Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2023 Sep 6.
Published in final edited form as: Stroke. 2022 Mar 8;53(4):1054–1064. doi: 10.1161/STROKEAHA.121.035850

New Insights into Cerebrovascular Pathophysiology and Hypertension

Alastair JS Webb *,, David J Werring
PMCID: PMC7615037  EMSID: EMS143698  PMID: 35255709

Abstract

Despite advances in acute management and prevention of cerebrovascular disease, stroke and vascular cognitive impairment together remain the world’s leading cause of death and neurological disability. Hypertension and its consequences are associated with over 50% of ischaemic and 70% of haemorrhagic strokes but despite good control of blood pressure there remains a 10% risk of recurrent cerebrovascular events and there is no proven strategy to prevent vascular cognitive impairment.

Hypertension evolves over the life-span, from predominant sympathetically-driven hypertension with elevated mean blood pressure (BP) in early and mid-life to a late-life phenotype of increasing systolic and falling diastolic pressures, associated with increased arterial stiffness and aortic pulsatility. This pattern may partially explain both the increasing incidence of stroke in younger adults as well as late-onset, chronic cerebrovascular injury associated with concurrent systolic hypertension and historic midlife diastolic hypertension. With increasing arterial stiffness and autonomic dysfunction, blood pressure variability increases, independently predicting the risk of ischaemic and intracerebral haemorrhage, and is potentially modifiable beyond control of mean blood pressure.

However, the interaction between hypertension and control of cerebral blood flow remains poorly understood. Cerebral small vessel disease is associated with increased pulsatility in large cerebral vessels and reduced reactivity to carbon dioxide, both of which are being targeted in early-phase clinical trials. Cerebral arterial pulsatility is mainly dependent upon increased transmission of aortic pulsatility via stiff vessels to the brain whilst cerebrovascular reactivity reflects endothelial dysfunction. In contrast, although cerebral autoregulation is critical to adapt cerebral tone to blood pressure fluctuations to maintain cerebral blood flow, its role as a modifiable risk factor for cerebrovascular disease is uncertain.

New insights into hypertension-associated cerebrovascular pathophysiology may provide key targets to prevent chronic cerebrovascular disease, acute events and vascular cognitive impairment.

Introduction

Hypertension is the risk factor with the highest global attributable risk for death and the second-highest attributable risk for morbidity.1 It affected over a quarter of the world’s population in 2000 and is set to rise to approximately 29% of the population by 2025, affecting 1.56 billion people.2,3 A mean systolic blood pressure (SBP) >115mmHg explains 50-60% of the population attributable risk of ischaemic stroke4,5 and over 70% of haemorrhagic stroke,6 whilst an increment of 20mmHg SBP doubles the risk of stroke between 40-69 years of age.4 Randomisation to a treatment which reduces mean SBP results in a significant reduction in vascular events with a 5-6mmHg decrease in diastolic pressure resulting in a 33-50% reduction in stroke and a 4-22% reduction in acute coronary events.7

However, a detailed understanding of the mechanistic relationship between hypertension and stroke has lagged behind that of other vascular outcomes due to heterogeneity in stroke aetiology,8 heterogeneous radiological manifestations of chronic cerebrovascular injury9 and a lack of stroke phenotyping in observational cohort studies and blood-pressure lowering clinical trials.10 There have been limited studies of the effects of hypertension on the cerebral circulation11 and there are no specific proven hypertension-related treatments for small vessel disease (SVD), beyond blood pressure lowering.

Hypertension is associated with all ischaemic stroke subtypes, whether cardioembolic stroke related to atrial fibrillation (AF),12 large artery stroke due to carotid stenosis,13 lacunar stroke in SVD14 or less common stroke aetiologies such as carotid dissection,15 with an even more powerful effect for intracerebral haemorrhage. These aetiologies depend upon intermediate disease phenotypes which can be treated, such as carotid endarterectomy for atherosclerosis16 or anticoagulation for AF.17 However, the intracranial physiological effects of hypertension are less well understood even though they may explain some of the residual risk of recurrent stroke despite effective BP lowering. In the PROGRESS study, a 12mmHg reduction in blood pressure with combination treatment reduced recurrent stroke by up to 43%,18 whilst any treatment reduced intracerebral haemorrhage by 50%, with similar efficacy in all secondary prevention trials,19 but there was still a 10% risk of recurrent events. In the multi-centre TIARegistry, there was a 14.3% risk of recurrent major vascular events (MACE) at 5 years with a 9.5% risk of recurrent stroke,20 despite high-quality secondary prevention.

The principal manifestation of chronic cerebrovascular injury is cerebral small vessel disease. Originally defined by pathological changes including vessel lipohyalinosis, fibrinoid necrosis and vessel rarefaction, cSVD is now primarily defined by imaging characteristics on MRI (figure 1) for both of its commonest two forms: cerebral amyloid angiopathy (CAA) and deep perforator arteriopathy.9 These represent further intermediate phenotypes linking hypertension with cerebrovascular outcomes, and are responsible for 30% of ischaemic strokes, 80% of intracerebral haemorrhage21,22 and 45% of dementia, as well as late-onset refractory depression and significant global functional impairment at older ages.23,24 SVD is evident on brain imaging in >50% of people over 65 years of age and nearly all people over 80 years of age25. However, no specific treatments exist to target its progression.

Figure 1. Radiological manifestations of small vessel disease.

Figure 1

A) An acute small subcortical infarct on DWI imaging; B) Deep and periventricular white matter hyperintensities (arrowed); C) Dilated perivascular spaces bilaterally in the basal ganglia (arrowed on the left); D) a slit-like haemosiderin-lined cavity due to a chronic right intracerebral haemorrhage, subcortical (deep) cerebral microbleeds (white arrow) and a cortical (lobar) (black arrow) microbleed.

Any original data that support the conclusions of this study are available from the authors upon reasonable request.

Epidemiological insights

Derivation of the statistical measure ‘usual’ blood pressure in the early 1990s allowed precise estimation of the blood pressure related vascular risk by adjusting for variation between clinic visits, to remove the resulting bias that caused underestimation of the effect of average blood pressure (‘regression dilution bias’).7,26 Randomised controlled trials then confirmed that a reduction in mean blood pressure reduced the risk of recurrent events by the predicted degree,7 irrespective of baseline cerebrovascular risk18 and at minimally increased baseline blood pressure levels.27,28 However, hypertension research for primary prevention has lacked a nuanced focus on stroke, with limited stroke phenotyping in large randomized controlled trials (RCTs), whilst stroke research has lacked a nuanced understanding of hypertensive physiology and its effect on the cerebral circulation.

Blood pressure evolution over the lifespan

At all ages, elevated mean blood pressure is associated with an increased risk of stroke.10 As preventative strategies including antihypertensive treatment and reduced salt intake have become more effective the incidence of stroke has reduced in older age groups.29 However, due to the aging population there is a marked overall increase in the prevalence of stroke and vascular cognitive impairment. Furthermore, in younger patients under 50 years, particularly women, age-standardised stroke incidence is increasing, associated with increasing rates of obesity and hypertension.30,31

In late life, stroke, SVD and vascular cognitive impairment reflect the cumulative cerebrovascular effect of hypertension over the whole life-span (figure 2). Although SBP rises linearly with age, at approximately 55 years of age diastolic blood pressure starts to fall, with a shift from mixed or diastolic hypertension to an increased frequency of isolated systolic hypertension. This reflects increasing aortic stiffness and the cerebrovascular effects of long-standing hypertension.32

Figure 2. Relationship between ‘Usual’ diastolic and systolic blood pressure and white matter hyperintensities in the UK Biobank Study, stratified by age group.

Figure 2

Severity of white matter hyperintensities is expressed as the ratio of their volume to the average volume in the youngest patients with the lowest blood pressure, plotted on a logarithmic scale. Wartolowska K, Webb AJS. EHJ (2020).

Similarly, SVD is uncommon before 55 years of age but then increases at an increasing rate,25 driven by aging and hypertension and with a strong, genetic contribution.33 White matter hyperintensities (WMH) and concurrent systolic blood pressure are strongly associated at all ages,34 but there is a stronger relationship with midlife blood pressures,35,36 whilst the strength of the relationship increases the earlier in life the blood pressure is measured.37 Early microstructural brain imaging changes on diffusion tensor imaging in midlife participants in the UK Biobank study are cross-sectionally associated predominantly with mean arterial pressure, independent of age, whilst the association with pulse pressure attenuates significantly after adjustment for age. However, this interaction is synergistic such that increased pulse pressure accentuates the age-related deterioration in cerebral microstructure.36

In contrast to the long-term association with chronic SVD, recent blood pressure elevations are more strongly associated with both acute intracerebral haemorrhage38 and acute ischaemic stroke due to small vessel occlusion,39 implying a potentially causative interaction between increasing blood pressure levels over a timescale of a year or less, and both rupture or occlusion of small cerebral vessels. This relationship is particularly strong for stroke subtypes most closely related to hypertension, including deep parenchymal vs lobar haemorrhage and lacunar versus non-lacunar stroke, and provides a strong rationale for early, consistent blood pressure control, particularly after intracerebral haemorrhage, as is being tested with telemetric home monitoring in PROHIBIT-ICH (NCT03863665).

