DZ2 Si Conditii Relationate in Rel Cu Dementa

15
Journal of Alzheimer’s Disease 20 (2010) 723–736  723 DOI 10.3233/JAD-2010-091687 IOS Press Review Type 2 Diabetes and Related Conditions in Relation to Dementia: An Opportunity for Prevention? Jos ´ e A. Luchsinger Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA Taub Institute for Research of Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA  Division of General Medicine, Department of Medicine, Columbia University College of Physicians and Surge ons,  New Yor k, NY, USA  Department of Epidemiology , Joseph P . Mailman School of Public Health, Columbia University , New York, NY , USA Accepted 11 February 2010 Abstract . This manuscript provides a comprehensive review of the epidemiologic evidence linking type 2 diabetes (T2D) and its precursor condi tions, ele vat ed adipo sity and hyper insulinemia, to deme ntia. The mechanisms rela ting these condi tions to dementia may be vascular and non-vascular. Elevated adiposity in middle age is related to a higher risk of dementia but the data on this association in old age is conicting. Several studies have shown that hyperinsulinemia, a consequence of higher adiposity and insulin resistance, is also related to a higher risk of dementia, including late onset Alzheimer’s disease (LOAD). Studies have consistently shown a relation of T2D with higher dementia risk, but the associations are stronger for vascular dementia compared to LOAD. A large proportion of the world population may be at increased risk of dementia given the trends for increasing preva lence of overweight, obesity, hyperinsulinemia, and T2D. However , these associations may present a unique opportunity for prevention and treatment of dementia. There are sever al known modalities that are effectiv e in the prevention and T2D and the reduction of hyperinsulinemia including lifestyle interventions, metformin, thiazolideniodones, and acarbose. Several studies in the prevention and treatment of T2D are currently measuring cognitive outcomes and will provide information on whether T2D treatment and prevention can prevent cognitive decline and dementia. Keywords: Adiposity, Alzheimer’s disease, cognitive impairment, glucose, hyperinsulinemia, insulin, obesity, overweight, type 2 diabetes INTRODUCTION Late onset Alzheimer’s disease (LOAD) is the most common form of deme ntia, accountin g for betwe en 70% to over 90% of all cases [1], and its prevalence is expected to quadruple by the year 2047 in the US [2]. As much as 50% of the population aged 85 years and older, the fastest growing segment of the population, Correspondence to: Jos ´ e A. Luchsinger, MD, 630 West 168th St., PH19, New Yo rk, NY 10032, USA. Tel.: +1 212 305 4730; Fax: +1 212 305 2526; E-mail: [email protected]. may have LOAD [3]. V ascular dementia (VaD) is the second most common form of dementia, although it va rie s wid ely dep end ingon the cri ter ia use d [4]. Mix ed dementia is a term that has been coined to describe the mix of clinical features of LOAD and VaD. Other types of dementia suc h as fronto tempor al dement ia and Lewy body dement ia occ ur les s commonly and wil l not be covered in this revie w. The reference to dementia in this review is a reference to LOAD, VaD, or mixed dement ia. It is import ant to point out that patho lo- gy studies suggest that dementia, including LOAD, is more heterogeneous than previously recognized [5], in ISSN 1387-287 7/10/$27.5 0 © 2010 – IOS Press and the authors. All rights reserved

Transcript of DZ2 Si Conditii Relationate in Rel Cu Dementa

Page 1: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 1/15

Journal of Alzheimer’s Disease 20 (2010) 723–736   723DOI 10.3233/JAD-2010-091687IOS Press

Review

Type 2 Diabetes and Related Conditions in

Relation to Dementia: An Opportunity for

Prevention?

Jose A. Luchsinger∗

Gertrude H. Sergievsky Center, Columbia University, New York, NY, USATaub Institute for Research of Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA

 Division of General Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons,

 New York, NY, USA

 Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY, USA

Accepted 11 February 2010

Abstract. This manuscript provides a comprehensive review of the epidemiologic evidence linking type 2 diabetes (T2D) and

its precursor conditions, elevated adiposity and hyperinsulinemia, to dementia. The mechanisms relating these conditions to

dementia may be vascular and non-vascular. Elevated adiposity in middle age is related to a higher risk of dementia but the data

on this association in old age is conflicting. Several studies have shown that hyperinsulinemia, a consequence of higher adiposity

and insulin resistance, is also related to a higher risk of dementia, including late onset Alzheimer’s disease (LOAD). Studies haveconsistently shown a relation of T2D with higher dementia risk, but the associations are stronger for vascular dementia compared

to LOAD. A large proportion of the world population may be at increased risk of dementia given the trends for increasing

prevalence of overweight, obesity, hyperinsulinemia, and T2D. However, these associations may present a unique opportunity for

prevention and treatment of dementia. There are several known modalities that are effective in the prevention and T2D and the

reduction of hyperinsulinemia including lifestyle interventions, metformin, thiazolideniodones, and acarbose. Several studies in

the prevention and treatment of T2D are currently measuring cognitive outcomes and will provide information on whether T2D

treatment and prevention can prevent cognitive decline and dementia.

Keywords: Adiposity, Alzheimer’s disease, cognitive impairment, glucose, hyperinsulinemia, insulin, obesity, overweight, type

2 diabetes

INTRODUCTION

Late onset Alzheimer’s disease (LOAD) is the mostcommon form of dementia, accounting for between70% to over 90% of all cases [1], and its prevalence isexpected to quadruple by the year 2047 in the US [2].As much as 50% of the population aged 85 years andolder, the fastest growing segment of the population,

∗Correspondence to: Jose A. Luchsinger, MD, 630 West 168thSt., PH19, New York, NY 10032, USA. Tel.: +1 212 305 4730; Fax:+1 212 305 2526; E-mail: [email protected].

may have LOAD [3]. Vascular dementia (VaD) is the

second most common form of dementia, although it

varies widely dependingon the criteria used [4]. Mixed

dementia is a term that has been coined to describe

the mix of clinical features of LOAD and VaD. Other

types of dementia such as frontotemporal dementia and

Lewy body dementia occur less commonly and will not

be covered in this review. The reference to dementia

in this review is a reference to LOAD, VaD, or mixed

dementia. It is important to point out that patholo-

gy studies suggest that dementia, including LOAD, is

more heterogeneous than previously recognized [5], in

ISSN 1387-2877/10/$27.50 © 2010 – IOS Press and the authors. All rights reserved

Page 2: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 2/15

724   J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia

addition to the observation that elderly peoplewith nor-

mal cognition have brain pathology usually ascribed todementia. Thus, some propose that the classification of 

dementia into LOAD, VaD, andmixeddementia should

no longer be used. This is particularly important for

the topic at hand because Type 2 diabetes (T2D) and

its related disorders are known to cause cerebrovas-

cular disease, and it is not surprising that they cause

VaD. Whether they cause LOAD or Alzheimer’s dis-

ease (AD) pathology is a matter of controversy. Pre-

dementia cognitive impairment is of importance as a

potential target for secondary prevention. Mild cog-

nitive impairment (MCI) has been used to describe a

transitional state between normal cognitive functionand LOAD dementia [6,7] and has thus been target-

ed for interventions [8]. Individuals with MCI do not

have dementia but have memory complaints without

loss of function in their daily activities [7]. While gen-

eral cognitive performance is well preserved, memory

performance on standardized tests falls below expec-

tations for age and education. MCI can be classified

in amnestic and non-amnestic MCI. It is believed that

amnestic MCI is an early stage of LOAD, while non-

amnestic MCI, such as executive MCI, is less specific

to LOAD [6]. Persons with amnestic MCI progress to

LOAD at the rate of nearly 10% to 15% per year [6]compared to 1 to 2% in elderly persons with normal

cognition [7]. The prevalence of amnestic MCI varies

between 3% and 20% depending on the criteria ap-

plied [9], and increases from about 1% in persons 60

years old to 25% at age 85 [10]. Risk factors for de-

mentia can be studied in epidemiologic studies through

the outcomes of cognitive impairment or decline, MCI,

anddementia itself dementia, and this review will cover

evidence examining these outcomes.

This review attempts to cover the prevention of de-

mentia, not its treatment. Thus far, trials of vitamin

E [8] and homocysteine related vitamins [11] did notprevent cognitive decline in persons without dementia.

One trial of hypertension demonstrated a reduced risk 

of dementia in its treatment arm [12,13], but several

were negative. Trials of statins in the prevention of 

cognitive decline have also been negative [14] despite

epidemiological data suggestive of a benefit [15]. T2D

and its related conditions have surfaced as potential

modifiable risk factors for dementia. This manuscript

reviews the evidence linking these conditions to de-

mentia and potential modalities of diabetes prevention

that may be used in the prevention of dementia.

BURDEN OF OBESITY, INSULIN

RESISTANCE, AND TYPE 2 DIABETES

According to 2007 prevalence data from the Centers

for Disease Control and Prevention (CDC), T2D now

affects nearly 24 million people in the US, an increase

of more than 3 million in approximately two years [16].

T2D disproportionately affects the elderly, and almost

25% of the population 60 years and older had T2D in

2007. Another 57 million people have pre-diabetes,

making the prevalence of T2D and prediabetes over

50% in persons 60 years and older. It is projected that

a third of Americans born in 2000 will develop T2D,

with the highest lifetime risk among Hispanics (males,

45.4% and women, 52.5%) [17]. A rise in adiposity, or

body fat, is the cause of the increase in T2D [18]. Ab-

dominal adiposity, the accumulation of body fat around

the waist, also named central adiposity, seems to be the

most important predictor of T2D [19] and cardiovas-

cular disease [20]. Two-thirds of American adults are

overweight or obese [21]. The prevalence of abdomi-

nal obesity among US adults has increased continuous-

ly during the past 15 years. T2D risk rises with over-

weight/obesity (body mass index, BMI  25 kg/m2),

lack of physical activity, increasing age (45 years),

and family history [22]. T2D is also higher in African

Americans (11.8%) and Hispanics (10.4%) comparedto non-Hispanic Whites (6.6%) [23]. Those with oth-

er metabolic syndrome components (high blood pres-

sure, low HDL cholesterol, and high triglycerides) are

at higher risk of T2D [24,25]. The common link of 

these conditions (obesity, prediabetes, T2D, high blood

pressure, low HDL cholesterol, and high triglycerides)

is insulin resistance [26]. There are two underlying

mechanisms which lead to the onset of clinical T2D,

the resistance of target tissues that dispose of glucose,

such as muscle, to the actions of insulin (insulin resis-

tance resulting in hyperinsulinemia) and inadequate in-

sulin secretion from pancreatic β -cells [27]. The most

important contributing factors to insulin resistance are

obesity and a sedentary lifestyle, largely because of a

decrease in insulin sensitivity that leads to hyperinsu-

linemia [28–30]. In the natural history of progression

to T2D, pancreatic β -cells initially increase insulin se-

cretion in response to insulin resistance causing hyper-

insulinemia and are able to effectively maintain glu-

cose levels below the T2D range. When β -cell func-

tion begins to decline, insulin production is inadequate

to overcome the insulin resistance, and blood glucose

levels rise, resulting in pre-diabetes and T2D. Insulin

resistance, once established, remains relatively stable

Page 3: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 3/15

 J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia   725

over time. Therefore, progression of T2D is a result

of worsening  β -cell function with preexisting insulinresistance and hyperinsulinemia.

