ArticlePDF Available

Midlife overweight and obesity increase late-life dementia risk: A population-based twin study

Authors:

Abstract

The relation of overweight to dementia is controversial. We aimed to examine the association of midlife overweight and obesity with dementia, Alzheimer disease (AD), and vascular dementia (VaD) in late life, and to verify the hypothesis that genetic and early-life environmental factors contribute to the observed association. From the Swedish Twin Registry, 8,534 twin individuals aged ≥65 (mean age 74.4) were assessed to detect dementia cases (DSM-IV criteria). Height and weight at midlife (mean age 43.4) were available in the Registry. Data were analyzed as follows: 1) unmatched case-control analysis for all twins using generalized estimating equation (GEE) models and 2) cotwin matched case-control approach for dementia-discordant twin pairs by conditional logistic regression taking into account lifespan vascular disorders and diabetes. Among all participants, dementia was diagnosed in 350 subjects, and 114 persons had questionable dementia. Overweight (body mass index [BMI] >25-30) and obesity (BMI >30) at midlife were present in 2,541 (29.8%) individuals. In fully adjusted GEE models, compared with normal BMI (20-25), overweight and obesity at midlife were related to dementia with odds ratios (ORs) (95% CIs) of 1.71 (1.30-2.25) and 3.88 (2.12-7.11), respectively. Conditional logistic regression analysis in 137 dementia-discordant twin pairs led to an attenuated midlife BMI-dementia association. The difference in ORs from the GEE and the matched case-control analysis was statistically significant (p = 0.019). Both overweight and obesity at midlife independently increase the risk of dementia, AD, and VaD. Genetic and early-life environmental factors may contribute to the midlife high adiposity-dementia association.
DOI 10.1212/WNL.0b013e3182190d09
2011;76;1568Neurology
W.L. Xu, A.R. Atti, M. Gatz, et al.
population-based twin study
Midlife overweight and obesity increase late-life dementia risk : A
June 8, 2011This information is current as of
http://www.neurology.org/content/76/18/1568.full.html
located on the World Wide Web at:
The online version of this article, along with updated information and services, is
rights reserved. Print ISSN: 0028-3878. Online ISSN: 1526-632X. Allsince 1951, it is now a weekly with 48 issues per year. Copyright © 2011 by AAN Enterprises, Inc.
® is the official journal of the American Academy of Neurology. Published continuouslyNeurology
by GUILLERMO GARCIA-RAMOS on June 8, 2011www.neurology.orgDownloaded from
Midlife overweight and obesity increase
late-life dementia risk
A population-based twin study
W.L. Xu, MD, PhD
A.R. Atti, MD, PhD
M. Gatz, PhD
N.L. Pedersen, PhD
B. Johansson, PhD
L. Fratiglioni, MD, PhD
ABSTRACT
Objective: The relation of overweight to dementia is controversial. We aimed to examine the asso-
ciation of midlife overweight and obesity with dementia, Alzheimer disease (AD), and vascular
dementia (VaD) in late life, and to verify the hypothesis that genetic and early-life environmental
factors contribute to the observed association.
Methods: From the Swedish Twin Registry, 8,534 twin individuals aged 65 (mean age 74.4)
were assessed to detect dementia cases (DSM-IV criteria). Height and weight at midlife (mean
age 43.4) were available in the Registry. Data were analyzed as follows: 1) unmatched case-
control analysis for all twins using generalized estimating equation (GEE) models and 2) cotwin
matched case-control approach for dementia-discordant twin pairs by conditional logistic regres-
sion taking into account lifespan vascular disorders and diabetes.
Results: Among all participants, dementia was diagnosed in 350 subjects, and 114 persons had
questionable dementia. Overweight (body mass index [BMI] 25–30) and obesity (BMI 30) at
midlife were present in 2,541 (29.8%) individuals. In fully adjusted GEE models, compared with
normal BMI (20–25), overweight and obesity at midlife were related to dementia with odds ratios
(ORs) (95% CIs) of 1.71 (1.30–2.25) and 3.88 (2.12–7.11), respectively. Conditional logistic
regression analysis in 137 dementia-discordant twin pairs led to an attenuated midlife BMI-
dementia association. The difference in ORs from the GEE and the matched case-control analysis
was statistically significant (p0.019).
Conclusions: Both overweight and obesity at midlife independently increase the risk of dementia,
AD, and VaD. Genetic and early-life environmental factors may contribute to the midlife high
adiposity–dementia association. Neurology
®
2011;76:1568–1574
GLOSSARY
AD Alzheimer disease; BMI body mass index; CERAD Consortium to Establish a Registry for Alzheimer’s Disease; CI
confidence interval; DSM-IV Diagnostic and Statistical Manual of Mental Disorders, 4th edition; DZ dizygotic; GEE
generalized estimating equation; ICD International Classification of Diseases; IDR Inpatient Discharge Registry;
MMSE Mini-Mental State Examination; MZ monozygotic; OR odds ratio; VaD vascular dementia.
Body mass index (BMI) provides an indirect measure of adiposity, and is strongly correlated with
total body fat tissue.
1
Adiposity may influence or be influenced by brain structures and functions,
which may be involved in dementing processes.
2
In the last decade, many population-based longi-
tudinal studies have evaluated the relationship between adiposity and dementia. Several reports have
shown that midlife obesity increases the risk of dementia in late life
3-10
; however, the effect of midlife
overweight on dementia is controversial.
3,5,7,9
Currently, the prevalence of overweight and obesity is
over 50% among adults in the United States and Europe.
11
Although adiposity may be linked to dementia through several biologically plausible path-
ways,
12
our understanding of the mechanisms for such an association is still limited. Both
From the Aging Research Center (W.L.X., A.R.A., L.F.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and
Stockholm University, Stockholm, Sweden; Department of Epidemiology (W.L.X.), Tianjin Medical University, Tianjin, China; Psychiatry Institute
Paolo Ottonello (A.R.A.), Bologna University, Bologna, Italy; Department of Psychology (M.G., N.L.P.), University of Southern California, Los
Angeles; Department of Medical Epidemiology and Biostatistics (M.G., N.L.P.), Karolinska Institutet, Stockholm; Department of Psychology (B.J.),
University of Gothenburg, Gothenburg; and Stockholm Gerontology Research Center (L.F.), Stockholm, Sweden.
Study funding: Funded by The National Institute on Aging (R01-AG08724), the Swedish Research Councils (FAS-09-0632), and the Swedish Brain
Power. Also supported in part by funds from the Gamla Tja¨narinnor, the Bertil Stohnes Foundation, the Demensfonden, the Loo and Hans
Ostermans Foundation, and the Foundation for Geriatric Diseases at Karolinska Institutet.
Disclosure: Author disclosures are provided at the end of the article.
Address correspondence and
reprint requests to Dr. Weili Xu,
Aging Research Center,
Karolinska Institutet, Ga¨vlegatan
16, S-113 30 Stockholm, Sweden
weili.xu@ki.se
1568 Copyright © 2011 by AAN Enterprises, Inc.
by GUILLERMO GARCIA-RAMOS on June 8, 2011www.neurology.orgDownloaded from
obesity and dementia are complex genetic and
lifestyle-related disorders. Evidence has shown
that early-life environments (such as child-
hood socioeconomic situation) may affect the
development of obesity and dementia.
13,14
The life course development of the adiposity-
dementia association has been suggested,
15
but the contribution of genetics and early life
environments to the relationship has not been
investigated. In the present study, we sought
to 1) verify the long-term effect of midlife
overweight and obesity measured by BMI on
the risk of dementia, Alzheimer disease (AD),
and vascular dementia (VaD) taking into ac-
count diabetes and lifespan vascular disorders
using unmatched case-control analysis; and 2)
explore whether genetic and early-life envi-
ronments could explain the observed associa-
tion between midlife adiposity and dementia
by cotwin matched case-control approach us-
ing data from the Swedish Twin Registry.