Mendelian randomisation has also demonstrated an age-dependent evolution of the relationship between blood pressure and cerebrovascular outcomes, with a likely causative association between high blood pressure and all stroke subtypes, except for lobar intracerebral haemorrhage.40 However, there were age-dependent differences between genetically determined drivers of mean versus pulsatile blood pressure. Both were associated with late-life large artery athero-thrombo-embolism, but elevated mean pressure was dominant across ages for other aetiological subtypes.41 Similar analyses have confirmed likely causative relationships between hypertension and the risk of incident all-cause dementia.42

Early life determinants of hypertension and cerebrovascular pathology

Early-life risk factors for late life cerebrovascular disease include low birth weight and pre-eclampsia,43 but vascular risk factors remain dominant after childhood.44 In particular, overactivity of the sympathetic nervous system is a key driver of hypertension, with increased muscle sympathetic nerve activity (MSNA) associated with obesity and blood pressure.45 This implies a potential treatment target with early, unblinded trials of renal sympathetic denervation (RDN) suggesting large reductions in systolic blood pressure (up to 40mmHg). Unfortunately, the properly blinded, sham-controlled Symplicity HTN-346 study failed to replicate this. However, more recent trials (SPYRAL-ON and OFF-MED, RADIANCE-HTN),47 with newer devices and 4-quadrant denervation, have suggested that a more rigorous interventional approach can achieve significant blood pressure reductions. However, the achieved BP reductions are equivalent only to the addition of one new drug, whilst the application of RDN to patients with stroke is yet to be tested.

Baroreceptor activation therapy is an alternative, whereby an implanted stimulator increases the parasympathetic:sympathetic balance, with a resulting reduction in blood pressure. Lagging behind RDN, uncontrolled studies have been prone to bias but a sustained blood pressure reduction was achieved in the sole RCT in resistant hypertension (RHEOS PIVOTAL).48

Arterial stiffness, pulse wave veloctity and wave reflection

Late-life hypertensive phenotypes are characterized by increasing arterial stiffness, with a reduction in elastin and increase in collagen in the aorta reducing vascular compliance and increasing speed of travel of the aortic pressure wave from the heart to the carotid and femoral arteries (pulse wave velocity, PWV). This marker of ‘vascular aging’ has a hazard ratio of 1.54 per standard deviation increase for stroke and 1.42 for all vascular events,4951 and its progression is hypertension-dependent, with a 20mmHg elevation of SBP associated with a 1.14m/s progression in PWV per decade.52 Furthermore, there are strong associations between aortic PWV and specific cerebrovascular phenotypes including acute lacunar stroke,53 deep intracerebral haemorrhage (by contrast with lobar),54 cerebral SVD and cognitive impairment 55,56, with a similar odds ratio of 1.25-1.3 per SD increase in PWV associated with an increase in severity of white matter hyperintensities on the Fazekas scale.

Increased arterial stiffness is also associated with increased wave reflection, whereby the pressure-wave generated with each cardiac impulse ‘reflects’ from points of increased impedance, either at the junction of large and medium vessels or between medium arteries and arterioles.57 Following reflection, the ‘reverse’ travelling wave then returns towards the heart to meet the forward travelling wave before it has dissipated. With increasing arterial stiffness, there is thus greater ‘augmentation’ of the peak of the waveform, increasing the aortic systolic pressure. Aortic BP is even more strongly associated with the risk of recurrent events than brachial pressure,58 since low resistance vascular beds (renal and cerebral) are more directly exposed to aortic pressures. Thus, a greater reduction in aortic pressure with an amlodipine-based regimen compared to an atenolol-based regimen potentially explained differences in vascular outcomes in the ASCOT-BPLA trial.59

Blood pressure variability

Mean blood pressure level over the lifespan is the strongest predictor of cerebrovascular disease.4 However, the statistical adjustment used to calculate ‘usual’ blood pressure depends upon adjusting for the variation in BP between assessments. The greater the variability in blood pressure (BPV) the greater the adjustment and the stronger the estimated relationship between mean blood pressure and subsequent risk. However, blood pressure is highly variable between clinic visits,60 and across five cohorts, the top decile of visit-to-visit BP variability was associated with 2.2-3.8 times the risk of recurrent stroke compared to the bottom decile.61 Variability was reduced in patients randomized to a calcium-channel blocker based regimen compared to an atenolol-based regimen, explaining the difference between treatment regimens that was not explained by mean blood pressure62 and consistent with effects of calcium channel blockers or thiazide-like diuretics in all large randomized controlled trials BPV and the risk of stroke.63

Visit-to-visit BP variability has subsequently been found to predict conditions from renal failure to dementia, with consistent associations with vascular risk64 on both visit-to-visit and home day-to-day variability,65 and worse outcome after both ischaemic66 and haemorrhagic stroke67 with increasing BPV. The association between BPV and cognitive impairment is greater than that for mean BP, although associations are highly heterogeneous between studies and may reflect reverse causation.68 Variability is predictive from visit-to-visit, day-to-day65,69 or beat-to-beat,70 is associated with arterial stiffness, baroreceptor dysfunction and increased reactivity to stress71 and progresses over the age of 5572 in parallel with progression of arterial stiffness and aortic pulsatility (figure 3).

Figure 3. Rate of progression of aortic pulse wave velocity (PWV) and middle cerebral pulsatility (PI) over 5 years in 188 patients after TIA or minor stroke, stratified by quartiles of age.

Figure 3

Individual changes are show, with summary results derived from a mixed effect linear model, stratified by quartiles of age and subdivided into men (red) and women (blue).56

The association between BPV, increased SVD and cognitive impairment73 supports the importance of an understudied, late-life complex phenotype of severe cerebral SVD, cognitive impairment, increased arterial stiffening and greater blood pressure variability. In this group, there may also be under-appreciated vulnerability to hypoperfusion of the brain due to low blood pressures,74 as suggested by the U-shaped association between cognitive impairment and diastolic blood pressure and the relationship with postural hypotension.75 In a recent analysis from the ARIC study, the highest risk of dementia was seen in patients with high mid-life blood pressure but late life hypotension.76

Pathophysiology of hypertensive cerebrovascular disease

Hypertension has broad vascular effects. It increases cardiac load, coronary artery disease and atrial dilatation, leading to AF, ventricular wall dyskinesia and heart failure. The increased shear stress on the endothelium contributes to the induction of atherosclerosis and carotid stenosis. These disease phenotypic manifestations have led to specific treatments to reduce stroke risk in the context of AF or carotid atherosclerosis. However, the intracranial effects of hypertension, particularly on cerebral small vessels, have not yet resulted in equivalent intermediate treatment targets, despite multiple identified pathways modifying the effect of hypertension and control of blood flow to the brain (figure 4).

Figure 4. Major mechanisms of cerebrovascular adaptation to changes in blood pressure and demand for blood flow to control cerebral perfusion.