An implication of the natural history describedabove

is that when an epidemiologic study finds a relation be-

tween the components of this continuum and dementia

we cannot be certain if we are looking at a surrogate

marker of one of the other components (e.g., T2D is a

marker of past adiposity or hyperinsulinemia, obesity

is a marker of hyperinsulinemia) or if the important ex-

posure is the one we are examining. The answer could

be that there is an aggregate effect of all the compo-

nents in the lifespan. Biessels has published a review

that describes how individual components of the nat-

ural history that lead to T2D may affect cognition in

different critical periods of the lifespan [31].

POTENTIAL MECHANISMS RELATING

ELEVATED ADIPOSITY,

HYPERINSULINEMIA, AND TYPE 2

DIABETES WITH DEMENTIA

These conditions are related to cerebrovascular dis-

ease [32–37]. Elevated adiposity [38], hyperinsuline-

mia, T2D [39], and their clustering with other vascular

risk factors [40] are risk factors for stroke. In addition,insulin or related byproducts may affect the amyloid

cascade [41]. Thus, we classify the mechanisms link-

ing this continuum with LOAD as cerebrovascular and

non-cerebrovascular.

Cerebrovascular mechanisms

 Brain infarcts

Strokes, ascertained by clinical history [42] or as

brain infarcts on MRI [43], are related to a higher risk 

of dementia including LOAD. The mechanisms for this

association are not clear. However, pathology studies

have demonstrated that the presence of amyloid plaques

is lower in brains of persons with dementia who also

have infarcts [44,45], suggesting that the presence of 

infarcts is an insult that lowers the threshold of amy-

loid in the brain that is necessary to cause dementia.

The Religious Orders Study, a study of religious or-

ders across the United States based at Rush Universi-

ty in Chicago, found that T2D was related to infarcts

on autopsy but not AD pathology in persons with de-

mentia [46]. This observation suggests that the main

mechanism linking T2D to dementia is the presence of 

infarcts, which lowers the burden of amyloid necessary

to cause memory decline and dementia. However, the

Honolulu-Asia Aging Study [47], a study of Japanese-Americans, found that T2D was related to AD pathol-

ogy, particularly in persons with the APOE-ε4 allele.

The Adult Changes in Thought Study, based at the Uni-

versity of Washington, reported that persons without

DM and with dementia had a greater amyloid-β  pep-

tide load and in the cerebral cortex, while those with

both T2D and dementia patients had more microvas-

cular infarcts. The number of microvascular infarcts

was greater in persons with dementia and treated T2D,

whereas amyloid plaque load tended to be greater for

persons with dementia with untreated T2D [48]. The

interaction between infarcts and amyloid pathology in

persons with dementia and T2D seems complicated and

more studies are needed.

White matter disease

White matter disease, ascertained as white matter

hyperintensities (WHI) or leukoaraiosis on brain imag-

ing represents microvascular disease in the brain or de-

myelination. Some studies show an increase of WHI

in persons with T2D [49], which may suggest a mi-

crovascular ischemic insult in the brain. However, the

nature of WHI is still a matter of controversy. WHI are

thought to be ischemic in origin in the same way that

infarcts are [50] and have thus been proposed as sur-rogate markers of cerebrovascular disease [50]. How-

ever, recent evidence shows that WHI are common in

LOAD and maybe related to cerebral amyloid angiopa-

thy [51–54]. Thus, some WHI may be due to amyloid

disease and contribute to the development of LOAD.

WHI are commoncorrelates of cognitive impairment in

T2D [49], but it is unclear whether this WHI are mark-

ers of microvascular injury or may represent a process

related to amyloid deposition.

 Non-cerebrovascular mechanisms

 Hyperinsulinemia and amyloid-β  ( Aβ ) clearance

Hyperinsulinemia is a plausible risk factor forLOAD

independent of cerebrovascular disease because a) in-

sulin can cross the blood brain barrier [55], and pe-

ripheral insulin infusion in the elderly may affect Aβ 42levels in the cerebrospinal fluid (CSF) [56], a surrogate

marker of Aβ   clearance in the brain and an indirect

marker of LOAD risk; b) there are insulin receptors

in the brain including the hippocampus and entorhinal

cortex [57], structures affected early in LOAD [58];

c) insulin degrading enzyme (IDE) has been linked to

clearance of Aβ  in the brain, and insulin and Aβ   are

Page 4: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 4/15

726   J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia

both competing substrates for IDE [59]; and d) insulin

in the brain can increase the deposition of Aβ  and tauprotein phosphorylation, which are central to the patho-genesis of LOAD [55]. The pathways relating insulinin the periphery with Aβ  clearance in the brain are mul-tiple and complex. Craft and colleagues have reviewedhow peripheral hyperinsulinemia affects Aβ  clearancein the brain [60]. A potential pathway is that peripheralhyperinsulinemia downregulates insulin uptake in theblood brain barrier due to saturation over physiolog-ic levels [61]. This may result in reduction of insulinlevels in the brain and downregulation of expression of IDE [62] and reduction in IDE mediated amyloid re-duction [59]. This complex observation has been used

to support the use of rosiglitazone, an insulin sensitiz-er [63,64], and intranasal insulin [65] in the treatmentof LOAD.

 Advanced products of glycosylation ( AGE )AGE are most closely linked with glycemia and di-

abetes, as elevated glucose concentration promotes theMaillard reaction and AGE accrual. In a hyperglycemicenvironment, diabetic animal and human tissues con-tain increased AGE and upregulation of its receptor(RAGE) [66–68]. In fact, the most recognized AGE,hemoglobin A1c, represents the standard-of-care fortracking T2D glycemic control. AGE contribute impor-

tantly to diabetic complications. AGE in the basementmembranes of vessels promote vascular leakage [69].AGE is associated with diabetic nephropathy [70,71],retinal neovascularization [72,73], and diabetic neu-ropathy [74]. Increased expression of RAGE is ob-served in LOAD [75–77]. Expression of RAGE is en-hanced in blood vessels near Aβ  deposits in LOADbrain [75,78]. Along with increased total amount of RAGE in AD brain, there is a shift of RAGE distri-bution from neuron to microvasculature [79]. Aβ 1−40and Aβ 1−42 transport in the blood brain barrier can beblocked by RAGE-specific IgG or sRAGE, providingevidence of a potential target for prevention of LOAD

in persons with T2D.

 Lipoprotein related proteins ( LRP)LRP is a family of lipoprotein receptors that affect

lipid metabolism. LRP-1, found in the liver and oth-er tissues, clears Aβ  from plasma and also mediatestransport of Aβ  out of the brain [79,80]. LRP-1 is di-minished in insulin resistance without affecting lipidlevels [80]. Thus, LRP-1 is a plausible mechanismlinking hyperinsulinemia with Aβ  and LOAD. SolubleLRP (sLRP) facilitates the clearance of Aβ  by LRP-1and may be a therapeutic candidate for the treatment of LOAD [81,82].

Specific products of adipose tissue

Adipose tissue used to be thought as a fat depot,and it is increasingly recognized as a functioning or-

gan. In fact, it produces important factors such as lep-

tin and adiponectin, in addition to inflammatory fac-

tors [83]. Leptin is positively correlated with insulin,

while adiponectin is inversely correlated. Because

these factors correlate with insulin, it is not clear at this

point if they have a direct role in cognitive impairment

or are simply markers of hyperinsulinemia. However,

a recent longitudinal analysis in the Framingham study

showed that high leptin levels, which correlate posi-

tively with insulin levels, were related to a lower risk 

or LOAD [84] and larger brain volumes. This is not

consistent with the hypothesis that high insulin levels

cause LOAD, but may indicate an independent effect

of leptin on neurodegeneration as suggested by animal

studies [85].

SUMMARY OF PROSPECTIVE

EPIDEMIOLOGICAL STUDIES LINKING

ADIPOSITY, HYPERINSULINEMIA, AND

DIABETES TO DEMENTIA

 Adiposity

Elevated BMI in middle age may be associated with

higher dementia risk [86,87]. A recent study showed

that central adiposity in middle age was related to a

higher risk of dementia in older age [88]. Higher BMI

at ages 70, 75, and 79 years may also predict higher

dementia risk [89]. However, there have been reports

of no association at mid-life [90] and of lower BMI

related to higher LOAD risk [91,92] at older ages. A

study in Northern New York City [93] found that in

younger elderly (65 to 76 years of age), the associa-

tion between BMI quartiles and LOAD resembles a U

shaped-curve, while in the oldest old (> 76 years) high-

er BMI is related to a lower LOAD risk. This U-shaped

association has been reported for the relation between

adiposity and cardiovascular mortality [94] and under-

scores the difficulty in studying the effects of adiposi-

ty in older age [95]. This study also found that high-

er waist circumference is related to higher LOAD risk 

in the younger elderly, but not in the oldest old. The

Cardiovascular Health Study recently reported that el-

evated self-reported BMI at age 50 years was associ-

ated with a higher risk of dementia, while BMI at age

65 or older in the same individuals did not [96]. This

study underscores the importance of the period in life at

Page 5: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 5/15

 J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia   727

which adiposity is ascertained in relation to dementia.

Themost importantexplanationfor the paradox linkinglow weight in old age to dementia seems to be weight

loss. The mechanisms for this are not entirely clear.

They may include loss of olfaction [97,98], one of the

earliest manifestations of LOAD, which may lead to

decreased caloric intake, forgetfulness of meals [99],

and metabolic changes related to LOAD that are not

well understood. In this regard, LOAD is accompanied

by abnormalities in brain insulin signaling [100] which

could affect appetite and food intake. It is not clear

if weight loss is a consequence of LOAD, a parallel

process, or if it is related to potential causes of LOAD,

such as insulin resistance [101].

 Hyperinsulinemia

Several cross-sectional studies show an association

between hyperinsulinemia and an increased risk of 

LOAD [102–104]. Two longitudinal studies, one in

elderly Japanese Americans in Hawaii [105] and an-

other in elderly Black, Caribbean Hispanic, and Non-

Hispanic Whites in New York City [106], found that

the risk of incident LOAD was higher in persons with

hyperinsulinemia independent of a history of stroke.