METHODS Participants and data collection. Partici-
pants were drawn from the nationwide Swedish Twin Registry.16
In 1998–2001, all living twins in the registry who were born in
1935 and earlier (aged 65 years) were invited to participate in a
telephone interview screening for most common diseases, which
included a brief cognitive assessment.17 In brief, a total of 20,206
individuals in the registry were eligible for screening. Of them,
13,693 twins completed the cognitive screening module, result-
ing in a participation rate of 67.8%. Information concerning
demographic factors, education, current height and weight,
health status and behavior, current and past diseases, and use of
medications was also obtained during the interview. In addition,
self-report information on height and weight at midlife (mean
age 43.4) was collected for all like-sexed twin pairs by the Regis-
try in 1967, i.e., on average 30 years preceding the screening
phase. Out of the 13,693 twins, 8,534 had data on height and
weight at midlife available for the current analysis (figure 1).
Screening phase. The screening test included the TELE ques-
tionnaire for participants18,19 and the Blessed Dementia Rating
Scale for informants when participants performed poorly on the
TELE.19,20 The TELE and BDRS were combined into an ordinal
scale with scores ranging from 0 (cognitively intact) to 3 (cogni-
tive dysfunction). Participants who screened positive (i.e., scored
3) and their cotwins were invited to a clinical examination (n
1,450).
Clinical phase and diagnosis. The full clinical workup
consisted of a visit by an assessment team composed of a nurse
and a physician using a protocol that generally followed the
Consortium to Establish a Registry for Alzheimer’s Disease
(CERAD). The protocol included physical and neurologic
examination, a review of medical history, informant interview,
and a neuropsychological assessment. The neuropsychological bat-
tery included the Mini-Mental State Examination (MMSE),
CERAD word list immediate and delayed recall, verbal flu-
ency, block design, figure copying, judgment, information,
symbol-digit, and prospective memory, as well as the Memory
in Reality test.
The assessment team made an initial diagnosis of dementia
and dementia subtypes following the DSM-IV criteria.21 All pro-
tocols were reviewed by a diagnostic board, consisting of a neu-
rologist and a neuropsychologist, who were blind to the
assessment team’s preliminary diagnosis, and verified the prelim-
inary diagnosis. More details of the clinical examination and di-
agnostic procedure have been reported previously.17 The cases
completely fulfilling the DSM-IV criteria were diagnosed as “de-
mentia,” in contrast with a category of “questionable dementia,”
which was used for individuals who did not fulfill one of the first
3DSM-IV diagnostic criteria, but did exhibit cognitive impair-
ment or functional disability. Participants were first classified as
no dementia, questionable, or dementia. A differential diagnosis
Figure 1 Flow chart of the study population
BMI body mass index.
Neurology 76 May 3, 2011 1569
by GUILLERMO GARCIA-RAMOS on June 8, 2011www.neurology.orgDownloaded from
was then given for dementia subtype according to the National
Institute of Neurological and Communicative Disorders and
Stroke–Alzheimer’s Disease and Related Disorders Association
criteria for AD22 and the National Institute of Neurological Dis-
orders and Stroke–Association Internationale pour la Recherche
et l’Enseignement en Neurosciences criteria for VaD.23 Through
the clinical workup, dementia was diagnosed in 350 subjects
including 232 AD and 74 VaD cases, and 114 had questionable
dementia. Among all participants, 8,070 did not have dementia.
Data on history of diabetes and vascular diseases obtained at
the screening interview were integrated with data from the Inpa-
tient Discharge Registry (IDR), which encompasses all hospital
discharges in Sweden since 1969. All discharge diagnoses for
participants who were hospitalized during 1969–2001 were ob-
tained. Each record in the IDR included up to 8 discharge diag-
noses according to the International Classification of Diseases,
8th revision (ICD-8) between 1969 and 1986 and 9th revision
(ICD-9) since 1987. Medical conditions derived from the inpa-
tient register database included diabetes (ICD-8, 9 code 250),
hypertension (ICD-8 codes 400404; ICD-9 codes 401–405),
ischemic heart disease (ICD-8, 9 codes 410414), cardiac dys-
rhythmia, heart failure or other myocardial insufficiency
(ICD-8, 9 codes 427 and 428), and stroke (ICD-8, 9 codes
430438). Diabetes and vascular diseases were deemed present
if reported by self or informant, or if recorded in the inpatient
registry. Survival status from midlife to the screening phase was
obtained from the Cause of Death Register.
Validation of self-reported height and weight. A previ-
ous study indicated that the correlations between self-reported
and measured height and weight were 0.97 for height and 0.95
for weight.24 The mean differences (SD) between self-reported
and measured values were 1.2 2.4 cm for height and 0.8 4.0
kg for weight. In the present study, the correlation between self-
reported height and weight at the screening phase and measured
height and weight at the clinical phase were 0.91 ( p0.001)
and 0.93 ( p0.001, n 864), respectively. Pearson correla-
tion coefficients were 0.87 ( p0.001, n 931) for the corre-
lation between the height at midlife recorded in the twin registry
and measured height at the clinical examination.
Standard protocol approvals. Informed consents were re-
quired from all participants during the telephone interview and
again in the clinical phase. The data collection procedures were
reviewed and approved by the Swedish Data Inspection Board,
Stockholm Sweden, the Regional Ethics Committee at Karolin-
ska Institutet, Stockholm, and the Institutional Review Board of
the University of Southern California.
Statistical analysis. BMI was calculated as self-reported
weight in kilograms divided by self-reported height in meters
squared, and categorized into 4 groups: underweight (20),
normal weight (2025), overweight (2530), and obese
(30).25 Midlife BMI was used as both continuous and categor-
ical variables in the analysis.
The characteristics of participants in different groups were
compared using
2
tests for categorical variables, one-way analy-
sis of variance for continuous variables. Multinomial logistic re-
gression was first performed to estimate the odds ratios (OR) and
95% confidence intervals (CI) of dementia (350 dementia cases
vs 8,070 controls) and questionable dementia (114 questionable
dementia cases vs 8,070 controls) separately in relation to BMI.
Further, 2 analytical strategies were applied to address the fol-
lowing aims: 1) unmatched case-control analyses was performed
in all subjects (464 dementia and questionable dementia cases vs
8,070 controls) using generalized estimating equations (GEE)
models, which are conceptually equivalent to logistic regression
for the analysis of classic case-control design, but control for the
clustering of twins within a pair; and 2) cotwin matched case-
control analyses was carried out using conditional logistic regres-
sion models among the 137 dementia-discordant twin pairs after
exclusion of 3,476 single twins, and 2,357 both no dementia and
35 both dementia twin pairs. The latter strategy allows matching
for unmeasured familial factors, which could reflect genetic
background or early life environment. Cases and controls are
comparable with respect to genetic and early life environmental
history (such as prenatal and postnatal nutritional status, and
childhood socioeconomic status). If the association found in
GEE analyses becomes attenuated in cotwin matched case-
control analyses, genetic factors or familial environments or both
are likely to contribute to the association. In contrast, if a signif-
icant association remains when using cotwin matched pairs, the
influences of genetic or early environmental factors on the asso-
ciation are likely to be marginal.
Logistic regression was used to test the difference in ORs
from GEE model and conditional logistic regression by examin-
ing the difference of overweight and obesity at midlife among
unmatched vs cotwin controls. Age, sex, education, zygosity, di-
abetes, and vascular diseases were considered as confounders in
all models. The statistical analyses were performed using SAS
statistical software version 9.1 and Stata SE 10.
RESULTS Population characteristics. Among the
8,534 participants, 350 (4.1%) were dementia cases
including 232 AD and 74 VaD, and 114 (1.3%)
were diagnosed with questionable dementia. Among
those included in the analysis, 6.0% of the females
and 4.6% of the males had dementia or questionable
dementia (
2
7.82, p0.005).
Compared to participants without dementia,
twins with dementia or questionable dementia were
older, had a lower level of education and current
BMI, had higher midlife BMI, and were more likely
to have diabetes, stroke, and heart disease. A total of
2,541 twins (29.8%) were overweight or obese at
midlife (table 1).
