Elsevier

Neurobiology of Disease

Volume 44, Issue 1, October 2011, Pages 116-124
Neurobiology of Disease

Dysregulation of SREBP2 induces BACE1 expression

https://doi.org/10.1016/j.nbd.2011.06.010Get rights and content

Abstract

β-Amyloid hyperproduction has been observed in response to alterations in neuronal intracellular cholesterol storage, efflux, and synthesis, induced in rats by a high-fat diet. It has been suggested that cholesterol homeostasis is altered in Alzheimer's disease resulting in higher β- and γ-secretase activity. In the current study the neuronal activation status of sterol regulatory element binding protein 2 (SREBP2) as well as its involvement in β-secretase BACE1 activity was investigated in high-fat fed rats (26% fat and 4% cholesterol for 20 weeks), and in SK-N-BE neuroblastoma cells exposed to 20 μM cholesterol. This work demonstrates that in the brain a hyperlipidic diet is able to induce a hyper-expression of BACE1 and determine an unbalance in cerebral cholesterol homeostasis so that SREBP2 is activated. In addition, we show for the first time the involvement of SREBP2 on expression of BACE1 in SK-N-BE cells exposed to high cholesterol. Although the enhanced risk of Alzheimer's disease in metabolic syndrome is related to several factors, our results suggest that SREBP2, which can be modulated by the impairment of cerebral cholesterol homeostasis, has a direct role on BACE1 expression and may be involved in Alzheimer's disease progression.

Highlights

► We propose a link between alterations of brain cholesterol homeostasis and BACE1. ► We utilized high-fat fed rats and high cholesterol-treated SK-N-BE cells. ► In both models we observed an increased expression of SREBP2 and BACE1. ► SREBP2 is able to bind BACE1 promoter and directly induce its expression. ► Changes in brain cholesterol represent a risk for BACE1 activation.

Introduction

Recent studies indicate that alterations in cholesterol metabolism induced by a high-fat diet influence some molecular mechanisms involved in neurodegenerative diseases, such as Niemann–Pick, Huntington's disease, prion diseases, and Alzheimer's disease (AD) (Bach et al., 2009, Bi and Liao, 2010, Block et al., 2010, Lütjohann et al., 2000, Solomon et al., 2009), even if controversial data have been reported (Hoyer and Riederer, 2007, Ishii et al., 2003, Zandi et al., 2005). Our study investigates a new potential link between diet-induced alterations in brain cholesterol metabolism and the β-site APP-cleaving enzyme 1 (BACE1) overexpression which represents an early event in AD development.

The CNS contains as much as 23% of total body cholesterol (Dietschy and Turley, 2004) and, since the blood brain barrier (BBB) is nearly impermeable to plasma lipoprotein-associated cholesterol, almost all of brain cholesterol is locally synthesized (Bjorkhem and Meaney, 2004). Excess of cholesterol is exported from the brain in the form of oxysterols which can cross the BBB. The major oxysterol in the brain is the 24S-hydroxycholesterol (24-OH-chol) which regulates genes involved in intracellular free-cholesterol modulation (Wang et al., 2008).

Brain cholesterol synthesis is tightly regulated by a feedback system based on the activity of sterol regulatory element binding protein 2 (SREBP2). In its inactivated form, SREBP2 is retained in the endoplasmic reticulum (ER) by two inhibitory proteins, SCAP and Insig. When cholesterol de novo synthesis is required, SREBP2 is cleaved and translocated to the nucleus, where it activates target genes involved in cholesterol synthesis and uptake (Bengoechea-Alonso and Ericsson, 2007, Goldstein et al., 2006). In contrast, when the intracellular levels of cholesterol increase, SREBP2 is down-regulated. Thus, a proper SREBP2 intracellular traffic is crucial for its activity. Indeed, an up-regulation of SREBP2 in prion-infected neuroblastoma cells has been recently reported. In this study authors suggest that changes in intracellular cholesterol distribution by prion-infection lead to its local decrease at the level of ER and this causes an activation of SREBP2 and cholesterol biosynthesis (Bach et al., 2009). SREBP2 controls the expression of enzymes involved in cholesterol synthesis. One of the target genes activated by SREBP2 is that encoding for the enzyme hydroxy-methylglutaryl-Coenzyme A reductase (HMG-CoA-R), the rate limiting enzyme in the process of cholesterol synthesis (Bengoechea-Alonso and Ericsson, 2007).