These studies also found that the risk of LOAD related

to hyperinsulinemia was higheramongpersonswith theAPOE-ε4. The Nurses’ health study found that higher

C-peptide levels, a measure of insulin secretion [107],

and fasting insulin levels arerelated to cognitive decline

in women [108–110]. There is a paucity of prospective

epidemiologic studies exploring the relation between

markers of hyperinsulinemia and LOAD and more are

needed.

Type 2 diabetes

T2D has been related to a two-fold higher risk of 

developing MCI amongpostmenopausal women [111].

A multiethnic study in elderly from New York city

found that T2D was related to a higher risk of cognitive

impairment-no dementia with stroke although the effect

on cognitive impairment-no dementia without stroke

was not evident after adjusting for demographic vari-

ables and the presence of APOE-ε4 allele [112]. An

Italian study showed a non-statistically significant in-

crease of MCI with T2D in an elderly population [113],

while a Canadian study found that T2D was related on-

ly to vascular cognitive impairment-no dementia [114].

A study inNew York City found that T2D was related to

a higher risk of both amnestic and non-amnestic MCI,

underliningthe importance of T2D for both LOAD and

vascular cognitive impairment [115]. A recent studyin Olmstead county, Minnesota found that presence of 

T2D was not related to MCI risk, but longer T2D dura-

tion and treatment with insulin, a surrogate marker of 

T2D duration, were related to higher MCI risk [116].

Numerous studies have examined the relation be-

tween T2D and dementia. Table 1 shows the results

of some representative prospective studies in different

countries and age groups. In general, the association

between T2D and dementia seems to be stronger for

vascular dementia compared to LOAD, but these ob-

servations are inconsistent. Some studies have also re-

ported an interaction between T2D and the APOE-ε4

allele, while others have not found this interaction. Im-

portantly, the same study in Japanese Americans re-

ported no associations between T2D and dementia at

midlife [117], but strong associations when T2D was

ascertained in old age [47], underlining the importance

of when in the lifespan T2D is examined. This is of 

particular importance because T2D is more common in

elderly people and may not be diagnosed in the same

subjects in younger age.

The diagnosis of T2D is somewhat arbitrary and

many cases go undetected. Few studies have examined

the relation between continuous measures of glycemia

and dementia. One study in postmenopausal womenfound that the risk of MCI and dementia increased with

each 1% elevation in glycosylated hemoglobin, a sta-

ble measure of glucose levels, even in women without

T2D [118]. Glycosylated hemoglobin in persons with-

out T2D correlates with both glucose intolerance and

insulin resistance, and this study underscores the con-

tinuous nature of the relation between these constructs

and higher dementia risk.

 Metabolic syndrome

There is limited evidence on the association between

the metabolic syndrome and dementia in the elderly.

One study in 2,632 black and white elders found that

the metabolic syndrome was associated with a higher

risk of cognitive decline, particularly among those with

high inflammatory markers [119]. A cross-sectional

study in Europeans found that LOAD prevalence was

higher in persons with the metabolic syndrome [120].

In Northern New York City, the metabolic syndrome

was not related to LOAD risk, while T2D and hyperin-

sulinemia were [121]. The discrepancy between these

studies could be due to the fact that the study in New

York City was conducted in an older population, eth-

Page 6: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 6/15

728   J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia

Table 1Summary of representative prospective epidemiologic studies relating Type 2 diabetes (T2D) with dementia

First author, Year of Publication

Setting Findings

Leibson, 1997 [159] Rates of dementia in 1455 persons 45 yearsand older with T2D in Rochester, Minnesotawere compared to population rates.

Relative risk (RR) relating T2D and all cause dementia was1.66 (95% confidence interval (CI): 1.34–2.05), RR relatingT2D with AD was 2.27 for men (95% CI:1.55–3.31) and 1.37for women, (95% CI: 0.94–2.01).

Brayne, 1998 [160] 2609 persons 75 years and older in Cam-bridge, England

Odds ratios (OR) relating T2D with all cause dementia was2.62 (0.89–7.75), and 1.44 (1.05–17.00) for AD.

Ott, 1999 [161] 6370 persons 55 years and older in Rotter-dam, The Netherlands

T2D related to both all cause dementia [RR   =  1.9 [95% CI= 1.3 to 2.8]) and AD (RR 1.9 [1.2–3.1]). Risk of dementiahighest in persons treated with insulin (RR 4.3; 95% CI: 1.7–10.5]).

Curb, 1999 [117] 3,774 Japanese American men in Hawaii,United States, aged 45 to 68 years at the timeof T2D ascertainment and between 71 to 93years at the time of dementia ascertainment.

RR relating T2D with VD was 1.48; 95%CI: 0.79,2.78), and0.98 (95% CI: 0.48,1.99) for AD

Peila, 2002 [47] 2,574 Japanese-American men aged 77yearson averageenrolledin the Honolulu-AsiaAg-ing Study, Hawaii, United States. T2D wasascertained in older age

RR for total dementia was 1.5 (95% CI: 1.01–2.2), 1.8 for AD(95% CI: 1.1–2.9), 2.3 for vascular dementia (95% CI: 1.1–5.0). Individuals with both T2D and the APOE  ε4 allele hadan RR of 5.5 (CI 2.2–13.7) for AD compared with those withneither risk factor.

Arvanitakis, 2004 [162] 824 persons older than 55 years from the Re-ligious Orders Study in the United States

Hazard ratio (HR) relating T2D with AD was 1.65 (95% CI:1.10–2.47).

Luchsinger, 2004 [122] 1138 persons aged 65 years and older fromNorthern Manhattan, United States

Hazard ratio relating T2D and AD was 2.4 (95% CI: 1.8–3.2).

Schnaider-Beeri,2004 [163]

1,892 male civil servants aged 40 to 65 attime of T2D ascertainment in Israel

OR relating T2D at midlife with dementia 30 years later was2.83 [95% CI = 1.40 to 5.71]).

Xu, 2004 [164] 1,301 persons aged 75 years and older inStockholm, Sweden

HR for T2D were 1.5 (95% CI 1.0 to 2.1) for dementia, 2.6(95% CI 1.2 to 6.1) for VaD, and 1.3 (95% CI 0.9 to 2.1) forAD.

Whitmer, 2005 [165] 8,845 participantsofa healthmaintenanceor-ganization in California, United States, whowere between the ages of 40 and 44 at thetime of T2D ascertainment

HR relating T2D with dementia was 1.46, (95% CI: 1.19 to1.79)

Xu, 2007 [166] 1,173 persons without known T2D aged 75years and older in Stockholm, Sweden

Borderline T2D diagnosed with plasma glucose was associatedwith adjusted hazard ratios (95% CIs) of 1.67 (1.04–2.67) fordementia and 1.77 (1.06–2.97) for AD.

Irie, 2008 [167] 2547 persons 65 years and from the Cardio-vascular Health Study in the United States.

RR for AD 1.42 (95% CI: 1.02–1.97) but was 4.53 (95% CI:2.47–8.30) when the APOE-ε4 allele was also present. Therewas no association with vascular dementia.

nically diverse, and with a high prevalence of vascularrisk factors [122]. In Japanese Americans,the metabol-ic syndrome in middle age was associated with VaD,but not LOAD [123].

CAN DEMENTIA BE PREVENTED THROUGH

THE PREVENTION OR TREATMENT OF T2D?

T2D prevention

There are four types of interventions that have beendemonstrated to decrease the risk of T2D in persons

at risk though the increase of insulin sensitivity and

the reduction of insulin levels: Lifestyle intervention

(diet, exercise), metformin, acarbose, and rosiglita-

zone [124]. These interventions are also used to treat

T2D. Other treatments of T2D, such as sulfonylureas

and insulin [125], do not increase insulin sensitivity

and are not used for T2D prevention. Thus, they are

not discussed.

 Lifestyle interventions

Lifestyle interventions are the most effective [126]

way to lose weight, improve insulin sensitivity, and

Page 7: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 7/15

 J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia   729

prevent T2D. The Finnish diabetes prevention study

(FDPS) was a trial of lifestyle intervention versus nointervention in 522 middle aged persons with over-

weight or obesity and glucose intolerance [127]. The

lifestyle intervention consisted of counseling to de-

crease weight, decreasedintake of total fats and saturat-

ed fats, increased intakeof fiber, andincreased physical

activity. The risk of T2D was decreased by approx-

imately 58% in the intervention group after approxi-

mately three years of follow-up. This improvement

was achieved mostly through improved insulin sensi-

tivity, not increased insulin secretion [128]. More im-

portantly, lifestyle improvements and the risk of T2D

remained lower in the intervention group several years

after stopping the counseling [129]. Although lifestyle

interventions are difficult to adopt and maintain [130],

the experience of the FDPS indicates not only that the

intervention is effective and feasible, but that its effects

remain years after stopping counseling.

Another landmark study of lifestyle intervention is

the Diabetes Prevention Program (DPP). The DPP was

a trial of lifestyle versus metformin versus placebo in

over 3,000 participants with glucose intolerance [131].

The lifestyle intervention, which consisted of a pro-

gram to achieve weight loss and increased physical ac-

tivity, was the most effective with a 58% reduction in

theincidence of T2Dcomparedto placebo after 3 years,a reduction similar to that achieved in the FDPS. After

3 years the DPP became an observational study called

the DPP outcomes study (DPPOS), but participants re-

mained in their randomization groups and persons in

the placebo groupreceived a lifestyle intervention. The

DPPOS recently reported that benefits in the prevention

of T2D continue after 10 years of follow-up [132].

Importantly, both the FDPS and DPPOS include

comprehensive neurocognitive batteries starting in

2009 which will permit the explorationof whether T2D

prevention through lifestyle interventions prevents cog-

nitive decline and dementia in middle aged persons.

There is a rich literature of relatively small short term

studies of exercise and its effects on cognition [133],

which demonstrate a clear benefit of exercise particu-

larly on executive functions. The assessment of cog-

nition in the FDPS and DPPOS will allow the specific

examination of the effects of long term improvements

of insulin sensitivity and T2D risk on memory, and

non-memory cognitive abilities.

