this review, the readers are directed to very good papers that 229]. These cytokines bind to their respective receptors in the brain and
focus on how these differences among strains, sex, age, and diet lead to neurodegeneration (Fig. 1). By using anti-inflammatory com­
composition can affect the development of pathological conditions [25, pounds in the HFD model, researchers have repeatedly shown that these
39, 81, 140]. compounds can ameliorate cognitive deficits and prevent neuronal
death [215, 230]. Not surprisingly, increased levels of these
4. Potential mechanisms of HFD mediated neuronal dysfunction pro-inflammatory cytokines have been well reported in AD brains [83,
and cognitive deficits 167] and blood samples [196]. Interestingly, studies have shown that
prolonged use of anti-inflammatory drugs such as nonsteroidal
4.1. HFD and oxidative stress anti-inflammatory drugs (NSAIDs) can reduce the risk of development of
AD by 50% [86].
The brain is considered as the most vulnerable organ to damage by Several studies have also shown that HFD fed mice exhibit activated
oxidative stress [67]. This is attributed to the higher lipid content, high microglial and astrocytic activity in the hippocampus along with an
demand for oxygen, and the limited availability of antioxidant enzymes elevated level of pro-inflammatory cytokines [158, 202]. An interesting
[21]. Continuous over-nutrition, as a result of HFD feeding, might lead study by Baufeld and colleagues showed that HFD feeding results in an
to the generation of excessive reactive oxygen species (ROS) and reac­ increased number of microglia cells (using Iba1 + cells count) and
tive nitrogen species (RNS) [207]. Overproduction of these species increased activity of astrocytes (using GFAP) specifically in the hypo­
damages the macromolecules such as DNA, membrane lipids, and pro­ thalamus region of HFD fed mice. They also suggested that these
tein structures [198]. Physiologically, these free radicals are quenched microglial and astrocytic responses increase with the duration of diet
by endogenous antioxidant enzymes including reduced glutathione, feeding [12].
catalase, and superoxide dismutase. However, with HFD feeding, ROS Additionally, recent studies have also suggested that cognitive
levels overwhelm the antioxidant enzyme capacity and lead to excessive decline as a result of HFD feeding could be associated with alteration of
ROS accumulation [199]. richness, diversity, and composition of gut microbiota [217, 230, 239].
Numerous studies have shown that HFD feeding results in an As such this alteration of microbiota has been found to be associated
elevated level of oxidative stress markers in the brain [115, 119, 139, with neuro-inflammation and resulting cognitive impairment [56, 180].
155, 156, 183, 198, 216, 224, 226]. The hippocampus and neocortex are Thus, it is likely that inflammation resulting from HFD feeding could be
among the most vulnerable parts of the brain that undergo selective responsible for neuronal dysfunction and cognitive decline through
damage by oxidative stress. Moreover, hippocampal oxidative stress is various possible mechanisms.
strongly linked to cognition dysfunction [47, 58, 59, 190]. To support
the hypothesis that oxidative stress might be a possible mechanism 4.3. HFD and insulin resistance
involved in HFD feeding associated cognitive decline, several re­
searchers have used antioxidant therapies and showed that cognitive For many decades after its discovery, insulin was considered just a
deficits can be rescued by using this approach in HFD fed animals [4, peripheral hormone that performs important functions including regu­
139, 228]. lation of glucose metabolism, food intake, and body weight but does not
Briefly, the mechanism by which HFD feeding can cause elevated affect the brain. However, later it was discovered that insulin is also vital
oxidative stress is stated here (Fig. 1). HFD feeding results in an for brain health as it performs an array of functions ranging from
increased generation of free fatty acids [151]. Under normal diet enhancing hippocampal long-term potentiation [88], modulation of
feeding, the mitochondrial β-oxidation of free fatty acids is associated synaptic plasticity [205, 241] and plays an important role in learning
with the transfer of electrons from cofactors (NADH and FADH2) to the and memory [240]. In addition, insulin has also been reported to

7
S. Sharma Physiology & Behavior 240 (2021) 113528

regulate structural plasticity in the brain [32], along with inducing the tropomyosin-related kinase B (TrkB) and activates different pathways
expression of PSD95, (a scaffold protein required for the formation of the involving phospholipase C (PLC)-γ, phosphatidylinositol 3-kinase
postsynaptic junction) [111]. Thus, keeping in mind the vast majority of (PI3K)/ protein kinase B (Akt) and mitogen-activated protein kinase
functions mediated by insulin in the brain, it can be assumed that any (MAPK)/ extracellular signal-regulated kinase (ERK) [225]. All of these
alteration in insulin signaling, especially during insulin resistance can converge on the transcription factor cyclic adenosine monophosphate
severely impact neuronal activity and possibly results in (cAMP) response element-binding protein (CREB), which can upregulate
neurodegeneration. genes related to cell survival, growth, and differentiation [101][176]. It
Insulin resistance has been well reported to play a critical role in the has been demonstrated that BDNF regulates phosphorylation of CREB
pathogenesis of T2DM and metabolic syndrome. Moreover, the litera­ [61, 181]. Importantly, CREB is also reported to be regulated by insulin
ture is rich with reports highlighting the role of insulin resistance in HFD signaling components [24, 51, 97]. Once activated, CREB is well re­
induced cognitive deficits [7, 37, 89, 118, 156]. Insulin resistance is ported to regulate the expression of proteins associated with learning
usually denoted by the increased homeostasis model assessment and memory [10, 14]. Various studies have demonstrated that HFD diet
(HOMA) index in most of the preclinical studies [8]. Despite much work, intake is associated with reduced CREB activity in the brain [119, 133,
the mechanisms by which HFD feeding results in development of insulin 224, 226, 229]. Thus, probably the reduction in BDNF level and
resistance are still not completely understood. According to recent cognitive deficits observed as a result of HFD feeding could in turn be a
literature, it has been proposed that HFD contributes to free fatty acid result of reduced CREB activity.
(FFA) elevation which induces insulin resistance in skeletal muscle.
Increased plasma FFA results in the accumulation of intramuscular 4.5. HFD and synaptic plasticity dysfunction
lipids, in particular: diacylglycerols (DAG), ceramide and long-chain
acyl-CoAs, which are proposed to be involved in the induction of insu­ In line with the neurotrophic factor hypothesis, impairment in brain
lin resistance [31, 54, 174, 188]. DAG has been well reported to impair morphological and structural abnormalities in relation to HFD feeding
insulin action via activation of novel PKCs (nPKCs), including PKCε in has also been suggested by few histological studies. Several animal
the liver and both PKCθ and PKCε in skeletal muscle [122]. Activation of studies have demonstrated that HFD feeding is associated with patho­
PKC subsequently impairs tyrosine phosphorylation of IRS-1/2 [123, logical changes in the hippocampus and cortex, including reduced
235]. dendritic spines in CA1 and impaired LTP along with memory impair­
From the insulin signaling standpoint, Glycogen synthase kinase-3 β ment [50, 74, 204]. Out of many brain structures, the hippocampus is
(GSK-3β) is a plausible candidate for mediating HFD induced compli­ extremely sensitive to changes in glucose homeostasis [124, 145, 163].
cations such as insulin resistance and cognitive decline. GSK-3β regu­ Stranahan et al. [191] demonstrated that feeding rats with a high-fat,
lates a plethora of biological processes and downstream signaling of high-glucose diet for eight months results in significant impairment of
insulin primarily through insulin receptor activation. Feeding HFD to spatial learning ability (assessed by MWM) along with reduced hippo­
experimental animals has been shown to increase the expression of campal dendritic spine density, and reduced LTP at Schaffer collater­
GSK3β and its association with cognitive decline [16, 82]. Alternatively, al–CA1 synapses. They also observed reduced BDNF level in the
several researchers have highlighted that GSK3β inhibitors could reverse hippocampus of high-fat high glucose fed animals as compared to con­
cognitive decline as a result of HFD feeding [43, 173, 178, 179]. trols. These authors described the changes could be a result of peripheral
insulin resistance or it may be due to direct effects of some components
4.4. HFD and transcriptional dysregulation of diet on brain health and hippocampal plasticity [191]. In addition,
impairment of stimulus-evoked LTP has been reported in granular cells
Brain-derived neurotrophic factor (BDNF) is a member of the neu­ of the DG of HFD fed rats [92]. Although many studies have shown that
rotrophin family of the growth factors that play a vital role in regulating HFD feeding result in a change of expression of protein-coding genes in
the survival, growth, and differentiation of peripheral and central ner­ the cortex, however, the alterations of non-coding RNAs of the brain
vous systems. BDNF has been reported to be critical for activity- cortex is relatively less explored. In this direction, a recent study by
dependent neuroplasticity [53] and can also induce long-term potenti­ Yoon and colleagues showed changes in expression of both coding and
ation (LTP) and thus plays a key role in learning and memory [110]. A non-coding RNAs in the brain cortex as a result of HFD feeding in mice.
reduction in BDNF expression has been associated with cognitive decline These researchers reported that HFD feeding for 8 weeks results in
and reduced mRNA and protein expression of BDNF has been found in decreased expression of genes associated with synaptogenesis and
AD brains [36, 154] and serum samples [107]. Moreover, studies have neurotransmitter release [234]. Arnold and colleagues also demon­
also demonstrated a reduction in circulating levels of BDNF in diabetes strated decreased expression of PSD-95, in the hippocampus of mice fed
and obesity [138, 144, 195]. BDNF has also been a focus of extensive with HFD [7]. Interestingly studies have also reported reduced neuro­
research in relation to HFD feeding. Various studies have highlighted the genesis in the dentate gyrus of HFD fed animals [113]. Thus, it is evident
deleterious effects of HFD feeding on BNDF profile in different brain that HFD feeding might severely affect the brain morphology and syn­
regions. Earlier studies by Molteni and colleagues found that rats fed aptic plasticity by altering the expression of various genes associated
with saturated fat and refined sugar diet for 2 months showed impaired with these processes.
performance in learning and memory tasks (morris water maze), which
was associated with reduced BDNF in the hippocampus [132]. These rats 5. Potential mechanisms of HFD mediated vascular dysfunction
also showed a decrease in mRNA and protein levels of synapsin I; cyclic and cognitive deficits
AMP-response element-binding protein (CREB) and growth-associated
protein 43 (mRNA only). Since these initial observations, several other 5.1. HFD and blood–brain barrier dysfunction
labs have demonstrated that HFD diet intake is associated with
decreased expression of BDNF in the brain ([224]; [94, 115, 191, 213, Many pathophysiological complications associated with HFD feeding
227]). Keeping in mind the important role played by BDNF in memory have profound effects on vascular function. The BBB is a complex unit
formation, it is plausible that the negative effects of HFD diet con­ comprising of endothelial cells joined together by tight junction proteins
sumption on learning and memory may also be mediated in part by (i.e., occludin and claudin), scaffolding proteins (i.e., ZO-1 and ZO-2),
alteration of BDNF-related synaptic plasticity. and astrocytes [150]. Continuous tight junctions form the
Although it is still not clear how exactly HFD feeding results in a adhesion-based connections between vascular endothelial cells, forming
reduction of BDNF levels, however, the BDNF receptor signaling can a seal around the vessel lumen and restricting the passage of anything
shed some light on this. BDNF binds to a tyrosine kinase receptor called larger than 180 Da [172]. Any damage to the BBB might lead to leakage

