You are on page 1of 8

Brief Review

Hypertension
A Harbinger of Stroke and Dementia
Giuseppe Faraco, Costantino Iadecola

D espite remarkable successes in prevention and treatment,


hypertension (HTN) remains a major cause of morbid-
ity and mortality worldwide.1 The brain is one of the preemi-
emerged as a potential risk factor for cardiovascular events
and cerebrovascular disease.6
Longstanding HTN induces deposition of collagen and
nent target organs in which high blood pressure is particularly fibronectin and elastin fragmentation, leading to an increase
damaging, contributing significantly to its burden of disease.1 in the stiffness of the wall of large arteries. Arterial stiffen-
HTN is the biggest risk factor for stroke, the second cause of ing is a good predictor of stroke and cognitive decline, and is
death worldwide and a major cause of long-term disability.2 associated with clinically silent brain lesions in patients with
HTN is also a leading risk factor for vascular cognitive impair- hypertension.3,7
ment (VCI),3 as well as Alzheimer disease (AD), the most
common cause of dementia in the elderly.3 Therefore, HTN is Atherosclerosis and Small Vessel Disease
involved in the pathogenesis of 2 major brain diseases: stroke HTN is a leading risk factor for atherosclerosis. A 10-mm Hg
and dementia. In this brief review, we first examine the role of increase in arterial pressure increases by 43% the odds of
HTN in the alterations in cerebrovascular structure and func- complex aortic atherosclerosis (protruding atheroma, ulcer-
tion underlying the pathological effects of HTN on the brain. ated plaques, mobiles debris), highly predictive of ischemic
Then, we examine the pathophysiological bases of these altera- strokes.8 Atherosclerotic lesions are also observed at sites of
tions, focusing on how high blood pressure promotes stroke turbulent flow, such as the carotid bifurcation and the verte-
and cognitive impairment. Finally, we discuss the impact of brobasilar system, and less frequently in intracranial arteries
preventive and therapeutic approaches to mitigate the deleteri- (Figure 2). A potential mechanism is related to vascular shear
ous effects of HTN on the brain. stress at those sites leading to expression of innate immunity
receptors on macrophages/monocytes and inflammation.9
Alterations in Cerebrovascular Structure Atherosclerotic plaques can cause stroke by releasing frag-
Downloaded from http://ahajournals.org by on October 13, 2022

Induced by HTN ments and leading to artery-to-artery embolism, or by rupture


Adaptive Changes: Hypertrophy and Remodeling and hemorrhage, resulting in acute cerebrovascular occlu-
Sustained elevations in blood pressure have profound effects sions (Figure 2).
on the structure of cerebral blood vessels, inducing adaptive HTN also promotes highly distinctive alterations in small
changes aimed at reducing the mechanical stress on the arte- arteries and arterioles supplying the deep hemispheric white
rial wall and protecting microvessels from pulsatile stress.4 In matter and basal ganglia, resulting in a condition known as
hypertrophic remodeling, the media thickens to encroach into small vessel disease (SVD; Figure 2). The susceptibility of
the lumen, resulting in increased media cross-sectional area these vessels to SVD may be related to their short linear path
and media/lumen ratio (Figure 1). The size of vascular smooth from larger vessels at the base of the brain, rendering them
muscle cells increases, and there is accumulation of extracel- more vulnerable to the mechanical stresses imposed by HTN.10
lular matrix proteins, such as collagen and fibronectin, in the The most common pathological substrates of SVD related to
vessel wall.5 In eutrophic remodeling, smooth muscle cells HTN is arteriolosclerosis.11 The pathological features of arte-
undergo a rearrangement that leads to a reduction in the outer riolosclerosis include loss of smooth muscle cells from the
and inner diameters, whereas media/lumen ratio is increased tunica media, deposits of fibro-hyaline material, narrowing of
and cross-sectional area is unaltered (Figure 1). In this case, the lumen, and thickening of the vessel wall (lipohyalinosis11;
the change in vascular smooth muscle cells size is negligi- Figures 1 and 2). In more advanced lesions, fibrinoid necrosis
ble, and the reduction in lumen is attributable to a reorgani- of the vessel wall facilitates the rupture of the vessel result-
zation of cellular and acellular material in the vascular wall, ing in microscopic hemorrhages or large hemorrhages typi-
accompanied by enhanced apoptosis in the outer regions of cally in basal ganglia or thalamus (Figure 2). In addition, HTN
the blood vessel.5 Remodeling is damaging because it reduces induced capillary rarefaction, which may also contribute to
the vessels’ lumen and increases vascular resistance, and has the associated brain lesions in the periventricular white matter.

Received July 17, 2013; first decision July 22, 2013; revision accepted August 2, 2013.
From the Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY.
Correspondence to Costantino Iadecola, Brain and Mind Research Institute, Weill Cornell Medical College, 407 E 61st St, RR-303, New York, NY
10065. E-mail coi2001@med.cornell.edu
(Hypertension. 2013;62:810-817.)
© 2013 American Heart Association, Inc.
Hypertension is available at http://hyper.ahajournals.org DOI: 10.1161/HYPERTENSIONAHA.113.01063

810
Faraco and Iadecola   Hypertension, Stroke, and Dementia   811

Figure 1. Effect of
hypertension (HTN) on cerebral
blood vessels. HTN induces
profound alterations of critical
regulatory mechanisms of the
cerebral circulation, which,
in concert with the structural
alterations, compromise the
blood supply to the brain and
increase the risk for stroke
and dementia. BBB indicates
blood–brain barrier; CBF,
cerebral blood flow; and ROS,
reactive oxygen species.

