ArticlePDF AvailableLiterature Review

Abstract

Purpose of review: Recent findings suggest that high-sugar diets can lead to cognitive impairment predisposing to neurodegenerative disorders such as Alzheimer's disease. This article discusses metabolic derangements induced by high-fructose/sucrose diets and presents evidence for the involvement of insulin resistance in sporadic Alzheimer's disease pathogenesis. Recent findings: There has been much concern regarding the role of dietary sugars (fructose/sucrose) in the development of type 2 diabetes (T2D). Accumulating evidence has also demonstrated a connection between T2D and Alzheimer's disease. The risk for developing T2D and Alzheimer's disease increases exponentially with age and having T2D increases the risk of developing Alzheimer's disease. Summary: The incidence of T2D increased dramatically over the last decades mainly due to Western lifestyle factors such as lack of exercise and high calorie diets. In fact, high-sugar diets are thought to promote weight gain and insulin resistance predisposing to T2D. To aggravate this scenario, it has been consistently shown that T2D is a risk factor for Alzheimer's disease and both disorders share similar demographic profiles, risk factors, and clinical and biochemical features (e.g. insulin resistance). Therefore, dietary changes can significantly reduce the risk of T2D and Alzheimer's disease and thereby increase the quality of life and improve longevity.
High-sugar diets, type 2 diabetes and Alzheimer’s disease
Paula I. Moreira
Laboratory of Physiology, Faculty of Medicine, University of Coimbra, 3000-354
Coimbra Portugal & Center for Neuroscience and Cell Biology, University of Coimbra,
3004-517 Coimbra, Portugal
Correspondence to: Paula I. Moreira, Laboratory of Physiology - Faculty of Medicine,
University of Coimbra, 3000-354 Coimbra, Portugal
Tel: 351-239480012
Fax: 351-239480034
Email: venta@ci.uc.pt / pimoreira@fmed.uc.pt
2
Purpose of review: Recent findings suggest that high-sugar diets can lead to cognitive
impairment predisposing to neurodegenerative disorders such as Alzheimer’s disease
(AD). This paper discusses metabolic derangements induced by high-fructose/sucrose
diets and presents evidence for the involvement of insulin resistance in sporadic AD
pathogenesis.
Recent findings: There has been much concern regarding the role of dietary sugars
(fructose/sucrose) in the development of type 2 diabetes (T2D). Accumulating evidence
has also demonstrated a connection between T2D and AD. The risk for developing
T2D and AD increases exponentially with age and having T2D increases the risk of
developing AD.
Summary: The incidence of T2D increased dramatically over the last decades mainly
due to western lifestyle factors such as lack of exercise and high calorie diets. In fact,
high-sugar diets are thought to promote weight gain and insulin resistance
predisposing to T2D. To aggravate this scenario, it has been consistently shown that
T2D is a risk factor for AD and both disorders share similar demographic profiles, risk
factors, and clinical and biochemical features (e.g. insulin resistance). Therefore,
dietary changes can significantly reduce the risk of T2D and AD and thereby increase
the quality of life and improve longevity.
Keywords: Alzheimer’s disease, high-sugar diets, insulin resistance, type 2 diabetes
3
Introduction
Type 2 diabetes (T2D) accounts for about 90% of all cases of diabetes mellitus
and is characterized by a reduction in the ability of insulin to stimulate glucose
utilization (insulin resistance) and inadequate pancreatic -cell insulin secretion in
response to hyperglycemia [1]. The prevalence and incidence of T2D augmented
dramatically over the last decades along with increased population obesity owing to
western lifestyle factors such as lack of exercise and high calorie diets. In fact, the
increase in sugar consumption has paralleled the increasing prevalence of obesity, and
high-sugar diets are thought to promote weight gain and insulin resistance
predisposing to T2D. As diabetes progresses without appropriate treatment, many
complications occur, which leads to increased risk of mortality. Among other
complications, T2D is related with a greater rate of cognitive deficits in comparison with
the general population. Studies of cerebral structure demonstrated a pronounced
cortical, subcortical, and hippocampal atrophy in T2D patients. Accumulating evidence
also shows that T2D is a risk factor for dementia, particularly vascular dementia and
Alzheimer’s disease (AD). Interestingly, both T2D and AD share similar demographic
profiles, risk factors and, clinical and biochemical features [2].
AD, the most common cause of dementia among older people, typically begins
with a subtle decline in memory and progresses to global deterioration in cognitive and
adaptive functioning. The neuropathological features associated with the disease
include the presence of extracellular senile plaques (SPs), intracellular neurofibrillary
tangles (NFTs) and the loss of basal forebrain cholinergic neurons that innervate the
hippocampus and the cortex. SPs are formed mostly from the deposition of amyloid
(A), a peptide generated through the proteolytic cleavage of amyloid precursor protein
(APP) by the - and -secretases. NFTs are mainly composed of neurofilaments and
hyperphosphorylated tau protein. A history of stroke can increase the prevalence of the
disease among elderly patients. The risk is highest when stroke is concomitant with
4
atherosclerotic vascular risk. Hypoxia is a direct consequence of hypoperfusion, a
common vascular component among the AD risk factors, and may play an important
role in the pathogenesis of this disorder. The great majority of all AD cases are
sporadic in origin, with old age and T2D considered main risk factors [3].
This review focus on the effects of high-sugar diets in the development of T2D
(and obesity), and how T2D, specifically insulin resistance, increases the risk of AD.
Type 2 diabetes: the influence of high-sugar diets
Unhealthy lifestyle, characterized by caloric overconsumption and physical
inactivity, is a risk factor for insulin resistance and T2D, often associated with obesity.
Foods containing simple sugars are prototypes of high-glycemic index foods that are
consumed in significant amounts worldwide. In fact, fructose found in sucrose and
high-fructose corn syrup has been the subject of intense debate. Products containing
fructose are preferred by consumers and cooks over those containing only glucose,
due to the intrinsically greater sweetness of fructose and its ability to improve the
texture and appearance of baked goods. As a result, fructose and high-fructose corn
syrup are present in several beverages (e.g. soda, sport and energy drinks) and food
(e.g. snacks, sauces, processed meats) that are highly consumed in developed and
developing countries.
An elevated consumption of sugar-enriched foods promotes insulin resistance
for two main reasons: induction of weight gain due to their high energy content and lack
of satiating effect, and higher postprandial blood glucose and insulin levels [4]. In fact,
high fructose intake and subsequent metabolism increase the synthesis of triglycerides,
which are assembled into very low-density lipoproteins (VLDL), and glucose that is
released into circulation culminating in increased plasma glucose. This increase
stimulates the secretion of insulin by the pancreatic cells. Insulin is an anabolic
hormone and, in an organism in positive energy balance, determines an increase in the
deposition of fat in adipose tissue and other organs.
