Hostname: page-component-76fb5796d-22dnz Total loading time: 0 Render date: 2024-04-26T14:05:43.834Z Has data issue: false hasContentIssue false

Nutrients in Alzheimer’s Disease: The Interaction of Diet, Drugs and Disease

Published online by Cambridge University Press:  28 January 2019

S. Imindu Liyanage
Affiliation:
Krembil Research Institute, University Health Network, Toronto, ON, Canada
Prachi Vilekar
Affiliation:
Krembil Research Institute, University Health Network, Toronto, ON, Canada
Donald F. Weaver*
Affiliation:
Krembil Research Institute, University Health Network, Toronto, ON, Canada Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
*
Correspondence to: Donald F. Weaver, Faculty of Medicine, University of Toronto, Toronto, ON, Canada. Email: donald.weaver@uhnresearch.ca
Rights & Permissions [Opens in a new window]

Abstract

In recent decades, clinical trials in Alzheimer’s disease (AD) have failed at an unprecedented rate. The etiology of AD has since come under renewed scrutiny, both to elucidate the underlying pathologies and to identify novel therapeutic strategies. Here, diet has emerged as a potential causative/protective agent. A variety of nutrients, including lipids, minerals, vitamins, antioxidants and sugars as well as broader dietary patterns and microbiotal interactions have demonstrated associations with AD. Although clinical trials have yet to definitively implicate any singular dietary element as therapeutic or causative, it is apparent that dietary preferences, likely in complex synergies, may influence the risk, onset and course of AD. This review catalogs the impact of major dietary elements on AD. It further examines an unexplored reciprocal association where AD may modulate diet, as well as how potential therapeutics may complicate these interactions. In doing so, we observe diet may have profound effects on the outcome of a clinical trial, either as a confounder of a drug/disease interaction or as a generally disruptive covariate. We therefore conclude that future clinical trials in AD should endeavor to control for diet, either in study design or subsequent analyses.

Résumé

Les interactions entre l’alimentation et la prise de médicaments dans le cas de la maladie d’Alzheimer. Au cours des dernières décennies, les essais cliniques liés à la maladie d’Alzheimer (MA) ont échoué à un rythme sans précédent. Depuis, l’étiologie de cette maladie a fait l’objet d’un examen plus attentif à la fois pour mieux comprendre les troubles sous-jacents de la MA et pour identifier de nouvelles stratégies thérapeutiques. À cet égard, l’alimentation est apparue au fil des années comme un agent potentiellement causal/protecteur dans le cas de la MA. On a ainsi pu démontrer qu’une variété de nutriments, ce qui inclut des lipides, des minéraux, des vitamines, des antioxydants et des sucres, de même que des habitudes alimentaires plus variées et des interactions propres à la flore intestinale, pouvaient être associées à l’évolution de la MA. Bien que l’on doive encore établir de façon définitive au moyen d’essais cliniques le rôle causal et thérapeutique de quelque élément nutritif que ce soit, il est clair que les choix alimentaires que l’on fait, probablement dans le cadre de synergies complexes, pourraient avoir une influence sur le risque d’être atteint de la MA, sur son apparition et son évolution. Cette étude entend donc se pencher sur l’impact des principaux éléments nutritifs en ce qui a trait à la MA. Elle examinera en outre les associations réciproques peu abordées en vertu desquelles il est possible que la MA ait un impact sur l’alimentation ; elle cherchera aussi à savoir dans quelle mesure d’éventuelles options thérapeutiques peuvent affecter ces interactions. Ce faisant, nous sommes d’avis que l’alimentation pourrait avoir de profonds effets sur les résultats d’essais cliniques, que ce soit à titre d’élément perturbant les interactions entre un médicament et la maladie ou, le plus souvent, à titre de covariable perturbatrice. En somme, nous sommes amenés à conclure que les essais cliniques du futur portant sur la MA devraient s’efforcer de tenir compte de l’alimentation, que ce soit dans leur méthodologie ou lors d’analyses ultérieures.

Type
Review Article
Copyright
Copyright © 2019 The Canadian Journal of Neurological Sciences Inc. 

Introduction

Alzheimer’s disease (AD) is the most common form of dementia,Reference Wong, Gilmour and Ramage-Morin 1 affecting 1 in 10 North American seniors (aged ≥65). 2 , Reference Qiu, Kivipelto and von Strauss 3 Although the search for a disease-modifying agent has been intense, 99.6% of compounds entering clinical trials have failed.Reference Cummings, Morstorf and Zhong 4 , Reference Becker, Greig and Giacobini 5 Typically, these failures are ascribed to an inadequate understanding of the multifactorial etiology of AD; studies have therefore urged renewed investigation of the underlying neuropathologies. However, the emerging complexity and prevalence of AD may suggest that broader systemic or environmental factors, not routinely considered in neurology, may also be relevant to the onset and outcomes of disease.

In early studies, environmental factors, like metals, toxins and diet, were thought to be the definitive causes of AD. Although the etiology has since proven more complex, this environmental hypothesis regained credence in the 1990s when studies of AD prevalence among Japanese migrants in the United States revealed rates assimilative to local populations, and significantly higher than in native Japanese.Reference White, Petrovitch, Ross, Masaki, Abbott and Teng 6 Similar trends were discerned among African-American and Hispanic-American populations, which cumulatively suggest a role for environment in AD pathogenesis.Reference Hendrie, Ogunniyi and Hall 7 Reference Tang, Cross and Andrews 9

Significant changes in AD prevalence were also emerging among static populations; of note again is Japan, which witnessed a dramatic rise in prevalence from 1985 to 2010, as reported by Grant.Reference Grant 10 This loosely correlated with a period of westernization, in which dietary habits underwent a transformation from traditional cuisines, rich in fish and fresh vegetables, to an appreciably more North-American diet, heavier in fats and processed foods.Reference Tanaka, Shimabukuro and Shimabukuro 11 , Reference Grant 12 Although purely speculative, these loose associations revived some interest in diet as a causative factor in AD.

At present, extensive studies have identified various mitigating and aggravating nutrients, in addition to regional diets and metabolic states, which may play a role in AD development and progression. This review first examines the major nutritional influences on AD, as currently understood, and catalogs the relevant lipids, carbohydrates, vitamins, minerals and broader dietary patterns associated with AD. In addition, we observe and report on a relatively unexplored reciprocal association, wherein AD may alter dietary preference and nutritional intake. This review also considers the implications of these interactions to therapeutic agents and clinical trials. Specifically, we present an argument that the multifaceted interactions among diet, drugs and AD may be sufficient to confound or obscure the efficacy of a clinical agent in a drug trial, and suggest strategies to abate such an influence.

Alzheimer’s Disease Pathology

Alzheimer’s disease is characterized by three dominant neuropathologies: oligomerization of β-amyloid (Aβ), aggregation of tau into neurofibrillary tangles and brain atrophy.Reference Perl 13 The etiology of these pathologies remains unclear; however, it is well established that no recognized singular dysfunction directly precedes the deposition of plaques and tangles. Rather, a complex interlay of proteopathic and immunopathic changes culminate in the observed pathologies.Reference Nelson, Braak and Markesbery 14 Reference Swerdlow 16

Aβ aggregates originate from transmembrane amyloid-precursor proteins (APP). APP is typically associated with essential signaling roles in cellular physiology,Reference van der Kant and Goldstein 17 but when cleaved by α/γ-secretase or β-site APP cleaving enzyme (BACE), they can misfold and aggregate into toxic oligomers.Reference Murphy and LeVine 18 This is attended by an inflammatory cascade, which causes further damage to the neural parenchyma, as well as elevate oxidative stress.Reference Akiyama, Barger and Barnum 19 Within neurons, AD is characterized by the accumulation of fibrillary tangles composed of microtubule-associated tau protein.Reference Iqbal, Liu, Gong and Grundke-Iqbal 20 Ordinarily, tau participates in intracellular transport and structural functions. In AD, it becomes hyper-phosphorylated and disassociates from the microtubules. Once free, tau aggregates to form a helical fiber, which impedes intracellular transport and communication.Reference Iqbal, Liu, Gong and Grundke-Iqbal 20 Together, Aβ and tau oligomerization lead to neuronal death and cerebral atrophy. This manifests in the deterioration of memory and cognition, as well as progressive declines of reasoning, language, and executive function. Alzheimer’s disease eventually evolves to an end-stage disease, typically resulting in mortality 4-8 years post diagnosis.Reference Helzner, Scarmeas, Cosentino, Tang, Schupf and Stern 21

Diet and Alzheimer’s Disease

Early associations between diet and AD were assumed to be ancillary to other primary disorders, like cerebrovascular disease. However, recent data have suggested a direct role for diet in modulating oxidative stress, inflammation, synaptic plasticity, amyloidogenesis and neurogenesis—processes fundamental to AD pathogenesisReference Wu, Ying and Gomez-Pinilla 22 Reference Gomez-Pinilla 24 and major treatment strategies.Reference Folch, Ettcheto and Petrov 25 Reference Vina, Lloret, Orti and Alonso 28 This has implicated diet as contributory in AD, and sparked numerous nutrient-based therapeutic trials. In this section, we review the interaction of major dietary elements, both protective and detrimental, with AD pathologies.

Lipids: Cholesterol and Fatty Acids

In the 1990s, a relationship between lipids and AD was suggested when mutations in the cholesterol carrier apolipoprotein E were identified as a major genetic predisposition for late-onset Alzheimer’s.Reference Strittmatter, Saunders and Schmechel 29 , Reference Strittmatter and Roses 30 Thereafter, cholesterol was shown to have a regulatory role on the proteolytic enzymes cleaving APP, where elevated membrane cholesterol increases both γ-secretase and BACE1 activity, ultimately upregulating Aβ production.Reference Xiong, Callaghan and Jones 31 , Reference Thirumangalakudi, Prakasam and Zhang 32 Cholesterol may also facilitate the aggregation of Aβ from its monomeric form to the neurotoxic oligomer.Reference Liu and Zhang 33 , Reference Di Scala, Chahinian, Yahi, Garmy and Fantini 34 In a mechanistic study, Kakio et al demonstrated that cholesterol elicits the binding of Aβ to cellular membranes, which can then facilitate a conformational change from a helical-rich Aβ structure to an aggregation prone β-pleated sheet.Reference Kakio, Nishimoto, Yanagisawa, Kozutsumi and Matsuzaki 35 Cholesterol may therefore seed the oligomerization of Aβ via membrane binding.Reference Kakio, Nishimoto, Yanagisawa, Kozutsumi and Matsuzaki 35

This cholesterol hypothesis, reviewed by Liu and Zhang,Reference Liu and Zhang 33 has yielded contradictory clinical outcomes. Certain statin-based therapies,Reference Lin, Chuang and Hsieh 36 , Reference Shepardson, Shankar and Selkoe 37 and the inhibition of cholesterol biosynthesis,Reference Kim, Kim, Jang and Mook-Jung 38 have offered protection against Aβ accumulation and AD development. However, cholesterol is known to decrease with AD progression, and a gradual decline of serum cholesterol was identified as a risk factor for dementia in a large, 32-year investigation.Reference Mielke, Zandi and Shao 39 , Reference Presecki, Muck-Seler and Mimica 40 Studies are further suggesting vital structural and functional roles for cholesterol in neurons.Reference Qin, Liao, Baudry and Bi 41 , Reference Mathews and Appel 42 Consequently, as an independent therapeutic strategy for AD, cholesterol management alone may prove challenging.

