ArticlePDF Available

Lessons from altitude: Cerebral perfusion insights and their potential clinical significance

Wiley
Experimental Physiology
Authors:

Abstract

New Findings What is the topic of this review? The long‐held assumption that transcranial Doppler middle cerebral artery velocity is a surrogate for cerebral blood flow has been questioned in certain circumstances, particularly where tissue oxygenation changes. What advances does it highlight? Cerebral venous outflow restriction appears to be implicated in the development of high‐altitude cerebral oedema. Rapid ascent to high altitude commonly results in acute mountain sickness and, on occasion, potentially fatal high‐altitude cerebral oedema. The exact pathophysiological mechanisms behind these syndromes remain to be determined. One of the main theories to explain the development of acute mountain sickness is an increase in intracranial pressure. Vasogenic (extracellular water accumulation attributable to increased permeability of the blood–brain barrier) and cytotoxic (intracellular) oedema have also been postulated as potential mechanisms that underlie high‐altitude cerebral oedema. Recently published findings derived from a very challenging field study (obtained at altitudes of up to 7950 m), substantiated by sea‐level hypoxic magnetic resonance angiography studies, have given new insights into the maintenance of cerebral blood flow at altitude. This report provides new perspectives and potential mechanisms to account for the maintenance of cerebral oxygen delivery at high and extreme altitude. In particular, the long‐held assumption that transcranial Doppler middle cerebral artery velocity is a surrogate for cerebral blood flow has been shown to be incorrect in certain circumstances. The emerging evidence for a potential third mechanism, namely the restrictive venous outflow hypothesis, in the development of high‐altitude cerebral oedema, over and above the accepted vasogenic and cytotoxic hypotheses, is also appraised.
Experimental Physiology
Exp Physiol 000.0 (2016) pp 1–6 1
Symposium Report
Symposium Report
Lessons from altitude: cerebral perfusion insights
and their potential translational clinical significance
Chris Imray
1,2,3
1
Department of Vascular Surgery, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
2
Warwick Medical School, Warwick University, Coventry, UK
3
Coventry University, Coventry, UK
New Findings
r
What is the topic of this review?
The long-held assumption that transcranial Doppler middle cerebral artery velocity is a
surrogate for cerebral blood flow has been questioned in certain circumstances, particularly
where tissue o xygenation changes.
r
What advances does it highlight?
Cerebral venous outflow restriction appears to be implicated in the development of
high-altitude cerebral oedema.
Rapid ascent to high altitude commonly results in acute mountain sickness and, on occasion,
potentially fatal high-altitude cerebral oedema. The exact pathophysiological mechanisms
behind these syndromes remain to be determined. One of the main theories to explain the
development of acute mountain sickness is an increase in intracranial pressure. Vasogenic
(extracellular water accumulation attributable to increased permeability of the blood–brain
barr i er) and cytotoxic (intracellular) oedema have also been postulated as potential mechanisms
that underlie high-altitude cerebral oedema. Recently published finding s derived from a very
challenging field study (obtained at altitudes of up to 7950 m), substantiated by sea-level hypoxic
magnetic resonance angiography studies, have given new insights into the maintenance of
cerebral blood flow at altitude. This report provides new perspectives and p otential mechanisms
to account for the maintenance of cerebral oxygen delivery at high and extreme altitude. In
particular, the long-held assumption that transcranial Doppler middle cerebral artery velocity
is a surrogate for cerebral blood flow has been shown to be incorrect in certain circumstances.
The emerging evidence for a potential third mechanism, namely the restrictive venous outflow
hypothesis, in the development of high-altitude cerebral oedema, over and above the accepted
vasogenic and cytotoxic hypotheses, is also appraised.
(Received 4 January 2016; accepted after revision 30 March 2016; first published online 8 April 2016)
Corresponding author C.H.E Imray: Department of Vascular Surgery, University Hospitals Coventry and Warwickshire
NHS Trust, Coventry CV2 2DX, UK. Email: chrisimray@aol.com
Altitude as a model for critical illness
Adapting knowledge derived from basic science research
that is relevant to clinical medicine is the goal of
translational research (Imray et al. 2015). Studies
of healthy individuals under extreme physiological
stress (such as extreme heat, cold or hypoxia) have
provided insigh ts into some of the basic but novel
pathophysiological mechanisms reported in acute severe
life-threatening illnesses. The response of healthy humans
to extreme altitude might be a useful model for hypoxic
patients in intensive care, and this model has recently
been explored extensively (Swenson et al. 2014).
C
2016 The Authors. Experimental Physiology
C
2016 The Physiological Society DOI: 10.1113/EP085813
2 Exp Physiol 000.0 (2016) pp 1–6
C. Imray
Vascular disease is a common and growing feature
of Western s ociety and accounts for a very significant
disease burden (Yusuf et al. 2001). Carotid artery disease
is common and results in significant morbidity and
mortality (Johnston et al. 2009). One of the pathological
processes observe d in carotid artery disease involves
narrowing of the internal carotid by atheromatous
plaque, and this can result in subsequent plaque rupture,
with downstream embolization. This can lead to focal
neurological events, such as transient ischaemic attacks
or stroke (Imray & Tiivas, 2005). Cerebral microemboli
can be detected using transcranial Doppler (TCD), and
thecerebralembolicloadappearstoberelatedtotherisk
of subsequent stroke (Molloy & Markus, 1999). Emboli
tend to be mainly platelet in origin and so may be
controllable with oral or intravenous antiplatelet agents
(van Dellen et al. 2008). These non-invasive portable
ultrasound techniques have subsequently proved to be
practical in the field.