Thiazolidinediones

Thiazolideniodones are PPAR-γ  agonists and potent

insulin sensitizers [134]. The Diabetes reduction as-

sessment with ramipril and rosiglitazone medication

(DREAM) trial demonstrated in over 5000 participantswith impaired glucose tolerance that the thiazolidine-

dione rosiglitazone was effective in preventing T2D

during 3 years of intervention. Rosiglitazone seems to

be as effective as lifestyle interventions in preventing

T2D and provides twice the risk reduction compared to

metformin or acarbose [124]. In addition, thiazolidine-

diones are potent anti-inflammatory drugs [135]. Giv-

en the common role of inflammation the pathogenesis

of T2D [136] and AD [60,137], this anti-inflammatory

effect could also decrease the risk of AD. However,

the thiazolidinediones have been consistently shown to

have a higher risk of heart failure and edema [138–

141], and there are recent concerns of other cardio-

vascular adverse effects with rosiglitazone [142]. Pi-

oglitazone, another thiazolideniodone,has an increased

risk of edema and congestive heart failure, but seems

to lacks the other cardiovascular side effects associat-

ed with rosiglitazone [143]. Based on their powerful

insulin sensitizing actions, they are being studied as a

potential treatment of AD based on the hypothesis that

treating hyperinsulinemia lowers Aβ   deposition and

AD progression. One pilot 6 month trial of rosiglita-

zone in 30 subjects with mild AD or AMCI showed

that persons receiving 4 mg daily had better delayed

recall at months 4 and 6 and better selective attentionat month 6, and plasma Aβ  decreased in persons on

placebo while there was no change in persons on treat-

ment [63]. There was a decrease in insulin levels at 6

months demonstrating the metabolic effects of rosigli-

tazone and better cognitive performance was related to

lower insulin levels. A randomized placebo controlled

trial lasting 24 weeks of rosiglitazone 2, 4, or 8 mg in

511 persons with mild to moderate LOAD found no

effect in their primary outcomes (ADAS-COG) in the

ITT analysis [64]. There was a significant interaction

between APOE-ε4 and ADAS-COG, and persons on 8

mg rosiglitazone without any APOE-ε4 allele showed

an improvement in ADAS-COG (and improvements in

insulin levels), while person with APOE-ε4 showed no

benefit. However a recent Phase III trial of rosiglita-

zone (NCT00428090) in mild to moderateLOAD failed

to show a benefit [144]. It is possible, however, that the

useof thiazolidinedionesin MCI could improve therisk 

of dementia. The Rosiglitazone Effects on Cognition

for Adults in Later Life (RECALL; NCT00242593)

study is examining the effects of rosiglitazone on cog-

nition in persons with MCI and is estimated to finish

in 2010. The Pioglitazone or Exercise to Treat Mild

Cognitive Impairment (POEM; NCT00736996) is ex-

Page 8: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 8/15

730   J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia

ploring the effects of pioglitazonecompared to exercise

or placebo in persons with MCI and is scheduled to endin 2011. The major limitation of thiazolidinediones in

the prevention of dementia is the class side effects of 

edema and congestive heart failure, and the concerns

with increased cardiovascular morbidity with rosiglita-

zone compared to pioglitazone, which are still a matter

of debate.

 Metformin

Metformin is a medication belonging to the biguan-

ide class [145,146]. It treats and prevents diabetes

by suppression of hepatic glucose output, increasing

insulin-mediated glucose disposal, by increasedintesti-

nal glucose use, and by decreasing fatty acid oxida-

tion [147]; this is accompanied by reduced require-

ments for pancreatic insulin secretion and relatively

lower insulin levels in blood in response to glucose

load. While the mechanisms for the action of met-

formin are not completely understood, it clearly re-

duces insulin levels [148], inflammation and thrombo-

sis [149], and the risk of the metabolic syndrome [150]

and diabetes [151] in persons without diabetes. The

largest and longest experience for metformin in per-

sons without diabetes has been in the DPP. Metformin

was more effective than placebo but less effective than

lifestyle interventions in preventing T2D [131], and itclearly improved insulin sensitivity an decreased in-

sulin levels [148]. While metformin is a less potent

insulin sensitizer compared to thiazolidinediones and

lifestyle intervention, it has been proven to effectively

and safely prevent T2D. Its usual side effect is gastroin-

testinal intolerance, and its worse side effect, acidosis,

is rare and occurs in persons with severe congestive

heart failure, liver disease, or renal disease [145]. One

recent study in cellular models showed that metformin

increases the production of Aβ  through upregulation of 

β -secretase [152], and the authors raised the concern

that metformin could increase the risk of LOAD. How-

ever, this study needs to be replicated, and the relevance

of its findings to humans demonstrated. The effect of 

metformin on cognition will be assessed in the met-

formin arm of the DPPOS. Additionally, there is an on-

going Phase II trial of metformin (NCT00620191) test-

ing whether metformin can decrease cognitive decline

and dementia in persons with MCI.

 Acarbose

Acarbose reduces the risk of diabetes by decreas-

ing carbohydrate absorption in the intestine and post-

prandial insulin levels [153] but adherence to acarbose

is poor due to gastrointestinal side effects [153]. To the

best of my knowledge, there are no trials of acarbosein the prevention of dementia.

 Diabetes treatment in the prevention of dementia

One of the implications of the body of work sum-

marized in this article is that persons with T2D are at

increased risk of dementia and that its treatment could

affect dementia risk. This could happen in two poten-

tial ways. It is possible that tighter T2D control could

improve the risk of dementia in T2D. The three control

parameters followed in clinical practice are glycemia,

measured with hemoglobin A1c, blood pressure, andlipids, measured with low density lipoprotein. The

recently finalized “Action to Control Cardiovascular

Risk in Diabetes–Memory in Diabetes” (ACCORD-

MIND; NCT00182910) study will be able to answer

whether tighter control in these parameters reduces

the risk of cognitive decline and dementia in persons

with T2D [154]. One of the consequences of tight

glycemic control is hypoglycemia, and a recent obser-

vational study in elderly persons with T2D demonstrat-

ed that hypoglycemia was related to higher risk of de-

mentia [155]. An as of yet unpublished analysis of 

data from the Informatics in Diabetes Education andTelemedicineStudy (IDEATel) [156], a randomizedtri-

al of telemedicineversus usual care in 2169 elderly per-

sons with T2D, showed that persons in the intervention

group, which showed better control parameters com-

pared to usual care, had less global cognitive decline

during a maximum of 6 years of follow-up. Impor-

tantly, the glycemic control goals of IDEATel followed

glycemic guidelines which are less stringent than the

goals in ACCORD, which showed increased mortality

in its tight glycemic control arm [157] and less likely

to lead to hypoglycemia.

The other way in which T2D control could affect

dementia risk is with the type of medication used to

achieve glycemic control. In general, medications used

to treat T2D can be classified into insulin or insulin sec-

retagogues (sulfonylureas) or insulin sensitizers (met-

formin or thiazolidinediones) [125]. A newer class of 

T2D medication, the incretins, increase insulin secre-

tion but also seem to improve insulin sensitivity and

induce weight loss [158]. Given the possible role of 

hyperinsulinemia in AD pathology, one could specu-

late that insulin sensitizing medications could decrease

the risk of LOAD compared to other medications. This

has not been demonstrated and needs to be tested.

Page 9: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 9/15

 J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia   731

SUMMARY AND CONCLUSIONS

Many epidemiologic studies have shown that T2D

and its precursors, elevated adiposity and hyperinsu-

linemia, are related to a higher risk of dementia. In

general, these associations seem to be stronger for VaD

compared with LOAD. Pathologic data seem to suggest

that cerebrovascular disease decrease the threshold of 

amyloid burden needed to manifest dementia. Howev-

er, experimental and basic studies have demonstrated

plausible links between hyperinsulinemia and products

of hyperglycemia (e.g., AGE) in amyloid. The possibil-

ity that elevated adiposity, hyperinsulinemia, and T2D

increase the risk of dementia is alarming because a ma- jority of the adult population in the US have these con-

ditions. However, these conditions can be treated and

prevented, presenting a potential opportunity for de-

mentia prevention. Ongoing clinical trials will answer

whether interventions proven to improve insulin sensi-

tivity and decrease T2D risk can prevent dementia, in-

cluding lifestyle interventions, thiazolidinediones, and

metformin. We will also soon find out whether tight

diabetes control can improve dementia risk in persons

with T2D. There are no proven ways to modify the

risk of dementia. Ongoing clinical trials of amyloid

specific treatments such as vaccines,  β -secretase, and

γ -secretase inhibitors will tell us in the next five years

if targeting amyloid can treat or prevent dementia. If 

these trials are negative or of modest success, we may

only have the treatment and prevention of vascular and

metabolic conditions as potential ways to prevent de-

mentia in the near future.

ACKNOWLEDGMENTS

Dr. Luchsinger’s work in this review was support-

ed by grants from the National Institute on Aging

(AG026413, AG07232), NCMHD (P60 MD00206),

ISOA/ADDF (270901) the American Diabetes Asso-

ciation (7-08-CR-41) and by the Florence and Herbert

Irving Clinical Research Scholar’s Award.

The author’s disclosure is available online (http:// 

www.j-alz.com/disclosures/view.php?id=335).

REFERENCES

[1] Ritchie K, Lovestone S (2002) The dementias. Lancet  360,1759-1766.

[2] Brookmeyer R, Gray S, Kawas C (1998) Projections of Alzheimer’s disease in the United States and the public healthimpact of delaying disease onset.   Am J Public Health   88,1337-1342.

[3] Evans DA, Funkenstein HH, Albert MS, Scherr PA, Cook NR, Chown MJ,Hebert LE, Hennekens CH,Taylor JO (1989)Prevalence of Alzheimer’s disease in a community popula-tion of older persons. Higher than previously reported. JAMA

262, 2551-2556.[4] Chui HC, Mack W, Jackson JE, Mungas D, Reed BR, Tin-

klenberg J, Chang FL, Skinner K, TasakiC, Jagust WJ (2000)Clinical criteria for the diagnosis of vascular dementia: amulticenter study of comparability and interrater reliability.

 Arch Neurol 57, 191-196.[5] Savva GM, Wharton SB, Ince PG, Forster G, Matthews FE,

Brayne C (2009) Age, neuropathology, and dementia. N Engl

 J Med  360, 2302-2309.

[6] Luis CA, Loewenstein DA, Acevedo A, Barker WW, DuaraR (2003) Mild cognitive impairment: directions for futureresearch. Neurology 61, 438-444.

[7] Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG,Kokmen E (1999) Mild cognitive impairment: clinical char-acterization and outcome.  Arch Neurol  56, 303-308.

[8] Petersen RC, Thomas RG, Grundman M, Bennett D, DoodyR, Ferris S, Galasko D, Jin S, Kaye J, Levey A, Pfeiffer E,Sano M, van Dyck CH, Thal LJ, the Alzheimer’s DiseaseCooperative Study Group (2005) Vitamin E and donepezilfor the treatment of mild cognitive impairment. NEngl J Med 

352, 2379-2388.[9] Busse A, Bischkopf J, Riedel-Heller SG, Angermeyer MC

(2003) Mild cognitive impairment: prevalence and inci-dence according to different diagnostic criteria. Results of the Leipzig Longitudinal Study of the Aged (LEILA75+). Br 

 J Psychiatry  182, 449-454.[10] Yesavage JA, O’Hara R, Kraemer H, Noda A, Taylor JL,

Ferris S, Gely-Nargeot MC, Rosen A, Friedman L, SheikhJ, Derouesne C (2002) Modeling the prevalence and inci-dence of Alzheimer’s disease and mild cognitive impairment.

