You are on page 1of 33

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/51095382

Diet and Alzheimer's disease risk factors or prevention: The current evidence

Article  in  Expert Review of Neurotherapeutics · May 2011


DOI: 10.1586/ern.11.56 · Source: PubMed

CITATIONS READS

134 1,609

7 authors, including:

Vincenzo Solfrizzi Francesco Panza


Università degli Studi di Bari Aldo Moro Università degli Studi di Bari Aldo Moro
317 PUBLICATIONS   12,688 CITATIONS    434 PUBLICATIONS   17,585 CITATIONS   

SEE PROFILE SEE PROFILE

Vincenza Frisardi Davide Seripa


Azienda Ospedaliera Carlo Poma Mantova IRCCS Ospedale Casa Sollievo della Sofferenza
92 PUBLICATIONS   3,298 CITATIONS    277 PUBLICATIONS   8,728 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Mutated form of Human Nerve Growth Factor View project

This work was supported by grants from the Shanghai Hospital Development Center (No. SHDC12014221), Shanghai Municipal Commission of Health and Family
Planning, Key developing disciplines (2015ZB0501) View project

All content following this page was uploaded by Bruno P. Imbimbo on 06 June 2014.

The user has requested enhancement of the downloaded file.


THEMED ARTICLE y Alzheimer’s disease Review
For reprint orders, please contact reprints@expert-reviews.com

Diet and Alzheimer’s disease


risk factors or prevention:
the current evidence
Expert Rev. Neurother. 11(5), 677–708 (2011)

Vincenzo Solfrizzi1*, Preventing or postponing the onset of Alzheimer’s disease (AD) and delaying or slowing its
Francesco Panza†2*, progression would lead to a consequent improvement of health status and quality of life in older
Vincenza Frisardi1, age. Elevated saturated fatty acids could have negative effects on age-related cognitive decline
and mild cognitive impairment (MCI). Furthermore, at present, epidemiological evidence
Davide Seripa2,
suggests a possible association between fish consumption, monounsaturated fatty acids and
Giancarlo Logroscino3, polyunsaturated fatty acids (PUFA; in particular, n-3 PUFA) and a reduced risk of cognitive decline
Bruno P Imbimbo4 and and dementia. Poorer cognitive function and an increased risk of vascular dementia (VaD) were
Alberto Pilotto2 found to be associated with a lower consumption of milk or dairy products. However, the
1
Department of Geriatrics, Center for consumption of whole-fat dairy products may be associated with cognitive decline in the elderly.
Aging Brain, Memory Unit, University Light-to-moderate alcohol use may be associated with a reduced risk of incident dementia and
of Bari, Bari, Italy
AD, while for VaD, cognitive decline and predementia syndromes, the current evidence is only
2
Geriatric Unit and Gerontology-
Geriatric Research Laboratory, IRCCS suggestive of a protective effect. The limited epidemiological evidence available on fruit and
Casa Sollievo della Sofferenza, vegetable consumption and cognition generally supports a protective role of these macronutrients
San Giovanni Rotondo, Italy against cognitive decline, dementia and AD. Only recently, higher adherence to a Mediterranean-
3
Department of Neurological and
Psychiatric Sciences, University of Bari,
type diet was associated with decreased cognitive decline, although the Mediterranean diet
Bari, Italy (MeDi) combines several foods, micro- and macro-nutrients already separately proposed as
4
Research & Development potential protective factors against dementia and predementia syndromes. In fact, recent
Department, Chiesi Farmaceutici, prospective studies provided evidence that higher adherence to a Mediterranean-type diet could
Parma, Italy

Author for correspondence: be associated with slower cognitive decline, reduced risk of progression from MCI to AD, reduced
Geriatric Unit and Gerontology- risk of AD and a decreased all-cause mortality in AD patients. These findings suggested that
Geriatric Research Laboratory, IRCCS adherence to the MeDi may affect not only the risk of AD, but also of predementia syndromes
Casa Sollievo della Sofferenza, Viale
and their progression to overt dementia. Based on the current evidence concerning these factors,
Cappuccini 1, 71013 San Giovanni
Rotondo, Foggia, Italy no definitive dietary recommendations are possible. However, following dietary advice for
Tel.: +39 882 416 260 lowering the risk of cardiovascular and metabolic disorders, high levels of consumption of fats
Fax: +39 882 416 264 from fish, vegetable oils, nonstarchy vegetables, low glycemic index fruits and a diet low in
geriat.dot@geriatria.uniba.it
f.panza@operapadrepio.it
foods with added sugars and with moderate wine intake should be encouraged. Hopefully this
will open new opportunities for the prevention and management of dementia and AD.
*Both authors contributed equally to
this article Keywords : alcohol consumption • Alzheimer’s disease • dairy products • dementia • fruits • Mediterranean diet
adherence • mild cognitive impairment • MUFA • predementia syndromes • PUFA • vegetables

Dementia & late-life cognitive disorders clinicians use the term AD to refer to a clini-
Owing to aging populations, dementia and cal entity that typically presents with a char-
late-life cognitive disorders are reaching epi- acteristic progressive amnestic disorder with
demic proportions. In Western countries, the the subsequent appearance of other cognitive,
most common forms of dementia are Alzheimer’s behavioral and neuropsychiatric changes that
disease (AD) and vascular dementia (VaD), impair social function and activities of daily
with respective frequencies of 70% and 15% living [4] . The initial presentation can also be
of all dementias [1] . AD is a progressive neuro­ atypical, with nonamnestic focal cortical cogni-
degenerative illness that affects 5.3  million tive symptoms [5] . AD is both multifactorial and
people in the USA [2] , with >26 million patients heterogeneous, involving aberrant protein pro-
with AD worldwide, and an expected increase cessing, and is characterized by the presence of
to more than 106 million by 2050 [3] . Currently, both intra­neuronal protein clusters composed of

www.expert-reviews.com 10.1586/ERN.11.56 © 2011 Expert Reviews Ltd ISSN 1473-7175 677


Review Solfrizzi, Panza, Frisardi et al.

paired helical filaments of hyperphosphorylated tau protein (neu- by cognitive impairment resulting from a cerebrovascular etiology.
rofibrillary tangles [NFTs]) and extracellular protein aggregates The main categories of VCD are vascular cognitive impairment
(senile plaques [SPs]). The SPs are the result of the misprocessing (i.e., vascular cognitive impairment with no dementia and vascular
of the amyloid precursor protein (APP), a type-1 transmembrane MCI), VaD and mixed AD plus cerebrovascular disease (CVD),
protein, by b- and g-secretases to form a toxic b-amyloid (Ab) previously termed ‘mixed dementia’ [14,15] . In fact, dementia is more
peptide of 40–42 amino acids [6] that aggregates and initiates a likely to be present when vascular and AD lesions coexist, a situa-
pathogenic self-perpetuating cascade, ultimately leading to neu- tion that is especially common with increasing age [16] . Therefore,
ronal loss and dementia. According to the updated version of the particularly in late life, we should be interested in preventing the
‘amyloid cascade hypothesis’ [7] , the development of SPs is thought ‘dementia syndrome’ per se with the distinct components being
to precede and precipitate the formation of NFTs as a result of the neuropathologic changes of AD, vascular changes and possibly
the cellular changes invoked, and the oligomeric forms of Ab1–42 other neurodegenerative processes [17,18] .
are the main cause of neuronal death in AD [8,9] .
The term ‘predementia syndrome’ identified all conditions with Dietary & vascular-related factors in
age-related deficits in cognitive function reported in the literature, dementia syndromes
including a mild stage of cognitive impairment based on a nor- In the last decade, advances in understanding the neurobiology
mality model and pathological conditions considered predictive of AD have translated into an increase in clinical trials assess-
of early stages of dementia [10] . Therefore, this ‘umbrella term’ ing various potential AD treatments [19] . However, drugs cur-
includes different conditions and, among them, mild cognitive rently used for the treatment of AD produce limited clinical
impairment (MCI) is currently the most widely used term to benefit and do not treat the underlying causes of the disease [19] .
indicate nondemented aged persons with no significant disabil- Therefore, at present, prevention appears to still be a mandatory
ity and a mild memory or cognitive impairment that cannot be option. The causes of dementia and predementia syndromes are
explained by any recognized medical or psychiatric condition [11] . unknown; however, some studies have suggested that it may be
There is now ample evidence that MCI is often a pathology-based preventable  [20–24] . In fact, epidemiological evidence supported
condition with a high rate of progression to AD [10] . However, the hypothesis that modifiable vascular and lifestyle-related fac-
in population-based studies, MCI classification is less consistent tors were associated with the development of dementia and pre­
than in clinical series, suggesting that MCI is a heterogeneous dementia syndromes in late life, opening new avenues for the pre-
descriptor and that the outcome at follow-up depends on which vention of these diseases [21] . The vascular and related factors that
population is studied and how MCI is defined, with a quota of have been associated with dementia and cognitive decline include
MCI subjects remaining cognitively stable or improving after a hypertension and elevated blood pressure [25] , total cholesterol [26] ,
brief follow-up [10] . Very recently, with the advances in the use diabetes mellitus [27] , BMI [27] and the metabolic syndrome [28] .
of reliable biomarkers of AD that provide in vivo evidence of the In addition, the presence of cardiovascular and other chronic
disease, new research criteria that reconceptualize the diagnosis diseases [29] , depression [30] and low levels of physical activity [21]
around both a specific pattern of cognitive changes and struc- have been identified as risk factors for cognitive decline. By con-
tural/biological evidence of AD pathology were proposed [12] . trast, physical exercise [21,31] and education [21,32] appear to have
The International Working Group for New Research Criteria clear protective effects on cognitive functioning and a possible
for the Diagnosis of AD proposed a common lexicon as a point reduction in incident dementia. For the latter, whether this is due
of reference for the clinical and research communities. The to education per se or related to lifetime occupation and/or level
corner­stone of this lexicon was to consider AD solely as a clini- of cognitive exercise is debatable [32] .
cal and symptomatic entity that encompasses both predementia Since several dietary factors affect the risk of cardiovascular
and dementia phases [13] . In this new lexicon, the term MCI is disease, it can be assumed that they also influence the risk of
applied to individuals with measurable MCI in the absence of a dementia [20,21,33–38] . This concept is further supported by recent
significant effect on instrumental activities of daily living. This evidence that certain diets have been associated with a lower
diagnostic label was applied if there was no disease to which MCI incidence of AD. In fact, recent findings demonstrate that main-
can be attributed. It remained a term of exclusion for individu- taining a healthy diet may impact on many of these possible risk
als who were suspected to have but did not meet the proposed factors for cognitive decline and the model to follow seems to be
new research criteria for AD [12] , in that they deviated from the the Mediterranean diet (MeDi) [20] . The typical dietary pattern
clinicobiological phenotype of prodromal AD because they had of MeDi is characterized by a high intake of vegetables, fruits
memory symptoms that were not characteristic of AD or because and nuts, legumes, cereals, fish and monounsaturated fatty acids
they were biomarker negative [13] . (MUFA); relatively low intakes of meat and dairy products; and
The clinical presentation of VaD varies greatly depending on moderate consumption of alcohol. In fact, higher levels of con-
the causes and location of cerebral damage. Given the definitional sumption of olive oil, very rich in MUFA, are considered the
heterogeneity of VaD, the term vascular cognitive disorder (VCD) hallmark of the traditional MeDi, and some recent studies have
has been proposed by Sachdev [14] and it would become the global suggested that dietary fatty acids, particularly high MUFA intake
diagnostic category for cognitive impairment of vascular origin [15] . and regular fish and n-3 polyunsaturated fatty acid (PUFA) con-
VCD includes the group of syndromes and diseases characterized sumption, may play a role in the prevention of cognitive decline

678 Expert Rev. Neurother. 11(5), (2011)


Diet & Alzheimer’s disease Review

associated with aging or dementia [39–42] . Elevated dietary MUFA (i.e., fish, unsaturated fatty acids [UFA], antioxidants, such as
and PUFA n-3 and high fish consumption, alongside high levels vitamin E, vitamin B12, folates, carotenes, flavonoids and moder-
of antioxidants from fruit and vegetables [43,44] , and moderate ate alcohol) may have potential protective effect against demen-
alcohol consumption [45,46] may have a beneficial effect on the tia or cognitive decline  [20,52] . In the last few years, the study
risk of dementia. approach has been to associate single micro- or macro-nutrients
In this article, we examine the possible role of macronutrients to MCI or AD. In this picture, several hallmarks of the MeDi
and food nutrients, with a particular focus on the MeDi and the were linked to an increased risk or a protective effect against
adherence to the Mediterranean dietary pattern in modulating cognitive impairment and AD [20,53,54] .
the risk of AD and dementia, as well as the possible mechanisms
behind the observed associations, from English literature pub- Dietary fatty acids, fish consumption & dementia
lished before January 2011. We review clinical and epidemio- Fatty acids can be categorized briefly into saturated fatty acids
logical studies from the international literature, including both (SFA) and UFA. SFA, such as stearic acid, are present in prod-
cross-sectional and longitudinal studies that provided a descrip- ucts such as meat, dairy products, cookies and pastries. MUFA
tion of the diagnostic criteria used for predementia or demen- are most frequently consumed in olive oil. The principal series
tia syndromes. We searched through the PubMed database of of PUFA are n-6 (i.e., linoleic acid) and n-3 (i.e., a-linolenic
NCBI (available at [301]) by author and the following keywords: acid, docosahexaenoic acid [DHA] and eicosapentaenoic acid
Mediterranean, diet, Mediterranean dietary pattern, adherence, [EPA]). In the Mediterranean dietary pattern, the main sources
MUFA, PUFA, alcohol consumption, cereals, fruits, vegetables, of n-6 PUFA are vegetable oils, while the principal sources of n-3
dairy products, mild cognitive impairment, dementia, Alzheimer’s PUFA are fatty fish (salmon, tuna and mackerel). In fact, olive oil
disease, predementia syndromes and MCI. contains 70–80% MUFA (oleic acid) and 8–10% PUFA (6–7%
linoleic acid and 1–2% a-linolenic acid) [39,41,42] .
Dietary macronutrients & AD An increasing number of cross-sectional and longitudinal
One of the most intriguing and appealing links hypothesized in epidemio­logical and clinical studies have addressed the link
recent years is the association between lifestyle factors, such as diet between UFA intake and cognitive function [39,41,42,55–65] . Future
and dietary habits, and the occurrence of AD [33,35] . Deficiencies studies on this topic will address different constructs of prede-
of some micronutrients (especially vitamins B1, B2, B6, B12, C mentia syndromes and subtypes of MCI, while also accounting
and folate) have been described quite frequently in elderly people for the possible role of the apolipoprotein E (APOE) e4 allele in
and found to be significantly associated with cognitive impair- modifying the possible associations between dietary fatty acids
ment [33] . However, the role of the diet in cognitive decline has and cognitive decline [41,42] . As discussed previously, MUFA rep-
not been extensively investigated, with few data available on the resented the most important fat in the MeDi as a consquence of
role of macronutrient intake in the pathogenesis of predementia the high consumption of extra-virgin olive oil. In US diets, a major
and dementia syndromes [20,21,33–38] . In this article, we focus on source of MUFA is canola oil, while olive oil consumption is much
the possible role of macronutrients from the MeDi in protecting less frequent. Cumulative evidence suggests that extra-virgin olive
against AD. The MeDi, from the south of Europe, represents the oil may have a role in the protection against cognitive decline, as
dietary pattern usually consumed among the populations border- well as against coronary artery disease (CAD) and several types
ing the Mediterranean sea, and it has been widely reported to be a of cancer, because of its high levels MUFA and polyphenolic com-
model of healthy eating for its contribution to a favorable health pounds [39,42] . Recently, in the Three-City (3C) Study, olive oil
status and a better quality of life [302] . Since the first data from consumption habits were significantly associated with selective
Keys from the Seven Countries’ Study in the 1950s to 1960s [47] , cognitive deficit and cognitive decline, independently of other
several studies in different populations have established a benefi- dietary intakes and after adjusting for potential confounders.
cial role for the main components of the MeDi on the occurrence Intensive use of olive oil in diet is associated with lower odds of
of cardiovascular diseases and chronic degenerative diseases [48,49] . cognitive deficit in visual memory and verbal fluency, even if the
The Mediterranean-style diet is characterized by abundant plant relationship is not significant when assessing global cognitive func-
food consumption in the form of fruits, vegetables, breads, other tioning with the Mini-Mental State Examination (MMSE) [65] .
forms of cereals, potatoes, beans, nuts and seeds; fresh fruit as In the analyses of 732 participants of the European Prospective
the typical dessert; olive oil as the main source of MUFA; dairy Investigation into Cancer and nutrition (EPIC)-Greece cohort,
products as principally cheese and yogurt; a low-to-moderate intake of olive oil, MUFA and SFA exhibited a weak, but not sig-
consumption of fish depending on the proximity of the sea; a nificant, positive association with cognitive function as assessed
low-to moderate consumption of poultry; fewer than four eggs by the MMSE, 6–13 years after enrolment [62] .
consumed per week; and low-to-moderate amounts of red meat The possible relationship between dietary fatty acids intake
and wine consumed, normally during meals [50] . However, there and risk of dementia and AD has also been evaluated in a series
is no single MeDi, but several definitions, because dietary habits of longitudinal studies with contrasting findings (Table 1) [66–73] .
vary considerably across the Mediterranean countries bordering However, the sum of evidence suggested that an increase of SFA
the sea [51] . Previous observational studies already indicated that could have negative effects on cognitive functions and incident
specific foods or nutrients that take part in the traditional MeDi dementia, while a clear reduction of risk for cognitive decline has

www.expert-reviews.com 679
Table 1. Principal longitudinal, clinical and epidemiological studies on the relationships among dietary fatty acids, fish consumption and

680
dementia (i.e., Alzheimer’s disease and vascular dementia) or predementia syndromes (i.e., age-related cognitive decline and mild
cognitive impairment).
Review

Study (year) Setting and Subjects Dietary Cognitive outcomes Results and conclusions Ref.
study design assessment
(duration)
Dietary fatty acids, fish consumption & predementia syndromes
Kalmijn et al. (1997) Longitudinal, 476 subjects, aged Evaluation of dietary Cognitive impairment defined as a High linoleic acid intake (PUFA) was [57]
The Zutphen Elderly population-based 69–89 years old intake with the MMSE score <25 points and cognitive positively associated with cognitive
Study, The (3 years) cross-check dietary decline as a drop of >2 points of MMSE impairment. High fish consumption was
Netherlands history method over a 3-year period inversely associated with cognitive
impairment
Morris et al. (2004) Longitudinal, 2560 subjects, Evaluation of dietary Cognitive change at 3-year and 6-year A diet high in saturated and trans- [58]
The Chicago Health population-based aged 65 years and intake with a follow-ups measured with the EBMT of unsaturated fat, or low in nonhydrogenated
and Aging Project (6 years) older 139-item FFQ Immediate and Delayed Recall, the unsaturated fats, may be associated with
Solfrizzi, Panza, Frisardi et al.

(CHAP), USA MMSE and the SDMT cognitive decline among older people
Morris et al. (2005) Longitudinal, 3718 subjects, Evaluation of dietary Cognitive change at 3-year and 6-year Dietary intake of fish was inversely [75]
CHAP, USA population-based aged 65 years and intake with a follow-ups measured with the EBMT of associated with a cognitive decline over
(6 years) older 139-item FFQ Immediate and Delayed Recall, the 6 years
MMSE and the SDMT There were no consistent associations with
the n-3 fatty acids, although the effect
estimates were in the direction of slower
decline
Solfrizzi et al. (2006) Longitudinal, 278 subjects, Evaluation of MUFA MMSE High MUFA, PUFA and total energy intake [60]
The Italian population-based 65–84 years old, and PUFA dietary were significantly associated with a better
Longitudinal Study (8.5 years) from a cohort of intake with a cognitive performance in time. The
on Aging (ILSA) 5632 subjects 77-item FFQ association between high MUFA and PUFA
Italy intake and cognitive performance remained
robust even after adjustment for potential
confounding variables, such as age, sex,
educational level, CCI, BMI and total energy
intakes
Solfrizzi et al. (2006) Longitudinal, 278 subjects, Evaluation of MUFA Incident MCI. Diagnostic criteria for Dietary fatty acids intakes were not [61]
ILSA, Italy population-based 65–84 years old, and PUFA dietary MCI: 1.5 SDs below age- and associated with incident MCI. However,
(2.6 years) from a cohort of intake with a education-adjusted mean on the MMSE high PUFA intake appeared to have
5632 subjects 77-item FFQ and 10th percentile below age- and borderline nonsignificant trend for a
education-adjusted mean on memory protective effect against the development
test, without SMC and intact ADL/IADL of MCI that may be important
AD: Alzheimer’s disease; ADL: Activities of daily living; APOE: Apolipoprotein E; CCI: Charlson Comorbidity Index; CFT: Category Fluency Test (semantic memory); DSM-III-R: Diagnostic and Statistical Manual of
Mental Disorders, third edition (revised); DSM-IV: Diagnostic and Statistical Manual of Mental Disorders, fourth edition; EBMT: East Boston Memory Test (immediate and delayed episodic memory); FFQ: Food
Frequency Questionnaire; IADL: Instrumental activities of daily living; LDST: Letter Digit Substitution Test (perceptual–motor speed); MCI: Mild cognitive impairment; MeDi: Mediterranean diet; MMSE: Mini-Mental
State Examination (global cognitive functioning); MUFA: Monounsaturated fatty acids; NINCDS–ADRDA: National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer’s Disease and
Related Disorders Association; NINCDS–AIREN: National Institute of Neurological and Communicative Disorders and Stroke and the Association Internationale pour la Recherche at l’Enseignement en Neurosciences;
PMSQ: Pfeiffer’s Mental State Questionnaire (global cognitive functioning); PPBt: Purdue Peg Board task (psychomotor speed); PUFA: Polyunsaturated fatty acids; SDMT: Symbol Digit Modalities Test
(perceptual–motor speed); SFA: Saturated fatty acids; SMC: Subjective memory complaint; VaD: Vascular dementia.

