Review
Dietary fats, cerebrovasculature integrity and Alzheimer’s disease risk

https://doi.org/10.1016/j.plipres.2009.10.004Get rights and content

Abstract

An emerging body of evidence is consistent with the hypothesis that dietary fats influence Alzheimer’s disease (AD) risk, but less clear is the mechanisms by which this occurs. Alzheimer’s is an inflammatory disorder, many consider in response to fibrillar formation and extracellular deposition of amyloid-beta (Aβ). Alternatively, amyloidosis could notionally be a secondary phenomenon to inflammation, because some studies suggest that cerebrovascular disturbances precede amyloid plaque formation. Hence, dietary fats may influence AD risk by either modulating Aβ metabolism, or via Aβ independent pathways. This review explores these two possibilities taking into consideration; (i) the substantial affinity of Aβ for lipids and its ordinary metabolism as an apolipoprotein; (ii) evidence that Aβ has potent vasoactive properties and (iii) studies which show that dietary fats modulate Aβ biogenesis and secretion. We discuss accumulating evidence that dietary fats significantly influence cerebrovascular integrity and as a consequence altered Aβ kinetics across the blood–brain barrier (BBB). Specifically, chronic ingestion of saturated fats or cholesterol appears to results in BBB dysfunction and exaggerated delivery from blood-to-brain of peripheral Aβ associated with lipoproteins of intestinal and hepatic origin. Interestingly, the pattern of saturated fat/cholesterol induced cerebrovascular disturbances in otherwise normal wild-type animal strains is analogous to established models of AD genetically modified to overproduce Aβ, consistent with a causal association. Saturated fats and cholesterol may exacerbate Aβ induced cerebrovascular disturbances by enhancing exposure of vessels of circulating Aβ. However, presently there is no evidence to support this contention. Rather, SFA and cholesterol appear to more broadly compromise BBB integrity with the consequence of plasma protein leakage into brain, including lipoprotein associated Aβ. The latter findings are consistent with the concept that AD is a dietary-fat induced phenotype of vascular dementia, reflecting the extraordinary entrapment of peripherally derived lipoproteins endogenously enriched in Aβ. Rather than being the initiating trigger for inflammation in AD, accumulation of extracellular lipoprotein-Aβ may be a secondary amplifier of dietary induced inflammation, or possibly, simply be consequential. Clearly, delineating the mechanisms by which dietary fats increase AD risk may be informative in developing new strategies for prevention and treatment of AD.

Introduction

Hallmark pathological characteristics of advanced Alzheimer’s disease (AD) include hyperphosphorylation of the microtubular protein tau in neurons and extracellular deposits of protein that are enriched in the protein amyloid-beta (Aβ) [1], [2]. The formation of tau-tangles results in neuronal synapse dysfunction and eventually loss of cell-cell communication, whereas disturbed Aβ kinetics may be pivotal to pro-inflammatory pathways that compromise cellular integrity [1], [3], [4]. Despite a substantive body of research, it is presently difficult to equivocally delineate if these pathological features of AD are causal or consequential [5], [6], emphasising the therapeutic challenge of identifying the inflammatory triggers that compromise cellular integrity.

Earlier research primarily focussed on the neuronal biogenesis of Aβ in the context that overproduction may initiate formation of fibrillar Aβ deposits and thereafter inflammation [7], [8], [9], [10]. All mutations known to cause AD increases the production of Aβ peptide. However, in sporadic and late onset AD, the most common form of AD, Aβ biosynthesis is comparable to otherwise healthy individuals [11]. Alternatively, insufficient removal of Aβ from cerebrospinal fluid (CSF) has also been proposed as a mechanism for Aβ oligomerization [12], [13], [14]. However, the brain seems potently equipped with substantive efflux processes that would otherwise prevent this. It is estimated that CSF is replenished some three times daily via the choroid plexus and indeed the epithelial cells of the choroid plexus host an array of enzymes which effectively hydrolyse potentially toxic proteins including Aβ [15]. In addition, the endothelial cells of the cerebrovasculature host receptor-proteins that permit reciprocal transfer of Aβ across the blood–brain barrier (BBB) [16], [17], [18], [19], [20]. Collectively, there seems to be exquisite cerebral Aβ homeostatic mechanisms and therefore the concept that cerebral Aβ-overload triggers inflammatory pathways seems physiologically unlikely.

