ArticlePDF AvailableLiterature Review

Breakdown of the Cerebrovasculature and Blood-Brain Barrier: A Mechanistic Link Between Diabetes Mellitus and Alzheimer’s Disease

Authors:

Abstract

Alzheimer’s disease (AD) and diabetes mellitus (DM) are among the most pervasive and devastating disorders that afflict people throughout the world. Although typically associated with older demographics, recent epidemiologic studies have reported parallel trends in decreasing age of onset and increasing incidence of these conditions. Promising research continues to implicate the cerebrovasculature and blood-brain barrier (BBB) as playing key roles in AD pathoetiology. Similarly, complications accompanying DM, such as diabetic nephropathy/retinopathy, cardiovascular disease, and stroke, have been rooted in vascular compromise. Not surprisingly, DM is now considered a major risk factor for AD. The purpose of this review is to highlight investigations into the role of the cerebrovasculature in the development and progression of AD. We give particular attention to studies on humans and a variety of animal model systems that have demonstrated a link between BBB dysfunction and pathological changes in the brain consistent with aging and AD. Together, these studies suggest that the vascular complications associated with chronic, poorly managed DM can lead to subclinical BBB breakdown that precedes and drives the pathological changes progressing to symptomatic AD, providing a common mechanistic thread connecting these two disorders. Furthermore, this emphasizes the need to focus on the vasculature as a potential therapeutic target with the intent of limiting BBB breakdown involved in disease initiation and progression. In conclusion, AD may be more than just an associated comorbidity of DM, and instead another manifestation of the underlying vascular pathology that is common to both.
Journal of Alzheimer’s Disease 54 (2016) 445–456
DOI 10.3233/JAD-160284
IOS Press
445
Review
Breakdown of the Cerebrovasculature
and Blood-Brain Barrier: A Mechanistic
Link Between Diabetes Mellitus
and Alzheimer’s Disease
Eric L. Goldwasera,b,1, Nimish K. Acharyad,1, Abhirup Sarkara,b, George Godseya,b
and Robert G. Nagelea,c,
aBiomarker Discovery Center, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic
Medicine, Stratford, NJ, USA
bGraduate School of Biomedical Sciences, Rowan University, Stratford, NJ, USA
cDepartment of Geriatrics and Gerontology, Rowan University School of Osteopathic Medicine, Stratford,
NJ, USA
dDepartment of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
Handling Associate Editor: Thomas Shea
Accepted 12 June 2016
Abstract. Alzheimer’s disease (AD) and diabetes mellitus (DM) are among the most pervasive and devastating disorders that
afflict people throughout the world. Although typically associated with older demographics, recent epidemiologic studies
have reported parallel trends in decreasing age of onset and increasing incidence of these conditions. Promising research
continues to implicate the cerebrovasculature and blood-brain barrier (BBB) as playing key roles in AD pathoetiology.
Similarly, complications accompanying DM, such as diabetic nephropathy/retinopathy, cardiovascular disease, and stroke,
have been rooted in vascular compromise. Not surprisingly, DM is now considered a major risk factor for AD. The purpose
of this review is to highlight investigations into the role of the cerebrovasculature in the development and progression of
AD. We give particular attention to studies on humans and a variety of animal model systems that have demonstrated a
link between BBB dysfunction and pathological changes in the brain consistent with aging and AD. Together, these studies
suggest that the vascular complications associated with chronic, poorly managed DM can lead to subclinical BBB breakdown
that precedes and drives the pathological changes progressing to symptomatic AD, providing a common mechanistic thread
connecting these two disorders. Furthermore, this emphasizes the need to focus on the vasculature as a potential therapeutic
target with the intent of limiting BBB breakdown involved in disease initiation and progression. In conclusion, AD may be
more than just an associated comorbidity of DM, and instead another manifestation of the underlying vascular pathology that
is common to both.
Keywords: Alzheimer’s disease, amyloid, blood-brain barrier, dementia, diabetes complications, diabetes mellitus
1These authors contributed equally to this work.
Correspondence to: Robert G. Nagele, PhD, Director of Trans-
lational Medicine, New Jersey Institute for Successful Aging,
Rowan University School of Osteopathic Medicine, Stratford, NJ
08084, USA. Tel.:+1 856 566 6083; Fax: +1 856 566 6419; E-mail:
nagelero@rowan.edu.
ISSN 1387-2877/16/$35.00 © 2016 – IOS Press and the authors. All rights reserved
446 E.L. Goldwaser et al. / Diabetes Disrupts the BBB Leading to AD
INTRODUCTION AND SCOPE
Diabetes mellitus (DM), a metabolic disorder char-
acterized by abnormally high blood glucose levels,
exists as two major types. Type 1 DM is described as
hyperglycemia and insulin deficiency resulting from
immune-mediated destruction of insulin-producing
-cells residing in the islets of Langerhans in
the pancreas. This pathoetiology is responsible for
only 5–10% of the total diabetic population (CDC,
National diabetes fact sheet, 2011). Type 2 DM
is primarily caused by insulin resistance, accounts
for about 90% of DM cases, and more tradition-
ally develops in older individuals. For these reasons,
type 2 DM will be included in the main focus of
this review. Presently, one in eleven Americans has
been diagnosed with DM, while one in three adults
is considered pre-diabetic. On the global scale, 346
million individuals have been diagnosed with DM
and, in 2010, 10.9 million Americans 65 years and
older were diabetic (CDC, National diabetes fact
sheet, 2011). This elderly age group also accounts
for the demographic at risk for Alzheimer’s dis-
ease (AD) (“2012 Alzheimer’s disease facts and
figures,” 2012). Epidemiologic studies hinting at a
close causal connection between DM and AD have
increased dramatically in number according to the
recent literature.
AD is the most prevalent neurodegenerative dis-
ease and the most common cause of dementia in the
elderly. One in nine Americans aged 65 years and
older is diagnosed with AD. Among major leading
causes of death in America, AD is the only chronic
disease with an ever increasing number of fatalities.
The 99.6% failure rate for AD drugs in clinical tri-
als has been both unfortunate and disheartening [1].
In light of this, the field as a whole is in desperate
need of a breakthrough, and serious attention to novel
potential drug targets may provide new opportunities
for successful therapeutic interventions. One promis-
ing and accessible target that has thus far received
little attention is the brain vasculature. A number
of recent studies has suggested a pivotal role for
an impaired blood vasculature in the pathogenesis
of both AD and DM. We conducted a search using
Pubmed indexed articles related to cognitive decline,
and cerebrovascular and blood-brain barrier (BBB)
compromise in diabetic patients, AD patients, and
normal aging. We also scoured the current litera-
ture concerning vascular contributions to dementia in
general, and cognitive-enhancing interventions that
are used for diabetics that may show benefits to the
comorbid AD population. Here, we present some of
the evidence supporting the possibility that an abnor-
mal cerebrovasculature is a common feature of AD
and DM, which may explain why DM is now being
recognized among the most important risk factors for
AD.
EPIDEMIOLOGICAL AND
NEUROPATHOLOGICAL EVIDENCE
SUPPORTS A CLOSE ASSOCIATION
BETWEEN DM, AGING, DEMENTIA,
AND AD
Several studies have demonstrated a greater inci-
dence of cognitive decline in individuals with DM.
Longitudinal studies carried out in Hashimaya, a
small town in Japan, have strongly established DM
as a risk factor for AD [2]. Similar research in the
United States, like the Rotterdam and Rochester stud-
ies, has also pointed to DM as a risk factor for AD
[3, 4]. Individuals diagnosed with DM at an earlier
age have a greater chance of developing AD com-
pared to those who became diabetic at age 65 or
older [5–8]. Similarly, metabolic syndrome, a con-
stellation of metabolic conditions that heighten the
risk for stroke, coronary artery diseases, and type
2 DM, also increases the prevalence of AD [5, 9].
Moreover, a five-year prospective study carried out
by Yaffe and colleagues has revealed an increased
incidence of cognitive decline in individuals with
metabolic syndrome [9].
A number of neuropathological changes are com-
mon to DM and AD. A state of chronic hyperglycemia
in DM patients can result in glycosylation of vari-
ous receptors, leading to the formation of “receptors
for advanced glycation end products” (RAGE) [10].
Several studies have reported an affinity between
amyloid-beta (A) peptides and RAGE, an interac-
tion that has been purported to trigger and propagate
chronic brain inflammation [10, 11]. Higher levels of
RAGE in neurons populating AD brains further bol-
ster a causal role for RAGE in AD pathology [11].
DM and AD are also often recognized as comorbid
diseases since pathological changes such as hyper-
insulinemia, glucose intolerance, hyperglycemia,
insulin resistance, atherosclerosis, hypertension, and
adiposity are common in afflicted patients [5,
12–14]. Aging, hypercholesterolemia, inflammation,
and obesity are additional risk factors associated with
both AD and DM [5, 12–15].
E.L. Goldwaser et al. / Diabetes Disrupts the BBB Leading to AD 447
AN ABNORMAL
CEREBROVASCULATURE IS COMMON
IN DM AND DEMENTIAS
Cerebrovascular disease is a general term most
often used to describe the consequences of an
unhealthy and abnormally functioning brain vascula-
ture. As the largest and most evolutionarily advanced
part of the human brain, the cerebral cortex requires
a profuse and steady supply of blood for normal
functioning. Blood vessels of different kinds and
sizes (structural hierarchies) are available to meet
this demand. Normally, during the transit of blood
through the brain vasculature, what leaves the blood
and enters into the brain parenchyma is stringently
regulated by the BBB, with a relatively small subset
of biomolecules being exchanged between brain cells
and blood via specialized receptors and transporters.
This helps to regulate and maintain overall brain
homeostasis as well as electrochemical balances that
are crucial for normal functioning of neuronal cir-
cuitry. Failure to properly control or regulate this
exchange, as in instances of local vascular inflam-
mation, atherosclerosis, hemorrhages, or infarctions
can lead to an indiscriminant leakage of blood compo-
nents, including a diverse array of proteins, metabolic
byproducts, and sometimes pathogens, into the brain
tissue [16].
