You are on page 1of 74

REVIEW ARTICLE

REGULATION OF CEREBRAL BLOOD FLOW IN


HUMANS: PHYSIOLOGY AND CLINICAL
IMPLICATIONS OF AUTOREGULATION
Cerebral Autoregulation AUTHORS
Jurgen A. H. R. Claassen, Dick H. J. Thijssen,
Static Dynamic
Vascular Diameter
Ronney B. Panerai, Frank M. Faraci

CBF 100 100


BP - CBF

50 50

BP - CBF
CORRESPONDENCE
0 0
0 50 100 150 jurgen.claassen@radboudumc.nl
(mmHg) Time

Chronic or steady state Rapid changes in BP


changes in BP

KEY WORDS
Alzheimer’s disease; hypertension; stroke;
microcirculation; neurovascular coupling

CLINICAL HIGHLIGHTS
1. Lassen’s classic autoregulation curve is often misinterpreted to mean that cerebral blood flow (CBF) is always kept con-
stant. However, this is only true for relatively gradual changes in blood pressure (BP), for example, during the normal noc-
turnal fall in BP or reductions in BP following treatment of hypertension. For progressively faster changes in BP (minutes,
seconds), CBF becomes incrementally more unstable and may show large fluctuations. Such rapid changes in BP may
occur, for example, during surgery, critical illness, everyday postural changes, and some forms of exercise.
2. It is commonly thought that chronic hypertension and aging, which often occur together, cause impairment in autoregula-
tion, such that brain blood vessels adapt and may even become dependent on higher BP to maintain perfusion. This mis-
conception carries the risk of undertreatment of hypertension, especially in older patients. Increasing evidence suggests
autoregulation is maintained in hypertension and aging, and treatment to lower BP is not associated with reductions in
CBF.
3. Autoregulation, CO2 reactivity, and neurovascular coupling (NVC) are sometimes confused or used interchangeably. For
example, a study that finds impairment in NVC may be reported as impaired autoregulation. However, impaired NVC or
CO2 reactivity do not automatically translate into impairment in how CBF responds to changes in BP (i.e., autoregulation).

CLAASSEN ET AL., 2021, Physiol Rev 101: 1487–1559


March 26, 2021; Copyright © 2021 the American Physiological Society
https://doi.org/10.1152/physrev.00022.2020
Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
Physiol Rev 101: 1487–1559, 2021
First published March 26, 2021; doi:10.1152/physrev.00022.2020

REVIEW ARTICLE

REGULATION OF CEREBRAL BLOOD FLOW IN


HUMANS: PHYSIOLOGY AND CLINICAL
IMPLICATIONS OF AUTOREGULATION
Jurgen A. H. R. Claassen,1 Dick H. J. Thijssen,2,3 Ronney B. Panerai,4,5 and Frank M. Faraci6
1
Department of Geriatrics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, The
Netherlands; 2Department of Physiology, Radboud Institute for Health Sciences, Nijmegen, The Netherlands; 3Research Institute for
Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom; 4Department of Cardiovascular Sciences,
University of Leicester, Leicester, United Kingdom; 5National Institute for Health Research Leicester Biomedical Research Centre,
University of Leicester, Leicester, United Kingdom; and 6Departments of Internal Medicine, Neuroscience, and Pharmacology, Carver
College of Medicine, University of Iowa, Iowa City, Iowa

Abstract
Brain function critically depends on a close matching between metabolic demands, appropriate delivery of
oxygen and nutrients, and removal of cellular waste. This matching requires continuous regulation of cere-
bral blood flow (CBF), which can be categorized into four broad topics: 1) autoregulation, which describes
the response of the cerebrovasculature to changes in perfusion pressure; 2) vascular reactivity to vasoac-
tive stimuli [including carbon dioxide (CO )]; 3) neurovascular coupling (NVC), i.e., the CBF response to
2
local changes in neural activity (often standardized cognitive stimuli in humans); and 4) endothelium-de-
pendent responses. This review focuses primarily on autoregulation and its clinical implications. To place
autoregulation in a more precise context, and to better understand integrated approaches in the cerebral
circulation, we also briefly address reactivity to CO and NVC. In addition to our focus on effects of perfu-
2
sion pressure (or blood pressure), we describe the impact of select stimuli on regulation of CBF (i.e., arterial
blood gases, cerebral metabolism, neural mechanisms, and specific vascular cells), the interrelationships
between these stimuli, and implications for regulation of CBF at the level of large arteries and the microcir-
culation. We review clinical implications of autoregulation in aging, hypertension, stroke, mild cognitive
impairment, anesthesia, and dementias. Finally, we discuss autoregulation in the context of common daily
physiological challenges, including changes in posture (e.g., orthostatic hypotension, syncope) and physical
activity.
Alzheimer’s disease; hypertension; stroke; microcirculation; neurovascular coupling

also named the “foramen of Monro” in the brain’s ven-


1. INTRODUCTION: REGULATION... 1487 tricles, deduced the brain’s high demand for blood flow.
2. AUTOREGULATION: THE STATIC... 1492 He did so by taking the weight of the arm and the diame-
3. OTHER MECHANISMS THAT REGULATE... 1504 ter of the subclavian artery, its source of blood supply,
4. OTHER FACTORS THAT INFLUENCE... 1510 and comparing this with the weight of the brain and its
5. REGULATION OF CEREBRAL BLOOD FLOW... 1512 blood supply, the combined diameters of the carotid
6. IMPACT OF DAILY LIFE CHALLENGES ON... 1531 and vertebral arteries (2). Monro, however, is perhaps
7. SUMMARY AND CONCLUSION 1536 best remembered for the Monro-Kellie doctrine (3),
which states that because the brain is enclosed in the
skull, there must be an equilibrium between its volumet-
ric components (brain parenchyma, interstitial and cere-
1. INTRODUCTION: REGULATION OF brospinal fluid, and arterial and venous blood volume).
CEREBRAL BLOOD FLOW IN HUMANS In the early 1900s, this doctrine led to the view that there
was no regulation of cerebral blood flow (CBF). Rather,
1.1. Historical Perspective CBF, and changes in CBF, were restrained by this equi-
librium. In other words, large increases in CBF were
Some of the earliest knowledge regarding cerebrovas- thought to be impossible because it would cause dise-
cular anatomy, and specifically the circle of Willis, dates quilibrium in intracranial pressure (ICP) (3), a view sup-
back to 1664 when Thomas Willis published his thesis ported by the influential physiologists Hill (4) and Bayliss
“Cerebri Anatome” (1). In 1783, Alexander Monro (2), who et al. (5). Their idea was that there was no active control

0031-9333/21 Copyright © 2021 the American Physiological Society 1487


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

An experiment by Roy and Sherrington in 1890 (9),


CLINICAL HIGHLIGHTS
wherein they injected a solution of homogenized
1. Lassen’s classic autoregulation curve is often misinterpreted to brain intravenously in a dog, revealed an increase in
mean that cerebral blood flow (CBF) is always kept constant. brain volume (used as an index of increased CBF)
However, this is only true for relatively gradual changes in blood without any increase in BP. This experiment formed
pressure (BP), for example, during the normal nocturnal fall in BP the first evidence for the metabolic hypothesis of
or reductions in BP following treatment of hypertension. For pro-
gressively faster changes in BP (minutes, seconds), CBF NVC (3). The interpretation was that this brain extract,
becomes incrementally more unstable and may show large fluc- from a dog that had died from hemorrhagic shock 4 h
tuations. Such rapid changes in BP may occur, for example, dur- earlier, contained chemical substances that induced
ing surgery, critical illness, everyday postural changes, and some
cerebral vasodilation. They hypothesized these sub-
forms of exercise.
2. It is commonly thought that chronic hypertension and aging, stances had been located in the perivascular fluid. Of
which often occur together, cause impairment in autoregulation, note, however, this experiment could not be repli-
such that brain blood vessels adapt and may even become de- cated by Bayliss et al. (5).
pendent on higher BP to maintain perfusion. This misconception
carries the risk of undertreatment of hypertension, especially in
These early experiments (by Donders, Mosso, Roy,
older patients. Increasing evidence suggests autoregulation is and Sherrington) were largely ignored in their time, or
maintained in hypertension and aging, and treatment to lower BP seen as incorrect, because Hill (4) found contradictory
is not associated with reductions in CBF. results and was convinced there was no relationship
3. Autoregulation, CO2 reactivity, and neurovascular coupling (NVC)
are sometimes confused or used interchangeably. For example, a
between brain function and its circulation and therefore
study that finds impairment in NVC may be reported as impaired no active regulation of CBF (10).
autoregulation. However, impaired NVC or CO2 reactivity do not While the study by Roy and Sherrington from 1890 (9)
automatically translate into impairment in how CBF responds to
is often cited for the experiment (based on just one ani-
changes in BP (i.e., autoregulation).
mal) that suggested the presence of NVC, their paper
contains many other experiments and represents one of
of CBF, rather CBF passively followed changes in blood the more comprehensive publications on early integra-
pressure (BP). If the brain demanded an increase in CBF, tive physiology of the cerebral circulation. In a series of
this was achieved by vasoconstriction in the rest of the studies using dogs, cats, and rabbits, they investigated
body, increasing BP and thereby CBF (4). effects of sensorimotor stimulation, vagal-sympathetic
In 1868, the physiologist F. C. Donders (6) may have stimulation, brain stem stimulation, hypoxia, hemor-
been the first to hint at active regulation of CBF in rhagic shock, acid-base changes, and several drugs (9).
response to cognitive activation (i.e., neuronal stimula- They may also have been the first to indirectly describe
tion), a concept now referred to as neurovascular cou- autoregulation, as will be discussed in sect. 2.1. It is also
pling (NVC) or functional hyperemia (see also sects. 1.3 interesting to read their description and recognition of
and 3.1 for further information on NVC). Donders (6) brain lymphatics and perivascular spaces, topics for
wrote “[because]. . . hemorrhage, or reduced cardiac which there is now renewed interest (11, 12).
function, are associated with loss of consciousness [we In 1895, Bayliss et al. (5) reported contrasting findings
conclude] that a regular supply of blood to the brain is a compared with the work from Roy and Sherrington (9).
prerequisite for cognitive processes, indicating a central Bayliss and colleagues (5) examined the brain circulation
role for brain metabolism [in cognitive activation]. . . by performing simultaneous recordings of arterial and
which consumes oxygen and produces carbon dioxide.” venous pressure, intracranial pressure (ICP), and cere-
In the late 19th century, the physiologist Angelo Mosso bral venous pressure with a design comparable to Roy
(7) published the first work related to regulation of CBF and Sherrington. They concluded “there is no evidence
in humans. Mosso recorded pulsations of the brain in a supporting the existence of cerebral local vasomotor
patient with skull defects following a neurosurgical pro- mechanisms”, and that “the cerebral circulation pas-
cedure, in which Mosso reported immediate increases sively follows the changes in the general arterial and ve-
in pulsations whenever the subject was spoken to, or nous pressures” (5). Bayliss is well known for the
when the subject began to think actively (e.g., arithme- “Bayliss effect,” which refers to the response of vascular
tic). Mosso (8) also introduced the “human circulation muscle to changes in pressure (see sect. 3.4) based on
balance,” which represents a measurement technique experiments in the peripheral circulation but not the cer-
using a balanced table that tipped downwards if the ebral circulation. Indeed, combining experiments,
weight on either end were increased. Using this bal- Bayliss et al. (5) concluded that “there is, up to the pres-
ance, he found that the moment emotional or intellectual ent, no satisfactory evidence of the presence of a vaso-
activity began in a subject, the balance would tip down motor supply to the brain; so that when an increased
at the head end, reflecting a redistribution of blood to- blood-supply is required in that organ it is provided by
ward the brain. constriction of vessels throughout the rest of the body,

1488 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

which is, in this respect, so to speak the slave of the (TCD) ultrasound in the 1980s (23, 24) helped resolve this
brain.” The Bayliss effect was in his opinion not present limitation because of its very high temporal resolution,
in the brain but only in the systemic circulation, most the method has its own limitations in that it measures
notably the splanchnic circulation. blood flow velocity, without knowledge of vessel diame-
Contrary to the belief of Hill and Bayliss, the concept ter (or cross-sectional area). Therefore, any changes in
that the cerebral circulation is responsive to neuronal vessel diameter during interventions and measurements
activation and changes in BP, as earlier suggested by affect estimates of CBF using TCD.
Donders and Mosso and (indirectly) Roy and Sherrington,
was confirmed in subsequent decades (13–15) [see 1.2. Relevance of CBF for Brain Function and
review by Rosenblum (16)]. This research may have been Consequences of Hypoperfusion
sparked by work from Fulton (17), who obtained evidence
of NVC in the occipital cortex upon visual stimulation in a The vertebrate brain is a unique organ. In the human,
patient with a cortical defect, very similar to the work of the brain represents only 2–3% of total body mass while
Mosso (3, 7, 10). requiring 15% of cardiac output and consuming 20%
Although the observations by Mosso (7) in humans of the available O2 under normal conditions. The high
are now 140 yr old, thorough insight into the integrative metabolic rate of the brain, combined with limited
nature of the regulation of CBF in humans has proven energy stores, highlights the importance of CBF for nutri-
difficult for a variety of reasons. For many years, the ent and O2 delivery but also for removal of cellular, met-
presence of the skull made the brain something of a abolic, or toxic by-products. The importance of
“black box” as it prohibited direct observation of blood controlling CBF is highlighted by the acute consequen-
vessels, measurements of blood flow, or blood flow- ces that occur following substantial reductions in CBF, a
mediated changes in volume or pulsatility, approaches change that can rapidly lead to unconsciousness, life-
that were feasible in the systemic circulation. threatening complications and brain damage if sus-
A milestone in this field was the nitrous oxide method tained (25, 26). In a clinical context, acute reductions in
developed by Seymour Kety in 1948 (18, 19). This method global CBF can occur with cardiac arrest (26) or with a
applies the Fick principle (conservation of mass) and sudden profound reduction in BP, which can lead to
measures changes in cerebral arterial and venous con- global reductions in CBF, termed syncope (for a recent
centrations of nitrous oxide following its inhalation. The review and overview of guidelines on syncope, see Ref.
Kety-Schmidt method yields an estimate of CBF aver- 27 and sect. 6.1.2). Focal reductions in CBF occur in
aged over 10–15 min. An adaptation of this method was stroke, where there is an acute vessel occlusion leading
introduced by Lassen and Ingvar in 1961 (20), who used to hypoperfusion and ischemia. Examples are the classic
intra-arterial injection of radioactive Krypton-85 or clinical stroke of the middle cerebral artery or down-
Xenon-133. Using a camera, the intracerebral concentra- stream microinfarcts in the case of small vessel disease
tion of the tracer could be recorded to derive both or migrating microthrombi. Experiments in nonhuman
global CBF and estimates of regional CBF. Other adap- primates and other preclinical models have shown that a
tations of the Kety-Schmidt method have been adopted reduction in CBF to below 50% of normal induces an
since, for example, in position emission tomography immediate loss of neuronal function (25, 26). Normal
(PET) and single-photon emission computed tomogra- global CBF in awake humans is 50 mL/100 g/min, and
phy (SPECT). loss of function is observed below 22 mL/100 g/min
A second important development was the introduction (25). This loss of function is responsible for the clinical
of advanced techniques from the 1970s onwards that symptoms of hypoperfusion, which include slowing of
could be applied safely in humans, starting with com- mental processing. These symptoms are followed by
puted tomography (CT), followed by PET and magnetic loss of consciousness, often within 10 s after the onset of
resonance imaging (MRI) (21, 22). These techniques have hypoperfusion or ischemia (26). With rapid recovery of
significantly contributed to improved understanding of CBF, function can be restored without permanent cellu-
the anatomy, physiology, and pathophysiology of the lar injury. In contrast, persistent lowering in CBF causes
cerebrovascular system and currently play a key role in irreversible damage. There are no exact thresholds
diagnosis and research focused on diseases that affect beyond which ischemic injury occurs, but estimates are
the cerebral circulation. Despite continuous technological that a sustained fall to 20–30% of baseline CBF causes
advances that provided more detailed insight, these tech- ischemia within minutes (26, 28, 29). This estimation is
niques are still limited by a poor temporal resolution, that obviously an oversimplification, because effects of is-
would be needed to record rapid changes in CBF in chemia vary between different populations of brain cells,
response to physiological or pathophysiological stimuli between the core and penumbra in the case of stroke,
(21). Although the introduction of transcranial Doppler and because apart from these immediate effects of

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1489


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

hypoperfusion on cell function, a cascade of responses autonomic nervous system. In addition, hypertension
are triggered that lead to further damage and delayed incr-eases the risk of orthostatic hypotension, which
ischemia, hours to days later (26). A detailed description poses an additional challenge to autoregulation (see
of such changes is beyond the scope for this review, we sects. 6.1.1 and 6.1.2). This example, focusing on hyper-
refer to the extensive review by Lipton (26). tension, highlights the importance of understanding the
Other than acute effects of ischemia, an increasing lit- integrative nature of CBF regulation and how alterations
erature describes the clinical relevance of chronic, but in BP may contribute to the development and progres-
more modest, reductions in CBF that potentially contrib- sion of a series of clinical conditions that share a final
ute to loss of brain health in neurodegenerative dis- common pathway involving CBF (FIGURE 1).
eases such as Alzheimer’s disease, vascular dementia,
or mixed dementia (see sect. 5). Here, hypertension is 1.3. Integrative Approach to Regulation of CBF
suggested to play a central role through its effects on
CBF. Hypertension is a key risk factor for overall disease Control of CBF involves a spectrum of overlapping regu-
burden and health loss worldwide (30, 31). Hypertension latory mechanisms that collectively facilitate optimal O2
is associated with chronic mild cerebral hypoperfusion, and nutrient delivery to individual brain cells (21, 33).
which may explain why hypertension is a risk factor for Mechanisms that influence CBF include arterial blood
Alzheimer’s disease. Recent research (see sect. 5.2) gases [partial pressure of arterial CO2 (PaCO2 ) and arte-
suggests that BP-lowering treatment in hypertension rial (PaO2 )] tissue levels of PCO2 and PO2 (34, 35), central
may restore CBF to normal, which may explain why hemodynamics (BP, including effects of hydrostatic pres-
antihypertensive treatment can reduce the risk of sure gradients), cerebral metabolism, and neural mecha-
Alzheimer’s disease. This clinical example illustrates the nisms including extrinsic autonomic and sensory nerves,
complexity and implications of regulation of cerebral and intrinsic neurons in close association with the vascu-
perfusion. Both hypertension itself and medications lature within the brain parenchyma (36). In addition to
used to treat hypertension can affect CBF, most likely their individual impact, there are strong interactions
via effects on vascular function, vascular structure, or between these regulators. Some of these mechanisms
vascular mechanics (32), as well as effects on the are unique to the brain, explaining the generalized

A Static Dynamic B Modifiers of


Autoregulation
Vessel Diameter
100 100 • hemodynamics
• O2, CO2, pH
CBF
• endothelium
50 50 • neurons
• metabolism
BP - CBF • vascular stiffness
0 0 • vascular structure
0 50 100 150
Time • behavior
BP (mmHg) • posture
• genetics
C D • temperature
Functional Autoregulation Dysfunctional Autoregulation • vascular risk factors
• vascular disease

• regulation of CBF • hypoperfusion or ischemia


• regulation of microvascular pressure • syncope
• protection of the BBB • hyperperfusion
• altered microvascular pressure
• loss of BBB integrity + edema
• microbleeds
• cognitive function
• non-cognitive function Brain Health
• cognitive dysfunction
• non-cognitive dysfunction

FIGURE 1. Overview of cerebral autoregulation. General overview of autoregulation and key summary points for this review. A: 2 graphs illustrating
the concepts of static and dynamic autoregulation. B: summary of modifiers or factors that have a direct influence on cerebral blood flow (CBF) or on
the relationship between blood pressure (BP) and CBF (i.e., autoregulation). Examples include hemodynamics (including the rate of rise or temporal pro-
file of changes in BP) and behavior (sedentary versus exercise). C: summary of the impact of normally functioning autoregulation. D: summary of conse-
quences of impairment in autoregulation in relation to brain health. See text for further details. BBB, blood brain barrier.

1490 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

observation that the peripheral vasculature represents a effects of autonomic and sensory nerves (extrinsic inner-
poor model or predictor for the cerebral vasculature in vation) on the extraparenchymal segments of the cere-
health or disease (37). bral vasculature (36).
An important characteristic of CBF regulation is mech- A fourth mechanism relates to the effects of vascular
anistic redundancy, i.e., overlapping mechanisms con- endothelial cells on vascular tone and therefore CBF. In
tribute to maintaining CBF under highly challenging response to hemodynamic stimuli (e.g., shear stress),
conditions. Studies exploring the regulation of CBF are ions, neurotransmitters, metabolic stimuli, and therapeu-
importantly impacted by this, because the overlap in tic agents, among other factors (32, 46–49). This ability
pathways makes it difficult to explore the relative impor- of endothelial cells to cause vasorelaxation importantly
tance of individual pathways or identify key contributors. contributes to NVC. Studies using selective injury of en-
From a teleological perspective, this redundancy makes dothelium or cell-specific genetic manipulation have
the regulation of CBF a robust system where multiple shown that this cell plays an essential role in propagated
strategies are present to ensure precise control and (or ascending) vasodilation during NVC, eventually
thus protect against potential brain damage. reaching arterioles and arteries on the pial surface (49,
The regulation of CBF can be affected by certain 50). Other studies that examined genetic alterations of
processes in the rest of the body, because they signaling molecules in endothelial cells but did not focus
can influence the sensitivity of the various systems on propagated vasodilation provide further direct evi-
that affect CBF. For example, PaCO2 and PaO2 are dence that endothelial cells play a major role in the vas-
determined primarily by pulmonary gas exchange, cular component of NVC in pial and parenchymal
local lung perfusion, and body posture, yet they have arterioles (51, 52). While still supporting a key role for en-
a major influence on cerebrovascular resistance (34, dothelium, one study (52) provided some evidence
35) (FIGURE 1). against a role for endothelial cells in propagated vasodi-
Using a simplified approach, one can divide mecha- lation (from capillaries to small parenchymal arterioles) in
nisms that regulate CBF into four distinct components or that vasodilation in response to whisker stimulation in
adaptive responses: autoregulation, chemoregulation, mice occurred in precapillary arterioles before changes
neuronal regulation, and endothelium-dependent regu- in diameter of capillaries. Endothelial dysfunction, a fea-
lation. The first mechanism, termed autoregulation, ture of many forms of large and small vessel disease,
relates to the response of the cerebral circulation to can therefore have multiple consequences, not only on
changes in cerebral perfusion pressure (21, 33, 38–42). CBF, but also on other endpoints related to brain health.
Cerebral perfusion pressure is determined by BP, ICP, Unlike myogenic responses (discussed below), in
and venous pressure. Under physiological conditions, which a single cell type mediates most (or all) of the
cerebral perfusion pressure is mainly determined by ar- response to changes in transmural pressure, endothe-
terial BP and body posture (supine or prone vs. seated lium-dependent responses cannot occur without endo-
or standing in humans), because venous BP and ICP are thelial cells exerting effects on vascular muscle or other
relatively low and normally exhibit only small changes. cellular targets via molecular or electrical signaling.
Some key exceptions are traumatic brain injury or intra- Furthermore, endothelium-dependent effects on plate-
cranial hemorrhage (ICH), where ICP can be substan- lets, immune cells, neurons, and glia, each involve target
tially elevated and affects perfusion pressure. Cerebral cells, highlighting that this signaling is not based on a
autoregulation (henceforward referred to simply as single cell response (45).
“autoregulation”) will be the focus of this review and will As a final short note to illustrate the complexity of en-
be discussed in detail in sects. 1, 5, and 6. dothelial function in the brain, endothelial cells normally
The second mechanism is chemoregulation, another also inhibit processing of b-amyloid and phosphorylation
class of vascular reactivity. This includes vascular of tau: both major contributors to Alzheimer’s disease
responses to changes in CO2 (and subsequently pH), (48, 53).
PO2, or O2 content. Changes in these variables (e.g., hy- Because autoregulation, i.e., the focus of this review,
pocapnia and hypercapnia) elicit strong CBF responses does not operate in isolation, we have attempted to
(34, 35, 43). Of these mechanisms, reactivity to CO2 has present an integrative approach, briefly discussing other
been the most widely studied in controlled laboratory- interacting mechanisms. Studies of autoregulation need
based setting in humans and has also been termed cer- to consider overall integrated responses. For example,
ebral vasomotor reactivity to CO2 (35). many studies of autoregulation only consider effects of
The third mechanism consists of the influence of BP on CBF. However, other parameters affecting CBF
nerves or neurons on CBF. This includes NVC, the local when measurements are made cause “physiological
CBF response to nearby intrinsic neuronal activation noise” in the BP-CBF relationship (54). For this reason,
within the brain parenchyma (44, 45), but also includes studies of autoregulation need to control variables such

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1491


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

as the amount of exercise and caffeine intake before (45, 59). The second mechanism is the perivascular
testing, room temperature, cognitive activation, or men- pathway. Here, toxins and waste that have diffused into
tal stress during the experiment and measure PaCO2 or the perivascular spaces that surround penetrating arte-
end-tidal CO2 during experiments to ascertain if this pa- rioles and are transported alongside these same arterio-
rameter remained stable or to take changes in CO2 into les toward arteries on the surface of the brain, followed
consideration when interpreting results (55). by drainage into cerebral lymphatics (located in the dura
Another reason for this integrative approach is that mater and meninges) and eventually into cervical lymph
mechanisms can be confused in the literature. For exam- nodes (45, 59). Pulsations in blood flow, influenced by
ple, some studies use the term autoregulation when the dynamics of vascular distention, or spontaneous vas-
describing vasomotor reactivity to CO2. Others use omotion, may have a role as a pump that drives this peri-
“pressure reactivity” to describe autoregulation, which vascular flow of brain interstitial fluid (ISF) (11, 62, 63).
can be confused with CO2 reactivity. The third proposed mechanism is paravascular transpor-
tation. This concept has recently been termed the “glym-
1.4. Other Roles for the Cerebral Circulation phatic system” (11). Here, cerebral spinal fluid (CSF) is
outside the Scope of This Review hypothesized to travel into the brain parenchyma, along-
side penetrating arteries and arterioles (therefore also in
The focus of this review is on the role of the cerebral the perivascular space, but in opposite direction from
circulation in delivering blood flow to the brain, with the perivascular pathway), where it would clear the ISF
the main purpose of delivering O2 and removing CO2. and then exit alongside venous perivascular spaces.
Other functions of the cerebral circulation receive little This paravascular pathway is thought to rely on aqua-
attention in this review. For example, the blood-brain porin-4 channels in astrocytic end-feet in the perivascu-
barrier (BBB) plays an essential role in controlling or lar spaces, to transport the CSF into the ISF (45, 59).
limiting the movement of ions, molecules, amino acids, Changes in hemodynamics, structure, or mechanics of
nutrients, cells, and so forth, into and out of the brain the cerebral circulation may affect these three pathways
parenchyma (56–58). Damage or dysfunction of the in several ways. For example, changes in the pulsatile
BBB can be caused by aging, abnormal hemodynam- dynamics, which occurs in hypertension with increased
ics (i.e., acute hypertension), and various diseases (e. vascular stiffness, may affect perivascular transportation.
g., stroke, chronic hypertension, dementias, traumatic A feature of small vessel disease are changes in the mi-
brain injury, multiple sclerosis, neuroinflammation, and crovascular perivascular spaces, associated with hyper-
so forth) (57, 58). Such damage or dysfunction can in tension and expansion of perivascular spaces (Virchow-
turn have multiple consequences, such as impaired Robin spaces), which may also contribute to impaired
transport of essential substances into the brain, ISF dynamics (12). Using an animal model, others have
reduced removal of waste products or toxins from the suggested recently that spontaneous slow oscillations
brain, and the entrance of molecules or cells (e.g., tox- (vasomotion) in arteriolar diameter may drive clearance
ins, infectious agents, drugs, proteins, and immune or drainage of solutes from the brain parenchyma (64).
cells) that are neurotoxic and/or cause cellular dam- This clearance was enhanced when vasomotion was
age. This topic is outside the scope of this review, so increased by visual activation of the occipital cortex and
we refer to other recent reviews for further reading was reduced in the context of vessel wall dysfunction (e.
(57, 58). g., cerebral amyloid angiopathy).
Another proposed function of the cerebral circulation
that has reemerged recently and has some overlap with
BBB function relates to clearance of waste and toxins 2. AUTOREGULATION: THE STATIC AND
from the brain (45, 59). Three mechanisms, or pathways, DYNAMIC RESPONSES OF CBF TO
have been proposed to play key roles. It should be CHANGES IN BP
noted that there is still controversy regarding the physio-
logical and pathophysiological role of these pathways 2.1. Historical Perspective
(60, 61).
One mechanism is the transvascular pathway, where Lassen’s publication in 1959 (38) in this journal can be
toxins are actively transported across the BBB, from the marked as the “birth” of autoregulation research in
brain and into the blood. An example is the transporta- humans, although Roy and Sherrington (9) provided a
tion of amyloid-b from the perivascular space into the description of what we now call autoregulation in 1890.
blood vessel by low-density receptor-related protein Roy and Sherrington were unable to directly measure
(LRP1). Amyloid-b and other waste/toxins reach the peri- CBF, but estimated changes in CBF from changes in
vascular space by diffusion from the brain parenchyma brain volume or increases in cerebral venous pressure.

1492 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

They noted that changes in CBF could be smaller than CBF values for a set of patients with a specific BP
changes in BP or show an earlier return toward baseline level, linked to specific medical conditions. Lassen’s
(9). Others such as Fog (13) provided early data that con- curve nonetheless was commonly interpreted as
firmed and extended the concept of autoregulation. reflecting what would happen to CBF if, in a group of
Using a cranial window approach in anesthetized cats, people or within a single individual, BP was reduced
Fog (13) described rapid changes in the diameter of pial or increased across the wide range depicted in the
arteries and arterioles in response to experimental figure.
manipulation in arterial BP. More direct evidence to support this interpretation
Lassen’s paper (38) introduced a more integrative of the autoregulatory curve came after this original
approach of linking central (systemic) hemodynamics publication, consisting mainly of findings from pre-
and regulation of CBF. Lassen presented a now very clinical models. A series of generally well-controlled
well-known figure that plotted CBF against BP for 11 experiments in nonhuman primates and common lab-
groups of patients with varying levels of BP. That figure oratory species confirmed the substantial autoregu-
revealed a plateau in CBF across a relatively large range latory capacity of the cerebral circulation, where
of BP (mean arterial BP between 50 and 150 mmHg) despite large changes in BP, CBF is held relatively
and was the first description of effective maintenance of stable (66–80). FIGURE 2 provides examples of such
CBF over a considerable range of BP, a concept that experiments (67, 72, 73, 75, 77, 79, 81–83). All these
Lassen termed autoregulation, to suggest the domi- studies reveal a range of BP in which CBF was rela-
nance of local mechanisms. Lassen’s conclusions tively stable. In some studies, there is a portion of the
regarding autoregulation, in combination with many sub- curve where a slope is not apparent. In others, there
sequent studies in the following decades, evolved into a is a small positive slope: an increase in CBF as BP is
widely accepted concept in vascular biology and medi- increased. Upper and lower limits of autoregulation
cine regarding regulation of CBF in humans and in pre- are suggested or can be seen in some of these data
clinical models. His work continues to be cited frequently (FIGURE 2).
in the literature. In conclusion, if one combines animal and human ex-
perimental data (discussed below), the overall concept
2.2. Critical Appraisal of Lassen’s Autoregulation of Lassen’s autoregulatory curve can be maintained,
Curve albeit with some important caveats and modifications.
The existence of an autoregulatory range of BP, where
Lassen’s original curve was an extrapolation, based autoregulation is highly effective and where CBF is rela-
on a cross-sectional comparison of 376 individuals tively stable, is undisputed. We suggest it should no lon-
from 7 publications. It featured a straight line drawn ger be called a “plateau phase,” because this implies a
across average CBF data from 9 of the 11 groups, horizontal line with zero slope. In reality, the relationship
which varied around a mean CBF of 55 mL/100g/ between BP and CBF within this autoregulatory range
min, for mean arterial BP between 50 and can vary between an upward or even a downward slope
175 mmHg. The graph also featured a downward line (85–87). A better description than “plateau” therefore
that connected two data points well below and to might be the term “gradient.” We suggest a more pre-
the left of the horizontal line, from two studies with cise description for this “gradient phase of autoregula-
mean CBF of 30 mL/100 g/min and mean BP values tion” would be that there is a range of BP where
of 40 mmHg. Lassen’s interpretation of these latter autoregulation minimizes variations in CBF when BP is
two data points was that they marked the “lower altered. How wide this range may be cannot be pre-
limit of autoregulation.” There were no data at cisely defined from the available studies, as a large num-
higher BP values, and therefore no “upper limit of ber only looked at one end of the autoregulatory curve
autoregulation”; that concept was added later. or a limited range of change in BP. Studies that would
Questions have been raised about the validity of be needed to obtain such data may be impossible to
some of the data points used to fit Lassen’s curve perform in humans, for both physiological and ethical
(65). For example, one of the seven publications reasons. Even if such data did exist, it is unlikely that a
could not be traced and verified, and one of the data group-averaged range would apply to all individuals.
points had been plotted in error. A corrected plot What we do know, however, is that in most studies rela-
would no longer feature a straight line but a line with a tively slow reductions or increases in mean arterial BP
slight upward slope (65). Furthermore, it is important to re- between approximately 20 and 120 mmHg are asso-
alize that the original curve contained no within-subject ciated with a stable CBF in humans (for details and refer-
data on changes in BP with repeated measurements of ences, see sect. 5.2.2 on hypertension and static
CBF. The curve consisted entirely of group averaged autoregulation).

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1493


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

Dog Rabbit Cat

100 100 100

50 50 50
Cerebral Blood Flow (ml/100g/min)

0 0 0
0 50 100 150 200 0 50 100 150 200 0 50 100 150 200

150
Baboon Mouse 250 Rat
100
200
100
150

50
100
50

50

0 0 0
0 50 100 150 200 0 50 100 150 200 0 50 100 150 200

Mean Arterial Pressure (mmHg)

FIGURE 2. Static autoregulation: summary of animal models. Data for a range of experimental models, replotted from the original publications, illus-
trate the relationship between cerebral blood flow (CBF) and mean arterial pressure (MAP). Many studies either examined the low end or the high end
of the autoregulatory curve, but a few quantified both ends of the curve within a single publication. For all species, there was a substantial range of
MAP where CBF was relatively stable (72,73, 75, 77, 79, 81–84). For clarity, we have not plotted all studies of this type, but for reference cite some addi-
tional examples (66, 78, 80). For the panels with data from dogs, cats, baboons, and rats (filled circles), CBF is expressed in ml/min/100 g. For panels
with data from rabbits, mice, and rats (open circles), CBF is expressed as percent change with the control value set at 100.

2.3. The Importance of Time: Autoregulation Is Numan et al. (87) recently reanalyzed 40 studies in
Strongly Affected by the Rate at Which BP healthy humans that examined the relation between BP
Changes and CBF above and below resting mean BP. Largely in-
dependent of the technique used to assess CBF (i.e.,
One of the reasons why it is difficult to study a wide range TCD, MRI, PET, or Xenon-133 technique), these studies
of BP changes in humans is the efficacy of the baroreflex. confirmed that the cerebrovasculature has autoregula-
The baroreflex normally helps to maintain BP within a nar- tory capacity but not that CBF was completely stable
row range during normal daily activities and under patho- over an autoregulatory range. In fact, they reported a rel-
physiological conditions (88). As such, this neural reflex atively small range of mean BP that is associated with a
will limit or at least dampen large fluctuations in BP (88). stable CBF within subjects. Moreover, the efficacy to
Commonly, this only allows for the assessment of rela- regulate CBF seemed to differ based on the direction of
tively small changes in BP that can be maintained for a change in BP, with a more effective capacity to stabilize
longer period. An example is lowering or increasing BP CBF during acute (transient) periods of hypertension,
over weeks by starting or stopping antihypertensive med- compared with hypotension. However, the studies sum-
ication. Larger, but acute, changes in BP are difficult to marized in that review used a variety of techniques to
achieve and can only be maintained for shorter periods, increase and decrease BP, leading to potential con-
usually only during an experimental setting. Examples are founders, such as effects of medications, autonomic acti-
administration of intravenous drugs to acutely lower or vation, and differences in PaCO2 . Many studies applied
increase BP (86, 89). Although pharmacological manipu- BP changes over relatively short periods of time (i.e.,
lations are a common experimental strategy to alter BP, it minutes), as in the study by Liu et al. (86). This affects the
is important to realize that depending on the agent used shape of the classic autoregulation curve, because
and whether it activates endothelial cell receptors or these faster changes in BP lead to a smaller range of BP
crosses the BBB to reach vascular muscle, pharmacologi- wherein CBF is kept relatively stable (the “gradient
cal agents sometimes have direct effects on the cerebral phase” noted above) and to a steeper gradient within
vasculature, independent of changes in BP. that range. The importance of time in autoregulatory

1494 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

mechanisms is captured in the terms static and dynamic when applied to between-subject changes in BP and
autoregulation (90). Understanding these two concepts, CBF, CBF is held relatively stable between various levels
and the difference between them, is important for a of BP under normal physiological conditions. This inter-
proper understanding of autoregulation. pretation is a close approximation of Lassen’s data pre-
sentation and conclusions (see also FIGURE 2).
2.4. Static and Dynamic Autoregulation
2.4.2. Dynamic autoregulation.
2.4.1. Static autoregulation.
The concept of dynamic autoregulation only emerged
The classic relationship between mean arterial BP and once techniques with high temporal resolution became
CBF, derived from Lassen’s publication (38), should now available to measure faster changes in CBF (40–42).
be referred to as “static” autoregulation (40, 41, 90–94). Now, instead of values that took 10 min or more to
This does not relate to physiological characteristics per obtain, these techniques allowed for measurements of
se but rather refers to experimental characteristics that changes in BP and CBF (or CBF velocity) occurring over
only measure the steady-state relationship between seconds (96–98). It became clear that during more
CBF and BP. The term static refers to the concept that rapid changes in BP, there was much greater variability
BP and CBF are measured under conditions where indi- in CBF than was hitherto assumed based on Lassen’s
vidual variables (BP, CBF) have reached a steady state concept of static autoregulation (FIGURES 3 and 4).
(40, 95). BP may have been experimentally increased or The ability of autoregulation to respond to rapid
decreased, but it has reached a new stable, steady-state changes in BP across seconds or minutes (e.g., during
level (40–42). BP and CBF are measured over longer postural changes, coughing, or physical activity) is
time intervals (10 min or more), and the resulting values referred to as dynamic autoregulation (40, 41, 90–94)
represent the average BP and CBF during that period. (FIGURES 3, 4, and 5).
This approach was a consequence of the fact that histor- TCD to measure CBF velocity (typically using the middle
ically, measurements of CBF in humans required at least cerebral artery), with simultaneous beat-to-beat recordings
10 min to perform. Due to this averaging, both BP and of BP, has become a popular technique to measure
CBF will not display their short-term variability. With the dynamic autoregulation (24). Aaslid and coworkers (24,
use of this definition of static autoregulation, especially 101, 102) examined the temporal relationships between

Cerebral blood flow

Blood pressure

Time scale: weeks days hours minutes seconds

FIGURE 3. Effect of the time scale of blood pressure (BP) changes on autoregulation of cerebral blood flow (CBF). Schematic representation explain-
ing how the time period in which changes in BP occur affects autoregulation of CBF. Slower changes (e.g., weeks, such as with chronic hypertension or
treatment of hypertension) have minimal effects on CBF, whereas with more rapid changes the effects on CBF can increase such that there is essen-
tially no effective autoregulation with very fast BP changes occurring within seconds. This presentation represents a hypothetical model of the transi-
tion from static autoregulation (slow changes in BP) to dynamic autoregulation (faster changes in BP).

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1495


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

80

CBFV R (cm/s)
FIGURE 4. Oscillations in blood pressure (BP) and
40 cerebral blood flow (CBF) induced by repeated sit to
stand maneuvers. An example of repeated sit-to-stand
80
maneuvers to induce strong oscillations in BP and CBF
from a patient in a study on autoregulation in
Alzheimer’s disease (99). Starting with a baseline re-
cording while seated, the patient was asked to stand
up for 10 s, then to sit down for 10 s, followed by stand-

CBFVL (cm/s)
60
ing up for 10 s, and so on [using methods proposed pre-
viously (100)]. With a cycle duration of 20 s, large
oscillations in BP and CBF (blood velocity in the left and
right middle cerebral artery) are induced at 0.05 Hz, a
220 40 frequency where dynamic autoregulation is active (see
text for details). Changes in systolic BP, which had a
200 seated baseline value of 150 mmHg, oscillated
between 110 and 190 mmHg and CBF velocity, which
180
SBP (mmHg)

20 had a seated baseline value of 50 cm/s, oscillated


between 40 and 70 cm/s. SBP, systolic blood pressure
160
recorded using Finapres; CBFV, blood velocity in the
140 left (L) and right (R) middle cerebral arteries, measured
using transcranial Doppler.
120

100

0 30 60 90 120 150 180 210 240 270 300

Time (seconds)

BP and CBF in response to deflation of BP cuffs placed transmitted or blocked depending on the frequency,
around the thigh to induce rapid, transient hypotension and is comparable to a high-pass filter that transmits
. The resulting drop in BP was accompanied by a pro- high frequencies but blocks low frequencies (104, 105).
portional decrease in middle cerebral artery blood This relationship between CBF and BP is analogous
flow velocity, which was assumed to provide an accu- to the application in acoustics, where a high-pass filter
rate index for changes in CBF. In healthy individuals, transmits high frequency signals from an audio source
CBF recovered more quickly than BP. These observa- to a tweeter. This led Giller (106) to apply transfer func-
tions highlighted in humans that autoregulatory mech- tion analysis (TFA) to dynamic autoregulation, a math-
anisms were unable to maintain CBF perfectly stable ematical concept based on this example of acoustic
during rapid changes in BP. Instead, dynamic autore- signals. TFA quantifies how different frequencies and
gulation results in a relative, time-dependent buffering amplitudes in BP are transmitted (transferred) to CBF.
of changes in CBF. Specifically, Giller (106) applied the concept of coher-
This idea of a relative CBF buffering capacity for ence [i.e., a measure of the strength of the relationship
rapid changes in BP was later expanded by Birch et al. between oscillations (frequencies) in BP and CBF]. As
(103). They identified that the capacity to buffer a simplified explanation, coherence can be compared
changes in CBF was strongly dependent on the speed with r in a linear regression. A coherence of 0 means
of BP changes (103). The slower the change in BP, the no relationship between oscillations in BP and CBF. A
smaller the impact on CBF, to a point where CBF coherence of 1 indicates that the oscillation in CBF is
becomes almost unaffected. However, for more rapid fully explained by the oscillation in BP. A coherence of
changes in BP, the buffering capacity is progressively 1 also means that CBF is passively following BP and
reduced and changes in CBF become larger, until a there is no dynamic autoregulation. Giller therefore
point where changes in CBF become as large as the proposed coherence as a measure of autoregulation
change in BP. At that point, CBF passively follows BP. efficacy: indeed, coherence increased for faster
This concept is illustrated in FIGURE 3, where oscilla- oscillations (higher frequencies). Further applications
tions (a change occurring at a specific frequency) are of TFA to dynamic autoregulation involved the use of

1496 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

TFA gain and phase by Panerai et al. (95, 107, 108), Blaber 2.5. How Is Dynamic Autoregulation Assessed?
et al. (109), and Zhang et al. (110). In summary, these groups
reported that TFA characteristics between oscillations TFA has become popular because it allows assessment
(fluctuations) in BP and CBF resemble a high-pass filter, of dynamic autoregulation from a 5-min (or longer) re-
wherein higher frequency fluctuations are more line- cording of BP and CBF, without the need for an experi-
arly transferred (i.e., higher coherence) to the cerebral mental intervention to change BP (55, 95, 110). This
circulation than lower frequency fluctuations (41). The approach is feasible because BP normally exhibits spon-
parameter gain, as in acoustics, quantifies damping (i. taneous changes (oscillations) that occur at specific fre-
e., smaller oscillations in CBF com-pared with BP oscil- quencies. These BP changes influence CBF, but how
lations). Phase, or more appropriate, phase shift, indi- they are transmitted is determined by dynamic autore-
cates that the oscillations in BP and CBF are no longer gulation (113). The benefits of this method are being able
synchronized. In other words, because in every oscil- to perform measurements while subjects rest quietly, ei-
lation, CBF returns toward baseline before BP does, ther supine or sitting. This makes the technique more
the CBF oscillations show a phase shift compared with suitable for patients who cannot tolerate or perform
the BP oscillations (see FIGURES 4 and 5). TFA, with interventions to experimentally manipulate BP (55).
estimates of coherence, phase, and gain, has become These benefits are in part offset by a number of disad-
the most widespread method to quantify dynamic vantages (114). The assessment of dynamic autoregula-
autoregulation (41, 55, 111, 112). tion can be hindered by a low signal-to-noise ratio. This

Phase shift ~ 45°

Damping (gain) ~ 0.8 (cm · s-1/mmHg)

0° 180° 360°

220

200 80

180

160 60
SBP (mmHg)

CBFV L (cm/s)

140

120
40

100

80
20
60
40
20
0 0
0 30 60 90
Time (seconds)

FIGURE 5. With the use of a zoomed-in section of the data presented in FIGURE 4, this graph illustrates the relationship between changes in blood
pressure (BP; black line) and cerebral blood flow (CBF) (blue line) induced by the repeated sit-to-stand maneuvers at 0.05 Hz, see details in the legend
for FIGURE 4. Bottom: the timing of these sit-stand maneuvers is schematically illustrated in the graph. Note that the BP and CBF graphs are not fully
synchronous; this is an effect of autoregulation. The leftward shift of CBF [here, represented by the left middle cerebral artery blood velocity (CBFVL)
signal recorded with transcranial Doppler] compared with BP [systolic BP (SBP) measured using Finapres] is referred to as phase shift. Phase shift is
one of the parameters that follows from transfer function analysis. Top: inserted graph is a schematic explanation of phase shift. If we consider the
repeated changes in BP and CBF as an oscillatory signal, resembling a sinusoid with a period of 360 , the leftward shift of CBF can be quantified in
degrees (or converted to radians), in this example 40–50 , which in this frequency (0.05 Hz, the very-low-frequency range) indicates normal dynamic
autoregulation. The parameter gain is also illustrated, representing damping, where effective dynamic autoregulation will result in relatively smaller
changes in CBF compared with BP. Because BP and CBF have different physical units, gain is not necessarily below 1.