 J Psychiatr Res 36, 281-286.[11] McMahon JA, Green TJ, Skeaff CM, Knight RG, Mann JI,

Williams SM (2006) A controlled trial of homocysteine low-ering and cognitive performance.  N Engl J Med  354, 2764-2772.

[12] Forette F, Seux ML, Staessen JA, Thijs L, Birkenhager WH,Babarskiene MR, Babeanu S, Bossini A, Gil-Extremera B,Girerd X, Laks T, Lilov E, Moisseyev V, Tuomilehto J, Van-hanen H, Webster J, Yodfat Y, Fagard R (1998) Preventionof dementia in randomised double-blind placebo-controlledSystolic Hypertension in Europe (Syst-Eur) trial. Lancet 352,1347-1351.

[13] Forette F, Seux ML, Staessen JA, Thijs L, Babarskiene MR,Babeanu S, Bossini A, Fagard R, Gil-Extremera B, Laks T,Kobalava Z, Sarti C, Tuomilehto J, Vanhanen H, WebsterJ, Yodfat Y, Birkenhager WH (2002) The prevention of de-mentia with antihypertensive treatment: new evidence fromthe Systolic Hypertension in Europe (Syst-Eur) study.  Arch

 Intern Med  162, 2046-2052.[14] ShepherdJ, BlauwGJ, Murphy MB, BollenEL,BuckleyBM,

Cobbe SM, Ford I, Gaw A, Hyland M, Jukema JW, KamperAM, Macfarlane PW, Meinders AE, Norrie J, Packard CJ,Perry IJ, Stott DJ, Sweeney BJ, Twomey C, Westendorp RG,Risk PsgPSoPitEa (2002) Pravastatin in elderly individuals atriskof vasculardisease(PROSPER): a randomised controlledtrial. Lancet  360, 1623-1630.

Page 10: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 10/15

732   J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia

[15] Jick H, Zornberg GL, Jick SS, Seshadri S, Drachman DA(2000) Statins and the risk of dementia.  Lancet  356, 1627-1631.

[16] Centers for Disease Control and Prevention, Fact SheetPress Release: Number of People with Diabetes Increas-es to 24 million, http://www.cdc.gov/media/pressrel/2008/ r080624.htm, Accessed June 27.

[17] Narayan KM, Boyle JP, Thompson TJ, Sorensen SW,Williamson DF (2003) Lifetime risk for diabetes mellitus inthe United States.  JAMA 290, 1884-1890.

[18] Mokdad AH, Ford ES, Bowman BA, Dietz WH, Vinicor F,Bales VS, Marks JS (2003) Prevalence of obesity, diabetes,and obesity-related health risk factors, 2001.  JAMA 289, 76-79.

[19] Freemantle N, Holmes J, Hockey A, Kumar S (2008) Howstrong is the association between abdominal obesity and theincidence of type 2 diabetes?  Int J Clin Pract 62, 1391-1396.

[20] Yusuf S, Hawken S, Ounpuu S, Dans T, Avezum A, Lanas F,McQueen M, Budaj A, Pais P, Varigos J, Lisheng L (2004)Effect of potentially modifiable risk factors associated withmyocardial infarction in 52 countries (the INTERHEARTstudy): case-control study. Lancet  364, 937-952.

[21] Ogden CL, Carroll MD, Curtin LR, McDowell MA, Tabak CJ, Flegal KM (2006) Prevalence of overweight and obesityin the United States, 1999-2004. JAMA 295, 1549-1555.

[22] (2008)Standardsof medical carein diabetes– 2008. Diabetes

Care 31 Suppl 1, S12-54.[23] Brickman AM, Schupf N, Manly JJ, Luchsinger JA, An-

drews H, Tang MX, Reitz C, Small SA, Mayeux R, DeCar-li C, Brown TR (2008) Brain morphology in older AfricanAmericans, Caribbean Hispanics, and whites from northernManhattan. Arch Neurol  65, 1053-1061.

[24] Wilson PW, Meigs JB, Sullivan L, Fox CS, Nathan DM,

D’Agostino RB, Sr. (2007) Prediction of incident diabetesmellitus in middle-aged adults: the Framingham OffspringStudy. Arch Intern Med  167, 1068-1074.

[25] Florez H (2008) Diabetes risk engine with clinical variables. Int Diabetes Monitor  20, 119-120.

[26] Reaven G (2005) Insulin resistance, type 2 diabetes melli-tus, and cardiovascular disease: the end of the beginning.Circulation 112, 3030-3032.

[27] DeFronzo RA (1988) Lilly lecture 1987. The triumvirate:beta-cell, muscle, liver. A collusion responsible for NIDDM.

 Diabetes 37, 667-687.[28] Mokdad AH, Ford ES, Bowman BA, Nelson DE, Engelgau

MM, Vinicor F, Marks JS (2000) Diabetes trends in the U.S.:1990-1998.  Diabetes Care 23, 1278-1283.

[29] Choi BC, Shi F (2001) Risk factors for diabetes mellitusby age and sex: results of the National Population HealthSurvey. Diabetologia  44, 1221-1231.

[30] Hu FB,Manson JE, Stampfer MJ, Colditz G, Liu S, SolomonCG, Willett WC (2001) Diet, lifestyle, and the risk of type 2diabetes mellitus in women. N Engl J Med  345, 790-797.

[31] Biessels GJ, Deary IJ, Ryan CM (2008) Cognition and dia-betes: a lifespan perspective.  Lancet Neurol  7, 184-190.

[32] Boden-Albala B, Cammack S, Chong J, Wang C, Wright C,Rundek T, Elkind MS, Paik MC, Sacco RL (2008) Diabetes,fasting glucose levels, and risk of ischemic stroke and vas-cular events: findings from the Northern Manhattan Study(NOMAS). Diabetes Care 31, 1132-1137.

[33] Suk SH, Sacco RL, Boden-Albala B, Cheun JF, Pittman JG,Elkind MS, Paik MC (2003) Abdominal obesity and risk of ischemic stroke: the Northern Manhattan Stroke Study.Stroke  34, 1586-1592.

[34] Sacco RL (2002) Reducing the risk of stroke in diabetes:what have we learned that is new?   Diabetes Obes Metab  4

Suppl 1, S27-34.[35] Sacco RL, Benson RT, Kargman DE, Boden-Albala B, Tuck 

C, Lin I-F, Cheng JF, Paik MC, Shea S, Berglund L (2001)High-density lipoprotein cholesterol and ischemic stroke inthe elderly: The Northern Manhattan Stroke Study.  JAMA

285, 2729-2735.[36] Benson RT, Sacco RL (2000) Stroke prevention: hyperten-

sion, diabetes, tobacco, and lipids. Neurol Clin  18, 309-319.[37] Boden-Albala B, Sacco RL (2000) Lifestyle factors and

stroke risk: exercise, alcohol, diet, obesity, smoking, druguse, and stress. Curr Atheroscler Rep 2, 160-166.

[38] Pi-Sunyer FX (2002) The Obesity Epidemic: Pathophysiol-ogy and Consequences of Obesity. Obes Res 10, 97S-104.

[39] Sacco RL, Benjamin EJ, Broderick JP, Dyken M, Easton JD,Feinberg WM, Goldstein LB, Gorelick PB, Howard G, Kit-

tner SJ, Manolio TA, Whisnant JP, Wolf PA (1997) AmericanHeart Association Prevention Conference. IV. Prevention andRehabilitation of Stroke. Risk factors.  Stroke 28, 1507-1517.

[40] Grundy SM, Cleeman JI, Daniels SR, Donato KA, EckelRH, Franklin BA, Gordon DJ, Krauss RM, Savage PJ, SmithSC, Jr., Spertus JA, Costa F (2005) Diagnosis and manage-ment of the metabolic syndrome: An American Heart Asso-ciation/National Heart, Lung, and Blood Institute ScientificStatement. Circulation  112, 2735-2752.

[41] Craft S, Watson GS (2004) Insulin and neurodegenerativedisease: shared and specific mechanisms. Lancet Neurol  3,169-178.

[42] Honig LS, Tang MX, Albert S, Costa R, Luchsinger J, ManlyJ, Stern Y, Mayeux R (2003) Stroke and the risk of Alzheimerdisease. Arch Neurol 60, 1707-1712.

[43] Vermeer SE, Prins ND, den Heijer T, Hofman A, Koudstaal

PJ, Breteler MM (2003) Silent brain infarcts and the risk of dementia and cognitive decline.  N Engl J Med   348, 1215-1222.

[44] Snowdon DA, Greiner LH, Mortimer JA, Riley KP, GreinerPA, Markesbery WR (1997) Brain infarction and the clinicalexpression of Alzheimer disease. The Nun Study. JAMA 277,813-817.

[45] Schneider JA, Arvanitakis Z, Bang W, Bennett DA (2007)Mixed brain pathologies account for most dementia casesin community-dwelling older persons.  Neurology  69, 2197-2204.

[46] Arvanitakis Z, Schneider JA, Wilson RS, Li Y, Arnold SE,WangZ, Bennett DA (2006) Diabetes is related to cerebral in-farction but not to AD pathology in older persons. Neurology

67, 1960-1965.[47] Peila R, Rodriguez BL, Launer LJ (2002) Type 2 Diabetes,

APOEGene, andthe Risk for Dementia and Related Patholo-gies: The Honolulu-Asia Aging Study. Diabetes  51, 1256-1262.

[48] Sonnen JA, Larson EB, Brickell K, Crane PK, Woltjer R,Montine TJ, Craft S (2009) Different patterns of cerebralinjury in dementia with or without diabetes.  Arch Neurol 66,315-322.

[49] Manschot SM, Brands AMA, van der Grond J, Kessels RPC,Algra A, Kappelle LJ, Biessels GJ, on behalf of the UtrechtDiabeticEncephalopathy Study G (2006) Brainmagneticres-onance imaging correlates of impaired cognition in patientswith type 2 diabetes.  Diabetes  55, 1106-1113.

[50] Pantoni L (2006) White matter ischemia: Time to beginintegrating experimental and clinical data.   Eur Neurol   56,71-73.

Page 11: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 11/15

 J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia   733

[51] Nakata-Kudo Y, Mizuno T, Yamada K, Shiga K, YoshikawaK, Mori S, Nishimura T, Nakajima K, Nakagawa M (2006)Microbleeds in Alzheimer disease are more related to cere-bral amyloid angiopathy than cerebrovascular disease.  De-

ment Geriatr Cogn Disord  22, 8-14.[52] Haglund M, Englund E (2002) Cerebral amyloid angiopa-

thy, white matter lesions and Alzheimer encephalopathy – ahistopathological assessment. Dement Geriatr Cogn Disord 

14, 161-166.[53] Gurol ME, Irizarry MC, Smith EE, Raju S, Diaz-Arrastia R,

Bottiglieri T, Rosand J, Growdon JH, Greenberg SM (2006)Plasma{beta}-amyloidand whitematter lesions in AD,MCI,and cerebral amyloid angiopathy. Neurology 66, 23-29.