Alterations in Cerebrovascular Function brain. Next, we examine the major mechanisms regulating the
Induced by HTN cerebral circulation and their perturbation by HTN.
Downloaded from http://ahajournals.org by on October 13, 2022

The brain is highly dependent on an adequate delivery of oxy-


gen and glucose from the circulation, and cerebral blood flow Cerebrovascular Autoregulation
(CBF) reductions impair neuronal function and, if protracted, Cerebrovascular autoregulation buffers the cerebrovascular
induce brain damage.12 Consequently, cerebrovascular control effects of the wide fluctuations in arterial pressure that occur
mechanisms assure that the delivery of oxygen and glucose is during the activities of daily living. Autoregulation allows the
well matched to the energy demands of the cellular constitu- brain to maintain a steady blood flow between 60 and 150
ents of the brain. HTN induces profound alterations of these mm Hg of mean arterial pressure.13 Within this range, blood
regulatory mechanisms, which, in concert with the structural pressure increases result in constriction, and blood pressure
changes described above, compromise the blood supply to the decreases result in dilatation of cerebral resistance vessels,
maintaining CBF relatively constant. Cerebral autoregulation
depends on the intrinsic ability of vascular smooth muscle
cells to constrict when transmural pressure increases (myo-
genic tone). Myogenic tone depends on the concerted action of
ion channels on the vascular smooth muscle cells membrane,
as well as stretch activated receptors, resulting in increases
in intracellular Ca2+ or increases in the Ca2+ sensitivity of the
contractile apparatus, ultimately leading to phosphorylation of
contractile proteins and constriction.13

Regulation by Endothelial Cells


Cerebral endothelial cells have powerful effects on vascular
tone and regulate CBF by releasing vasodilators (NO, prostacy-
clin, bradykinin, etc) and vasoconstrictors (endothelin-1 [ET-1],
Figure 2. Ischemic brain lesions associated with hypertension endothelium-derived constrictor factor, etc).14 Endothelium-
(HTN). HTN induces atherosclerosis of major extracranial and
intracranial arteries, which could lead to major infarcts. HTN causes derived vasoactive factors participate in the maintenance of
both microbleeds and macrobleeds and is the main pathogenetic resting CBF and may play a role in coordinating the vasodi-
factor for small vessel disease responsible for lacunar infarcts, latation of intraparenchymal arterioles with that of upstream
white matter lesions (leukoaraiosis), and microinfarcts. The pial arteries, and in local adjustments of flow in response to
subcortical white matter at the boundary between different vascular
territories (middle cerebral artery [MCA] and anterior cerebral artery mechanical forces.12 Another important aspect of endothelial
[ACA]) is particularly susceptible to damage. cell function is the regulation of the blood–brain barrier (BBB).
812  Hypertension  November 2013

Cerebral endothelial cells have a low vesicular transport and hypertensive rats.22,23 The cerebrovascular dysfunction pre-
are linked to each other by tight junctions, which prevent the cedes the HTN induced by slow-pressor Ang II infusion and
entry of hydrophilic substances into the brain.15 Specialized persists beyond the elevation in blood pressure at the end
transport proteins on the endothelial cell membrane regulate of the infusion.21 Furthermore, doses of Ang II that do not
the bidirectional transfer of substances into and from the brain elevate blood pressure produce cerebrovascular alterations
parenchyma.15 The integrity of the BBB is vitally important to comparable with those observed with slow-pressor or pressor
maintain the homeostasis of the cerebral microenvironments, doses,21,24,25 suggesting that the cerebrovascular actions of Ang
which is a prerequisite for normal brain function. II are independent of the elevation in blood pressure.
Consistent with findings in animal models, the increase
Functional Hyperemia in CBF induced by brain activation is attenuated in patients
Functional hyperemia is a homeostatic mechanism by which with chronic HTN, and the cerebrovascular dysfunction cor-
the vascular delivery of oxygen and glucose and the removal relates with cognitive deficits.26 Furthermore, the increase in
of metabolites produced by brain activity are coupled to the retinal blood flow produced by light flickering, a model of
energy requirements of neurons and glia.12 Therefore, CBF is function hyperemia, is blunted in individuals with hyperten-
dynamically related to the level of neural activity of the differ- sion.27 Direct evidence of altered cerebrovascular endothelial
ent brain regions. A growing body of evidence indicates that responses in humans with HTN is lacking. But the NO syn-
neurons, glia, and cerebrovascular cells, acting as an integrated thase inhibitor L-NAME (NG-nitro-L-arginine methyl ester)
unit, mediate the increase in CBF produced by neural activ- does not reduce retinal blood flow in patients with HTN, a
ity.12,16 Neural activity induces the release of vasoactive media- finding consistent with NO-dependent endothelial dysfunc-
tors such as NO, prostanoids, carbon monoxide, cytochrome tion.27 These observations, collectively, indicate that HTN
p450 metabolites, H+, and K+ ions that act at different levels of disrupts key cerebrovascular control mechanisms aimed at
the cerebral vasculature to mediate the hemodynamic changes maintaining the energy homeostasis of the brain, which act in
underlying the CBF increase.12 Vasodilation of arterioles at the concert with the structural alterations of cerebral blood vessels
site of activation is accompanied by vasodilation of upstream described earlier to produce brain dysfunction and damage.
pial arteries that supply the activated area, and this coordinated
response is essential for increasing CBF efficiently.12 Key Role of Oxidative Stress in the Cerebrovascular
Effects of HTN
HTN and Cerebrovascular Dysfunction Several lines of evidence suggest that reactive oxygen spe-
HTN has profound effects on all aspects of the regulation of cies (ROS) are key mediators of the cerebrovascular damage
Downloaded from http://ahajournals.org by on October 13, 2022