Cerebral perfusion on ascent to altitude
The barometric pressure falls with increasing altitude
and with it also the partial pressure of atmospheric and
inspired oxygen. Acclimatization to this environmental
hypobaric hypoxic stress involves a number of adaptive
processes, including hyperventilation and an increase in
haematocrit, which tend to restore arterial oxygen content
toward sea level values (Imray et al. 2011). In addition,
increased cerebral blood flow (CBF) is believed to be one
compensatory mechanism ser ving to maintain normal
ox ygen flux to the br ain in the face of arterial hypoxaemia.
Although other measures of cerebral perfusion, such as
nitrous oxide washout (Severinghaus et al. 1966) and
xenon (Jensen et al. 1985), have been used in the past,
TCD has the attraction of being both non-invasive and
portable, making it a potentially attractive investigative
tool for altitude studies. The technique uses a 2 MHz
probe focused at 5 cm to measure the velocity of the
blood within the middle cerebra l artery (MCA).
Over the years, the term ‘flow’ velocity has been widely
incorporated, and makes the assumption that there is no
change in the MCA diameter (and so velocity would be
an accurate surrogate of flow; Serrador et al. 2000). The
TCD measurement of ‘flow velocity’ in the MCA has been
extensively used to assess CBF dynamics both at rest and
during exercise at altitude, and this was reviewed by Wilson
et al. (2009). However, the assumption that velocity is
a surrogate for flow has been disputed (Giller, 2003).
Furthermore, an opposite or contradictory assumption
is made in many clinical situations; in the management
of subarachnoid hemorrhage, for example, changes in
TCD-derived blood velocity are assumed to represent
changes in vessel diameter (vasospasm). In the past, the
measurement of MCA diameter has only been possible
by direct vision at surgery (Giller et al. 1993), by use
of contrast angiography (Du Boulay & Symon, 1971) or
magnetic resonance angiography (Valdueza et al. 1997);
techniques inappropriate for field studies.
Transcranial colour Doppler power signal has
previously been used to infer MCA cross-sectional area
indirectly in a laboratory setting (Poulin, 1996). The
recent development of portable ultrasound devices that
incorporate both two-dimensional colour flow mapping
and concurrent pulse wave Doppler ultrasonography
permits measurement of both the vessel diameter and
the velocity of the blood within it. The two-dimensional
ultrasound ensures that the same segment of the artery
can be reliably visualized and assessed. This approach has
nove l utility, but there are potential serious theoretical
limitations with the use of colour Doppler when
measuring vessel diameter, including the influence of gain
settings. To that end, a validation study directly comparing
transcranial colour Doppler and magnetic resonance
angiog raphy was performed (Wilson et al. 2011a), and
transcranial Doppler and magnetic resonance imaging
(MRI)-measured vessel diameters correlated closely [r =
0.82 (Pearson’s), r
2
= 0.67].
Using transcr anial colour Doppler in the first published
field study of cerebral perfusion above 5500 m (at 7950 m),
it was found that exposure to extreme hypobaric hypoxia
was associated with an increase in MCA diameter
(Wilson et al. 2011a). The MCA dilatation was rapidly
reversed by inhaled supplemental oxygen. These field
transcranial colour Doppler findings have subsequently
been replicated using mag netic resonance angiography
in acute hypoxia at sea level. The increased diameter,
as opposed to increased blood velocity, was the major
factor increasing CBF and maintaining cerebral oxygen
delivery at extreme altitude (>5300 m). Others have
independently confirmed these observations in bulk flow
within extracranial vessels internal carotid artery (i.e.
internal carotid artery; Willie et al. 2012; Ogoh et al.
2013). Middle cerebral artery vasodilatation may have
implications for the pathogenesis of cerebral high-altitude
illness and the acclimatization process. Future studies
inferri ng CBF from TCD velocity measurements at
altitude and clinical studies where oxygenation may
change should take vessel caliber into account.
It is important to acknowledge that the culmination of
the findings presented challenge current dogma regarding
cerebral vessel function during physiological hypoxia.
More research is needed to define the physiological range
over which cerebral vessel dilatation may occur or in what
populations this may be observed (or not). Putting this
into context, it should be noted that interpretation of
data collected using other methods, and in particular
TCD, may be misleading. For instance, the increase in
flow velocity measured during hypoxia using TCD is not
wrong, but may be an underrepresentation. Researchers
C
2016 The Authors. Experimental Physiology
C
2016 The Physiological Society
Exp Physiol 000.0 (2016) pp 1–6 3
Cerebral perfusion insights and their potential clinical significance
and/or clinicians should be aware of the limitations when
interpreting results that do not take into account possible
changes in MCA diameter.