8
S. Sharma Physiology & Behavior 240 (2021) 113528

of unwanted substances in the brain, which can result in neuronal weeks after HFD feeding. Interestingly, these levels recovered to normal
damage and neurodegenerative diseases including AD [22, 212]. For after 10 weeks of diet feeding. These studies highlight the importance of
instance, Wisniewski and Kozlowski initially reported extensive peri­ insulin signaling in BBB integrity and suggesting that changes in the
vascular leakage of IgG and albumin in AD brain samples compared to status of insulin signaling/resistance could influence the expression of
controls [222]. Since then, several studies have found histopathological BBB transporters, which in turn may alter the expression of cognitive
evidence of BBB abnormalities in AD [34, 76, 203]. function-related proteins. [141].
Similarly, alterations of BBB integrity have also been reported in
T2DM [84]. In a clinical study, Starr and colleagues showed increased 5.2. HFD and impaired cerebral blood flow
BBB permeability to gadolinium diethylenetriamine pentaacetic acid in
patients with well-controlled T2DM [187]. Although the clinical studies Keeping in mind, the high metabolic demand of brain, a continuous
are rare, the literature is rich with preclinical studies demonstrating the blood supply is required for its proper functioning. Thus, cerebral blood
damaging effects of hyperglycemia and the advanced glycation flow (CBF) is tightly regulated and any disturbance in blood flow to
end-products (AGEs) on BBB integrity in T2DM [160, 175]. Stranahan brain might result in severe neurological or neurodegenerative condi­
and colleagues used the db/db mouse model of leptin deficiency (a tion. An important review by David Attwell’s group highlighted the role
widely used mouse model of T2DM) and showed that the BBB break­ of decreased CBF in cognitive impairments observed in AD patients
down results in cerebral macrophage infiltration in these mice [189]. [102]. In addition, the impact of diabetes has also been studied in
Another study used AD transgenic APP/PS1 mice crossed with diabetic relation to CBF and cognitive impairments, and findings across these
db/db mice, and showed that F1 progeny of these mice demonstrate BBB limited studies are contradictory. Dandona et al. [41] reported no dif­
alterations associated with the onset of T2DM [162]. ference in age-related perfusion reductions in T2DM patients and control
Most importantly, in the context of this review, the effects of HFD subjects by measuring global CBF using 133-Xe inhalation method. On
feeding on BBB integrity and the associated cognitive decline has also the contrary, other studies employing single photon emission computed
been demonstrated in various studies ([45, 90, 91][64, 197]). Seminal tomography (SPECT) found that diabetic subjects showed reduced CBF
work from Davidson lab revealed that rats fed with saturated fats and [137, 168, 209]. Moreover, recent studies also demonstrated regional
sugar-rich western diet showed impaired performance on a cerebral hypoperfusion in individuals with diabetes using Arterial spin
hippocampal-dependent serial feature negative discrimination task that labeling (ASL) [40, 108, 226, 244]. Although the effect of HFD feeding
is accompanied by increased BBB permeability as detected by higher on CBF changes has not been studied extensively but a recent study
sodium fluorescein density in hippocampus of these rats, as compared to explored the effect of HFD induced T2DM on cognitive performance in
chow-fed rats [44]. In another study, where they used a lard-based mice [243]. These researchers found that vascular cognitive impairment
saturated fat rich diet in rats, Kanoski and colleagues showed mice fed a HFD showed higher cognitive deficits in behavioral tasks (fear
decreased mRNA expression for tight junction proteins (claudin-5 and conditioning and Morris water maze), increased neuronal loss, glial
claudin-12) as compared to chow fed rats [91]. In a more recent study, activation, and most importantly global decrease in CBF as compared to
Takechi and colleagues showed that 4 weeks of feeding a diet rich in fats control diet-fed vascular cognitive impairment mice. They described
and fructose (HFF) to C57BL/6 J mice results in a significant BBB that HFD exacerbates vascular cognitive impairment pathology. In
dysfunction along with associated neuro-inflammation in cortex and contrast, a recent study found no significant change in CBF level in
hippocampal regions. In addition, they also reported the effects of APP/PS1 mice fed a HFD as compared to controls [23]. Schaffer lab used
long-term diet feeding (24 weeks) and showed that mice fed with HFF adult APP/PS1 transgenic mice and fed them with HFD (42% calories
diet showed significant cognitive decline associated with increased BBB from fat) for a long duration (over 6 months) and then tested their CBF
permeability [197]. Another interesting study reveals the long-term ef­ along with other experimental protocols. Surprisingly, they did not find
fects (6 months) of a diet rich in saturated fat and cholesterol on the a significant difference in CBF in HFD fed or control-fed mice [23]. Thus,
hippocampus in middle-aged Fischer 344 rats [64]. These researchers further studies are required to confirm the role of CBF changes in HFD
used SMI-71 immunofluorescence to evaluate the integrity of BBB and induced cognitive decline.
showed reduced SMI-71 and occludin protein immunoreactivity on
blood vessels of these rats, indicating a loss in tight junction protein and 6. Conclusion and future directions
a consequent increase in BBB permeability. Surprisingly, they found
increased levels of occludin and ZO-1, along with an increased number In conclusion, the HFD feeding animal model has proven an
of activated microglia in the hippocampus of HFHC diet-fed rats as invaluable tool not only for studying the pathology of metabolic con­
compared with control rats [64]. A recent study by Salameh and col­ ditions such as obesity and diabetes, but also for the identification of
leagues showed that HFD feeding in mice results in increased entry of molecular pathways involved in co-morbid metabolic and cognitive
14C-sucrose (in the hypothalamus and hippocampus), and 99mTc-albu­ dysfunctions. Researchers across the globe have manipulated the diet
min (in the whole brain) as compared to the normal diet fed mice. composition to study different objectives based upon their research
Moreover, they demonstrated that treatment with topiramate (an goals and to better mimic the human disease condition. The fact that
anti-epileptic drug) significantly attenuate the increased permeability these diets have not been standardized makes it very hard to replicate
through BBB, decreased oxidative stress markers, and also increased the the findings of the previous studies. Most of the studies have used lard
expression of ZO-1 and claudin-12 (tight junction proteins) [170]. based saturated fat diet (60% kcal). The characteristic features of HFD
Two different studies using the same in vitro model, human cerebral feeding induced cognitive decline have been reported to be influenced
microvascular endothelial cell line (hCMEC/D3) for assessing human by several factors such as the sex, strain and age of mice as well as
BBB integrity showed the effects of insulin on tight junction proteins composition an duration of HFD feeding. Although the detrimental ef­
[87, 104]. They demonstrated that insulin treatment enhances the fects of HFD feeding on neuronal health have been reported as early as
integrity of tight junction by increasing the expression of tight junction 24 h after the initiation of diet feeding. However, the duration can be
transmembrane proteins. In contrast, they showed that inhibiting decided based on the research questions for particular study. Among
insulin/insulin-like growth factor-1 receptor kinase activity using various behavioural tasks used to evaluate the impact of diet feeding on
AG1024, results in blocking of the increase of tight-junction integrity learning and memory paradigms, the Morris water maze is considered as
[87]. Similarly, in a recent study, Ogata and colleagues investigated a gold standard and has been used extensively by researchers. It is
changes in BBB and brain parenchymal protein expression as a result of mostly a hippocampal based task where a mice swims in a water tank
HFD feeding, where they found reduced expression levels of BBB and tries to find the hidden platform by using external clues. Numerous
transporters as well as tight-junction proteins (claudin-5, occludin) 2 behavioural studies suggest that the effect of HFD feeding is not just

9
S. Sharma Physiology & Behavior 240 (2021) 113528

Fig. 2. Schematic diagram of possible mechanisms responsible for HFD mediated cognitive decline by affecting neuronal and vascular components of
brain. Note: BBB: Blood Brain Barrier; BDNF: Brain-derived neurotrophic factor; CREB: cAMP Responsive element binding protein; HFD: High fat diet.