CBF (Figure 1). HTN alters cerebrovascular autoregulation, produced by HTN. HTN promotes ROS production in cere-
leading to a shift of the pressure–flow relationship to the right, bral blood vessels and ROS scavenger counteracts the effects
such that higher pressures are needed to maintain the same of HTN on functional hyperemia and endothelial dysfunction,
level of cerebral perfusion (Figure 1). The lateral shift may including alterations of the BBB.21,24,25,28 In models of Ang II
be also associated with a reduction in resting CBF, resulting HTN, ROS production is elevated in brain regions regulat-
in a downward displacement of the curve17 (Figure 1). These ing cardiovascular function.22 Suppression of ROS in one of
alterations reduce cerebral perfusion at each level of blood these regions, the subfornical organ, prevents the alterations
pressure, compromising the ability of the brain to maintain in functional hyperemia and endothelium-dependent responses
adequate CBF in the face of hypotension or arterial occlusion. induced by slow-pressor administration of Ang II.22 In this
The mechanisms of the effects of HTN on autoregulation are model, the cerebrovascular dysfunction involves, in addition
not completely understood, but are likely to include a combi- to direct vascular effects of circulating Ang II, also activation
nation of effects on myogenic tone and on the changes in the of neurohumoral mechanisms leading to vasopressin release
mechanical characteristics of cerebral blood vessels induced and upregulation of ET-1 in cerebral blood vessels.22 ET-1, in
by remodeling and stiffening.4,13 These changes in autoregu- turn, leads to vascular ROS production through activation of
lation are particularly damaging to the periventricular white ET type A receptors.22 In contrast, the cerebrovascular altera-
matter, which is located at the boundary between different arte- tions induced by acute administration of pressor Ang II doses
rial territories18 and, as such, is most susceptible to hypoperfu- are entirely depended on a direct action of Ang II on cerebral
sion (Figure 2). Accordingly, the magnitude of autoregulatory blood vessels leading to oxidative and nitrosative stress.24,25
dysfunction induced by HTN correlates with the severity of Cerebrovascular ET-1, via ET type A receptor and ROS, is also
periventricular white matter injury.19 The HTN-induced auto- involved in the cerebrovascular effects of the HTN induced
regulatory impairment also leads to more severe brain damage by chronic intermittent hypoxia, a model of obstructive sleep
after arterial occlusion in stroke models, which may underlie apnea.29 Oxidative stress has also been implicated in other vas-
the increased susceptibility to large artery stroke. cular effects of HTN including vascular remodeling and inflam-
HTN also alters functional hyperemia and endothelial mation.30 The evidence for a role of ROS in the white matter
function. Attenuations in cerebrovascular responses to endo- damage produced by SVD is suggestive but limited. Increased
thelium-dependent vasodilators have been reported in rodent markers of oxidative stress have been described in white matter
models of chronic HTN.20–22 Similarly, functional hyper- lesions in autopsy studies.31 Furthermore, reduced circulating
emia, produced in the somatosensory cortex by activation of levels of NO metabolites, possibly reflecting increased oxida-
the facial whiskers, is attenuated in mice treated with slow- tive stress, have been reported in patients with white matter
pressor doses of angiotensin II (Ang II) and in spontaneously lesions detected by MRI.32 More studies are clearly needed.
Faraco and Iadecola   Hypertension, Stroke, and Dementia   813

Among the potential sources of ROS in cerebral blood ves- appearing periventricular white matter of patients with leuko-
sels,6 the enzyme nicotinamide adenine dinucleotide phosphate araiosis,38 and hypoxia inducible genes are expressed in white
(NADPH)-oxidase has emerged as a major contributor in the matter lesions.39 Endothelial dysfunction and BBB alterations
cerebrovascular effects of HTN.33 Importantly, cerebral blood produced by HTN could cause leakage of plasma proteins,
vessels have a greater capacity to produce NADPH oxidase– leading to oxidative stress, inflammation, and edema, which
derived ROS than systemic vessels.34 The cerebrovascular compresses the tissue and contributes to hypoperfusion and
dysfunction induced by Ang II is not observed in mice lack- demyelination.36 Hypoxia could induce production of metal-
ing Nox2, and NADPH inhibition abrogates the alterations in loproteases in oligodendrocyte precursors contributing to the
endothelium-dependent vasodilation and functional hyperemia BBB opening.31,40,41
induced by Ang II.24,25 Nox2-derived radicals are also involved
in the cerebrovascular remodeling induced by Ang II HTN.30 Microinfarcts
SVD is also associated with ischemic lesions not visible to the
Brain Lesions Underlying VCI naked eyes (microinfarcts).42 Cerebral microinfarcts are small
VCI includes a wide spectrum of cognitive alterations infarcts (<1 mm diameter) thought to be very common in the
caused by cerebrovascular factors, ranging from mild cog- elderly, although their detection in vivo is challenging because
nitive impairment affecting a single cognitive domain, of their small size.42 It is estimated that 1 or 2 microinfarcts in
such as executive function, to full blown vascular dementia routine postmortem examination suggest the presence of hun-
affecting multiple domains and impairing the activities of dreds of microinfarcts throughout the brain.43 Microinfarcts
daily living.3 Stroke is a major cause of cognitive impair- are common in patients with vascular or mixed dementia but
ment. Up to one third of patients with stroke have cognitive are an independent predictor of VCI.42 Number and the loca-
impairment within 3 months, and having a stroke doubles tion of the microinfarcts are major determinants of cognitive
the risk of dementia (poststroke dementia).3 Furthermore, dysfunction, the cortical location being more closely associ-
the risk of VCI is increased in patients with no history ated with dementia.42
of stroke in which imaging shows brain infarcts (silent
infarcts).3 A single stroke affecting a region important for Cerebral Microbleeds and Macrobleeds
cognition, like the thalamus or the frontal lobe, can lead SVD also produces hemorrhagic manifestations. Cerebral
to cognitive impairment (strategic-infarct dementia).3 VCI microbleeds are small perivascular hemorrages (2–10 mm)
and dementia can also result from multiple strokes destroy- that can be detected histologically at autopsy or in vivo using
ing large amounts of brain tissue (multi-infarct dementia).3 iron-sensitive MRI sequences.44 Microbleeds, which are
Downloaded from http://ahajournals.org by on October 13, 2022