BMI in relation to questionable dementia and demen-
tia. In the multinomial logistic analysis of the entire
sample, midlife BMI as a continuous variable was
significantly associated with an increased risk of de-
mentia (OR 1.08, 95% CI 1.03–1.14) as well as
questionable dementia (OR 1.07, 95% CI 1.04
1.11) after adjustment for potential confounders.
Figure 2 shows the multi-adjusted ORs of dementia
and questionable dementia related to BMI as cate-
gorical variable. As the midlife BMI-related ORs for
dementia and questionable dementia were similar,
these 2 categories were combined as an outcome of
dementia in subsequent analyses.
Unmatched analysis. In GEE models, higher midlife
BMI was associated with greater risk of all dementia,
AD, and VaD using BMI as a continuous variable.
1570 Neurology 76 May 3, 2011 by GUILLERMO GARCIA-RAMOS on June 8, 2011www.neurology.orgDownloaded from
When BMI was analyzed as a categorical variable,
compared with normal weight, overweight was asso-
ciated with an increased risk of dementia and AD,
while obesity increased the risk of dementia, AD, and
VaD controlling for age, sex, and education. Further,
the association was not attenuated by additional ad-
justing for diabetes and vascular diseases (table 2).
However, the statistical power was limited for the
BMI-VaD association due to the few VaD cases.
Matched analysis. Among the 137 dementia discor-
dant twin pairs, 44 were monozygotic (MZ), 90
were dizygotic (DZ), and 3 were unknown zygos-
ity. In the cotwin matched case-control analyses,
the association of midlife overweight and obesity
(BMI 25) with dementia was attenuated, and no
longer significant (table 3).
The difference between the ORs of dementia re-
lated to BMI 25 (overweight and obesity) from
GEE model based on the entire sample (age- and
sex-adjusted OR 1.56 [95% CI 1.28–1.91]) and
from conditional logistic model based on dementia-
discordant pairs (sex-adjusted OR 0.88 [95% CI
0.64–1.21]) was tested by comparing high BMI
(25) among the unmatched and matched controls.
The difference was statistically significant (OR 1.62
[95% CI 1.12–2.66 p0.019]), indicating that ge-
netic and family environmental factors may contrib-
ute to the high adiposity-dementia association.
Supplementary analyses. The dropout rate in the
screening phase of this study was 32.2% (6,531/
20,206) mainly due to refusal (74.8%) and to
death (4.6%). Multivariable GEE model showed
that being a dropout was associated with ORs of
1.05 (95% CI 1.02–1.06) for old age, 1.06 (95%
CI 0.86–1.39) for female gender, 0.77 (95% CI
0.72–0.81) for education (per 1-year increase),
1.05 (95% CI 0.81–1.35) for stroke, 0.95 (95%
CI 0.72–1.27) for diabetes, 0.79 (95% CI 0.63–
1.03) for heart disease, and 0.98 (95% CI 0.74
1.29) for hypertension.
Figure 2 Odds ratio (OR) and 95% confidence interval (CI) of dementia and questionable dementia related
to midlife body mass index (BMI), after adjustment for age, sex, education, zygosity, diabetes,
stroke, hypertension, and heart disease (results from Multinomial Logistic Regression)
Table 1 Characteristics of the study participants (n 8,435) by
dementia diagnosis
a
Characteristics
No dementia
(n 8,070)
Questionable
dementia
(n 114)
Dementia
(n 350) pValue
Age, y, mean (SD) 74.0 (6.7) 80.5 (7.1) 82.3 (6.5) 0.001
Female sex 4,669 (57.9) 58 (50.9) 241 (68.9) 0.001
Education, y, mean (SD)b12.5 (62.3) 8.2 (2.9) 7.3 (2.5) 0.001
Zygotic status
Monozygotic 2,892 (35.8) 44 (38.6) 133 (38.0)
Dizygotic 5,041 (62.5) 70 (61.4) 204 (58.3)
Unknown 137 (1.7) 0 13 (3.7) 0.001
Midlife BMI, mean (SD) 23.7 (2.9) 24.9 (4.9) 25.0 (3.1) 0.001
Underweight (<20) 610 (7.5) 6 (5.3) 11 (3.1)
Normal (20–25) 5,126 (63.5) 61 (53.5) 179 (51.1)
Overweight (25–30) 2,120 (26.3) 41 (36.0) 136 (38.9)
Obese (>30) 214 (2.7) 6 (5.3) 24 (6.9) 0.001
Current BMI, mean (SD) 25.1 (3.7) 24.4 (3.6) 23.9 (4.7) 0.001
Diabetes 805 (10.0) 22 (19.3) 84 (24.0) 0.001
Stroke 621 (7.7) 35 (30.7) 103 (29.4) 0.001
Heart disease 1,111 (13.8) 48 (42.1) 245 (70.0) 0.001
Hypertension 2,727 (33.8) 42 (36.8) 126 (36.0) 0.05
Abbreviation: BMI body mass index.
a
Values are n (%) of participants unless indicated otherwise.
b
A total of 32 participants had missing value for education.
Neurology 76 May 3, 2011 1571
by GUILLERMO GARCIA-RAMOS on June 8, 2011www.neurology.orgDownloaded from
Further, we repeated the analysis of the BMI–
dementia association leaving out the twins who
scored 3 at cognitive screening but were diagnosed
as no dementia or not referred to clinical workup
(n 655). This produced results similar to those
from the initial analysis. Finally, we examined the
effect of BMI 25 on mortality from midlife to
late life (the screening phase). Among all like-
sexed twins born before 1936, 4,825 died before
the screening phase. In the GEE model, BMI 25
(overweight and obesity) at midlife was related to
elevated risk of death with multiadjusted OR of
1.30 (95% CI 1.19–1.42).
DISCUSSION In this nationwide Swedish twin
study, overweight and obesity at midlife increase the
risk of all dementia, AD, and VaD, independently of
lifespan diabetes and vascular diseases. Findings from
the cotwin matched analyses suggest that familial fac-
tors (genetic factors and early life environments) con-
tribute to the association between midlife high
adiposity and dementia in late life.
A growing body of evidence suggests that a high
level of adiposity is associated with cognitive decline
and dementia
10,26,27
; however, the relation between
BMI and dementia among people aged over 65 is
controversial.
28-30
Several population-based studies
have reported an effect of obesity at middle age on
dementia risk.
3,4,7,9
Only one study reported an in-
creased dementia risk in people with overweight.
5
Despite the fact that high adiposity is associated with
dementia, there remains a debate whether this con-
cerns only AD or also VaD. Some prospective studies
did find an association between obesity and increased
risk of VaD,
7,29,31
while others did not.
9,32-34
In this
Swedish twin cohort, we found that having dementia
or AD was associated with more than a 70% higher
odds of being overweight at midlife, while the odds
of being obese at midlife were higher for those with
AD as well as those with VaD. Although the effect of
midlife overweight on dementia is not as substantial
as that of obesity, its impact on public health and
clinical practice is significant due to the fact that
there are 1.6 billion overweight adults worldwide.
35
Several potential biological mechanisms may ex-
plain the association between adiposity and demen-
tia. First, higher BMI is associated with diabetes and
vascular diseases, which are related to dementia
risk.
12
Nonetheless, in our study, the association be-
tween midlife high BMI and dementia remained sig-
nificant after controlling for lifespan vascular
diseases, suggesting that nonvascular pathways might
play an important role in the adiposity-dementia as-
sociation. Second, higher adiposity at midlife may
reflect a lifetime exposure to an altered metabolic and
inflammatory state. Adiposity is one component of
the metabolic syndrome which has been related to
cognitive decline.