High-altitude cerebral oedema and current concepts
Ross (1985) proposed that variations in cerebral and spinal
compliance (the ability to accommodate hypoxia- induced
cerebral swelling) between individuals could account for
the ‘random nature’ of the headache that occurs at
altitude (Hackett & Roach, 2001). This formed the basis
of the currently accepted pathogenesis of acute mountain
sickness (AMS). An additional physiological component
of exercise-exacerbated hypoxaemia (that is hypothesized
to worsen cerebral swelling) has been incorporated into
the theory, and likewise, hypoxia during sleep may
compound the situation (Bailey et al. 2006). Free radical
formation, nitric oxide and inflammatory m diators (e.g.
vascular endothelial growth factor) have been proposed as
mechanistic links between hypoxia and oedema for mation
(Wilson et al. 2009). Unfortunately, there is little evidence
to suggest that hypoxia causes cerebral oedema sufficiently
(at least in AMS) to account for the severe headache, and
such swelling does not correlate with headache symptoms
(Bailey et al. 2006; Kall enberg et al. 2007). Evidence
for a rise in intracranial pressure (ICP) is also sparse
(Wilson et al. 2009). Cerebral oedema itself is far more
common in conjunction with high-altitude pulmonary
oedema (Hackett et al. 1998; Fagenholz et al. 2007).
Currently, high-altitude cerebral oedema is a clinical
diagnosis defined as symptoms of AMS plus gait ataxia
or mental status changes, or both ataxia and mental status
changes. The term cerebral oedema’ normally implies an
underlying pathological process that may not be the case
in all patients with this clinical diagnosis. This misnomer
may potentially distra ct from other pathological processes
(including hypoxia itself) that could account for altered
neurology in some people at altitude.
The initial cause of high-altitude headache might
theoretically be venous congestion. This occurs before the
limits of cranial compliance are reached (hence, before a
rise in ICP) and before cerebral oedema forms. A rise in
ICP and vasogenic oedema formation, if and when they
do occur, may well be sequelae to early cerebral venous
congestion.
The retinal vasculature, because of its direct connection,
is often considered to reflect changes in cerebral
vasculature (Patton et al. 2005). A number of studies
have demonstrated retinal artery dilatation/increased
tortuosity and vein distension with hypobaric hypoxia. In
24 subjects ascending to 5300 m, retinal venous distension
correlated with the sum of an individual’s headache scores
during the ascent (Wilson et al. 2013). Although arterial
distension was also noted, there was no correlation with
headache. Willmann et al. (2013) also found evidence
of retinal artery and venous dilatation on ascent to
4559 m, but have questioned the restricted venous
outflow hypothesis because they found no association with
headache.
Initial interest in venous changes with hypoxia resulted
from a pilot study using susceptibility weighted MRI after
3 h of 12% hypoxia (Wilson et al. 2013). The dramatic
increase in venous caliber, however, could have been
an artifact and influenced by the altered susceptibility
weighting (MRI signal characteristics) of deoxygenated
blood. The study was therefore repeated with 1 h of
11% hypoxia using gadolinium (which does not alter
susceptibility in hypoxia) enhanced T1 imaging,andagain,
venous prominence increased with hypoxia.
In a recent hypoxic study (Sagoo et al. 2016), there
is further evidence implicating the venous system in the
development of hig h-altitude cerebral oedema. Subjects
were exposed to 22 h of continuous normobaric hypoxia
(fractional inspired O
2
= 0.12, approximately equivalent
to an altitude of 4400 m) inside a 2 m × 2m× 1.6 m
hypoxic tent. Hypoxic generators were used, and build-up
of carbon dioxide within the tent was controlled with
an air pump, which pumped the tent air through soda
lime scrubbers. Baseline physiological measurements,
venous blood samples and MRI scans were obtained
at normoxia (fractional inspired O
2
= 0.21). Inspired
oxygen and carbon dioxide concentrations were checked
every 15 min for the first 2 h, every 30 min from 2 to
10 h, and then every 2 h from 12 to 22 h. Extended
MRI-compatible tubing and a tight-fitting mask that were
connected to one of the three hypoxic generators enabled
the subjects to remain hypoxic during the MRI studies.
The subjects were scanned at rest and did not undertake
exercise during the study period. Subjects ate and drank
ad libitum,werekeptwellhydratedandsleptatvarious
points between the 12 and 22 h time points.
Thenovelfeatureofthisstudywasthatarterial
inflow, large and small vessel venous outflow, cerebral
oedema [apparent diffusion coefficient (ADC)], brain
parenchymal volumes and intracranial cerebrospinal fluid
volumes were measured repeatedly and simultaneously
over a 22 h period of sustained hypoxia. As a consequence,
the study g ives insights into the time course of both
the adaptive and the potentially maladaptive processes
on acute exposure to a fractional inspired O
2
of 0.12.
The combination of time (including overnight) and a
simulated altitude of 4400 m represents the sort of stimulus
that would be expected to cause a significant level of
acute high-altitude illness. Cerebral oxygen delivery was
maintained by increases in MCA velocity and diameter.
In addition, this study has shown, for the first time,
a possible mechanistic link between hypoxia, increased
arterial inflow, a hypothesized rise in Starling pressures
and the subsequent development of parenchymal brain
oedema and the clinical development of AMS.
C
2016 The Authors. Experimental Physiology
C
2016 The Physiological Society
4 Exp Physiol 000.0 (2016) pp 1–6
C. Imray
This study appears to unify the arterial inflow and
restricted venous outflow hypotheses. With the demon-
strated increase in arterial inflow, there will be an
associated increase in venous outflow. This has previously
been hypothesized and then shown to be associated with
high-altitude headache (Wilson & Imray, 2016; Sagoo et al.