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1497


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

can occur because BP oscillations can sometimes be precisely, dynamic autoregulation may not reflect static
too small (115) in relation to the level of noise that is autoregulation (89), even though this was originally sug-
always present when performing noninvasive record- gested when dynamic autoregulation was introduced
ings (54, 116, 117). The signal-to-noise ratio is also (90). Methodological differences may explain the oppo-
reduced because CBF is affected not only by BP, but by site findings of de Jong et al. (89), who found no associa-
various other parameters during these measurements, tion between static and dynamic autoregulation when
such as changes in PaCO2 or cognitive activation (54). tested in the same subjects, versus Tiecks et al. (90)
This has led to the development of methods to enhance who found a strong linear relationship between static
BP oscillations (114, 118). Several methods are now used, and dynamic autoregulation. The main methodological
including repeated squat-stand (118, 119) or repeated sit- difference is that Tiecks et al. performed experiments
stand maneuvers (99, 100), paced breathing (104, 120), during surgery, where isoflurane anesthesia was used
repeated occlusion/deflation of thigh cuffs (121), or oscil- to impair autoregulation (see sect. 5.7). This drug-
latory lower body negative pressure (122). Each proce- induced impairment of autoregulation may have driven
dure has in common that the frequency of the maneuver the correlation between static and dynamic autoregula-
determines the frequency of the BP oscillation. tion (89). Further research is needed to evaluate how
An example of how this method can be used to study assessment of dynamic autoregulation relates to static
dynamic autoregulation is illustrated in FIGURE 4. This autoregulation. For example, can a finding of normal
figure shows an example of repeated sit-stand maneu- dynamic autoregulation be used to predict a normal
vers that cause large oscillations in BP and CBF (middle static autoregulation response (e.g., to BP-lowering
cerebral artery blood velocity) at a frequency of 0.05 Hz. treatment)? It is conceivable that in an individual, imp-
Visual inspection might indicate that BP changes are airment in autoregulation manifests as a slowing of
passively transmitted to CBF, implying absent or poor the adaptive response, starting with impairment to coun-
autoregulation. Upon closer inspection, with each oscil- teract faster changes in BP (i.e., impaired dynamic autor-
lation in BP, CBF recovers before BP. This leads to a egulation), while the adaptation to slow changes in BP is
phase shift between the BP and CBF signals (FIGURE still intact (i.e., normal static autoregulation) (125).
5). FIGURE 5 also illustrates the concept of gain (or the Nonetheless, in the study by de Jong et al. (89), the lack
ability for damping) by comparing the relative magnitude of association between dynamic and static autoregula-
of CBF oscillations to those in BP. With normal dynamic tion was observed in subjects with normal static and nor-
autoregulation, a faster return toward baseline of CBF mal dynamic autoregulation. Ideally, studies performing
compared with BP will result in damping of CBF oscilla- repeated within-subject comparisons of static and
tions (41). dynamic autoregulation could shed more light on this
The frequency of 0.05 Hz is one where dynamic autor- controversy.
egulation is very effective. Autoregulation can be even A second consequence of the “two ends of a spec-
more effective with slower changes in BP (lower fre- trum” concept is that the outcome of studies in humans
quencies). This phenomenon can, at least theoretically, that try to measure static autoregulation are affected by
be extended to changes in BP that become progres- the timing and magnitude of BP changes. A clear exam-
sively slower, occurring over hours, days, and weeks ple of this is the study by Tan (126) that plotted a static
and resulting in progressively smaller changes in CBF. autoregulation curve using induced BP (and CBF) oscil-
These very low frequency fluctuations in BP represent lations at a frequency of 0.03 Hz. This frequency is much
the transition between dynamic (faster changes) and more in the dynamic range than in the static range,
static (very slow changes) autoregulation, represented which explains why that study found a very narrow gra-
in FIGURE 3. Empirical evidence supporting this notion dient phase (“autoregulation plateau”) of only 10 mmHg,
was recently published, in experiments wherein pro- and a steep slope for this gradient (126).
gressively slower oscillations in BP were induced with
oscillatory lower body negative pressure (123) but is also 2.7. Revisiting Historical Experiments
supported by long-duration observations of spontane-
ous changes in BP (124). When we revisit early studies with the current knowl-
edge of dynamic autoregulation, we can explain initial
2.6. Static versus Dynamic Autoregulation observations of Roy and Sherrington (9) and Bayliss et
al. (5). Based on the heart beat traces in the figures in
The concept that dynamic and static autoregulation are the Bayliss and Hill publication, we can tell that the per-
two ends of a spectrum has several consequences. fusion pressure changes in the various experiments
First, a measurement at one end of the spectrum may were of short duration (i.e., seconds). We know now that
not correlate with measurements at the other end. More for such rapid changes in perfusion pressure, isolated

1498 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

cerebral blood vessels and CBF passively, but transi- response to experimental manipulation of arterial BP.
ently, follow such BP changes, without effective regula- These changes included vasoconstriction, with vascular
tion of CBF. FIGURE 6 compares one of the figures of diameter decreasing below baseline values, in response
Bayliss et al. (5) with a recent human experiment to a rise in BP and vasodilation following a reduction in
wherein rapid changes in BP were provoked by BP (13). This work provided direct early examples of the
repeated transitions between squatting and standing rapidity of the cerebrovascular response following
(118). In both experiments, the sudden increases and increases or decreases in arterial BP. In addition to
decreases in BP occur within 15 s, and these transient insight regarding the temporal nature of the response,
changes in BP appear to be passively followed by CBF, these studies revealed that BP-induced changes in the
as interpreted by Bayliss et al. (5). Closer inspection diameter of smaller arterioles were greater on a percent-
however indicates a more rapid return of the CBF trace age basis than in larger arteries. Although it was not yet
in both experiments. In retrospect, a more precise inter- known that arteries and arterioles in this segment of the
pretation of Bayliss’s experiments should have been cerebral circulation are resistance vessels (45, 72, 127,
that this control was unable to maintain a stable CBF 128), Fog (13) speculated that reductions in vessel diam-
under the experimental conditions that were used eter during increases in BP would prevent a rise in capil-
(rather than that there was no active vasomotor control lary pressure and thus protect against brain edema. This
in the cerebral circulation). Another important message concept remains widely accepted today (FIGURE 1) (129,
from the experiments of the Bayliss et al. is the value of 130).
presenting individual findings, something that is often With respect to autoregulation-related terminology,
omitted in the current literature. In this case, presenting myogenic tone is defined as the state of partial vasocon-
individual data made it possible to reinterpret findings, striction that occurs in an isolated blood vessel when
even >100 yr after the original publication. maintained at a constant pressure (129). Arteries and
arterioles in the pial and parenchymal circulation
2.8. Mechanisms That Underlie Autoregulation respond to pressurization with development of myo-
genic tone (129). As an example, isolated brain paren-
Some key phenotypic features of autoregulation have chymal arterioles from both humans and mice develop
been known for many decades. For example, using a substantial myogenic tone when pressurized to physio-
cranial window to measure vascular responses in anes- logical levels (127, 131–133). Myogenic reactivity reflects
thetized cats, Fog (13) described rapid changes in the di- changes in vascular tone in response to changes in
ameter of pial arteries and arterioles (within 1–2 min) in pressure (129). The term myogenic response seems to

A B
160

FIGURE 6. Comparison of hemodynamic


traces from one of Bayliss’s 1895 animal
120
experiments with those from a human experi-
ment performed in 2009. A: original figure
BP (mmHg)

80 from Bayliss et al. (5). The transient increase


BP

in carotid arterial blood pressure (BP)


appears to be passively followed by the trace
40 labeled cerebral venous pressure (CVP),
which was used in this experiment as a proxy
for cerebral blood fluid (CBF). Systemic ve-
0 nous pressure (SVP) was recorded simultane-
SVP

ously and remained stable (to indicate that


the cerebrovascular changes were not sec-
ondary to systemic changes). B: tracings from
160
a human experiment from Claassen et al.
(118). The transient increase in BP (evoked by
CBFV (cm/s)

120 a squat-stand maneuver) also appears to be


passively followed by middle cerebral artery
CVP

80 blood velocity (CBFV; see also the legends


for FIGURES 4 and 5), a proxy for CBF.
Closer inspection and analysis, as described
40
in FIGURES 4 and 5, are required to appreci-
ate effects of autoregulation.
0

18 beats 12 s

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1499


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

be used as a more general or global term, representing myogenic tone and reduce vascular diameter, thus
alterations in vascular tone that result from changes in increasing vascular resistance. This suggests that rather
intravascular or transmural pressure in vitro or in vivo. than regulating vessel diameter per se, the physiological
Myogenic responses of isolated cerebral blood vessels variable that is being regulated is transmural wall tension
are more commonly studied during increases in pres- (129). With increases in BP, a reduction in vessel diameter
sure (rather than reductions in pressure). Many in vitro helps to normalize wall tension. This concept is supported
studies have confirmed the basic concept described in by the fact that similar phenotypic changes are seen in
vivo, that changes in vessel diameter occur relatively isolated vessels studied under pressurized conditions in
rapidly in response to changes in intravascular BP (127, vitro, but where blood flow is absent. Such behavior of
132–138). isolated arteries or arterioles in vitro during increases or
Arteries or arterioles from the pial or parenchymal cir- decreases in transmural pressure (127, 132–138) confirm
culation exhibit myogenic reactivity following changes in the changes in vascular diameter that have been
transmural pressure (129, 139). In response to reductions described by many laboratories performing in vivo experi-
in arterial BP, these vessels respond by reducing myo- ments in which arterial BP was altered (for examples, see
genic tone and increasing lumen diameter, thus reducing Refs. 13, 71, 72, 76, 83, 134). It is important to stress that,
resistance through each vessel. Conversely, an increase as the name implies, the myogenic response is intrinsic to
in arterial BP activates mechanisms that increase vascular muscle. For example, this response remains

A Vascular Tone
Diameter

Pressure
C Candidate D Modulators of
Sensors Autoregulation
• integrins • endothelium E Distribution of Pressure
Membrane Potential • GPCRs: • extrinsic innervation
AT1R, S1P, P2Y • intrinsic innervation aorta
Ca2+ • G12/G13 • genetics

% of Aortic Pressure
BKCa Mechanotransducer
• TRPC6 • glia? arteries
CaV2.1 • TRPM4 • brain metabolism
• βENaC • O2, CO2, pH
∆ tone arterioles
Ca2+
sparks
hyperpolarization depolarization RhoGEF

capillaries
Ca2+ Ca2+
RhoA Resistance Arteries venules
SR MLC and Arterioles

MLCK MLCP ROCK


F Autoregulation of CBF
MLC-P
Cerebral Blood Flow

Contraction

Vascular Smooth
Muscle
cell membrane

B Mechanisms of Contraction
Mean Arterial Pressure

FIGURE 7. Mechanisms that contribute to myogenic responses and the impact of these mechanisms on regulation of vascular tone, the distribution
of intravascular pressure in brain, and autoregulation of cerebral blood flow (CBF). Several mechanisms have been implicated in regulation of myogenic
responses. Vascular diameter is highly sensitive to changes in the cellular membrane potential (A). Membrane potential is determined by the integrated
effects of mechanisms that produce depolarization or hyperpolarization of the cell membrane (A). Two key regulators in the context of pressure-
induced changes in vascular tone are voltage-dependent calcium channels (e.g., CaV2.1) and large conductance potassium channels (BKCa) (A). Several
molecular mechanotransducers have been proposed to function as sensors for changes in pressure and initiators of depolarization of vascular muscle,
resulting in increased intracellular Ca21 (B). Increases in intracellular Ca21 can also occur due to Ca21 release from the sarcoplasmic reticulum (SR)
21
(local Ca signals) resulting in activation of contractile proteins [myosin light chain kinase (MLCK)], phosphorylation of myosin (MLCP), contraction of
21
vascular muscle, and a reduction in diameter of resistance vessels (B). Local release of Ca sparks from the SR can activate BKCa, producing local hy-
perpolarization and feedback that limits the degree of vasoconstriction. In addition to activating CaV2.1, mechanotransduction activates guanine nucle-
otide exchange factors that activate RhoA (RhoGEF) and a RhoA target, Rho kinase (ROCK). ROCK exerts inhibitory effects on MLC phosphatase. As
discussed in the main text, there are several candidate sensors (or mechanotransducers) for pressure changes (C) as well as modulators of autoregula-
tion (D). Integrated changes in myogenic responses and thus vascular tone influence the distribution of intravascular pressure that normally occurs
along the vascular tree in brain (E) as well as the efficacy of autoregulation of CBF (F). See text for further details. GPCR, G protein-coupled receptor;
TRP, transient receptor potential.

1500 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

intact if endothelial cells are removed or selectively mechanosensitive G protein-coupled receptors (GPCRs)
injured (129, 134–138). [e.g., angiotensin AT1, sphingosine-1-phosphate (S1P),
An important step in the development of the myo- purinergic (P2Y)], G-protein subunits (G12/G13) linked to
genic response relates to depolarization of vascular activation of ROCK, and stretch-activated ion channels
muscle, which has profound effects on vascular tone such as transient receptor potential (TRP) (e.g., TRPC6,
(FIGURE 7, A and B). Work by Nelson and others (140, TRPM4) and epithelial Na1 channels (bENaC) (FIGURE
141) demonstrated the high sensitivity of vascular tone to 7C) (51, 129, 139, 144–150). Another potential mediator,
changes in membrane potential of vascular muscle. the PIEZO1 stretch-activated ion channel in vascular
Data based on direct recordings during alterations in muscle, has been investigated but does not appear to
intravascular pressure demonstrated changes in mem- play a role in regulating myogenic tone in isolated cere-
brane potential of only a few millivolts. Under physiologi- bral arteries (151).
cal conditions (i.e., baseline 40 mV), such changes In addition to multiple pressure-sensitive candidates,
cause significant alterations in intracellular Ca21 and defining myogenic mechanisms is further complicated
subsequently vascular diameter (FIGURE 7A) (140–142). by evidence that the role of putative mechanotran-
Increases in intraluminal pressure within an isolated ducers may vary depending on the vascular segment or
artery or arteriole results in mechanotransduction, depo- zone in question. For example, pharmacological inhibi-
larization of vascular muscle, and Ca21 mobilization tion of the AT1 receptor has been reported to reduce
(FIGURE 7B). The increase in intracellular Ca21 follow- myogenic tone in cerebral arteries but not parenchymal
ing a rise in intravascular pressure is due primarily to arterioles (144), despite the fact that parenchymal arte-
influx of Ca21 across the plasmalemma (mainly via volt- rioles develop greater levels of myogenic tone (127, 129,
age-dependent Ca21 channel, CaV1.2) or release of 131, 143, 152, 153).
Ca21-stores from the sarcoplasmic reticulum (local Although myogenic responses are intrinsic to vascular
Ca21-signals) via IP3 or ryanodine receptors (129, 140), muscle, there are additional determinants or modulators
resulting in activation of contractile proteins [phosphoryl- of this response including genomics, endothelial-, meta-
ation of myosin light chain (MLC)] and a reduction in vas- bolic-, neural-, and immune-related signaling, as well as
cular diameter (FIGURE 7B). When voltage-dependent other ion channels and signaling networks (FIGURE 7D)
Ca21 channels are blocked, or when extracellular Ca21 (129, 140, 150). For example, large conductance Ca21-
is removed, the vasoconstrictor response to pressure activated K1 channels (BKCa) in the cell membrane are
(development of myogenic tone) is abolished, thus con- activated by local release of Ca21 sparks from the sarco-
firming an essential role of Ca21 (141). Conversely, hyper- plasmic reticulum, resulting in local membrane hyperpo-
polarization of the cell membrane, due to stimuli that larization and attenuation of myogenic tone (FIGURE
include Ca21 sparks, closes CaV1.2 channels, reducing 7B) (140, 142). The cystic fibrosis transmembrane regula-
Ca21 entry and intracellular Ca21 concentrations, result- tor (a membrane protein and chloride channel) exerts in-
ing in vasodilation (FIGURE 7, A and B) (129, 140, 141). hibitory effects on myogenic tone by attenuating S1P-
Together, these changes are believed to be major con- dependent signaling (154). Collectively, such mecha-
tributors to the maintenance or relative stability of CBF nisms may limit the development of excessive myogenic
during increases or decreases in perfusion pressure in tone in vivo.
vivo (129, 140) (FIGURE 7). In addition to changes in sub- While vascular muscle and the myogenic response
cellular concentrations of Ca21, myogenic tone is regu- appear to play a predominant role in relation to autore-
lated by mechanisms that influence Ca21 sensitivity, gulation of CBF during changes in BP in vivo, it is not the
including protein kinase C and Rho kinase (ROCK) only mechanism or cell type involved, making its precise
(FIGURE 7B) (129, 139). For example, in brain parenchy- contribution to overall autoregulation difficult to quantify.
mal arterioles, pharmacological inhibition of ROCK, or Thus, when new mechanotransducers, cell types, or sig-
the ROCK2 subtype, eliminates the vast majority of myo- naling events are implicated, based on studies of myo-
genic tone (127, 143). genic responses in isolated vessels, one cannot assume
In relation to the control of myogenic responses and that the relative impact of a given mechanism studied in
autoregulation of CBF, a major unanswered question vitro is the same in vivo. For example, during increases
continues to be: “what is the sensor responsible for in arterial BP (the upper half of the autoregulatory curve),
mechanotransduction underlying the myogenic res- several lines of evidence suggest sympathetic and tri-
ponse and subsequent depolarization of vascular geminal nerves influence the vascular response (see
muscle?” Although this aspect of myogenic responses sect. 3.4). Mechanisms that control vessel diameter and
remains relatively poorly defined (particularly in vivo), vascular resistance during reductions in arterial BP are
multiple candidates have been proposed to be involved. likely not mediated through inhibition of these mecha-
These candidates include integrins (e.g., a5b1), nisms that are activated when BP is increased. With

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1501


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

decreases in perfusion pressure, mechanisms involving and G12/G13 G protein subunits, along with ROCK-de-
tissue hypoxia, adenosine, calcitonin gene-related pep- pendent signaling in vascular muscle (FIGURE 7B)
tide (CGRP), ATP-sensitive K1 channels, and endothelial (145). Additionally, links between ROCK and TRPM4
nitric oxide synthase (eNOS) have all been implicated to had been suggested previously (143). The investiga-
play a role (47, 76, 150, 155–159). In this context, genetic tors also observed that genetic deficiency in G12/G13
deficiency in NOS3 (eNOS) shifts the lower end of the subunits in vascular muscle increased baseline CBF
autoregulatory curve to the right, consequently leading (145), a phenotype that is consistent with reduced
to augmented reductions in CBF in response to myogenic tone, although autoregulatory responses
decreases in BP (155). A loss of function mutation in the per se were not examined in vivo.
NOS3 gene is associated with impaired autoregulation
in response to carotid artery compression in humans 2.9. Where in the Vascular Bed Does
(150). In contrast, other genetic variants enhance NO Autoregulation Take Place?
(eNOS-derived) signaling and are associated with long-
term beneficial effects, including reducing the risk for Basic laws of hemodynamics dictate that blood flow is
stroke in humans (160). We speculate that such protec- determined by pressure gradients and vascular resist-
tive effects may relate, in part, to positive effects on ance. As a consequence, autoregulation operates by
autoregulation of CBF. changing vascular resistance in response to alterations
Based on a model in which vessels within a brain slice in BP, to maintain blood flow (168, 169). For blood ves-
are perfused and pressurized in vitro, a role for astro- sels with a fixed length and blood with a fixed viscosity,
cytes in modulating myogenic tone has been suggested vascular resistance is determined by vessel diameter
(FIGURE 7D) (161). The potential impact of these cells in (170). Changes in the radius “r” of a blood vessel (i.e.,
this context is difficult to define because the model ele- due to vasodilation, vasoconstriction, or vascular remod-
vated intravascular pressure by increasing vascular eling) affect its resistance by r4. This means that even
perfusion within the slice. In other words, arteriolar small changes in vessel diameter can lead to large
responses were not studied at constant pressure (with changes in vascular resistance, and therefore in blood
constant flow) making interpretation less clear. The slice flow (33, 168, 169, 171, 172). For example, an increase in
model is also complicated by the fact it was studied vessel diameter of only 20% (at a constant pressure),
under hypothermic conditions, which are known to which is within the physiological ability of arteries for
affect myogenic tone and CBF (162). acute changes in diameter, will increase blood flow by
Recent studies continue to provide insight into novel 207%.
potential sensors or signaling events that may contribute The degree of myogenic tone that develops under
to myogenic responses in isolated vessels. Unfortunately, normal conditions varies along the vascular tree. Based
the majority have not tested the relative contribution of on work using isolated blood vessels, there is less myo-
each candidate mechanism in vivo. For example, the AT1 genic tone under baseline conditions in larger cerebral
receptor is reported to be mechanosensitive, with activa- arteries than in smaller pial and parenchymal arterioles
tion of the receptor increasing myogenic tone in isolated (127, 129, 131, 143, 152, 153). These findings are consist-
cerebral arteries (144, 163). In other studies, however, inhi- ent with the observation that effects of changes in arte-
bition of this receptor does not significantly alter baseline rial pressure on vessel diameter are size dependent in
diameter of pial arterioles or resting CBF or impair autore- pial arteries and arterioles in vivo (13, 76, 83). This sug-
gulation of CBF during increases in arterial pressure gests that, during autoregulation (i.e., changes in vascu-
(79, 164–167). It seems logical to assume that in vivo lar resistance during changes in perfusion pressure in
myogenic responses contribute to resting tone of cer- vivo), differences may exist in the relative importance
ebral arterioles, baseline CBF, and changes in CBF between segments. Direct measurements of CBF and
with increased BP. Thus the contribution of AT1 recep- intravascular pressure under baseline conditions indi-
tors in relation to autoregulation of CBF remains cate that vascular resistance is distributed over different
unclear (FIGURE 7B). vascular segments (or zones), resulting in a reduction in
There are exceptions to the approach of only studying pressure along the vascular tree. For example, intravas-
myogenic mechanisms in isolated vessels in vitro. For cular pressure in pial arterioles on the surface of the
example, a recent study examined effects of smooth brain is approximately half of systemic arterial BP (i.e.,
muscle-specific deficiency in G12/G13 protein subunits central aortic BP) (FIGURE 7E) (45, 46, 71, 72, 127). As
or the Rho guanine nucleotide exchange factor part of this arrangement, pulsatile effects of the beating
ARHGEF12, both in vitro and in vivo (145). Overall, the heart on intravascular pressure (pulse pressure) are atte-
study suggested a dual mechanism for development nuated as blood flows down the vascular tree and into
of myogenic tone, one that involves Ca21-dependent the parenchymal circulation and pial venules (67, 71, 72,

1502 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

128, 173). Furthermore, the hemodynamic profile within autoregulate is exceeded during acute severe hyperten-
the parenchyma may be influenced by precapillary sion, CBF increases substantially along with elevated
sphincters or other structures at the transition from small pressure in pial venules (FIGURE 7, E and F) (84, 173,
arterioles to capillaries (174, 175). Venules and larger 182). These microvascular hemodynamic changes are im-
veins contribute modestly to overall vascular resistance portant because acute increases in venular pressure are
under normal conditions (67, 71, 72, 173). key determinants of disruption of the BBB, much of which
Increases or decreases in BP evoke changes in resist- occurs at the levels of venules (173, 182, 183). Although
ance of large arteries, arterioles on the pial surface, and the BBB is present in cerebral arterioles, capillaries, and
within the brain parenchyma (13, 72, 73, 76, 83, 84, 128, venules (184), proteins that comprise tight and adherent
176). From these experiments, we have learned that junctions between endothelial cells exhibit heterogenous
small vessel resistance, which was calculated based on features, including looser junctional strands in venules
measurements of intravascular pressure in pial arterioles (184). These looser venular junctional strands may explain
and pial venules, accounts for approximately half of total why the BBB in venules appears to be predisposed to
cerebrovascular resistance (FIGURE 7E) (84, 128). disruption during acute episodes of hypertension that
To our knowledge, the relative contribution of arterio- exceeds the autoregulatory capacity of the circulation
les, capillaries, and venules within the parenchyma to (FIGURE 7, E and F). The clinical aspects of hypertensive
overall vascular resistance has never been directly emergencies are discussed in sect. 5.2.
determined in vivo. Because most of the decline in intra- Anatomical differences in the cerebrovascular bed or
vascular pressure in the pial circulation occurs at the other factors, either in physiological or pathological con-
level of arteries and arterioles (FIGURE 7E) (45, 127, 128, ditions, can result in regional differences in intravascular
177), we assume arterioles and capillaries within the pa- pressure. For example, intravascular pressure is pre-
renchyma are the major contributors to the small vessel dicted to be higher in lenticulostriate arteries than in the
resistance component of total cerebrovascular resist- intraparenchymal arteries in the parietal lobe (185). In
ance (84, 128). Indeed, during moderate increases in ar- addition, regional differences in vascular anatomy and
terial BP (40 mmHg), small vessel resistance increased patterns of the vascular tree can affect the control of mi-
by 50% and CBF was maintained at baseline levels crovascular pressure and vascular resistance. This could
(84). cause regional differences in susceptibility to ischemic
Although myogenic tone and reactivity have been events, such as lacunar infarction, which is predomi-
studied in isolated parenchymal arterioles in vitro, little is nantly seen in brain regions supplied by the lenticulostri-
known regarding the responses of these same arterioles ate arteries (185, 186). Under pathophysiological con-
to changes in BP in vivo. In one study that used multi- ditions, such as small vessel disease, distinct changes in
photon imaging, a reduction in cerebral perfusion pres- structure and function of small arterioles in the paren-
sure was produced by increasing ICP by 10–15 mmHg, chyma may increase local intravascular pressure in
resulting in vasodilation of penetrating arterioles by upstream arterioles (175). This increase in arteriolar pres-
12% (178). Although this experiment demonstrates an sure may increase local wall stress and contribute to the
autoregulatory response in vivo, details regarding its development of arteriolar rupture and the pathogenesis
influence on CBF and effects of changes in perfusion of microbleeds (175).
pressure on the various subtypes of parenchymal arte- In some, but not all studies, changes in diameter of
rioles (179, 180) are lacking. capillaries have also been described during vasoactive
As described elsewhere (sect. 5), the clinical conse- stimuli (49, 174, 187–190). When changes in capillary di-
quences of failed autoregulation and the mechanisms ameter are detected in vivo, to what extent these
involved differ depending on which portion of the auto- changes are active or passive, that is, due to alterations
regulatory curve is affected (FIGURE 7F). Failure at the in the contractile state of local pericytes versus changes
left side of the curve results in hypoperfusion, potential in transmural capillary pressure, remains largely uns-
ischemia, and even death (FIGURE 1). If autoregulatory tudied and a possibility that is not generally considered
mechanisms are overwhelmed and fail at the right end (191).
of the curve, hyperperfusion, increases in pressure in Under some conditions, changes in diameter or
arterioles, capillaries, and venules, disruption of the cross-sectional area of venules may also occur (192).
BBB, edema, increased ICP, and death can result For example, increases in diameter of small venules
(FIGURE 1) (129, 130, 150, 181). With moderate increases occur during hypercapnia (192). A caveat when meas-
in arterial pressure (40 mmHg), elevations in vascular uring venous or venular diameter is that veins often
resistance are sufficient to maintain CBF at normotensive do not maintain a circular shape at low pressures (or
levels and prevent increases in microvascular pressure in when not pressurized), exhibiting an oval or col-
pial venules (84). However, when the ability to lapsed shape (193). A similar limitation is a potential

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1503


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

issue when measuring diameter of any vessel using perfusion pressure that follow from the various postural
in vitro brain slice preparations, where no pressuriza- challenges in daily living, it is not surprising that autore-
tion is typically present. gulation is normally so effective and that multiple mech-
Overall, multiple lines of evidence indicate that vascu- anisms and cell types modulate the response (FIGURE
lar muscles in cerebral arteries and arterioles are sensi- 7). From an evolutionary perspective, the choice for rela-
tive to changes in BP and that smooth muscle is the tively simple (but robust) myogenic responses as the
primary effector cell driving acute changes in vascular main mechanism for autoregulation is logical, as they
resistance in response to increases or decreases in BP appear well positioned to withstand the effects of aging
(FIGURE 7, A, B, E, and F). Based on the assumption and some diseases.
that the primary mechanism contributing to autoregula-
tion of CBF is myogenic responses of vascular muscle, 2.10. Summary
then cerebral arteries and arterioles (pial and parenchy-
mal) play a significant role in mediating substantial Various methods currently exist to 1) assess CBF (or CBF
changes in myogenic tone. Capillaries, despite their velocity), 2) measure and manipulate BP, and 3) quantify
large surface area, make a smaller contribution to total the efficacy of autoregulatory mechanisms through sev-
vascular resistance (FIGURE 7) and are unlikely to have eral analytical approaches, each with their respective
a major contribution to autoregulation as capillaries do limitations and caveats that must be considered.
not actively alter their diameter in response to changes Consequently, research examining autoregulation can
in transmural pressure. Whether pericytes can respond be challenging, which may contribute to results that
to physiological changes in capillary pressure by actively appear to be conflicting at times. Nonetheless, collec-
adjusting local capillary diameter is an area of ongoing tively these approaches have provided strong evidence
investigation. that autoregulatory mechanisms maintain CBF relatively
In relation to potential neural control of autoregulation, stable across a range of BP values in humans.
a few studies have examined whether specific regions In the remainder of this review, we focus on improving
within the brain may influence autoregulation of CBF our understanding of dynamic autoregulation of CBF in
globally. For example, electrolytic lesions of the fastigial humans under resting conditions, in the presence of rel-
nucleus in the cerebellum had little or no effect on rest- evant disease states, as well as during daily life chal-
ing CBF or autoregulation of CBF during reductions in lenges. We first address other important mechanisms
arterial BP (produced by hemorrhagic hypotension) in that affect CBF (e.g., arterial blood gases, cerebral me-
conscious rats (194). In contrast, electrolytic lesions of tabolism, neurogenic stimuli) but also the interrelation-
the nucleus tractus solitarii (NTS) in the medulla impaired ships between these stimuli. We discuss how and where
autoregulation in multiple brain regions during phenyl- these mechanisms or stimuli regulate CBF (i.e., at the
ephrine-induced increases in arterial BP in anesthetized level of large arteries and/or the microcirculation). With
rats (195). This suggests that neurons that originate or that background, we then focus on autoregulation, dis-
pass through the NTS may play an important role. What cussing clinical implications that may arise as a conse-
mechanism(s) could account for such an effect remains quence of impaired autoregulation. We address clinical
unclear as the neural pathways from the NTS do not pro- conditions where CBF seems to play an important role:
ject to most of the brain regions that were affected (195). hypertension, vascular dementia, Alzheimer’s disease,
The hypothesis that central pathways might be essen- mixed dementia, ischemic cerebrovascular disease, and
tial for intact autoregulation contrasts with the concept cerebral hemorrhage (FIGURE 1). Finally, we discuss the
that local myogenic responses play a major role in this consequences of daily life challenges for CBF, such as
adaptive vascular response. That isolated arteries and postural challenges (e.g., orthostatic hypotension, syn-
arterioles studied in vitro exhibit myogenic responses cope) and the impact of physical activity (or inactivity).
that phenocopy key elements of autoregulation argues
that the presence of specific brain nuclei or dependent
signaling pathways are not essential. Having said that, 3. OTHER MECHANISMS THAT REGULATE
modulation of autoregulation by neural pathways (e.g., CEREBRAL BLOOD FLOW
sympathetic nerves) has been described and will be fur-
ther discussed (sect. 3.4). Thus neural modulation of Factors that we discuss below collectively influence CBF
autoregulation via specific central pathways remains a regionally or globally, serving to match CBF to demands
possibility, albeit one that remains poorly defined at this for O2 and removal of CO2, or other mechanisms includ-
time. ing the impact of blood gases or neural mechanisms
In conclusion, because of the physiological and evolu- (21). Many textbooks and reviews state that autoregula-
tionary relevance of maintaining CBF during changes in tion operates through “myogenic, metabolic, and

1504 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

neurogenic” mechanisms, implying that all these mecha- CBF of O2, CO2, and associated changes in pH will be
nisms are involved. It may therefore appear confusing if discussed in sect. 3.2. In addition to O2 and CO2,
we list metabolic and neurogenic mechanisms as “addi- changes in metabolism may also affect CBF through
tional factors.” For autoregulation, in the definition of the local production of other vasoactive substances, includ-
adaptation of CBF to changes in BP, it remains uncertain ing adenosine or potassium ion (45).
whether all these mechanisms contribute in an orches- The metabolic “coupling” pathway has been described
trated manner or whether they represent redundant or as a “feedback system,” where changes in O2, CO2, pH,
fail-safe mechanisms (134, 196–199). For example, if BP and other vasoactive substances [i.e., adenosine, nitric ox-
falls, and a major mechanism influencing autoregulation ide (NO), prostanoids, and potassium ion] drive local vaso-
(myogenic responses) should fail to maintain CBF, cere- dilation and increase CBF (45). For at least some brain
bral metabolism may be affected. Such a change could regions, these factors alone are unlikely to account for the
prompt metabolic mechanisms to promote vasodilation local increase in CBF that occurs in response to increases
and restore CBF. Similarly, in response to a large rise in in neural activity (45). Indeed, the increase in CBF following
BP, increases in myogenic tone and activation of sym- neural activation can exceed metabolic demand (212).
pathetic nerves promote protective vasoconstrictor Moreover, increases in CBF following neuronal activation
responses, thus attenuating increases in CBF and hemo- also occur under conditions of an abundance in glucose
dynamic consequences that include disruption of the and O2 (213). Metabolic processes alone may be too slow
BBB and edema. With such a combination of mecha- to explain the fast increase in CBF following changes in
nisms, all working synergistically and showing redun- neural activity. Therefore, a “feedforward” theory has also
dancy, it is difficult to discern which could be viewed as been proposed, wherein coupling largely takes place
a primary mechanism in autoregulation or simply an in- between neurons, glia (responsible for consuming O2 and
dependent mechanism that provides redundancy or the demand for nutrients), and the microvasculature (re-
synergy due to overlapping functional effects. For exam- sponsible for supplying blood, nutrients, and O2), within
ple, the role of the autonomic nervous system in autore- elements of what is commonly referred to as a neurovas-
gulation can be investigated through inhibition or cular unit (45). Excitatory and inhibitory neurons form syn-
activation (e.g., pharmacological, denervation, or genetic apses on both astrocytes and GABAergic interneurons
approaches), followed by studying the response of CBF (36). With neural activation and NVC, endothelium-depend-
to changes in BP under such contrasting conditions ent propagation of vascular signals occurs, which leads to
(200–205). However, such experiments can be riddled remote vasodilation of upstream arterioles and arteries,
with confounding factors. For example, sympathetic including within the pial circulation (49, 50, 214, 215).
blockade causes hypotension and affects respiratory These actions participate in mechanisms that maintain a
control, causing changes in PaCO2 (9). Similar “side close coupling between neuronal activation and local CBF.
effects” may be observed in experiments that aim to in- For example, activation of the occipital cortex by visual
hibit endothelial function (134, 206, 207). This means that, stimulation leads to a rapid increase in posterior cerebral
even though some experiments have suggested that artery blood flow, which supplies the occipital lobe (216,
myogenic responses are independent of endothelial cells 217). This coupling can be observed through various tech-
or the autonomic nervous system in vitro, experimental niques and allows for detailed insight into the ability to
designs that can definitely exclude a role for endothelial closely match regional demand with the supply of blood
cells or nerves are difficult to achieve in vivo. We have and O2 (217). Recent work indicates that there may be re-
therefore chosen to discuss these factors or determinants gional differences in the timing and the precise nature of
of CBF that may operate independently from autoregula- vascular responses to neural activation, but it should be
tion (even leading to confounding, or “physiological noted that it remains uncertain how experimental condi-
noise,” under some conditions) (54, 55, 117). tions (e.g., anesthesia) may affect these results (187).
It is now thought that the coupling of CBF to brain activ-
3.1. Cerebral Metabolism and NVC ity derives from a combination of the “feedback system”
and “feedforward” mechanisms (45). Our overview of all
The regulation of CBF is tightly coupled to cerebral me- factors that influence CBF includes factors that play a role
tabolism (208–211). The observation that both variables in the CBF response to changes in metabolic or neural
are closely linked has been acknowledged for more activity (NVC): arterial blood gasses, pH, endothelium,
than a century (6, 9), although the precise mechanisms vascular muscle, and (autonomic) neural innervation.
underlying this coupling are not fully understood. The Research into defining precise mechanisms that
term metabolism suggests a role for reductions in tissue underlie NVC is an active area that is still unfolding (45,
molecular O2 and increases in CO2 (which result from 49, 51, 174) and has implicated a role for neurons, endo-
metabolic activity) in this coupling (6). The effects on thelial cells, astrocytes, pericytes, and more recently,

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1505


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

precapillary sphincters (45, 49, 51, 174). These topics are CBF following hypocapnia and hypercapnia showed no
beyond the scope for this review. Instead, we refer to relevant regional differences across various brain
two excellent reviews covering these topics (45, 187). regions including gray and white matter (244). Note
In summary, cerebral metabolism and NVC regulate however that regional differences in baseline CBF (e.g.,
local CBF in response to neural activation. The constant much lower CBF in white matter compared with gray
and rapid changes in brain activity explain the relevance matter) cause regional differences in absolute CBF
of this system, including a hypothetical condition where responses to hypocapnia and hypercapnia. More
BP is fully stable and autoregulation has no active role in recently, MRI techniques have been developed to mea-
regulating CBF. Therefore, we consider metabolic con- sure global and regional responses in CBF to changes
trol of CBF and NVC as mechanisms of their own, inde- in PaCO2 (for review see Ref. 245).
pendent from autoregulation, despite some obvious Acidosis is known to be responsible for many of the
redundancies. Components of NVC will be discussed in biological effects of increased CO2 (34). Classic work by
sects. 3.2.2, 3.2.3, and 3.2.4. Kontos et al. (243) demonstrated that reduced extracel-
lular pH, not increases in CO2 per se, are responsible for
3.2. Regulation by Arterial Blood Gases and pH vasodilation to local changes in PCO2. This suggests that
vascular responses activated by increases in PaCO2
3.2.1. Partial pressure of carbon dioxide. relate to the diffusion of CO2 across the BBB, subse-
quently leading to reductions in pH in the extracellular
Brain perfusion is highly sensitive to changes in PaCO2 space and cerebrospinal fluid. Ultimately, a reduction
or tissue levels of CO2 (34, 35, 43, 218–221). An increase in pH represents the primary stimulus that causes vaso-
in PaCO2 (hypercapnia) produces vasodilation, reduc- dilation of vascular muscle. While intermediate pathways
tions in cerebrovascular resistance, and increases in involved in the response to hypercapnia have been
CBF, whereas a drop in PaCO2 (hypocapnia) increases studied for years, and include NO and prostanoids (34,
cerebrovascular resistance and reduces CBF (34, 43, 230, 234, 246, 247), molecular sensors that detect
219–222). As summarized recently (35, 223), studies changes in extracellular pH have been more difficult to
adopting TCD for assessment of the middle and poste- define. Recent studies suggest activation of a proton-
rior cerebral arteries or duplex ultrasound of internal ca- gated cation channel [acid-sensing ion channel-1A
rotid and vertebral arteries (extracranial conduit arteries) (ASIC1A)], particularly in neurons, by extracellular acido-
all demonstrated an 3–6% increase in CBF per mmHg sis plays a critical role in CO2-induced vasodilation in
rise in PaO2 and a 1–3% decrease in CBF per mmHg brain (248). With very high levels of hypercapnia, addi-
reduction in PaCO2 (43, 102, 222, 224). Differences in tional mechanisms may be recruited and contribute to
methodological design, assessment of CBF, analysis of increases in CBF (248, 249).
the responses, magnitude of manipulation of PaCO2 , and
correction for PaO2 , and especially differences in BP, as 3.2.2. Partial pressure of oxygen.
changes PaCO2 can affect BP (43, 225, 226), may con-
tribute to varying results between studies. Nonetheless, The cerebrovasculature is sensitive to variations in PaO2
the primary observation has consistently been that CBF (or tissue PO2). When PaO2 drops below 50 mmHg
shows a high sensitivity to changes in PaCO2 . This (equivalent to an arterial O2 saturation of 80%), this
behavior of the cerebral circulation is clearly distinct leads to cerebral vasodilation. This response also
from most of the systemic circulation (227–229). This ba- depends on the prevailing PaCO2 . The simultaneous
sic observation in humans is consistent with many stud- presence of hypercapnia (i.e., another vasodilator)
ies using preclinical models and multiple species, with increases the sensitivity to hypoxia, strengthening the
only a small representation presented here (34, 74, vasodilator response, whereas the presence of hypo-
230–236). capnia (i.e., a vasoconstrictor) attenuates the CBF
Sensitivity to changes in PaCO2 can be detected response to hypoxia (101, 236, 250–257). The practical
throughout the cerebrovascular tree, from large arteries, consequence of this interdependency of cerebrovascu-
pial arteries and arterioles, and parenchymal arterioles lar responses to changes in PaCO2 and PaO2 is that
(67, 230, 235, 237–242). Based on changes relative to examining the impact of hypoxia in humans leads to a
the original baseline diameter, the greatest response to ventilatory response (i.e., hyperventilation), with result-
hypercapnia occurs in the smaller arterioles (230, 231, ing hypocapnia and vasoconstriction. In addition, re-
239, 241, 243). This may be related, in part, to a relatively gional differences in sensitivity to PaO2 may be present.
larger content of vascular muscle and to a higher degree As a result, studies on the topic of cerebrovascular reac-
of resting tone under baseline conditions. In a study tivity to hypoxia have produced results with significant
using SPECT, the relative reductions and increases in variations (252). Although the overall impact of hypoxia

1506 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

is clear, these variations and dependency on alterations 3.3. Role of Endothelium, Vascular Muscle, and
in PaCO2 highlight the importance for simultaneous Pericytes
assessment of levels of PaCO2 and PaO2 in experimental
conditions to correctly interpret outcomes. All arteries, arterioles, capillaries, venules, and veins
A discussion of the impact of PaO2 on cerebral are lined by endothelial cells (45–47). Arteries con-
perfusion should not omit the role of arterial oxygen tain multiple layers of vascular muscle, with the small-
content (CaO2). The magnitude of changes in CBF est arterioles containing a single layer of smooth
during hypoxia are such that O 2 delivery (the prod- muscle (261, 262). During functional hyperemia or
uct of CBF and CaO2) is maintained at or near nor- NVC (see also 3.1), arterioles in the parenchyma
mal levels despite reductions in CaO2, and appear dilate and this response is propagated in a retro-
linked to CaO2, rather than PaO2 per se (258). grade direction so that the integrated response
Situations that produce a reduction in CaO2 (even in includes vasodilation of arterioles and arteries on the
the presence of preserved PaO2 ), such as carbon brain surface (49, 215, 217). Whether the diameter of
monoxide exposure, acute or chronic anemia, or capillaries increases or not during NVC (or other
hemodilution procedures, lead to an increased CBF increases in local CBF) has been considered, but is
that maintains CaO2 delivery at or near normal lev- currently controversial (49, 187–191). For example,
els (258). recent studies indicate that red blood cells can rap-
Several processes are suggested to play a role in the idly deform under conditions of reduced O2 (i.e.,
increase in CBF during hypoxia. The first relates to local increased O2 consumption during activation), thereby
decreases in PO2 within neurons, glia, or vascular cells. increasing the flow of red blood cells through the
Second, lack of local oxygen (PO2 or O2 delivery) may capillary, without the necessity for capillary dilation
contribute to anaerobic metabolism, leading to extracel- (187). Below, the role of endothelium and vascular
lular acidosis, another vasodilator stimulus. Third, direct muscle are discussed in relation to regulation of re-
vascular mechanisms may be in place, where local hy- gional CBF.
poxia leads to the production and release of vasodilator Regulation of vascular tone by endothelial cells
substances. In this respect, adenosine is a popular and occurs via several mechanisms, including the release of
frequently studied vasodilator, partly because of various signaling molecules that affect adjacent vascular muscle
studies reporting increases in adenosine in response (47, 129, 263) but also the spread of electrical signals
to hypoxemia (259). Indeed, several studies have from endothelial cells to smooth muscle via myoendo-
observed increases in the CBF response to hypoxia thelial gap junctions (263, 264). A family of endothelium-
when adenosine is enhanced and reduced CBF res- derived molecules have been identified that directly
ponses to hypoxia when adenosine is antagonized (e.g., affect the contractile state of vascular muscle. The major
by theophylline, caffeine, or more specific antagonists), molecular messenger in this context is NO. In addition,
although not all studies show similar results (259, 260). potassium ion, hydrogen peroxide, ATP, and other, yet
These partly conflicting data suggest that, although unidentified, endothelium-derived hyperpolarizing fac-
adenosine may contribute to cerebral vascular respo- tors may contribute to affecting vascular smooth muscle
nses to hypoxia, redundancy in vasodilator pathways tone under some circumstances (46). These vasoactive
may be present and vasodilation may also be mediated substances are released by the endothelium in
through alternative mechanisms. response to mechanical shear stress, activation of endo-
In summary, arterial blood gases can markedly thelial cell receptors, and other mechanisms (46, 263).
affect CBF but are not considered to be a mechanistic Collectively, these factors play an important role in the
component of the autoregulatory response. These endothelium-dependent control of vascular tone, along
variables are however of clear relevance for studies with elements of vascular structure, vascular mechanics,
of autoregulation, as changes in arterial blood gases antithrombotic activity, as well as function of neurons,
affect autoregulatory control of CBF. This interaction synapses, and glia (32, 45–50, 129, 138, 263, 265, 266).
is important in preclinical studies, as well as in clinical Despite these advances, our understanding of this
conditions where blood gases are altered, such as system is still relatively limited, particularly in relation
cardiopulmonary diseases, artificial ventilation (inten- to integration of responses between and within differ-
sive care), anesthesia, exposure to high altitude, or ent cell types and segments (or zones) of the vascula-
exercise. Even everyday changes in posture, from ture. Studies that specifically focus on cerebral
lying to standing for example, cause changes in venti- arteries, arterioles, capillaries, and venules, preferably
lation and lung perfusion, affecting blood gases and in an in vivo setting (187), are required to truly under-
thereby CBF (and hence valid evaluation of autoregu- stand the diverse impact of endothelium-dependent
lation). mechanisms. The importance of better understanding

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1507


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

these mechanisms is emphasized by overwhelming 3.4. Sympathetic, Parasympathetic, and Sensory


evidence from experimental and clinical studies high- (Trigeminal) Innervation
lighting the central role of endothelial dysfunction in
mediating and accelerating the process of atheroscle- Large arteries and arterioles in the pial circulation are
rosis (267, 268). richly innervated by adrenergic (neurotransmitters: epi-
Emerging evidence suggests endothelial dysfunction nephrine, norepinephrine, dopamine, and neuropeptide
is also a major player in the pathogenesis of small vessel Y), cholinergic (neurotransmitters: acetylcholine and vaso-
disease in brain (58, 265, 269). As one example, endo- active intestinal polypeptide), and sensory (neurotrans-
thelial-dependent vasodilation plays a key role in NVC mitters: calcitonin gene-related peptide, substance P, and
(50–52). To what extent endothelial cells modulate pituitary adenylate cyclase-activity peptide) fibers within
autoregulation in vivo is less studied (155), but most evi- the sympathetic, parasympathetic, and trigeminal nerve
dence to date suggests that myogenic responses in systems, respectively (36, 278, 279). This innervation
vitro are largely independent from endothelial function does not follow blood vessels as they dive into the brain
(129, 134–138, 270–272). parenchyma (36, 279). The presence of adventitial sym-
Morphological and molecular characteristics of peri- pathetic, parasympathetic, and sensory fibers, innervating
cytes have been reviewed in detail (273, 274). a major segment of the cerebrovasculature, makes them
Although anatomical relationships of pericytes in the distinct from most peripheral organs (280). In addition to
cerebral circulation are reasonably well defined (with this extrinsic innervation of the pial circulation, intrinsic
the exception of transition zones such as precapillary innervation of parenchymal vessels is also present in
arterioles and postcapillary venules), uncertainties brain (36). Extrinsic innervation of extraparenchymal
regarding their molecular characteristics and key arteries and arterioles originates mainly from the superior
aspects of their potential functional importance cervical ganglia (sympathetic innervation), otic, and sphe-
remain unclear (187, 274, 275). In contrast to vascular nopalatine ganglia (parasympathetic innervation), and the
muscle, where closely associated cells are circularly trigeminal ganglion (sensory nerves). Upon entry into the
arranged on a regular basis and oriented perpendicu- brain parenchyma, arterioles and other microvessels are
lar to blood flow with essentially a zero-degree pitch under influence of innervation from brain stem and other
(261), pericytes often extend processes down the long nuclei, including the nucleus basalis of Meynert (36, 281,
axis of capillaries, occasionally encircling some or all 282). In addition to this innervation, sympathetic nerves
of its circumference (273, 275). Activation of contract- potentially influence CBF through effects of circulating
ile pericytes at the capillary level could potentially neurotransmitters (283), although an intact BBB may sub-
reduce local diameter at specific sites and thus affect stantially limit such effects.
individual capillary blood flow. However, the concept
that changes in capillary diameter due to activation or 3.4.1. Sympathetic nervous system.
relaxation of pericytes occur in response to physiological
stimuli is supported by some studies but not by all (49, When exploring effects of the sympathetic nervous
187–190, 274). In addition, much of the work on pericyte system in the regulation of CBF, activation of these
control of capillary diameter has used brain slices in vitro, nerves is sometimes associated with a decrease in
a model that some experts conclude is not adequate to CBF. Studies that have examined patients that, as part
mimic in vivo conditions (187). Furthermore, changes in of their treatment, underwent superior cervical gangli-
capillary diameter are often quite modest in magnitude, onectomy demonstrate that this procedure leads to
raising questions as to whether commonly used micro- an increase in CBF (284–286). Studies adopting local
scopic techniques (multiphoton microscopy for example) or systemic pharmacological ganglionic blockade
can reliably detect physiological changes in capillary di- have produced mixed results, although the majority
ameter (187). With respect to autoregulation, we are not report an increase in CBF (42). Methodological factors
aware of direct evidence supporting an active role for related to the pharmacological procedure used to
pericytes in regulation of CBF during changes in BP. block the ganglia (including potentially only partial in-
However, pericytes might affect capillary blood flow hibition but also effects on BP and PaCO2 ) may con-
under conditions of ischemia, where some studies sug- tribute to variations in findings. Nonetheless, it is
gest pericytes are sensitive to ischemia and respond with generally accepted that the sympathetic nervous sys-
prolonged contraction, causing local entrapment of red tem prevents increases in CBF under resting condi-
blood cells (81, 276, 277). This concept is controversial, as tions (77, 278, 279, 287). However, mechanisms other
others found that precapillary arteriolar smooth muscles, than the sympathetic nervous system (e.g., endothe-
rather than capillary pericytes, contract under ischemic lial, chemical, or metabolic) are likely to be more im-
conditions (189). portant for the regulation of resting CBF (283).

1508 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

While the contribution of the sympathetic nervous blockade with glycopyrrolate led to increased transfer
system to resting CBF may be modest, a key role for the function coherence between BP and CBF, which could
sympathetic nervous system is its influence during rapid indicate a reduction in dynamic autoregulation (295). In
or acute increases in arterial BP. For example, sympa- addition, Seifert et al. (296) found that glycopyrrolate
thetic blockade doubled CBF during a Valsalva maneu- abolished the increase in middle cerebral artery blood
ver (204) and prevented the decrease in CBF velocity during cycling and static handgrip exercise, with
associated with head-up tilt (288), although this finding no change in cerebral metabolism. Although these data
was not replicated (289). In addition, nonselective a-ad- support a role for cholinergic effects on the regulation of
renergic receptor blockade led to a stronger increase in CBF, the question remains whether a role for the para-
CBF when BP was rapidly increased using intravenous sympathetic nervous system per se is indicated.
phenylephrine (290). Inhibiting sympathetic innervation The role of the extrinsic parasympathetic nervous
through ganglionic blockade also led to increased trans- system must not be confused with the brain’s intrinsic
fer function gain and reduced transfer function phase, cholinergic innervation that derives from the basal fore-
implying reduced dynamic autoregulation (205). A com- brain (nucleus basalis of Meynert) (36, 297, 298). In rat
plicating aspect for this and related studies is that gan- models and in human experiments, stimulation of the
glionic blockade reduced BP and BP variability, thus basal forebrain increases CBF through direct choliner-
affecting TFA. This was addressed by increasing BP gic and indirect interneuron (that release NO) vascular
using phenylephrine and by increasing BP variability innervation (297). Of note, Alzheimer’s disease and
using oscillatory lower body negative pressure (205). Lewy body dementia are characterized by early and
Such an example illustrates that it is nearly impossible to prominent degeneration of these intrinsic cholinergic
study effects of sympathetic activation or blockade in neurons originating from the nucleus basalis of Meynert
isolation, without affecting other confounding variables. (281, 282). It has been hypothesized that the reduction
Several other studies, adopting different paradigms or in CBF that occurs in these dementias may be related,
pharmaceutical strategies, but with the same caveats, in part, to this loss of cholinergic innervation and
suggest the sympathetic nervous system contributes to thereby its vasodilator influence and that part of the
autoregulation of CBF during rapid changes in BP clinical benefit of cholinesterase inhibitors may be
[reviewed by ter Laan et al. (283)] For example, during explained by increased cholinergic-mediated vasodila-
acute hypertension, the vasoconstrictor effects of the tion (281, 282). However, these relationships await fur-
sympathetic nervous system buffers surges in down- ther study. The current view is that in humans,
stream microvascular pressure, thus contributing to cholinergic-mediated vasodilation is recognized for pe-
preservation of CBF and potentially the BBB (77, 278). It ripheral blood vessels but not generally considered
is important to realize that both the direct innervation of physiologically important within the brain (36).
cerebral arteries, but also stimulation of adrenergic In summary, cholinergic projections influence paren-
receptors by circulating neurotransmitters, provided chymal neurons and the vasculature by releasing their
they cross the BBB, may contribute to dampening neurotransmitters [i.e., acetylcholine; a potent vasodila-
increases in CBF associated with acute hypertension. tor (294, 297)]. In addition, cholinergic projections inner-
Previous publications provide a more extensive discus- vate interneurons that release NO locally (230, 281, 282,
sion on whether or not autoregulation is influenced by 297). This intrinsic cholinergic system can be pharmaco-
the sympathetic nervous system (291, 292). logically influenced by cholinergic or anticholinergic
drugs. Nonetheless, at this point, it seems unlikely that
3.4.2. Parasympathetic nervous system. this system plays a major role in autoregulation.