Alzheimer’s disease is a chronic disorder and shares risk factors with other diseases such as non-insulin dependent diabetes and cardiovascular disease (CVD) [21], [22], [23], [24], [25]. However, chronic diseases are often ‘spectrum disorders’ with multiple aetiology. For example, obesity is a major risk factor for diabetes and CVD [26], [27], but not a requisite feature per se and 40% of subjects who experience a coronary event are normolipaemic [26], [27]. Indeed, cholesterol infiltration is not always found in atherosclerotic plaque and there is substantial heterogeneity in the extent of smooth muscle cell proliferation and tissue calcification [28], [29]. Such paradoxes raise the possibility that amyloidosis is simply one of many ‘triggers’ for dementia per se.

Common to chronic disorders, there is ample evidence that lifestyle influences AD risk and progression. Good nutrition, physical activity and environmental enrichment confer synergistic reduction in AD risk [30], [31], [32], [33], [34], [35], [36], [37]. However, in a therapeutic context, less is known of the efficacy of lifestyle interventions on disease progression, perhaps confounded by the diversity of dementia phenotypes. Given that within 20 years the expected global health burden for dementia, of which AD accounts for 80%, will exceed treatment of any other chronic disease [38], [39], [40], exploring lifestyle therapies has become as much an economic imperative as a therapeutic priority.

Most AD research has focused on damage of neurons, however there is an increasing effort to understand the possibility of cerebrovascular dysfunction as a primary risk factor for AD. This paradigm shift is arguably warranted because vascular alterations including endothelial and smooth muscle cell proliferation precede frank amyloidosis [41]. Blood plasma proteins have been detected in the parenchyma of AD brains [42], [43] and inflammatory sequelae are commonly reported [44], [45], observations that are consistent with breakdown of the BBB. Targeting vascular disturbances rather than Aβ deposition may therefore be an appropriate first-focus strategy for prevention and treatment of AD.

It is reasonable to suggest that diet is important in maintaining cerebrovascular integrity [46] particularly given the overwhelming evidence that it contributes substantially to coronary artery health and CVD risk [47], [48], [49], [50], [51], [52], [53]. Population studies also generally support this contention. Saturated fats and cholesterol are both positively associated with AD risk [54], [55], [56], [57], [58], [59], [60] and in animal models, including amyloid transgenic mice, saturated fat (SFA) and cholesterol induce or exacerbate cerebral amyloidoisis [61], [62], [63], [64]. The studies in transgenic amyloid mice are certainly consistent with a vascular contribution to disease over and above exaggerated Aβ biogenesis.

The purpose of this review is to provide contemporary consideration of the mechanisms by which dietary fats influence AD risk. Specifically, this article will focus on the putative interrelationship between plasma lipoproteins, peripheral Aβ kinetics and cerebrovasculature integrity.

Section snippets

Population, clinical and animal model studies

Population studies support a role of dietary fats in AD, although this remains controversial. Laitinen reported that intake of unsaturated fats is protective, whereas intake of saturates increases risk of AD [60]. In the Framingham study, the top quartile of plasma docosahexanoic acid (DHA) (profoundly influenced by diet) was associated with a 47% reduction in risk of all-cause dementia [65]. Strong evidence continues to come from animal studies. Many studies show that cerebral amyloid burden

Receptor mediated cerbrovascular amyloid-beta kinetics

The receptor for advanced glycosylation end products (RAGE) is one endothelial cell protein found to facilitate Aβ transfer from blood-to-brain [16], [17]. However, there is no evidence that TRL or lipoproteins per se bind to RAGE, requiring therefore transfer of Aβ from the lipoprotein particle to the aqueous mileu prior to transport via this pathway. However, in vivo and in vitro studies suggest that Aβ binds tightly to TRL and is not shed or transferred to other chaperone transporters [93],

Triacylglycerol-rich-lipoprotein amyloid-beta-induced cerbrovascular disturbances

It is proposed that post-prandial hyperamyloidemia is one possible mechanism for SFA-induced BBB dysfunction and delivery of TRL-Aβ from blood-to-brain, but presently this remains to be substantiated. Rather, we found that the plasma concentration of Aβ1–40 and Aβ1–42 in SFA-fed mice was similar to mice maintained on either MUFA, PUFA or low-fat (control) diets [106] (Fig. 2). However, caution must be exercised with this interpretation. Post-prandial hyperamyloidemia may not have been apparent

Apolipoprotein E isoforms and Alzheimer’s disease risk

Inheriting one or two alleles for apo E4 increase the risk of AD by 17% and 43%, respectively, compared to individuals hetero- or homo-zygous for apo E2 and E3 isoforms [130]. A number of hypotheses have been put forward for the positive association of AD with apo E4 and reviewed extensively in the literature [92], [131], [132], [133], [134], [135], [136], [137], [138], [139]. Briefly, key concepts include; poorer sequestration of soluble Aβ and hence a propensity for oligomers to form;