2016).
Small vessel venocompression appeared towards the
endofthisstudyandisprobablyaresultofthe
simultaneous cerebral oedema that was developing.
Exploring this further, and based upon Poiseuille’s Law,
in order to sustain the increased arterial inflow there
must be either an increase in driving pressure or an
increase in venous diameter. Based upon this study, the
change observed in the large intracerebral veins is, at best,
modest, and in fact, there appeared to be an element of
small vessel venous restriction, implying that the increased
CBF seen in hypoxia is maintained by increased driving
pressures. The changes in driving pressures would result
in changes in the Starling pressures, with increases in
the venous capillary hydrostatic pressure. It should be
noted that in this study the ‘venous outflow restric tion’
appears to be associated with smaller venous vessels
rather than the large vessels/anatomical variants described
by Wilson et al. (2013). These postulated changes in
Starling pressures could, in turn, account for early oedema
formation. The observed rise in oedema between 11
and 22 h might be a time-based effect, or alternatively,
the formation of oedema might be exacerbated by 8 h
of recumbency, which would further increase capillary
backpressure.
Late rises in ADC values within the genu and splenium
of the corpus callosum confirm the build-up of regional
water content and are an indirect measure of cerebral
oedema. This supports the findings of Hackett et al.
(1998). We found that the observed increase in brain
parenchymal volumes at 22 h was associated with
decreased brain cerebrospinal fluid volumes (confirming
‘internal validation’ of our methodology of independently
measuring the segmented cerebrospinal fluid and brain
parenchymal volumes) and also a reduction in s mall
venous vessel volumes [susceptibility weighted imaging
(SWI) sequence] at 22 h, further increasing the Starling
pressure driving oedema formation. A decreased ability
to buffer these parenchymal brain volume changes will be
reflected by increases in ICP and, eventually, high-altitude
cerebral oedema (Imray et al. 2011). A possible novel
mechanism that involves local oedema causing a reduction
in postcapillary venous diameters has b een observed,
and the same mechanism may be implicated in raised
ICP from other causes. Interestingly, a correlation was
found between the cumulative Lake Louise score and
an increase in brain white matter volume at 22 h. This
study supports certain aspects of the publication by
Lawley et al. (2014), where they found that an increase
in brain volume (predominantly in the grey matter) at
10 h was associated with a reduction in the cerebral
component of cerebrospinal fluid. They also found a
statistically significant relationship between the change
in ICP (measured non-invasively by MRI) and severity of
AMS after 10 h. We studied our subjects for a further 12 h
and found, as would be predicted, higher levels of AMS
symptoms. In addition, we found a later increase in total
brain parenchymal volume (grey and white matter) at 22
h compared with baseline.
As a consequence, increases in cerebral vessel
diameter, which would potentially augment cerebral
blood flow, might have an important effect on ICP. It
follows that the use of TCD may underestimate the
interpretation/prediction of changes in ICP and have
implications on associated conditions.
Wider clinical relevance
The results of this study may have important
translational implications. Intracranial pressure is the
primary neurological parameter that guides therapy on
neuro-intensive care. Understanding the cerebrovascular
interactions that affect ICP is vital in the management
of both t rauma and stroke patients. The optimization of
cerebral perfusion pressure is determined by ICP (cerebral
perfusion pressure = mean arterial pressure minus ICP).
Intracranial venous outflow pressure closely correlates
with ICP (Johnston & Rowan, 1974).
The effects of carbon dioxide and hyperventilation
on CBF are well known. The effects of hypoxia, and
in particular, the changes that occur over a period of
time, may aid the understanding of the interplay between
chest and intracranial physiology. Hypoxia, for example
secondary to chest sepsis or adult respiratory distress
syndrome, is common in the neurocritical patient (Lee
& Rincon, 2012).
As such, the management of cerebral oxygenation and
carbon dioxide is integr al to good neurocritical care and
is likely to be crucial to optimizing the correct therap eutic
regimen. Hypoxia resulted in parenchymal brain swelling
(Sagoo et al. 2016). Subsequent development of cerebral
oedema was found, which could eventually result in an
increase in ICP. This was attri buted to compression of
the cerebral venous system, restricting venous outflow,
and possibly, a Starling resistor mechanism (Simard et al.
2007). In an animal model, venous hypertension has been
associated with increases in brain volume, and this was
caused by vessel dilatation. Cerebral oedema only occurred
if there was also associated cerebral tissue injury (Cuypers
et al. 1976). Acute hypoxia can cause tissue injury through
a number of different mechanisms, such as direct damage
to basal membrane structures (Miserocchi et al. 2001),
vascular endothelial growth factor (Dorward et al. 2007)
or free radicals (Bailey et al. 2004).
C
2016 The Authors. Experimental Physiology
C
2016 The Physiological Society
Exp Physiol 000.0 (2016) pp 1–6 5
Cerebral perfusion insights and their potential clinical significance
References
Bailey DM, Kleger GR, Holzgraefe M, Ballmer PE & B
¨
artsch P
(2004). Pathophysiological significance of peroxidative
stress, neuronal damage, and membrane permeability in
acute mountain sickness. JApplPhysiol96, 1459–1463.
Bailey DM, Roukens R, Knauth M, Kallenberg K, Christ S,
Mohr A, Genius J, Storch-Hagenlocher B, Meisel F,
McEnenyJ,YoungIS,SteinerT,HessK&B
¨
artsch P (2006).