Figure 4

Pathophysiological changes of hypertension

Chronic hypertension is associated with characteristic changes in small vessels throughout the body. Shear stress promotes intracranial atherosclerosis and intracranial stenoses, whilst in smaller vessels it induces smooth muscle hypertrophy, reorganization (or disorganization) of smooth muscle cells, a reduction in the wall:lumen ratio, reduced vascular compliance and, over time, rarefaction of vessels. This increases intracranial cerebrovascular resistance, balancing increased perfusion pressures, but is associated with impaired resting perfusion is evident in the white matter of patients with severe cSVD.74 This hypertrophic remodeling is partly dependent upon sympathetic nerve fibre innervation, and ameliorated (in rat models) by antagonism of the angiotensin-aldosterone system. Other potential targets include PPARy receptors, with reduced remodeling of large cerebral vessels in rat models under the influence of rosiglitazone or pioglitazone,77 with could explain the reduction in cerebrovascular events with pioglitazone in the IRIS trial.78

Endothelium derived nitric oxide (NO) is a key driver of vasodilatation, with reduced functional hyperaemia in older hypertensive patients, reduced vascular reactivity in cerebral small vessel disease and increased circulating markers of endothelial cell dysfunction in patients with cSVD and hypertension.79 Furthermore, break down of the blood brain barrier (BBB) in cerebral small vessel disease is seen both in white matter hyperintensities and in areas of normal appearing white matter in patients with either lacunar stroke or established white matter hyperintensities elsewhere in the brain.80 BBB dysfunction may be induced directly by chronic hypertension as a potential mechanism for development of SVD or cognitive decline, either through haemodynamic effects or through the induction of oxidative stress and low-grade inflammation81 or through Amyloid beta-mediated pathways.82

Phenotypes of Cerebral Small Vessel disease

Cerebral small vessel disease is associated with 30% of ischaemic stroke, 80% of intracerebral haemorrhage and 45% of dementia.39 Recurrent stroke was reduced in patients with lacunar events in the SPS3 trial, whilst haemorrhagic stroke was reduced by 63%. Moreover, intensive blood pressure lowering was associated with a reduction in progression of white matter hyperintensities (PROGRESS, SPRINT-MIND; ACCORD-MIND), with a dose-response relationship between blood pressure reduction and rate of progression. However, there was also increased cerebral atrophy, and in the majority of these trials baseline white matter hyperintensities were not severe, limiting generalizability to patients with advanced cerebral SVD. However, reassuringly, there was no evidence of impairment of MRI-measured cerebral perfusion with intensive treatment in the more severely affected participants in the PRESERVE trial.83

Cerebral microbleeds are the most prevalent haemorrhagic radiological manifestation of cerebral SVD. These areas of ‘blooming’ artefact most often reflect deposition of paramagnetic material, usually microscopic areas of subclinical blood extravasation (haemorrhage), though a range of lesions from haemosiderin deposition to intact erythrocytes to vessel wall calcification have been demonstrated.84,85 The pattern of distribution of microbleeds is indicative of the underlying pathology, with a strictly lobar distribution associated with cerebral amyloid angiopathy, reflecting deposition of β-amyloid in small cortical and leptomeningeal vessels. In contrast, deep microbleeds in the basal ganglia and brainstem are associated with hypertension, left ventricular hypertrophy, arterial stiffening and pulse pressure with a dose-response relationship between hypertension and severity of microbleeds.86 Although hypertension may trigger symptomatic lobar haemorrhage in CAA, there is a weaker overall association with hypertension, suggesting hypertension may aggravate CAA but not cause it. Studies to intensively control blood pressure after haemorrhage, either by intensive monitoring (PROHIBIT-ICH) or early triple, combination therapy (TRIDENT), may help to determine the effects of improved hypertension control.

Similar to cerebral microbleeds, there is a stronger association between hypertension and dilated perivascular spaces in the basal ganglia than with PVS in the centrum semiovale.87 This distribution of SVD markers associated with hypertension reflects brain regions most exposed to intraarterial pressures and pulsatility,88 and vulnerability to these factors is at least partially genetically determined, with a heritability of basal ganglia PVS of up to 0.6 and significant shared heritability with white matter hyperintensities and shared genetic associations in large scale genome-wide association studies.89 This indicates a common hypertension-associated genetic predisposition across different manifestations of cSVD, and may also contribute to observed associations between PVS and both ischaemic90 and intracerebral haemorrhage.91

Cerebral Pulsatility

Cerebral vessels modify the blood pressure to maintain perfusion of the brain throughout the cardiac cycle (via arterial pulsatility), from beat-to-beat and hour-to-hour (via cerebral autoregulation) and in response to endogenous and exogenous stimuli (via reactivity). Cerebral artery pulsatility is measured on transcranial ultrasound (Gosling’s pulsatility index), 4d-phase contrast MRI sequences,92 or by high temporal-resolution multiband imaging.93 Cerebral pulsatility is strongly associated with severity of white matter hyperintensities,94 acute lacunar infarcts, cerebral microbleeds95 and dilated perivascular spaces.96 It increases with age and is tightly associated with aortic stiffness and aortic pulsatility,56,97 reflecting increased transmission of the aortic waveform to the brain, modified by dampening of the waveform at points of tortuosity and impedance and by the distal resistance of smaller cerebral vessels. It is highly reproducible, progresses over the age of 65 and independently predicts the risk of recurrent cerebrovascular events. However, methods to modify it independent of mean blood pressure are not clear. In the ECLIPSE study a phosphodiesterase 3 inhibitor (cilostazol)98 reduced cerebral pulsatility, potentially explaining beneficial effects of cilostazol in clinical trials beyond its known antiplatelet activity.11

Perivascular Clearance

Arterial pulsatility may also be key to drainage of interstitial fluid from the brain along perivascular spaces, sometimes called ‘glymphatics’. This recently identified pathway challenged the axiom that CSF reabsorption only happened via the arachnoid granulations, with intracisternal injection of radiolabeled tracers appearing in the CSF filled perivascular spaces around arteries, and then around veins,99 exiting via newly identified meningeal lymphatic vessels.100 This pathyway has been linked to drainage of amyloid β and other solutes. Despite limitations in experimental techniques101, 2-photon imaging tracer studies and ligation studies of the carotid102 indicate that arterial pulsatility provides motive force for perivascular drainage in healthy animals. However, it is unclear whether hypertension-associated increases in pulsatility and vascular stiffness affect perivascular drainage in humans, given a lack of in vivo glymphatic imaging techniques that do not require intrathecal injection of contrast.103

Cerebrovascular Reactivity

The endothelium controls the cerebrovascular response to stimuli, regulating smooth muscle tone via balanced vasodilator and vasoconstrictor pathways. The archetypal, eNOS vasodilator pathway induces smooth muscle cGMP upregulation and cell relaxation to increase local CBF, which antagonises vasoconstrictor pathways such as endothelin-mediated vasocontriction.

Cerebrovascular reactivity (CVR) is most commonly measured in vivo by altering carbon dioxide levels, (inhalation of CO2, breath-holding, hyperventilation, acetazolamide) and measuring the vascular response (BOLD-MRI, ASL-MRI, MCA cerebral blood flow velocity on TCD, near infrared spectroscopy).104 CVR is an attractive therapeutic target, being simple to measure, modifiable by established medications, and with a biologically plausible relationship to disease. It is associated with severity of white matter hyperintensities, dilated perivascular spaces105 and microbleeds, and progresses in parallel with progression of SVD. Recent studies have also demonstrated impaired CVR in the basal ganglia in patients with deep intracerebral haemorrhages.106

However, studies of the relationship between hypertension and CVR, independent of established cerebrovascular injury remain sparse with inconsistencies in methods of measurement of CVR, limited data in different cerebrovascular manifestations and uncertainty regarding the temporal order of the components of the phenotypes. This critical question may be answered by large collaborative studies such the MarkVCID and SVDs@TARGET consortia, and by interventional studies targeting improvements in CVR: TREAT-SVDs (NCT03082014), OxHARP (NCT03855332), LACI-2 (NCT03451591).

Cerebral Autoregulation

Cerebral autoregulation maintains cerebral blood flow despite changes in blood pressure, with sustained BP elevation inducing vasoconstriction and increased resistance to blood flow. Hypertension is associated with a rise in the ‘set-point’ of this curve to compensate for elevations in blood pressure, although the dynamic range achieved may still be less than the upper limit of blood pressure variability.

With technical advances to measure beat-to-beat blood pressure and cerebral blood flow, dynamic cerebral autoregulation (dCA) reflects rapid adaptation to changes in blood pressure.107 dCA is measured either by induced blood pressure changes (thigh-cuff release, pharmacologically-induced hypertension) or by resting-state relationships between spontaneous fluctuations in blood pressure and cerebral blood flow in time-domain (‘Mx’), or frequency-domain transfer function analysis to estimate the magnitude (gain), delay (phase) and strength of the relationship (coherence), or by modeling the response to an acute step change (autoregulation index, ARI). The underlying changes in vascular tone depend upon myogenic, metabolic and autonomic/neurogenic responses to maintain stable CBF.

Despite the critical biological importance of cerebral autoregulation, evidence for its role in the aetiology of stroke is limited. Autoregulation is commonly impaired in the ipsilateral hemisphere in severe ischaemic stroke,108 is associated with an increased risk of early neurological deterioration109 and haemorrhage110 and can predict a poor response to reperfusion therapies despite recanalisation,111 but studies have been small with inconsistent findings, partly due to difficulties in consistent measurement of dCA, although international collaborations (INFOMATAS) seek to better define its role. 112

In comparison, the population-based effect of hypertension and age on dCA are even less well documented. Limited studies have not demonstrated a consistent decline in autoregulation with age, although with conflicting results. Similarly, there is limited evidence of the effects of chronic hypertension on dCA and its relevance to development of cerebrovascular pathology, despite significant potential for new interventional targets.