Apolipoprotein B association with agrin, perlecan, biglycan and decorin

Proteoglycans are major components of the extracellular matrices, comprised of one or more glycosaminoglycans chains covalently attached to a core protein [166]. Proteoglycans may serve as binding sites for receptors, or as mediators of cell adhesion, migration and proliferation [166]. Studies over the past decade suggest that proteoglycans, in particularly heparin sulfate proteoglycans, contribute towards the formation and thereafter stability of amyloid plaques [167]. However, their putative

Apolipoprotein B isoforms and triacylglycerol-rich-lipoprotein kinetics

In man, hepatically derived TRL can be distinguished from chylomicrons based on the apo B100 and apo B48 isoforms respectively [184], [185]. Apo B48 is synthesized in enterocytes as a consequence of mRNA processing and essentially represents half of the apo B100 amino acid sequence. It’s not clear why this editing process occurs specifically in absorptive epithelial cells of the small intestine of man, suffice to say that this may be responsible for constitutive rates of chylomicron biogenesis

Conclusion

The critical observations considered in this review are that dietary saturated fats and cholesterol cause BBB dysfunction, resulting in the blood-to-brain delivery of apo B lipoprotein-Aβ. In some individuals, dietary-induced disturbances in BBB integrity may be the initiating event for AD. If cerebrovascular disturbances are central to AD aetiology and progression, then considering strategies to positively influence integrity is a therapeutic priority. Presently, drug strategies used to treat

References (192)

  • S. Itagaki et al.

    Relationship of microglia and astrocytes to amyloid deposits of Alzheimer disease

    J Neuroimmunol

    (1989)
  • S. Oba et al.

    Diet based on the Japanese food guide spinning top and subsequent mortality among men and women in a general Japanese population

    J Am Diet Assoc

    (2009)
  • S. Yusuf et al.

    Effect of potentially modifiable risk factors associated with myocardial infarction in 52 countries (the INTERHEART study): case-control study

    Lancet

    (2004)
  • R.B. Singh et al.

    Effect of an Indo-Mediterranean diet on progression of coronary artery disease in high risk patients (Indo-Mediterranean diet heart study): a randomised single-blind trial

    Lancet

    (2002)
  • M. De Lorgeril et al.

    Effect of a mediterranean type of diet on the rate of cardiovascular complications in patients with coronary artery disease. Insights into the cardioprotective effect of certain nutriments

    J Am Coll Cardiol

    (1996)
  • J.A. Luchsinger et al.

    Dietary factors and Alzheimer’s disease

    Lancet Neurol

    (2004)
  • V. Solfrizzi et al.

    Dietary fatty acids intake: possible role in cognitive decline and dementia

    Exp Gerontol

    (2005)
  • D.L. Sparks et al.

    Induction of Alzheimer-like beta-amyloid immunoreactivity in the brains of rabbits with dietary cholesterol

    Exp Neurol

    (1994)
  • M. Oksman et al.

    Impact of different saturated fatty acid, polyunsaturated fatty acid and cholesterol containing diets on beta-amyloid accumulation in APP/PS1 transgenic mice

    Neurobiol Dis

    (2006)
  • L.M. Refolo et al.

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

    Neurobiol Dis

    (2000)
  • C.R. Hooijmans et al.

    DHA and cholesterol containing diets influence Alzheimer-like pathology, cognition and cerebral vasculature in APPswe/PS1dE9 mice

    Neurobiol Dis

    (2009)
  • C.R. Hooijmans et al.

    Changes in cerebral blood volume and amyloid pathology in aged Alzheimer APP/PS1 mice on a docosahexaenoic acid (DHA) diet or cholesterol enriched typical western diet (TWD)

    Neurobiol Dis

    (2007)
  • G.J. Petot et al.

    Lipids, diet and Alzheimer disease: an extended summary

    J Neurol Sci

    (2004)
  • A. Wu et al.

    Docosahexaenoic acid dietary supplementation enhances the effects of exercise on synaptic plasticity and cognition

    Neuroscience

    (2008)
  • S.C. Cunnane et al.

    Fish, docosahexaenoic acid and Alzheimer’s disease

    Prog Lipid Res

    (2009)
  • G.W. Arendash et al.

    A diet high in omega-3 fatty acids does not improve or protect cognitive performance in Alzheimer’s transgenic mice

    Neuroscience

    (2007)
  • M. Sarsilmaz et al.

    Potential role of dietary omega-3 essential fatty acids on some oxidant/antioxidant parameters in rats’ corpus striatum

    Prostaglandins Leukot Essent Fatty Acids

    (2003)
  • A.R. Koudinov et al.