Intracellular cholesterol dynamics can be influenced by several factors and a high fat diet is among these. High cholesterol diet leads to metabolic disturbances including obesity, insulin resistance and dislipidemia and may contribute, at least in part, to the development of brain diseases through the action of key mediators such as trophic factors support alterations, mitochondrial abnormalities, oxidative stress increase, and intracellular cholesterol homeostasis failure (Koudinov and Koudinova, 2005, Zhang et al., 2009). Moreover, several studies report that alterations in both plasma and in brain cholesterol levels are related to AD (Kivipelto and Solomon, 2006, Puglielli et al., 2003, Simons et al., 2001, Solomon et al., 2009). Other studies, demonstrating a possible beneficial effect of cholesterol lowering drugs, such as statins or 7-dehydrocholesterol reductase inhibitors, against AD development, support this hypothesis (Kirsch et al., 2003, Parsons et al., 2007, Tong et al., 2009). Indeed, in hypercholesterolemic patients, statins reduce serum β-amyloid peptide (Aβ) levels (Buxbaum et al., 2002, Friedhoff et al., 2001), even if other data are conflicting (Ishii et al., 2003, Serrano-Pozo et al., 2010). However, a recent therapeutic approach to ameliorate amyloid pathology has proposed to act on cholesterol synthesis or on inhibition of the activity of specific enzymes correlated to conversion between free cholesterol and cholesterol esters (Bryleva et al., 2010, Parsons et al., 2007).

The abnormal generation and deposition of Aβ are the pathologic hallmark of AD. Aβ is generated by two sequential proteolytic cleavage steps from the β-amyloid precursor protein (βAPP) (Haass, 2004) that is initially cleaved by the β-secretase BACE1, which has been identified as a β-site APP-cleaving transmembrane aspartic protease. This cleavage is followed by the subsequent proteolysis of the membrane-bound C-terminal fragment catalyzed by the γ-secretase complex (Vassar, 2001). Hypercholesterolemia is linked to increased Aβ production and deposition in the brain both in humans (Kivipelto and Solomon, 2006, Pappolla et al., 2003) and in animals (Ghribi et al., 2006, Thirumangalakudi et al., 2008) and is linked to an increased risk of developing AD (Rushworth and Hooper, 2010). Moreover, Aβ hyperproduction has been observed in response to alterations in neuronal intracellular cholesterol storage, efflux, trafficking, and synthesis, induced in rats by a high-fat diet (Koudinov and Koudinova, 2005) and it has been suggested that cholesterol homeostasis is altered in AD brains resulting in higher β- and γ-secretase activity (Xiong et al., 2008).

In the current study the neuronal activation status of SREBP2 as well as its involvement in BACE1 activity was investigated in in vivo experimental conditions mimicking a cholesterol-rich Western diet feeding and in high cholesterol-exposed SK-N-BE neuroblastoma cells.

Section snippets

Materials and methods

All compounds were purchased from Sigma Chemical Co. (St. Louis, MO, USA) and materials for real-time PCR were from Bio-Rad Laboratories (Hercules, CA, USA), unless otherwise stated.

High fat diet in rats induces alterations in body weight, glucose tolerance, plasma lipid profile and brain cholesterol metabolism

As shown in Fig. 1, panel A, rats fed a high-fat diet (HF rats) for 20 weeks showed a significant increase in body weight compared to control rats (CTRL) (+ 30%), paralleled at insulin resistance onset (insulin plasma levels + 270%, HOMA index + 150%). Fasting blood glucose concentration was unchanged in both experimental groups.

Before the sacrifice of the animals, an oral glucose tolerance test (OGTT) was performed. As reported in Fig. 1, panel B, the glycemic curve in HF rats significantly moved

Discussion

High cholesterol levels may be detrimental to human neurological functions, although either positive, negative or null correlations between cholesterol levels and impaired cognitive functions have been reported by several studies (Hoyer and Riederer, 2007, Ishii et al., 2003, Kivipelto and Solomon, 2006, Pappolla et al., 2003, Zandi et al., 2005).