It is not surprising that vascular abnormalities are
more readily detectable when they involve larger
blood vessels, as they can be more readily visual-
ized by currently available neuroimaging techniques,
such as computed tomography (CT) and magnetic
resonance imaging (MRI). Using MRI, increased
deep white matter lesions, cerebral atrophy, sub-
cortical brain atrophy, white matter hyperintensities,
hippocampal and amygdalar atrophy, and lacunar
infarctions have been found to be more prevalent in
type 2 DM patients compared to controls [17]. The
extent of damage and specific brain regions where
these changes occur dictates the nature of the symp-
tomatology. Acute and extensive leakage of blood
components can be detected using MRI and CT.
Cognitive decline with clear evidence of vascular
compromise has been named vascular dementia, vas-
cular cognitive impairment, or the vascular disease
(VaD) subtype of neurocognitive disorders. However,
currently employed neuroimaging techniques fail to
detect subacute or subclinical lesions/leakages with
reliable and reproducible accuracy [18–20]. We pro-
pose that vascular compromise of the smaller blood
vessels that comprise the microvasculature, such as
arterioles, is chronic and prevalent in AD patients,
while that of larger blood vessels has been linked to
VaD. The latter may present acutely or in a stepwise
degenerative manner as additional individual tribu-
taries become involvedthat service larger segments of
brain tissue. Unfortunately, because the smaller blood
vessels associated with AD are generally beyond the
limits of resolution for commonly used neuroimag-
ing devices, the causal role of a compromised or
dysfunctional cerebrovasculature in AD pathogene-
sis and its progression is more difficult to demonstrate
directly, and thus is constantly being challenged
[20].
As in other parts of the body, capillaries in the
brain exchange oxygen and nutrients for waste and
carbon dioxide. These tiny vessels have a relatively
simple architecture in that they lack the rather elabo-
rate organization of circumferential tunics composed
of smooth muscle layers and connective tissue, other-
wise typical of larger arterioles, venules, and all other
blood vessels greater in size. Instead, capillaries are
lined by a monolayer of vascular endothelial cells that
serves as a physical barrier between the blood and the
brain known as the BBB. Continuous tight junctions
that form between adjacent brain vascular endothelial
cells are the main structural correlate of this barrier. A
number of tight junction proteins, including claudin -
5, -3, and -12, occludin, zonula occludens (ZO) -1 and
-2, and Vascular Endothelial-Cadherin, are known
to be involved in their assembly and maintenance.
Pericytes, astrocytic foot processes, and endothelial
cell basement membranes also appear to support the
structure and function of the BBB, although the exact
nature of their individual contributions to the barrier
functions are unknown and currently under investi-
gation. Along with neurons, this complex is often
referred to as the neurovascular unit, which is cur-
rently depicted as a functional entity that conveys
information between the vasculature and the neurons
that it supports. Further structural details of the BBB
fall outside the scope of this review, but can be found
in a number of excellent articles [21, 22].
THE BBB MAINTAINS NEURONAL
HOMEOSTASIS
As blood vessels penetrate into deeper brain
structures, they progressively decrease in diameter
and eventually feed into a capillary plexus that is
responsible for nourishing a given volume of brain tis-
448 E.L. Goldwaser et al. / Diabetes Disrupts the BBB Leading to AD
sue. At this level of proximity to neurons, proper BBB
functionality is crucial for maintaining an optimal
electrochemical milieu and neuronal microenviron-
ment. Failure to have such an effective segregation
of the blood from the neuronal parenchyma can
have devastating results. The BBB is tasked with
carefully regulating which blood components enter
into the immunoprivileged brain tissue [23, 24]. As
in AD, there is a rapidly growing body of evi-
dence implicating a dysfunctional BBB as being
responsible, at least in part, for initiating and/or
propagating many conditions, including epilepsy,
multiple sclerosis, Parkinson’s disease, depression,
and schizophrenia [25–28]. This makes it reason-
able to implicate BBB breakdown as a common
mechanistic link in the initiation and progression of
many, if not all, common neurocognitive disorders,
with the nature of the symptoms being dictated by
the exact brain location and severity of the BBB
compromise.
INCREASED BBB PERMEABILITY IN
DIABETIC PATIENTS AND ANIMAL
MODELS
Several studies have reported increased BBB
permeability in diabetic patients. Starr and col-
leagues used gadolinium and MRI to show significant
increases in BBB permeability in type 2 DM patients
compared to age-matched controls [29]. They also
reported significantly increased BBB permeability in
deeper brain regions (basal ganglia), more so than in
the superficial areas (cortex), but other brain regions
failed to show similar changes in these diabetic sub-
jects.
In streptozotocin-induced diabetic rats, Huber and
colleagues observed an increased BBB permeability
for smaller molecules (e.g., sucrose, 342Da) com-
pared to somewhat larger molecules (e.g., inulin,
5000Da) [30]. The observed increase in BBB per-
meability was region- specific, with the midbrain
exhibiting the greatest extent of BBB compro-
mise. Furthermore, levels of BBB permeability
increased directly with time after induction of dia-
betes. They also reported that insulin treatment was
effective in curtailing BBB compromise, especially
at earlier stages of DM. In another study, BBB
functional integrity was diminished in streptozotocin-
treated diabetic rats [31]. Interestingly, this increased
BBB permeability was coincident with lowered
expression of occludin and ZO-1 proteins. Upon
receiving insulin treatment, hyperglycemia and BBB
permeability were successfully attenuated. These
streptozotocin-treated rats also showed increased
matrix metalloproteinase activity in their plasma.
Altered matrix metalloproteinase activity has been
reported in various neurodegenerative diseases such
as AD and epilepsy [32–34].
EARLIER STUDIES DEMONSTRATING
A DEFECTIVE BBB IN AD PATIENTS
A large number of studies have reported a com-
promised BBB in AD patients. Earlier human studies
have measured and used the levels of certain plasma
proteins normally absent in cerebrospinal fluid (CSF)
as markers of BBB breach. Serum albumin and
immunoglobulin (Ig) G are the two most widely
used plasma protein markers for this purpose. More
recently, Chalbot and colleagues have suggested
the use of secretory Ca2+-dependent phospholipase
A2(sPLA2) activity as a more sensitive marker of
BBB compromise than CSF albumin levels [35].
Levels of sPLA2in AD patients were found to
increase in association with BBB compromise as
compared to controls, suggesting its potential util-
ity as a biomarker. Increased levels of albumin in the
CSF of AD and multi-infarct dementia (a subtype of
VaD) patients compared to controls have also been
reported [36]. Similarly, increased CSF/serum IgG
and albumin ratios (which compares albumin levels
in the blood and CSF) have been shown in both ambu-
latory and institutionalized AD patients [37]. Also
supporting this notion is the higher mean albumin
ratios in the AD group, and overall reduced levels
of serum albumin and IgG in patients with major
depression and early- and late-onset AD [37, 38].
Furthermore, there are significantly higher levels of
serum albumin in late-onset AD or major depres-
sive patients compared to early-onset AD [38]. Along
these same lines, significantly elevated levels of CSF
albumin and albumin quotients were exhibited in the
AD cohort compared to controls [39]. In a three-year
longitudinal study performed on 65 individuals at 85
years of age, there was an increased mean CSF/serum
albumin ratio in the patients diagnosed with AD
and dementia compared to controls [40]. From the
same study, three of the women who later developed
dementia had an increased CSF/serum albumin ratio
compared to those who did not go on to develop
E.L. Goldwaser et al. / Diabetes Disrupts the BBB Leading to AD 449
dementia, again signifying this measurement’s util-
ity in even pre-symptomatic diagnostic assays
[40].
These in vivo reports have been further supported
by histopathological studies carried out on post-
mortem AD brains. As in animal studies, the presence
of blood components in the brain parenchyma that
are normally absent are commonly used as indica-
tors of BBB compromise, with IgG and albumin
leakage being the two most widely used for this
purpose. Wisniewski and Kozlowski first reported
extensive perivascular leakage of IgG and albumin
in AD brain samples compared to controls [41].
In agreement with these early reports, additional
work revealed an increased prevalence of micro- or
mini-bleeds, a type of cerebrovascular lesion, in AD
brains compared to controls [42]. Neuroradiological
modalities employed gadolinium-diethylenetriamine
pentaacetic acid (Gd-DTPA) and MRI to demon-
strate the presence of a compromised BBB even at
an early stage of AD [20]. In this study, however,
they failed to demonstrate any significant difference
in the extent of BBB compromise between AD and
age-matched controls. Given the recent understand-
ing that AD pathology can be ongoing decades before
symptoms manifest and the disease is diagnosed,
this finding is not surprising. The ability to accu-
rately establish a “pathologically controlled” non-AD
cohort, rather than a “symptomatically controlled”
non-AD cohort, will be crucial to proper interpreta-
tion of these findings. Unfortunately, at present, it is
impossible to detect accruing AD pathology early on
in the disease development in vivo, even though such
findings are readily uncovered on postmortem brain
tissue.
Our group has also investigated the integrity
of the BBB in age-matched non-demented control
and AD brains by utilizing the direct detection of
extravasation of plasma components such as IgG,
complement protein C1q, and Apeptides as tissue
indicators of BBB breach [43, 44]. AD brains demon-
strated increased incidence of focal BBB compromise
in the cerebral cortex and hippocampus compared
to age-matched controls [43, 44]. Additionally,
in agreement with the earlier findings of Wis-
niewski and Kozlowski, we also commonly observed
regions of BBB compromise in age-matched con-
trol brains [41, 44]. Furthermore, our group and
others have reported increased cerebral amyloid
angiopathy in AD, with the implicated blood ves-
sels exhibiting a greater extent of perivascular leakage
[41, 42, 45, 46].
THE CONCEPT OF A PATHOGENIC
CONNECTION BETWEEN A LEAKY
BRAIN VASCULATURE AND THE
INITIATION AND PROGRESSION OF AD
REMAINS CONTENTIOUS
Contrary to the aforementioned studies and
reports, there are also several studies that have failed
to observe any significant differences in the extent
of BBB permeability between AD and age-matched
controls [47–51]. Skepticism on the causal role of an
abnormal BBB in AD pathogenesis has been widely
cited, as some neuroimaging studies using CT and
MRI have failed to detect evidence of BBB com-
promise [20, 52–54]. More recently, using advanced
dynamic contrast-enhanced MRI, Montagne and col-
leagues were able to demonstrate the accelerated rate
of BBB compromise in the hippocampus of patients
with mild cognitive impairment, a well-recognized
early stage of AD pathology [55]. However, the
understanding that AD pathology begins to develop
decades prior to the emergence of symptoms chal-
lenges the concept of who may serve as appropriate
controls, as mentioned previously [56]. Considering
that up to 50% of individuals over the age of 85 have
symptomatic AD, it is likely that a large proportion
of those not exhibiting symptoms at this age actu-
ally have ongoing pathology, but as yet at a level
insufficient to lead to clear-cut AD-related symptoms
[20].