The role of fatty acids has garnered similar attention. These are among the best-studied lipids, and have shared conclusive associations to various diseases, including AD.Reference Liu and Zhang 33 Moreover, following early work by Foot et al, which demonstrated that diet can directly alter the fat composition of the brain, fatty acid management has long been purported to possess therapeutic potential.Reference Foot, Cruz and Clandinin 43

Dietary fats are classified by length, degree of saturation and isomerism (cis or trans). Saturated fatty acids and trans-unsaturated fatty acids have been associated with an adverse risk of AD. An observational study by Morris et al discerned that diets high in these fats could more than double the risk of AD over an average of 4 years.Reference Morris, Evans and Bienias 44 Unlike cholesterol, a clear interaction with AD pathologies has not been discerned. Rather, saturated and trans-fats have been linked to altered gene expression, elevated inflammatory signaling, disrupted metabolism, protein pathologies and higher oxidative stress, all of which have been linked to cognitive decline and behavioral changes.Reference Freeman, Haley-Zitlin, Rosenberger and Granholm 45 Reference Julien, Tremblay and Phivilay 48 Prominent associations between these fats and traditional AD risk factors such as cerebrovascular disease, obesity and diabetes may also mediate the observed correlations.Reference Takechi, Galloway, Pallebage-Gamarallage, Lam and Mamo 49 , Reference Pugazhenthi, Qin and Reddy 50 Although occasional studies have revealed null or insignificant associations after extended follow-up,Reference Engelhart, Geerlings and Ruitenberg 51 the link between saturated and trans-fats with AD appears conclusive and detrimental.

Conversely, some polyunsaturated fatty acids (PUFAs) have revealed protective associations. Omega(ω)-3 PUFAs in particular, typically obtained from fish, are associated with beneficial outcomes in both AD and dementia.Reference Hooijmans, Pasker-de Jong, de Vries and Ritskes-Hoitinga 52 , Reference Cole, Ma and Frautschy 53 Studies have repeatedly demonstrated that consumption of these fats (such as docosahexaenoic acid) are inversely associated with cognitive decline and AD.Reference Yurko-Mauro 54 Reference Siegel and Ermilov 58 Furthermore, an exhaustive review by Cole et al identified 11 pleiotropic mechanisms by which ω-3 fats may confer this protection, ranging from regulating apoptosis to diminishing local pro-inflammatory markers.Reference Cole, Ma and Frautschy 53 Yet, the effect is not without contradiction, particularly among carriers of the ApoE4 allele, as well as otherwise well-nourished subjects, where no effect has been observed.Reference Freund-Levi, Hjorth and Lindberg 59 Reference Araya-Quintanilla, Gutierrez-Espinoza and Sanchez-Montoya 63 Clinical trials and meta-analyses have also failed to demonstrate a significant benefit in AD, although other cognitive parameters may be improved in unrelated cohorts.Reference Araya-Quintanilla, Gutierrez-Espinoza and Sanchez-Montoya 63 , Reference Mazereeuw, Lanctot, Chau, Swardfager and Herrmann 64

Omega-6 PUFAs, however, derived from meat, and often more abundant in a western diet, may be deleterious to an AD brain.Reference Creegan, Hunt, McManus and Rainey-Smith 65 Reference Baierle, Vencato and Oldenburg 67 Specifically, the phospholipid product of ω-6 fatty acids, arachidonic acid, is implicated in upregulating inflammation and promoting Aβ aggregation.Reference Otsuka, Yamaguchi and Ueki 66 , Reference Amtul, Uhrig, Wang, Rozmahel and Beyreuther 68 This claim has been occasionally disputed, and studies have suggested that a therapeutic ratio between ω-6 and ω-3 PUFAs may exist. Hosono et al showed that in a 2:1 dietary ratio of arachidonic acid to docosahexaenoic acid, Tg2576 mice had lower deposition of amyloid plaques and altered APP processing;Reference Hosono, Nishitsuji and Nakamura 69 ω-6 and ω-3 fats have also been shown to preserve neural stem cells in rats.Reference Tokuda, Kontani, Kawashima, Kiso, Shibata and Osumi 70 This suggests PUFAs may operate under a dynamic synergy. However, without further study it remains unclear whether combinations of these fats are simply confounding the observation of each other’s effects, or whether there is a genuine additive association.

Broadly, cholesterol, saturated fats, trans-fats and ω-6 fatty acids are likely deleterious in the context of AD, while ω-3 fatty acids have demonstrated some potential benefits. Although in all cases associations have been obscured by clinical inefficacy and contradictory findings, dietary fats may alter Aβ dynamics, influence neural inflammation and present numerous additional mechanisms of interaction with AD.

Carbohydrates: Sugars and Insulin

High sugar and high calorie diets are well-established risk factors for dementia and AD.Reference Otsuka 71 Reference Luchsinger, Tang, Shea and Mayeux 74 Further, as contributors to diabetes and other metabolic syndromes, which themselves are risk factors for AD, the association between elevated sugar levels and Alzheimer’s is strong.Reference Mushtaq, Khan, Kumosani and Kamal 75 Reference Peila, Rodriguez, Launer and Honolulu-Asia Aging 78 Paradoxically, AD-afflicted neurons are typically starved of glucose with marked reductions in glucose metabolism (up to 44%) across brain regions most associated with AD pathology.Reference Ishii, Sasaki and Kitagaki 79 Reference Hoyer, Oesterreich and Wagner 81 This pattern has become so robust that topographic patterns of impaired glucose metabolism can serve as an early marker of AD, and act as a criterion for staging progression.Reference Wattamwar and Mathuranath 82

One explanation for this apparent paradox implicates glucose transport across the blood-brain barrier (BBB). AD brains have lower levels of the glucose transporters GLUT1 and GLUT3, and therefore diminished shuttling of glucose across the BBB.Reference Szablewski 83 Winkler et al demonstrated this transporter-mediated glucose starvation can aggravate AD neuropathologies, vascular degeneration and impair cognition.Reference Iadecola 80 , Reference Winkler, Nishida and Sagare 84 An alternative theory implicates insulin resistance for the observed glucose dysregulation. Insulin resistance occurs when tissues loose sensitivity to insulin signaling after protracted exposure, triggered by chronically elevated blood sugar. The breakdown of signaling prevents glucose from entering cells, leading simultaneously to hyperglycemia, and diminished glucose within cells. Insulin resistance has also been implicated in triggering inflammation, which may exacerbate AD pathologies.Reference Creegan, Hunt, McManus and Rainey-Smith 65 In addition, insulin signaling is integral to neural function, maintenance, plasticity and is increasingly associated with cognition (in the context of diabetes) all of which have been compromised when insulin signaling is perturbed.Reference McNay and Recknagel 85 Reference Akintola and van Heemst 87 More recently, components of insulin signaling were implicated in regulating tau phosphorylation and tangling,Reference Hong and Lee 88 Reference Planel, Tatebayashi and Miyasaka 90 as well as Aβ synthesis and degradation.Reference Vekrellis, Ye and Qiu 91 Reference Craft 93 This has led to speculation as to whether altered glucose metabolism may be causative in AD. Evidence of glucose dysregulation preceding clinical symptoms, as well as its high prevalence among AD cohorts provide some support for this notion.Reference Mosconi, Pupi and De Leon 94

Micronutrients: Vitamins and Minerals

The rise of commercial vitamins and dietary supplements reflects a growing appreciation of the importance of micronutrients in health maintenance. The accessibility of these supplements also offers a practical strategy to manage AD on a large scale. However, in spite of exhaustive investigations, including several well-powered clinical trials, no isolated micronutrient has yet been definitively demonstrated as an effective therapy for AD.Reference Luchsinger, Noble and Scarmeas 95 Yet, deficiencies of various vitamins and minerals have been associated with an elevated risk of AD, cognitive impairment and other dementias.Reference Creegan, Hunt, McManus and Rainey-Smith 65 ,95

The B vitamin family is critical to the metabolism of most macronutrients, including carbohydrates and lipids, and has therefore garnered some interest in the context of AD.Reference Huskisson, Maggini and Ruf 96 Vitamin B1 or thiamine was among the first nutrients associated with dementia when it was conclusively linked to Wernicke’s encephalopathy in long-term alcoholics.Reference Butterworth 97 , Reference Rodríguez, Qizilbash, López-Arrieta and Rodríguez 98 Earlier studies have noted conspicuous similarities between the symptoms of Wernicke’s and Alzheimer’s, including shared deficiencies of thiamine-dependent processes, cognitive decline and aberrant glucose metabolism.Reference Gibson, Hirsch, Cirio, Jordan, Fonzetti and Elder 99 Reference Zhang, Yang and Li 101 In vitro studies have since shown that in a state of acute thiamine insufficiency, neurons produce markers of stress, including reactive oxygen species, and begin apoptotic processes, implying a physiologic role for thiamine in neuronal function.Reference Ba 102 , Reference Wang, Wang, Fan, Shi, Ke and Luo 103 This has been corroborated by Zhang et alReference Zhang, Yang and Li 101 and othersReference Karuppagounder, Xu and Shi 104 who demonstrated that thiamine deficiency promoted β-secretase activity, Aβ accumulation and oxidative stress. In clinical trials, however, thiamine supplementation has not borne significant outcomes. An early systematic review did not identify a single trial with significant positive associations between thiamine and AD.Reference Rodríguez, Qizilbash, López-Arrieta and Rodríguez 98 Koh et al also demonstrated that thiamine deficiency lost its associations with dementia when subjects were otherwise well nourished.Reference Koh, Charlton and Walton 105 This may suggest that the initial correlations were in fact between malnourishment, in which thiamine deficiency is often concomitant, and AD.