Physiologically guided treatment

Hypertension-related secondary prevention of cerebrovascular disease will be the focus of subsequent articles in this series, including intracerebral haemorrhage, recurrent ischaemic events and cognitive impairment. These intermediate pathophysiological targets between hypertension and clinical events provide new potential treatment targets. For example, calcium channel blockers and thiazide-like diuretics appear to reduce BP variability, but effects at different baseline blood pressure levels and by stroke aetiology are needed and there remains a need to define which antihypertensive medications are optimal in the secondary prevention of stroke.113

A number of trials are currently testing repurposed interventions to target specific aspects of these phenotypes. TREAT-SVDs is comparing the effects of amlodipine versus atenolol and losartan on cerebrovascular reactivity to inhaled carbon dioxide on MRI, and is expected to report in 2022/2023. OxHARP is testing phosphodiesterase inhibitors (sildenafil and cilostazol) on cerebral pulsatility and reactivity to carbon dioxide, using both transcranial ultrasound and MRI, with dCA as a secondary outcome. The completed but unreported PASTIS study (NCT02450253)assessed the effect of a single dose of tadalafil on white matter perfusion in the early phase after a lacunar stroke. LACI-1 tested implied improved cerebrovascular reactivity on MRI with either cilostazol or isosorbide mononitrate in lacunar stroke, leading to LACI 2 to test the same agents to prevent recurrent lacunar strokes and progression of white matter hyperintensities. This follows suggested benefits in larger clinical trials (PICASSO,114 CSPS115) with cilostazol reducing ischaemic stroke to an extent greater than expected from its antiplatelet effects, with no increased risk of haemorrhage. Finally, XILO-FIST has recently reported a null effect of allopurinol on progression of white matter hyperintensities, despite a small reduction in blood pressure.116 However, no trial has yet identified a reliable intervention that has lead on to definitive trials showing efficacy.

Conclusion

The relationship between hypertension and stroke, and its reduction by appropriate blood pressure control, has significantly reduced its incidence. However, our developing understanding of the complex physiological effects of hypertension on the cerebral circulation, particularly in the context of cerebral SVD and its consequences, promises new treatment paradigms for the future.

Sources of Funding

AJSW is funded by a Wellcome Trust Clinical Research Development Fellowship (206589/Z/17/Z).

Non-standard Abbreviations and Acronyms

AF

Atrial fibrillation

ASL

Arterial spin labelling

ARI

Autoregulatory index

BBB

Blood brain barrier

BPV

Blood pressure variability

CAA

Cerebral amyloid angiopathy

CBF

Cerebral blood flow

CSF

Cerebrospinal fluid

CVR

Cerebrovascular reactivity

dCA

Dynamic cerebral autoregulation

MCA

Middle cerebral artery

MSNA

Muscle sympathetic nerve activity

NO

Nitric oxide

PVS

Perivascular spaces

PWV

Pulse wave velocity

RCT

Randomised controlled trial

RDN

Renal sympathetic denervation

SBP

Systolic blood pressure

SVD

Small vessel disease

TCD

Transcranial Doppler ultrasound

WMH

White matter hyperintensities

Footnotes

Disclosures

Prof Werring reports compensation from Alexion Pharmaceuticals, Inc. for other services; compensation from Alnylam Pharmaceuticals for other services; compensation from Novo Nordisk for other services; compensation from Portola Pharmaceuticals for other services; and compensation from Bayer for other services.