Expert Rev. Neurother. 11(5), (2011)


Table 1. Principal longitudinal, clinical and epidemiological studies on the relationships among dietary fatty acids, fish consumption and
dementia (i.e., Alzheimer’s disease and vascular dementia) or predementia syndromes (i.e., age-related cognitive decline and mild
cognitive impairment).
Study (year) Setting and Subjects Dietary Cognitive outcomes Results and conclusions Ref.
study design assessment
(duration)

www.expert-reviews.com
Dietary fatty acids, fish consumption & predementia syndromes
Psaltopoulou et al. Longitudinal, 732 subjects, Evaluation of dietary MMSE No significant association between MeDi [62]
(2008), European population-based 60 years or older intakes with a score and MMSE scores, whereas a
Prospective (median 8 years) 150-item FFQ. statistically significant inverse association
Investigation into A dietary composite was found between MMSE performance
Cancer and Nutrition score (MeDi score) and some individual dietary components,
(EPIC), Greece evaluated adherence such as seed oil or PUFA intake
to MeDi
Eskilinen et al. (2008) Longitudinal, 1449 subjects, Evaluation of dietary The Mayo Clinic AD Research Center Elevated SFA intake at midlife was [63]
Cardiovascular Risk population-based aged 65–80 years intakes with a criteria were applied for diagnosing associated with poorer global cognitive
Factors Aging and (21 years) old 208-item FFQ MCI; MMSE, CFT, PPBt, LDST, episodic function and prospective memory and with
Dementia (CAIDE) memory with immediate word recall an increased risk of MCI. High intake of
Finland tests; executive function with the PUFA was associated with better semantic
Stroop test; and prospective memory memory. In addition, frequent fish
with a task by Einstein consumption was associated with better
global cognitive function and semantic
memory. Furthermore, higher PUFA:SFA
ratio was associated with better
psychomotor speed and executive function
Dietary fatty acids, fish consumption & dementia syndromes
Engelhart et al. (2002) Longitudinal, 5395 subjects, Evaluation of dietary Diagnosis of dementia (DSM-III-R High intakes of total fat, saturated fat, trans [67]
The Rotterdam Study, population-based aged 55 years intakes with a criteria), AD (NINCDS–ADRDA criteria), fat and cholesterol, and low intake of
The Netherlands (6 years) and older 100-item FFQ and VaD (NINCDS–AIREN criteria) MUFA, PUFA, n-6 PUFA and n-3 PUFA were
not associated with an increased risk of
dementia, AD or VaD
Luchsinger et al. Longitudinal, 980 subjects, Evaluation of dietary Diagnosis of prevalent dementia Higher intake of calories and fats may be [68]
(2002) population-based mean age: intake with a 61-item (DSM-IV criteria) and incident AD associated with higher risk of AD in
Washington Heights- (4 years) 75.3 ± 5.8 years FFQ (NINCDS–ADRDA criteria) subjects carrying the APOE e4 allele
Inwood Columbia old
Aging Project, USA
AD: Alzheimer’s disease; ADL: Activities of daily living; APOE: Apolipoprotein E; CCI: Charlson Comorbidity Index; CFT: Category Fluency Test (semantic memory); DSM-III-R: Diagnostic and Statistical Manual of
Diet & Alzheimer’s disease

Mental Disorders, third edition (revised); DSM-IV: Diagnostic and Statistical Manual of Mental Disorders, fourth edition; EBMT: East Boston Memory Test (immediate and delayed episodic memory); FFQ: Food
Frequency Questionnaire; IADL: Instrumental activities of daily living; LDST: Letter Digit Substitution Test (perceptual–motor speed); MCI: Mild cognitive impairment; MeDi: Mediterranean diet; MMSE: Mini-Mental
State Examination (global cognitive functioning); MUFA: Monounsaturated fatty acids; NINCDS–ADRDA: National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer’s Disease and
Related Disorders Association; NINCDS–AIREN: National Institute of Neurological and Communicative Disorders and Stroke and the Association Internationale pour la Recherche at l’Enseignement en Neurosciences;
PMSQ: Pfeiffer’s Mental State Questionnaire (global cognitive functioning); PPBt: Purdue Peg Board task (psychomotor speed); PUFA: Polyunsaturated fatty acids; SDMT: Symbol Digit Modalities Test
(perceptual–motor speed); SFA: Saturated fatty acids; SMC: Subjective memory complaint; VaD: Vascular dementia.
Review

681
Table 1. Principal longitudinal, clinical and epidemiological studies on the relationships among dietary fatty acids, fish consumption and

682
dementia (i.e., Alzheimer’s disease and vascular dementia) or predementia syndromes (i.e., age-related cognitive decline and mild
cognitive impairment).
Review

Study (year) Setting and Subjects Dietary Cognitive outcomes Results and conclusions Ref.
study design assessment
(duration)
Dietary fatty acids, fish consumption & dementia syndromes
Morris et al. (2003) Longitudinal, 815 subjects, Evaluation of dietary Incident diagnosis of AD Higher intake of n-3 PUFA and weekly fish [70]
Chicago Health and population-based aged 65 years intake with a 154-item (NINCDS–ADRDA criteria) consumption may reduce the risk of
Aging Project (3.9 years) and older FFQ incident AD
(CHAP), USA
Huang et al. (2005) Longitudinal, 5201 Evaluation of dietary Incident diagnosis of dementia Fatty fish, such as tuna or ‘other fish’, was [78]
Cardiovascular Health population-based participants, intake with a 99-item (DSM-IV criteria) and AD associated with a lower risk of developing
Cognition Study (5.4 years) aged 65 years FFQ (NINCDS–ADRDA criteria) dementia and AD with a dose–response
(CHCS), USA and older relationship, whereas lean, fried fish was
Solfrizzi, Panza, Frisardi et al.

not. Those without an APOE e4 allele had a


35–45% lower risk with consumption of
fatty fish, whereas there was little or no
difference for APOE e4 allele carriers
Laitinen et al., (2006) Longitudinal, 1449 subjects, Evaluation of dietary Incident diagnosis of AD Moderate intake of PUFA at midlife was [72]
Cardiovascular Risk population-based aged 65–80 years intake with a 154-item (NINCDS–ADRDA criteria) protective, whereas a moderate intake of
Factors Aging and (21 years) FFQ SFA may increase the risk of dementia and
Dementia (CAIDE), AD, especially among APOE e4 carriers
Finland
Barbeger-Gateau et al. Longitudinal, 9294 subjects, Evaluation of dietary Incident diagnosis of dementia Frequent consumption of fruits and [73]
(2007) population-based aged 65 years intakes with a FFQ (DSM-IV criteria) and AD vegetables, fish and n-3-rich oils may
Three-City Study, (4 years) and older (NINCDS–ADRDA criteria) decrease the risk of dementia and AD,
France especially among APOE e4 noncarriers

AD: Alzheimer’s disease; ADL: Activities of daily living; APOE: Apolipoprotein E; CCI: Charlson Comorbidity Index; CFT: Category Fluency Test (semantic memory); DSM-III-R: Diagnostic and Statistical Manual of
Mental Disorders, third edition (revised); DSM-IV: Diagnostic and Statistical Manual of Mental Disorders, fourth edition; EBMT: East Boston Memory Test (immediate and delayed episodic memory); FFQ: Food
Frequency Questionnaire; IADL: Instrumental activities of daily living; LDST: Letter Digit Substitution Test (perceptual–motor speed); MCI: Mild cognitive impairment; MeDi: Mediterranean diet; MMSE: Mini-Mental
State Examination (global cognitive functioning); MUFA: Monounsaturated fatty acids; NINCDS–ADRDA: National Institute of Neurological and Communicative Disorders and Stroke and the Alzheimer’s Disease and
Related Disorders Association; NINCDS–AIREN: National Institute of Neurological and Communicative Disorders and Stroke and the Association Internationale pour la Recherche at l’Enseignement en Neurosciences;
PMSQ: Pfeiffer’s Mental State Questionnaire (global cognitive functioning); PPBt: Purdue Peg Board task (psychomotor speed); PUFA: Polyunsaturated fatty acids; SDMT: Symbol Digit Modalities Test
(perceptual–motor speed); SFA: Saturated fatty acids; SMC: Subjective memory complaint; VaD: Vascular dementia.

Expert Rev. Neurother. 11(5), (2011)


Diet & Alzheimer’s disease Review

been found in population samples with elevated fish consumption, of n-3 PUFA supplementation on patients with VaD, AD, MCI or
and high intake of MUFA and PUFA, particularly n-3 PUFA [42] . age-related cognitive decline (ARCD) in cognitively unimpaired
In fact, fish, particularly fatty fish (e.g., herring, mackerel, salmon older subjects [79–85] . These RCTs suggested a positive effect of
or trout), is the principal source of n-3 PUFA in the MeDi. Very this intervention only in very mild AD or MCI patients, or in
recently, the baseline data from the Older People And n-3 Long- subgroups (e.g., APOE- e4 carriers), for cognitive performance in
chain polyunsaturated fatty acid (OPAL) study suggested that nondemented subjects or for neuropsychiatric symptoms in mild-
higher fish consumption is associated with better cognitive func- to-moderate AD patients [79] . Supplemental DHA may not neces-
tion in later life [74] . Epidemiological, longitudinal, observational sarily be recommended when the decline is more severe, possibly
studies reporting associations of fish consumption with cognitive because additional DHA may contribute to degenerative processes
function have demonstrated mixed results; some longitudinal in the brain related to lipid peroxidation [41] .
studies have reported a positive association with higher fish con- Different pathways could contribute to the neuroprotective,
sumption [63,75–77] , while others have found no association  [57] . as well as the neurotrophic, properties of UFA (Figure 1) [38,39,42] .
Nevertheless, a lower risk of incident dementia in subjects con- An underlying biological mechanism for the observed associa-
suming more fish has been reported in several other independent tion between UFA and dementia and predementia syndromes is
prospective cohort studies [69,71,73,78] . Therefore, despite these con- currently unknown. In particular, the prolonged protection of
trasting findings, there is now considerable evidence suggesting MUFA intake against ARCD and AD may be linked to the rel-
that fatty acid intake and fish consumption may influence demen- evant quota of antioxidant compounds in olive oil, including low-
tia and AD risk, but the direction (protection or risk) and the level molecular-weight phenols (Figure 1) [38,39,42] . Moreover, substitution
of this effect remain unclear [42] . Finally, some recent randomized of caloric energy from SFA with UFA has been demonstrated to
controlled trials (RCTs) assessed the cognitive or functional effect lower low-density lipoprotein (LDL) cholesterol levels and increase

Mediterranean diet

Olive oil Fruits and


Dairy products Moderate
Fish consumption consumption vegetables
alcohol intake

n-3 PUFA MUFA

Blood pressure
regulation

Antioxidant Insulin sensitivity


compounds Vascular changes
Lacunar infarcts
Structural WMLs Weight
integrity of Inflammation reduction
neuronal Oxidative stress
membranes Dendritic
pathology

sAAPα sAAPα
Aβ AD VaD
β-secretase pathology
γ-secretase
α-secretase BACE1

c83 APP c99 AICD


Nonamyloidogenic pathway Amyloidogenic pathway

Figure 1. Overview of the principal underlying mechanisms linking the Mediterranean diet and its principal components to
Alzheimer’s disease and vascular dementia.
Ab: b-amyloid; AD: Alzheimer’s disease; AICD: APP intracellular domain; APP: Amyloid precursor protein; BACE1: b-site APP-cleaving
enzyme 1; MUFA: Monounsaturated fatty acids; PUFA: Polyunsaturated fatty acids; sAPPa: Soluble amyloid precursor protein a;
sAPPb: Soluble amyloid precursor protein b; VaD: Vascular dementia; WML: White matter lesion.

www.expert-reviews.com 683
Review Solfrizzi, Panza, Frisardi et al.

high-density lipoprotein (HDL) cholesterol levels [86] . Other stud- for cardiovascular disease, and therefore for cognitive decline,
ies have found that high-fat/high-cholesterol diets increased, and VaD and AD [38,39,42] . A diet high in SFA and cholesterol may
cholesterol-lowering drugs decreased, Ab peptide deposition and increase hippocampal accumulation of Ab [9] . In addition, a diet
AD-related abnormalities  [41,42] . The neuroprotective effects of high in fat/SFA may lead to cognitive impairment through its
dietary UFA could also rely on their impact on membrane architec- effects on, for example, hyperinsulinemia [86] , oxidative stress or
ture: maintaining the structural integrity of neuronal membranes, endothelial damage [38,39,42] . In experimental studies, SFA have
determining the fluidity of synaptosomal membranes and thereby decreased the levels of hippocampal brain-derived neurotrophic
regulating neuronal transmission (Figure 1) . Furthermore, essential factor (BDNF) that are relevant for neuronal plasticity and the
fatty acids can modify the activity of certain membrane-bound maintenance of cognitive functions [97] . On the contrary, treat-
enzymes (phospho­lipase A2, protein kinase C and acetyltranfer- ment for 4 weeks with a Mediterranean-inspired diet rich in n-3
ase), and the function of the neurotransmitter receptors. Moreover, PUFA decreased blood lipids in healthy individuals with a low-
free fatty acids, lipid metabolites and phospholipids modify the risk profile for cardio­vascular disease, with a beneficial effect also
function of membrane proteins, including ion channels [38,39,42] . on vascular function and oxidative stress [98] . Finally, the inverse
Fatty acid composition of neuronal membranes in advancing age relationship between fish consumption and dementia could be
also demonstrated an increase in MUFA content and a decrease explained by the biological hypothesis that the potentially protec-
in PUFA content  [38,39,42] . In adult rats, learning and cognitive tive active ingredients are the n-3 PUFA content in fish, with fatty
behavior are related to brain DHA status, which, in turn, is related fish containing considerably greater amounts of n-3 PUFA than
to the levels of the dietary n-3 PUFA [87] . In fact, administration of white fish: approximately 2 g and 0.3 g n-3 PUFA per 100 g of
DHA seems to improve learning ability in Ab-infused rats [88] and average fish, respectively [99] . However, other constituents of fish
inhibit decline in avoidance learning ability in the AD model rats, could prevent dementia. For instance, fish is also a good source
which was associated with an increase in the corticohippocampal of selenium, a powerful anti­oxidant, and fatty fish traditionally
n-3:n-6 ratio, and a decrease in neuronal apoptotic products [89] . consumed in Northern Europe are not only rich in n-3 PUFA, but
In other transgenic AD mouse models, DHA also protects against also in vitamin D, which is a recently recognized neurosteroid hor-
dendritic path­ology  [90] , prevents neuronal apoptosis induced by mone [100,101] . Vitamin D is essential for neurophysiological func-
soluble Ab peptides [91] , increases synaptic protein and phospho- tion, regulation of neuro­transmitters or neurotrophic factors, and
lipid densities [92] , reduces the intraneuronal accumulation of both cerebral antioxidant, anti-inflammatory and anti-ischemic mecha-
Ab and tau protein [93] , and inhibits degradative endopeptidase nisms [100,101] . A recent systematic literature review suggested that
activities [94] . Some experimental evidence suggests that essential vitamin D insufficiency may instead cause neuropathological dys-
n-3 PUFA effectively lower Ab production in transgenic mice, function, such as cognitive impairment [100] . Therefore, the appar-
as reported in studies from several laboratories (Figure 1) [92,95,96] . ent protective effect of fish consumption against dementia could
There are several published studies on human infant subjects be explained by the impact of a health-promoting lifestyle or by
in which breastfeeding – which leads to higher DHA concen- the fish itself. In this view, the intake of n-3 PUFA, antioxidant
trations in the brain – or n-3 PUFA supplementation, is related compounds and vitamin D should be taken into account.
to better cognitive performance at a later age [38,39,42] . The n-3 In conclusion, epidemiological evidence has suggested a pos-
PUFA from fish may be inversely associated with dementia because sible association among fish consumption, MUFA and PUFA
it lowers the risk of thrombosis, stroke, cardiovascular disease (particularly, n-3 PUFA) and reduced risk of cognitive decline,
and cardiac arrhythmia, reducing the risk of thromboembolism AD and dementia. However, due to the small number of stud-
in the brain and consequently of lacunar and large infarcts that ies that inform this topic, further research is necessary before a
can lead to VaD and AD [38,39,42] . Furthermore, the n-3 PUFA strong conclusion can be drawn. Based on the current evidence
may be important as lipids in the brain, particularly for the pos- from human and animal studies, it is not possible to make defini-
sible influence of DHA on the physical properties of the brain tive dietary recommendations in relation to the AD risk on fish
that are essential for its function [38,39,42] . Furthermore, fish oil consumption and the lower intake of saturated fat from meat
was a better source than a-linolenic acid for the incorporation and dairy products [102] , as well as on UFA consumption or lower
of n-3 PUFA into rat brain phospholipid subclasses [38,39,42] . On intake of saturated fat in relation to the risk for dementia and
the contrary, high linoleic acid intake (n-6 PUFA) may increase cognitive decline. However, a high consumption of fats from
the susceptibility of LDL cholesterol to oxidation, which makes fish, vegetable oils, vegetables and nuts should be encouraged as
it more atherogenic, even if the association between linoleic acid this dietary advice is in accordance with recommendations for
and atherosclerosis is controversial  [38,39,42] . Therefore, the ratio lowering the risk of cardiovascular disease, obesity, diabetes and
of dietary n-3:n-6 PUFA intake may influence the potential role hypertension [42,102] .
of PUFA on cognitive decline and dementia, and the optimal
ratio of n-6:n-3 for a healthier diet should be <5:1 [38,39,42] . At Dairy products & risk of cognitive impairment
present, similar indications for cognitive decline or dementia are & Alzheimer’s disease
not available, but n-3 PUFA, particularly DHA status, should While positive associations have been shown between a number
not be considered independently of n-6 PUFA intake [80] . Finally, of macronutrients and cognitive performance, little attention
a high dietary intake of SFA and cholesterol increases the risk has been paid to the potential role of dairy foods in modulating

684 Expert Rev. Neurother. 11(5), (2011)


Diet & Alzheimer’s disease Review

the risk of cognitive impairment or AD [103] . This is despite foods are involved in blood pressure regulation [113] . Magnesium
recent epidemiological, observational and interventional studies may also modulate vascular function through its antioxidant
providing evidence for the role of dairy products in improving and anti-inflammatory properties [114] , and increasing dietary
cardiovascular risk factors and lowering the prevalence of the intake of magnesium may help reduce cholesterol and triglycer-
metabolic syndrome [103] . Dairy consumption may reduce the ides [115] , and improve diabetes control, owing to its role in glu-
likelihood of cognitive decline, either directly, or via mediating cose homeostasis [116] . The beneficial effects of these components
effects on cardio-metabolic health [103] . Among the cross-sectional of dairy foods on vascular disease may also be mediated by an
epidemiological studies (Table 2) [104–106] , one report suggested improvement in the inflammatory state. Individuals with greater
that women with poor cognitive function had significantly lower adiposity have an increased inflammatory state in comparison to
intakes of milk and dairy products than those with inadequate or their leaner counterparts [117] , which has been linked with adverse
normal cognitive function [104] , and another study reported that cognitive function [118] . Any decrease in inflammation (via weight
higher cheese intake was associated with a reduced likelihood loss or improved regulation of glucose and blood pressure) may
of cognitive impairment, while milk intake was not associated subsequently reduce the risk for cognitive decline (Figure 1) [103] .
with cognitive impairment [105] . Among longitudinal studies
(Table 2) [63,64,72,107,108] , only one out of the five principal prospec- Alcohol consumption in predementia
tive reports found a beneficial effect from dairy consumption & dementia syndromes
in regards of cognitive function or MCI/dementia risk [108] . In An additional interesting link between diet and AD is in rela-
fact, a Japanese prospective study indicated that milk intake was tion to alcohol intake. Recent studies with older samples have
associated with a significantly lower likelihood of VaD in older indicated that a U- or J-shaped curve may best describe the rela-
age amongst both men and women [108] . On the other hand, three tionship between the level of alcohol consumption and cognitive
longitudinal studies found adverse effects on cognitive function performance [119–123] . In fact, in several cross-sectional studies,
from dairy fat [63,64,72] . The Cardiovascular Risk Factors, Aging moderate drinking, from up to one drink per day (up to 14 g of
and Incidence of Dementia (CAIDE) study did not examine dairy alcohol) to four drinks per day (52 g of alcohol) has been associated
intake per se, but evaluated associations of the fat from milk prod- with better function in many cognitive domains compared with
ucts and spreads with the risk of MCI, dementia, AD and a num- nondrinking [124–127] . Furthermore, in cross-sectional studies,
ber of cognitive domains [63,72] . High saturated or total fat intakes both greater [128–132] and poorer [133] cognitive performances have
were associated with an increased risk for MCI, poorer global been observed with higher levels of alcohol drinking. The studies
cognitive function and poorer psychomotor speed [63] , but not that have assessed changes in cognition prospectively have had
with risk of dementia or AD [72] . Similarly, an Australian study contradictory results (Table 3) [46] . Drinkers have been proposed
on older men demonstrated that whole-fat milk consumption was to have a greater decrease in global cognitive function [134] and
associated with impaired cognitive function [107] , while the final attention [135] , compared with nondrinkers, but moderate drink-
study reported that higher consumption of dairy desserts and ice ers (~one drink, or less than 15 ml of alcohol per day) have less
cream was associated with an increased risk of cognitive decline decline in general cognition [136] or psychomotor speed [137] than
amongst elderly French women [64] . No significant associations nondrinkers. Furthermore, some studies demonstrated no asso-
were found between milk and yogurt, or cheese consumption and ciation at all [123,130,138,139] . In particular, the association between
cognitive decline [64] . A very recent systematic review on the pos- current cognitive performance and alcohol drinking 5–24 years
sible association between dairy consumption and cognitive func- earlier have also been studied, with contrasting results. In fact, the
tion concluded that methodological variability and study limita- global cognitive function may be poorer among both alcoholics
tions did not enable conclusions to be drawn regarding optimal and past drinkers, compared with nondrinkers or moderate drink-
dairy intake and cognitive performance [103] . The limited available ers [121] , and better among moderate drinkers and poorer among
literature supported the concept that high dairy consumption may heavy drinkers as compared with nondrinkers [140] . Moreover,
be associated with a lower likelihood of cognitive impairment; smoking or the APOE e4 allele often modified the association
however, high intakes of full-fat dairy and/or dairy fats may be between alcohol drinking and cognitive functions [46,141] .
associated with declines in cognitive performance [103] . Dairy Alcohol drinking is also one of the possible determinants of
foods may reduce the risk for cognitive impairment by modifying clinical dementia [46,133,135,137,138] . Epidemiological studies have
vascular factors linked to detrimental brain changes, particularly reported an association between red wine consumption and the
via weight reduction (Figure 1) . Epidemiological studies, animal incidence of AD and dementia in cross-sectional and longitudinal
studies and a small number of randomized trials provide evidence studies (Table 3) [142,143] . These results were consistent with the
for an antiobesity effect of calcium and dairy foods [109–111] . In findings from some cohort studies [144–147] , but not with oth-
addition to calcium, bioactive compounds derived from whey ers  [133,135,137,138,148,149] . Other population-based, prospective
protein in dairy may contribute to accelerating weight and fat studies, with longer follow-up periods, have studied the effects
loss [109,111] . Furthermore, dairy products are the primary dietary of different patterns of alcohol intake on dementia [150–152] . For
source of vitamin D [112] , which, through its role in regulating predementia syndromes, in 1445 noncognitively impaired indi-
calcium homeostasis, may improve insulin sensitivity and glucose viduals aged 65–84 years from the Italian Longitudinal Study
homeostasis [112] . Phosphorus and magnesium contained in dairy on Aging (ILSA), patients with MCI who consumed up to one

www.expert-reviews.com 685
Table 2. Principal longitudinal, clinical and epidemiological studies on the relationship between dietary dairy products and dementia