36
Further, adipose tissue is the larg-
est endocrine organ and secretes inflammatory cyto-
kines and growth hormones; some of them (such as
Table 2 Adjusted odds ratio (OR) and 95% confidence interval (CI) of dementia, Alzheimer disease, and vascular dementia related to
midlife BMI (results from generalized estimating equation models)
Midlife
BMI No. of twins
All dementia Alzheimer disease Vascular dementia
No. OR (95% CI)aOR (95% CI)bNo. OR (95% CI)aOR (95% CI)bNo. OR (95% CI)aOR (95% CI)b
Continuous 8,534 464 1.09 (1.06–1.12) 1.06 (1.03–1.10) 232 1.09 (1.04–1.13) 1.06 (1.01–1.10) 74 1.14 (1.08–1.21) 1.11 (1.04–1.19)
Categorical
<20 627 17 0.74 (0.44–1.25) 0.79 (0.45–1.38) 8 0.89 (0.64–1.23) 0.66 (0.31–1.41) 0
20–25 5,366 240 1 (Reference) 1 (Reference) 120 1 (Reference) 1 (Reference) 36 1 (Reference) 1 (Reference)
>25 2,541 207 1.50 (1.22–1.84) 1.80 (1.37–2.35) 104 1.52 (1.15–2.02) 1.98 (1.36–2.88) 38 1.62 (1.01–2.59) 1.35 (0.81–2.24)
25–30 2,297 177 1.37 (1.11–1.70) 1.71 (1.30–2.25) 90 1.41 (1.05–1.89) 1.91 (1.30–2.80) 31 1.39 (0.85–2.29) 1.17 (0.69–2.00)
>30 244 30 3.01 (1.95–4.64) 3.88 (2.12–7.11) 14 2.87 (1.57–5.26) 3.43 (1.49–7.90) 7 4.38 (1.89–10.14) 3.50 (1.36–8.99)
Abbreviations: BMI body mass index; CI confidence interval; OR odds ratio.
a
Adjusted for age, sex, and education.
b
Adjusted for age, sex, education, diabetes, hypertension, stroke, and heart disease.
Table 3 Midlife BMI in relation to dementia in dementia-discordant twin pairs
(n 137) (results from conditional logistic regression models)
Midlife BMI No. of twins Pairs
All dementia (n 137)
OR (95% CI)aOR (95% CI)b
Continuous 274 137 1.07 (0.97–1.21) 1.09 (0.95–1.26)
Categorical
<20 15 8 1.19 (0.40–3.56) 1.07 (0.23–4.96)
20–25 154 73 1 (Reference) 1 (Reference)
>25 105 56 1.26 (0.77–2.67) 1.24 (0.71–2.87)
25–30 92 47 1.14 (0.71–2.32) 1.10 (0.68–3.30)
>30 13 9 2.24 (0.77–6.72) 1.62 (0.65–6.94)
Abbreviations: BMI body mass index; CI confidence interval; OR odds ratio.
a
Adjusted for sex and education.
b
Adjusted for sex, education, diabetes, stroke, heart disease, and hypertension.
1572 Neurology 76 May 3, 2011 by GUILLERMO GARCIA-RAMOS on June 8, 2011www.neurology.orgDownloaded from
leptin, interleukin-6, and C-reactive protein) may af-
fect cognitive functioning. Leptin is involved in de-
position of amyloid
-42, and plays a role in
neurodegenerative process.
37
Twin studies involving a life-course approach
may help to identify genetic and environmental in-
fluences on the development of chronic diseases, as
twins provide naturally matched pairs, in which con-
founding effects of a large number of potentially
causal factors (e.g., genetics and childhood environ-
ments) may be removed when comparisons are made
within twin pairs. In cotwin control analyses, the as-
sociation between higher midlife BMI and dementia
was significantly attenuated, and thus may be attrib-
uted in part to genetic and early environmental fac-
tors such as childhood socioeconomic situation.
However, twins may also experience similar expo-
sures even at midlife and late life. Studies have re-
ported that early life exposure to an imbalanced
nutrition and disadvantaged economic status are re-
lated to a greater risk of obesity in adult life
38
and
dementia in late life.
39
Our findings suggest that
early-life environmental and genetic factors contrib-
ute to the link between adiposity and dementia.
Some limitations of the study should be men-
tioned. First, the use of prevalent dementia cases may
have introduced some confounding effect due to dif-
ferential survival among cases and controls. In addi-
tion, midlife obesity is associated with elevated
mortality in our study, which would probably lead to
an underestimation of the strength of the observed
association. Second, compared to participants, the
nonresponders were older, less educated, and more
likely to be women, but did not differ from partici-
pants in terms of vascular risk factors. As old age, low
education, and female sex are risk factors for demen-
tia, possible selection bias could arise if the preva-
lence of dementia in this cohort differs from that in
the general population. However, the prevalence of
dementia in this study was comparable with several
studies of dementia prevalence in Europe and the
United States.
17
Third, midlife height and weight re-
lied on self-reported information, which, however,
were validated by a previous study. In addition, BMI
alone might not be an ideal representation of body
composition, although BMI has been widely used in
population-based studies. Additional anthropomet-
ric measures, such as waist circumference, would be
useful. Finally, both obesity and AD are genetically
influenced disorders with substantial concordance in
twins. Thus, the matched pairs could be regarded as
overmatched, as twin pairs are similar on many as-
pects. Nevertheless, the comparison of the results
from the entire sample and from matched pairs pro-
vides information about the potential role of genetic
and familial influences in the observed association.
Finally, in the matched analysis, both MZ and DZ
twins were included due to limited number of
dementia-discordant MZ twin pairs. Hence, genetic
effects were not perfectly controlled for.
Our results provide further support for the im-
portant role of high adiposity at midlife in the devel-
opment of dementia, and highlight the need to
control body weight as early as midlife for prevention
of dementia in late life. These findings have relevant
implications for public health, as the risk of dementia
could be reduced by midlife weight loss.
40
Genetic
and early-life environmental factors may contribute
to the link of overweight and obesity to dementia,
suggesting that the high adiposity-dementia associa-
tion might develop across the lifespan.
AUTHOR CONTRIBUTIONS
Study concept and design: W.L.X., L.F., M.G., N.L.P.; analysis and inter-
pretation of the study: W.L.X., L.F., M.G., N.L.P.; drafting the manu-
script: W.L.X.; critical revision of the manuscript: L.F., M.G., N.L.P.,
A.R.A., B.J. Statistical analysis was conducted by Weili Xu.
ACKNOWLEDGMENT
The authors thank the members of the HARMONY study group for data
collection and management.
DISCLOSURE
Dr. Xu and Dr. Atti report no disclosures. Dr. Gatz received a medical
education grant from Forest Laboratories, Inc.; serves on the editorial
board of the Journal of Community Psychology; receives publishing royalties
for Handbook of the Psychology of Aging, Fifth Edition (Academic Press,
2001) and Assessing and Treating Late-life Depression: A Casebook and Re-
source Guide (Basic Books, 2002); and has received research support from
Forest Laboratories, Inc. and the NIH. Dr. Pedersen serves as an Associate
Editor of International Journal of Molecular Epidemiology and Genetics. Dr.
Johansson serves on scientific advisory boards for Aging Research Center,
Stockholm, Danish Aging Research Center, Denmark, and Kavli Re-
search Center for Ageing and Dementia, Norway; and serves on editorial
advisory boards for Aging and Mental Health,European Journal of Ageing,
Journal of Gerontopsychology and Geriatric Psychiatry, the Journal of Aging
Research, and the Journal of European Psychology Students. Dr. Fratiglioni
reports no disclosures.
Received June 22, 2010. Accepted in final form January 28, 2011.
REFERENCES
1. Taylor RW, Keil D, Gold EJ, Williams SM, Goulding A.
Body mass index, waist girth, and waist-to-hip ratio as in-
dexes of total and regional adiposity in women: evaluation
using receiver operating characteristic curves. Am J Clin
Nutr 1998;67:4449.
2. Gustafson D. A life course of adiposity and dementia. Eur
J Pharmacol 2008;585:163–175.
3. Kivipelto M, Ngandu T, Fratiglioni L, et al. Obesity and
vascular risk factors at midlife and the risk of dementia and
Alzheimer disease. Arch Neurol 2005;62:1556–1560.