References

  • 1.Collaborators GBDRF. Global burden of 87 risk factors in 204 countries and territories, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet. 2020;396:1223–1249. doi: 10.1016/S0140-6736(20)30752-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lawes CM, Vander Hoorn S, Rodgers A. Global burden of blood-pressure-related disease, 2001. Lancet. 2008;371:1513–1518. doi: 10.1016/S0140-6736(08)60655-8. [DOI] [PubMed] [Google Scholar]
  • 3.Kearney PM, Whelton M, Reynolds K, Muntner P, Whelton PK, He J. Global burden of hypertension: analysis of worldwide data. Lancet. 2005;365:217–223. doi: 10.1016/S0140-6736(05)17741-1. [DOI] [PubMed] [Google Scholar]
  • 4.Lewington S, Clarke R, Qizilbash N, Peto R, Collins R. Age-specific relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies. Lancet. 2002;360:1903–1913. doi: 10.1016/s0140-6736(02)11911-8. [DOI] [PubMed] [Google Scholar]
  • 5.O’Donnell MJ, Xavier D, Liu L, Zhang H, Chin SL, Rao-Melacini P, Rangarajan S, Islam S, Pais P, McQueen MJ, et al. Risk factors for ischaemic and intracerebral haemorrhagic stroke in 22 countries (the INTERSTROKE study): a case-control study. Lancet. 2010;376:112–123. doi: 10.1016/S0140-6736(10)60834-3. [DOI] [PubMed] [Google Scholar]
  • 6.OD M, Hankey GJ, Rangarajan S, Chin SL, Rao-Melacini P, Ferguson J, Xavier D, Lisheng L, Zhang H, Pais P, et al. Variations in knowledge, awareness and treatment of hypertension and stroke risk by country income level. Heart. 2020 doi: 10.1136/heartjnl-2019-316515. [DOI] [PubMed] [Google Scholar]
  • 7.Collins R, Peto R, MacMahon S, Hebert P, Fiebach NH, Eberlein KA, Godwin J, Qizilbash N, Taylor JO, Hennekens CH. Blood pressure, stroke, and coronary heart disease. Part 2, Short-term reductions in blood pressure: overview of randomised drug trials in their epidemiological context. Lancet. 1990;335:827–838. doi: 10.1016/0140-6736(90)90944-z. [DOI] [PubMed] [Google Scholar]
  • 8.Lovett JK, Coull AJ, Rothwell PM. Early risk of recurrence by subtype of ischemic stroke in population-based incidence studies. Neurology. 2004;62:569–573. doi: 10.1212/01.wnl.0000110311.09970.83. [DOI] [PubMed] [Google Scholar]
  • 9.Wardlaw JM, Smith EE, Biessels GJ, Cordonnier C, Fazekas F, Frayne R, Lindley RI, O’Brien JT, Barkhof F, Benavente OR, et al. Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration. Lancet Neurol. 2013;12:822–838. doi: 10.1016/S1474-4422(13)70124-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lewington S, Clarke R, Qizilbash N, Peto R, Collins R, Prospective Studies C Age-specific relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies. Lancet. 2002;360:1903–1913. doi: 10.1016/s0140-6736(02)11911-8. [DOI] [PubMed] [Google Scholar]
  • 11.Webb AJS. Effects of vasodilating medications on cerebral haemodynamics in health and disease: systematic review and meta-analysis. J Hypertens. 2019;37:1119–1125. doi: 10.1097/HJH.0000000000002033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Tohgi H, Tajima T, Konno T, Towada S, Kamata A, Yamazaki M. The risk of cerebral infarction in non-valvular atrial fibrillation: effects of age, hypertension and antihypertensive treatment. Eur Neurol. 1991;31:126–130. doi: 10.1159/000116661. [DOI] [PubMed] [Google Scholar]
  • 13.Park JH, Ovbiagele B, Hong KS, Kwon SU. Association of Systolic Blood Pressure with Progression of Symptomatic Intracranial Atherosclerotic Stenosis. J Stroke. 2017;19:304–311. doi: 10.5853/jos.2017.00136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Group SPSS. Benavente OR, Coffey CS, Conwit R, Hart RG, McClure LA, Pearce LA, Pergola PE, Szychowski JM. Blood-pressure targets in patients with recent lacunar stroke: the SPS3 randomised trial. Lancet. 2013;382:507–515. doi: 10.1016/S0140-6736(13)60852-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Pezzini A, Caso V, Zanferrari C, Del Zotto E, Paciaroni M, Bertolino C, Grassi M, Agnelli G, Padovani A. Arterial hypertension as risk factor for spontaneous cervical artery dissection. A case-control study. J Neurol Neurosurg Psychiatry. 2006;77:95–97. doi: 10.1136/jnnp.2005.063107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Rothwell PM. ACST: which subgroups will benefit most from carotid endarterectomy? Lancet. 2004;364:1122–1123. doi: 10.1016/S0140-6736(04)17090-6. author reply 1125-1126. [DOI] [PubMed] [Google Scholar]
  • 17.Mant J, Hobbs FD, Fletcher K, Roalfe A, Fitzmaurice D, Lip GY, Murray E, investigators B, Midland Research Practices N Warfarin versus aspirin for stroke prevention in an elderly community population with atrial fibrillation (the Birmingham Atrial Fibrillation Treatment of the Aged Study, BAFTA): a randomised controlled trial. Lancet. 2007;370:493–503. doi: 10.1016/S0140-6736(07)61233-1. [DOI] [PubMed] [Google Scholar]
  • 18.Group PC. Randomised trial of a perindopril-based blood-pressure-lowering regimen among 6,105 individuals with previous stroke or transient ischaemic attack. Lancet. 2001;358:1033–1041. doi: 10.1016/S0140-6736(01)06178-5. [DOI] [PubMed] [Google Scholar]
  • 19.Katsanos AH, Filippatou A, Manios E, Deftereos S, Parissis J, Frogoudaki A, Vrettou AR, Ikonomidis I, Pikilidou M, Kargiotis O, et al. Blood Pressure Reduction and Secondary Stroke Prevention: A Systematic Review and Metaregression Analysis of Randomized Clinical Trials. Hypertension. 2017;69:171–179. doi: 10.1161/HYPERTENSIONAHA.116.08485. [DOI] [PubMed] [Google Scholar]
  • 20.Amarenco P, Lavallee PC, Monteiro Tavares L, Labreuche J, Albers GW, Abboud H, Anticoli S, Audebert H, Bornstein NM, Caplan LR, et al. Five-Year Risk of Stroke after TIA or Minor Ischemic Stroke. N Engl J Med. 2018 doi: 10.1056/NEJMoa1802712. [DOI] [PubMed] [Google Scholar]
  • 21.Pasi M, Cordonnier C. Clinical Relevance of Cerebral Small Vessel Diseases. Stroke. 2020;51:47–53. doi: 10.1161/STROKEAHA.119.024148. [DOI] [PubMed] [Google Scholar]
  • 22.Pasi M, Charidimou A, Boulouis G, Auriel E, Ayres A, Schwab KM, Goldstein JN, Rosand J, Viswanathan A, Pantoni L, et al. Mixed-location cerebral hemorrhage/microbleeds: Underlying microangiopathy and recurrence risk. Neurology. 2018;90:e119–e126. doi: 10.1212/WNL.0000000000004797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Teodorczuk A, Firbank MJ, Pantoni L, Poggesi A, Erkinjuntti T, Wallin A, Wahlund LO, Scheltens P, Waldemar G, Schrotter G, et al. Relationship between baseline white-matter changes and development of late-life depressive symptoms: 3-year results from the LADIS study. Psychol Med. 2010;40:603–610. doi: 10.1017/S0033291709990857. [DOI] [PubMed] [Google Scholar]
  • 24.Inzitari D, Pracucci G, Poggesi A, Carlucci G, Barkhof F, Chabriat H, Erkinjuntti T, Fazekas F, Ferro JM, Hennerici M, et al. Changes in white matter as determinant of global functional decline in older independent outpatients: three year follow-up of LADIS (leukoaraiosis and disability) study cohort. BMJ. 2009;339:b2477. doi: 10.1136/bmj.b2477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Simoni M, Li L, Paul NL, Gruter BE, Schulz UG, Kuker W, Rothwell PM. Age- and sex-specific rates of leukoaraiosis in TIA and stroke patients: population-based study. Neurology. 2012;79:1215–1222. doi: 10.1212/WNL.0b013e31826b951e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.MacMahon S, Peto R, Cutler J, Collins R, Sorlie P, Neaton J, Abbott R, Godwin J, Dyer A, Stamler J. Blood pressure, stroke, and coronary heart disease. Part 1, Prolonged differences in blood pressure: prospective observational studies corrected for the regression dilution bias. Lancet. 1990;335:765–774. doi: 10.1016/0140-6736(90)90878-9. [DOI] [PubMed] [Google Scholar]
  • 27.Group SR, Wright JT, Jr, Williamson JD, Whelton PK, Snyder JK, Sink KM, Rocco MV, Reboussin DM, Rahman M, Oparil S, et al. A Randomized Trial of Intensive versus Standard Blood-Pressure Control. N Engl J Med. 2015;373:2103–2116. doi: 10.1056/NEJMoa1511939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Blood Pressure Lowering Treatment Trialists C. Pharmacological blood pressure lowering for primary and secondary prevention of cardiovascular disease across different levels of blood pressure: an individual participant-level data meta-analysis. Lancet. 2021;397:1625–1636. doi: 10.1016/S0140-6736(21)00590-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Rothwell PM, Coull AJ, Giles MF, Howard SC, Silver LE, Bull LM, Gutnikov SA, Edwards P, Mant D, Sackley CM, et al. Change in stroke incidence, mortality, case-fatality, severity, and risk factors in Oxfordshire, UK from 1981 to 2004 (Oxford Vascular Study) Lancet. 2004;363:1925–1933. doi: 10.1016/S0140-6736(04)16405-2. [DOI] [PubMed] [Google Scholar]