confined to one brain area, however, it affects the functioning of [9] W.A. Banks, A.J. Kastin, R.D. Broadwell, Passage of cytokines across the blood-
brain barrier, Neuroimmunomodulation. 2 (4) (1995) 241–248.
different brain areas, including hippocampus, cortex, hypothalamus etc.
[10] A. Barco, H. Marie, Genetic approaches to investigate the role of CREB in
Thus, instead of using just one behavioural task that mostly relies on neuronal plasticity and memory, Mol. Neurobiol. 44 (3) (2011) 330–349. Dec.
functioning of particular brain area, it is advisable to use a battery of [11] S. Basu, P. Yoffe, N. Hills, R.H. Lustig, The relationship of sugar to population
behavioural tasks which rely on different brain areas to study a more level diabetes prevalence: an econometric analysis of repeated cross-sectional
data, PLoS One. 8 (2) (2013) e57873.
diverse action of HFD feeding on overall brain functioning. Overall, from [12] C. Baufeld, A. Osterloh, S. Prokop, K.R. Miller, F.L. Heppner, High-fat diet-
these preclinical studies, it has been postulated that oxidative stress, induced brain region-specific phenotypic spectrum of CNS resident microglia,
neuro-inflammation, insulin resistance and BBB disruption are among Acta Neuropathol. 132 (3) (2016) 361–375, https://doi.org/10.1007/s00401-
016-1595-4.
the most important factors that underlies the HFD induced cognitive [13] J.L. Bayer-Carter, P.S. Green, T.J. Montine, B. VanFossen, L.D. Baker, G.
decline (Fig. 2). However, further studies are still warranted to fully S. Watson, et al., Diet intervention and cerebrospinal fluid biomarkers in amnestic
understand the relationship between fat intake and cognitive mild cognitive impairment, Arch. Neurol. 68 (2011) 743–752.
[14] E. Benito, A. Barco, CREB’s control of intrinsic and synaptic plasticity:
performance. implications for CREB-dependent memory models, Trends Neurosci. 33 (5)
(2010) 230–240. May.
[15] Y. Beulen, M.A. Martínez-González, O. van de Rest, et al., Quality of dietary fat
Declaration of Competing Interest intake and body weight and obesity in a mediterranean population: secondary
analyses within the PREDIMED trial, Nutrients. 10 (12) (2018) 2011.
The authors have declared that no conflict of interest exists. [16] N.R. Bhat, L. Thirumangalakudi, Increased tau phosphorylation and impaired
brain insulin/IGF signaling in mice fed a high fat/high cholesterol diet,
J. Alzheimers Dis. 36 (4) (2013) 781–789.
References [17] G.J. Biessels, F. Despa, Cognitive decline and dementia in diabetes mellitus:
mechanisms and clinical implications, Nat. Rev. Endocrinol. 14 (10) (2018)
[1] J. Ai, N. Wang, M. Yang, Z.M. Du, Y.C. Zhang, B.F. Yang, Development of wistar 591–604.
rat model of insulin resistance, World J. Gastroenterol. 11 (2005) 3675–3679. [18] R.R. Biswas, C.D. M, A S R S Rao, S.R Kadali, Effect of atorvastatin on memory in
[2] K. Almind, C.R. Kahn, Genetic determinants of energy expenditure and insulin albino mice, J. Clin. Diagn. Res. 8 (11) (2014) HF01–HF04.
resistance in diet-induced obesity in mice, Diabetes. 53 (2004) 3274–3285. [19] I. Björkhem, S. Meaney, Brain cholesterol: long secret life behind a barrier,
[3] M.A. Alsaif, M.M.S. Duwaihy, Influence of dietary fat quantity and composition Arterioscler. Thromb. Vasc. Biol. 24 (2004) 806–815.
on glucose tolerance and insulin sensitivity in rats, Nutr. Res. 24 (2004) 417–425. [20] C. Boitard, A. Cavaroc, J. Sauvant, et al., Impairment of hippocampal-dependent
[4] K.H. Alzoubi, O.F. Khabour, H.A. Salah, Z. Hasan, Vitamin E prevents high-fat memory induced by juvenile high-fat diet intake is associated with enhanced
high-carbohydrates diet-induced memory impairment: the role of oxidative hippocampal inflammation in rats, Brain Behav. Immun. 40 (2014) 9–17.
stress, Physiol. Behav. 119 (2013) 72–78. [21] M.P. Bordone, M.M. Salman, H.E. Titus, et al., The energetic brain - a review from
[5] R.A. Anderson, B. Qin, F. Canini, L. Poulet, A.M. Roussel, Cinnamon counteracts students to students, J. Neurochem. 151 (2) (2019) 139–165.
the negative effects of a high fat/high fructose diet on behavior, brain insulin [22] G.L. Bowman, J.A. Kaye, M. Moore, D. Waichunas, N.E. Carlson, J.F. Quinn,
signaling and Alzheimer-associated changes, PLoS One. 8 (12) (2013) e83243. Blood-brain barrier impairment in Alzheimer disease: stability and functional
[6] C. Andre, A.L. Dinel, G. Ferreira, S. Laye, N. Castanon, Diet-induced obesity significance, Neurol. 68 (21) (2007) 1809–1814.
progressively alters cognition, anxiety-like behavior and lipopolysaccharide- [23] O. Bracko, L.K. Vinarcsik, J.C. Cruz Hernández, et al., High fat diet worsens
induced depressive-like behaviour: focus on brain indoleamine 2,3-dioxygenase Alzheimer’s disease-related behavioral abnormalities and neuropathology in
activation, Brain Behav. Immunol. 41 (2014) 10–21. APP/PS1 mice, but not by synergistically decreasing cerebral blood flow, Sci.
[7] S.E. Arnold, I. Lucki, B.R. Brookshire, et al., High fat diet produces brain insulin Rep. 10 (2020) 9884.
resistance, synaptodendritic abnormalities and altered behavior in mice, [24] K. Brami-Cherrier, E. Valjent, M. Garcia, C. Pagès, R.A. Hipskind, J. Caboche,
Neurobiol. Dis. 67 (2014) 79–87. Dopamine induces a PI3-kinase-independent activation of Akt in striatal neurons:
[8] D. Avtanski, V.A. Pavlov, K.J. Tracey, L. Poretsky, Characterization of a new route to cAMP response element-binding protein phosphorylation,
inflammation and insulin resistance in high-fat diet-induced male C57BL/6 J J. Neurosci. 22 (20) (2002) 8911–8921. Oct 15.
mouse model of obesity, Animal Model Exp. Med. 2 (2019) 252–258.