However, SVD remains a major cause of VCI contributing observed in 10% to 20% of the elderly often in association
up to 45% of dementia cases.3 Next, we examine the neuro- with other SVD pathologies, are an independent predictor of
pathological alterations caused by cerebral SVD that have cognitive decline.44 HTN is a major risk factor of microbleeds,
been linked to VCI. which in this condition are typically located in the basal gan-
glia, thalamus, brain stem, and cerebellum.45 Microbleeds are
Lacunar Infarcts frequently associated also with cerebral amyloid angiopathy,
Lacunar infarcts, small (<20 mm in diameter) rounded lesion but in this case they tend to have a lobar distribution.44 HTN
most commonly found in the basal ganglia, are commonly is well known to cause large cerebral hemorrhages, typically
associated with SVD and are a strong predictor of VCI.35 They in the basal ganglia or thalamus (Figure 2). The relationship
have been attributed to acute occlusion of small perforat- between microbleeds and cerebral hemorrhages is not entirely
ing cerebral arteries (40–200 µm diameter) because of SVD clear, but microbleeds are associated with an increased risk of
pathology or, less likely, embolism from upstream vessels.36 brain hemorrhage.46
More recently, evidence of white matter BBB disruption has
been provided in patients with lacunes, raising the possibility HTN and AD
that BBB alterations are early pathogenic events that could AD and VCI are, respectively, the first and second most com-
lead to secondary ischemia and inflammation.36 Therefore, mon causes of dementia in the elderly.3 Although traditionally
multiple factors are likely to be involved in the pathogenesis considered separate pathologies, increasing evidence indicates
of lacunar infarcts. that AD and VCI share common pathogenetic mechanisms.16
Vascular risk factors, such as HTN, smoking, hyperlipidemia,
Diffuse White Matter Damage and diabetes mellitus, are also risk factors for AD.16 In par-
Another manifestation of SVD is diffuse white matter dam- ticular, midlife HTN doubles the risk of AD later in life and
age or leukoaraiosis, indicating a reduction in white matter accelerates the progression of the dementia.47 Up to 50% of
density.3,11 High systolic blood pressure precedes the devel- cases of dementia have mixed pathology featuring both vas-
opment of leukoaraiosis, and blood pressure lowering slows cular (SVD) and neurodegenerative lesions (amyloid plaques
down its progression.37 Often present in the periventricular and neurofibrillary tangles).48 Several studies have shown that
white matter, leukoaraiosis could result from hypoxia-hypo- HTN-induced lesions and AD may have an additive or syner-
perfusion. The periventricular white matter is thought to be gistic effect and produce a more severe cognitive impairment
more susceptible to hypoperfusion because it is located at the than either process alone.16
boundary between separate arterial territories18 (Figure 2). Several postmortem studies have also reported more ath-
Supporting this hypothesis, CBF is reduced in normal erosclerosis in cerebral vessels of AD patients compared with
814  Hypertension  November 2013

nondemented controls, a finding associated with increased These observations, collectively, suggest that HTN may
amyloid plaques and neurofibrillary tangles.49,50 Furthermore, promote the development of AD through several mechanisms
an increased amount of amyloid plaques and neurofibrillary (Figure 3). First, HTN could impair the vascular clearance
tangles have been reported in the brain of patients with hyper- of the peptide, enhancing Aβ accumulation in brain and ves-
tension.51 β-amyloid (Aβ), a peptide involved in the patho- sels. Second, HTN could increase the cleavage of Aβ from
genesis of AD, is elevated in the blood of patients with VCI the amyloid precursor protein. Both of these effects would
and could potentially contribute to vascular insufficiency and lead to increased Aβ concentration in the brain parenchyma
white matter injury.52,53 Furthermore, increases in diastolic and blood vessels, aggravating the attendant vascular and syn-
blood pressure in midlife are linked to an increase in AD risk aptic dysfunction. Speculations aside, our understanding of
and to a reduction in plasma Aβ decades later,54 suggesting human HTN and AD is far from complete and, consequently,
that the vascular dysfunction induced by HTN may inhibit the the relationship between these 2 conditions is even less well
vascular transport of brain Aβ into the plasma. HTN interacts understood. Nevertheless, because the pathological processes
with Apoε4 to promote amyloid deposition in healthy individ- underlying AD starts decades before their clinical manifesta-
uals, suggesting an interaction between vascular and genetic tion,64 HTN is likely to exert its effects in the presymptomatic
factors in increasing the susceptibility to AD.51 Patients with phase of the disease, stressing the need for early diagnosis and
HTN and elevated AD biomarkers have increased gray mat- blood pressure lowering.
ter atrophy, indicating that HTN may exacerbate gray matter
damage in AD.55 Treatment of HTN and Prevention of Stroke
Experimental studies also support an interaction between vand Dementia
HTN and Aβ. In mouse models of AD, Aβ attenuates func- The National High Blood Pressure Education Program of the
tional hyperemia and endothelium-dependent vasodilata- National Heart, Lung, and Blood Institute, introduced >40
tion through mechanisms involving Nox2-derived ROS.56–58 years ago, has led to remarkable advances in the public aware-
However, unlike Ang II, the vascular effects of Aβ require ness and treatment of HTN. This highly successful program is
the scavenger receptor and Aβ receptor CD36.59 It is con- credited with a 70% to 80% reduction in the morbidity attrib-
ceivable that HTN and Aβ have an additive or synergistic utable to HTN, mainly heart attack and stroke.1 The effect of
pathogenic effect on cerebrovascular function. Consistent HTN control on the incidence of stroke is indeed substantial,
with this hypothesis, brain deposition of Aβ is increased in and for each 10-mm Hg decrease in systolic blood pressure,
hypertensive mice,60 an effect attributed to inhibition of the there has been a one-third decrease in stroke risk.2
perivascular and transvascular clearance of Aβ secondary to The effects of HTN treatment on VCI have been more dif-
Downloaded from http://ahajournals.org by on October 13, 2022