Other B vitamins, including pyridoxine (B6), cobalamin (B12), folate (B9) and homocysteine, have also come under scrutiny for potential links to AD. Homocysteine is a metabolic precursor of the amino acids methionine and cysteine; however, before metabolism, homocysteine is a potent neurotoxin.Reference Obeid and Herrmann 106 Multiple studies have demonstrated a persistent correlation between dietary homocysteine and AD, with physiological levels exceeding 14 µmol/L estimated to double the risk of AD.Reference Baierle, Vencato and Oldenburg 67 , Reference Seshadri, Beiser and Selhub 107 Reference Li and Pratico 109 Further, in 3xTg transgenic mice, homocysteine was directly associated with cognitive decline, as well as promoting Aβ and tau pathologies.Reference Li, Chu, Barrero, Merali and Pratico 110

As an upstream metabolite, homocysteine levels do not necessarily have to be controlled by reducing dietary intake. Instead, increasing consumption of vitamins B6, B12 and folate can perpetuate the conversion of homocysteine into its amino acid products. Wang et al evidenced this by demonstrating that when vitamin B12 and folate levels were impaired, subjects exhibited a higher risk of AD (over a 3-year follow-up).Reference Wang, Wahlin, Basun, Fastbom, Winblad and Fratiglioni 111 Yet, the association has been repeatedly proven insignificant and current evidence on the role of these vitamins has been deemed insufficient.Reference Morris, Evans, Schneider, Tangney, Bienias and Aggarwal 112 Reference Nelson, Wengreen, Munger and Corcoran 114

Additional B vitamins including riboflavin (B2) and niacin/nicotinamide (B3) have also received attention owing to their metabolic roles in the brain.Reference Creegan, Hunt, McManus and Rainey-Smith 65 Niacin, in particular, has shown promise with high doses demonstrating some protective and even curative capacity in mouse models.Reference Green, Steffan and Martinez-Coria 115 , Reference Morris, Evans and Bienias 116 Green et al demonstrated that specific phosphorylated species of tau were reduced and proteins associated with microtubule stability were elevated upon exposure to a high dose of vitamin B3.Reference Green, Steffan and Martinez-Coria 115 This rescued cognition in their murine model (3xTg) raising the possibility of a curative treatment, rather than a typical arresting strategy. However, clinical trials have not demonstrated any tangible improvements to cognition from vitamin B3, particularly when given as supplements rather than in their natural form.Reference Phelan 117 , Reference Morris 118 The optimal dosage of vitamins is further controversial. Studies have often examined high doses, well in-excess of established guidelines. It has been suggested that this may exceed a therapeutic benefit, if any, and potentially expose patients to toxic side effects. The excretion of supplemental vitamins may also diminish a therapeutic efficacy. As such, a re-examination of the optimal form, dose and functional levels may be required.

Nutrients with antioxidant properties have also been an area of intense study. Oxidative damage, elevated production of reactive oxygen species and impeded clearance of oxidative stress markers are well recognized in AD neurons.Reference Akiyama, Barger and Barnum 19 , Reference Verdile, Keane and Cruzat 119 , Reference Wyss-Coray and Rogers 120 Furthermore, accumulating evidence suggests that this oxidative damage may precede the deposition of Aβ or tau.Reference Verdile, Keane and Cruzat 119 Accordingly, antioxidants have been proposed as a potential preventative and therapeutic strategy. Common dietary antioxidants include vitamin C, vitamin E, polyphenols and carotenes.Reference Luchsinger, Noble and Scarmeas 95 The Cache County Study involving over 3000 participants demonstrated that in combination, vitamins C and E supplements were associated with a lower incidence of AD.Reference Zandi, Anthony and Khachaturian 121 Several other studies of comparable scope demonstrated similar associations,Reference Luchsinger, Noble and Scarmeas 95 though randomized control trials have largely failed to demonstrate any therapeutic utility for antioxidants.Reference Polidori and Nelles 122 Rather, a double-blind trial by Galasko et al noted that cognitive decline appeared accelerated in AD subjects given a combination of antioxidants, in spite of diminished oxidative stress markers.Reference Galasko, Peskind and Clark 123 As with other vitamins, these inconsistencies may arise from the form of antioxidant under consideration, where supplements, as opposed to dietary antioxidants, may prove less effective; though further study is required to ascertain the validity of this supposition.

The most controversial debate in the realm of antioxidants has been relating to the minerals zinc and copper. With some variability, serum zinc deficiency has been repeatedly correlated with AD.Reference Tully, Snowdon and Markesbery 124 Reference Ventriglia, Brewer and Simonelli 127 The zinc cation (Zn2+) is integral to neural enzymes mediating clearance of Aβ, APP processing and the neutralization of reactive oxidative species, all of which imply a protective role for zinc in AD.Reference Nuttall and Oteiza 125 , Reference Watt, Whitehouse and Hooper 128 However, Zn2+ is often elevated in AD brains and has also been directly implicated in potentiating the neurotoxicity of Aβ and tau; further, by binding to Aβ, Zn2+ may promote oligomerization.Reference Watt, Whitehouse and Hooper 128 Reference Huang, Wu, Cao, Lang, Lu and Zhou 130 This inconsistency has brought the role of Zn2+ into dispute, and studies have speculated on a potential dual role in the context of AD.Reference Huang, Cuajungco, Atwood, Moir, Tanzi and Bush 131

However, it is more likely that Zn2+ levels are elevated in the brain as a physiological countermeasure against the oxidative and inflammatory stress of AD. This would decrease serum Zn2+ concentrations, as observed, but may elevate cerebral Zn2+ to toxic levels, perpetuating the observed pathological functions. This phenomenon, termed zinc flooding, implies that dietary zinc should be reduced, in spite of the observed zinc deficiency among Alzheimer’s patients. The copper ion (Cu2+) has similarly been implicated in AD, sharing much the same associations as Zn2+. Studies have shown that binding of Cu2+ to Aβ may lead to the creation of neurotoxic products and that it may have further roles in mediating the characteristic oxidative signature of AD.Reference Mathys and White 132 Reference Syme, Nadal, Rigby and Viles 134 However, as with zinc, Cu2+ deficiencies have also been linked to AD.Reference Xu, Church and Patassini 135 As reported by Quinn et al, these contradictions have led to the investigation of both copper-sequestering and copper-supplementing agents in clinical trials.Reference Quinn, Crane, Harris and Wadsworth 136

In general, inconsistencies are ubiquitous when considering the role of individual micronutrients in AD. It is likely that variations in dosing, disease stage and the form of nutrient may influence any perceived utility on AD risk or progression. Furthermore, few studies have attempted to consider any broad synergistic effects among differing nutrients, preferring instead to focus on a single cause-and-effect relationship. As diets seldom provide nutrients in isolation, traditional trials may fail to capture the inherent interactions among different dietary elements.

Dietary Patterns

Analysis of larger dietary patterns may alleviate some of the uncertainties raised by single-nutrient trials. In particular, they offer the ability to examine the role of nutrients in situ or as components of a larger system rather than distinct entities. Among the best studied has been the Mediterranean diet (MeDi). In studies of other diseases, the MeDi has demonstrated significant reductions in mortality and morbidity.Reference Martinez-Gonzalez and Bes-Rastrollo 137 , Reference Grosso, Mistretta and Frigiola 138 The MeDi may have benefit in AD as it features many nutrients suggested to have beneficial impacts on AD risk and progression. This includes a high intake of fruits and vegetables, rich in natural antioxidants and ω-3 unsaturated fatty acids (in the form of fresh fish and olive oil). Moreover, the diet is low in saturated fats, cholesterol, high-fat dairy products and red meats.Reference Huhn, Kharabian Masouleh, Stumvoll, Villringer and Witte 139 It therefore excludes many of the known foods associated with AD risk, and includes several protective nutrients.Reference Gu, Nieves, Stern, Luchsinger and Scarmeas 140 It is thus unsurprising that in numerous studies and meta-analyses, adherence to the MeDi was correlated with a reduced risk of AD.Reference Scarmeas, Stern, Tang, Mayeux and Luchsinger 141 Reference Sofi, Cesari, Abbate, Gensini and Casini 143 Scarmeas et al were further able to demonstrate that in addition to AD onset, the MeDi also reduced the risk of mortality after diagnosis.Reference Scarmeas, Luchsinger, Mayeux and Stern 144

Other diets, such as the “Dietary Approaches to Stop Hypertension” (DASH) diet were associated with improved cognition in hypertensive cohorts.Reference Smith, Blumenthal and Babyak 145 Observational studies have also correlated general diet quality to cognition in non-demented cohorts.Reference Correa Leite, Nicolosi, Cristina, Hauser and Nappi 146 , Reference Wengreen, Neilson, Munger and Corcoran 147 It is noteworthy that when attempting to resolve this effect, no single element of the diet could be independently associated with the purported benefits. Even supplementation of multiple discrete nutrients have proved clinically insignificant, as was the case in a recent trail of Souvenaid (branded Fortasyn Connect) which provided a preformulated combination ω-3 PUFAs, synaptic membrane precursors, cofactors and vitamins.Reference Ritchie, Bajwa and Coleman 148 , Reference Soininen, Solomon and Visser 149 Therefore, it is likely that a complex additive relationship may sustain the observed benefits of the MeDi and other diets.

Perspectives on the Microbiota

Although the human microbiota has recently emerged as a major contributor to various disease states, it remained largely unexplored in the field of neurology until the recognition of the gut-brain axis, which revealed the existence of complex bilateral communication between the brain and the intestines.Reference Carabotti, Scirocco, Maselli and Severi 150 To date, investigations have catalogued a variety of signaling molecules originating from the gut as modulators of pain, stress and possibly emotions.Reference Mayer, Tillisch and Gupta 151 Reference Cryan and Dinan 153 Murine models have also demonstrated significant behavioral and biochemical differences in animals raised with a sterile gut, which can be restored after colonization with probiotic bacteria.Reference Sampson and Mazmanian 154

The role of microbiota in AD is an emerging area of study. From preliminary data, we glean three gross mechanisms by which the microbiota may influence AD. The first involves the well-studied ability of the microbiota to affect nutrient absorption and activation.Reference Moco, Martin and Rezzi 155 Jandhyala et al report that absorption and activation of polyphenols, a class of organic antioxidants with protective associations in AD, is contingent on enteric microbial enzymes.Reference Jandhyala, Talukdar, Subramanyam, Vuyyuru, Sasikala and Nageshwar Reddy 156 Likewise, the absorption of ω-3 fatty acidsReference Menni, Zierer and Pallister 157 and the bioavailability of macronutrients may also depend on the state and composition of the microbiome.Reference Krajmalnik-Brown, Ilhan, Kang and DiBaise 158 It is therefore conceivable that the microbiota may mediate the physiological levels of the nutrients implicated in AD. Thus, the microbiota may cause identical quantities of administered nutrients to result in substantially different concentrations within the body. The failure of most trials to account for this may therefore offer some explanation for the inability of nutrient-based trials to demonstrate associations on AD. However, this remains speculative pending definitive quantification of the potential influence and variability imposed by the microbiota.