Cholinergic nerve terminals are richly distributed 3.4.3. Trigeminal sensory nerves.
throughout intracranial vessels, proximal to the Virchow-
Robin spaces (36). Despite the anatomical presence of In addition to sympathetic and parasympathetic innerva-
this innervation, a clear view on the role of the parasym- tion, cerebral arteries and pial arterioles are innervated
pathetic nervous system in the regulation of CBF in by the trigeminovascular system (36, 299, 300). Roy and
humans is still lacking. In animal models, cholinergic Sherrington (9) provided evidence that stimulation of the
control of CBF seems species specific, with a role trigeminal nerve increased CBF without a change in BP.
observed for the parasympathetic nervous system in Trigeminal nerve fibers release calcitonin gene-related
dogs and rats (293, 294), among others. Although the peptide (CGRP), substance P, and pituitary adenylate cy-
traditional view considered cholinergic-mediated vaso- clase-activity peptide (36, 299, 300). Released by stimula-
dilation to be of limited importance, some recent studies tion of meningeal afferents, CGRP is an extremely potent
suggest the opposite. For example, systemic cholinergic peptide that mediates most of the effects of the trigeminal

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1509


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

nerve on vascular tone. It has therefore become a new The common and internal carotid arteries have been
therapeutic target for treatment of migraine (299). a popular site for ultrasound-based assessment of arte-
Using several approaches, including unilateral trigeminal rial characteristics, largely because they are relatively
ganglionectomy in a preclinical model, insight into vascular easy to access and exhibit a high susceptibility for devel-
effects of the trigeminal vascular system have emerged. For opment of atherosclerotic plaques (310). In addition, for
example, increases in pial arteriolar diameter and CBF in clinical diagnostic purposes (e.g., to detect stenosis),
response to severe acute hypertension (levels of BP that ultrasound-based assessment of the common or internal
exceed the upper limit of autoregulation) are mediated in carotid artery wall thickness demonstrates independent
part by these sensory fibers (301, 302). One implication of prognostic value for future cardiovascular or cerebro-
this work is that increases in CBF during severe acute hyper- vascular disease (311). These structural characteristics
tension are not only caused by autoregulatory breakthrough, may also affect blood flow profiles in the internal carotid
with pressure-driven passive vasodilation (301, 302). In con- arteries. Indeed, assessment of blood flow profiles is fre-
trast, trigeminal ganglionectomy had no effect on regional quently used to evaluate the impact of a stenosis on
CBF under baseline conditions (301, 302), suggesting the downstream blood flow toward the brain. Analysis of
influence of this neural system on resting CBF is minimal. intra- or extracranial artery blood flow and shear stress
waveforms contribute to our understanding the develop-
ment of atherosclerosis in these arterial segments, as
low and turbulent levels of shear stress are associated
4. OTHER FACTORS THAT INFLUENCE with increased atherosclerotic plaque development and
CEREBRAL BLOOD FLOW complexity in the internal carotid artery (312).
Structural (e.g., wall thickening, stenosis) or functional
4.1. Patterns of Blood Flow impairment (e.g., waveform) in large extracranial arteries
supplying the brain may also affect regulation of
In the peripheral circulation, a significant literature CBF. Indirect evidence for this idea came from a meta-
focuses on patterns of blood flow in large arteries. While analysis that reported that carotid atherosclerosis is
resistance arteries and microvessels demonstrate a rela- associated with white matter lesions and silent brain
tively smooth, continuous flow of blood, large conduit infarctions (313). Moreover, in a 7-yr prospective study in
arteries exhibit fluctuations in blood flow across the car- 6,025 dementia-free subjects, carotid artery plaque pre-
diac cycle. These fluctuations relate to the functional dicted development of vascular or mixed dementias
and structural characteristics of large conduit arteries, (314). Whether development of atherosclerosis and pla-
blood volume, pressure gradients (within the artery and ques with resulting changes in blood flow patterns was
across the arterial wall), and blood characteristics (e.g., a direct cause of progression of cognitive impairment
viscosity). remains uncertain. In this context, recent work on doli-
The pattern of blood flow represents an important he- choectasia, an age-associated vascular disease character-
modynamic stimulus in relation to adaptation of conduit ized by increased vessel diameter, increased tortuosity,
arteries in the systemic circulation (303). Blood flow in and altered blood flow, is of interest. Older people with
peripheral arteries is characterized by a large antegrade dolichoectasia carried a higher risk of progression to
component during systole (i.e., blood flows toward the Alzheimer’s dementia (315).
periphery), followed by a short retrograde component More direct evidence for a link between cranial artery
during diastole (i.e., blood flows back to the heart). waveforms and cerebrovascular health was provided in
Acute increases in antegrade blood flow are linked to a study reporting a correlation between diastolic (but
immediate vasodilation, upregulation of antiatherogenic not systolic) carotid artery wall shear stress and cerebral
gene expression and improvement of vascular function infarcts (316). Others confirmed that low carotid artery
(304–306). In contrast, periods of increased retrograde wall shear stress was associated with white-matter
blood flow led to stimulus-dependent impairment, up- lesions and cognitive impairment in older humans (317).
regulation of proatherogenic genes and impairment of Moreover, it was recently reported that carotid artery
vascular health (304, 307–309). Sustained exposure to wall shear stress is independently related to progression
antegrade or retrograde blood flow can ultimately lead of cognitive decline and occurrence of white matter
to changes in vascular structure or stiffness (303). These lesions across a 5.4-yr follow-up in 689 older humans
findings highlight the impact of changes in blood flow (318).
patterns on vascular health in the peripheral circulation. The majority of work on blood flow and shear stress
To date, few studies have focused on blood flow wave- patterns in arteries supplying CBF comes from studies
form patterns in the cerebral circulation, and to what of the common and internal carotid arteries. Current
extent they impact cerebrovascular health. techniques that lack high spatial and temporal resolution

1510 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

to measure cerebral artery diameter make it challenging controls (325). More work will be required to better
to examine shear stress patterns in intracranial arteries. understand if and how patterns of blood flow, rather
CBF pulsatility, estimated from waveform characteristics than just the quantity of blood flow per se, affect the
of the middle cerebral artery using TCD, was investi- effectiveness of autoregulation and brain health.
gated across a large group of healthy subjects ranging
from 22 to 80 yr (319). Advancing age was associated 4.2. Brain-Heart Hydrostatic Gradient
with a linear decline in middle cerebral artery CBF veloc-
ity but also an increase in pulsatility of the waveform in The brain-heart hydrostatic gradient describes the
this artery that started after midlife. A reduction in wall potential influence of gravity on CBF. The influence of
shear stress in cerebral arteries with aging was associ- the brain-heart hydrostatic gradient has been debated
ated with increases in vessel diameter and increased BP since the earliest literature on the cerebral circulation
(320). The potential clinical relevance of these findings (326–329). For an overview and detailed explanation of
is highlighted by the observation that higher CBF pulsa- arguments for and against the relevance of a brain-heart
tility correlated with a greater white matter lesion volume hydrostatic gradient, we refer to a Point-Counterpoint
in older adults (321) and the finding that subjects with discussion (327) and exchange in Anesthesia Patient
Alzheimer’s disease exhibit increased levels of middle Safety Foundation Newsletters in 2007–09. It is not the
cerebral artery pulsatility (322). As a caveat, a “chicken presence of the gradient itself that is debated, but
or the egg” dilemma on causality applies here. Do whether or not the gradient affects CBF. The hydrostatic
altered flow patterns cause vascular injury and then cog- gradient between heart and brain refers to the pressure
nitive deficits, or is it the other way around where gradient in an imaginary fluid column between the level
increased pulsatility is caused by increased vascular re- of the heart and the brain for a person in the upright
sistance due to vascular pathology? position. There are currently two scientific opinions on
Some work in this area has also been performed on how this gradient affects CBF. The first claims that the
the basilar artery. A cross-sectional study reported that pressure gradient on the arterial side is compensated by
lower diastolic (but not systolic or mean) wall shear the gradient on the venous side, so that the net effect
stress in the basilar artery was found in patients with on CBF is null (329, 330). The second opinion claims this
mild cognitive impairment and Alzheimer’s disease, assumption is not valid in the cerebral circulation, and
while levels of wall shear stress correlated with the level therefore perfusion pressure is reduced in the upright
of cognitive decline (323). Here, it is interesting to note body posture by the hydrostatic gradient (i.e., BP-hydro-
that work on dolichoectasia (with potential effects on static pressure) (302, 326, 327).
CBF) hinted at a relationship between a larger basilar ar- Scientists that propose that there are no hydro-
tery diameter and markers of cerebrovascular disease static effects on CBF refer to the cerebral circulation
(324). as a closed-loop system, also referred to as a siphon
Taken together, these studies suggest a link between model (329, 330). The siphon model is the concept of
conduit artery waveforms and cerebrovascular health. a length of rigid tubing containing fluid, where the
The exact underlying mechanisms are difficult to sum- pressure at either end is determined by the vertical
marize at this stage. Changes in waveforms in arteries position of these ends but not by the position of the
may result from a combination of proximal, upstream rest of the tubing. The closed-loop system is similar
alterations (e.g., systolic or diastolic and pulse pressure), but contains a pump that circulates fluid through a
local factors (e.g., endothelial functional, mechanical or length of tubing. If this system is brought from a hori-
structural characteristics), and downstream (distal) varia- zontal plane to a vertical plane, the pump will not
bles in the cerebrovascular tree (e.g., increased resist- require extra energy against the hydrostatic pressure
ance). Whether information on wall shear stress from gradient, because the returning flow now returns with
different intracranial arteries provide distinct prognostic a higher pressure.
information for cerebrovascular health is currently Opponents of this view indicate that this comparison
unknown and should be a topic for future research. For does not hold for the cerebral circulation, for reasons
the purposes of this review, an interesting question is that include nonrigid tubing (collapsible veins), pressure
whether blood flow patterns play a role in autoregula- differences across the system (intrathoracic pressure,
tion. Left ventricular assist devices (LVAD) for patients ICP), and the cerebral circulation itself with its wide
with heart failure offer an opportunity to investigate the range of vessel diameters (326, 327).
effects of different flow patterns. Loss of pulsatile flow in This ongoing debate extends from humans to giraffes
patients with continuous-flow devices had no effect on to sauropods (with their extraordinary hydrostatic gradi-
dynamic autoregulation in a study of 9 patients with con- ent) (330–332) to species of snakes [the heart-brain dis-
tinuous flow LVAD, 5 with pulsatile flow LVAD, and 10 tance is longer in snakes with a mostly horizontal habitat

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1511


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

(i.e., aquatic) vs. snakes in a vertical habitat (i.e., arbo- incorrectly dismissing the siphon hypothesis. For exam-
real)] (333). ple, in a patient with hypotension (mean arterial BP of
It is very complex to perform measurements to prove 50 mmHg), if the siphon hypothesis is correct, the posi-
or disprove either theory. For example, one could study tion of the head compared with the rest of the body
the effect of a change in posture from supine to upright (e.g., upright, supine or head-down tilt for example) has
on CBF. If the only parameter to change were the hydro- no effect on perfusion pressure, and therefore no effect
static gradient, a reduction in CBF following the reduc- on CBF. However, if the siphon hypothesis is false,
tion in perfusion pressure (BP minus hydrostatic the upright position may reduce perfusion pressure
gradient) from supine to upright would suggest the to 30 mmHg, whereas a head-down tilt could increase
hydrostatic gradient affects CBF. Indeed, CBF decreases it to 70 mmHg. This simplified example ignores effects
in human subjects between supine and standing of body position on BP and ICP, but hopefully serves to
(upright) position (331, 332, 334–337). Unfortunately, this explain our point. For further reading on such clinical
change in body position affects multiple factors that also implications, see Pohl and Cullen (345) and the call-out
influence CBF, such as BP, central blood volume, PaCO2 , text for clinicians.
and ICP (334–337). This array of changes makes it diffi-
cult to isolate the single effect of a hydrostatic reduction
in perfusion pressure on CBF. In addition, dynamic 5. REGULATION OF CEREBRAL BLOOD FLOW
autoregulation will modify the effect of a change in per- AND CLINICAL IMPLICATIONS
fusion pressure on CBF (see sect. 2.4 on dynamic
autoregulation). Despite redundancy in mechanisms that regulate CBF,
Parabolic flight experiments offer the unique opportu- diverse clinical conditions have been linked to acute or
nity to modify effects of gravity without changing pos- chronic changes in brain perfusion. In this section, we
ture (338, 339). In a parabola following normal gravity describe the clinical implications of autoregulation in
(1 G), a period of 22 s of 0 G is preceded and followed diseases commonly associated with cerebrovascular
by 30 s of hypergravity (1.8 G). These changes in grav- pathology [e.g., ischemic stroke, hypertension, and
ity occur quickly (in 1 s). For a hydrostatic gradient in a dementias (vascular dementia, Alzheimer’s disease,
subject seated upright in this plane, this pressure gradi- and mixed dementia)] and dysregulation of CBF. Since
ent suddenly nearly doubles for 30 s (from 1 to 1.8 G) and older age is one of the most common risk factors in
then equally suddenly drops to zero for 20 s (from 1.8 G these clinical conditions, we start by discussing the
to 0 G), only to rapidly return to double for another 30 s impact of aging on regulation of CBF.
(from 0 to 1.8 G), after which it returns to normal (338–
342). For autoregulation, these conditions create a se- 5.1. Cerebral Perfusion Across the Age Span
ries of step-wise, very fast changes in perfusion pres-
sure (under the hypothesis that the gradient affects Although the impact of aging on CBF has been fre-
perfusion pressure) of 30 mmHg. Under the opposing quently studied, our understanding of the biology of vas-
hypothesis (siphon), these changes in gravity should cular aging is relatively modest compared with its
have no effect on perfusion pressure. Unfortunately (yet clinical impact (265). Independent of the techniques and
unsurprisingly), the sudden changes in gravity affect designs used to measure CBF, blood flow to gray and
multiple physiological processes (338, 339, 341–343). white matter (or global CBF) decreases by 0.5% per
For example, hypergravity increases BP whereas zero year from early adulthood (346–348). The potential rele-
gravity reduces BP, but also ICP, and PaCO2 (344). vance of a lower CBF may not only relate to its regula-
Nonetheless, in 16 healthy volunteers, with 15 parabo- tion but may also be associated with age-related
las for each subject, the averaged traces of BP and CBF changes in cognitive function (349) and specific
velocity revealed stepwise changes in CBF velocity dur- domains of cognitive performance (350). Recently, a
ing the transitions from hypergravity to zero-gravity and prospective cohort study in 4,759 participants from the
back, while BP remained stable during these changes general Dutch population with a 6.9-yr follow-up found
(343). Plots for BP corrected for changes in hydrostatic that cerebral hypoperfusion at baseline was related to
gradient identified stepwise changes in perfusion pres- accelerated cognitive decline and increased risk for de-
sure that could explain (from the viewpoint of autore- mentia, independent of known risk factors for dementia
gulation) the stepwise changes and subsequent adapta- (351). Separate findings from the Alzheimer’s Disease
tion in CBF velocity (343). This is circumstantial evidence Neuroimaging Initiative reinforced these conclusions,
that is not meant to end this debate. However, from a reporting that a higher index of cerebrovascular resist-
clinical perspective, incorrectly dismissing effects of a ance (ergo lower CBF) predicted cognitive decline (inde-
hydrostatic gradient on CBF carries more risk than pendent of reduced metabolism) and brain atrophy

1512 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

(independent of b-amyloid) in older adults across a 2-yr Another factor that may contribute to age-related
period (352). reductions in CBF is impairment of autoregulation or
Mechanisms that account for age-related decreases related mechanisms. Oudegeest-Sander et al. (362)
in CBF are not fully understood but are likely multifacto- examined the dynamic responses of CBF velocity (using
rial. One potential explanation relates to changes in the TCD) and cortical oxygenation (using near infrared spec-
cerebral metabolic rate (see sect. 3.1), which has been troscopy) to changes in BP in response to sit-to-stand
demonstrated to decline by 0.5% per annum (348). maneuvers and PaCO2 in cognitively healthy individuals:
Older age may impair neuronal or glial mitochondrial young, elderly, and very elderly. While this study con-
metabolism. Indeed, metabolic rates for neuronal tricar- firmed the presence of an age-related decrease in mid-
boxylic acid and glutamate-glutamine cycles are lower dle cerebral artery CBF velocity, the relative changes in
in older adults (353). However, other studies found pro- CBF in response to these paradigms were similar across
gressive reductions in CBF that were independent of age groups, suggesting preserved dynamic autoregula-
changes in cerebral O2 consumption (318, 350, 352, tion as well as preserved CO2 reactivity with older age.
354). These findings suggest that age-related changes Others have also reported preserved dynamic autoregu-
in metabolism cannot fully explain the progressive lation in healthy older populations (100, 363–370).
decline in CBF that occurs as individuals age. Previous studies have linked older age and neurode-
Somewhat related to cerebral metabolic rate is the generation with impaired baroreflex sensitivity (88).
presence of cerebral atrophy (and therefore lower cere- Interestingly, such work suggested that baroreflex and
bral metabolism). Even in healthy individuals, advanced dynamic autoregulation may interact in the regulation of
age is associated with cerebral atrophy. Interestingly, CBF to counteract acute changes in arterial BP. One
age-related atrophy shows regional variability and study found an inverse correlation between baroreflex
seems more prominent in frontal and temporal regions sensitivity and autoregulation (371), suggesting counter-
(355, 356). Regional differences in cerebral hypoperfu- regulatory functionalities between these mechanisms.
sion may also be present with older age (357, 358). However, a recent study examined both dynamic autor-
These findings raise the hypothesis that regions of cere- egulation and baroreflex sensitivity in a cohort of 136
bral atrophy and hypoperfusion with older age may healthy individuals (370). This study confirmed a reduc-
match. To understand this potential link, the spatial pat- tion in CBF and impaired baroreflex sensitivity in the
tern of age-related changes in cerebral atrophy and CBF older population. However, autoregulation was unim-
were explored (358, 359). While these studies con- paired with aging, and there was no relationship
firmed the presence of regional variation in age-related between baroreflex function and autoregulation. Similar
changes in CBF and brain atrophy, regional effects of observations were found in a large data set of healthy
age on CBF differed from that of gray-matter atrophy participants across a wide age range, showing pre-
and were not related to metabolism. Therefore, dissocia- served autoregulation and no relationship between
tion may be present between age-related hypoperfusion autoregulation and baroreflex function (372). Together,
and cerebral atrophy in older humans. Atrophy may also these data do not support a link between baroreflex
affect the reliability of imaging through partial volume sensitivity and autoregulation in the older population.
effects. This may affect imaging-based measurements Studies have also examined the impact of aging on
of CBF and lead to either underestimation or overesti- cerebrovascular reactivity to changes in PaCO2 . While
mation of CBF (348). some studies found that increases in CBF during hyper-
capnia are impaired with aging (46, 373), others did not
5.1.1. Aging and autoregulation, CO2 reactivity, (265, 362). These conflicting findings may result from
and baroreflex function. differences in the methods used to measure CBF (TCD,
MRI), the range of PaCO2 tested, the extent of aging, and
Changes in hemodynamics may also contribute to the the protocol used to induce hypo- and/or hypercapnia
age-related decline in CBF. Aging is associated with a (223). Although some studies do not provide clear
gradual increase in arterial BP, which may be related to insight into the impact of older age on vascular reactiv-
several mechanisms including endothelial dysfunction, ity, these results are largely in line with studies that
aortic stiffening, and an increase in systemic vascular re- examined dynamic autoregulation, in that older age
sistance (360). In addition, assuming ICP does not does not demonstrate a clear age-related decline in the
change significantly (361), an increase in arterial BP ability to regulate CBF in response to stimuli affecting
leads to an increase in cerebral perfusion pressure with BP, arterial blood gases, or pH.
aging. To compensate for such changes and prevent It is important to note that studies of CBF in aging
hyperperfusion, an increase in cerebrovascular resist- have a potential confounder due to age-associated neu-
ance is required. rodegenerative disease (e.g., Alzheimer’s disease,

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1513


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

vascular dementia, and mixed dementia). Because the (72 yr of age) underwent assessment of dynamic autore-
prevalence of neurodegenerative diseases increases gulation during an active stand from sitting. This maneu-
sharply with advancing age (374), it is likely that some ver causes a transient reduction in BP, which usually
studies have unknowingly included older people with reaches a maximum around 15 s after standing and
lower CBF caused by neurodegenerative disease. This recovers within 30–40 s (100, 369, 383). Mean BP fell by
can occur because many of these diseases remain 25%, leading to a reduction in CBF of 15% in old adults
asymptomatic or at least not clinically recognized, in and 19% in young adults. Autoregulation during sponta-
their early stages. neous fluctuations in BP was also assessed using TFA,
during 5 min of quiet sitting or quiet standing. An impair-
5.2. Autoregulation in Hypertension ment in dynamic autoregulation would manifest as a
lower phase and a higher gain (see section 2.4).
There are two widespread, yet incorrect, concepts about However, phase was normal and gain was lower in hy-
autoregulation in hypertension. The first is that chronic pertensive older adults. In contrast with the hypothesis,
hypertension (which is often combined with aging) leads the authors concluded that there were no differences in
to impaired autoregulation (static and dynamic), through dynamic autoregulation between young, older, and
hypertension-associated vascular dysfunction (see Ref. older hypertensive adults (369).
375 or Ref. 376 for examples from review articles where These results were confirmed by a study in 21 hyper-
this concept is presented as a fact). The second, in par- tensive patients (49 yr of age) and 21 normotensive con-
tial contrast with the first, is that static autoregulation trols (384). Here, transient hypotension was evoked by
remains intact in hypertension; however, its lower and standing up from a squatting position, which leads to a
upper levels are shifted toward higher pressure levels stronger drop in BP compared with sit-to-stand. The av-
(377–379). In other words, the autoregulation curve has erage 35–40% transient decrease in BP led to a reduc-
shifted rightwards (376, 380). With this concept, the tion in CBF of 21% in hypertensive adults and 28% in
brain can tolerate higher BP but becomes more sensi- controls (384). In the hypertensive group, there were no
tive to low BP (380). The translation of these two con- differences between those with well controlled BP (136/
cepts to clinical practice is explained in the following 78 mmHg) and uncontrolled BP (154/95 mmHg) (384).
example. A 75-yr-old hypertensive patient has a habitual In a study of acute BP lowering in hypertensive crisis
systolic BP of 160 mmHg. According to the first concept, (385), 28 patients (55 yr of age) with a mean systolic BP
further increases or decreases in BP, albeit fast or slow, of 200 mmHg were randomized to rapid BP reduction
would affect CBF because static and dynamic autoregu- with sublingual captopril or nifedipine. CBF velocity was
lation is impaired. According to the second concept, an not significantly reduced following treatment with capto-
increase in systolic BP to 180 mmHg will not affect CBF, pril but was modestly reduced by nifedipine (385).
because the upper limit of the static autoregulation These three studies with relatively small sample sizes
curve has shifted upward. However, a reduction in sys- were followed by a larger study in 35 normotensive
tolic BP to 120 mmHg may fall below the lower limit of (mean BP, 124/76 mmHg) and 45 hypertensive older
autoregulation, which has also shifted to a higher pres- adults (mean BP, 152/89 mmHg, 68 yr of age) (386). In
sure, leading to a reduction in CBF. With both concepts, this study, the range of systolic BP was wide in the hy-
older hypertensive patients are considered to be vulner- pertensive adults, up to 206 mmHg. The authors investi-
able to cerebral hypoperfusion when BP is reduced gated a wider spectrum of dynamic autoregulation by
(380–382). Common clinical examples of BP reduction looking at effects of decreases and increases in BP that
are those associated with antihypertensive treatment were sustained for up to 3 min, as well as more rapid
(mostly slow, gradual changes) and with postural decreases and increases in BP. For all procedures, the
changes in BP (fast changes, see also sect. 6.1 on pos- changes in BP (up or down) were between 15 and
tural changes). 20 mmHg. The authors found no differences in dynamic
autoregulation between hypertensives and controls
5.2.1. Dynamic autoregulation. (386).
A second large study recruited 60 older adults (72 yr
One of the first studies of dynamic autoregulation in of age); 22 normotensives (BP <140/90 mmHg, no medi-
hypertension tested the following hypothesis: “. . . that cation), 20 controlled hypertension (BP <140/90 mmHg
age and hypertension would impair dynamic autoregula- on medication), and 18 uncontrolled hypertension (sys-
tion, resulting in relative cerebral hypoperfusion during tolic BP >160 mmHg with or without medication) (387).
acute hypotensive stress” (369). Ten young (24 yr of Dynamic autoregulation was examined using TFA of
age), 10 normotensive (mean BP of 125/68 mmHg), and spontaneous changes in BP at rest and by studying tran-
10 hypertensive (mean BP of 153/90 mmHg) older adults sient BP changes during a single sit to stand protocol.

1514 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

This study was comparable in design to the smaller the uncontrolled hypertensive group (389). Thus pre-
study by Lipsitz et al. (369). Following standing, the tran- served static autoregulation can be deduced from the
sient decrease in BP ranged between 20 and 25 mmHg. observation that there was no decline in CBF following
A transient reduction in CBF was observed, 15% in nor- BP reduction. The increase in CBF following BP reduc-
motensives and 10% in controlled and uncontrolled tion is not explained by autoregulation but may point to-
hypertensives (387). TFA found similar values for phase ward a negative effect of chronic hypertension on CBF.
in all groups, whereas, as in the earlier study by Lipsitz Chronic hypertension could impair vascular compliance
et al. (369), the gain was lower in hypertensives. and through this mechanism reduce CBF. Indeed, anti-
hypertensive treatment led to increased carotid artery
5.2.1.1. SUMMARY OF DYNAMIC AUTOREGULATION STUDIES IN distensibility, a measure of vascular compliance (389).
HYPERTENSION. In five studies with 240 participants and a Static combined with dynamic autoregulation were
range of ages from young, middle aged, to older, addressed in a study in 21 newly diagnosed (but
dynamic autoregulation was intact in hypertension dur- untreated) hypertensive patients, 49 yr of age (range
ing nonpharmacological changes in BP. Limitations of from 27 to 66), and 9 controls (390). Of the 21 hyperten-
these studies are that no patients over 80 yr old were sive patients, 12 had mild (BP of 143/88 mmHg), and 9
included and that participants were nonfrail and had lim- had moderate (BP of 163/101 mmHg) hypertension, diag-
ited comorbidity. nosed with 24-h ambulatory BP monitoring. Within 2 wk,
BP was reduced using antihypertensive medications
5.2.2. Static autoregulation. (losartan-hydrochlorothiazide) to 126/77 mmHg in mild
and to 134/84 mmHg in moderately hypertensive
5.2.2.1. STUDIES USING TCD. For clinical translation, the patients, (i.e., an average reduction of systolic BP of 20–
studies described above related mainly to faster BP 30 mmHg), thus lowering BP to or near the lower limit of
changes evoked by daily life challenges such as pos- autoregulation. At that point, CBF was measured dur-
tural changes. An unexplored area was how slower, ing an orthostatic stress test using head-up tilt. This
more gradual reductions in BP using antihypertensive was repeated after longer term treatment (3–4 mo).
treatment would affect CBF. There are two arguments Acute (1–2 wk) reductions in BP brought about by anti-
why this is relevant. First, antihypertensive medication hypertensive treatment did not reduce CBF. Fur-ther-
could have direct effects on cerebrovascular function more, CBF remained stable during the orthostatic stress
that may impair autoregulation. Second, BP lowering test. After 3–4 mo of treatment, BP remained at its
using antihypertensive treatment could reduce BP to the reduced level, and CBF remained stable (390). Thus,
lower limit of autoregulation, such that any further reduc- combining assessments of static and dynamic autoregu-
tion in BP (e.g., during a postural challenge) would lead lation, this study found no evidence for impairment in
to hypoperfusion. Ideally, therefore, studies exploring autoregulation. There was also no evidence of an
these relationships would combine measures of static upward shift of the lower limit of autoregulation (FIGURE 8).
and dynamic autoregulation. Similar to the findings of Lipsitz et al. (390) and Serrador
In one study, normal static autoregulation was et al. (387), the patients with hypertension (but not those
observed in 42 young (34 yr of age) hypertensive with only mild hypertension) had lower gain values than
patients (388). BP was reduced with atenolol from a controls at baseline. An interesting observation is that
mean arterial BP value between 100 and 105 mmHg to gain increased to similar values as in the control group
between 80 and 85 mmHg. CBF velocity was measured following treatment. Lower gain could be interpreted as
after 30 and 60 days and was not reduced (388). In better autoregulation; however, it may also be explained
another study, older hypertensive patients were studied by differences in vascular compliance, supported by the
(389) in three groups: normotensive (n = 19), controlled observation in this study, but also several other studies
hypertension (n = 18), and uncontrolled hypertension of autoregulation (99, 387, 389, 390, 395), that
(n = 14), with a mean age of 70.4, 72.4, and 72.4 yr increases in cerebrovascular resistance are associated
and 50% female. At baseline, mean systolic BP in the with lower gain and that normalization of cerebrovascu-
three groups was 124, 135, and 160 mmHg, respectively. lar resistance increases gain (387, 390).
Only the uncontrolled hypertensive group was treated,
to a systolic BP target <140 mmHg. After 6 mo of antihy- 5.2.2.2. SUMMARY OF STUDIES ON STATIC (COMBINED
pertensive therapy, the group with uncontrolled hyper- WITH DYNAMIC) AUTOREGULATION IN HYPERTENSION.
tension had, on average, a 17-mmHg decline in systolic In n = 123 subjects, including young, middle-aged, and
BP, while CBF velocity increased significantly. At base- old individuals, static and dynamic autoregulation was
line, dynamic autoregulation was similar between investigated before and after pharmacological treatment
groups, and this did not change following BP lowering in to lower BP. Treatment had no effect on dynamic

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1515


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

cerebral arteries. That artery supplies 70% of the blood


flow to the cerebral cortex. Investigators in this area use
TCD because it has the high temporal resolution that is
needed to measure dynamic autoregulation in humans.
For static autoregulation, additional techniques are avail-
able that can provide measurements of global or re-
gional CBF, some of which have been around for more
than half a century. Perhaps the first studies to dispel
the notion of impaired static autoregulation in hyperten-
sion were published in the late 1970s and 1980s (394,
396) but appear to have received little attention. Conen
et al. (396) investigated the effects of short-term (hours)
and longer term (4 wk) BP lowering on CBF, where CBF
was measured using the Xenon-133 method: the same
technique used by Lassen (38) and Harper et al. (70) in
their work on static autoregulation. Patients (n = 10, 37 to
70 yr of age) with a mean arterial BP of  130 mmHg,
requiring emergency BP lowering (for hypertensive
encephalopathy, ICH, fundal hemorrhage, or diabetic
retinopathy) were studied. BP was reduced on average
from 220 mmHg to 150 mmHg systolic in 1 h using in-
travenous nifedipine or clonidine, whereas CBF (60
mL/100 g/min) remained stable overall (396). In that
same study, 21 patients (10 female) with mild to mod-
erate hypertension (50–89 yr of age), were treated
for 4wk with nitrendipine, verapamil, or chlorthali-
done. Systolic BP was reduced from 170 mmHg to
145 mmHg, while CBF remained stable (50 mL/100
g/min) (396).
In 1979, Griffith et al. (394) investigated the effects of
BP lowering using different antihypertensive agents (all
b-blockers) in a large number of hypertensive patients
(27 per group, >100 in total). Mean BP was lowered from
FIGURE 8. Effects of antihypertensive treatment on cerebral blood
values between 135–140 mmHg to between 110–
flow (CBF) in hypertensive patients. Summary of studies that meas-
ured effects of antihypertensive treatment on blood pressure (BP) 115 mmHg. CBF was measured using Xenon-133. In all
and CBF, using different techniques to measure CBF: transcranial patients, and for all b-blockers, despite this substantial
Doppler (TCD; A), MRI arterial spin labeling (B), and Xenon-133 CT BP lowering, CBF remained stable (394) (see also
(C). A: data from a study investigating antihypertensive treatment in
FIGURE 8).
patients with mild or moderate hypertension, after 2–3 wk and after
3 mo of treatment (390). B: summary from 3 studies. Two of these A small study in 1987 using Xenon-133 in eight older
studies (label 1: Ref. 391; label 3: Ref. 392) investigated effects of hypertensive patients found no reduction in CBF follow-
standard versus intensive BP-lowering treatment. One study (label ing BP lowering with prazosin (397). In addition, in 23
2: Ref. 393) investigated effects of stopping antihypertensive treat-
ment (causing BP to increase). C: the study that compared different older hypertensives (66–91 yr of age), systolic BP was
b-blocking agents to lower BP (394). See text for details on all these lowered using amlodipine from 177 to 156 mmHg (24-h
studies. CBFV, middle cerebral artery blood velocity. ambulatory BP) for daytime measurements, and from
157 to 140 mmHg for overnight measurements, in 8 wk.
autoregulation, and follow-up CBF remained stable, sug- There was no reduction in global or regional CBF
gesting normal static autoregulation. Beneficial effects described using SPECT (Tc-HM-PAO) (398). However, in
of BP lowering on CBF (i.e., increases in CBF) were most instances (as in this study) SPECT only allows quali-
observed, possibly due to improvement in arterial tative estimates of CBF patterns, not quantitative CBF,
compliance. unless tracer kinetics are applied (398). Lastly, in 15
patients (9 men, 60–79 yr of age), with hypertension and
5.2.2.3. STUDIES USING XENON-133. In all studies carotid artery stenosis, CBF (measured using Xenon-
described above, CBF was evaluated using TCD to mea- 133) before and 2 h after BP lowering (mean arterial BP
sure changes in cerebral blood velocity in middle from 110 to 102 mmHg) remained stable (399).

1516 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

5.2.2.4. STUDIES USING MRI. In 37 older (74 yr of age) sive crisis, malignant hypertension, or hypertensive ence-
hypertensive patients (systolic BP >150 mmHg), CBF phalopathy, BP levels are extremely high and exceed
was measured using MRI (arterial spin labeling) before the upper limit of autoregulation, (e.g., systolic BP
and after antihypertensive treatment in two different reg- of >180 mmHg) (130). In this situation, an excessive
imens: standard and intensive (392). After 12 wk, systolic increase in CBF and ICP may occur, which explains the
BP was reduced by an average of 15 mmHg in the stand- signs and symptoms associated with hypertensive emer-
ard treatment group and 27 mmHg in the intensive treat- gency: altered mental state, encephalopathy, infarction or
ment group. BP lowering did not result in lower CBF bleeding, optic disk swelling, and retinal bleeds. The pres-
(FIGURE 8). In fact, in the intensive BP-lowering group, ence of signs or symptoms of acute end-organ damage in
CBF actually increased by 10% (392). a patient with very high BP levels (for example, diastolic
The following is an illustration of how persistent the BP >130 mmHg, systolic >200 mmHg) should alert the cli-
concept of impaired autoregulation in hypertension is in nician that the upper limit of autoregulation may have
the literature. Despite accumulating evidence indicating been exceeded, and therefore, autoregulation is impaired
preserved autoregulation in older hypertensive patients, (130). Impaired autoregulation (static and dynamic) in
a study was initiated in 2013 with the hypothesis that patients with hypertensive emergency was demonstrated
older hypertensive patients have impaired autoregula- in a small study using TCD (401), which reported a reduc-
tion, and their antihypertensive treatment would therefore tion in CBF following BP-lowering treatment. It is possible
cause cerebral hypoperfusion (400). It was also hypothe- however that the magnitude of reduction in CBF was over-
sized that stopping their medication would increase BP estimated due to changes in minimum alveolar concentra-
and CBF, leading to improved cognitive function (400). tion (MCA) diameter, affecting blood-velocity measure-
This was a large important study in 102 older, hyperten- ments (401). CBF was unaffected in a small study using
sive adults (81 yr of age). Participants were randomized to Xenon-133, where systolic BP was reduced from 220 to
continuation of antihypertensive treatment (n = 47) or to 150 mmHg in 1 h (396). The clinical relevance is that in a
stopping all antihypertensive medication (n = 55). BP and patient with a hypertensive emergency, a very fast and
CBF (arterial spin labeling) were evaluated at baseline very large reduction in BP must be avoided to prevent cer-
and after 4 mo. It was noteworthy that already at baseline, ebral hypoperfusion. Current guidelines recommend a
the findings were in contrast with the original hypothesis 20–25% reduction in BP (e.g., from 220 to 165 mmHg) in
(393). Hypertension (i.e., higher systolic BP) was associ- the first hour, followed by a target BP of 160/(100–110)
ated with lower, not higher, CBF. At follow-up, discontinu- mmHg in hours 2–6, using intra-arterial BP measurements
ation of antihypertensive medication led to an expected and intravenous antihypertensive agents (130). The vascu-
increase in BP of 10 mmHg systolic. Again, in contrast lar biology in hypertensive emergency is discussed in
with the original hypothesis, this did not lead to an sect. 2.8.
increase in CBF (FIGURE 8) (393). An important contribu- A hypertensive crisis is not only determined by how
tion of this study is that it included subgroups of people high BP levels are but also by how fast they have risen
with small vessel disease, diabetes, and mild cognitive (130). This follows logically from autoregulation: with a
impairment. In these subgroups, there was no evidence slow or gradual increase in BP, autoregulation adapts by
of impaired autoregulation (393). a rightward shift of the upper limit of autoregulation, the
Whether autoregulation is impaired in older hyperten- result being that autoregulation can function effectively,
sive patients with cerebrovascular disease (small vessel even at very high BP levels (e.g., systolic BP of
disease) was further explored in the PRESERVE trial 200 mmHg). In contrast, in an individual with a habitual
(391). Patients with severe small vessel disease were systolic BP of 120 mmHg, a sudden sustained increase
included, i.e., a combination of clinically defined lacunar in BP to 180 mmHg can exceed the upper limit of autore-
infarcts and white matter disease (a Fazekas score of 2 gulation and trigger a hypertensive crisis. In clinical prac-
or 3 out of 3). Sixty-two patients (69 yr of age) were tice, hypertensive crisis tends to be overdiagnosed
included, with relevant comorbidity such as diabetes, because there are many (older) hypertensive patients
cognitive impairment, depression, and smoking. With with chronically elevated BP and because the clinical
treatment, mean BP was reduced from 150/83 to 141/ symptoms in such individuals (altered mental state, con-
79 mmHg in the standard group and from 154/88 to 126/ fusion, encephalopathy) fully overlap with those charac-
75 mmHg in the intensive treatment group over a period teristics of delirium and dementia, which have a high
of 3 mo. In both groups, CBF (measured using MRI arte- prevalence in older patients in acute care settings.
rial spin labeling) was unaffected by BP lowering (391).
5.2.2.6. SUMMARY OF ALL STUDIES ON AUTOREGULATION
5.2.2.5. HYPERTENSIVE EMERGENCY. In hypertensive IN HYPERTENSION. Between 1979 and 2018, 747 hyper-
emergencies, also described in the literature as hyperten- tensive patients have been investigated. In 240 patients,

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1517


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

dynamic autoregulation was studied (without evaluation There have also been prospective, randomized trials
of treatment). All studies found that autoregulation was that have evaluated the outcome of BP lowering on cog-
similar to normotensive controls. In 507 older patients, nitive decline. The SPRINT trial was a study of intensive
autoregulation was evaluated before and after BP-low- versus standard antihypertensive therapy. It included
ering treatment. In 384 patients, this was an evaluation patients >75 yr old, with a subgroup of moderately frail
of static autoregulation. In 160 patients, CBF was meas- participants. The SPRINT-MIND substudy (n = 9361) was
ured using Xenon-133 CT, in 23 with SPECT, and in 201 specifically designed to evaluate cognitive function
patients with MRI (arterial spin labeling). In 123 patients, (405). If autoregulation were impaired in hypertension,
both static and dynamic autoregulation were evaluated the expected outcome would have been that intensive
before and after treatment, using TCD to measure BP lowering, through cerebral hypoperfusion, would
changes in CBF velocity. Individuals that were included promote cognitive decline. The contrary was found. The
were not only otherwise healthy and nonfrail but intensive treatment group had less risk of developing
included a substantial number of people over 70 and mild cognitive impairment or dementia than the standard
even 80 yr of age with comorbidities (vascular disease, treatment group (405). Equally, there was no increase in
including cerebral small vessel disease, diabetes, risk of falls or fractures in the SPRINT study (405, 406).
depression) and cognitive impairment. All these studies The prevalence of syncope was not increased (see sect.
came to the conclusion that autoregulation is not 6.1.2: note that syncope is a disorder of BP regulation,
impaired in hypertension. FIGURE 8 provides a sum- much more than a disorder of autoregulation). In the
mary of studies that investigated static autoregulation in subgroup of patients >80 yr old, intensive treatment
hypertension. was associated with increased risk of a decline in kidney
function (406). The clinical consequences of this reduc-
5.2.3. Hypertension and adverse outcomes. tion in kidney function (which was defined as a 30% or
more reduction in estimated clearance) remain uncer-
Evidence suggesting normal autoregulation during hyper- tain. A recent study indicates that these reductions in
tension can also be obtained from studies that did not estimated creatinine clearance do no really represent
measure CBF. Such studies investigated adverse out- clinically relevant reductions in renal function (407). In
comes associated with cerebral hypoperfusion, the addition, this older subgroup demonstrated reductions
feared outcome of BP lowering if autoregulation is in cardiovascular events and mortality and reductions in
impaired. Examples of adverse outcomes related to cere- cognitive decline with intensive BP lowering, without
bral hypoperfusion include falls, dizziness, cerebrovascu- increased risk of reduced mobility (gait speed) or falls
lar lesions, mild cognitive impairment, or dementias. (406). Of note, the benefit was influenced by baseline
Interestingly, in a prospective cohort of almost 600 older cognitive function: in those with poor cognitive function
hypertensive patients (70–97 yr of age), the use of antihy- at baseline (defined as a Montreal Cognitive Assess-
pertensive medication to lower BP was associated with a ment score below 18 or 20 depending on level of educa-
substantially lower odds ratio (0.6) for the risk of falls tion), there was no or limited benefit of intensive treat-
(402). This result strongly argues against the concept of ment on all endpoints, including cardiovascular events.
impaired autoregulation with aging and hypertension and This seems to emphasize the role of early start of antihy-
also against the concept of an upward shift of the lower pertensive treatment in the prevention of cognitive
limit of autoregulation (402). The increased risk of falls in decline.
patients not using antihypertensive medication may be The INFINITY trial also investigated effects of inten-
explained by the observation that untreated hypertension sive versus standard antihypertensive therapy in older
(in older people) increases orthostatic hypotension. patients (199 randomized patients, 54% women, 80.4 yr
Orthostatic hypotension is a strong risk factor for falls of age) (408). At study entry, subjects had a systolic BP
(403, 404). The findings by Lipsitz at al. (138) have subse- >170 mmHg, or between 150 and 170 mmHg on antihy-
quently been confirmed by others, as summarized else- pertensive medication. All subjects had evidence of
where (404). These studies highlight that hesitance to white matter disease on MRI. They were then treated
treat hypertension in older people, based on the assump- from a mean systolic BP (based on 24-h ambulatory
tion of impaired autoregulation and fear of induced cere- monitoring) of 149 mmHg to 128 mmHg (intensive) or
bral hypoperfusion, is unsupported and may in fact have 144 mmHg (standard), with a 3-yr follow-up (408).
the opposite effect on the risk of falls (402, 404). Outcome measures were gait speed, cerebrovascular
Untreated hypertension can increase the risk for ortho- lesions, volume of white matter lesions, falls, and syn-
static hypotension, with large transient reductions in BP, cope. Impairment of autoregulation would be predicted
which may not be prevented by autoregulation and cause to manifest as a slowing of gait, an increase in white mat-
cerebral hypoperfusion and falls. ter lesion volume, and/or an increase in falls. However,

1518 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

these were not found. In contrast, the time-dependent autoregulation a top research priority to inform clinical
increase in white matter lesion volume was smallest in decision-making, with the aim of preventing secondary
the intensively treated group (408). Finally, a meta-analy- damage due to extremes of hypo- or hyperperfusion
sis of 31,090 individual participant data from trials per- (FIGURE 1) (410, 411). The need for a better understand-
formed between 1980 and 2019 investigated the risk of ing of autoregulation following stroke has led to a sub-
development of dementia associated with use of antihy- stantial literature in this area as reflected by the number
pertensive medication (409). Participants were 55 yr and of reviews dedicated to the topic (32, 41, 139, 379, 410,
older (50% were >75 yr, but only 5% were >85) and 412–421). The main aim of this section is to assess pro-
were followed for at least 5 yr. In 15,000 participants gress with the understanding of autoregulation patho-
with a baseline systolic BP <140 mmHg, antihyperten- physiology in human stroke and to identify priorities for
sive use did not increase dementia risk. In the 15,000 future research.
participants with baseline systolic BP >140 mmHg, use Ischemia impairs CBF regulatory mechanisms, includ-
of antihypertensive medication even reduced dementia ing autoregulation (44, 416, 418, 422), and hence, it is
risk (RR 0.88) (409). There were no differences between not surprising that most studies of autoregulation have
classes of antihypertensive medication. shown that autoregulatory mechanisms are often
impaired following both ischemic stroke and ICH (379,
5.2.3.1. CONCLUSIONS. Despite the evidence presented 410, 412, 414, 415, 417, 420, 423). In addition, autoregula-
above, statements claiming that autoregulation is tion has been shown to be impaired in most studies fol-
impaired in hypertension, and that older hypertensive lowing aneurysmal subarachnoid hemorrhage (SAH)
patients require higher BP because of a rightward shift (424–440). The finding that autoregulation tends to be
of the autoregulation curve, are still found frequently in impaired in the main types of stroke (ischemic or hemor-
the literature. Very often, they are presented as facts, rhagic) has come from controlled and observational
sometimes without references. Equally, there is an abun- studies where patients have been compared with
dance of cross-sectional studies dealing with BP levels healthy controls as distinct groups. However, this cannot
and treatment of hypertension, sometimes with, but of- be generalized to all stroke patients as some studies,
ten without, measurements of CBF, that nevertheless mainly involving mild strokes, have shown that many
draw inferences regarding the relationship between BP stroke patients maintain intact autoregulation (410, 412,
and CBF. The bias in such studies almost always leads 423, 441–447). Moreover, even in the case of patients
to observations that people with low BP have worse out- with moderate to severe strokes, autoregulation is not
comes in mortality or cognition than people with high impaired at all time points. Indeed, one of the main items
BP. Such anticipated outcomes may be explained of interest is the temporal evolution of autoregulatory ef-
because BP levels often decline in cancer, heart failure, ficacy poststroke to inform treatment decisions in a
or dementia. Nevertheless, and almost without excep- timely fashion (423). In addition to the need to under-
tion, such studies end with a conclusion that the nega- stand the time course of impairment of autoregulation
tive outcome can be explained by a low CBF, caused by following stroke, individual heterogeneity due to pheno-
low BP and impaired autoregulation. type, stroke etiology, severity, subtype, location, comor-
However, based on prospective studies that measured bidities, and early intervention, as well as the different
CBF as an outcome in >700 older hypertensive patients, protocols and techniques used for assessment of autor-
there is no convincing evidence that autoregulation is egulation, represents a highly complex, multifaceted
impaired in hypertension, nor is there evidence that older problem that has not been addressed in a comprehen-
hypertensive patients require higher levels of BP to pre- sive manner. The extent of impairment of autoregula-
serve CBF. This conclusion is further supported by the tion, and its poststroke temporal evolution, also likely
outcomes of three large prospective studies, and one depend on the volumes of the core and penumbra
recent large meta-analysis, of antihypertensive treatment regions (sect. 1.2) and patterns of reperfusion (26, 415,
(combining >25,000 patients >75 yr of age) that found 448, 449). The role of these multiple factors is well illus-
no evidence for adverse effects that could be attributed trated by one of the first studies of autoregulation in is-
to cerebral hypoperfusion, including falls, cerebrovascu- chemic stroke that proposed a quantitative measure of
lar lesions, or cognitive decline and risk of dementia. static autoregulation, as the ratio of changes in CBF
[measured with hydrogen clearance and cerebral (A-V)
5.3. Cerebral Perfusion and Stroke: Immediate CaO2] and cerebral perfusion pressure (invasive meas-
and Long-Term Changes urements of BP and ICP) (450). Meyer et al. (450) found
that dysregulation was associated with severity and
The key role of BP control in the management of acute location. In other words, there was worse autoregulation
ischemic and hemorrhagic stroke makes assessment of with brainstem or subcortical lesions as compared with

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1519


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

cortical strokes. Moreover, they reported an inverse cor- By far, the main approach has been TFA, in conjunction
relation with time of measurement (day 1 to 44) after with spontaneous fluctuations in BP, with the use of the
ictus, suggesting less dysregulation with longer duration amplitude (gain) and phase frequency responses as
since ictus, without an influence of age or hypertension. metrics of dynamic autoregulation (95, 110, 434, 464–
467). At each frequency, gain reflects the ratio of output
5.3.1. Methodological aspects of autoregulation (i.e., CBF or CBF velocity) to input (i.e., BP) amplitudes,
assessment in stroke. while the phase expresses the time delay between out-
put and input. With failing autoregulation, one would
Before reviewing the different factors that can influ- expect the gain to increase (that is reduced buffering of
ence autoregulation in ischemic or hemorrhagic BP fluctuations) and the phase to decrease (that is the
stroke, it is important to call attention to the divers- tendency of output to follow the input). A recent system-
ity of protocols and measures of both static and atic review and meta-analysis has shown that phase is a
dynamic autoregulation that have been used in these more reliable parameter than gain to detect changes in
studies and their potential to confound findings. autoregulation in ischemic stroke (414), something that
Assessment of autoregulation based on static meth- had been highlighted in previous reviews (55, 91). When
ods requires induction of changes in mean BP that comparing studies that used phase or gain, it is impor-
have been performed by means of tilting (450) or the tant to consider the different settings that are required in
use of vasoactive drugs such as nicardipine or labe- TFA, which may confound comparability. To address this
talol (442, 443). In addition to changes in physio- problem, a White Paper proposed standardization of
logical processes that can result from these manipu- TFA settings aimed at improving comparability of future
lations of BP, such as changes in PaCO2 with tilting, studies (55). When BP changes were induced by
there are also concerns about estimating the slope of means of thigh cuffs, efficiency of autoregulation
Lassen’s static autoregulation curve using only two was expressed with the rate of regulation (101) and
points, which can potentially lead to substantial autoregulation index (ARI) (90, 429, 451–454) indexes.
errors (40). The rate of regulation was adapted to express the rate
The majority of the literature is dominated by studies of recovery of the CBF velocity response to a step
using the dynamic autoregulation approach. Although change in BP (calculated via TFA) (464, 468, 469) and
originally proposed in combination with sudden reduc- also to studies of autoregulation in stroke based on
tions in BP induced by the rapid release of compressed MRI estimates of CBF (470). Following the demonstra-
thigh cuffs (90, 101), dynamic autoregulation assessment tion that the ARI can be calculated by means of TFA,
has been increasingly based on spontaneous fluctua- using spontaneous fluctuations in BP (108), several
tions in BP, whose convenience is particularly relevant in studies have used this approach in both ischemic and
the acute stroke setting (113), where patients may be hemorrhagic stroke (441, 446, 454, 459, 471–473).
unable to perform or tolerate postural changes or other This alternative has a number of advantages as it
interventions to manipulate their BP. Concern about the abbreviates several choices that need to be made
sufficiency of BP variability in spontaneous cardiovascu- when using phase or gain directly (55).
lar fluctuations (114) has led many investigators to revert When using spontaneous fluctuations in BP, one alter-
to the thigh cuff approach (429, 451–454) or to use alter- native to TFA is the mean velocity index, usually referred
native ways to provoke rapid changes in BP, such as to as Mx (or Mxa) that has been adopted in autoregula-
rhythmic handgrip (455), tilting (453), paced breathing tion studies of both ischemic and hemorrhagic stroke
(434, 456, 457), Valsalva maneuver (458), rapid changes (including SAH) (421, 426, 428, 437, 440, 444, 445, 449,
in head position (459), brief carotid artery occlusion 467). A variant of the Mx, the Sx index, using the systolic
(424, 433, 435, 438, 460), or elbow flexion (461). The value of CBF velocity, instead of the mean value, has
concomitant changes that these maneuvers can induce been adopted in SAH studies (426, 427, 437, 460). The
in covariates of autoregulation, such as PaCO2 , heart Mx/Mxa is correlated with the ARI (474), but hitherto it
rate, cardiac output, and activity of the sympathetic nerv- has not been scrutinized regarding its sensitivity to
ous system, need to be kept in mind when comparing noise, measurement errors, and choice of parameter
studies using different protocols (113). A few studies settings. In situations when measurements of ICP are
used spontaneous fluctuation recordings but attempted available, the PRx index, expressed by the correlation
to improve reliability of autoregulation parameters by coefficient between mean arterial BP and ICP, has also
using only larger pressor or depressor transients in BP been used, mainly in studies of SAH (430, 475–477). In a
identified in the data (452, 453, 462, 463). few studies, indirect measures of CBF were obtained
A relatively large number of indexes have been with methods reflecting the utilization of O2, leading to
adopted for estimation of autoregulation efficiency (91). correlation indexes, similar to the Mx or Sx, labeled TOx

1520 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

(427, 440, 460, 476) or ORx (430, 432), from measure- phase, derived with the MMPF method, was lower in
ments with near-infrared spectroscopy (NIRS) or an inva- comparison with controls, an average of 18 and 6 mo af-
sive tissue O2 tension probe, respectively. ter stroke onset, respectively, but other investigators
Finally, modeling of autoregulation in stroke has been reported normal values of autoregulation parameters 1
performed with the multimodal pressure-flow (MMPF) mo up to 63 mo after ictus (453, 482). In summary, most
method (458, 478, 479). This approach generates esti- studies assessing the evolution of autoregulation param-
mates of phase differences between fluctuations in BP eters in ischemic stroke, have shown a deterioration of
and CBF velocity (either spontaneous or from Valsalva autoregulation metrics in the first 2 wk after ictus, with
maneuvers), but these should not be confounded with return to normal values between 1 and 3 mo later.
phase differences derived by TFA as there are substan- Although the corresponding number of studies in ICH
tial differences in the data processing involved. In fact, is much smaller, there seems to be a similar pattern.
the authors have shown that MMPF seems to provide Based on TFA phase and the ARI, respectively, Oeinck
greater sensitivity than TFA in detecting deterioration of et al. (465) and Minhas et al. (472) have not observed
autoregulation in stroke and other conditions (95, 110, any changes in the TFA phase and ARI, respectively, in
458, 464, 465, 467, 480, 481). the first 5 to 14 days after ictus. Reinhard et al. (467)
reported Mx index values not different from controls, on
5.3.2. Temporal course of changes in day 1, but by day 5, higher values of the Mx index (indi-
autoregulation following stroke. cating worse dynamic autoregulation) were a significant
predictor of outcome. A longer follow up (464) indicated
Most studies of autoregulation in ischemic stroke have that TFA phase was lower than controls from days 1–2,
performed assessments in the acute (<48 h) and suba- reaching a minimum by days 10–12, but then recovering
cute phases (<14 days after ictus) (410, 412), with a much to comparable values as controls at 30 days after ictus.
smaller number of studies performing measurements Therefore, these data on TFA phase and ARI support
several weeks or months after stroke onset (450, 452, the earlier temporal changes in autoregulation after
453, 455, 458, 459, 473, 478, 479, 482, 483). Given the stroke, with a decline in the first 2 wk after ictus and a
considerable heterogeneity of patient conditions and return to normal values thereafter.
autoregulation assessment methodology, inferences In SAH, several studies reported impairment of autor-
about longitudinal changes in autoregulatory metrics egulation from days 1 to 4 after admission (424–426,
can only be made from a relatively small number of stud- 428–430, 432, 435, 436, 439, 440, 466), in some cases
ies that performed two or more intrapatient measure- with temporary improvement, followed by further deteri-
ments. In addition to the time period encompassing oration around days 7–14 (427, 429, 430, 434, 435). The
measurements, it is also relevant to take into considera- occurrence of vasospasm, often followed by delayed
tion whether autoregulation parameters were found to cerebral ischemia (DCI), shows an interaction with autor-
be altered in comparison with controls, or not. In studies egulation impairment.
that did not have a control group, or indications that The temporal evolution of effectiveness of autoregula-
autoregulation was impaired at any time following tion, as suggested by different metrics, needs to be
stroke, autoregulatory metrics were found to deteriorate taken into account to inform the clinical management of
within the first 5 days following stroke (444, 449). In con- patients with either ischemic or hemorrhagic stroke but
trast, others detected improvement over time, mainly in cannot be considered in isolation from a number of
the acute and subacute stages (445, 484), although other factors, such as the severity of stroke.
Kwan et al. (455) found increased values of TFA phase
and decreased gain 3 mo after stroke onset. When con- 5.3.3. Influence of stroke severity and subtype on
trol groups were included, measures of autoregulation estimates of autoregulation.
indicated that impairment was present from the acute to
the subacute stages, up to 28 days after stroke onset When examining the influence of stroke severity, con-
(452, 471, 485, 486). Noteworthy, two studies reported trolled studies in ischemic and hemorrhagic stroke indi-
normalization of the ARI after 3 mo, following signifi- cate that indexes of autoregulation show normal values
cantly reduced values of this index at approximately 5 in mild strokes but have a greater tendency to present
days (459) and after 2 wk (473), respectively. On the abnormal values in moderate and severe strokes (410,
other hand, Guo et al. (483) obtained reduced values of 412, 415, 416, 421). The volumes of the core infarct and
TFA phase, suggesting impaired dynamic autoregula- penumbral regions (26) likely influence the relationship
tion, at <48 h after onset that were not altered 6 mo between autoregulation and stroke severity, but distin-
later. Although serial assessments were not performed, guishing the core and penumbral regions has been
Novak et al. (458) and Hu et al. (479, also found that the elusive (415). In many studies, the association of