5
Fructose-enriched diets promote an increase in plasma triglycerides mainly by
the stimulation of hepatic de novo lipogenesis, and decrease in VLDL-triglyceride
clearance [5]. The increase in plasma triglyceride induced by fructose is significantly
attenuated in women supporting the idea that estrogen/progesterone may exert a
protective effect [6,7]. It was reported that 1 week of a high-fructose, hypercaloric diet
led to ectopic fat deposition in liver and skeletal muscle [8], and a high-fructose diet
maintained during 10 weeks impaired glucose homeostasis in overweight individuals
[9]. A recent randomized controlled trial [10●●] showed that even moderate amounts of
fructose and sucrose during a period of 3 weeks significantly alter hepatic insulin
sensitivity and lipid metabolism in healthy normal-weight male volunteers, compared
with similar amounts of glucose. High fructose intake has been shown to increase
energy intake via an increase in orexigenic signaling peptides levels in the
hypothalamus [11]. Recently, Page and collaborators [12●●], using arterial spin labeling,
a magnetic resonance imaging technique, quantified regional cerebral blood flow as a
surrogate for brain activity in 20 healthy, young, normal-weighed subjects before and
after drinking a 75g solution of pure glucose or fructose. The authors observed that the
hypothalamic brain signal generated after fructose ingestion was statistically different
from that generated by glucose ingestion. This difference was accompanied by a
sensation of satiety and fullness after glucose, but not fructose, ingestion [12●●]. A
previous study based on blood oxygenation level dependent functional brain magnetic
resonance imaging (BOLD fMRI) also showed that in normal weight humans, cortical
responses to infused glucose are opposite to those of fructose [13]. These findings
support the idea that glucose and fructose modulate different neurobiological
pathways, and, in the case of fructose, it stimulates pathways involved in food intake.
In rodents there is overwhelming evidence that high-sugar diets decrease both
liver and muscle insulin sensitivity predisposing to obesity, diabetes, and dyslipidemia
[14]. Similarly, young adult male baboons exposed to a high-sugar/high-fat diet
presented increased body fat and triglyceride levels, altered adipokine levels, and
6
impaired glucose metabolism [15]. The adverse effects of fructose on glucose
metabolism are closely linked to alterations in lipid metabolism. In rats, 6 weeks of
high-fructose diet promoted an increase in plasma VLDL-triglycerides levels and
intrahepatic fat content, while intramuscular fat content increased after 3 months of
high-fructose diet [14]. Interestingly, hepatic insulin resistance was observed early after
the introduction of high-fructose diet, while the muscle insulin resistance appeared
latter [14]. These results suggest that fructose-induced insulin resistance is closely
linked to ectopic lipid deposition and tissue-specific lipotoxicity. Moreover, mice fed a
20% sucrose solution during 7 months presented hyperglycemia, glucose intolerance,
hyperinsulinemia, hypertriglyceridemia, and increased glycated hemoglobin and body
weight [16].
Altogether these studies show that high-sugar diets promote several metabolic
derangements predisposing to T2D and/or obesity.
Insulin resistance A link between type 2 diabetes and Alzheimer’s
disease
Insulin, long known as an important regulator of blood glucose levels, plays key
roles in the brain. This hormone contributes to several neurobiological processes, in
particular energy homeostasis and cognition.
Accumulating evidence supports the involvement of impaired insulin signaling in AD
etiopathogenesis: 1) reduced insulin levels and insulin receptor (IR) expression were
observed in AD brains, 2) increasing AD Braak stage was associated with
progressively reduced expression levels of insulin, insulin growth factor (IGF) 1 and 2
and respective receptors, 3) AD patients show increased fasting plasma insulin levels,
decreased cerebrospinal fluid (CSF) insulin levels, and/or decreased CSF/plasma
insulin ratio, besides increased A levels, which suggest a decrease in insulin
clearance that may provoke an elevation of plasma A levels, 4) disruption of brain
7
insulin signaling by the intracerebroventricular administration of the diabetogenic drug
streptozotocin (STZ) leads to the development of numerous behavioral, neurochemical
and structural features that resemble those found in human sporadic AD, and 5)
administration of insulin and glucose enhances memory of AD patients [for review see
1,2].
Evidence from animal studies
Animal studies show that high-fructose diets induce rapid insulin resistance,
compensatory hyperinsulinemia and memory impairment. Rats exposed to a high-
fat/high-sucrose diet presented impaired memory due to an alteration in the signaling
pathways involved in hippocampal neuronal plasticity [17]. A subsequent study
demonstrated that sucrose- and fat-enriched diets promoted a weight gain and
deposition of visceral fat in rats, but only animals fed with the sucrose diet displayed
deficits in long-term spatial memory [18]. It was also shown that rats fed diets
supplemented with high-fructose corn syrup were more insulin resistant and cognitively
impaired, due to a reduction in dendritic spine density and hippocampal levels of brain
derived neurotrophic factor [19]. Hamsters fed with 60% fructose presented a decrease
in neuronal insulin signaling in the cerebral cortex and hippocampus, and a subsequent
impairment of synaptic function [20]. Overall, this evidence suggests that high-fructose
diets-induced insulin resistance is linked to cognitive decline and predisposes to
neurodegeneration. In fact, the intake of sucrose-sweetened water was shown to in-
duce insulin resistance and exacerbate memory deficits and amyloidosis in a mouse
model of AD [21]. In the same line, a previous study from our laboratory showed that
3xTg-AD and sucrose-induced diabetic mice presented a similar profile of behavioral
and cognitive alterations, vascular anomalies, oxidative imbalance and mitochondrial
abnormalities [16●●,22●●]. Interestingly, sucrose-induced diabetic mice, showed a
significant increase in the levels of A in brain cortex and hippocampus [16●●,22●●],
supporting the idea that metabolic alterations induced by sucrose intake increase the
8
risk of AD. AD mice exposed to a dietary regimen that induced insulin resistance and a
hyperinsulinemic state induced a significant accumulation of A and reduction in the
levels of insulin-degrading enzyme (IDE; ubiquitously expressed zinc-metalloprotease
involved in the clearance of insulin and A) [23]. Conversely, treatment with
rosiglitazone, an insulin sensitizer, was shown to reduce A levels and improve
learning and memory deficits [24] suggesting that insulin sensitizers might increase
insulin signaling and decrease the levels of insulin available to compete with A for
degradation by IDE.
The regulation of tau protein phosphorylation is another potential mechanism by
which insulin links T2D to AD. Both chronic peripheral hyperinsulinemia and central
insulin resistance can modulate tau protein phosphorylation. In IRS2-disrupted mice, a
model of T2D, an accumulation of NFTs was detected in the hippocampus [25] and the
neuronal/brain-specific deletion of IR in mice (NIRKO mice) led to tau protein
hyperphosphorylation [26]. Meanwhile, the pattern of tau protein phosphorylation
differed between NIRKO and IRS-2 knockout mice, suggesting that not only insulin
resistance, but also hyperinsulinemia could mediate site-specific, differential tau protein
phosphorylation. In fact, Freude and collaborators [27] found that peripherally injected
insulin directly targets the brain and causes rapid cerebral IR signal transduction and
site-specific tau protein phosphorylation. Of note, in insulin-stimulated NIRKO mice,
cerebral IR signaling and tau protein phosphorylation were completely abolished [27].