4. Rosengren A, Skoog I, Gustafson D, Wilhelmsen L. Body
mass index, other cardiovascular risk factors, and hospital-
ization for dementia. Arch Intern Med 2005;165:321–
326.
5. Whitmer RA, Gunderson EP, Barrett-Connor E, Quesen-
berry CP Jr, Yaffe K. Obesity in middle age and future risk
Neurology 76 May 3, 2011 1573
by GUILLERMO GARCIA-RAMOS on June 8, 2011www.neurology.orgDownloaded from
of dementia: a 27 year longitudinal population based
study. BMJ 2005;330:1360.
6. Chiang CJ, Yip PK, Wu SC, et al. Midlife risk factors for
subtypes of dementia: a nested case-control study in Tai-
wan. Am J Geriatr Psychiatry 2007;15:762–771.
7. Whitmer RA, Gunderson EP, Quesenberry CP Jr, Zhou J,
Yaffe K. Body mass index in midlife and risk of Alzheimer
disease and vascular dementia. Curr Alzheimer Res 2007;
4:103–109.
8. Whitmer RA, Gustafson DR, Barrett-Connor E, Haan
MN, Gunderson EP, Yaffe K. Central obesity and in-
creased risk of dementia more than three decades later.
Neurology 2008;71:1057–1064.
9. Fitzpatrick AL, Kuller LH, Lopez OL, et al. Midlife and
late-life obesity and the risk of dementia: Cardiovascular
Health Study. Arch Neurol 2009;66:336–342.
10. Hassing LB, Dahl AK, Thorvaldsson V, et al. Overweight
in midlife and risk of dementia: a 40-year follow-up study.
Int J Obes 2009;33:893–898.
11. Flegal KM, Carroll MD, Ogden CL, Curtin LR. Preva-
lence and trends in obesity among US adults, 1999–2008.
JAMA 2010;303:235–241.
12. Whitmer RA. The epidemiology of adiposity and demen-
tia. Curr Alzheimer Res 2007;4:117–122.
13. Moceri VM, Kukull WA, Emanuel I, van Belle G, Larson
EB. Early-life risk factors and the development of Alzhei-
mer’s disease. Neurology 2000;54:415–420.
14. Teasdale TW, Sorensen TI, Stunkard AJ. Genetic and
early environmental components in sociodemographic in-
fluences on adult body fatness. BMJ 1990;300:1615–
1618.
15. Gustafson D. Adiposity indices and dementia. Lancet
Neurol 2006;5:713–720.
16. Lichtenstein P, Sullivan PF, Cnattingius S, et al. The
Swedish Twin Registry in the third millennium: an up-
date. Twin Res Hum Genet 2006;9:875–882.
17. Gatz M, Fratiglioni L, Johansson B, et al. Complete ascer-
tainment of dementia in the Swedish Twin Registry: the
HARMONY study. Neurobiol Aging 2005;26:439447.
18. Gatz M, Reynolds CA, John R, Johansson B, Mortimer
JA, Pedersen NL. Telephone screening to identify poten-
tial dementia cases in a population-based sample of older
adults. Int Psychogeriatr 2002;14:273–289.
19. Gatz M, Reynolds C, Nikolic J, Lowe B, Karel M, Pedersen
N. An empirical test of telephone screening to identify poten-
tial dementia cases. Int Psychogeriatr 1995;7:429438.
20. Blessed G, Tomlinson BE, Roth M. The association be-
tween quantitative measures of dementia and of senile
change in the cerebral grey matter of elderly subjects. Br J
Psychiatry 1968;114:797–811.
21. American Psychiatric Association. Diagnostic and Statistical
Manual of Mental Disorders, Fourth Edition (DSM-IV).
Washington, DC: American Psychiatric Association; 1994.
22. McKhann G, Drachman D, Folstein M, Katzman R, Price
D, Stadlan EM. Clinical diagnosis of Alzheimer’s disease:
report of the NINCDS-ADRDA Work Group under the
auspices of Department of Health and Human Services
Task Force on Alzheimer’s Disease. Neurology 1984;34:
939–944.
23. Roman GC, Tatemichi TK, Erkinjuntti T, et al. Vascular
dementia: diagnostic criteria for research studies: report of
the NINDS-AIREN International Workshop. Neurology
1993;43:250–260.
24. Stunkard AJ, Harris JR, Pedersen NL, McClearn GE. The
body-mass index of twins who have been reared apart.
N Engl J Med 1990;322:1483–1487.
25. Diehr P, Newman AB, Jackson SA, Kuller L, Powe N.
Weight-modification trials in older adults: what should the
outcome measure be? Curr Control Trials Cardiovasc Med
2002;3:1.
26. Hassing LB, Dahl AK, Pedersen NL, Johansson B. Over-
weight in midlife is related to lower cognitive function 30
years later: a prospective study with longitudinal assess-
ments. Dement Geriatr Cogn Disord 2010;29:543–552.
27. Gustafson DR, Backman K, Waern M, et al. Adiposity
indicators and dementia over 32 years in Sweden. Neurol-
ogy 2009;73:1559–1566.
28. Knopman DS, Edland SD, Cha RH, Petersen RC, Rocca
WA. Incident dementia in women is preceded by weight
loss by at least a decade. Neurology 2007;69:739–746.
29. Luchsinger JA, Patel B, Tang MX, Schupf N, Mayeux R.
Measures of adiposity and dementia risk in elderly persons.
Arch Neurol 2007;64:392–398.
30. Atti AR, Palmer K, Volpato S, Winblad B, De RD,
Fratiglioni L. Late-life body mass index and dementia inci-
dence: nine-year follow-up data from the Kungsholmen
Project. J Am Geriatr Soc 2008;56:111–116.
31. Kalmijn S, Foley D, White L, et al. Metabolic cardiovascu-
lar syndrome and risk of dementia in Japanese-American
elderly men: The Honolulu-Asia Aging Study. Arterioscler
Thromb Vasc Biol 2000;20:2255–2260.
32. Gustafson D, Rothenberg E, Blennow K, Steen B, Skoog I.
An 18-year follow-up of overweight and risk of Alzheimer
disease. Arch Intern Med 2003;163:1524–1528.
33. Hayden KM, Zandi PP, Lyketsos CG, et al. Vascular risk
factors for incident Alzheimer disease and vascular demen-
tia: the Cache County study. Alzheimer Dis Assoc Disord
2006;20:93–100.
34. Yoshitake T, Kiyohara Y, Kato I, et al. Incidence and risk
factors of vascular dementia and Alzheimer’s disease in a
defined elderly Japanese population: the Hisayama Study.
Neurology 1995;45:1161–1168.
35. WHO. Obesity and overweight 2010. Available at: http://
www.who.int/dietphysicalactivity/publications/facts/
obesity/en/. Accessed June 22, 2010.
36. Yaffe K, Haan M, Blackwell T, Cherkasova E, Whitmer
RA, West N. Metabolic syndrome and cognitive decline in
elderly Latinos: findings from the Sacramento Area Latino
Study of Aging study. J Am Geriatr Soc 2007;55:758
762.
37. Lieb W, Beiser AS, Vasan RS, et al. Association of plasma
leptin levels with incident Alzheimer disease and MRI
measures of brain aging. JAMA 2009;302:2565–2572.
38. Salsberry PJ, Reagan PB. Comparing the influence of
childhood and adult economic status on midlife obesity in
Mexican American, white, and African American women.
Public Health Nurs 2009;26:14–22.
39. Borenstein AR, Copenhaver CI, Mortimer JA. Early-life
risk factors for Alzheimer disease. Alzheimer Dis Assoc
Disord 2006;20:63–72.
40. Luchsinger JA, Gustafson DR. Adiposity, type 2 diabetes,
and Alzheimer’s disease. J Alzheimers Dis 2009;16:693–
704.