[54] Alonzo NC, Hyman BT, Rebeck GW, Greenberg SM (1998)Progression of cerebral amyloid angiopathy: accumulationof amyloid-beta40 in affected vessels.   J Neuropathol Exp

 Neurol 57, 353-359.

[55] Park CR (2001) Cognitive effects of insulin in the centralnervous system. Neurosci Biobehav Rev  25, 311-323.

[56] Watson GS, Peskind ER, Asthana S, Purganan K, Wait C,Chapman D, Schwartz MW, Plymate S, Craft S (2003) In-sulin increases CSF A{beta}42 levels in normal older adults.

 Neurology 60, 1899-1903.[57] Frolich L, Blum-Degen D, Bernstein HG, Engelsberger S,

Humrich J, Laufer S, Muschner D, Thalheimer A, Turk A,Hoyer S, Zochling R, Boissl KW, Jellinger K, Riederer P(1998) Brain insulin and insulin receptors in aging and spo-radic Alzheimer’s disease. J Neural Transm  105, 423-438.

[58] SmallSA,PereraGM,DeLaPazR, MayeuxR, SternY (1999)Differential regional dysfunction of the hippocampal forma-tion among elderly with memory decline and Alzheimer’sdisease. Ann Neurol 45, 466-472.

[59] Farris W, Mansourian S, Chang Y, Lindsley L, Eckman EA,

Frosch MP, Eckman CB, Tanzi RE, Selkoe DJ, GuenetteS (2003) Insulin-degrading enzyme regulates the levels of insulin, amyloid beta-protein, andthe beta-amyloidprecursorprotein intracellular domain  in vivo.  Proc Natl Acad Sci U S 

 A 100, 4162-4167.[60] Craft S (2007) Insulin resistance and Alzheimer’s disease

pathogenesis: potential mechanisms and implications fortreatment. Curr Alzheimer Res 4, 147-152.

[61] Banks WA, Jaspan JB, Huang W, Kastin AJ (1997) Trans-port of insulin across the blood-brain barrier: saturability ateuglycemic doses of insulin.  Peptides  18, 1423-1429.

[62] Zhao L, Teter B, Morihara T, Lim GP, Ambegaokar SS, Ube-da OJ, Frautschy SA, Cole GM (2004) Insulin-degrading en-zyme as a downstream target of insulin receptor signalingcascade: implications for Alzheimer’s disease intervention.

 J Neurosci  24, 11120-11126.[63] Watson GS, Cholerton BA, Reger MA, Baker LD, Plymate

SR, Asthana S, Fishel MA, Kulstad JJ, Green PS, Cook DG,Kahn SE, Keeling ML, Craft S (2005) Preserved cognitionin patients with early alzheimer disease and amnestic mildcognitive impairment during treatment with rosiglitazone: apreliminary study. Am J Geriatr Psychiatry  13, 950-958.

[64] Risner ME, Saunders AM, Altman JF, Ormandy GC, Craft S,Foley IM, Zvartau-Hind ME, Hosford DA, Roses AD (2006)Efficacy of rosiglitazone in a genetically defined populationwith mild-to-moderate Alzheimer’s disease.   Pharmacoge-

nomics J  6, 246-254.[65] Reger MA, Watson GS, Frey WH, 2nd, Baker LD, Choler-

ton B, Keeling ML, Belongia DA, Fishel MA, Plymate SR,Schellenberg GD, Cherrier MM, Craft S (2006) Effects of intranasal insulin on cognition in memory-impaired older

adults: modulation by APOE genotype. Neurobiol Aging 27,451-458.

[66] Goldin A, Beckman JA, Schmidt AM, Creager MA (2006)Advanced glycation end products: sparking the developmentof diabetic vascular injury. Circulation  114, 597-605.

[67] Basta G, Schmidt AM, De Caterina R (2004) Advanced gly-cation end products and vascular inflammation: implicationsfor accelerated atherosclerosis in diabetes.   Cardiovasc Res

63, 582-592.[68] Negrean M, Stirban A, Stratmann B, Gawlowski T,

Horstmann T, Gotting C, Kleesiek K, Mueller-Roesel M,Koschinsky T, Uribarri J, Vlassara H, Tschoepe D (2007) Ef-fects of low- and high-advanced glycation endproduct mealson macro- and microvascular endothelial function and ox-idative stress in patients with type 2 diabetes mellitus.  Am J 

Clin Nutr  85, 1236-1243.[69] Vlassara H, Striker LJ,Teichberg S, FuhH, Li YM,Steffes M

(1994) Advanced glycation end products induce glomerularsclerosis and albuminuria in normal rats.  Proc Natl Acad Sci

U S A 91, 11704-11708.[70] Peppa M, Brem H, Cai W, Zhang JG, Basgen J, Li Z, Vlas-

sara H, Uribarri J (2006) Prevention and reversal of diabeticnephropathy in db/db mice treated with alagebrium (ALT-711).  Am J Nephrol 26, 430-436.

[71] Thallas-Bonke V, Lindschau C, Rizkalla B, Bach LA, Bon-er G, Meier M, Haller H, Cooper ME, Forbes JM (2004)Attenuation of extracellular matrix accumulation in diabeticnephropathy by the advanced glycation end product cross-linkbreaker ALT-711via a protein kinase C-alpha-dependentpathway. Diabetes  53, 2921-2930.

[72] Stitt AW (2001) Advanced glycation: an important patho-logical event in diabetic and age related ocular disease. Br J 

Ophthalmol 85, 746-753.

[73] Schalkwijk CG, Lieuw-a-Fa M, van Hinsbergh VW, Ste-houwer CD (2002) Pathophysiological role of Amadori-glycated proteins in diabetic microangiopathy.   Semin Vasc

 Med  2, 191-197.[74] Sullivan KA, Feldman EL (2005) New developments in dia-

betic neuropathy. Curr Opin Neurol  18, 586-590.[75] Yan SD, Chen X, Fu J, Chen M, Zhu H, Roher A, Slattery

T, Zhao L, Nagashima M, Morser J, Migheli A, NawrothP, Stern D, Schmidt AM (1996) RAGE and amyloid-betapeptide neurotoxicity in Alzheimer’s disease.   Nature   382,685-691.

[76] Schmidt AM, Yan SD, Yan SF, SternDM (2000) The biologyof the receptor for advanced glycation end products and itsligands.  Biochim Biophys Acta  1498, 99-111.

[77] Lue LF, Walker DG, Brachova L, Beach TG, Rogers J,Schmidt AM, Stern DM, Yan SD (2001) Involvement of microglial receptor for advanced glycation endproducts(RAGE) in Alzheimer’s disease: identification of a cellularactivation mechanism. Exp Neurol 171, 29-45.

[78] Deane R, Du Yan S, Submamaryan RK, LaRue B, JovanovicS,Hogg E,Welch D,MannessL, Lin C,YuJ, Zhu H,GhisoJ,Frangione B, Stern A, Schmidt AM, Armstrong DL, ArnoldB, Liliensiek B, Nawroth P, Hofman F, Kindy M, Stern D,Zlokovic B (2003) RAGE mediates amyloid-beta peptidetransport across the blood-brain barrier and accumulation inbrain. Nat Med  9, 907-913.

[79] Donahue JE, Flaherty SL, Johanson CE, Duncan JA, 3rd, Sil-verberg GD, Miller MC, Tavares R, Yang W, Wu Q, Sabo E,Hovanesian V, Stopa EG (2006) RAGE, LRP-1, and amyloid-beta protein in Alzheimer’s disease.  Acta Neuropathol ( Berl)112, 405-415.

Page 12: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 12/15

734   J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia

[80] Tamaki C, Ohtsuki S, Terasaki T (2007) Insulin facilitatesthe hepatic clearance of plasma amyloid beta-peptide (140) by intracellular translocation of low-density lipoproteinreceptor-related protein 1 (LRP-1) to the plasma membranein hepatocytes.  Mol Pharmacol 72, 850-855.

[81] Sagare A, Deane R, Bell RD, Johnson B, Hamm K, PenduR, Marky A, Lenting PJ, Wu Z, Zarcone T, Goate A, MayoK, Perlmutter D, Coma M, Zhong Z, Zlokovic BV (2007)Clearance of amyloid-beta by circulating lipoprotein recep-tors. Nat Med  13, 1029-1031.

[82] Deane R,WuZ, Sagare A,DavisJ, DuYanS, HammK, XuF,ParisiM, LaRue B, HuHW,Spijkers P, GuoH, Song X, Lent-ing PJ, Van Nostrand WE, Zlokovic BV (2004) LRP/amyloidbeta-peptide interaction mediates differential brain efflux of Abeta isoforms. Neuron 43, 333-344.

[83] Yu YH, Ginsberg HN (2005) Adipocyte signaling and lipidhomeostasis: sequelae of insulin-resistant adipose tissue.

Circ Res 96, 1042-1052.[84] Lieb W, Beiser AS, Vasan RS, Tan ZS, Au R, Harris TB,

Roubenoff R, Auerbach S, Decarli C, Wolf PA, SeshadriS (2009) Association of plasma leptin levels with incidentAlzheimer disease and MRI measures of brain aging.  JAMA

302, 2565-2572.[85] Carro EM (2009) Therapeutic approaches of leptin in

Alzheimer’s disease.  Recent Pat CNS Drug Discov   4, 200-208.

[86] Kivipelto M, Ngandu T, Fratiglioni L, Viitanen M, Kareholt I,Winblad B, Helkala EL, Tuomilehto J, Soininen H, NissinenA (2005) Obesity and vascular risk factors at midlife andthe risk of dementia and Alzheimer disease.  Arch Neurol 62,1556-1560.

[87] Whitmer RA, Gunderson EP, Barrett-Connor E, QuesenberryCP, Jr, Yaffe K (2005) Obesity in middle age and future risk 

of dementia: a 27 year longitudinal population based study. BMJ  330, 1360.

[88] Whitmer RA, Gustafson DR, Barrett-Connor E, Haan MN,Gunderson EP, Yaffe K (2008) Central obesity and increasedrisk of dementia more than three decades later. Neurology 71,1057-1064.

[89] Gustafson D, Rothenberg E, Blennow K, Steen B, SkoogI (2003) An 18-year follow-up of overweight and risk of Alzheimer disease. Arch Intern Med  163, 1524-1528.

