Vous êtes sur la page 1sur 20

Compendium on the Pathophysiology and

Treatment of Hypertension
Neurovascular and Cognitive Dysfunction in Hypertension
Epidemiology, Pathobiology, and Treatment
Costantino Iadecola, Rebecca F. Gottesman

Abstract: Hypertension has emerged as a leading cause of age-related cognitive impairment. Long known to be
associated with dementia caused by vascular factors, hypertension has more recently been linked also to Alzheimer
disease—the major cause of dementia in older people. Thus, although midlife hypertension is a risk factor for late-
life dementia, hypertension may also promote the neurodegenerative pathology underlying Alzheimer disease.
The mechanistic bases of these harmful effects remain to be established. Hypertension is well known to alter
in the structure and function of cerebral blood vessels, but how these cerebrovascular effects lead to cognitive
impairment and promote Alzheimer disease pathology is not well understood. Furthermore, critical questions
also concern whether treatment of hypertension prevents cognitive impairment, the blood pressure threshold for
treatment, and the antihypertensive agents to be used. Recent advances in neurovascular biology, epidemiology,
brain imaging, and biomarker development have started to provide new insights into these critical issues. In this
review, we will examine the progress made to date, and, after a critical evaluation of the evidence, we will highlight
questions still outstanding and seek to provide a path forward for future studies.   (Circ Res. 2019;124:1025-1044.
DOI: 10.1161/CIRCRESAHA.118.313260.)
Key Words: Alzheimer’s disease ◼ biomarkers ◼ brain blood supply ◼ dementia
◼ risk factors ◼ white matter pathology

E levated arterial blood pressure (BP) contributes signifi-


cantly to the global burden of disease, its harmful effects
Subsequent population and prospective studies clearly es-
tablished the relationship between elevated BP and cognitive
Downloaded from http://ahajournals.org by on June 30, 2019

on the brain being a major culprit.1Well established as the impairment7 and unveiled an unanticipated link between hy-
leading risk factor for stroke, hypertension has also emerged pertension and AD8. With the development of safe and effective
as a pathogenic factor in cognitive impairment with a vas- antihypertensive drugs and their widespread use, during the past
cular basis and Alzheimer disease (AD)—the major causes several decades, a dramatic reduction in the morbidity and mor-
of dementia in the elderly.2 The harmful effects of hyperten- tality caused by cardiovascular pathologies, stroke in particular,
sion on cognitive function were already noted at the end of was observed in a number of large clinical trials.9 Surprisingly,
the 19th century, in patients who later developed massive however, similar beneficial effects on cognition were not u-
cerebral hemorrhages, but systematic investigations on the niformly noted.10 Owing to the rapid rise in the prevalence of
phenomenon did not appear until the 1950s to 1960s. In par- age-related dementia worldwide, hypertension has received
ticular, pioneering observations on the neuropsychological increasing attention as a treatable condition that, if properly
performance of air traffic controllers with hypertension by controlled, could potentially stave off the onset of the cognitive
Spieth3 and the longitudinal study on cognitive function in deficits. Consequently, an increasing number of studies have
hypertensives of Wilkie and Eisdorfer4 provided initial ev- focused on the mechanisms by which hypertension impairs
idence that hypertension deteriorates cognition. The intro- cognitive function, on the temporal relationships between hyper-
duction of methods to measure cerebral blood flow (CBF) tension and cognitive deficits during the life course, and on the
let to the discovery that individuals with hypertension have ideal BP targets to achieve to minimize its impact on the brain.
increased cerebrovascular resistance, which correlated with Although this body of new knowledge has provided a deeper
the degree of hypertensive retinopathy,5 substantiating earlier insight into the relationship between hypertension and the
neuropathological findings linking cerebrovascular damage brain, new questions have emerged that need to be addressed.
with the effects of hypertension on brain health. However, The present review seeks to provide an assessment of
the prevailing belief at the time was that the elevation in BP the state of the art of the effects of hypertension on cognitive
was a beneficial compensatory mechanism aimed at main- function, on the underlying structural and functional altera-
taining normal cerebral perfusion, and attempts to lower BP tions and their mechanisms, and on potential preventive and
were considered dangerous.6 therapeutic strategies to minimize them. After a critical review

From the Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York (C.I.); and Departments of Neurology (R.F.G.) and
Epidemiology (R.F.G.), Johns Hopkins University, Baltimore, MD.
Correspondence to Costantino Iadecola, MD, Feil Family Brain and Mind Research Institute, 407 E 61st St, RR-303, New York, NY 10065. Email
coi2001@med.cornell.edu
© 2019 American Heart Association, Inc.
Circulation Research is available at https://www.ahajournals.org/journal/res DOI: 10.1161/CIRCRESAHA.118.313260

1025
1026  Circulation Research  March 29, 2019

have a stronger public health message because these concepts


Nonstandard Abbreviations and Acronyms
are easier to define and understand, and cases of preventable
Aβ amyloid-β disease are easily embraced, as opposed to changes in steep-
ACE angiotensin-converting enzyme ness of cognitive slopes. For hypertension, evidence from
AD Alzheimer disease epidemiological studies supports all of these outcomes: hyper-
Ang II angiotensin II
tension is clearly associated with steeper cognitive decline,12,13
ApoE apolipoprotein-E
poor cognitive performance,14 as well as incident MCI15 and
dementia.16–19 Furthermore, among adults with MCI, elevated
APP amyloid precursor protein
BP is associated with increased progression and worsening
ARIC Atherosclerosis Risk in Communities
of cognition.20 Prehypertension, previously considered as a
AT1R angiotensin type-1 receptor
systolic BP (SBP) <140 mm Hg, has even been reported as a
BBB blood-brain barrier
risk factor for dementia in several studies, including the ARIC
BP blood pressure
(Atherosclerosis Risk in Communities) cohort,18 in which both
BPH blood pressure high
midlife hypertension and prehypertension conferred a similar
CBF cerebral blood flow risk for dementia, at about 40% higher than among individuals
CCB calcium channel blocker with normotension. New definitions of hypertension now con-
Cdk5 cyclin-dependent kinase-5 sider SBP in this range consistent with stage 1 hypertension.21
CSF cerebrospinal fluid Although epidemiological data emphasize the importance
DBP diastolic blood pressure of hypertension in the development of stroke, the impact on
EVA Epidemiology of Vascular Aging cognitive outcomes appears to be independent of stroke. This
GSK3β glycogen synthase kinase-3 is the case when stroke is defined clinically, with similar as-
HAAS Honolulu-Asia Aging Study sociations when individuals with incident stroke are excluded
ISF interstitial fluid from analyses on cognition,7,18 but fewer studies have system-
LYVE1 lymphatic vessel endothelial hyaluronan receptor-1 atically excluded subclinical stroke or small vessel disease
MCI mild cognitive impairment (SVD; including white matter lesions, lacunes, and enlarged
MRI magnetic resonance imaging perivascular spaces), important mediators of the impact of hy-
NOX nicotinamide adenine dinucleotide phosphate oxidase pertension on cognitive outcomes.
PET positron emission tomography The primary focus of this review is hypertension and its com-
PROGRESS Perindopril Protection Against Recurrent Stroke Study ponents, SBP and diastolic BP (DBP), but different components
Downloaded from http://ahajournals.org by on June 30, 2019

PVM perivascular macrophage or aspects of BP may also play an important role in dementia.
ROS reactive oxygen species Although pulse pressure was not significantly associated with
SBP systolic blood pressure dementia in the HAAS (Honolulu-Asia Aging Study),17 it was
SHR spontaneously hypertensive rat associated with cognitive decline among APOE (apolipoprotein-
SVD small vessel disease
E)-ε4 carriers in the Framingham Offspring Study.22 Most hu-
man studies do not differentiate between primary and secondary
causes of hypertension; although most studies primarily enroll
individuals with essential hypertension, rodent models consid-
of the evidence, we will attempt to identify outstanding ques- ering several mechanisms of hypertension (including modeling
tions and knowledge gaps that remain to be addressed. renovascular hypertension along with SHRs [spontaneously hy-
pertensive rats]) have demonstrated similar cognitive deficits and
Impact of Hypertension on Cognition white matter hyperintensities. Orthostatic hypotension, which
A growing body of evidence supports the role of hyperten- occurs more frequently in individuals with hypertension, was
sion as a risk factor for adverse cognitive outcomes. This ev- associated with a 54% higher risk (95% CI, 1.20–1.97), with
idence primarily comes from epidemiological studies, which relationships persisting after censoring for ischemic stroke.23 In
consider hypertension in communities as a risk factor for a Uppsala, Sweden, using ambulatory BP monitoring, nondip-
range of adverse outcomes, including cognitive decline (wors- ping circadian BP patterns and persistently high 24-hour BP (at
ening of cognitive function over years to decades, steeper 70 years of age) was associated with worse cognitive perfor-
than expected because of age alone), mild cognitive impair- mance.24 BP variability has also been described in several stud-
ment (MCI; reduced function in memory, thinking, and other ies as an important risk factor for reduced cognitive function.25
cognitive domains but not impacting daily functioning), and
dementia (impairments in cognition, including memory and Age-Dependent Risk (Midlife Versus Late Life)
other cognitive domains, but with adverse impacts on daily Hypertension’s impact on late-life cognitive outcomes appears
functioning). Studies considering cognitive decline allow e- the greatest when considered in middle age. Several studies
valuation of change, which captures more subtle cases of have demonstrated cognitive decline in people aged ≥70 years
cognitive dysfunction, without requiring frank thresholds for when BP is elevated in the fourth or fifth decade of life. In the
dementia or MCI to be met but also may be a less confounded HAAS, risk of reduced cognitive function in late life increased
measure of cognitive status, allowing a more pure evaluation as midlife SBP increased,14 and elevated SBP (>140 mm Hg)
of the association between hypertension and cognition.11 In in midlife was associated with a 1.77× higher risk of dementia
contrast, studies considering MCI and dementia as outcomes at ages 71 to 93.17 Furthermore in HAAS, elevated midlife BP
Iadecola and Gottesman   Hypertension and Cognition   1027

(DBP, especially) and decrease in plasma Aβ (amyloid-β) 1–40 and the hippocampus, in this case).34 Other studies have also
interacted to increase risk of late-life dementia.26 In the National reported strong associations with hypertension for SVD, in-
Heart, Lung, and Blood Institute Twin Study, higher midlife cluding white matter hyperintensities35 and their progression.36
SBP was associated with steeper 10-year cognitive decline.13 In
the ARIC cohort, hypertension in midlife was associated with Hypertension During the Life Course
steeper 20-year cognitive decline on 3 repeated cognitive tests, Although the above studies emphasize the impact of midlife
without similar associations with late-life hypertension, which hypertension as a risk factor for cognitive decline and demen-
was not associated with cognitive decline during the preceding tia, change in BP may be relevant as well. As discussed later
20 years.12 Other studies have failed to find an association be- in this review, BP changes are particularly important when
tween BP and dementia or cognitive decline,27 but this may be considering shifts that occur in an individual’s autoregula-
because they primarily focused on BP in older adults and with tory curve in response to that individual’s baseline value. In
a shorter follow-up period, without evaluation of hypertension the HAAS study, men who developed dementia had a gradual
during its maximal risk period (middle age). increase in SBP from midlife to late life, with a subsequent
Some studies have suggested a U-shaped relationship be- decrease of 1.36 mm Hg/year in late life; in addition, of the
tween BP and cognition, but these studies seem isolated to 109 participants in this subcohort who developed dementia,
studies of older adults, further reflecting the potential detri- the majority (58%) had had a recent fall in SBP during the pre-
mental effect of midlife hypertension but perhaps even a pro- ceding 6 years.37 Similar patterns were shown in the Göteborg
tective effect of late-life hypertension. There is no convincing Longitudinal Population Study, where 70 year olds with el-
evidence that low BP, in younger people, is associated with evated BP had more dementia at 79 to 85 years of age, but
bad cognitive outcomes, but several studies do suggest worse many also experienced decline in BP in the years more prox-
outcomes in older individuals with low BP. In 1 study, cogni- imal to a dementia diagnosis.38 In the Framingham Offspring
tive performance was worst among individuals with either SBP Cohort, elevated midlife BP was an important risk factor for
<130 mm Hg or with elevated SBP (≥160 mm Hg), compared dementia, but a steep decline in BP from midlife to late life
with people at 130 to 139 mm Hg, but this cohort consisted was associated with a 2.4× higher risk of dementia in later life
of adults aged 65 to 102 years.28 In an analysis including data (95% CI, 1.4–4.2).39
from the Rotterdam study and the Göteborg H-70 study, hyper- One complicating factor in studies of BP trajectories is the
tension in old age appeared protective: elderly adults had an fact that BP often falls in response to incident cognitive impair-
inverse association between BP and dementia, with a reduced ment and dementia, making it difficult to determine whether
relative risk for dementia of 0.93 (95% CI, 0.88–0.99) per 10 the apparent relationship with recent decline in BP is actually
Downloaded from http://ahajournals.org by on June 30, 2019

mm Hg higher SBP, but only among elderly adults taking anti- because of reverse causation. Studies support lower and declin-
hypertensive medications.29 In the Bronx Aging Study, adults ing BP in elderly individuals with lower cognitive performance,
>75 years of age with SBP between 140 and 179 mm Hg had a some of whom went on to develop dementia,40 consistent with
lower risk for AD (hazard ratio, 0.55; 95% CI, 0.32–0.96), and this concept of reverse causation possibly playing a role. This
risk for dementia was especially high in individuals with low may also explain the above-cited Rotterdam/Göteborg H-70
DBP at baseline (age >75 years).30 A U-shaped relationship result,29 where hypertension in old age appeared protective.
between BP and cognition was also reported in the Baltimore Studies considering brain volumes as an outcome have also
Longitudinal Study on Aging, among adults >80 years of age found evidence that a late-life decline in BP may be a marker
who had decline at both extremes of BP.31 for brain volume loss, with more cortical atrophy in individuals
Although most observed associations are with demen- with a drop in DBP of >10 mm Hg over 20 years in 1 study.41 In
tia broadly defined, several studies have attempted to cap- the ARIC study, the pattern of midlife hypertension with sub-
ture dementia subtypes. Given the lack of gold standard for sequent late-life hypotension was associated with smaller brain
subtyping dementia, it is plausible that these etiologic clas- volumes in regions typically affected in AD.42
sifications actually represent mixed pathogenesis, so still may Persistently elevated BP, from midlife and continuing
point to a role of hypertension in dementias of other types. In through later life, does appear to be consistently associated
the Hisayama study, midlife (more so than late life) hyper- with worse cognitive outcomes: Yaffe et al,43 evaluated cu-
tension was associated with vascular dementia, but not AD,32 mulative SBP and DBP level over 25 years, finding worse
with similar findings in the Adult Health Study in Hiroshima, cognitive performance on several tests among individuals
which evaluated SBP in association with subsequent 25- with higher areas under the curve based on cumulative BP.
to 30-year development of vascular dementia.33 In a large However, using a Mendelian randomization design, genet-
Finnish study, midlife SBP ≥160 mm Hg was associated with ically predicted systolic hypertension was associated with
a >2-fold higher risk of AD, independent of other vascular and lower risk of AD, possibly indicative of subsequent changes
demographic risk factors.19 in lifestyle or management in response to this elevated risk.44
Studies using imaging surrogates for cognitive or demen-
tia status also support the importance of midlife hypertension. Cognitive Domains Affected
In the National Heart, Lung, and Blood Institute Twin Study, Although several studies have suggested that the impact of
midlife SBP was associated with not only more white matter hypertension on cognition is global, this is mostly the case
hyperintensities in later life but also smaller brain parenchy- because not all studies broadly consider several distinct cogni-
mal volumes.13 Furthermore, BP appears particularly related tive domains or because the most impacted domains in hyper-
to atrophy in regions highly relevant for AD (the amygdala tension-associated brain disease are also represented in global
1028  Circulation Research  March 29, 2019

cognitive measures. When more detailed neuropsychological enter the skull and merge the base of the brain to form the
batteries are performed and hypertension-specific effects are circle of Willis, from which the major intracranial vessels
considered and compared, the biggest impact of hypertension supplying the brain originate (anterior, middle, and poste-
appears to be on executive function,12 motor speed, and at- rior cerebral arteries; Figure 1).50 Branches of these arteries
tention,45 which are classically considered domains involved travel along the brain surface giving rise to a highly anas-
with subcortical disease, such as typical vascular disease or tomotic vascular network (pial arteries).50 Branches of the
pure vascular dementia.46 Even scores on the Mini-Mental pial arteries penetrate the substance of the brain (penetrating
State Examination and Montreal Cognitive Assessment, 2 arterioles) surrounded by a virtual space, the perivascular
global cognitive measures, although lower in individuals with space, or Virchow-Robin space,50 separating the vascular ba-
more vascular risk factors, including hypertension, are prima- sal lamina from the astrocytic processes enveloping the brain
rily driven by impairments in items related to attention (for parenchyma (glial limitans). As the vessels get smaller, the
the Mini-Mental State Examination) and visuoexecutive func- perivascular space disappears and the vascular basal lamina
tion (for lower scoring individuals on the Montreal Cognitive enters in direct contact with the astrocytic foot processes,
Assessment).47 which completely envelop the vessel’s outer surface.50 At
The role of memory impairments in hypertension-relat- this level, the vessel wall includes a single layer of smooth
ed dementias is complicated by the likely overlap between muscle cells resting on the endothelial basement mem-
pathogeneses and the high prevalence of mixed dementias. brane.50 As the vessels transition into capillaries, the smooth
Although memory is more typically involved in AD-related muscle cell layer becomes discontinuous and is gradually
cognitive impairments than in vascular types of dementia, replaced by pericytes covering ≈30% of the abluminal cap-
it has been noted to have impairments in individuals with a illary surface.51 Venules are endowed with a discontinuous
combination of hypertension and elevated brain amyloid smooth muscle cell layer, which becomes more pronounced
by Pittsburgh compound-B positron emission tomography in veins, although not as marked as in the media of arter-
(PET).48 However, many of these studies of domain-specific ies. Recent single-cell RNA sequencing studies have started
cognitive deficits are influenced by misdiagnosis or difficulty to unveil the molecular bases of the diversity of the cellu-
determining pathogenesis during life. Using a study neuro- lar composition of the cerebrovascular tree.52 These efforts,
pathologic data, domains did not differ by pathogenesis as still in an initial stage, will provide critical insight into the
might be expected: individuals with pure AD neuropathology cellular, molecular, and signaling mechanisms underlying
had reduced memory scores, more so than executive function, the segmental susceptibility to hypertension of the cerebral
but individuals with cerebrovascular disease had similarly re- vasculature.
Downloaded from http://ahajournals.org by on June 30, 2019

duced premorbid cognitive function in executive function and


verbal and nonverbal memory.49 Effects of Hypertension on Cerebrovascular
In summary, epidemiological data strongly support that Structure
hypertension is associated with worse cognitive outcomes, es- Chronic hypertension exerts a profound influence on the struc-
pecially when BP is considered in midlife. A pattern of midlife ture of the cerebral vasculature. These structural changes re-
hypertension followed by later life hypotension or normoten- flect an adaptive response to protect downstream microvessels
sion may be especially problematic, and consideration of BP from the increased transmural pressure. However, this process
variability not only during the course of a day but during years over time becomes maladaptive resulting in different patholo-
and decades may be important in understanding what compo- gies depending on the segment of the cerebrovascular tree in-
nents of BP are the most important in increasing risk of cogni- volved (Figure 1).
tive decline, MCI, and dementia. Although hypertension may
impact all aspects of cognition, its impact is the greatest on Large Cerebral Arteries
motor speed, attention, and executive functioning. The likely Hypertension is a leading risk factor for extracranial and
overlap with AD pathology, however, in the large group of in- intracranial atherosclerosis.53 Extracranial lesions are char-
dividuals with mixed pathology of MCI and dementia, makes acterized by lipid accumulation in carotid and vertebral
differentiation of dementia subtype on the basis of patterns of arteries, often associated with ulceration and atheroma in-
impaired cognitive domains challenging. stability and protrusions, all linked to artery-to-artery em-
bolism.54 Intracranial lesions, affecting the circle of Willis
and its major branches, have a more fibrous component,
Pathobiology of the Effects of Hypertension more likely to result in local vascular occlusions and focal
on the Brain ischemic stroke.55
The cerebral vasculature is the prime target of the deleterious Remodeling is an important structural change induced
effects of hypertension on the brain. In this section, we will by hypertension and other vascular risk factors in extra-
briefly review basic concepts on the blood supply of the brain cranial and intracranial cerebral arteries.56 Hypertrophic
and its regulation and examine how chronic hypertension af- remodeling leads to increased wall thickness and reduction
fects the structure and function of cerebral blood vessels. in vascular lumen, with an increase in media-to-lumen ra-
tio and in the total volume of wall tissue.56 Eutrophic (in-
Blood Supply of the Brain ward) remodeling consists of an increased media thickness,
The brain’s blood supply is provided by the internal carotid a reduced lumen, and external diameter, without changes in
arteries and the vertebral arteries. These extracranial vessels the total amount of wall tissue.56 In a recent study assessing
Iadecola and Gottesman   Hypertension and Cognition   1029

Figure 1. Cerebrovascular anatomy and segmental pathology induced by hypertension. Large extracranial cerebral arteries (internal carotid arteries and
vertebral arteries) enter the skull and converge at the base of the brain to form the circle of Willis from which the major intracranial cerebral arteries emanate.
Downloaded from http://ahajournals.org by on June 30, 2019

Branches of these arteries form a dense anastomotic network (pial arteries and arterioles), which give rise to arterioles that dive into the substance of the
brain (penetrating arterioles). Some vessels, like the lenticulostriate arteries, arise from the circle of Willis and proximal branches and vascularize the deep
brain structures, including the white matter (WM). The predominant pathology caused by hypertension in the different segment of the cerebrovascular tree is
indicated. Because of its precarious blood supply from terminal arterioles particularly vulnerable to hypertensive damage, the subcortical WM is highly likely
to be harmed by hypoxia-ischemia. ICA indicates internal carotid artery; and MCA, middle cerebral artery.

remodeling in the carotid arteries using magnetic resonance that hypertrophic remodeling and eutrophic remodeling
imaging (MRI), the pattern of remodeling varied according are the predominant alterations in pial and penetrating ves-
to the arterial segment.57 Whereas the common carotid artery sels, stiffening being less common than in large arteries.56
demonstrated hypertrophy, the carotid bifurcation exhibited These vessels are often surrounded by enlarged perivascu-
eutrophic remodeling. In contrast, the internal carotid artery lar spaces—a hallmark of SVD.63 Particularly vulnerable
exhibited a mixed pattern of inward and outer remodeling.57 are small arteries and arterioles of the deep cortical, peri-
Sex and comorbidities had a significant effect on the pattern ventricular, and basal ganglia white matter. These vessels
of remodeling.57 arise either from the first segment of the middle cerebral
Hypertension is also associated with stiffening of large artery, for example, lenticulostriate arteries, or from termi-
arteries. In humans as in animal models, stiffening precedes nal branches of the pial arteries, and converge on the white
the development of hypertension, raising the possibility that matter deep into the hemisphere (Figure 2).64 Because of
it may contribute to the BP elevation by reducing vascular the limited anastomosis and collateralization among pene-
compliance and increasing pulse pressure.58 At the same time, trating arterioles, their dysfunction or occlusion is particu-
stiffening may also reduce CBF,59 deplete cerebrovascular larly damaging to the white matter.62,65,66 Hypertension also
reserves,60 and increase the hydrodynamic impact of the ele- induces degenerative changes of the vessel wall, including
vated pressure (pulsatility) on the cerebral microvasculature.61 accumulation of an amorphous material consisting of de-
Perhaps because of these deleterious microvascular actions, generated smooth muscle and collagen (lipohyalinosis) or
arterial stiffening is a sensitive predictor of subsequent white infiltration of fibrin and fibrin degradation products (fibrin-
matter lesions and cognitive impairment. oid necrosis).67 Possibly because of their shorter length and
higher pressures, fibrinoid necrosis is more common in ba-
Pial and Penetrating Arteries and Arterioles sal ganglia arterioles and is frequently associated with ce-
These vessels are responsible for a sizable component of rebral hemorrhage.67 Microatheromas can also be observed
the pressure drop observed between large arteries and the in larger arterioles, which can lead to vascular occlusion.64
cerebral capillaries50,62 and, as such, are a prime target of the Alterations are also observed in capillaries, with reduction
effects of chronic elevations in BP. Animal studies suggest in their number, swelling and loss of endothelial cells and
1030  Circulation Research  March 29, 2019

Figure 2. Cerebrovascular autoregulation and hypertension. Autoregulation keeps cerebral blood flow (CBF) relatively stable during fluctuations in blood
pressure (BP). Autoregulation was first examined by measuring CBF during stepwise changes in BP, once a steady state is reached (static autoregulation;
left). Based largely on animal data, chronic hypertension was found to shift the pressure-flow curve to the right, such that the autoregulated range moves
toward higher BP. The development of methods to assess CBF dynamically like transcranial doppler flowmetry enabled to study the flow velocity changes in
large cerebral arteries during BP transients induced by different maneuvers, for example, standing from a sitting position. As illustrated in the right, middle
cerebral artery (MCA) flow drops in sync with mean arterial pressure (MAP). But, owing to an autoregulatory drop in cerebrovascular resistance (CVR), it
recovers faster. Evidence suggests that hypertension does not impair dynamic autoregulation, except in severe hypertension (see text for further details).
Right, Data derived from Aaslid et al.69

pericytes (string vessels), tortuosity, increased thickness of Impact of Hypertension on Cerebrovascular


the basal lamina, and fibrin deposition.68 Function
Endowed with limited energy reserves, the brain is highly de-
What Causes Remodeling and Stiffening?
pendent on the timely delivery of oxygen and glucose through
Remodeling and stiffening result from interacting mechanical,
blood flow to maintain its functional and structural integrity.50
Downloaded from http://ahajournals.org by on June 30, 2019

humoral, and cellular factors, including hemodynamic stress,


To this end, sophisticated neurovascular regulatory mecha-
endothelial dysfunction, vascular inflammation, immune cell
nisms assure that the blood supply is well matched to the
infiltration, and calcium deposition.70,71 Both processes are
brain’s regionally and temporally diverse metabolic require-
driven by overlapping mediators, including inflammatory cy-
ments, balancing energy delivery with removal of unwanted
tokines, sex hormones, Ang II (angiotensin II), endothelin,
byproducts of brain activity.50 In this section, we will first re-
aldosterone, and oxidative stress.72 In addition, modifications
view these mechanisms and then examine their disruption by
of the extracellular matrix play an essential role in stiffening.
hypertension.
Accumulation of collagen and fibronectin, elastin fragmenta-
tion by matrix metalloproteases, and the profibrotic effects of Neurovascular Coupling and Endothelial
transforming growth factor-β are critical factors in the pro- Regulation
cess.70 A deficit in endothelial NO and free radicals may play Neurons, astrocytes, and vascular cells work as a single func-
an important role in remodeling.56,72 In the aorta, perivascu- tional unit (neurovascular unit) to maintain the homeostasis
lar macrophages (PVM) expressing the lymphatic hyaluronan of the brain’s internal milieu.50 Brain activity is a major reg-
receptor LYVE1 (lymphatic vessel endothelial hyaluronan ulator of local cerebral perfusion, such that increased synap-
receptor-1) safeguard the elasticity of the vascular wall by tic activity leads to increases in CBF highly localized to the
governing collagen content in smooth muscle cells through activated area.50 This fundamental property of the cerebral
hyaluronan and pericellular matrix metalloprotease-9.73 circulation (neurovascular coupling) results from the close in-
Although LYVE1+ PVMs are present also in cerebral arter- teraction among cells of the neurovascular unit at all levels of
ies,73,74 it remains unclear whether they serve a similar role in the cerebrovascular tree. Although the mechanistic bases of
the homeostasis of the cerebrovascular wall. the hemodynamic response remain to be completely elucidat-
In summary, stiffening, hypertrophic remodeling, and eu- ed, it is now clear that neurons, astrocytes, endothelial cells,
trophic remodeling result from the concerted action of me- vascular smooth muscle cells, and pericytes all participate in
chanical, cellular, and molecular factors that converge on the this carefully orchestrated process through multiple diffusible
vascular wall to alter its structure and composition, as well mediators (NO, prostanoids, adenosine, ions, etc), ion chan-
as the balance between collagen and elastin. Intended to pro- nels, and segment-specific signaling mechanisms.50 Thus, ac-
tect the downstream microvessels from the mechanical stress tivation of neurons deep in the substance of the brain initiates
associated with elevated pressure, these responses become a series of highly coordinated vascular changes that start at
maladaptive and, in concert with the functional alterations de- the level of the capillary endothelium and are transmitted to
scribed in the next section, contribute to the harmful effects of upstream arterioles through intramural vascular signaling re-
hypertension on the brain. sulting in their relaxation. The retrograde propagation of the
Iadecola and Gottesman   Hypertension and Cognition   1031

vasodilation ultimately involves larger vessels at the brain’s flow in patients with hypertension, indicating a deficit of NO
surface, which is required to induce a vigorous increase in and endothelial dysfunction.87 Furthermore, postmortem anal-
flow to the activated area.50,75 Endothelial cells are important ysis of arterioles of patients with SVD, most often caused by
regulator of cerebral perfusion by producing powerful vasodi- hypertension, showed a reduced response to the endothelium-
lators, for example, NO, prostacylin, or vasoconstrictors, for dependent vasodilator acetylcholine.89 Independent evidence
example, endothelin-1.76 These vasoactive mediators are re- for endothelial dysfunction is also provided by the observation
leased in response to mechanical or chemical stimuli resulting that hypertension alters the BBB90—a property of the cerebral
in increased or decreased in blood flow (endothelium-depen- microvasculature attributable to endothelial cells.77
dent vasodilatation or vasoconstriction).76 The endothelium is The implications of cerebral endothelial dysfunction
also the site of the blood-brain barrier (BBB) governing the for the effects of hypertension on cognition are paramount.
bidirectional molecular exchange between brain and blood.77 Endothelial dysfunction is a prelude to atherosclerosis76 and,
Furthermore, the endothelium exerts vital trophic effects on as mentioned above, contributes to vascular remodeling and
brain cells and contributes to maintain the health of neurons, stiffening. Failure of endothelium-dependent vasodilation in
glia, and oligodendrocytes.78,79 peripheral arteries correlates with white matter lesion bur-
Hypertension reduces resting CBF and suppresses neu- den91 and microbleeds,92 whereas in a population of elderly
rovascular coupling and endothelium-dependent responses. hypertensives with subjective memory complaints, white mat-
Contrary to early observations,5 several studies have shown ter lesion burden was associated with circulating levels of von
focal or global reductions in resting CBF in individuals with Willebrand factor— an index of endothelial dysfunction.93
hypertension.80,81 In a large-scale longitudinal study of hyper- The BBB dysfunction is also likely to have a devastating ef-
tensives with ischemic lesions and other vascular risk factors, fect on the brain, especially in the white matter, wherein in-
a reduction in global CBF was observed with uncontrolled or creased BBB permeability has been implicated in the damage
untreated hypertension but not with satisfactory BP control.82 produced by hypertension.90
In cognitively normal people with hypertension, but no other
risk factors or evidence of brain damage, focal reductions in Cerebrovascular Autoregulation
CBF were observed in the prefrontal, cingulate, temporal, and Cerebrovascular autoregulation refers to the ability of brain
occipital cortex,83 suggesting that the reduction in CBF may vessels to maintain a relatively stable CBF, despite changes
precede the development of brain pathology. It remains un- in BP within a certain range.94 Owing to cerebrovascular au-
clear whether these reductions in CBF are a consequence of toregulation, CBF does not follow passively the BP swings
reduced brain energy metabolism or of a direct effect of hy- that occur during activities of daily living, protecting the brain
Downloaded from http://ahajournals.org by on June 30, 2019

pertension on the cerebral vasculature. Although reductions from excessive CBF fluctuations. Autoregulation has tradition-
in glucose utilization—an index of energy metabolism—have ally been assessed using stepwise changes in BP and measur-
been reported in neocortical and subcortical brain regions of ing the corresponding changes in CBF when a steady state is
patients with well-controlled longstanding hypertension,84 it reached (static autoregulation).95,96 In the normal state, plot-
has not been established whether CBF is also reduced in these ting the CBF change as a function of the BP changes leads to
regions. However, in patients with severe hypertension, oxy- an italic S-shaped curve with a plateau in which CBF remains
gen consumption is reduced in regions with reduced CBF,81 relatively stable, despite changes in BP (the autoregulated
suggesting that with disease progression, the vascular effect range: 60–150 mm Hg mean BP; Figure 2).95 More recently,
could be related to reduced metabolic demands possibly re- methods have been developed to assess cerebrovascular per-
lated to brain dysfunction and damage. fusion dynamics in response to fast changes in BP (dynamic
Relatively few studies have investigated neurovascular autoregulation; Figure 2).94 These studies have shown that
coupling in patients with hypertension. The cerebrocortical during pressure changes on a timescale of seconds, blood flow
CBF response produced by cognitive tasks (memory and at- does not remain stable but follows BP until the autoregulatory
tention), assessed by O15 PET, was reduced in patients with adjustments in vascular resistance counteract the BP change
untreated hypertension.85 Furthermore, in patients with re- (Figure 2). In general, the faster the change in BP, the less ef-
paired aortic coarctation, in whom only pulse pressure was ficient is autoregulation.97
significantly elevated, the flow velocity increase produced The cellular bases of autoregulation rest, in part, on the
by photic stimulation in the posterior cerebral artery, which intrinsic property of the smooth muscle cells to constrict in
supplies the visual cortex, was suppressed.86 Consistent with response to increases in transmural pressure (the myogenic re-
altered neurovascular coupling, the increase in retinal blood sponse of Bayliss).96 Accordingly, if BP increases, the smooth
flow induced by light flickering was absent in individuals muscle cells constrict the vessels and increase vascular re-
with hypertension.87 These findings, albeit limited, support sistance, thereby preventing the CBF increase resulting from
the view that neurovascular uncoupling may compromise sub- the increased pressure. Conversely, decreases in BP relax
strate delivery to the brain and render the brain more vulnera- the smooth muscle, reducing vascular resistance and coun-
ble to vascular insufficiency. teracting the anticipated CBF decrease. Multiple redundant
Endothelial dysfunction has been studied extensively in pe- mechanisms underlie the myogenic response, including (1)
ripheral arteries and was found to precede the elevation in BP mechanosensors that modulate intracellular Ca2+ levels in re-
and, once developed, to correlate with its severity.88 Although sponse to changes in transmural pressure; selected candidates
direct assessment of cerebral endothelial function in humans is include: transient receptor potential channels98 and G-coupled
not feasible, inhibition of NO synthesis does not reduce retinal receptors, including AT1Rs (Ang II type-1 receptors)99 and,
1032  Circulation Research  March 29, 2019

possibly, tissue necrosis factor receptors100; (2) amplification carry solutes to the brain surface. In particular, a perivascular
of the Ca2+ signal by depolarization and opening of voltage- pathway has been proposed to travel retrogradely within the
gated Ca2+ channels leading to further Ca2+ entry, as well as arterial wall,112 whereas a paravascular glymphatic pathway,
intracellular Ca2+ release101; (3) modulation of the Ca2+ sensi- involving aquaporin-4 in astrocytic end-feet, reaches the brain
tivity of the contractile apparatus by regulating the balance be- surface through perivenous spaces.113 However, this field is
tween kinases and phosphatases responsible for myosin light still in evolution, and additional evidence is needed to assess
chain phosphorylation and involving Rho kinase and protein the relative weight of these clearance pathways in health and
kinase-C.102 The mechanisms regulating the vasodilatation disease. In the end, solutes drain into the cervical lymphnodes,
that occurs during BP lowering are less well understood. It probably through dural lymphatics.114 Cerebral blood vessels
has been suggested that the vasodilatation occurring with and perivascular spaces are also critical for the trafficking of
reduced BP may be an active phenomenon driven by meta- immune cells in and out of the brain required to maintain im-
bolic byproducts, such as adenosine, which accumulates as mune homeostasis and protect the brain from invading patho-
a result of the reduced tissue oxygen tension caused by the gens.115 Innate and adaptive immune cells enter the brain
hypoperfusion.103 through transvascular, meningeal, choroid plexus routes and
Hypertension results in the chronic adaptation of the ce- play a critical role in immune surveillance and in a wide va-
rebral circulation to higher levels of BP, such that the au- riety of pathologies.115 In particular, as described in the next
toregulated range of CBF is higher than in normotensive section, PVMs located in the perivascular space have emerged
individuals.104 The implication of this right shift of the curve as a critical source of reactive oxygen species (ROS) in hyper-
is that, if the BP is lowered to a level that would be safe in tension with a profound impact on neurovascular function.116
nonhypertensive individuals, the brain would be more suscep-
tible to hypoperfusion.95 The shift of the autoregulatory curve, How Does Hypertension Cause Neurovascular
first described in a baboon model105 and verified in a limited Dysfunction?
number of hypertensive individuals, has been attributed to the Several animal models have been used to investigate the
increase in vascular resistance induced by remodeling in ce- neurovascular effects of acute or chronic elevations in BP.117
rebral resistance vessels.95 More recent studies of dynamic Better-studied models involve administration of pressor doses
autoregulation have failed to report major alterations in the of Ang II, which induce acute elevations in BP, or administra-
moment-to-moment adjustments of CBF to BP changes.106–108 tion of subpressor doses (slow-pressor Ang II hypertension),
Rather, in patients with moderate hypertension (mean SBP, which produce delayed elevations in BP over several days. The
163±11 mm Hg), an enhancement of dynamic autoregulation slow-pressor model has gained popularity since is thought to
reflect the progressive increase in BP observed in essential hy-
Downloaded from http://ahajournals.org by on June 30, 2019

was found.109 Furthermore, lowering BP with antihypertensive


therapy did not reduce flow velocity108,109 but improved carotid pertension, although with a more compressed timescale (2–4
artery distensibility after 6 months of treatment.108 However, in weeks).118 In hypertension induced by acute or chronic Ang II
patients with malignant hypertension (SBP, 180–260 mm Hg), administration, there is attenuation of neurovascular coupling
dynamic autoregulation was altered and flow velocity fol- and endothelium-dependent responses.119–121 Interestingly, the
lowed passively the reduction in BP during treatment with so- mechanical effects of the elevation in transmural pressure are
dium nitroprusside,110 suggesting that severe disease can have not required for the cerebrovascular dysfunction, at least in
a profound impact on both static and dynamic autoregulation. the short term.119,120 Neurovascular dysfunction and cognitive
These observations, collectively, indicate that while hy- deficits have also been reported in other models of hyperten-
pertension may shift the autoregulatory range toward higher sion, including lifelong hypertension in SHR or mice (BPH
BPs, the compensatory adjustments taking place during BP [BP high] mice),74 intermittent hypoxia (a model of sleep ap-
variations within this range are not impaired, except in ma- nea),122 hypertension induced by administration of deoxycorti-
lignant hypertension. In addition, in moderate hypertension, costerone acetate+salt,123 eNOS inhibition, etc.117,124
lowering BP does not seem to lead to cerebral hypoperfu- The pathways mediating the neurovascular effects of hy-
sion. However, autoregulation data during the life course and pertension induced by slow-pressor Ang II administration
in the presence of vascular comorbidities are not available. have been studied in some detail and involve both central
Therefore, it remains unclear how magnitude and duration of effects on the brain and peripheral effects on cerebral blood
the BP elevation influence dynamic autoregulation and what is vessels. The evidence suggests that circulating Ang II acts on
the impact of antihypertensive therapy on CBF. AT1R in the subfornical organs—one of the circumventricular
organs—leading to ROS-mediated production of vasopressin
Perivascular Spaces, Clearance, Neuroimmune from the hypothalamic paraventricular nucleus. Vasopressin,
Trafficking in turn, acts on cerebral blood vessels to trigger local endo-
The brain vascular system and surrounding structures are also thelin-1 production which, in concert with circulating Ang
responsible for the clearance of waste products generated by II, contributes to the neurovascular dysfunction by inducing
brain activity, including toxic proteins, such as Aβ and tau.111 vascular oxidative stress.125 Evidence from systemic vessels
A transvascular pathway utilizes abluminal transporters that also indicates a critical role of vascular oxidative stress.126
carry solutes through the vascular wall and drain into the cir- However, how oxidative stress alters vascular function re-
culating blood.77 Other pathways are thought to use the per- mains to be defined. In Ang II hypertension, peroxynitrite—
ivascular space—a site of exchange between the interstitial the reaction product of NO and superoxide—is required for the
fluid (ISF) and the cerebrospinal fluid (CSF)—as a conduit to neurovascular dysfunction,127 but whether the effect depends
Iadecola and Gottesman   Hypertension and Cognition   1033

on reduction of NO bioavailability by the reaction with super- In summary, experimental studies and human data indi-
oxide, or on downstream effects of peroxynitrite, remains to cate that oxidative stress and inflammation are critical factors
be established. Evidence from the Ang II hypertension model in the alterations in neurovascular and endothelial function
indicates that the latter possibility is more likely because a produced by hypertension. Although other enzymatic sourc-
peroxynitrite decomposition catalyst rescues the neurovascu- es cannot be excluded, NOX2 has been identified as a major
lar dysfunction.127 Peroxynitrite could alter vascular function source of ROS in hypertension involving Ang II. Furthermore,
through several mechanisms.128 In the neurovascular dysfunc- innate immune cells, the PVM in particular, have emerged as
tion produced by Aβ, peroxynitrite-induced DNA damage ac- a powerful source of vascular ROS production.
tivates the DNA repair enzyme poly-ADP ribose polymerase
and leads poly-ADP ribose production, which, in turn, opens Brain Lesions Induced by Hypertension and
transient receptor potential melastatin-2 channels resulting in Their Impact on Cognition
endothelial Ca2+ overload and dysfunction.129 However, it re- The alterations in cerebrovascular structure and function in-
mains to be established whether this mechanism is also in- duced by hypertension described in the previous sections
volved in the neurovascular effects of Ang II hypertension. predispose the brain to dysfunction and damage, which, in
There are several potential enzymatic sources of vascu- turn, alters cognition. Chronic hypertension is the major risk
lar oxidative stress, including mainly mitochondrial com- factor for ischemic and hemorrhagic stroke, which is asso-
plexes, xanthine oxidase, and eNOS uncoupling—a condition ciated with a 3- to 6-fold increase in cognitive impairment,
in which the enzyme generates ROS instead of NO.130,131 especially when multiple strokes are involved (multi-infarct
However, a NOX-2 containing nicotinamide adenine dinucle- dementia).66 Additional lesions associated with hypertension
otide phosphate (NADPH) oxidase has emerged as a critical include microinfarcts and microhemorrhages, microscopic is-
source of the ROS involved in neurovascular dysfunction, es- chemic or hemorrhagic lesions most common in the cerebral
pecially in Ang II hypertension. NOX2 is present in vascu- cortex, which portend cognitive deficits.133–135 Hypertension is
lar and perivascular cells in close association with AT1R.74,120 also associated with enlarged perivascular spaces, which also
Furthermore, pharmacological inhibition or genetic deletion correlate with cognitive impairment.63 The expansion of the
of NOX2 rescues the neurovascular dysfunction in models perivascular spaces may be because of fluid stagnation and
of hypertension.120,121,125 Other isoforms of NOX, particularly protein accumulation reflecting impaired ISF/CSF clearance,
NOX1 and NOX4, may also present in the cerebral vascula- as well as the mechanical stresses on the perivascular tissue
ture, but their cellular localization and role in neurovascular by the increased pulsatility and stiffening.63 Of note, enlarged
dysfunction has not been elucidated.130 perivascular spaces can be confused with small cavitated le-
Downloaded from http://ahajournals.org by on June 30, 2019

As for the cellular source of ROS, endothelial cells, smooth sions (lacunae) on conventional brain imaging, and it has been
muscle cells, and pericytes have the potential to produce ROS. proposed that lacunes rather than enlarged perivascular spaces
However, recent data suggest that PVMs are a critical source correlate with cognitive impairment.136 The mechanisms of
of the ROS initiating the vascular dysfunction in Ang II hy- the brain atrophy induced by hypertension remain unclear,
pertension. PVMs are endowed with AT1R and, as phagocytic but (1) chronic hypoperfusion leading to neuronal loss,80 (2)
cells, express large amounts of NOX2 and have a greater po- retrograde neurodegeneration from deafferentation because
tential for ROS production than vascular cells.116 In slow-pres- of lesions in other brain regions,137 or (3) vascular oxidative
sor Ang II hypertension and in BPH mice—a genetic model stress and inflammation,138 leading to neurovascular trophic
of lifelong hypertension—PVM depletion prevents vascular failure,139 are potential contributing factors.
oxidative stress, neurovascular dysfunction, and cognitive The most common brain lesions associated with hyper-
impairment, as well as middle cerebral artery remodeling in tension are white matter lesions, especially in the frontal cor-
hypertensive rats.74,132 Using bone marrow transplantation to tex, which appear as areas of hyperintensity on T2-weighted
replace PVM with bone marrow–derived macrophages lack- MRI.140 SVD is considered the major cause of white matter
ing the AT1R or NOX2 abrogates vascular oxidative stress and disease, but the mechanisms have not been completely elu-
neurovascular dysfunction, pointing to PVM as the cellular cidated.141 Cerebral hypoxia-ischemia because of stenosis or
site of the AT1R and NOX2.74 Because in BPH mice and in occlusion of vessels feeding the white matter, which are par-
the slow-pressor model Ang II is elevated in the circulation ticularly susceptible to the effect of hypertension, is consid-
but not in brain, circulating Ang II was found to cross the BBB ered to be a major contributor.66 In support of this hypothesis,
and gain access to PVM on the abluminal side of the vessels reduction in CBF in normal-appearing white matter and ex-
(Figure 3).74 Although PVMs are unlikely to be the sole source pression of hypoxic markers have been reported.142,143 Other
of vascular oxidative stress, the observation that their elimina- studies have not supported this view144,145 and have suggested
tion or genetic modification blocks vascular ROS production that hypoperfusion is not the cause but the consequence of
suggests that they are the primary source, which may trigger the white matter lesion.146 Inflammation, upregulation of me-
oxidative chain reactions in adjacent vascular cells. Whether talloproteases, and increased BBB permeability in the white
PVMs are also involved in the neurovascular dysfunction in matter may lead to water shift and local edema resulting in
human hypertension remain to be established. However, as microvascular compression and occlusion.146,147 A link be-
discussed in the next sections, the observation of perivascular tween vascular health and white matter maintenance and
space abnormalities, vascular oxidative stress, and inflamma- repair by oligodendrocyte has also been established. A pop-
tion in SVD is consistent with findings emerged from animal ulation of oligodendrocytes associated specifically with cere-
models implicating PVM. bral blood vessels has been identified148 and oligodendrocyte
1034  Circulation Research  March 29, 2019

Figure 3. Putative mechanisms of neurovascular dysfunction in slow-pressor Ang II (angiotensin II) hypertension, BPH (blood pressure high) mice and
deoxycorticosterone acetate+salt hypertension. Circulating or brain Ang II reaches AT1R (angiotensin type-1 receptor) in perivascular macrophage (PVM)
wherein it activates NOX2 leading the vascular oxidative stress and neurovascular dysfunction. AT1R and Nox2 are also present in other vascular cells, but
they do not seem to play a primary role in these hypertension models. The cartoon depicts a penetrating arteriole, but brain macrophages are also present
in the meninges, wherein they may play a role in pial and meningeal vessel vasomotor function and permeability. Whether PVM has a similar role in models
Downloaded from http://ahajournals.org by on June 30, 2019

of Ang II–independent hypertension remains to be established. EC indicates endothelial cell; ROS, reactive oxygen species; and SMC, smooth muscle cell.
Illustration Credit: Ben Smith.

proliferation and survival requires vascular growth factors.149 connectivity between the anterior thalamus to the frontal cor-
These observations have suggested an oligovascular niche es- tex.134,152 Indeed, studies using white matter tractography and
sential for the maintenance of white matter health. Indeed, resting-state MRI have provided evidence for loss of connec-
arrested development of oligodendrocyte precursors has been tivity and network efficiency degradation, which correlate
observed human white matter in SVD resulting in faulty re- with a reduction in processing speed—a typical feature of vas-
myelination of damaged white matter (Figure 4).150 cular cognitive impairment.152 In addition, brain atrophy could
In summary, oxidative stress, hypoxia-ischemia, inflam- also be a contributing factor by reducing gray matter in critical
mation, and BBB dysfunction are critical vascular factors brain regions, such as hippocampus and prefrontal cortex.153
threatening the health of the subcortical white matter. There The impact of enlarged perivascular spaces on cognitive
might be regional differences on the impact of hypertension impairment of hypertension remains uncertain. The morpho-
on white matter, and the white matter of the frontal lobe may logical alterations of the perivascular space raise the possibil-
be more susceptible.134,140 However, the mechanistic bases for ity that perivascular and paravascular clearance systems may
such predilection remain to be established. be altered and have a role in the white matter damage.63,141
Perivascular spaces are enlarged and distorted in hyperten-
How Does Hypertension Cause Cognitive sion63—an alteration that could hamper the disposal of po-
Dysfunction? tentially toxic byproducts of brain activity. Hypertension is
Atrophy, brain damage caused by macroinfarcts and microin- associated with Aβ and tau pathology (see Hypertension and
farcts, and hemorrhages are important determinants of cogni- Alzheimer Pathology), suggesting that clearance of these
tive impairment. Furthermore, infarcts strategically placed in proteins may be reduced. Hypertension-induced large artery
brain regions involved in cognition, for example, hippocam- stiffening increases pulsatility in microvessels, which has
pus, medial thalamus, and frontal lobe, can produce cognitive been proposed to alter perivascular spaces and reduce ISF/
dysfunction, despite relatively small volume of damage,66,134 CSF clearance.63 However, a transient increase in pulsatil-
and the estimated functional impact of microinfarcts is greater ity induced by administration of dobutamine increases CSF
than anticipated by the volume of injury.151 The volume and clearance in mice,154 suggesting that hypertension could in-
location of white matter lesions also correlate with the degree crease ISF/CSF clearance. In support of this hypothesis, ISF
and temporal evolution of cognitive dysfunction.134 These flow is increased in SHR, but such increase is associated with
white matter lesions may affect cognition by impairing the greater retention of tracers at the ISF/CSF interface, indicating
Iadecola and Gottesman   Hypertension and Cognition   1035

Figure 5. Brain lesions produced by hypertension that underlie cognitive


impairment. Brain atrophy, microinfarcts, and microbleeds cause neuronal
loss and brain dysfunction. In addition, microinfarcts and microbleeds
disrupt brain connectivity and reduce network efficiency. Damage to
white matter lesions (WML) also degrades connectivity, especially
in thalamo-cortico networks. Alteration in perivascular spaces (PVS)
impairs brain clearance and may promote protein accumulation in brain
and vessels, and dysfunction of the neurovascular unit (NVU) leads to
vascular insufficiency and BBB damage. Potential interactions between
different pathogenic mechanisms are indicated by the dashed arrows. CBF
indicates cerebral blood flow.

Figure 4. Potential mechanisms of white matter (WM) damage by between AD pathology and hypertension in vivo, in an attempt
hypertension. Vascular oxidative stress and inflammation disrupt the to explore the nature, causal or incidental, of their association.
blood-brain barrier (BBB), induce neurovascular unit (NVU) dysfunction
and damage, and impair oligodendrocyte development and function. The
resulting alterations in tissue homeostasis, hypoxia-ischemia, reduced Clinical-Pathological Biomarkers of AD in
brain clearance, and impaired remyelination lead to WM damage. Hypertension
Although hypertension’s impact on cognition is fairly clearly
Downloaded from http://ahajournals.org by on June 30, 2019

a reduction in clearance.155 On the contrary, carotid ligation established, whether this acts directly on AD neuropathology
reduced CSF clearance,154 suggesting a negative impact of ce- or simply as a parallel contributor to cognitive impairment and
rebral hypoperfusion and reduced pulsatility, which is relevant dementia is still somewhat controversial.8
to large artery stenosis or occlusion often associated with hy- In the ARIC-PET study, nondemented participants with
pertension. Therefore, direct evidence linking hypertension to a greater number of vascular risk factors in middle age, in-
brain clearance pathways has not been provided, in humans as cluding hypertension, had more elevated brain amyloid in late
in animal models, and additional work is needed in this area. life, suggesting a possible direct impact of these risk factors
These observations, collectively, suggest that hypertension on amyloid; hypertension by itself, however, was not signifi-
may lead to cognitive impairment through several pathogenic cantly associated with elevated amyloid so may not have an
factors (Figure 5). Gray matter loss, loss of connectivity and independent role.157 Other studies have failed to find strong re-
network efficiency from white matter lesions, reduced peri- lationships between hypertension and brain amyloid, although
vascular clearance, and neurovascular dysfunction all have the many of these studied hypertension in older age. Elevated vas-
potential to impair brain function, but their relative impact on cular risk, including hypertension, was associated more with
cognitive processes remain to be established. atrophy and neurodegeneration than with brain amyloid, a-
mong 430 adults studied at >60 years of age with both tau and
Hypertension and Alzheimer Pathology amyloid PET, as well as brain MRI.158 In a neuropathologic
A large body of evidence indicates that hypertension is a risk sample, late-life BP was not associated with brain amyloid,
factor for AD—a condition once considered purely a neuro- but SBP was associated with neurofibrillary tangles.159
degenerative disease. AD is characterized neuropathologi- Using CSF biomarkers, the potential interaction of BP and
cally by extracellular accumulation of Aβ (amyloid plaques) APOE genotype was identified on tau levels, in one memory
and intraneuronal aggregates of hyperphosphorylated tau clinic cohort, in which hypertension was not associated with
(neurofibrillary tangles).2 Aβ is derived from the proteolytic amyloid but rather was related to tau among APOE-ε4 homo-
cleavage of the APP (amyloid precursor protein) by β- and zygotes.160 Although not specific for AD, associations between
γ-secretase enzymes.2 Vascular brain lesions similar to those hypertension and regional brain atrophy also support a poten-
associated with vascular cognitive impairment are also present tial role of elevated BP in Alzheimer neurodegeneration. In
in AD, such that mixed AD-vascular pathology is present in addition to the above-cited studies of elevated BP and brain
the majority of AD cases diagnosed clinically.156 The recent atrophy,42 midlife BP has also been associated with hippocam-
availability of imaging and biochemical biomarkers of AD pal atrophy by MRI, with the strongest associations among
has provided the opportunity to investigate the relationship untreated hypertensives.161
1036  Circulation Research  March 29, 2019

Figure 6. Potential mechanisms underlying the relationship between hypertension and Alzheimer disease (AD). The vascular damage produced by
hypertension leads to brain dysfunction through hypoxia-ischemia, as well as increase in Aβ (amyloid-β) production because of increased APP (amyloid
precursor protein) processing by secretase enzymes and reduced clearance of AD-related proteins. In turn, Aβ and tau alter vascular function amplifying the
deleterious vascular impact of hypertension. BBB indicates blood-brain barrier. Illustration Credit: Ben Smith. Adapted from Iadecola et al8 with permission.
Copyright ©2018, the American Heart Association.
Downloaded from http://ahajournals.org by on June 30, 2019

Association Between Hypertension and AD main kinases phosphorylating tau169—and consequently the
Pathology: Experimental Studies NO deficit associated with hypertension could promote tau
Several studies have suggested that experimental hyperten- phosphorylation. Central administration of Ang II induces tau
sion promotes Aβ accumulation and tau phosphorylation. phosphorylation by activation of the tau kinase GSK3β (glyco-
Hypertension, induced either by administration of Ang II,123 gen synthase kinase-3β) via AT1R-dependent mechanisms.170
aortic coarctation,162,163 renal artery stenosis,162 or in genetic These clinical and experimental observations, collectively,
models164 increases microvascular deposition of Aβ or neu- suggest that hypertension has the potential to promote AD pa-
ronal tau phosphorylation. At the same time, administration of thology by acting at different levels. However, it remains un-
renin-angiotensin system blockers or AT1R genetic deletion clear whether hypertension is a pathogenic factor in AD, and,
ameliorates amyloid deposition and behavioral dysfunction conversely, whether the AD pathology associated with hyper-
in APP-overexpressing mice.165,166 There are several mecha- tension is a contributor to the cognitive dysfunction.8
nisms by which hypertension could worsen AD pathology
(Figure 6). As discussed in a previous section, Aβ is cleared, Prevention and Therapy
in part, through vascular and perivascular pathways, and, Given the strong and consistent associations seen in the ep-
consequently, the vascular and perivascular dysfunction and idemiological literature between hypertension and dementia,
damage induced by hypertension could impair Aβ disposal and given the strong biological plausibility for a link between
the two, the inevitable next question is whether treatment of
leading to its accumulation. On the contrary, the loss of NO
hypertension reduces risk of dementia. Certainly, this is the
and prostacyclin bioavailability associated with vascular dys-
major reason why the study of hypertension as a risk factor for
function may also contribute to promote amyloidogenic APP
dementia is of particular interest: in the absence of other ways
processing and increase Aβ. Thus, NO reductions upregulate
to treat or prevent dementia and AD specifically, hypertension
APP and β-secretase, whereas reduction in prostacyclin may
is an especially appealing target.
favor amyloidogenic APP processing.167 Mediators of hyper-
tension may also promote amyloidogenic APP processing. For Does Treating Hypertension Reduce
example, Ang II increases APP cleavage by β- and γ-secretase Dementia Risk?
and increases Aβ production in vitro and in vivo.123,168 Less is Several epidemiological studies have considered the role of an-
known about a potential effect of hypertension on tau phos- tihypertensive medication treatment, although all remain sus-
phorylation. Endothelial NO prevents tau phosphorylation ceptible to some indication bias: individuals who are prescribed
by inhibiting Cdk5 (cyclin-dependent kinase-5)—one of the and take antihypertensives are different than people who do not,
Iadecola and Gottesman   Hypertension and Cognition   1037

in many ways beyond which can be adjusted statistically. Thus, Heart Study, hypertension was associated with worse decline
clinical trials would be the ideal forum in which to test this ques- in the presence of diabetes mellitus compared with its effect
tion, but given the above reviewed evidence that relationships in nondiabetics,178 and in the Framingham Offspring Study,
between hypertension and dementia are the strongest when hy- cognitive outcomes were the worst among participants with
pertension is considered in middle age, decades before the de- hypertension and elevated waist-hip ratio.179 In 1449 adults
velopment of dementia, and that clinical trials cannot randomize aged 65 to 79 years in Finland, the combination of SBP at
and follow participants for that long duration, some reliance on ≥160 mm Hg and elevated serum cholesterol, each in midlife,
longer term observational study designs is needed to consider was associated with a markedly increased risk of AD (odds ra-
these life span considerations. These studies also allow consid- tio, 3.5; 95% CI, 1.6–7.9), when compared with the presence
eration of age of treatment or duration of treatment, which can of either risk factor alone.19
extend beyond the windows allowed by clinical trials. Similar relationships are seen when consideration is made
In several observational studies, antihypertensive medica- of any additional risk factor: in 8945 participants of the Kaiser
tion use is associated with less cognitive decline; in the ARIC HMO (Health Maintenance Organization), an increasing num-
study, participants taking antihypertensive medications had ber of midlife vascular risk factors was associated with ele-
a 20-year cognitive decline equivalent to a prehypertensive vated risk of dementia in late life, with a hazard ratio of 1.27
group (higher than the normotensives but lower than untreated for individuals with only 1 midlife vascular risk factor and 2.37
hypertensives).12 In the EVA study group (Epidemiology of for 4 midlife vascular risk factors (considering smoking, hy-
Vascular Aging), treated hypertension was associated with less pertension, hypercholesterolemia, and diabetes mellitus).16 The
cognitive decline over only 4 years compared with untreated CAIDE score (Cardiovascular Risk Factors Aging and Incidence
hypertension.171 Furthermore, duration of antihypertensive of Dementia), which considers several vascular risk factors as a
therapy appears important in risk reduction. In the HAAS dementia risk score, including hypertension, has been reliably
study, per additional year of antihypertensive treatment, de- associated with dementia,180 with reasonably strong predictive
mentia risk was lower (hazard ratio, 0.94; 95% CI, 0.89–0.99), power (area under the curve, 0.74).181 In parallel, better Life’s
with the greatest reduction for individuals with treatment over Simple 7 scores, which consider optimal control of vascular (in-
12 years in duration, with overall risk nearing that of normo- cluding hypertension) and lifestyle risk factors, were associated
tensives.172 In the Rotterdam study, each additional year of an- with less cognitive decline, in the ARIC study.182
tihypertensive use, before age 75, was associated with an 8% As the primary genetic risk factor for AD, carriage of the
reduced risk of dementia; after age 75, the additional risk was APOE-ε4 allele independently increases risk for AD, but the
reduced and was no longer statistically significant.173 combination of the ε4 genotype and hypertension may fur-
Downloaded from http://ahajournals.org by on June 30, 2019

Trial data consists of several randomized clinical trials fo- ther increase risk of AD.183 In the Personality and Total Health
cusing primarily on antihypertensive therapy, with most trials Through Life study, the combination of an APOE-ε4 allele
to date showing negative results, with the exception of Syst- with hypertension was associated with greater decline over 8
Eur (Systolic Hypertension in Europe),174 and the most recent years in 1474 cognitively normal adults aged 60 to 64 years.184
SPRINT-MIND trial (Systolic Blood Pressure Intervention In ARIC-PET, as cited above, although no statistically signif-
Trial-Memory and Cognition In Decreased Hypertension).175 icant difference was found among APOE-ε4 carriers versus
These negative trials include those in individuals with a his- noncarriers for risk of elevated BP on late-life brain amyloid,
tory of stroke, in which a Cochrane review yielded a pooled there was a suggestion of more brain amyloid in individuals
relative risk of 0.88 (not significant) for the use of antihyper- with at least 1 ε4 allele, as midlife BP increased.157 This poten-
tensives to reduce dementia,176 as well as studies of individuals tial interaction was further supported by a study by Rodrigue
without prior known cerebrovascular disease.177 et al185 in which the combination of hypertension and at least
As one of the few positive clinical trials, the Syst-Eur trial 1 APOE-ε4 allele was associated with greater brain amyloid,
showed that dementia-free hypertensive individuals (SBP, by Pittsburgh compound-B PET, with a similar pattern with
160–219 mm Hg, and DBP, <95 mm Hg) aged ≥60 years who greater cortical thinning among individuals with both hyper-
received medications (a dihydropyridine calcium channel tension and at least 1 ε4 allele.186
blocker [CCB] plus other antihypertensive drugs as needed)
to lower BP to <150 mm Hg systolic, compared with placebo, Class-Specific Effects of Antihypertensive Drugs
had less incident dementia during a median follow-up of 2 There are conflicting data as to the potential role of distinct an-
years.174 The preliminary SPRINT-MIND data175 do suggest a tihypertensive medications: in HAAS, the only antihyperten-
benefit in progression of cerebral SVD for individuals treated sive drug category when considered at mean age of 77 years,
with intensive BP control versus standard control, as well as which was associated with reduced cognitive impairment, was
for cognitive outcomes, but only for the combination of MCI β-blockers (incidence rate ratio, 0.69; 95% CI, 0.50–0.94).187
and dementia and not for dementia itself. Syst-Eur primarily had a CCB-based intervention, and
showed reduction in dementia rates,174 and the PROGRESS
Importance of Other Comorbid Vascular Disease (Perindopril Protection Against Recurrent Stroke Study)
and Risk Factors showed a benefit in dementia reduction among individuals
Although the focus of this review is on hypertension, elevated with a history of stroke or transient ischemic attack who were
BP as a risk factor rarely occurs in isolation, and several risk treated with active therapy (perindopril [an ACE (angiotensin-
factors co-occurring in the same individuals may further in- converting enzyme) inhibitor] or indapamide [a diuretic]) for
crease accelerated cognitive decline. In the Framingham 3.9 years but only among individuals who also experienced
1038  Circulation Research  March 29, 2019

recurrent stroke.188 In the 3-City cohort study, participants involved in cognitive function, such as the hippocampus, en-
who took a combination of selective serotonin reuptake inhib- torinal cortex, and prefrontal cortex. A better understanding of
itors with CCBs, compared with selective serotonin reuptake the natural history of the impact of hypertension on cerebro-
inhibitors combined with other antihypertensives, had im- vascular function, network degradation, and cognition would
proved cognitive performance, as well as improved depression be needed to address this question, which is relevant to the
scores, at 2-year follow-up,189 with a similar beneficial effect initiation of antihypertensive therapy. In parallel, a more nu-
on cognitive decline from CCBs noted in the Newcastle 85+ anced understanding of the signaling mechanisms by which
Study of elderly adults, without a similar benefit from other the neurovascular dysfunction interferes with neuronal func-
antihypertensives.190 This is in direct contrast to findings from tion would also be desirable. Is CBF insufficiency the major
the Canadian Study of Health and Aging, in which Canadians factor? Or, are there other aspects of cerebrovascular biology,
aged ≥65 years who took CCBs had steeper decline, meas- such as trophic support by endothelial factors or perivascular
ured by a modified Mini-Mental State Examination repeated clearance, also at play?
serially for 5 years, than did their counterparts who used other
antihypertensive drugs (75% on CCBs declined versus 59% When Does Antihypertensive Therapy Need
on other antihypertensive drugs).191
to be Initiated to Minimize Risk of Cognitive
Impairment?
A growing body of evidence supports the biological im-
As summarized above, the benefits of antihypertensive ther-
portance of the renin-angiotensin system and points to the
apy are likely to be the greatest when initiated in midlife, and
potential importance of drugs within this family for demen-
continued over decades, although preliminary results from the
tia prevention in individuals with hypertension. Intervention
SPRINT-MIND trial suggest that benefits may still be possible
trials in SHR have shown reduced poststroke cognitive im-
with a shorter duration of antihypertensive therapy. In this re-
pairment in rodents treated with renin-angiotensin system
gard, it would be important to assess whether the appearance
modifiers (specifically candesartan—an AT1R blocker).192
of brain lesions diminishes the impact of hypertensive therapy
The ongoing HEART (History ECG Age Risk Factors and
on cognitive health. Although difficult to study in clinical tri-
Troponin) phase 1B randomized controlled trial will eval-
als, further consideration will need to be made of optimal age
uate the renin-angiotensin system specifically, in nonde-
and duration of antihypertensive therapy, and correlation with
mented adults at risk for AD, in which participants will be
cardiovascular and structural-functional imaging biomarkers,
randomized to placebo versus 1 of 2 doses of telmisartan.193
to best prevent cognitive decline, MCI, and dementia.
The optimal treatment regimen to prevent future cogni-
tive impairment or dementia is difficult to ascertain because Are There Individuals Especially Susceptible to the
Downloaded from http://ahajournals.org by on June 30, 2019

nearly all antihypertensives have studies supporting their po- Cognitive Effects of Hypertension and in Whom
tential benefit. Again, these are still likely confounded by More Aggressive Therapy Might Be Warranted?
indication bias, similar to the overall issue of indication bias Individuals with more vascular risk factors appear especially
regarding any antihypertensive use versus none, although to vulnerable to hypertension, with worse outcomes among indi-
a lesser degree (considering one antihypertensive versus an- viduals with several vascular risk factors. This suggests that
other may be influenced by comorbidities or demographic individuals with other known risk factors, such as diabetes
characteristics but are less profoundly impacted by socioec- mellitus, may need better screening of BP with a lower thresh-
onomic or other factors influencing access to medical care old for initiation of antihypertensive therapy. This is consist-
more broadly). ent with the management of vascular risk factors because they
relate to other cardiovascular outcomes. Beyond vascular
Outstanding Questions and Future Prospects risk, there may be genetic susceptibilities, such as has been
Although the body of knowledge relating hypertension to seen with the APOE data, leading to greater risk and, there-
cognitive outcomes, dementia, and AD has expanded tremen- fore, greater potential benefit from antihypertensive therapy,
dously in recent years, several key questions remain. Answers in individuals with a known risk allele or even of a particular
to some of these questions, listed below, will be critical to race or sex. Focused trials with enrollment of these higher risk
gain a better insight into how hypertension impacts cognitive groups could identify a subgroup in whom prevention might
function and to best recommend prevention and management be especially effective.
strategies.
What Class of Antihypertensives Is the Most
Is the Neurovascular Dysfunction Induced by Effective for Prevention of Cognitive Impairment?
Hypertension Sufficient to Cause Cognitive Although ongoing studies are directly addressing the potential
Impairment? benefit of particular antihypertensives, at least on surrogate
Hypertension induces alterations in neurovascular function, end points, at the time of this manuscript preparation, no con-
which are thought to induce brain lesions associated with vincing data point to a clinical difference with the use of par-
cognitive impairment (Figure 5). However, it remains to be ticular antihypertensive medications. This question—whether
established whether the neurovascular dysfunction alone any antihypertensive is as effective as the next—will be crit-
is sufficient to induced cognitive impairment. Reduced ce- ical as evidence is translated into practice. Furthermore, it is
rebral perfusion, alterations in brain clearance and BBB, as possible that the same recommended therapeutics may not be
well as vascular growth factor deficiency have the potential to ideal for all individuals. Race-based differences in therapeutic
alter neuronal function in metabolically active brain regions effect for distinct antihypertensives have been noted for other
Iadecola and Gottesman   Hypertension and Cognition   1039

cardiovascular outcomes,194 and similar differences by race, or References


other demographic or genetic factors, may be found for reduc- 1. Forouzanfar MH, Liu P, Roth GA, et al. Global burden of hypertension and
tion of adverse cognitive outcomes. systolic blood pressure of at least 110 to 115 mm Hg, 1990-2015. JAMA.
2017;317:165–182. doi: 10.1001/jama.2016.19043
What Are the Best Surrogate End Points or 2. Scheltens P, Blennow K, Breteler MM, de Strooper B, Frisoni GB, Salloway
S, Van der Flier WM. Alzheimer’s disease. Lancet. 2016;388:505–517.
Biomarkers for Measuring a Cerebral or Cognitive doi: 10.1016/S0140-6736(15)01124-1
Benefit in Treating Hypertension? 3. Spieth W. Cardiovascular health status, age, and psychological perfor-
Biomarkers or surrogate end points are valuable in studying mance. J Gerontol. 1964;19:277–284.
long-term relationships as seen between hypertension and 4. Wilkie F, Eisdorfer C. Intelligence and blood pressure in the aged. Science.
1971;172:959–962.
dementia because they allow for earlier assessment of thera- 5. Kety S, Hafkenschiel J, Jeffers W, Leopold I, Shenkin H. The blood flow,
peutic effect, both for research purposes, as well as clinically, vascular resistance, and oxygen consumption of the brain in essential hy-
to evaluate benefits of treatment. Ongoing studies and con- pertension. J Clin Invest. 1948;27:511–514.
6. Saklayen MG, Deshpande NV. Timeline of history of hypertension treat-
sortia, including MarkVCID (Mark Vascular Contributions to
ment. Front Cardiovasc Med. 2016;3:3. doi: 10.3389/fcvm.2016.00003
Cognitive Impairment and Dementia), are evaluating biomark- 7. Elias MF, Wolf PA, D’Agostino RB, Cobb J, White LR. Untreated
ers for the vascular contribution to cognitive impairment and blood pressure level is inversely related to cognitive functioning: the
dementia, which likely would be candidates for the specific Framingham Study. Am J Epidemiol. 1993;138:353–364.
8. Iadecola C, Gottesman RF. Cerebrovascular alterations in alzheimer disease.
effect of hypertension on the brain as well, and may include Circ Res. 2018;123:406–408. doi: 10.1161/CIRCRESAHA.118.313400
serum, CSF, or imaging markers to examine the contribution 9. Moser M, Roccella EJ. The treatment of hypertension: a remarka-
from AD pathology. ble success story. J Clin Hypertens (Greenwich). 2013;15:88–91. doi:
10.1111/jch.12033
Is There a Synergistic Relationship Between 10. Iadecola C, Yaffe K, Biller J, Bratzke LC, Faraci FM, Gorelick PB, Gulati
M, Kamel H, Knopman DS, Launer LJ, Saczynski JS, Seshadri S, Zeki
Hypertension and AD Pathology? Al Hazzouri A; American Heart Association Council on Hypertension;
As discussed in a previous section, there is biological plau- Council on Clinical Cardiology; Council on Cardiovascular Disease in
sibility that hypertension and other vascular risk factors may the Young; Council on Cardiovascular and Stroke Nursing; Council on
promote AD pathology and vice versa. However, the clinical- Quality of Care and Outcomes Research; and Stroke Council. Impact
of hypertension on cognitive function: a scientific statement from the
pathological evidence remains contradictory. The recent de- American Heart Association. Hypertension. 2016;68:e67–e94. doi:
velopment and validation of amyloid and tau PET imaging 10.1161/HYP.0000000000000053
provides the opportunity to examine in vivo the reciprocal 11. Gottesman RF, Rawlings AM, Sharrett AR, et al. Impact of differential
interaction between markers of AD and hypertensive neuro- attrition on the association of education with cognitive change over 20
years of follow-up: the ARIC neurocognitive study. Am J Epidemiol.
pathology and their relative contribution to cognitive impair-
Downloaded from http://ahajournals.org by on June 30, 2019

2014;179:956–966. doi: 10.1093/aje/kwu020


ment. These findings would have important implications for 12. Gottesman RF, Schneider AL, Albert M, Alonso A, Bandeen-Roche K,
targeting treatments to the relevant pathology responsible for Coker L, Coresh J, Knopman D, Power MC, Rawlings A, Sharrett AR,
Wruck LM, Mosley TH. Midlife hypertension and 20-year cognitive
the cognitive deficits in a particular individual. At the same change: the atherosclerosis risk in communities neurocognitive study.
time, in-depth investigations on the impact of AD and hyper- JAMA Neurol. 2014;71:1218–1227. doi: 10.1001/jamaneurol.2014.1646
tension on vessel-associated clearance pathways would be in- 13. Swan GE, DeCarli C, Miller BL, Reed T, Wolf PA, Jack LM, Carmelli D.
valuable to provide insight into the role of failure of Aβ and Association of midlife blood pressure to late-life cognitive decline and
brain morphology. Neurology. 1998;51:986–993.
tau disposal on disease onset and progression. 14. Launer LJ, Masaki K, Petrovitch H, Foley D, Havlik RJ. The association
These are a few of the outstanding questions that remain between midlife blood pressure levels and late-life cognitive function. The
to be addressed. Answering these questions will require en- Honolulu-Asia Aging Study. JAMA. 1995;274:1846–1851.
15. Kivipelto M, Helkala EL, Hänninen T, Laakso MP, Hallikainen M,

gaging both the preclinical and clinical scientific communi-
Alhainen K, Soininen H, Tuomilehto J, Nissinen A. Midlife vascular risk
ties in a concerted effort to advance the understanding of factors and late-life mild cognitive impairment: a population-based study.
how hypertension affects brain function leading to cognitive Neurology. 2001;56:1683–1689.
impairment. Thus, epidemiological, biomarker, and clini- 16. Whitmer RA, Sidney S, Selby J, Johnston SC, Yaffe K. Midlife car-
diovascular risk factors and risk of dementia in late life. Neurology.
cal-pathological studies should be closely related to basic 2005;64:277–281. doi: 10.1212/01.WNL.0000149519.47454.F2
science efforts to unravel mechanisms and provide new tar- 17. Freitag MH, Peila R, Masaki K, Petrovitch H, Ross GW, White

gets to be tested in clinical trials. Such cooperation will be LR, Launer LJ. Midlife pulse pressure and incidence of demen-
essential for the development of new diagnostic tools and tia: the Honolulu-Asia Aging Study. Stroke. 2006;37:33–37. doi:
10.1161/01.STR.0000196941.58869.2d
treatment strategies for one of the most devastating health 18. Gottesman RF, Albert MS, Alonso A, Coker LH, Coresh J, Davis SM,
challenges of our times. Deal JA, McKhann GM, Mosley TH, Sharrett AR, Schneider ALC,
Windham BG, Wruck LM, Knopman DS. Associations between midlife
vascular risk factors and 25-year incident dementia in the atherosclerosis
Sources of Funding risk in communities (ARIC) cohort. JAMA Neurol. 2017;74:1246–1254.
This study was supported by National Institutes of Health grants doi: 10.1001/jamaneurol.2017.1658
R37-NS089323 (C. Iadecola), R01-NS100447 (C. Iadecola), R01- 19. Kivipelto M, Helkala EL, Laakso MP, Hänninen T, Hallikainen M,

NS09544 (C. Iadecola), R01-NS/HL037853 (C. Iadecola), K24- Alhainen K, Soininen H, Tuomilehto J, Nissinen A. Midlife vascular
AG052573 (R.F. Gottesman), RF1-AG050745 (R.F. Gottesman), and risk factors and Alzheimer’s disease in later life: longitudinal, population
R01-AG040282 (R.F. Gottesman). based study. BMJ. 2001;322:1447–1451.
20. Goldstein FC, Levey AI, Steenland NK. High blood pressure and cognitive
decline in mild cognitive impairment. J Am Geriatr Soc. 2013;61:67–73.
Disclosures doi: 10.1111/jgs.12067
C. Iadecola serves on the Scientific Advisory Board of Broadview 21. Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/
Ventures. R.F. Gottesman is Associate Editor for the journal Neurology. ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for
1040  Circulation Research  March 29, 2019

the prevention, detection, evaluation, and management of high blood and risk of incident dementia. Neurology. 2017;89:2447–2454. doi:
pressure in adults: executive summary: a report of the American 10.1212/WNL.0000000000004741
College of Cardiology/American Heart Association task force on clin- 40. Guo Z, Viitanen M, Winblad B, Fratiglioni L. Low blood pressure and in-
ical practice guidelines. Hypertension. 2018;71:1269–1324. doi: cidence of dementia in a very old sample: dependent on initial cognition.
10.1161/HYP.0000000000000066 J Am Geriatr Soc. 1999;47:723–726.
22. Nation DA, Preis SR, Beiser A, Bangen KJ, Delano-Wood L, Lamar 41. Heijer Td, Skoog I, Oudkerk M, de Leeuw FE, de Groot JC, Hofman A,
M, Libon DJ, Seshadri S, Wolf PA, Au R. Pulse pressure is associ- Breteler MM. Association between blood pressure levels over time and
ated with early brain atrophy and cognitive decline: modifying ef- brain atrophy in the elderly. Neurobiol Aging. 2003;24:307–313.
fects of APOE-ε4. Alzheimer Dis Assoc Disord. 2016;30:210–215. doi: 42. Power MC, Schneider AL, Wruck L, Griswold M, Coker LH, Alonso A,
10.1097/WAD.0000000000000127 Jack CR Jr, Knopman D, Mosley TH, Gottesman RF. Life-course blood
23. Rawlings AM, Juraschek SP, Heiss G, Hughes T, Meyer ML, Selvin pressure in relation to brain volumes. Alzheimers Dement. 2016;12:890–
E, Sharrett AR, Windham BG, Gottesman RF. Association of ortho- 899. doi: 10.1016/j.jalz.2016.03.012
static hypotension with incident dementia, stroke, and cognitive decline. 43. Yaffe K, Vittinghoff E, Pletcher MJ, Hoang TD, Launer LJ, Whitmer
Neurology. 2018;91:e759–e768. doi: 10.1212/WNL.0000000000006027 R, Coker LH, Sidney S. Early adult to midlife cardiovascular risk fac-
24. Kilander L, Nyman H, Boberg M, Hansson L, Lithell H. Hypertension tors and cognitive function. Circulation. 2014;129:1560–1567. doi:
is related to cognitive impairment: a 20-year follow-up of 999 men. 10.1161/CIRCULATIONAHA.113.004798
Hypertension. 1998;31:780–786. 44. Østergaard SD, Mukherjee S, Sharp SJ, et al; Alzheimer’s Disease

25. Sabayan B, Wijsman LW, Foster-Dingley JC, Stott DJ, Ford I, Buckley Genetics Consortium; GERAD1 Consortium; EPIC-InterAct Consortium.
BM, Sattar N, Jukema JW, van Osch MJ, van der Grond J, van Buchem Associations between potentially modifiable risk factors and Alzheimer
MA, Westendorp RG, de Craen AJ, Mooijaart SP. Association of visit- disease: a mendelian randomization study. PLoS Med. 2015;12:e1001841;
to-visit variability in blood pressure with cognitive function in old age: discussion e1001841. doi: 10.1371/journal.pmed.1001841
prospective cohort study. BMJ. 2013;347:f4600. doi: 10.1136/bmj.f4600 45. Goldstein FC, Hajjar IM, Dunn CB, Levey AI, Wharton W. The relation-
26. Shah NS, Vidal JS, Masaki K, Petrovitch H, Ross GW, Tilley C,
ship between cognitive functioning and the JNC-8 guidelines for hyper-
DeMattos RB, Tracy RP, White LR, Launer LJ. Midlife blood pres- tension in older adults. J Gerontol A Biol Sci Med Sci. 2017;72:121–126.
sure, plasma β-amyloid, and the risk for Alzheimer disease: the doi: 10.1093/gerona/glw181
Honolulu Asia Aging Study. Hypertension. 2012;59:780–786. doi: 46. Smits LL, van Harten AC, Pijnenburg YA, Koedam EL, Bouwman FH,
10.1161/HYPERTENSIONAHA.111.178962 Sistermans N, Reuling IE, Prins ND, Lemstra AW, Scheltens P, van der
27. Shah RC, Wilson RS, Bienias JL, Arvanitakis Z, Evans DA, Bennett DA. Flier WM. Trajectories of cognitive decline in different types of dementia.
Relation of blood pressure to risk of incident Alzheimer’s disease and Psychol Med. 2015;45:1051–1059. doi: 10.1017/S0033291714002153
change in global cognitive function in older persons. Neuroepidemiology. 47. Sikaroodi H, Yadegari S, Miri SR. Cognitive impairments in patients with
2006;26:30–36. doi: 10.1159/000089235 cerebrovascular risk factors: a comparison of mini mental status exam and
28. Glynn RJ, Beckett LA, Hebert LE, Morris MC, Scherr PA, Evans DA. montreal cognitive assessment. Clin Neurol Neurosurg. 2013;115:1276–
Current and remote blood pressure and cognitive decline. JAMA. 1280. doi: 10.1016/j.clineuro.2012.11.026
1999;281:438–445. 48. Smith EE, Muzikansky A, McCreary CR, Batool S, Viswanathan A,

29. Ruitenberg A, Skoog I, Ott A, Aevarsson O, Witteman JC, Lernfelt B, van Dickerson BC, Johnson K, Greenberg SM, Blacker D. Impaired mem-
Harskamp F, Hofman A, Breteler MM. Blood pressure and risk of de- ory is more closely associated with brain beta-amyloid than leuko-
mentia: results from the Rotterdam study and the Gothenburg H-70 Study. araiosis in hypertensive patients with cognitive symptoms. PLoS One.
Downloaded from http://ahajournals.org by on June 30, 2019

Dement Geriatr Cogn Disord. 2001;12:33–39. doi: 10.1159/000051233 2018;13:e0191345. doi: 10.1371/journal.pone.0191345
30. Verghese J, Lipton RB, Hall CB, Kuslansky G, Katz MJ. Low blood 49. Reed BR, Mungas DM, Kramer JH, Ellis W, Vinters HV, Zarow C,

pressure and the risk of dementia in very old individuals. Neurology. Jagust WJ, Chui HC. Profiles of neuropsychological impairment in au-
2003;61:1667–1672. topsy-defined Alzheimer’s disease and cerebrovascular disease. Brain.
31. Waldstein SR, Giggey PP, Thayer JF, Zonderman AB. Nonlinear re-
2007;130:731–739. doi: 10.1093/brain/awl385
lations of blood pressure to cognitive function: the Baltimore 50. Iadecola C. The neurovascular unit coming of age: a journey through neu-
Longitudinal Study of Aging. Hypertension. 2005;45:374–379. doi: rovascular coupling in health and disease. Neuron. 2017;96:17–42. doi:
10.1161/01.HYP.0000156744.44218.74 10.1016/j.neuron.2017.07.030
32. Ninomiya T, Ohara T, Hirakawa Y, Yoshida D, Doi Y, Hata J, Kanba S, 51. Armulik A, Genové G, Betsholtz C. Pericytes: developmental, physio-
Iwaki T, Kiyohara Y. Midlife and late-life blood pressure and dementia in logical, and pathological perspectives, problems, and promises. Dev Cell.
Japanese elderly: the Hisayama study. Hypertension. 2011;58:22–28. doi: 2011;21:193–215. doi: 10.1016/j.devcel.2011.07.001
10.1161/HYPERTENSIONAHA.110.163055 52. Vanlandewijck M, He L, Mäe MA, et al. A molecular atlas of cell types
33. Yamada M, Kasagi F, Sasaki H, Masunari N, Mimori Y, Suzuki G.
and zonation in the brain vasculature. Nature. 2018;80:844–480.
Association between dementia and midlife risk factors: the Radiation 53. Hollander W, Prusty S, Kemper T, Rosene DL, Moss MB. The effects
Effects Research Foundation Adult Health Study. J Am Geriatr Soc. of hypertension on cerebral atherosclerosis in the cynomolgus monkey.
2003;51:410–414. Stroke. 1993;24:1218–1226; discussion 1226.
34. den Heijer T, Launer LJ, Prins ND, van Dijk EJ, Vermeer SE, Hofman 54. Agmon Y, Khandheria BK, Meissner I, Schwartz GL, Petterson TM,
A, Koudstaal PJ, Breteler MM. Association between blood pressure, O’Fallon WM, Gentile F, Whisnant JP, Wiebers DO, Seward JB.
white matter lesions, and atrophy of the medial temporal lobe. Neurology. Independent association of high blood pressure and aortic atherosclerosis:
2005;64:263–267. doi: 10.1212/01.WNL.0000149641.55751.2E a population-based study. Circulation. 2000;102:2087–2093.
35. Dufouil C, de Kersaint-Gilly A, Besançon V, Levy C, Auffray E,
55.
Qureshi AI, Caplan LR. Intracranial atherosclerosis. Lancet.
Brunnereau L, Alpérovitch A, Tzourio C. Longitudinal study of blood 2014;383:984–998. doi: 10.1016/S0140-6736(13)61088-0
pressure and white matter hyperintensities: the EVA MRI Cohort. 56. Hu X, De Silva TM, Chen J, Faraci FM. Cerebral vascular disease and
Neurology. 2001;56:921–926. neurovascular injury in ischemic stroke. Circ Res. 2017;120:449–471. doi:
36. Gottesman RF, Coresh J, Catellier DJ, Sharrett AR, Rose KM, Coker 10.1161/CIRCRESAHA.116.308427
LH, Shibata DK, Knopman DS, Jack CR, Mosley TH Jr. Blood pres- 57. Watase H, Sun J, Hippe DS, Balu N, Li F, Zhao X, Mani V, Fayad ZA,
sure and white-matter disease progression in a biethnic cohort: athero- Fuster V, Hatsukami TS, Yuan C. Carotid artery remodeling is seg-
sclerosis risk in communities (ARIC) study. Stroke. 2010;41:3–8. doi: ment specific: an in vivo study by vessel wall magnetic resonance
10.1161/STROKEAHA.109.566992 imaging. Arterioscler Thromb Vasc Biol. 2018;38:927–934. doi:
37. Stewart R, Xue QL, Masaki K, Petrovitch H, Ross GW, White
10.1161/ATVBAHA.117.310296
LR, Launer LJ. Change in blood pressure and incident dementia: 58. Kaess BM, Rong J, Larson MG, Hamburg NM, Vita JA, Levy D,

a 32-year prospective study. Hypertension. 2009;54:233–240. doi: Benjamin EJ, Vasan RS, Mitchell GF. Aortic stiffness, blood pressure
10.1161/HYPERTENSIONAHA.109.128744 progression, and incident hypertension. JAMA. 2012;308:875–881. doi:
38. Skoog I, Lernfelt B, Landahl S, Palmertz B, Andreasson LA, Nilsson L, 10.1001/2012.jama.10503
Persson G, Odén A, Svanborg A. 15-year longitudinal study of blood pres- 59. Jefferson AL, Cambronero FE, Liu D, Moore EE, Neal JE, Terry JG, Nair
sure and dementia. Lancet. 1996;347:1141–1145. S, Gifford KA, Hohman TJ, Bell SP, Wang TJ, Beckman JA, Carr JJ. Higher
39. McGrath ER, Beiser AS, DeCarli C, Plourde KL, Vasan RS,
aortic stiffness is related to lower cerebral blood flow and preserved cere-
Greenberg SM, Seshadri S. Blood pressure from mid- to late life brovascular reactivity in older adults. Circulation. 2018;138:1951–1962.
Iadecola and Gottesman   Hypertension and Cognition   1041

60. DuBose LE, Boles Ponto LL, Moser DJ, Harlynn E, Reierson L, Pierce 82. Muller M, van der Graaf Y, Visseren FL, Mali WP, Geerlings MI;
GL. Higher aortic stiffness is associated with lower global cerebrovas- SMART Study Group. Hypertension and longitudinal changes in cere-
cular reserve among older humans. Hypertension. 2018;72:476–482. doi: bral blood flow: the SMART-MR study. Ann Neurol. 2012;71:825–833.
10.1161/HYPERTENSIONAHA.118.11143 doi: 10.1002/ana.23554
61. Schnerr RS, Jansen JFA, Uludag K, Hofman PAM, Wildberger JE, van 83. Beason-Held LL, Moghekar A, Zonderman AB, Kraut MA, Resnick SM.
Oostenbrugge RJ, Backes WH. Pulsatility of lenticulostriate arteries as- Longitudinal changes in cerebral blood flow in the older hypertensive brain.
sessed by 7 tesla flow MRI-measurement, reproducibility, and applicability Stroke. 2007;38:1766–1773. doi: 10.1161/STROKEAHA.106.477109
to aging effect. Front Physiol. 2017;8:961. doi: 10.3389/fphys.2017.00961 84. Mentis MJ, Salerno J, Horwitz B, Grady C, Schapiro MB, Murphy DG,
62. Blanco PJ, Müller LO, Spence JD. Blood pressure gradients in cerebral Rapoport SI. Reduction of functional neuronal connectivity in long-term
arteries: a clue to pathogenesis of cerebral small vessel disease. Stroke treated hypertension. Stroke. 1994;25:601–607.
Vasc Neurol. 2017;2:108–117. doi: 10.1136/svn-2017-000087 85. Jennings JR, Muldoon MF, Ryan C, Price JC, Greer P, Sutton-Tyrrell K,
63. Brown R, Benveniste H, Black SE, Charpak S, Dichgans M, Joutel A, van der Veen FM, Meltzer CC. Reduced cerebral blood flow response and
Nedergaard M, Smith KJ, Zlokovic BV, Wardlaw JM. Understanding the compensation among patients with untreated hypertension. Neurology.
role of the perivascular space in cerebral small vessel disease. Cardiovasc 2005;64:1358–1365. doi: 10.1212/01.WNL.0000158283.28251.3C
Res. 2018;114:1462–1473. doi: 10.1093/cvr/cvy113 86. Wong R, Ahmad W, Davies A, Spratt N, Boyle A, Levi C, Howe P,
64. Marinković S, Milisavljević M, Puskas L. Microvascular anatomy of the Collins N. Assessment of cerebral blood flow in adult patients with
hippocampal formation. Surg Neurol. 1992;37:339–349. aortic coarctation. Cardiol Young. 2017;27:1606–1613. doi: 10.1017/
65. Blinder P, Tsai PS, Kaufhold JP, Knutsen PM, Suhl H, Kleinfeld D. S1047951117000920
The cortical angiome: an interconnected vascular network with non- 87. Delles C, Michelson G, Harazny J, Oehmer S, Hilgers KF,
columnar patterns of blood flow. Nat Neurosci. 2013;16:889–897. doi: Schmieder RE. Impaired endothelial function of the retinal vascu-
10.1038/nn.3426 lature in hypertensive patients. Stroke. 2004;35:1289–1293. doi:
66. Iadecola C. The pathobiology of vascular dementia. Neuron. 2013;80:844– 10.1161/01.STR.0000126597.11534.3b
866. doi: 10.1016/j.neuron.2013.10.008 88. Daiber A, Steven S, Weber A, Shuvaev VV, Muzykantov VR, Laher I, Li
67. Rosenblum WI. Fibrinoid necrosis of small brain arteries and arteri-
H, Lamas S, Münzel T. Targeting vascular (endothelial) dysfunction. Br
oles and miliary aneurysms as causes of hypertensive hemorrhage: J Pharmacol. 2017;174:1591–1619. doi: 10.1111/bph.13517
a critical reappraisal. Acta Neuropathol. 2008;116:361–369. doi: 89. Bagi Z, Brandner DD, Le P, McNeal DW, Gong X, Dou H, Fulton DJ,
10.1007/s00401-008-0416-9 Beller A, Ngyuen T, Larson EB, Montine TJ, Keene CD, Back SA.
68. Brown WR, Moody DM, Thore CR, Challa VR, Anstrom JA. Vascular de- Vasodilator dysfunction and oligodendrocyte dysmaturation in aging
mentia in leukoaraiosis may be a consequence of capillary loss not only white matter. Ann Neurol. 2018;83:142–152. doi: 10.1002/ana.25129
in the lesions, but in normal-appearing white matter and cortex as well. J 90. Muñoz Maniega S, Chappell FM, Valdés Hernández MC, Armitage PA,
Neurol Sci. 2007;257:62–66. doi: 10.1016/j.jns.2007.01.015 Makin SD, Heye AK, Thrippleton MJ, Sakka E, Shuler K, Dennis MS,
69. Aaslid R, Lindegaard KF, Sorteberg W, Nornes H. Cerebral autoregulation Wardlaw JM. Integrity of normal-appearing white matter: influence
dynamics in humans. Stroke. 1989;20:45–52. of age, visible lesion burden and hypertension in patients with small-
70. Iulita MF, Noriega de la Colina A, Girouard H. Arterial stiffness, cognitive vessel disease. J Cereb Blood Flow Metab. 2017;37:644–656. doi:
impairment and dementia: confounding factor or real risk? J Neurochem. 10.1177/0271678X16635657
2018;144:527–548. doi: 10.1111/jnc.14235 91. Hoth KF, Tate DF, Poppas A, Forman DE, Gunstad J, Moser DJ, Paul
71. Norlander AE, Madhur MS, Harrison DG. The immunology of hyperten- RH, Jefferson AL, Haley AP, Cohen RA. Endothelial function and white
Downloaded from http://ahajournals.org by on June 30, 2019

sion. J Exp Med. 2018;215:21–33. doi: 10.1084/jem.20171773 matter hyperintensities in older adults with cardiovascular disease.
72. Ogola BO, Zimmerman MA, Clark GL, Abshire CM, Gentry KM, Miller Stroke. 2007;38:308–312. doi: 10.1161/01.STR.0000254517.04275.3f
KS, Lindsey SH. New insights into arterial stiffening: does sex matter? Am 92. Nezu T, Hosomi N, Aoki S, Kubo S, Araki M, Mukai T, Takahashi T,
J Physiol Heart Circ Physiol. 2018;6:71. Maruyama H, Higashi Y, Matsumoto M. Endothelial dysfunction is asso-
73. Lim HY, Lim SY, Tan CK, et al. Hyaluronan receptor LYVE-1-expressing ciated with the severity of cerebral small vessel disease. Hypertens Res.
macrophages maintain arterial tone through hyaluronan-mediated reg- 2015;38:291–297. doi: 10.1038/hr.2015.4
ulation of smooth muscle cell collagen. Immunity. 2018;49:1191. doi: 93. Kearney-Schwartz A, Rossignol P, Bracard S, Felblinger J, Fay R,
10.1016/j.immuni.2018.12.009 Boivin JM, Lecompte T, Lacolley P, Benetos A, Zannad F. Vascular
74. Faraco G, Sugiyama Y, Lane D, Garcia-Bonilla L, Chang H, Santisteban structure and function is correlated to cognitive performance and
MM, Racchumi G, Murphy M, Van Rooijen N, Anrather J, Iadecola C. white matter hyperintensities in older hypertensive patients with
Perivascular macrophages mediate the neurovascular and cognitive dys- subjective memory complaints. Stroke. 2009;40:1229–1236. doi:
function associated with hypertension. J Clin Invest. 2016;126:4674– 10.1161/STROKEAHA.108.532853
4689. doi: 10.1172/JCI86950 94. Castro P, Azevedo E, Sorond F. Cerebral autoregulation in stroke. Curr
75. Longden TA, Dabertrand F, Koide M, Gonzales AL, Tykocki NR, Brayden Atheroscler Rep. 2018;20:37. doi: 10.1007/s11883-018-0739-5
JE, Hill-Eubanks D, Nelson MT. Capillary K+-sensing initiates retrograde 95. Paulson OB, Strandgaard S, Edvinsson L. Cerebral autoregulation.
hyperpolarization to increase local cerebral blood flow. Nat Neurosci. Cerebrovasc Brain Metab Rev. 1990;2:161–192.
2017;20:717–726. doi: 10.1038/nn.4533 96. Lassen NA. Cerebral blood flow and oxygen consumption in man.
76. Vanhoutte PM, Shimokawa H, Feletou M, Tang EH. Endothelial dysfunc- Physiol Rev. 1959;39:183–238. doi: 10.1152/physrev.1959.39.2.183
tion and vascular disease - a 30th anniversary update. Acta Physiol (Oxf). 97. Tan CO. Defining the characteristic relationship between arterial pres-
2017;219:22–96. doi: 10.1111/apha.12646 sure and cerebral flow. J Appl Physiol (1985). 2012;113:1194–1200. doi:
77. Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-brain 10.1152/japplphysiol.00783.2012
barrier: from physiology to disease and back. Physiol Rev. 2019;99:21–78. 98. Longden TA, Hill-Eubanks DC, Nelson MT. Ion channel networks in the
doi: 10.1152/physrev.00050.2017 control of cerebral blood flow. J Cereb Blood Flow Metab. 2016;36:492–
78. Marie C, Pedard M, Quirié A, Tessier A, Garnier P, Totoson P, Demougeot 512. doi: 10.1177/0271678X15616138
C. Brain-derived neurotrophic factor secreted by the cerebral endothelium: 99. Mederos y Schnitzler M, Storch U, Meibers S, Nurwakagari P, Breit
A new actor of brain function? J Cereb Blood Flow Metab. 2018;38:935– A, Essin K, Gollasch M, Gudermann T. Gq-coupled receptors as
949. doi: 10.1177/0271678X18766772 mechanosensors mediating myogenic vasoconstriction. EMBO J.
79. Tran KA, Zhang X, Predescu D, Huang X, Machado RF, Göthert JR, 2008;27:3092–3103.
Malik AB, Valyi-Nagy T, Zhao YY. Endothelial β-catenin signaling is 100. Kroetsch JT, Levy AS, Zhang H, Aschar-Sobbi R, Lidington D,

required for maintaining adult blood-brain barrier integrity and cen- Offermanns S, Nedospasov SA, Backx PH, Heximer SP, Bolz SS.
tral nervous system homeostasis. Circulation. 2016;133:177–186. doi: Constitutive smooth muscle tumour necrosis factor regulates micro-
10.1161/CIRCULATIONAHA.115.015982 vascular myogenic responsiveness and systemic blood pressure. Nat
80. Dai W, Lopez OL, Carmichael OT, Becker JT, Kuller LH, Gach HM. Commun. 2017;8:14805. doi: 10.1038/ncomms14805
Abnormal regional cerebral blood flow in cognitively normal eld- 101. Knot HJ, Nelson MT. Regulation of arterial diameter and wall [Ca2+] in
erly subjects with hypertension. Stroke. 2008;39:349–354. doi: cerebral arteries of rat by membrane potential and intravascular pressure.
10.1161/STROKEAHA.107.495457 J Physiol. 1998;508(pt 1):199–209.
81. Fujishima M, Ibayashi S, Fujii K, Mori S. Cerebral blood flow and brain 102. Jarajapu YP, Knot HJ. Relative contribution of Rho kinase and pro-
function in hypertension. Hypertens Res. 1995;18:111–117. tein kinase C to myogenic tone in rat cerebral arteries in hypertension.
1042  Circulation Research  March 29, 2019

Am J Physiol Heart Circ Physiol. 2005;289:H1917–H1922. doi: β-secretase activity, and leads to amyloidogenic processing of APP. J
10.1152/ajpheart.01012.2004 Cereb Blood Flow Metab. 2016;36:241–252. doi: 10.1038/jcbfm.2015.79
103. Kusano Y, Echeverry G, Miekisiak G, Kulik TB, Aronhime SN, Chen 124. Yang Y, Kimura-Ohba S, Thompson J, Rosenberg GA. Rodent models of
JF, Winn HR. Role of adenosine A2 receptors in regulation of cerebral vascular cognitive impairment. Transl Stroke Res. 2016;7:407–414. doi:
blood flow during induced hypotension. J Cereb Blood Flow Metab. 10.1007/s12975-016-0486-2
2010;30:808–815. doi: 10.1038/jcbfm.2009.244 125. Capone C, Faraco G, Peterson JR, Coleman C, Anrather J, Milner TA,
104. Strandgaard S. Autoregulation of cerebral blood flow in hypertensive Pickel VM, Davisson RL, Iadecola C. Central cardiovascular circuits con-
patients. The modifying influence of prolonged antihypertensive treat- tribute to the neurovascular dysfunction in angiotensin II hypertension. J
ment on the tolerance to acute, drug-induced hypotension. Circulation. Neurosci. 2012;32:4878–4886. doi: 10.1523/JNEUROSCI.6262-11.2012
1976;53:720–727. 126. Touyz RM, Briones AM. Reactive oxygen species and vascular biology:
105. Strandgaard S, Jones JV, MacKenzie ET, Harper AM. Upper limit of ce- implications in human hypertension. Hypertens Res. 2011;34:5–14. doi:
rebral blood flow autoregulation in experimental renovascular hyperten- 10.1038/hr.2010.201
sion in the baboon. Circ Res. 1975;37:164–167. 127. Girouard H, Park L, Anrather J, Zhou P, Iadecola C. Cerebrovascular
106. Birns J, Jarosz J, Markus HS, Kalra L. Cerebrovascular reactivity
nitrosative stress mediates neurovascular and endothelial dysfunction in-
and dynamic autoregulation in ischaemic subcortical white matter duced by angiotensin II. Arterioscler Thromb Vasc Biol. 2007;27:303–
disease. J Neurol Neurosurg Psychiatry. 2009;80:1093–1098. doi: 309. doi: 10.1161/01.ATV.0000253885.41509.25
10.1136/jnnp.2009.174607 128. Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxyni-

107. Traon AP, Costes-Salon MC, Galinier M, Fourcade J, Larrue V. Dynamics trite in health and disease. Physiol Rev. 2007;87:315–424. doi:
of cerebral blood flow autoregulation in hypertensive patients. J Neurol 10.1152/physrev.00029.2006
Sci. 2002;195:139–144. 129. Park L, Wang G, Moore J, Girouard H, Zhou P, Anrather J, Iadecola
108. Lipsitz LA, Gagnon M, Vyas M, Iloputaife I, Kiely DK, Sorond F, C. The key role of transient receptor potential melastatin-2 chan-
Serrador J, Cheng DM, Babikian V, Cupples LA. Antihypertensive nels in amyloid-β-induced neurovascular dysfunction. Nat Commun.
therapy increases cerebral blood flow and carotid distensibility in hy- 2014;5:5318. doi: 10.1038/ncomms6318
pertensive elderly subjects. Hypertension. 2005;45:216–221. doi: 130. Ago T, Kuroda J, Kamouchi M, Sadoshima J, Kitazono T.

10.1161/01.HYP.0000153094.09615.11 Pathophysiological roles of NADPH oxidase/nox family proteins
109. Zhang R, Witkowski S, Fu Q, Claassen JA, Levine BD. Cerebral hemo- in the vascular system. -Review and perspective-. Circ J. 2011;75:
dynamics after short- and long-term reduction in blood pressure in mild 1791–1800.
and moderate hypertension. Hypertension. 2007;49:1149–1155. doi: 131. Santhanam AV, d’Uscio LV, Smith LA, Katusic ZS. Uncoupling of eNOS
10.1161/HYPERTENSIONAHA.106.084939 causes superoxide anion production and impairs NO signaling in the ce-
110. Immink RV, van den Born BJ, van Montfrans GA, Koopmans RP,
rebral microvessels of hph-1 mice. J Neurochem. 2012;122:1211–1218.
Karemaker JM, van Lieshout JJ. Impaired cerebral autoregulation in doi: 10.1111/j.1471-4159.2012.07872.x
patients with malignant hypertension. Circulation. 2004;110:2241–2245. 132. Pires PW, Girgla SS, McClain JL, Kaminski NE, van Rooijen N,

doi: 10.1161/01.CIR.0000144472.08647.40 Dorrance AM. Improvement in middle cerebral artery structure and
111. Tarasoff-Conway JM, Carare RO, Osorio RS, et al. Clearance sys-
endothelial function in stroke-prone spontaneously hypertensive rats
tems in the brain-implications for Alzheimer disease. Nat Rev Neurol. after macrophage depletion. Microcirculation. 2013;20:650–661. doi:
2015;11:457–470. doi: 10.1038/nrneurol.2015.119 10.1111/micc.12064
112. Morris AW, Sharp MM, Albargothy NJ, Fernandes R, Hawkes CA, 133. van Veluw SJ, Shih AY, Smith EE, Chen C, Schneider JA, Wardlaw JM,
Downloaded from http://ahajournals.org by on June 30, 2019

Verma A, Weller RO, Carare RO. Vascular basement membranes as path- Greenberg SM, Biessels GJ. Detection, risk factors, and functional con-
ways for the passage of fluid into and out of the brain. Acta Neuropathol. sequences of cerebral microinfarcts. Lancet Neurol. 2017;16:730–740.
2016;131:725–736. doi: 10.1007/s00401-016-1555-z doi: 10.1016/S1474-4422(17)30196-5
113. Nedergaard M. Neuroscience. Garbage truck of the brain. Science.
134. Biesbroek JM, Weaver NA, Biessels GJ. Lesion location and cognitive
2013;340:1529–1530. doi: 10.1126/science.1240514 impact of cerebral small vessel disease. Clin Sci (Lond). 2017;131:715–
114. Absinta M, Ha S-K, Nair G, Sati P, Luciano NJ, Palisoc M, Louveau A, 728. doi: 10.1042/CS20160452
Zaghloul KA, Pittaluga S, Kipnis J, Reich DS. Human and nonhuman 135. Akoudad S, Wolters FJ, Viswanathan A, de Bruijn RF, van der Lugt A,
primate meninges harbor lymphatic vessels that can be visualized nonin- Hofman A, Koudstaal PJ, Ikram MA, Vernooij MW. Association of ce-
vasively by mri. Elife. 2017;6:780. rebral microbleeds with cognitive decline and dementia. JAMA Neurol.
115. Engelhardt B, Vajkoczy P, Weller RO. The movers and shapers in
2016;73:934–943. doi: 10.1001/jamaneurol.2016.1017
immune privilege of the CNS. Nat Immunol. 2017;18:123–131. doi: 136. Benjamin P, Trippier S, Lawrence AJ, Lambert C, Zeestraten E, Williams
10.1038/ni.3666 OA, Patel B, Morris RG, Barrick TR, MacKinnon AD, Markus HS.
116. Faraco G, Park L, Anrather J, Iadecola C. Brain perivascular macro- Lacunar infarcts, but not perivascular spaces, are predictors of cognitive
phages: characterization and functional roles in health and disease. J Mol decline in cerebral small-vessel disease. Stroke. 2018;49:586–593. doi:
Med (Berl). 2017;95:1143–1152. doi: 10.1007/s00109-017-1573-x 10.1161/STROKEAHA.117.017526
117. Santisteban MM, Iadecola C. Hypertension, dietary salt, and cognitive 137. Seo SW, Lee JM, Im K, Park JS, Kim SH, Kim ST, Ahn HJ, Chin J,
impairment. J Cereb Blood Flow Metab. 2018;38:2112–2128. Cheong HK, Weiner MW, Na DL. Cortical thinning related to peri-
118. Reckelhoff JF, Romero JC. Role of oxidative stress in angiotensin-
ventricular and deep white matter hyperintensities. Neurobiol Aging.
induced hypertension. Am J Physiol Regul Integr Comp Physiol. 2012;33:1156–1167. doi: 10.1016/j.neurobiolaging.2010.12.003
2003;284:R893–R912. doi: 10.1152/ajpregu.00491.2002 138. Rosenberg GA. Extracellular matrix inflammation in vascular cogni-
119. Kazama K, Wang G, Frys K, Anrather J, Iadecola C. Angiotensin II tive impairment and dementia. Clin Sci (Lond). 2017;131:425–437. doi:
attenuates functional hyperemia in the mouse somatosensory cortex. 10.1042/CS20160604
Am J Physiol Heart Circ Physiol. 2003;285:H1890–H1899. doi: 139. Guo S, Kim WJ, Lok J, Lee SR, Besancon E, Luo BH, Stins MF, Wang
10.1152/ajpheart.00464.2003 X, Dedhar S, Lo EH. Neuroprotection via matrix-trophic coupling be-
120. Kazama K, Anrather J, Zhou P, Girouard H, Frys K, Milner TA, Iadecola tween cerebral endothelial cells and neurons. Proc Natl Acad Sci USA.
C. Angiotensin II impairs neurovascular coupling in neocortex through 2008;105:7582–7587. doi: 10.1073/pnas.0801105105
NADPH oxidase-derived radicals. Circ Res. 2004;95:1019–1026. doi: 140. Habes M, Sotiras A, Erus G, Toledo JB, Janowitz D, Wolk DA, Shou
10.1161/01.RES.0000148637.85595.c5 H, Bryan NR, Doshi J, Völzke H, Schminke U, Hoffmann W, Resnick
121. Girouard H, Park L, Anrather J, Zhou P, Iadecola C. Angiotensin II SM, Grabe HJ, Davatzikos C. White matter lesions: spatial heteroge-
attenuates endothelium-dependent responses in the cerebral microcir- neity, links to risk factors, cognition, genetics, and atrophy. Neurology.
culation through nox-2-derived radicals. Arterioscler Thromb Vasc Biol. 2018;91:e964–e975. doi: 10.1212/WNL.0000000000006116
2006;26:826–832. doi: 10.1161/01.ATV.0000205849.22807.6e 141. Joutel A, Chabriat H. Pathogenesis of white matter changes in cerebral
122. Capone C, Faraco G, Coleman C, Young CN, Pickel VM, Anrather J, small vessel diseases: beyond vessel-intrinsic mechanisms. Clin Sci
Davisson RL, Iadecola C. Endothelin 1-dependent neurovascular dys- (Lond). 2017;131:635–651. doi: 10.1042/CS20160380
function in chronic intermittent hypoxia. Hypertension. 2012;60:106– 142. Fernando MS, Simpson JE, Matthews F, Brayne C, Lewis CE, Barber R,
113. doi: 10.1161/HYPERTENSIONAHA.112.193672 Kalaria RN, Forster G, Esteves F, Wharton SB, Shaw PJ, O’Brien JT, Ince
123. Faraco G, Park L, Zhou P, Luo W, Paul SM, Anrather J, Iadecola C. PG; MRC Cognitive Function and Ageing Neuropathology Study Group.
Hypertension enhances Aβ-induced neurovascular dysfunction, promotes White matter lesions in an unselected cohort of the elderly: molecular
Iadecola and Gottesman   Hypertension and Cognition   1043

pathology suggests origin from chronic hypoperfusion injury. Stroke. biomarkers for Alzheimer’s disease. J Alzheimers Dis. 2010;20:1083–
2006;37:1391–1398. doi: 10.1161/01.STR.0000221308.94473.14 1090. doi: 10.3233/JAD-2010-091198
143. Sam K, Crawley AP, Conklin J, Poublanc J, Sobczyk O, Mandell
161. Korf ES, White LR, Scheltens P, Launer LJ. Midlife blood pressure and the
DM, Venkatraghavan L, Duffin J, Fisher JA, Black SE, Mikulis DJ. risk of hippocampal atrophy: the HonoluluAsiaAging Study. Hypertension.
Development of white matter hyperintensity is Ppreceded by reduced 2004;44:29–34. doi: 10.1161/01.HYP.0000132475.32317.bb
cerebrovascular reactivity. Ann Neurol. 2016;80:277–285. doi: 10.1002/ 162. Shih YH, Wu SY, Yu M, Huang SH, Lee CW, Jiang MJ, Lin PY, Yang TT,
ana.24712 Kuo YM. Hypertension accelerates Alzheimer’s disease-related pathol-
144. van der Veen PH, Muller M, Vincken KL, Hendrikse J, Mali WP, van ogies in pigs and 3xTg mice. Front Aging Neurosci. 2018;10:73. doi:
der Graaf Y, Geerlings MI; SMART Study Group. Longitudinal re- 10.3389/fnagi.2018.00073
lationship between cerebral small-vessel disease and cerebral blood 163. Carnevale D, Mascio G, D’Andrea I, Fardella V, Bell RD, Branchi I,
flow: the second manifestations of arterial disease-magnetic reso- Pallante F, Zlokovic B, Yan SS, Lembo G. Hypertension induces brain
nance study. Stroke. 2015;46:1233–1238. doi: 10.1161/STROKEAHA. β-amyloid accumulation, cognitive impairment, and memory dete-
114.008030 rioration through activation of receptor for advanced glycation end
145. Shi Y, Thrippleton MJ, Makin SD, Marshall I, Geerlings MI, de Craen products in brain vasculature. Hypertension. 2012;60:188–197. doi:
AJM, van Buchem MA, Wardlaw JM. Cerebral blood flow in small ves- 10.1161/HYPERTENSIONAHA.112.195511
sel disease: a systematic review and meta-analysis. J Cereb Blood Flow 164. Schreiber S, Drukarch B, Garz C, Niklass S, Stanaszek L, Kropf S,
Metab. 2016;36:1653–1667. doi: 10.1177/0271678X16662891 Bueche C, Held F, Vielhaber S, Attems J, Reymann KG, Heinze HJ,
146. Farrall AJ, Wardlaw JM. Blood-brain barrier: ageing and microvas-
Carare RO, Wilhelmus MM. Interplay between age, cerebral small vessel
cular disease–systematic review and meta-analysis. Neurobiol Aging. disease, parenchymal amyloid-β, and tau pathology: longitudinal stud-
2009;30:337–352. doi: 10.1016/j.neurobiolaging.2007.07.015 ies in hypertensive stroke-prone rats. J Alzheimers Dis. 2014;42(suppl
147. Maclullich AM, Ferguson KJ, Reid LM, Deary IJ, Starr JM, Seckl JR, 3):S205–S215. doi: 10.3233/JAD-132618
Bastin ME, Wardlaw JM. Higher systolic blood pressure is associated 165. Wang J, Ho L, Chen L, Zhao Z, Zhao W, Qian X, Humala N, Seror I,
with increased water diffusivity in normal-appearing white matter. Bartholomew S, Rosendorff C, Pasinetti GM. Valsartan lowers brain
Stroke. 2009;40:3869–3871. doi: 10.1161/STROKEAHA.109.547877 beta-amyloid protein levels and improves spatial learning in a mouse
148. Marques S, Zeisel A, Codeluppi S, et al. Oligodendrocyte heteroge- model of Alzheimer disease. J Clin Invest. 2007;117:3393–3402. doi:
neity in the mouse juvenile and adult central nervous system. Science. 10.1172/JCI31547
2016;352:1326–1329. doi: 10.1126/science.aaf6463 166. Royea J, Zhang L, Tong XK, Hamel E. Angiotensin IV receptors me-
149. Arai K, Lo EH. An oligovascular niche: cerebral endothelial cells pro- diate the cognitive and cerebrovascular benefits of losartan in a mouse
mote the survival and proliferation of oligodendrocyte precursor cells. J model of Alzheimer’s disease. J Neurosci. 2017;37:5562–5573. doi:
Neurosci. 2009;29:4351–4355. doi: 10.1523/JNEUROSCI.0035-09.2009 10.1523/JNEUROSCI.0329-17.2017
150. Back SA, Tuohy TM, Chen H, Wallingford N, Craig A, Struve J, Luo 167. d’Uscio LV, He T, Katusic ZS. Expression and processing of amyloid
NL, Banine F, Liu Y, Chang A, Trapp BD, Bebo BF Jr, Rao MS, Sherman precursor protein in vascular endothelium. Physiology (Bethesda).
LS. Hyaluronan accumulates in demyelinated lesions and inhibits oli- 2017;32:20–32. doi: 10.1152/physiol.00021.2016
godendrocyte progenitor maturation. Nat Med. 2005;11:966–972. doi: 168. Zhu D, Shi J, Zhang Y, Wang B, Liu W, Chen Z, Tong Q. Central angio-
10.1038/nm1279 tensin II stimulation promotes β amyloid production in Sprague Dawley
151. Summers PM, Hartmann DA, Hui ES, Nie X, Deardorff RL, McKinnon rats. PLoS One. 2011;6:e16037. doi: 10.1371/journal.pone.0016037
ET, Helpern JA, Jensen JH, Shih AY. Functional deficits induced by cor- 169. Austin SA, Katusic ZS. Loss of endothelial nitric oxide synthase

Downloaded from http://ahajournals.org by on June 30, 2019

tical microinfarcts. J Cereb Blood Flow Metab. 2017;37:3599–3614. doi: promotes p25 generation and tau phosphorylation in a murine
10.1177/0271678X16685573 model of Alzheimer’s disease. Circ Res. 2016;119:1128–1134. doi:
152. Carnevale L, D’Angelosante V, Landolfi A, Grillea G, Selvetella G, 10.1161/CIRCRESAHA.116.309686
Storto M, Lembo G, Carnevale D. Brain MRI fiber-tracking reveals 170. Tian M, Zhu D, Xie W, Shi J. Central angiotensin II-induced Alzheimer-
white matter alterations in hypertensive patients without damage at con- like tau phosphorylation in normal rat brains. FEBS Lett. 2012;586:3737–
ventional neuroimaging. Cardiovasc Res. 2018;361:878–811. 3745. doi: 10.1016/j.febslet.2012.09.004
153. Beauchet O, Celle S, Roche F, Bartha R, Montero-Odasso M, Allali G, 171. Tzourio C, Dufouil C, Ducimetière P, Alpérovitch A. Cognitive decline
Annweiler C. Blood pressure levels and brain volume reduction: a sys- in individuals with high blood pressure: a longitudinal study in the eld-
tematic review and meta-analysis. J Hypertens. 2013;31:1502–1516. doi: erly. EVA Study Group. Epidemiology of Vascular Aging. Neurology.
10.1097/HJH.0b013e32836184b5 1999;53:1948–1952.
154. Iliff JJ, Wang M, Zeppenfeld DM, Venkataraman A, Plog BA, Liao Y, 172. Peila R, White LR, Masaki K, Petrovitch H, Launer LJ. Reducing the
Deane R, Nedergaard M. Cerebral arterial pulsation drives paravas- risk of dementia: efficacy of long-term treatment of hypertension. Stroke.
cular CSF-interstitial fluid exchange in the murine brain. J Neurosci. 2006;37:1165–1170. doi: 10.1161/01.STR.0000217653.01615.93
2013;33:18190–18199. doi: 10.1523/JNEUROSCI.1592-13.2013 173. Haag MD, Hofman A, Koudstaal PJ, Breteler MM, Stricker BH.

155. Bedussi B, Almasian M, de Vos J, VanBavel E, Bakker EN. Paravascular Duration of antihypertensive drug use and risk of dementia: A prospec-
spaces at the brain surface: low resistance pathways for cerebro- tive cohort study. Neurology. 2009;72:1727–1734. doi: 10.1212/01.wnl.
spinal fluid flow. J Cereb Blood Flow Metab. 2018;38:719–726. doi: 0000345062.86148.3f
10.1177/0271678X17737984 174. Forette F, Seux ML, Staessen JA, et al. Prevention of dementia in ran-
156. Azarpazhooh MR, Avan A, Cipriano LE, Munoz DG, Sposato LA,
domised double-blind placebo-controlled Systolic Hypertension in
Hachinski V. Concomitant vascular and neurodegenerative pathologies Europe (Syst-Eur) trial. Lancet. 1998;352:1347–1351.
double the risk of dementia. Alzheimers Dement. 2018;14:148–156. doi: 175. The SPRINT MIND Investigators for the SPRINT Research Group,
10.1016/j.jalz.2017.07.755 Williamson JD, Pajewski NM, Auchus AP, et al. Effect of intensive vs
157. Gottesman RF, Schneider AL, Zhou Y, Coresh J, Green E, Gupta N, standard blood pressure control on probable dementia. JAMA. 2019;1–9.
Knopman DS, Mintz A, Rahmim A, Sharrett AR, Wagenknecht LE, doi: 10.1001/jama.2018.21442
Wong DF, Mosley TH. Association between midlife vascular risk factors 176. Zonneveld TP, Richard E, Vergouwen MD, Nederkoorn PJ, de Haan R,
and estimated brain amyloid deposition. JAMA. 2017;317:1443–1450. Roos YB, Kruyt ND. Blood pressure-lowering treatment for preventing
doi: 10.1001/jama.2017.3090 recurrent stroke, major vascular events, and dementia in patients with a
158. Vemuri P, Lesnick TG, Przybelski SA, Knopman DS, Lowe VJ,
history of stroke or transient ischaemic attack. Cochrane Database Syst
Graff-Radford J, Roberts RO, Mielke MM, Machulda MM, Petersen Rev. 2018;7:CD007858. doi: 10.1002/14651858.CD007858.pub2
RC, Jack CR Jr. Age, vascular health, and Alzheimer disease bio- 177. McGuiness B, Todd S, Passmore P, Bullock R. The effects of blood pres-
markers in an elderly sample. Ann Neurol. 2017;82:706–718. doi: sure lowering on development of cognitive impairment and dementia
10.1002/ana.25071 in patients without apparent prior cerebrovascular disease. Cochrane
159. Arvanitakis Z, Capuano AW, Lamar M, Shah RC, Barnes LL, Bennett Database Systemic Reviews. 2006;19:CD004034.
DA, Schneider JA. Late-life blood pressure association with cerebrovas- 178. Elias PK, Elias MF, D’Agostino RB, Cupples LA, Wilson PW, Silbershatz
cular and Alzheimer disease pathology. Neurology. 2018;91:e517–e525. H, Wolf PA. NIDDM and blood pressure as risk factors for poor cognitive
doi: 10.1212/WNL.0000000000005951 performance. The Framingham Study. Diabetes Care. 1997;20:1388–1395.
160. Kester MI, van der Flier WM, Mandic G, Blankenstein MA, Scheltens 179. Wolf PA, Beiser A, Elias MF, Au R, Vasan RS, Seshadri S. Relation
P, Muller M. Joint effect of hypertension and APOE genotype on CSF of obesity to cognitive function: importance of central obesity and
1044  Circulation Research  March 29, 2019

synergistic influence of concomitant hypertension. The Framingham 188. Tzourio C, Anderson C, Chapman N, Woodward M, Neal B,

Heart Study. Curr Alzheimer Res. 2007;4:111–116. MacMahon S, Chalmers J; PROGRESS Collaborative Group. Effects
180. Kivipelto M, Ngandu T, Laatikainen T, Winblad B, Soininen H,
of blood pressure lowering with perindopril and indapamide therapy
Tuomilehto J. Risk score for the prediction of dementia risk in 20 years on dementia and cognitive decline in patients with cerebrovascular di-
among middle aged people: a longitudinal, population-based study. sease. Arch Intern Med. 2003;163:1069–1075. doi: 10.1001/archinte.
Lancet Neurol. 2006;5:735–741. doi: 10.1016/S1474-4422(06)70537-3 163.9.1069
181. Virta JJ, Heikkilä K, Perola M, Koskenvuo M, Räihä I, Rinne JO, Kaprio 189. Tully PJ, Peters R, Pérès K, Anstey KJ, Tzourio C. Effect of SSRI and cal-
J. Midlife cardiovascular risk factors and late cognitive impairment. Eur cium channel blockers on depression symptoms and cognitive function
J Epidemiol. 2013;28:405–416. doi: 10.1007/s10654-013-9794-y in elderly persons treated for hypertension: three city cohort study. Int
182. González HM, Tarraf W, Harrison K, Windham BG, Tingle J, Alonso Psychogeriatr. 2018;30:1345–1354. doi: 10.1017/S1041610217002903
A, Griswold M, Heiss G, Knopman D, Mosley TH. Midlife cardio- 190. Peters R, Collerton J, Granic A, Davies K, Kirkwood T, Jagger C.
vascular health and 20-year cognitive decline: Atherosclerosis Risk in Antihypertensive drug use and risk of cognitive decline in the very
Communities Study results. Alzheimers Dement. 2018;14:579–589. doi: old: an observational study - the Newcastle 85+ Study. J Hypertens.
10.1016/j.jalz.2017.11.002 2015;33:2156–2164. doi: 10.1097/HJH.0000000000000653
183. Qiu C, Winblad B, Fastbom J, Fratiglioni L. Combined effects of APOE 191. Maxwell CJ, Hogan DB, Ebly EM. Calcium-channel blockers and cogni-
genotype, blood pressure, and antihypertensive drug use on incident AD. tive function in elderly people: results from the Canadian Study of Health
Neurology. 2003;61:655–660. and Aging. CMAJ. 1999;161:501–506.
184. Andrews S, Das D, Anstey KJ, Easteal S. Interactive effect of APOE gen- 192. Ahmed HA, Ishrat T, Pillai B, Fouda AY, Sayed MA, Eldahshan W,
otype and blood pressure on cognitive decline: the PATH Through Life Waller JL, Ergul A, Fagan SC. RAS modulation prevents progres-
study. J Alzheimers Dis. 2015;44:1087–1098. doi: 10.3233/JAD-140630 sive cognitive impairment after experimental stroke: a randomized,
185. Rodrigue KM, Rieck JR, Kennedy KM, Devous MD Sr, Diaz-Arrastia blinded preclinical trial. J Neuroinflammation. 2018;15:229. doi:
R, Park DC. Risk factors for β-amyloid deposition in healthy aging: 10.1186/s12974-018-1262-x
vascular and genetic effects. JAMA Neurol. 2013;70:600–606. doi: 193. Wharton W, Goldstein FC, Tansey MG, Brown AL, Tharwani

10.1001/jamaneurol.2013.1342 SD, Verble DD, Cintron A, Kehoe PG. Rationale and Design of
186. Rast P, Kennedy KM, Rodrigue KM, Robinson PRAW, Gross
the Mechanistic Potential of Antihypertensives in Preclinical
AL, McLaren DG, Grabowski T, Schaie KW, Willis SL. APOEε4 Alzheimer’s (HEART) Trial. J Alzheimers Dis. 2018;61:815–824. doi:
Genotype and Hypertension Modify 8-year Cortical Thinning: Five 10.3233/JAD-161198
Occasion Evidence from the Seattle Longitudinal Study. Cereb Cortex. 194. Wright JT Jr, Dunn JK, Cutler JA, Davis BR, Cushman WC, Ford CE,
2018;28:1934–1945. doi: 10.1093/cercor/bhx099 Haywood LJ, Leenen FH, Margolis KL, Papademetriou V, Probstfield
187. Gelber RP, Ross GW, Petrovitch H, Masaki KH, Launer LJ, White LR. JL, Whelton PK, Habib GB; ALLHAT Collaborative Research Group.
Antihypertensive medication use and risk of cognitive impairment: Outcomes in hypertensive black and nonblack patients treated with
the Honolulu-Asia Aging Study. Neurology. 2013;81:888–895. doi: chlorthalidone, amlodipine, and lisinopril. JAMA. 2005;293:1595–1608.
10.1212/WNL.0b013e3182a351d4 doi: 10.1001/jama.293.13.1595
Downloaded from http://ahajournals.org by on June 30, 2019

Vous aimerez peut-être aussi