1574 Neurology 76 May 3, 2011 by GUILLERMO GARCIA-RAMOS on June 8, 2011www.neurology.orgDownloaded from
DOI 10.1212/WNL.0b013e3182190d09
2011;76;1568Neurology
W.L. Xu, A.R. Atti, M. Gatz, et al.
population-based twin study
Midlife overweight and obesity increase late-life dementia risk : A
June 8, 2011This information is current as of
Services
Updated Information & http://www.neurology.org/content/76/18/1568.full.html
including high resolution figures, can be found at:
References
1
http://www.neurology.org/content/76/18/1568.full.html#ref-list-
at:
This article cites 38 articles, 19 of which can be accessed free
Subspecialty Collections
http://www.neurology.org/cgi/collection/cohort_studies
Cohort studies http://www.neurology.org/cgi/collection/alzheimers_disease
Alzheimer's disease
s_dementia
http://www.neurology.org/cgi/collection/all_cognitive_disorder
All Cognitive Disorders/Dementia
following collection(s):
This article, along with others on similar topics, appears in the
Permissions & Licensing
http://www.neurology.org/misc/about.xhtml#permissions
tables) or in its entirety can be found online at:
Information about reproducing this article in parts (figures,
Reprints http://www.neurology.org/misc/addir.xhtml#reprintsus
Information about ordering reprints can be found online:
by GUILLERMO GARCIA-RAMOS on June 8, 2011www.neurology.orgDownloaded from
... Numerous health consequences are associated with obesity [8], and obesity has been recognized as a factor contributing to neurocognitive disorders [1,9,10]. According to recent studies, having an excessive amount of body fat in middle age (between 40 and 59 years old) poses a considerable threat of developing dementia in the future [8,11]. The risk of dementia is approximately 2-4 times higher in middleaged individuals with an elevated body mass index (BMI) in comparison with those who have a BMI within the normal range [11]. ...
... According to recent studies, having an excessive amount of body fat in middle age (between 40 and 59 years old) poses a considerable threat of developing dementia in the future [8,11]. The risk of dementia is approximately 2-4 times higher in middleaged individuals with an elevated body mass index (BMI) in comparison with those who have a BMI within the normal range [11]. ...
Article
Full-text available
Background Some studies suggest that the metabolic alterations linked to obesity in humans may increase the likelihood of developing dementia. However, there are currently no existing meta‐analyses exploring the possible occurrence of dementia in patients with metabolically healthy obesity (MHO). In the present study, we aimed to investigate a potential correlation between individuals with MHO and the likelihood of developing dementia. Method Our research focused on searching three databases to locate pertinent studies starting from January 10, 2023. We used sensitivity analysis, the Egger's test, and a visual funnel plot to separately assess the reliability of the research results and publication bias. We conducted this study according to a pre‐established protocol in the PROSPERO registry (CRD42023449459). Results We incorporated three potential cohort investigations. Individuals with good metabolic health and obesity showed a decreased likelihood of developing dementia compared with those who were metabolically healthy but not obese (hazard ratio (HR) = 0.79; 95% confidence interval (CI) 0.68–0.92). In contrast, individuals who were metabolically unhealthy and nonobese had an increased likelihood of developing dementia (HR = 1.33; 95% CI 1.25–1.42), whereas the population with metabolically unhealthy obesity did not exhibit a notable association with dementia onset (HR = 1.06; 95% CI 0.83–1.37). Conclusion According to our meta‐analysis, patients with MHO had a reduced likelihood of developing general dementia and Alzheimer disease. Maintaining a well‐functioning metabolism can provide advantages in reducing the occurrence of dementia.
... ). Among the set of modifiable risk variables, physical health factors including obesity, especially in midlife (Albanese et al. 2017;Xu et al. 2011), cardiovascular disease (Eriksson et al. 2010;Stefanidis et al. 2018), hypertension (McGrath et al. 2017), and diabetes (Xue et al. 2019) are considered to be alterable at most stages of the lifespan and are associated with multiple dementia etiologies (Bir et al. 2021;Profenno et al. 2010;Stampfer 2006). Subjective health ratings of overall health may encompass various modifiable risk factors of dementia but have been infrequently studied in this context (Stephan et al. 2021). ...
Article
Full-text available
Subjective health ratings are associated with dementia risk such that those who rate their health more poorly have increased risk for dementia. The genetic and environmental mechanisms underlying this association are unclear, as prior research cannot rule out whether the association is due to genetic confounds. The current study addresses this gap in two samples of twins, one from Sweden (N = 548) and one from Denmark (N = 4,373). Using genetically-informed, bivariate regression models, we assessed whether additive genetic effects explained the association between subjective health and dementia risk as indexed by a latent variable proxy measure. Age at intake, sex, education, depressive symptomatology, and follow-up time between subjective health and dementia risk assessments were included as covariates. Results indicate that genetic variance and other sources of confounding accounted for the majority of the effect of subjective health ratings on dementia risk. After adjusting for genetic confounding and other covariates, a small correlation was observed between subjective health and latent dementia risk in the Danish sample (rE = − .09, p < .05). The results provide further support for the genetic association between subjective health and dementia risk, and also suggest that subjective ratings of health measures may be useful for predicting dementia risk.
... In longitudinal studies mid-life obesity in overweight and obese individuals has been linked to changing lifestyles, characterized by reduced physical activity and poor dietary choices. These alterations result in metabolic disorders such as type 2 diabetes, and cardiovascular disease have been identified as risk factors for developing dementia and cognitive decline decades later [2][3][4][5]. Numerous investigations described the systemic alterations through high-caloric diets like Western diets (WDs) [6][7][8]. In response to obesity and associated chronic oversupply of fatty acids and sugar, a low-grade chronic inflammation develops, which if persisting over time, leads to a constant release of inflammatory effectors into the periphery [9][10][11]. ...
Article
Full-text available
Diet-induced increase in body weight is a growing health concern worldwide. Often accompanied by a low-grade metabolic inflammation that changes systemic functions, diet-induced alterations may contribute to neurodegenerative disorder progression as well. This study aims to non-invasively investigate diet-induced metabolic and inflammatory effects in the brain of an APPPS1 mouse model of Alzheimer’s disease. [¹⁸F]FDG, [¹⁸F]FTHA, and [¹⁸F]GE-180 were used for in vivo PET imaging in wild-type and APPPS1 mice. Ex vivo flow cytometry and histology in brains complemented the in vivo findings. ¹H- magnetic resonance spectroscopy in the liver, plasma metabolomics and flow cytometry of the white adipose tissue were used to confirm metaflammatory condition in the periphery. We found disrupted glucose and fatty acid metabolism after Western diet consumption, with only small regional changes in glial-dependent neuroinflammation in the brains of APPPS1 mice. Further ex vivo investigations revealed cytotoxic T cell involvement in the brains of Western diet-fed mice and a disrupted plasma metabolome. ¹H-magentic resonance spectroscopy and immunological results revealed diet-dependent inflammatory-like misbalance in livers and fatty tissue. Our multimodal imaging study highlights the role of the brain-liver-fat axis and the adaptive immune system in the disruption of brain homeostasis in amyloid models of Alzheimer’s disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-024-03080-0.
... A sedentary lifestyle, excessive consumption of saturated fat, or both bring on a major global health concern, obesity, which is defined by as Body Mass Index (BMI) being ≥30 kg/m 2 . Obesity is considered as a significant risk factor of AD [199,200]. Adipose tissue secretes the adipokine leptin, which rises with obesity and is thought to be the mediator of inflammation that leads to dementia [201]. Several prospective studies suggested midlife obesity as a risk factor for dementia. ...