686
(i.e., Alzheimer’s disease and vascular dementia) or predementia syndromes (i.e., age-related cognitive decline and mild
cognitive impairment).
Review

Study (year) Setting and Subjects Dietary Cognitive outcomes Results and conclusions Ref.
study design assessment
(duration)
Dietary dairy products & predementia syndromes
Almeida et al. (2006) Longitudinal, 601 men, aged Self-report ‘Preserved cognitive function’: MMSE ≥24 Consumption of full-cream milk associated [107]
Australia population- 80 years and older questionnaire, ‘Preserved mood’: GDS ≤5 with impaired cognitive function and with
based including ‘Successful mental health aging’: poor mental health
(4.8 years) dietary measures MMSE ≥24 and GDS ≤5
Vercambre et al. (2009) Longitudinal, 4809 elderly Evaluation of DECO and IADL No significant association between milk [64]
Etude Epidémiologique population- women, dietary intakes and yogurt, and cheese consumption with
de Femmes de la based 76–82 years old with a 208-item cognitive decline
Mutuelle Générale de (13 years) FFQ
Solfrizzi, Panza, Frisardi et al.

l’Education Nationale
(E3N), France
Eskilinen et al. (2008) Longitudinal, 1449 subjects, Evaluation of The Mayo Clinic AD Research Center High SFA intake from milk products and [63]
Cardiovascular Risk population- aged 65–80 years dietary intake criteria were applied for diagnosing MCI: spreads (>21.6 g) associated with increased
Factors Aging and based with a 208-item MMSE, CFT, PPBt, LDST, episodic memory risk for MCI compared with those with
Dementia (CAIDE), (21 years) FFQ with immediate word recall tests; lower intakes. High SFA from milk products
Finland executive function with the Stroop test,; associated with poorer global cognitive
and prospective memory with a task by function (MMSE).
Einstein High total fat from milk products and
spreads associated with poorer
psychomotor speed
Dietary dairy products & dementia syndromes
Yamada et al. (2003) Longitudinal, 1774 subjects, Evaluation of Clinical diagnosis of dementia (DSM-IV Almost-daily milk intake associated with [108]
The Adult Health Study population- born before dietary intake criteria), CASI, IQCDE (by the caregiver), significantly lower likelihood of VaD,
Japan based September 1932 with a FFQ Hachinski’s Ischemic Score, Clinical compared with consuming milk less than
(25–30 years) Dementia Rating four-times per week
Laitinen et al. (2006) Longitudinal, 1449 subjects, Evaluation of Incident diagnosis of AD Fat intake from milk products (milk and [72]
CAIDE, Finland population- aged 65–80 years dietary intake (NINCDS– ADRDA criteria) sour milk) not significantly associated with
based (21 years) with a 154-item risk of dementia or AD. Moderate intakes
FFQ of PUFA from spreads associated with
decreased risk of dementia. Moderate
intake of SFA from spreads associated with
increased risk of dementia and AD
AD: Alzheimer’s disease; CASI: Cognitive Ability Screening Instrument; CFT: Category Fluency Test (semantic memory); DECO: DEtérioration Cognitive Observéè scale (observed cognitive deterioration);
DSM-IV: Diagnostic and Statistical Manual of Mental Disorders, fourth edition; EBMT: East Boston Memory Test (immediate and delayed episodic memory); FFQ: Food Frequency Questionnaire; GDS: Geriatric
Depression Scale; IADL: Instrumental activities of daily living; IQCDE: Informative Questionnaire on Cognitive Decline in the Elderly; LDST: Letter–Digit Substitution Test (perceptual–motor speed); MCI: Mild
cognitive impairment; MMSE: Mini-Mental State Examination (global cognitive functioning); MUFA: Monounsaturated fatty acids; NINCDS–ADRDA: National Institute of Neurological and Communicative Disorders
and Stroke and the Alzheimer’s Disease and Related Disorders Association; PPBt: Purdue Peg Board task (psychomotor speed); PUFA: Polyunsaturated fatty acids; SFA: Saturated fatty acids; VaD: Vascular dementia.

Expert Rev. Neurother. 11(5), (2011)


Table 3. Principal longitudinal clinical and epidemiological studies on the relationship between alcohol consumption and dementia
(i.e., Alzheimer’s disease and vascular dementia) or predementia syndromes (i.e., age-related cognitive decline and mild
cognitive impairment).
Study (year) Setting and study Subjects Methods Results and conclusions Ref.
design (duration)

www.expert-reviews.com
Alcohol consumption & predementia syndromes
Launer et al. (1996) Cross-sectional 489 men, aged MMSE, evaluation of alcohol intake Men with CVD/diabetes and low-to-moderate alcohol intake [123]
The Zutphen Elderly Study, and longitudinal, 69–89 years and smoking habits had a significantly lower risk for poor cognitive function than
The Netherlands population-based abstainers. Alcohol intake was not associated with cognitive
(3 years) decline
Elias et al. (1999) Cross-sectional 1786 subjects, Eight cognitive tests of verbal Women who drank moderately (two to four drinks/day) [124]
The Framingham Heart and longitudinal, aged memory, learning, visual demonstrated superior performance in many cognitive domains
Study, USA population-based 55–88 years organization and memory, attention, relative to abstainers. For men, superior performance was
(24 years) abstract reasoning and concept found within the range of four to eight drinks/day, although
formation; evaluation of weekly fewer significant relations were observed. These results were
alcohol intake confirmed by prospective analyses of 24-year drinking history
Dufouil et al. (2000) Longitudinal, 1389 subjects, MMSE, evaluation of alcohol Alcohol consumption was associated with a decreased risk of [134]
Epidemiology of Vascular population-based aged consumption, APOE genotyping and cognitive decline in individuals without the APOE e4 allele,
Aging (EVA) Study, France (4 years) 59–71 years smoking habits whereas moderate drinking increased the risk of decline in
APOE e4 allele carriers. In addition, lifetime smoking was a risk
factor for cognitive decline in individuals without the APOE e4
allele. The data also suggested a slight protective effect of
smoking in APOE e4 allele carriers
Ngandu et al. (2007) Longitudinal, 1341 MMSE and neuropsychological tests The nondrinkers, both at midlife and later, had a poorer [141]
Cardiovascular Risk population-based participants, evaluating episodic memory, cognitive performance than drinkers, especially in the domains
Factors Aging and (21 years) aged semantic memory, psychomotor related to fluid intelligence – that is, executive function,
Dementia (CAIDE), 65–79 years speed, executive function, psychomotor speed, as well as episodic memory – whereas the
Finland prospective memory and subjective other cognitive functions showed little association with alcohol
memory. Evaluation of alcohol drinking. No interactions between APOE e4 and alcohol, or sex
intake, smoking habits and APOE and alcohol were found
genotyping
Anttila et al. (2004) Longitudinal, 1464 men and Diagnosis of incident dementia and Alcohol drinking in middle-age demonstrated a U-shaped [154]
CAIDE, Finland population-based women, aged MCI, and subjects classified as those relationship with risk of MCI in old age. Only the carriers of
study (23 years) 65–79 years who never drank alcohol, those who APOE e4 had an increased risk of dementia with increasing
drank ‘infrequently’ (less than once a alcohol consumption
month) and those who drank
‘frequently’ (several times a month)
Diet & Alzheimer’s disease

Espeland et al. (2005) Longitudinal, 4451 Diagnosis of incident MCI and Moderate alcohol intake was associated with an approximately [155]
Women’s Health Initiative population-based community- dementia, and annual modified 50% reduced risk of combined probable-dementia and/or MCI.
Memory Study, USA study (4.2 years) dwelling MMSE. Subjects classified as Significant decline in global cognition was less common among
women, aged abstainers, <one drink/day and >one women who reported alcohol intake at baseline, and also after
65–79 years drink/day adjustment for both demographic/social and clinical factors
AD: Alzheimer’s disease; APOE: Apolipoprotein E; CVD: Cerebrovascular disease; MCI: Mild cognitive impairment; MMSE: Mini-Mental State Examination; OR: Odds ratio; VaD: Vascular dementia.
Review

687
Table 3. Principal longitudinal clinical and epidemiological studies on the relationship between alcohol consumption and dementia

688
(i.e., Alzheimer’s disease and vascular dementia) or predementia syndromes (i.e., age-related cognitive decline and mild
cognitive impairment).
Review

Study (year) Setting and study Subjects Methods Results and conclusions Ref.
design (duration)
Alcohol consumption & predementia syndromes
Solfrizzi et al. (2007) Longitudinal, 1445 men and Diagnosis of incident MCI and In patients with MCI, up to one drink/day of alcohol or wine [153]
The Italian Longitudinal population-based women, aged progression from MCI to dementia; may decrease the rate of progression to dementia. No
Study on Aging (ILSA), study (3.5 years) 65–84 years subjects classified as abstainers, zero significant associations were found between any levels of
Italy to one drink/day, one to two drinking and the incidence of MCI in noncognitively impaired
drink/day, and >two drink/day individuals versus abstainers
Alcohol consumption & dementia syndromes
Yoshitake et al. (1995) Longitudinal, 828 subjects, Diagnosis of dementia, AD and VaD; Multivariate ana­lysis demonstrated that age, systolic blood [133]
The Hisayama Study, population-based aged 65 years evaluation of alcohol consumption pressure, drinking habits and prior stroke episodes were
Japan (7 years) and over significant independent precursors of VaD
Solfrizzi, Panza, Frisardi et al.

Orgogozo et al. (1997) Longitudinal, 3777 subjects, Diagnosis of dementia and AD; In the 318 moderate drinkers, the ORs were respectively 0.19 [142]
Personnes Agees QUID population-based aged 65 years evaluation of alcohol intake for incident dementia and 0.28 for AD as compared with the
(PAQUID) Study, France (3 years) and over 971 nondrinkers, and after adjusting for possible confounders.
Among the 922 mild drinkers, the relative risk for AD was
reduced significantly with respect to the abstainers
Ruitenberg et al. (2002) Longitudinal, 5395 subjects, Diagnosis of AD, VaD or other Light-to-moderate drinking (one to three drinks per day) was [144]
The Rotterdam Study, The population-based aged 55 years dementia; evaluation of alcohol significantly associated with a lower risk of any dementia and
Netherlands study (6 years) and older intake and APOE genotyping VaD. No evidence was found that the relationship between
alcohol and dementia varied by type of alcoholic beverage
Mukamal et al. (2003) Nested case-control 373 cases with Diagnosis of incident dementia (AD Compared with abstention, consumption of one to six drinks [165]
The Cardiovascular Health of a longitudinal, incident and VaD), average alcohol weekly was associated with a lower risk of incident dementia
Study (CHS), USA population-based dementia and consumption and MRI findings among older adults. A trend toward greater odds of dementia
study (6 years) 373 controls associated with heavier alcohol consumption was most
apparent among men and participants with an APOE e4 allele,
with similar relationships of alcohol use with AD and VaD
Deng et al. (2006) Longitudinal, 3286 subjects, Diagnosis of incident dementia, AD Light-to-moderate drinkers had a lower risk of dementia than [147]
Chongqing City Study, population-based aged 60 years and VaD and evaluation of alcohol nondrinkers, but a nonsignificant increased risk was observed in
China study (2 years) and older intake heavy drinkers
Mehlig et al. (2008) Longitudinal, 1462 women, Diagnosis of dementia; evaluation of Wine was protective for dementia, and the association was [152]
The Prospective population-based aged alcohol intake and smoking habits strongest among women who consumed wine only. By
Population Study of study (34 years) 38–60 years contrast, consumption of spirits at baseline was associated with
Women in Göteborg, a slightly increased risk of dementia
Sweden
AD: Alzheimer’s disease; APOE: Apolipoprotein E; CVD: Cerebrovascular disease; MCI: Mild cognitive impairment; MMSE: Mini-Mental State Examination; OR: Odds ratio; VaD: Vascular dementia.

Expert Rev. Neurother. 11(5), (2011)


Diet & Alzheimer’s disease Review

drink of alcohol per day had a reduction in the rate of progression participants evaluated for AD, 10,225 participants evaluated for
to dementia, in comparison with patients with MCI who never VaD and 11,875 followed for any type of dementia [157] . This
consumed alcohol. Overall, patients with MCI who consumed less meta-ana­lysis indicated that light-to-moderate alcohol intake
than one drink of wine per day versus nondrinkers had a decrease was associated with a 25–28% reduction in risk of AD, VaD
in the rate of progression to dementia of approximately 85%. No and any dementia compared with alcohol abstinence in older
significant association was found between any levels of drinking adults. Heavy drinking was not associated with increased risk
and the incidence of MCI in noncognitively impaired individuals of dementia in the present study [157] .
versus abstainers [153] . Only a few other studies have examined The mechanism by which low-to-moderate alcohol intake could
the effect of alcohol consumption on the risk for the incidence of be protective against cognitive impairment or decline in older
MCI [154,155] . After an average follow-up of 23 years, non­drinkers age or against predementia and dementia syndromes is, at pres-
and frequent drinkers were both more than twice as likely to ent, unclear. Given that drinking alcohol influenced measures
have MCI in old age than occasional drinkers [154] . However, the of psychomotor speed, episodic memory and executive function,
APOE genotype seemed to modify the relationship, such that the in particular, it was supposed that white matter lesions (WMLs)
risk of old age dementia increased with increasing midlife alcohol would play a neuropathological role. In fact, alcohol drinking has
consumption only among carriers of the APOE e4 allele [154] . On been associated with fewer brain infarcts and was demonstrated
the other hand, the findings from the ILSA were consistent with to have a U-shaped relationship with WMLs [158] . WMLs and
those obtained in the Women’s Health Initiative Memory Study infarcts, in turn, may reflect a vascular mechanism responsible
with a 4.2-year follow-up, which found that moderate alcohol for the observed association between alcohol and cognitive func-
intake (≥one drink per day) was associated with an approximately tions (Figure 1) . Furthermore, different mechanisms may underlie
50% reduced risk of combined probable dementia and MCI [155] . the adverse effects of heavy drinking and the potential beneficial
However, after adjusting for demographic and socioeconomic effects of low-to-moderate drinking. In fact, higher doses of alco-
factors and baseline Modified Mini-Mental State Examination hol may affect cognitive functioning through increased release of
(3MSE), the significance disappeared [155] . acetylcholine from the hippocampus [159] , which was linked to
Recently, a systematic review with meta-ana­lysis was carried problems with memory and attention [160] . On the other hand,
out to investigate any relationship between incident cognitive animal evidence indicated that low doses of alcohol may stimulate
decline or dementia in the elderly and alcohol consumption the release of hippocampal acetylcholine [161] . Finally, moder-
between 1995 and March 2006, with only longitudinal studies ate alcohol consumption was associated with reduced cardiovas-
of subjects aged ≥65 years included [156] . Definitions of outcomes cular risk factors [162] and may be protective against ischemic
varied with some studies reporting on dementia, or AD alone, stroke  [163] , lowering rates of cardiovascular disease, protecting
or AD and dementia, or VaD, or AD and VaD, or all demen- brain vasculature and preventing subclinical strokes, resulting
tia, VaD and AD [156] . This meta-ana­lysis suggested that, at in better cognitive performance. Moreover, light-to-moderate
least in epidemiological studies, light-to-moderate alcohol use alcohol use is associated with a lower prevalence of MRI-defined
was associated with a 38% reduced risk of unspecified incident WMLs and subclinical infarcts  [164] , although MRI abnormali-
dementia. In the studies included in this meta-ana­lysis, there ties, HDL cholesterol levels and fibrinogen levels only marginally
was also no close agreement as to the optimal level of alcohol influenced the association of alcohol consumption and dementia
consumption and the classification of light-to-moderate drinking in the Cardiovascular Heart Study [165] . The suggested protection
varied very widely. In fact, for dementia, benefit was shown for of low-to-moderate alcohol use against CVD may also explain
more than one drink per day, weekly or monthly wine consump- the possible protection against VaD. In the Rotterdam study,
tion, 250–500 ml (usually wine) or more, which is equivalent the protective effect of alcohol consumption was mainly found
to three drinks per day, and from 1 to 28 units per week [46] . for VaD, and the authors suggested that moderate alcohol intake
For AD, light-to-moderate alcohol was associated with a sig- may protect against dementia via a reduction in vascular risk fac-
nificantly reduced risk of 32%, defining optimal amounts as tors [144] . In fact, moderate doses of alcohol may increase prosta-
weekly consumption of wine, 1–6 units or more than two drinks cyclin concentrations, reduce the generation of thromboxane A2
per week, or more than three drinks/250–500 ml per day (usu- and inhibit platelet function [46] . They may increase plasma levels
ally wine), or where studied by gender, one to three per day in of endogenous tissue-type plasminogen activator, a serine protease
males  [46] . Although the point estimates were also in a similar that regulates intravascular fibrinolysis and fibrinolytic activity,
direction for VaD and cognitive decline (0.82 and 0.89, respec- while decreasing plasma fibrinogen levels [46] . It is also known that
tively), the results were not statistically significant [156] . With alcohol is associated with increased levels of HDL cholesterol, its
regard to cognitive function, results for optimal consumption sub­fractions HDL2 and HDL3, and its associated apolipoproteins
were mixed, either above, below or equal to one drink a month A-I and A-II [46] . The association with HDL cholesterol is deemed
or day, in subjects with cardiovascular disease or diabetes, one to account for up to half of the reduction in coronary events
to two drinks per week, while for VaD, one to three drinks per associated with moderate alcohol consumption [46] .
day in males appeared to be beneficial [46] . Finally, another very While the above-cited factors affect the risk of unspeci-
recent meta-ana­lysis included 15 prospective studies (follow- fied dementia and, probably, of VaD, other experimental and
ups ranged from 2 to 8 years), with samples including 14,646 clinical findings may partly explain the suggested protection of

www.expert-reviews.com 689
Review Solfrizzi, Panza, Frisardi et al.

low-to-moderate alcohol consumption against AD. In fact, small reach next-to-therapeutic concentrations [173] . In fact, even after
amounts of alcohol have been reported to be associated with a a 25-mg oral dose, unconjugated resveratrol blood level was less
lower prevalence of vascular brain findings and, in APOE e4 than 5 ng/ml, and a resveratrol intake of 25 mg corresponds to
carriers, with hippocampal and amygdalar atrophy as assessed the consumption of more than 4 l of red wine (assuming a mean
by MRI  [164] . Experimental studies found that ethanol initially resveratrol content of 6 mg/l red wine) [174] .
increases hippo­campal acetylcholine release, which could conceiv- In conclusion, low-to-moderate alcohol drinking has been pro-
ably improve memory performance [165] . Processes that originate, posed as a protective factor against MCI and dementia in several
modulate or precipitate the deposition of Ab in the brain (e.g., longitudinal studies, but contrasting findings also exist  [46] . In
oxidative stress) rather than vascular processes, may better explain fact, many of these studies were limited by cross-sectional design,
the development of AD and the vascular effects of the alcohol com- restriction by age or sex, or incomplete ascertainment. Moreover,
ponent of alcoholic beverages may not be enough to explain the different outcomes, beverages, drinking patterns or follow-up
protective effects of the moderate intake of alcohol from dementia. periods, or possible interactions with other lifestyle-related (i.e.,
Wine consumption may exert a protective effect, either through smoking) or genetic factors (i.e., APOE genotyping) may be
alcohol intake itself, through the antioxidant effects of polyphe- sources of great variability. In fact, the body of evidence within
nols richly represented in red wine [46] or through both (Figure 1) . the last 10 years suggested that low-to-moderate alcohol use may
The latter effects, of course, are independent of alcohol and, in be associated with a reduced risk of unspecified incident demen-
fact, have been also associated with alcohol-free red wine [166] . tia and AD, while for VaD, cognitive decline and predementia
The constituents of red wine also have potentially beneficial vas- syndromes the current evidence is only suggestive of a protective
cular effects, enhance endothelial nitric oxide release and reduce effect [46] . Therefore, at present, the preferable outcomes for these
athero­sclerosis in APOE-deficient mice [46] . Red wine polyphenols kind of studies appear to be unspecified dementia and AD, given
are a complex mixture of flavonoids (mainly anthocyanins and the small number of studies reporting VaD as an outcome and
flavan-3-ols) and nonflavonoids (such as resveratrol and gallic the difficulties in the classification of cognitive decline and pure
acid). Flavan-3-ols are the most abundant, with oligomeric and VaD,; often including cases with vascular factors in the AD cat-
polymeric procyanidins (condensed tannins) often representing egory. On the other hand, the cardiovascular mechanisms behind
25–50% of the total phenolic constituents [167] . Given the link the protective effects of alcohol may have an even greater effect on
between VaD, vascular risk and the increasing body of evidence VaD. For the different types of beverages, several studies suggest
suggesting that AD may be influenced by vascular factors [21] , it that low-to-moderate wine consumption may be protective against
may be concluded that the vascular protection associated with dementia and cognitive decline and not total alcohol intake, beer
wine consumption decreases the risk of incident dementia/AD. In or spirits, with strong but not conclusive evidence [46] . In this
fact, in the 5-year follow-up Personnes Agées QUID (PAQUID) context, it would be desirable to differentiate red and white wine,
cohort, a significant inverse association between flavonoid intake but this information is not currently available. Whether the pres-
and the risk of dementia was found [168] . It has also been sug- ent divergent findings can be explained by drinking patterns has
gested that the antioxidant properties of the flavonoids in wine not been extensively investigated, and studies with long follow-up
may help prevent the oxidative damage implicated in dementia. periods are few [46] . Therefore, the role of alcohol drinking for
In fact, oxidative stress may also develop in the brain, leading cognitive functions may be somewhat different at different time
to neuronal death by various mechanisms such as formation of points. Nonetheless, the current evidence suggests that alcohol
Ab, DNA damage, mitochondrial dysfunction and abnormal tau consumption in old age is substantially in agreement with midlife
protein [169,170] . The presence in wine of nonalcoholic compo- consumption in relation to cognitive decline [46] . At present, there
nents, such as particular antioxidants, could explain a differential is no evidence indicating that starting to drink at a later age
effect of wine on dementia. In fact, liquor has been demonstrated would be beneficial against predementia or dementia syndromes.
to have less antioxidant activity than wine [171] . Nonetheless, in Therefore, as interventional studies are not feasible in this area,
some studies on the neuroprotective role of moderate alcohol con- the best evidence comes from an overview of epidemiological
sumption, the most typically consumed alcohol types were beer studies. At present, there is no indication that low-to-moderate
and spirits [105,119] . Finally, sirtuins, conserved proteins with a alcohol drinking would be harmful to cognition and dementia.
NAD + -dependent deacetylase and mono-ADP-ribosyltransferase However, it is not possible to define a specific beneficial level
activity, and particularly SIRT1, has been linked to the protec- of alcohol intake in relation to cognitive functions and demen-
tive effects of red wine and calorie restriction in in vivo models of tia. Taking into account the well-known harms related to heavy
AD, and this has shed new light on the importance of moderate alcohol drinking and the lack of long-term follow-up studies or
wine consumption and reduced food intake as AD-preventing RCTs, it would be too early to recommend alcohol to abstainers
behavioral strategies, suggesting a possible molecular mechanism to prevent cognitive decline or predementia syndromes or delay
underlying these AD risk modulators [172] . However, the benefi- the onset of dementia. In moderate-to-heavy alcohol drinkers
cial effects of wine drinking and calorie restriction may involve experiencing memory difficulty, or with suggested diagnoses of
molecular pathways other than sirtuins, and the bioavailability of MCI or early AD [175] , among management options, given the
resveratrol, the most potent natural SIRT1 activator, is quite low challenge in achieving total abstinence, a viable alternative option
and would require more than a ‘moderate’ wine consumption to could also be low-to-moderate drinking [176] .