10
S. Sharma Physiology & Behavior 240 (2021) 113528

[25] R. Buettner, K.G. Parhofer, M. Woenckhaus, et al., Defining high-fat-diet rat [53] M.F. Egan, M. Kojima, J.H. Callicott, T.E. Goldberg, B.S. Kolachana, A. Bertolino,
models: metabolic and molecular effects of different fat types, J. Mol. Endocrinol. et al., The BDNF val66met polymorphism affects activity-dependent secretion of
36 (3) (2006) 485–501. BDNF and human memory and hippocampal function, Cell. 2 (2) (2003)
[26] E. Calvo-Ochoa, K. Hernández-Ortega, P. Ferrera, S. Morimoto, C. Arias, Short- 257–269. Jan 24;11.
term high-fat-and-fructose feeding produces insulin signaling alterations [54] B.A. Ellis, A. Poynten, A.J. Lowy, S.M. Furler, D.J. Chisholm, E.W. Kraegen, et al.,
accompanied by neurite and synaptic reduction and astroglial activation in the Long-chain acyl-CoA esters as indicators of lipid metabolism and insulin
rat hippocampus, J. Cereb. Blood Flow Metab. 34 (6) (2014) 1001–1008. sensitivity in rat and human muscle, Am. J. Physiol. Endocrinol. Metab. 279
[27] G.Y. Cao, M. Li, L. Han, et al., Dietary fat intake and cognitive function among (2000) E554–E560, https://doi.org/10.1152/ajpendo.2000.279.3.E554.
older populations: a systematic review and meta-analysis, J. Prev. Alzheimers Dis. [55] J.R. Erion, M. Wosiski-Kuhn, A. Dey, et al., Obesity elicits interleukin 1-mediated
6 (3) (2019) 204–211. deficits in hippocampal synaptic plasticity, J. Neurosci. 34 (7) (2014) 2618–2631.
[28] I. Casserly, E. Topol, Convergence of atherosclerosis and Alzheimer’s disease: [56] D. Erny, A.L. Hrabe de Angelis, D. Jaitin, P. Wieghofer, O. Staszewski, E. David, et
inflammation, cholesterol, and misfolded proteins, Lancet. 363 (9415) (2004) al., Host microbiota constantly control maturation and function of microglia in
1139–1146. the CNS, Nat. Neurosci. 18 (2015) 965–977, https://doi.org/10.1038/nn.4030.
[29] G. Cavaliere, G. Trinchese, E. Penna, et al., High-Fat diet induces [57] M.H. Eskelinen, T. Ngandu, E.L. Helkala, et al., Fat intake at midlife and cognitive
neuroinflammation and mitochondrial impairment in mice cerebral cortex and impairment later in life: a population-based CAIDE study, Int. J. Geriatr.
synaptic fraction, Front. Cell Neurosci. 13 (2019) 509, https://doi.org/10.3389/ Psychiatry. 23 (7) (2008) 741–747.
fncel.2019.00509. Published 2019 Nov 12. [58] J.L. Evans, I.D. Goldfine, B.A. Maddux, G.M. Grodsky, Are oxidative stress
[30] S. Chatterjee, S.A. Peters, M. Woodward, S. Mejia Arango, G.D. Batty, N. Beckett, activated signaling pathways mediators of insulin resistance and β-cell
et al., Type 2 diabetes as a risk factor for dementia in women compared with men: dysfunction? Diabetes. 52 (2003) 1–8.
a pooled analysis of 2.3 million people comprising more than 100,000 cases of [59] S.A. Farr, K.A. Yamada, D.A. Butterfield, H.M. Abdul, L. Xu, N.E. Miller, Obesity
dementia, Diabetes Care 39 (2) (2016) 300–307. and hypertriglyceridemia produce cognitive impairment, Endocrinology. 149 (5)
[31] J.A. Chavez, T.A. Knotts, L.P. Wang, G. Li, R.T. Dobrowsky, G.L. Florant, et al., (2008) 2628–2636.
A role for ceramide, but not diacylglycerol, in the antagonism of insulin signal [60] S.T. Ferreira, J.R. Clarke, T.R. Bomfim, F.G. De Felice, Inflammation, defective
transduction by saturated fatty acids, J. Biol. Chem. 278 (2003) 10297–10303, insulin signaling, and neuronal dysfunction in Alzheimer’s disease, Alzheimers
https://doi.org/10.1074/jbc.M21230720. Dement. 10 (2014) S76–S83.
[32] S.L. Chiu, C.M. Chen, H.T. Cline, Insulin receptor signaling regulates synapse [61] S. Finkbeiner, CREB couples neurotrophin signals to survival messages, Neuron.
number, dendritic plasticity, and circuit function in vivo, Neuron. 58 (2008) 25 (1) (2000) 11–14. Jan.
708–719. [62] T.C. Foster, Biological markers of age-related memory deficits: treatment of
[33] M. Cifre, A. Palou, P. Oliver, Cognitive impairment in metabolically-obese, senescent physiology, CNS Drugs. 20 (2) (2006) 153–166.
normal-weight rats: identification of early biomarkers in peripheral blood [63] H.M. Francis, M. Mirzaei, M.C. Pardey, P.A. Haynes, J.L. Cornish, Proteomic
mononuclear cells, Mol. Neurodegener. 13 (1) (2018) 14. analysis of the dorsal and ventral hippocampus of rats maintained on a high fat
[34] L Claudio, Ultrastructural features of the blood-brain barrier in biopsy tissue from and refined sugar diet, Proteomics. 13 (20) (2013) 3076–3091.
Alzheimer’s disease patients, Acta Neuropathol. 91 (1) (1996) 6–14. [64] L.R. Freeman, A.C. Granholm, Vascular changes in rat hippocampus following a
[35] D.J. Clegg, K. Gotoh, C. Kemp, et al., Consumption of a high-fat diet induces high saturated fat and cholesterol diet, J. Cereb. Blood Flow Metab. 32 (4) (2012)
central insulin resistance independent of adiposity, Physiol. Behav. 103 (1) 643–653.
(2011) 10–16. [65] S.J. Gainey, K.A. Kwakwa, J.K. Bray, et al., Short-term high-fat diet (HFD)
[36] B. Connor, D. Young, Q. Yan, R.L. Faull, B. Synek, M. Dragunow, Brain-derived induced anxiety-like behaviors and cognitive impairment are improved with
neurotrophic factor is reduced in Alzheimer’s disease, Brain Res. Mol. Brain Res. treatment by glyburide, Front. Behav. Neurosci. 10 (2016) 156.
49 (1–2) (1997) 71–81. Oct 3. [66] L. Gan, E. England, J.Y. Yang, N. Toulme, S. Ambati, D.L. Hartzell, et al., A 72-
[37] Z.A. Cordner, K.L. Tamashiro, Effects of high-fat diet exposure on learning & hour high fat diet increases transcript levels of the neuropeptide galanin in the
memory, Physiol. Behav. 152 (Pt B) (2015) 363–371. dorsal hippocampus of the rat, BMC Neurosci. 16 (2015) 51.
[38] P.K. Crane, R. Walker, R.A. Hubbard, et al., Glucose levels and risk of dementia [67] V.R. Garbarino, M.E. Orr, K.A. Rodriguez, R. Buffenstein, Mechanisms of
[published correction appears in N Engl J Med. 2013 Oct 10;369(15):1476], oxidative stress resistance in the brain: lessons learned from hypoxia tolerant
N. Engl. J. Med. 369 (6) (2013) 540–548. extremophilic vertebrates, Arch. Biochem. Biophys. 576 (2015) 8–16.
[39] R. Crescenzo, F. Bianco, A. Mazzoli, et al., Fat quality influences the obesogenic [68] S.L.C. Geijselaers, S.J.S. Sep, C.D.A. Stehouwer, G.J. Biessels, Glucose regulation,
effect of high fat diets, Nutrients. 7 (11) (2015) 9475–9491. Published 2015 Nov cognition, and brain MRI in type 2 diabetes: a systematic review, Lancet Diabetes
16. Endocrinol. 3 (1) (2015) 75–89.
[40] Y. Cui, X. Liang, H. Gu, et al., Cerebral perfusion alterations in type 2 diabetes and [69] R. Godar, Y. Dai, H. Bainter, C. Billington, C.M. Kotz, C.F. Wang, Reduction of
its relation to insulin resistance and cognitive dysfunction, Brain Imaging Behav. high-fat diet-induced obesity after chronic administration of brain-derived
11 (5) (2017) 1248–1257. neurotrophic factor in the hypothalamic ventromedial nucleus, Neurosci. 194
[41] P. Dandona, I.M. James, P.A. Newbury, M.L. Woollard, A.G. Beckett, Cerebral (2011) 36–52.
blood flow in diabetes mellitus: evidence of abnormal cerebrovascular reactivity, [70] F. Gomez-Pinilla, E. Tyagi, Diet and cognition: interplay between cell metabolism
Br Med. J. 2 (6133) (1978) 325–326. and neuronal plasticity, Curr. Opin. Clin. Nutr. Metab. Care. 16 (6) (2013)
[42] R. Dantzer, J.C. O’Connor, G.G. Freund, R.W. Johnson, K.W. Kelley, From 726–733.
inflammation to sickness and depression: when the immune system subjugates [71] A.C. Granholm, H.A. Bimonte-Nelson, A.B. Moore, M.E. Nelson, L.R. Freeman,
the brain, Nat. Rev. Neurosci. 9 (2008) 46–56. K. Sambamurti, Effects of a saturated fat and high cholesterol diet on memory and
[43] A.K. Datusalia, S.S. Sharma, Amelioration of diabetes-induced cognitive deficits hippocampal morphology in the middle-aged rat, J. Alzheimers Dis. 14 (2) (2008)
by GSK-3β inhibition is attributed to modulation of neurotransmitters and 133–145.
neuroinflammation, Mol. Neurobiol. 50 (2) (2014) 390–405. [72] C.E. Greenwood, G. Winocur, High-fat diets, insulin resistance and declining
[44] T.L. Davidson, S.L. Hargrave, S.E. Swithers, et al., Inter-relationships among diet, cognitive function, Neurobiol. Aging. 26 (2005) 42–45. Suppl 1.
obesity and hippocampal-dependent cognitive function, Neurosci. 253 (2013) [73] C.E. Greenwood, G. Winocur, Learning and memory impairment in rats fed a high
110–122. saturated fat diet, Behav. Neural. Biol. 53 (1) (1990) 74–87.
[45] T.L. Davidson, A. Monnot, A.U. Neal, A.A. Martin, J.J. Horton, W. Zheng, The [74] S. Hao, A. Dey, X. Yu, A.M. Stranahan, Dietary obesity reversibly induces synaptic
effects of a high-energy diet on hippocampal-dependent discrimination stripping by microglia and impairs hippocampal plasticity, Brain Behav. Immun.
performance and blood–brain barrier integrity differ for diet-induced obese and 51 (2016) 230–239.
diet-resistant rats, Physiol. Behav. 107 (1) (2012) 26–33. [75] S. Hao, A. Dey, X. Yu, A.M. Stranahan, Dietary obesity reversibly induces synaptic
[46] F.G. De Felice, M.V. Lourenco, S.T. Ferreira, How does brain insulin resistance stripping by microglia and impairs hippocampal plasticity, Brain Behav. Immun.
develop in Alzheimer’s disease? Alzheimers Dement. 10 (1 Suppl) (2014) 51 (2016) 230–239. Jan.
S26–S32. [76] S.I. Harik, R.N. Kalaria, Blood-brain barrier abnormalities in Alzheimer’s disease,
[47] M.R. de Oliveira, R.F. da Rocha, L. Stertz, G.R. Fries, D.L. de Oliveira, Ann. N Y Acad Sci. 640 (1991) 47–52.
F. Kapczinski, Total and mitochondrial nitrosative stress, decreased brain derived [77] N. Hariri, R. Gougeon, L. Thibault, A highly saturated fat-rich diet is more
neurotrophic factor (BDNF) levels and glutamate uptake, and evidence of obesogenic than diets with lower saturated fat content, Nutr Res 30 (9) (2010)
endoplasmic reticulum stress in the hippocampusof vitamin A treated rats, 632–643. Sep.
Neurochem. Res. 36 (3) (2011) 506–517. [78] E. Head, Oxidative damage and cognitive dysfunction: antioxidant treatments to
[48] J.P. DeLany, M.M. Windhauser, C.M. Champagne, G.A. Bray, Differential promote healthy brain aging, Neurochem. Res. 34 (2009) 670–678.
oxidation of individual dietary fatty acids in humans, Am. J. Clin. Nutr. 72 (4) [79] J. Henkel, C.D. Coleman, A. Schraplau, et al., Induction of steatohepatitis (NASH)
(2000) 905–911. Oct. with insulin resistance in wildtype B6 mice by a western-type diet containing
[49] G.A. Dienel, Fueling and imaging brain activation, ASN Neuro. 4 (5) (2012). soybean oil and cholesterol, Mol. Med. 23 (2017) 70–82.
[50] P.M. Dingess, R.A. Darling, E. Kurt Dolence, B.W. Culver, T.E Brown, Exposure to [80] B. Herculano, M. Tamura, A. Ohba, M. Shimatani, N. Kutsuna, T. Hisatsune,
a diet high in fat attenuates dendritic spine density in the medial prefrontal β-alanyl-L-histidine rescues cognitive deficits caused by feeding a high fat diet in a
cortex, Brain Struct. Funct. 222 (2) (2017) 1077–1085. transgenic mouse model of Alzheimer’s disease, J. Alzheimers Dis. 33 (4) (2013)
[51] K. Du, M. Montminy, CREB is a regulatory target for the protein kinase Akt/PKB, 983–997.
J. Biol. Chem. 273 (49) (1998) 32377–32379. Dec 4. [81] A. Heydemann, An overview of murine high fat diet as a model for type 2 diabetes
[52] L.M. Edwards, A.J. Murray, C.J. Holloway, E.E. Carter, G.J. Kemp, I. Codreanu, et mellitus, J. Diabetes Res. 2016 (2016) 1–14.
al., Short-term consumption of a high-fat diet impairs whole-body efficiency and
cognitive function in sedentary men, FASEB J. 25 (2011) 1088–1096.