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1521


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

autoregulation with severity is incidental without formal 486, 494). Most of these studies have not found a signifi-
statistical testing. However, a reasonable number of cant, multiclass association of autoregulation status with
studies have used a nominal or ordinal scale to assess either the OCSP or the TOAST classifications, with the
the relationship between indexes of autoregulation and exception of Ma et al. (486) who reported lower values
corresponding measures of severity. The National of TFA phase in strokes due to large artery atherosclero-
Institutes of Health Stroke Scale (NIHSS) is the main tool sis when compared with small vessel occlusions. A
for quantifying severity in stroke, with values 1–5 indicat- larger number of studies however investigated interhe-
ing a mild stroke and very severe strokes corresponding mispherical comparisons of autoregulation parameters
to values >25 (441, 444, 446, 462, 486). Other measures for different subtypes of ischemic stroke. In general,
of severity that have been used are the Glasgow Coma small vessel disease and lacunar strokes result in bilat-
Score (GCS) (443, 464, 465, 467), the Barthel Index eral impairment of autoregulation, as might be expected
(462), degree of stenosis (456, 468, 487, 488), or other given their more diffuse distribution (191, 458, 463, 469,
scales (450). With the use of MRI or CT imaging, classifi- 483, 495). For large vessel occlusions though, a more
cation of severity has also been based on the volume of complex pattern emerges, given the expectation that
infarct or hematoma (442, 443, 448, 449, 467, 489, 490), unilateral lesions should lead to impairment of autoregu-
the presence of brain atrophy (478), hemorrhagic trans- lation in the affected hemisphere, when compared with
formation in ischemic stroke (489), or ventricular hemor- the unaffected hemisphere. This was shown to be the
rhage in ICH (467, 472). In SAH, the most common case in some studies (463, 469, 484, 490), but in other
measures of severity are the GCS, the Hunt and Hess studies autoregulation was depressed in both hemi-
score, the World Federation of Neurological Surgeons spheres, despite unilateral occlusion (486, 495). In the
Scale (WFNS), or the Fisher scale (491). In summary, in- majority of studies though, the population included a
dependent of the measures used to express stroke se- mixture of stroke subtypes, with small vessel disease or
verity, or the parameter adopted for assessment of lacunar infarcts representing a variable proportion (usu-
autoregulation, these studies found no association ally <50%) of the sample. What is startling is that in all
between autoregulatory metrics and stroke severity for these studies, both hemispheres had depressed autore-
mild strokes (442–444, 446, 462, 486, 487). However, in gulation, independently of the metric adopted for autor-
moderate and severe strokes, there is a positive associ- egulation (441, 442, 451, 454, 455, 459, 462, 470, 471,
ation between stroke severity and worse autoregulation, 473, 478, 479, 496). In ischemic strokes of undeter-
independently of stroke etiology (441, 446, 448–450, mined etiology, Tutaj et al. (457) found impaired autore-
456, 464, 465, 467, 468, 478, 487–489, 492). The influ- gulation only in the unaffected hemisphere. In ICH, most
ence of stroke severity on autoregulation in SAH is more studies did not find a difference in autoregulation pa-
complex, due to the development of vasospasm and rameters between sides, and an association with hema-
DCI, entities that by themselves are dependent on se- toma location was not reported (448, 464, 465, 467,
verity. Direct association of the Hunt and Hess, WFNS, 472). Of note, in some of these studies, the autoregula-
Fisher, or GCS with autoregulation was reported in only tion metric was not different from controls. For SAH, we
a few cases (435). On the other hand, multiple studies could not find any reports of an association between the
suggested an association of indexes of autoregulation extent of impairment of autoregulation and the location
with the occurrence of vasospasm (424, 429, 434, 436, of the ruptured aneurysm. One possible explanation for
437, 477) or DCI (426, 428, 430, 433, 435, 436, 493). this lack of specificity, is that the diffusion of blood in the
Given that autoregulation was found to be depressed af- subarachnoid space, and its effects of triggering endo-
ter SAH, often before the development of vasospasm thelial dysfunction, reactive oxygen species formation,
and DCI, several authors have proposed predictive mod- and hypersensitivity of vascular muscle to contraction
els, showing that alterations in autoregulation indexes (139, 419, 420, 497), make the corresponding loss of
were independent predictors of vasospasm and/or DCI autoregulation independent of the site of bleeding.
(424, 428, 435, 436, 460, 466). The interdependence of
stroke subtypes, severity, phenotype, and comorbidity 5.3.3.1. INTERIM SUMMARY. Autoregulation parameters
makes it difficult to unravel the separate influences of after ischemic or hemorrhagic stroke tend to show simi-
these factors on autoregulation. Studies specifically lar values in both hemispheres, independently of loca-
designed to assess the influence of stroke subtypes on tion or subtype, with only a few studies reporting
autoregulation have adopted either the Oxford unilateral impairment. The difficulty of explaining the
Community Stroke Project (OCSP) or the TOAST criteria extent of autoregulation impairment based on stroke
for classifying stroke subtype (494) but have not con- subtype is undoubtedly related to the intervening effects
trolled for co-factors and usually involved a relatively of stroke severity. A few studies highlight this interde-
small number of participants (441, 451, 454, 462, 469, pendence (446, 488, 495), but much more work is

1522 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

needed to improve our understanding of how autoregu- newborns through the concept of optimal cerebral per-
lation is influenced by the confluence of stroke subtype fusion pressure or optimal BP (493, 500). Given the long
and severity. monitoring times required by this approach, indexes of
autoregulation have been estimated with NIRS or from
5.3.4. The association of autoregulation with the BP-ICP relationship, with the pressure-reactivity
stroke outcome. index (501), rather than with indexes derived from CBF
velocity (TCD). By observing changes in pressure-reac-
The potential association of autoregulation efficiency tivity index or a number of different indexes obtained
with the outcome of stroke is of considerable interest, from NIRS (500), a U-shaped curve can be derived as a
not only for its more immediate importance to guide function of BP or cerebral perfusion pressure, with its
patient management, but also to shed light on the causal minimum indicating the optimal value to maximize autor-
mechanisms involved in the pathophysiology underlying egulation efficacy (502). A recent NIRS-based study has
alterations of autoregulation. The relevance of this asso- demonstrated the feasibility of using the optimal-BP
ciation is reflected by the recent increase in the number approach in ischemic stroke, in patients undergoing me-
of studies describing the relationship between autore- chanical thrombectomy (503). A similar approach in SAH
gulation and outcomes. In these studies, outcome has has shown a strong association between outcome and
been nearly unanimously expressed with the modified the time patients spent outside the boundaries of the
Rankin Scale (mRS) (498) at 90 days after stroke onset, optimal BP interval (475, 476). The possibility of individu-
dichotomized as good (0–2) or poor (3–6). In ischemic alizing optimal BP management, as compared with the
stroke, poor outcomes were associated with parameters adoption of fixed BP targets, and the better outcomes of
indicating impairment of autoregulation; reduced TFA the former (475, 476, 503), warrant further research,
phase (456, 486, 490, 496), increasing Mx index (445, including large scale clinical trials. On the other hand,
449), increased TFA gain (499), or lower ARI (446). The the fairly long observation times required with the opti-
short-term prognostic value of depressed dynamic mal BP approach, which was 28 6 18 h (503), 68 6 51 h
autoregulation, lower TFA phase 6 h after stroke onset, (476), or an average of 95 h per patient (475), might be a
was shown by its association with hemorrhagic transfor- critical limitation for extending this approach to all stroke
mation and cerebral edema (489). Similar associations patients. Moreover, unless mean BP is manipulated, its
of TFA phase and the Mx index with outcome were spontaneous variation might not necessarily cover a suf-
observed in ICH (464, 465, 467). In patients with SAH, ficient range to show a U-shaped curve and hence the
outcome was often assessed with the mRS at discharge, identification of the optimal BP target. Alternatively, a
as well as at 3, or sometimes 6, mo after discharge, with simpler approach to restoring autoregulation to normal-
the majority of studies reporting a strong association ity was proposed in the context of ICH (472), by using
with autoregulation (426, 429, 430, 432, 434, 435, 475, hyperventilation to improve dynamic autoregulation by
476). A measure of functional independence and the ef- means of hypocapnia (101, 223). The possibility of adopt-
ficiency of rehabilitation were also associated to severity ing a similar strategy in ischemic stroke, could be limited
of SAH and the shape of the autoregulation curve (425). by the observation that spontaneous hypocapnia al-
ready seems to be prevalent in this population (504).
5.3.4.1. INTERIM SUMMARY. Taken together, the accu- Furthermore, the effects of hypocapnia on CBF are tran-
mulated evidence that autoregulation is often impaired sient, as CBF starts to return to normal after 4-6 h of
in ischemic stroke, ICH, and SAH, with significant associ- hyperventilation (505).
ations to severity and outcome, suggests that further Although restoration of autoregulation efficacy seems
consideration should be given to incorporate autoregu- an intuitive target in the treatment of ischemic and hem-
latory assessment as part of routine protocols to man- orrhagic stroke, its judicious application to patients with
age patients with stroke. To progress in that direction different phenotypes, stroke subtypes, location, and se-
though, there are important gaps in our knowledge verity, will undoubtedly require more work to fill in the
about the pathophysiology of autoregulation in humans gaps in knowledge identified in the sections above.
that need to be addressed. Considerably more data are needed to characterize the
The association of different indexes of autoregulation temporal course of autoregulation stratified by stroke
with severity and outcome of ischemic and hemorrhagic subtypes, location, and severity, for example. Likewise,
stroke would suggest that one possible strategy to the recovery in autoregulation, observed between 1 and
improve stroke treatment is to take autoregulation effi- 3 mo after ischemic stroke onset (459, 473), needs to be
cacy into account in treatment protocols. This approach better understood, in particular how individual time
has been gaining momentum in the critical care of courses relate to outcome, which hitherto remains
patients with severe brain injury and also in premature unexplored.

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1523


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

Although targeting improvement of autoregulation as placebo or other controls, but could not demonstrate a
a potential coadjutant in stroke therapy is an avenue difference in favorable clinical outcomes, usually based
that needs further exploration, the overall conclusion so on mRS <3 (507–514, 516). On the other hand, most tri-
far is that advances in clinical or surgical management of als have not detected any adverse effects of antihyper-
patients with ischemic or hemorrhagic stroke are tensive therapy either. Noteworthy, the Controlling
unlikely to be achieved without due consideration for Hypertension and Hypotension Immediately PostStroke
the key role of autoregulation in poststroke pathophysi- (CHHIPS) trial, comparing the use of labetalol or lisinopril
ology and management. to placebo, reported a reduction in mortality at 3 mo
from 20.3% (placebo) to 9.7% in the treatment group
5.3.5. Autoregulation and blood pressure control (509). A pooled analysis of two previous large RCTs
in stroke. showed that achieving early and stable systolic BP was
associated with favorable outcomes in patients with ICH
Although the role of autoregulation has not been directly (513).
investigated in large randomized controlled trials (RCT) The lack of impact of BP-lowering therapy on the out-
of BP management in ischemic or hemorrhagic stroke, come from stroke, as reported by most trials, would sug-
some indirect evidence about autoregulation efficacy in gest that, contrary to the underlying assumptions,
stroke can be inferred from the results of several RCTs, autoregulation was intact in the populations studied and
and important lessons can be learned for improving the the reductions in BP did not influence cerebral perfu-
design of future trials. Following ischemic stroke, a sig- sion. This interpretation, however, needs reconsidera-
nificant number of patients present with hypertension tion, taking into account three key aspects of the
(411, 506), which has been the main drive to test different involvement of autoregulation in ischemic and hemor-
approaches to lowering BP in the acute and subacute rhagic stroke: 1) severity of stroke, 2) extent of BP reduc-
stages (507–511). Theories about autoregulation in tion achieved, and 3) timing of intervention and
stroke play an important role in the rationale behind assessment. The association of stroke severity with
such trials. For example, the increase in BP can be impairment of autoregulation would suggest that most
hypothesized as serving a purpose of maintaining CBF large RCT samples would involve a mixture of patients
(specifically in the penumbra) and lowering BP may with autoregulation ranging from intact to severely
therefore be disadvantageous. On the other hand, if affected and that the accruing benefits of BP lowering
autoregulation is impaired in the penumbra, the increase could then be diluted by the prevalence of less severe
in BP following stroke may cause hyperperfusion injury strokes. As an example, in the pooled analysis of the
in the infarcted area, and BP-lowering interventions may INTERACT-2 and ATACH-2 trials, the patients showing
prove beneficial. However, excessive BP lowering may improved outcomes were mainly those with mild-to-
cause hypoperfusion injury if autoregulation is impaired. moderate ICH (513). Although most trials demonstrated
This reasoning also extends to ICH, where antihyperten- a significant reduction in BP in the treated group com-
sive therapy has been the focus of many trials in ICH in pared with controls, the amplitude of the difference is
the attempt to reduce the risk of hematoma expansion likely to have an association with outcome. In the
and intraventricular hemorrhage due to elevated BP RIGHT-2 trial, early application of transdermal glyceryl
(512–514). Many other hypotheses on what would consti- trinitrate led to a mean reduction in BP of only
tute optimal BP management in stroke can be formu- 5.8 mmHg in comparison with patients receiving a sham
lated based on assumptions of autoregulation in the patch (508), while the trial in ICH, where improvements
affected infarct area, affected hemisphere, or unaffected in outcome were observed, had a sustained mean sys-
hemisphere. The setting of optimal BP targets, following tolic BP reduction of 29 mmHg (513). In line with this lat-
ischemic stroke, is also complicated by differences in ter observation, the CHHIPS study, where a difference in
the extent of the collateral circulation in individual mortality was detected, had a systolic BP reduction of
patients, which will play a key role in reperfusion of the 31 mmHg (at 2 wk) compared with placebo (509).
penumbra (411, 515). In summary, in both types of stroke, Although a “dose-response” relationship remains to be
the underlying assumption for many trials has been that investigated, it is more likely that differences in outcome
autoregulation would be impaired and CBF would would be observed with larger BP reductions, as long as
then need to be maintained within safe limits by con- the mean BP remains above the lower limit of autoregu-
trol of BP. lation. Following this, positive effects of larger BP reduc-
Overall, different strategies for lowering BP, involving tions may have been obscured in these trials by patients
different BP target levels, timing of intervention, or types in whom BP was lowered too much, i.e., below the lower
of vasodepressor drugs, have shown significant reduc- limit of autoregulation. Knowledge of a patient’s pre-
tions in systolic and diastolic BP, in comparison with stroke habitual BP is essential to define optimal BP

1524 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

targets poststroke, but this information is often not strokes followed by a stepwise deterioration in cognitive
known or not taken into account. The most critical as- function. On examination, these patients have signs of
pect of the trials under discussion is the timing of inter- stroke (e.g., hemiparesis) and have characteristic neuro-
ventions, especially in relation to our current knowledge psychological deficits, specifically in executive function
of the temporal evolution of autoregulation poststroke. and processing speed. Their memory deficits benefit
Autoregulation tends to be impaired at 2 wk after from cues, and typically they function well in a structured
stroke onset, followed by recovery by 3 mo. Given this environment. In contrast, the common but more atypical
time course, it could be expected that maximum benefit presentation of vascular dementia is that of a gradual
would be obtained by achieving BP targets in the most cognitive decline (more similar to Alzheimer’s disease)
critical period, that is between 1 and 3 wk after onset. combined with the identification on MRI of significant
This was the case with the CHHIPS trial (509), but in vascular brain injury (e.g., diffuse white matter lesions)
most RCTs intervention took place <36 h after onset (374, 376, 518, 523, 526). Often, this vascular brain injury
(507, 508, 511, 512), and it was not demonstrated that has been clinically silent in the sense that there is no his-
reduced BP levels were sustained for more than a few tory of transient ischemia attacks or stroke. Clinically,
days following treatment. the vascular lesions may not manifest as hemiparesis
This mismatch between BP control and the time-de- but as a more subtle disorder in balance, reduced motor
pendent deterioration of autoregulation poststroke is coordination, vascular Parkinsonism, or loss of urinary
likely to have contributed to the absence of significant control (518). A complicating factor is that there is con-
impact of BP-lowering therapy on stroke outcomes (517). siderable overlap in cognitive profiles with which
An ongoing RCT (CAARBS) will perform multiple obser- patients with vascular dementia or Alzheimer’s disease
vations of BP, following administration of Ca21 channel may present. For example, vascular dementia patients
blockers or angiotensin-converting enzyme inhibitors, can have predominant memory dysfunction, which
which might allow a better assessment of outcome tak- is a characteristic of Alzheimer’s disease, whereas
ing into account the timing of BP reduction in compari- Alzheimer’s disease patients can have prominent execu-
son with the expected evolution of autoregulation tive dysfunction, which is characteristic of vascular de-
efficacy poststroke (510). mentia (527).
In summary, the main lesson learned from scrutiny of In line with coronary and peripheral arterial disease,
RCTs aiming to improve outcome by means of BP con- impaired endothelial function contributes to cerebrovas-
trol is that inclusion of autoregulation assessment pro- cular disease with its unique features (32, 46, 48, 50,
vides the “missing link” to allow objective interpretation 263, 265, 521, 523, 528). Not surprisingly, a large num-
of results that could lead to better informed, individual- ber of studies, independent of the technique used to
ized treatment of patients with ischemic or hemorrhagic examine CBF, have demonstrated that patients with vas-
stroke. cular dementia present with lower CBF compared with
cognitively healthy peers (529, 530). Two review papers
5.4. Autoregulation in Cognitive Impairment and have provided overviews of TCD studies in vascular de-
Dementias mentia (compared with Alzheimer’s disease and con-
trols) and found that resting CBF is reduced (both in
5.4.1. Vascular dementia. vascular dementia and Alzheimer’s disease) and that
pulsatility index (a marker of vascular compliance) is
Vascular dementia represents a heterogeneous group increased in both dementias but more so in vascular de-
of brain disorders in which cognitive impairment is attrib- mentia. CO2 reactivity is impaired in both dementias, but
utable to cerebrovascular abnormalities and is responsi- again, more so in vascular dementia (531, 532). In vascu-
ble for 20% of all cases of dementia (518). When mixed lar dementia, resting CBF seems especially depressed
dementia is taken into consideration (the combination of in areas of white matter lesions. Interestingly, cerebral
neurodegeneration and vascular disease), the contribu- hypoperfusion has also been described in normal
tion of vascular changes to all dementias is much more appearing white matter (533), suggesting that local
substantial (374, 519–523). Vascular dementia can pres- blood flow reductions precede and, accordingly, may
ent in several ways clinically (376, 518, 524–526). For contribute to damage of white matter and its connec-
brevity, we will divide them broadly into two forms: 1) a tome (http://www.humanconnectomeproject.org). To
typical, easily recognized presentation; and 2) a com- support this hypothesis, cerebral hypoperfusion and
mon, but atypical and less distinct presentation. In the impaired cerebrovascular reactivity to hypercapnia are
typical presentation and characteristic form, the patient associated with a larger volume of white matter lesions
will have a history of clinical stroke followed by the onset (534, 535), suggesting that reductions in CBF are pres-
of cognitive impairment, or a history of repeated clinical ent before the onset of dementias, supporting an

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1525


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

important role for cerebral hypoperfusion in the patho- disease and other dementias. Previous work from ani-
genesis of vascular (and potentially other) dementias. mal models found that mild-to-moderate cerebral hypo-
To date, there have been no studies on autoregula- perfusion impairs neuronal protein synthesis, while
tion in vascular dementia to our knowledge, only studies cerebral ischemia ultimately leads to an increase in local
on autoregulation in stroke patients and in patients with glutamate concentrations and contributes to the accu-
small vessel disease described in the sections on autor- mulation of neurotoxic molecules (e.g., amyloid-b) (545).
egulation in stroke and hypertension. Reported increases in amyloid-b following ischemia
appear to be short-lived however, and it remains
5.4.2. Alzheimer’s disease. debated whether this mechanism can explain the sus-
tained widespread accumulation of amyloid-b in
There is a vast body of literature, mainly preclinical, that Alzheimer’s disease (550). Nonetheless, these vascular-
describes impairment of cerebrovascular function in derived insults might contribute to neuronal degenera-
Alzheimer’s disease (44, 58, 323, 374, 536–545). tion in Alzheimer’s disease. A reduction in CBF in
Changes in both cerebrovascular function and structure Alzheimer’s disease compared with healthy controls has
appear to be involved in this form of dementia (58). This been observed in multiple studies using different imag-
literature started with the rare genetic forms of ing modalities: Xenon-133, SPECT, PET, TCD, extracra-
Alzheimer’s disease and covers animal models mimick- nial ultrasound, and arterial spin labeling MRI (551, 552).
ing genetic forms in humans, as well as clinical studies Importantly, reductions in CBF have repeatedly been
addressing genetic causes of human Alzheimer’s dis- shown in the preclinical stages of Alzheimer’s disease
ease. More recently, evidence of cerebrovascular (541, 548). The reduction in CBF shows a regional pat-
involvement has also been repeatedly demonstrated tern and has been proposed as a diagnostic biomar-
in the much more common late onset, sporadic form ker, comparable to FDG-PET, which is a labeled-
of Alzheimer’s disease (58, 539, 546). Because cerebro- glucose imaging modality that can identify regional
vascular disease is commonly present in late onset hypometabolism (551, 553). Such data raise the question
Alzheimer’s disease, it can be difficult to distinguish if lower levels of CBF in Alzheimer’s disease may reflect
whether cerebrovascular dysfunction is a consequence synaptic failure (359, 554), resulting in reduced metabo-
of Alzheimer’s pathology or vascular disease independ- lism and reduced demand for CBF. This synaptic failure
ent of Alzheimer’s pathology (547, 548). Advances in in then continues throughout the course of the disease
vivo imaging now allow better phenotyping of patients, and is associated with the cognitive decline in the later
which helps in differentiating vascular effects caused by stages (555). In this scenario, reduced CBF is not a
Alzheimer’s disease from vascular effects that result sim- causal factor in neurodegeneration but a consequence
ply from vascular comorbidities associated with aging of loss of synaptic function. Support for this notion
(549). Also, ongoing research on rare human genetic comes from recent studies that reported that cerebral
forms of Alzheimer’s disease may help to shed light on global hypoperfusion in the general population is rela-
this issue, because cerebrovascular dysfunction can be ted to accelerated cognitive decline (351, 352) and
studied in the early stages of Alzheimer’s disease in a increased risk for dementia (351). Specifically related to
population with, because of the young age of onset, lim- Alzheimer’s disease, cross-sectional studies found that
ited vascular comorbidities. lower CBF is present in those with worse cognition (556)
Before we discuss what is currently known about and in patients that are rapidly declining (557). In a pro-
autoregulation in Alzheimer’s disease, we will first briefly spective study, cerebral hypoperfusion, measured using
summarize the literature on global changes in CBF, CO2 MRI (arterial spin labeling), predicted progression from
reactivity, and NVC in Alzheimer’s disease. These mild cognitive impairment to Alzheimer’s disease (558).
aspects of CBF regulation have been studied much Moreover, reduced CBF was also associated with a
more extensively than autoregulation. However, these faster 2-yr cognitive decline in a cohort of 88 patients
concepts are sometimes confused, with the result that with dementia due to Alzheimer’s disease (559). After
some studies of NVC or CO2 reactivity have been adjustment for subject demographics (e.g., age, sex, and
reported to reflect changes in autoregulation, which is education) and brain structure (e.g., gray matter volume,
incorrect. atrophy, white matter lesions), whole brain and parietal
hypoperfusion is associated with accelerated cognitive
5.4.2.1. CEREBRAL BLOOD FLOW. CBF is essential to decline. In another study, these authors confirmed that
support changes in activity of neurons and other brain reduced regional CBF was associated with a decline in
cells. Disruption of CBF regulation, baseline, temporal, global cognitive function but also executive function
or regional homeostasis, may represent a major factor in (560). These studies may reflect a close matching
the development and progression of Alzheimer’s of CBF to synaptic function, making CBF an early

1526 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

biomarker of disease and a predictor of disease pro- commonly encountered reductions in BP in patients with
gression. Alzheimer’s disease are those that occur in the context of
Alternatively, reduced CBF may be caused by cere- hypertension, antihypertensive treatment, daily postural
brovascular dysfunction and contribute to loss of brain changes including orthostatic hypotension, and during
function and disease progression through a mismatch of perioperative care.
CBF to neuronal demand. Reduced levels of CBF, meas-
ured by MRI-arterial spin labeling, represented the ear- 5.4.2.3.1. Studies of autoregulation in
liest sign of late-onset Alzheimer’s disease, present Alzheimer’s disease. The oldest study of autoregu-
even before traditional biomarkers (e.g., cerebrospinal lation in dementia known to us was from 1971 using the
fluid b-amyloid, cerebral hypometabolism, or brain atro- methods proposed by Simard et al. (567). Global and re-
phy) (541). These findings and others (529, 548) leave gional CBF was measured using the intra-arterial
open the possibility that cerebral hypoperfusion has a Xenon-133. Baseline CBF was studied in 24 dementia
direct causal role in the development of Alzheimer’s patients (39–74 yr of age); 13 with early onset dementia
disease. (<65 yr), of whom 5 had biopsy-confirmed Alzheimer pa-
thology; and 9 patients with onset >65 yr, including four
5.4.2.2. CO2 REACTIVITY AND NVC. Impaired vascular Korsakoff and three vascular dementia patients. In 15
reactivity to CO2 may already be present at an early, pre- patients (6 young, 9 old), autoregulation was measured
clinical stage of Alzheimer’s disease. Indeed, individuals before and after pharmacologically induced BP
carrying the APOE4 gene (561), and those with early changes. It is uncertain if any of these 15 patients had
stage probable Alzheimer’s disease (562), exhibit Alzheimer’s disease. The average reduction in mean ar-
impaired cerebrovascular responses to hypercapnia terial BP was 28%, with an associated mean reduction in
measured with TCD, compared with cognitively normal CBF of 4%. In two cases (young onset dementia), large
controls. These findings were confirmed in a study in reductions in BP were obtained (69 and 44%). These
younger, cognitively healthy APOE4 carriers that sho- changes were associated with reductions in CBF of 25
wed reduced responses to hypercapnia using blood O2 and 19%, respectively. Increases in BP (in 2 cases) had
level-dependent (BOLD)-functional (f)MRI (563). In addi- no effect on CBF. No control group was included,
tion to changes in cerebrovascular reactivity, other stud- although the authors could draw from their numerous
ies suggest impaired NVC in Alzheimer’s disease, which previous experiments using this technique for compari-
may also already be present in the preclinical stage (44, son. Despite the obvious limitations related to the heter-
45, 548). With the use of BOLD-fMRI, diminished local ogeneity of the sample (in both age and diagnosis), the
CBF responses in brain regions engaged during cogni- authors found no evidence for significant impairment of
tively demanding tasks in individuals with genetic pre- autoregulation in any of the 15 patients with dementia,
disposition for Alzheimer’s disease or in early stage regardless of age or underlying diagnosis (567).
Alzheimer’s disease have been observed (564). The first study of autoregulation in patients with a cer-
Moreover, fMRI studies indicate disrupted resting-state tain clinical diagnosis of Alzheimer’s disease was pub-
neural connectivity in brain areas susceptible to atrophy lished in 2009 (568). Nine patients with Alzheimer’s
in Alzheimer’s disease (e.g., hippocampus, medial pre- disease (3 with mild cognitive impairment, CDR 0.5, 6
frontal, and cingulate cortex) (564). with mild dementia, CDR 1, mean age 68 yr) and eight
age- and sex-matched controls were studied. The diag-
5.4.2.3. AUTOREGULATION. Evidence pointing toward nosis was confirmed by neuropsychological evaluation,
progressive impairment in cerebrovascular function and MRI, and CSF biomarkers. Dynamic autoregulation was
structure in Alzheimer’s disease led to the hypothesis evaluated using both spontaneous changes in BP and
that autoregulation is impaired in Alzheimer’s disease changes induced by repeated squatting and standing
(565). Clinically, the most relevant consequence of impair- maneuvers (118). These maneuvers induce large (20–
ment in autoregulation in patients with Alzheimer’s dis- 30%) changes in BP and mimic the effects of daily life
ease would be their increased vulnerability to cerebral postural changes on BP and CBF (118). CBF velocity, and
hypoperfusion following reductions in BP. This increa- its changes, were measured using TCD. Alzheimer’s dis-
sed vulnerability could be a double-edged sword. With ease patients had lower CBF and higher cerebrovascu-
impaired autoregulation for a given reduction in BP, the lar resistance, while there were no significant
reduction in CBF would be greater in patients with differences in total brain volume between patients and
Alzheimer’s disease. Added to this, effects of Alzheimer’s controls. Despite these cerebrovascular differences,
pathology and hypoperfusion on neurons, glia, and other dynamic autoregulation was equally effective in counter-
cell types could be greater than in healthy brain (58, 518, acting BP changes in Alzheimer’s patients and controls
548, 566). From a clinical perspective, the most (118).

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1527


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

Preserved autoregulation was confirmed in a study in Thus, after combinination of these smaller studies, 58
20 Alzheimer’s disease patients (no controls), 74.6 yr of patients with Alzheimer’s disease (38 dementia, 20 mild
age, 16 with CDR 0.5, 4 with CDR 1, and 17 with a positive cognitive impairment) were studied with TCD, indicating
amyloid-PET (569). This study evaluated static autoregu- normal dynamic autoregulation, except for 1 study with
lation, where CBF was measured using O15-PET before partially conflicting results (562). In addition, 20 patients
and after BP lowering induced by intravenous nicardi- with mild cognitive impairment were studied using PET
pine. CBF was measured after a reduction in BP that and had normal static autoregulation.
was maintained for 7–10 min. While mean BP was Recently, the largest studies on autoregulation in
reduced on average from 109 to 92 mmHg, global CBF Alzheimer’s disease to date were published (99, 572)
was unaffected (44 vs. 43 mL/100 g/min). Moreover, wherein both dynamic and static autoregulation were
there were no regional reductions in CBF, including investigated. In 53 patients with Alzheimer’s dementia
areas with more white matter lesions, in watershed (73 yr of age), 37 patients with mild cognitive impairment
regions, and in regions with the highest b-amyloid accu- (suspected early stage Alzheimer’s disease, 69 yr of age)
mulation (569). and 47 controls (69 yr of age), dynamic autoregulation,
A study of dynamic autoregulation used TCD and measured with TCD, was evaluated during spontaneous
evaluated 10 Alzheimer’s disease patients (73 yr of age) changes in BP and during larger induced changes in BP
and 17 controls during simple, every day postural chal- using repeated sit-to-stand maneuvers (99). These
lenges: standing up from sitting (100, 570). Standing up induced changes led to increases and decreases in
from sitting causes transient (<30 s) reductions in BP, mean BP of on average 25%. This large study confirmed
between 15 and 20 mmHg in mean arterial BP. When that dynamic autoregulation was similar in Alzheimer’s
performed as repeated maneuvers, similar to repeated disease patients (mild cognitive impairment and demen-
squat-stand maneuvers, they induce strong oscillations tia) and controls. This conclusion was despite clear evi-
in BP and CBF, with the benefit that sit-stand maneuvers dence for other cerebrovascular changes in Alzheimer’s
are easier to perform in older patients than squat-stand disease (reduced CBF, higher cerebrovascular resist-
maneuvers (100). CBF was lower and cerebrovascular ance, and reduced CO2 reactivity).
resistance was higher in Alzheimer’s disease patients Next, from the 53 patients with Alzheimer’s dementia,
compared with controls. Despite these baseline differen- 44 patients also underwent CBF measurements using
ces, the CBF response to the orthostatic challenges was MRI-arterial spin labeling with a repeated measurement
similar in patients and controls (570, 571). Of note, corti- after six months of BP lowering with nilvadipine versus
cal oxygenation [measured using near-infrared spectros- placebo (572). In 22 patients on nilvadipine, systolic BP
copy (NIRS)] decreased more in Alzheimer’s disease was reduced by an average of 11 mmHg, while global
patients, despite similar changes in CBF, which may and regional CBF were not reduced (572). There was a
point toward reduced O2 supply or increased O2 extrac- nonsignificant trend toward a 10% increase in global
tion in the microcirculation (571). However, the NIRS CBF following the BP reduction, which is of interest
technique requires further validation. because two other studies of antihypertensive treatment
Other authors confirmed normal dynamic autoregu- in patients of similar age, but without Alzheimer’s dis-
lation in 17 patients with Alzheimer’s dementia, 19 with ease, found (in larger sample sizes) a significant increase
mild cognitive impairment and 20 controls (395). in CBF of 10% following BP-lowering treatment (see sect.
However, the first potentially discrepant observation 5.2.2).
was made in a study in 12 patients (of whom 10 were 5.4.2.3.2. Autoregulation: indirect evidence
also included in Refs. 570, 571). Even though a normal of normal autoregulation in Alzheimer’s dis-
CBF response to a BP reduction following standing ease.There have been several large prospective stud-
was confirmed, the transient increase in BP (20% on ies that have measured outcomes that offer indirect
average) that followed the return to sitting after stand- evidence suggesting autoregulation is normal, similar to
ing was associated with a greater increase in CBF in what was discussed in sect. 4.2 in relation to hypertension
Alzheimer’s disease patients versus controls (24 vs. and autoregulation. These are outcomes that capture the
13%). Such a difference was also observed during expected consequences of impaired autoregulation, such
repeated sit- stand maneuvers, which led to repeated as cognitive decline, cerebrovascular events, and falls or
20% increases and decreases in BP from baseline. In syncope. Regarding falls and particularly regarding syn-
Alzheimer patients, these BP changes led to slightly cope, we repeat the caveat that these often represent a
larger changes in CBF than in controls (27% vs. 22%). failure of BP regulation (88) and not necessarily a failure of
However, assessment of dynamic autoregulation autoregulation (sect. 6.1).
using TFA again revealed no differences between Recent meta-analyses of antihypertensive treat-
patients and controls (562). ment show that BP lowering is associated with a lower

1528 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

risk to develop dementia’s, including Alzheimer’s dis- cognitive decline. These and other limitations of preclini-
ease (409). In addition, the SPRINT-MIND study found a cal models for Alzheimer’s disease have been discussed
lower risk of mild cognitive impairment and possibly de- elsewhere (574) and likely contribute to the discrepan-
mentia when intensive BP-lowering treatment (to a sys- cies described to date between studies performed in
tolic BP of 120 mmHg or below) was compared with animals and humans. This also highlights the difficulty of
standard treatment (405). Given the relatively short fol- translating observations from animals to clinical popula-
low-up times in these studies, and the knowledge that tions in humans.
Alzheimer’s disease is a slowly progressive disease with
5.4.2.3.4. Summary and clinical interpreta-
an asymptomatic, clinically unrecognized phase that can
tion.There is consistent evidence for cerebrovascular
last 10–20 yr, we can assume that many of the partici-
changes in Alzheimer’s disease, which can be summar-
pants with an outcome of mild cognitive impairment or
ized as an increase in vascular resistance, an early reduc-
dementia due to Alzheimer’s disease already had
tion in global (and regional) CBF, and impaired NVC and
asymptomatic Alzheimer’s disease at the time of inclu-
CO2 reactivity. These changes appear to be somewhat
sion. If Alzheimer’s disease were associated with
selective, because there is now substantial evidence that
impaired autoregulation already in an early stage of the
autoregulation is preserved in patients with Alzheimer’s
disease, as suggested by animal models (573), impair-
disease. The available studies were performed under con-
ment in autoregulation would be predicted to be present
ditions wherein BP was increased or decreased by 20–
at study inclusion in these participants. If so, exposure to
25% or less. This was done through BP-lowering medica-
BP-lowering treatment would have increased the risk for
tion or by postural challenges to decrease or increase BP.
cerebral hypoperfusion and therefore increased the risk
Thus we can translate these studies to clinical situations
to develop progressive cognitive decline and reach the
such as treatment of mild to moderate hypertension, or to
end-point of mild cognitive impairment or dementia. In
orthostatic hypotension, but not to more extreme condi-
reality however, the reverse was found, with a reduced
tions such as malignant hypertension (hypertensive crisis),
risk (hazard ratio 0.84) to develop dementia (405).
or septic or hypovolemic shock. The clinical implication is
Moreover, in the subgroup of older (>80 yr old) patients
that patients with Alzheimer’s disease have no increased
with low cognitive scores on a validated screening test
vulnerability to BP-lowering treatment and therefore do
(Montreal Cognitive Assessment), indicating a subgroup
not require higher levels of BP to maintain normal CBF.
that will include many patients with mild cognitive
For example, when a hypertension guideline advises a BP
impairment or even unrecognized dementia, there was
target of <140 mmHg systolic and <90 mmHg diastolic for
no increased risk of cognitive deterioration following in-
a hypertensive patient, there is no evidence that if this
tensive BP reduction (406).
patient also has Alzheimer’s disease, these targets should
5.4.2.3.3 Contrast with animal models. These be raised to higher levels of BP to preserve CBF. There
findings of preserved autoregulation in patients with late may of course be other arguments not to follow such
onset Alzheimer’s disease are in contrast with observa- guidelines, for example, time to benefit, patient prefer-
tions from an animal model (573). Indications that autore- ence, severe frailty, and so forth. However, available data
gulation was impaired in Alzheimer’s disease came from indicated that fear of causing cerebral hypoperfusion
other studies performed in preclinical models [reviewed should no longer be an argument. Of note, but not
elsewhere (565)]. For example, impaired static autoregu- reviewed here (as it is beyond the current scope), there is
lation was observed at a young age in transgenic mice also no evidence that Alzheimer’s disease patients have
that overexpress amyloid precursor protein, even before increased risk of orthostatic hypotension related to antihy-
the occurrence of parenchymal amyloid-b deposition pertensive treatment (402, 570, 575, 576).
(573). Impaired autoregulation in these animals was The impairment in NVC or CO2 reactivity that is
attributed to vascular dysfunction due to effects of associated with Alzheimer’s disease is not directly
b-amyloid, impaired contraction of vascular muscle (with related to autoregulation. Higher levels of BP may not
increased arterial BP), and altered vascular architecture translate into better NVC, nor will lower levels (within the
(564, 565, 573). ranges described) further impair NVC. In contrast, long-
There are several potential explanations for the dis- standing untreated hypertension can promote cerebro-
crepancy between the animal model results and human vascular disease and thereby further impairment in NVC
observations. The animal model for Alzheimer’s disease or CO2 reactivity. Some studies even suggest that it is
is not a model for late onset Alzheimer’s disease, but not BP per se, but factors associated with hypertension,
rather for the rare young onset genetic form. Even there, that cause impairment in NVC. For example, elevated
this represents only a partial model, as they do not levels of tissue or circulating angiotensin II (165, 528)
involve tau pathology. In addition, animal models some- may directly affect endothelial function, which could
times do not develop significant neurodegeneration or then contribute to impairment in NVC, independent of

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1529


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

changes in BP. The clinical relevance of this discussion 5.6. Direct Control of the Brain on Blood
is that hypertension and Alzheimer’s disease often coex- Pressure?
ist. Around 40% of Alzheimer’s disease patients also
have hypertension (577). Vice versa, the prevalence of Recently, the idea has resurfaced that the brain may
hypertension increases to 60% with aging. Because directly control BP to preserve its function. This was the
aging is the strongest risk factor for Alzheimer’s disease idea formulated by Hill (4) and Bayliss et al. (5) around
(578), many older hypertensive patients may develop 1890, who were convinced that there was no active
Alzheimer’s disease. autoregulation to maintain CBF, and therefore (para-
phrased), the rest of the body functioned as a slave to
5.5. Autoregulation in Neurocritical Care the brain, increasing BP if the brain needed more CBF.
Indeed, in more recent times, common examples such
We have explained the strong capacity of the cerebro- as the increase in BP following stroke has been inter-
vasculature and dynamic autoregulation to protect preted as direct control of the brain on BP in response
against significant increases in CBF (hyperperfusion) to the failing CBF. In a similar vein, the higher BP that is
during acute increases in BP or hypertensive emergen- associated with aging has been considered to be
cies. Clinical conditions that can cause such increases in caused by the brain in response to the waning CBF with
BP include drug abuse (i.e., cocaine, amphetamines), aging (588). This idea was recently reframed (120 yr after
anxiety or panic disorders, stroke, traumatic brain injury, Hill) as the “selfish brain” hypothesis (note that the “self-
aortic dissection, and hypertensive disorders of preg- ish brain” term has also been used for very different
nancy (130). End-organ damage to brain (and the retina) hypotheses for mood disorders, addiction, etc.) (588).
can result if autoregulatory capacity is exceeded, result- A recent study provided evidence for a direct effect
ing in marked increases in CBF, endothelial dysfunction, of the brain on BP, following a change in ICP and cere-
increased microvascular pressure, loss of blood-brain bral hemodynamics (178). This study identified astro-
barrier integrity, and edema (130). cytes that lie adjacent to brain autonomic centers as
Neurointensive care is a field where protection of potential modifiers of BP to maintain brain function.
the brain against effects of either reduced or increased There are however several methodological caveats to
perfusion pressure is critical (130, 500, 579–581). In this study. First, the results are presented as if it is a
the majority of this review, we have discussed autore- reduction in CBF that triggered the astrocytes’ response
gulation and diseases related to autoregulation, gen- (178). However, the reduction in CBF was brought about
erally in the context of conditions where ICP is normal by an increase in ICP, raising the question of whether
and relatively stable. In neurocritical care, this situation the astrocytic response was due to increased ICP or to
can be very different. For example, in ICH or in trau- reduced CBF (178). Raising ICP to lower CBF was per-
matic brain injury, brain damage can result from ele- haps the only reasonable design, because other options
vated ICP. Under these conditions, monitoring and to reduce CBF (i.e., lowering BP) would have triggered
treatment are therefore not only focused on maintain- baroreflex responses (88). Activation of the classic bar-
ing adequate CBF to supply O2 and glucose but also oreflex response would have confounded the results, as
on adapting BP to changes in ICP. The importance of the authors wished to test if astrocytes functioned as a
ICP monitoring and the availability of long-term meas- “brain baroreflex.” The increase in ICP (and subsequent
urements of ICP in the neurointensive care unit has led reduction in CBF) led to an increase in BP with a delay of
to monitoring of autoregulation based on the relation- 30 s, persisting for 30 min: the time scale of these
ship between BP and ICP, as well as the relationship changes clearly differs from the faster changes associ-
between BP and CBF (500, 579–581). This field of clini- ated with the classic baroreflex. Interpretation of this
cal research is unique in that it has provided long-term work is further complicated by the fact that the some-
(hours) monitoring, where changes in autoregulation what moderate increased in ICP (10–15 mmHg) was
over time can be linked to clinical prognosis (500, associated with a 40% reduction in CBF, suggesting
581–587). In this setting, additional factors come into autoregulation in this model was rather ineffective. In
play in relation to autoregulation (e.g., brain swelling contrast, multiple other studies have reported that CBF
and edema, CSF circulation, vascular or neuroinflam- is maintained quite effectively during increases in ICP of
mation, or changes in the meninges or choroid plexus: this magnitude or more. These issues aside, this study
key sites of immune cell trafficking), making BP and of astrocytes is of potential interest, perhaps applicable
ICP regulation, versus CBF regulation, topics that to the neurointensive care unit setting, where elevated
would require a review of its own. Fortunately, several ICP is frequently observed (see sect. 5.5). Second, with
excellent recent reviews on this topic are available regard to the discussion above, most other instances
(493, 500, 581). where CBF is reduced (e.g., following BP lowering,

1530 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

antihypertensive treatment, postural changes, hypovole- impaired autoregulation above 1.5 MCA. For sevoflurane,
mic, or septic shock), ICP decreases in parallel with CBF autoregulation is already impaired at a dose above 1
(344, 589). Therefore, it remains uncertain if a reduction MCA (593, 596).
in CBF, without an increase in ICP, would trigger a Effects of fluranes or anesthetics are modified by CO2,
“brain-initiated” increase in BP, mediated through the where hypercapnia lowers the threshold for impairment
autonomic nervous system. in autoregulation, and hypocapnia increases it. These
anesthetics all lower brain metabolism (CMRO2) and
5.7. Anesthesia and Autoregulation because they also induce vasodilation cause uncoupling
between metabolism and CBF (i.e., CBF is higher than
From a clinical perspective, the topic of anesthesia and metabolism would demand) (593). Nitrous oxide, when
autoregulation involves not only the use of anesthetic used as a single agent (which is no longer in practice),
drugs but also control of respiration and respiratory increases CBF while CMRO2 remains stable; however,
gases, BP, combined with the potential hemodynamic when used in combination with other anesthetics,
effects of surgery. The relevance of taking autoregula- results are inconclusive. There is no information on its
tion into account to prevent cerebral ischemia during effects on autoregulation (593, 596). Propofol reduces
surgery has recently been reviewed (590) and is also brain metabolism, which in turn leads to a reduction in
explained in our call-out text box for clinicians. A discus- CBF. Direct in vitro vascular application of propofol
sion of complex cardiovascular or cardiothoracic sur- causes vasodilation. Autoregulation remains intact
gery, with extracorporeal circulation and anterograde unless doses of propofol exceed 200 mgram/kg/min
selective cerebral perfusion (591), is beyond the scope (593, 596). Ketamine increases CBF through direct vaso-
of this review. This also applies to the use of anesthesia dilation, but there is no evidence for effects on autoregu-
in traumatic brain injury, where a recent review identified lation (593, 596). Etomidate reduces CMRO2 and
important knowledge gaps (592). For the effects of thereby CBF. This agent has a potential direct vasocon-
changes in concentration of respiratory gases (O2 and strictor effect, but there are no known effects on autore-
CO2) on CBF, we refer to sects. 1.3 and 3.2 of this review. gulation. Unique among anesthetics, etomidate does
Here we briefly discuss the effects of anesthetic agents not lower BP (593). Dexmedetomidine reduces CBF,
on CBF and autoregulation and refer to a recent review potentially by activation of postsynaptic a2-adrenergic
for further reading (593). receptors that induce vasoconstriction, and in addition
Anesthetic agents affect brain metabolism (593, 594). reduce CMRO2 (525). Benzodiazepines (e.g., midazo-
For most agents, the suppression of consciousness lam), nonbenzodiazepine agents that target GABA (e.g.,
reduces metabolism and hence demand for O2 con- zolpidem), and opioids (e.g., fentanyl) reduce CRMO2
sumption. If, as under normal conditions, the tight cou- and thereby CBF but do not affect autoregulation (593,
pling between metabolism and CBF is maintained (sect. 596).
1.2), this reduction in the cerebral metabolic rate of O2 Because of their effects on cerebral metabolism and
(CMRO2) will equally reduce CBF. However, some anes- on cerebrovascular tone, anesthetic agents can also
thetic agents cause an uncoupling of metabolism and affect NVC. From a research perspective, this has impli-
blood flow (593, 594). In some cases, there may also be cations for the interpretation of studies on NVC in pre-
direct effects of anesthetics on the cerebral vasculature, clinical models, which are most commonly performed
changing vascular tone and thus cerebrovascular resist- under anesthesia (594). Anesthesia may also affect NVC
ance. The net outcome of such changes on CBF during in humans (597, 598). New approaches that allow stud-
anesthesia depends on whether metabolism and blood ies in awake animals have been proposed to address
flow remain coupled, and on interaction with changes in this potential limitation (594).
respiratory gases. Although anesthetics affect CBF,
most agents do not impair autoregulation, with the
exception of volatile anesthetics (the fluranes) (593). The 6. IMPACT OF DAILY LIFE CHALLENGES ON
effects of commonly used anesthetic agents on CBF, CEREBRAL BLOOD FLOW
metabolism (CMRO2), and autoregulation are briefly
summarized below. 6.1. Postural Challenges
Isoflurane, desflurane, and sevoflurane (volatile agents)
have a direct vasodilatory effect on cerebral blood ves- 6.1.1. Orthostatic hypotension.
sels, and this effect is independent of the dose or depth
of anesthesia (595). The effects on autoregulation how- Whenever we stand up from a lying or sitting position,
ever are dose dependent, with normal autoregulation at the combination of our upright posture and Earth’s grav-
lower dose for isoflurane or desflurane (1 MCA), but ity causes large shifts in circulatory volume (300–500

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1531


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

mL) from the central body and head to the lower abdo- (pre)syncope earlier than someone with normal autoregu-
men and legs (599). These fluid shifts lead to reductions lation. However, the influence of BP regulation in syncope
in venous return to the heart, reducing cardiac output outweighs that of autoregulation (603).
and BP. Actively standing up also causes reflex arterial Syncope is caused by a failure to maintain BP at a
vasodilation (to support muscle perfusion) that contrib- level that is required to maintain sufficient CBF (604,
utes to the reduction in BP. Maximum decreases in BP af- 605). Autoregulation operates within two boundaries:
ter standing are reached within 15 s and can easily be as the magnitude (static) and the speed (dynamic) of BP
large as 15–25% (599). The baroreflex usually restores changes. The magnitude refers to the limits of static
BP within 30–40 s (88). Failure to normalize BP after 60 s autoregulation. When BP falls below the lower limit of
is termed orthostatic hypotension, defined as a sustained autoregulation (e.g., below a systolic BP of 70–
reduction in systolic BP of 20 mmHg or more. 80 mmHg), CBF will fall progressively with further reduc-
Even without orthostatic hypotension, normal BP tions in BP. The speed refers to limitations in response
responses pose daily challenges for autoregulation. Their time of dynamic autoregulation. Slow changes in BP are
speed of onset limits the potential for autoregulation to much more efficiently buffered than fast changes, and
buffer these BP changes. This explains the common very fast changes (e.g., those occurring within 1–5 s) can-
subtle symptoms we all may experience upon standing not be buffered at all.
up. In orthostatic hypotension however, sustained reduc- BP is determined by cardiac output and total periph-
tions in systolic BP can be as large as 60 mmHg and eral resistance (170, 604, 606–608). Cardiac output is in
exceed the lower limits of autoregulation, causing cere- turn determined by stroke volume and heart rate,
bral hypoperfusion, dizziness, falls, or syncope. whereas peripheral vascular resistance is determined by
The longer we lie or sit, the stronger is this transient vascular structure, vascular mechanics, and vascular
decrease in BP. Other postural challenges may also tone of the systemic circulation (170, 604, 606–608).
cause large reductions in BP. For example, getting up af- The baroreflex system plays an important role in short-
ter squatting leads to strong reductions in BP (118). term BP regulation, as it controls heart rate and periph-
Combining standing with a Valsalva maneuver (e.g., eral vascular resistance through the parasympathetic
singing, blowing a horn) may also elicit a strong fall in (mainly vagus nerve) and sympathetic arms of the auto-
BP. Bending forward may cause abdominal vascular nomic nervous system (88).
compression, more so in pregnant women or in obesity, Even if baroreflex function is normal, syncope can
and provoke hypotension (599). occur when certain challenges exceed baroreflex
These challenges occur on a daily basis and are an capacity (27, 88, 609, 610). Examples are severe hypo-
important reminder that BP is not a stable entity per se. volemia (bleeding, excessive fluid loss due to diarrhea,
Steady-state BP, measured sitting after 5 min of rest, vomiting, exercise, or hot environments) or large volume
should indeed be more or less constant in a healthy per- shifts (when standing up after prolonged squatting, e.g.,
son if repeated with an interval of days or weeks. during gardening). This leads to a fast and large reduc-
However, when measured over shorter time intervals tion in BP, that due to its speed of onset cannot be buf-
throughout a day, BP constantly varies with postural fered by autoregulation and leads to hypoperfusion.
changes. The strong impact of postural changes on both Many cases of syncope occur during temporary barore-
BP and CBF can easily be learned from studies using flex dysfunction (88). Called neurally mediated reflex, or
repeated sit-stand or squat-stand maneuvers (118, 570) vasovagal syncope (88), such disorders can cause the
(FIGURE 4). baroreflex to initiate bradycardia and peripheral vasodi-
lation, leading to prolonged reductions in BP below the
6.1.2. Syncope. lower limit of autoregulation, and loss of consciousness.
These circumstances include pain, fear, strong emotion,
Syncope (or fainting) is defined as a brief loss of con- sustained orthostatic stress (standing still for long peri-
sciousness caused by a transient reduction in CBF (600– ods), among others. Baroreflex failure associated with
602). This definition clearly sets syncope apart from the disease is seen in autonomic failure (e.g., diabetes,
two other most common causes of transient loss of con- Parkinson’s disease, primary autonomic failure, or spinal
sciousness: seizures (epilepsy) and psychogenic syn- cord lesions (88). Finally, cardiac disorders (structural or
cope. The definition of syncope may have contributed to rhythm) can cause syncope through their effects on BP.
a common misconception that syncope is caused by a
disorder in autoregulation. Syncope is however primarily 6.2. Physical Activity
caused by a failure to maintain stable and adequate BP
(600–602). Theoretically, for the same reduction in BP, a The spectrum of physical activity encompasses the
person with impaired autoregulation could experience areas of low-, moderate-, and vigorous-intensity physical

1532 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

activity but also the lower end of the spectrum including frequency range (618). The subsequent study that
sedentary behavior (e.g., sitting and lying). The interest adopted a 6 h uninterrupted sitting protocol found an
in understanding the impact of exercise-like physical ac- increase in normalized gain in the very-low-fre-
tivity on health (611), including the regulation of CBF (612) quency range (618). The differential impairment in au-
and its impact on brain health, dates back to the middle toregulatory parameters between both studies may
of the previous century. During the most recent decade, relate to a time-dependent, progressive impact of sit-
studies have started to acknowledge the importance of ting on autoregulation. It is hypothesized that impair-
examining the impact of lifestyle or behavior, driven by ment in autoregulation may first affect phase (i.e., the
the observation that increasing sedentary behavior, in- latency of response), before affecting gain (i.e., the
dependent of exercise performance, is related to all- efficiency of the response) (624). However, a series
cause mortality and morbidity (613, 614). More recent of studies from our laboratory in an older population
work also suggested the potential of sedentary behavior with or without increased cardiometabolic risk found
to affect cardiovascular and cerebrovascular health and no impact of prolonged, uninterrupted sitting on
thus brain health (615–617). autoregulation, despite a significant decrease in CBF
(621, 625). Therefore, it is unlikely that a change
6.2.1. Sedentary behavior. in autoregulation is responsible for the reductions in
CBF during prolonged sitting that were found in
In studies designed to better understand the immediate some studies. One should also consider the potential
impact of prolonged sedentary behavior in healthy indi- impact of sitting on alternative interacting pathways
viduals, Carter et al. (618) examined the impact of 4- and or mechanisms, including baroreflex sensitivity, cere-
6-h periods of prolonged, uninterrupted sitting on CBF. bral metabolism, endothelial function, or NVC.
With the use of TCD, prolonged, uninterrupted sitting Population (626)- and laboratory-based studies (627)
led to a small (1.4–3.4 cm/s, equivalent to 2–5%) but sig- aimed at better understanding the impact of prolonged
nificant decline in middle cerebral artery CBF velocity in sitting on health outcomes highlight the relative impor-
healthy volunteers. Because there was a small tance of breaking up sedentary behavior, independent
(0.9 mmHg) reduction in end-tidal CO2, which may be of the duration and intensity of this physical activity
the result of changes in pulmonary ventilation during break. For this purpose, studies have interrupted pro-
prolonged sitting, it remains uncertain if a change in longed sitting and found that breaking up sitting pre-
PaCO2 could have contributed to the reduction in CBF vents the decline in CBF in healthy volunteers (618).
(618). In this range, even small changes in PaCO2 (e.g., Interestingly, this is not a universal finding across all pop-
1 mmHg) can cause significant changes in CBF (3–8%) ulations, since a recent study found that effects of 3 h of
(223, 619). The effect of sitting on CBF has also been sitting on CBF were not prevented by physical activity
explored in groups at increased risk for cerebrovascular breaks in subjects with prediabetes (625) or older indi-
disease. A study in 12 older adults (70 yr of age) found a viduals (621). Possibly, in subjects with a priori increased
small reduction in CBF (620); however, PaCO2 or end- risk for cerebrovascular disease, low-intensity physical
tidal CO2 was not measured in that study. In 22 older activity breaks are insufficient. Some support for this hy-
adults (78 yr of age, 9 women), there was no reduction in pothesis is provided by a recent study that found that 30
CBF after 3 h of sitting (621). CO2 was measured in that min of moderate-intensity walking exercise prevented
study and remained stable. Of note, there was an the decline in CBF associated with prolonged sitting
increase in BP with sitting. In 25 middle-aged workers (620). However, this study did not measure and control
with prehypertension (622), sitting, but not sitting alter- for changes in PaCO2 .
nated with periods of standing, was associated with a Taken together, recent studies suggest that pro-
decline in CBF over the day (without measurements of longed sitting is associated with a stimulus-dependent
PaCO2 or end-tidal CO2). Finally, a recent cross-sectional decrease in CBF but with no consistent effect on
study found in 52 healthy older adults that greater sed- dynamic autoregulation. The exact mechanisms underly-
entary time was significantly associated with lower CBF ing these changes in CBF are unclear but appear to
in lateral and medial frontal regions (623). result from the prolonged period of physical inactivity,
Some of these studies have also investigated if since regularly interrupting sitting with short bouts of
sedentary behavior may affect autoregulation or CO2 low-intensity physical activity or moderate-intensity
reactivity. While cerebrovascular responses to hyper- walking exercise prevents these effects. However,
capnia were preserved, changes in autoregulation changes in PaCO2 due to changes in ventilation and
during sit-to-stand maneuvers were found in one lung perfusion are expected with prolonged sitting and
study (618). Prolonged uninterrupted sitting for 4 h can be altered by the bouts of physical activity, and
resulted in a decrease in phase in the very-low- these changes alone may explain the effects on CBF.