690 Expert Rev. Neurother. 11(5), (2011)


Diet & Alzheimer’s disease Review

High intakes of cereals, fruits & vegetables in AD AD clinical trials, antioxidants have demonstrated only a mar-
In vitro and in vivo findings demonstrated that chronic accumula- ginal positive effect on disease progression [190,191] , and a recent
tion of reactive oxygen species (ROS) in the brain may exhaust MCI trial with antioxidants indicated that vitamin E ingestion
antioxidant capacity, including antioxidant vitamins, and lead to over 3 years has no benefit on the risk of progression to AD [192] .
the onset and progression of AD [169,170] . Therefore, anti­oxidant There are several reasons explaining this discrepancy. As recently
vitamins from dietary fruits and vegetables may also play a role reviewed [185,193] , the failure of these trials probably arises from a
in delaying the onset of AD. Some findings demonstrated that combination of factors, including not monitoring the drug levels
older African–Americans [177] and Japanese individuals living in and surrogate markers for the in vivo therapeutic effect of the drug
the USA have a much higher prevalence of AD (6.24 and 4.1%, of interest and starting the therapy very late in the disease stage.
respectively) [178] than those still living in their ethnic homelands In accordance with convergent data, oxidative stress is a very early
(<2%), suggesting that the prevalence of AD is more greatly influ- event in AD, occurring in presymptomatic phases, and so anti-
enced by diet and nutrition, environment and/or lifestyle, than by oxidant strategies should start years before the dementia age risk.
genetics. However, the simple comparison of prevalence data may The limited epidemiological evidence available on fruit and
be misleading, since there is large difference in selective survival vegetable consumption and cognition generally supported a
and other cultural effects. A more valuable comparison would be protective role of these macronutrients against cognitive decline
that of the overall dementia rate between different countries, after (Table 4) . Associations between greater intake of fruits and veg-
it had been culturally adjusted. In a cross-sectional study on AD, etables and better cognitive performance have been found in two
regression analyses have been performed in the 65+ age popula- cross-sectional studies [43,194] . In the Nurses’ Health Study, when
tions of 11 countries obtained from 18 community-wide studies fruit and vegetable intake in relation to cognitive function and
versus the components of the national diets [33,34] . The primary decline among aging women were prospectively followed from
finding is that fat and total caloric supply have the highest correla- 1976 to 2003 with biennial telephone questionnaires, women in
tions with AD prevalence rates. A significant inverse correlation the highest quintile of cruciferous vegetable consumption declined
was found between the fraction of calories derived from cereals slower compared with the lowest quintile [195] . In the same study,
and AD prevalence [33,34] . While whole grains have antioxidant women consuming the highest quantity of green leafy vegetables
vitamins and minerals, it is not clear whether the cereals generally also experienced slower decline than women consuming the small-
consumed are whole grains. Therefore, this relationship could be est amount [195] . These findings were consistent with those from a
due to the fact that countries with a low fat supply have a high subset of subjects participating in the Chicago Health and Aging
cereal supply, rather than to a direct therapeutic effect of the cere- Project (CHAP) [196] – a high vegetable consumption was associ-
als. These ecological findings on the possible role of cereals as a ated with a slower rate of cognitive decline over 6 years. Of the dif-
risk reduction factor were confirmed in a 4-year cohort study in ferent types of vegetables, green leafy vegetables had the strongest
New York (USA), the Washington Heights–Inwood Columbia association. Fruit consumption was not associated with cognitive
Aging Project (WHICAP), that also found that dietary fat was an change [196] . High intake of b carotene, flavonoids, vitamins C and
important risk factor for AD for those with the APOE e4 allele, E, thiamine and folate from dietary fruit and vegetables was also
but not for those without that allele [179] . associated with a lower risk of AD in the Rotterdam Study [186] .
Given the possible role of vascular factors in contributing to Furthermore, in the Kame Project cohort (a population-based
cognitive decline [21] , high intake of fruits and vegetables appear prospective study on 1836 Japanese–Americans aged 65 years or
to decrease the risk of CAD [180] and stroke [181] , and thus may older based in King County, WA, USA) follow-up surveys found
also decrease cognitive decline. In fact, bioactive compounds, that 63 participants had developed AD after an average of 6 years.
such as antioxidants, mostly contained in fruits and vegetables, After adjusting for age, sex, education and other relevant factors,
are important for the protection against oxidative and nitrosative those who reported drinking at least three glasses of juice a week
stress, and these micronutrients have been related with aging itself had a 76% lower risk of developing AD than those who reported
and the pathophysiology of some age-related illnesses, including drinking juice less than once a week [43] . Data from the 3C Study
cognitive impairment and dementia [182] . While a growing body of demonstrated that a diet rich in fish, n-3 PUFA-rich oils, fruits
knowledge demonstrates both the importance of oxidative stress and vegetables could contribute to decrease the risk of dementia
in the etiology of dementia and the efficacy of antioxidant treat- and AD in older persons, whereas consumption of n-6-rich oils
ment in animal and cellular models, studies in humans are pres- could exert detrimental effects when not counterbalanced by suf-
ently inconclusive [183,184] . In fact, epidemiological studies have ficient n-3 intake. These effects seem more pronounced among
demonstrated that dietary intake of natural or synthetic products APOE e4 noncarriers [73] . Finally, among the 3779 members of
with a putative antioxidant effect, mainly vitamin E, improve the Swedish Twin Registry who completed a diet questionnaire
cognitive function and reduce the risk of AD [185,186] . Moreover, approximately 30 years before cognitive screening and full clinical
anti­oxidative intervention studies in animal models of AD-like evaluation for dementia as part of the study of dementia in Swedish
amyloidosis demonstrate a significant reduction in oxidative stress, Twins (HARMONY) study, a medium or great proportion of
Ab deposition and behavioral improvements [187,188] . However, fruits and vegetables in the diet, compared with no or small, was
intervention trials of antioxidant supplementation have demon- associated with a decreased risk of dementia and AD. This effect
strated no major benefit against cognitive impairment [185,189] . In was observed among women and individuals with angina [197] .

www.expert-reviews.com 691
Review Solfrizzi, Panza, Frisardi et al.

Table 4. Principal longitudinal, clinical and epidemiological studies on the relationships between fruit and
vegetable intake and dementia (i.e., Alzheimer’s disease) or predementia syndromes (i.e., age-related
cognitive decline).
Study (year) Setting and Subjects Methods Results and conclusions Ref.
study design
(duration)
Kang et al. (2005) Longitudinal, 13,388 women, The TICS and five other cognitive tests High consumption of [195]
The Nurses’ Health population- aged 70 years (immediate and delayed recalls of the vegetables, but not fruit, was
Study, USA based and over EBMT, category fluency, delayed recall of associated with less cognitive
(10–16 years) the TICS and DSB) and evaluation of decline among older woman
dietary intake of food with a
semiquantitative FFQ
Morris et al. Longitudinal, 3718 subjects, Immediate and delayed recalls of the High vegetable, but not fruit, [196]
(2006) population- aged 65 years EBMT, MMSE, and SDMT and evaluation consumption was associated
Chicago Health and based and older of dietary intake of food with a with slower rate of cognitive
Aging Project (3–6 years) semiquantitative FFQ decline with older age
(CHAP), USA
Dai et al. (2006) Longitudinal, 3045 Japanese– Diagnosis of incident AD and evaluation Frequent drinking of fruit and [43]
The Kame Project, population- American of dietary intake of food with a vegetable juices was associated
USA based subjects, semiquantitative FFQ with a substantially decreased
(6.3 years) aged 65 or over risk of AD. This inverse
association was stronger after
adjustments for potential
confounding factors
Hughes et al. (2010) Longitudinal 3779 members Diagnosis of incident dementia and AD These findings suggested that [197]
The HARMONY (30 years) of the Swedish and evaluation of dietary habits higher fruit and vegetable
study, Twin Registry intake assessed at midlife was
Sweden associated with a lower risk of
dementia and later AD
AD: Alzheimer’s disease; DSB: Digit Span Backward; EBMT: East Boston Memory Test; FFQ: Food Frequency Questionnaire; MMSE: Mini-Mental State Examination;
SDMT: Symbol Digit Modalities Test; TICS: Telephone Interview for Cognitive Status.

One important unanswered question relates to the largely unex- involved in neurodegenerative processes. This is in accordance
plored relationship between intake of fruits and vegetables, anti- with previous finding of a protective effect for higher consump-
oxidant micronutrient status, a condition of oxidative stress and tion of dietary flavonoids against dementia [168] and cognitive
cognitive performance in healthy subjects [198] . In a very recent decline  [202] in the PAQUID study. However, in the full Adult
cross-sectional study, 193 healthy community dwellers consum- Changes in Thought (ACT) cohort, the use of supplemental
ing a diet rich in fruits and vegetables had higher plasma levels of vitamin E and C, alone or in combination, was not associated
lipophilic antioxidant micronutrients, lower levels of biomarkers with reduced risk for dementia over 5.5 years of follow-up [203] .
related to oxidative stress and better scores on neuropsycho­logical Examining brains from the same cohort, typical antioxidant sup-
evaluation compared with subjects with low intakes of fruits and plement use was not associated with suppressed basal or increased
vegetables. This result was independent of gender, education, levels of free radical damage to the cerebral cortex from AD,
BMI, lipid profile and albumin levels but, most importantly, was microvascular brain injury or smoking [204] . Evidence suggested
independent of age. Therefore, micronutrient levels appeared to that the combined intake of tocopherol forms may be important
be a good indicator for fruit and vegetable intake and important for AD protection, especially the combination of a- and g-tocoph-
in influencing the in vivo oxidant/antioxidant balance in healthy erol [205] ; thus, dietary sources of vitamin E may be preferred over
subjects [199] . Furthermore, individuals who consume a diet high supplementation. In contrast to supplemental antioxidant use,
in fruits and vegetables, as well as nuts or fish, would probably more evidence exists for a protective effect of dietary antioxidants
have other lifestyle characteristics that may be effective in reducing for AD development  [168,186,206] , although study results remain
AD or VaD risk (e.g., physical activity) [200] . conflicting. Therefore, future clinical trials for AD should con-
Fruits and vegetables reflect distinct food groups in the MeDi sider dietary sources rather than supplements, other antioxidants,
and in the American Diabetes Association food pyramid, and combinations or doses other than those used by ACT partici-
the benefits of these food groups could be provided through the pants [203] . Furthermore, there is growing evidence indicating that
antioxidant properties that foods in this class provide [201] . In fact, oxidative damage caused by the Ab peptide in the pathogenesis
fruits and vegetables are rich sources of antioxidant nutrients and of AD may be hydrogen peroxide (H2O2)-mediated [207] . Recent
bioactive compounds, such as flavonoids, that are thought to be studies have shown that flavonols, a class of flavonoids, from apple

692 Expert Rev. Neurother. 11(5), (2011)


Diet & Alzheimer’s disease Review

and citrus juices, such as quercetin, are able to cross the BBB [208] activator, a marker of endothelial dysfunction [201] . Furthermore,
and demonstrate neuro­protection against H2O2 [209] in human aside from their antioxidant properties, many polyphenols, such as
neuroblastoma cells, which may or may not behave like neurons quercetin, have potent anti-inflammatory properties [220] . Recent
in the intact brain, also preventing Ab-induced calcium influx clinical trials have demonstrated that intake of orange juice sig-
and apoptosis, each of which results, in part, owing to increased nificantly reduces plasma concentrations of F2-isoprostanes, a
ROS. The effect of flavonols from citrus is similar to vitamin C, valid biomarker of oxidative stress [221] . In addition to antioxidant
but quercetin from apple juice confers stronger neuroprotection compounds, fruit and vegetable juices may also possess other pro-
than vitamin C [210] . Early in vitro studies reported that polyphe- tective components, such as folate and minerals [222] , and a high
nol flavonols, such as quercetin, protect mammalian and bacterial serum level of folate was found to be associated with a decreased
cells from toxicity induced by H 2O2, but that a-tocopherol is risk of AD [223] .
not effective [211] . Recent in vitro studies demonstrate that many
polyphenols, including flavonols (e.g., quercetin and others), AD risk & the whole-diet & dietary pattern approaches
protect mouse hippocampal cells from oxidative glutamate and to the Mediterranean diet model
H2O2 toxicity [209] . Additional animal studies demonstrate that In the last 5 years, research interest on diet and nutrition in rela-
chronic administration of flavonols or apple juice protects against tion to the occurrence of AD has been focused in exploring whole
aging and cognitive impairment induced by lipopolysaccharide, types of foods (e.g., tomatoes, fruits and vegetables) and whole
genetic and dietary vitamin deficiency in animal models [212,213] . dietary patterns rather than a single nutrient [53] . Among the
However, the Nurses’ Health study [195] and the CHAP [196] found broad spectrum of dietary patterns analyzed with this aim, a
a protective effect for higher vegetable consumption, but not for special role has been assigned to the MeDi [51] . In fact, the MeDi
higher fruit consumption, against cognitive decline. A partial could be an interesting model to further study the association
explanation of this finding could be that vegetables contain more between dietary patterns and cognitive functioning, given the
vitamin E than fruits, and this antioxidant nutrient was previously suggested role of many components of this diet (MUFA, n-3
demonstrated to be inversely related to risk of cognitive decline in PUFA, fish and olive oil consumption, cereals and red wine) in
the CHAP study population [214] . This explanation is supported contrasting cognitive impairment. The evidence reviewed thus far
by the fact that green leafy vegetables (which generally contain is based on the hypothesis that individual nutrients contained in
more vitamin E than other vegetable types) had the strongest foods may have effects on predementia and dementia syndromes,
inverse relationship with cognitive decline in the CHAP [196] . neglecting the possible importance of the additive or synergistic
Vegetables, but not fruits, are also typically consumed with added effects of other nutrients within and across particular foods that
fats (e.g., salad dressings, mayonnaise, margarine or butter), and form part of a diet [33–38] . However, as seen extensively above,
fats increase the absorption of vitamin E and other fat soluble the results regarding isolated nutrients or foods appear to be
antioxidant nutrients, such as carotenoids and flavonoids [196] . conflicting. Compared with traditional single-food or nutrient
Other possible mechanisms linking fruit and vegetable intake methods, the whole-diet or dietary pattern approach is appeal-
with neuroprotection could be of vascular origin. In fact, a meta- ing for several reasons. Indeed, the analyses of single nutrients
ana­lysis demonstrated that regular consumption of fruits and ignore important interactions (additive, synergistic or antago-
vegetables decreased the risk for stroke [215] , which could explain nist effects) between components of diet and, more importantly,
their protective effect against the vascular component of demen- people do not eat isolated nutrients [224,225] . The evaluation of
tia. In addition to antioxidant and vascular mechanisms, anti- the adherence to a Mediterranean-style diet was conducted to
inflammatory mechanisms have been invoked as contributing develop scores or indexes named the ‘Mediterranean Diet Scale’
to the benefit of these food groups. In fact, compelling epide- or ‘Mediterranean Diet Score’ or ‘Mediterranean Adequacy Index’
miological data have repeatedly demonstrated that protracted or even ‘Mediterranean-Style Dietary Pattern Score’ based on
use of NSAID inhibitors of COX isoforms prior to the onset of a priori hypotheses and intended to be close to the traditional
dementia can substantially lower the risk of subsequently devel- one [224,226,227] . In particular, a scale indicating the degree of adher-
oping dementia (50% or more), especially in subjects carrying ence to the traditional MeDi was constructed by Trichopoulou
one or more e4 allele of the APOE [216] . However, clinical trials and colleagues [228] , revised to include fish intake [229] , and used
and other observational studies, including the ACT study [217] , in recent studies investigating cognitive decline on US, French
demonstrated no protection or promotion of AD [216] . In par- and Greek cohorts [62,230–236] . A value of 0 or 1 was assigned to
ticular, neuro­pathological findings from the ACT cohort indi- each of nine indicated components (vegetables, fruits, legumes,
cated that heavy use of NSAIDs was associated with increased cereals, the ratio of MUFA to SFA, alcohol and fish presumed to
neuritic plaque score, a hallmark feature of AD that contains be beneficial; meat and dairy products presumed to be detrimen-
Ab peptides, abnormal neurites and reactive glia [218] . Therefore, tal) with the use of a sex-specific median as the cut-off. The total
while epidemiological evidence have demonstated an inconsistent MeDi score ranged from 0 (minimal adherence to the traditional
protective effect of chronic NSAID use, some studies have dem- MeDi) to 9 (maximal adherence).
onstrated that dietary levels of fruit intake and of vitamin C are The use of diet-scoring systems, such as the MeDi score,
inversely related to levels of C-reactive protein (CRP) [219] , and have undeniable advantages in understanding the role of diet in
vegetable intake is inversely associated with tissue plasminogen chronic disease [237] . Scarmeas and colleagues, using data from

www.expert-reviews.com 693
Review Solfrizzi, Panza, Frisardi et al.

the WHICAP with an inclusive dietary score (MeDi score) but findings. Our first concern was on the comparison between the
studying a population with a substantial difference in dietary French and US findings. In fact, from a methodological point of
habits compared with Greek (European Prospective Investigation view, French data were left-truncated (in contrast to the US data),
into Cancer and Nutrition [EPIC]) [62] , Italian (ILSA) [60,61] , and and the results of the MeDi score of the 3C Study and WHICAP
French (3C Study) [234] cohorts, found that a higher adherence were noncomparable (the medians used as cut-offs are generally
to the MeDi is associated with a trend for reduced risk of inci- study-specific). Moreover, from a clinical point of view, the French
dent MCI and also with reduced risk of MCI progression to AD findings were on ARCD, which was measured by MMSE per-
(Table 5) [233] . Individuals older than 65 years of age were enrolled formance, and the US findings had incident MCI as a primary
in the WHICAP in 1992 and 1999 and have been followed on outcome [233] , which is a more structured clinical construct and
average for 4 years. Among individuals free from dementia at not just a cognitive test. The second concern regards the role of
baseline, 2258 individuals had a complete assessment of cognitive drop-out dependence on the outcome of interest that was not
functions and of dietary habits (Table 5) . These findings confirmed addressed, particularly because the slower MMSE performance
previous reports from the WHICAP in which higher adherence across time was only borderline significant, and follow-up was
to the MeDi reduced the risk of developing AD [230,231] , and was over 5 years. Féart and colleagues correctly acknowledged among
associated with lower mortality in AD (Table 5) [232] . They may potential limitations of their study that, as already reported in the
account for the complex biological interactions between differ- non-Mediterranean population of the WHICAP [230,233] , they
ent components of a composite dietary pattern, such as MeDi, found a relatively low ratio of MUFA to SFA [234] . The hallmark
that may be difficult to detect in analyses focusing on only the of the traditional MeDi is a high consumption of olive oil, leading
individual components. The potentially beneficial effect of the to a high ratio. In the 3C Study, it was verified that consumption
MeDi for cognitive decline may be the result of some of its indi- of olive oil was positively correlated with the diet score and with
vidual food components. For example, as seen above, potentially the ratio (data available on request). Nonetheless, these positive
beneficial effects for MCI and its progression to dementia and associations did not give any indication about the adherence to the
for AD or dementia have been reported for alcohol [46] , fish con- MeDi pattern. The ratio of MUFA to SFA intake in the context
sumption [42] , PUFA [42] and lower SFA [42] . Interestingly, in other of other parameters of the MeDi score could be very useful as a
studies, alcohol and PUFA have failed to be associated with protec- marker of adherence to the MeDi pattern.
tion against MCI, its progression to dementia and AD or demen- Other original findings from the WHICAP suggested that
tia [42,46] . These contrasting findings about the impact of MeDi both high Mediterranean-type diet adherence and participating
score or individual macronutrients on predementia and dementia in physical activity were independently associated with a lower
syndromes may suggest an approach not only confined to cogni- risk of AD over time [235] . Very recently, findings from the CHAP
tive skills, but extended to functional status and co­morbidity. suggested that another dietary index based on the traditional
However, we should not renounce a priori the work for a correct MeDi  [239] was associated with slower rates of cognitive decline,
estimate of the validity of the MeDi score for cognitive impair- while the Healthy Eating Index (HEI)-2005 dietary quality index,
ment as a criterion. While several prospective cohort studies have a 12-component measure reflecting diet-related recommendations
consistently indicated that adherence to the traditional MeDi is of the 2005 US Dietary Guidelines [240] , was not associated with
associated with longevity or specific health outcomes, no study has cognitive change [241] . Finally, in an ana­lysis of food combination
investigated the relative importance of individual components of from the WHICAP, a dietary pattern that explained variation
the MeDi score in the generation of this association. Very recent of AD-related nutrients strongly protective against the develop-
findings from the EPIC on mortality suggested that the con- ment of AD was identified. This dietary pattern reflected a diet
tribution of the nine components of the MeDi to the apparent rich in n-3 PUFA, n-6 PUFA, vitamin E and folate, but with
protective effect of the score assessing adherence to traditional lower SFA and vitamin B12 [242] . Furthermore, dietary habits of
Mediterranean diet is approximately additive. The dominant com- subjects adhering more to this dietary pattern were characterized
ponents of the MeDi score as a predictor of lower mortality were by a high intake of salad dressing, nuts, fish, tomatoes, poultry,
moderate consumption of alcohol, low consumption of meat and cruciferous vegetables, fruits and dark and green leafy vegetables
meat products, and high consumption of vegetables, fruits and and low intake of high-fat dairy, red meat, organ meat and butter.
nuts, olive oil and legumes [238] . However, the evidence about the Notwithstanding the undeniable advantages of the whole-diet
role of the whole MeDi on cognitive decline is still scarce [224] . approach, using such a score in a US and a multiethnic popula-
Recently, a report from the 3C Study from over a 5-year period tion may not adequately represent conformity with the traditional
in a sample of older community dwellers in France suggested that MeDi. Furthermore, these recent findings from the WHICAP
higher adherence to a MeDi was associated with slower cognitive further underlined the possible protective role against cognitive
decline (MMSE score), while it was not associated with a lower decline of both n-3 and n-6 PUFA intake and the detrimental role
risk of incident dementia or AD (Table 5) [234] . These findings of SFA. In fact, taken together with the recent findings from the
were compared with those of the WHICAP from the USA on the WHICAP and the ILSA [60,61,242] , it should be advisable to include
possible role of adherence to a Mediterranean dietary pattern on elevated PUFA (both n-3 and n-6) intake, and lower MUFA:SFA
the risk of MCI and AD [230,233] . However, some concern could ratio or SFA intake, in the food combination presumed to be
be expressed regarding the ana­lysis and discussion of the study beneficial against AD and cognitive decline.