Article
Full-text available
Alzheimer's disease (AD) is a age-related neurodegenerative disease and is a major public health concern both in Texas, US and Worldwide. This neurodegenerative disease is mainly characterized by amyloid-beta (Aβ) and phosphorylated Tau (p-Tau) accumulation in the brains of patients with AD and increasing evidence suggests that these are key biomarkers in AD. Both Aβ and p-tau can be detected through various imaging techniques (such as positron emission tomography, PET) and cerebrospinal fluid (CSF) analysis. The presence of these biomarkers in individuals, who are asymptomatic or have mild cognitive impairment can indicate an increased risk of developing AD in the future. Furthermore, the combination of Aβ and p-tau biomarkers is often used for more accurate diagnosis and prediction of AD progression. Along with AD being a neurodegenerative disease, it is associated with other chronic conditions such as cardiovascular disease, obesity, depression, and diabetes because studies have shown that these comorbid conditions make people more vulnerable to AD. In the first part of this review, we discuss that biofluid-based biomarkers such as Aβ, p-Tau in cerebrospinal fluid (CSF) and Aβ & p-Tau in plasma could be used as an alternative sensitive technique to diagnose AD. In the second part, we discuss the underlying molecular mechanisms of chronic conditions linked with AD and how they affect the patients in clinical care.
... WD and obesity are risk factors for neuropsychiatric and psychological disorders, such as mild cognitive impairment, dementia, and depression (Castanon et al. 2014;Pedditzi et al. 2016;Pistell et al. 2010;Xu et al. 2011). WD components could induce neurochemical changes in specific brain regions; in particular, the dysfunction of the hippocampus leads to an impaired cognitive state, disrupting normal intake control and memory tasks (Francis and Stevenson 2013;Kanoski and Davidson 2011;Stevenson et al. 2020). ...
Article
Full-text available
Diet is one of the most important external factor shaping the composition and metabolic activities of the gut microbiome. The gut microbiome plays a crucial role in host health, including immune system development, nutrients metabolism, and the synthesis of bioactive molecules. In addition, the gut microbiome has been described as critical for the development of several mental disorders. Nutritional psychiatry is an emerging field of research that may provide a link between diet, microbial function, and brain health. In this study, we have reviewed the influence of different diet types, such as Western, Mediterranean, vegetarian, and ketogenic, on the gut microbiota composition and function, and their implication in various neuropsychiatric and psychological disorders.
Article
Full-text available
Air pollution is an urgent concern linked to numerous health problems in low- and middle-income countries, where 92% of air pollution-related deaths occur. Particulate matter 2.5 (PM2.5) is the most harmful component of air pollutants, increasing inflammation and changing gut microbiota, favoring obesity, type 2 diabetes, and Alzheimer’s Disease (AD). PM2.5 contains lipopolysaccharides (LPS), which can activate the Toll-like receptor 4 (TLR4) signaling pathway. This pathway can lead to the release of pro-inflammatory markers, including interleukins, and suppressor of cytokine signaling-3 (SOCS3), which inhibits leptin action, a hormone that keeps the energy homeostasis. Leptin plays a role in preventing amyloid plaque deposition and hyperphosphorylation of tau-protein (p-tau), mechanisms involved in the neurodegeneration in AD. Approximately 50 million people worldwide are affected by dementia, with a significant proportion living in low—and middle-income countries. This number is expected to triple by 2050. This mini-review focuses on the potential impact of PM2.5 exposure on the TLR4 signaling pathway, its contribution to leptin resistance, and dysbiosis that exacerbates the link between obesity and AD.
Article
Once considered passive cells of the central nervous system (CNS), glia are now known to actively maintain the CNS parenchyma; in recent years, the evidence for glial functions in CNS physiology and pathophysiology has only grown. Astrocytes, a heterogeneous group of glial cells, play key roles in regulating the metabolic and inflammatory landscape of the CNS and have emerged as potential therapeutic targets for a variety of disorders. This review will outline astrocyte functions in the CNS in healthy ageing, obesity, and neurodegeneration, with a focus on the inflammatory responses and mitochondrial function, and will address therapeutic outlooks.
Article
Full-text available
Lifestyle choices leading to obesity, hypertension and diabetes in mid-life contribute directly to the risk of late-life Alzheimer’s disease (AD). However, in late-life or in late-stage AD conditions, obesity reduces the risk of AD and disease progression. To examine the mechanisms underlying this paradox, TgF344-AD rats were fed a varied high-carbohydrate, high-fat (HCHF) diet to induce obesity from nine months of age representing early stages of AD to twelve months of age in which rats exhibit the full spectrum of AD symptomology. We hypothesized regions primarily composed of gray matter, such as the somatosensory cortex (SSC), would be differentially affected compared to regions primarily composed of white matter, such as the striatum. We found increased myelin and oligodendrocytes in the somatosensory cortex of rats fed the HCHF diet with an absence of neuronal loss. We observed decreased inflammation in the somatosensory cortex despite increased AD pathology. Compared to the somatosensory cortex, the striatum had fewer changes. Overall, our results suggest that the interaction between diet and AD progression affects myelination in a brain region specific manner such that regions with a lower density of white matter are preferentially affected. Our results offer a possible mechanistic explanation for the obesity paradox.
Article
Context Studies have indicated a link between leptin, Alzheimer disease (AD), and Parkinson disease (PD). However, the causal relationship among them remains not well established due to confounders and reverse causation. Objective This Mendelian randomization (MR) study was performed to evaluate the impact and potential mechanism of leptin and its soluble receptor on AD and PD. Methods Utilizing genome-wide association studies summary-level data, a comprehensive MR was conducted to assess the causal effect of leptin and soluble leptin receptor (sLEPR) on AD and PD. Additionally, we also explored the role of body mass index (BMI) and insulin sensitivity index (ISI) in using the multivariable MR. The primary analysis was performed using the inverse variance-weighted method. Results Pooled estimates showed that genetically proxied higher leptin levels was significantly associated with a decreased risk of AD (OR 0.838, 95% CI 0.741-0.948, P = .005), but not PD. In contrast, no significant associations were observed between sLEPR levels, AD, and PD. Moreover, the effect of leptin on AD was attenuated to null after adjustment of ISI (OR 0.879, 95% CI 0.758-1.018, P = .086), but not BMI. There was no causal impact of AD and PD on circulating levels of leptin and sLEPR, indicating the absence of reverse causation. Sensitivity analyses confirmed the robustness of these associations, with no obvious pleiotropy and heterogeneity. Conclusion The study offers evidence supporting a potential protective effect of leptin in AD, but not PD, via the enhancement of insulin sensitivity. Our findings underscore the distinct roles of leptin in AD and PD.
Article
Full-text available
To examine if the body mass index (BMI) in midlife is related to cognitive function 30 years later in a dementia-free sample. BMI was reported in 1963 at age 50-60 years, and cognitive abilities were examined 30 years later in a longitudinal design with 5 measurement occasions at 2-year intervals (n = 417). The cognitive abilities examined included tests of long-term memory, short-term memory, speed, verbal and spatial ability. Multilevel modeling adjusting for demographic and lifestyle factors, and relevant diseases showed that a higher BMI in midlife predicted lower test performance 30 years later. Significant associations between BMI and level of performance were found in all cognitive abilities; however, a higher midlife BMI was not associated with steeper cognitive decline. Our results indicate that midlife overweight is related to lower overall cognitive function in old age. The fact that BMI-related effects were noted in mean-level cognitive performance, whereas only one ability showed differences in slopes, suggests that the negative effect of overweight has an onset before the entry into very old age.