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1533


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

PaCO2 was not always measured and accounted for in elevates the CBF response, possibly as a compensatory
these studies. Further work is required to better under- response to a lower PaO2 and maintenance of O2 deliv-
stand the impact of sedentary behavior at different lev- ery. In contrast, lower CBF responses to exercise are
els of the cerebrovasculature, including the blood-brain observed during chronic hypoxia. At a minimum, these
barrier, or more chronic changes such as vascular studies highlight the importance of measuring both
remodeling or changes in vascular mechanics. Whether PaCO2 and PaO2 when attempting to understand the
the effects of sitting are distinct in populations with regulation of CBF during exercise.
impaired cerebrovascular health and hypoperfusion and During exercise, marked increases in BP (30% or
translate to detrimental long-term effects is unclear at more), cardiac output (300–600%), and neural activity
this time and requires further examination. may support appropriate regional distribution of blood
flow of the peripheral circulation and the brain. These
6.2.2. Exercise: role of intensity. factors all show a clear temporal linearity with increases
in activity level, suggesting that BP, cardiac output, or
Exercise can be regarded as an ultimate, integrative neural activity alone are unlikely to dictate the biphasic
stimulus for the brain to regulate CBF, especially since CBF response during exercise. Moreover, any increases
exercise is known to evoke large changes in key stimuli in BP should be buffered by autoregulation. Regarding
that affect CBF-O2 consumption, BP, cardiac output, cardiac output, studies have examined whether altered
blood gases (especially PaCO2 ), activity of sympathetic cardiac output during exercise, either within individuals
nerves, and brain activity. All these variables change [e.g., manipulation (632)] or between subjects [e.g., heart
with exercise, often with different magnitudes and pat- transplantation (633)], is followed by comparable
terns, constantly challenging the sufficient delivery of changes in CBF. Despite increases in cardiac output
CBF to meet metabolic demand (but also to prevent with exercise, CBF remains largely unaffected, suggest-
hyperperfusion) (347, 628). Changes in CBF in response ing a relatively modest role for cardiac output in contrib-
to incremental levels of exercise are typically described uting to CBF changes with exercise. In addition, studies
as biphasic. There is a progressive increase in CBF in aimed at better understanding neurogenic mechanisms
response to whole body exercise, up to an exercise in- (e.g., by blocking activity or effects of parasympathetic
tensity of 60% of maximal O2 consumption, followed nerves) provide somewhat conflicting results that may
by a plateau in CBF and a decrease toward resting lev- relate to methodological issues pertaining to these
els when reaching maximal intensity exercise (see experiments (e.g., effectiveness of blockade). There is
review in Ref. 347). Our understanding of regulatory currently limited evidence supporting a key role for neu-
mechanisms that contribute to the biphasic CBF rogenic mechanisms in mediating CBF changes during
response to incremental levels of exercise is limited by exercise (628).
the complexity and redundancy in processes that regu- A final factor for regulation of CBF relates to cerebral
late blood flow to the brain. Below, we have summarized metabolism. While various techniques, exercise proto-
what are thought to be the most important factors con- cols and methodological designs have been used, the
tributing to the biphasic CBF response. general consensus is that exercise leads to a gradual,
Early hypotheses assumed there was a key role for dose-dependent increase in cerebral O2 uptake, reflec-
exercise-induced changes in blood gases, especially tive of elevated cerebral metabolism (628). Accordingly,
PaCO2 , contributing to changes in CBF with exercise. increases in cerebral metabolism may contribute to the
This idea was supported by the sensitivity of the cere- increased CBF at low- to moderate-intensity exercise.
brovasculature to changes in PaCO2 but also the obser- However, the lower CBF at near maximal exercise was
vation that incremental levels of exercise first caused an not reflected by a decrease in cerebral metabolism
increase in PaCO2 , followed by a (hyperventilation- (634). Possibly, a compensatory increase in O2 extrac-
induced) decline in PaCO2 toward (near) maximal exer- tion and/or cerebral delivery of O2 may contribute to a
cise intensity. Indeed, CBF was increased with increas- further increase in cerebral metabolism with higher-in-
ing PaCO2 levels at baseline (629) or when mitigating tensity exercise. Alternatively, studies have suggested
the drop in PaCO2 during near maximal exercise (630). changes in substrate utilization may occur during exer-
During submaximal exercise, however, increases in CBF cise. More specifically, cerebral metabolism normally
seem less dependent on changes in PaCO2 , although (under resting conditions) is thought to mainly depend
this stimulus may still contribute to the regional distribu- on O2 and glucose. However, during exercise, there is
tion of CBF (631). In addition to PaCO2 , PaO2 may also an increased uptake of lactate, accompanied by a
play a role in regulating CBF during exercise, especially reduced uptake of O2 and glucose, which may suggest
when exercise is performed during acute or chronic hy- that the brain uses lactate as fuel, thus being less de-
poxia. For example, exercise during acute hypoxia pendent on the consumption of glucose and O2 (635).

1534 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

Since older age is an important risk factor for cerebro- exercise training on regional or global CBF. A recent
vascular pathology, several studies have explored the study in healthy older men examined the impact of 8 wk
impact of aging on CBF during exercise. Studies examin- of exercise training and examined global and regional
ing these responses have previously been reviewed in CBF using MRI (641). Interestingly, this study found
detail (347) and suggest the presence of a similar bipha- improved regional CBF in the frontal lobe, particularly
sic CBF response to incremental levels of exercise as the subcallosal and anterior cingulate gyrus, whereas a
observed in healthy young individuals. However, CBF is significant decrease was found in the temporal fusiform
markedly lower in older individuals compared with gyrus. These regional differences may explain the ab-
healthy young subjects, both at rest and during exercise sence of an effect on global CBF in this study (641).
(636–638). Despite these differences in absolute CBF Others also found regular exercise training to improve
between both groups, the magnitude of change in cere- local CBF (642), including elevated bilateral blood flow
bral metabolism, including the dependency on the vari- to the hippocampus (643). When exercise training is
ous metabolic substrates at different levels of exercise, causally linked to increases in local CBF, one may
are comparable between younger and older subjects. expect that cessation of exercise training would
Similarly, comparable responses are found between decrease CBF. Indeed, a study of 10-day cessation of
younger and older individuals in central hemodynamics exercise training in older master athletes found reduc-
(e.g., BP, cardiac output) and neural activity. One poten- tions in CBF in the hippocampus and gray matter (644).
tial factor, partly contributing to the age-related differen- These results suggest that exercise training increases
ces, may involve a lower PaCO2 in older individuals local CBF, specifically in regions that are linked to cogni-
(638). Other factors, such as brain atrophy or altered va- tive function.
sodilator mechanisms, may also contribute to these age- In line with studies performed in healthy individuals,
related differences in CBF during exercise. exercise training also seems to improve local CBF in
Taken together, the biphasic pattern of CBF during clinical populations. For example, exercise training for 6
exercise seems to relate to multiple physiological mech- mo in stroke survivors improved CBF to grey matter and
anisms that have a complex integration, synergism, and perfusion in the parietal lobe (645). However, increased
redundancy to prevent impairment of CBF during exer- local CBF is not a universal finding. For example, Alfini
cise. Based on current evidence, the balance between and coworkers (646) found increased CBF in the left
cerebral metabolism and PaCO2 seem to contribute insula in subjects with mild cognitive impairment, while
most to the biphasic pattern of CBF during incremental 12 wk of endurance exercise training decreased CBF in
levels of exercise, which can be present in healthy the left insula and improved cognitive function. In sub-
young as well as in older populations. Nonetheless, a jects with cognitive impairment, changes in local CBF
lower CBF in older individuals may be present at rest should be interpreted with caution, especially when
and during exercise, which in part may relate to a lower examining brain regions that may be related to impaired
PaCO2 . Better insight into these responses of acute exer- cognitive function. Previous work has linked decreased
cise may help to understand, but also to guide and opti- regional CBF to the pathophysiology of cognitive impair-
mize, the effectiveness of exercise training strategies to ment (351, 352). Future studies are warranted, preferably
improve cerebrovascular and brain health. adopting a longitudinal design and concomitant assess-
ment of cognitive function, to better understand the
6.2.3. Exercise training. impact of exercise training on regional CBF and the
impact of these regional changes in CBF for clinical
Several studies have examined the potential clinical populations.
impact of exercise training and/or physical activity in pri- Related to the central topic of this review, one may
mary and secondary prevention of cerebrovascular dis- question whether regular exercise training impacts
eases and have largely provided convincing evidence autoregulation. Aengevaeren et al. (363) was among the
for a role of regular exercise in the clinical management first to examine this question and compared autoregula-
of these diseases. Since this is beyond the scope of this tion between older master athletes and age-matched
review, we refer to excellent reviews in the literature sedentary controls. In this cross-sectional comparison,
that have covered this topic (639, 640). Here we discuss no differences in autoregulation were found between
the impact of regular exercise training on CBF and autor- groups (363). Subsequent cross-sectional studies also
egulation but also whether these effects depend on the found no significant differences in autoregulation
population examined. between highly trained versus sedentary individuals
In contrast to a relatively large number of studies (647–649). It is important to highlight that some recent
examining the acute effects of exercise on CBF, few studies suggest that changes in autoregulation may
have thoroughly examined the impact of regular be present after high-intensity exercise (650). Such

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1535


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

changes consisted of very small reductions in TFA flow. A second important characteristic is that the brain
phase at 0.1 Hz. Another study found no effects of high- is enclosed in the skull, which requires a much tighter
intensity training on autoregulation (651). While work on control of tissue pressure (ICP) compared with other
exercise and autoregulation, largely coming from cross- organs that have more room to expand. Autoregulation
sectional comparisons, suggests that regular exercise also plays an important role in the homeostasis of ICP,
training has little impact on autoregulation in healthy because an increase in cerebral blood volume will
individuals, one may question whether this conclu- increase ICP and vice versa.
vsion can be extrapolated to clinical populations. In one Autoregulation is the mechanism that safeguards
study, patients with chronic obstructive pulmonary dis- blood flow to the brain in the context of changes in BP,
ease exhibited preserved autoregulation compared with the most important determinant of CBF. In this review,
healthy controls, and 8 wk of exercise training did not al- we have discussed physiological and pathophysiologi-
ter autoregulation in either group (652). cal examples where changes in BP occur. These include
Taken together, exercise training seems to have no everyday changes in body position including effects of
major impact on autoregulation. However, regular exer- gravity on hemodynamics, physical activity, hyperten-
cise training is a potent stimulus for changes in global sion, and antihypertensive treatment. Autoregulation
and regional CBF, in both healthy individuals as well as operates by regulating cerebrovascular resistance to
in those with cerebrovascular disease. Specifically, regu- counteract changes in BP. In relation to acute adjust-
lar exercise training is associated with increased re- ments, these changes are thought to mainly occur due
gional CBF, such as in the hippocampus and gray matter to actions of pressure-sensitive vascular muscle that al-
(i.e., regions that match with cognitive abilities like mem- ter vascular resistance through vasodilation or vasocon-
ory and attention). Within this respect, previous studies striction (FIGURE 7).
have linked cellular biological factors, e.g., brain-derived The time frame over which changes in BP occur deter-
neurotrophic factor and myokines, to contribute to (hip- mines the effectiveness and stabilization of CBF by autor-
pocampal) neuroplasticity (653). Although aerobic exer- egulation. The effect of relatively slow changes in BP on
cise is known to have beneficial effects on the cerebral CBF is best described by the model of static autoregula-
vasculature, brain health, and cognitive function, mecha- tion, where it has been shown that CBF is held stable
nisms that underlie these changes are still relatively (within a margin of about 610%) across a wide range of
poorly understood (654, 655). Several mechanisms are BP levels (50–150 mmHg in MAP). The effects of faster
likely to be involved. Part of the benefit of exercise may changes in BP on CBF are captured in the model of
be hemodynamic, including effects of shear stress on dynamic autoregulation, where progressively faster
phosphorylation and activity of eNOS, reductions in oxi- changes in BP lead to larger changes in CBF. A conse-
dative stress, angiogenesis, and increased CBF (654, quence of dynamic autoregulation is that even with opti-
655). Another mechanism may relate to effects of neuro- mally functioning autoregulatory mechanisms, there can
trophic factors including brain-derived neurotrophic fac- still be variability in CBF caused by changes in BP.
tor (BDNF). Exercise increases expression of BDNF, In contrast to what is often stated, autoregulation is a
known to have positive effects on neuronal plasticity, robust mechanism that is not generally affected by aging
neurogenesis, synaptogenesis, hippocampal and cogni- or hypertension. Evolving research suggests it also
tive function (654, 655). Other data suggest that BDNF remains relatively intact in Alzheimer’s disease. However,
signaling may be dependent on activity of eNOS and autoregulation does not function in isolation. We have
production of NO by endothelial cells (654, 655). described that CBF is affected and controlled by multiple
Considering the importance of the topic for brain health, factors, such as blood gases, neurovascular innervation,
further research is warranted to better define pathways endothelial function, ICP, metabolism, and NVC.
explaining and facilitating optimization of exercise train- Conditions such as hypertension and Alzheimer’s disease
ing to improve brain health. can therefore affect CBF, even if they do not impair autor-
egulation. As an example, endothelial dysfunction associ-
ated with hypertension may lead to a chronic reduction in
7. SUMMARY AND CONCLUSION CBF, while autoregulation (which is independent from en-
dothelial function) is still intact, and therefore, when BP is
The brain is a richly perfused organ with a dense vascu- lowered by antihypertensive treatment, there is no risk of
lar supply, characterized by relatively high blood flow hypoperfusion, despite the lower baseline CBF. Thus a
and low vascular resistance. Its high metabolic demand, reduction in CBF or impairment in NVC, as has been
roughly one-fifth of the total body metabolism, in an observed in hypertension and in Alzheimer’s disease,
organ that represents <2% of total body weight, should not be interpreted as evidence of failed autoregu-
depends on autoregulation to maintain adequate blood lation and should not justify attempts to raise BP levels to

1536 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

restore CBF or NVC. Vice versa, a finding of normal autor- Utrechtsche Hoogeschool. Tweede Reeks 92-120: 1868–1869,
egulation in a clinical population (e.g., stroke, hyperten- 1868.

sion, Alzheimer’s disease) should not be interpreted as a 7. Mosso A. Ueber den Kreislauf des Blutes im Menschlichen
guarantee that global or regional CBF, or NVC, will be Gehirn: Untersuchungen. Leipzig: Von Veit, 1881.
normal. Instead, we proposed an integrative approach to 8. Sandrone S, Bacigaluppi M, Galloni MR, Cappa SF, Moro A, Catani
study the regulation of CBF, hoping to stimulate research M, Filippi M, Monti MM, Perani D, Martino G. Weighing brain activity
and clinical efforts to disentangle autoregulation from the with the balance: Angelo Mosso’s original manuscripts come to
multiple other mechanisms that influence CBF. light. Brain 137: 621–633, 2014. doi:10.1093/brain/awt091.

9. Roy CS, Sherrington CS. On the regulation of the blood-supply of


the brain. J Physiol 11: 85–158, 1890. doi:10.1113/jphysiol.1890.
sp000321.
CORRESPONDENCE
10. Raichle ME. Behind the scenes of functional brain imaging: a histori-
J.A.H.R. Claassen (e-mail: jurgen.claassen@radboudumc.nl). cal and physiological perspective. Proc Natl Acad Sci U S A 95:
765–772, 1998. doi:10.1073/pnas.95.3.765.

11. Rasmussen MK, Mestre H, Nedergaard M. The glymphatic pathway


GRANTS in neurological disorders. Lancet Neurol 17: 1016–1024, 2018.
doi:10.1016/S1474-4422(18)30318-1.
During the preparation of this article, F.M.F. was supported by 12. Wardlaw JM, Benveniste H, Nedergaard M, Zlokovic BV, Mestre H,
National Institute of Neurological Disorders and Stroke Grants Lee H, Doubal FN, Brown R, Ramirez J, MacIntosh BJ, Tannenbaum
NS-096465 and NS-108409 and J.A.C. was supported by A, Ballerini L, Rungta RL, Boido D, Sweeney M, Montagne A,
Alzheimer Nederland Grant WE.03-2015-15040. Charpak S, Joutel A, Smith KJ, Black SE, colleagues from the
Fondation Leducq Transatlantic Network of Excellence on the Role
of the Perivascular Space in Cerebral Small Vessel Disease.
Perivascular spaces in the brain: anatomy, physiology and pathol-
DISCLOSURES ogy. Nat Rev Neurol 16: 137–153, 2020. doi:10.1038/s41582-020-
0312-z.
No conflicts of interest, financial or otherwise, are declared by
13. Fog M. The relationship between the blood pressure and the tonic
the authors. regulation of the pial arteries. J Neurol Psychiatry 1: 187–197, 1938.
doi:10.1136/jnnp.1.3.187.

14. Forbes HS, Cobb SS. Vasomotor control of cerebral vessels. Brain
AUTHOR CONTRIBUTIONS 61: 221–232, 1938. doi:10.1093/brain/61.2.221.

J.A.C. conceived and designed the review; J.A.C. and F.M.F. 15. Sokoloff L. The action of drugs on the cerebral circulation.
Pharmacol Rev 11: 1–85, 1959.
prepared figures; J.A.C., D.H.T., R.B.P., and F.M.F. drafted
manuscript; J.A.C., D.H.T., R.B.P., and F.M.F. edited and re- 16. Rosenblum WI. Cerebral microcirculation: a review emphasizing the
vised manuscript; J.A.C., D.H.T., R.B.P., and F.M.F. approved interrelationship of local blood flow and neuronal function.
final version of manuscript. Angiology 16: 485–507, 1965. doi:10.1177/000331976501600809.

17. Fulton JF. Vasomotor and reflex sequelae of unilateral cervical


and lumbar ramisectomy in a case of Raynaud's disease, with
observations on tonus. Ann Surg 88: 827–841, 1928. doi:10.1097/
REFERENCES
00000658-192811000-00003.

1. Willis T. Cerebri Anatome: Cui Accessit Nervorum Descriptio Et 18. Kety SS. Quantitative determination of cerebral blood flow in man.
Usus. London: James Flesher, Joseph Martyn and James Allestry, Methods Med Res 1: 204–217, 1948.
1664.
19. Kety SS, Schmidt CF. The nitrous oxide method for the quantita-
2. Monro A. Observations on the Structure and Functions of the tive determination of cerebral blood flow in man: theory, proce-
Nervous System. Edinburgh, Scotland: William Creech, 1783. dure and normal values. J Clin Invest 27: 476–483, 1948. doi:
10.1172/JCI101994.
3. Friedland RP, Iadecola C. Roy and Sherrington (1890): a centennial
reexamination of “On the regulation of the blood-supply of the 20. Lassen NA, Ingvar DH. The blood flow of the cerebral cortex deter-
brain”. Neurology 41: 10–14, 1991. doi:10.1212/WNL.41.1.10. mined by radioactive krypton. Experientia 17: 42–43, 1961. doi:10.
1007/BF02157946.
4. Hill L. The Physiology and Pathology of the Cerebral Circulation. An
Experimental Research. London: J&A Churchill, 1896. 21. Payne S. Cerebral Blood Flow and Metabolism: A Quantitative
Approach. Hackensack, NJ: World Scientific, 2018, p. 1–440.
5. Bayliss WM, Hill L, Gulland GL. On intra-cranial pressure and the cer-
ebral circulation: Part I. Physiological; Part II. Histological. J Physiol 22. Wintermark M, Sesay M, Barbier E, Borbe ly K, Dillon WP, Eastwood
18: 334–362, 1895. doi:10.1113/jphysiol.1895.sp000572. JD, Glenn TC, Grandin CB, Pedraza S, Soustiel JF, Nariai T,
 JM, Dousset V, Yonas H. Comparative overview
Zaharchuk G, Caille
6. Donders FC. Over de snelheid van psychische processen. of brain perfusion imaging techniques. J Neuroradiol 32: 294–314,
Onderzoekingen gedaan in het Physiologisch Laboratorium der 2005. doi:10.1016/s0150-9861(05)83159-1.

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1537


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

23. Aaslid R, Huber P, Nornes H. Evaluation of cerebrovascular spasm special focus on the elderly. J Cereb Blood Flow Metabol 28: 1071–
with transcranial Doppler ultrasound. J Neurosurg 60: 37–41, 1984. 1085, 2008. doi:10.1038/jcbfm.2008.13.
doi:10.3171/jns.1984.60.1.0037.
42. Willie CK, Tzeng YC, Fisher JA, Ainslie PN. Integrative regulation of
24. Aaslid R, Markwalder TM, Nornes H. Noninvasive transcranial human brain blood flow. J Physiol 592: 841–859, 2014. doi:10.1113/
Doppler ultrasound recording of flow velocity in basal cerebral jphysiol.2013.268953.
arteries. J Neurosurg 57: 769–774, 1982. doi:10.3171/jns.1982.57.6.
0769. 43. Claassen JA, Zhang R, Fu Q, Witkowski S, Levine BD. Transcranial
Doppler estimation of cerebral blood flow and cerebrovascular con-
25. del Zoppo GJ, Sharp FR, Heiss WD, Albers GW. Heterogeneity in ductance during modified rebreathing. J Appl Physiol 102: 870–
the penumbra. J Cereb Blood Flow Metab 31: 1836–1851, 2011. 877, 2007. doi:10.1152/japplphysiol.00906.2006.
doi:10.1038/jcbfm.2011.93.
44. Girouard H, Iadecola C. Neurovascular coupling in the normal brain
26. Lipton P. Ischemic cell death in brain neurons. Physiol Rev 79: 1431– and in hypertension, stroke, and Alzheimer disease. J Appl Physiol
1568, 1999. doi:10.1152/physrev.1999.79.4.1431. (1985) 100: 328–335, 2006. doi:10.1152/japplphysiol.00966.2005.
27. Goldberger ZD, Petek BJ, Brignole M, Shen WK, Sheldon RS, 45. Iadecola C. The neurovascular unit coming of age: a journey
Solbiati M, Deharo JC, Moya A, Hamdan MH. ACC/AHA/HRS versus through neurovascular coupling in health and disease. Neuron 96:
ESC guidelines for the diagnosis and management of syncope: 17–42, 2017. doi:10.1016/j.neuron.2017.07.030.
JACC guideline comparison. J Am Coll Cardiol 74: 2410–2423,
2019. doi:10.1016/j.jacc.2019.09.012. 46. Faraci FM. Protecting against vascular disease in brain. Am J
Physiol Heart Circ Physiol 300: H1566–H1582, 2011. doi:10.1152/
28. Astrup J, Siesjo BK, Symon L. Thresholds in cerebral ischemia–the ajpheart.01310.2010.
ischemic penumbra. Stroke 12: 723–725, 1981. doi:10.1161/01.
STR.12.6.723. doi:10.1161/01.str.12.6.723. 47. Faraci FM, Heistad DD. Regulation of the cerebral circulation: Role
of endothelium and potassium channels. Physiol Rev 78: 53–97,
29. Siesjo BK. Pathophysiology and treatment of focal cerebral ische- 1998. doi:10.1152/physrev.1998.78.1.53.
mia. Part I: Pathophysiology. J Neurosurg 77: 169–184, 1992. doi:10.
3171/jns.1992.77.2.0169. 48. Katusic ZS, Austin SA. Endothelial nitric oxide: protector of a healthy
mind. Eur Heart J 35: 888–894, 2014. doi:10.1093/eurheartj/eht544.
30. Fuchs FD, Whelton PK. High blood pressure and cardiovascular disease.
Hypertension 75: 285–292, 2020. doi:10.1161/HYPERTENSIONAHA. 49. Longden TA, Dabertrand F, Koide M, Gonzales AL, Tykocki NR,
119.14240. Brayden JE, Hill-Eubanks D, Nelson MT. Capillary K1-sensing ini-
tiates retrograde hyperpolarization to increase local cerebral blood
31. Mills KT, Stefanescu A, He J. The global epidemiology of hyperten-
flow. Nat Neurosci 20: 717–726, 2017. doi:10.1038/nn.4533.
sion. Nat Rev Nephrol 16: 223–237, 2020. doi:10.1038/s41581-019-
0244-2. 50. Chen BR, Kozberg MG, Bouchard MB, Shaik MA, Hillman EA. A criti-
cal role for the vascular endothelium in functional neurovascular
32. Hu X, De Silva TM, Chen J, Faraci FM. Cerebral vascular disease
coupling in the brain. J Am Heart Assoc 3: e000787, 2014.
and neurovascular injury in ischemic stroke. Circ Res 120: 449–471,
doi:10.1161/jaha.114.000787.
2017. doi:10.1161/CIRCRESAHA.116.308427.
51. Chow BW, Nunez V, Kaplan L, Granger AJ, Bistrong K, Zucker HL,
33. Payne S. Cerebral autoregulation: control of blood flow in the brain.
Kumar P, Sabatini BL, Gu C. Caveolae in CNS arterioles mediate
In: SpringerBriefs in Bioengineering. New York: Springer, 2016, p.
neurovascular coupling. Nature 579: 106–110, 2020. doi:10.1038/
1–125.
s41586-020-2026-1.
34. Brian JE Jr. Carbon dioxide and the cerebral circulation. Anesthesiology
52. Hogan-Cann AD, Lu P, Anderson CM. Endothelial NMDA receptors
88: 1365–1386, 1998. doi:10.1097/00000542-199805000-00029.
mediate activity-dependent brain hemodynamic responses in mice.
35. Hoiland RL, Fisher JA, Ainslie PN. Regulation of the cerebral circula- Proc Natl Acad Sci U S A 116: 10229–10231, 2019. doi:10.1073/
tion by arterial carbon dioxide. Compr Physiol 9: 1101–1154, 2019. pnas.1902647116.
doi:10.1002/cphy.c180021.
53. Austin SA, Katusic ZS. Loss of endothelial nitric oxide synthase pro-
36. Hamel E. Perivascular nerves and the regulation of cerebrovascular motes p25 generation and tau phosphorylation in a murine model
tone. J Appl Physiol (1985) 100: 1059–1064, 2006. doi:10.1152/ of Alzheimer's disease. Circ Res 119: 1128–1134, 2016. doi:10.1161/
japplphysiol.00954.2005. CIRCRESAHA.116.309686.

37. Edvinsson L, Krause DN. Cerebral Blood Flow and Metabolism. 54. Sanders ML, Elting JW, Panerai RB, Aries M, Bor-Seng-Shu E,
London: Lippincott Williams & Wilkins: 2002, p. 1–520. Caicedo A, Chacon M, Gommer ED, Van Huffel S, Jara JL, Kostoglou
K, Mahdi A, Marmarelis VZ, Mitsis GD, Muller M, Nikolic D, Nogueira
38. Lassen NA. Cerebral blood flow and oxygen consumption in man. RC, Payne SJ, Puppo C, Shin DC, Simpson DM, Tarumi T, Yelicich B,
Physiol Rev 39: 183–238, 1959. doi:10.1152/physrev.1959.39.2.183. Zhang R, Claassen J. Dynamic cerebral autoregulation reproducibil-
39. Lassen NA. Control of cerebral circulation in health and disease. ity is affected by physiological variability. Front Physiol 10: 865,
Circ Res 34: 749–760, 1974. doi:10.1161/01.RES.34.6.749. 2019. doi:10.3389/fphys.2019.00865.

40. Panerai RB. Assessment of cerebral pressure autoregulation in 55. Claassen JA, Meel-van den Abeelen AS, Simpson DM, Panerai RB,
humans–a review of measurement methods. Physiol Meas 19: on behalf of the International Cerebral Autoregulation Research
305–338, 1998. doi:10.1088/0967-3334/19/3/001. Network (CARNet). Transfer function analysis of dynamic cerebral
autoregulation: a white paper from the International Autoregulation
41. van Beek AH, Claassen JA, Rikkert MG, Jansen RW. Cerebral autor- Research Network (CARNet). J Cereb Blood Flow Metab 36: 665–
egulation: an overview of current concepts and methodology with 680, 2016. doi:10.1177/0271678X15626425.

1538 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

56. Abbott NJ, Ronnback L, Hansson E. Astrocyte-endothelial interac- 71. Harper SL, Bohlen HG. Microvascular adaptation in the cerebral cor-
tions at the blood-brain barrier. Nat Rev Neurosci 7: 41–53, 2006. tex of adult spontaneously hypertensive rats. Hypertension 6: 408–
doi:10.1038/nrn1824. 419, 1984. doi:10.1161/01.HYP.6.3.408. doi:10.1161/01.hyp.6.3.408.

57. Profaci CP, Munji RN, Pulido RS, Daneman R. The blood-brain barrier 72. Harper SL, Bohlen HG, Rubin MJ. Arterial and microvascular contri-
in health and disease: Important unanswered questions. J Exp Med butions to cerebral cortical autoregulation in rats. Am J Physiol
217: e20190062, 2020. doi:10.1084/jem.20190062. Heart Circ Physiol 246: H17–H24, 1984. doi:10.1152/ajpheart.
1984.246.1.H17.
58. Sweeney MD, Montagne A, Sagare AP, Nation DA, Schneider LS,
Chui HC, et al. Vascular dysfunction–The disregarded partner of 73. Heistad DD, Marcus ML, Abboud FM. Role of large arteries in regula-
Alzheimer's disease. Alzheimers Dement 15: 158–167, 2019. tion of cerebral blood flow in dogs. J Clin Invest 62: 761–768, 1978.
doi:10.1016/j.jalz.2018.07.222. doi:10.1172/JCI109187.

59. Tarasoff-Conway JM, Carare RO, Osorio RS, Glodzik L, Butler T, 74. Heistad DD, Marcus ML, Piegors DJ, Armstrong ML. Regulation of
Fieremans E, Axel L, Rusinek H, Nicholson C, Zlokovic BV, cerebral blood flow in atherosclerotic monkeys. Am J Physiol Heart
Frangione B, Blennow K, Menard J, Zetterberg H, Wisniewski T, de Circ Physiol 239: H539–H544, 1980. doi:10.1152/ajpheart.1980.
Leon MJ. Clearance systems in the brain-implications for Alzheimer 239.4.H539.
disease. Nat Rev Neurol 11: 457–470, 2015. doi:10.1038/nrneurol.
75. Hoffman WE, Edelman G, Kochs E, Werner C, Segil L, Albrecht RF.
2015.119.
Cerebral autoregulation in awake versus isoflurane-anesthetized
60. Jin BJ, Smith AJ, Verkman AS. Spatial model of convective solute rats. Anesth Analg 73: 753–757, 1991. doi:10.1213/00000539-
transport in brain extracellular space does not support a “glym- 199112000-00013.
phatic” mechanism. J Gen Physiol 148: 489–501, 2016. doi:10.1085/
76. Kontos HA, Wei EP, Navari RM, Levasseur JE, Rosenblum WI,
jgp.201611684.
Patterson JL Jr. Responses of cerebral arteries and arterioles to
61. Smith AJ, Yao X, Dix JA, Jin BJ, Verkman AS. Test of the “glym- acute hypotension and hypertension. Am J Physiol Heart Circ
phatic” hypothesis demonstrates diffusive and aquaporin-4-inde- Physiol 234: H371–H383, 1978. doi:10.1152/ajpheart.1978.234.4.
pendent solute transport in rodent brain parenchyma. eLife 6: H371.
e27679, 2017. doi:10.7554/eLife.27679.
77. Marcus ML, Heistad DD. Effects of sympathetic nerves on cerebral
62. Bakker EN, Bacskai BJ, Arbel-Ornath M, Aldea R, Bedussi B, Morris blood flow in awake dogs. Am J Physiol Heart Circ Physiol 236:
AW, Weller RO, Carare RO. Lymphatic clearance of the brain: peri- H549–H553, 1979. doi:10.1152/ajpheart.1979.236.4.H549.
vascular, paravascular and significance for neurodegenerative dis- 78. Sadoshima S, Heistad DD. Regional cerebral blood flow during hy-
eases. Cell Mol Neurobiol 36: 181–194, 2016. doi:10.1007/s10571- potension in normotensive and stroke-prone spontaneously hyper-
015-0273-8. tensive rats: effect of sympathetic denervation. Stroke 14: 575–579,
63. Preston SD, Steart PV, Wilkinson A, Nicoll JA, Weller RO. Capillary 1983. doi:10.1161/01.STR.14.4.575.
and arterial cerebral amyloid angiopathy in Alzheimer's disease: 79. Strandgaard S, MacKenzie ET, Sengupta D, Rowan JO, Lassen NA,
defining the perivascular route for the elimination of amyloid b from Harper AM. Upper limit of autoregulation of cerebral blood flow in
the human brain. Neuropathol Appl Neurobiol 29: 106–117, 2003. the baboon. Circ Res 34: 435–440, 1974. doi:10.1161/01.RES.34.4.
doi:10.1046/j.1365-2990.2003.00424.x. 435.
64. van Veluw SJ, Hou SS, Calvo-Rodriguez M, Arbel-Ornath M, Snyder 80. Tureen JH, Dworkin RJ, Kennedy SL, Sachdeva M, Sande MA. Loss
AC, Frosch MP, Greenberg SM, Bacskai BJ. Vasomotion as a driving of cerebrovascular autoregulation in experimental meningitis in rab-
force for paravascular clearance in the awake mouse brain. Neuron bits. J Clin Invest 85: 577–581, 1990. doi:10.1172/JCI114475.
105: 549–561, 2020. doi:10.1016/j.neuron.2019.10.033.
81. Dalkara T, Irikura K, Huang Z, Panahian N, Moskowitz MA.
65. Heistad DD, Kontos HA. Cerebral circulation. In: Handbook of Cerebrovascular responses under controlled and monitored physio-
Physiology, edited by Shepherd JT. Bethesda, Maryland: American logical conditions in the anesthetized mouse. J Cereb Blood Flow
Physiological Society, 1983, p. 137–182. Metab 15: 631–638, 1995. doi:10.1038/jcbfm.1995.78.
66. Chemtob S, Beharry K, Rex J, Varma DR, Aranda JV. Prostanoids 82. Jones JV, Fitch W, MacKenzie ET, Strandgaard S, Harper AM. Lower
determine the range of cerebral blood flow autoregulation of new- limit of cerebral blood flow autoregulation in experimental renovas-
born piglets. Stroke 21: 777–784, 1990. doi:10.1161/01.STR.21.5.777. cular hypertension in the baboon. Circ Res 39: 555–557, 1976.
doi:10.1161/01.RES.39.4.555.
67. Faraci FM, Heistad DD, Mayhan WG. Role of large arteries in regula-
tion of blood flow to brain stem in cats. J Physiol 387: 115–123, 1987. 83. Tuor UI, Farrar JK. Pial vessel caliber and cerebral blood flow during
doi:10.1113/jphysiol.1987.sp016566. hemorrhage and hypercapnia in the rabbit. Am J Physiol Heart Circ
Physiol 247: H40–H51, 1984. doi:10.1152/ajpheart.1984.247.1.H40.
68. Fitch W, Ferguson GG, Sengupta D, Garibi J, Harper AM.
Autoregulation of cerebral blood flow during controlled hypotension 84. Faraci FM, Mayhan WG, Heistad DD. Segmental vascular responses
in baboons. J Neurol Neurosurg Psychiatry 39: 1014–1022, 1976. to acute hypertension in cerebrum and brain stem. Am J Physiol
doi:10.1136/jnnp.39.10.1014. Heart Circ Physiol 252: H738–H742, 1987. doi:10.1152/
ajpheart.1987.252.4.H738.
69. Fitch W, Jones JV, MacKenzie ET, Harper AM. Autoregulation of cer-
ebral blood flow in chronically hypertensive baboons. Br J Surg 63: 85. Claassen JA. The plateau phase is a slippery slope: Raising blood
663, 1976. pressure may lower brain perfusion. J Physiol 594: 2783, 2016.
doi:10.1113/JP272121.
70. Harper AM, Lassen NA, MacKenzie ET, Rowan JO, Sengupta D,
Strandgaard S. Proceedings: the upper limit of ‘autoregulation’ of 86. Liu J, Tseng BY, Khan MA, Tarumi T, Hill C, Mirshams N, Hodics TM,
cerebral blood flow in the baboon. J Physiol 234: 61P–62P, 1973. Hynan LS, Zhang R. Individual variability of cerebral autoregulation,

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1539


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

posterior cerebral circulation and white matter hyperintensity. J Doppler study. J Cereb Blood Flow Metab 4: 368–372, 1984.
Physiol 594: 3141–3155, 2016. doi:10.1113/JP271068. doi:10.1038/jcbfm.1984.54.

87. Numan T, Bain AR, Hoiland RL, Smirl JD, Lewis NC, Ainslie PN. Static 103. Birch AA, Dirnhuber MJ, Hartley-Davies R, Iannotti F, Neil-Dwyer G.
autoregulation in humans: A review and reanalysis. Med Eng Phys Assessment of autoregulation by means of periodic changes in
36: 1487–1495, 2014. doi:10.1016/j.medengphy.2014.08.001. blood pressure. Stroke 26: 834–837, 1995. doi:10.1161/01.STR.26.5.
834.
88. Kaufmann H, Norcliffe-Kaufmann L, Palma JA. Baroreflex dysfunction.
N Engl J Med 382: 163–178, 2020. doi:10.1056/NEJMra1509723. 104. Diehl RR, Linden D, L€
uCke D, Berlit P. Phase relationship between
cerebral blood flow velocity and blood pressure. A clinical test of
89. de Jong DL, Tarumi T, Liu J, Zhang R, Claassen J. Lack of linear cor- autoregulation. Stroke 26: 1801–1804, 1995. doi:10.1161/01.STR.26.
relation between dynamic and steady-state cerebral autoregulation. 10.1801.
J Physiol 595: 5623–5636, 2017. doi:10.1113/JP274304.
105. Diehl RR, Linden D, Lucke D, Berlit P. Spontaneous blood pressure
90. Tiecks FP, Lam AM, Aaslid R, Newell DW. Comparison of static and oscillations and cerebral autoregulation. Clin Auton Res 8: 7–12,
dynamic cerebral autoregulation measurements. Stroke 26: 1014– 1998. doi:10.1007/BF02267598.
1019, 1995. doi:10.1161/01.STR.26.6.1014.
106. Giller CA. The frequency-dependent behavior of cerebral autoregu-
91. Panerai RB. Cerebral autoregulation: from models to clinical applica- lation. Neurosurgery 27: 362–368, 1990. doi:10.1227/00006123-
tions. Cardiovasc Eng 8: 42–59, 2008. doi:10.1007/s10558-007- 199009000-00004.
9044-6.
107. Panerai RB, Kelsall AW, Rennie JM, Evans DH. Analysis of cerebral
92. Panerai RB, Carey BJ, Potter JF. Short-term variability of cerebral blood flow autoregulation in neonates. IEEE Trans Biomed Eng 43:
blood flow velocity responses to arterial blood pressure transients. 779–788, 1996. doi:10.1109/10.508541.
Ultrasound Med Biol 29: 31–38, 2003. doi:10.1016/S0301-5629(02)
00698-1. 108. Panerai RB, White RP, Markus HS, Evans DH. Grading of cerebral
dynamic autoregulation from spontaneous fluctuations in arterial
93. Panerai RB, Dawson SL, Eames PJ, Potter JF. Cerebral blood flow blood pressure. Stroke 29: 2341–2346, 1998. doi:10.1161/01.STR.29.
velocity response to induced and spontaneous sudden changes in 11.2341.
arterial blood pressure. Am J Physiol Heart Circ Physiol 280:
H2162–H2174, 2001. doi:10.1152/ajpheart.2001.280.5.H2162. 109. Blaber AP, Bondar RL, Stein F, Dunphy PT, Moradshahi P, Kassam
MS, Freeman R. Transfer function analysis of cerebral autoregula-
94. Panerai RB, Eames PJ, Potter JF. Variability of time-domain indices tion dynamics in autonomic failure patients. Stroke 28: 1686–1692,
of dynamic cerebral autoregulation. Physiol Meas 24: 367–381, 1997. doi:10.1161/01.STR.28.9.1686.
2003. doi:10.1088/0967-3334/24/2/312.
110. Zhang R, Zuckerman JH, Giller CA, Levine BD. Transfer function
95. Panerai RB, Rennie JM, Kelsall AW, Evans DH. Frequency-domain analysis of dynamic cerebral autoregulation in humans. Am J
analysis of cerebral autoregulation from spontaneous fluctuations in Physiol Heart Circ Physiol 274: H233–H241, 1998. doi:10.1152/
arterial blood pressure. Med Biol Eng Comput 36: 315–322, 1998. ajpheart.1998.274.1.H233.
doi:10.1007/BF02522477.
111. Meel-van den Abeelen AS, Simpson DM, Wang LJ, Slump CH,
96. Greenfield JC Jr, Rembert JC, Tindall GT. Transient changes in cere- Zhang R, Tarumi T, Rickards CA, Payne S, Mitsis GD, Kostoglou K,
bral vascular resistance during the Valsalva maneuver in man. Marmarelis V, Shin D, Tzeng YC, Ainslie PN, Gommer E, Muller M,
Stroke 15: 76–79, 1984. doi:10.1161/01.STR.15.1.76. Dorado AC, Smielewski P, Yelicich B, Puppo C, Liu X, Czosnyka M,
97. Nornes H, Knutzen HB, Wikeby P. Cerebral arterial blood flow and Wang CY, Novak V, Panerai RB, Claassen JA. Between-centre vari-
aneurysm surgery. J Neurosurg 47: 819–827, 1977. doi:10.3171/ ability in transfer function analysis, a widely used method for linear
jns.1977.47.6.0819. quantification of the dynamic pressure-flow relation: The CARNet
Study. Med Eng Phys 36: 620–627, 2014. doi:10.1016/j.medengphy.
98. Nornes H, Wikeby P. Cerebral arterial blood flow and aneurysm sur- 2014.02.002.
gery. Part 1: Local arterial flow dynamics. J Neurosurg 47: 810–818,
1977. doi:10.3171/jns.1977.47.6.0810. 112. Meel-van den Abeelen AS, Van Beek AH, Slump CH, Panerai RB,
Claassen JA. Transfer function analysis for the assessment of cere-
99. de Heus RA, de Jong DL, Sanders ML, van Spijker GJ, Oudegeest- bral autoregulation using spontaneous oscillations in blood pres-
Sander MH, Hopman MT, Lawlor BA, Olde Rikkert MG, Claassen J. sure and cerebral blood flow. Med Engineering Physics 36: 563–
Dynamic regulation of cerebral blood flow in patients with 575, 2014. doi:10.1016/j.medengphy.2014.02.001.
Alzheimer disease. Hypertension 72: 139–150, 2018. doi:10.1161/
HYPERTENSIONAHA.118.10900. 113. Tzeng YC, Panerai RB. CrossTalk proposal: dynamic cerebral autor-
egulation should be quantified using spontaneous blood pressure
100. van Beek AH, Rikkert O, Pasman JW, Hopman MT, Claassen JA. fluctuations. J Physiol 596: 3–5, 2018. doi:10.1113/JP273899.
Dynamic cerebral autoregulation in the old using a repeated sit-
stand maneuver. Ultrasound Med Biol 36: 192–201, 2010. doi:10. 114. Simpson D, Claassen J. CrossTalk opposing view: dynamic cerebral
1016/j.ultrasmedbio.2009.10.011. autoregulation should be quantified using induced (rather than
spontaneous) blood pressure fluctuations. J Physiol 596: 7–9, 2018.
101. Aaslid R, Lindegaard KF, Sorteberg W, Nornes H. Cerebral autore- doi:10.1113/JP273900.
gulation dynamics in humans. Stroke 20: 45–52, 1989. [PMC
doi:10.1161/01.STR.20.1.45. 115. van den Brule JM, van Kaam CR, van der Hoeven JG, Claassen J,
Hoedemaekers CW. Influence of induced blood pressure variability
102. Markwalder TM, Grolimund P, Seiler RW, Roth F, Aaslid R. on the assessment of cerebral autoregulation in patients after car-
Dependency of blood flow velocity in the middle cerebral artery on diac arrest. Biomed Res Int 2018: 8153241, 2018. doi:10.1155/2018/
end-tidal carbon dioxide partial pressure–a transcranial ultrasound 8153241.