  • 30.Mitchell AB, Cole JW, McArdle PF, Cheng YC, Ryan KA, Sparks MJ, Mitchell BD, Kittner SJ. Obesity increases risk of ischemic stroke in young adults. Stroke. 2015;46:1690–1692. doi: 10.1161/STROKEAHA.115.008940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kernan WN, Dearborn JL. Obesity increases stroke risk in young adults: opportunity for prevention. Stroke. 2015;46:1435–1436. doi: 10.1161/STROKEAHA.115.009347. [DOI] [PubMed] [Google Scholar]
  • 32.Webb AJS. Progression of Arterial Stiffness is Associated With Midlife Diastolic Blood Pressure and Transition to Late-Life Hypertensive Phenotypes. J Am Heart Assoc. 2020;9:e014547. doi: 10.1161/JAHA.119.014547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Malik R, Chauhan G, Traylor M, Sargurupremraj M, Okada Y, Mishra A, Rutten-Jacobs L, Giese AK, van der Laan SW, Gretarsdottir S, et al. Multiancestry genome-wide association study of 520,000 subjects identifies 32 loci associated with stroke and stroke subtypes. Nat Genet. 2018;50:524–537. doi: 10.1038/s41588-018-0058-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wilkinson I, Webb AJS. Consistency of associations of systolic and diastolic blood pressure with white matter hyperintensities: A meta-analysis. Int J Stroke. 2021:17474930211043364. doi: 10.1177/17474930211043364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Lane CABJ, Nicholas JM, Sudre CH, Cash DM, Parker TD, Malone IB, Lu K, James SN, Keshavan A, Murray-Smith H, Wong A, et al. Associations between blood pressure across adulthood and late-life brain structure and pathology in the neuroscience substudy of the 1946 British birth cohort (Insight 46): an epidemiological study. Lancet Neurol. 2019 doi: 10.1016/S1474-4422(19)30228-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Wartolowska KA, Webb AJS. Midlife blood pressure is associated with the severity of white matter hyperintensities: analysis of the UK Biobank cohort study. Eur Heart J. 2020 doi: 10.1093/eurheartj/ehaa756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lau KK, Li L, Simoni M, Mehta Z, Kuker W, Rothwell PM, Oxford Vascular S Long-Term Premorbid Blood Pressure and Cerebral Small Vessel Disease Burden on Imaging in Transient Ischemic Attack and Ischemic Stroke. Stroke. 2018;49:2053–2060. doi: 10.1161/STROKEAHA.118.021578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Fischer U, Cooney MT, Bull LM, Silver LE, Chalmers J, Anderson CS, Mehta Z, Rothwell PM. Acute post-stroke blood pressure relative to premorbid levels in intracerebral haemorrhage versus major ischaemic stroke: a population-based study. Lancet Neurol. 2014;13:374–384. doi: 10.1016/S1474-4422(14)70031-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Li L, Welch SJV, Gutnikov SA, Mehta Z, Rothwell PM, Oxford Vascular S Time course of blood pressure control prior to lacunar TIA and stroke: Populationbased study. Neurology. 2018;90:e1732–e1741. doi: 10.1212/WNL.0000000000005526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Georgakis MK, Gill D, Webb AJS, Evangelou E, Elliott P, Sudlow CLM, Dehghan A, Malik R, Tzoulaki I, Dichgans M. Genetically determined blood pressure, antihypertensive drug classes, and risk of stroke subtypes. Neurology. 2020;95:e353–e361. doi: 10.1212/WNL.0000000000009814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Georgakis MK, Gill D, Malik R, Protogerou AD, Webb AJS, Dichgans M. Genetically Predicted Blood Pressure Across the Lifespan: Differential Effects of Mean and Pulse Pressure on Stroke Risk. Hypertension. 2020;76:953–961. doi: 10.1161/HYPERTENSIONAHA.120.15136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Malik R, Georgakis MK, Neitzel J, Rannikmae K, Ewers M, Seshadri S, Sudlow CLM, Dichgans M. Midlife vascular risk factors and risk of incident dementia: Longitudinal cohort and Mendelian randomization analyses in the UK Biobank. Alzheimers Dement. 2021;17:1422–1431. doi: 10.1002/alz.12320. [DOI] [PubMed] [Google Scholar]
  • 43.Lewandowski AJ, Raman B, Bertagnolli M, Mohamed A, Williamson W, Pelado JL, McCance A, Lapidaire W, Neubauer S, Leeson P. Association of Preterm Birth With Myocardial Fibrosis and Diastolic Dysfunction in Young Adulthood. J Am Coll Cardiol. 2021;78:683–692. doi: 10.1016/j.jacc.2021.05.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Williamson W, Lewandowski AJ, Forkert ND, Griffanti L, Okell TW, Betts J, Boardman H, Siepmann T, McKean D, Huckstep O, et al. Association of Cardiovascular Risk Factors With MRI Indices of Cerebrovascular Structure and Function and White Matter Hyperintensities in Young Adults. JAMA. 2018;320:665–673. doi: 10.1001/jama.2018.11498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Grassi G, Biffi A, Seravalle G, Trevano FQ, Dell’Oro R, Corrao G, Mancia G. Sympathetic Neural Overdrive in the Obese and Overweight State. Hypertension. 2019;74:349–358. doi: 10.1161/HYPERTENSIONAHA.119.12885. [DOI] [PubMed] [Google Scholar]
  • 46.Bakris GL, Townsend RR, Liu M, Cohen SA, D’Agostino R, Flack JM, Kandzari DE, Katzen BT, Leon MB, Mauri L, et al. Impact of renal denervation on 24-hour ambulatory blood pressure: results from SYMPLICITY HTN-3. J Am Coll Cardiol. 2014;64:1071–1078. doi: 10.1016/j.jacc.2014.05.012. [DOI] [PubMed] [Google Scholar]
  • 47.Kjeldsen SE, Narkiewicz K, Burnier M, Oparil S. The five RADIANCE-HTN and SPYRAL-HTN randomised studies suggest that the BP lowering effect of RDN corresponds to the effect of one antihypertensive drug. Blood Press; 2021. pp. 1–5. [DOI] [PubMed] [Google Scholar]
  • 48.Bisognano JD, Bakris G, Nadim MK, Sanchez L, Kroon AA, Schafer J, de Leeuw PW, Sica DA. Baroreflex activation therapy lowers blood pressure in patients with resistant hypertension: results from the double-blind, randomized, placebo-controlled rheos pivotal trial. J Am Coll Cardiol. 2011;58:765–773. doi: 10.1016/j.jacc.2011.06.008. [DOI] [PubMed] [Google Scholar]
  • 49.Laurent S, Cockcroft J, Van Bortel L, Boutouyrie P, Giannattasio C, Hayoz D, Pannier B, Vlachopoulos C, Wilkinson I, Struijker-Boudier H, et al. Expert consensus document on arterial stiffness: methodological issues and clinical applications. Eur Heart J. 2006;27:2588–2605. doi: 10.1093/eurheartj/ehl254. [DOI] [PubMed] [Google Scholar]
  • 50.Ben-Shlomo Y, Spears M, Boustred C, May M, Anderson SG, Benjamin EJ, Boutouyrie P, Cameron J, Chen CH, Cruickshank JK, et al. Aortic pulse wave velocity improves cardiovascular event prediction: an individual participant meta-analysis of prospective observational data from 17,635 subjects. J Am Coll Cardiol. 2014;63:636–646. doi: 10.1016/j.jacc.2013.09.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Vlachopoulos C, Aznaouridis K, Stefanadis C. Prediction of cardiovascular events and all-cause mortality with arterial stiffness: a systematic review and meta-analysis. J Am Coll Cardiol. 2010;55:1318–1327. doi: 10.1016/j.jacc.2009.10.061. [DOI] [PubMed] [Google Scholar]
  • 52.Wilson J, Webb AJS. Systolic Blood Pressure and Longitudinal Progression of Arterial Stiffness: A Quantitative Meta-Analysis. J Am Heart Assoc. 2020;9:e017804. doi: 10.1161/JAHA.120.017804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Tuttolomondo A, Di Sciacca R, Di Raimondo D, Serio A, D’Aguanno G, Pinto A, Licata G. Arterial stiffness indexes in acute ischemic stroke: relationship with stroke subtype. Atherosclerosis. 2010;211:187–194. doi: 10.1016/j.atherosclerosis.2010.02.010. [DOI] [PubMed] [Google Scholar]
  • 54.Acampa M, Guideri F, Di Donato I, Tassi R, Marotta G, Lo Giudice G, D’Andrea P, Martini G. Arterial stiffness in patients with deep and lobar intracerebral hemorrhage. J Stroke. 2014;16:184–188. doi: 10.5853/jos.2014.16.3.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Meyer ML, Palta P, Tanaka H, Deal JA, Wright J, Knopman DS, Griswold ME, Mosley TH, Heiss G. Association of Central Arterial Stiffness and Pressure Pulsatility with Mild Cognitive Impairment and Dementia: The Atherosclerosis Risk in Communities Study-Neurocognitive Study (ARIC-NCS) J Alzheimers Dis. 2017;57:195–204. doi: 10.3233/JAD-161041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Alastair JS, Webb AL, Wartolowska Karolina, Mazzucco Sara, Rothwell PeterM, on behalf of the OXVASC study cohort Aortic stiffness, pulse pressure and cerebral pulsatility progress despite best medical management: the OXVASC cohort. Stroke. 2021 doi: 10.1161/STROKEAHA.121.035560. (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Westerhof N, Sipkema P, van den Bos GC, Elzinga G. Forward and backward waves in the arterial system. Cardiovasc Res. 1972;6:648–656. doi: 10.1093/cvr/6.6.648. [DOI] [PubMed] [Google Scholar]