In the current study we demonstrate an up-regulation of SREBP2 and BACE1 in the brain of rats fed a high cholesterol diet as well as in SK-N-BE

Acknowledgments

This study was supported by the Regione Piemonte “Progetto di Ricerca Sanitaria Finalizzata 2009”, by the CRT Foundation 2009: “Il diabete come fattore di rischio per l'insorgenza della malattia di Alzheimer”, Turin, Italy, and by the PRIN 2007.

References (51)

  • L.M. Refolo et al.

    Hypercholesterolemia accelerates the Alzheimer's amyloid pathology in a transgenic mouse model

    Neurobiol. Dis.

    (2000)
  • L.A. Shobab et al.

    Cholesterol in Alzheimer's disease

    Lancet Neurol.

    (2005)
  • X.K. Tong et al.

    Simvastatin improves cerebrovascular function and counters soluble amyloid-beta, inflammation and oxidative stress in aged APP mice

    Neurobiol. Dis.

    (2009)
  • C. Ullrich et al.

    Hypercholesterolemia in rats impairs the cholinergic system and leads to memory deficits

    Mol. Cell. Neurosci.

    (2010)
  • B. Wolozin

    Cholesterol and the biology of Alzheimer's disease

    Neuron

    (2004)
  • H. Xiong et al.

    Cholesterol retention in Alzheimer's brain is responsible for high beta- and gamma-secretase activities and Abeta production

    Neurobiol. Dis.

    (2008)
  • L. Zhang et al.

    Diet-induced metabolic disturbances as modulators of brain homeostasis

    Biochim. Biophys. Acta.

    (2009)
  • X. Bi et al.

    Cholesterol in Niemann–Pick type C disease

    Subcell. Biochem.

    (2010)
  • I. Björkhem

    Do oxysterols control cholesterol homeostasis?

    J. Clin. Invest.

    (2002)
  • I. Bjorkhem et al.

    Brain cholesterol: long secret life behind a barrier

    Aterioscler. Tromb. Vasc. Biol.

    (2004)
  • I. Björkhem et al.

    Oxysterols and Alzheimer's disease

    Acta Neurol. Scand. Suppl.

    (2006)
  • K.Z. Bourne et al.

    Differential regulation of BACE1 promoter activity by nuclear factor-kappaB in neurons and glia upon exposure to beta-amyloid peptides

    J. Neurosci. Res.

    (2007)
  • E.Y. Bryleva et al.

    ACAT1 gene ablation increases 24(S)-hydroxycholesterol content in the brain and ameliorates amyloid pathology in mice with AD

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

    (2010)
  • J.D. Buxbaum et al.

    Pharmacological concentrations of the HMG-CoA reductase inhibitor lovastatin decrease the formation of the Alzheimer beta-amyloid peptide in vitro and in patients

    Front. Biosci.

    (2002)
  • J.M. Cordy et al.

    Exclusively targeting beta-secretase to lipid rafts by GPI-anchor addition up-regulates beta-site processing of the amyloid precursor protein

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

    (2003)
  • Cited by (19)

    • Lead exposure induces Alzheimers's disease (AD)-like pathology and disturbes cholesterol metabolism in the young rat brain

      2018, Toxicology Letters
      Citation Excerpt :

      Barbero et al. found that SREBP2 expression were increased in APP/PS1 mouse and overexpression of SREBP2 result in AD pathologies (Barbero-Camps et al., 2013). Meanwhile, SREBP2 is one of the transcription factors for BACE1 and dysregulated activation of SREBP2 induced an increase of BACE1 expression (Mastrocola et al., 2011). In our study, we found that lead exposure significantly increased the mRNA and protein expression of SREBP2 compared with those in the control.

    • Investigation of Potential Drug Targets for Cholesterol Regulation to Treat Alzheimer’s Disease

      2023, International Journal of Environmental Research and Public Health
    View all citing articles on Scopus
    View full text