1540 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

116. Elting JW, Sanders ML, Panerai RB, Aries M, Bor-Seng-Shu E, 130. Peixoto AJ. Acute severe hypertension. N Engl J Med 381: 1843–
Caicedo A, Chacon M, Gommer ED, Van Huffel S, Jara JL, Kostoglou 1852, 2019. doi:10.1056/NEJMcp1901117.
K, Mahdi A, Marmarelis VZ, Mitsis GD, Muller M, Nikolic D, Nogueira
RC, Payne SJ, Puppo C, Shin DS, Simpson DM, Tarumi T, Yeliich B, 131. Dabertrand F, Kroigaard C, Bonev AD, Cognat E, Dalsgaard T,
Zhang R, Claassen JA. Assessment of dynamic cerebral autoregula- Domenga-Denier V, Hill-Eubanks DC, Brayden JE, Joutel A, Nelson
tion in humans: is reproducibility dependent on blood pressure vari- MT. Potassium channelopathy-like defect underlies early-stage
ability? PLoS One 15: e0227651, 2020. doi:10.1371/journal.pone. cerebrovascular dysfunction in a genetic model of small vessel dis-
0227651. ease. Proc Natl Acad Sci USA 112: E796–805, 2015. doi:10.1073/
pnas.1420765112.
117. Sanders ML, Claassen J, Aries M, Bor-Seng-Shu E, Caicedo A,
Chacon M, Gommer ED, Van Huffel S, Jara JL, Kostoglou K, Mahdi 132. De Silva TM, Modrick ML, Dabertrand F, Faraci FM. Changes in cere-
A, Marmarelis VZ, Mitsis GD, Muller M, Nikolic D, Nogueira RC, bral arteries and parenchymal arterioles with aging: role of Rho ki-
Payne SJ, Puppo C, Shin DC, Simpson DM, Tarumi T, Yelicich B, nase 2 and impact of genetic background. Hypertension 71: 921–
Zhang R, Panerai RB, Elting JW. Reproducibility of dynamic cerebral 927, 2018. doi:10.1161/HYPERTENSIONAHA.118.10865.
autoregulation parameters: a multi-centre, multi-method study. 133. Elhusseiny A, Hamel E. Muscarinic–but not nicotinic–acetylcholine
Physiol Meas 39: 125002, 2018. doi:10.1088/1361-6579/aae9fd. receptors mediate a nitric oxide-dependent dilation in brain cortical
arterioles: a possible role for the M5 receptor subtype. J Cereb
118. Claassen JA, Levine BD, Zhang R. Dynamic cerebral autoregulation
Blood Flow Metabol 20: 298–305, 2000. doi:10.1097/00004647-
during repeated squat-stand maneuvers. J Appl Physiol 106: 153–
200002000-00011.
160, 2009. doi:10.1152/japplphysiol.90822.2008.
134. Faraci FM, Baumbach GL, Heistad DD. Myogenic mechanisms in the
119. Smirl JD, Hoffman K, Tzeng YC, Hansen A, Ainslie PN.
cerebral circulation. J Hypertens Suppl 7: S61–64, 1989. 2681598
Methodological comparison of active- and passive-driven oscilla-
tions in blood pressure; implications for the assessment of cerebral 135. Knot HJ, Nelson MT. Regulation of membrane potential and diame-
pressure-flow relationships. J Appl Physiol 119: 487–501, 2015. ter by voltage-dependent K1 channels in rabbit myogenic cerebral
doi:10.1152/japplphysiol.00264.2015. arteries. Am J Physiol Heart Circ Physiol 269: H348–H355, 1995.
doi:10.1152/ajpheart.1995.269.1.H348.
120. Diehl RR. Cerebral autoregulation studies in clinical practice. Eur J
Ultrasound 16: 31–36, 2002. doi:10.1016/s0929-8266(02)00048-4. 136. McCarron JG, Osol G, Halpern W. Myogenic responses are inde-
pendent of the endothelium in rat pressurized posterior cerebral
121. Katsogridakis E, Simpson DM, Bush G, Fan L, Birch AA, Allen R,
arteries. Blood Vessels 26: 315–319, 1989. doi:10.1159/000158780.
Potter JF, Panerai RB. Coherent averaging of pseudorandom binary
stimuli: is the dynamic cerebral autoregulatory response symmetri- 137. Wallis SJ, Firth J, Dunn WR. Pressure-induced myogenic responses
cal? Physiol Meas 38: 2164–2175, 2017. doi:10.1088/1361-6579/ in human isolated cerebral resistance arteries. Stroke 27: 2287–
aa9086. 2290, 1996. doi:10.1161/01.str.27.12.2287.
122. Hamner JW, Cohen MA, Mukai S, Lipsitz LA, Taylor JA. Spectral indi- 138. Ward ME, Yan L, Kelly S, Angle MR. Flow modulation of pressure-
ces of human cerebral blood flow control: responses to augmented sensitive tone in rat pial arterioles: Role of the endothelium.
blood pressure oscillations. J Physiol 559: 965–973, 2004. doi:10. Anesthesiology 93: 1456–1464, 2000. doi:10.1097/00000542-
1113/jphysiol.2004.066969. 200012000-00018.
123. Hamner JW, Ishibashi K, Tan CO. Revisiting human cerebral blood 139. Lidington D, Kroetsch JT, Bolz SS. Cerebral artery myogenic reactiv-
flow responses to augmented blood pressure oscillations. J Physiol ity: the next frontier in developing effective interventions for subar-
597: 1553–1564, 2019. doi:10.1113/JP277321. achnoid hemorrhage. J Cereb Blood Flow Metabol 38: 17–37, 2018.
doi:10.1177/0271678X17742548.
124. Zhang R, Zuckerman JH, Levine BD. Spontaneous fluctuations in
cerebral blood flow: Insights from extended-duration recordings in 140. Harraz OF, Hill-Eubanks D, Nelson MT. PIP2: a critical regulator of
humans. Am J Physiol Heart Circ Physiol 278: H1848–1855, 2000. vascular ion channels hiding in plain sight. Proc Natl Acad Sci U S A
doi:10.1152/ajpheart.2000.278.6.H1848. 117: 20378–20389, 2020. doi:10.1073/pnas.2006737117.

125. Panerai RB, Robinson TG, Minhas JS. The upper frequency limit of 141. Knot HJ, Nelson MT. Regulation of arterial diameter and wall [Ca21]
dynamic cerebral autoregulation. J Physiol 597: 5821–5833, 2019. in cerebral arteries of rat by membrane potential and intravascular
doi:10.1113/JP278710. pressure. J Physiol 508: 199–209, 1998. doi:10.1111/j.1469-7793.
1998.199br.x.
126. Tan CO. Defining the characteristic relationship between arterial
pressure and cerebral flow. J Appl Physiol (1985) 113: 1194–1200, 142. Nelson MT, Cheng H, Rubart M, Santana LF, Bonev AD, Knot HJ,
2012. doi:10.1152/japplphysiol.00783.2012. Lederer WJ. Relaxation of arterial smooth muscle by calcium sparks.
Science 270: 633–637, 1995. doi:10.1126/science.270.5236.633.
127. De Silva TM, Faraci FM. Microvascular dysfunction and cognitive
impairment. Cell Mol Neurobiol 36: 241–258, 2016. doi:10.1007/ 143. Li Y, Brayden JE. Rho kinase activity governs arteriolar myogenic
s10571-015-0308-1. depolarization. J Cereb Blood Flow Metab 37: 140–152, 2017. doi:
10.1177/0271678X15621069.
128. Faraci FM, Heistad DD. Regulation of large cerebral arteries and cer-
ebral microvascular pressure. Circ Res 66: 8–17, 1990. doi:10.1161/01. 144. Brayden JE, Li Y, Tavares MJ. Purinergic receptors regulate myo-
RES.66.1.8. genic tone in cerebral parenchymal arterioles. J Cereb Blood Flow
Metab 33: 293–299, 2013. doi:10.1038/jcbfm.2012.169.
129. Cipolla MJ. The cerebral circulation. Colloquium Series on
Integrated Systems Physiology: from Molecule to Function 145. Chennupati R, Wirth A, Favre J, Li R, Bonnavion R, Jin YJ,
(2nd ed.). London: Morgan & Claypool, 2016, p. 1–67. doi:10.4199/ Wietelmann A, Schweda F, Wettschureck N, Henrion D, Offermanns
C00141ED2V01Y201607ISP066. S. Myogenic vasoconstriction requires G12/G13 and LARG to maintain

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1541


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

local and systemic vascular resistance. Elife 8: e49374, 2019. doi:10. 159. Toyoda K, Fujii K, Ibayashi S, Kitazono T, Nagao T, Fujishima M. Role
7554/eLife.49374. of ATP-sensitive potassium channels in brain stem circulation during
hypotension. Am J Physiol Heart Circ Physiol 273: H1342–H1346,
146. Earley S, Waldron BJ, Brayden JE. Critical role for transient receptor 1997. doi:10.1152/ajpheart.1997.273.3.H1342.
potential channel TRPM4 in myogenic constriction of cerebral
arteries. Circ Res 95: 922–929, 2004. doi:10.1161/01.RES.0000147 160. Emdin CA, Khera AV, Klarin D, Natarajan P, Zekavat SM, Nomura A,
311.54833.03. Haas M, Aragam K, Ardissino D, Wilson JG, Schunkert H, McPherson
R, Watkins H, Elosua R, Bown MJ, Samani NJ, Baber U, Erdmann J,
147. Lim M, Choi SK, Cho YE, Yeon SI, Kim EC, Ahn DS, Lee YH. The role Gormley P, Palotie A, Stitziel NO, Gupta N, Danesh J, Saleheen D,
of sphingosine kinase 1/sphingosine-1-phosphate pathway in the Gabriel S, Kathiresan S. Phenotypic consequences of a genetic pre-
myogenic tone of posterior cerebral arteries. PLoS One 7: e35177, disposition to enhanced nitric oxide signaling. Circulation 137: 222–
2012. doi:10.1371/journal.pone.0035177. 232, 2018. doi:10.1161/CIRCULATIONAHA.117.028021.
148. VanLandingham LG, Gannon KP, Drummond HA. Pressure-induced 161. Kim KJ, Iddings JA, Stern JE, Blanco VM, Croom D, Kirov SA, Filosa
constriction is inhibited in a mouse model of reduced bENaC. Am J JA. Astrocyte contributions to flow/pressure-evoked parenchymal
Physiol Regul Integr Comp Physiol 297: R723–R728, 2009. arteriole vasoconstriction. J Neurosci 35: 8245–8257, 2015.
doi:10.1152/ajpregu.00212.2009. doi:10.1523/JNEUROSCI.4486-14.2015.
149. Welsh DG, Morielli AD, Nelson MT, Brayden JE. Transient receptor 162. Kuluz JW, Prado R, Chang J, Ginsberg MD, Schleien CL, Busto R.
potential channels regulate myogenic tone of resistance arteries. Selective brain cooling increases cortical cerebral blood flow in rats.
Circ Res 90: 248–250, 2002. doi:10.1161/hh0302.105662. Am J Physiol Heart Circ Physiol 265: H824–H827, 1993. doi:10.
1152/ajpheart.1993.265.3.H824.
150. Zeiler FA, Thelin EP, Donnelly J, Stevens AR, Smielewski P,
Czosnyka M, Hutchinson PJ, Menon DK. Genetic drivers of cerebral 163. Mederos y Schnitzler M, Storch U, Meibers S, Nurwakagari P, Breit
blood flow dysfunction in TBI: a speculative synthesis. Nat Rev A, Essin K, Gollasch M, Gudermann T. Gq-coupled receptors as
Neurol 15: 25–39, 2019. doi:10.1038/s41582-018-0105-9. mechanosensors mediating myogenic vasoconstriction. EMBO J
27: 3092–3103, 2008. doi:10.1038/emboj.2008.233.
151. Retailleau K, Duprat F, Arhatte M, Ranade SS, Peyronnet R, Martins
JR, Jodar M, Moro C, Offermanns S, Feng Y, Demolombe S, Patel A, 164. Arrick DM, Sharpe GM, Sun H, Mayhan WG. Losartan improves
Honore E. Piezo1 in smooth muscle cells is involved in hypertension- impaired nitric oxide synthase-dependent dilatation of cerebral arte-
dependent arterial remodeling. Cell Rep 13: 1161–1171, 2015. doi:10. rioles in type 1 diabetic rats. Brain Res 1209: 128–135, 2008. doi:10.
1016/j.celrep.2015.09.072. 1016/j.brainres.2008.03.020.
152. Coucha M, Abdelsaid M, Ward R, Abdul Y, Ergul A. Impact of meta- 165. Girouard H, Park L, Anrather J, Zhou P, Iadecola C. Angiotensin II
bolic diseases on cerebral circulation: Structural and functional con- attenuates endothelium-dependent responses in the cerebral micro-
sequences. Compr Physiol 8: 773–799, 2018. doi:10.1002/cphy. circulation through Nox-2-derived radicals. Arterioscler Thromb
c170019. Vasc Biol 26: 826–832, 2006. doi:10.1161/01.ATV.0000205849.
22807.6e.
153. De Silva TM, Ketsawatsomkron P, Pelham C, Sigmund CD, Faraci
FM. Genetic interference with peroxisome proliferator-activated re- 166. Matulla B, Streit G, Pieh S, Findl O, Entlicher J, Graselli U, Eichler HG,
ceptor c in smooth muscle enhances myogenic tone in the cerebro- Wolzt M, Schmetterer L. Effects of losartan on cerebral and ocular
vasculature via a Rho kinase-dependent mechanism. Hypertension circulation in healthy subjects. Br J Clin Pharmacol 44: 369–375,
65: 345–351, 2015. doi:10.1161/HYPERTENSIONAHA.114.04541. 1997. doi:10.1046/j.1365-2125.1997.t01-1-00598.x.
154. Lidington D, Fares JC, Uhl FE, Dinh DD, Kroetsch JT, Sauve M, Malik 167. Warrington JP, Fan F, Duncan J, Cunningham MW, LaMarca BB,
FA, Matthes F, Vanherle L, Adel A, Momen A, Zhang H, Aschar- Dechend R, Wallukat G, Roman RJ, Drummond HA, Granger JP,
Sobbi R, Foltz WD, Wan H, Sumiyoshi M, Macdonald RL, Husain M, Ryan MJ. The angiotensin II type I receptor contributes to impaired
Backx PH, Heximer SP, Meissner A, Bolz SS. CFTR therapeutics nor- cerebral blood flow autoregulation caused by placental ischemia in
malize cerebral perfusion deficits in mouse models of heart failure pregnant rats. Biol Sex Differ 10: 58, 2019. doi:10.1186/s13293-019-
and subarachnoid hemorrhage. JACC Basic Transl Sci 4: 940–958, 0275-1.
2019. doi:10.1016/j.jacbts.2019.07.004.
168. Early CB, Dewey RC, Pieper HP, Hunt WE. Dynamic pressure-flow
155. Huang Z, Huang PL, Ma J, Meng W, Ayata C, Fishman MC, relationships of brain blood flow in the monkey. J Neurosurg 41:
Moskowitz MA. Enlarged infarcts in endothelial nitric oxide synthase 590–596, 1974. doi:10.3171/jns.1974.41.5.0590.
knockout mice are attenuated by nitro-L-arginine. J Cereb Blood
Flow Metab 16: 981–987, 1996. doi:10.1097/00004647-199609000- 169. Kety SS, Hafkenschiel JH, Jeffers WA, Leopold IH, Shenkin HA. The
00023. blood flow, vascular resistance, and oxygen consumption of the
brain in essential hypertension. J Clin Invest 27: 511–514, 1948.
156. Kusano Y, Echeverry G, Miekisiak G, Kulik TB, Aronhime SN, Chen doi:10.1172/JCI101998.
JF, Winn HR. Role of adenosine A2 receptors in regulation of cere-
bral blood flow during induced hypotension. J Cereb Blood Flow 170. Burton AC. Physiology and Biophysics of the Circulation. Chicago,
Metab 30: 808–815, 2010. doi:10.1038/jcbfm.2009.244. IL: Year Book Medical Publishers, 1972, p. 1–226.

157. Rocha MP, Campos MO, Mattos JD, Mansur DE, Rocha HN, Secher 171. Panerai RB, Coughtrey H, Rennie JM, Evans DH. A model of the in-
NH, Nobrega AC, Fernandes IA. KATP channels modulate cerebral stantaneous pressure-velocity relationships of the neonatal cerebral
blood flow and oxygen delivery during isocapnic hypoxia in circulation. Physiol Meas 14: 411–418, 1993. doi:10.1088/0967-3334/
humans. J Physiol 598: 3343–3356, 2020. doi:10.1113/JP279751. 14/4/002.

158. Shin HK, Hong KW. Importance of calcitonin gene-related peptide, 172. Payne S, Morris H, Rowley A. A combined haemodynamic and bio-
adenosine and reactive oxygen species in cerebral autoregulation chemical model of cerebral autoregulation. Conf Proc IEEE Eng
under normal and diseased conditions. Clin Exp Pharmacol Physiol Med Biol Soc 2005: 2295–2298, 2005. doi:10.1109/iembs.2005.
31: 1–7, 2004. doi:10.1111/j.1440-1681.2004.03943.x. 1616923.

1542 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

173. Mayhan WG, Faraci FM, Heistad DD. Mechanisms of protection of 188. Hall CN, Reynell C, Gesslein B, Hamilton NB, Mishra A, Sutherland
the blood-brain barrier during acute hypertension in chronically hy- BA, O'Farrell FM, Buchan AM, Lauritzen M, Attwell D. Capillary peri-
pertensive rats. Hypertension 9: 101–105, 1987. doi:10.1161/01. cytes regulate cerebral blood flow in health and disease. Nature
HYP.9.6_Pt_2.III101. 508: 55–60, 2014. doi:10.1038/nature13165.

174. Grubb S, Cai C, Hald BO, Khennouf L, Murmu RP, Jensen AG, 189. Hill RA, Tong L, Yuan P, Murikinati S, Gupta S, Grutzendler J.
Fordsmann J, Zambach S, Lauritzen M. Precapillary sphincters main- Regional blood flow in the normal and ischemic brain is controlled
tain perfusion in the cerebral cortex. Nat Commun 11: 395, 2020. by arteriolar smooth muscle cell contractility and not by capillary
doi:10.1038/s41467-020-14330-z. pericytes. Neuron 87: 95–110, 2015. doi:10.1016/j.neuron.2015.06.
001.
175. Ratelade J, Klug NR, Lombardi D, Angelim M, Dabertrand F,
Domenga-Denier V, Al-Shahi Salman R, Smith C, Gerbeau JF, 190. Wei HS, Kang H, Rasheed ID, Zhou S, Lou N, Gershteyn A,
Nelson MT, Joutel A. Reducing hypermuscularization of the transi- McConnell ED, Wang Y, Richardson KE, Palmer AF, Xu C, Wan J,
tional segment between arterioles and capillaries protects against Nedergaard M. Erythrocytes are oxygen-sensing regulators of the
spontaneous intracerebral hemorrhage. Circulation 141: 2078– cerebral microcirculation. Neuron 91: 851–862, 2016. doi:10.1016/j.
2094, 2020. doi:10.1161/CIRCULATIONAHA.119.040963. neuron.2016.07.016.
176. Baumbach GL, Heistad DD. Effects of sympathetic stimulation and 191. Faraci FM. Watching small vessel disease grow. Circ Res 122: 810–
changes in arterial pressure on segmental resistance of cerebral 812, 2018. doi:10.1161/CIRCRESAHA.118.312762.
vessels in rabbits and cats. Circ Res 52: 527–533, 1983. doi:10.1161/
01.RES.52.5.527. 192. Mayhan WG, Faraci FM, Heistad DD. Effects of vasodilatation and
acidosis on the blood-brain barrier. Microvasc Res 35: 179–192,
177. Laurent S, Boutouyrie P. The structural factor of hypertension: large 1988. doi:10.1016/0026-2862(88)90061-1.
and small artery alterations. Circ Res 116: 1007–1021, 2015. doi:10.
1161/CIRCRESAHA.116.303596. 193. Katz AI, Chen Y, Moreno AH. Flow through a collapsible tube.
Experimental analysis and mathematical model. Biophys J 9: 1261–
178. Marina N, Christie IN, Korsak A, Doronin M, Brazhe A, Hosford PS, 1279, 1969. doi:10.1016/S0006-3495(69)86451-9.
Wells JA, Sheikhbahaei S, Humoud I, Paton JF, Lythgoe MF,
Semyanov A, Kasparov S, Gourine AV. Astrocytes monitor cerebral 194. Takahashi S, Crane AM, Jehle J, Cook M, Kennedy C, Sokoloff L.
perfusion and control systemic circulation to maintain brain blood Role of the cerebellar fastigial nucleus in the physiological regula-
flow. Nature Commun 11: 131, 2020. doi:10.1038/s41467-019-13956-y. tion of cerebral blood flow. J Cereb Blood Flow Metab 15: 128–142,
1995. doi:10.1038/jcbfm.1995.15.
179. Hariharan A, Weir N, Robertson C, He L, Betsholtz C, Longden TA.
The ion channel and GPCR toolkit of brain capillary pericytes. Front 195. Ishitsuka T, Iadecola C, Underwood MD, Reis DJ. Lesions of nucleus
Cell Neurosci 14: 601324, 2020. doi:10.3389/fncel.2020.601324. tractus solitarii globally impair cerebrovascular autoregulation. Am J
Physiol Heart Circ Physiol 251: H269–H281, 1986. doi:10.1152/
180. Rungta RL, Chaigneau E, Osmanski BF, Charpak S. Vascular com-
ajpheart.1986.251.2.H269.
partmentalization of functional hyperemia from the synapse to the
pia. Neuron 99: 362–375, 2018. doi:10.1016/j.neuron.2018.06.012. 196. Liu Y, Harder DR, Lombard JH. Interaction of myogenic mechanisms
and hypoxic dilation in rat middle cerebral arteries. Am J Physiol
181. Gonzalez-Marrero I, Hernandez-Abad LG, Castaneyra-Ruiz L,
Heart Circ Physiol 283: H2276–H2281, 2002. doi:10.1152/ajpheart.
Carmona-Calero EM, Castaneyra-Perdomo A. Changes in the cho-
00635.2002.
roid plexuses and brain barriers associated with high blood pres-
sure and ageing. Neurologia S0213-4853(18)30175-0, 2018. 197. Panerai RB. System identification of human cerebral blood flow reg-
doi:10.1016/j.nrl.2018.06.001. ulatory mechanisms. Cardiovasc Eng 4: 59–71, 2004. doi:10.1023/B:
182. Mayhan WG, Heistad DD. Role of veins and cerebral venous pres- CARE.0000025123.43747.e1.
sure in disruption of the blood-brain barrier. Circ Res 59: 216–220,
198. Panerai RB, Hanby MF, Robinson TG, Haunton VJ. Alternative repre-
1986. doi:10.1161/01.RES.59.2.216.
sentation of neural activation in multivariate models of neurovascu-
183. Auer L. Origin and localization of Evans blue extravasations in lar coupling in humans. J Neurophysiol 122: 833–843, 2019. doi:10.
acutely-induced hypertension in cats. Eur Neurol 17: 211–215, 1978. 1152/jn.00175.2019.
doi:10.1159/000114947.
199. Payne SJ. Identifying the myogenic and metabolic components of
184. Wilhelm I, Nyul-Toth A, Suciu M, Hermenean A, Krizbai IA. cerebral autoregulation. Med Eng Phys 58: 23–30, 2018. doi:10.
Heterogeneity of the blood-brain barrier. Tissue Barriers 4: 1016/j.medengphy.2018.04.018.
e1143544, 2016. doi:10.1080/21688370.2016.1143544.
200. Ainslie PN, Brassard P. Why is the neural control of cerebral autore-
185. Spence JD. Blood pressure gradients in the brain: their importance gulation so controversial? F1000Prime Rep 6: 14, 2014. doi:10.
to understanding pathogenesis of cerebral small vessel disease. 12703/P6-14.
Brain Sci 9: 21, 2019. doi:10.3390/brainsci9020021.
201. Bernardi L, Cencetti S, Cipriani M, Fattorini L, Bandinelli G,
186. Blanco PJ, Muller LO, Spence JD. Blood pressure gradients in cere- WdowczyckSzulc JL, Lagi A. Assessment of autonomic control of
bral arteries: A clue to pathogenesis of cerebral small vessel dis- cerebral blood flow: comparison with peripheral circulation.
ease. Stroke Vasc Neurol 2: 108–117, 2017. doi:10.1136/svn-2017- Cerebrovasc Dis 7: 303–304, 1997.
000087.
202. Goadsby PJ. Autonomic nervous system control of the cerebral
187. Grutzendler J, Nedergaard M. Cellular control of brain capillary circulation. In: Handbook of Clinical Neurology Autonomic
blood flow: in vivo imaging veritas. Trends Neurosci 42: 528–536, Nervous System, edited by Buuijs RM, Swaab DF. Amsterdam,
2019. doi:10.1016/j.tins.2019.05.009. The Netherlands: Elsevier, 2013, p. 193–201.

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1543


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

203. Hamner JW, Tan CO, Lee K, Cohen MA, Taylor JA. Sympathetic con- 220. Kety SS, Schmidt CF. The effects of altered arterial tensions of car-
trol of the cerebral vasculature in humans. Stroke 41: 102–109, 2010. bon dioxide and oxygen on cerebral blood flow and cerebral oxy-
doi:10.1161/STROKEAHA.109.557132. gen consumption of normal young men. J Clin Invest 27: 484–492,
1948. doi:10.1172/JCI101995.
204. Zhang R, Crandall CG, Levine BD. Cerebral hemodynamics during
the Valsalva maneuver: Insights from ganglionic blockade. Stroke 221. Reivich M. Arterial Pco2 and cerebral hemodynamics. Am J Physiol
35: 843–847, 2004. doi:10.1161/01.STR.0000120309.84666.AE. 206: 25–35, 1964. doi:10.1152/ajplegacy.1964.206.1.25.

205. Zhang R, Zuckerman JH, Iwasaki K, Wilson TE, Crandall CG, 222. Ide K, Poulin MJ. A protocol to study the relationship between mid-
Levine BD. Autonomic neural control of dynamic cerebral autore- dle cerebral artery blood velocity and end-tidal Pco2 in the hypo-
gulation in humans. Circulation 106: 1814–1820, 2002. doi:10.1161/ capnic-hypercapnic range in humans. FASEB J 17: A872–A872,
01.CIR.0000031798.07790.FE. 2003.

206. Ngai AC, Winn HR. Modulation of cerebral arteriolar diameter by in- 223. Minhas JS, Panerai RB, Robinson TG. Modelling the cerebral hae-
traluminal flow and pressure. Circ Res 77: 832–840, 1995. doi:10. modynamic response in the physiological range of PaCO2. Physiol
1161/01.res.77.4.832. Meas 39: 065001, 2018. doi:10.1088/1361-6579/aac76b.

207. White RP, Vallance P, Markus HS. Effect of inhibition of nitric oxide 224. Dahl A, Russell D, Nyberg-Hansen R, Rootwelt K, Mowinckel P.
synthase on dynamic cerebral autoregulation in humans. Clin Sci Simultaneous assessment of vasoreactivity using transcranial
(Lond) 99: 555–560, 2000. doi:10.1042/CS20000064, 10.1042/ Doppler ultrasound and cerebral blood flow in healthy subjects. J
cs0990555. doi:10.1042/CS20000064. Cereb Blood Flow Metab 14: 974–981, 1994. doi:10.1038/jcbfm.
1994.130.
208. Buxton RB, Uludağ K, Dubowitz DJ, Liu TT. Modeling the hemody-
225. Edwards MR, Lin DC, Hughson RL. Modeling the interaction
namic response to brain activation. Neuroimage 23, Suppl 1: S220–
between perfusion pressure and CO2 on cerebral blood flow. Front
233, 2004. doi:10.1016/j.neuroimage.2004.07.013.
Modeling Control Breathing 499: 285–290, 2001. doi:10.1007/978-
209. Gotoh J, Kuang TY, Nakao Y, Cohen DM, Melzer P, Itoh Y, Pak H, 1-4615-1375-9_45.
Pettigrew K, Sokoloff L. Regional differences in mechanisms of cere-
226. Hetzel A, Braune S, Guschlbauer B, Dohms K. CO2 reactivity testing
bral circulatory response to neuronal activation. Am J Physiol Heart
without blood pressure monitoring? Stroke 30: 398–401, 1999.
Circ Physiol 280: H821–H829, 2001. doi:10.1152/ajpheart.2001.
doi:10.1161/01.str.30.2.398.
280.2.H821.
227. Ainslie PN, Ashmead JC, Ide K, Morgan BJ, Poulin MJ. Differential
210. Raichle ME. Functional brain imaging and human brain function. J
responses to CO2 and sympathetic stimulation in the cerebral and
Neurosci 23: 3959–3962, 2003. doi:10.1523/JNEUROSCI.23-10- femoral circulations in humans. J Physiol 566: 613–624, 2005.
03959.2003. doi:10.1113/jphysiol.2005.087320.
211. Sokoloff L. Energetic of functional activation in neural tissues. 228. Kontos HA, Richardson DW, Patterson JL Jr. Effects of hypercapnia
Neurochem Res 24: 321–329, 1999. doi:10.1023/A:1022534709672. on human forearm blood vessels. Am J Physiol 212: 1070–1080,
1967. doi:10.1152/ajplegacy.1967.212.5.1070.
212. Raichle ME, Mintun MA. Brain work and brain imaging. Annu Rev
Neurosci 29: 449–476, 2006. doi:10.1146/annurev.neuro.29.051605. 229. Raper AJ, Richardson DW, Kontos HA, Patterson JL Jr. Circulatory
112819. responses to breath holding in man. J Appl Physiol 22: 201–206,
1967. doi:10.1152/jappl.1967.22.2.201.
213. Attwell D, Iadecola C. The neural basis of functional brain imaging
signals. Trends Neurosci 25: 621–625, 2002. doi:10.1016/s0166- 230. Faraci FM, Brian JE Jr. Nitric oxide and the cerebral circulation.
2236(02)02264-6. Stroke 25: 692–703, 1994. doi:10.1161/01.STR.25.3.692.
214. Fujii K, Heistad DD, Faraci FM. Flow-mediated dilatation of the basi- 231. Golding EM, Steenberg ML, Contant CF Jr, Krishnappa I, Robertson
lar artery in vivo. Circ Res 69: 697–705, 1991. doi:10.1161/01.res.69. CS, Bryan RM Jr. Cerebrovascular reactivity to CO2 and hypotension
3.697. after mild cortical impact injury. Am J Physiol Heart Circ Physiol
277: H1457–H1466, 1999. doi:10.1152/ajpheart.1999.277.4.H1457.
215. Iadecola C, Yang G, Ebner TJ, Chen G. Local and propagated vascu-
lar responses evoked by focal synaptic activity in cerebellar cortex. 232. Harris AP, Ohata H, Koehler RC. Role of nitric oxide in cerebrovascu-
J Neurophysiol 78: 651–659, 1997. doi:10.1152/jn.1997.78.2.651. lar reactivity to NMDA and hypercapnia during prenatal develop-
ment in sheep. Int J Dev Neurosci 26: 47–55, 2008. doi:10.1016/j.
216. Aaslid R. Visually evoked dynamic blood flow response of the ijdevneu.2007.08.011.
human cerebral circulation. Stroke 18: 771–775, 1987. doi:10.1161/01.
STR.18.4.771. 233. Helou SM, Hudak ML, Jones MD Jr. Cerebral blood flow response to
hypercapnia in immature fetal sheep. Am J Physiol Heart Circ
217. O'Herron P, Chhatbar PY, Levy M, Shen Z, Schramm AE, Lu Z, Kara Physiol 261: H1366–H1370, 1991. doi:10.1152/ajpheart.1991.261.5.
P. Neural correlates of single-vessel haemodynamic responses in H1366.
vivo. Nature 534: 378–382, 2016. doi:10.1038/nature17965.
234. Iadecola C, Zhang F. Permissive and obligatory roles of NO in cere-
218. Ainslie PN, Duffin J. Integration of cerebrovascular CO2 reactivity brovascular responses to hypercapnia and acetylcholine. Am J
and chemoreflex control of breathing: mechanisms of regulation, Physiol Regul Integr Comp Physiol 271: R990–R1001, 1996.
measurement, and interpretation. Am J Physiol Regul Integr Comp doi:10.1152/ajpregu.1996.271.4.R990.
Physiol 296: R1473–R1495, 2009. doi:10.1152/ajpregu.91008.2008.
235. Kidoguchi K, Tamaki M, Mizobe T, Koyama J, Kondoh T, Kohmura E,
219. Greenberg JH, Alavi A, Reivich M, Kuhl D, Uzzell B. Local cerebral Sakurai T, Yokono K, Umetani K. In vivo X-ray angiography in the
blood volume response to carbon dioxide in man. Circ Res 43: mouse brain using synchrotron radiation. Stroke 37: 1856–1861,
324–331, 1978. doi:10.1161/01.res.43.2.324. 2006. doi:10.1161/01.STR.0000226904.96059.a6.

1544 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

236. Massik J, Jones MD Jr, Miyabe M, Tang YL, Hudak ML, Koehler RC, neural mediation of hypoxia-induced cerebral vasodilation. In:
Traystman RJ. Hypercapnia and response of cerebral blood flow to Cerebral Blood Flow: Effects of Nerves and Neurotransmitters,
hypoxia in newborn lambs. J Appl Physiol (1985) 66: 1065–1070, edited by Heistad DD, Marcus ML. Amsterdam, The Natherlands:
1989. doi:10.1152/jappl.1989.66.3.1065. Elsevier North Holland, Inc., 1982, p. 463–471.
237. Coverdale NS, Badrov MB, Shoemaker JK. Impact of age on cere- 251. Fortune JB, Feustel PJ, deLuna C, Graca L, Hasselbarth J, Kupinski
brovascular dilation versus reactivity to hypercapnia. J Cereb Blood AM. Cerebral blood flow and blood volume in response to O2 and
Flow Metab 37: 344–355, 2017. doi:10.1177/0271678X15626156. CO2 changes in normal humans. J Trauma 39: 463–471, 1995.
238. Hotta H, Masamoto K, Uchida S, Sekiguchi Y, Takuwa H, Kawaguchi doi:10.1097/00005373-199509000-00012.
H, Shigemoto K, Sudo R, Tanishita K, Ito H, Kanno I. Layer-specific
252. Hoiland RL, Bain AR, Rieger MG, Bailey DM, Ainslie PN. Hypoxemia,
dilation of penetrating arteries induced by stimulation of the nucleus
oxygen content, and the regulation of cerebral blood flow. Am J
basalis of Meynert in the mouse frontal cortex. J Cereb Blood Flow
Physiol Regul Integr Comp Physiol 310: R398–R413, 2016.
Metab 33: 1440–1447, 2013. doi:10.1038/jcbfm.2013.92.
doi:10.1152/ajpregu.00270.2015.
239. Oizumi XS, Akisaki T, Kouta Y, Song XZ, Takata T, Kondoh T,
Umetani K, Hirano M, Yamasaki K, Kohmura E, Yokono K, Sakurai T. 253. Hudetz AG. Regulation of oxygen supply in the cerebral circulation.
Impaired response of perforating arteries to hypercapnia in chronic In: Oxygen Transport to Tissue XIX, edited by Harrison D. New
hyperglycemia. Kobe J Med Sci 52: 27–35, 2006. York: Plenum Press, 1997, pp. 513–520.

240. Verbree J, Bronzwaer AS, Ghariq E, Versluis MJ, Daemen MJ, van 254. Ogoh S, Nakahara H, Ainslie PN, Miyamoto T. The effect of oxygen
Buchem MA, Dahan A, van Lieshout JJ, van Osch MJ. Assessment on dynamic cerebral autoregulation: critical role of hypocapnia. J
of middle cerebral artery diameter during hypocapnia and hyper- Appl Physiol (1985) 108: 538–543, 2010. doi:10.1152/japplphysiol.
capnia in humans using ultra-high-field MRI. J Appl Physiol (1985) 01235.2009.
117: 1084–1089, 2014. doi:10.1152/japplphysiol.00651.2014.
255. Poulin MJ, Liang PJ, Robbins PA. Dynamics of the cerebral blood
241. Wei EP, Kontos HA, Patterson JL Jr. Dependence of pial arteriolar flow response to step changes in end-tidal Pco2 and Po2 in humans.
response to hypercapnia on vessel size. Am J Physiol 238: 697– J Appl Physiol (1985) 81: 1084–1095, 1996. doi:10.1152/jappl.
703, 1980. 1996.81.3.1084.
242. Willie CK, Macleod DB, Shaw AD, Smith KJ, Tzeng YC, Eves ND, 256. Rosenberg AA, Narayanan V, Jones MD Jr. Comparison of anterior
Ikeda K, Graham J, Lewis NC, Day TA, Ainslie PN. Regional brain cerebral artery blood flow velocity and cerebral blood flow during
blood flow in man during acute changes in arterial blood gases. J hypoxia. Pediatr Res 19: 67–70, 1985. doi:10.1203/00006450-
Physiol 590: 3261–3275, 2012. doi:10.1113/jphysiol.2012.228551.
198501000-00018.
243. Kontos HA, Raper AJ, Patterson JL. Analysis of vasoactivity of local
257. Ursino M, Magosso E. Acute cardiovascular response to isocapnic
pH, Pco2, and bicarbonate on pial vessels. Stroke 8: 358–360,
hypoxia. I. A mathematical model. Am J Physiol Heart Circ Physiol
1977. doi:10.1161/01.str.8.3.358.
279: H149–H165, 2000. doi:10.1152/ajpheart.2000.279.1.H149.
244. Rostrup E, Knudsen GM, Law I, Holm S, Larsson HB, Paulson OB. The
relationship between cerebral blood flow and volume in humans. 258. Jones MD Jr, Traystman RJ, Simmons MA, Molteni RA. Effects of
Neuroimage 24: 1–11, 2005. doi:10.1016/j.neuroimage.2004.09.043. changes in arterial O2 content on cerebral blood flow in the lamb.
Am J Physiol Heart Circ Physiol 240: H209–H215, 1981.
245. Liu P, De Vis JB, Lu H. Cerebrovascular reactivity (CVR) MRI with doi:10.1152/ajpheart.1981.240.2.H209.
CO2 challenge: a technical review. Neuroimage 187: 104–115, 2019.
doi:10.1016/j.neuroimage.2018.03.047. 259. Phillis JW. Adenosine and adenine nucleotides as regulators of cere-
bral blood flow: roles of acidosis, cell swelling, and KATP channels.
246. Hurn PD, Traystman RJ. Changes in arterial gas tension. In: Crit Rev Neurobiol 16: 237–270, 2004. doi:10.1615/CritRevNeurobiol.
Cerebral Blood Flow and Metabolism, edited by Edvinsson LK. v16.i4.20.
New York: Lippincott Williams & Wilkens, 2001, p. 384–394.
260. Kontos HA, Wei EP. Oxygen-dependent mechanisms in cerebral
247. Okamoto H, Hudetz AG, Roman RJ, Bosnjak ZJ, Kampine JP.
autoregulation. Ann Biomed Eng 13: 329–334, 1985. doi:10.1007/
Neuronal NOS-derived NO plays permissive role in cerebral blood
BF02584251.
flow response to hypercapnia. Am J Physiol Heart Circ Physiol
272: H559–H566, 1997. doi:10.1152/ajpheart.1997.272.1.H559. 261. Chan SL, Sweet JG, Bishop N, Cipolla MJ. Pial collateral reactivity
248. Faraci FM, Taugher RJ, Lynch C, Fan R, Gupta S, Wemmie JA. Acid- during hypertension and aging: Understanding the function of col-
sensing ion channels: Novel mediators of cerebral vascular laterals for stroke therapy. Stroke 47: 1618–1625, 2016. doi:10.1161/
responses. Circ Res 125: 907–920, 2019. doi:10.1161/CIRCRESAHA. STROKEAHA.116.013392.
119.315024.
262. Holm A, Heumann T, Augustin HG. Microvascular mural cell organo-
249. Wenzel J, Hansen CE, Bettoni C, Vogt MA, Lembrich B, Natsagdorj typic heterogeneity and functional plasticity. Trends Cell Biol 28:
R, Huber G, Brands J, Schmidt K, Assmann JC, Stolting I, Saar K, 302–316, 2018. doi:10.1016/j.tcb.2017.12.002.
Sedlacik J, Fiehler J, Ludewig P, Wegmann M, Feller N, Richter M,
Muller-Fielitz H, Walther T, Konig GM, Kostenis E, Raasch W, Hubner 263. Hill-Eubanks DC, Gonzales AL, Sonkusare SK, Nelson MT. Vascular
N, Gass P, Offermanns S, de Wit C, Wagner CA, Schwaninger M. TRP channels: performing under pressure and going with the flow.
Impaired endothelium-mediated cerebrovascular reactivity pro- Physiology (Bethesda) 29: 343–360, 2014. doi:10.1152/physiol.
motes anxiety and respiration disorders in mice. Proc Natl Acad Sci 00009.2014.
U S A 117: 1753–1761, 2020. doi:10.1073/pnas.1907467117.
264. Aydin F, Rosenblum WI, Povlishock JT. Myoendothelial junctions in
250. Aubineau P, Sercombe R, Seylaz J. Synergy of effects of PaO2 and human brain arterioles. Stroke 22: 1592–1597, 1991. doi:10.1161/01.
PaCO2 on local cerebral blood flow: Evidence against extrinsic STR.22.12.1592.

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1545


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

265. De Silva TM, Faraci FM. Contributions of aging to cerebral small ves- 281. Claassen JA, Jansen RW. Cholinergically mediated augmentation of
sel disease. Annu Rev Physiol 82: 275–295, 2020. doi:10.1146/ cerebral perfusion in Alzheimer's disease and related cognitive dis-
annurev-physiol-021119-034338. orders: the cholinergic-vascular hypothesis. J Gerontol A Biol Sci
Med Sci 61: 267–271, 2006. doi:10.1093/gerona/61.3.267.
266. Sobey CG, Faraci FM. Effect of nitric oxide and potassium channel
agonists and inhibitors on basilar artery diameter. Am J Physiol 282. Van Beek AH, Claassen JA. The cerebrovascular role of the cholin-
Heart Circ Physiol 272: H256–H262, 1997. doi:10.1152/ajpheart. ergic neural system in Alzheimer's disease. Behav Brain Res 221:
1997.272.1.H256. 537–542, 2011. doi:10.1016/j.bbr.2009.12.047.

267. Scha€Chinger V, Britten MB, Zeiher AM. Prognostic impact of coro- 283. ter Laan M, van Dijk JM, Elting JW, Staal MJ, Absalom AR.
nary vasodilator dysfunction on adverse long-term outcome of coro- Sympathetic regulation of cerebral blood flow in humans: a review.
nary heart disease. Circulation 101: 1899–1906, 2000. doi:10.1161/01. Br J Anaesth 111: 361–367, 2013. doi:10.1093/bja/aet122.
CIR.101.16.1899.
284. Hafkenschiel JH, Crumpton CW, Shenkin HA, Moyer JH, Zintel HA,
268. Widlansky ME, Gokce N, Keaney JF Jr, Vita JA. The clinical implica- Wendel H, Jeffers WA. The effects of twenty degree head-up tilt
tions of endothelial dysfunction. J Am Coll Cardiol 42: 1149–1160, upon the cerebral circulation of patients with arterial hypertension
2003. doi:10.1016/S0735-1097(03)00994-X. before and after sympathectomy. J Clin Invest 30: 793–798, 1951.
doi:10.1172/JCI102494.
269. Wardlaw JM, Smith C, Dichgans M. Mechanisms of sporadic cere-
bral small vessel disease: Insights from neuroimaging. Lancet 285. Jeng JS, Yip PK, Huang SJ, Kao MC. Changes in hemodynamics of
Neurol 12: 483–497, 2013. doi:10.1016/S1474-4422(13)70060-7. the carotid and middle cerebral arteries before and after endo-
scopic sympathectomy in patients with palmar hyperhidrosis: prelim-
270. Adib-Samii P, Rost N, Traylor M, Devan W, Biffi A, Lanfranconi S, et inary results. J Neurosurg 90: 463–467, 1999. doi:10.3171/jns.1999.
al. 17q25 Locus is associated with white matter hyperintensity vol- 90.3.0463.
ume in ischemic stroke, but not with lacunar stroke status. Stroke
44: 1609–1615, 2013. doi:10.1161/STROKEAHA.113.679936. 286. Suzuki J, Iwabuchi T, Hori S. Cervical sympathectomy for cerebral
vasospasm after aneurysm rupture. Neurol Med Chir (Tokyo) 15:
271. Jahshan S, Dayan L, Jacob G. Nitric oxide-sensitive guanylyl cyclase 41–50, 1975. doi:10.2176/nmc.15pt1.41.
signaling affects CO2-dependent but not pressure-dependent regu-
287. Edvinson L. Neurogenic mechanisms in the cerebrovascular bed.
lation of cerebral blood flow. Am J Physiol Regul Integr Comp
Autonomic nerves, amine receptors and their effects on cerebral
Physiol 312: R948–R955, 2017. doi:10.1152/ajpregu.00241.2016.
blood flow. Acta Physiol Scand Suppl 427: 1–35, 1975.
272. Zhang R, Wilson TE, Witkowski S, Cui J, Crandall GG, Levine BD.
288. Jordan J, Shannon JR, Diedrich A, Black B, Costa F, Robertson D,
Inhibition of nitric oxide synthase does not alter dynamic cerebral
Biaggioni I. Interaction of carbon dioxide and sympathetic nervous
autoregulation in humans. Am J Physiol Heart Circ Physiol 286:
system activity in the regulation of cerebral perfusion in humans.
H863–H869, 2004. doi:10.1152/ajpheart.00373.2003.
Hypertension 36: 383–388, 2000. doi:10.1161/01.HYP.36.3.383.
273. Armulik A, Genove G, Betsholtz C. Pericytes: developmental, physio-
289. Zhang R, Levine BD. Autonomic ganglionic blockade does not pre-
logical, and pathological perspectives, problems, and promises.
vent reduction in cerebral blood flow velocity during orthostasis in
Dev Cell 21: 193–215, 2011. doi:10.1016/j.devcel.2011.07.001.
humans. Stroke 38: 1238–1244, 2007. doi:10.1161/01.STR.0000260
274. Berthiaume AA, Hartmann DA, Majesky MW, Bhat NR, Shih AY. 095.94175.d0.
Pericyte structural remodeling in cerebrovascular health and home-
290. Kimmerly DS, Tutungi E, Wilson TD, Serrador JM, Gelb AW,
ostasis. Front Aging Neurosci 10: 210, 2018. doi:10.3389/fnagi.2018. Hughson RL, Shoemaker JK. Circulating norepinephrine and cere-
00210. brovascular control in conscious humans. Clin Physiol Funct
275. Uemura MT, Maki T, Ihara M, Lee VM, Trojanowski JQ. Brain micro- Imaging 23: 314–319, 2003. doi:10.1046/j.1475-0961.2003.00507.x.
vascular pericytes in vascular cognitive impairment and dementia. 291. Levine BD, Zhang R. Comments on Point:Counterpoint: Sympathetic ac-
Front Aging Neurosci 12: 80, 2020. doi:10.3389/fnagi.2020.00080. tivity does/does not influence cerebral blood flow. J Appl Physiol 105:
1369–1373, 2008. doi:10.1152/japplphysiol.zdg-8199.pcpcomm.2008.
276. Dalkara T. Pericytes: A novel target to improve success of recanaliza-
tion therapies. Stroke 50: 2985–2991, 2019. doi:10.1161/STROKEAHA. 292. van Lieshout JJ, Secher NH, Strandgaard S, Sigurdsson ST. Point:
118.023590. Counterpoint: Sympathetic activity does/does not influence cerebral
blood flow. J Appl Physiol 105: 1364–1368, 2008. doi:10.1152/
277. Yemisci M, Gursoy-Ozdemir Y, Vural A, Can A, Topalkara K, Dalkara
japplphysiol.90597.2008.
T. Pericyte contraction induced by oxidative-nitrative stress impairs
capillary reflow despite successful opening of an occluded cerebral 293. D'Alecy LG, Rose CJ. Parasympathetic cholinergic control of cere-
artery. Nat Med 15: 1031–1037, 2009. doi:10.1038/nm.2022. bral blood flow in dogs. Circ Res 41: 324–331, 1977. doi:10.1161/01.
res.41.3.324.
278. Faraci FM, Mayhan WG, Werber AH, Heistad DD. Cerebral circula-
tion: effects of sympathetic nerves and protective mechanisms dur- 294. Scremin OU, Rovere AA, Raynald AC, Giardini A. Cholinergic control
ing hypertension. Circulation Res 61: 102–106, 1987. of blood flow in the cerebral cortex of the rat. Stroke 4: 233–239,
1973. doi:10.1161/01.STR.4.2.232.
279. Heistad DD. Protection of cerebral vessels by sympathetic nerves.
Physiologist 23: 44–49, 1980. 295. Hamner JW, Tan CO, Tzeng YC, Taylor JA. Cholinergic control of
the cerebral vasculature in humans. J Physiol 590: 6343–6352,
280. Parati G, Mancia G, Rienzo MD, Castiglioni P. Point:Counterpoint: 2012. doi:10.1113/jphysiol.2012.245100.
Cardiovascular variability is/is not an index of autonomic control of
circulation. J Appl Physiol 101: 676–682, 2006. doi:10.1152/ 296. Seifert T, Fisher JP, Young CN, Hartwich D, Ogoh S, Raven PB,
japplphysiol.00446.2006. Fadel PJ, Secher NH. Glycopyrrolate abolishes the exercise-

1546 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

induced increase in cerebral perfusion in humans. Exp Physiol 95: asymptomatic carotid artery stenosis in the general population: An
1016–1025, 2010. doi:10.1113/expphysiol.2010.054346. individual participant data meta-analysis. Stroke 41: 1294–1297,
2010. doi:10.1161/STROKEAHA.110.581058.
297. Lecrux C, Hamel E. Neuronal networks and mediators of cortical
neurovascular coupling responses in normal and altered brain 311. Lorenz MW, Schaefer C, Steinmetz H, Sitzer M. Is carotid intima
states. Philos Trans R Soc Lond B Biol Sci 371: 20150350, 2016. media thickness useful for individual prediction of cardiovascular
doi:10.1098/rstb.2015.0350. risk? Ten-year results from the Carotid Atherosclerosis Progression
Study (CAPS). Eur Heart J 31: 2041–2048, 2010. doi:10.1093/
298. Vaucher E, Hamel E. Cholinergic basal forebrain neurons project to eurheartj/ehq189.
cortical microvessels in the rat: Electron microscopic study with an-
terogradely transported Phaseolus vulgaris leucoagglutinin and 312. Seneviratne A, Hulsmans M, Holvoet P, Monaco C. Biomechanical
choline acetyltransferase immunocytochemistry. J Neurosci 15: factors and macrophages in plaque stability. Cardiovasc Res 99:
7427–7441, 1995. doi:10.1523/JNEUROSCI.15-11-07427.1995. 284–293, 2013. doi:10.1093/cvr/cvt097.
299. Ashina H, Schytz HW, Ashina M. CGRP in human models of mi- 313. Moroni F, Ammirati E, Magnoni M, D'Ascenzo F, Anselmino M,
graine. Handb Exp Pharmacol 255: 109–120, 2019. doi:10.1007/ Anzalone N, Rocca MA, Falini A, Filippi M, Camici PG. Carotid ather-
164_2018_128. osclerosis, silent ischemic brain damage and brain atrophy: A sys-
tematic review and meta-analysis. Int J Cardiol 223: 681–687, 2016.
300. Ashina M, Hansen JM, Do TP, Melo-Carrillo A, Burstein R, Moskowitz
doi:10.1016/j.ijcard.2016.08.234.
MA. Migraine and the trigeminovascular system–40 years and
counting. Lancet Neurol 18: 795–804, 2019. doi:10.1016/S1474-4422 314. Carcaillon L, Plichart M, Zureik M, Rouaud O, Majed B, Ritchie K,
(19)30185-1. Tzourio C, Dartigues JF, Empana JP. Carotid plaque as a predictor
of dementia in older adults: The Three-City Study. Alzheimers
301. Moskowitz MA, Wei EP, Saito K, Kontos HA. Trigeminalectomy modi-
Dement 11: 239–248, 2015. doi:10.1016/j.jalz.2014.07.160.
fies pial arteriolar responses to hypertension or norepinephrine. Am
J Physiol Heart Circ Physiol 255: H1–H6, 1988. doi:10.1152/ 315. Gutierrez J, Guzman V, Khasiyev F, Manly J, Schupf N, Andrews H,
ajpheart.1988.255.1.H1. Mayeux R, Brickman AM. Brain arterial dilatation and the risk of
Alzheimer's disease. Alzheimers Dement 15: 666–674, 2019.
302. Sakas DE, Moskowitz MA, Wei EP, Kontos HA, Kano M, Ogilvy CS.
doi:10.1016/j.jalz.2018.12.018.
Trigeminovascular fibers increase blood flow in cortical gray matter
by axon reflex-like mechanisms during acute severe hypertension 316. Mutsaerts HJ, Palm-Meinders IH, de Craen AJ, Reiber JH, Blauw GJ,
or seizures. Proc Natl Acad Sci U S A 86: 1401–1405, 1989. doi:10. van Buchem MA, van der Grond J, Box FM, PROSPER Study Group.
1073/pnas.86.4.1401. Diastolic carotid artery wall shear stress is associated with cerebral
303. Green DJ, Hopman MT, Padilla J, Laughlin MH, Thijssen DH. infarcts and periventricular white matter lesions. Stroke 42: 3497–
Vascular adaptation to exercise in humans: Role of hemodynamic 3501, 2011. doi:10.1161/STROKEAHA.111.614453.
stimuli. Physiol Rev 97: 495–528, 2017. doi:10.1152/physrev.00014. 317. Liu Z, Zhao Y, Wang X, Zhang H, Cui Y, Diao Y, Xiu J, Sun X, Jiang
2016. G. Low carotid artery wall shear stress is independently associated
304. Newcomer SC, Thijssen DH, Green DJ. Effects of exercise on endo- with brain white-matter hyperintensities and cognitive impairment in
thelium and endothelium/smooth muscle cross talk: role of exercise- older patients. Atherosclerosis 247: 78–86, 2016. doi:10.1016/j.
induced hemodynamics. J Appl Physiol (1985) 111: 311–320, 2011. atherosclerosis.2016.02.003.
doi:10.1152/japplphysiol.00033.2011.
318. Zhang H, Liu H, Dong Y, Wang J, Zhao Y, Cui Y, Chai Q, Liu Z. Low
305. Tinken TM, Thijssen DH, Hopkins N, Black MA, Dawson EA, Minson carotid wall shear stress independently accelerates the progression
CT, Newcomer SC, Laughlin MH, Cable NT, Green DJ. Impact of of cognitive impairment and white matter lesions in the elderly.
shear rate modulation on vascular function in humans. Hypertension Oncotarget 9: 11402–11413, 2018. doi:10.18632/oncotarget.23191.
54: 278–285, 2009. doi:10.1161/HYPERTENSIONAHA.109.134361.
319. Tarumi T, Ayaz KM, Liu J, Tseng BY, Parker R, Riley J, Tinajero C,
306. Tinken TM, Thijssen DH, Hopkins N, Dawson EA, Cable NT, Green Zhang R. Cerebral hemodynamics in normal aging: central artery
DJ. Shear stress mediates endothelial adaptations to exercise train- stiffness, wave reflection, and pressure pulsatility. J Cereb Blood
ing in humans. Hypertension 55: 312–318, 2010. doi:10.1161/ Flow Metabol 34: 971–978, 2014. doi:10.1038/jcbfm.2014.44.
HYPERTENSIONAHA.109.146282.
320. Zhao X, Zhao M, Amin-Hanjani S, Du X, Ruland S, Charbel FT. Wall
307. Schreuder TH, Green DJ, Hopman MT, Thijssen DH. Impact of retro- shear stress in major cerebral arteries as a function of age and gen-
grade shear rate on brachial and superficial femoral artery flow- der–a study of 301 healthy volunteers. J Neuroimaging 25: 403–
mediated dilation in older subjects. Atherosclerosis 241: 199–204, 407, 2015. doi:10.1111/jon.12133.
2015. doi:10.1016/j.atherosclerosis.2015.04.017.
321. Webb AJ, Simoni M, Mazzucco S, Kuker W, Schulz U, Rothwell PM.
308. Thijssen DH, Dawson EA, Tinken TM, Cable NT, Green DJ. Increased cerebral arterial pulsatility in patients with leukoaraiosis:
Retrograde flow and shear rate acutely impair endothelial func- Arterial stiffness enhances transmission of aortic pulsatility. Stroke
tion in humans. Hypertension 53: 986–992, 2009. doi:10.1161/ 43: 2631–2636, 2012. doi:10.1161/STROKEAHA.112.655837.
HYPERTENSIONAHA.109.131508.
322. Roher AE, Garami Z, Tyas SL, Maarouf CL, Kokjohn TA, Belohlavek
309. Thijssen DH, Schreuder TH, Newcomer SW, Laughlin MH, Hopman M, Vedders LJ, Connor D, Sabbagh MN, Beach TG, Emmerling MR.
MT, Green DJ. Impact of 2-weeks continuous increase in retrograde Transcranial Doppler ultrasound blood flow velocity and pulsatility
shear stress on brachial artery vasomotor function in young and index as systemic indicators for Alzheimer's disease. Alzheimers
older men. J Am Heart Assoc 4: e001968, 2015. doi:10.1161/jaha.115. Dement 7: 445–455, 2011. doi:10.1016/j.jalz.2010.09.002.
001968.
323. van Es AC, van der Flier WM, Box FM, Middelkoop HA, Westendorp
310. de Weerd M, Greving JP, Hedblad B, Lorenz MW, Mathiesen EB, RG, van Buchem MA, van der Grond J. Carotid and basilar artery
O'Leary DH, Rosvall M, Sitzer M, Buskens E, Bots ML. Prevalence of wall shear stress in Alzheimer's disease and mild cognitive