These results indicate that tau protein modification caused by insulin dysfunction and
hyperglycemia may contribute to the increased incidence of AD in diabetic subjects.
Loss of insulin-mediated activation of phosphatidylinositol 3-kinase (PI3-K) and
subsequent reduction of phosphorylation of Akt and glycogen synthase kinase (GSK)-
3 are reported to result in a substantial increase in the levels of phosphorylated tau
protein in the brains of neuron specific IR knockout mice [26]. Insulin also regulated
soluble APP release via PI3-K-mediated pathway, suggesting that PI3-K involvement in
9
APP metabolism may occur at vesicular trafficking level [28]. It was also shown that
GSK-3 inhibition attenuated APP processing and inhibited hyperphosphorylated tau
protein-associated neurodegeneration [28].
Evidence from human studies
Evidence shows that individuals with T2D have nearly a twofold higher risk of
AD, independent of the risk for vascular dementia, than non-diabetic individuals [29].
Prospective and cross-sectional analyses suggest that diabetes may accelerate the
onset of AD, rather than increasing the long-term risk [30]. In fact, cross-sectional
studies report more frequent structural brain lesions, greater cortical atrophy and more
white matter hyperintensities in people with diabetes compared with non-diabetic
controls [31-33]. These studies have found relationships between these structural
abnormalities and underlying hypertension and vascular disease [31], but also
diabetes-specific parameters such as diabetes duration and fasting glucose levels [33].
These findings support the idea that diabetic subjects are more prone to develop
dementia, particularly AD.
The impairment of insulin signaling was demonstrated by postmortem studies
that revealed that mitogen-activated protein kinase (MAPK) activation occurs at very
early stages (Braak stages IV-V) [34]. A correlation between Akt activity/protein levels
and Braak staging has also been documented in human AD postmortem analyses,
which indicates a time-dependent and insulin-stimulated PI3-K signaling-dependent
pattern of changes [35].
Altogether, animal and human studies clearly show that central insulin resistance
contributes to the pathogenesis of AD.
10
Conclusion
There is no doubt that high-sugar diets provoke drastic metabolic derangements
predisposing to obesity, dyslipidemia and T2D; conditions that have reached epidemic
proportions. T2D increases the incidence of life-threatening complications including an
enhanced risk of cognitive decline and dementia. In fact, accumulating evidence shows
that T2D is a main risk factor for AD, with insulin signaling abnormalities playing a key
role. Similarly to T2D, also AD is growing exponentially in our aging society. Because
many metabolic disorders arise from unhealthy diets and sedentary lifestyles, we have
in our hands (or mouth and feet!) the opportunity to reverse this dark scenario.
Key points
1. High-sugar (fructose/sucrose) diets lead to metabolic derangements predisposing to
T2D, normally associated with obesity.
2. T2D is associated with an increased risk of cognitive impairment and dementia,
particularly AD.
3. Insulin resistance is a common feature between T2D and sporadic AD.
Acknowledgements
The authors’ work is supported by the Fundação para a Ciência e a Tecnologia (FCT)
and Fundo Europeu de Desenvolvimento Regional (FEDER) (PTDC/SAU-
NEU/103325/2008) and Quadro de Referência Estratégico Nacional (QREN DO-IT
DIAMARKER Project).
11
References
1 Correia SC, Santos RX, Carvalho C, et al. Insulin signaling, glucose metabolism and
mitochondria: major players in Alzheimer's disease and diabetes interrelation.
Brain Res 2012; 1441:64-78.
This paper reviews epidemiological and clinical studies showing that impaired insulin signaling is deeply involved in AD
pathogenesis.
2 Moreira PI. Alzheimer's disease and diabetes: an integrative view of the role of
mitochondria, oxidative stress, and insulin. J Alzheimers Dis 2012; 30:S199-
215.
This paper reviews epidemiological and clinical studies showing that impaired insulin signaling is deeply involved in AD
pathogenesis.
3 Correia SC, Santos RX, Cardoso S, et al. Alzheimer disease as a vascular disorder:
where do mitochondria fit? Exp Gerontol 2012; 47:878-886.
4 Atkinson FS, Foster-Powell K, Brand-Miller JC. International tables of glycemic index
and glycemic load values. Diabetes Care 2008; 31:22812283.
5 Chong MF, Fielding BA, Frayn KN. Mechanisms for the acute effect of fructose on
postprandial lipemia. Am J Clin Nutr 2007; 85:15111520.
6 Couchepin C, Le KA, Bortolotti M, et al. Markedly blunted metabolic effects of
fructose in healthy young female subjects compared with male subjects.
Diabetes Care 2008; 31:12541256.
7 Stanhope KL, Griffen SC, Bair BR, et al. Twenty-four-hour endocrine and metabolic
profiles following consumption of high-fructose corn syrup-, sucrose-, fructose-,
and glucose-sweetened beverages with meals. Am J Clin Nutr 2008; 87:1194
203.
12
8 Le KA, Ith M, Kreis R, et al. Fructose overconsumption causes dyslipidemia and
ectopic lipid deposition in healthy subjects with and without a family history of
type 2 diabetes. Am J Clin Nutr 2009; 89:17601765.
9 Stanhope KL, Schwarz JM, Keim NL, et al. Consuming fructose-sweetened, not
glucose-sweetened, beverages increases visceral adiposity and lipids and
decreases insulin sensitivity in overweight/obese humans. J Clin Invest 2009;
119:13221334.
●●10 Aeberli I, Hochuli M, Gerber PA, et al. Moderate amounts of fructose consumption
impair insulin sensitivity in healthy young men: a randomized controlled trial.
Diabetes Care 2013; 36:150-156.
This is a double-blind, randomized, cross-over trial that demonstrates that moderate amounts of fructose and sucrose
significantly alter hepatic insulin sensitivity and lipid metabolism compared with similar amounts of glucose.
11 Cha SH, Wolfgang M, Tokutake Y, et al. Differential effects of central fructose and
glucose on hypothalamic malonyl-CoA and food intake. Proc Natl Acad Sci U S
A 2008; 105:1687116875.
●●12 Page KA, Chan O, Arora J, et al. Effects of fructose vs glucose on regional
cerebral blood flow in brain regions involved with appetite and reward pathways.
JAMA 2013; 309:63-70.
This study shows that fructose and glucose differently affects cerebral blood flow in brain regions involved with appetite
and reward pathways.
13 Purnell JQ, Klopfenstein BA, Stevens AA, et al. Brain functional magnetic
resonance imaging response to glucose and fructose infusions in humans.
Diabetes Obes Metab 2011; 13:229-234.
13
14 Bizeau ME, Pagliassotti MJ. Hepatic adaptations to sucrose and fructose. Metab
Clin Exp 2005; 54:1189201.
15 Higgins PB, Bastarrachea RA, Lopez-Alvarenga JC, et al. Eight week exposure to a
high sugar high fat diet results in adiposity gain and alterations in metabolic
biomarkers in baboons (Papio hamadryas sp.). Cardiovasc Diabetol. 2010;
9:71-77.
●●16 Carvalho C, Cardoso S, Correia SC, et al. Metabolic alterations induced by
sucrose intake and Alzheimer's disease promote similar brain mitochondrial
abnormalities. Diabetes 2012; 61:1234-1242.
This paper shows that in mice, metabolic alterations induced by sucrose intake predispose to AD-like
neurodegeneration.
17 Goldbart AD, Row BW, Kheirandish-Gozal L, et al. High fat/refined carbohydrate
diet enhances the susceptibility to spatial learning deficits in rats exposed to
intermittent hypoxia. Brain Res 2006; 1090:190-196.
18 Jurdak N, Lichtenstein AH, Kanarek RB. Diet-induced obesity and spatial cognition
in young male rats. Nutr Neurosci 2008; 11:4854.
19 Stranahan AM, Norman ED, Lee K, et al. Diet-induced insulin resistance impairs
hippocampal synaptic plasticity and cognition in middle-aged rats. Hippocampus
2008; 18:10851088.
20 Mielke JG, Taghibiglou C, Liu L, et al. A biochemical and functional characterization
of diet-induced brain insulin resistance. J Neurochem 2005; 93:15681578.
21 Cao D, Lu H, Lewis TL, Li L. Intake of sucrose-sweetened water induces insulin
resistance and exacerbates memory deficits and amyloidosis in a transgenic
mouse model of Alzheimer disease. J Biol Chem 2007; 282:3627536282.
14
●●22 Carvalho C, Machado N, Mota P, et al. Type 2 diabetic and Alzheimer’s disease
mice present similar behavioral, cognitive and vascular anomalies. J Alzheimers
Dis 2013 (in press)
This study shows that diabetes induced by sucrose intake promote behavioral, cognitive, vascular similar to those found
in AD.
24 Ho L, Qin W, Pompl PN, et al. Diet-induced insulin resistance promotes amyloidosis
in a transgenic mouse model of Alzheimer’s disease. FASEB J 2004; 18:902
904.
24 Pedersen WA, McMillan PJ, Kulstad JJ, et al. Rosiglitazone attenuates learning and
memory deficits in Tg2576 Alzheimer mice. Exp Neurol 2006; 199:265-273.
25 Schubert M, Brazil DP, Burks DJ, et al. Insulin receptor substrate-2 deficiency
impairs brain growth and promotes tau phosphorylation. J Neurosci 2003;
23:7084-92.
26 Schubert M, Gautam D, Surjo D, et al. Role for neuronal insulin resistance in
neurodegenerative diseases. Proc Natl Acad Sci U S A 2004; 101:3100-3105.
27 Freude S, Plum L, Schnitker J, et al. Peripheral hyperinsulinemia promotes tau
phosphorylation in vivo. Diabetes 2005; 54:3343-3348.
28 Noble W, Planel E, Zehr C, et al. Inhibition of glycogen synthase kinase-3 by lithium
correlates with reduced tauopathy and degeneration in vivo. Proc Natl Acad Sci
U S A 2005;102:6990-6995.
29 Kroner Z. The relationship between Alzheimer's disease and diabetes: Type 3
diabetes? Altern Med Rev 2009; 14:373-379.
15
30 Pasquier F, Boulogne A, Leys D, Fontaine P. Diabetes mellitus and dementia.
Diabetes Metab 2006; 32:403-414.
31 Manschot SM, Biessels GJ, de Valk H, et al. Metabolic and vascular determinants of
impaired cognitive performance and abnormalities on brain magnetic resonance
imaging in patients with type 2 diabetes. Diabetologia 2007; 50:2388-2397.
32 Manschot SM, Brands AM, van der Grond J, et al. Brain magnetic resonance
imaging correlates of impaired cognition in patients with type 2 diabetes.
Diabetes 2006;55:1106-1113.
33 Tiehuis AM, van der Graaf Y, Visseren FL, et al. Diabetes increases atrophy and
vascular lesions on brain MRI in patients with symptomatic arterial disease.
Stroke 2008;39:1600-1603.
34 Sun A, Liu M, Nguyen XV, Bing G. P38 MAP kinase is activated at early stages in
Alzheimer's disease brain. Exp Neurol 2003; 183:394-405.
35 Pei JJ, Khatoon S, An WL, et al. Role of protein kinase B in Alzheimer's
neurofibrillary pathology. Acta Neuropathol 2003; 105:381-392.
16
Legends
Figure 1. Insulin resistance and Alzheimer’s disease. Central insulin resistance
(impairment of brain insulin signaling) is closely associated with the two
neuropathological hallmarks of Alzheimer’s disease; senile plaques (SPs) and
neurofibrillary tangles (NFTs). Under hyperinsulinemic conditions, insulin competes
with amyloid protein (A) for insulin-degrading enzyme (IDE), leading to the
accumulation of A and deposition of SPs. Impaired insulin/insulin receptor (IR)
signaling culminates in loss of insulin-mediated activation of phosphoinositide 3-kinase
(PI3-K)/Akt pathway, and subsequent dephosphorylation of glycogen synthase kinase-
3 (GSK-3), which potentiates tau protein hyperphosphorylation and formation of
NFTs. Overall, the impairment of brain insulin signaling is a contributing factor in AD
pathogenesis. IRS insulin receptor substrates.
17
... contrast to healthy controls [29,30]. Moreover, the APOE4-status appears to have an influence 23 on the gut microbiome in AD patients as well as APOE transgenic mice, particularly regarding 24 butyrate producing microbes [31]. ...
... In general, the slightly decreased diversity in the fecal microbiome parallels results from other 5 studies investigating alterations in fecal microbiome in patients with AD [29,39]. Although the 6 majority of the studies in patients with AD report a decreased diversity in the fecal microbiome 7 (reviewed in [22]), the differently abundant genera widely differ. ...
... accordance to previously reported findings [40], but also conflicts with other reports [29,39,41]. 10 However, increased abundances were detected in several genera within Bacteroidetes, 11 ...