11
S. Sharma Physiology & Behavior 240 (2021) 113528

[82] L. Ho, W. Qin, P.N. Pompl, et al., Diet-induced insulin resistance promotes [109] L.S. Le, G. Simard, M.C. Martinez, R. Andriantsitohaina, Oxidative stress and
amyloidosis in a transgenic mouse model of Alzheimer’s disease, FASEB J. 18 (7) metabolic pathologies: from an adipocentric point of view, Oxid. Med. Cell
(2004) 902–904. Longev. 2014 (2014), 908539.
[83] C. Holmes, C. Cunningham, E. Zotova, J. Woolford, C. Dean, S. Kerr, et al., [110] G. Leal, P.M. Afonso, I.L. Salazar, C.B. Duarte, Regulation of hippocampal
Systemic inflammation and disease progression in Alzheimer disease, Neurol 73 synaptic plasticity by BDNF, Brain Res. 1621 (2015) 82–101. Sep 24.
(10) (2009) 768–774. [111] C.C. Lee, C.C. Huang, M.Y. Wu, K.S. Hsu, Insulin stimulates postsynaptic density-
[84] J.D. Huber, Diabetes, cognitive function, and the blood-brain barrier, Curr. 95 protein translation via the phosphoinositide 3-kinase-Akt-mammalian target of
Pharm. Des. 14 (16) (2008) 1594–1600. rapamycin signaling pathway, J. Biol. Chem. 280 (2005) 18543–18550.
[85] L.L. Hwang, C.H. Wang, T.L. Li, et al., Sex differences in high-fat diet-induced [112] C.L. Leibson, W.A. Rocca, V.A. Hanson, et al., Risk of dementia among persons
obesity, metabolic alterations and learning, and synaptic plasticity deficits in with diabetes mellitus: a population-based cohort study, Am. J. Epidemiol. 145
mice, Obesity. 18 (3) (2010) 463–469. (4) (1997) 301–308.
[86] B.A. in ‘t Veld, L.J. Launer, A.W. Hoes, A. Ott, A. Hofman, M.M. Breteler, et al., [113] A. Lindqvist, P. Mohapel, B. Bouter, H. Frielingsdorf, D. Pizzo, P. Brundin,
NSAIDs and incident Alzheimer’s disease, The Rotterdam Study. Neurobiol. C. Erlanson-Albertsson, High-fat diet impairs hippocampal neurogenesis in male
Aging. 19 (6) (1998) 607–611. rats, Eur. J. Neurol. 13 (12) (2006) 1385–1388. Dec.
[87] S. Ito, M. Yanai, S. Yamaguchi, P.O. Couraud, S. Ohtsuki, Regulation of tight- [114] X. Liu, Z. Zhu, M. Kalyani, J.M. Janik, H. Shi, Effects of energy status and diet on
junction integrity by insulin in an in vitro model of human blood-brain barrier, Bdnf expression in the ventromedial hypothalamus of male and female rats,
J. Pharm. Sci. 106 (9) (2017) 2599–2605. Sep. Physiol. Behav. 130 (2014) 99–107, a.
[88] Y. Izumi, K.A. Yamada, M. Matsukawa, C.F. Zorumski, Effects of insulin on long- [115] Y. Liu, X. Fu, N. Lan, et al., Luteolin protects against high fat diet-induced
term potentiation in hippocampal slices from diabetic rats, Diabetologia. 46 (7) cognitive deficits in obesity mice, Behav. Brain Res. 267 (2014) 178–188, b.
(2003) 1007–1012. [116] Z. Liu, I.Y. Patil, T. Jiang, et al., High-fat diet induces hepatic insulin resistance
[89] B.T. Jeon, E.A. Jeong, H.J. Shin, Y. Lee, D.H. Lee, H.J. Kim, S.S. Kang, G.J. Cho, and impairment of synaptic plasticity, PLoS One. 10 (5) (2015), e0128274.
W.S. Choi, G.S. Roh, Resveratrol attenuates obesity-associated peripheral and [117] I. Lozano, R. Van der Werf, W. Bietiger, et al., High-fructose and high-fat diet-
central inflammation and improves memory deficit in mice fed a high-fat diet, induced disorders in rats: impact on diabetes risk, hepatic and vascular
Diabetes. 61 (2012) 1444–1454. complications, Nutr. Metab. (Lond) 13 (2016) 15.
[90] S.E. Kanoski, T.L. Davidson, Western diet consumption and cognitive impairment: [118] J. Lu, D.M. Wu, Y.L. Zheng, et al., Ursolic acid improves high fat diet-induced
links to hippocampal dysfunction and obesity, Physiol Behav. 103 (1) (2011) cognitive impairments by blocking endoplasmic reticulum stress and IκB kinase
59–68. β/nuclear factor-κB-mediated inflammatory pathways in mice, Brain Behav.
[91] S.E. Kanoski, Y. Zhang, W. Zheng, T.L. Davidson, The effects of a high-energy diet Immun. 25 (8) (2011) 1658–1667, a.
on hippocampal function and blood-brain barrier integrity in the rat, [119] J. Lu, D.M. Wu, Z.H. Zheng, Y.L. Zheng, B. Hu, Z.F. Zhang, Troxerutin protects
J. Alzheimers Dis. 21 (1) (2010) 207–219. against high cholesterol-induced cognitive deficits in mice, Brain. 134 (Pt 3)
[92] S.A. Karimi, I. Salehi, A. Komaki, A. Sarihi, M. Zarei, S. Shahidi, Effect of high-fat (2011) 783–797, b.
diet and antioxidants on hippocampal long-term potentiation in rats: an in vivo [120] C. Luo, H. Yang, C. Tang, et al., Kaempferol alleviates insulin resistance via
study, Brain Res. 1539 (2013) 1–6. Nov 20. hepatic IKK/NF-κB signal in type 2 diabetic rats, Int. Immunopharmacol. 28 (1)
[93] A.R. Kessler, B. Kessler, S. Yehuda, In vivo modulation of brain cholesterol level (2015) 744–750.
and learning performance by a novel plant lipid: indications for interactions [121] J. Luo, J. Quan, J. Tsai, et al., Nongenetic mouse models of non-insulin-dependent
between hippocampal-cortical cholesterol and learning, Life Sci. 38 (13) (1986) diabetes mellitus, Metabolism. 47 (6) (1998) 663–668.
1185–1192. Mar 31. [122] M.C. Petersen, G.I. Shulman, Roles of diacylglycerols and ceramides in hepatic
[94] T. Kishi, Y. Hirooka, T. Nagayama, K. Isegawa, M. Katsuki, K. Takesue, et al., insulin resistance, Trends Pharmacol. Sci. 38 (2017) 649–665.
Calorie restriction improves cognitive decline via up-regulation of brain-derived [123] M. Roden, et al., Mechanism of free fatty acid-induced insulin resistance in
neurotrophic factor: tropomyosin-related kinase B in hippocampus ofobesity- humans, J. Clin. Invest. 97 (1996) 2859–2865.
induced hypertensive rats, Int. Heart J. 56 (1) (2015) 110–115. [124] A.M. Magariños, B.S. McEwen, Experimental diabetes in rats causes hippocampal
[95] M. Kivimäki, A. Singh-Manoux, Prevention of dementia by targeting risk factors, dendritic and synaptic reorganization and increased glucocorticoid reactivity to
LancetLancet 391 (2018) 1574–1575. stress, Proc. Natl. Acad Sci. U S A. 97 (20) (2000) 11056–11061. Sep 26.
[96] M. Kivipelto, T. Ngandu, L. Fratiglioni, et al., Obesity and vascular risk factors at [125] N. Matsuzawa-Nagata, T. Takamura, H. Ando, S. Nakamura, S. Kurita, H. Misu, et
midlife and the risk of dementia and Alzheimer disease, Arch. Neurol. 62 (10) al., Increased oxidative stress precedes the onset of high-fat diet-induced insulin
(2005) 1556–1560. resistance and obesity, Metabolism. 57 (8) (2008) 1071–1077.
[97] D.J. Klemm, W.J. Roesler, T. Boras, L.A. Colton, K. Felder, J.E. Reusch, Insulin [126] M.P. Mattson, Energy intake and exercise as determinants of brain health and
stimulates cAMP-response element binding protein activity in HepG2 and 3T3-L1 vulnerability to injury and disease, Cell Metab. 16 (2012) 706–722.
cell lines, J. Biol. Chem. 273 (2) (1998) 917–923. Jan 9. [127] J.C. McCann, B.N. Ames, Is docosahexaenoic acid, an n-3 long-chain
[98] E.M. Knight, I.V. Martins, S. Gümüsgöz, S.M. Allan, C.B. Lawrence, High-fat diet- polyunsaturated fatty acid, required for development of normal brain function?
induced memory impairment in triple-transgenic Alzheimer’s disease (3xTgAD) an overview of evidence from cognitive and behavioral tests in humans and
mice is independent of changes in amyloid and tau pathology, Neurobiol. Aging. animals. Am. J. Clin. Nutr. 82 (2005) 281–295.
35 (8) (2014) 1821–1832. [128] F.H. McLean, C. Grant, A.C. Morris, et al., Rapid and reversible impairment of
[99] M. Kohjima, Y. Sun, L. Chan, Increased food intake leads to obesity and insulin episodic memory by a high-fat diet in mice, Sci. Rep. 8 (1) (2018) 11976.
resistance in the tg2576 Alzheimer’s disease mouse model, Endocrinology. 151 [129] A.D. McNeilly, R. Williamson, C. Sutherland, D.J. Balfour, C.A. Stewart, High fat
(4) (2010) 1532–1540. feeding promotes simultaneous decline in insulin sensitivity and cognitive
[100] R. Kohli, M. Kirby, S.A. Xanthakos, et al., High-fructose, medium chain trans fat performance in a delayed matching and non-matching to position task, Behav.
diet induces liver fibrosis and elevates plasma coenzyme Q9 in a novel murine Brain Res. 217 (1) (2011) 134–141.
model of obesity and nonalcoholic steatohepatitis, Hepatology. 52 (3) (2010) [130] J.G. Mielke, K. Nicolitch, V. Avellaneda, et al., Longitudinal study of the effects of
934–944. a high-fat diet on glucose regulation, hippocampal function, and cerebral insulin
[101] E. Koponen, M. Lakso, E. Castrén, Overexpression of the full-length neurotrophin sensitivity in C57BL/6 mice, Behav. Brain Res. 175 (2) (2006) 374–382.
receptor trkB regulates the expression of plasticity-related genes in mouse brain, [131] A.A. Miller, S.J. Spencer, Obesity and neuroinflammation: a pathway to cognitive
Brain Res. Mol. Brain Res. 130 (1–2) (2004) 81–94. Nov 4. impairment, Brain Behav. Immun. 42 (2014) 10–21.
[102] N. Korte, R. Nortley, D. Attwell, Cerebral blood flow decrease as an early [132] R. Molteni, J.R. Barnard, Z. Ying, C.K. Roberts, F. Gomez-Pinilla, A high-fat,
pathological mechanism in Alzheimer’s disease, Acta Neuropathol. 140 (2020) refined sugar diet reduces hippocampal brain-derived neurotrophic factor,
793–810. neuronal plasticity, and learning, Neurosci. 112 (2002) 803–814.
[103] E.W. Kraegen, P.W. Clark, A.B. Jenkins, E.A. Daley, D.J. Chisholm, L.H. Storlien, [133] R. Molteni, A. Wu, S. Vaynman, Z. Ying, R.J. Barnard, F. Gómez-Pinilla, Exercise
Development of muscle insulin resistance after liver insulin resistance in high-fat- reverses the harmful effects of consumption of a high-fat diet on synaptic and
fed rats, Diabetes. 40 (11) (1991) 1397–1403. behavioral plasticity associated to the action of brain-derived neurotrophic factor,
[104] Z. Kuai, Y. Xu, Q. Zhao, J. Liu, S. Guan, Y. Qiao, X. Gong, J. Nie, P. Li, D. Liu, Neurosci. 123 (2) (2004) 429–440.
Y. Xing, H. Li, Z. Sun, W. Wang, C. Ning, Y. Shi, W. Kong, Y. Shan, Effects of [134] P.I. Moreira, M.S. Santos, C.R. Oliveira, J.C. Shenk, A. Nunomura, M.A. Smith,
insulin on transcriptional response and permeability in an in vitro model of X. Zhu, G. Perry, Alzheimer disease and the role of free radicals in the
human blood-brain barrier, J. Cell Biochem. 119 (7) (2018) 5657–5664. Jul. pathogenesis of the disease, CNS Neurol. Disord. Drug. Targets. 7 (2008) 3–10.
[105] M.H. Laitinen, T. Ngandu, S. Rovio, et al., Fat intake at midlife and risk of [135] C.D. Morrison, P.J. Pistell, D.K. Ingram, et al., High fat diet increases hippocampal
dementia and Alzheimer’s disease: a population-based study, Dement. Geriatr. oxidative stress and cognitive impairment in aged mice: implications for
Cogn. Disord. 22 (1) (2006) 99–107. decreased Nrf2 signaling, J. Neurochem. 114 (6) (2010) 1581–1589.
[106] P. Lang, S. Hasselwander, H. Li, et al., Effects of different diets used in diet- [136] Nagai, A., Mizushige, T., Matsumura, S., Inoue, K., & Ohinata, K. (2019). Orally
induced obesity models on insulin resistance and vascular dysfunction in C57BL/ administered milk-derived tripeptide improved cognitive decline in mice fed a
6 mice, Sci. Rep. 9 (2019) 19556. high-fat diet. FASEB J., fj.201900621R.
[107] C. Laske, E. Stransky, T. Leyhe, G.W. Eschweiler, A. Wittorf, E. Richartz, [137] S. Nagamachi, T. Nishikawa, S. Ono, et al., Regional cerebral blood flow in
M. Bartels, G. Buchkremer, K. Schott, Stage-dependent BDNF serum diabetic patients: evaluation by N-isopropyl-123I-IMP with SPECT, Nucl. Med.
concentrations in Alzheimer’s disease, J. Neural. Transm. (Vienna) 113 (9) (2006) Commun. 15 (6) (1994) 455–460.
1217–1224. Sep. [138] T. Nakagawa, M. Ono-Kishino, E. Sugaru, M. Yamanaka, M. Taiji, H. Noguchi,
[108] D. Last, D.C. Alsop, A.M. Abduljalil, et al., Global and regional effects of type 2 Brain-derived neurotrophic factor (BDNF) regulates glucose and energy
diabetes on brain tissue volumes and cerebral vasoreactivity, Diabetes Care. 30 metabolism in diabetic mice, Diabetes Metab. Res. Rev. 18 (3) (2002) 185–191.
(5) (2007) 1193–1199. May-Jun.