Free radical-mediated damage to barrier function is not
associated with altered brain morphology in high-altitude
headache. J Cereb Blood Flow Metab 26, 99–111.
Cuypers J, Matakas F & Potolicchio SJ (1976). Effect of central
venous pressure on brain tissue pressure and brain volume.
JNeurosurg45, 89–94.
Dorward DA, Thompson AA, Baillie JK, MacDougall M &
Hirani N (2007). Change in plasma vascular endothelial
growth factor during onset and recovery from acute
mountain sickness. Respir Med 101, 587–594.
Du Boulay GH & Symon L (1971). The anaesthetist’s effect
upon the cerebral arteries. Proc R Soc Med 64, 77–80.
Fagenholz PJ, Murray AF, Gutman JA, Findley JK & Harris NS
(2007). New- onset anxiety disorders at high altitude.
Wilderness Environ Med 18, 312–316.
Giller CA (2003). The Emperor has no clothes: velocity, flow,
and the use of TCD. J Neuroimaging 13, 97–98.
Giller CA, Bowman G, Dyer H, Mootz L & Krippner W (1993).
Cerebral arterial diameters during changes in blood pressure
and carbon dioxide duri ng craniotomy. Neurosurgery 32,
737–741.
Hackett PH & Roach RC (2001). High-altitude illness. NEngl
JMed345, 107–114.
Hackett PH, Yarnell PR, Hill R, Reynard K, Helt J &
McCormick J (1998). High-altitude cerebral edema
evaluated with magnetic resonance imaging: clinical
correlation and pathophysiology. JAMA 280, 1920–1925.
Imray C, Booth A, Wright A & Br a dwell A (2011). Acute
altitude illnesses. BMJ 343, d4943.
Imray CHE, Grocott MPW, Wilson MH, Hughes A & Auerbach
PS (2015). Extreme, expedition and wilder ness medicine.
Lancet 386, 2520–2525.
Imray CHE & Tiivas CAS (2005). Are some strokes pre ventable?
The potential role of transcranial doppler in transient
ischaemic attacks of carotid origin. Lancet Neurol 4, 580–586.
Jensen JB, Wright AD, Lassen NA, Harvey TC, Winterborn
MH, Raichle ME & Bradwell AR (1985). Cerebral blood flow
in acute mountain sickness. JApplPhysiol69, 430–433.
Johnston IH & Rowan JO (1974). Raised intracranial pressure
and cerebral blood flow. 3. Venous outflow tract pressures
and vascular resistances in experimental intracr a nial
hypertension. J Neurol Neurosurg Psychiatry 37, 392–402.
Johnston SC, Mendis S & Mathers CD (2009). Global variation
in stroke burden and mortalit y: estimates from monitoring,
surveillance, and modelling. Lancet Neurol 8, 345–354.
Kallenberg K, Bailey DM, Christ S, Mohr A, Roukens R,
Menold E, Steiner T, B
¨
artsch P & Knauth M (2007).
Magnetic resonance imaging evidence of cytotoxic cerebral
edema in acute mountain sickness. J Cereb Blood Flow Metab
27, 1064–1071.
Lawley JS, Alperin N, Bagci AM, Lee SH, Mullins PG, Oliver SJ
& Macdonald JH (2014). Normobaric hypoxia and
symptoms of acute mountain sickness: elevated brain
volume and intracranial hypertension. Ann Neurol 75,
890–898.
Lee K & Rincon F (2012). Pulmonary complications in
patients with severe brain injury. Crit Care Res Pract 2012,
207247.
Miserocchi G, Passi A, Negrini D, Del Fabbro M & De Luca G
(2001). Pulmonary interstitial pressure and tissue matrix
structure in acute hypoxia. Am J Physiol Lung Cell Mol
Physiol 280, L881–L887.
Molloy J & Markus HS (1999). Asymptomatic embolization
predicts stroke and TIA risk in patients with carotid artery
stenosis. Stroke 30, 1440–1443.
Ogoh S, Sato K, Nakahara H, Okazaki K, Subudhi AW &
Miyamoto T (2013). Effect of acute hypoxia on blood flow in
vertebral and internal carotid arteries. Exp Physiol 98,
692–698.
Patton N, Aslam T, MacGillivray T, Pattie A, Deary IJ &
Dhillon B (2005). Retinal vascular image analysis as a
potential screening tool for cerebrovascular disease: a
rationale based on homology between cerebral and retinal
microvasculatures. JAnat206, 319–348.
Poulin MJ & Robbins PA (1996). Indexes of flow and
cross-sectional area of the middle cerebral artery using
Doppler ultrasound during hypoxia and hypercapnia in
humans. Stroke 27, 2244–2250.
Ross RT (1985). The random nature of cerebral mountain
sickness. Lancet 1, 990–991.
Sagoo RS, Hutchinson CE, Wright A, Handford C, Parsons H,
Sherwood V, Wayte S, Nagaraja S, Ng’Andwe E, Wilson MH
& Imray CH; Birmingham Medical Research and Expedition
Society (2016). Magnetic resonance investigation into the
mechanisms involved in the development of high-altitude
cerebral edema. J Cereb Blood Flow Metab doi:
0271678X15625350. [Epub ahead of print].