  • 58.McEniery CM, Cockcroft JR, Roman MJ, Franklin SS, Wilkinson IB. Central blood pressure: current evidence and clinical importance. Eur Heart J. 2014;35:1719–1725. doi: 10.1093/eurheartj/eht565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Williams B, Lacy PS, Thom SM, Cruickshank K, Stanton A, Collier D, Hughes AD, Thurston H, O’Rourke M, Investigators C, et al. Differential impact of blood pressure-lowering drugs on central aortic pressure and clinical outcomes: principal results of the Conduit Artery Function Evaluation (CAFE) study. Circulation. 2006;113:1213–1225. doi: 10.1161/CIRCULATIONAHA.105.595496. [DOI] [PubMed] [Google Scholar]
  • 60.Cuffe RL, Howard SC, Algra A, Warlow CP, Rothwell PM. Medium-term variability of blood pressure and potential underdiagnosis of hypertension in patients with previous transient ischemic attack or minor stroke. Stroke. 2006;37:2776–2783. doi: 10.1161/01.STR.0000244761.62073.05. [DOI] [PubMed] [Google Scholar]
  • 61.Rothwell PM, Howard SC, Dolan E, O’Brien E, Dobson JE, Dahlof B, Sever PS, Poulter NR. Prognostic significance of visit-to-visit variability, maximum systolic blood pressure, and episodic hypertension. Lancet. 2010;375:895–905. doi: 10.1016/S0140-6736(10)60308-X. [DOI] [PubMed] [Google Scholar]
  • 62.Rothwell PM, Howard SC, Dolan E, O’Brien E, Dobson JE, Dahlof B, Poulter NR, Sever PS, Ascot B, Investigators MRCT Effects of beta blockers and calcium-channel blockers on within-individual variability in blood pressure and risk of stroke. Lancet Neurol. 2010;9:469–480. doi: 10.1016/S1474-4422(10)70066-1. [DOI] [PubMed] [Google Scholar]
  • 63.Webb AJ, Fischer U, Mehta Z, Rothwell PM. Effects of antihypertensive-drug class on interindividual variation in blood pressure and risk of stroke: a systematic review and meta-analysis. Lancet. 2010;375:906–915. doi: 10.1016/S0140-6736(10)60235-8. [DOI] [PubMed] [Google Scholar]
  • 64.Stevens SL, Wood S, Koshiaris C, Law K, Glasziou P, Stevens RJ, McManus RJ. Blood pressure variability and cardiovascular disease: systematic review and meta-analysis. BMJ. 2016;354:i4098. doi: 10.1136/bmj.i4098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Johansson JK, Niiranen TJ, Puukka PJ, Jula AM. Prognostic value of the variability in home-measured blood pressure and heart rate: the Finn-Home Study. Hypertension. 2012;59:212–218. doi: 10.1161/HYPERTENSIONAHA.111.178657. [DOI] [PubMed] [Google Scholar]
  • 66.Manning LS, Mistri AK, Potter J, Rothwell PM, Robinson TG. Short-term blood pressure variability in acute stroke: post hoc analysis of the controlling hypertension and hypotension immediately post stroke and continue or stop post-stroke antihypertensives collaborative study trials. Stroke. 2015;46:1518–1524. doi: 10.1161/STROKEAHA.115.009078. [DOI] [PubMed] [Google Scholar]
  • 67.Manning L, Hirakawa Y, Arima H, Wang X, Chalmers J, Wang J, Lindley R, Heeley E, Delcourt C, Neal B, et al. Blood pressure variability and outcome after acute intracerebral haemorrhage: a post-hoc analysis of INTERACT2, a randomised controlled trial. Lancet Neurol. 2014;13:364–373. doi: 10.1016/S1474-4422(14)70018-3. [DOI] [PubMed] [Google Scholar]
  • 68.de Heus RAA, Tzourio C, Lee EJL, Opozda M, Vincent AD, Anstey KJ, Hofman A, Kario K, Lattanzi S, Launer LJ, et al. Association Between Blood Pressure Variability With Dementia and Cognitive Impairment: A Systematic Review and Meta-Analysis. Hypertension. 2021;78:1478–1489. doi: 10.1161/HYPERTENSIONAHA.121.17797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kikuya M, Ohkubo T, Metoki H, Asayama K, Hara A, Obara T, Inoue R, Hoshi H, Hashimoto J, Totsune K, et al. Day-by-day variability of blood pressure and heart rate at home as a novel predictor of prognosis: the Ohasama study. Hypertension. 2008;52:1045–1050. doi: 10.1161/HYPERTENSIONAHA.107.104620. [DOI] [PubMed] [Google Scholar]
  • 70.Webb AJS, Mazzucco S, Li L, Rothwell PM. Prognostic Significance of Blood Pressure Variability on Beat-to-Beat Monitoring After Transient Ischemic Attack and Stroke. Stroke. 2018;49:62–67. doi: 10.1161/STROKEAHA.117.019107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Webb AJ, Rothwell PM. Physiological correlates of beat-to-beat, ambulatory, and day-to-day home blood pressure variability after transient ischemic attack or minor stroke. Stroke. 2014;45:533–538. doi: 10.1161/STROKEAHA.113.003321. [DOI] [PubMed] [Google Scholar]
  • 72.Webb AJS, Lawson A, Wartolowska K, Mazzucco S, Rothwell PM. Progression of Beat-to-Beat Blood Pressure Variability Despite Best Medical Management. Hypertension. 2020 doi: 10.1161/HYPERTENSIONAHA.120.16290. HYPERTENSIONAHA12016290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.van Middelaar T, van Vught LA, van Gool WA, Simons EMF, van den Born BH, Moll van Charante EP, Richard E. Blood pressure-lowering interventions to prevent dementia: a systematic review and meta-analysis. J Hypertens. 2018;36:1780–1787. doi: 10.1097/HJH.0000000000001829. [DOI] [PubMed] [Google Scholar]
  • 74.Markus HS, Lythgoe DJ, Ostegaard L, O’Sullivan M, Williams SC. Reduced cerebral blood flow in white matter in ischaemic leukoaraiosis demonstrated using quantitative exogenous contrast based perfusion MRI. J Neurol Neurosurg Psychiatry. 2000;69:48–53. doi: 10.1136/jnnp.69.1.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wolters FJ, Mattace-Raso FU, Koudstaal PJ, Hofman A, Ikram MA, Heart Brain Connection Collaborative Research G Orthostatic Hypotension and the Long-Term Risk of Dementia: A Population-Based Study. PLoS Med. 2016;13:e1002143. doi: 10.1371/journal.pmed.1002143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Walker KA, Sharrett AR, Wu A, Schneider ALC, Albert M, Lutsey PL, Bandeen-Roche K, Coresh J, Gross AL, Windham BG, et al. Association of Midlife to Late-Life Blood Pressure Patterns With Incident Dementia. JAMA. 2019;322:535–545. doi: 10.1001/jama.2019.10575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cipolla MJ, Bishop N, Vinke RS, Godfrey JA. PPAR{gamma} activation prevents hypertensive remodeling of cerebral arteries and improves vascular function in female rats. Stroke. 2010;41:1266–1270. doi: 10.1161/STROKEAHA.109.576942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kernan WN, Viscoli CM, Furie KL, Young LH, Inzucchi SE, Gorman M, Guarino PD, Lovejoy AM, Peduzzi PN, Conwit R, et al. Pioglitazone after Ischemic Stroke or Transient Ischemic Attack. N Engl J Med. 2016;374:1321–1331. doi: 10.1056/NEJMoa1506930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Poggesi A, Pasi M, Pescini F, Pantoni L, Inzitari D. Circulating biologic markers of endothelial dysfunction in cerebral small vessel disease: A review. J Cereb Blood Flow Metab. 2016;36:72–94. doi: 10.1038/jcbfm.2015.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Wardlaw JM, Doubal FN, Valdes-Hernandez M, Wang X, Chappell FM, Shuler K, Armitage PA, Carpenter TC, Dennis MS. Blood-brain barrier permeability and long-term clinical and imaging outcomes in cerebral small vessel disease. Stroke. 2013;44:525–527. doi: 10.1161/STROKEAHA.112.669994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Pires PW, Dams Ramos CM, Matin N, Dorrance AM. The effects of hypertension on the cerebral circulation. Am J Physiol Heart Circ Physiol. 2013;304:H1598–1614. doi: 10.1152/ajpheart.00490.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Zhang M, Mao Y, Ramirez SH, Tuma RF, Chabrashvili T. Angiotensin II induced cerebral microvascular inflammation and increased blood-brain barrier permeability via oxidative stress. Neuroscience. 2010;171:852–858. doi: 10.1016/j.neuroscience.2010.09.029. [DOI] [PubMed] [Google Scholar]
  • 83.Croall ID, Tozer DJ, Moynihan B, Khan U, O’Brien JT, Morris RG, Cambridge VC, Barrick TR, Blamire AM, Ford GA, et al. Effect of Standard vs Intensive Blood Pressure Control on Cerebral Blood Flow in Small Vessel Disease: The PRESERVE Randomized Clinical Trial. JAMA Neurol. 2018;75:720–727. doi: 10.1001/jamaneurol.2017.5153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.van Veluw SJ, Biessels GJ, Klijn CJ, Rozemuller AJ. Heterogeneous histopathology of cortical microbleeds in cerebral amyloid angiopathy. Neurology. 2016;86:867–871. doi: 10.1212/WNL.0000000000002419. [DOI] [PubMed] [Google Scholar]
  • 85.Shoamanesh A, Kwok CS, Benavente O. Cerebral microbleeds: histopathological correlation of neuroimaging. Cerebrovasc Dis. 2011;32:528–534. doi: 10.1159/000331466. [DOI] [PubMed] [Google Scholar]
  • 86.Lyu L, Shen J, Zeng C, Ji J, Hu W, Wei T, Mao W. Cerebral microbleeds are associated with blood pressure levels in individuals with hypertension. Clin Exp Hypertens. 2020;42:328–334. doi: 10.1080/10641963.2019.1665673. [DOI] [PubMed] [Google Scholar]