694 Expert Rev. Neurother. 11(5), (2011)


Table 5. Principal epidemiological studies on the relationship between adherence to the Mediterranean diet and dementia (i.e.,
Alzheimer’ disease) or predementia syndromes (i.e., age-related cognitive decline and mild cognitive impairment) in older people.
Study (year) Setting and Subjects Dietary assessment Cognitive Results and conclusions Ref.
study design outcomes
(duration)

www.expert-reviews.com
Scarmeas et al. (2006) Longitudinal, 2258 subjects, Evaluation of dietary intake with Diagnosis of incident Higher adherence to MeDi reduced risk for [230]
Washington Heights-Inwood population- aged 65 years and a 61-item FFQ. A dietary AD probable AD, either with or without
Columbia Aging Project based over composite score (MeDi score) coexisting stroke
(WHICAP), USA (4 years) evaluated adherence to MeDi
Scarmeas et al. (2007) Longitudinal, 192 community- Evaluation of dietary intake with All-cause mortality Higher adherence to the MeDi was [232]
Washington Heights-Inwood population- based individuals, a 61-item FFQ. A dietary associated with lower mortality in AD. The
Columbia Aging Project based aged 65 years and composite score (MeDi score) gradual reduction in mortality risk for
(WHICAP), USA (4.4 years) older, who were evaluated adherence to MeDi higher MeDi adherence tertiles suggested
diagnosed with AD a possible dose–response effect
Psaltopoulou et al. (2008) Longitudinal, 732 subjects, Evaluation of dietary intakes with MMSE No significant association between MeDi [62]
European Prospective population- aged 60 years or a 150-item FFQ. A dietary score and MMSE scores, whereas a
Investigation into Cancer and based older composite score (MeDi score) statistically significant inverse association
nutrition (EPIC), Greece (median 8 years) evaluated adherence to MeDi was found between MMSE performance
and some individual dietary components,
such as seed oil or PUFA intakes
Scarmeas et al. (2009) Longitudinal, 2258 subjects, Evaluation of dietary intake with Incidence of MCI and Higher adherence to the MeDi was [233]
Washington Heights-Inwood population- aged 65 years and a 61-item FFQ. A dietary progression associated with a trend for reduced risk of
Columbia Aging Project based over composite score (MeDi score) from MCI to AD developing MCI and with reduced risk of
(WHICAP), USA (4.5 years) evaluated adherence to MeDi MCI progression to AD
Scarmeas et al. (2009) Longitudinal, 2247 subjects, Evaluation of dietary intake with Diagnosis of incident Both higher Mediterranean-type diet [235]
Washington Heights-Inwood population- aged 65 years and a 61-item FFQ. A dietary AD adherence and higher physical activity
Columbia Aging Project based over composite score (MeDi score) were independently associated with
(WHICAP), USA (5.4 years) evaluated adherence to MeDi. reduced risk for AD
Two slightly different versions of
the Godin leisure time exercise
questionnaire were also used
Féart et al. (2009) Longitudinal, 1410 subjects, Evaluation of dietary intakes with Cognitive Adherence to a Mediterranean-type diet [234]
Three-City Study, France population- aged 65 years and a FFQ. A dietary composite score performance was was associated with slower decline on the
based older (MeDi score) evaluated adherence assessed with MMSE, but not other cognitive tests, and
(5 years) to MeDi MMSE, IST, BVRT and was not associated with the risk for
FCSRT. incident dementia over 5 years of
Incident cases of follow-up
dementia were also
Diet & Alzheimer’s disease

diagnosed
AD: Alzheimer’s disease; BVRT: Benton Visual Retention Test; EBMT: East Boston Memory Test; FCSRT: Free and Cued Selective Reminding Test; FFQ: Food Frequency Questionnaire; HEI-2005: Healthy Eating
Index 2005; IST: Isaacs Set Test; MCI: Mild cognitive impairment; MeDi: Mediterranean diet; MMSE: Mini-Mental State Examination (global cognitive functioning); SDMT: Symbol Digit Modalities Test.
Review

695
Review Solfrizzi, Panza, Frisardi et al.

At present, no clear synergistic mecha-


Ref.

[236]

[241]
nism of the whole MeDi on predementia and
dementia syndromes has been proposed, but

AD: Alzheimer’s disease; BVRT: Benton Visual Retention Test; EBMT: East Boston Memory Test; FCSRT: Free and Cued Selective Reminding Test; FFQ: Food Frequency Questionnaire; HEI-2005: Healthy Eating
the magnitude of the association between
type diet was significantly associated with

them into the COX model did not change


high-sensitivity C reactive protein, fasting

Mediterranean-type diet and incident AD


different biological mechanisms could be
Alzheimer’ disease) or predementia syndromes (i.e., age-related cognitive decline and mild cognitive impairment) in older people.

Mediterranean diet was associated with

index was not associated with cognitive


A dietary index based on the traditional
Diagnosis of incident Greater adherence to a Mediterranean-

insulin, adiponectin or combinations of

contrast, the HEI-2005 dietary quality


evoked to explain the association between

Index 2005; IST: Isaacs Set Test; MCI: Mild cognitive impairment; MeDi: Mediterranean diet; MMSE: Mini-Mental State Examination (global cognitive functioning); SDMT: Symbol Digit Modalities Test.
lower risk for AD. Introduction of the
Table 5. Principal epidemiological studies on the relationship between adherence to the Mediterranean diet and dementia (i.e.,

slower rates of cognitive decline. By


foods or nutrients of the MeDi and poten-
tial neuroprotective properties. First of all,
vascular comorbidities, including diabetes,
Results and conclusions

hypertension, dyslipidemia and WMLs, have


been related to dementia, AD and MCI [21–23] .
Furthermore, the role of nutrition on cardio-
vascular events has been well documented [243],
and there is strong evidence relating the MeDi
change
to a lower risk of vascular risk factors, such
as dyslipidemia [244,245] , hypertension [244–246]
and CAD  [245,247] . Therefore, a protective
delayed recalls of the

effect of the MeDi against VaD, the vascular


EBMT, MMSE and

component of AD, or MCI variants particu-


Immediate and

larly related to CVD [248] could be expected.


outcomes

Nevertheless, nonvascular biological-mediat-


Cognitive

ing mechanisms (i.e., metabolic, oxidative and


SDMT

inflammatory) may be also responsible for the


AD

epidemiological MeDi–MCI associations [231] .


food with a semiquantitative FFQ;

In fact, although vascular variables are likely


diet-related recommendations of
Evaluation of dietary intake with

scores) evaluated adherence to


MeDi; the HEI-2005 evaluated

to be in the causal pathway between MeDi


Evaluation of dietary intake of
composite score (MeDi score)
evaluated adherence to MeDi

(MedDiet and MedDiet–wine

the 2005 Dietary Guidelines

and AD and should be considered as pos-


a dietary composite score

dietary quality reflecting


a 61-item FFQ. A dietary

sible mediators, when the WHICAP vascular


Dietary assessment

risk factors were considered in the models,


the association between MeDi and AD did
not change [231] . Furthermore, dementia and
predementia syndromes and the MeDi have
been associated with metabolic abnormalities.
Insulin resistance and subsequent hyperinsu-
linemia have been found to increase the risk of
aged 65 years and

aged 65 years and

AD and promote decline in memory and cog-


nitive dysfunction [249,250] . Furthermore, MCI
3790 subjects,
1219 subjects,

has been linked to the dysregulation of glucose


Subjects

and insulin homeostasis  [251,252] and diabe-


older

older

tes [253,254] . At the same time, higher adherence


to the MeDi seems to improve carbohydrate
metabolism, and in interventional studies has
study design
Setting and

been associated with significant reductions in


Longitudinal,

Longitudinal,
(duration)

population-

population-

plasma glucose levels [245,255] , serum insulin


(4 years)

(3 years)

levels and insulin resistance [255,256] .


based

based

Oxidative stress could be another bio-


logical mechanism relating dementia, pre-
Washington Heights-Inwood

dementia syndromes and the MeDi owing


Chicago Health and Aging

to its antioxidant properties [257] . A grow-


Columbia Aging Project

ing body of evidence implicated oxidative


Tangney et al. (2011)

Project (CHAP), USA

damage in the pathogenesis of AD, with


(WHICAP), USA
Gu et al. (2010)

increased presence of lipid peroxidation,


Study (year)

nitration, free carbonyls and nucleic acid


oxidation [169,258] . Higher oxidative stress has
also been clearly invoked in MCI [170,259,260] .
Complex phenols and many other substances

696 Expert Rev. Neurother. 11(5), (2011)


Diet & Alzheimer’s disease Review

with important antioxidant properties, such as olive oil [261] , wine, the direct effect of the MeDi or MeDi components (UFA, elevated
fruits and vegetables, vitamins C and E, and carotenoids [262–264] , fish consumption, moderate alcohol intake and fruit and vegetable
are found in high concentrations in the typical components of intake) or specific substances in MeDi components (antioxidant
the MeDi  [265] . Typical Mediterranean meals [266] or meals rich or anti-inflammatory compounds) that provide the protection,
in typical Mediterranean food elements, such as olive oil [267] but the higher adherence to the MeDi may be an indicator of
or pomegranate juice [268] , have been demonstrated to increase a complex set of favorable social and lifestyle factors. In fact,
enzymes with antioxidant properties, such as paraoxonase and very recent findings from the WHICAP demonstrated that high-
plasma carotenoids [266] . Food intervention studies with either sensitivity CRP, fasting insulin and adiponectin (as biomarkers of
different tomato products, a very commonly used food in metabolic abnormalities) did not mediate the association between
Mediterranean areas [265] , or a typical Mediterranean dish such MeDi and AD risk (Table 5) [236] . These results suggest that greater
as gazpacho – a cold vegetable soup that contains approximately adherence to MeDi may not be associated with reduced risk for
75% vegetables (tomato, cucumber, pepper), 2–10% olive oil AD via inflammatory or metabolic pathways.
and other minor components (onion, garlic, wine vinegar and
sea salt) [269] – have indicated significant reductions of markers of Expert commentary
oxidative stress, such as isoprostanes. Therefore, the MeDi could At present, there is no curative treatment for dementia and AD, or
be capturing the composite effect of dietary antioxidants and this a therapeutic approach to prevent the conversion of MCI to demen-
could, at least partially, explain the association with a lower risk tia. Given the very limited therapeutic value of drugs currently used
of dementia and predementia syndromes. Finally, inflammation in the treatment of cognitive impairment and dementia, it remains
is another mechanism involved in the pathogenesis of dementia necessary to potentially individualize new strategies able to prevent
and predementia syndromes that is, in general, reduced with a and to slow down the progression of pre­dementia and dementia
higher adherence to the MeDi [270,271] . In fact, inflammation is syndromes. Recently, higher adherence to a Mediterranean-type
another potential mechanism for AD pathogenesis [272,273] and diet was associated with cognitive decline, although the MeDi
has been found to be associated with a higher risk for AD, VaD combines several foods and nutrients already separately proposed
and cognitive decline  [28] , and links between MCI and higher as potential protective factors against dementia and predementia
inflammatory states have been also demonstrated [274–276] . In fact, syndromes. Epidemiological evidence suggested a possible associa-
CRP, an inflammatory marker that has been detected in SPs and tion among fish consumption, MUFA and PUFA (particularly,
NFTs in the brains of patients with AD [277,278] and is up­regulated n-3 PUFA) and reduced risk of cognitive decline and dementia.
in AD brains [279,280] and serum [281] , has been proposed as a pos- Furthermore, light-to-moderate alcohol use may be associated with
sible biomarker for AD [282] , MCI and nonamnestic MCI [283] . a reduced risk of incident dementia and AD, while for VaD, cogni-
Higher adherence to the MeDi has been associated with lower tive decline and predementia syndromes the current evidence is
CRP levels in both observational [244,284,285] and interventional only suggestive of a protective effect. Finally, some lines of evidence
studies  [255,256] . Furthermore, IL-6, a cytokine that mediates have suggested that older healthy subjects consuming a diet rich
inflammatory reactions, has been consistently detected in diffuse in fruits and vegetables have higher plasma levels of lipophilic
early SPs without neuritic pathological abnormalities of cortical antioxidant micronutrients, lower levels of biomarkers related to
regions of patients with AD [282] , and has been associated with oxidative stress and better scores on neuropsycho­logical evaluation
greater cognitive decline [286] and an increased risk of dementia compared with subjects with low intakes of fruits and vegetables.
during follow-up [287] . Tyrosol and caffeic acid, both found in On the basis of this evidence, the evaluation of the MeDi on cogni-
extra-virgin olive oil and in wine (which are essential components tive outcomes seems of particular interest and recent prospective
of the MeDi), have been demonstrated to significantly reduce studies focused on dementia and predementia syndromes appeared
IL-6 production from peripheral blood mononuclear cells of to be promising. In fact, higher adherence to a Mediterranean-
healthy volunteers [288] . In the ATTICA study, subjects in the type diet has been associated with slower cognitive decline, with a
highest tertile of MeDi adherence have been reported to have 17% trend towards a reduced risk for developing MCI, reduced risk of
lower IL-6 serum levels [244] , and in the Nurses’ Health Study, progression from MCI to AD, decreased risk for AD and a reduced
there was a 16% reduction in IL-6 levels for subjects belonging to all-cause mortality in AD patients. These findings suggested that
the upper quintile of a MeDi adherence index [285] . In a clinical adherence to the MeDi may affect not only the risk for AD, but
trial, as compared with subjects assigned to the control diet, the also that for predementia syndromes, probably influencing the
subjects assigned to the MeDi had significantly reduced levels of evolution of cognitive performances over time and subsequent
IL-6 [255] . Higher adherence to the MeDi is in general associated disease progression.
with a significant reduction in a series of other inflammatory However, while for the vascular hypothesis there was clear evi-
markers, including white blood cell counts and others [244] . Thus, dence about a protective role of MeDi and its nutrients in prevent-
it is possible that the MeDi may, at least partially, lower the risk for ing all cardiovascular conditions linked to dementia, for the other
dementia and predementia syndromes by affecting inflammatory possible mechanisms it is only possible to suggest hypothetical
processes. It is also possible that moderate lifestyles in general, biological pathways, taking into account the results from animal
which obviously vary according to different cultural environ- studies. Therefore, the lack of reproducibility in some results and
ments, protect from cognitive impairment. Thus, it may not be the speculative aspect of the biological pathways presented, suggest

www.expert-reviews.com 697
Review Solfrizzi, Panza, Frisardi et al.

caution. However, although it is difficult to define the real impact for AD in the existing literature may be a result of failure to con-
of these biological findings, these studies provided another possible sider possible additive and interactive (antagonistic or synergistic)
explanation of the neuroprotective effect of the MeDi and its micro- effects among nutritional components, which may be captured
and macro-nutrients that are not only linked to inflammatory and in a composite dietary pattern, such as the MeDi. Focusing on
oxidative-related mechanisms. In fact, the efficacy of nutritionally single MeDi macronutrients, recommendations regarding future
derived compounds as neuroprotective agents is increasingly sup- research on the effects of n-3 PUFA supplementation on pre­
ported by empirical evidence. Plant-derived molecules, including dementia syndromes and very mild AD include properly designed
polyphenols, have demonstrated neuroprotective activities in cell RCTs that are sufficiently powered and with an adequate length
culture and animal models. However, the molecular mechanisms (e.g., 3–5 years of follow-up). In particular, the future RCTs on
underlying their protective effects are generally not completely PUFA supplementation should address the effects of different
understood and further investigation will be essential to provide types of n-3 PUFA (i.e., DHA, EPA, arachidonic acid and total
links between MeDi nutrients and brain function in a mechanistic, n-3 PUFA), as well as the ratio of n-6 to n-3 PUFA. Finally,
dynamic and quantitative way. Based, in part, on the evidence dis- these RCTs should be designed to include a baseline assessment
cussed in this article, it appears possible that in the near future we of dietary n-3 and n-6 PUFA intake and to evaluate the effect
may consider employing dietary intervention in preventative and of dose, treatment duration and the sustainment of effect after
possibly therapeutic strategies for dementia and predementia syn- discontinuation of n-3 PUFA consumption. In particular, the
dromes. Nonetheless, at present, no definitive dietary recommenda- dose of n-3 PUFA that might work for prevention may be very
tions are possible. However, high levels of consumption of fats from high. Furthermore, moderate alcohol consumption seems to be
fish, vegetable oils, nonstarchy vegetables and low glycemic index associated with a decreased risk of predementia and dementia syn-
fruits, and a diet low in foods with added sugars and with moder- dromes, but, at present, it is still questionable whether the effect
ate wine intake should be encouraged. In fact, this dietary advice of alcohol may be due to social and lifestyle factors. Furthermore,
provides a variety of antioxidant nutrients, is in accordance with studies on the interaction between smoking and drinking alcohol
recommendations for lowering the risk of cardiovascular disease, have yielded inconclusive results [46] . The effect of alcohol on
obesity, diabetes and hypertension, and may open new routes for cognition may be modified by the presence of the APOE e4 allele,
the prevention and management of cognitive decline and dementia but the patterns of interaction suggested have yielded different
to supplement existing symptomatic approaches. results. Some studies investigating the effect of different types
of alcohol on the cognitive function found no beverage-specific
Five-year view differences. There is evidence that binge drinking is associated
In previous years, extensive research has increased our knowledge with negative cardiovascular outcomes, and possibly also with
of the etiology of AD, other dementing disorders and pre­dementia an increased risk of dementia. The role of drinking patterns for
syndromes and several hypotheses have already emerged from the the development of cognitive impairment needs to receive more
epidemiological research. The reviewed evidence supports the attention in the future: trying to understand whether starting to
hypothesis that dietary-related factors may be associated with the drink at a later age would be beneficial and the role of the interac-
development of dementia and predementia syndromes in late life, tion between APOE e4 allele and alcohol consumption. Finally,
opening new avenues for the prevention of these diseases. However, the long-term effect of alcohol drinking on cognitive functions
many questions remain for future research, addressing possible has not been extensively investigated. Therefore, we need studies
confounders. For example, if a change of dietary habits may play to collect information on alcohol drinking collected from 5 to
a major part, what is the role of genetic risk factors, and will these 20 years earlier, while also examining possible interaction with
associations valid in populations with different dietary patterns? APOE e4 and other lifestyle factors.
Other questions address the underlying mechanisms and which is In future studies on the MeDi and AD, it would be indicated,
the most relevant component, among dietary micro- and macro- along with measuring this effect by a dietary composite score, to
nutrients and their possible interactions. Answers to these questions also report the estimates and the impact of the individual compo-
will help us to better define the target populations for future preven- nents of the diet (particularly fats) versus the whole-diet approach,
tive and therapeutic strategies. Therefore, in the future, addressing to better understand if the whole MeDi is important and not as
possible interactions in different populations with genetic factors only sources of beneficial fats (e.g., MUFA and n-3 PUFA). At
and other possible confounders, and whether change in dietary pat- present, only the EPIC cohort has been studied for a whole-diet
terns may protect against cognitive decline and AD would require approach, plus individual nutrients ana­lysis, with no significant
longitudinal studies on larger populations and RCTs. association found between MeDi score and global cognitive scores,
Among different dietary patterns, studies in support of MeDi whereas a statistically significant inverse association was found
as an optimal diet for prevention of cardiovascular and major between cognitive performance and some individual dietary com-
chronic diseases has rapidly evolved. As discussed extensively ponents [62] . In a very recent reana­lysis from the ILSA cohort, high
above, the association between a high adherence to the MeDi and PUFA were associated with reduced risk of incident MCI among
lower risk of AD and cognitive decline may be mediated by the those who consumed a low MUFA:SFA ratio intake [289] . In fact,
composite effect of some of its beneficial components. Some of while an increasing body of evidence suggested that elevated fish
the inconsistencies regarding the above dietary elements and risk consumption and high intake of n-3 PUFA may be protective

698 Expert Rev. Neurother. 11(5), (2011)


Diet & Alzheimer’s disease Review

against ARCD and MCI, the traditional Cretan diet, although the MeDi as a potential preventive approach against dementia and
strongly dependent on high olive oil intake, was never centred on AD. Recommendations regarding future research on the effects of
fish consumption [290] . In this context, n-6 PUFA could poten- adherence to the MeDi on predementia and dementia syndromes
tially exert some health benefits. In fact, in the MeDi, some foods include properly designed RCTs that are sufficiently powered and
are rich in n-6 PUFA (e.g., walnuts, almonds and hazelnuts), while with an adequate length, although this approach could have some
other foods, although poor in n-6 PUFA, are highly consumed, potential limitations. In fact, given the long preclinical phase of
such as cereals, legumes and, in a lesser amount, some types of dementia, it may be difficult to execute appropriate RCTs over a
meat (pork) and poultry. Furthermore, olive oil contains n-6 and long enough time period and with large enough samples to draw
n-3 PUFA in a ratio of 10:1. Therefore, it should be advisable to accurate and repeatable conclusions.
include PUFA in the MeDi score as well as the other individual
macronutrients (i.e., MUFA:SFA ratio), among the components Financial & competing interests disclosure
presumed to be beneficial, in evaluating the relationship between This work was supported by the Italian Longitudinal Study on Aging (ILSA;
adherence to the MeDi and ARCD or MCI. Future studies on this Italian National Research Council – CNR-Targeted Project on Ageing –
topic should probably address different constructs of predementia Grants 9400419PF40 and 95973PF40) and Ministero della Salute, IRCCS
syndromes and subtypes of MCI, while also accounting for the Research Program 2009–2011, Line 2: “Malattie complesse”. The authors
possible role of the APOE e4 allele, a major genetic risk factor for have no other relevant affiliations or financial involvement with any organiza-
AD, in modifying the possible associations between the MeDi tion or entity with a financial interest in or financial conflict with the subject
and cognitive decline. Further research is also needed to allow the matter or materials discussed in the manuscript apart from those disclosed.
generalization of these results to other populations and to propose No writing assistance was utilized in the production of this manuscript.