Serrador JM, Picot PA, Rutt BK, Shoemaker JK & Bondar RL
(2000). MRI measures of middle cerebral artery diameter in
conscious humans during simulated orthostasis. Stroke 31,
1672–1678.
Severinghaus JW, Chiodi H, Eger EI 2nd, Brandstater B &
Hornbein TF (1966). Cerebral blood flow in man at high
altitude. Role of cerebrospinal fluid pH in normalization of
flow in chronic hypocapnia. Circ Res 19, 274–282.
Simard JM, Kent TA, Chen M, Tarasov KV & Gerzanich V
(2007). Brain oedema in focal ischaemia: molecular
pathophysiology and theoretical implications. Lancet Neurol
6, 258–268.
Swenson ER & Bartsch P (eds.) (2014). High Altitude: Human
Adaptation to Hypoxia.SpringerScience+Business Media,
New York. doi: 10.1007/978-1-4614-8772-2_7.
Valdueza JM, B alzer JO, Villringer A, Vogl TJ, Kutter R &
Einh
¨
aupl KM (1997). Changes in blood flow velocity and
diameter of the middle cerebral artery during
hyperventilation: assessment with MR and transcranial
Doppler sonography. AJNR Am J Neuroradiol 18,
1929–1934.
C
2016 The Authors. Experimental Physiology
C
2016 The Physiological Society
6 Exp Physiol 000.0 (2016) pp 1–6
C. Imray
van Dellen D, Tiivas CA, Jarvi K, Marshall C, Higman DJ &
Imray CH (2008). Transcranial Doppler ultrasonography-
directed intravenous glycoprotein IIb/IIIa receptor antago-
nist therapy to control transient cerebral microemboli before
and after carotid endarterectomy. Br J Surg 95, 709–713.
Willie CK, Macleod DB, Shaw AD, Smith KJ, Tzeng YC, Eves
ND, I keda K, Graham J, Lewis NC, Day TA & Ainslie PN
(2012). Regional brain blood flow in man during acute
changes in arterial blood gases. JPhysiol590, 3261–3275.
Willmann G, Fischer MD, Schommer K, B
¨
artsch P, Gekeler F &
Schatz A (2013). Missing correlation of retinal vessel
diameter with high-altitude headache. Ann Clin Transl
Neurol 1, 59–63.
Wilson MH, Davagnanam I, Holland G, Dattani RS, Tamm A,
Hirani SP, Kolfschoten N, Strycharczuk L, Green C,
Thornton JS, Wright A, Edsell M, Kitchen ND, Sharp DJ,
Ham TE, M urray A, Holloway CJ, Clarke K, Grocott MPW,
Montgomer y H & Imray C; Birmingham Medical Research
Expeditionary Society & Caudwell Xtreme Everest Research
Group (2013). Cerebral venous system and anatomical
predisposition to high-altitude headache. Ann Neurol 73,
381–389.
Wilson MH, Edsell MEG, Davagnanam I, Hirani SP, Martin
DS, Levett DZH, Thornton JS, Golay X, Strycharczuk L,
Newman SP, Montgomery HE, Grocott MPW & Imray
CHE; Caudwell Xtreme Everest Research Group (2011a).
Cerebral artery dilatation maintains cerebral oxygenation at
extreme altitude and in acute hypoxia—an ultrasound and
MRI study. J Cereb Blood Flow Metab 31, 2019–2029.
Wilson MH, Imray CHE & Hargens AR (2011b). The headache
of high altitude and microgravity—similarities with clinical
syndromes of cerebral venous hypertension. High Alt Med
Biol 12, 379–386.
Wilson MH, Newman S & Imray CH (2009). The cerebral
effects of ascent to high altitudes. Lancet Neurol 8,
175–191.
Wilson MH & Imray CHE (2016). The cerebral venous system
and hypoxia. JApplPhysiol120, 244–250.
Yusuf S, Reddy S,
ˆ
Ounpuu S & Anand S (2001). Global
burden of cardiovascular diseases: part I: general
considerations, the epidemiologic transition, risk factors,
and impact of urbanization. Circulation 104,
2746–2753.
Additional information
Competing interests
None declared.
Acknowledgements
I would like to acknowledge Mark Wilson, Alex Wright, the
Birmingham Research Expeditionary Society and the Centre for
Altitude, Space and Extreme Environment Medicine for their
support and input.
C
2016 The Authors. Experimental Physiology
C
2016 The Physiological Society
... Increasing myocardial contractility and systemic vascular resistance, hypocapnia enhances cardiac oxygen demand [19]. Originally, compensatory-oriented mechanisms at HA, thus, may finally reduce tissue oxygen provision [20,21]. Whether imbalances of cardiovascular adjustments at HA may also affect testicular vessels motivated us to investigate the prevalence of VC in subjects intermittently exposed to hypobaric hypoxia. ...