A second emerging mechanism lies in the capacity of the microbiota to modulate inflammation. The gut is known to be a source for various oxidative stressors and signals,Reference Hakansson and Molin 159 , Reference Boulange, Neves, Chilloux, Nicholson and Dumas 160 and recent data by Bruce-Keller et al and others have demonstrated that varying the microbiota can directly alter inflammation, both in the brain and systemically.Reference Bruce-Keller, Salbaum and Luo 161 , Reference Mu, Yang and Zhu 162 Studies have narrowed this tendency to specific bacterial species, which can secrete or otherwise generate pro-inflammatory cytokines; others may counteract this signaling by producing anti-inflammatory agents.Reference Hakansson and Molin 159 , Reference Boulange, Neves, Chilloux, Nicholson and Dumas 160 , Reference Mu, Yang and Zhu 162 Notably, it was microbes thriving in a high-fat environment that were directly associated with elevated neuroinflammation.Reference Bruce-Keller, Salbaum and Luo 161 Mice enriched with these bacteria further demonstrated significant behavioral changes in several domains, in comparison to healthy controls.Reference Bruce-Keller, Salbaum and Luo 161 As high fat consumption and inflammation are directly associated with AD, it is conceivable that the microbiota may, at least in part, moderate the pathogenic effects of fats in AD.

Data have also suggested a role for the microbiota in amyloidogenesis. Bacteria may perpetuate the leakage of dietary amyloids into the blood, where they may eventually enter the nervous system and seed further amyloid transformations.Reference Walker and Jucker 163 , Reference Pistollato, Sumalla Cano, Elio, Masias Vergara, Giampieri and Battino 164 The gut may therefore facilitate Aβ oligomerization in a manner similar to transmissible encephalopathies or prion diseases.Reference Walker and Jucker 163 However, this theory is again speculative and has yet to be proven in patients. It is also unlikely any pathogenic amyloids derived from diet may preserve their unique misfolded conformations as they pass through the gastrointestinal tract. Nonetheless, it is now known that in transgenic murine models (3xTg) of AD, a probiotic formula can slow cognitive decline and decrease amyloid plaques in the brain.Reference Bonfili, Cecarini and Berardi 165 A role for the microbiota in AD is therefore possible. In future, this should be a field of study, and trials, particularly in nutrition should begin to consider the state of the microbiota as a potential disrupting variable.

Alzheimer’s Disease, Drugs and Diet

We have thus far summarized how diet may mediate the onset and progression of AD. Although this has been the principal focus of nutritional studies, the reciprocal relationship, wherein AD may alter diet, is also an emerging area of investigation. As early as 1990, Mungas et al demonstrated that patients with dementia displayed significantly altered dietary preferences when compared with healthy controls.Reference Mungas, Cooper, Weiler, Gietzen, Franzi and Bernick 166 Specifically, they preferred high-fat and/or high-sugar foods, which may arise from diminished inhibition of cravings.Reference Mungas, Cooper, Weiler, Gietzen, Franzi and Bernick 166 More recent data suggest that various forms of dementia are associated with broader changes in appetite, food preference, eating habits and swallowing, with up to 80% of AD patients displaying some dietary disturbance (although data have varied heavily by form and stage of dementia considered).Reference Ikeda, Brown, Holland, Fukuhara and Hodges 167 Reference Cipriani, Carlesi, Lucetti, Danti and Nuti 170 These conclusions posit an intriguing scenario: AD may drive patients toward a diet that may worsen outcomes and prognoses. The fact that sizable fractions of AD cohorts are routinely observed with hyperphagia,Reference Hwang, Yang, Tsai and Liu 171 Reference Tsang, Keene and Hope 175 and poorer adherence to beneficial diets,Reference Gardener, Gu and Rainey-Smith 176 add suggestive evidence to this hypothesis.

An alternate scenario suggests that AD and dementia, particularly in its latter stages, may be associated with malnutrition and nutrient deficiencies.Reference Tombini, Sicari, Pellegrino, Ursini, Insarda and Di Lazzaro 177 , Reference Sun, Wen, Zhou and Ding 178 Both are near endemic among elderly and AD populations, and are well associated with deleterious outcomes and prognosis.Reference Sparre-Sorensen and Kristensen 179 , Reference Yildiz, Buyukkoyuncu Pekel, Kilic, Tolgay and Tufan 180 It is currently unclear whether AD may be more strongly associated with malnourishment or higher fat and sugar consumption, or a complex mix of both. However, given that both dietary states are known to impact AD, either case implies that AD and diet may exert a self-modifying interaction, wherein each may modulate the other.

The impact of these associations may also be significant in the context of drug metabolism. Nutrition has long shared associations to the efficacy and bioactivity of drugs, and it is well known that diet is an important determinant of drug metabolism.Reference Walter-Sack and Klotz 181 A review by Walter-Sack and Klotz summarized many known interactions, including changes in excretion, enzyme function and clearance that have been demonstrated by alterations in diet.Reference Walter-Sack and Klotz 181 In the context of AD, perhaps the most relevant observation is that a high-fat diet may moderate the clearance of drugs.Reference Walter-Sack and Klotz 181 Reference Gupta and Benet 183 Studies have also indicated that malnutrition may decrease drug efficacy by impeding requisite biotransformation and activation reactions.Reference Krishnaswamy 184 , Reference Speerhas 185 States of elevated inflammation may similarly hamper crucial drug activation and metabolism systems.Reference Morgan 186 , Reference Renton 187 As evidenced previously, inflammation as well as both high-fat diets and malnutrition are endemic in AD, and may be partly perpetuated by AD itself. Though the reported associations have been largely from isolated studies for unrelated disease and drug models—donepezil, rivastigmine and memantine—three major drugs used to manage AD symptoms, are all associated with gastrointestinal reactions and/or appetite changes.Reference Farlow, Grossberg, Sadowsky, Meng and Somogyi 188 Reference Popik, Kos, Zhang and Bisaga 190 As such, drugs and diet may conceivably interact in yet another multifaceted, reciprocal interaction, where drugs can modify diet and diet can in turn modify the efficacy of drugs. Although it is premature to know whether this interaction may have any role in AD, these relationships underlie a definitive web of dynamic interactions between diet, drugs and disease (Figure 1).

Diet in Alzheimer’s Clinical Trials

An ideal clinical trial would administer a singular intervention, in isolation, and discern the effect on a discrete therapeutic outcome. However, the complexity of AD inevitably means that numerous interceding factors may modulate outcome measures, irrespective of the action of a potential therapy. In cataloguing the interaction of diet and AD, this review has identified various dietary elements as potential intervening factors (Table 1).

Table 1 Summary of mechanistic interactions between major nutrients and AD pathologies

AD=Alzheimer’s disease; PUFAs=polyunsaturated fatty acids.

To the best of authors’ knowledge, the role of diet has not been systematically considered in the context of a non-nutritional therapeutic agent in AD. Although an array of extensive interactions has now been established, along with a potentially reciprocal self-modifying interactions, studies have largely relied on the control group to mitigate any variability induced by diet. Yet, in raising the variability of cohorts, studies can compromise statistical power, and hinder the observance of the efficacy of a therapy. We therefore suggest that trials in AD proactively consider strategies to account for diet in their study design and analysis.

Shim et al have reviewed approaches to assess diet in the context of epidemiology.Reference Shim, Oh and Kim 191 They note that diet may be tracked by objective observation or subjective reports;Reference Shim, Oh and Kim 191 however, in AD both may prove challenging. Alzheimer’s disease trials require extended follow-up to observe the manifestation of symptoms; thus consistent observations of diet by study personnel is impractical—although it should be considered the gold standard. Likewise, patients with advancing dementia cannot reliably be asked to track food intake or adhere to a prescribed diet. Caregivers may be a potential substitute for self-reporting, yet typical concerns about recall, bias and overburdening may be raised.

A more feasible strategy may be to track baseline dietary preferences in domains like fat, sugar, vitamin and mineral status. In secondary or sensitivity analyses, this data could stratify patients in the exposure and control cohorts into subgroups of comparable diet. Similar results could be attained by multivariate modeling for studies of sufficient size, and appropriate design. Alternatively, for pair-matched control trials, diet may be a factor of consideration when matching subjects and controls. Either strategy may strain study resources, and stratification is known to diminish statistical power. However, these measures may be performed as sensitivity analyses, and accompany primary data, rather than detract from it.

Studies should further consider substantial changes in these nutrients’ levels during the study period as a potential confounding interaction. For example, a dramatic rise of cholesterol, an ostensible Aβ pro-aggregate, or perturbations in micronutrients (including the use of nutritional supplements) may combat the efficacy of an anti-aggregative agent. Likewise, intra-study variations of ω-6 fatty acid levels, or the development of glucose/insulin dysregulation may alter neuroinflammation, thereby confounding anti-inflammatory therapies. Data on these parameters may serve to refine participant inclusion and exclusion criteria, or shape data analysis and the selection of control subjects as outlined previously.

Conclusions

A comprehensive analysis of the repeated failure of AD clinical trials must consider secondary factors which may diminish the apparent efficacy of these drugs. Since diet may be a major factor in regulating the onset and progression of AD, as well as influencing the ultimate bioavailability of drugs, we suggest that diet may be an important uncontrolled variable in conventional AD clinical trial design.

The role of diet in altering AD risk is well established. Nutrients including fats, sugars, minerals and vitamins have been conclusively associated with varying AD risk and disease progression via a range of mechanisms. Larger dietary patterns have also been definitively linked to AD risk, and the microbiota is emerging as a potent variable in this context. A reciprocal association in which AD modulates diet is also conceivable. Notably, the diets that AD predisposes are those linked with worse prognoses, and impeded drug metabolism. As such, a highly complex set of interactions between diet, AD, and drugs is evident.

This review cannot claim that the failure of experimental drugs in AD is attributable to diet; however, it does suggest that diet may play a role in the outcomes of AD, and the perceived clinical efficacy of novel agents. We therefore suggest that future studies, particularly clinical trials make a concerted effort to monitor, regulate or account for diet among AD patients. This may require significant investment of resources, and may burden patients and caregivers. Studies may also diminish statistical power depending on the mode of analysis undertaken. However, the extent of diet’s interactions with AD and its apparent role modulating outcomes justifies greater scrutiny in clinical trials.

Figure 1 Summary of the interactions of diet, drugs and Alzheimer’s disease (AD). Dietary nutrients (fats or sugars) may medicate inflammatory signaling, or contribute toward underlying metabolic states associated with AD. This interaction may be modified by the actions of the microbiota. Likewise, cholesterol and vitamins may interact directly with Aβ, contributing to AD risk. Antioxidants and drugs may counteract some of the exacerbating signaling, even though further reciprocal interactions among diet and nutritional status may affect their efficacy.

Acknowledgements

DFW acknowledges salary support from a Tier 1 Canada Research Chair.

Disclosures

The authors have no relevant financial disclosures.

Statement of Authorship

All authors contributed equally to the conception, design and drafting of this work.