POPULAR RODENT MODELS OF AD ARE
INADEQUATE FOR STUDYING
VASCULAR CHANGES AND EARLY
STAGES OF AD PATHOLOGY
Over 100 rodent models are available for studies
on AD (http://www.alzforum.org/research-models).
Each of these models were developed for the purpose
of studying one or more aspects of AD-associated
neurodegenerative changes or pathological features
of the disease, such as amyloid plaques, neurofibril-
lary tangles, reactive gliosis, neuronal loss, synaptic
loss, long-term depression/long-term potentiation,
and cognitive decline. Unfortunately, these animals
do not live long enough to develop the same types
of aging-associated changes in the brain vascula-
ture that afflict humans, including BBB breakdown,
and therefore do not naturally develop AD-associated
pathologies within their relatively short lifespan.
Using radiotracers and endogenous mouse IgG
450 E.L. Goldwaser et al. / Diabetes Disrupts the BBB Leading to AD
leakage as markers of BBB compromise, vascular
integrity and BBB function were tested in six of the
most commonly used rodent models of AD [57]. Dur-
ing this study, the authors failed to observe significant
incidence of BBB breach in these mouse models com-
pared to the controls.
It is generally thought that symptoms associated
with AD emerge after many years of progressive
neurodegeneration. The best hope for being able to
effectively treat this disease almost certainly lies in
early detection and intervention. The implementation
of early treatments, in turn, will depend on early diag-
noses as well as our ability to definitively identify
causal factors that drive the early stages of disease
initiation. Based on the translational studies carried
out by our group and others, cerebrovascular break-
down appears to be one of the common early triggers
of AD that is rarely considered while designing ani-
mal models to recapitulate human pathology. Instead,
the most widely used animal models of AD involve
forced neuronal overexpression of mutant genes that
does not occur during typical disease pathogenesis in
humans. This provides a possible explanation for the
observation that most AD models fail to show BBB
leak [57]. One must also consider that the structural
hierarchy of affected vessels exhibiting BBB compro-
mise seen in the much larger human brain is virtually
absent in the mouse brain.
To test the effects of BBB and cerebrovascula-
ture compromise on the development of AD-relevant
pathology, we have studied acute [43] and chronic
(manuscript submitted) mouse models. In both acute
and chronic states of BBB compromise, we have
observed significantly elevated levels of extravasated
plasma proteins in the mouse brain parenchyma.
In both models, fluorescein isothiocyanate-labeled
A42 was injected in the general circulation and
found to enter into the brain parenchyma, bind selec-
tively to hippocampal neurons and cortical pyramidal
neurons, and accumulate in the lysosomal compart-
ment of these cells (which are the same cells that
are particularly vulnerable to early stages of AD
pathology) [58]. Additionally, the mouse model with
chronic (i.e., three-month) BBB breach maintained
by repeated injections of Pertussis toxin also demon-
strated hippocampal astrogliosis and a decline in
long-term memory. These rodent models, however,
failed to demonstrate features typical of the termi-
nal stages of AD pathology such as senile plaques,
tangles, and neuronal loss that otherwise accompany
the clinical presentation. In view of the fact that AD
pathology can be underway in humans for a decade
or more before symptoms become apparent and the
disease is considered to be at an early stage, these
findings suggest that mouse models may only be suf-
ficient to represent the very earliest stages of disease
progression, despite clinical correlates in cognitive
tests performed. It is noteworthy to point out that
the pathological changes resulting from acute and
chronic BBB compromise in the rodent target the
same brain regions and closely resemble the changes
seen in human brains at early disease stages.
A PORCINE MODEL IS BETTER SUITED
TO MIMIC HUMAN AD, AND
ESTABLISHES A LINK BETWEEN DM,
BBB COMPROMISE, AND THE ONSET OF
AD-RELATED PATHOLOGY
In a recent study, we investigated the question of
whether or not chronic BBB compromise could lead
to pathological changes consistent with early-stage
AD using a pig model of DM and hypercholes-
terolemia (HC) [59]. This animal model was orig-
inally developed to investigate the pathogenesis of
atherosclerosis in the great vessels, but was subse-
quently found to also increase BBB permeability, thus
enabling relatively long-term studies of the effects of
chronic BBB breakdown on the brain [59]. HC is
often comorbid in humans with DM and also serves
as its own independent risk factor for developing
AD and VaD [5, 10, 13, 14, 60–63]. An associa-
tion between HC and cognitive decline has also been
strongly supported by epidemiological, neuropatho-
logical, and animal studies [63, 64]. It was even
possible to increase the number and size of amy-
loid plaques by feeding a cholesterol-rich diet to AD
transgenic mice [65].
Histopathological analyses of DMHC porcine
brains revealed an increased incidence of BBB breach
as visualized by a significantly higher level of free
IgG present in the brain parenchyma compared to
controls [59]. The extravasation of IgG occurred
primarily in arterioles, with little evidence of signif-
icant leaks from venules or capillaries. Interestingly,
extravasated IgG was also found to selectively tar-
get the surfaces of pyramidal neurons resident in
the cerebral cortex. Along with IgG leakage signif-
icant for AD-like pathology, the DMHC pigs also
exhibited a greater density of A42-positive neurons
compared to controls [59]. Most importantly, pyrami-
dal neurons accumulating A42 also demonstrated
IgG co-labeling, perhaps alluding to a common
E.L. Goldwaser et al. / Diabetes Disrupts the BBB Leading to AD 451
mechanistic underpinning shared by these two
events. An “antibody-mediated internalization” phe-
nomenon of A42 is the current focus of a novel
mechanism proposed and investigated for the patho-
genesis of AD [44]. In addition, the vasculature in the
DMHC pigs revealed increased instances of cerebral
amyloid angiopathy and “corkscrew dendrites” in
association with pyramidal cells, two key histopatho-
logical hallmark features of AD brains [44, 59].
CURTAILING VASCULAR
INFLAMMATION THAT FAVORS BBB
COMPROMISE SHOULD BE A MAJOR
THERAPEUTIC TARGET
An important outcome of the above-mentioned
DMHC porcine study was in revealing the benefi-
cial effects of darapladib, a potent anti-inflammatory
inhibitor of lipoprotein-associated phospholipase A2
developed by GlaxoSmithKline, on BBB func-
tional integrity [66]. Treatment of DMHC pigs
with darapladib resulted in a reduction of AD-
associated pathological hallmarks described above,
and included a diminution of IgG leakage from blood
vessels and a decreased number of amyloid-burdened
pyramidal neurons compared to untreated DMHC
controls [66]. Essentially, darapladib successfully
rescued the DMHC animals from the AD pheno-
type that was otherwise observed in untreated DMHC
controls, and restored the pathological indices that
measured back to nearly non-DMHC control lev-
els. By inhibiting the BBB breakdown, this drug
contributed to the maintenance of the proper corti-
cal microenvironment in DMHC pigs. This suggests
that darapladib and other drugs that promote BBB
integrity may have similar utility in both diabetic
and non-diabetic humans, and may also reduce the
incidence and magnitude of the neurodegenerative
changes triggered by chronic DMHC.
These outcomes are also in agreement with pre-
vious studies that implicate abnormalities in the
cerebrovasculature in the pathogenesis of AD and
VaD. Furthermore, we have recently found that
the efficacy of darapladib in inhibiting vascular
anomalies rooted in inflammation may also extend
its beneficial effects to another common afflic-
tion of the diabetic population—diabetic retinopathy
(manuscript submitted). Here, key pathological fea-
tures of diabetic retinopathy, such as edematous
plexiform layers and shrinkage of various cellular
layers, were evident in the same DMHC porcine
model. M¨
uller cell activation also alluded toward the
pathological state and incurred cellular damage. We
have shown increased blood-retinal barrier perme-
ability, as evidenced by selective binding of IgG to
ganglion cells of the retina, which closely resembled
the situation where pyramidal cells in the cerebral
cortex displayed IgG-positivity within or near regions
of BBB breach. As in the cerebral cortex, pathologi-
cal changes observed in the retinal layers of DMHC
pigs were ameliorated by darapladib to near control
levels in the cohort of DMHC pigs. This finding lends
further credence to the concept that chronic inflam-
matory changes in the walls of blood vessels within
the brain can trigger BBB breakdown, initiating a
cascade of pathological events that can culminate in
AD and perhaps other neurodegenerative diseases. It
also suggests that darapladib and other agents that
can block vascular inflammation should be inves-
tigated and evaluated for their capacity to assuage
the rampant neurodegeneration accompanying these
diseases. In fact, rilapladib, a structurally related
compound to that of darapladib with similar pharma-
cokinetic and pharmacodynamic profiles, efficacies,
and tolerances across all age brackets, was studied
in a Phase II clinical trial for early AD. Here, the
researchers, using cognitive tests and biochemical
assays, detected significant benefits for rilapladib-
treated patients to that of placebo [67–69].
ANTI-DIABETIC DRUGS AND OTHER
THERAPEUTIC EFFORTS TARGETING
THE VASCULATURE
It is natural to progress from the mechanistic
underpinnings shared by AD and DM to potential
for overlap in therapeutic interventions (medical or
behavioral). Statins are an obvious choice consider-
ing its many years in use and post-marketing statistics
gathered. A research group in Taiwan followed
elderly DM patients for nearly a decade after starting
statin therapies and found a decrease in the incidence
of AD in those taking statin therapy for DM than in
those not on a statin therapy [70]. Meanwhile, another
study found that although statin use decreased lipid
levels in patients already diagnosed with AD, their
cognitive scores did not improve [71]. The finding
that these therapies are largely ineffective in patients
that have been already diagnosed with AD is not
surprising since we now know that a considerable
amount of AD-related pathology and neuronal death
has already occurred in these individuals prior to
452 E.L. Goldwaser et al. / Diabetes Disrupts the BBB Leading to AD
Fig. 1. Diabetes induces blood-brain barrier (BBB) breakdown, triggering Alzheimer’s disease (AD) and perhaps other neurodegenerative
disease. Many complications of diabetes mellitus (DM) are related to a globally impaired vascular integrity, spanning vessel hierarchy and
structure from large to small, and resulting in conditions such as peripheral neuropathy, retinopathy, nephropathy, and coronary artery disease.
These vascular abnormalities extend throughout the body, and include the supply to the cortical and deep brain structures. Here, macrovascular
lesions of the arterial blood supply result in stroke and vascular dementia (VaD). On the other hand, chronic leakage of plasma components
from the microvasculature occurs at the level of arterioles and capillaries as a result of BBB breakdown and contributes to the pathogenesis
and development of AD. Following BBB breakdown, various plasma components, including Ig species (IgG) and amyloid peptides (e.g.,
A42), leak out of the blood vessels, enter into the brain tissue, and bind to pyramidal neurons in regions of vascular compromise. Chronic
BBB leakage drives the binding of Ig and Ato neuronal surfaces, followed by their internalization and accumulation within these cells,
leading to neuronal damage, synaptic loss, cell death, and the appearance of plaques and tangles as pathological hallmarks commonly
observed at autopsy. DM, diabetes mellitus; AD, Alzheimer’s disease; BBB, blood-brain barrier; Ig, immunoglobulin; A42 , amyloid-142;
VaD, vascular dementia.
E.L. Goldwaser et al. / Diabetes Disrupts the BBB Leading to AD 453
symptom manifestation and treatment initiation. This
is the most likely explanation for the unfortunately
high failure rate of AD clinical trials. Naturally, as
the disease progresses, a greater volume of the brain
parenchyma is afflicted, which underscores why such
failures have been met with clinical trials for patients
already diagnosed with AD. Recent literature sug-
gests that statin use may be effective in delaying the
onset of AD and slowing the progression from mild
cognitive impairment to AD, but only if treatment
begins prior to overt AD. Pro-inflammatory media-
tors are implicated in many studies linking aberrant
and exaggerated inflammatory processes to AD [72].
Along those lines, pro-resolving mediators, includ-
ing lipoxin A4,may play a key anti-inflammatory
role that can reduce AD symptoms and even promote
amyloid phagocytosis by microglial cells [73, 74].
On the other hand, insulin administered intranasally
showed efficacy in mild cognitive impairment and
early AD patients leading to improved cognition
[75, 76]. Aerobic exercise was demonstrated in an
established mouse model to delay the onset and pro-
gression of AD [77, 78]. Furthermore, behavioral,
diet, and lifestyle modifications are strongly advo-
cated as the best non-pharmacological methods to
prevent the onset and progression AD [79].
CONCLUSION AND PERSPECTIVES
AD and DM are two disorders of growing con-
cern in the US for a multitude of reasons. Given
the alarming rate at which AD is being diagnosed in
the diabetic population, along with a startlingly high
failure rate of AD drugs, it may be time to recon-
sider prevailing models of the pathogenesis of AD.
To more fully understand the mechanisms underly-
ing AD pathology, it is time to devote considerable
attention to the role of the blood vasculature, and
promote research designed to curtail the kinds of vas-
cular pathology that contribute to the disease with the
goal of restoring BBB functionality. We propose that
an early stage in the pathogenesis of AD, preceding
the formation of amyloid plaques, is a compromised
brain microvasculature that includes breakdown of
the BBB. In DM patients, it is suggested that this BBB
compromise arises from the chronic vascular inflam-
mation that also predisposes individuals to dementia
(Fig. 1). This may evolve into a decades-long pre-
symptomatic phase of AD, during which patients are
continuously accruing brain pathology. Here, brain
neurons are metabolically taxed and homeostatically
perturbed by the chronic influx of plasma components
into the brain, but individuals remain asymptomatic.
This early stage of disease presents a prime window
of opportunity to achieve an effective and lasting ther-
apy before extensive brain devastation has occurred.
Toward this end, early treatment with drugs that block
vascular inflammation would be of great benefit, as
they would act to maintain or repair BBB integrity.
Revitalization of a functional BBB would be expected
to help restore brain homeostasis, promote neuronal
health and function, and shed light on a pathoetiol-
ogy that may prove to span multiple neurological and
neurodegenerative diseases.
DISCLOSURE STATEMENT
Authors’ disclosures available online (http://j-alz.
com/manuscript-disclosures/16-0284r1).
REFERENCES
[1] Cummings JLMT, Zhong K (2014) Alzheimer’s disease
drug-development pipeline: Few candidates, frequent fail-
ures. Alzheimers Res Ther 6, 37.
[2] Ohara T, Doi Y, Ninomiya T, Hirakawa Y, Hata J, Iwaki T,
Kanba S, Kiyohara Y (2011) Glucose tolerance status and
risk of dementia in the community: The Hisayama study.
Neurology 77, 1126-1134.
[3] Ott A, Stolk RP, Hofman A, van Harskamp F, Grobbee
DE, Breteler MM (1996) Association of diabetes melli-
tus and dementia: The rotterdam study. Diabetologia 39,
1392-1397.
[4] Leibson CL, Rocca WA, Hanson VA, Cha R, Kokmen E,
O’Brien PC, Palumbo PJ (1997) Risk of dementia among
persons with diabetes mellitus: A population-based cohort
study. Am J Epidemiol 145, 301-308.
[5] Carlsson CM (2010) Type 2 diabetes mellitus, dyslipi-
demia, and Alzheimer’s disease. J Alzheimers Dis 20, 711-
722.
[6] Xu W, Qiu C, Gatz M, Pedersen NL, Johansson B,
Fratiglioni L (2009) Mid- and late-life diabetes in rela-
tion to the risk of dementia: A population-based twin study.
Diabetes 58, 71-77.
[7] Xu W, Qiu C, Winblad B, Fratiglioni L (2007) The effect of
borderline diabetes on the risk of dementia and Alzheimer’s
disease. Diabetes 56, 211-216.
[8] Xu WL, von Strauss E, Qiu CX, Winblad B, Fratiglioni
L (2009) Uncontrolled diabetes increases the risk of
Alzheimer’s disease: A population-based cohort study. Dia-
betologia 52, 1031-1039.
[9] Yaffe K, Kanaya A, Lindquist K, Simonsick EM, Har-
ris T, Shorr RI, Tylavsky FA, Newman AB (2004) The
metabolic syndrome, inflammation, and risk of cognitive
decline. JAMA 292, 2237-2242.
[10] Taguchi A (2009) Vascular factors in diabetes and
Alzheimer’s disease. J Alzheimers Dis 16, 859-864.
[11] Yan SD, Chen X, Fu J, Chen M, Zhu H, Roher A, Slattery
T, Zhao L, Nagashima M, Morser J, Migheli A, Nawroth
P, Stern D, Schmidt AM (1996) RAGE and amyloid-beta
454 E.L. Goldwaser et al. / Diabetes Disrupts the BBB Leading to AD
peptide neurotoxicity in Alzheimer’s disease. Nature 382,
685-691.
[12] Craft S (2009) The role of metabolic disorders in Alzheimer
disease and vascular dementia: Two roads converged. Arch
Neurol 66, 300-305.
[13] Kalaria RN (2010) Vascular basis for brain degeneration:
Faltering controls and risk factors for dementia. Nutr Rev
68(Suppl 2), S74-S87.
[14] Sims-Robinson C, Kim B, Rosko A, Feldman EL (2010)
How does diabetes accelerate Alzheimer disease pathology?
Nat Rev Neurol 6, 551-559.
[15] Bowman GL, Kaye JA, Quinn JF (2012) Dyslipidemia and
blood-brain barrier integrity in Alzheimer’s disease. Curr
Gerontol Geriatr Res 2012, 184042.
[16] Huber JD (2008) Diabetes, cognitive function, and the
blood-brain barrier. Curr Pharm Des 14, 1594-1600.
[17] den Heijer T, Vermeer SE, van Dijk EJ, Prins ND, Koudstaal
PJ, Hofman A, Breteler MM (2003) Type 2 diabetes and
atrophy of medial temporal lobe structures on brain MRI.
Diabetologia 46, 1604-1610.
[18] Snowdon DAGL, Mortimer JA, Riley KP, Greiner PA,
Markesbery WR (1997) Brain infarction and the clinical
expression of Alzheimer disease. The Nun Study. JAMA
277, 813-817.
[19] Ikram MA VdIA, Niessen WJ, Krestin GP, Koudstaal PJ,
Hofman A, Breteler MMB, Vernooij MW (2011) The rot-
terdam scan study: Design and update up to 2012. Eur J
Epidemiol 26, 811-824.
[20] Starr JM, Farrall AJ, Armitage P, McGurn B, Wardlaw
J (2009) Blood-brain barrier permeability in Alzheimer’s
disease: A case-control MRI study. Psychiatry Res 171,
232-241.
[21] Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley
DJ (2010) Structure and function of the blood-brain barrier.
Neurobiol Dis 37, 13-25.
[22] Zlokovic BV (2008) The blood-brain barrier in health
and chronic neurodegenerative disorders. Neuron 57, 178-
201.
[23] Abbott NJPA, Dolman DEM, Yusof SR, Begley DJ (2010)
Structure and function of the blood-brain barrier. Neurobiol
Dis 37, 13-25.
[24] Zlokovic B (2008) The blood-brain barrier in health and
chronic neurodegenerative disorders. Neuron 57, 178-201.
[25] Suit YT BK, Erickson MA, Zhang J, Banks WA (2014)
Alpha synuclein is transported into and out of the brain by
the blood-brain barrier. Peptides 62, 197-202.
[26] Patel JPFB (2015) Disruption in the blood-brain barrier:
The missing link between brain and body inflammation in
bipolar disorder? Neural Plast 2015, 708306.
[27] Michalak ZLA, DiMiceli M, Rousset M, Crespel A, Coubes
P, Henshall DC, Lerner-Natoli M, Rigau V (2012) IgG
leakage may contribute to neuronal dysfunction in drug-
refractory epilepsies with blood-brain barrier disruption. J
Neuropathol Exp Neurol 71, 826-838.
[28] Hammer CSB, Schneider A, Papiol S, Tantra M, Begemann
M, Siren A-L, Pardo AL, Sperling S, Mohd Jofrry S, Gur-
vich A, Jensen N, Ostmeier K, Lunder F, Probst C, Martens
H, Gillis M, Saher G, Assogna F, Spalletta G, Stocker W,
Schulz TF, Nave K-A, Ehrenreich H (2014) Neuropsychi-
atric disease relevance of circulating anti-NMDA receptor
autoantibodies depends on blood-brain barrier integrity.Mol
Psychiatry 19, 1143-1149.
[29] Starr JM, Wardlaw J, Ferguson K, MacLullich A, Deary IJ,
Marshall I (2003) Increased blood-brain barrier permeabil-
ity in type II diabetes demonstrated by gadolinium magnetic
resonance imaging. J Neurol Neurosurg Psychiatry 74,
70-76.
[30] Huber JD, VanGilder RL, Houser KA (2006)
Streptozotocin-induced diabetes progressively increases
blood-brain barrier permeability in specific brain regions
in rats. Am J Physiol Heart Circ Physiol 291, H2660-
H2668.
[31] Hawkins BT,Lundeen TF, Norwood KM, Brooks HL, Egle-
ton RD (2007) Increased blood-brain barrier permeability
and altered tight junctions in experimental diabetes in the
rat: Contribution of hyperglycaemia and matrix metallopro-
teinases. Diabetologia 50, 202-211.
[32] Mizoguchi H, Yamada K, Nabeshima T (2011) Matrix
metalloproteinases contribute to neuronal dysfunction in
animal models of drug dependence, Alzheimer’s disease,
and epilepsy. Biochem Res Int 2011, 681385.
[33] Mizoguchi H, Yamada K (2013) Roles of matrix metallo-
proteinases and their targets in epileptogenesis and seizures.
Clin Psychopharmacol Neurosci 11, 45-52.
[34] Wang XX, Tan MS, Yu JT, Tan L (2014) Matrix metallo-
proteinases and their multiple roles in Alzheimer’s disease.
Biomed Res Int 2014, 908636.
[35] Chalbot S, Zetterberg H, Blennow K, Fladby T, Andreasen
N, Grundke-Iqbal I, Iqbal K (2011) Blood-cerebrospinal
fluid barrier permeability in Alzheimer’s disease. J
Alzheimers Dis 25, 505-515.
[36] Alafuzoff I, Adolfsson R, Bucht G, Winblad B (1983) Albu-
min and immunoglobulin in plasma and cerebrospinal fluid,
and blood-cerebrospinal fluid barrier function in patients
with dementia of Alzheimer type and multi-infarct demen-
tia. J Neurol Sci 60, 465-472.
[37] Elovaara I, Icen A, Palo J, Erkinjuntti T (1985) CSF in
Alzheimer’s disease. Studies on blood-brain barrier func-
tion and intrathecal protein synthesis. J Neurol Sci 70,
73-80.
[38] Hampel H, Kotter HU, Moller HJ (1997) Blood-
cerebrospinal fluid barrier dysfunction for high molecular
weight proteins in Alzheimer disease and major depression:
Indication for disease subsets. Alzheimer Dis Assoc Disord
11, 78-87.
[39] Wada H (1998) Blood-brain barrier permeability of the
demented elderly as studied by cerebrospinal fluid-serum
albumin ratio. Intern Med 37, 509-513.
[40] Skoog I, WallinA, Fredman P, Hesse C, AevarssonO, Karls-
son I, Gottfries CG, Blennow K (1998) A population study
on blood-brain barrier function in 85-year-olds: Relation to
Alzheimer’s disease and vascular dementia. Neurology 50,
966-971.
[41] Wisniewski HM, Kozlowski PB (1982) Evidence for blood-
brain barrier changes in senile dementia of the Alzheimer
type (SDAT). Ann N Y Acad Sci 396, 119-129.
[42] De Reuck JL (2012) The significance of small cerebral
bleeds in neurodegenerative dementia syndromes. Aging
Dis 3, 307-312.
[43] Clifford PM, Zarrabi S, Siu G, Kinsler KJ, Kosciuk MC,
Venkataraman V, D’Andrea MR, Dinsmore S, Nagele RG
(2007) Abeta peptides can enter the brain through a defective
blood-brain barrier and bind selectively to neurons. Brain
Res 1142, 223-236.
[44] Nagele RG, Clifford PM, Siu G, Levin EC, Acharya NK,
Han M, Kosciuk MC, Venkataraman V, Zavareh S, Zarrabi
S, Kinsler K, Thaker NG, Nagele EP, Dash J, Wang HY,
Levitas A (2011) Brain-reactive autoantibodies prevalent
in human sera increase intraneuronal amyloid-beta(1-42)
deposition. J Alzheimers Dis 25, 605-622.
E.L. Goldwaser et al. / Diabetes Disrupts the BBB Leading to AD 455
[45] Tian J, Shi J, Mann DM (2004) Cerebral amyloid angiopathy
and dementia. Panminerva Med 46, 253-264.
[46] Clifford PM, Siu G, Kosciuk M, LevinEC, Venkataraman V,
D’Andrea MR, Nagele RG (2008) Alpha7 nicotinic acetyl-
choline receptor expression by vascular smooth muscle cells
facilitates the deposition of Abeta peptides and promotes
cerebrovascular amyloid angiopathy. Brain Res 1234, 158-
171.
[47] Bowman GL, Kaye JA, Moore M, Waichunas D, Carlson
NE, Quinn JF (2007) Blood-brain barrier impairment in
Alzheimer disease: Stability and functional significance.
Neurology 68, 1809-1814.
[48] Alafuzoff I, Adolfsson R, Grundke-Iqbal I, Winblad B
(1987) Blood-brain barrier in Alzheimer dementia and in
non-demented elderly.An immunocytochemical study. Acta
Neuropathol 73, 160-166.
[49] Rozemuller JM, Eikelenboom P, Kamphorst W, Stam FC
(1988) Lack of evidence for dysfunction of the blood-brain
barrier in Alzheimer’s disease: An immunohistochemical
study. Neurobiol Aging 9, 383-391.
[50] Munoz DG, Erkinjuntti T, Gaytan-Garcia S, Hachinski V
(1997) Serum protein leakage in Alzheimer’s disease revis-
ited. Ann N Y Acad Sci 826, 173-189.
[51] Tomimoto H, Akiguchi I, Suenaga T, Nishimura M, Wakita
H, Nakamura S, Kimura J (1996) Alterations of the blood-
brain barrier and glial cells in white-matter lesions in
cerebrovascular and Alzheimer’s disease patients. Stroke 27,
2069-2074.
[52] Erickson MA, Banks WA (2013) Blood-brain barrier dys-
function as a cause and consequence of Alzheimer’s disease.
J Cereb Blood Flow Metab 33, 1500-1513.
[53] Rosenberg GA (2012) Neurological diseases in relation to
the blood-brain barrier.J Cereb Blood Flow Metab 32, 1139-
1151.
[54] Rosenberg GA (2014) Blood-brain barrier permeability in
aging and Alzheimer’s disease. J Prev Alzheimers Dis 1,
138-139.
[55] Montagne A, Barnes SR, Sweeney MD, Halliday MR,
Sagare AP, Zhao Z, Toga AW, Jacobs RE, Liu CY, Amezcua
L, Harrington MG, Chui HC, Law M, Zlokovic BV (2015)
Blood-brain barrier breakdown in the aging human hip-
pocampus. Neuron 85, 296-302.
[56] Blennow K, de Leon MJ, Zetterberg H (2006) Alzheimer’s
disease. Lancet 368, 387-403.
[57] Bien-Ly N, Boswell CA, Jeet S, Beach TG, Hoyte K, Luk W,
Shihadeh V, Ulufatu S, Foreman O, Lu Y, DeVoss J, van der
Brug M, Watts RJ (2015) Lack of widespread BBB disrup-
tion in Alzheimer’s disease models: Focus on therapeutic
antibodies. Neuron 88, 289-297.
[58] Fitch MT, Silver J (1997) Glial cell extracellular matrix:
Boundaries for axon growth in development and regenera-
tion. Cell Tissue Res 290, 379-384.
[59] Acharya NK, Levin EC, Clifford PM, Han M, Tourtel-
lotte R, Chamberlain D, Pollaro M, Coretti NJ, Kosciuk
MC, Nagele EP, Demarshall C, Freeman T, Shi Y, Guan C,
Macphee CH, Wilensky RL, Nagele RG (2013) Diabetes
and hypercholesterolemia increase blood-brain barrier per-
meability and brain amyloid deposition: Beneficial effects
of the LpPLA2 inhibitor darapladib. J Alzheimers Dis 35,
179-198.
[60] Ledesma MD, Dotti CG (2012) Peripheral cholesterol,
metabolic disorders and Alzheimer’s disease. Front Biosci
(Elite Ed) 4, 181-194.
[61] Luchsinger JA, Gustafson DR (2009) Adiposity, type 2
diabetes, and Alzheimer’s disease. J Alzheimers Dis 16,
693-704.
[62] Harris MI (1991) Hypercholesterolemia in diabetes and glu-
cose intolerance in the U.S. population. Diabetes Care 14,
366-374.
[63] Shepardson NE, Shankar GM, Selkoe DJ (2011) Choles-
terol level and statin use in Alzheimer disease: I. Review
of epidemiological and preclinical studies. Arch Neurol 68,
1239-1244.
[64] Jiang X, Guo M, Su J, Lu B, Ma D, Zhang R, Yang L, Wang
Q, Ma Y, Fan Y (2012) Simvastatin blocks blood-brain bar-
rier disruptions induced by elevated cholesterol both in vivo
and in vitro.Int J Alzheimers Dis 2012, 109324.
[65] Refolo LM, Malester B, LaFrancois J, Bryant-Thomas T,
Wang R, Tint GS, Sambamurti K, Duff K, Pappolla MA
(2000) Hypercholesterolemia accelerates the Alzheimer’s
amyloid pathology in a transgenic mouse model. Neurobiol
Dis 7, 321-331.
[66] Acharya NKLE, Clifford PM, Han M, Tourtellotte R, Cham-
berlain D, Pollaro M, Coretti N, Kosciuk MC, Nagele EP,
DeMarshall C, Freeman T, Shi Y, Guan C, Macphee CH,
Wilensky RL, Nagele RG (2013) Diabetes and hypercholes-
terolemia increase blood-brain barrier permeability and
brain amyloid deposition: Beneficial effects of the LpPLA2
inhibitor darapladib. J Alzheimers Dis 35, 179-198.
[67] Shaddinger BC, Xu Y, Roger JH, Macphee CH, Handel
M, Baidoo CA, Magee M, Lepore JJ, Sprecher DL (2014)
Platelet aggregation unchanged by lipoprotein-associated
phospholipase A(2) inhibition: Results from an in vitro
study and two randomized phase I trials. PLoS One 9,
e83094.
[68] Tawakol A, Singh P, Rudd JH, Soffer J, Cai G, Vucic
E, Brannan SP, Tarka EA, Shaddinger BC, Sarov-Blat
L, Matthews P, Subramanian S, Farkouh M, Fayad ZA
(2014) Effect of treatment for 12 weeks with rilapladib, a
lipoprotein-associated phospholipase A2 inhibitor, on arte-
rial inflammation as assessed with 18F-fluorodeoxyglucose-
positron emission tomography imaging. J Am Coll Cardiol
63, 86-88.
[69] Maher-Edwards G, De’Ath J, Barnett C, Lavrov A, Lock-
hart A (2015) A 24-week study to evaluate the effect of
rilapladib on cognition and cerebrospinal fluid biomark-
ers of Alzheimer’s disease. Alzheimers Dement (N Y) 1,
131-140.
[70] Chen JM, Chang CW, Chang TH, Hsu CC, Horng JT,
Sheu WH (2014) Effects of statins on incident dementia in
patients with type 2 DM: A population-based retrospective
cohort study in Taiwan. PLoS One 9, e88434.
[71] Sano M, Bell KL, Galasko D, Galvin JE, Thomas RG, van
Dyck CH, Aisen PS (2011) A randomized, double-blind,
placebo-controlled trial of simvastatin to treat Alzheimer
disease. Neurology 77, 556-563.
[72] Zhu M, Wang X, Hjorth E, Colas RA, Schroeder L,
Granholm AC, Serhan CN, Schultzberg M (2016) Pro-
resolving lipid mediators improve neuronal survival and
increase Abeta42 phagocytosis. Mol Neurobiol 53, 2733-
2749.
[73] Dunn HC, Ager RR, Baglietto-Vargas D, Cheng D,
Kitazawa M, Cribbs DH, Medeiros R (2015) Restoration
of lipoxin A4 signaling reduces Alzheimer’s disease-like
pathology in the 3xTg-AD mouse model. J Alzheimers Dis
43, 893-903.
[74] Medeiros R, Kitazawa M, Passos GF, Baglietto-Vargas D,
Cheng D, Cribbs DH, LaFerla FM (2013) Aspirin-triggered
lipoxin A4 stimulates alternative activationof microglia and
reduces Alzheimer disease-like pathology in mice. Am J
Pathol 182, 1780-1789.
456 E.L. Goldwaser et al. / Diabetes Disrupts the BBB Leading to AD
[75] Claxton A, Baker LD, Hanson A, Trittschuh EH, Choler-
ton B, Morgan A, Callaghan M, Arbuckle M, Behl C, Craft
S (2015) Long acting intranasal insulin detemir improves
cognition for adults with mild cognitive impairment or
early-stage Alzheimer’s disease dementia. J Alzheimers Dis
45, 1269-1270.
[76] Craft S, Baker LD, Montine TJ, Minoshima S, Watson GS,
Claxton A, Arbuckle M, Callaghan M, Tsai E, Plymate SR,
Green PS, Leverenz J, Cross D, Gerton B (2012) Intranasal
insulin therapy for Alzheimer disease and amnestic mild
cognitive impairment: A pilot clinical trial. Arch Neurol 69,
29-38.
[77] Huttenrauch M, Brauss A, KurdakovaA, Borgers H, Klinker
F, Liebetanz D, Salinas-Riester G, Wiltfang J,Klafki HW,
Wirths O (2016) Physical activity delays hippocampal
neurodegeneration and rescues memory deficits in an
Alzheimer disease mouse model. Transl Psychiatry 6, e800.
[78] Isla AG, Vazquez-Cuevas FG, Pena-Ortega F (2016) Exer-
cise prevents amyloid-beta-induced hippocampal network
disruption by inhibiting GSK3beta activation. J Alzheimers
Dis 52, 333-343.
[79] Kobe T, Witte AV, Schnelle A, Lesemann A, Fabian S, Tesky
VA, Pantel J, Floel A (2016) Combined omega-3 fatty acids,
aerobic exercise and cognitive stimulation prevents decline
in gray matter volume of the frontal, parietal and cingulate
cortex in patients with mild cognitive impairment. Neuroim-
age 131, 226-238.
... It is not surprising, therefore, that multiple epidemiological studies have revealed a marked overlap among risk factors for small-vessel cerebrovascular disease and late-onset Alzheimer's disease. [2] It has been reported continually that endothelial modulation and/or repair is practicable by pharmacological targeting [1,2,[7][8][9][10][11][12][13] via cell-surface scavenger receptors (mainly class B type I, i.e., SR-BI). [13][14][15] Since SR-BI has already been identified as a major receptor for high-density lipoprotein or HDL (with their major apolipoprotein (apo)A-I), as well as for the recently reviewed [1,2] "lipid-coated microbubble/nanoparticle-derived" (LCM/ND) nanoemulsion (see below), this multitasking lipid nanoemulsion can arguably serve as a targeted, apoA-I-based, (SR-BI mediated) therapeutic agent for common (late-onset) dementias. ...
... [2] It has been reported continually that endothelial modulation and/or repair is practicable by pharmacological targeting [1,2,[7][8][9][10][11][12][13] via cell-surface scavenger receptors (mainly class B type I, i.e., SR-BI). [13][14][15] Since SR-BI has already been identified as a major receptor for high-density lipoprotein or HDL (with their major apolipoprotein (apo)A-I), as well as for the recently reviewed [1,2] "lipid-coated microbubble/nanoparticle-derived" (LCM/ND) nanoemulsion (see below), this multitasking lipid nanoemulsion can arguably serve as a targeted, apoA-I-based, (SR-BI mediated) therapeutic agent for common (late-onset) dementias. [16][17][18] Documented similarities in the lipid composition of HDL versus these nanoemulsion (drug-carrier) particles may enable such LCM/ND nanoemulsions to mimic, in part, HDL-particle heterogeneity. ...
... Accordingly, endothelial modulation and repair become feasible by pharmacological targeting [8][9][10][11][12][86][87][88][89][90] via SR-BI receptors (cf. [13,90] ). While SR-BI is a major receptor highly expressed in the liver, SR-BI is also amply expressed by endothelial cells -including those comprised within the BBB -and this receptor is involved in the endothelial protective effects of HDL. ...
... It is not surprising, therefore, that multiple epidemiological studies have revealed a marked overlap among risk factors for small-vessel cerebrovascular disease and late-onset Alzheimer's disease [2]. It has been reported continually that endothelial modulation and/or repair is practicable by pharmacological targeting [1,2,[7][8][9][10][11][12][13] via cell-surface scavenger receptors (mainly class B type I, i.e., SR-BI) [13][14][15]. Since SR-BI has already been identified as a major receptor for highdensity lipoprotein or HDL (with their major apolipopro-tein (apo)A-I), as well as for the recently reviewed [1,2] "lipid-coated microbubble/nanoparticle-derived" (LCM/ ND) nanoemulsion (see below), this multitasking lipid nanoemulsion can arguably serve as a targeted, apoA-Ibased, (SR-BI mediated) therapeutic agent for common (late-onset) dementias [16][17][18]. ...
... It is not surprising, therefore, that multiple epidemiological studies have revealed a marked overlap among risk factors for small-vessel cerebrovascular disease and late-onset Alzheimer's disease [2]. It has been reported continually that endothelial modulation and/or repair is practicable by pharmacological targeting [1,2,[7][8][9][10][11][12][13] via cell-surface scavenger receptors (mainly class B type I, i.e., SR-BI) [13][14][15]. Since SR-BI has already been identified as a major receptor for highdensity lipoprotein or HDL (with their major apolipopro-tein (apo)A-I), as well as for the recently reviewed [1,2] "lipid-coated microbubble/nanoparticle-derived" (LCM/ ND) nanoemulsion (see below), this multitasking lipid nanoemulsion can arguably serve as a targeted, apoA-Ibased, (SR-BI mediated) therapeutic agent for common (late-onset) dementias [16][17][18]. Documented similarities in the lipid composition of HDL versus these nanoemulsions (drug-carrier) particles may enable such LCM/ND nanoemulsions to mimic, in part, HDL-particle heterogeneity [1,2]. ...
... Accordingly, endothelial modulation and repair become feasible by pharmacological targeting [8][9][10][11][12][86][87][88][89][90] via SR-BI receptors (cf. [13,90]). The proposed multitasking combination therapeutic, described herein, appears likely to display greater efficacy at different stages of Alzheimer's disease (cf. ...
Article
Microvascular endothelial dysfunction precedes, often by decades, the cognitive decline associated with Alzheimer’s disease. Accordingly, cerebrovascular risk factors (e.g., atherosclerosis, diabetes, aging) contribute to the pathogenesis of this common neurodegenerative disease. By incorporating appropriate drug(s) into biobased (lipid cubic phase) nanocarriers, one obtains a multitasking combination therapeutic which targets certain cell-surface scavenger receptors, mainly class B type I (i.e., SR-BI), and crosses the blood-brain barrier. This (intravenous) combination therapeutic, known to be a successful drug carrier, would make it possible for various cell types within the brain (all potentially implicated [see below] in Alzheimer’s disease) to be simultaneously nanotargeted for localized drug delivery via cell-surface SR-BI. Hence, such colloidal-nanocarrier targeting allows for various Alzheimer’s-related cell types to be simultaneously searched in a holistic integrative approach. This (colloidal-nanocarrier) targeting advantage, in vivo, may be particularly important when delivering pleiotropic natural substances (e.g., a flavonoid) or for repurposing an FDA-approved drug. For example, the described/proposed colloidal (nanoemulsion) nanocarrier is especially useful for the delivery of low-molecular-weight compounds having a significant degree of lipophilicity (i.e., low water solubility), such as various pleiotropic natural substances (e.g., the plant polyphenol known as “resveratrol”, commonly used as a dietary supplement).
... Thus, it is not clear whether T2D favors AD [135,136], or vice versa [137,138], or whether both conditions evolve in parallel [139]. Among the favored mechanisms underlying this connection, T2D-induced brain Aβ accumulation [140], tau hyperphosphorylation [141], Aβ-induced loss of insulin sensitivity [142], T2D-associated oxidative stress and inflammation [134], subclinical blood-brain barrier (BBB) breakdown [143], and defective insulin signaling [144] have been proposed. ...
Article
Full-text available
Despite decades of intense research, disease-modifying therapeutic approaches for Alzheimer’s disease (AD) are still very much needed. Apart from the extensively analyzed tau and amyloid pathological cascades, two promising avenues of research that may eventually identify new druggable targets for AD are based on a better understanding of the mechanisms of resilience and vulnerability to this condition. We argue that insulin-like growth factor I (IGF-I) activity in the brain provides a common substrate for the mechanisms of resilience and vulnerability to AD. We postulate that preserved brain IGF-I activity contributes to resilience to AD pathology as this growth factor intervenes in all the major pathological cascades considered to be involved in AD, including metabolic impairment, altered proteostasis, and inflammation, to name the three that are considered to be the most important ones. Conversely, disturbed IGF-I activity is found in many AD risk factors, such as old age, type 2 diabetes, imbalanced diet, sedentary life, sociality, stroke, stress, and low education, whereas the Apolipoprotein (Apo) E4 genotype and traumatic brain injury may also be influenced by brain IGF-I activity. Accordingly, IGF-I activity should be taken into consideration when analyzing these processes, while its preservation will predictably help prevent the progress of AD pathology. Thus, we need to define IGF-I activity in all these conditions and develop a means to preserve it. However, defining brain IGF-I activity cannot be solely based on humoral or tissue levels of this neurotrophic factor, and new functionally based assessments need to be developed.
... Diabetes-related cognitive decline (DACD) is an important chronic complication of diabetes, and studies have shown that people with diabetes mellitus have a higher risk of developing Alzheimer's disease (AD) than the general population, especially in Eastern populations [97]. The current findings support that the pathogenesis of DACD is related to a chronic neuroinflammatory response and blood-brain barrier (BBB) disruption due to hyperglycemia [98] related, but the exact mechanism remains unclear. High-mobility group protein b1 (HMGB1) is a ubiquitous nuclear protein that is released extracellularly after cell activation, stress, injury, or apoptosis and plays an important role in neurodegenerative diseases by mediating neuroinflammation [99,100]; an in vivo trial demonstrated that [101] that inhibition of HMGB1 may alleviate NLRP3 inflammatory vesiclemediated inflammatory responses in hippocampal neurons via the TLR4/MAPKs/NF-κB signaling pathway. ...
Article
Full-text available
Objective To visualize and analyze the published literature on diabetes mellitus and pyroptosis based on a bibliometric approach, so as to provide a comprehensive picture of the hot research directions and dynamic progress in this field. Methods This study was based on the web of science core collection database to conduct a comprehensive search of the published literature in the field of diabetes mellitus and Pyroptosis from January 1985 to August 2022, including the published research literature in this field, as well as a visual analysis of the number of citations, year of publication, journal, author, research institution, country, and research topic. Results A total of 139 literature on research related to diabetes mellitus and cellular scorch from 2011 to 2022 were retrieved, with a total of 3009 citations and a maximum of 255 citations for a single article, which had a first author Schmid-Burgk, JL The first author of this article is from Germany; among 20 publishing countries, China leads with 100 articles; among 222 publishing institutions, Harbin Medical University leads with 18 articles and 184 citations; among 980 authors, Chen, X from China tops the list of high-impact authors with 5 articles and 29 citations. Among the 98 journals, "CELL DEATH DISEASE" ranked first in both volume and high-impact journals with 4 articles and 29 citations. Among 349 keywords, "pyroptosis" ranked first with a cumulative frequency of 65 times. The cluster analysis was divided into three categories, chronic complications of diabetes mellitus and pyroptosis (67 articles), diabetes mellitus and pyroptosis (60 articles), and diabetes mellitus combined with other diseases and pyroptosis (12 articles), and the number of articles related to diabetes mellitus and its chronic complications increased rapidly from 2019, among which, diabetic cardiomyopathy (27 articles) had the highest number of articles. Conclusions Based on a comprehensive analysis of published literature in the field of diabetes mellitus and pyroptosis from 2011 to 2022, this study achieved a visual analysis of studies with significant and outstanding contributions to the field, thus framing a picture showing the development and changes in the field. At the same time, this study provides research information and direction for clinicians and investigators to conduct diabetes mellitus and pyroptosis-related research in the future.
... 150 In other words, BBB breakdown makes a bridge between diabetes and dementia. 151 Consequently, it appears to be a logical goal to seek synchronous changes in the BBB and BRB. An in-vivo study showed that recombinant annexin A1, a drug that targets cell membrane proteins involved in membrane dynamics, decreases inflammation and repairs BBB. ...
Article
Diabetic retinopathy (DR) is a major contributor to permanent vision loss and blindness. Changes in retinal neurons, glia, and microvasculature have been the focus of intensive study in the quest to better understand DR. However, the impact of diabetes on the rest of the visual system has received less attention. There are reports of associations of changes in the visual system with preclinical and clinical manifestations of diabetes. Simultaneous investigation of the retina and the brain may shed light on the mechanisms underlying neurodegeneration in diabetics. Additionally, investigating the links between DR and other neurodegenerative disorders of the brain including Alzheimer's and Parkinson's disease may reveal shared mechanisms for neurodegeneration and potential therapy options.
Article
Full-text available
Background: Increased blood-brain barrier (BBB) permeability and amyloid-β (Aβ) peptides (especially Aβ1-42) (Aβ42) have been linked to Alzheimer's disease (AD) pathogenesis, but the nature of their involvement in AD-related neuropathological changes leading to cognitive changes remains poorly understood. Objective: To test the hypothesis that chronic extravasation of bloodborne Aβ42 peptide and brain-reactive autoantibodies and their entry into the brain parenchyma via a permeable BBB contribute to AD-related pathological changes and cognitive changes in a mouse model. Methods: The BBB was rendered chronically permeable through repeated injections of Pertussis toxin (PT), and soluble monomeric, fluorescein isothiocyanate (FITC)-labeled or unlabeled Aβ42 was injected into the tail-vein of 10-month-old male CD1 mice at designated intervals spanning ∼3 months. Acquisition of learned behaviors and long-term retention were assessed via a battery of cognitive and behavioral tests and linked to neuropathological changes. Results: Mice injected with both PT and Aβ42 demonstrated a preferential deficit in the capacity for long-term retention and an increased susceptibility to interference in selective attention compared to mice exposed to PT or saline only. Immunohistochemical analyses revealed increased BBB permeability and entry of bloodborne Aβ42 and immunoglobulin G (IgG) into the brain parenchyma, selective neuronal binding of IgG and neuronal accumulation of Aβ42 in animals injected with both PT and Aβ42 compared to controls. Conclusion: Results highlight the potential synergistic role of BBB compromise and the influx of bloodborne Aβ42 into the brain in both the initiation and progression of neuropathologic and cognitive changes associated with AD.
Preprint
Full-text available
Objective: To visualize and analyze the published literature on diabetes and pyroptosis based on a bibliometric approach, so as to provide a comprehensive picture of the hot research directions and dynamic progress in this field. Methods: This study was based on the web of science core collection database to conduct a comprehensive search of the published literature in the field of diabetes and Pyroptosis from January 1985 to August 2022, including the published research literature in this field, as well as a visual analysis of the number of citations, year of publication, journal, author, research institution, country, and research topic. Results: A total of 139 literature on research related to diabetes and cellular scorch from 2011 to 2022 were retrieved, with a total of 3009 citations and a maximum of 255 citations for a single article, which had a first authorSchmid-Burgk, JLThe first author of this article is from Germany; among 20 publishing countries, China leads with 100 articles; among 222 publishing institutions, Harbin Medical University leads with 18 articles and 184 citations; among 980 authors, Chen, X from China tops the list of high-impact authors with 5 articles and 29 citations; Among the 98 journals, "CELL DEATH DISEASE" ranked first in both volume and high impact journals with 4 articles and 29 citations; among 349 keywords, "pyroptosis" ranked first with a cumulative frequency of 65 times. The cluster analysis was divided into three categories, chronic complications of diabetes and pyroptosis (67 articles), diabetes and pyroptosis (60 articles), and diabetes combined with other diseases and pyroptosis (12 articles), and the number of articles related to diabetes and its chronic complications increased rapidly from 2019, among which, diabetic cardiomyopathy (27 articles) had the highest number of articles. Conclusions: Based on a comprehensive analysis of published literature in the field of diabetes and pyroptosis from 2011-2022, this study achieved a visual analysis of studies with significant and outstanding contributions to the field, thus framing a picture showing the development and changes in the field. At the same time, this study provides research information and direction for clinicians and investigators to conduct diabetes and pyroptosis related research in the future.
Preprint
Objective: To visualize and analyze the published literature on diabetes and pyroptosis based on a bibliometric approach, so as to provide a comprehensive picture of the hot research directions and dynamic progress in this field. Methods: This study was based on the web of science core collection database to conduct a comprehensive search of the published literature in the field of diabetes and Pyroptosis from January 1985 to August 2022, including the published research literature in this field, as well as a visual analysis of the number of citations, year of publication, journal, author, research institution, country, and research topic. Results: A total of 139 literature on research related to diabetes and cellular scorch from 2011 to 2022 were retrieved, with a total of 3009 citations and a maximum of 255 citations for a single article, which had a first authorSchmid-Burgk, JL The first author of this article is from Germany; among 20 publishing countries, China leads with 100 articles; among 222 publishing institutions, Harbin Medical University leads with 18 articles and 184 citations; among 980 authors, Chen, X from China tops the list of high-impact authors with 5 articles and 29 citations; Among the 98 journals, "CELL DEATH DISEASE" ranked first in both volume and high impact journals with 4 articles and 29 citations; among 349 keywords, "pyroptosis" ranked first with a cumulative frequency of 65 times. The cluster analysis was divided into three categories, chronic complications of diabetes and pyroptosis (67 articles), diabetes and pyroptosis (60 articles), and diabetes combined with other diseases and pyroptosis (12 articles), and the number of articles related to diabetes and its chronic complications increased rapidly from 2019, among which, diabetic cardiomyopathy (27 articles) had the highest number of articles. Conclusions: Based on a comprehensive analysis of published literature in the field of diabetes and pyroptosis from 2011-2022, this study achieved a visual analysis of studies with significant and outstanding contributions to the field, thus framing a picture showing the development and changes in the field. At the same time, this study provides research information and direction for clinicians and investigators to conduct diabetes and pyroptosis related research in the future.
Chapter
According to existing scientific evidence, diabetes mellitus is associated with mild to modest decrements in cognitive function. Domains of psychomotor speed, mental flexibility, attention, and general intelligence are those most frequently affected. Hypoglycemia is not usually risk factor for cognitive decline; however, this may not be true for children with young age at onset of diabetes. Relevant risk factors for the development of cognitive decline are an early age of onset and the presence of microvascular complications. Since age and duration of diabetes are important contributors to the changes in cognitive function, we need longitudinal studies looking at cognitive function, especially in elderly subject with type 1 diabetes mellitus. In addition, more information is needed in order to better understand the clinical implications of these mild-moderate decrements in cognitive function, also in terms of impact on daily lives and habits. In addition, the underlying mechanism and the risk factors that may lead to the development of more severe cognitive dysfunction like dementia in some, but not all diabetic patients are not clear. Diabetic autonomic neuropathy (DAN) represents a particular aspect of diabetic neuropathy, which may lead to impairment of several organs, including the heart, both in type 1 and type 2 diabetic patients. The pathogenesis of DAN is not entirely clear, but metabolic, genetic, and hormonal factors may be involved; however, the final pathway probably involves oxidative stress and inflammation caused by hyperglycemia. Since no therapy was demonstrated to effectively reverse DAN, prevention with close glycemic control, multifactorial intervention, and lifestyle modification remain crucial.
Article
Full-text available
The evidence for a protective role of physical activity on the risk and progression of Alzheimer's disease (AD) has been growing in the last years. Here we studied the influence of a prolonged physical and cognitive stimulation on neurodegeneration, with special emphasis on hippocampal neuron loss and associated behavioral impairment in the Tg4-42 mouse model of AD. Tg4-42 mice overexpress Aβ4-42 without any mutations, and develop an age-dependent hippocampal neuron loss associated with a severe memory decline. We demonstrate that long-term voluntary exercise diminishes CA1 neuron loss and completely rescues spatial memory deficits in different experimental settings. This was accompanied by changes in the gene expression profile of Tg4-42 mice. Deep sequencing analysis revealed an upregulation of chaperones involved in endoplasmatic reticulum protein processing, which might be intimately linked to the beneficial effects seen upon long-term exercise. We believe that we provide evidence for the first time that enhanced physical activity counteracts neuron loss and behavioral deficits in a transgenic AD mouse model. The present findings underscore the relevance of increased physical activity as a potential strategy in the prevention of dementia.
Article
Full-text available
Inflammation in the brain is a prominent feature in Alzheimer's disease (AD). Recent studies suggest that chronic inflammation can be a consequence of failure to resolve the inflammation. Resolution of inflammation is mediated by a family of lipid mediators (LMs), and the levels of these specialized pro-resolving mediators (SPMs) are reduced in the hippocampus of those with AD. In the present study, we combined analysis of LMs in the entorhinal cortex (ENT) from AD patients with in vitro analysis of their direct effects on neurons and microglia. We probed ENT, an area affected early in AD pathogenesis, by liquid chromatography-tandem mass spectrometry (LC-MS-MS), and found that the levels of the SPMs maresin 1 (MaR1), protectin D1 (PD1), and resolvin (Rv) D5, were lower in ENT of AD patients as compared to age-matched controls, while levels of the pro-inflammatory prostaglandin D2 (PGD2) were higher in AD. In vitro studies showed that lipoxin A4 (LXA4), MaR1, resolvin D1 (RvD1), and protectin DX (PDX) exerted neuroprotective activity, and that MaR1 and RvD1 down-regulated β-amyloid (Aβ)42-induced inflammation in human microglia. MaR1 exerted a stimulatory effect on microglial uptake of Aβ42. Our findings give further evidence for a disturbance of the resolution pathway in AD, and indicate that stimulating this pathway is a promising treatment strategy for AD.
Article
Full-text available
Background The lipoprotein-associated phospholipase A2 inhibitor (Lp-PLA2), rilapladib (SB659032), is being evaluated as a potential treatment to slow the progression of Alzheimer's disease (AD). Methods One hundred twenty-four subjects with possible mild AD and with neuroimaging evidence of cerebrovascular disease were randomized to placebo or 250-mg rilapladib once daily, for 24 weeks, in addition to stable background acetylcholinesterase inhibitor and/or memantine. The study assessed the safety and tolerability of rilapladib and its effects on cognition, mechanistic, and disease-related biomarkers. Although the overall intent behind the study was to take a broad exploratory view of the data, two primary end points of interest (cerebrospinal fluid [CSF] amyloid beta peptide 1–42 [Aβ1–42] and CogState executive function/working memory [EF/WM] composite score at week 24) were prespecified in the analysis plan for inferential statistical analysis. Results Rilapladib was well tolerated with no significant safety concerns. A significant difference from placebo was observed for rilapladib on change from baseline in EF/WM (effect size, 0.45; P = .026). There was no significant difference between groups on the change from baseline in CSF Aβ1–42 (P = .133). Preliminary evidence of effects was detected on other mechanistic (albumin quotient) and disease-related biomarkers (tau/P-tau and neurofilament light chain). Conclusion These data provide initial evidence supporting Lp-PLA2 inhibition as a novel treatment for dementia. Clinical Trial Registration Clinicaltrials.gov identifier: NCT01428453.
Article
Full-text available
The blood-brain barrier (BBB) regulates the transport of micro- and macromolecules between the peripheral blood and the central nervous system (CNS) in order to maintain optimal levels of essential nutrients and neurotransmitters in the brain. In addition, the BBB plays a critical role protecting the CNS against neurotoxins. There has been growing evidence that BBB disruption is associated with brain inflammatory conditions such as Alzheimer’s disease and multiple sclerosis. Considering the increasing role of inflammation and oxidative stress in the pathophysiology of bipolar disorder (BD), here we propose a novel model wherein transient or persistent disruption of BBB integrity is associated with decreased CNS protection and increased permeability of proinflammatory (e.g., cytokines, reactive oxygen species) substances from the peripheral blood into the brain. These events would trigger the activation of microglial cells and promote localized damage to oligodendrocytes and the myelin sheath, ultimately compromising myelination and the integrity of neural circuits. The potential implications for research in this area and directions for future studies are discussed.
Article
Exercise is becoming a promising therapeutic approach to prevent alterations both in Alzheimer's disease (AD) patients and in transgenic models of AD. This neuroprotection has been associated with changes in hippocampal structure and function, as well as with the reduction of amyloid-β (Aβ) production and accumulation. However, whether exercise produces lasting changes in hippocampal population activity and renders it resistant to Aβ-induced network dysfunction is still unknown. Thus, we tested whether voluntary exercise changes hippocampal population activity and prevents its alteration in the presence of Aβ, which has been associated to glycogen synthase kinase-3β (GSK3β) activation. We found that the hippocampal population recorded in slices obtained from mice that exercised voluntarily (with free access to a running wheel for 21 days) exhibits higher power and a faster frequency composition than in slices obtained from sedentary animals. Moreover, the hippocampal network of mice that exercised becomes insensitive to Aβ-induced inhibition of spontaneous population activity. This protective effect correlates with the inability of Aβ to activate GSK3β, is mimicked by GSK3β inhibition with SB126763 (in slices obtained from sedentary mice), and is abolished by the inhibition of PI3K with LY294002 (in slices obtained from mice that exercised). We conclude that voluntary exercise produces a lasting protective state in the hippocampus, maintained in hippocampal slices by a PI3K-dependent mechanism that precludes its functional disruption in the presence of Aβ by avoiding GSK3β activation.
Article
The blood-brain barrier (BBB) limits brain uptake of therapeutic antibodies. It is believed that the BBB is disrupted in Alzheimer's disease (AD), potentially increasing drug permeability de facto. Here we compared active versus passive brain uptake of systemically dosed antibodies (anti-transferrin receptor [TfR] bispecific versus control antibody) in mouse models of AD. We first confirmed BBB disruption in a mouse model of multiple sclerosis as a positive control. Importantly, we found that BBB permeability was vastly spared in mouse models of AD, including PS2-APP, Tau transgenics, and APOE4 knockin mice. Brain levels of TfR in mouse models or in human cases of AD resembled controls, suggesting target engagement of TfR bispecific is not limited. Furthermore, infarcts from human AD brain showed similar occurrences compared to age-matched controls. These results question the widely held view that the BBB is largely disrupted in AD, raising concern about assumptions of drug permeability in disease. It is generally believed that neurodegeneration is accompanied by BBB dysfunction, potentially increasing drug bioavailability in the CNS. Bien-Ly et al. report that AD mouse models lack widespread BBB disruption and display restricted passive permeability to therapeutic antibodies.
Article
Previous studies in older adults suggested beneficial effects of omega-3 fatty acid (FA) supplementation, aerobic exercise, or cognitive stimulation on brain structure and function. However, combined effects of these interventions in patients suffering from mild cognitive impairment (MCI) are unknown. Using a randomized interventional design, we evaluated the effect of combined omega-3 FA supplementation, aerobic exercise and cognitive stimulation (target intervention) versus omega-3 FA supplementation and non-aerobic exercise (control intervention) on cognitive function and gray matter volume in patients with MCI. Moreover, we analyzed potential vascular, metabolic or inflammatory mechanisms underlying these effects. Twenty-two MCI patients (8 females; 60–80 years) successfully completed six months of omega-3 FA intake, aerobic cycling training and cognitive stimulation (n = 13) or omega-3 FA intake and non-aerobic stretching and toning (n = 9). Before and after the interventions, cognitive performance, magnetic resonance imaging of the brain at 3 T (n = 20), intima-media thickness of the internal carotid artery and serum markers of glucose control, lipid and B-vitamin metabolism, and inflammation were assessed. Intervention-related changes in gray matter volume of Alzheimer's disease (AD)-related brain regions, i.e., frontal, parietal, temporal and cingulate cortex were examined using voxel-based morphometry of high resolution T1-weighted images.