Article
Background Testicular aches have been reported to occur on exposure to high altitude (HA). As a painful expression of venous congestion at the pampiniform plexus, varicocele (VC) might be a consequence of cardiovascular adjustments at HA. Chile’s National Social Security Regulatory Body (SUSESO) asked to evaluate this condition in the running follow-up study “Health effects of exposure to chronic intermittent hypoxia in Chilean mining workers.” Objectives To investigate the prevalence of VC in a population usually shifting between sea level and HA, thereby intermittently being exposed to hypobaric hypoxia. Methodology Miners (n=492) agreed to be examined at their working place by a physician, in the context of a general health survey, for the presence of palpable VC, either visible or not. Among them was a group exposed to low altitude (LA) <2,400 m; n=123; another one, exposed to moderate high altitude (MHA) working 3,050 m; n=70, and a third one exposed to very high altitude (VHA) >3,900 m, n=165. The Chi2 test and Kruskal-Wallis test were used in the descriptive analysis, and logistic regression was applied to evaluate the association of VC with exposure to HA. The Ethics Committee for Research in Human Beings, Faculty of Medicine, University of Chile, approved this project. Results VC prevalence (grades 2 and 3) was found to be 10% at LA, 4.1% at MHA and 16.7% at VHA (p≤0.05). Hemoglobin oxygen saturation (SaO2) was lower, and hemoglobin concentrations were higher in workers with high grade VC at VHA compared with LA and MHA (Wilcoxon tests, p<0.001). Odds ratios (OR) for association of VC with HA were 3.7 (95%CI: 1.26 to 12.3) and 4.06 (95%CI: 1.73 to 11.2) for MHA and VHA, respectively Conclusions Association of VC with HA, a clinically relevant finding for its own sake, may be related to blood volume centralization as mediated by hypobaric hypoxia.
... In the case of cancer there is concomitant increased pressure and decreased electromagnetic fields. During hepatic biopsy, the (Hasiloglu et al., 2012) High-altitude cerebral edema (Imray, 2016) High-altitude and microgravity headache (Wilson et al., 2011) Post-lumbar puncture headache and brain herniation (Kongstad and Grände, 1999 Parkinson's disease (Goldman et al., 2006) Amyotrophic lateral sclerosis (Pupillo et al., 2012) Noise-induced hearing loss (Sun et al., 2015) Goldman et al., 2006Wilson et al., 2011;Levy Nogueira et al., 2015a,b Kongstad andGrände, 1999;McKee et al., 2013;Stein et al., 2014Pupillo et al., 2012Sun et al., 2015 Kongstad andGrände, 1999;Wilson et al., 2011;Hasiloglu et al., 2012;Imray, 2016 Orešković andKlarica, 2011;Sun et al., 2015 Systemic conditions Fluid shifts * (Bloomfield et al., 2016;Johnson and Luks, 2016) Soft tissue changes due to microgravity (Avula, 1994) Fluid shifts* (Bloomfield et al., 2016;Johnson and Luks, 2016) Soft tissue damage due to traumatic and non-traumatic loads (Shoham and Gefen, 2012;Valdez and Balachandran, 2013) End-organ damage due to blood pressure (Safar et al., 2012) Inflammation Schwartz et al., 2008;Levy Nogueira et al., 2016) Cancer (Schwartz et al., 2002;Schwartz, 2004;Levy Nogueira et al., 2016) Avula, 1994Schwartz et al., 2002;Schwartz, 2004;Abolhassani et al., 2008;Safar et al., 2012;Shoham and Gefen, 2012;Valdez and Balachandran, 2013;Bloomfield et al., 2016;Johnson and Luks, 2016;Levy Nogueira et al., 2016Schwartz et al., 2002Schwartz, 2004;Safar et al., 2012;Levy Nogueira et al., 2016 * Fluid shifts: edema, ascites, pleural, pericardial and joint effusion. ** Numbers in square are the related references. ...
Article
Full-text available
Background: Diseases and health conditions have been classified according to anatomical site, etiological, and clinical criteria. Physico-chemical mechanisms underlying the biology of diseases, such as the flow of energy through cells and tissues, have been often overlooked in classification systems. Objective: We propose a conceptual framework toward the development of an energy-oriented classification of diseases, based on the principles of physical chemistry. Methods: A review of literature on the physical chemistry of biological interactions in a number of diseases is traced from the point of view of the fluid and solid mechanics, electricity, and chemistry. Results: We found consistent evidence in literature of decreased and/or increased physical and chemical forces intertwined with biological processes of numerous diseases, which allowed the identification of mechanical, electric and chemical phenotypes of diseases. Discussion: Biological mechanisms of diseases need to be evaluated and integrated into more comprehensive theories that should account with principles of physics and chemistry. A hypothetical model is proposed relating the natural history of diseases to mechanical stress, electric field, and chemical equilibria (ATP) changes. The present perspective toward an innovative disease classification may improve drug-repurposing strategies in the future.
Article
This is a mini-review of potential wearable physiological sensors and algorithms (process and equations) for detection of acute mountain sickness (AMS). Given the emerging status of this effort, the focus of the review is on the current clinical assessment of AMS, known risk factors (environmental, demographic and physiological), and current understanding of AMS pathophysiology. Studies that have examined a range of physiological variables to develop AMS prediction and/or detection algorithms are reviewed to provide insight and potential technological roadmaps for future development of real-time physiological sensors and algorithms to detect AMS. Given the lack of signs and non-specific symptoms associated with AMS, development of wearable physiologic sensors and embedded algorithms to predict in near term or detect established AMS will be challenging. Prior work using SaO2, HR or HRV has not provided the sensitivity and specificity for useful application to predict or detect AMS. Rather than using spot checks as most prior studies have, wearable systems that continuously measure SpO2 and HR are commercially available. Employing other statistical modeling approaches such as general linear and logistic mixed models or time series analysis to these continuously measured variables is the most promising approach for developing algorithms that are sensitive and specific for physiologic prediction or detection of AMS.