Article
Full-text available
The prevalence of obesity increased in the United States between 1976-1980 and 1988-1994 and again between 1988-1994 and 1999-2000. To examine trends in obesity from 1999 through 2008 and the current prevalence of obesity and overweight for 2007-2008. Analysis of height and weight measurements from 5555 adult men and women aged 20 years or older obtained in 2007-2008 as part of the National Health and Nutrition Examination Survey (NHANES), a nationally representative sample of the US population. Data from the NHANES obtained in 2007-2008 were compared with results obtained from 1999 through 2006. Estimates of the prevalence of overweight and obesity in adults. Overweight was defined as a body mass index (BMI) of 25.0 to 29.9. Obesity was defined as a BMI of 30.0 or higher. In 2007-2008, the age-adjusted prevalence of obesity was 33.8% (95% confidence interval [CI], 31.6%-36.0%) overall, 32.2% (95% CI, 29.5%-35.0%) among men, and 35.5% (95% CI, 33.2%-37.7%) among women. The corresponding prevalence estimates for overweight and obesity combined (BMI > or = 25) were 68.0% (95% CI, 66.3%-69.8%), 72.3% (95% CI, 70.4%-74.1%), and 64.1% (95% CI, 61.3%-66.9%). Obesity prevalence varied by age group and by racial and ethnic group for both men and women. Over the 10-year period, obesity showed no significant trend among women (adjusted odds ratio [AOR] for 2007-2008 vs 1999-2000, 1.12 [95% CI, 0.89-1.32]). For men, there was a significant linear trend (AOR for 2007-2008 vs 1999-2000, 1.32 [95% CI, 1.12-1.58]); however, the 3 most recent data points did not differ significantly from each other. In 2007-2008, the prevalence of obesity was 32.2% among adult men and 35.5% among adult women. The increases in the prevalence of obesity previously observed do not appear to be continuing at the same rate over the past 10 years, particularly for women and possibly for men.
Article
Full-text available
The adipokine leptin facilitates long-term potentiation and synaptic plasticity in the hippocampus, promotes beta-amyloid clearance, and improves memory function in animal models of aging and Alzheimer disease (AD). To relate baseline circulating leptin concentrations in a community-based sample of individuals without dementia to incident dementia and AD during follow-up and magnetic resonance imaging (MRI) measures of brain aging in survivors. Prospective study of plasma leptin concentrations measured in 785 persons without dementia (mean [SD] age, 79 [5] years; 62% female), who were in the Framingham original cohort at the 22nd examination cycle (1990-1994). A subsample of 198 dementia-free survivors underwent volumetric brain MRI between 1999 and 2005, approximately 7.7 years after leptin was assayed. Two measures of brain aging, total cerebral brain volume and temporal horn volume (which is inversely related to hippocampal volume) were assessed. Incidence of dementia and AD during follow-up until December 31, 2007. During a median follow-up of 8.3 years (range, 0-15.5 years), 111 participants developed incident dementia; 89 had AD. Higher leptin levels were associated with a lower risk of incident dementia and AD in multivariable models (hazard ratio per 1-SD increment in log leptin was 0.68 [95% confidence interval, 0.54-0.87] for all-cause dementia and 0.60 [95% confidence interval, 0.46-0.79] for AD). This corresponds to an absolute AD risk over a 12-year follow-up of 25% for persons in the lowest quartile (first quartile) vs 6% for persons in the fourth quartile of sex-specific leptin levels. In addition, a 1-SD elevation in plasma leptin level was associated with higher total cerebral brain volume and lower temporal horn volume, although the association of leptin level with temporal horn volume did not reach statistical significance. Circulating leptin was associated with a reduced incidence of dementia and AD and with cerebral brain volume in asymptomatic older adults.
Article
Criteria for the diagnosis of vascular dementia (VaD) that are reliable, valid, and readily applicable in a variety of settings are urgently needed for both clinical and research purposes. To address this need, the Neuroepidemiology Branch of the National Institute of Neurological Disorders and Stroke (NINDS) convened an International Workshop with support from the Association Internationale pour la Recherche et l'Enseignement en Neurosciences (AIREN), resulting in research criteria for the diagnosis of VaD. Compared with other current criteria, these guidelines emphasize (1) the heterogeneity of vascular dementia syndromes and pathologic subtypes including ischemic and hemorrhagic strokes, cerebral hypoxic-ischemic events, and senile leukoencephalopathic lesions; (2) the variability in clinical course, which may be static, remitting, or progressive; (3) specific clinical findings early in the course (eg, gait disorder, incontinence, or mood and personality changes) that support a vascular rather than a degenerative cause; (4) the need to establish a temporal relationship between stroke and dementia onset for a secure diagnosis; (5) the importance of brain imaging to support clinical findings; (6) the value of neuropsychological testing to document impairments in multiple cognitive domains; and (7) a protocol for neuropathologic evaluations and correlative studies of clinical, radiologic, and neuropsychological features. These criteria are intended as a guide for case definition in neuroepidemiologic studies, stratified by levels of certainty (definite, probable, and possible). They await testing and validation and will be revised as more information becomes available.
Article
Article
Clinical criteria for the diagnosis of Alzheimer's disease include insidious onset and progressive impairment of memory and other cognitive functions. There are no motor, sensory, or coordination deficits early in the disease. The diagnosis cannot be determined by laboratory tests. These tests are important primarily in identifying other possible causes of dementia that must be excluded before the diagnosis of Alzheimer's disease may be made with confidence. Neuropsychological tests provide confirmatory evidence of the diagnosis of dementia and help to assess the course and response to therapy. The criteria proposed are intended to serve as a guide for the diagnosis of probable, possible, and definite Alzheimer's disease; these criteria will be revised as more definitive information become available.
Article
Prior work has suggested that obesity and overweight as measured by body mass index (BMI) increases risk of dementia. It is unknown if there is a difference in the risk of developing Alzheimer disease (AD) versus vascular dementia (VaD) associated with high body weight. The goal of this study was to examine the association between midlife BMI and risk of both AD and VaD an average of 36 years later in a large (N= 10,136) and diverse cohort of members of a health care delivery system. Participants aged 40-45 participated in health exams between 1964 and 1968. AD and VaD diagnoses were obtained from Neurology visits between January 1, 1994 and June 15, 2006. Those with diagnoses of general dementia from primary care providers were excluded from the study. BMI was analyzed in WHO categories of underweight, overweight and obese, as well as in subdivisions of WHO categories. All models were fully adjusted for age, education, race, sex, marital status, smoking, hyperlipidemia, hypertension, diabetes, ischemic heart disease and stroke. Cox proportional hazard models showed that compared to those with a normal BMI (18.5-24.9), those obese (BMI > or = 30) at midlife had a 3.10 fold increase in risk of AD (fully adjusted model, Hazard Ratio=3.10, 95% CI 2.19-4.38), and a five fold increase in risk of VaD (fully adjusted model, HR=5.01, 95% CI 2.98-8.43) while those overweight ( BMI > or = 25 and <30) had a two fold increase in risk of AD and VaD (fully adjusted model, HR=2.09, 95% CI 1.69-2.60 for AD and HR=1.95, 95% CI 1.29-2.96 for VaD). These data suggest that midlife BMI is strongly predictive of both AD and VaD, independent of stroke, cardiovascular and diabetes co morbidities. Future studies need to unveil the mechanisms between adiposity and excess risk of AD and VaD.
Article
High midlife and late-life adiposity may increase risk for dementia. Late-life decrease in body mass index (BMI) or body weight within several years of a dementia diagnosis has also been reported. Differences in study designs and analyses may provide different pictures of this relationship. Thirty-two years of longitudinal body weight, BMI, waist circumference, and waist-to-hip ratio (WHR) data, from the Prospective Population Study of Women in Sweden, were related to dementia. A representative sample of 1,462 nondemented women was followed from 1968 at ages 38-60 years, and subsequently in 1974, 1980, 1992, and 2000, using neuropsychiatric, anthropometric, clinical, and other measurements. Cox proportional hazards regression models estimated incident dementia risk by baseline factors. Logistic regression models including measures at each examination were related to dementia among surviving participants 32 years later. While Cox models showed no association between baseline anthropometric factors and dementia risk, logistic models showed that a midlife WHR greater than 0.80 increased risk for dementia approximately twofold (odds ratio 2.22, 95% confidence interval 1.00-4.94, p = 0.049) among surviving participants. Evidence for reverse causality was observed for body weight, BMI, and waist circumference in years preceding dementia diagnosis. Among survivors to age 70, high midlife waist-to-hip ratio may increase odds of dementia. Traditional Cox models do not evidence this relationship. Changing anthropometric parameters in years preceding dementia onset indicate the dynamic nature of this seemingly simple relationship. There are midlife and late-life implications for dementia prevention, and analytical considerations related to identifying risk factors for dementia.