12
S. Sharma Physiology & Behavior 240 (2021) 113528

[139] Kumar A Neha, A.S. Jaggi, R.K. Sodhi, N Singh, Silymarin ameliorates memory VGlut1 expression in high-fat-fed rats: implications for Alzheimer’s disease,
deficits and neuropathological changes in mouse model of high-fat-diet-induced Biochim. Biophys. Acta. 1862 (4) (2016) 511–517.
experimental dementia, Naunyn. Schmiedebergs Arch. Pharmacol. 387 (8) (2014) [167] J. Rogers, D. Mastroeni, B. Leonard, J. Joyce, A. Grover, Neuroinflammation in
777–787. Alzheimer’s disease and Parkinson’s disease: are microglia pathogenic in either
[140] S. Nishikawa, A. Yasoshima, K. Doi, H. Nakayama, K. Uetsuka, Involvement of disorder? Int. Rev. Neurobiol. 82 (2007) 235–246.
sex, strain and age factors in high fat diet-induced obesity in C57BL/6 J and [168] O. Sabri, D. Hellwig, M. Schreckenberger, et al., Influence of diabetes mellitus on
BALB/cA mice, Exp. Anim. 56 (4) (2007) 263–272. regional cerebral glucose metabolism and regional cerebral blood flow. Nucl.
[141] S. Ogata, S. Ito, T. Masuda, S. Ohtsuki, Changes of blood-brain barrier and brain Med. Commun. 21 (1) (2000) 19–29.
parenchymal protein expression levels of mice under different insulin-resistance [169] S.K. Sah, C. Lee, J.H. Jang, G.H. Park, Effect of high-fat diet on cognitive
conditions induced by high-fat diet, Pharm. Res. 36 (10) (2019) 141. Jul 31. impairment in triple-transgenic mice model of Alzheimer’s disease, Biochem.
[142] H. Oh, S. Boghossian, D.A. York, M. Park-York, The effect of high fat diet and Biophys. Res. Commun. 493 (1) (2017) 731–736.
saturated fatty acids on insulin signaling in the amygdala and hypothalamus of [170] T.S. Salameh, G.N. Shah, T.O. Price, M.R. Hayden, W.A. Banks, Blood-brain
rats, Brain research. 1537 (2013) 191–200. barrier disruption and neurovascular unit dysfunction in diabetic mice: protection
[143] T. Ohara, Y. Doi, T. Ninomiya, Y. Hirakawa, J. Hata, T. Iwaki, et al., Glucose with the mitochondrial carbonic anhydrase inhibitor topiramate, J. Pharmacol.
tolerance status and risk of dementia in the community: the hisayama study, Exp. Ther. 359 (3) (2016) 452–459.
NeurologyNeurol. 77 (2011) 1126–1134. [171] L.T. Samuels, R.M. Reinecke, H.A. Ball, Effect of diet on glucose tolerance and
[144] M.S. Ola, M.I. Nawaz, A.A. El-Asrar, M. Abouammoh, A.S. Alhomida, Reduced liver and muscle glycogen of hypophysectomized and normal rats, Endocrinol. 31
levels of brain derived neurotrophic factor (BDNF) in the serum of diabetic (1942) 42–53.
retinopathy patients and in the retina of diabetic rats, Cell Mol. Neurobiol. 33 (3) [172] K.E. Sandoval, K.A. Witt, Blood-brain barrier tight junction permeability and
(2013) 359–367. Apr. ischemic stroke, Neurobiol. Dis. 32 (2) (2008) 200–219.
[145] N. Pamidi, B.N. Satheesha Nayak, Effect of streptozotocin induced diabetes on rat [173] C. Sathiya Priya, R. Vidhya, K. Kalpana, C.V. Anuradha, Indirubin-3′ -monoxime
hippocampus, Bratisl. Lek. Listy. 113 (10) (2012) 583–588. prevents aberrant activation of GSK-3β/NF-κB and alleviates high fat-high
[146] H.R. Park, M. Park, J. Choi, K.Y. Park, H.Y. Chung, J. Lee, A high-fat diet impairs fructose induced Aβ-aggregation, gliosis and apoptosis in mice brain, Int.
neurogenesis: involvement of lipid peroxidation and brain-derived neurotrophic Immunopharmacol. 70 (2019) 396–407.
factor, Neurosci. Lett. 482 (3) (2010) 235–239. [174] C. Schmitz-Peiffer, Protein kinase C and lipid-induced insulin resistance in
[147] H.S. Park, S.S. Park, C.J. Kim, M.S. Shin, T.W. Kim, Exercise alleviates cognitive skeletal muscle, Ann. N. Y. Acad Sci. 967 (2002) 146–157, https://doi.org/
functions by enhancing hippocampal insulin signaling and neuroplasticity in 10.1111/j.1749-6632.2002.tb04272.x.
high-fat diet-induced obesity, Nutrients. 11 (7) (2019) 1603. [175] L. Shahriyary, G. Riazi, M.R. Lornejad, M. Ghezlou, B. Bigdeli, B. Delavari,
[148] A.R. Pathan, A.B. Gaikwad, B. Viswanad, P. Ramarao, Rosiglitazone attenuates F. Mamashli, S. Abbasi, J. Davoodi, A.A. Saboury, Effect of glycated insulin on the
the cognitive deficits induced by high fat diet feeding in rats, Eur. J. Pharmacol. blood-brain barrier permeability: an in vitro study, Arch. Biochem. Biophys. 647
589 (1–3) (2008) 176–179. (2018) 54–66. Jun 1.
[149] G. Perry, A. Nunomura, K. Hirai, et al., Is oxidative damage the fundamental [176] S. Sharma, K. Kumar, R. Deshmukh, P.L. Sharma, Phosphodiesterases: regulators
pathogenic mechanism of Alzheimer’s and other neurodegenerative diseases? of cyclic nucleotide signals and novel molecular target for movement disorders,
Free Radic. Biol. Med. 33 (11) (2002) 1475–1479. Eur. J. Pharmacol. 714 (1–3) (2013) 486–497.
[150] Y. Persidsky, S. Ramirez, J. Haorah, G. Kanmogne, Blood-brain barrier: structural [177] S. Sharma, R. Taliyan, Epigenetic modifications by inhibiting histone deacetylases
components and function under physiologic and pathologic conditions, reverse memory impairment in insulin resistance induced cognitive deficit in
J. Neuroimmune. Pharmacol. 1 (2006) 223–236. mice, Neuropharmacol. 105 (2016) 285–297.
[151] D. Pessayre, A. Berson, B. Fromenty, A. Mansouri, Mitochondria in [178] S. Sharma, R. Taliyan, Neuroprotective role of Indirubin-3′ -monoxime, a GSKβ
steatohepatitis, Semin. Liver Dis. 21 (2001) 57–69. inhibitor in high fat diet induced cognitive impairment in mice, Biochem.
[152] D. Pessayre, A. Mansouri, B. Fromenty, Nonalcoholic steatosis and steatohepatitis, Biophys. Res. Commun. 452 (4) (2014) 1009–1015.
Mitochondrial dysfunction in steatohepatitis. Am. J. Physiol. Gastrointest. Liver [179] S. Sharma, R. Taliyan, Synergistic effects of GSK-3β and HDAC inhibitors in
Physiol. 282 (2) (2002) G193–G199. intracerebroventricular streptozotocin-induced cognitive deficits in rats, Naunyn.
[153] A.E. Petro, J. Cotter, D.A. Cooper, J.C. Peters, S.J. Surwit, R.S. Surwit, Fat, Schmiedebergs Arch. Pharmacol. 388 (3) (2015) 337–349.