Key issues
• Vascular- and dietary-related factors appeared to be associated with increased risk or protection against predementia syndromes and
Alzheimer’s disease (AD).
• Among dietary factors, epidemiological evidence suggested a possible association among fish consumption, monounsaturated fatty
acids and polyunsaturated fatty acids (PUFA; particularly, n-3 PUFA) and reduced risk of cognitive decline and dementia.
• Limited epidemiological evidence suggested that poorer cognitive function and an increased risk for vascular dementia (VaD) were
found to be associated with a lower consumption of milk or dairy products, while whole-fat dairy products may be associated with
late-life cognitive decline.
• Light-to-moderate alcohol use may be associated with a reduced risk of incident dementia and AD, while for VaD, cognitive decline and
predementia syndromes, the current evidence is only suggestive of a protective effect.
• The limited epidemiological evidence available on fruit and vegetable consumption and cognition generally supported a protective role
of these macronutrients against cognitive decline, dementia and AD.
• Among different dietary patterns, recent prospective studies provided evidence that a higher adherence to a Mediterranean-type diet
could be associated with slower cognitive decline, reduced risk of progression from MCI to AD, reduced risk of AD and decreased
all-cause mortality in AD patients.
• Although this impressive amount of convincing epidemiological evidence, at present, no definitive dietary recommendations for
preventing dementia and AD are possible.
• In accordance with recommendations for lowering the risk of cardiovascular and metabolic disorders, high levels of consumption of fats
from fish, vegetable oils, nonstarchy vegetables and low glycemic index fruits, and a diet low in foods with added sugars and with
moderate wine intake should be encouraged, possibly opening new routes for the prevention and management of cognitive decline
and dementia.

References 4 Cummings JL. Alzheimer’s disease. 8 Lesne S, Koh MT, Kotilinek L et al.
Papers of special note have been highlighted as: N. Engl. J. Med. 351(1), 56–67 (2004). A specific amyloid-b protein assembly in
• of interest 5 Alladi S, Xuereb J, Bak T et al. Focal the brain impairs memory. Nature
•• of considerable interest cortical presentations of Alzheimer’s 440(7082), 352–357 (2006).
1 Qiu C, De Ronchi D, Fratiglioni L. disease. Brain 130(Pt 10), 2636–2645 9 Shankar GM, Li S, Mehta TH et al.
The epidemiology of the dementias: an (2007). Amyloid-b protein dimers isolated directly
update. Curr. Opin. Psychiatry 20(4), 6 Walter J, Kaether C, Steiner H, Haass C. from Alzheimer's brains impair synaptic
380–385 (2007). The cell biology of Alzheimer’s disease: plasticity and memory. Nat. Med. 14(8)
2 Alzheimer’s Association. Alzheimer’s uncovering the secrets of secretases. Curr. 837–842 (2008).
disease facts and figures. Alzheimers Opin. Neurobiol. 11(5), 585–590 (2001). 10 Panza F, D’Introno A, Colacicco AM et al.
Dement. 6(2), 158–194 (2010). 7 Hardy J, Allsop D. Amyloid deposition as Current epidemiology of mild cognitive
3 Brookmeyer R, Johnson E, the central event in the aetiology of impairment and other predementia
Ziegler-Graham K, Arrighi HM. Forecasting Alzheimer’s disease. Trends Pharmacol. Sci. syndromes. Am. J. Geriatr. Psychiatry
the global burden of Alzheimer’s disease. 12(10), 383–388 (1991). 13(8), 633–644 (2005).
Alzheimers Dement. 3(3), 186–191 (2007).

www.expert-reviews.com 699
Review Solfrizzi, Panza, Frisardi et al.

11 Winblad B, Palmer K, Kivipelto M et al. 25 Qiu C, Winblad B, Fratiglioni L. The age- 38 Frisardi V, Panza F, Seripa D et al.
Mild cognitive impairment – beyond dependent relation of blood pressure to Nutraceutical properties of Mediterranean
controversies, towards a consensus: report cognitive function and dementia. Lancet diet and cognitive decline: possible
of the International Working Group on Neurol. 4(8), 487–499 (2005). underlying mechanisms. J. Alzheimers Dis.
Mild Cognitive Impairment. J. Intern. 26 Anstey KJ, Lipnicki DM, Low LF. 22(3), 715–740 (2010).
Med. 256(3), 240–246 (2004). Cholesterol as a risk factor for dementia 39 Solfrizzi V, D’Introno A, Colacicco AM
12 Dubois B, Feldman HH, Jacova C et al. and cognitive decline: a systematic review et al. Dietary fatty acids intake: possible
Research criteria for the diagnosis of of prospective studies with meta-analysis. role in cognitive decline and dementia.
Alzheimer’s disease: revising the Am. J. Geriatr. Psychiatry. 16(5), 343–354 Exp. Gerontol. 40(4), 257–270 (2005).
NINCDS–ADRDA criteria. Lancet Neurol. (2008). 40 Assisi A, Banzi R, Buonocore C et al. Fish
6(8), 734–746 (2007). 27 Luchsinger JA, Gustafson DR. Adiposity, oil and mental health. the role of n-3
13 Dubois B, Feldman HH, Jacova C et al. Type 2 diabetes, and Alzheimer’s disease. long-chain polyunsaturated fatty acids in
Revising the definition of Alzheimer’s J. Alzheimers Dis. 16(4), 693–704 (2009). cognitive development and neurological
disease: a new lexicon. Lancet Neurol. 28 Frisardi V, Solfrizzi V, Seripa D et al. disorders. Int. Clin. Psychpharmacol. 21(6),
9(11), 1118–1127 (2010). Metabolic-cognitive syndrome: a cross-talk 319–336 (2006).
14 Sachdev P. Vascular cognitive disorder. between metabolic syndrome and 41 Cunnane SC, Plourde M, Pifferi F,
Int. J. Geriatr. Psychiatry 14(5), 402–403 Alzheimer’s disease. Ageing Res. Rev. 9(4), Bégin M, Féart C, Barberger-Gateau P.
(1999). 399–417 (2010). Fish, docosahexaenoic acid and Alzheimer‘s
15 Roman GC, Sachdev P, Royall DR et al. 29 Kalaria RN. Vascular basis for brain disease. Prog. Lipid Res. 48(5), 239–256
Vascular cognitive disorder: a new diagnostic degeneration: faltering controls and risk (2009).
category updating vascular cognitive factors for dementia. Nutr. Rev. 42 Solfrizzi V, Frisardi V, Capurso C et al.
impairment and vascular dementia. 68(Suppl. 2) S74–S87 (2010). Dietary fatty acids in dementia and
J. Neurol. Sci. 226(1–2), 81–87 (2004). 30 Panza F, Frisardi V, Capurso C et al. predementia syndromes: epidemiological
16 Langa KM, Foster NL, Larson EB. Mixed Late-life depression, mild cognitive evidence and possible underlying
dementia: emerging concepts and impairment, and dementia: possible mechanisms. Ageing Res. Rev. 9(2),
therapeutic implications. JAMA 292(23), continuum? Am. J. Geriatr. Psychiatry 184–199 (2010).
2901–2908 (2004). 18(2), 98–116 (2010). 43 Dai Q, Borenstein A, Wu Y, Jackson JC,
17 Lim A, Tsuang D, Kukull W et al. 31 Kramer AF, Hahn S, Cohen NJ et al. Larson EB. Fruit and vegetable juices and
Clinico-neuropathological correlation of Ageing, fitness and neurocognitive Alzheimer’s disease. the Kame project.
Alzheimer’s disease in a community-based function. Nature 400(6743), 418–419 Am. J. Med. 119(9), 751–759 (2006).
case series. J. Am. Geriatr. Soc. 47(5), (1999). 44 Parrott MD, Greenwood CE. Dietary
564–569 (1999). 32 Ngandu T, von Strauss E, Helkala EL et al. influences on cognitive function with aging.
18 Sonnen JA, Postupna N, Larson EB et al. Education and dementia what lies beneath From high-fat diets to healthful eating.
Pathologic correlates of dementia in the association? Neurology 69(14), Ann. NY Acad. Sci. 1114, 389–397 (2007).
individuals with Lewy body disease. Brain 1442–1450 (2007). 45 Peters R, Peters J, Warner J, Beckett N,
Pathol. 20(3), 654–659 (2010). 33 Solfrizzi V, Panza F, Capurso A. The role of Bulpitt C. Alcohol, dementia and cognitive
19 Frisardi V, Solfrizzi V, Imbimbo BP et al. diet in cognitive decline. J. Neural Transm. decline in the elderly. a systematic review.
Towards disease-modifying treatment of 110(1), 95–110 (2003). Age Ageing 37(5), 505–512 (2008).
Alzheimer’s disease: drugs targeting 34 Grant WB. Dietary links to Alzheimer’s 46 Panza F, Capurso C, D’Introno A et al.
b-amyloid. Curr. Alzheimer Res. 7(1), disease: 1999 update. J. Alzheimers Dis. Alcohol drinking, cognitive functions in
40–55 (2010). 1(4–5), 197–201 (1999). older age, predementia, and dementia
20 Panza F, Solfrizzi V, Colacicco AM et al. syndromes. J. Alzheimers Dis. 17(1), 7–31
• Update on a cross-sectional study on (2009).
Mediterranean diet and cognitive decline. Alzheimer’s disease (AD), suggesting that
Public Health Nutr. 7(7), 959–963 (2004). 47 Keys A. Seven Countries: A Multivariate
fat and total caloric supply have the
21 Solfrizzi V, Capurso C, D’Introno A et al. Analysis of Death and Coronary Heart
highest correlations with AD prevalence
Lifestyle-related factors in predementia and Disease. Harvard University Press, MA,
rates. A significant inverse correlation was
dementia syndromes. Expert Rev. USA (1980).
found between the fraction of calories
Neurother. 8(1), 133–158 (2008). derived from cereals and AD prevalence. 48 Serra-Majem L, Roman B, Estruch R.
22 Peters R. The prevention of dementia. Scientific evidence of interventions using
35 Luchsinger JA, Mayeux R. Dietary factors the Mediterranean diet: a systematic
Int. J. Geriatr. Psychiatry 24(5), 452–458 and Alzheimer’s disease. Lancet Neurol.
(2009). review. Nutr. Rev. 64(2 Pt 2), S27–S47
3(10), 579–587 (2004). (2006).
23 de la Torre JC. Alzheimer’s disease is 36 Luchsinger JA, Noble JM, Scarmeas N.
incurable but preventable. J. Alzheimers 49 De Lorgeril M, Salen P, Martin J,
Diet and Alzheimer’s disease. Curr. Neurol. Monjaud I, Delaye J, Mamelle N.
Dis. 20(3), 861–870 (2010). Neurosci. Rep. 7(5), 366–372 (2007). Mediterranean diet, traditional risk factors
24 Frisardi V, Solfrizzi V, Capurso C et al. Coley N, Andrieu S, Gardette V et al.
37 and the rate of cardiovascular
Aluminum in the diet and Alzheimer’s Dementia prevention: methodological complications after myocardial infarction:
disease: from current epidemiology to explanations for inconsistent results. final report of the Lyon diet heart study.
possible disease-modifying treatment. Epidemiol. Rev. 30, 35–66 (2008). Circulation 99(6), 799–785 (1999).
J. Alzheimers Dis. 20(1), 17–30 (2010).

700 Expert Rev. Neurother. 11(5), (2011)


Diet & Alzheimer’s disease Review

50 Willett WC, Sacks F, Trichopoulou A et al. cognitive impairment among elders: the among older people in the UK: baseline
Mediterranean diet pyramid: a cultural EPIC-Greece cohort (European Prospective data from the Opal Study. J. Nutr. Health
model for healthy eating. Am. J. Clin. Nutr. Investigation into Cancer and Nutrition). Aging 13(3), 198–202 (2009).
61(6 Suppl.), S1402–S1406 (1995). Public Health Nutr. 11(10), 1054–1062 75 Morris MC, Evans DA, Tangney CC,
51 Sofi F. The Mediterranean diet revisited: (2008). Bienias JL, Wilson RS. Fish consumption
evidence of its effectiveness grows. 63 Eskelinen MH, Ngandu T, Helkala EL and cognitive decline with age in a large
Curr. Opin. Cardiol. 24(5), 442–446 et al. Fat intake at midlife and cognitive community study. Arch. Neurol. 62(12),
(2009). impairment later in life: a population-based 1849–1853 (2005).
52 Gomez-Pinilla F. Brain foods: the effects of CAIDE study. Int. J. Geriatr. Psychiatry 76 Nurk E, Drevon CA, Refsum H et al.
nutrients on brain function. Nat. Rev. 23(7), 741–747 (2008). Cognitive performance among the elderly
Neurosci. 9(7), 568–578 (2008). 64 Vercambre MN, Boutron-Ruault MC, and dietary fish intake: the Hordaland
53 Sofi F, Macchi C, Abbate R, Gensini GF, Ritchie K, Clavel-Chapelon F, Berr C. Health Study. Am. J. Clin. Nutr. 86(5),
Casini A. Effectiveness of the Long-term association of food and nutrient 1470–1478 (2007) .
Mediterranean diet: can it help delay or intakes with cognitive and functional 77 van Gelder BM, Tijhuis M, Kalmijn S,
prevent Alzheimer’s disease? J. Alzheimers decline: a 13-year follow-up study of elderly Kromhout D. Fish consumption, n-3 fatty
Dis. 20(3), 795–801 (2010). French women. Br. J. Nutr. 102(3), acids, and subsequent 5-y cognitive decline
419–427 (2009). in elderly men: the Zutphen Elderly Study.
54 Solfrizzi V, Frisardi V, Seripa D et al.
Mediterranean diet in predementia and 65 Berr C, Portet F, Carriere I et al. Olive oil Am. J. Clin. Nutr. 85(4), 1142–1147
dementia syndromes. Curr. Alzheimer Res. and cognition: results from the Three-City (2007).
(2011) (In press). study. Dement. Geriatr. Cogn. Disord. 78 Huang TL, Zandi PP, Tucker KL et al.
28(4), 357–364 (2009). Benefits of fatty fish on dementia risk are
55 Ortega RM, Requejo AM, Andres P et al.
Dietary intake and cognitive function in a 66 Kalmijn S, Launer LJ, Ott A, stronger for those without APOE e4.
group of elderly people. Am. J. Clin. Nutr. Witteman JC, Hofman A, Breteler MM. Neurology 65(9), 1409–1414 (2005) .
66(4), 803–809 (1997). Dietary fat intake and the risk of incident 79 Panza F, Frisardi V, Capurso C et al.
dementia in the Rotterdam study. Polyunsaturated fatty acid and
56 Solfrizzi V, Panza F, Torres F et al.
Ann. Neurol. 42(5), 776–782 (1997). S-adenosylmethionine supplementation in
High monounsaturated fatty acids intake
protects against age-related cognitive 67 Engelhart MJ, Geerlings MI, Ruitenberg A predementia syndromes and Alzheimer’s
decline. Neurology 52(8), 1563–1569 (1999). et al. Diet and risk of dementia: does fat disease: a review. ScientificWorldJournal 9,
matter? The Rotterdam Study. Neurology 373–389 (2009).
57 Kalmijn S, Feskens EJ, Launer LJ,
59(12), 1915–1921 (2002). 80 Freund-Levi Y, Eriksdotter-Jönhagen M,
Kromhout D. Polyunsaturated fatty acids,
antioxidants, and cognitive functions in 68 Luchsinger JA, Tang MX, Shea S, Cederholm T et al. w-3 fatty acid treatment
very old men. Am. J. Epidemiol. 145(1), Mayeux R. Caloric intake and the risk of in 174 patients with mild to moderate
33–41 (1997). Alzheimer disease. Arch. Neurol. 59(8), Alzheimer disease: Omegad Study:
1256–1263 (2002). a randomized double-blind trial.
58 Morris MC, Evans DA, Bienias JL,
69 Barberger-Gateau P, Letenneur L, Arch. Neurol. 63(10), 1402–1408 (2006).
Tangney CC, Bennett DA, Wilson RS.
Dietary fat intake and 6-year cognitive Deschamps V, Peres K, Dartigues JF, • In this randomized double-blind trial,
change in an older biracial community Renaud S. Fish, meat, and risk of dementia: after the treatment period the authors
population. Neurology 62(9), 1573–1579 cohort study. BMJ 325(7370), 932–933 found that the supplementation did not
(2004). (2002). delay the rate of cognitive decline but, in
59 Kalmijn S, van Boxtel MP, Ocke M, 70 Morris MC, Evans DA, Bienias JL et al. the group of 32 patients with the most
Verschuren WM, Kromhout D, Launer LJ. Dietary fats and the risk of incident mild AD (Mini-Mental State
Dietary intake of fatty acids and fish in Alzheimer disease. Arch. Neurol. 60(2), Examination [MMSE] >27, Clinical
relation to cognitive performance at middle 194–200 (2003). Dementia Rating Score 0.5–1), n-3
age. Neurology 62(2), 275–280 (2004). 71 Morris MC, Evans DA, Bienias JL et al. polyunsaturated fatty acid
60 Solfrizzi V, Colacicco AM, D’Introno A Consumption of fish and n-3 fatty acids supplementation slowed the decline in
et al. Dietary intake of unsaturated fatty and risk of incident Alzheimer disease. MMSE scores.
acids and age-related cognitive decline: Arch. Neurol. 60(7), 940–946 (2003). 81 Kotani S, Sakaguchi E, Warashina S et al.
a 8.5-year follow-up of the Italian 72 Laitinen MH, Ngandu T, Rovio S et al. Dietary supplementation of arachidonic
Longitudinal Study on Aging. Neurobiol. Fat intake at midlife and risk of dementia and docosahexaenoic acids improves
Aging 27(11), 1694–1704 (2006). and Alzheimer’s disease: a population- cognitive dysfunction. Neurosci. Res. 56(2),
61 Solfrizzi V, Colacicco AM, D’Introno A based study. Dement. Geriatr. Cogn. Disord. 159–164 (2006).
et al. Dietary polyunsaturated fatty acids 22(1), 99–107 (2006). 82 Chiu CC, Su KP, Cheng TC et al.
intakes and rate of mild cognitive 73 Barberger-Gateau P, Raffaitin C, The effects of omega-3 fatty acids
impairment. The Italian Longitudinal Letenneur L et al. Dietary patterns and risk monotherapy in Alzheimer’s disease and
Study on Aging. Exp. Gerontol. 41(6), of dementia: the Three-City cohort study. mild cognitive impairment: a preliminary
619–627 (2006). Neurology 69(20), 1921–1930 (2007). randomized double-blind placebo-
62 Psaltopoulou T, Kyrozis A, Stathopoulos P, 74 Dangour AD, Allen E, Elbourne D, controlled study. Prog.
Trichopoulos D, Vassilopoulos D, Fletcher A, Richards M, Uauy R. Fish Neuropsychopharmacol. Biol. Psychiatry
Trichopoulou A. Diet, physical activity and Consumption and cognitive function 32(6), 1538–1544 (2008).

www.expert-reviews.com 701
Review Solfrizzi, Panza, Frisardi et al.

83 Freund-Levi Y, Basun H, Cederholm T 94 Park IH, Hwang EM, Hong HS et al. 105 Rahman A, Sawyer Baker P, Allman RM,
et al. Omega-3 supplementation in mild to Lovastatin enhances Ab production and Zamrini E. Dietary factors and cognitive
moderate Alzheimer’s disease: effects on senile plaque deposition in female Tg2576 impairment in community-dwelling
neuropsychiatric symptoms. Int. J. Geriatr. mice. Neurobiol. Aging 24(5), 637–643 elderly. J. Nutr. Health Aging 11(1), 49–54
Psychiatry 23(2), 161–169 (2008). (2003). (2007).
84 van de Rest O, Geleijnse JM, Kok FJ et al. 95 Lim GP, Calon F, Morihara T et al. A diet 106 Avila-Funes JA, Garant MP,
Effect of fish-oil supplementation on enriched with the omega-3 fatty acid Aguilar-Navarro S. Relationship between
mental well-being in older subjects: docosahexaenoic acid reduces amyloid determining factors depressive symptoms
a randomized, double-blind, placebo- burden in an aged Alzheimer mouse model. and for dietary habits in older adults in
controlled trial. Am. J. Clin. Nutr. 88(3), J. Neurosci. 25(12), 3032–3040 (2005). Mexico. Rev. Panam. Salud Publica 19(5),
706–713 (2008). 96 Oksman M, Iivonen H, Hogyes E et al. 321–330 (2006).
85 van de Rest O, Geleijnse JM, Kok FJ et al. Impact of different saturated fatty acid, 107 Almeida OP, Norman P, Hankey G,
Effect of fish oil on cognitive performance polyunsaturated fatty acid and cholesterol Jamrozik K, Flicker L. Successful mental
in older subjects: a randomized, controlled containing diets on b-amyloid health aging: results from a longitudinal
trial. Neurology 71(6), 430–438 (2008). accumulation in APP/PS1 transgenic mice. study of older Australian men. Am. J.
86 Greenwood CE, Winocur G. Cognitive Neurobiol. Dis. 23(3), 563–572 (2006). Geriatr. Psychiatry 14(1), 27–35 (2006).
impairment in rats fed high-fat diets: 97 Molteni R, Barnard RJ, Ying Z, 108 Yamada M, Kasagi F, Sasaki H,
a specific effect of saturated fatty acid Roberts CK, Gómez-Pinilla F. A high-fat, Masunari N, Mimori Y, Suzuki G.
intake. Behav. Neurosci. 110(3), 451–459 refined sugar diet reduces hippocampal Association between dementia and midlife
(1996). brain-derived neurotrophic factor, neuronal risk factors: the radiation effects research
87 Moriguchi T, Greiner RS, Salem N. plasticity, and learning. Neuroscience foundation adult health study. J. Am.
Behavioral deficits associated with dietary 112(4), 803–814 (2002). Geriatr. Soc. 51(3), 410–414 (2003).
induction of decreased brain 98 Ambring A, Friberg P, Axelsen M et al. 109 Zemel MB. Role of calcium and dairy
docosahexaenoic acid concentration. Effects of a Mediterranean-inspired diet on products in energy partitioning and weight
J. Neurochem. 75(6), 2563–2573 (2000). blood lipids; vascular function and management. Am. J. Clin. Nutr. 79(5),
88 Hashimoto M, Tanabe Y, Fujii Y, Kikuta T, oxidative stress in healthy subjects. S907–S912 (2004).
Shibata H, Shido O. Chronic Clin. Sci. 106(5), 519–525 (2004). 110 Zemel MB, Teegarden D, Van Loan MD
administration of docosahexaenoic acid 99 Scientific Advisory Committee on et al. Dairy-rich diets augment fat loss on
ameliorates the impairment of spatial Nutrition and the Committee on Toxicity. an energy-restricted diet: a multicenter
cognition learning ability in amyloid Advice on Fish Consumption: Benefits and trial. Nutrients 1(1), 83–100 (2009).
{b}-infused rats. J. Nutr. 135(3), 549–555 Risks. The Stationery Office, London, UK 111 Teegarden D. The influence of dairy
(2005). (2004). product consumption on body
89 Hashimoto M, Hossain S, Shimada T et al. 100 Annweiler C, Allali G, Allain P et al. composition. J. Nutr. 135(12), 2749–2752
Docosahexaenoic acid provides protection Vitamin D and cognitive performance in (2005).
from impairment of learning ability in adults: va systematic review. Eur. J. Neurol. 112 Teegarden D, Donkin SS. Vitamin D:
Alzheimer’s disease model rats. 16(10), 1083–1089 (2009). emerging new roles in insulin sensitivity.
J. Neurochem. 81(5), 1084–1091 (2002). 101 Annweiler C, Schott AM, Allali G et al. Nutr. Res. Rev. 22(1), 82–92 (2009).
90 Calon F, Lim GP, Yang F et al. Association of vitamin D deficiency with 113 Alonso A, Nettleton JA, Ix JH et al.
Docosahexaenoic acid protects from cognitive impairment in older women. Dietary phosphorus, blood pressure, and
dendritic pathology in an Alzheimer’s Cross-sectional study. Neurology 74(1), incidence of hypertension in the
disease mouse model. Neuron 43(5), 27–32 (2010). atherosclerosis risk in communities study
633–645 (2004). 102 Friedland RP. Fish consumption and the and the multi-ethnic study of
91 Florent S, Malaplate-Armand C, Youssef I risk of Alzheimer disease: is it time to make atherosclerosis. Hypertension 55(3),
et al. Docosahexaenoic acid prevents dietary recommendations? Arch. Neurol. 776–784 (2010).
neuronal apoptosis induced by soluble 60(7), 923–924 (2003). 114 Weglicki WB, Mak IT, Kramer JH et al.
amyloid oligomers. J. Neurochem. 96(2), 103 Crichton GE, Bryan J, Murphy KJ, Role of free radicals and substance P in
385–395 (2006). Buckley J. Review of dairy consumption magnesium deficiency. Cardiovasc. Res.
92 Cole GM, Lim GP, Yang F et al. Prevention and cognitive performance in adults: 31(5), 677–682 (1996).
of Alzheimer’s disease: omega-3 fatty acid findings and methodological issues. 115 Singh RB, Rastogi SS, Sharma VK,
and phenolic anti-oxidant interventions. Dement. Geriatr. Cogn. Disord. 30(4), Saharia RB, Kulshretha SK. Can dietary
Neurobiol. Aging 26(Suppl. 1), 133–136 352–361 (2010). magnesium modulate lipoprotein
(2005). • Comprehensive and unique review on the metabolism? Magnes. Trace Elem. 9(5),
93 Green KN, Martinez-Coria H, possible association between dairy intake 255–264 (1990).
Khashwji H et al. Dietary and cognitive functioning. 116 Ma B, Lawson AB, Liese AD, Bell RA,
docosahexaenoic acid and Mayer-Davis EJ. Dairy, magnesium, and
104 Lee L, Kang SA, Lee HO et al.
docosapentaenoic acid ameliorate calcium intake in relation to insulin
Relationships between dietary intake and
amyloid-b and tau pathology via a sensitivity: approaches to modeling a
cognitive function level in Korean elderly
mechanism involving presenilin 1 levels. dose–dependent association. Am. J.
people. Public Health 115(2), 133–138
J. Neurosci. 27(16), 4385–4395 (2007). Epidemiol. 164(5), 449–458 (2006).
(2001).

702 Expert Rev. Neurother. 11(5), (2011)


Diet & Alzheimer’s disease Review

117 Festa A, D’Agostino R, Williams K et al. performance. EVA Study Group. 139 Cervilla JA, Prince M, Mann A. Smoking,
The relation of body fat mass and Epidemiology of Vascular Aging. Am. J. drinking, and incident cognitive
distribution to markers of chronic Epidemiol. 146(5), 405–412 (1997). impairment: a cohort community based
inflammation. Int. J. Obes. Relat. Metab. 129 Schinka JA, Vanderploeg RD, Rogish M, study included in the Gospel Oak Project.
Disord. 25(10), 1407–1415 (2001). Graves AB, Mortimer JA, Ordoric PI. J. Neurol. Neurosurg. Psychiatry 68(5),
118 Yaffe K, Lindquist K, Penninx BW et al. Effects of the use of alcohol and cigarettes 622–626 (2000).
Inflammatory markers and cognition in on cognition in elderly adults. J. Int. 140 Galanis DJ, Joseph C, Masaki KH,
well-functioning African–American and Neuropsychol. Soc. 8(6), 811–818 (2002). Petrovitch H, Ross GW, White L.
white elders. Neurology 61(1), 76–80 130 Verhaegen P, Borchelt M, Smith J. Relation A longitudinal study of drinking and
(2003). between cardiovascular and metabolic cognitive performance in elderly Japanese
119 Solfrizzi V, Frisardi V, Capurso C et al. disease and cognition in very old age: American men: the Honolulu-Asia aging
Moderate alcohol consumption, cross-sectional and longitudinal findings study. Am. J. Public Health 90(8),
apolipoprotein E, and neuroprotection. from the Berlin aging study. Health Psychol. 1254–1259 (2000).
Arch. Neurol. 66(4), 541–542 (2009). 22(6), 559–569 (2003). 141 Ngandu T, Helkala EL, Soininen H et al.
120 Goodwin JS, Sanchez CJ, Thomas P, 131 Lindeman RD, Wayne SJ, Baumgartner RN, Alcohol drinking and cognitive functions:
Hunt C, Garry PJ, Goodwin JM. Alcohol Garry PJ. Cognitive function in drinkers findings from the Cardiovascular Risk
intake in a healthy elderly population. compared to abstainers in the New Mexico Factors Aging and Dementia (CAIDE)
Am. J. Public Health 77(2), 173–177 Elder Health Survey. J. Gerontol. A Biol. Sci. Study. Dement. Geriatr. Cogn. Disord.
(1987). Med. Sci. 60(8), 1065–1070 (2005). 23(3), 140–149 (2007).
121 Christian JC, Reed T, Carmelli D, 132 Reid MC, Van Ness PH, Hawkins KA, 142 Orgogozo JM, Dartigues JF, Lafont S et al.
Page WF, Norton JA Jr, Breitner JC. Towle V, Concato J, Guo Z. Light to Wine consumption and dementia in the
Self-reported alcohol intake and cognition moderate alcohol consumption is associated elderly: a prospective community study in
in aging twins. J. Stud. Alcohol 56(4), with better cognitive function among older the Bordeaux area. Rev. Neurol. (Paris)
414–416 (1995). male veterans receiving primary care. 153(3), 185–192 (1997).
122 Hendrie HC, Gao S, Hall KS, Hui SL, J. Geriatr. Psychiatry Neurol. 19(2), 98–105 143 Lemeshow S, Letenneur L, Dartigues JF,
Unverzagt FW. The relationship between (2006). Lafont S, Orgogozo JM, Commenges D.
alcohol consumption, cognitive 133 Yoshitake T, Kiyohara Y, Kato I et al. Illustration of analysis taking into account
performance, and daily functioning in an Incidence and risk factors of vascular complex survey considerations:
urban sample of older Black Americans. dementia and Alzheimer’s disease in a the association between wine consumption
J. Am. Geriatr. Soc. 44(10), 1158–1165 defined elderly Japanese population: and dementia in the PAQUID Study. Am.
(1996). the Hisayama Study. Neurology 45(6), J. Epidemiol. 148(3), 298–306 (1998).
123 Launer LJ, Feskens EJ, Kalmijn S, 1161–1168 (1995). 144 Ruitenberg A, van Swieten JC,
Kromhout D. Smoking; drinking; and 134 Dufouil C, Tzourio C, Brayne C, Berr C, Witteman JC et al. Alcohol consumption
thinking. The Zutphen Elderly Study. Amouyel P, Alperovitch A. Influence of and risk of dementia: the Rotterdam Study.
Am. J. Epidemiol. 143(3), 219–227 (1996). apolipoprotein E genotype on the risk of Lancet 359(9303), 281–286 (2002).
124 Elias PK, Elias MF, D’Agostino RB, cognitive deterioration in moderate 145 Lindsay J, Laurin D, Verreault R et al. Risk
Silbershatz H, Wolf PA. Alcohol drinkers and smokers. Epidemiology 11(3), factors for Alzheimer’s disease:
consumption and cognitive performance in 280–284 (2000). a prospective analysis from the Canadian
the Framingham Heart Study. Am. J. 135 Leibovici D, Ritchie K, Ledesert B, Study of Health and Aging. Am. J.
Epidemiol. 150(6), 580–589 (1999). Touchon J. The effects of wine and tobacco Epidemiol. 156(5), 445–453 (2002).
125 Carmelli D, Swan GE, Reed T, consumption on cognitive performance in 146 Huang W, Qiu C, Winblad B,
Schellenberg GD, Christian JC. The effect the elderly: a longitudinal study of relative Fratiglioni L. Alcohol consumption and
of apolipoprotein E e4 in the relationships risk. Int. J. Epidemiol. 28(1), 77–81 (1999). incidence of dementia in a community
of smoking and drinking to cognitive 136 Stampfer MJ, Kang JH, Chen J, Cherry R, sample aged 75 years and older. J. Clin.
function. Neuroepidemiology 18(3), Grodstein F. Effects of moderate alcohol Epidemiol. 55(10), 959–964 (2002).
125–133 (1999). consumption on cognitive function in 147 Deng J, Zhou DH, Li J, Wang YJ, Gao C,
126 Bond GE, Burr R, McCurry SM, women. N. Engl. J. Med. 352(3), 245–253 Chen M. A 2-year follow-up study of
Graves AB, Larson EB. Alcohol, aging, and (2005). alcohol consumption and risk of dementia.
cognitive performance in a cohort of 137 Herbert LE, Taylor JO, Evans DA. Relation Clin. Neurol. Neurosurg. 108(4), 378–383
Japanese Americans aged 65 and older: of smoking and low-to-moderate alcohol (2006).
the Kame Project. Int. Psychogeriatr. 13(2), consumption to change in cognitive 148 Kivipelto M, Helkala EL, Hänninen T
207–223 (2001). function: a longitudinal study in a defined et al. Midlife vascular risk factors and
127 Bond GE, Burr R., Rice MM et al. community of older persons. Am. J. Alzheimer’s disease in later life:
Alcohol, aging, and cognitive performance: Epidemiol. 137(8), 881–891 (1993). longitudinal; population based study. BMJ
a cross-cultural comparison. J. Aging Health 138 Broe GA, Creasey H, Jorm AF et al. Health 322(7300), 1447–1451 (2001).
15(2), 371–390 (2003). habits and risk of cognitive impairment and 149 Fujishima M, Kiyohara Y. Incidence and
128 Dufouil C, Ducimetière P, Alperovitch A. dementia in old age: a prospective study on risk factors of dementia in a defined elderly
Sex differences in the association between the effects of exercise, smoking and alcohol Japanese population the Hisayama study.
alcohol consumption and cognitive consumption. Aust. N. Z. J. Public Health Ann. NY Acad. Sci. 977, 1–8 (2002).
22(5), 621–623 (1998).

www.expert-reviews.com 703
Review Solfrizzi, Panza, Frisardi et al.

150 Järvenpää T, Rinne JO, Koskenvuo M, 159 Fadda F, Rossetti ZL. Chronic ethanol 172 Albani D, Polito L, Forloni G. Sirtuins as
Räihä I, Kaprio J. Binge drinking in consumption: from neuroadaptation to novel targets for Alzheimer’s disease and
midlife and dementia risk. Epidemiology neurodegeneration. Prog. Neurobiol. 56(4), other neurodegenerative disorders:
16(6), 766–771 (2005). 385–431 (1998). experimental and genetic evidence.
151 Simons LA, Simons J, McCallum J, 160 National Institute on Alcohol Abuse and J. Alzheimers Dis. 19(1), 11–26 (2010).
Friedlander Y. Lifestyle factors and risk of Alcoholism. Alcohol Alert No. 46: 173 Wenzel E, Somoza V. Metabolism and
dementia: Dubbo study of the elderly. Are Women More Vulnerable to Alcohol’s bioavailability of trans-resveratrol.
Med. J. Aust. 184(2), 68–70 (2006). Effects? National Institutes of Health, MD, Mol. Nutr. Food Res. 49(5), 472–481 (2005).
152 Mehlig K, Skoog I, Guo X et al. Alcoholic USA (1999). 174 Walle T, Hsieh F, DeLegge MH, Oatis JE,
beverages and incidence of dementia: 161 Stancampiano R, Carta M, Cocco S, Walle UK. High absorption but very low
34-year follow-up of the prospective Curreli R, Rossetti ZL, Fadda F. Biphasic bioavailability of oral resveratrol in
population study of women in Goteborg. effects of ethanol on acetylcholine release humans. Drug Metab. Dispos. 32(12),
Am. J. Epidemiol. 167(6), 684–691 (2008). in the rat prefrontal cortex. Brain Res. 1377–1382 (2004).
153 Solfrizzi V, D’Introno A, Colacicco AM 997(1), 128–132 (2004). 175 Brust JC. A 74-year-old man with memory
et al, Italian Longitudinal Study on Aging 162 Naimi TS, Brown DW, Brewer RD et al. loss and neuropathy who enjoys alcoholic
Working Group. Alcohol consumption; Cardiovascular risk factors and beverages. JAMA 299(9), 1046–1054
mild cognitive impairment; and confounders among nondrinking and (2008).
progression to dementia. Neurology 68(10), moderate-drinking US adults. Am. J. Prev. 176 Panza F, Capurso C, Solfrizzi V. Alcohol
1790–1789 (2007). Med. 28(4), 369–373 (2005). use, thiamine deficiency, and cognitive
154 Anttila T, Helkala EL, Viitanen M et al. 163 Palomaki H, Kaste M. Regular light-to- impairment. JAMA 299(24), 2853–2854
Alcohol drinking in middle age and moderate intake of alcohol and the risk of (2008).
subsequent risk of mild cognitive ischemic stroke: is there a beneficial effect? 177 Hendrie HC, Osuntokun BO, Hall KS et al.
impairment and dementia in old age: Stroke 24(12), 1828–1832 (1993). Prevalence of Alzheimer’s disease and
a prospective population based study. BMJ 164 den Heijer T, Vermeer SE, van Dijk EJ dementia in two communities:
329(7465), 539 (2004). et al. Alcohol intake in relation to brain Nigerian–Africans and African–Americans.
•• First article in which alcohol drinking in magnetic resonance imaging findings in Am. J. Psychiatry 152(10), 1485–1492 (1995).
middle age demonstrated a U-shaped older persons without dementia. Am. J. 178 White L, Petrovitch H, Ross GW et al.
relationship with risk of mild cognitive Clin. Nutr. 80(4), 992–997 (2004). Prevalence of dementia in older
impairment in old age. Only the carriers 165 Mukamal KJ, Kuller LH, Fitzpatrick AL, Japanese–American men in Hawaii. JAMA
of apolipoprotein E e4 had an increased Longstreth WT Jr, Mittleman MA, 276(12), 955–960 (1996).
risk of dementia with increasing Siscovick DS. Prospective study of alcohol 179 Luchsinger JA, Tang MX, Shea S,
alcohol consumption. consumption and risk of dementia in older Mayeux R. Caloric intake and the risk of
155 Espeland MA, Gu L, Masaki KH et al. adults. JAMA 289(11), 1405–1413 (2003). Alzheimer disease. Arch. Neurol. 59(8),
Association between reported alcohol 166 Serafini M, Maiani G, Ferro-Luzzi A. 1256–1263 (2002).
intake and cognition: results from the Alcohol-free red wine enhances plasma 180 Steffen LM, Jacobs DR Jr, Stevens J,
Women’s Health Initiative Memory Study. antioxidant capacity in humans. J. Nutr. Shahar E, Carithers T, Folsom AR.
Am. J. Epidemiol. 161(3), 228–238 (2005). 128(6), 1003–1007 (1998). Associations of whole-grain, refined-grain,
156 Peters R, Peters J, Warner J, Beckett N, 167 Waterhouse AL. Wine phenolics. Ann. NY and fruit and vegetable consumption with
Bulpitt C. Alcohol, dementia and cognitive Acad. Sci. 957(5), 21–36 (2002). risks of all-cause mortality and incident
decline in the elderly: a systematic review. 168 Commenges D, Scotet V, Renaud S, coronary artery disease and ischemic
Age Ageing 37(5), 505–512 (2008). Jacqmin-Gadda H, Barberger-Gateau P, stroke: the Atherosclerosis Risk in
Dartigues J-F. Intake of flavonoids and risk Communities (ARIC) Study. Am. J. Clin.
• First systematic review with meta-analysis
of dementia. Eur. J. Epidemiol. 16(4), Nutr. 78(3), 383–390 (2003).
carried out to investigate any relationship
between incident cognitive decline or 357–363 (2000). 181 Mizrahi A, Knekt P, Montonen J, Laaksonen
dementia in the elderly and 169 Giasson BI, Ischiropoulos H, Lee VM, MA, Heliövaara M, Järvinen R. Plant foods
alcohol consumption, with only Trojanowski JQ. The relationship between and the risk of cerebrovascular diseases: a
oxidative/nitrative stress and pathological potential protection of fruit consumption.
longitudinal studies of subjects aged
inclusions in Alzheimer’s and Parkinson’s Br. J. Nutr. 102(7), 1075–1083 (2009).
≥65 years included.
diseases. Free Radic. Biol. Med. 32(12), 182 La Rue A, Koehler KM, Wayne SJ,
157 Anstey KJ, Mack HA, Cherbuin N.
1264–1275 (2002). Chiulli SJ, Haaland KY, Garry PJ.
Alcohol consumption as a risk factor for
170 Ding Q, Dimayuga E, Keller JN. Nutritional status and cognitive
dementia and cognitive decline: meta-
Oxidative damage, protein synthesis, and functioning in a normally aging sample:
analysis of prospective studies. Am. J.
protein degradation in Alzheimer’s a 6-y reassessment. Am. J. Clin. Nutr.
Geriatr. Psychiatry 17(7), 542–555 (2009).
disease. Curr. Alzheimer Res. 4(1), 73–79 65(1), 20–29 (1997).
158 Mukamal KJ, Longstreth WT Jr, Mittleman
(2007). 183 Salerno-Kennedy R, Cashman KD.
MA, Crum RM, Siscovick DS. Alcohol
171 Heinonen IM, Lehtonen PJ, Hopia AI. Relationship between dementia and
consumption and subclinical findings on
Antioxidant activity of berry and fruit nutrition-related factors and disorders:
magnetic resonance imaging of the brain in
wines and liquors. J. Agric. Food Chem. an overview. Int. J. Vit. Nutr. Res. 75(2),
older adults: the cardiovascular health study.
46(1), 25–31 (1998). 83–95 (2005).
Stroke 32(9), 1939–1946 (2001).