  • 87.Yang S, Qin W, Yang L, Fan H, Li Y, Yin J, Hu W. The relationship between ambulatory blood pressure variability and enlarged perivascular spaces: a cross-sectional study. BMJ Open. 2017;7:e015719. doi: 10.1136/bmjopen-2016-015719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Gutierrez J, Elkind MS, Cheung K, Rundek T, Sacco RL, Wright CB. Pulsatile and steady components of blood pressure and subclinical cerebrovascular disease: the Northern Manhattan Study. J Hypertens. 2015;33:2115–2122. doi: 10.1097/HJH.0000000000000686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Knol MJ, Lu D, Traylor M, Adams HHH, Romero JRJ, Smith AV, Fornage M, Hofer E, Liu J, Hostettler IC, et al. Association of common genetic variants with brain microbleeds: A genome-wide association study. Neurology. 2020;95:e3331–e3343. doi: 10.1212/WNL.0000000000010852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lau KK, Li L, Schulz U, Simoni M, Chan KH, Ho SL, Cheung RTF, Kuker W, Mak HKF, Rothwell PM. Total small vessel disease score and risk of recurrent stroke: Validation in 2 large cohorts. Neurology. 2017;88:2260–2267. doi: 10.1212/WNL.0000000000004042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Best JG, Barbato C, Ambler G, Du H, Banerjee G, Wilson D, Shakeshaft C, Cohen H, Yousry TA, Al-Shahi Salman R, et al. Association of enlarged perivascular spaces and anticoagulant-related intracranial hemorrhage. Neurology. 2020;95:e2192–e2199. doi: 10.1212/WNL.0000000000010788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Schnell S, Ansari SA, Wu C, Garcia J, Murphy IG, Rahman OA, Rahsepar AA, Aristova M, Collins JD, Carr JC, et al. Accelerated dual-venc 4D flow MRI for neurovascular applications. J Magn Reson Imaging. 2017;46:102–114. doi: 10.1002/jmri.25595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Viessmann O, Moller HE, Jezzard P. Dual regression physiological modeling of resting-state EPI power spectra: Effects of healthy aging. Neuroimage. 2019;187:68–76. doi: 10.1016/j.neuroimage.2018.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Webb AJ, Simoni M, Mazzucco S, Kuker W, Schulz U, Rothwell PM. Increased cerebral arterial pulsatility in patients with leukoaraiosis: arterial stiffness enhances transmission of aortic pulsatility. Stroke. 2012;43:2631–2636. doi: 10.1161/STROKEAHA.112.655837. [DOI] [PubMed] [Google Scholar]
  • 95.Chou KH, Wang PN, Peng LN, Liu LK, Lee WJ, Chen LK, Lin CP, Chung CP. Location-Specific Association Between Cerebral Microbleeds and Arterial Pulsatility. Front Neurol. 2019;10:1012. doi: 10.3389/fneur.2019.01012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Brown R, Benveniste H, Black SE, Charpak S, Dichgans M, Joutel A, Nedergaard M, Smith KJ, Zlokovic BV, Wardlaw JM. Understanding the role of the perivascular space in cerebral small vessel disease. Cardiovasc Res. 2018;114:1462–1473. doi: 10.1093/cvr/cvy113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Webb AJ, Lawson A, Li L, Mazzucco S, Rothwell PM, Oxford Vascular Study Phenotyped C Physiological determinants of residual cerebral arterial pulsatility on best medical treatment after TIA or minor stroke. J Cereb Blood Flow Metab. 2020:271678X20969984. doi: 10.1177/0271678X20969984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Han SW, Lee SS, Kim SH, Lee JH, Kim GS, Kim OJ, Koh IS, Lee JY, Suk SH, Lee SI, et al. Effect of cilostazol in acute lacunar infarction based on pulsatility index of transcranial Doppler (ECLIPse): a multicenter, randomized, double-blind, placebo-controlled trial. Eur Neurol. 2013;69:33–40. doi: 10.1159/000338247. [DOI] [PubMed] [Google Scholar]
  • 99.Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, Benveniste H, Vates GE, Deane R, Goldman SA, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med. 2012;4:147ra111. doi: 10.1126/scitranslmed.3003748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, Derecki NC, Castle D, Mandell JW, Lee KS, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523:337–341. doi: 10.1038/nature14432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Plog BA, Nedergaard M. The Glymphatic System in Central Nervous System Health and Disease: Past, Present, and Future. Annu Rev Pathol. 2018;13:379–394. doi: 10.1146/annurev-pathol-051217-111018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Iliff JJ, Wang M, Zeppenfeld DM, Venkataraman A, Plog BA, Liao Y, Deane R, Nedergaard M. Cerebral arterial pulsation drives paravascular CSF-interstitial fluid exchange in the murine brain. J Neurosci. 2013;33:18190–18199. doi: 10.1523/JNEUROSCI.1592-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Eide PK, Vatnehol SAS, Emblem KE, Ringstad G. Magnetic resonance imaging provides evidence of glymphatic drainage from human brain to cervical lymph nodes. Sci Rep. 2018;8:7194. doi: 10.1038/s41598-018-25666-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Sleight E, Stringer MS, Marshall I, Wardlaw JM, Thrippleton MJ. Cerebrovascular Reactivity Measurement Using Magnetic Resonance Imaging: A Systematic Review. Front Physiol. 2021;12:643468. doi: 10.3389/fphys.2021.643468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Blair GW, Thrippleton MJ, Shi Y, Hamilton I, Stringer M, Chappell F, Dickie DA, Andrews P, Marshall I, Doubal FN, et al. Intracranial hemodynamic relationships in patients with cerebral small vessel disease. Neurology. 2020;94:e2258–e2269. doi: 10.1212/WNL.0000000000009483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lee BC, Tsai HH, Huang AP, Lo YL, Tsai LK, Chen YF, Wu WC. Arterial Spin Labeling Imaging Assessment of Cerebrovascular Reactivity in Hypertensive Small Vessel Disease. Front Neurol. 2021;12:640069. doi: 10.3389/fneur.2021.640069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Claassen J, Thijssen DHJ, Panerai RB, Faraci FM. Regulation of cerebral blood flow in humans: physiology and clinical implications of autoregulation. Physiol Rev. 2021;101:1487–1559. doi: 10.1152/physrev.00022.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Nogueira RC, Aries M, Minhas JS, N HP, Xiong L, Kainerstorfer JM, Castro P. Review of studies on dynamic cerebral autoregulation in the acute phase of stroke and the relationship with clinical outcome. J Cereb Blood Flow Metab. 2021:271678X211045222. doi: 10.1177/0271678X211045222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Nogueira RC, Lam MY, Llwyd O, Salinet ASM, Bor-Seng-Shu E, Panerai RB, Robinson TG. Cerebral autoregulation and response to intravenous thrombolysis for acute ischemic stroke. Sci Rep. 2020;10:10554. doi: 10.1038/s41598-020-67404-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Castro P, Azevedo E, Serrador J, Rocha I, Sorond F. Hemorrhagic transformation and cerebral edema in acute ischemic stroke: Link to cerebral autoregulation. J Neurol Sci. 2017;372:256–261. doi: 10.1016/j.jns.2016.11.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Tian G, Ji Z, Huang K, Lin Z, Pan S, Wu Y. Dynamic cerebral autoregulation is an independent outcome predictor of acute ischemic stroke after endovascular therapy. BMC Neurol. 2020;20:189. doi: 10.1186/s12883-020-01737-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Beishon L, Minhas JS, Nogueira R, Castro P, Budgeon C, Aries M, Payne S, Robinson TG, Panerai RB. INFOMATAS multi-center systematic review and meta-analysis individual patient data of dynamic cerebral autoregulation in ischemic stroke. Int J Stroke. 2020;15:807–812. doi: 10.1177/1747493020907003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Schrader J, Luders S, Kulschewski A, Hammersen F, Plate K, Berger J, Zidek W, Dominiak P, Diener HC, Group MS Morbidity and Mortality After Stroke, Eprosartan Compared with Nitrendipine for Secondary Prevention: principal results of a prospective randomized controlled study (MOSES) Stroke. 2005;36:1218–1226. doi: 10.1161/01.STR.0000166048.35740.a9. [DOI] [PubMed] [Google Scholar]
  • 114.Kim BJ, Kwon SU, Park JH, Kim YJ, Hong KS, Wong LKS, Yu S, Hwang YH, Lee JS, Lee J, et al. Cilostazol Versus Aspirin in Ischemic Stroke Patients With High-Risk Cerebral Hemorrhage: Subgroup Analysis of the PICASSO Trial. Stroke. 2020;51:931–937. doi: 10.1161/STROKEAHA.119.023855. [DOI] [PubMed] [Google Scholar]
  • 115.Hankey GJ. CSPS.com Trial of Adding Cilostazol to Antiplatelet Therapy to Reduce Recurrent Stroke. Stroke. 2020;51:696–698. doi: 10.1161/STROKEAHA.119.028409. [DOI] [PubMed] [Google Scholar]
  • 116.Dawson J, Broomfield N, Dani K, Dickie DA, Doney A, Forbes K, Houston G, Kean S, Lees K, McConnachie A, et al. Xanthine oxidase inhibition for the improvement of long-term outcomes following ischaemic stroke and transient ischaemic attack (XILO-FIST) - Protocol for a randomised double blind placebo-controlled clinical trial. Eur Stroke J. 2018;3:281–290. doi: 10.1177/2396987318771426. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES