Elsevier

Neurobiology of Aging

Volume 35, Issue 6, June 2014, Pages 1233-1242
Neurobiology of Aging

Regular article
Mammalian target of rapamycin hyperactivity mediates the detrimental effects of a high sucrose diet on Alzheimer's disease pathology

https://doi.org/10.1016/j.neurobiolaging.2013.12.006Get rights and content

Abstract

High sugar consumption and diabetes increase the risk of developing Alzheimer's disease (AD) by unknown mechanisms. Using an animal model of AD, here we show that high sucrose intake induces obesity with changes in central and peripheral insulin signaling. These pre-diabetic changes are associated with an increase in amyloid-β production and deposition. Moreover, high sucrose ingestion exacerbates tau phosphorylation by increasing Cdk5 activity. Mechanistically, the sucrose-mediated increase in AD-like pathology results from hyperactive mammalian target of rapamycin (mTOR), a key nutrient sensor important in regulating energy homeostasis. Specifically, we show that rapamycin, an mTOR inhibitor, prevents the detrimental effects of sucrose in the brain without altering changes in peripheral insulin resistance. Overall, our data suggest that high sucrose intake and dysregulated insulin signaling, which are known to contribute to the occurrence of diabetes, increase the risk of developing AD by upregulating brain mTOR signaling. Therefore, early interventions to modulate mTOR activity in individuals at high risk of developing diabetes may decrease their AD susceptibility.

Introduction

Alzheimer's disease (AD) is an untreatable neurodegenerative disorder currently affecting more than 5 million Americans. The vast majority of AD cases are sporadic and of unknown etiology; nevertheless, epidemiologic studies have provided crucial insight into disease risk factors (Mayeux and Stern, 2012). Growing evidence shows that certain lifestyle choices, such as a high sugar or high fat diet, increase the risk of developing AD (Biessels et al., 2006). The Rotterdam study, a historic prospective population-based longitudinal study, identified that diabetes alone increases the risk of developing AD by ∼2-fold (Ott et al., 1999); this finding has been widely confirmed (reviewed by Sims-Robinson et al., 2010). Treating midlife health conditions known to increase the risk for developing AD, such as diabetes, may provide an opportunity for managing the projected increase in AD incidence.

Clinically, type 2 diabetic (T2D), metabolic syndrome and AD patients share many common pathophysiological features. These include hyperglycemia, hyperinsulinemia, insulin resistance, glucose intolerance, dyslipidemia and inflammation; these traits reportedly correlate with attention and memory deficits (Jones et al., 2009). Mounting evidence suggests that insulin resistance and concomitant elevated blood glucose is a key metabolic dysfunction contributing to AD (reviewed by Cholerton et al., 2013). Namely, recent studies have reported evidence for insulin resistance in AD brains independent of the patients' diabetic status (e.g., Talbot et al., 2012). Additionally, preliminary results from an ongoing clinical study have revealed that T2D patients taking the insulin sensitizing drug, metformin, were less likely to develop AD then T2D patients taking other anti-diabetic medications (ClinicalTrials.gov Identifier: NCT01595646).

Neuropathologically, the AD brain is characterized by the accumulation of plaques, comprised mainly of amyloid-β (Aβ), and neurofibrillary tangles that are largely formed of hyperphosphorylated protein tau (Querfurth and LaFerla, 2010). Notably, patients with T2D have increased levels of hyperphosphorylated tau in their brains (Liu et al., 2009), further strengthening the link between diabetes and AD. The use of AD mouse models with either genetically or chemically induced-diabetes have also reported a link between diabetes and AD pathologies (Jolivalt et al., 2010, Ke et al., 2009, Plaschke et al., 2010, Takeda et al., 2010). For example, inducing type I diabetes by streptozotocin exacerbated the development of neurofibrillary tangles in a mouse model overexpressing mutant human tau (Ke et al., 2009). Similarly, induction of experimental diabetes with streptozotocin or analogous drugs increased Aβ and tau levels in wild-type mice and rabbits (Bitel et al., 2012, Ke et al., 2009). Collectively, studies provide compelling evidence that these prevalent age-associated diseases share alterations in common molecular pathways associated with glucose metabolism. However, the molecular mechanisms underlying the link between abnormal glucose homeostasis and AD remain to be elucidated.