References

1. Wong, SL, Gilmour, H, Ramage-Morin, PL. Alzheimer’s disease and other dementias in Canada. Health Rep. 2016;27(5):11-16.Google Scholar
2. Alzheimer’s Association. Alzheimer’s disease facts and figures. Alzheimers Dement. 2016;12(4):459-509.Google Scholar
3. Qiu, C, Kivipelto, M, von Strauss, E. Epidemiology of Alzheimer’s disease: occurrence, determinants, and strategies toward intervention. Dialogues Clin Neurosci. 2009;11(2):111-128.Google Scholar
4. Cummings, JL, Morstorf, T, Zhong, K. Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res Ther. 2014;6(4):37.Google Scholar
5. Becker, RE, Greig, NH, Giacobini, E. Why do so many drugs for Alzheimer’s disease fail in development? Time for new methods and new practices? J Alzheimers Dis. 2008;15(2):303-325.Google Scholar
6. White, L, Petrovitch, H, Ross, GW, Masaki, KH, Abbott, RD, Teng, EL, et al. Prevalence of dementia in older Japanese-American men in Hawaii: The Honolulu-Asia aging study. JAMA. 1996;276(12):955-960.Google Scholar
7. Hendrie, HC, Ogunniyi, A, Hall, KS, et al. Incidence of dementia and Alzheimer disease in 2 communities: Yoruba residing in Ibadan, Nigeria, and African Americans residing in Indianapolis, Indiana. JAMA. 2001;285(6):739-747.Google Scholar
8. Reitz, C, Mayeux, R. Genetics of Alzheimer’s disease in Caribbean Hispanic and African American populations. Biol Psychiatry. 2014;75(7):534-541.Google Scholar
9. Tang, MX, Cross, P, Andrews, H, et al. Incidence of AD in African-Americans, Caribbean Hispanics, and Caucasians in northern Manhattan. Neurology. 2001;56(1):49-56.10.1212/WNL.56.1.49Google Scholar
10. Grant, WB. Using multicountry ecological and observational studies to determine dietary risk factors for Alzheimer’s disease. J Am Coll Nutr. 2016;35(5):476-489.Google Scholar
11. Tanaka, H, Shimabukuro, T, Shimabukuro, M. High prevalence of metabolic syndrome among men in Okinawa. J Atheroscler Thromb. 2005;12(5):284-288.Google Scholar
12. Grant, WB. Trends in diet and Alzheimer’s disease during the nutrition transition in Japan and developing countries. J Alzheimers Dis. 2014;38(3):611-620.Google Scholar
13. Perl, DP. Neuropathology of Alzheimer’s disease. Mt Sinai J Med. 2010;77(1):32-42.Google Scholar
14. Nelson, PT, Braak, H, Markesbery, WR. Neuropathology and cognitive impairment in Alzheimer disease: a complex but coherent relationship. J Neuropathol Exp Neurol. 2009;68(1):1-14.10.1097/NEN.0b013e3181919a48Google Scholar
15. Farlow, MR. Etiology and pathogenesis of Alzheimer’s disease. Am J Health Syst Pharm. 1998;55(Suppl 2):S5-S10.Google Scholar
16. Swerdlow, RH. Pathogenesis of Alzheimer’s disease. Clin Interv Aging. 2007;2(3):347-359.Google Scholar
17. van der Kant, R, Goldstein, LS. Cellular functions of the amyloid precursor protein from development to dementia. Dev Cell. 2015;32(4):502-515.10.1016/j.devcel.2015.01.022Google Scholar
18. Murphy, MP, LeVine, H 3rd., Alzheimer’s disease and the amyloid-beta peptide. J Alzheimers Dis. 2010;19(1):311-323.10.3233/JAD-2010-1221Google Scholar
19. Akiyama, H, Barger, S, Barnum, S, et al. Inflammation and Alzheimer’s disease. Neurobiol Aging. 2000;21(3):383-421.10.1016/S0197-4580(00)00124-XGoogle Scholar
20. Iqbal, K, Liu, F, Gong, CX, Grundke-Iqbal, I. Tau in Alzheimer disease and related tauopathies. Curr Alzheimer Res. 2010;7(8):656-664.10.2174/156720510793611592Google Scholar
21. Helzner, EP, Scarmeas, N, Cosentino, S, Tang, MX, Schupf, N, Stern, Y. Survival in Alzheimer disease: a multiethnic, population-based study of incident cases. Neurology. 2008;71(19):1489-1495.10.1212/01.wnl.0000334278.11022.42Google Scholar
22. Wu, A, Ying, Z, Gomez-Pinilla, F. The interplay between oxidative stress and brain-derived neurotrophic factor modulates the outcome of a saturated fat diet on synaptic plasticity and cognition. Eur J Neurosci. 2004;19(7):1699-1707.10.1111/j.1460-9568.2004.03246.xGoogle Scholar
23. Murphy, T, Dias, GP, Thuret, S. Effects of diet on brain plasticity in animal and human studies: mind the gap. Neural Plast. 2014;2014:563160.Google Scholar
24. Gomez-Pinilla, F. Brain foods: the effects of nutrients on brain function. Nat Rev Neurosci. 2008;9(7):568-578.Google Scholar
25. Folch, J, Ettcheto, M, Petrov, D, et al. Review of the advances in treatment for Alzheimer disease: strategies for combating beta-amyloid protein. Neurologia. 2018;33(1):47-58.10.1016/j.nrl.2015.03.012Google Scholar
26. Bronzuoli, MR, Iacomino, A, Steardo, L, Scuderi, C. Targeting neuroinflammation in Alzheimer’s disease. J Inflamm Res. 2016;9:199-208.Google Scholar
27. DeCarolis, NA, Eisch, AJ. Hippocampal neurogenesis as a target for the treatment of mental illness: a critical evaluation. Neuropharmacology. 2010;58(6):884-893.Google Scholar
28. Vina, J, Lloret, A, Orti, R, Alonso, D. Molecular bases of the treatment of Alzheimer’s disease with antioxidants: prevention of oxidative stress. Mol Aspects Med. 2004;25(1-2):117-123.10.1016/j.mam.2004.02.013Google Scholar
29. Strittmatter, WJ, Saunders, AM, Schmechel, D, et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci U S A. 1993;90(5):1977-1981.Google Scholar
30. Strittmatter, WJ, Roses, AD. Apolipoprotein E and Alzheimer disease. Proc Natl Acad Sci U S A. 1995;92(11):4725-4727.Google Scholar
31. Xiong, H, Callaghan, D, Jones, A, et al. Cholesterol retention in Alzheimer’s brain is responsible for high beta- and gamma-secretase activities and Abeta production. Neurobiol Dis. 2008;29(3):422-437.10.1016/j.nbd.2007.10.005Google Scholar
32. Thirumangalakudi, L, Prakasam, A, Zhang, R, et al. High cholesterol-induced neuroinflammation and amyloid precursor protein processing correlate with loss of working memory in mice. J Neurochem. 2008;106(1):475-485.Google Scholar
33. Liu, Q, Zhang, J. Lipid metabolism in Alzheimer’s disease. Neurosci Bull. 2014;30(2):331-345.Google Scholar
34. Di Scala, C, Chahinian, H, Yahi, N, Garmy, N, Fantini, J. Interaction of Alzheimer’s beta-amyloid peptides with cholesterol: mechanistic insights into amyloid pore formation. Biochemistry. 2014;53(28):4489-4502.Google Scholar
35. Kakio, A, Nishimoto, SI, Yanagisawa, K, Kozutsumi, Y, Matsuzaki, K. Cholesterol-dependent formation of GM1 ganglioside-bound amyloid beta-protein, an endogenous seed for Alzheimer amyloid. J Biol Chem. 2001;276(27):24985-24990.10.1074/jbc.M100252200Google Scholar
36. Lin, FC, Chuang, YS, Hsieh, HM, et al. Early statin use and the progression of Alzheimer disease: a total population-based case-control study. Medicine (Baltimore). 2015;94(47):e2143.10.1097/MD.0000000000002143Google Scholar
37. Shepardson, NE, Shankar, GM, Selkoe, DJ. Cholesterol level and statin use in Alzheimer disease: II. Review of human trials and recommendations. Arch Neurol. 2011;68(11):1385-1392.Google Scholar
38. Kim, Y, Kim, C, Jang, HY, Mook-Jung, I. Inhibition of cholesterol biosynthesis reduces gamma-secretase activity and amyloid-beta generation. J Alzheimers Dis. 2016;51(4):1057-1068.Google Scholar
39. Mielke, MM, Zandi, PP, Shao, H, et al. The 32-year relationship between cholesterol and dementia from midlife to late life. Neurology. 2010;75(21):1888-1895.Google Scholar
40. Presecki, P, Muck-Seler, D, Mimica, N, et al. Serum lipid levels in patients with Alzheimer’s disease. Coll Antropol. 2011;35(Suppl 1):115-120.Google Scholar
41. Qin, Q, Liao, G, Baudry, M, Bi, X. Cholesterol perturbation in mice results in p53 degradation and axonal pathology through p38 MAPK and Mdm2 activation. PLoS One. 2010;5(4):e9999.10.1371/journal.pone.0009999Google Scholar
42. Mathews, ES, Appel, B. Cholesterol biosynthesis supports myelin gene expression and axon ensheathment through modulation of P13K/Akt/mTor signaling. J Neurosci. 2016;36(29):7628-7639.Google Scholar
43. Foot, M, Cruz, TF, Clandinin, MT. Influence of dietary fat on the lipid composition of rat brain synaptosomal and microsomal membranes. Biochem J. 1982;208(3):631-640.Google Scholar
44. Morris, MC, Evans, DA, Bienias, JL, et al. Dietary fats and the risk of incident Alzheimer disease. Arch Neurol. 2003;60(2):194-200.Google Scholar
45. Freeman, LR, Haley-Zitlin, V, Rosenberger, DS, Granholm, AC. Damaging effects of a high-fat diet to the brain and cognition: a review of proposed mechanisms. Nutr Neurosci. 2014;17(6):241-251.Google Scholar
46. Nakamura, MT, Cheon, Y, Li, Y, Nara, TY. Mechanisms of regulation of gene expression by fatty acids. Lipids. 2004;39(11):1077-1083.10.1007/s11745-004-1333-0Google Scholar
47. Lloyd, HM, Green, MW, Rogers, PJ. Mood and cognitive performance effects of isocaloric lunches differing in fat and carbohydrate content. Physiol Behav. 1994;56(1):51-57.Google Scholar
48. Julien, C, Tremblay, C, Phivilay, A, et al. High-fat diet aggravates amyloid-beta and tau pathologies in the 3xTg-AD mouse model. Neurobiol Aging. 2010;31(9):1516-1531.10.1016/j.neurobiolaging.2008.08.022Google Scholar