carbohydrate, and calories in the development of diabetes and obesity in the [180] G. Sharon, T.R. Sampson, D.H. Geschwind, S.K. Mazmanian, The central nervous
C57BL/6 J mouse, Metabolism. 53 (2004) 454–457. system and the gut microbiome, Cell 167 (2016) 915–932, https://doi.org/
[154] H.S. Phillips, J.M. Hains, M. Armanini, G.R. Laramee, S.A. Johnson, J. 10.1016/j.cell.2016.10.027.
W. Winslow, BDNF mRNA is decreased in the hippocampus of individuals with [181] A.J. Shaywitz, M.E. Greenberg, CREB: a stimulus-induced transcription factor
Alzheimer’s disease, Neuron. 7 (5) (1991) 695–702. Nov. activated by a diverse array of extracellular signals, Annu. Rev. Biochem. 68
[155] H. Pintana, N. Apaijai, N. Chattipakorn, S.C. Chattipakorn, DPP-4 inhibitors (1999) 821–861.
improve cognition and brain mitochondrial function of insulin-resistant rats, [182] P.W. Siri-Tarino, Q. Sun, F.B. Hu, R.M. Krauss, Saturated fat, carbohydrate, and
J. Endocrinol. 218 (1) (2013) 1–11. cardiovascular disease, Am. J. Clin. Nutr. 91 (3) (2010) 502–509.
[156] H. Pintana, N. Apaijai, W. Pratchayasakul, N. Chattipakorn, S.C. Chattipakorn, [183] R.K. Sodhi, N. Singh, Liver X receptor agonist T0901317 reduces
Effects of metformin on learning and memory behaviors and brain mitochondrial neuropathological changes and improves memory in mouse models of
functions in high fat diet induced insulin resistant rats, Life Sci. 91 (11–12) (2012) experimental dementia, Eur. J. Pharmacol. 732 (2014) 50–59.
409–414. [184] A. Solomon, I. Kåreholt, T. Ngandu, B. Winblad, A. Nissinen, J. Tuomilehto,
[157] N. Pipatpiboon, H. Pintana, W. Pratchayasakul, N. Chattipakorn, S. H. Soininen, M. Kivipelto, Serum cholesterol changes after midlife and late-life
C. Chattipakorn, DPP4-inhibitor improves neuronal insulin receptor function, cognition: twenty-one-year follow-up study, Neurol. 68 (10) (2007) 751–756. Mar
brain mitochondrial function and cognitive function in rats with insulin resistance 6.
induced by high-fat diet consumption, Eur. J. Neurosci. 37 (5) (2013) 839–849. [185] A. Solomon, M. Kivipelto, B. Wolozin, J. Zhou, R.A. Whitmer, Midlife serum
[158] P.J. Pistell, C.D. Morrison, S. Gupta, et al., Cognitive impairment following high cholesterol and increased risk of Alzheimer’s and vascular dementia three
fat diet consumption is associated with brain inflammation, J. Neuroimmunol. decades later, Dement. Geriatr. Cogn. Disord. 28 (1) (2009) 75–80, https://doi.
219 (1–2) (2010) 25–32. org/10.1159/000231980.
[159] D.W. Porter, B.D. Kerr, P.R. Flatt, C. Holscher, V.A. Gault, Four weeks [186] K. Srinivasan, B. Viswanad, L. Asrat, C.L. Kaul, P. Ramarao, Combination of high-
administration of liraglutide improves memory and learning as well as glycaemic fat diet-fed and low-dose streptozotocin-treated rat: a model for type 2 diabetes
control in mice with high fat dietary-induced obesity and insulin resistance, and pharmacological screening, Pharmacol. Res. 52 (4) (2005) 313–320.
Diabetes Obes. Metab. 12 (10) (2010) 891–899. [187] J.M. Starr, J. Wardlaw, K. Ferguson, A. MacLullich, I.J. Deary, I. Marshall,
[160] S. Prasad, R.K. Sajja, P. Naik, L. Cucullo, Diabetes mellitus and blood-brain barrier Increased blood-brain barrier permeability in type II diabetes demonstrated by
dysfunction: an overview, J. Pharmacovigil. 2 (2) (2014) 125. Jun. gadolinium magnetic resonance imaging, J. Neurol. Neurosurg. Psychiatry. 74 (1)
[161] L. Puglielli, R.E. Tanzi, D.M. Kovacs, Alzheimer’s disease: the cholesterol (2003) 70–76.
connection, Nat. Neurosci. 6 (4) (2003) 345–351. [188] M. Straczkowski, I. Kowalska, M. Baranowski, A. Nikolajuk, E. Otziomek,
[162] J.J. Ramos-Rodriguez, M. Jimenez-Palomares, M.I. Murillo-Carretero, C. Infante- P. Zabielski, et al., Increased skeletal muscle ceramide level in men at risk of
Garcia, E. Berrocoso, F. Hernandez-Pacho, et al., Central vascular disease and developing type 2 diabetes, Diabetologia. 50 (2007) 2366–2373, https://doi.org/
exacerbated pathology in a mixed model of type 2 diabetes and Alzheimer’s 10.1007/s00125-007-0781-2.
disease, PsychoneuroendocrinologyPsychoneuroendocrinology 62 (2015) 69–79. [189] A.M. Stranahan, S. Hao, A. Dey, X. Yu, B. Baban, Blood-brain barrier breakdown
[163] J.J. Ramos-Rodriguez, S. Molina-Gil, O. Ortiz-Barajas, M. Jimenez-Palomares, promotes macrophage infiltration and cognitive impairment in leptin receptor-
G. Perdomo, I. Cozar-Castellano, A.M. Lechuga-Sancho, M. Garcia-Alloza, Central deficient mice, J. Cereb. Blood Flow Metab. 36 (12) (2016) 2108–2121. Dec.
proliferation and neurogenesis is impaired in type 2 diabetes and prediabetes [190] A.M. Stranahan, R.G. Cutler, C. Button, R. Telljohann, M.P. Mattson, Diet induced
animal models, PLoS One. 9 (2) (2014) e89229. Feb 20. elevations in serum cholesterol are associated with alterations in hippocampa
[164] P. Reddy, D. Lent-Schochet, N. Ramakrishnan, M. McLaughlin, I. Jialal, Metabolic lipid metabolism and increased oxidative stress, J. Neurochem. 118 (4) (2011)
syndrome is an inflammatory disorder: a conspiracy between adipose tissue and 611–615.
phagocytes, Clin. Chim. Acta. 496 (2019) 35–44, https://doi.org/10.1016/j. [191] A.M. Stranahan, E.D. Norman, K. Lee, R.G. Cutler, R.S. Telljohann, J.M. Egan, et
cca.2019.06.019. Sep. al., Diet-induced insulin resistance impairs hippocampal synaptic plasticity and
[165] M.J. Reed, K. Meszaros, L.J. Entes, et al., A new rat model of type 2 diabetes: the cognition in middle-aged rats, Hippocampus. 18 (11) (2008) 1085–1088.
fat-fed, streptozotocin-treated rat, Metabolism. 49 (11) (2000) 1390–1394. [192] B. Su, X. Wang, A. Nunomura, et al., Oxidative stress signaling in Alzheimer’s
[166] M. Rodriguez-Perdigon, M. Solas, M.J. Moreno-Aliaga, M.J. Ramirez, Lipoic acid disease, Curr. Alzheimer Res. 5 (6) (2008) 525–532.
improves neuronal insulin signalling and rescues cognitive function regulating