Mammalian target of rapamycin (mTOR) is a protein kinase that plays a key role in maintaining energy homeostasis in the brain and other tissue types (Mannaa et al., 2013). As an energy sensor, mTOR regulates numerous cellular pathways including protein translation, cell growth, and proliferation. To modulate insulin signaling in times of high nutrient exposure, mTOR directly phosphorylates the insulin receptor leading to its internalization; this, in turn, results in a decrease of mTOR signaling (Wullschleger et al., 2006). However, through the same mechanisms, chronic mTOR hyperactivity leads to insulin resistance, a key feature of T2D (Saha et al., 2011). mTOR hyperactivity is also found in AD brains and in AD mouse models (Caccamo et al., 2010, Caccamo et al., 2011, Oddo, 2012, Pei and Hugon, 2008). We, and others, have shown that restoring mTOR activity using an mTOR inhibitor, rapamycin, mitigates AD-associated pathology and cognitive deficits (Caccamo et al., 2010, Majumder et al., 2011, Majumder et al., 2012, Spilman et al., 2010). As mTOR hyperactivity is common to both diabetes and AD, here, we explored the role of mTOR signaling as a molecular link between these two age-related diseases.

Section snippets

Mice

The 3xTg-AD mice used in these studies have been previously described (Oddo et al., 2003); only female mice were used for this study. Mice were housed in 4–5 to cages. Twice weekly, fresh 20% sucrose dissolved in water was given to all 3xTg-ADSuc and 3xTgADSuc + Rapa mice. Additionally, microencapsulated rapamycin (Harrison et al., 2009) was added at a concentration of 14 mg/kg to mouse chow for 3xTgADSuc + Rapa mice. The 3xTg-ADCTL mice were on regular water and regular chow. Mice were kept on

Prolonged sucrose intake altered insulin signaling in 3xTg-AD mice

To better understand the relation among AD, insulin resistance, and diabetes we used a candidate approach and focused on the role of mTOR signaling, given its involvement in AD pathogenesis, energy homeostasis, and insulin signaling. Specifically, 12-month-old 3xTg-AD mice, a widely used animal model of AD, were randomly assigned to one of the following groups: (1) 15 mice had ad libitum access to 20% sucrose water as the only source of water, these mice will be referred to as 3xTg-ADSuc

Discussion

Currently there are no pharmacologic interventions that effectively cure, prevent, or delay AD progression. Midlife health conditions known to increase the risk for developing AD, such as diabetes, are treatable and may provide an opportunity for intervention. While many studies have independently confirmed an increased risk of AD and diabetes (reviewed by Sims-Robinson et al., 2010), since the pivotal Rotterdam study was originally published in 1999, little progress has been made to understand

Disclosure statement

The authors declare no competing financial interests. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Acknowledgements

This work was supported by an National Institutes of Health grant to Salvatore Oddo, AG037637-03. Miranda Orr is supported by a training grant from the National Institute of Aging (T32 AG021890).

References (61)

  • D.W. Lamming et al.

    A central role for mTOR in lipid homeostasis

    Cell Metab.

    (2013)
  • L. Martin et al.

    Tau protein kinases: involvement in Alzheimer's disease

    Ageing Res. Rev.

    (2013)
  • S. Oddo et al.

    Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular abeta and synaptic dysfunction

    Neuron

    (2003)
  • D. Tang et al.

    An isoform of the neuronal cyclin-dependent kinase 5 (Cdk5) activator

    J. Biol. Chem.

    (1995)
  • J.F. Tanti et al.