49. Takechi, R, Galloway, S, Pallebage-Gamarallage, MM, Lam, V, Mamo, JC. Dietary fats, cerebrovasculature integrity and Alzheimer’s disease risk. Prog Lipid Res. 2010;49(2):159-170.Google Scholar
50. Pugazhenthi, S, Qin, L, Reddy, PH. Common neurodegenerative pathways in obesity, diabetes, and Alzheimer’s disease. Biochim Biophys Acta. 2017;1863(5):1037-1045.Google Scholar
51. Engelhart, MJ, Geerlings, MI, Ruitenberg, A, et al. Diet and risk of dementia: does fat matter? The Rotterdam Study. Neurology. 2002;59(12):1915-1921.Google Scholar
52. Hooijmans, CR, Pasker-de Jong, PC, de Vries, RB, Ritskes-Hoitinga, M. The effects of long-term omega-3 fatty acid supplementation on cognition and Alzheimer’s pathology in animal models of Alzheimer’s disease: a systematic review and meta-analysis. J Alzheimers Dis. 2012;28(1):191-209.Google Scholar
53. Cole, GM, Ma, QL, Frautschy, SA. Omega-3 fatty acids and dementia. Prostaglandins Leukot Essent Fatty Acids. 2009;81(2-3):213-221.Google Scholar
54. Yurko-Mauro, K. Cognitive and cardiovascular benefits of docosahexaenoic acid in aging and cognitive decline. Curr Alzheimer Res. 2010;7(3):190-196.Google Scholar
55. Morris, MC, Evans, DA, Bienias, JL, et al. Consumption of fish and n-3 fatty acids and risk of incident Alzheimer disease. Arch Neurol. 2003;60(7):940-946.Google Scholar
56. Gu, Y, Schupf, N, Cosentino, SA, Luchsinger, JA, Scarmeas, N. Nutrient intake and plasma beta-amyloid. Neurology. 2012;78(23):1832-1840.Google Scholar
57. Olivera-Perez, HM, Lam, L, Dang, J, et al. Omega-3 fatty acids increase the unfolded protein response and improve amyloid-beta phagocytosis by macrophages of patients with mild cognitive impairment. FASEB J. 2017;31(10):4359-4369.Google Scholar
58. Siegel, G, Ermilov, E. Omega-3 fatty acids: benefits for cardio-cerebro-vascular diseases. Atherosclerosis. 2012;225(2):291-295.Google Scholar
59. Freund-Levi, Y, Hjorth, E, Lindberg, C, et al. Effects of omega-3 fatty acids on inflammatory markers in cerebrospinal fluid and plasma in Alzheimer’s disease: the OmegAD study. Dement Geriatr Cogn Disord. 2009;27(5):481-490.Google Scholar
60. Devore, EE, Grodstein, F, van Rooij, FJ, et al. Dietary intake of fish and omega-3 fatty acids in relation to long-term dementia risk. Am J Clin Nutr. 2009;90(1):170-176.Google Scholar
61. Arendash, GW, Jensen, MT, Salem, N Jr., et al. A diet high in omega-3 fatty acids does not improve or protect cognitive performance in Alzheimer’s transgenic mice. Neuroscience. 2007;149(2):286-302.Google Scholar
62. Barberger-Gateau, P, Samieri, C, Feart, C, Plourde, M. Dietary omega 3 polyunsaturated fatty acids and Alzheimer’s disease: interaction with apolipoprotein E genotype. Curr Alzheimer Res. 2011;8(5):479-491.Google Scholar
63. Araya-Quintanilla, F, Gutierrez-Espinoza, H, Sanchez-Montoya, U, et al. Effectiveness of omega-3 fatty acid supplementation in patients with Alzheimer disease: A systematic review and meta-analysis. Neurologia. 2017.Google Scholar
64. Mazereeuw, G, Lanctot, KL, Chau, SA, Swardfager, W, Herrmann, N. Effects of omega-3 fatty acids on cognitive performance: a meta-analysis. Neurobiol Aging. 2012;33(7):1482 e17-29.Google Scholar
65. Creegan, R, Hunt, W, McManus, A, Rainey-Smith, SR. Diet, nutrients and metabolism: cogs in the wheel driving Alzheimer’s disease pathology? Br J Nutr. 2015;113(10):1499-1517.Google Scholar
66. Otsuka, M, Yamaguchi, K, Ueki, A. Similarities and differences between Alzheimer’s disease and vascular dementia from the viewpoint of nutrition. Ann N Y Acad Sci. 2002;977:155-161.Google Scholar
67. Baierle, M, Vencato, PH, Oldenburg, L, et al. Fatty acid status and its relationship to cognitive decline and homocysteine levels in the elderly. Nutrients. 2014;6(9):3624-3640.Google Scholar
68. Amtul, Z, Uhrig, M, Wang, L, Rozmahel, RF, Beyreuther, K. Detrimental effects of arachidonic acid and its metabolites in cellular and mouse models of Alzheimer’s disease: structural insight. Neurobiol Aging. 2012;33(4):831 e21-831 e31.Google Scholar
69. Hosono, T, Nishitsuji, K, Nakamura, T, et al. Arachidonic acid diet attenuates brain Abeta deposition in Tg2576 mice. Brain Res. 2015;1613:92-99.Google Scholar
70. Tokuda, H, Kontani, M, Kawashima, H, Kiso, Y, Shibata, H, Osumi, N. Differential effect of arachidonic acid and docosahexaenoic acid on age-related decreases in hippocampal neurogenesis. Neurosci Res. 2014;88:58-66.Google Scholar
71. Otsuka, M. [Prevention of Alzheimer’s Disease and Nutrients]. Brain Nerve. 2016;68(7):809-817.Google Scholar
72. Gustaw-Rothenberg, K. Dietary patterns associated with Alzheimer’s disease: population based study. Int J Environ Res Public Health. 2009;6(4):1335-1340.10.3390/ijerph6041335Google Scholar
73. Crane, PK, Walker, R, Hubbard, RA, et al. Glucose levels and risk of dementia. N Engl J Med. 2013;369(6):540-548.Google Scholar
74. Luchsinger, JA, Tang, MX, Shea, S, Mayeux, R. Caloric intake and the risk of Alzheimer disease. Arch Neurol. 2002;59(8):1258-1263.Google Scholar
75. Mushtaq, G, Khan, JA, Kumosani, TA, Kamal, MA. Alzheimer’s disease and type 2 diabetes via chronic inflammatory mechanisms. Saudi J Biol Sci. 2015;22(1):4-13.10.1016/j.sjbs.2014.05.003Google Scholar
76. Moreira, PI. High-sugar diets, type 2 diabetes and Alzheimer’s disease. Curr Opin Clin Nutr Metab Care. 2013;16(4):440-445.Google Scholar
77. Akter, K, Lanza, EA, Martin, SA, Myronyuk, N, Rua, M, Raffa, RB. Diabetes mellitus and Alzheimer’s disease: shared pathology and treatment? Br J Clin Pharmacol. 2011;71(3):365-376.Google Scholar
78. Peila, R, Rodriguez, BL, Launer, LJ, Honolulu-Asia Aging, S. Type 2 diabetes, APOE gene, and the risk for dementia and related pathologies: The Honolulu-Asia Aging Study. Diabetes. 2002;51(4):1256-1262.Google Scholar
79. Ishii, K, Sasaki, M, Kitagaki, H, et al. Reduction of cerebellar glucose metabolism in advanced Alzheimer’s disease. J Nucl Med. 1997;38(6):925-928.Google Scholar
80. Iadecola, C. Sugar and Alzheimer’s disease: a bittersweet truth. Nat Neurosci. 2015;18(4):477-478.10.1038/nn.3986Google Scholar
81. Hoyer, S, Oesterreich, K, Wagner, O. Glucose metabolism as the site of the primary abnormality in early-onset dementia of Alzheimer type? J Neurol. 1988;235(3):143-148.Google Scholar
82. Wattamwar, PR, Mathuranath, PS. An overview of biomarkers in Alzheimer’s disease. Ann Indian Acad Neurol. 2010;13(Suppl 2):S116-S123.Google Scholar
83. Szablewski, L. Glucose transporters in brain: in health and in Alzheimer’s disease. J Alzheimers Dis. 2017;55(4):1307-1320.Google Scholar
84. Winkler, EA, Nishida, Y, Sagare, AP, et al. GLUT1 reductions exacerbate Alzheimer’s disease vasculo-neuronal dysfunction and degeneration. Nat Neurosci. 2015;18(4):521-530.Google Scholar
85. McNay, EC, Recknagel, AK. Brain insulin signaling: a key component of cognitive processes and a potential basis for cognitive impairment in type 2 diabetes. Neurobiol Learn Mem. 2011;96(3):432-442.Google Scholar
86. Chiu, SL, Cline, HT. Insulin receptor signaling in the development of neuronal structure and function. Neural Dev. 2010;5:7.Google Scholar
87. Akintola, AA, van Heemst, D. Insulin, aging, and the brain: mechanisms and implications. Front Endocrinol (Lausanne). 2015;6:13.Google Scholar
88. Hong, M, Lee, VM. Insulin and insulin-like growth factor-1 regulate tau phosphorylation in cultured human neurons. J Biol Chem. 1997;272(31):19547-19553.Google Scholar
89. El Khoury, NB, Gratuze, M, Papon, MA, Bretteville, A, Planel, E. Insulin dysfunction and Tau pathology. Front Cell Neurosci. 2014;8:22.Google Scholar
90. Planel, E, Tatebayashi, Y, Miyasaka, T, et al. Insulin dysfunction induces in vivo tau hyperphosphorylation through distinct mechanisms. J Neurosci. 2007;27(50):13635-13648.Google Scholar
91. Vekrellis, K, Ye, Z, Qiu, WQ, et al. Neurons regulate extracellular levels of amyloid beta-protein via proteolysis by insulin-degrading enzyme. J Neurosci. 2000;20(5):1657-1665.Google Scholar
92. Qiu, WQ, Walsh, DM, Ye, Z, et al. Insulin-degrading enzyme regulates extracellular levels of amyloid beta-protein by degradation. J Biol Chem. 1998;273(49):32730-32738.Google Scholar
93. Craft, S. Insulin resistance and Alzheimer’s disease pathogenesis: potential mechanisms and implications for treatment. Curr Alzheimer Res. 2007;4(2):147-152.Google Scholar
94. Mosconi, L, Pupi, A, De Leon, MJ. Brain glucose hypometabolism and oxidative stress in preclinical Alzheimer’s disease. Ann N Y Acad Sci. 2008;1147:180-195.Google Scholar
95. Luchsinger, JA, Noble, JM, Scarmeas, N. Diet and Alzheimer’s disease. Curr Neurol Neurosci Rep. 2007;7(5):366-372.Google Scholar
96. Huskisson, E, Maggini, S, Ruf, M. The role of vitamins and minerals in energy metabolism and well-being. J Int Med Res. 2007;35(3):277-289.Google Scholar
97. Butterworth, RF. Effects of thiamine deficiency on brain metabolism: implications for the pathogenesis of the Wernicke-Korsakoff syndrome. Alcohol Alcohol. 1989;24(4):271-279.Google Scholar