vascular dysfunction and damage (Figure 3). HTN may also ficult to assess,65 and there is still a debate as to whether HTN
facilitate the transfer of Aβ from blood to brain through the control leads to a better cognitive outcome,10,65 except for the
receptor for advanced glycation products (RAGE).61 Cerebral prevention of poststroke dementia.3 Some studies have shown
hypoxia-ischemia, which can occur with HTN, can also facili- a cognitive benefit from blood pressure lowering especially
tate β-secretase–mediated cleavage of Aβ from its precursor in the younger of the old, for example, Perindopril pROtec-
protein and increases the brain Aβ burden.62,63 tion aGainst REcurrent Stroke (PROGRESS), Hypertension
in the Very Elderly (HYVET), Systolic Hypertension in
the Elderly (SHEP), Systolic Hypertension in Europe
(SYST-EUR), whereas other studies have not, for example,
Study on Cognition and Prognosis in the Elderly (SCOPE),
Action in Diabetes and Vascular Disease (ADVANCE), and
Prevention Regimen for Effectively Avoiding Second Strokes
(PRoFESS).10,65 No specific class of antihypertensive agents
has been consistently found to be more effective.3 Limitations
of all these studies have been short follow-up, low rates of
incident dementia, lack of accounting for cotreatments, and
heterogeneity of the cognitive assessment, among others.3,65
Despite the controversy, a recent American Heart Association
statement strongly recommends blood pressure lowering
in patients who have suffered a stroke (Class I, level of evi-
dence level B), and encourages treatment in the younger of the
elderly (Class IIa, level of evidence B).3 The degree of blood
pressure lowering remains uncertain, but there is evidence that
more aggressive lowering may lead to a greater improvement
Figure 3. Interaction between hypertension (HTN) and Alzheimer
disease. HTN increases the deposition of β-amyloid (Aβ) and in cerebral perfusion.17 The blood pressure threshold for start-
might aggravate the cerebrovascular dysfunction induced by ing treatment remains to be defined. Data from the Honolulu
Aβ. HTN could impair the vascular clearance of Aβ and increase Heart Program/Honolulu Asian Aging Study suggest that 17%
its cleavage from the amyloid precursor protein (APP). Both of of late-life dementia cases are attributable to midlife systolic
these effects would lead to increased Aβ concentration in the
brain parenchyma and blood vessels, aggravating the attendant pressure levels between 120 and 140 mm Hg.66 Similarly, a
vascular and synaptic dysfunction. progressive cognitive decline has been reported for blood
Faraco and Iadecola   Hypertension, Stroke, and Dementia   815

pressures between 120 and 140 mm Hg,67 arguing for starting reduction needed to maximize benefits and reduce risks.
treatment at prehypertensive blood pressure levels. Whatever the impact on cognition, however, the great benefits
Treatments targeting oxidative stress have long been consid- for general health afforded by blood pressure control justify
ered to treat HTN and related complications,68 but it has been early and aggressive intervention. Nonpharmacological strate-
difficult to develop agents that cross the BBB and effectively gies based on diet and exercise may be particularly valuable in
reduce oxidative stress in brain.69 Similarly, epidemiological younger patients facing treatment for a lifetime.
studies have not shown a beneficial impact of dietary antioxi-
dants on late-life dementia.70 NADPH oxidase inhibitors would Sources of Funding
be valuable, and there is great interest in developing agents The work was supported by National Institutes of Health grant
that could be used for cardiovascular and brain diseases.69 On HL96571 and Alzheimer’s Association grant ZEN-11-202707.
the other hand, the link between HTN and AD provides clues
to potential therapeutic interventions that could benefit both
Disclosures
None.
diseases. Calcium-channel antagonists and inhibitors of the
renin–angiotensin system increase Aβ removal from the brain
References
and protect against cognitive dysfunction in mouse models.71,72 1. Moser M, Roccella EJ. The treatment of hypertension: a remarkable suc-
The renin–angiotensin system has been of particular interest cess story. J Clin Hypertens (Greenwich). 2013;15:88–91.
in light of the observation that angiotensin-converting enzyme 2. Lawes CM, Bennett DA, Feigin VL, Rodgers A. Blood pressure and
stroke: an overview of published reviews. Stroke. 2004;35:1024.
activity is increased in patients with AD, and that this enzyme
3. Gorelick PB, Scuteri A, Black SE, et al; American Heart Association
may degrade Aβ, but there are many unresolved issues.72 Stroke Council, Council on Epidemiology and Prevention, Council on
Nevertheless, these preclinical observations raise the possibil- Cardiovascular Nursing, Council on Cardiovascular Radiology and
ity of using specific blood pressure–lowering agents in patients Intervention, and Council on Cardiovascular Surgery and Anesthesia.
Vascular contributions to cognitive impairment and dementia: a state-
with HTN at risk for AD. Furthermore, dietary interventions ment for healthcare professionals from the American Heart Association/
and exercise have been shown to be promising, and would also American Stroke Association. Stroke. 2011;42:2672–2713.
be valuable as a primary or adjuvant therapeutic approach.73 4. Scuteri A, Nilsson PM, Tzourio C, Redon J, Laurent S. Microvascular
brain damage with aging and hypertension: pathophysiological consider-
ation and clinical implications. J Hypertens. 2011;29:1469–1477.
Conclusions 5. Mulvany MJ. Small artery remodelling in hypertension. Basic Clin
The evidence reviewed in this article indicates that HTN, Pharmacol Toxicol. 2012;110:49–55.
despite resounding successes in controlling its impact on 6. Faraci FM. Protecting against vascular disease in brain. Am J Physiol
Heart Circ Physiol. 2011;300:H1566–H1582.
morbidity and mortality, remains one of most pervasive and 7. Henskens LH, Kroon AA, van Oostenbrugge RJ, Gronenschild EH, Fuss-
Downloaded from http://ahajournals.org by on October 13, 2022

devastating diseases with broad consequences for worldwide Lejeune MM, Hofman PA, Lodder J, de Leeuw PW. Increased aortic pulse
health. HTN has profound effects on the brain and contributes wave velocity is associated with silent cerebral small-vessel disease in
hypertensive patients. Hypertension. 2008;52:1120–1126.
in a substantive manner to stroke and dementia, highly preva-
8. Agmon Y, Khandheria BK, Meissner I, Schwartz GL, Petterson TM,
lent diseases projected to have an even greater public heath O’Fallon WM, Gentile F, Whisnant JP, Wiebers DO, Seward JB.
impact in decades to come because of population aging. Large Independent association of high blood pressure and aortic atherosclerosis:
and small cerebral vessels are key targets of HTN, resulting a population-based study. Circulation. 2000;102:2087–2093.
9. Sakamoto H, Aikawa M, Hill CC, Weiss D, Taylor WR, Libby P, Lee
in pathological alteration of the vascular wall, impairment of RT. Biomechanical strain induces class a scavenger receptor expres-
vital hemodynamic responses regulating cerebral perfusion, sion in human monocyte/macrophages and THP-1 cells: a potential
and disruption of BBB permeability leading to major altera- mechanism of increased atherosclerosis in hypertension. Circulation.
tions in the brain microenvironment. Although oxidative stress 2001;104:109–114.
10. Sörös P, Whitehead S, Spence JD, Hachinski V. Antihypertensive
and inflammation are important factors in the pathogenesis of treatment can prevent stroke and cognitive decline. Nat Rev Neurol.
these events, the cellular and molecular bases of the suscep- 2013;9:174–178.
tibility of the white matter to HTN and other cerebrovascu- 11. Pantoni L. Cerebral small vessel disease: from pathogenesis and clinical
characteristics to therapeutic challenges. Lancet Neurol. 2010;9:689–701.
lar risk factors have not been fully elucidated and represent a 12. Iadecola C. Neurovascular regulation in the normal brain and in
fruitful area of future research. The vascular changes induced Alzheimer’s disease. Nat Rev Neurosci. 2004;5:347–360.
by HTN not only increase the susceptibility of the brain to 13. Cipolla MJ. The Cerebral Circulation. San Rafael, CA: Morgan &
ischemic-hypoxic damage in vulnerable white matter regions Claypool Life Sciences; 2009.
14. Andresen J, Shafi NI, Bryan RM Jr. Endothelial influences on cerebrovas-
but also promote the expression of AD neuropathology. The cular tone. J Appl Physiol. 2006;100:318–327.
realization that HTN has an impact on AD represents a major 15. Neuwelt EA, Bauer B, Fahlke C, Fricker G, Iadecola C, Janigro D,
departure from traditional views on the pathogenesis of AD, Leybaert L, Molnár Z, O’Donnell ME, Povlishock JT, Saunders NR,
Sharp F, Stanimirovic D, Watts RJ, Drewes LR. Engaging neuroscience to
a disease in which vascular factors were thought not to play a
advance translational research in brain barrier biology. Nat Rev Neurosci.
role.16 However, our understanding of the interaction of HTN 2011;12:169–182.
with AD is rudimentary at best, and much needs to be learned 16. Iadecola C. The overlap between neurodegenerative and vascular factors
on the vascular biology of their respective effects on the cere- in the pathogenesis of dementia. Acta Neuropathol. 2010;120:287–296.
17. Tryambake D, He J, Firbank MJ, O’Brien JT, Blamire AM, Ford GA.
bral vasculature and resulting cognitive impact. Although cur- Intensive blood pressure lowering increases cerebral blood flow in older
rent approaches to treat HTN have dramatically reduced stroke subjects with hypertension. Hypertension. 2013;61:1309–1315.
incidence and mortality, the role of blood pressure control in 18. De Reuck J. The human periventricular arterial blood supply and the anat-
omy of cerebral infarctions. Eur Neurol. 1971;5:321–334.
the prevention of late-life dementia has been more difficult to
19. Matsushita K, Kuriyama Y, Nagatsuka K, Nakamura M, Sawada T, Omae
assess. Questions remain on the blood pressure threshold for T. Periventricular white matter lucency and cerebral blood flow autoregu-
therapy initiation, length of treatment, and the blood pressure lation in hypertensive patients. Hypertension. 1994;23:565–568.
816  Hypertension  November 2013

20. Didion SP, Sigmund CD, Faraci FM. Impaired endothelial function in 41. Rosenberg GA. Matrix metalloproteinases and their multiple roles in neu-
transgenic mice expressing both human renin and human angiotensino- rodegenerative diseases. Lancet Neurol. 2009;8:205–216.
gen. Stroke. 2000;31:760–764; discussion 765. 42. Smith EE, Schneider JA, Wardlaw JM, Greenberg SM. Cerebral microin-
21. Capone C, Faraco G, Park L, Cao X, Davisson RL, Iadecola C. The cere- farcts: the invisible lesions. Lancet Neurol. 2012;11:272–282.
brovascular dysfunction induced by slow pressor doses of angiotensin 43. Westover MB, Bianchi MT, Yang C, Schneider JA, Greenberg SM.
II precedes the development of hypertension. Am J Physiol Heart Circ Estimating cerebral microinfarct burden from autopsy samples.
Physiol. 2011;300:H397–H407. Neurology. 2013;80:1365–1369.
22. Capone C, Faraco G, Peterson JR, Coleman C, Anrather J, Milner TA, 44. Greenberg SM, Vernooij MW, Cordonnier C, Viswanathan A, Al-Shahi
Pickel VM, Davisson RL, Iadecola C. Central cardiovascular circuits con- Salman R, Warach S, Launer LJ, Van Buchem MA, Breteler MM;
tribute to the neurovascular dysfunction in angiotensin II hypertension. J Microbleed Study Group. Cerebral microbleeds: a guide to detection and
Neurosci. 2012;32:4878–4886. interpretation. Lancet Neurol. 2009;8:165–174.
23. Calcinaghi N, Wyss MT, Jolivet R, Singh A, Keller AL, Winnik S, 45. Fazekas F, Kleinert R, Roob G, Kleinert G, Kapeller P, Schmidt R,
Fritschy JM, Buck A, Matter CM, Weber B. Multimodal imaging in Hartung HP. Histopathologic analysis of foci of signal loss on gradient-
rats reveals impaired neurovascular coupling in sustained hypertension. echo T2*-weighted MR images in patients with spontaneous intracerebral
Stroke. 2013;44:1957–1964. hemorrhage: evidence of microangiopathy-related microbleeds. AJNR Am
24. Kazama K, Anrather J, Zhou P, Girouard H, Frys K, Milner TA, Iadecola J Neuroradiol. 1999;20:637–642.
C. Angiotensin II impairs neurovascular coupling in neocortex through 46. Greenberg SM, Eng JA, Ning M, Smith EE, Rosand J. Hemorrhage bur-
NADPH oxidase-derived radicals. Circ Res. 2004;95:1019–1026. den predicts recurrent intracerebral hemorrhage after lobar hemorrhage.
25. Girouard H, Park L, Anrather J, Zhou P, Iadecola C. Angiotensin II Stroke. 2004;35:1415–1420.
attenuates endothelium-dependent responses in the cerebral microcir- 47. Skoog I, Gustafson D. Update on hypertension and Alzheimer’s disease.
culation through nox-2-derived radicals. Arterioscler Thromb Vasc Biol. Neurol Res. 2006;28:605–611.
2006;26:826–832. 48. Schneider JA, Arvanitakis Z, Leurgans SE, Bennett DA. The neuropathol-
26. Jennings JR, Muldoon MF, Ryan C, Price JC, Greer P, Sutton-Tyrrell K, ogy of probable Alzheimer disease and mild cognitive impairment. Ann
van der Veen FM, Meltzer CC. Reduced cerebral blood flow response and Neurol. 2009;66:200–208.
compensation among patients with untreated hypertension. Neurology. 49. Roher AE, Kuo YM, Esh C, Knebel C, Weiss N, Kalback W, Luehrs
2005;64:1358–1365. DC, Childress JL, Beach TG, Weller RO, Kokjohn TA. Cortical and
27. Delles C, Michelson G, Harazny J, Oehmer S, Hilgers KF, Schmieder RE. leptomeningeal cerebrovascular amyloid and white matter pathology in
Impaired endothelial function of the retinal vasculature in hypertensive Alzheimer’s disease. Mol Med. 2003;9:112–122.
patients. Stroke. 2004;35:1289–1293. 50. Yarchoan M, Xie SX, Kling MA, Toledo JB, Wolk DA, Lee EB, Van
28. Zhang M, Mao Y, Ramirez SH, Tuma RF, Chabrashvili T. Angiotensin Deerlin V, Lee VM, Trojanowski JQ, Arnold SE. Cerebrovascular ath-
II induced cerebral microvascular inflammation and increased erosclerosis correlates with Alzheimer pathology in neurodegenerative
blood-brain barrier permeability via oxidative stress. Neuroscience. dementias. Brain. 2012;135(pt 12):3749–3756.
2010;171:852–858. 51. Rodrigue KM, Rieck JR, Kennedy KM, Devous MD Sr, Diaz-Arrastia R,
29. Capone C, Faraco G, Coleman C, Young CN, Pickel VM, Anrather J, Park DC. Risk factors for β-amyloid deposition in healthy aging: vascular
Davisson RL, Iadecola C. Endothelin 1-dependent neurovascular dysfunc- and genetic effects. JAMA Neurol. 2013;70:600–606.
tion in chronic intermittent hypoxia. Hypertension. 2012;60:106–113. 52. Gomis M, Sobrino T, Ois A, Millán M, Rodríguez-Campello A, Pérez de
30. De Silva TM, Faraci FM. Effects of angiotensin II on the cerebral circula- la Ossa N, Rodríguez-González R, Jiménez-Conde J, Cuadrado-Godia E,
Downloaded from http://ahajournals.org by on October 13, 2022

tion: role of oxidative stress. Front Physiol. 2012;3:484. Roquer J, Dávalos A. Plasma beta-amyloid 1-40 is associated with the
31. Back SA, Kroenke CD, Sherman LS, Lawrence G, Gong X, Taber diffuse small vessel disease subtype. Stroke. 2009;40:3197–3201.
EN, Sonnen JA, Larson EB, Montine TJ. White matter lesions 53. Park L, Zhou P, Koizumi K, El Jamal S, Previti ML, Van Nostrand WE, Carlson
defined by diffusion tensor imaging in older adults. Ann Neurol. G, Iadecola C. Brain and circulating levels of Aβ1-40 differentially contribute
2011;70:465–476. to vasomotor dysfunction in the mouse brain. Stroke. 2013;44:198–204.
32. Shibata H, Nabika T, Moriyama H, Masuda J, Kobayashi S. Correlation of 54. Shah NS, Vidal JS, Masaki K, Petrovitch H, Ross GW, Tilley C, DeMattos
NO metabolites and 8-iso-prostaglandin F2a with periventricular hyperin- RB, Tracy RP, White LR, Launer LJ. Midlife blood pressure, plasma
tensity severity. Arterioscler Thromb Vasc Biol. 2004;24:1659–1663. β-amyloid, and the risk for Alzheimer disease: the Honolulu Asia Aging
33. Iadecola C, Davisson RL. Hypertension and cerebrovascular dysfunction. Study. Hypertension. 2012;59:780–786.
Cell Metab. 2008;7:476–484. 55. Glodzik L, Mosconi L, Tsui W, de Santi S, Zinkowski R, Pirraglia E, Rich
34. Miller AA, Drummond GR, Schmidt HH, Sobey CG. NADPH oxidase KE, McHugh P, Li Y, Williams S, Ali F, Zetterberg H, Blennow K, Mehta
activity and function are profoundly greater in cerebral versus systemic P, de Leon MJ. Alzheimer’s disease markers, hypertension, and gray mat-
arteries. Circ Res. 2005;97:1055–1062. ter damage in normal elderly. Neurobiol Aging. 2012;33:1215–1227.
35. Jokinen H, Gouw AA, Madureira S, Ylikoski R, van Straaten EC, van 56. Niwa K, Younkin L, Ebeling C, Turner SK, Westaway D, Younkin S, Ashe
der Flier WM, Barkhof F, Scheltens P, Fazekas F, Schmidt R, Verdelho KH, Carlson GA, Iadecola C. Abeta 1-40-related reduction in functional
A, Ferro JM, Pantoni L, Inzitari D, Erkinjuntti T; LADIS Study Group. hyperemia in mouse neocortex during somatosensory activation. Proc
Incident lacunes influence cognitive decline: the LADIS study. Neurology. Natl Acad Sci U S A. 2000;97:9735–9740.
2011;76:1872–1878. 57. Iadecola C, Zhang F, Niwa K, Eckman C, Turner SK, Fischer E, Younkin
36. Wardlaw JM, Smith C, Dichgans M. Mechanisms of sporadic cere- S, Borchelt DR, Hsiao KK, Carlson GA. SOD1 rescues cerebral endothe-
bral small vessel disease: insights from neuroimaging. Lancet Neurol. lial dysfunction in mice overexpressing amyloid precursor protein. Nat
2013;12:483–497. Neurosci. 1999;2:157–161.
37. Verhaaren BF, Vernooij MW, de Boer R, Hofman A, Niessen WJ, van 58. Park L, Zhou P, Pitstick R, Capone C, Anrather J, Norris EH, Younkin L,
der Lugt A, Ikram MA. High blood pressure and cerebral white mat- Younkin S, Carlson G, McEwen BS, Iadecola C. Nox2-derived radicals con-
ter lesion progression in the general population. Hypertension. tribute to neurovascular and behavioral dysfunction in mice overexpressing the
2013;61:1354–1359. amyloid precursor protein. Proc Natl Acad Sci U S A. 2008;105:1347–1352.
38. O’Sullivan M, Lythgoe DJ, Pereira AC, Summers PE, Jarosz JM, Williams 59. Park L, Zhou J, Zhou P, Pistick R, El Jamal S, Younkin L, Pierce J,
SC, Markus HS. Patterns of cerebral blood flow reduction in patients with Arreguin A, Anrather J, Younkin SG, Carlson GA, McEwen BS, Iadecola
ischemic leukoaraiosis. Neurology. 2002;59:321–326. C. Innate immunity receptor CD36 promotes cerebral amyloid angiopa-
39. Fernando MS, Simpson JE, Matthews F, Brayne C, Lewis CE, Barber R, thy. Proc Natl Acad Sci U S A. 2013;110:3089–3094.
Kalaria RN, Forster G, Esteves F, Wharton SB, Shaw PJ, O’Brien JT, Ince 60. Gentile MT, Poulet R, Di Pardo A, Cifelli G, Maffei A, Vecchione C,
PG; MRC Cognitive Function and Ageing Neuropathology Study Group. Passarelli F, Landolfi A, Carullo P, Lembo G. Beta-amyloid deposition
White matter lesions in an unselected cohort of the elderly: molecular in brain is enhanced in mouse models of arterial hypertension. Neurobiol
pathology suggests origin from chronic hypoperfusion injury. Stroke. Aging. 2009;30:222–228.
2006;37:1391–1398. 61. Carnevale D, Mascio G, D’Andrea I, Fardella V, Bell RD, Branchi I,
40. Seo JH, Miyamoto N, Hayakawa K, Pham LD, Maki T, Ayata C, Pallante F, Zlokovic B, Yan SS, Lembo G. Hypertension induces brain
Kim KW, Lo EH, Arai K. Oligodendrocyte precursors induce early β-amyloid accumulation, cognitive impairment, and memory deteriora-
blood-brain barrier opening after white matter injury. J Clin Invest. tion through activation of receptor for advanced glycation end products in
2013;123:782–786. brain vasculature. Hypertension. 2012;60:188–197.
Faraco and Iadecola   Hypertension, Stroke, and Dementia   817