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1547


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

impairment. Dement Geriatr Cogn Disord 28: 220–224, 2009. microgravity. J Appl Physiol 81: 408–412, 1996. doi:10.1152/jappl.
doi:10. 1996.81.1.408.
1159/000237740.
339. Latham RD, Fanton JW, Vernalis MN, Gaffney FA, Crisman RP.
324. Tanaka M, Sakaguchi M, Miwa K, Okazaki S, Furukado S, Yagita Y, Central hemodynamics in a baboon model during microgravity
Mochizuki H, Kitagawa K. Basilar artery diameter is an independent induced by parabolic flight. Adv Space Res 14: 349–358, 1994.
predictor of incident cardiovascular events. Arterioscler Thromb doi:10.1016/0273-1177(94)90422-7.
Vasc Biol 33: 2240–2244, 2013. doi:10.1161/ATVBAHA.113.301467.
340. Klein T, Wollseiffen P, Sanders M, Claassen J, Carnahan H, Abeln V,
325. Cornwell WK 3rd, Tarumi T, Aengevaeren VL, Ayers C, Divanji P, Fu Vogt T, Struder HK, Schneider S. The influence of microgravity on
Q, Palmer D, Drazner MH, Meyer DM, Bethea BT, Hastings JL, cerebral blood flow and electrocortical activity. Exp Brain Res 237:
Fujimoto N, Shibata S, Zhang R, Markham DW, Levine BD. Effect of 1057–1062, 2019. doi:10.1007/s00221-019-05490-6.
pulsatile and nonpulsatile flow on cerebral perfusion in patients with
341. Ogoh S, Hirasawa A, Raven PB, Rebuffat T, Denise P, Lericollais R,
left ventricular assist devices. J Heart Lung Transplant 33: 1295–
Sugawara J, Normand H. Effect of an acute increase in central blood
1303, 2014. doi:10.1016/j.healun.2014.08.013.
volume on cerebral hemodynamics. Am J Physiol Regul Integr
326. Dawson EA, Secher NH, Dalsgaard MK, Ogoh S, Yoshiga CC, Comp Physiol 309: R902–R911, 2015. doi:10.1152/ajpregu.00137.
Gonzalez-Alonso J, Steensberg A, Raven PB. Standing up to the 2015.
challenge of standing: a siphon does not support cerebral blood
342. Waki H, Shimizu T, Katahira K, Nagayama T, Yamasaki M, Katsuda S.
flow in humans. Am J Physiol Regul Integr Comp Physiol 287:
Effects of microgravity elicited by parabolic flight on abdominal aor-
R911–R914, 2004. doi:10.1152/ajpregu.00196.2004.
tic pressure and heart rate in rats. J Appl Physiol 93: 1893–1899,
327. Gisolf J, Gisolf A, van Lieshout JJ, Karemaker JM. The siphon contro- 2002. doi:10.1152/japplphysiol.01064.2001.
versy: an integration of concepts and the brain as baffle. Am J
343. Klein T, Sanders M, Wollseiffen P, Carnahan H, Abeln V, Askew CD,
Physiol Regul Integr Comp Physiol 289: R627–R629, 2005.
Claassen JA, Schneider S. Transient cerebral blood flow responses
doi:10.1152/ajpregu.00709.2004.
during microgravity. Life Sci Space Res (Amst) 25: 66–71, 2020.
328. Hill L. The influence of the force of gravity on the circulation of doi:10.1016/j.lssr.2020.03.003.
the blood. J Physiol 18: 15–53, 1895. doi:10.1113/jphysiol.1895.
344. Lawley JS, Petersen LG, Howden EJ, Sarma S, Cornwell WK, Zhang
sp000556.
R, Whitworth LA, Williams MA, Levine BD. Effect of gravity and micro-
329. Munis JR, Lozada LJ. Giraffes, siphons, and starling resistors. gravity on intracranial pressure. J Physiol 595: 2115–2127, 2017.
Cerebral perfusion pressure revisited. J Neurosurg Anesthesiol 12: doi:10.1113/JP273557.
290–296, 2000. doi:10.1097/00008506-200007000-00029.
345. Pohl A, Cullen DJ. Cerebral ischemia during shoulder surgery in the
330. Hicks JW, Badeer HS. Siphon mechanism in collapsible tubes: appli- upright position: A case series. J Clin Anesth 17: 463–469, 2005.
cation to circulation of the giraffe head. Am J Physiol Regul Integr doi:10.1016/j.jclinane.2004.09.012.
Comp Physiol 256: R567–R571, 1989. doi:10.1152/ajpregu.1989.2 346. Ainslie PN, Cotter JD, George KP, Lucas S, Murrell C, Shave R,
56.2.R567. Thomas KN, Williams MJ, Atkinson G. Elevation in cerebral blood
331. Seymour RS. Cardiovascular physiology of dinosaurs. Physiology flow velocity with aerobic fitness throughout healthy human ageing.
(Bethesda) 31: 430–441, 2016. doi:10.1152/physiol.00016.2016. J Physiol 586: 4005–4010, 2008. doi:10.1113/jphysiol.2008.158279.

332. Seymour RS, Lillywhite HB. Why vascular siphons with sub-atmos- 347. Braz ID, Fluck D, Lip GY, Lundby C, Fisher JP. Impact of aerobic fit-
pheric pressures are physiologically impossible in sauropod dino- ness on cerebral blood flow and cerebral vascular responsiveness
saurs. J Exp Biol 219: 2078–2079, 2016. doi:10.1242/jeb.140988. to CO2 in young and older men. Scand J Med Sci Sports 27: 634–
642, 2017. doi:10.1111/sms.12674.
333. Lillywhite HB. Snakes, blood-circulation and gravity. Sci Am 259:
348. Leenders KL, Perani D, Lammertsma AA, Heather JD, Buckingham
92–98, 1988. doi:10.1038/scientificamerican1288-92.
P, Jones T, Healy MJ, Gibbs JM, Wise RJ, Hatazawa J, Herold S,
334. Gisolf J, van Lieshout JJ, van Heusden K, Pott F, Stok WJ, Beaney RP, Brooks DJ, Spinks T, Rhodes C, Frackowiak RS.
Karemaker JM. Human cerebral venous outflow pathway depends Cerebral blood flow, blood volume and oxygen utilization. Normal
on posture and central venous pressure. J Physiol 560: 317–327, values and effect of age. Brain 113: 27–47, 1990. doi:10.1093/brain/
2004. doi:10.1113/jphysiol.2004.070409. 113.1.27.

335. Gisolf J, Wilders R, Immink RV, van Lieshout JJ, Karemaker JM. Tidal 349. Xekardaki A, Rodriguez C, Montandon ML, Toma S, Tombeur E,
volume, cardiac output and functional residual capacity determine Herrmann FR, Zekry D, Lovblad KO, Barkhof F, Giannakopoulos P,
end-tidal CO2 transient during standing up in humans. J Physiol Haller S. Arterial spin labeling may contribute to the prediction of
554: 579–590, 2004. doi:10.1113/jphysiol.2003.056895. cognitive deterioration in healthy elderly individuals. Radiology 274:
490–499, 2015. doi:10.1148/radiol.14140680.
336. Van Lieshout JJ, Wieling W, Karemaker JM, Secher NH. Syncope,
cerebral perfusion, and oxygenation. J Appl Physiol (1985) 94: 350. De VJ, Peng SL, Chen X, Li Y, Liu P, Sur S, Rodrigue KM, Park DC, Lu
833–848, 2003. doi:10.1152/japplphysiol.00260.2002. H. Arterial-spin-labeling (ASL) perfusion MRI predicts cognitive func-
tion in elderly individuals: a 4-year longitudinal study. J Magn Reson
337. Wieling W, Van Lieshout JJ, Ten Harkel AD. Dynamics of circulatory Imaging 48: 449–458, 2018. doi:10.1002/jmri.25938.
adjustments to head-up tilt and tilt-back in healthy and sympatheti-
cally denervated subjects. Clin Sci (Lond) 94: 347–352, 1998. 351. Wolters FJ, Zonneveld HI, Hofman A, van der Lugt A, Koudstaal PJ,
doi:10.1042/cs0940347. Vernooij MW, Ikram MA, Heart-Brain Connection Collaborative
Research Group. Cerebral perfusion and the risk of dementia. A
338. Foldager N, Andersen TA, Jessen FB, Ellegaard P, Stadeager C, population-based study. Circulation 136: 719–728, 2017. doi:10.1161/
Videbaek R, Norsk P. Central venous pressure in humans during CIRCULATIONAHA.117.027448.

1548 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

352. Yew B, Nation DA, Alzheimer's Disease Neuroimaging Initiative. 367. Dineen NE, Brodie FG, Panerai RB, Robinson TG. Effects of ageing
Cerebrovascular resistance: effects on cognitive decline, cortical at- on cerebral autoregulation assessed during respiratory manoeu-
rophy, and progression to dementia. Brain 140: 2001–2017, 1987. vres. Age Ageing 40: 199–204, 2011. doi:10.1093/ageing/afq170.
doi:10.1093/brain/awx112.
368. Edlow BL, Kim MN, Durduran T, Zhou C, Putt ME, Yodh AG,
353. Boumezbeur F, Mason GF, de Graaf RA, Behar KL, Cline GW, Greenberg JH, Detre JA. The effects of healthy aging on cerebral
Shulman GI, Rothman DL, Petersen KF. Altered brain mitochondrial hemodynamic responses to posture change. Physiol Meas 31: 477–
metabolism in healthy aging as assessed by in vivo magnetic reso- 495, 2010. doi:10.1088/0967-3334/31/4/002.
nance spectroscopy. J Cereb Blood Flow Metab 30: 211–221, 2010.
doi:10.1038/jcbfm.2009.197. 369. Lipsitz LA, Mukai S, Hamner J, Gagnon M, Babikian V. Dynamic reg-
ulation of middle cerebral artery blood flow velocity in aging and
354. Amin-Hanjani S, Du X, Pandey DK, Thulborn KR, Charbel FT. Effect hypertension. Stroke 31: 1897–1903, 2000. doi:10.1161/01.STR.31.8.
of age and vascular anatomy on blood flow in major cerebral ves- 1897.
sels. J Cereb Blood Flow Metab 35: 312–318, 2015. doi:10.1038/
jcbfm.2014.203. 370. Xing CY, Tarumi T, Meijers RL, Turner M, Repshas J, Xiong L, Ding K,
Vongpatanasin W, Yuan LJ, Zhang R. Arterial pressure, heart rate,
355. Mander BA, Rao V, Lu B, Saletin JM, Lindquist JR, Ancoli-Israel S, and cerebral hemodynamics across the adult life span.
Jagust W, Walker MP. Prefrontal atrophy, disrupted NREM slow Hypertension 69: 712–720, 2017. doi:10.1161/HYPERTENSIONAHA.
waves and impaired hippocampal-dependent memory in aging. Nat 116.08986.
Neurosci 16: 357–364, 2013. doi:10.1038/nn.3324.
371. Tzeng YC, Lucas SJ, Atkinson G, Willie CK, Ainslie PN. Fundamental
356. Oh H, Madison C, Villeneuve S, Markley C, Jagust WJ. Association relationships between arterial baroreflex sensitivity and dynamic
of gray matter atrophy with age, b-amyloid, and cognition in aging. cerebral autoregulation in humans. J Appl Physiol (1985) 108: 1162–
Cereb Cortex 24: 1609–1618, 2014. doi:10.1093/cercor/bht017. 1168, 2010. doi:10.1152/japplphysiol.01390.2009.

357. Jefferson AL, Liu D, Gupta DK, Pechman KR, Watchmaker JM, 372. Maxwell JD. Novel Interventions to Improve Cerebral and
Gordon EA, Rane S, Bell SP, Mendes LA, Davis LT, Gifford KA, Peripheral Vascular Function. Liverpool, UK: Liverpool John
Hohman TJ, Wang TJ, Donahue MJ. Lower cardiac index levels Moores University, 2020.
relate to lower cerebral blood flow in older adults. Neurology 89:
2327–2334, 2017. doi:10.1212/WNL.0000000000004707. 373. Kastrup A, Dichgans J, Niemeier M, Schabet M. Changes of cerebro-
vascular CO2 reactivity during normal aging. Stroke 29: 1311–1314,
358. Lu H, Xu F, Rodrigue KM, Kennedy KM, Cheng Y, Flicker B, Hebrank 1998. doi:10.1161/01.STR.29.7.1311.
AC, Uh J, Park DC. Alterations in cerebral metabolic rate and blood
supply across the adult lifespan. Cereb Cortex 21: 1426–1434, 2011. 374. Wiesmann M, Kiliaan AJ, Claassen JA. Vascular aspects of cognitive
doi:10.1093/cercor/bhq224. impairment and dementia. J Cereb Blood Flow Metab 33: 1696–
1706, 2013. doi:10.1038/jcbfm.2013.159.
359. Chen JJ, Rosas HD, Salat DH. Age-associated reductions in cerebral
blood flow are independent from regional atrophy. Neuroimage 55: 375. Shekhar S, Liu R, Travis OK, Roman RJ, Fan F. Cerebral autoregula-
468–478, 2011. doi:10.1016/j.neuroimage.2010.12.032. tion in hypertension and ischemic stroke: a mini review. J Pharm Sci
Exp Pharmacol 2017: 21–27, 2017.
360. Wahlin A, Nyberg L. At the heart of cognitive functioning in aging.
Trends Cogn Sci 23: 717–720, 2019. doi:10.1016/j.tics.2019.06.004. 376. Toth P, Tarantini S, Csiszar A, Ungvari Z. Functional vascular contri-
butions to cognitive impairment and dementia: mechanisms and
361. Czosnyka M, Balestreri M, Steiner L, Smielewski P, Hutchinson PJ, consequences of cerebral autoregulatory dysfunction, endothelial
Matta B, Pickard JD. Age, intracranial pressure, autoregulation, and impairment, and neurovascular uncoupling in aging. Am J Physiol
outcome after brain trauma. J Neurosurg 102: 450–454, 2005. Heart Circ Physiol 312: H1–H20, 2017. doi:10.1152/ajpheart.00581.
doi:10.3171/jns.2005.102.3.0450. 2016.
362. Oudegeest-Sander MH, van Beek AH, Abbink K, Olde Rikkert MG, 377. Strandgaard S. Autoregulation of cerebral blood flow in hyperten-
Hopman MT, Claassen JA. Assessment of dynamic cerebral autore- sive patients. The modifying influence of prolonged antihyperten-
gulation and cerebrovascular CO2 reactivity in ageing by measure- sive treatment on the tolerance to acute, drug-induced hypotension.
ments of cerebral blood flow and cortical oxygenation. Exp Physiol Circulation 53: 720–727, 1976. doi:10.1161/01.cir.53.4.720.
99: 586–598, 2014. doi:10.1113/expphysiol.2013.076455.
378. Strandgaard S, Paulson OB. Cerebral autoregulation. Stroke 15:
363. Aengevaeren VL, Claassen JA, Levine BD, Zhang R. Cardiac barore- 413–416, 1984. doi:10.1161/01.str.15.3.413.
flex function and dynamic cerebral autoregulation in elderly Masters
athletes. J Appl Physiol (1985) 114: 195–202, 2013. doi:10.1152/ 379. Strandgaard S, Paulson OB. Regulation of cerebral blood flow in
japplphysiol.00402.2012. health and disease. J Cardiovasc Pharmacol 19: S89–93, 1992.
doi:10.1097/00005344-199219006-00014.
364. Brodie FG, Panerai RB, Foster S, Evans DH, Robinson TG. Long-term
changes in dynamic cerebral autoregulation: a 10 years follow up 380. Strandgaard S, Paulson OB. Cerebral blood flow in untreated and
study. Clin Physiol Funct Imaging 29: 366–371, 2009. doi:10.1111/ treated hypertension. Neth J Med 47: 180–184, 1995. doi:10.1016/
j.1475-097X.2009.00880.x. 0300-2977(95)00065-u.

365. Carey BJ, Eames PJ, Blake MJ, Panerai RB, Potter JF. Dynamic cere- 381. Goodwin JS. Embracing complexity: a consideration of hypertension
bral autoregulation is unaffected by aging. Stroke 31: 2895–2900, in the very old. J Gerontol A Biol Sci Med Sci 58: 653–658, 2003.
2000. doi:10.1161/01.STR.31.12.2895. doi:10.1093/gerona/58.7.m653.

366. Carey BJ, Panerai RB, Potter JF. Effect of aging on dynamic cerebral 382. Rigaud AS, Forette B. Hypertension in older adults. J Gerontol A
autoregulation during head-up tilt. Stroke 34: 1871–1875, 2003. Biol Sci Med Sci 56: M217–225, 2001. doi:10.1093/gerona/56.4.
doi:10.1161/01.STR.0000081981.99908.F3. m217.

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1549


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

383. Sorond FA, Serrador JM, Jones RN, Shaffer ML, Lipsitz LA. The sit- patients. J Cardiovasc Pharmacol 12: S64–68, 1988. doi:10.1097/
to-stand technique for the measurement of dynamic cerebral autor- 00005344-198806124-00012.
egulation. Ultrasound Med Biol 35: 21–29, 2009. doi:10.1016/j.
ultrasmedbio.2008.08.001. 397. Ram CV, Meese R, Kaplan NM, Devous MD Sr, Bonte FJ, Forland
SC, Cutler RE. Antihypertensive therapy in the elderly. Effects on
384. Traon AP, Costes-Salon MC, Galinier M, Fourcade J, Larrue V. blood pressure and cerebral blood flow. Am J Med 82: 53–57,
Dynamics of cerebral blood flow autoregulation in hypertensive 1987. doi:10.1016/0002-9343(87)90145-8.
patients. J Neurol Sci 195: 139–144, 2002. doi:10.1016/s0022-510x
(02)00010-2. 398. Pandita-Gunawardena ND, Clarke SE. Amlodipine lowers blood
pressure without affecting cerebral blood flow as measured by sin-
385. Gemici K, Baran I, Bakar M, Demircan C, Ozdemir B, Cordan J. gle photon emission computed tomography in elderly hypertensive
Evaluation of the effect of the sublingually administered nifedipine subjects. Age Ageing 28: 451–457, 1999. doi:10.1093/ageing/28.5.
and captopril via transcranial doppler ultrasonography during hyper- 451.
tensive crisis. Blood Press 12: 46–48, 2003.
399. Patel RV, Ramadan NM, Levine SR, Welch KM, Fagan SC. Effects of
386. Eames PJ, Blake MJ, Panerai RB, Potter JF. Cerebral autoregulation ramipril and enalapril on cerebral blood flow in elderly patients with
indices are unimpaired by hypertension in middle aged and older asymptomatic carotid artery occlusive disease. J Cardiovasc
people. Am J Hypertens 16: 746–753, 2003. doi:10.1016/s0895- Pharmacol 28: 48–52, 1996. doi:10.1097/00005344-199607000-
7061(03)00947-6. 00008.

387. Serrador JM, Sorond FA, Vyas M, Gagnon M, Iloputaife ID, Lipsitz 400. Moonen JE, Foster-Dingley JC, de Ruijter W, van der Grond J,
LA. Cerebral pressure-flow relations in hypertensive elderly humans: Bertens AS, van Buchem MA, Gussekloo J, Middelkoop HA,
Transfer gain in different frequency domains. J Appl Physiol (1985) Wermer MJ, Westendorp RG, de Craen AJ, van der Mast RC.
98: 151–159, 2005. doi:10.1152/japplphysiol.00471.2004. Effect of discontinuation of antihypertensive treatment in elderly
people on cognitive functioning–the DANTE Study Leiden: a
388. Troisi E, Attanasio A, Matteis M, Bragoni M, Monaldo BC, randomized clinical trial. JAMA Intern Med 175: 1622–1630,
Caltagirone C, Silvestrini M. Cerebral hemodynamics in young hy- 2015. doi:10.1001/jamainternmed.2015.4103.
pertensive subjects and effects of atenolol treatment. J Neurol Sci
159: 115–119, 1998. doi:10.1016/S0022-510X(98)00147-6. 401. Immink RV, van den Born BJ, van Montfrans GA, Koopmans RP,
Karemaker JM, van Lieshout JJ. Impaired cerebral autoregulation in
389. Lipsitz LA, Gagnon M, Vyas M, Iloputaife I, Kiely DK, Sorond F, patients with malignant hypertension. Circulation 110: 2241–2245,
Serrador J, Cheng DM, Babikian V, Cupples LA. Antihypertensive 2004. doi:10.1161/01.CIR.0000144472.08647.40.
therapy increases cerebral blood flow and carotid distensibility in
hypertensive elderly subjects. Hypertension 45: 216–221, 2005. 402. Lipsitz LA, Habtemariam D, Gagnon M, Iloputaife I, Sorond F, Tchalla
doi:10.1161/01.HYP.0000153094.09615.11. AE, Dantoine TF, Travison TG. Reexamining the effect of antihyper-
tensive medications on falls in old age. Hypertension 66: 183–189,
390. Zhang R, Witkowski S, Fu Q, Claassen JA, Levine BD. Cerebral 2015. doi:10.1161/HYPERTENSIONAHA.115.05513.
hemodynamics after short- and long-term reduction in blood pres-
sure in mild and moderate hypertension. Hypertension 49: 1149– 403. Claassen J. Orthostatic hypotension in elderly patients. Ned
1155, 2007. doi:10.1161/HYPERTENSIONAHA.106.084939. Tijdschr Geneeskd 162: D1943, 2018.

391. Croall ID, Tozer DJ, Moynihan B, Khan U, O'Brien JT, Morris RG, 404. Kahlaee HR, Latt MD, Schneider CR. Association between chronic
Cambridge VC, Barrick TR, Blamire AM, Ford GA, Markus HS, Team or acute use of antihypertensive class of medications and falls in
PS, for the PRESERVE Study Team. Effect of standard vs intensive older adults. A systematic review and meta-analysis. Am J
blood pressure control on cerebral blood flow in small vessel dis- Hypertens 31: 467–479, 2018. doi:10.1093/ajh/hpx189.
ease: The PRESERVE randomized clinical trial. JAMA Neurol 75:
405. Group S, Williamson JD, Pajewski NM, Auchus AP, Bryan RN,
720–727, 2018. doi:10.1001/jamaneurol.2017.5153.
Chelune G, Cheung AK, Cleveland ML, et al. Effect of intensive vs
392. Tryambake D, He J, Firbank MJ, O’Brien JT, Blamire AM, Ford GA. standard blood pressure control on probable dementia: A random-
Intensive blood pressure lowering increases cerebral blood flow in ized clinical trial. JAMA 321: 553–561, 2019. doi:10.1001/jama.2018.
older subjects with hypertension. Hypertension 61: 1309–1315, 21442.
2013. doi:10.1161/HYPERTENSIONAHA.112.200972.
406. Pajewski NM, Berlowitz DR, Bress AP, Callahan KE, Cheung AK, Fine
393. Foster-Dingley JC, Moonen JE, de Craen AJ, de Ruijter W, van der LJ, Gaussoin SA, Johnson KC, King J, Kitzman DW, Kostis JB, Lerner
Mast RC, van der Grond J. Blood pressure is not associated with cer- AJ, Lewis CE, Oparil S, Rahman M, Reboussin DM, Rocco MV,
ebral blood flow in older persons. Hypertension 66: 954–960, Snyder JK, Still C, Supiano MA, Wadley VG, Whelton PK, Wright JT
2015. doi:10.1161/HYPERTENSIONAHA.115.05799. Jr, Williamson JD. Intensive vs standard blood pressure control in
adults 80 years or older: A secondary analysis of the systolic blood
394. Griffith DN, James IM, Newbury PA, Woollard ML. The effect of ß-ad- pressure intervention trial. J Am Geriatr Soc 68: 496–504, 2020.
renergic receptor blocking drugs on cerebral blood flow. Br J Clin doi:10.1111/jgs.16272.
Pharmacol 7: 491–494, 1979. doi:10.1111/j.1365-2125.1979.tb00991.x.
407. Collard D, Brouwer TF, Olde Engberink RH, Zwinderman AH, Vogt L,
395. Gommer ED, Martens EG, Aalten P, Shijaku E, Verhey FR, Mess WH, van den Born BJ. BH. Initial estimated glomerular filtration rate
Ramakers IH, Reulen JP. Dynamic cerebral autoregulation in sub- decline and long-term renal function during intensive antihyperten-
jects with Alzheimer’s disease, mild cognitive impairment, and con- sive therapy: A post hoc analysis of the SPRINT and ACCORD-BP
trols: evidence for increased peripheral vascular resistance with randomized controlled trials. Hypertension 75: 1205–1212, 2020.
possible predictive value. J Alzheimers Dis 30: 805–813, 2012. doi:10.1161/HYPERTENSIONAHA.119.14659.
doi:10.3233/JAD-2012-111628.
408. White WB, Wakefield DB, Moscufo N, Guttmann CR, Kaplan RF,
396. Conen D, Ruttimann S, Noll G, Schneider K, Muller J. Short- and Bohannon RW, Fellows D, Hall CB, Wolfson L. Effects of intensive
long-term cerebrovascular effects of nitrendipine in hypertensive versus standard ambulatory blood pressure control on

1550 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

cerebrovascular outcomes in older people (INFINITY). Circulation 424. Al-Jehani H, Angle M, Marcoux J, Teitelbaum J. Early abnormal tran-
140: 1626–1635, 2019. doi:10.1161/CIRCULATIONAHA.119.041603. sient hyperemic response test can predict delayed ischemic neuro-
logic deficit in subarachnoid hemorrhage. Crit Ultrasound J 10: 1,
409. Ding J, Davis-Plourde KL, Sedaghat S, Tully PJ, Wang W, Phillips C, 2018. doi:10.1186/s13089-017-0079-7.
Pase MP, Himali JJ, Gwen WB, Griswold M, Gottesman R, Mosley
TH, White L, Guethnason V, Debette S, Beiser AS, Seshadri S, Ikram 425. Brooks FA, Ughwanogho U, Henderson GV, Black-Schaffer R,
MA, Meirelles O, Tzourio C, Launer LJ. Antihypertensive medica- Sorond FA, Tan CO. The link between cerebrovascular hemody-
tions and risk for incident dementia and Alzheimer's disease: Ad namics and rehabilitation outcomes after aneurysmal subarachnoid
meta-analysis of individual participant data from prospective cohort hemorrhage. Am J Phys Med Rehabil 97: 309–315, 2018. doi:10.
studies. Lancet Neurol 19: 61–70, 2020. doi:10.1016/S1474-4422(19) 1097/PHM.0000000000000886.
30393-X.
426. Budohoski KP, Czosnyka M, Kirkpatrick PJ, Reinhard M, Varsos GV,
410. Aries MJ, Elting JW, De Keyser J, Kremer BP, Vroomen PC. Cerebral Kasprowicz M, Zabek M, Pickard JD, Smielewski P. Bilateral failure
autoregulation in stroke: a review of transcranial Doppler studies. of cerebral autoregulation is related to unfavorable outcome after
Stroke 41: 2697–2704, 2010. doi:10.1161/STROKEAHA.110.594168. subarachnoid hemorrhage. Neurocrit Care 22: 65–73, 2015.
doi:10.1007/s12028-014-0032-6.
411. Vitt JR, Trillanes M, Hemphill JC. Management of blood pressure
during and after recanalization therapy for acute ischemic stroke. 427. Budohoski KP, Czosnyka M, Smielewski P, Kasprowicz M, Helmy A,
Front Neurol 10: 138, 2019. doi:10.3389/fneur.2019.00138. Bulters D, Pickard JD, Kirkpatrick PJ. Impairment of cerebral autore-
gulation predicts delayed cerebral ischemia after subarachnoid
412. Castro P, Azevedo E, Sorond FA. Cerebral autoregulation in stroke. hemorrhage: A prospective observational study. Stroke 43: 3230–
Curr Atheroscler Rep 20: 37, 2018. doi:10.1007/s11883-018-0739-5. 3237, 2012. doi:10.1161/STROKEAHA.112.669788.
413. Guo ZN, Jin H, Sun H, Zhao Y, Liu J, Ma H, Sun X, Yang Y. 428. Calviere L, Nasr N, Arnaud C, Czosnyka M, Viguier A, Tissot B, Sol
Antioxidant melatonin: Potential functions in improving cerebral JC, Larrue V. Prediction of delayed cerebral ischemia after subarch-
autoregulation after subarachnoid hemorrhage. Front Physiol 9: noid hemorrhage using cerebral blood flow velocities and cerebral
1146, 2018. doi:10.3389/fphys.2018.01146. autoregulation assessment. Neurocrit Care 23: 253–258, 2015.
doi:10.1007/s12028-015-0125-x.
414. Intharakham K, Beishon L, Panerai RB, Haunton VJ, Robinson TG.
Assessment of cerebral autoregulation in stroke: a systematic 429. Fontana J, Moratin J, Ehrlich G, Scharf J, Weib C, Schmieder K, Barth
review and meta-analysis of studies at rest. J Cereb Blood Flow M. Dynamic autoregulatory response after aneurysmal subarachnoid
Metabol 39: 2105–2116, 2019. doi:10.1177/0271678X19871013. hemorrhage and its relation to angiographic vasospasm and clinical
outcome. Neurocrit Care 23: 355–363, 2015. doi:10.1007/s12028-
415. Jordan JD, Powers WJ. Cerebral autoregulation and acute ischemic
014-0104-7.
stroke. Am J Hypertens 25: 946–950, 2012. doi:10.1038/ajh.
2012.53. 430. Gaasch M, Schiefecker AJ, Kofler M, Beer R, Rass V, Pfausler B,
Thome C, Schmutzhard E, Helbok R. Cerebral autoregulation in the
416. Kunz A, Iadecola C. Cerebral vascular dysregulation in the ischemic
prediction of delayed cerebral ischemia and clinical outcome in poor-
brain. Handb Clin Neurol 92: 283–305, 2009. doi:10.1016/S0072-
grade aneurysmal subarachnoid hemorrhage patients. Crit Care Med
9752(08)01914-3.
46: 774–780, 2018. doi:10.1097/ccm.0000000000003016.
417. Minhas JS, Panerai RB, Ghaly G, Divall O, Robinson TG. Cerebral
431. Hattingen E, Blasel S, Dettmann E, Vatter H, Pilatus U, Seifert V,
autoregulation in hemorrhagic stroke: A systematic review and
Zanella FE, Weidauer S. Perfusion-weighted MRI to evaluate cere-
meta-analysis of transcranial Doppler ultrasonographic studies. J
bral autoregulation in aneurysmal subarachnoid haemorrhage.
Clin Ultrasound 47: 14–21, 2019. doi:10.1002/jcu.22645.
Neuroradiology 50: 929–938, 2008. doi:10.1007/s00234-008-
418. Palomares SM, Cipolla MJ. Myogenic tone as a therapeutic target 0424-4.
for ischemic stroke. Curr Vasc Pharmacol 12: 788–800, 2014.
432. Jaeger M, Soehle M, Schuhmann MU, Meixensberger J. Clinical sig-
doi:10.2174/15701611113116660155.
nificance of impaired cerebrovascular autoregulation after severe
419. Sobey CG, Faraci FM. Subarachnoid haemorrhage: what happens aneurysmal subarachnoid hemorrhage. Stroke 43: 2097–2101,
to the cerebral arteries? Clin Exp Pharmacol Physiol 25: 867–876, 2012. doi:10.1161/STROKEAHA.112.659888.
1998. doi:10.1111/j.1440-1681.1998.tb02337.x.
433. Lam JM, Smielewski P, Czosnyka M, Pickard JD, Kirkpatrick PJ.
420. Xiao M, Li Q, Feng H, Zhang L, Chen Y. Neural vascular mechanism Predicting delayed ischemic deficits after aneurysmal subarachnoid
for the cerebral blood flow autoregulation after hemorrhagic stroke. hemorrhage using a transient hyperemic response test of cerebral
Neural Plast 2017: 5819514, 2017. doi:10.1155/2017/5819514. autoregulation. Neurosurgery 47: 819–825, 2000. doi:10.1097/
00006123-200010000-00004.
421. Xiong L, Liu X, Shang T, Smielewski P, Donnelly J, Guo ZN, Yang Y,
Leung T, Czosnyka M, Zhang R, Liu J, Wong KS. Impaired cerebral 434. Lang EW, Diehl RR, Mehdorn HM. Cerebral autoregulation testing af-
autoregulation: Measurement and application to stroke. J Neurol ter aneurysmal subarachnoid hemorrhage: the phase relationship
Neurosurg Psychiatry 88: 520–531, 2017. doi:10.1136/jnnp-2016- between arterial blood pressure and cerebral blood flow velocity. Crit
314385. Care Med 29: 158–163, 2001. doi:10.1097/00003246-200101000-
00031.
422. Jackman K, Iadecola C. Neurovascular regulation in the ischemic
brain. Antioxid Redox Signal 22: 149–160, 2015. doi:10.1089/ars. 435. Ratsep T, Asser T. Cerebral hemodynamic impairment after aneur-
2013.5669. ysmal subarachnoid hemorrhage as evaluated using transcranial
Doppler ultrasonography: relationship to delayed cerebral ischemia
423. Markus HS. Transcranial Doppler ultrasound. Br Med Bull 56: 378– and clinical outcome. J Neurosurg 95: 393–401, 2001. doi:10.3171/
388, 2000. doi:10.1258/0007142001903021. jns.2001.95.3.0393.

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1551


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

436. Santos GA, Petersen N, Zamani AA, Du R, LaRose S, Monk A, size and outcome after ischemic stroke. Acta Neurol Scand 125:
Sorond FA, Tan CO. Pathophysiologic differences in cerebral autor- 156–162, 2012. doi:10.1111/j.1600-0404.2011.01515.x.
egulation after subarachnoid hemorrhage. Neurology 86: 1950–
1956, 2016. doi:10.1212/WNL.0000000000002696. 450. Meyer JS, Shimazu K, Fukuuchi Y, Ouchi T, Okamoto S, Koto A.
Impaired neurogenic cerebrovascular control and dysautoregula-
437. Soehle M, Czosnyka M, Pickard JD, Kirkpatrick PJ. Continuous tion after stroke. Stroke 4: 169–186, 1973. doi:10.1161/01.str.4.2.169.
assessment of cerebral autoregulation in subarachnoid hemor-
rhage. Anesth Analg 98: 1133–1139, 2004. doi:10.1213/01. 451. Dawson SL, Blake MJ, Panerai RB, Potter JF. Dynamic but not static
ane.0000111101.41190.99. cerebral autoregulation is impaired in acute ischaemic stroke.
Cerebrovasc Dis 10: 126–132, 2000. doi:10.1159/000016041.
438. Tseng MY, Czosnyka M, Richards H, Pickard JD, Kirkpatrick PJ.
Effects of acute treatment with pravastatin on cerebral vasospasm, 452. Eames PJ, Robinson TG, Panerai RB, Potter JF. The systemic hae-
autoregulation, and delayed ischemic deficits after aneurysmal sub- modynamic and cerebral autoregulatory effects of bendrofluazide in
arachnoid hemorrhage: a phase II randomized placebo-controlled the subacute post-stroke period. J Hypertens 22: 2017–2024,
trial. Stroke 36: 1627–1632, 2005. doi:10.1161/01.STR.0000176743. 2004. doi:10.1097/00004872-200410000-00026.
67564.5d.
453. Gierthmuhlen J, Allardt A, Sawade M, Baron R, Wasner G. Dynamic
439. Yundt KD, Grubb RL Jr, Diringer MN, Powers WJ. Autoregulatory va- cerebral autoregulation in stroke patients with a central sympathetic
sodilation of parenchymal vessels is impaired during cerebral vaso- deficit. Acta Neurol Scand 123: 332–338, 2011. doi:10.1111/j.1600-
spasm. J Cereb Blood Flow Metab 18: 419–424, 1998. doi:10.1097/ 0404.2010.01424.x.
00004647-199804000-00010.
454. Saeed NP, Panerai RB, Horsfield MA, Robinson TG. Does stroke
440. Zweifel C, Castellani G, Czosnyka M, Carrera E, Brady KM, subtype and measurement technique influence estimation of cere-
Kirkpatrick PJ, Pickard JD, Smielewski P. Continuous assessment of bral autoregulation in acute ischaemic stroke? Cerebrovasc Dis 35:
cerebral autoregulation with near-infrared spectroscopy in adults af- 257–261, 2013. doi:10.1159/000347075.
ter subarachnoid hemorrhage. Stroke 41: 1963–1968, 2010. doi:10.
1161/STROKEAHA.109.577320. 455. Kwan J, Lunt M, Jenkinson D. Assessing dynamic cerebral autoregu-
lation after stroke using a novel technique of combining transcranial
441. Llwyd O, Salinet AS, Panerai RB, Lam MY, Saeed NP, Brodie F, Bor- Doppler ultrasonography and rhythmic handgrip. Blood Press
Seng-Shu E, Robinson TG, Nogueira RC. Cerebral haemodynamics Monit 9: 3–8, 2004. doi:10.1097/00126097-200402000-00002.
following acute ischaemic stroke: effects of stroke severity and
stroke subtype. Cerebrovasc Dis Extra 8: 80–89, 2018. doi:10.1159/ 456. Gong X, Liu J, Dong P, Zhang P, Li N, Zhao X, Wang Y.
000487514. Assessment of dynamic cerebral autoregulation in patients with
basilar artery stenosis. PLoS One 8: e77802, 2013. doi:10.1371/
442. Powers WJ, Videen TO, Diringer MN, Aiyagari V, Zazulia AR. journal.pone.0077802.
Autoregulation after ischaemic stroke. J Hypertens 27: 2218–2222,
2009. doi:10.1097/HJH.0b013e328330a9a7. 457. Tutaj M, Miller M, Krakowska-Stasiak M, Piaôtek A, Hebda J, Laôtka
M, Strojny J, Szczudlik A, Słowik A. Dynamic cerebral autoregulation
443. Powers WJ, Zazulia AR, Videen TO, Adams RE, Yundt KD, Aiyagari
is compromissed in ischaemic stroke of undetermined aetiology
V, Grubb RL, Jr, Diringer MN. Autoregulation of cerebral blood flow
only in the non-affected side. Neurol Neurochir Pol 48: 91–97, 2014.
surrounding acute (6 to 22 hours) intracerebral hemorrhage.
doi:10.1016/j.pjnns.2013.12.006.
Neurology 57: 18–24, 2001. doi:10.1212/WNL.57.1.18.
458. Novak V, Yang AC, Lepicovsky L, Goldberger AL, Lipsitz LA, Peng
444. Reinhard M, Roth M, Guschlbauer B, Harloff A, Timmer J, Czosnyka M,
CK. Multimodal pressure-flow method to assess dynamics of cere-
Hetzel A. Dynamic cerebral autoregulation in acute ischemic stroke
bral autoregulation in stroke and hypertension. Biomed Eng Online
assessed from spontaneous blood pressure fluctuations. Stroke 36:
3: 39, 2004. doi:10.1186/1475-925X-3-39.
1684–1689, 2005. doi:10.1161/01.STR.0000173183.36331.ee.
459. Lam MY, Haunton VJ, Robinson TG, Panerai RB. Dynamic cerebral
445. Reinhard M, Wihler C, Roth M, Harloff A, Niesen WD, Timmer J,
autoregulation measurement using rapid changes in head position-
Weiller C, Hetzel A. Cerebral autoregulation dynamics in acute is-
chemic stroke after rtPA thrombolysis. Cerebrovasc Dis 26: 147– ing: Experiences in acute ischemic stroke and healthy control popu-
155, 2008. doi:10.1159/000139662. lations. Am J Physiol Heart Circ Physiol 316: H673–H683, 2019.
doi:10.1152/ajpheart.00550.2018.
446. Salinet AS, Silva NC, Caldas J, de Azevedo DS, de-Lima-Oliveira M,
Nogueira RC, Conforto AB, Texeira MJ, Robinson TG, Panerai RB, 460. Budohoski KP, Czosnyka M, Smielewski P, Varsos GV, Kasprowicz
Bor-Seng-Shu E. Impaired cerebral autoregulation and neurovascu- M, Brady KM, Pickard JD, Kirkpatrick PJ. Cerebral autoregulation af-
lar coupling in middle cerebral artery stroke: influence of severity? J ter subarachnoid hemorrhage: comparison of three methods. J
Cereb Blood Flow Metab 39: 2277–2285, 2019. doi:10.1177/ Cereb Blood Flow Metabol 33: 449–456, 2013. doi:10.1038/jcbfm.
0271678X18794835. 2012.189.

447. Truijen J, Rasmussen LS, Kim YS, Stam J, Stok WJ, Pott FC, van 461. Salinet AS, Robinson TG, Panerai RB. Effects of cerebral ischemia
Lieshout JJ. Cerebral autoregulatory performance and the cerebro- on human neurovascular coupling, CO2 reactivity, and dynamic cer-
vascular response to head-of-bed positioning in acute ischaemic ebral autoregulation. J Appl Physiol 118: 170–177, 2015. doi:10.1152/
stroke. Eur J Neurol 25: 1365–e1117, 2018. doi:10.1111/ene.13737. japplphysiol.00620.2014.

448. Ma H, Guo ZN, Sun X, Liu J, Lv S, Zhao L, Guo W, Jin H, Yang Y. 462. Eames PJ, Blake MJ, Dawson SL, Panerai RB, Potter JF. Dynamic
Hematoma volume is a predictive factor of disturbed autoregulation cerebral autoregulation and beat to beat blood pressure control are
after spontaneous intracerebral hemorrhage. J Neurol Sci 382: 96– impaired in acute ischaemic stroke. J Neurol Neurosurg Psychiatry
100, 2017. doi:10.1016/j.jns.2017.09.035. 72: 467–472, 2002. doi:10.1136/jnnp.72.4.467.

449. Reinhard M, Rutsch S, Lambeck J, Wihler C, Czosnyka M, Weiller C, 463. Immink RV, van Montfrans GA, Stam J, Karemaker JM, Diamant M,
Hetzel A. Dynamic cerebral autoregulation associates with infarct van Lieshout JJ. Dynamic cerebral autoregulation in acute lacunar

1552 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

and middle cerebral artery territory ischemic stroke. Stroke 36: with worse outcome after subarachnoid hemorrhage. Stroke 50:
2595–2600, 2005. doi:10.1161/01.STR.0000189624.06836.03. 2729–2737, 2019. doi:10.1161/STROKEAHA.119.026282.

464. Ma H, Guo ZN, Liu J, Xing Y, Zhao R, Yang Y. Temporal course of 477. Tseng MY, Czosnyka M, Richards H, Pickard JD, Kirkpatrick PJ.
dynamic cerebral autoregulation in patients with intracerebral hem- Effects of acute treatment with statins on cerebral autoregulation in
orrhage. Stroke 47: 674–681, 2016. doi:10.1161/STROKEAHA.115. patients after aneurysmal subarachnoid hemorrhage. Neurosurg
011453. Focus 21: E10, 2006. doi:10.3171/foc.2006.21.3.10.

465. Oeinck M, Neunhoeffer F, Buttler KJ, Meckel S, Schmidt B, 478. Aoi MC, Hu K, Lo MT, Selim M, Olufsen MS, Novak V. Impaired cere-
Czosnyka M, Weiller C, Reinhard M. Dynamic cerebral autoregula- bral autoregulation is associated with brain atrophy and worse func-
tion in acute intracerebral hemorrhage. Stroke 44: 2722–2728, tional status in chronic ischemic stroke. PLoS One 7: e46794, 2012.
2013. doi:10.1161/STROKEAHA.113.001913. doi:10.1371/journal.pone.0046794.

466. Otite F, Mink S, Tan CO, Puri A, Zamani AA, Mehregan A, Chou S, 479. Hu K, Lo MT, Peng CK, Liu Y, Novak V. A nonlinear dynamic
Orzell S, Purkayastha S, Du R, Sorond FA. Impaired cerebral autore- approach reveals a long-term stroke effect on cerebral blood flow
gulation is associated with vasospasm and delayed cerebral ische- regulation at multiple time scales. PLoS Comput Biol 8: e1002601,
mia in subarachnoid hemorrhage. Stroke 45: 677–682, 2014. 2012. doi:10.1371/journal.pcbi.1002601.
doi:10.1161/STROKEAHA.113.002630.
480. Hu K, Peng CK, Czosnyka M, Zhao P, Novak V. Nonlinear assess-
467. Reinhard M, Neunhoeffer F, Gerds TA, Niesen WD, Buttler KJ, ment of cerebral autoregulation from spontaneous blood pressure
Timmer J, Schmidt B, Czosnyka M, Weiller C, Hetzel A. Secondary and cerebral blood flow fluctuations. Cardiovasc Eng 8: 60–71,
decline of cerebral autoregulation is associated with worse outcome 2008. doi:10.1007/s10558-007-9045-5.
after intracerebral hemorrhage. Intensive Care Med 36: 264–271,
481. Hu K, Peng CK, Huang NE, Wu Z, Lipsitz LA, Cavallerano J, Novak V.
2010. doi:10.1007/s00134-009-1698-7.
Altered phase interactions between spontaneous blood pressure
468. Chen J, Liu J, Xu WH, Xu R, Hou B, Cui LY, Gao S. Impaired dynamic and flow fluctuations in type 2 diabetes mellitus: Nonlinear assess-
cerebral autoregulation and cerebrovascular reactivity in middle ment of cerebral autoregulation. Physica A 387: 2279–2292, 2008.
cerebral artery stenosis. PLoS One 9: e88232, 2014. doi:10.1371/ doi:10.1016/j.physa.2007.11.052.
journal.pone.0088232.
482. Gommer ED, Staals J, van Oostenbrugge RJ, Lodder J, Mess WH,
469. Guo ZN, Liu J, Xing Y, Yan S, Lv C, Jin H, Yang Y. Dynamic cerebral Reulen JP. Dynamic cerebral autoregulation and cerebrovascular
autoregulation is heterogeneous in different subtypes of acute is- reactivity: A comparative study in lacunar infarct patients. Physiol
chemic stroke. PLoS One 9: e93213, 2014. doi:10.1371/journal. Meas 29: 1293–1303, 2008. doi:10.1088/0967-3334/29/11/005.
pone.0093213. 483. Guo ZN, Xing Y, Wang S, Ma H, Liu J, Yang Y. Characteristics of
470. Panerai RB, Jara JL, Saeed NP, Horsfield MA, Robinson TG. dynamic cerebral autoregulation in cerebral small vessel disease:
Dynamic cerebral autoregulation following acute ischaemic stroke: diffuse and sustained. Sci Rep 5: 15269, 2015. doi:10.1038/
comparison of transcranial Doppler and magnetic resonance imag- srep15269.
ing techniques. J Cereb Blood Flow Metabol 36: 2194–2202, 2016. 484. Petersen NH, Ortega-Gutierrez S, Reccius A, Masurkar A, Huang A,
doi:10.1177/0271678X15615874. Marshall RS. Dynamic cerebral autoregulation is transiently impaired
for one week after large-vessel acute ischemic stroke. Cerebrovasc
471. Atkins ER, Brodie FG, Rafelt SE, Panerai RB, Robinson TG. Dynamic
Dis 39: 144–150, 2015. doi:10.1159/000368595.
cerebral autoregulation is compromised acutely following mild is-
chemic stroke but not transient ischaemic attack. Cerebrovasc Dis 485. Dawson SL, Panerai RB, Potter JF. Serial changes in static and
29: 228–235, 2010. doi:10.1159/000267845. dynamic cerebral autoregulation after acute ischaemic stroke.
Cerebrovasc Dis 16: 69–75, 2003. doi:10.1159/000070118.
472. Minhas JS, Panerai RB, Swienton D, Robinson TG. Feasibility of
improving cerebral autoregulation in acute intracerebral hemor- 486. Ma H, Guo ZN, Yan JH, Liu X, Lv J, Zhang S, Sun P, Yang XY.
rhage (BREATHE-ICH) study: results from an experimental interven- Preliminary study of dynamic cerebral autoregulation in acute ische-
tional study. Int J Stroke 15: 627–637, 2020. doi:10.1177/17474930 mic stroke: association with clinical factors. Front Neurol 9: 1006,
19873690. 2018. doi:10.3389/fneur.2018.01006.
473. Salinet AS, Panerai RB, Robinson TG. The longitudinal evolution of 487. Haubrich C, Kruska W, Diehl RR, Moller-Hartmann W, Klotzsch
cerebral blood flow regulation after acute ischaemic stroke. C. Dynamic autoregulation testing in patients with middle cere-
Cerebrovasc Dis Extra 4: 186–197, 2014. doi:10.1159/000366017. bral artery stenosis. Stroke 34: 1881–1885, 2003. doi:10.1161/01.
STR.0000080936.36601.34.
474. Czosnyka M, Smielewski P, Lavinio A, Pickard JD, Panerai R. An
assessment of dynamic autoregulation from spontaneous fluctua- 488. Wang S, Guo ZN, Xing Y, Ma H, Jin H, Liu J, Yang Y. Dynamic cere-
tions of cerebral blood flow velocity: a comparison of two models, bral autoregulation in asymptomatic patients with unilateral middle
index of autoregulation and mean flow index. Anesth Analg 106: cerebral artery stenosis. Medicine 94: e2234, 2015. doi:10.1097/MD.
234–239, 2008. doi:10.1213/01.ane.0000295802.89962.13. 0000000000002234.