    Cellular mechanisms of insulin resistance: role of stress-regulated serine kinases and insulin receptor substrates (IRS) serine phosphorylation

    Curr. Opin. Pharmacol.

    (2009)
  • S. Wullschleger et al.

    TOR signaling in growth and metabolism

    Cell

    (2006)
  • J.C. Augustinack et al.

    Specific tau phosphorylation sites correlate with severity of neuronal cytopathology in Alzheimer's disease

    Acta Neuropathol.

    (2002)
  • J.M. Backer et al.

    Phosphatidylinositol 3'-kinase is activated by association with IRS-1 during insulin stimulation

    EMBO J.

    (1992)
  • C.L. Bitel et al.

    Amyloid-beta and tau pathology of Alzheimer's disease induced by diabetes in a rabbit animal model

    J. Alzheimers Dis.

    (2012)
  • A. Caccamo et al.

    mTOR regulates tau phosphorylation and degradation: implications for Alzheimer's disease and other tauopathies

    Aging Cell

    (2013)
  • A.Y. Choo et al.

    Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation

    Proc. Natl. Acad. Sci. U.S.A

    (2008)
  • K.D. Copps et al.

    Regulation of insulin sensitivity by serine/threonine phosphorylation of insulin receptor substrate proteins IRS1 and IRS2

    Diabetologia

    (2012)
  • C.J. Cortes et al.

    Rapamycin delays disease onset and prevents PrP plaque deposition in a mouse model of Gerstmann-Straussler-Scheinker disease

    J. Neurosci.

    (2012)
  • L. Devi et al.

    Mechanisms underlying insulin deficiency-induced acceleration of beta-amyloidosis in a mouse model of Alzheimer's disease

    PLoS One

    (2012)
  • Y. Feng et al.

    Cleavage of GSK-3beta by calpain counteracts the inhibitory effect of Ser9 phosphorylation on GSK-3beta activity induced by H2O2

    J. Neurochem.

    (2013)
  • D.E. Harrison et al.

    Rapamycin fed late in life extends lifespan in genetically heterogeneous mice

    Nature

    (2009)
  • M. Jimenez-Palomares et al.

    Increased abeta production prompts the onset of glucose intolerance and insulin resistance

    Am. J. Physiol. Endocrinol. Metab.

    (2012)
  • C.G. Jolivalt et al.

    Defective insulin signaling pathway and increased glycogen synthase kinase-3 activity in the brain of diabetic mice: parallels with Alzheimer's disease and correction by insulin

    J. Neurosci. Res.

    (2008)
  • A. Jones et al.

    Common pathological processes and transcriptional pathways in Alzheimer's disease and type 2 diabetes

    J. Alzheimers Dis.

    (2009)
  • Y.D. Ke et al.

    Experimental diabetes mellitus exacerbates tau pathology in a transgenic mouse model of Alzheimer's disease

    PLoS One

    (2009)
  • Cited by (64)

    • A high sucrose diet modifies brain oxylipins in a sex-dependent manner

      2022, Prostaglandins Leukotrienes and Essential Fatty Acids
    • A high-sucrose diet aggravates Alzheimer's disease pathology, attenuates hypothalamic leptin signaling, and impairs food-anticipatory activity in APPswe/PS1dE9 mice

      2020, Neurobiology of Aging
      Citation Excerpt :

      Two-way ANOVA indicated that HSD interacts with APP/PS1 genetic background on the upregulation of GFAP and IL-1β mRNA in the mouse cortex (Table 2). Previous studies have shown that insulin resistance can be induced by HSD in WT rodents (Apolzan and Harris, 2013; Oliveira et al., 2014; Schultz et al., 2015) and by sucrose-sweetened water in 3×Tg AD mice (Orr et al., 2014). Whether or not glucose homeostasis in APP/PS1 mice is altered by HSD was assessed in this study.

    View all citing articles on Scopus
    View full text