98. Rodríguez, J-L, Qizilbash, N, López-Arrieta, J, Rodríguez, J-L. Thiamine for Alzheimer’s disease. Cochrane Database of Systematic Reviews. 2001.Google Scholar
99. Gibson, GE, Hirsch, JA, Cirio, RT, Jordan, BD, Fonzetti, P, Elder, J. Abnormal thiamine-dependent processes in Alzheimer’s disease. Lessons from diabetes. Mol Cell Neurosci. 2013;55:17-25.Google Scholar
100. Gold, M, Hauser, RA, Chen, MF. Plasma thiamine deficiency associated with Alzheimer’s disease but not Parkinson’s disease. Metab Brain Dis. 1998;13(1):43-53.Google Scholar
101. Zhang, Q, Yang, G, Li, W, et al. Thiamine deficiency increases beta-secretase activity and accumulation of beta-amyloid peptides. Neurobiol Aging. 2011;32(1):42-53.Google Scholar
102. Ba, A. Metabolic and structural role of thiamine in nervous tissues. Cell Mol Neurobiol. 2008;28(7):923-931.Google Scholar
103. Wang, X, Wang, B, Fan, Z, Shi, X, Ke, ZJ, Luo, J. Thiamine deficiency induces endoplasmic reticulum stress in neurons. Neuroscience. 2007;144(3):1045-1056.Google Scholar
104. Karuppagounder, SS, Xu, H, Shi, Q, et al. Thiamine deficiency induces oxidative stress and exacerbates the plaque pathology in Alzheimer’s mouse model. Neurobiol Aging. 2009;30(10):1587-1600.Google Scholar
105. Koh, F, Charlton, KE, Walton, K, et al. Protein and thiamin intakes are not related to cognitive function in well-nourished community-living older adults. Nutr Diet. 2015;72(1):47-53.Google Scholar
106. Obeid, R, Herrmann, W. Mechanisms of homocysteine neurotoxicity in neurodegenerative diseases with special reference to dementia. FEBS Lett. 2006;580(13):2994-3005.Google Scholar
107. Seshadri, S, Beiser, A, Selhub, J, et al. Plasma homocysteine as a risk factor for dementia and Alzheimer’s disease. N Engl J Med. 2002;346(7):476-483.Google Scholar
108. Beydoun, MA, Beydoun, HA, Gamaldo, AA, Teel, A, Zonderman, AB, Wang, Y. Epidemiologic studies of modifiable factors associated with cognition and dementia: systematic review and meta-analysis. BMC Public Health. 2014;14:643.Google Scholar
109. Li, JG, Pratico, D. High levels of homocysteine results in cerebral amyloid angiopathy in mice. J Alzheimers Dis. 2015;43(1):29-35.Google Scholar
110. Li, JG, Chu, J, Barrero, C, Merali, S, Pratico, D. Homocysteine exacerbates beta-amyloid pathology, tau pathology, and cognitive deficit in a mouse model of Alzheimer disease with plaques and tangles. Ann Neurol. 2014;75(6):851-863.Google Scholar
111. Wang, HX, Wahlin, A, Basun, H, Fastbom, J, Winblad, B, Fratiglioni, L. Vitamin B(12) and folate in relation to the development of Alzheimer’s disease. Neurology. 2001;56(9):1188-1194.Google Scholar
112. Morris, MC, Evans, DA, Schneider, JA, Tangney, CC, Bienias, JL, Aggarwal, NT. Dietary folate and vitamins B-12 and B-6 not associated with incident Alzheimer’s disease. J Alzheimers Dis. 2006;9(4):435-443.Google Scholar
113. Doets, EL, van Wijngaarden, JP, Szczecinska, A, et al. Vitamin B12 intake and status and cognitive function in elderly people. Epidemiol Rev. 2013;35:2-21.Google Scholar
114. Nelson, C, Wengreen, HJ, Munger, RG, Corcoran, CD. Dietary folate, vitamin B-12, vitamin B-6 and incident Alzheimer’s disease: the cache county memory, health and aging study. J Nutr Health Aging. 2009;13(10):899-905.Google Scholar
115. Green, KN, Steffan, JS, Martinez-Coria, H, et al. Nicotinamide restores cognition in Alzheimer’s disease transgenic mice via a mechanism involving sirtuin inhibition and selective reduction of Thr231-phosphotau. J Neurosci. 2008;28(45):11500-11510.Google Scholar
116. Morris, MC, Evans, DA, Bienias, JL, et al. Dietary niacin and the risk of incident Alzheimer’s disease and of cognitive decline. J Neurol Neurosurg Psychiatry. 2004;75(8):1093-1099.Google Scholar
117. Phelan, MJ. Phase II clinical trial of nicotinamide for the treatment of mild to moderate Alzheimer’s disease. J Geriatr Med Gerontol. 2017;3:1.Google Scholar
118. Morris, MC. Diet and Alzheimer’s disease: what the evidence shows. MedGenMed. 2004;6(1):48.Google Scholar
119. Verdile, G, Keane, KN, Cruzat, VF, et al. Inflammation and oxidative stress: the molecular connectivity between insulin resistance, obesity, and Alzheimer’s disease. Mediators Inflamm. 2015;2015:105828.Google Scholar
120. Wyss-Coray, T, Rogers, J. Inflammation in Alzheimer disease: a brief review of the basic science and clinical literature. Cold Spring Harb Perspect Med. 2012;2(1):a006346.Google Scholar
121. Zandi, PP, Anthony, JC, Khachaturian, AS, et al. Reduced risk of Alzheimer disease in users of antioxidant vitamin supplements: the Cache County Study. Arch Neurol. 2004;61(1):82-88.Google Scholar
122. Polidori, MC, Nelles, G. Antioxidant clinical trials in mild cognitive impairment and Alzheimer’s disease – challenges and perspectives. Curr Pharm Des. 2014;20(18):3083-3092.Google Scholar
123. Galasko, DR, Peskind, E, Clark, CM, et al. Antioxidants for Alzheimer disease: a randomized clinical trial with cerebrospinal fluid biomarker measures. Arch Neurol. 2012;69(7):836-841.Google Scholar
124. Tully, CL, Snowdon, DA, Markesbery, WR. Serum zinc, senile plaques, and neurofibrillary tangles: findings from the Nun Study. Neuroreport. 1995;6(16):2105-2108.Google Scholar
125. Nuttall, JR, Oteiza, PI. Zinc and the aging brain. Genes Nutr. 2014;9(1):379.Google Scholar
126. Brewer, GJ, Kaur, S. Zinc deficiency and zinc therapy efficacy with reduction of serum free copper in Alzheimer’s disease. Int J Alzheimers Dis. 2013;2013:586365.Google Scholar
127. Ventriglia, M, Brewer, GJ, Simonelli, I, et al. Zinc in Alzheimer’s disease: a meta-analysis of serum, plasma, and cerebrospinal fluid studies. J Alzheimers Dis. 2015;46(1):75-87.Google Scholar
128. Watt, NT, Whitehouse, IJ, Hooper, NM. The role of zinc in Alzheimer’s disease. Int J Alzheimers Dis. 2010;2011:971021.Google Scholar
129. Miller, Y, Ma, B, Nussinov, R. Zinc ions promote Alzheimer Abeta aggregation via population shift of polymorphic states. Proc Natl Acad Sci U S A. 2010;107(21):9490-9495.Google Scholar
130. Huang, Y, Wu, Z, Cao, Y, Lang, M, Lu, B, Zhou, B. Zinc binding directly regulates tau toxicity independent of tau hyperphosphorylation. Cell Rep. 2014;8(3):831-842.Google Scholar
131. Huang, X, Cuajungco, MP, Atwood, CS, Moir, RD, Tanzi, RE, Bush, AI. Alzheimer’s disease, beta-amyloid protein and zinc. J Nutr. 2000;130(5S Suppl):1488S-1492SS.Google Scholar
132. Mathys, ZK, White, AR. Copper and Alzheimer’s disease. Adv Neurobiol. 2017;18:199-216.Google Scholar
133. Squitti, R, Siotto, M, Polimanti, R. Low-copper diet as a preventive strategy for Alzheimer’s disease. Neurobiol Aging. 2014;35(Suppl 2):S40-S50.Google Scholar
134. Syme, CD, Nadal, RC, Rigby, SE, Viles, JH. Copper binding to the amyloid-beta (Abeta) peptide associated with Alzheimer’s disease: folding, coordination geometry, pH dependence, stoichiometry, and affinity of Abeta-(1-28): insights from a range of complementary spectroscopic techniques. J Biol Chem. 2004;279(18):18169-18177.Google Scholar
135. Xu, J, Church, SJ, Patassini, S, et al. Evidence for widespread, severe brain copper deficiency in Alzheimer’s dementia. Metallomics. 2017;9(8):1106-1119.Google Scholar
136. Quinn, JF, Crane, S, Harris, C, Wadsworth, TL. Copper in Alzheimer’s disease: too much or too little? Expert Rev Neurother. 2009;9(5):631-637.Google Scholar
137. Martinez-Gonzalez, MA, Bes-Rastrollo, M. Dietary patterns, Mediterranean diet, and cardiovascular disease. Curr Opin Lipidol. 2014;25(1):20-26.Google Scholar
138. Grosso, G, Mistretta, A, Frigiola, A, et al. Mediterranean diet and cardiovascular risk factors: a systematic review. Crit Rev Food Sci Nutr. 2014;54(5):593-610.Google Scholar
139. Huhn, S, Kharabian Masouleh, S, Stumvoll, M, Villringer, A, Witte, AV. Components of a Mediterranean diet and their impact on cognitive functions in aging. Front Aging Neurosci. 2015;7:132.Google Scholar
140. Gu, Y, Nieves, JW, Stern, Y, Luchsinger, JA, Scarmeas, N. Food combination and Alzheimer disease risk: a protective diet. Arch Neurol. 2010;67(6):699-706.Google Scholar
141. Scarmeas, N, Stern, Y, Tang, MX, Mayeux, R, Luchsinger, JA. Mediterranean diet and risk for Alzheimer’s disease. Ann Neurol. 2006;59(6):912-921.Google Scholar
142. Scarmeas, N, Stern, Y, Mayeux, R, Luchsinger, JA. Mediterranean diet, Alzheimer disease, and vascular mediation. Arch Neurol. 2006;63(12):1709-1717.Google Scholar
143. Sofi, F, Cesari, F, Abbate, R, Gensini, GF, Casini, A. Adherence to Mediterranean diet and health status: meta-analysis. BMJ. 2008;337:a1344.Google Scholar
144. Scarmeas, N, Luchsinger, JA, Mayeux, R, Stern, Y. Mediterranean diet and Alzheimer disease mortality. Neurology. 2007;69(11):1084-1093.Google Scholar
145. Smith, PJ, Blumenthal, JA, Babyak, MA, et al. Effects of the dietary approaches to stop hypertension diet, exercise, and caloric restriction on neurocognition in overweight adults with high blood pressure. Hypertension. 2010;55(6):1331-1338.Google Scholar