Article
Full-text available
Microbial communities in the intestinal tract are suggested to impact the ethiopathogenesis of Alzheimer’s disease (AD). The human microbiome might modulate neuroinflammatory processes and contribute to neurodegeneration in AD. However, the microbial compositions in AD patients at different stages of the disease are still not fully characterized. We used 16S rRNA analyses to investigate the oral and fecal microbiota in patients with AD and mild cognitive impairment (MCI) (n=84), at-risk individuals (APOE4 carriers) (n=17) and healthy controls (n=50) and investigated the relationship of microbial communities and disease specific markers via multivariate- and network-based approaches. We found a slightly decreased diversity in the fecal microbiota of AD patients (average Chao1 diversity for AD = 212 (SD=66); for Controls = 215 (SD=55)) and identified differences in bacterial abundances including Bacteroidetes, Ruminococcus, Sutterella, and Porphyromonadaceae. The diversity in the oral microbiota was increased in AD patients and at-risk individuals (average Chao1 diversity for AD = 174 (SD=60), for at-risk group = 195 (SD=49)). Gram-negative pro-inflammatory bacteria including Haemophilus, Neisseria, Actinobacillus and Porphyromonas were dominant oral bacteria in AD and MCI patients and the abundance correlated with the cerebrospinal fluid (CSF) biomarker. Taken together, we observed a strong shift in the fecal and the oral communities of patients with AD already prominent in prodromal and, in case of the oral microbiota, in at-risk stages. This indicates stage-dependent alterations in oral and fecal microbiota in AD which may contribute to the pathogenesis via a facilitated intestinal and systemic inflammation leading to neuroinflammation and neurodegeneration.
... These aspects raise the question of how the requirements of appropriate metabolic regulation and diabetes management, which set strict rules, can affect the quality of life of T1DM children and their parents [26]. Several studies have shown that adolescents with chronic diseases such as kidney disease [27,28], epilepsy [29], obesity [29], rheumatoid arthritis [30], and sickle cell anaemia [30] have a lower quality of life compared to their healthy peers. This finding is also consistent with the results of research examining the quality of life of adolescents with T1DM [31 -33]. ...
... These aspects raise the question of how the requirements of appropriate metabolic regulation and diabetes management, which set strict rules, can affect the quality of life of T1DM children and their parents [26]. Several studies have shown that adolescents with chronic diseases such as kidney disease [27,28], epilepsy [29], obesity [29], rheumatoid arthritis [30], and sickle cell anaemia [30] have a lower quality of life compared to their healthy peers. This finding is also consistent with the results of research examining the quality of life of adolescents with T1DM [31 -33]. ...
Chapter
Frontiers in Clinical Drug Research-Diabetes and Obesity is a book series that brings updated reviews to readers interested in advances in the development of pharmaceutical agents for the treatment of two metabolic diseases - diabetes and obesity. The scope of the series covers a range of topics including the medicinal chemistry, pharmacology, molecular biology and biochemistry of natural and synthetic drugs affecting endocrine and metabolic processes linked with diabetes and obesity. Reviews in this series also include research on specific receptor targets and pre-clinical / clinical findings on novel pharmaceutical agents. Frontiers in Clinical Drug Research - Diabetes and Obesity is a valuable resource for pharmaceutical scientists and postgraduate students seeking updated and critically important information for developing clinical trials and devising research plans in the field of diabetes and obesity research. The seventh volume of this series features 6 reviews on diabetes related topics for both medical specialists and pharmacologists. Clinical and diagnostic implications of glycated albumin in diabetes mellitus Development of novel therapeutic groups and bioactive compounds from herbs for diabetes management Aspartame as a sugar substitute Mental health, adherence, and self-management among children with diabetes Cardioprotective effects of new generation anti-diabetic and lipid-lowering agents Epidemiology, pathophysiology, and treatment of diabesity
... Numerous research have proven that dietary factors can play a quintessential role in the development of human and animal risk observation. Cognitive impairment in animals, excessive concentrations of AD and Taupathology in rodents predicting AD pathogenesis are associated with high concentrations of sucrose and fructose in Western diets (François B. Vialatte et al: 2006, Moreira PI et al: 2013, whereas trans and saturated fatty acids are associated with extended threat of AD and mild cognitive impairment (Barnard ND et al: 2014, Morris MC et al: 2014. Another rodent test (Grimm MO et al: 2012, Oksman M et al: 2006 shows that trans and saturated fatty acids have accelerated concentrations of AD. ...
Article
Full-text available
The study discusses various methods suggested, replicated or performed by scientists, researchers which are mainly based on identification of Dementia which can lead to Alzheimer's Disease (AD). The study also provides various remarks or personal findings of particular specific methods provided by the authors of this paper, where methods are ranging from hardware based models to software based models.
... Upon ingestion, sucrose is hydrolyzed into glucose and fructose, leading to an increase in blood sugar levels [11]. However, excessive consumption of sucrose can lead to obesity and an increased risk of heart disease and type II diabetes [12]. As people's living standards and health awareness improve, there is a growing interest in seeking healthier alternatives to sucrose as functional sweeteners [13]. ...
Article
Full-text available
Isomaltulose is a novel sweetener and is considered healthier than the common sugars, such as sucrose or glucose. It has been internationally recognized as a safe food product and holds vast potential in pharmaceutical and food industries. Sucrose isomerase is commonly used to produce isomaltulose from the substrate sucrose in vitro and in vivo. However, free cells/enzymes were often mixed with the product, making recycling difficult and leading to a significant increase in production costs. Immobilized cells/enzymes have the following advantages including easy separation from products, high stability, and reusability, which can significantly reduce production costs. They are more suitable than free ones for industrial production. Recently, immobilized cells/enzymes have been encapsulated using composite materials to enhance their mechanical strength and reusability and reduce leakage. This review summarizes the advancements made in immobilized cells/enzymes for isomaltulose production in terms of refining traditional approaches and innovating in materials and methods. Moreover, innovations in immobilized enzyme methods include cross-linked enzyme aggregates, nanoflowers, inclusion bodies, and directed affinity immobilization. Material innovations involve nanomaterials, graphene oxide, and so on. These innovations circumvent challenges like the utilization of toxic cross-linking agents and enzyme leakage encountered in traditional methods, thus contributing to enhanced enzyme stability.
... Experimental organisms have been widely utilized to investigate the potential molecular links between diabetes and memory impairments. One of the most accurate and reproducible ways to mimic T2DM in animal models is by the use of high sugar diets (HSDs), where the excess of sugar induces hyperglycemia and insulin resistance in baboons, rodents, and alternative organism such as Drosophila melanogaster [25][26][27][28]. D. melanogaster reared on HSD develops several hallmarks of T2DM, including increased levels of glucose/trehalose, obesity-like phenotypes, insulin resistance and changes in insulin-like peptide mRNA levels, renal tubule impairments, and even impaired immune responses [28][29][30][31][32][33]. ...
Article
Full-text available
Type 2 diabetes mellitus (T2DM) has been shown to affect a series of cognitive processes including memory, increasing the risk for dementia, particularly Alzheimer’s disease (AD). Although increasing evidence has supported that both diseases share common features, the pathophysiological mechanisms connecting these two disorders remain to be fully elucidated. Herein, we used Drosophila melanogaster fed on a high-sugar diet (HSD) to mimic T2DM, and investigate its effects on memory as well as identify potential molecular players associated with the memory deficits induced by HSD. Flies hatched from and reared on HSD for 7 days had a substantial decrease in short-term memory (STM). The screening for memory-related genes using transcriptome data revealed that HSD altered the expression of 33% of memory genes in relation to the control. Among the differentially expressed genes (DEGs) with a fold change (FC) higher than two, we found five genes, related to synapse and memory trace formation, that could be considered strong candidates to underlie the STM deficits in HSD flies: Abl tyrosine kinase (Abl), bruchpilot (Brp), minibrain (Mnb), shaker (Sh), and gilgamesh (Gish). We also analyzed genes from the dopamine system, one of the most relevant signaling pathways for olfactory memory. Interestingly, the flies fed on HSD presented a decreased expression of the Tyrosine hydroxylase (Ple) and Dopa decarboxylase (Ddc) genes, signals of a possible dopamine deficiency. In this work, we present promising biomarkers to investigate molecular networks shared between T2DM and AD. Graphical Abstract
... For example, blood sugar levels are controlled by two major feedback loops involving insulin and glucagon. Since glucose control by insulin is based by an activation of beta cells via glucose (see Supporting Material in Ref. [11]), constantly high glucose levels ("glucose overload") [64,65], for example, may lead to a slower resetting of the insulin-based control loop in comparison with more rapid anticipated adaptations at lower glucose levels. Such a slowing-down response may be one of the causes that could participate in the mechanisms leading to insulin resistance and early diabetes. ...
Article
Full-text available
We have studied the resetting behavior of eight basic integral controller motifs with respect to different but constant backgrounds. We found that the controllers split symmetrically into two classes: one class, based on derepression of the compensatory flux, leads to more rapid resetting kinetics as backgrounds increase. The other class, which directly activates the compensatory flux, shows a slowing down in the resetting at increased backgrounds. We found a striking analogy between the resetting kinetics of vertebrate photoreceptors and controllers based on derepression, i.e. vertebrate rod or cone cells show decreased sensitivities and accelerated response kinetics as background illuminations increase. The central molecular model of vertebrate photoadaptation consists of an overlay of three negative feedback loops with cytosolic calcium (Ca i 2 +), cyclic guanosine monophosphate (cGMP) and cyclic nucleotide-gated (CNG) channels as components. While in one of the feedback loops the extrusion of Ca i 2 + by potassium-dependent sodium-calcium exchangers (NCKX) can lead to integral control with cGMP as the controlled variable, the expected robust perfect adaptation of cGMP is lost, because of the two other feedback loops. They avoid that Ca i 2 + levels become too high and toxic. Looking at psychophysical laws, we found that in all of the above mentioned basic controllers Weber’s law is followed when a “just noticeable difference” (threshold) of 1% of the controlled variable’s set-point was considered. Applying comparable threshold pulses or steps to the photoadaptation model we find, in agreement with experimental results, that Weber’s law is followed for relatively high backgrounds, while Stephens’ power law gives a better description when backgrounds are low. Limitations of our photoadaption model, in particular with respect to potassium/sodium homeostasis, are discussed. Finally, we discuss possible implication of background perturbations in biological controllers when compensatory fluxes are based on activation.
Preprint
Full-text available
Type two diabetes mellitus (T2DM) has been shown to affect a series of cognitive processes including memory, increasing the risk for dementia, particularly Alzheimer's disease (AD). Although increasing evidence has supported that both diseases share common features, the pathophysiological mechanisms connecting these two disorders remain to be fully elucidated. Herein, we utilized Drosophila melanogaster fed on a high-sugar diet (HSD) to mimic T2DM, and investigate its effects on memory as well as identify potential molecular players associated with the memory deficits induced by HSD. Flies hatched from and reared on HSD for 7 days had a substantial decrease in short-term memory (STM). The screening for memory-related genes using transcriptome data revealed that HSD altered the expression of 33% of memory genes in relation to the control. Among the differentially expressed genes (DEGs) with a fold-change (FC) higher than two, we found five genes, related to synapse and memory trace formation, that could be considered strong candidates to underlie the STM deficits in HSD flies: Abl tyrosine kinase (Abl), Bruchpilot (Brp), Minibrain (Mnb), Skaker (Sh), and Gilgamesh (Gish). We also analyzed genes from the dopamine system, one the most relevant signaling pathways for olfactory memory. Interestingly, the flies fed on HSD presented a decreased expression of the Tyrosine hydroxylase (Ple) and Dopa decarboxylase (Ddc) genes, signals of a possible dopamine deficiency. In this work, we present promising “biomarkers” to investigate molecular networks shared between T2DM and AD.
Article
Ethnopharmacological relevance: Recent studies claim that Type-2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) overlap in several common pathological pathways which from neuronal damage to impaired memory performance. It is known that the use of Rosa canina L. (R. canina) as medicine in folk medicine dates back to ancient times and is used in the treatment of nervous diseases in Persian medicine. However, the effect of R. canina on diabetes-related cognitive decline and memory impairment has not yet been studied. Aim of the study: We evaluated the impact of T2DM on AD-like alterations and examined the molecular mechanism of a possible effect of R. canina on cognitive alterations in diabetic rats. Materials&methods: R. canina ethanol extract was obtained by maceration method. This study was performed with male Spraque-Dawley rats fed with a high-fat diet (HFD) for 8 weeks, low-dose streptozotocin (STZ; 35 mg/kg IP) injection for 4 weeks, and R. canina (250 mg/kg; per oral) and metformin (400 mg/kg; per oral) administration for 4 weeks. The weight and blood glucose of rats were measured weekly. To evaluate glucose tolerance area under the curve (AUC) was calculated by performing an oral glucose tolerance test. Then the rats were subjected to behavioural tests, and their hippocampus and cortex tissues were obtained for biochemical and morphological analyses. Results: R. canina could manage glucose responsiveness by reducing post-prandial blood glucose levels, preventing weight loss, and raising serum insulin levels in T2DM-induced rats. Behavioural tests showed that R. canina significantly improves diabetes-related cognitive decline in recall and long-term memory. Treatment with R. canina significantly reversed HFD/STZ-induced increases in insulin, amyloid-β, amyloid precursor protein levels, and acetylcholinesterase activity in the prefrontal cortex and hippocampus. Furthermore, histological analyzes revealed the protection of R. canina against neuronal disruption in the cortical and hippocampal CA3 region caused by chronic hyperglycemia. Conclusion: Analyzed collectively, these results suggest that R. canina can correct T2DM-related cognitive decline may be attributed to insulin pathway modulation, prevention of amyloid deposition, and increased cholinergic transmission.
Article
The topic of gut microbiota and the microbiota-gut-brain (MGB) axis has become the forefront of research and reports in the past few years. The gut microbiota is a dynamic interface between the environment, food, and the host, reflecting the health status as well as maintaining normal physiological metabolism. Modern ultra-processed foods (UPF) contain large quantities of saturated and trans fat, added sugar, salt, and food additives that seriously affect the gut and physical health. In addition, these unhealthy components directly cause changes in gut microbiota functions and microbial metabolism, subsequently having the potential to impact the neural network. This paper reviews an overview of the link between UPF ingredients and the MGB axis. Considerable studies have examined that high intake of trans fat, added sugar and salt have deleterious effects on gut and brain functions, but relatively less focus has been placed on the impact of food additives on the MGB axis. Data from several studies suggest that food additives might be linked to metabolic diseases and inflammation. They may also alter the gut microbiota composition and microbial metabolites, which potentially affect cognition and behavior. Therefore, we emphasize that food additives including emulsifiers, artificial sweeteners, colorants, and preservatives interact with the gut microbiota and their possible effects on altering the brain and behavior based on the latest research. Future studies should further investigate whether gut dysbiosis mediates the effect of UPF on brain diseases and behavior. This thesis here sheds new light on future research pointing to the potentially detrimental effects of processed food consumption on brain health.
Article
Full-text available
Type 2 diabetes (T2D) is considered a major risk factor for Alzheimer's disease (AD). To elucidate the links between both pathological conditions, we compared behavioral and cognitive functions, cerebral amyloid-β peptide (Aβ) levels and vasculature integrity of 11-month-old T2D and AD mice. For this purpose, we performed behavioral tests (open field, object recognition, Y-maze, and elevated plus maze tests), ELISA to assess plasma markers of endothelial/vascular dysfunction, spectrophotometric assays to evaluate cerebral vascular permeability and enzymatic activities, and immunohistochemistry for the assessment of Aβ levels. Both T2D and AD showed similar behavioral and cognitive anomalies characterized by increased fear and anxiety and decreased learning and memory abilities. Interestingly, both groups of animals presented increased plasma markers of endothelial/vascular dysfunction and permeability of cerebral vasculature and impaired mitochondrial enzymatic activities. In addition, a significant increase in Aβ levels was observed in the cortex and hippocampus of T2D mice. These results support the notion that T2D predisposes to cerebrovascular alterations, cognitive decline, and development of AD.
Article
Full-text available
Increases in fructose consumption have paralleled the increasing prevalence of obesity, and high-fructose diets are thought to promote weight gain and insulin resistance. Fructose ingestion produces smaller increases in circulating satiety hormones compared with glucose ingestion, and central administration of fructose provokes feeding in rodents, whereas centrally administered glucose promotes satiety. To study neurophysiological factors that might underlie associations between fructose consumption and weight gain. Twenty healthy adult volunteers underwent 2 magnetic resonance imaging sessions at Yale University in conjunction with fructose or glucose drink ingestion in a blinded, random-order, crossover design. Relative changes in hypothalamic regional cerebral blood flow (CBF) after glucose or fructose ingestion. Secondary outcomes included whole-brain analyses to explore regional CBF changes, functional connectivity analysis to investigate correlations between the hypothalamus and other brain region responses, and hormone responses to fructose and glucose ingestion. There was a significantly greater reduction in hypothalamic CBF after glucose vs fructose ingestion (-5.45 vs 2.84 mL/g per minute, respectively; mean difference, 8.3 mL/g per minute [95% CI of mean difference, 1.87-14.70]; P = .01). Glucose ingestion (compared with baseline) increased functional connectivity between the hypothalamus and the thalamus and striatum. Fructose increased connectivity between the hypothalamus and thalamus but not the striatum. Regional CBF within the hypothalamus, thalamus, insula, anterior cingulate, and striatum (appetite and reward regions) was reduced after glucose ingestion compared with baseline (P < .05 significance threshold, family-wise error [FWE] whole-brain corrected). In contrast, fructose reduced regional CBF in the thalamus, hippocampus, posterior cingulate cortex, fusiform, and visual cortex (P < .05 significance threshold, FWE whole-brain corrected). In whole-brain voxel-level analyses, there were no significant differences between direct comparisons of fructose vs glucose sessions following correction for multiple comparisons. Fructose vs glucose ingestion resulted in lower peak levels of serum glucose (mean difference, 41.0 mg/dL [95% CI, 27.7-54.5]; P < .001), insulin (mean difference, 49.6 μU/mL [95% CI, 38.2-61.1]; P < .001), and glucagon-like polypeptide 1 (mean difference, 2.1 pmol/L [95% CI, 0.9-3.2]; P = .01). CONCLUSION AND RELEVANCE: In a series of exploratory analyses, consumption of fructose compared with glucose resulted in a distinct pattern of regional CBF and a smaller increase in systemic glucose, insulin, and glucagon-like polypeptide 1 levels.
Article
Full-text available
OBJECTIVE Adverse effects of hypercaloric, high-fructose diets on insulin sensitivity and lipids in human subjects have been shown repeatedly. The implications of fructose in amounts close to usual daily consumption, however, have not been well studied. This study assessed the effect of moderate amounts of fructose and sucrose compared with glucose on glucose and lipid metabolism.RESEARCH DESIGN AND METHODS Nine healthy, normal-weight male volunteers (age 21-25 years) were studied in this double-blind, randomized, cross-over trial. All subjects consumed four different sweetened beverages (600 mL/day) for 3 weeks each: medium fructose (MF) at 40 g/day, and high fructose (HF), high glucose (HG), and high sucrose (HS) each at 80 g/day. Euglycemic-hyperinsulinemic clamps with [6,6]-(2)H(2) glucose labeling were used to measure endogenous glucose production. Lipid profile, glucose, and insulin were measured in fasting samples.RESULTSHepatic suppression of glucose production during the clamp was significantly lower after HF (59.4 ± 11.0%) than HG (70.3 ± 10.5%, P < 0.05), whereas fasting glucose, insulin, and C-peptide did not differ between the interventions. Compared with HG, LDL cholesterol and total cholesterol were significantly higher after MF, HF, and HS, and free fatty acids were significantly increased after MF, but not after the two other interventions (P < 0.05). Subjects' energy intake during the interventions did not differ significantly from baseline intake.CONCLUSION This study clearly shows that moderate amounts of fructose and sucrose significantly alter hepatic insulin sensitivity and lipid metabolism compared with similar amounts of glucose.
Article
Full-text available
Although the precise culprit in the etiopathogenesis of Alzheimer disease (AD) is still obscure, defective mitochondria functioning has been proposed to be an upstream event in AD. Mitochondria fulfill a number of essential cellular functions, and it is recognized that the strict regulation of the structure, function and turnover of these organelles is an immutable control node for the maintenance of neuronal and vascular homeostasis. Extensive research in postmortem brain tissue from AD subjects, and AD animal and cellular models revealed that mitochondria undergo multiple malfunctions during the course of this disease. The present review summarizes the current views on how mitochondria are implicated in both AD-related neuronal and cerebrovascular degeneration. The understanding of the mitochondrial mechanisms underlying AD pathology is critical to design more effective strategies to halt or delay disease progression.