13
S. Sharma Physiology & Behavior 240 (2021) 113528

[193] L. Sun, X. Diao, X. Gang, et al., Risk factors for cognitive impairment in patients [219] G. Winocur, C.E. Greenwood, The effects of high fat diets and environmental
with type 2 diabetes, J. Diabetes Res. 2020 (2020), 4591938. influences on cognitive performance in rats, Behav. Brain Res. 101 (1999)
[194] R.S. Surwit, C.M. Kuhn, C. Cochrane, J.A. McCubbin, M.N. Feinglos, Diet-induced 153–161.
type II diabetes in C57BL/6 J mice, Diabetes. 37 (1988) 1163–1167. [220] G. Winocur, C.E. Greenwood, Studies of the effects of high fat diets on cognitive
[195] M. Suwa, H. Kishimoto, Y. Nofuji, H. Nakano, H. Sasaki, Z. Radak, S. Kumagai, function in a rat model, Neurobiol. Aging. 26 (2005) 46–49. Suppl 1.
Serum brain-derived neurotrophic factor level is increased and associated with [221] G. Winocur, C.E. Greenwood, G.G. Piroli, C.A. Grillo, L.R. Reznikov, L.P. Reagan,
obesity in newly diagnosed female patients with type 2 diabetes mellitus, B.S. McEwen, Memory impairment in obese Zucker rats: an investigation of
Metabolism. 55 (7) (2006) 852–857. Jul. cognitive function in an animal model of insulin resistance and obesity, Behav.
[196] W. Swardfager, K. Lanctot, L. Rothenburg, A. Wong, J. Cappell, N. Herrmann, Neurosci. 119 (5) (2005) 1389–1395.
A meta-analysis of cytokines in Alzheimer’s disease, Biol. Psychiatry. 68 (2010) [222] H.M. Wisniewski, P.B. Kozlowski, Evidence for blood-brain barrier changes in
930–941. senile dementia of the Alzheimer type (SDAT), Ann. N Y Acad Sci. 396 (1982)
[197] R. Takechi, V. Lam, E. Brook, C. Giles, N. Fimognari, A. Mooranian, et al., 119–129.
Blood–brain barrier dysfunction precedes cognitive decline and [223] A. Wu, Z. Ying, F. Gomez-Pinilla, Omega-3 fatty acids supplementation restores
neurodegeneration in diabetic insulin resistant mouse model: an implication for mechanisms that maintain brain homeostasis in traumatic brain injury,
causal link, Front. Aging Neurosci. 9 (2017) 399. J. Neurotrauma. 24 (2007) 1587–1595.
[198] B.L. Tan, M.E. Norhaizan, Effect of high-fat diets on oxidative stress, cellular [224] A. Wu, Z. Ying, F. Gomez-Pinilla, The interplay between oxidative stress and
inflammatory response and cognitive function, Nutrients. 11 (11) (2019) 2579. brain-derived neurotrophic factor modulates the outcome of a saturated fat diet
[199] S. Tangvarasittichai, Oxidative stress, insulin resistance, dyslipidemia and type 2 on synaptic plasticity and cognition, Eur. J. Neurosci. 19 (7) (2004) 1699–1707.
diabetes mellitus, World J Diabetes. 6 (3) (2015) 456–480. [225] K. Wu, J.L. Xu, P.C. Suen, E. Levine, Y.Y. Huang, H.T. Mount, S.Y. Lin, I.B. Black,
[200] M.M. Teixeira, V.M.A. Passos, S.M. Barreto, et al., Association between diabetes Functional trkB neurotrophin receptors are intrinsic components of the adult
and cognitive function at baseline in the brazilian longitudinal study of adult brain postsynaptic density, Brain Res. Mol. Brain Res. 43 (1–2) (1996) 286–290.
health (ELSA- Brasil), Sci. Rep. 10 (1) (2020) 1596. Dec 31.
[201] P. Thériault, A. El Ali, S. Rivest, High fat diet exacerbates Alzheimer’s disease- [226] S.F. Xia, Z.X. Xie, Y. Qiao, et al., Differential effects of quercetin on hippocampus-
related pathology in APPswe/PS1 mice, Oncotarget. 7 (42) (2016) 67808–67827. dependent learning and memory in mice fed with different diets related with
[202] L. Thirumangalakudi, A. Prakasam, R. Zhang, et al., High cholesterol-induced oxidative stress, Physiol. Behav. 138 (2015) 325–331.
neuroinflammation and amyloid precursor protein processing correlate with loss [227] W. Xia, H. Rao, A.M. Spaeth, et al., Blood pressure is associated with cerebral
of working memory in mice, J. Neurochem. 106 (1) (2008) 475–485. blood flow alterations in patients with T2DM as revealed by perfusion functional
[203] M. Ujiie, D.L. Dickstein, D.A. Carlow, W.A. Jefferies, Blood-brain barrier MRI, Medicine (Baltimore). 94 (48) (2015) e2231.
permeability precedes senile plaque formation in an Alzheimer disease model, [228] S.F. Xia, Z.X. Xie, Y. Qiao, L.R. Li, X.R. Cheng, X.M. Duan, et al., Salvianolic acid B
Microcirculation. 10 (6) (2003) 463–470. counteracts cognitive decline triggered by oxidative stress in mice fed with high-
[204] R.M. Uranga, A.J. Bruce-Keller, C.D. Morrison, S.O. Fernandez-Kim, P. fat diets, J. Funct. Foods 11 (2014) 278–292.
J. Ebenezer, L. Zhang, K. Dasuri, J.N. Keller, Intersection between metabolic [229] J. Xu, H. Gao, L. Zhang, et al., Melatonin alleviates cognition impairment by
dysfunction, high fat diet consumption, and brain aging, J. Neurochem. 114 (2) antagonizing brain insulin resistance in aged rats fed a high-fat diet, J. Pineal.
(2010) 344–361. Jul. Res. 67 (2) (2019) e12584.
[205] L.P. van der Heide, A. Kamal, A. Artola, W.H. Gispen, G.M. Ramakers, Insulin [230] X. Yang, M. Zheng, S. Hao, H. Shi, D. Lin, X. Chen, A. Becvarovski, W. Pan,
modulates hippocampal activity-dependent synaptic plasticity in a N-methyl-D- P. Zhang, M. Hu, X.-.F. Huang, K. Zheng, Y. Yu, Curdlan prevents the cognitive
aspartate receptor and phosphatidyl-inositol-3-kinase-dependent manner, deficits induced by a high-fat diet in mice via the gut-brain axis. front, Neurosci.
J. Neurochem. 94 (4) (2005) 1158–1166. 14 (2020) 384, https://doi.org/10.3389/fnins.2020.00384.
[206] M. Vandal, P.J. White, C. Tremblay, et al., Insulin reverses the high-fat diet- [231] S. Yehuda, S. Rabinovitz, D.I. Motofsky, Modulation of learning and neuronal
induced increase in brain Aβ and improves memory in an animal model of membrane composition in the rat by essential fatty acid preparation: time-course
Alzheimer disease, Diabetes. 63 (12) (2014) 4291–4301. analysis, Neurochem. Res. 23 (1998) 627–634.
[207] G. Verdile, K.N. Keane, V.F. Cruzat, S. Medic, M. Sabale, J. Rowles, et al., [232] D.Y. Yoo, W. Kim, S.M. Nam, et al., Reduced cell proliferation and neuroblast
Inflammation and oxidative stress:the molecular connectivity between insulin differentiation in the dentate gyrus of high fat diet-fed mice are ameliorated by
resistance, obesity, and Alzheimer’s disease, Mediators Inflamm. 2015 (2015), metformin and glimepiride treatment, Neurochem. Res. 36 (12) (2011)
105828. 2401–2408.
[208] B. Viswanad, K. Srinivasan, C.L. Kaul, P. Ramarao, Effect of tempol on altered [233] D.Y. Yoo, W. Kim, K.Y. Yoo, et al., Effects of pyridoxine on a high-fat diet-induced
angiotensin II and acetylcholine-mediated vascular responses in thoracic aorta reduction of cell proliferation and neuroblast differentiation depend on cyclic
isolated from rats with insulin resistance, Pharmacol. Res. 53 (2006) 209. adenosine monophosphate response element binding protein in the mouse
[209] M. Wakisaka, S. Nagamachi, K. Inoue, Y. Morotomi, K. Nunoi, M. Fujishima, dentate gyrus, J. Neurosci. Res. 90 (8) (2012) 1615–1625.
Reduced regional cerebral blood flow in aged noninsulin-dependent diabetic [234] G. Yoon, K.A. Cho, J. Song, Y.K. Kim, Transcriptomic analysis of high fat diet fed
patients with no history of cerebrovascular disease: evaluation by N-isopropyl- mouse brain cortex, Front. Genet. 10 (2019) 83. Feb 19.
123I-p-iodoamphetamine with single-photon emission computed tomography, [235] C. Yu, Y. Chen, G.W. Cline, D. Zhang, H. Zong, Y. Wang, et al., Mechanism by
J. Diabet. Complications. 4 (4) (1990) 170–174. which fatty acids inhibit activation of insulin receptor substrate-1 (IRS-1)-
[210] J.M. Walker, S. Dixit, A.C. Saulsberry, J.M. May, F.E. Harrison, Reversal of high associated phosphatidylinositol 3-kinase activity in muscle, J. Biol. Chem. 277
fat diet-induced obesity improves glucose tolerance, inflammatory response, (2002) 50230–50236.
β-amyloid accumulation and cognitive decline in the APP/PSEN1 mouse model of [236] M.Q. Zaman, V. Leray, J. Le Blocíh, C. Thorin, K. Ouguerram, P. Nguyen, Lipid
Alzheimer’s disease, Neurobiol. Dis. 100 (2017) 87–98. profile and insulin sensitivity in rats fed with high-fat or high-fructose diets, Br. J.
[211] J.M. Walker, F.E. Harrison, Shared neuropathological characteristics of obesity, Nutr. 106 (2011) S206–S210.
type 2 diabetes and Alzheimer’s disease: impacts on cognitive decline, Nutrients. [237] F. Zhang, C. Ye, G. Li, W. Ding, W. Zhou, H. Zhu, G. Chen, T. Luo, M. Guang,
7 (9) (2015) 7332–7357. Y. Liu, D. Zhang, S. Zheng, J. Yang, Y. Gu, X. Xie, M. Luo, The rat model of type 2
[212] H. Wang, E.J. Golob, M.Y. Su, Vascular volume and blood-brain barrier diabetic mellitus and its glycometabolism characters, Exp. Anim. 52 (5) (2003)
permeability measured by dynamic contrast enhanced MRI in hippocampus and 401–407. Oct.
cerebellum of patients with MCI and normal controls, J. Magn. Reson. Imaging. [238] Z. Zhang, S. Wang, S. Zhou, et al., Sulforaphane prevents the development of
24 (3) (2006) 695–700. cardiomyopathy in type 2 diabetic mice probably by reversing oxidative stress-
[213] J. Wang, D. Freire, L. Knable, W. Zhao, B. Gong, P. Mazzola, et al., Childhood and induced inhibition of LKB1/AMPK pathway, J. Mol. Cell Cardiol. 77 (2014)
adolescent obesity and long-term cognitive consequences during aging, J. Comp. 42–52.
Neurol. 523 (5) (2015) 757–768. [239] P. Zhang, Y. Yu, Y. Qin, Y. Zhou, R. Tang, Q. Wang, et al., Alterations to the
[214] J. Wang, B. Gong, W. Zhao, C. Tang, M. Varghese, T. Nguyen, et al., Epigenetic microbiota-colon-brain axis in high-fat-diet-induced obese mice compared to diet-
mechanisms linking diabetes and synaptic impairments, Diabetes. 63 (2) (2014) resistant mice, J. Nutr. Biochem. 65 (2019) 54–65, https://doi.org/10.1016/j.
645–654. jnutbio.2018.08.016.
[215] Q. Wang, J. Yuan, Z. Yu, L. Lin, Y. Jiang, Z. Cao, P. Zhuang, M.J. Whalen, B. Song, [240] W.Q. Zhao, D.L. Alkon, Role of insulin and insulin receptor in learning and
X.J. Wang, X. Li, E.H. Lo, Y. Xu, X. Wang, FGF21 attenuates high-fat diet-induced memory, Mol. Cell Endocrinol. 177 (2001) 125–134.
cognitive impairment via metabolic regulation and anti-inflammation of obese [241] W.Q. Zhao, H. Chen, M.J. Quon, D.L. Alkon, Insulin and the insulin receptor in
mice, Mol. Neurobiol. 55 (6) (2018) 4702–4717. Jun. experimental models of learning and memory, Eur. J. Pharmacol. 490 (2004)
[216] Z. Wang, Q. Ge, Y. Wu, J. Zhang, Q. Gu, J. Han, Impairment of long-term memory 71–81.
by a short-term high-fat diet via hippocampal oxidative stress and alterations in [242] X. Zhou, L. He, S. Zuo, et al., Serine prevented high-fat diet-induced oxidative
synaptic plasticity, Neurosci. 424 (2020) 24–33. stress by activating AMPK and epigenetically modulating the expression of
[217] S. Wang, X.F. Huang, P. Zhang, K.A. Newell, H. Wang, K. Zheng, et al., Dietary glutathione synthesis-related genes, Biochim. Biophys. Acta Mol. Basis Dis. 1864
teasaponin ameliorates alteration of gut microbiota and cognitive decline in diet- (2) (2018) 488–498.
induced obese mice, Sci. Rep. 7 (2017) 12203. [243] K.L. Zuloaga, L.A. Johnson, N.E. Roese, et al., High fat diet-induced diabetes in
[218] R.A. Whitmer, S. Sidney, J. Selby, S.C. Johnston, K. Yaffe, Midlife cardiovascular mice exacerbates cognitive deficit due to chronic hypoperfusion, J. Cereb. Blood
risk factors and risk of dementia in late life, Neurol. 64 (2) (2005) 277–281. Jan Flow Metab. 36 (7) (2016) 1257–1270.
25. [244] Weiying Dai, Wenna Duan, Freddy Alfaro, et al., The Resting Perfusion Pattern
Associates with Functional Decline in Type 2 Diabetes, Neurobiology of aging 60
(2017) 192–202, https://doi.org/10.1016/j.neurobiolaging.2017.09.004.

14

You might also like