704 Expert Rev. Neurother. 11(5), (2011)


Diet & Alzheimer’s disease Review

184 Ancelin ML, Christen Y, Ritchie K. 195 Kang JH, Ascherio A, Grodstein F. Fruit 207 Smith MA, Hirai K, Hsiao K et al.
Is antioxidant therapy a viable alternative and vegetable consumption and cognitive Amyloid-b deposition in Alzheimer
for mild cognitive impairment? decline in aging women. Ann. Neurol. transgenic mice is associated with oxidative
Examination of the evidence. Dem. Geriatr. 57(5), 713–720 (2005). stress. J. Neurochem. 70(5), 2212–2215
Cogn. Disord. 24(1), 1–19 (2007). 196 Morris MC, Evans DA, Tangney CC, (1998).
185 Grodstein F, Chen J, Willet WC. Bienias JL, Wilson RS. Associations of 208 Youdim KA, Qaiser MZ, Begley DJ,
High-dose antioxidant supplements and vegetable and fruit consumption with Rice-Evans CA, Abbott NJ. Flavonoid
cognitive function in community-dweling age-related cognitive change. Neurology permeability across an in situ model of the
elderly women. Am. J. Clin. Nutr. 77(4), 67(8), 1370–1376 (2006). blood-brain barrier. Free Radic. Biol. Med.
3223–3229 (2003). 197 Hughes TF, Andel R, Small BJ et al. 36(5), 592–604 (2004).
186 Engelhart MJ, Geerlinks MI, Ruitenberg A Midlife fruit and vegetable consumption 209 Ortiz D, Shea TB. Apple juice prevents
et al. Dietary intake of antioxidants and and risk of dementia in later life in Swedish oxidative stress induced by amyloid-b in
risk of Alzheimer disease. JAMA 287(24), twins. Am. J. Geriatr. Psychiatry 18(5), culture. J. Alzheimer Dis. 6(1), 27–30 (2004).
3223–3229 (2002). 413–420 (2010). 210 Heo HJ, Lee CY. Protective effects of
187 Lim GP, Chu T, Yang F, Beech W, • Findings of this recent article suggested quercetin and vitamin C against oxidative
Frautschy SA, Cole GM. The curry spice that higher fruit and vegetable intake stress-induced neurodegeneration. J. Agric.
curcumin reduces oxidative damage and assessed at midlife was associated with a Food Chem. 52(6), 7514–7517 (2004).
amyloid pathology in an Alzheimer´s lower risk of dementia and AD in late life. 211 Nakayama T. Suppression of
transgenic mouse. J. Neurosci. 21(21), hydroperoxide-induced cytotoxicity by
198 Mecocci P, Mariani E, Polidori MC,
8370–8377 (2001). polyphenols. Cancer Res. 54(7 Suppl.),
Hensley K, Butterfield DA. Antioxidant
188 Conte V, Uryu K, Fujimoto S et al. agents in Alzheimer’s disease. Centr. Nerv. S1991–S1993 (1994).
Vitamin E reduces amyloidosis and Syst. Ag. Med. Chem. 8, 48–63 (2008). 212 Patil CS, Singh VP, Satyanarayan PS,
improves cognitive function in Tg2576 Jain NK, Singh A, Kulkarni SK. Protective
199 Polidori MC, Praticó D, Mangialasche F
mice following repetitive concussive brain effect of flavonoids against aging- and
et al. High fruit and vegetable intake is
injury. J. Neurochem. 90(3), 758–764 lipopolysaccharide-induced cognitive
positively correlated with antioxidant status
(2004). impairment in mice. Pharmacology 69(2),
and cognitive performance in healthy
189 Huang HY, Caballero B, Chang S et al. subjects. J. Alzheimer Dis. 17(4), 921–927 59–67 (2003).
The efficacy and safety of multivitamin (2009). 213 Rogers EJ, Milhalik S, Orthiz D, Shea TB.
and mineral supplement use to prevent Apple juice prevents oxidative stress and
200 Fratiglioni L, Paillard-Borg S, Winblad B.
cancer and chronic disease in adults: a impaired cognitive performance caused by
An active and socially integrated lifestyle in
systematic review for a National Institutes genetic and dietary deficiencies in mice.
late life might protect against dementia.
of Health state-of-the-science conference. J. Nutr. Health Aging 8(2), 92–97 (2004).
Lancet Neurol. 3(6), 343–353 (2004).
Ann. Intern. Med. 145(5), 372–385 (2006).
201 Van Dyk K, Sano M. The impact of 214 Morris MC, Evans DA, Bienias JL,
190 Sano M, Ernesto C, Thomas RG et al. Tangney CC, Wilson RS. Vitamin E and
nutrition on cognition in the elderly.
A controlled trial of selegiline, cognitive decline in older persons.
Neurochem. Res. 32(4–5), 893–904 (2007).
a-tocopherol, or both as treatment for Arch. Neurol. 59(7), 1125–1132 (2002).
Alzheimer´s disease. N. Engl. J. Med. 202 Letenneur L, Proust-Lima C, Le Gouge A,
Dartigues J, Barberger-Gateau P. Flavonoid 215 Dauchet L, Amouyel P, Dallongeville J.
336(17), 1216–1222 (1997).
intake and cognitive decline over a 10-year Fruit and vegetable consumption and risk
191 Le Bars PL, Velasco VM, Fergunson JM, of stroke: a meta-analysis of cohort studies.
period. Am. J. Epidemiol. 165(12),
Dessain EC, Keiser M, Hoerr R. Influence Neurology 65(8), 1193–1197 (2005).
1364–1371 (2007).
of the severity of cognitive impairment on
203 Gray SL, Anderson ML, Crane PK et al. 216 Imbimbo BP, Solfrizzi V, Panza F.
the effect of Gingko biloba extract Egb 761
Antioxidant vitamin supplement use and Are NSAIDs useful to treat Alzheimer’s
in Alzheimer´s disease. Neuropsychobiology
risk of dementia or Alzheimer’s disease in disease or mild cognitive impairment?
45(1), 19–26 (2002).
older adults. J. Am. Geriatr. Soc. 56(2), Front. Aging Neurosci. 2. Pii, 19 (2010).
192 Petersen RC, Thomas RG, Grundman M
291–295 (2008). 217 Breitner JC, Haneuse SJ, Walker R et al.
et al. Vitamin E and donepezil for the
204 Sonnen JA, Larson EB, Gray SL et al. Free Risk of dementia and AD with prior
treatment of mild cognitive impairment
radical damage to cerebral cortex in exposure to NSAIDs in an elderly
Alzheimer’s Disease Cooperative Study
Alzheimer’s disease, microvascular brain community-based cohort. Neurology
Group. N. Engl. J. Med. 352(23),
injury, and smoking. Ann. Neurol. 65(2), 72(22), 1899–1905 (2009).
2379–2388 (2005).
226–229 (2009). 218 Sonnen JA, Larson EB, Walker RL et al.
193 Kamat CD, Gadal S, Mhatre M,
205 Morris MC, Evans DA, Tangney CC et al. Nonsteroidal anti-inflammatory drugs are
Williamson KS, Pye QN, Hensley K.
Relation of the tocopherol forms to incident associated with increased neuritic plaques.
Antioxidants in central nervous system
Alzheimer disease and to cognitive change. Neurology 75(13), 1203–1210 (2010).
diseases: preclinical promise and
translational challenges. J. Alzheimer Dis. Am. J. Clin. Nutr. 81(2), 508–514 (2005). 219 Wannamethee SG, Lowe GD, Rumley A,
15(3), 473–493 (2008). 206 Morris MC, Evans DA, Bienias JL et al. Bruckdorfer KR, Whincup PH.
Dietary intake of antioxidant nutrients and Associations of vitamin C status, fruit and
194 Lee L, Kang SA, Lee HO et al. Relationships
the risk of incident Alzheimer disease in a vegetable intakes, and markers of
between dietary intake and cognitive
biracial community study. JAMA 287(24), inflammation and hemostasis. Am. J. Clin.
function level in Korean elderly people.
3230–3237 (2002). Nutr. 83(3), 567–574 (2006).
Public Health 115(2), 133–128 (2001).

www.expert-reviews.com 705
Review Solfrizzi, Panza, Frisardi et al.

220 Manach C, Scalbert A, Morand C, 232 Scarmeas N, Luchsinger JA, Mayeux R, polyunsaturated fatty acids, n-6 PUFA,
Remesy C, Jimenez L. Polyphenols: food Stern Y. Mediterranean diet, and Alzheimer vitamin E, and folate, but with less
sources and bioavailability. Am. J. Clin. disease mortality. Neurology 69(11), saturated fatty acids and vitamin B12.
Nutr. 79(5), 727–747 (2004). 1084–1093 (2007). 243 Renaud S, de Lorgeril M, Delaye J et al.
221 Sanchez-Moreno C, Cano MP et al. 233 Scarmeas N, Stern Y, Mayeux R, Manly JJ, Cretan Mediterranean diet for prevention
High-pressurized orange juice consumption Schupf N, Luchsinger JA. Mediterranean of coronary heart disease. Am. J. Clin.
affects plasma vitamin C, antioxidative diet and mild cognitive impairment. Nutr. 61(6 Suppl.), S1360–S1367 (1995).
status and inflammatory markers in healthy Arch. Neurol. 66(2), 216–225 (2009). 244 Chrysohoou C, Panagiotakos DB,
humans. J. Nutr. 133(7), 2204–2209 234 Féart C, Samieri C, Rondeau V, Amieva H, Pitsavos C, Das UN, Stefanadis C.
(2003). Portet F, Dartigues JF et al. Adherence to a Adherence to the Mediterranean diet
222 Kiefer I, Prock P, Lawrence C et al. Mediterranean diet, cognitive decline, and attenuates inflammation and coagulation
Supplementation with mixed fruit and risk of dementia. JAMA 302(6), 638–648 process in healthy adults: the ATTICA
vegetable juice concentrates increased serum (2009). Study. J. Am. Coll. Cardiol. 44(1), 152–158
antioxidants and folate in healthy adults. 235 Scarmeas N, Luchsinger JA, Schupf N et al. (2004) .
J. Am. Coll. Nutr. 23(3), 205–211 (2004). Physical activity, diet, and risk of 245 Singh RB, Dubnov G, Niaz MA et al.
223 Wang HX, Wahlin A, Basun H, Fastbom J, Alzheimer disease. JAMA 302(6), 627–637 Effect of an Indo–Mediterranean diet on
Winblad B, Fratiglioni L. Vitamin B(12) (2009). progression of coronary artery disease in
and folate in relation to the development of 236 Gu Y, Luchsinger JA, Stern Y, Scarmeas N. high risk patients (Indo–Mediterranean
Alzheimer’s disease. Neurology 56(9), Mediterranean diet, inflammatory and Diet Heart Study): a randomised single-
1188–1194 (2001). metabolic biomarkers, and risk of blind trial. Lancet 360(9344), 1455–1461
224 Féart C, Samieri C, Barberger-Gateau P. Alzheimer’s disease. J. Alzheimers Dis. (2002).
Mediterranean diet and cognitive 22(2), 483–492 (2010). 246 Psaltopoulou T, Naska A, Orfanos P,
function in older adults. Curr. Opin. 237 Solfrizzi V, Capurso C, D’Introno A, Trichopoulos D, Mountokalakis T,
Clin. Nutr. Metab. Care 13(1), 14–18 Colacicco AM, Capurso A, Panza F. Trichopoulou A. Olive oil, the
(2010). Whole-diet approach and risk of chronic Mediterranean diet, and arterial blood
225 Kant AK. Dietary patterns and health disease: limits and advantages. J. Am. pressure: the Greek European Prospective
outcomes. J. Am. Diet. Assoc. 104(4), Geriatr. Soc. 54(11), 1800–1802 (2006). Investigation into Cancer and Nutrition
615–635 (2004). (EPIC) study. Am. J. Clin. Nutr. 80(4),
238 Trichopoulou A, Bamia C, Trichopoulos D.
1012–1018 (2004).
226 Kourlaba G, Panagiotakos DB. Dietary Anatomy of health effects of Mediterranean
quality indices and human health: a review. diet: Greek EPIC prospective cohort study. 247 de Lorgeril M, Salen P, Martin JL,
Maturitas 62(1), 1–8 (2009). BMJ 338, b2337 (2009). Monjaud I, Delaye J, Mamelle N.
Mediterranean diet, traditional risk factors,
227 Rumawas ME, Dwyer JT, McKeown NM, 239 Panagiotakos DB, Pitsavos C, Arvaniti F,
and the rate of cardiovascular
Meigs JB, Rogers G, Jacques PF. Stefanadis C. Adherence to the
complications after myocardial infarction:
The development of the Mediterranean- Mediterranean food pattern predicts the
final report of the Lyon Diet Heart Study.
style dietary pattern score and its prevalence of hypertension,
Circulation 99(6), 779–785 (1999).
application to the American diet in the hypercholesterolemia, diabetes and obesity,
Framingham Offspring Cohort. J. Nutr. among healthy adults; the accuracy of the 248 Gauthier S, Reisberg B, Zaudig M et al.
139(6), 1150–1156 (2009). MedDietScore. Prev. Med. 44(4), 335–340 Mild cognitive impairment. Lancet
(2007). 367(9518), 1262–1270 (2006).
228 Trichopoulou A, Kouris-Blazos A, Wahlqvist
ML et al. Diet and overall survival in elderly 240 Guenther PM, Reedy J, Krebs-Smith SM. 249 Luchsinger JA, Tang MX, Shea S,
people. BMJ 311(7018), 1457–1460 (1995). Development of the Healthy Eating Mayeux R. Hyperinsulinemia and risk of
Index-2005. J. Am. Diet. Assoc. 108(11), Alzheimer disease. Neurology 63(7),
229 Hu FB, Bronner L, Willett WC et al. Fish
1896–1901 (2008). 1187–1192 (2004).
and omega-3 fatty acid intake and risk of
coronary heart disease in women. JAMA 241 Tangney CC, Kwasny MJ, Li H, 250 Watson GS, Craft S. The role of insulin
287(14), 1815–1821 (2002). Wilson RS, Evans DA, Morris MC. resistance in the pathogenesis of
Adherence to a Mediterranean-type dietary Alzheimer’s disease: implications for
230 Scarmeas N, Stern Y, Tang MX, Mayeux R,
pattern and cognitive decline in a treatment. CNS Drugs 17(1), 27–45
Luchsinger JA. Mediterranean diet and risk
community population. Am. J. Clin. Nutr. (2003).
for Alzheimer’s disease. Ann. Neurol. 59(6),
912–921 (2006). 93(3), 601–607 (2011). 251 Reger MA, Watson GS, Green PS et al.
242 Gu Y, Nieves JW, Stern Y, Luchsinger JA, Intranasal insulin improves cognition and
•• Provides another reminder that the
Scarmeas N. Food combination and modulates b-amyloid in early AD.
preservation of certain dietary and lifestyle
Alzheimer disease risk: a protective diet. Neurology 70(6), 440–448 (2008).
traditions may have important health
Arch. Neurol. 67(6), 699–706 (2010). 252 Zhao Z, Xiang Z, Haroutunian V,
consequences, and that the advantages of
•• In an analysis of food combination, a Buxbaum JD, Stetka B, Pasinetti GM.
the Mediterranean diet may be potentially
dietary pattern that explained variations Insulin degrading enzyme activity
transferable to other populations.
in AD-related nutrients and was strongly selectively decreases in the hippocampal
231 Scarmeas N, Stern Y, Mayeux R, formation of cases at high risk to develop
Luchsinger JA. Mediterranean diet, protective against the development of AD
Alzheimer’s disease. Neurobiol. Aging
Alzheimer disease, and vascular mediation. was identified. This dietary pattern
28(6), 824–830 (2007).
Arch. Neurol. 63(12), 1709–1717 (2006). reflected a diet rich in n-3

706 Expert Rev. Neurother. 11(5), (2011)


Diet & Alzheimer’s disease Review

253 Yaffe K, Blackwell T, Kanaya AM, radical generation, and serum lipoprotein 276 Magaki S, Mueller C, Dickson C,
Davidowitz N, Barrett-Connor E, oxidation of several natural antioxidants. Kirsch W. Increased production of
Krueger K. Diabetes, impaired fasting J. Agric. Food Chem. 50(8), 2464–2469 inflammatory cytokines in mild cognitive
glucose, and development of cognitive (2002). impairment. Exp. Gerontol. 42(3), 233–240
impairment in older women. Neurology 265 Mancini M, Parfitt VJ, Rubba P. (2007).
63(4), 658–663 (2004). Antioxidants in the Mediterranean diet. 277 Iwamoto N, Nishiyama E, Ohwada J,
254 Luchsinger JA, Reitz C, Patel B, Tang MX, Can. J. Cardiol. 11(Suppl. G), G105–G109 Arai H. Demonstration of CRP
Manly JJ, Mayeux R. Relation of diabetes (1995). immunoreactivity in brains of Alzheimer’s
to mild cognitive impairment. 266 Blum S, Aviram M, Ben-Amotz A, Levy Y. disease: immunohistochemical study using
Arch. Neurol. 64(4), 570–575 (2007). Effect of a Mediterranean meal on formic acid pretreatment of tissue sections.
255 Esposito K, Marfella R, Ciotola M et al. postprandial carotenoids, paraoxonase Neurosci. Lett. 177(1–2), 23–26 (1994).
Effect of a Mediterranean-style diet on activity and C-reactive protein levels. 278 Duong T, Nikolaeva M, Acton PJ.
endothelial dysfunction and markers of Ann. Nutr. Metab. 50(1), 20–24 (2006). C-reactive protein-like immunoreactivity
vascular inflammation in the metabolic 267 Wallace AJ, Sutherland WH, Mann JI, in the neurofibrillary tangles of Alzheimer’s
syndrome: a randomized trial. JAMA Williams SM. The effect of meals rich in disease. Brain Res. 749(1), 152–156 (1997).
292(12), 1440–1446 (2004). thermally stressed olive and safflower oils 279 McGeer EG, Yasojima K, Schwab C,
256 Jula A, Marniemi J, Huupponen R, Virtanen on postprandial serum paraoxonase activity McGeer PL. The pentraxins: possible role
A, Rastas M, Rönnemaa T. Effects of diet in patients with diabetes. Eur. J. Clin. in Alzheimer’s disease and other innate
and simvastatin on serum lipids, insulin, and Nutr. 55(11), 951–958 (2001). inflammatory diseases. Neurobiol. Aging
antioxidants in hypercholesterolemic men: a 268 Aviram M, Dornfeld L, Rosenblat M et al. 22(6), 843–848 (2001).
randomized controlled trial. JAMA 287(5), Pomegranate juice consumption reduces 280 Wood JA, Wood PL, Ryan R et al.
598–605 (2002). oxidative stress, atherogenic modifications Cytokine indices in Alzheimer’s temporal
257 Dai J, Jones DP, Goldberg J et al. to LDL, and platelet aggregation: studies in cortex: no changes in mature IL-1 b or
Association between adherence to the humans and in atherosclerotic IL-1RA but increases in the associated
Mediterranean diet and oxidative stress. apolipoprotein E-deficient mice. Am. J. acute phase proteins IL-6,
Am. J. Clin. Nutr. 88(5), 1364–1370 Clin. Nutr. 71(5), 1062–1076 (2000). a 2-macroglobulin and C-reactive protein.
(2008). 269 Sanchez-Moreno C, Cano MP, de Ancos B Brain Res. 629(2), 245–252 (1993).
258 Harris MI, Flegal KM, Cowie CC et al. et al. Mediterranean vegetable soup 281 Schmidt R, Schmidt H, Curb JD,
Prevalence of diabetes, impaired fasting consumption increases plasma vitamin C Masaki K, White LR, Launer LJ. Early
glucose, and impaired glucose tolerance in and decreases F2-isoprostanes, inflammation and dementia: a 25-year
U.S. adults: the Third National Health and prostaglandin E2 and monocyte follow-up of the Honolulu-Asia Aging
Nutrition Examination Survey, 1988–1994. chemotactic protein-1 in healthy humans. Study. Ann. Neurol. 52(2), 168–174
Diabetes Care 21(4), 518–524 (1998). J. Nutr. Biochem. 17(3), 183–189 (2006). (2002).
259 Keller JN, Schmitt FA, Scheff SW et al. 270 Giugliano D, Esposito K. Mediterranean 282 Frank RA, Galasko D, Hampel H et al.
Evidence of increased oxidative damage in diet and metabolic diseases. Curr. Opin. Biological markers for therapeutic trials in
subjects with mild cognitive impairment. Lipidol. 19(1), 63–68 (2008). Alzheimer’s disease: proceedings of the
Neurology 64(7), 1152–1156 (2005). 271 Panagiotakos DB, Dimakopoulou K, biological markers working group; NIA
260 Mecocci P. Oxidative stress in mild Katsouyanni K et al. Mediterranean diet initiative on neuroimaging in Alzheimer’s
cognitive impairment and Alzheimer and inflammatory response in myocardial disease. Neurobiol. Aging 24(4), 521–536
disease: a continuum. J. Alzheimers Dis. infarction survivors. Int. J. Epidemiol. (2003).
6(2), 159–163 (2004). 38(3), 856–866 (2009). 283 Roberts RO, Geda YE, Knopman DS et al.
261 Fito M, Cladellas M, de la Torre R et al. 272 Weninger SC, Yankner BA. Inflammation Association of C-reactive protein with mild
Antioxidant effect of virgin olive oil in and Alzheimer disease: the good, the bad, cognitive impairment. Alzheimers Dement.
patients with stable coronary heart disease: and the ugly. Nat. Med. 7(5), 527–528 5(5), 398–405 (2009).
a randomized, crossover, controlled, (2001). 284 Paschos GK, Rallidis LS, Liakos GK et al.
clinical trial. Atherosclerosis 181(1), 149–158 273 Walsh S, Aisen PS. Inflammatory processes Background diet influences the anti-
(2005). and Alzheimer’s disease. Expert Rev. inflammatory effect of a-linolenic acid in
262 Szeto YT, Tomlinson B, Benzie IF. Total Neurother. 4(5), 793–798 (2004). dyslipidaemic subjects. Br. J. Nutr. 92(4),
antioxidant and ascorbic acid content of 649–655 (2004).
274 Galimberti D, Schoonenboom N,
fresh fruits and vegetables: implications for Scheltens P et al. Intrathecal chemokine 285 Fung TT, McCullough ML, Newby P et al.
dietary planning and food preservation. synthesis in mild cognitive impairment and Diet-quality scores and plasma
Br. J. Nutr. 87(1), 55–59 (2002). Alzheimer disease. Arch. Neurol. 63(4), concentrations of markers of inflammation
263 Byers T, Perry G. Dietary carotenes, 538–543 (2006). and endothelial dysfunction. Am. J. Clin.
vitamin C, and vitamin E as protective Nutr. 82(1), 163–173 (2005).
275 Guerreiro RJ, Santana I, Bras JM,
antioxidants in human cancers. Annu. Rev. Santiago B, Paiva A, Oliveira C. Peripheral 286 Weaver JD, Huang MH, Albert M,
Nutr. 12, 139–159 (1992). inflammatory cytokines as biomarkers in Harris T, Rowe JW, Seeman TE.
264 Stupans I, Kirlich A, Tuck KL, Hayball PJ. Alzheimer’s disease and mild cognitive Interleukin-6 and risk of cognitive decline:
Comparison of radical scavenging effect, impairment. Neurodegener. Dis. 4(6), MacArthur studies of successful aging.
inhibition of microsomal oxygen free 406–412 (2007). Neurology 59(3), 371–378 (2002).

www.expert-reviews.com 707
Review Solfrizzi, Panza, Frisardi et al.

287 Engelhart MJ, Geerlings MI, Meijer J 289 Solfrizzi V, Frisardi V, Capurso C et al. Websites
et al. Inflammatory proteins in plasma Whole-diet approach: working on a
301 National Center for
and the risk of dementia: the Rotterdam criterion validity for age-related cognitive
Biotechnology Information
study. Arch. Neurol. 61(5), 668–672 decline and mild cognitive impairment.
www.ncbi.nlm.nih.gov
(2004). J. Am. Geriatr. Soc. 57(10), 1944–1946
(2009). 302 US Department of Health and Human
288 Bertelli A A, Migliori M, Panichi V et al.
Services, US Department of Agriculture.
Oxidative stress and inf lammatory 290 Jacques PF, Tucker KL. Are dietary
Dietary guidelines for Americans 2005. 6th
reaction modulation by white wine. patterns useful for understanding the role
ed. Washington, DC: US Government
Ann. NY Acad. Sci. 957, 295–301 of diet in chronic disease? Am. J. Clin.
Printing Office (2005)
(2002). Nutr. 73(1), 1–2 (2001).
http://health.gov/dietaryguidelines/

708 Expert Rev. Neurother. 11(5), (2011)

View publication stats

You might also like