Article
Full-text available
Many epidemiological studies have shown that diabetes, particularly type 2 diabetes, significantly increases the risk to develop Alzheimer's disease. Both diseases share several common abnormalities including impaired glucose metabolism, increased oxidative stress, insulin resistance and deposition of amyloidogenic proteins. It has been suggested that these two diseases disrupt common cellular and molecular pathways and each disease potentiates the progression of the other. This review discusses clinical and biochemical features shared by Alzheimer's disease and diabetes, giving special attention to the involvement of insulin signaling, glucose metabolism and mitochondria. Understanding the key mechanisms underlying this deleterious interaction may provide opportunities for the design of effective therapeutic strategies.
Article
Full-text available
An increasing number of studies have demonstrated a connection between Alzheimer's disease (AD) and diabetes, particularly type 2 diabetes (T2D). The risk for developing T2D and AD increases exponentially with age and having T2D increases the risk of developing AD. This has propelled researchers to investigate the mechanism(s) underlying this connection. This review critically discusses the involvement of mitochondrial abnormalities and oxidative stress in AD and diabetes highlighting the similarities between both pathologies. The impact of insulin resistance/insulin signaling impairment in AD pathogenesis will be also debated. A better understanding of the key mechanisms underlying the interaction between AD and diabetes is needed for the design of effective preventive and therapeutic strategies.
Article
Full-text available
Evidence shows that diabetes increases the risk of developing Alzheimer's disease (AD). Many efforts have been done to elucidate the mechanisms linking diabetes and AD. To demonstrate that mitochondria may represent a functional link between both pathologies, we compared the effects of AD and sucrose-induced metabolic alterations on mouse brain mitochondrial bioenergetics and oxidative status. For this purpose, brain mitochondria were isolated from wild-type (WT), triple transgenic AD (3xTg-AD), and WT mice fed 20% sucrose-sweetened water for 7 months. Polarography, spectrophotometry, fluorimetry, high-performance liquid chromatography, and electron microscopy were used to evaluate mitochondrial function, oxidative status, and ultrastructure. Western blotting was performed to determine the AD pathogenic protein levels. Sucrose intake caused metabolic alterations like those found in type 2 diabetes. Mitochondria from 3xTg-AD and sucrose-treated WT mice presented a similar impairment of the respiratory chain and phosphorylation system, decreased capacity to accumulate calcium, ultrastructural abnormalities, and oxidative imbalance. Interestingly, sucrose-treated WT mice presented a significant increase in amyloid β protein levels, a hallmark of AD. These results show that in mice, the metabolic alterations associated to diabetes contribute to the development of AD-like pathologic features.
Article
Full-text available
Baboons (Papio hamadryas Sp.) develop features of the cardiometabolic syndrome and represent a clinically-relevant animal model in which to study the aetiology of the disorder. To further evaluate the baboon as a model for the study of the cardiometabolic syndrome, we developed a high sugar high fat diet and hypothesized that it could be used to induce adiposity gain and affect associated circulating biomarkers. We developed a diet enriched with monosaccharides and saturated fatty acids that was composed of solid and liquid energy sources. We provided a group of baboons (n = 9) ad libitum access to this diet for 8 weeks. Concurrently, a control group (n = 6) was maintained with ad libitum access to a low sugar low fat baseline diet and normal water for 8 weeks. Body composition was determined by dual-energy X-ray absorptiometry and circulating metabolic biomarkers were measured using standard methodology before and after the 8 week study period. Neither body composition nor circulating biomarkers changed in the control group. Following the 8 weeks, the intervention group had a significant increase in fat mass (1.71 ± 0.98 vs. 3.23 ± 1.70 kg, p = 0.004), triglyceride (55 ± 13 vs. 109 ± 67 mg/dL, p = 0.006,), and leptin (1.19 ± 1.40 vs. 3.29 ± 2.32 ng/mL, p = 0.001) and a decline in adiponectin concentrations (33530 ± 9744 vs. 23330 ± 7863 ng/mL, p = 0.002). Percentage haemoglobin A1C (4.0 ± 0.3 vs. 6.0 ± 1.4, p = 0.002) also increased in the intervention group. Our findings indicate that when exposed to a high sugar high fat diet, young adult male baboons develop increased body fat and triglyceride concentrations, altered adipokine concentrations, and evidence of altered glucose metabolism. Our findings are in keeping with observations in humans and further demonstrate the potential utility of this highly clinically-relevant animal model for studying diet-induced metabolic dysregulation.
Article
Overall dietary energy intake, particularly the consumption of simple sugars such as fructose, has been increasing steadily in Western societies, but the effects of such diets on the brain are poorly understood. Here, we used functional and structural assays to characterize the effects of excessive caloric intake on the hippocampus, a brain region important for learning and memory. Rats fed with a high-fat, high-glucose diet supplemented with high-fructose corn syrup showed alterations in energy and lipid metabolism similar to clinical diabetes, with elevated fasting glucose and increased cholesterol and triglycerides. Rats maintained on this diet for 8 months exhibited impaired spatial learning ability, reduced hippocampal dendritic spine density, and reduced long-term potentiation at Schaffer collateral--CA1 synapses. These changes occurred concurrently with reductions in levels of brain-derived neurotrophic factor in the hippocampus. We conclude that a high-calorie diet reduces hippocampal synaptic plasticity and impairs cognitive function, possibly through BDNF-mediated effects on dendritic spines.
Article
In animals, intracerebroventricular glucose and fructose have opposing effects on appetite and weight regulation. In humans, functional brain magnetic resonance imaging (fMRI) studies during glucose ingestion or infusion have demonstrated suppression of hypothalamic signalling, but no studies have compared the effects of glucose and fructose. We therefore sought to determine if the brain response differed to glucose vs. fructose in humans independently of the ingestive process. Nine healthy, normal weight subjects underwent blood oxygenation level dependent (BOLD) fMRI measurements during either intravenous (IV) glucose (0.3 mg/kg), fructose (0.3 mg/kg) or saline, administered over 2 min in a randomized, double-blind, crossover study. Blood was sampled every 5 min during a baseline period and following infusion for 60 min in total for glucose, fructose, lactate and insulin levels. No significant brain BOLD signal changes were detected in response to IV saline. BOLD signal in the cortical control areas increased during glucose infusion (p = 0.002), corresponding with increased plasma glucose and insulin levels. In contrast, BOLD signal decreased in the cortical control areas during fructose infusion (p = 0.006), corresponding with increases of plasma fructose and lactate. Neither glucose nor fructose infusions significantly altered BOLD signal in the hypothalamus. In normal weight humans, cortical responses as assessed by BOLD fMRI to infused glucose are opposite to those of fructose. Differential brain responses to these sugars and their metabolites may provide insight into the neurologic basis for dysregulation of food intake during high dietary fructose intake.