[90] Stewart R, Masaki K, Xue Q-L, Peila R, Petrovitch H, WhiteLR, Launer LJ (2005) A 32-year prospective study of changein body weight and incident dementia: The Honolulu-AsiaAging Study. Arch Neurol 62, 55-60.

[91] Atti AR, Palmer K, Volpato S, Winblad B, De Ronchi D,Fratiglioni L (2008) Late-life body mass index and dementiaincidence: nine-year follow-up data from the KungsholmenProject. J Am Geriatr Soc  56, 111-116.

[92] Nourhashemi F, Deschamps V, Larrieu S, Letenneur L, Dar-tigues JF, Barberger-Gateau P, Quid PsPA (2003) Body massindex and incidence of dementia: the PAQUID study.  Neu-

rology  60, 117-119.[93] Luchsinger JA, Patel B, Tang MX, Schupf N, Mayeux R

(2007) Measures of adiposity and dementia risk in elderlypersons. Arch Neurol 64, 392-398.

[94] Stevens J, Cai J, Pamuk ER, Williamson DF, ThunMJ, WoodJL (1998) The effect of age on the association between body-mass index and mortality. N Engl J Med , 1-7.

[95] Stevens J (2000) Impact of age on associations betweenweight and mortality.  Nutr Rev 58, 129-137.

[96] Fitzpatrick AL, Kuller LH, Lopez OL, Diehr P, O’Meara ES,Longstreth WT, Jr., Luchsinger JA (2009) Midlife and late-

life obesity and the risk of dementia: cardiovascular healthstudy. Arch Neurol 66, 336-342.

[97] Tabert MH, Liu X,DotyRL,SerbyM, ZamoraD, PeltonGH,Marder K, Albers MW, Stern Y, Devanand DP (2005) A 10-item smell identification scale related to risk for Alzheimer’sdisease. Ann Neurol 58, 155-160.

[98] Devanand DP, Michaels-Marston KS, Liu X, Pelton GH,Padilla M, Marder K, Bell K, Stern Y, Mayeux R (2000)Olfactory deficits in patients with mild cognitive impairmentpredict Alzheimer’s disease at follow-up.  Am J Psychiatry

157, 1399-1405.[99] Gustafson D (2008) A life course of adiposity and dementia.

 Eur J Pharmacol 585, 163-175.[100] Steen E, Terry BM, Rivera EJ, Cannon JL, Neely TR,Tavares

R, Xu XJ, Wands JR, de la Monte SM (2005) Impaired in-sulin and insulin-like growth factor expression and signalingmechanisms in Alzheimer’s disease–is this type 3 diabetes?

 J Alzheimers Dis 7, 63-80.[101] Wedick NM, Mayer-Davis EJ, Wingard DL, Addy CL,

Barrett-Connor E (2001) Insulin resistance precedes weightloss in adults without diabetes: The Rancho Bernardo Study.

 Am J Epidemiol 153, 1199-1205.[102] Razay G, Wilcock GK (1994) Hyperinsulinaemia and

Alzheimer’s disease. Age Ageing  23, 396-399.[103] Kuusisto J, Koivisto K, Mykkanen L, Helkala EL, Vanhanen

M, Hanninen T, Kervinen K, Kesaniemi YA, Riekkinen PJ,Laakso M (1997) Association between features of the insulinresistance syndrome and Alzheimer’s disease independentlyof apolipoprotein E4 phenotype: cross sectional populationbased study. BMJ  315, 1045-1049.

[104] Stolk RP, Breteler MM, Ott A, Pols HA, Lamberts SW,Grobbee DE, Hofman A (1997) Insulin and cognitive func-tion in an elderly population. The Rotterdam Study. Diabetes

Care 20, 792-795.[105] Peila R, Rodriguez BL, White LR, Launer LJ (2004) Fasting

insulin and incident dementia in an elderly population of Japanese-American men. Neurology 63, 228-233.

[106] Luchsinger JA, Tang M-X, Shea S, Mayeux R (2004) Hy-perinsulinemia and risk of Alzheimer disease.  Neurology 63,1187-1192.

[107] HarrisMI, CowieCC, GuK, Francis ME,Flegal K, EberhardtMS (2002) Higher fasting insulin but lower fasting C-peptidelevels in African Americans in the US population.  Diabetes

 Metab Res Rev 18, 149-155.[108] Okereke OI, Pollak MN, Hu FB, Hankinson SE, Selkoe DJ,

Grodstein F (2008) Plasma C-peptide levels and rates of cog-nitive decline in older, community-dwelling women withoutdiabetes. Psychoneuroendocrinology  33, 455-461.

[109] van Oijen M, Okereke OI, Kang JH, Pollak MN, Hu FB,Hankinson SE, Grodstein F (2008) Fasting insulin levels andcognitive decline in olderwomenwithout diabetes. Neuroepi-

demiology 30, 174-179.[110] Okereke O, Hankinson SE, Hu FB, Grodstein F (2005) Plas-

ma C peptide level and cognitive function among older wom-en without diabetes mellitus.  Arch Intern Med   165, 1651-1656.

[111] Yaffe K, Blackwell T, Kanaya AM, Davidowitz N, Barrett-Connor E, Krueger K (2004) Diabetes, impaired fasting glu-cose, and development of cognitive impairment in olderwomen. Neurology 63, 658-663.

[112] Luchsinger JA, Tang MX, Stern Y, Shea S, Mayeux R (2001)Diabetes mellitus and risk of Alzheimer’s disease and de-mentia with stroke in a multiethnic cohort.  Am J Epidemiol

154, 635-641.

Page 13: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 13/15

 J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia   735

[113] Solfrizzi V, Panza F, Colacicco AM, D’Introno A, CapursoC, Torres F, Grigoletto F, Maggi S, Del Parigi A,Reiman EM,Caselli RJ, Scafato E, Farchi G, Capurso A, for the ItalianLongitudinal Study on Aging Working Group (2004) Vascu-lar risk factors, incidence of MCI, and rates of progressionto dementia. Neurology  63, 1882-1891.

[114] MacKnight C, Rockwood K, Awalt E, McDowellI (2002)Di-abetes mellitus and the risk of dementia, Alzheimer’s diseaseand vascular cognitive impairment in the Canadian Study of Health and Aging. Dement Geriatr Cogn Disord  14, 77-83.

[115] Luchsinger JA, Reitz C, Patel B, Tang M-X, Manly JJ,Mayeux R (2007) Relation of diabetes to mild cognitive im-pairment. Arch Neurol 64, 570-575.

[116] Roberts RO, Geda YE, Knopman DS, Christianson TJ,Pankratz VS, Boeve BF, Vella A, Rocca WA, Petersen RC(2008) Association of duration and severity of diabetes mel-litus with mild cognitive impairment.  Arch Neurol 65, 1066-

1073.[117] Curb JD, Rodriguez BL, Abbott RD, Petrovitch H, Ross

GW, Masaki KH, Foley D, Blanchette PL, Harris T, ChenR, White LR (1999) Longitudinal association of vascularand Alzheimer’s dementias, diabetes, and glucose tolerance.

 Neurology 52, 971-975.[118] Yaffe K, Blackwell T, Whitmer RA, Krueger K, Barrett Con-

nor E (2006) Glycosylated hemoglobin level and develop-mentof mildcognitive impairment or dementia in olderwom-en. J Nutr Health Aging 10, 293-295.

[119] Yaffe K, Kanaya A, Lindquist K, Simonsick EM, Harris T,Shorr RI, Tylavsky FA, Newman AB (2004) The metabolicsyndrome, inflammation, and riskof cognitive decline. JAMA

292, 2237-2242.[120] Vanhanen M, Koivisto K, Moilanen L, Helkala EL, Hanni-

nen T, Soininen H, Kervinen K, Kesaniemi YA, Laakso M,

Kuusisto J (2006) Association of metabolic syndrome withAlzheimer disease: A population-based study. Neurology 67,843-847.

[121] Muller M, Tang MX, Schupf N, Manly JJ, Mayeux R,Luchsinger JA(2007) Metabolic syndrome and dementia risk in a multiethnic elderly cohort.  Dement Geriatr Cogn Disord 

24, 185-192.[122] Luchsinger JA, Reitz C, Honig LS, Tang MX, Shea S,

Mayeux R (2005) Aggregation of vascular risk factors andrisk of incident Alzheimer disease. Neurology 65, 545-551.

[123] Kalmijn S, Foley D, White L, Burchfiel CM, Curb JD, Petro-vitch H, Ross GW, Havlik RJ, Launer LJ (2000) Metaboliccardiovascular syndrome and risk of dementia in Japanese-American elderly men. The Honolulu-Asia aging study.  Ar-

terioscler Thromb Vasc Biol  20, 2255-2260.[124] Nathan DM, Berkwits M (2007) Trials that matter: rosigli-

tazone, ramipril, and the prevention of type 2 diabetes.  Ann

 Intern Med  146, 461-463.[125] DeFronzo RA (2000) Pharmacologic therapy for type 2 dia-

betes mellitus.  Ann Int Med  133, 73-74.[126] Orchard TJ, Temprosa M, Goldberg R, Haffner S, Ratner R,

Marcovina S, Fowler S, for the Diabetes Prevention ProgramResearch G (2005) The effect of metformin and intensivelifestyle intervention on the metabolic syndrome: The Dia-betes Prevention Program Randomized Trial. Ann Intern Med 

142, 611-619.[127] Tuomilehto J, Lindstrom J, Eriksson JG, Valle TT,

Hamalainen H, Ilanne-Parikka P, Keinanen-Kiukaanniemi S,Laakso M, Louheranta A, Rastas M, Salminen V, UusitupaM (2001) Prevention of type 2 diabetes mellitus by changes

in lifestyle among subjects with impaired glucose tolerance. N Eng l J Med  344, 1343-1350.

[128] Uusitupa M, Lindi V, Louheranta A, Salopuro T, LindstromJ, Tuomilehto J (2003) Long-term improvement in insulinsensitivity by changing lifestyles of people with impairedglucose tolerance: 4-year results from the Finnish diabetesprevention study. Diabetes  52, 2532-2538.

[129] Lindstrom J, Ilanne-Parikka P, Peltonen M, Aunola S, Eriks-son JG, Hemio K, Hamalainen H, Harkonen P, Keinanen-Kiukaanniemi S, Laakso M, Louheranta A, Mannelin M,Paturi M, Sundvall J, Valle TT, Uusitupa M, Tuomilehto J(2006) Sustained reduction in the incidence of type2 diabetesby lifestyle intervention: follow-up of the Finnish DiabetesPrevention Study. Lancet  368, 1673-1679.