    Biochemical characterization of Alzheimer’s soluble amyloid beta protein in human cerebrospinal fluid: association with high density lipoproteins

    Biochem Biophys Res Commun

    (1996)
  • K. Yanagisawa et al.

    Amyloid beta-protein (A beta) associated with lipid molecules: immunoreactivity distinct from that of soluble A beta

    FEBS Lett

    (1997)
  • A.L. Biere et al.

    Amyloid beta-peptide is transported on lipoproteins and albumin in human plasma

    J Biol Chem

    (1996)
  • A.R. Koudinov et al.

    Alzheimer’s soluble amyloid beta protein is secreted by HepG2 cells as an apolipoprotein

    Cell Biol Int

    (1997)
  • S. Galloway et al.

    Beta-amyloid or its precursor protein is found in epithelial cells of the small intestine and is stimulated by high-fat feeding

    J Nutr Biochem

    (2007)
  • R. Takechi et al.

    Chylomicron amyloid-beta in the aetiology of Alzheimer’s disease

    Atheroscler Suppl

    (2008)
  • K.G. Jackson et al.

    Saturated fat-induced changes in Sf 60–400 particle composition reduces uptake of LDL by HepG2 cells

    J Lipid Res

    (2006)
  • C.K. Roberts et al.

    Effect of diet on adipose tissue and skeletal muscle VLDL receptor and LPL: implications for obesity and hyperlipidemia

    Atherosclerosis

    (2002)
  • K.C. Hayes et al.

    Saturated fatty acids and LDL receptor modulation in humans and monkeys

    Prostaglandins Leukot Essent Fatty Acids

    (1997)
  • I.J. Goldberg

    Hypertriglyceridemia: impact and treatment

    Endocrinol Metab Clin North Am

    (2009)
  • F. Karpe et al.

    Differences in postprandial concentrations of very-low-density lipoprotein and chylomicron remnants between normotriglyceridemic and hypertriglyceridemic men with and without coronary heart disease

    Metabolism

    (1999)
  • J.C. Mamo et al.

    Retention of chylomicron remnants by arterial tissue; importance of an efficient clearance mechanism from plasma

    Atherosclerosis

    (1998)
  • C.L. Joachim et al.

    Clinically diagnosed Alzheimer’s disease: autopsy results in 150 cases

    Ann Neurol

    (1988)
  • C. Duyckaerts et al.

    Classification and basic pathology of Alzheimer disease

    Acta Neuropathol

    (2009)
  • D.J. Selkoe

    Alzheimer’s disease is a synaptic failure

    Science

    (2002)
  • M. Citron et al.

    Mutant presenilins of Alzheimer’s disease increase production of 42-residue amyloid beta-protein in both transfected cells and transgenic mice

    Nat Med

    (1997)
  • M.D. Kirkitadze et al.

    Molecular mechanisms initiating amyloid beta-fibril formation in Alzheimer’s disease

    Acta Biochim Pol

    (2005)
  • J.A. Hardy et al.

    Alzheimer’s disease: the amyloid cascade hypothesis

    Science

    (1992)
  • J.L. Cummings et al.

    Alzheimer’s disease: etiologies, pathophysiology, cognitive reserve, and treatment opportunities

    Neurology

    (1998)
  • R. Deane et al.

    IgG-assisted age-dependent clearance of Alzheimer’s amyloid beta peptide by the blood–brain barrier neonatal Fc receptor

    J Neurosci

    (2005)
  • R. Deane et al.

    Clearance of amyloid-beta peptide across the blood–brain barrier: implication for therapies in Alzheimer’s disease

    CNS Neurol Disord Drug Targets

    (2009)
  • J.S. Crossgrove et al.

    The choroid plexus removes beta-amyloid from brain cerebrospinal fluid

    Exp Biol Med (Maywood)

    (2005)
  • N. Strazielle et al.

    Choroid plexus in the central nervous system: biology and physiopathology

    J Neuropathol Exp Neurol

    (2000)
  • Cited by (71)

    • Long-term exposition to a high fat diet favors the appearance of β-amyloid depositions in the brain of C57BL/6J mice. A potential model of sporadic Alzheimer's disease

      2017, Mechanisms of Ageing and Development
      Citation Excerpt :

      It degrades damaged or unwanted components and recycles those destined for use in energy production and other biosynthetic reactions. Therefore, we can suggest that the increase of βA depositions in the cortex and hippocampus of the elder mice could be the result of a dysfunction of autophagy (Takechi et al., 2010a, 2010b). Thus, we theorized that autophagy is involved in the aging of the brain and in age-related neurodegenerative disorders.

    View all citing articles on Scopus
    View full text