146. Correa Leite, ML, Nicolosi, A, Cristina, S, Hauser, WA, Nappi, G. Nutrition and cognitive deficit in the elderly: a population study. Eur J Clin Nutr. 2001;55(12):1053-1058.Google Scholar
147. Wengreen, HJ, Neilson, C, Munger, R, Corcoran, C. Diet quality is associated with better cognitive test performance among aging men and women. J Nutr. 2009;139(10):1944-1949.Google Scholar
148. Ritchie, CW, Bajwa, J, Coleman, G, et al. Souvenaid(R): a new approach to management of early Alzheimer’s disease. J Nutr Health Aging. 2014;18(3):291-299.Google Scholar
149. Soininen, H, Solomon, A, Visser, PJ, et al. 24-month intervention with a specific multinutrient in people with prodromal Alzheimer’s disease (LipiDiDiet): a randomised, double-blind, controlled trial. Lancet Neurol. 2017;16(12):965-975.Google Scholar
150. Carabotti, M, Scirocco, A, Maselli, MA, Severi, C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 2015;28(2):203-209.Google Scholar
151. Mayer, EA, Tillisch, K, Gupta, A. Gut/brain axis and the microbiota. J Clin Invest. 2015;125(3):926-938.Google Scholar
152. Foster, JA, McVey Neufeld, KA. Gut-brain axis: how the microbiome influences anxiety and depression. Trends Neurosci. 2013;36(5):305-312.Google Scholar
153. Cryan, JF, Dinan, TG. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci. 2012;13(10):701-712.Google Scholar
154. Sampson, TR, Mazmanian, SK. Control of brain development, function, and behavior by the microbiome. Cell Host Microbe. 2015;17(5):565-576.Google Scholar
155. Moco, S, Martin, FP, Rezzi, S. Metabolomics view on gut microbiome modulation by polyphenol-rich foods. J Proteome Res. 2012;11(10):4781-4790.Google Scholar
156. Jandhyala, SM, Talukdar, R, Subramanyam, C, Vuyyuru, H, Sasikala, M, Nageshwar Reddy, D. Role of the normal gut microbiota. World J Gastroenterol. 2015;21(29):8787-8803.Google Scholar
157. Menni, C, Zierer, J, Pallister, T, et al. Omega-3 fatty acids correlate with gut microbiome diversity and production of N-carbamylglutamate in middle aged and elderly women. Sci Rep. 2017;7(1):11079.Google Scholar
158. Krajmalnik-Brown, R, Ilhan, ZE, Kang, DW, DiBaise, JK. Effects of gut microbes on nutrient absorption and energy regulation. Nutr Clin Pract. 2012;27(2):201-214.Google Scholar
159. Hakansson, A, Molin, G. Gut microbiota and inflammation. Nutrients. 2011;3(6):637-682.Google Scholar
160. Boulange, CL, Neves, AL, Chilloux, J, Nicholson, JK, Dumas, ME. Impact of the gut microbiota on inflammation, obesity, and metabolic disease. Genome Med. 2016;8(1):42.Google Scholar
161. Bruce-Keller, AJ, Salbaum, JM, Luo, M, et al. Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity. Biol Psychiatry. 2015;77(7):607-615.Google Scholar
162. Mu, C, Yang, Y, Zhu, W. Gut microbiota: the brain peacekeeper. Front Microbiol. 2016;7:345.Google Scholar
163. Walker, LC, Jucker, M. Neurodegenerative diseases: expanding the prion concept. Annu Rev Neurosci. 2015;38:87-103.Google Scholar
164. Pistollato, F, Sumalla Cano, S, Elio, I, Masias Vergara, M, Giampieri, F, Battino, M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev. 2016;74(10):624-634.Google Scholar
165. Bonfili, L, Cecarini, V, Berardi, S, et al. Microbiota modulation counteracts Alzheimer’s disease progression influencing neuronal proteolysis and gut hormones plasma levels. Sci Rep. 2017;7(1):2426.Google Scholar
166. Mungas, D, Cooper, JK, Weiler, PG, Gietzen, D, Franzi, C, Bernick, C. Dietary preference for sweet foods in patients with dementia. J Am Geriatr Soc. 1990;38(9):999-1007.Google Scholar
167. Ikeda, M, Brown, J, Holland, AJ, Fukuhara, R, Hodges, JR. Changes in appetite, food preference, and eating habits in frontotemporal dementia and Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2002;73(4):371-376.Google Scholar
168. Kai, K, Hashimoto, M, Amano, K, Tanaka, H, Fukuhara, R, Ikeda, M. Relationship between eating disturbance and dementia severity in patients with Alzheimer’s disease. PLoS One. 2015;10(8):e0133666.Google Scholar
169. Fernandez, M, Gobartt, AL, Balana, M, Group, CS. Behavioural symptoms in patients with Alzheimer’s disease and their association with cognitive impairment. BMC Neurol. 2010;10:87.Google Scholar
170. Cipriani, G, Carlesi, C, Lucetti, C, Danti, S, Nuti, A. Eating behaviors and dietary changes in patients with dementia. Am J Alzheimers Dis Other Demen. 2016;31(8):706-716.Google Scholar
171. Hwang, JP, Yang, CH, Tsai, SJ, Liu, KM. Behavioural disturbances in psychiatric inpatients with dementia of the Alzheimer’s type in Taiwan. Int J Geriatr Psychiatry. 1997;12(9):902-906.Google Scholar
172. Adebakin, A, Bradley, J, Gumusgoz, S, Waters, EJ, Lawrence, CB. Impaired satiation and increased feeding behaviour in the triple-transgenic Alzheimer’s disease mouse model. PLoS One. 2012;7(10):e45179.Google Scholar
173. Bhagavati, S. Marked hyperphagia associated with total loss of satiety in Alzheimer’s disease. J Neuropsychiatry Clin Neurosci. 2008;20(2):248-249.Google Scholar
174. Burns, A, Jacoby, R, Levy, R. Psychiatric phenomena in Alzheimer’s disease. IV: Disorders of behaviour. Br J Psychiatry. 1990;157:86-94.Google Scholar
175. Tsang, SW, Keene, J, Hope, T, et al. A serotoninergic basis for hyperphagic eating changes in Alzheimer’s disease. J Neurol Sci. 2010;288(1-2):151-155.Google Scholar
176. Gardener, S, Gu, Y, Rainey-Smith, SR, et al. Adherence to a Mediterranean diet and Alzheimer’s disease risk in an Australian population. Transl Psychiatry. 2012;2:e164.Google Scholar
177. Tombini, M, Sicari, M, Pellegrino, G, Ursini, F, Insarda, P, Di Lazzaro, V. Nutritional status of patients with Alzheimer’s disease and their caregivers. J Alzheimers Dis. 2016;54(4):1619-1627.Google Scholar
178. Sun, J, Wen, S, Zhou, J, Ding, S. Association between malnutrition and hyperhomocysteine in Alzheimer’s disease patients and diet intervention of betaine. J Clin Lab Anal. 2017;31(5):e22090.Google Scholar
179. Sparre-Sorensen, M, Kristensen, G. Alzheimer’s disease in the Danish malnutrition period 1999-2007. J Alzheimers Dis. 2015;48(4):979-985.Google Scholar
180. Yildiz, D, Buyukkoyuncu Pekel, N, Kilic, AK, Tolgay, EN, Tufan, F. Malnutrition is associated with dementia severity and geriatric syndromes in patients with Alzheimer disease. Turk J Med Sci. 2015;45(5):1078-1081.Google Scholar
181. Walter-Sack, I, Klotz, U. Influence of diet and nutritional status on drug metabolism. Clin Pharmacokinet. 1996;31(1):47-64.Google Scholar
182. Tan, KK, Trull, AK, Uttridge, JA, et al. Effect of dietary fat on the pharmacokinetics and pharmacodynamics of cyclosporine in kidney transplant recipients. Clin Pharmacol Ther. 1995;57(4):425-433.Google Scholar
183. Gupta, SK, Benet, LZ. High-fat meals increase the clearance of cyclosporine. Pharm Res. 1990;7(1):46-48.Google Scholar
184. Krishnaswamy, K. Drug metabolism and pharmacokinetics in malnourished children. Clin Pharmacokinet. 1989;17(Suppl 1):68-88.Google Scholar
185. Speerhas, R. Drug metabolism in malnutrition and obesity: clinical concerns. Cleve Clin J Med. 1995;62(1):73-75.Google Scholar
186. Morgan, ET. Impact of infectious and inflammatory disease on cytochrome P450-mediated drug metabolism and pharmacokinetics. Clin Pharmacol Ther. 2009;85(4):434-438.Google Scholar
187. Renton, KW. Regulation of drug metabolism and disposition during inflammation and infection. Expert Opin Drug Metab Toxicol. 2005;1(4):629-640.Google Scholar
188. Farlow, MR, Grossberg, GT, Sadowsky, CH, Meng, X, Somogyi, M. A 24-week, randomized, controlled trial of rivastigmine patch 13.3 mg/24 h versus 4.6 mg/24 h in severe Alzheimer’s dementia. CNS Neurosci Ther. 2013;19(10):745-752.Google Scholar
189. Yatabe, Y, Hashimoto, M, Kaneda, K, et al. Efficacy of increasing donepezil in mild to moderate Alzheimer’s disease patients who show a diminished response to 5 mg donepezil: a preliminary study. Psychogeriatrics. 2013;13(2):88-93.Google Scholar
190. Popik, P, Kos, T, Zhang, Y, Bisaga, A. Memantine reduces consumption of highly palatable food in a rat model of binge eating. Amino Acids. 2011;40(2):477-485.Google Scholar
191. Shim, JS, Oh, K, Kim, HC. Dietary assessment methods in epidemiologic studies. Epidemiol Health. 2014;36:e2014009.Google Scholar
Figure 0

Table 1 Summary of mechanistic interactions between major nutrients and AD pathologies

Figure 1

Figure 1 Summary of the interactions of diet, drugs and Alzheimer’s disease (AD). Dietary nutrients (fats or sugars) may medicate inflammatory signaling, or contribute toward underlying metabolic states associated with AD. This interaction may be modified by the actions of the microbiota. Likewise, cholesterol and vitamins may interact directly with Aβ, contributing to AD risk. Antioxidants and drugs may counteract some of the exacerbating signaling, even though further reciprocal interactions among diet and nutritional status may affect their efficacy.