[130] Norris SL, Zhang X, Avenell A, Gregg E, Schmid CH, LauJ (2005) Long-term non-pharmacological weight loss inter-ventionsfor adults with prediabetes. Cochrane Database Syst 

 Rev, CD005270.[131] Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF,

Lachin JM, Walker EA, Nathan DM (2002) Reduction in theincidence of type 2 diabetes with lifestyle intervention ormetformin. N Engl J Med  346, 393-403.

[132] Knowler WC, Fowler SE,Hamman RF, Christophi CA,Hoff-man HJ, Brenneman AT,Brown-Friday JO, GoldbergR, Ven-ditti E, Nathan DM (2009) 10-year follow-up of diabetes in-cidence and weight loss in the Diabetes Prevention ProgramOutcomes Study. Lancet  374, 1677-1686.

[133] Colcombe S, Kramer AF (2003) Fitness effects on the cogni-tive function of older adults: a meta-analytic study. Psychol

Sci 14, 125-130.[134] Yki-Jarvinen H (2004) Thiazolidinediones.   N Engl J Med 

351, 1106-1118.[135] Nesto R (2004) C-reactive protein, its role in inflammation,

Type 2 diabetes and cardiovascular disease, and the effects of insulin-sensitizing treatment with thiazolidinediones. Diabet 

 Med  21, 810-817.[136] Cefalu WT (2009) Inflammation, insulin resistance, and type

2 diabetes: back to the future?   Diabetes 58, 307-308.[137] Tuppo EE, Arias HR (2005) The role of inflammation in

Alzheimer’s disease. Int J Biochem Cell Biol 37, 289-305.[138] Dormandy JA, Charbonnel B, Eckland DJ, Erdmann E,

Massi-Benedetti M, Moules IK, Skene AM, Tan MH, Lefeb-vre PJ, Murray GD, Standl E, Wilcox RG, Wilhelmsen L,Betteridge J, Birkeland K, Golay A, Heine RJ, Koranyi L,Laakso M, Mokan M, Norkus A, Pirags V, Podar T, ScheenA, Scherbaum W, Schernthaner G, Schmitz O, Skrha J, SmithU, Taton J (2005) Secondary prevention of macrovascularevents in patients with type 2 diabetes in the PROactive Study(PROspective pioglitAzone Clinical Trial In macroVascularEvents): a randomised controlled trial.   Lancet   366, 1279-1289.

[139] Nesto RW, Bell D, Bonow RO, Fonseca V, Grundy SM,Horton ES, Le Winter M, Porte D, Semenkovich CF, SmithS, Young LH, Kahn R (2003) Thiazolidinedione use, fluidretention, and congestive heart failure: a consensus statementfromthe AmericanHeart Associationand American DiabetesAssociation. Circulation 108, 2941-2948.

[140] Kahn SE, Haffner SM, Heise MA, Herman WH, HolmanRR, Jones NP, Kravitz BG, Lachin JM, O’Neill MC, ZinmanB, Viberti G (2006) Glycemic durability of rosiglitazone,metformin, or glyburide monotherapy.   N Engl J Med   355,2427-2443.

[141] Nissen SE, Wolski K, Topol EJ (2005) Effect of muraglitazar

Page 14: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 14/15

736   J.A. Luchsinger / Type 2 Diabetes and Related Conditions in Relation to Dementia

on death and major adverse cardiovascular events in patientswith type 2 diabetes mellitus.  JAMA 294, 2581-2586.

[142] Nissen SE, Wolski K (2007) Effect of rosiglitazone on therisk of myocardial infarction and death from cardiovascularcauses. N Engl J Med  356, 2457-2471.

[143] Lincoff AM, WolskiK, Nicholls SJ,Nissen SE (2007)Piogli-tazone and risk of cardiovascular events in patients with type2 diabetes mellitus: a meta-analysis of randomized trials.

 JAMA 298, 1180-1188.[144] Rabiner EA, Tzimopoulou S, Cunningham VJ, Jeter B,

Zvartau-Hind M, Castiglia M, Mistry P, Bird NP, MatthewsJ, Whitcher B, Nichols TE, Lai R, Lotay N, Saunders A,Reiman E, Chen K, Gold M, Matthews PM (2009) Effectsof 12 months of treatment with the PPARγ  agonist rosigli-tazone on brain glucose metabolism in Alzheimer’s disease:A 18F-FDG PET study. Alzheimers Dement  5, 207.

[145] Bailey CJ, Turner RC (1996) Metformin. N Engl J Med  334,

574-579.[146] Dunn CJ, Peters DH (1995) Metformin. A review of its phar-

macological properties and therapeutic use in non-insulin-dependent diabetes mellitus.  Drugs 49, 721-749.

[147] Tian J, Shi J, Bailey K, Lendon CL, Pickering-Brown SM,Mann DMA (2004) Association between apolipoprotein Ee4 allele and arteriosclerosis, cerebral amyloid angiopathy,and cerebral white matter damage in Alzheimer’s disease.  J 

 Neurol Neurosurg Psychiatry 75, 696-699.[148] The Diabetes Prevention Program Research G (2005) Role

of Insulin Secretion and Sensitivity in the Evolution of Type2 Diabetes in the Diabetes Prevention Program: Effects of Lifestyle Intervention and Metformin.  Diabetes   54, 2404-2414.

[149] The Diabetes Prevention Program Research Group (2005)Intensive Lifestyle Intervention or Metformin on Inflamma-

tion and Coagulation in Participants With Impaired GlucoseTolerance.  Diabetes 54, 1566-1572.

[150] Orchard TJ, Temprosa M, Goldberg R, Haffner S, Ratner R,Marcovina S, Fowler S, for the Diabetes Prevention ProgramResearch Group (2005) The effect of metformin and inten-sive lifestyle intervention on the metabolic syndrome: TheDiabetes Prevention Program Randomized Trial.  Ann Intern

 Med  142, 611-619.[151] Diabetes Prevention Program Research Group (2002) Re-

duction in the incidence of type 2 diabetes with lifestyleintervention or metformin. N Engl J Med  346, 393-403.

[152] Chen Y, Zhou K, Wang R, Liu Y, Kwak YD, Ma T, Thomp-son RC, Zhao Y, Smith L, Gasparini L, Luo Z, Xu H, LiaoFF (2009) Antidiabetic drug metformin (GlucophageR) in-creases biogenesis of Alzheimer’s amyloid peptides via up-regulating BACE1 transcription. Proc Natl Acad Sci U S A

106, 3907-3912.[153] Chiasson J-L, Josse RG, Gomis R, Hanefeld M, Karasik A,

Laakso M (2002) Acarbose for prevention of type 2 diabetesmellitus: the STOP-NIDDM randomised trial. The Lancet 

359, 2072-2077.[154] Williamson JD, Miller ME, Bryan RN, Lazar RM, Coker LH,

Johnson J, Cukierman T, Horowitz KR, Murray A, LaunerLJ (2007) The Action to Control Cardiovascular Risk in Di-abetes Memory in Diabetes Study (ACCORD-MIND): ratio-nale, design, and methods.  Am J Cardiol  99, 112i-122i.

[155] Whitmer RA, Karter AJ, Yaffe K, Quesenberry CP, Jr., SelbyJV (2009) Hypoglycemic episodes and risk of dementia inolder patients withtype 2 diabetes mellitus. JAMA 301, 1565-1572.

[156] Shea S, Weinstock RS, Teresi JA, Palmas W, Starren J, Cimi-no JJ, Lai AM, Field L, Morin PC, Goland R, Izquierdo RE,Ebner S, Silver S, Petkova E, Kong J, Eimicke JP (2009) Arandomized trial comparing telemedicine case managementwith usual care in older, ethnically diverse, medically under-served patients with diabetes mellitus: 5 year results of theIDEATel study. J Am Med Inform Assoc  16, 446-456.

[157] The Action to Control Cardiovascular Risk in Diabetes StudyG (2008) Effects of intensive glucose lowering in type 2

diabetes. N Engl J Med  358, 2545-2559.[158] Kendall DM, Riddle MC, Rosenstock J, Zhuang D, Kim

DD, Fineman MS, Baron AD (2005) Effects of Exenatide(exendin-4) on glycemic control over 30 weeks in patientswith type 2 diabetes treated with metformin and a sulfony-lurea. Diabetes Care 28, 1083-1091.

[159] Leibson CL, Rocca WA, Hanson VA, Cha R, Kokmen E,O’Brien PC, Palumbo PJ (1997) Risk of dementia amongpersons with diabetes mellitus: a population-based cohortstudy. Am J Epidemiol 145, 301-308.

[160] Brayne C,GillC, Huppert FA,BarkleyC, Gehlhaar E,GirlingDM, O’Connor DW, Paykel ES (1998) Vascular risks andincident dementia: results from a cohort study of the veryold. Dement Geriatr Cogn Disord  9, 175-180.

[161] Ott A, Stolk RP, van Harskamp F, Pols HA, Hofman A,Breteler MM (1999) Diabetes mellitus and the risk of de-

mentia: The Rotterdam Study. Neurology 53, 1937-1942.[162] Arvanitakis Z, Wilson RS, Bienias JL, Evans DA, Bennett

DA (2004) Diabetes mellitus and risk of Alzheimer diseaseand decline in cognitive function.  Arch Neurol  61, 661-666.

[163] Schnaider Beeri M, Goldbourt U, Silverman JM, Noy S,Schmeidler J, Ravona-Springer R, Sverdlick A, Davidson M(2004) Diabetes mellitus in midlife and the risk of dementiathree decades later.  Neurology 63, 1902-1907.

[164] Xu WL, Qiu CX, Wahlin A, Winblad B, Fratiglioni L (2004)Diabetes mellitus and risk of dementia in the Kungsholmenproject: a 6-year follow-up study. Neurology 63, 1181-1186.

[165] Whitmer RA,Sidney S, Selby J,Johnston SC,Yaffe K (2005)Midlife cardiovascular risk factors and risk of dementia inlate life. Neurology 64, 277-281.

[166] Xu W, Qiu C, Winblad B, Fratiglioni L (2007) The effect of borderline diabetes on the risk of dementia and Alzheimer’sdisease. Diabetes 56, 211-216.

[167] Irie F, Fitzpatrick AL, Lopez OL, Kuller LH, Peila R, New-man AB, Launer LJ (2008) Enhanced risk for Alzheimer dis-ease in persons with type 2 diabetes and APOE epsilon4: theCardiovascular Health Study Cognition Study.  Arch Neurol

65, 89-93.

Page 15: DZ2 Si Conditii Relationate in Rel Cu Dementa

8/10/2019 DZ2 Si Conditii Relationate in Rel Cu Dementa

http://slidepdf.com/reader/full/dz2-si-conditii-relationate-in-rel-cu-dementa 15/15

Copyright of Journal of Alzheimer's Disease is the property of IOS Press and its content may not be copied or

emailed to multiple sites or posted to a listserv without the copyright holder's express written permission.

However, users may print, download, or email articles for individual use.