475. Rasulo FA, Girardini A, Lavinio A, De Peri E, Stefini R, Cenzato M, 489. Castro P, Azevedo E, Serrador J, Rocha I, Sorond F. Hemorrhagic
Nodari I, Latronico N. Are optimal cerebral perfusion pressure and transformation and cerebral edema in acute ischemic stroke: link to
cerebrovascular autoregulation related to long-term outcome in cerebral autoregulation. J Neurol Sci 372: 256–261, 2017. doi:10.
patients with aneurysmal subarachnoid hemorrhage? J Neurosurg 1016/j.jns.2016.11.065.
Anesthesiol 24: 3–8, 2012. doi:10.1097/ANA.0b013e318224030a.
490. Castro P, Serrador JM, Rocha I, Sorond F, Azevedo E. Efficacy of cer-
476. Silverman A, Kodali S, Strander S, Gilmore EJ, Kimmel A, Wang A, ebral autoregulation in early ischemic stroke predicts smaller
Cord B, Falcone G, Hebert R, Matouk C, Sheth KN, Petersen NH. infarcts and better outcomes. Front Neurol 8: 113, 2017. doi:10.3389/
Deviation from personalized blood pressure targets is associated fneur.2017.00113.

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1553


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

491. Rosen DS, Macdonald RL. Subarachnoid hemorrhage grading 505. Raichle ME, Posner JB, Plum F. Cerebral blood flow during and after
scales: a systematic review. Neurocrit Care 2: 110–118, 2005. doi:10. hyperventilation. Arch Neurol 23: 394–403, 1970. doi:10.1001/
1385/NCC:2:2:110. archneur.1970.00480290014002.

492. Dohmen C, Bosche B, Graf R, Reithmeier T, Ernestus RI, Brinker G, 506. Willmot M, Leonardi-Bee J, Bath PM. High blood pressure in acute
Sobesky J, Heiss WD. Identification and clinical impact of impaired stroke and subsequent outcome: a systematic review. Hypertension
cerebrovascular autoregulation in patients with malignant middle 43: 18–24, 2004. doi:10.1161/01.HYP.0000105052.65787.35.
cerebral artery infarction. Stroke 38: 56–61, 2007. doi:10.1161/01.
STR.0000251642.18522.b6. 507. Anderson CS, Huang Y, Lindley RI, Chen X, Arima H, Chen G, et al.
Intensive blood pressure reduction with intravenous thrombolysis
493. Aries MJ, Czosnyka M, Budohoski KP, Steiner LA, Lavinio A, Kolias therapy for acute ischaemic stroke (ENCHANTED): an international,
AG, Hutchinson PJ, Brady KM, Menon DK, Pickard JD, Smielewski P. randomised, open-label, blinded-end-point, phase 3 trial. Lancet
Continuous determination of optimal cerebral perfusion pressure in 393: 877–888, 2019. doi:10.1016/S0140-6736(19)30038-8.
traumatic brain injury. Crit Care Med 40: 2456–2463, 2012.
doi:10.1097/CCM.0b013e3182514eb6. 508. Bath PM, Scutt P, Anderson CS, Ankolekar S, Appleton JP, Berge E,
Cala L, et al. Prehospital transdermal glyceryl trinitrate in patients
494. Amarenco P, Bogousslavsky J, Caplan LR, Donnan GA, Hennerici with ultra-acute presumed stroke (RIGHT-2): an ambulance-based,
MG. Classification of stroke subtypes. Cerebrovasc Dis 27: 493– randomised, sham-controlled, blinded, phase 3 trial. Lancet 393:
501, 2009. doi:10.1159/000210432. 1009–1020, 2019. doi:10.1016/S0140-6736(19)30194-1.
495. Xiong L, Tian G, Lin W, Wang W, Wang L, Leung T, Mok V, Liu J, Chen
509. Potter JF, Robinson TG, Ford GA, Mistri A, James M, Chernova J,
X, Wong KS. Is dynamic cerebral autoregulation bilaterally impaired
Jagger C. Controlling hypertension and hypotension immediately
after unilateral acute ischemic stroke? J Stroke Cerebrovasc Dis 26:
post-stroke (CHHIPS): A randomised, placebo-controlled, double-
1081–1087, 2017. doi:10.1016/j.jstrokecerebrovasdis.2016.12.024.
blind pilot trial. Lancet Neurol 8: 48–56, 2009. doi:10.1016/S1474-
496. Chi NF, Hu HH, Wang CY, Chan L, Peng CK, Novak V, Hu CJ. 4422(08)70263-1.
Dynamic cerebral autoregulation is an independent functional out-
510. Robinson TG, Davison WJ, Rothwell PM, Potter JF. Randomised
come predictor of mild acute ischemic stroke. Stroke 49: 2605–
controlled trial of a calcium channel or angiotensin converting
2611, 2018. doi:10.1161/STROKEAHA.118.022481.
enzyme inhibitor/angiotensin receptor blocker regime to reduce
497. Deng W, Kandhi S, Zhang B, Huang A, Koller A, Sun D. Extravascular blood pressure variability following ischaemic stroke (CAARBS):
blood augments myogenic constriction of cerebral arterioles: impli- a protocol for a feasibility study. BMJ Open 9: e025301, 2019.
cations for hemorrhage-induced vasospasm. J Am Heart Assoc 7: doi:10.1136/bmjopen-2018-025301.
e008623, 2018. doi:10.1161/jaha.118.008623.
511. Robinson TG, Potter JF, Ford GA, Bulpitt CJ, Chernova J, Jagger C,
498. Banks JL, Marotta CA. Outcomes validity and reliability of the modi- James MA, Knight J, Markus HS, Poulter NR, Cossacs I. Effects of ani-
fied Rankin scale: Implications for stroke clinical trials: a literature thypertensive treatment after acute stroke in the Continue or Stop
review and synthesis. Stroke 38: 1091–1096, 2007. doi:10.1161/01. Post-Stroke Antihypertensives Collaborative Study (COSSACS): a pro-
STR.0000258355.23810.c6. spective, randomised, open, blinded end-point trial. Lancet Neurol 9:
767–775, 2010. doi:10.1016/S1474-4422(10)70163-0.
499. Castro P, Azevedo E, Rocha I, Sorond F, Serrador JM. Chronic kid-
ney disease and poor outcomes in ischemic stroke: Is impaired cer- 512. Chan E, Anderson CS, Wang X, Arima H, Saxena A, Moullaali TJ,
ebral autoregulation the missing link? BMC Neurol 18: 21, 2018. Delcourt C, Wu G, Wang J, Chen G, Lavados PM, Stapf C, Robinson
doi:10.1186/s12883-018-1025-4. T, Chalmers J, The INTERACT Investigators. Early blood pressure
lowering does not reduce growth of intraventricular hemorrhage fol-
500. Czosnyka M, Hutchinson P, Smielewski P. Treatment targets based on
lowing acute intracerebral hemorrhage: results of the INTERACT
autoregulation parameters in neurocritical care patients. Curr Opin Crit
studies. Cerebrovasc Dis Extra 6: 71–75, 2016. doi:10.1159/
Care 26: 109–114, 2020. doi:10.1097/MCC.0000000000000704.
000448897.
501. Czosnyka M, Smielewski P, Kirkpatrick P, Laing RJ, Menon D,
513. Moullaali TJ, Wang X, Martin RH, Shipes VB, Robinson TG, Chalmers
Pickard JD. Continuous assessment of the cerebral vasomotor reac-
J, Suarez JI, Qureshi AI, Palesch YY, Anderson CS. Blood pressure
tivity in head injury. Neurosurgery 41: 11–17, 1997. doi:10.1097/
00006123-199707000-00005. control and clinical outcomes in acute intracerebral haemorrhage: a
preplanned pooled analysis of individual participant data. Lancet
502. Steiner LA, Czosnyka M, Piechnik SK, Smielewski P, Chatfield D, Neurol 18: 857–864, 2019. doi:10.1016/S1474-4422(19)30196-6.
Menon DK, Pickard JD. Continuous monitoring of cerebrovascular
pressure reactivity allows determination of optimal cerebral perfu- 514. Qureshi AI, Palesch YY, Barsan WG, Hanley DF, Hsu CY, Martin RL,
sion pressure in patients with traumatic brain injury. Crit Care Med Moy CS, Silbergleit R, Steiner T, Suarez JI, Toyoda K, Wang Y,
30: 733–738, 2002. doi:10.1097/00003246-200204000-00002. Yamamoto H, Yoon BW, Investigators ATACH-2 Trial Investigators
and the Neurological Emergency Treatment Trials Network.
503. Petersen NH, Silverman A, Strander SM, Kodali S, Wang A, Sansing Intensive blood-pressure lowering in patients with acute cerebral
LH, Schindler JL, Falcone GJ, Gilmore EJ, Jasne AS, Cord B, Hebert hemorrhage. N Engl J Med 375: 1033–1043, 2016. doi:10.1056/
RM, Johnson M, Matouk CC, Sheth KN. Fixed compared with autore- NEJMoa1603460.
gulation-oriented blood pressure thresholds after mechanical
thrombectomy for ischemic stroke. Stroke 51: 914–921, 2020. 515. Palomares SM, Cipolla MJ. Vascular protection following cerebral is-
doi:10.1161/STROKEAHA.119.026596. chemia and reperfusion. J Neurol Neurophysiol 2011: S1–S004,
2011. doi:10.4172/2155-9562.S1-004.
504. Salinet AS, Minhas JS, Panerai RB, Bor-Seng-Shu E, Robinson TG.
Do acute stroke patients develop hypocapnia? A systematic review 516. Johnston KC, Durkalski-Mauldin VL. Considering prehospital stroke
and meta-analysis. J Neurol Sci 402: 30–39, 2019. doi:10.1016/j. trials: Did TIGHT-2 get it right? Lancet 393: 963–965, 2019. doi:10.
jns.2019.04.038. 1016/S0140-6736(19)30276-4.

1554 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

517. Starke RM, Peterson EC, Komotar RJ, Connolly ES. A randomized 529. Iadecola C. The pathobiology of vascular dementia. Neuron 80:
clinical trial of aggressive blood pressure control in patients with 844–866, 2013. doi:10.1016/j.neuron.2013.10.008.
acute cerebral hemorrhage. Neurosurgery 79: N17–N18, 2016.
doi:10.1227/01.neu.0000508604.61565.ba. 530. Vinciguerra L, Lanza G, Puglisi V, Pennisi M, Cantone M, Bramanti A,
Pennisi G, Bella R. Transcranial Doppler ultrasound in vascular cog-
518. Gorelick PB, Scuteri A, Black SE, Decarli C, Greenberg SM, Iadecola nitive impairment–no dementia. PLoS One 14: e0216162, 2019.
C, Launer LJ, Laurent S, Lopez OL, Nyenhuis D, Petersen RC, doi:10.1371/journal.pone.0216162.
Schneider JA, Tzourio C, Arnett DK, Bennett DA, Chui HC,
Higashida RT, Lindquist R, Nilsson PM, Roman GC, Sellke FW, 531. Keage HA, Churches OF, Kohler M, Pomeroy D, Luppino R, Bartolo
ML, Elliott S. Cerebrovascular function in aging and dementia: a sys-
Seshadri S, American Heart Association Stroke Council, Council on
tematic review of transcranial Doppler studies. Dement Geriatr
Epidemiology and Prevention, Council on Cardiovascular Nursing,
Cogn Dis Extra 2: 258–270, 2012. doi:10.1159/000339234.
Council on Cardiovascular Radiology and Intervention, and Council
on Cardiovascular Surgery and Anesthesia. Vascular contributions 532. Sabayan B, Jansen S, Oleksik AM, van Osch MJ, van Buchem MA,
to cognitive impairment and dementia: a statement for healthcare van Vliet P, de Craen AJ, Westendorp RG. Cerebrovascular hemo-
professionals from the American Heart Association/American dynamics in Alzheimer's disease and vascular dementia: a meta-
Stroke Association. Stroke 42: 2672–2713, 2011. doi:10.1161/STR. analysis of transcranial Doppler studies. Ageing Res Rev 11: 271–
0b013e3182299496. 277, 2012. doi:10.1016/j.arr.2011.12.009.
519. Arvanitakis Z, Capuano AW, Leurgans SE, Bennett DA, Schneider 533. O'Sullivan M, Lythgoe DJ, Pereira AC, Summers PE, Jarosz JM,
JA. Relation of cerebral vessel disease to Alzheimer's disease de- Williams SC, Markus HS. Patterns of cerebral blood flow reduction in
mentia and cognitive function in elderly people: a cross-sectional patients with ischemic leukoaraiosis. Neurology 59: 321–326,
study. Lancet Neurol 15: 934–943, 2016. doi:10.1016/S1474-4422(16) 2002. doi:10.1212/wnl.59.3.321.
30029-1.
534. Bakker SL, de Leeuw FE, de Groot JC, Hofman A, Koudstaal PJ,
520. Cuadrado-Godia E, Dwivedi P, Sharma S, Ois Santiago A, Roquer Breteler MM. Cerebral vasomotor reactivity and cerebral white mat-
Gonzalez J, Balcells M, Laird J, Turk M, Suri HS, Nicolaides A, Saba ter lesions in the elderly. Neurology 52: 578–583, 1999. doi:10.1212/
L, Khanna NN, Suri JS. Cerebral small vessel disease: A review fo- wnl.52.3.578.
cusing on pathophysiology, biomarkers, and machine learning strat-
egies. J Stroke 20: 302–320, 2018. doi:10.5853/jos.2017.02922. 535. Vernooij MW, van der Lugt A, Ikram MA, Wielopolski PA, Vrooman
HA, Hofman A, Krestin GP, Breteler MM. Total cerebral blood flow
521. Poggesi A, Inzitari D, Pantoni L. Atrial fibrillation and cognition: epi- and total brain perfusion in the general population: The Rotterdam
demiological data and possible mechanisms. Stroke 46: 3316–3321, Scan Study. J Cereb Blood Flow Metabol 28: 412–419, 2008.
2015. doi:10.1161/STROKEAHA.115.008225. doi:10.1038/sj.jcbfm.9600526.

522. Qiu C, Fratiglioni L. A major role for cardiovascular burden in age- 536. Bowler JV, Gorelick PB. Advances in vascular cognitive impairment.
related cognitive decline. Nat Rev Cardiol 12: 267–277, 2015. Stroke 40: e315-318–e318, 2009. doi:10.1161/STROKEAHA.108.544650.
doi:10.1038/nrcardio.2014.223.
537. Claassen JA. Alzheimer's disease: trade-off for increased survival
523. Wardlaw JM, Smith EE, Biessels GJ, Cordonnier C, Fazekas F, with atherosclerosis? Ann Neurol 64: 475, 2008. doi:10.1002/
Frayne R, STandards for ReportIng Vascular changes on ana.21285.
nEuroimaging (STRIVE v1), et al. Neuroimaging standards for
research into small vessel disease and its contribution to ageing 538. Claassen JA. Cognitive decline and dementia: are we getting to the
and neurodegeneration. Lancet Neurol 12: 822–838, 2013. vascular heart of the matter? Hypertension 65: 505–506, 2015.
doi:10.1161/HYPERTENSIONAHA.114.04706.
doi:10.1016/S1474-4422(13)70124-8.
539. Claassen JA. New cardiovascular targets to prevent late onset
524. Ballard C, Burns A, Corbett A, Livingston G, Rasmussen J. Helping
Alzheimer disease. Eur J Pharmacol 763: 131–134, 2015. doi:10.1016/
you to assess cognition. A practical toolkit for clinicians. Alzheimer's
j.ejphar.2015.05.022.
Society March 2013: 1–40, 2013.
540. Iadecola C. Neurovascular regulation in the normal brain and in
525. Makin SD, Turpin S, Dennis MS, Wardlaw JM. Cognitive impairment
Alzheimer's disease. Nat Rev Neurosci 5: 347–360, 2004. doi:10.
after lacunar stroke: Systematic review and meta-analysis of inci-
1038/nrn1387.
dence, prevalence and comparison with other stroke subtypes. J
Neurol Neurosurg Psychiatry 84: 893–900, 2013. doi:10.1136/jnnp- 541. Iturria-Medina Y, Sotero RC, Toussaint PJ, Mateos-Perez JM, Evans
2012-303645. AC, Alzheimer’s Disease Neuroimaging Initiative. Alzheimer's dis-
ease neuroimaging I. Early role of vascular dysregulation on late-
526. Sachdev P, Kalaria R, O’Brien J, Skoog I, Alladi S, Black SE, Blacker
onset Alzheimer's disease based on multifactorial data-driven analy-
D, Blazer DG, Chen C, Chui H, Ganguli M, Jellinger K, Jeste DV,
sis. Nat Commun 7: 11934, 2016. doi:10.1038/ncomms11934.
Pasquier F, Paulsen J, Prins N, Rockwood K, Roman G, Scheltens P.
Diagnostic criteria for vascular cognitive disorders: a VASCOG state- 542. Nicolakakis N, Hamel E. Neurovascular function in Alzheimer's dis-
ment. Alzheimer Dis Assoc Disord 28: 206–218, 2014. doi:10.1097/ ease patients and experimental models. J Cereb Blood Flow Metab
WAD.0000000000000034. 31: 1354–1370, 2011. doi:10.1038/jcbfm.2011.43.

527. Mathias JL, Burke J. Cognitive functioning in Alzheimer's and vascu- 543. Pasha EP, Rutjes E, Tomoto T, Tarumi T, Stowe A, Claassen J,
lar dementia: a meta-analysis. Neuropsychology 23: 411–423, 2009. Munro Cullum C, Zhu DC, Zhang R. Carotid stiffness is associated
doi:10.1037/a0015384. with brain amyloid-b burden in amnestic mild cognitive impairment.
J Alzheimers Dis 74: 925–935, 2020. doi:10.3233/JAD-191073.
528. Park L, Anrather J, Girouard H, Zhou P, Iadecola C. Nox2-derived re-
active oxygen species mediate neurovascular dysregulation in the 544. Roher AE, Esh C, Rahman A, Kokjohn TA, Beach TG.
aging mouse brain. J Cereb Blood Flow Metab 27: 1908–1918, Atherosclerosis of cerebral arteries in Alzheimer disease. Stroke 35:
2007. doi:10.1038/sj.jcbfm.9600491. 2623–2627, 2004. doi:10.1161/01.STR.0000143317.70478.b3.

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1555


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

545. Zlokovic BV. Neurovascular pathways to neurodegeneration in 557. Nagahama Y, Nabatame H, Okina T, Yamauchi H, Narita M,
Alzheimer's disease and other disorders. Nat Rev Neurosci 12: Fujimoto N, Murakami M, Fukuyama H, Matsuda M. Cerebral corre-
723–738, 2011. doi:10.1038/nrn3114. lates of the progression rate of the cognitive decline in probable
Alzheimer's disease. Eur Neurol 50: 1–9, 2003. doi:10.1159/
546. Iadecola C, Yaffe K, Biller J, Bratzke LC, Faraci FM, Gorelick PB, 000070851.
Gulati M, Kamel H, Knopman DS, Launer LJ, Saczynski JS, Seshadri
S, Hazzouri ZA, American Heart Association Council on 558. Chao LL, Buckley ST, Kornak J, Schuff N, Madison C, Yaffe K, Miller
Hypertension; Council on Clinical Cardiology; Council on BL, Kramer JH, Weiner MW. ASL perfusion MRI predicts cognitive
Cardiovascular Disease in the Young; Council on Cardiovascular decline and conversion from MCI to dementia. Alzheimer Dis Assoc
and Stroke Nursing; Council on Quality of Care and Outcomes Disord 24: 19–27, 2010. doi:10.1097/WAD.0b013e3181b4f736.
Research; and Stroke Council. Impact of hypertension on cognitive
function: a scientific statement from the American Heart Assoc- 559. Benedictus MR, Leeuwis AE, Binnewijzend MA, Kuijer JP, Scheltens
iation. Hypertension 68: e67–e94, 2016. doi:10.1161/hyp.00000 P, Barkhof F, van der Flier WM, Prins ND. Lower cerebral blood flow
00000000053. is associated with faster cognitive decline in Alzheimer's disease.
Eur Radiol 27: 1169–1175, 2017. doi:10.1007/s00330-016-4450-z.
547. Gustavsson AM, van Westen D, Stomrud E, Engstrom G, Nagga K,
Hansson O. Midlife atherosclerosis and development of Alzheimer 560. Leeuwis AE, Benedictus MR, Kuijer JP, Binnewijzend MA,
or vascular dementia. Ann Neurol 87: 52–62, 2020. doi:10.1002/ Hooghiemstra AM, Verfaillie SC, Koene T, Scheltens P, Barkhof F,
ana.25645. Prins ND, van der Flier WM. Lower cerebral blood flow is associated
with impairment in multiple cognitive domains in Alzheimer's dis-
548. Iadecola C, Gottesman RF. Cerebrovascular alterations in Alzheimer
ease. Alzheimers Dement 13: 531–540, 2017. doi:10.1016/j.jalz.2016.
disease. Circ Res 123: 406–408, 2018. doi:10.1161/CIRCRESAHA.118.
08.013.
313400.
561. Hajjar I, Sorond F, Lipsitz LA. Apolipoprotein E, carbon dioxide vaso-
549. Jack CR Jr., Bennett DA, Blennow K, Carrillo MC, Dunn B,
reactivity, and cognition in older adults: effect of hypertension. J Am
Haeberlein SB, Holtzman DM, Jagust W, Jessen F, Karlawish J, Liu
Geriatr Soc 63: 276–281, 2015. doi:10.1111/jgs.13235.
E, Molinuevo JL, Montine T, Phelps C, Rankin KP, Rowe CC,
Scheltens P, Siemers E, Snyder HM, Sperling R, Contributors. NIA- 562. Abeelen A, Lagro J, Beek A, Claassen J. Impaired cerebral autore-
AA Research Framework: toward a biological definition of gulation and vasomotor reactivity in sporadic Alzheimer’s disease.
Alzheimer’s disease. Alzheimer's Dement 14: 535–562, 2018. Curr Alzheimer Res 11: 11–17, 2014. doi:10.2174/156720501066613111
doi:10.1016/j.jalz.2018.02.018. 9234845.
550. Garcia-Alloza M, Gregory J, Kuchibhotla KV, Fine S, Wei Y, Ayata C,
563. Suri S, Mackay CE, Kelly ME, Germuska M, Tunbridge EM, Frisoni
Frosch MP, Greenberg SM, Bacskai BJ. Cerebrovascular lesions
GB, Matthews PM, Ebmeier KP, Bulte DP, Filippini N. Reduced cere-
induce transient ß-amyloid deposition. Brain 134: 3697–3707, 2011.
brovascular reactivity in young adults carrying the APOE epsilon4 al-
doi:10.1093/brain/awr300.
lele. Alzheimers Dement 11: 648–657, 2015. doi:10.1016/j.jalz.
551. Alsop DC, Dai W, Grossman M, Detre JA. Arterial spin labeling blood 2014.05.1755.
flow MRI: Its role in the early characterization of Alzheimer's disease.
564. Kisler K, Nelson AR, Montagne A, Zlokovic BV. Cerebral blood flow
JAD 20: 871–880, 2010. doi:10.3233/JAD-2010-091699.
regulation and neurovascular dysfunction in Alzheimer disease. Nat
552. Wolk DA, Detre JA. Arterial spin labeling MRI: an emerging bio- Rev Neurosci 18: 419–434, 2017. doi:10.1038/nrn.2017.48.
marker for Alzheimer's disease and other neurodegenerative condi-
tions. Curr Opin Neurol 25: 421–428, 2012. doi:10.1097/WCO.0b013 565. Claassen JA, Zhang R. Cerebral autoregulation in Alzheimer's dis-
e328354ff0a. ease. J Cereb Blood Flow Metab 31: 1572–1577, 2011. doi:10.1038/
jcbfm.2011.69.
553. Xu G, Rowley HA, Wu G, Alsop DC, Shankaranarayanan A, Dowling
M, Christian BT, Oakes TR, Johnson SC. Reliability and precision of 566. Zhang F, Eckman C, Younkin S, Hsiao KK, Iadecola C. Increased sus-
pseudo-continuous arterial spin labeling perfusion MRI on 3.0 T and ceptibility to ischemic brain damage in transgenic mice overex-
comparison with 15O-water PET in elderly subjects at risk for pressing the amyloid precursor protein. J Neurosci 17: 7655–7661,
Alzheimer's disease. NMR Biomed 23: 286–293, 2010. doi:10.1002/ 1997. doi:10.1523/JNEUROSCI.17-20-07655.1997.
nbm.1462.
567. Simard D, Olesen J, Paulson OB, Lassen NA, Skinhoj E. Regional
554. Musiek ES, Chen Y, Korczykowski M, Saboury B, Martinez PM, cerebral blood flow and its regulation in dementia. Brain 94: 273–
Reddin JS, Alavi A, Kimberg DY, Wolk DA, Julin P, Newberg AB, 288, 1971. doi:10.1093/brain/94.2.273.
Arnold SE, Detre JA. Direct comparison of fluorodeoxyglucose posi-
tron emission tomography and arterial spin labeling magnetic reso- 568. Claassen JA, Diaz-Arrastia R, Martin-Cook K, Levine BD, Zhang R.
nance imaging in Alzheimer's disease. Alzheimers Dement 8: 51– Altered cerebral hemodynamics in early Alzheimer disease: a pilot
59, 2012. doi:10.1016/j.jalz.2011.06.003. study using transcranial Doppler. J Alzheimers Dis 17: 621–629,
2009. doi:10.3233/JAD-2009-1079.
555. Landau SM, Mintun MA, Joshi AD, Koeppe RA, Petersen RC, Aisen
PS, Weiner MW, Jagust WJ, Alzheimer's Disease Neuroimaging 569. Zazulia AR, Videen TO, Morris JC, Powers WJ. Autoregulation of
Initiative. Amyloid deposition, hypometabolism, and longitudinal cerebral blood flow to changes in arterial pressure in mild
cognitive decline. Ann Neurol 72: 578–586, 2012. doi:10.1002/ Alzheimer's disease. J Cereb Blood Flow Metab 30: 1883–1889,
ana.23650. 2010. doi:10.1038/jcbfm.2010.135.

556. Binnewijzend MA, Benedictus MR, Kuijer JP, van der Flier WM, 570. van Beek AH, Sijbesma JC, Jansen RW, Rikkert MG, Claassen JA.
Teunissen CE, Prins ND, Wattjes MP, van Berckel BN, Scheltens P, Cortical oxygen supply during postural hypotension is further
Barkhof F. Cerebral perfusion in the predementia stages of decreased in Alzheimer's disease, but unrelated to cholinesterase-
Alzheimer's disease. Eur Radiol 26: 506–514, 2016. doi:10.1007/ inhibitor use. J Alzheimers Dis 21: 519–526, 2010. doi:10.3233/JAD-
s00330-015-3834-9. 2010-100288.

1556 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

571. van Beek AH, Lagro J, Olde-Rikkert MG, Zhang R, Claassen JA. 585. Czosnyka M, Smielewski P, Piechnik S, Schmidt EA, Seeley H, Al-
Oscillations in cerebral blood flow and cortical oxygenation in Rawi P, Matta BF, Kirkpatrick PJ, Pickard JD. Continuous assess-
Alzheimer’s disease. Neurobiol Aging 33: e421-431, 2012. ment of cerebral autoregulation–clinical verification of the method
doi:10.1016/j.neurobiolaging.2010.11.016. in head injured patients. Acta Neurochir Suppl 76: 483–484, 2000.
doi:10.1007/978-3-7091-6346-7_101.
572. de Jong DL, de Heus RA, Rijpma A, Donders R, Olde Rikkert MG,
Gunther M, Lawlor BA, van Osch MJ, Claassen J. Effects of nilvadi- 586. Kirkpatrick PJ, Czosnyka M, Pickard JD. Multimodal monitoring in
pine on cerebral blood flow in patients with Alzheimer disease. neurointensive care. J Neurol Neurosurg Psychiatry 60: 131–139,
Hypertension 74: 413–420, 2019. doi:10.1161/HYPERTENSIONAHA. 1996. doi:10.1136/jnnp.60.2.131.
119.12892.
587. Lu CW, Czosnyka M, Shieh JS, Smielewska A, Pickard JD,
573. Niwa K, Kazama K, Younkin L, Younkin SG, Carlson GA, Iadecola C. Smielewski P. Complexity of intracranial pressure correlates with
Cerebrovascular autoregulation is profoundly impaired in mice over- outcome after traumatic brain injury. Brain 135: 2399–2408, 2012.
expressing amyloid precursor protein. Am J Physiol Heart Circ doi:10.1093/brain/aws155.
Physiol 283: H315–H323, 2002. doi:10.1152/ajpheart.00022.2002.
588. McBryde FD, Malpas SC, Paton JF. Intracranial mechanisms for pre-
574. King A. The search for better animal models of Alzheimer's disease. serving brain blood flow in health and disease. Acta Physiol (Oxf)
Nature 559: S13–S15, 2018. doi:10.1038/d41586-018-05722-9. 219: 274–287, 2017. doi:10.1111/apha.12706.
575. de Heus RA, Donders R, Santoso AM, Olde RM, Lawlor BA, 589. Wilson MH. Monro-Kellie 2.0: the dynamic vascular and venous
Claassen J, Nilvad S. G. Blood pressure lowering with nilvadipine in pathophysiological components of intracranial pressure. J Cereb
patients with mild-to-moderate Alzheimer disease does not increase Blood Flow Metabol 36: 1338–1350, 2016. doi:10.1177/0271678X
the prevalence of orthostatic hypotension. J Am Heart Assoc 8:
16648711.
e011938, 2019. doi:10.1161/jaha.119.011938.
590. Zhou ZB, Meng L, Gelb AW, Lee R, Huang WQ. Cerebral ischemia
576. van Beek AH, Sijbesma JC, Olde Rikkert MG, Claassen JA.
during surgery: an overview. J Biomed Res 30: 83–87, 2016.
Galantamine does not cause aggravated orthostatic hypotension in
doi:10.7555/jbr.30.20150126.
people with Alzheimer's disease. J Am Geriatr Soc 58: 409–410,
2010. doi:10.1111/j.1532-5415.2009.02712.x. 591. Spielvogel D, Kai M, Tang GH, Malekan R, Lansman SL. Selective
cerebral perfusion: a review of the evidence. J Thorac Cardiovasc
577. de Heus RA, Olde Rikkert MG, Tully PJ, Lawlor BA, Claassen J,
Surg 145: S59–62, 2013. doi:10.1016/j.jtcvs.2012.11.073.
Group NS, NILVAD Study Group. Blood pressure variability and pro-
gression of clinical Alzheimer disease. Hypertension 74: 1172–1180, 592. Froese L, Dian J, Batson C, Gomez A, Unger B, Zeiler FA.
2019. doi:10.1161/HYPERTENSIONAHA.119.13664. Cerebrovascular response to propofol, fentanyl, and midazolam in
578. Livingston G, Sommerlad A, Orgeta V, Costafreda SG, Huntley J, moderate/severe traumatic brain injury: a scoping systematic review
Ames D, Ballard C, Banerjee S, Burns A, Cohen-Mansfield J, Cooper of the human and animal literature. Neurotrauma Rep 1: 100–112,
C, Fox N, Gitlin LN, Howard R, Kales HC, Larson EB, Ritchie K, 2020. doi:10.1089/neur.2020.0040.
Rockwood K, Sampson EL, Samus Q, Schneider LS, Selbaek G, Teri
593. Slupe AM, Kirsch JR. Effects of anesthesia on cerebral blood flow,
L, Mukadam N. Dementia prevention, intervention, and care. Lancet
metabolism, and neuroprotection. J Cereb Blood Flow Metab 38:
390: 2673–2734, 2017. doi:10.1016/S0140-6736(17)31363-6.
2192–2208, 2018. doi:10.1177/0271678X18789273.
579. Czosnyka M, Brady K, Reinhard M, Smielewski P, Steiner LA.
594. Gao YR, Ma Y, Zhang Q, Winder AT, Liang Z, Antinori L, Drew PJ,
Monitoring of cerebrovascular autoregulation: Facts, myths, and
Zhang N. Time to wake up: Studying neurovascular coupling and
missing links. Neurocrit Care 10: 373–386, 2009. doi:10.1007/
brain-wide circuit function in the un-anesthetized animal. Neuroimage
s12028-008-9175-7.
153: 382–398, 2017. doi:10.1016/j.neuroimage.2016.11.069.
580. Liu X, Czosnyka M, Donnelly J, Budohoski KP, Varsos GV, Nasr N,
595. Matta BF, Heath KJ, Tipping K, Summors AC. Direct cerebral vasodi-
Brady KM, Reinhard M, Hutchinson PJ, Smielewski P. Comparison of
latory effects of sevoflurane and isoflurane. Anesthesiology 91:
frequency and time domain methods of assessment of cerebral
autoregulation in traumatic brain injury. J Cereb Blood Flow Metab 677–680, 1999. doi:10.1097/00000542-199909000-00019.
35: 248–256, 2015. doi:10.1038/jcbfm.2014.192. 596. Minhas JS, Rook W, Panerai RB, Hoiland RL, Ainslie PN, Thompson
581. Rivera-Lara L, Zorrilla-Vaca A, Geocadin R, Ziai W, Healy R, JP, Mistri AK, Robinson TG. Pathophysiological and clinical consider-
Thompson R, Smielewski P, Czosnyka M, Hogue CW. Predictors of ations in the perioperative care of patients with a previous ischae-
outcome with cerebral autoregulation monitoring: A systematic mic stroke: a multidisciplinary narrative review. Br J Anaesth 124:
review and meta-analysis. Crit Care Med 45: 695–704, 2017. 183–196, 2020. doi:10.1016/j.bja.2019.10.021.
doi:10.1097/CCM.0000000000002251.
597. Saxena N, Gili T, Diukova A, Huckle D, Hall JE, Wise RG. Mild propo-
582. Brady KM, Lee JK, Kibler KK, Smielewski P, Czosnyka M, Easley RB, fol sedation reduces frontal lobe and thalamic cerebral blood flow:
Koehler RC, Shaffner DH. Continuous time-domain analysis of cere- an arterial spin labeling study. Front Physiol 10: 1541, 2019.
brovascular autoregulation using near-infrared spectroscopy. doi:10.3389/fphys.2019.01541.
Stroke 38: 2818–2825, 2007. doi:10.1161/STROKEAHA.107.485706.
598. Wu GR, Di Perri C, Charland-Verville V, Martial C, Carriere M,
583. Czosnyka M, Smielewski P, Kirkpatrick P, Menon DK, Pickard JD. Vanhaudenhuyse A, Laureys S, Marinazzo D. Modulation of the
Monitoring of cerebral autoregulation in head-injured patients. spontaneous hemodynamic response function across levels of
Stroke 27: 1829–1834, 1996. doi:10.1161/01.STR.27.10.1829. consciousness. Neuroimage 200: 450–459, 2019. doi:10.1016/j.
neuroimage.2019.07.011.
584. Czosnyka M, Smielewski P, Piechnik S, Schmidt EA, Al-Rawi PG,
Kirkpatrick PJ, Pickard JD. Hemodynamic characterization of intra- 599. Gutkin M, Stewart JM. Orthostatic circulatory disorders: from nosol-
cranial pressure plateau waves in head-injury patients. J Neurosurg ogy to nuts and bolts. Am J Hypertens 29: 1009–1019, 2016.
91: 11–19, 1999. doi:10.3171/jns.1999.91.1.0011. doi:10.1093/ajh/hpw023.

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1557


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CLAASSEN ET AL.

600. O' Brien H, Anne Kenny R. Syncope in the elderly. Eur Cardiol 9: Intern Med 172: 494–500, 2012. doi:10.1001/archinternmed.2011.
28–36, 2014. doi:10.15420/ecr.2014.9.1.28. 2174.

601. Sutton R. Reflex syncope: diagnosis and treatment. J Arrhythm 33: 615. Carter S, Hartman Y, Holder S, Thijssen DH, Hopkins ND. Sedentary
545–552, 2017. doi:10.1016/j.joa.2017.03.007. behavior and cardiovascular disease risk: mediating mechanisms.
Exerc Sport Sci Rev 45: 80–86, 2017. doi:10.1249/JES.00000000
602. Moya A, Sutton R, Ammirati F, Blanc JJ, Brignole M, Dahm JB, 00000106.
Deharo JC, Gajek J, Gjesdal K, Krahn A, Massin M, Pepi M, Pezawas
T, Ruiz GR, Sarasin F, Ungar A, van Dijk JG, Walma EP, Wieling Task 616. Falck RS, Davis JC, Liu-Ambrose T. What is the association between
Force for the Diagnosis and Management of Syncope, European sedentary behaviour and cognitive function? A systematic review. Br
Society of Cardiology, European Heart Rhythm Association, Heart J Sports Med 51: 800–811, 2017. doi:10.1136/bjsports-2015-095551.
Failure Association, Heart Rhythm Society. W. Guidelines for the di-
617. Wheeler MJ, Dempsey PC, Grace MS, Ellis KA, Gardiner PA, Green
agnosis and management of syncope (version 2009). Eur Heart J
DJ, Dunstan DW. Sedentary behavior as a risk factor for cognitive
30: 2631–2671, 2009. doi:10.1093/eurheartj/ehp298.
decline? A focus on the influence of glycemic control in brain health.
603. Schondorf R, Stein R, Roberts R, Benoit J, Cupples W. Dynamic cere- Alzheimers Dement (N Y) 3: 291–300, 2017. doi:10.1016/j.trci.
bral autoregulation is preserved in neurally mediated syncope. J 2017.04.001.
Appl Physiol (1985) 91: 2493–2502, 2001. doi:10.1152/jappl.2001. 618. Carter SE, Draijer R, Holder SM, Brown L, Thijssen DH, Hopkins ND.
91.6.2493. Regular walking breaks prevent the decline in cerebral blood flow
604. Fu Q, Levine BD. Pathophysiology of neurally mediated syncope: associated with prolonged sitting. J Appl Physiol (1985) 125: 790–
role of cardiac output and total peripheral resistance. Auton 798, 2018. doi:10.1152/japplphysiol.00310.2018.
Neurosci 184: 24–26, 2014. doi:10.1016/j.autneu.2014.07.004. 619. Claassen JA, Levine BD, Zhang R. Cerebral vasomotor reactivity
before and after blood pressure reduction in hypertensive patients.
605. van Wijnen VK, Ten Hove D, Gans RO, Nieuwland W, van Roon AM,
Am J Hypertens 22: 384–391, 2009. doi:10.1038/ajh.2009.2.
Ter Maaten JC, Harms MP. Orthostatic blood pressure recovery pat-
terns in suspected syncope in the emergency department. Emerg 620. Wheeler MJ, Dunstan DW, Smith B, Smith KJ, Scheer A, Lewis J,
Med J 35: 226–230, 2018. doi:10.1136/emermed-2017-207207. Naylor LH, Heinonen I, Ellis KA, Cerin E, Ainslie PN, Green DJ.
Morning exercise mitigates the impact of prolonged sitting on cere-
606. Iwasaki K, Zhang R, Perhonen MA, Zuckerman JH, Levine BD.
bral blood flow in older adults. J Appl Physiol 126: 1049–1055, 2019.
Reduced baroreflex control of heart period after bed rest is normal-
doi:10.1152/japplphysiol.00001.2019.
ized by acute plasma volume restoration. Am J Physiol Regul
Integr Comp Physiol 287: R1256–R1262, 2004. doi:10.1152/ 621. Maasakkers CM, Melis RJ, Kessels RP, Gardiner PA, Olde RM,
ajpregu.00613.2002. Thijssen DH, Claassen J. The short-term effects of sedentary behav-
iour on cerebral hemodynamics and cognitive performance in older
607. Levine BD, Giller CA, Lane LD, Buckey JC, Blomqvist CG. Cerebral adults: a cross-over design on the potential impact of mental and/or
versus systemic hemodynamics during graded orthostatic stress in physical activity. Alzheimers Res Ther 12: 76, 2020. doi:10.1186/
humans. Circulation 90: 298–306, 1994. doi:10.1161/01.cir.90.1.298. s13195-020-00644-z.
608. Zhang R, Zuckerman JH, Pawelczyk JA, Levine BD. Effects of head- 622. Perdomo SJ, Gibbs BB, Kowalsky RJ, Taormina JM, Balzer JR.
down-tilt bed rest on cerebral hemodynamics during orthostatic Effects of alternating standing and sitting compared with prolonged
stress. J Appl Physiol (1985) 83: 2139–2145, 1997. doi:10.1152/ sitting on cerebrovascular hemodynamics. Sport Sci Health 15:
jappl.1997.83.6.2139. 375–383, 2019. doi:10.1007/s11332-019-00526-4.
609. Benditt DG, Fabian W, Iskos D, Lurie KG. Review article: Heart rate 623. Zlatar ZZ, Hays CC, Mestre Z, Campbell LM, Meloy MJ, Bangen KJ,
and blood pressure control in vasovagal syncope. J Interv Card Liu TT, Kerr J, Wierenga CE. Dose-dependent association of accel-
Electrophysiol 2: 25–32, 1998. doi:10.1023/a:1009756521965. erometer-measured physical activity and sedentary time with brain
perfusion in aging. Exp Gerontol 125: 110679, 2019. doi:10.1016/j.
610. Thijs RD, Wieling W, Kaufmann H, van Dijk G. Defining and classify-
exger.2019.110679.
ing syncope. Clin Auton Res 14: i4–8, 2004. doi:10.1007/s10286-
004-1002-4. 624. Wright AD, Smirl JD, Bryk K, van Donkelaar P. Systolic and diastolic
regulation of the cerebral pressure-flow relationship differentially
611. Morris JN, Heady JA, Raffle PA, Roberts CG, Parks JW. Coronary affected by acute sport-related concussion. Acta Neurochir Suppl
heart-disease and physical activity of work. Lancet 262: 1053–1057, 126: 303–308, 2018. doi:10.1007/978-3-319-65798-1_59.
1953. doi:10.1016/s0140-6736(53)90665-5.
625. Hartman YA, Tillmans LC, Benschop DL, Hermans AN, Nijssen KM,
612. Lambertsen CJ, Owen SG, Wendel H, Stroud MW, Lurie AA, Lochner Eijsvogels TM, Willems P, Tack CJ, Hopman MT, Claassen J,
W, Clark GF. Respiratory and cerebral circulatory control during Thijssen DH. Long-term and acute benefits of reduced sitting on
exercise at .21 and 2.0 atmospheres inspired PO2. J Appl Physiol 14: vascular flow and function. Med Sci Sports Exerc 53: 341–350,
966–982, 1959. doi:10.1152/jappl.1959.14.6.966. 2021. doi:10.1249/MSS.0000000000002462.
613. Ekelund U, Steene-Johannessen J, Brown WJ, Fagerland MW, 626. Healy GN, Winkler EA, Brakenridge CL, Reeves MM, Eakin EG.
Owen N, Powell KE, Bauman A, Lee IM, Lancet Physical Activity Accelerometer-derived sedentary and physical activity time in over-
Series 2 Executive Committee, Lancet Sedentary Behaviour weight/obese adults with type 2 diabetes: cross-sectional associa-
Working Group. Does physical activity attenuate, or even eliminate, tions with cardiometabolic biomarkers. PLoS One 10: e0119140,
the detrimental association of sitting time with mortality? A harmon- 2015. doi:10.1371/journal.pone.0119140.
ised meta-analysis of data from more than 1 million men and women.
Lancet 388: 1302–1310, 2016. doi:10.1016/S0140-6736(16)30370-1. 627. Dunstan DW, Kingwell BA, Larsen R, Healy GN, Cerin E, Hamilton MT,
Shaw JE, Bertovic DA, Zimmet PZ, Salmon J, Owen N. Breaking up pro-
614. van der Ploeg HP, Chey T, Korda RJ, Banks E, Bauman A. Sitting longed sitting reduces postprandial glucose and insulin responses.
time and all-cause mortality risk in 222 497 Australian adults. Arch Diabetes Care 35: 976–983, 2012. doi:10.2337/dc11-1931.

1558 Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.
CEREBRAL AUTOREGULATION

628. Smith KJ, Ainslie PN. Regulation of cerebral blood flow and metabolism 642. Lehmann N, Villringer A, Taubert M. Colocalized white matter plas-
during exercise. Exp Physiol 102: 1356–1371, 2017. doi:10.1113/EP086249. ticity and increased cerebral blood flow mediate the beneficial effect
of cardiovascular exercise on long-term motor learning. J Neurosci
629. Siebenmann C, Sorensen H, Jacobs RA, Haider T, Rasmussen P, 40: 2416–2429, 2020. doi:10.1523/JNEUROSCI.2310-19.2020.
Lundby C. Hypocapnia during hypoxic exercise and its impact on
cerebral oxygenation, ventilation and maximal whole body O2 643. Chapman SB, Aslan S, Spence JS, Keebler MW, DeFina LF,
uptake. Respir Physiol Neurobiol 185: 461–467, 2013. doi:10.1016/j. Didehbani N, Perez AM, Lu H. D'Esposito S. Distinct brain and be-
resp.2012.08.012. havioral benefits from cognitive vs. physical training: a randomized
trial in aging adults. Front Hum Neurosci 10: 338, 2016. doi:10.3389/
630. Olin JT, Dimmen AC, Subudhi AW, Roach RC. Cerebral blood flow fnhum.2016.00338.
and oxygenation at maximal exercise: the effect of clamping carbon
dioxide. Respir Physiol Neurobiol 175: 176–180, 2011. doi:10.1016/j. 644. Alfini AJ, Weiss LR, Leitner BP, Smith TJ, Hagberg JM, Smith JC.
resp.2010.09.011. Hippocampal and cerebral blood flow after exercise cessation in
master athletes. Front Aging Neurosci 8: 184, 2016. doi:10.3389/
631. Braz ID, Fisher JP. The impact of age on cerebral perfusion, oxygen- fnagi.2016.00184.
ation and metabolism during exercise in humans. J Physiol 594:
4471–4483, 2016. doi:10.1113/JP271081. 645. Robertson AD, Marzolini S, Middleton LE, Basile VS, Oh PI,
MacIntosh BJ. Exercise training increases parietal lobe cerebral
632. Ogoh S, Brothers RM, Barnes Q, Eubank WL, Hawkins MN, blood flow in chronic stroke: An observational study. Front Aging
Purkayastha S, O-Yurvati A, Raven PB. The effect of changes in car- Neurosci 9: 318, 2017. doi:10.3389/fnagi.2017.00318.
diac output on middle cerebral artery mean blood velocity at rest
and during exercise. J Physiol 569: 697–704, 2005. doi:10.1113/ 646. Alfini AJ, Weiss LR, Nielson KA, Verber MD, Smith JC. Resting cere-
jphysiol.2005.095836. bral blood flow after exercise training in mild cognitive impairment. J
Alzheimers Dis 67: 671–684, 2019. doi:10.3233/JAD-180728.
633. Smirl JD, Haykowsky MJ, Nelson MD, Altamirano-Diaz LA, Ainslie
PN. Resting and exercise cerebral blood flow in long-term heart 647. Ichikawa D, Miyazawa T, Horiuchi M, Kitama T, Fisher JP, Ogoh S.
transplant recipients. J Heart Lung Transplant 31: 906–908, 2012. Relationship between aerobic endurance training and dynamic cer-
doi:10.1016/j.healun.2012.04.003. ebral blood flow regulation in humans. Scand J Med Sci Sports 23:
e320-329–n/a, 2013. doi:10.1111/sms.12082.
634. Smith KJ, MacLeod D, Willie CK, Lewis NC, Hoiland RL, Ikeda K,
Tymko MM, Donnelly J, Day TA, MacLeod N, Lucas SJ, Ainslie PN. 648. Lind-Holst M, Cotter JD, Helge JW, Boushel R, Augustesen H, Van
Influence of high altitude on cerebral blood flow and fuel utilization Lieshout JJ, Pott FC. Cerebral autoregulation dynamics in endur-
during exercise and recovery. J Physiol 592: 5507–5527, 2014. ance-trained individuals. J Appl Physiol 110: 1327–1333, 2011.
doi:10.1113/jphysiol.2014.281212. doi:10.1152/japplphysiol.01497.2010.

635. Quistorff B, Secher NH, Van Lieshout JJ. Lactate fuels the human 649. Perry BG, Cotter JD, Korad S, Lark S, Labrecque L, Brassard P,
brain during exercise. FASEB J 22: 3443–3449, 2008. doi:10.1096/ Paquette M, Le Blanc O, Lucas SJ. Implications of habitual endur-
fj.08-106104. ance and resistance exercise for dynamic cerebral autoregulation.
Exp Physiol 104: 1780–1789, 2019. doi:10.1113/EP087675.
636. Fisher JP, Hartwich D, Seifert T, Olesen ND, McNulty CL, Nielsen
HB, van Lieshout JJ, Secher NH. Cerebral perfusion, oxygenation 650. Drapeau A, Labrecque L, Imhoff S, Paquette M, Le Blanc O,
and metabolism during exercise in young and elderly individuals. J Malenfant S, Brassard P. Six weeks of high-intensity interval training
Physiol 591: 1859–1870, 2013. doi:10.1113/jphysiol.2012.244905. to exhaustion attenuates dynamic cerebral autoregulation without
influencing resting cerebral blood velocity in young fit men. Physiol
637. Fisher JP, Ogoh S, Young CN, Raven PB, Fadel PJ. Regulation of Rep 7: e14185, 2019. doi:10.14814/phy2.14185.
middle cerebral artery blood velocity during dynamic exercise in
humans: influence of aging. J Appl Physiol 105: 266–273, 2008. 651. Tsukamoto H, Hashimoto T, Olesen ND, Petersen LG, Sorensen H,
doi:10.1152/japplphysiol.00118.2008. Nielsen HB, Secher NH, Ogoh S. Dynamic cerebral autoregulation is
maintained during high-intensity interval exercise. Med Sci Sports
638. Fluck D, Beaudin AE, Steinback CD, Kumarpillai G, Shobha N, Exerc 51: 372–378, 2019. doi:10.1249/MSS.0000000000001792.
McCreary CR, Peca S, Smith EE, Poulin MJ. Effects of aging on the
association between cerebrovascular responses to visual stimula- 652. Lewis N, Gelinas JC, Ainslie PN, Smirl JD, Agar G, Melzer B, Rolf JD,
tion, hypercapnia and arterial stiffness. Front Physiol 5: 49, 2014. Eves ND. Cerebrovascular function in patients with chronic obstruc-
doi:10.3389/fphys.2014.00049. tive pulmonary disease: The impact of exercise training. Am J
Physiol Heart Circ Physiol 316: H380–H391, 2019. doi:10.1152/
639. Duzel E, van Praag H, Sendtner M. Can physical exercise in old age ajpheart.00348.2018.
improve memory and hippocampal function? Brain 139: 662–673,
2016. doi:10.1093/brain/awv407. 653. Kim TW, Park SS, Park JY, Park HS. Infusion of plasma from exer-
cised mice ameliorates cognitive dysfunction by increasing hippo-
640. Erickson KI, Hillman C, Stillman CM, Ballard RM, Bloodgood B, campal neuroplasticity and mitochondrial functions in 3xTg-AD
Conroy DE, Macko R, Marquez DX, Petruzzello SJ, Powell KE, for mice. Int J Mol Sci 21: 3291, 2020. doi:10.3390/ijms21093291.
Physical Activity Guidelines Advisory Council. Physical activity, cog-
nition, and brain outcomes: a review of the 2018 Physical Activity 654. Bliss ES, Wong RH, Howe PR, Mills DE. Benefits of exercise training
Guidelines. Med Sci Sports Exerc 51: 1242–1251, 2019. doi:10.1249/ on cerebrovascular and cognitive function in ageing. J Cereb Blood
MSS.0000000000001936. Flow Metab 41: 447–470, 2021. doi:10.1177/0271678X20957807.

641. Kleinloog JP, Mensink RP, Ivanov D, Adam JJ, Uludag K, Joris PJ. 655. Stillman CM, Esteban-Cornejo I, Brown B, Bender CM, Erickson KI.
Aerobic exercise training improves cerebral blood flow and executive Effects of exercise on brain and cognition across age groups and
function: a randomized, controlled cross-over trial in sedentary older health states. Trends Neurosci 43: 533–543, 2020. doi:10.1016/j.
men. Front Aging Neurosci 11: 333, 2019. doi:10.3389/fnagi.2019.00333. tins.2020.04.010.

Physiol Rev  VOL 101  OCTOBER 2021  www.prv.org 1559


Downloaded from journals.physiology.org/journal/physrev (181.141.214.118) on June 1, 2023.

You might also like