62. Sun X, He G, Qing H, Zhou W, Dobie F, Cai F, Staufenbiel M, Huang 68. Delles C, Miller WH, Dominiczak AF. Targeting reactive oxygen species
LE, Song W. Hypoxia facilitates Alzheimer’s disease pathogenesis in hypertension. Antioxid Redox Signal. 2008;10:1061–1077.
by up-regulating BACE1 gene expression. Proc Natl Acad Sci U S A. 69. Drummond GR, Selemidis S, Griendling KK, Sobey CG. Combating oxi-
2006;103:18727–18732. dative stress in vascular disease: NADPH oxidases as therapeutic targets.
63. Tesco G, Koh YH, Kang EL, Cameron AN, Das S, Sena-Esteves M, Nat Rev Drug Discov. 2011;10:453–471.
Hiltunen M, Yang SH, Zhong Z, Shen Y, Simpkins JW, Tanzi RE. 70. Laurin D, Masaki KH, Foley DJ, White LR, Launer LJ. Midlife dietary
Depletion of GGA3 stabilizes BACE and enhances beta-secretase activ- intake of antioxidants and risk of late-life incident dementia: the
ity. Neuron. 2007;54:721–737. Honolulu-Asia Aging study. Am J Epidemiol. 2004;159:959–967.
64. Bateman RJ, Xiong C, Benzinger TL, et al; Dominantly Inherited 71. Paris D, Bachmeier C, Patel N, Quadros A, Volmar CH, Laporte V,
Alzheimer Network. Clinical and biomarker changes in dominantly inher- Ganey J, Beaulieu-Abdelahad D, Ait-Ghezala G, Crawford F, Mullan MJ.
ited Alzheimer’s disease. N Engl J Med. 2012;367:795–804. Selective antihypertensive dihydropyridines lower Aβ accumulation by
65. Dichgans M, Zietemann V. Prevention of vascular cognitive impairment. targeting both the production and the clearance of Aβ across the blood-
Stroke. 2012;43:3137–3146. brain barrier. Mol Med. 2011;17:149–162.
66. Launer LJ, Hughes T, Yu B, Masaki K, Petrovitch H, Ross GW, White 72. Kehoe PG, Miners S, Love S. Angiotensins in Alzheimer’s disease—
LR. Lowering midlife levels of systolic blood pressure as a public health friend or foe? Trends Neurosci. 2009;32:619–628.
strategy to reduce late-life dementia: perspective from the Honolulu Heart 73. Smith PJ, Blumenthal JA, Babyak MA, Craighead L, Welsh-Bohmer
Program/Honolulu Asia Aging study. Hypertension. 2010;55:1352–1359. KA, Browndyke JN, Strauman TA, Sherwood A. Effects of the dietary
67. Knecht S, Wersching H, Lohmann H, Bruchmann M, Duning T, Dziewas approaches to stop hypertension diet, exercise, and caloric restric-
R, Berger K, Ringelstein EB. High-normal blood pressure is associated tion on neurocognition in overweight adults with high blood pressure.
with poor cognitive performance. Hypertension. 2008;51:663–668. Hypertension. 2010;55:1331–1338.
Downloaded from http://ahajournals.org by on October 13, 2022

You might also like