Article
Video slideshow introduction to the symposium by symposium organiser Damian M. Bailey can be found here.
Article
Full-text available
We agree with Dr. Basnyat that dehydration in the mountains is a concern, but no compelling data confirm that dehydration has a role in acute mountain sickness. The report he cites demonstrated a correlation between lower water intake and acute mountain sickness, but causality could not be established; persons with acute mountain sickness are often nauseated and therefore have reduced fluid intake. The only well-controlled investigation found no effect of hydration status on the development of acute mountain sickness.1 As for the rate of ascent, 600 m per day may indeed be too rapid for some, whereas for others, 400 m per day is agonizingly slow. Such is the problem with offering general guidelines. We cited the article by Dumont et al. only to acknowledge the uncertainty regarding the lowest effective dose of acetazolamide. Unlike Dr. Ogilvie, we consider the conclusion by Dumont et al. invalid, for reasons that have already been described elsewhere.2,3 In fact, 500 mg of acetazolamide per day is helpful during rapid ascent.2,3 Only studies directly comparing different doses of acetazolamide will be able to establish the optimal dose; the meta-analysis of Dumont et al. does not. Dr. White highlights the risk of dexamethasone abuse. Dexamethasone should not be used for routine prophylaxis, since it does not enhance acclimatization, as acetazolamide does. However, it is useful for those who have an intolerance to acetazolamide, preferably in the setting of rapid ascent to a high altitude with no further ascent until acclimatization has occurred. We agree that its use as a performance-enhancing agent at high altitude is dangerous. In addition, dexamethasone does not prevent high-altitude pulmonary edema, a deadly risk for those who push their limit of acclimatization. In response to Dr. O’Brien: we did not mention nitric oxide as a therapeutic agent for high-altitude pulmonary edema because no clinical advantage over oxygen has yet been demonstrated, and its use is impractical in the field.Oxygen is remarkably effective for the rapid resolution of high-altitude pulmonary edema. Whether the finding of Anand et al.4 that oxygen combined with nitric oxide is more effective than either alone for decreasing pulmonary vascular resistance will translate into a clinical benefit remains unknown. Perhaps the combination will prove useful for the occasional victim who does not have a prompt response to oxygen. Only a clinical trial can answer this question.
Article
Full-text available
Rapid ascent to high altitude commonly results in acute mountain sickness, and on occasion potentially fatal high-altitude cerebral edema. The exact pathophysiological mechanisms behind these syndromes remain to be determined. We report a study in which 12 subjects were exposed to a FiO2 = 0.12 for 22 h and underwent serial magnetic resonance imaging sequences to enable measurement of middle cerebral artery velocity, flow and diameter, and brain parenchymal, cerebrospinal fluid and cerebral venous volumes. Ten subjects completed 22 h and most developed symptoms of acute mountain sickness (mean Lake Louise Score 5.4; p < 0.001 vs. baseline). Cerebral oxygen delivery was maintained by an increase in middle cerebral artery velocity and diameter (first 6 h). There appeared to be venocompression at the level of the small, deep cerebral veins (116 cm3 at 2 h to 97 cm3 at 22 h; p < 0.05). Brain white matter volume increased over the 22-h period (574 ml to 587 ml; p < 0.001) and correlated with cumulative Lake Louise scores at 22 h (p < 0.05). We conclude that cerebral oxygen delivery was maintained by increased arterial inflow and this preceded the development of cerebral edema. Venous outflow restriction appeared to play a contributory role in the formation of cerebral edema, a novel feature that has not been observed previously.
Article
Full-text available
Most hypobaric hypoxia studies have focused on oxygen delivery and therefore cerebral blood inflow. Few have studied venous outflow. However, the volume of blood entering and leaving the skull (approximately 700mls/min) is considerably greater than CSF production (0.35mls/min) or edema formation rates and slight imbalances of in- and out-flow have considerable effects on intracranial pressure (ICP). This dynamic phenomenon is not necessarily appreciated in the currently taught static "Monro-Kellie" doctrine which forms the basis of the "Tight-Fit" hypothesis thought to under lie high altitude headache, acute mountain sickness (AMS) and High Altitude Cerebral Edema (HACE). Investigating both sides of the cerebral circulation was an integral part of the 2007 Xtreme Everest Expedition. The results of the relevant studies performed as part of and subsequent to this expedition are reviewed here. The evidence from recent studies suggests a relative venous outflow insufficiency is an early step in the pathogenesis of high altitude headache. Translation of knowledge gained from high altitude studies is important. Many patients in a critical care environment develop hypoxemia akin to that of high altitude exposure. An inability to drain the hypoxemic induced increase in cerebral blood flow could be an underappreciated regulatory mechanism of intracranial pressure. Copyright © 2015, Journal of Applied Physiology.
Article
Full-text available
Acute altitude illnesses are potentially serious conditions that can affect otherwise fit individuals who ascend too rapidly to altitude. They include high altitude headache, acute mountain sickness, high altitude cerebral oedema, and high altitude pulmonary oedema. The number of people travelling to altitude for work (soldiers, miners, construction workers, and astronomers) or for recreation (skiing, trekking, mountain biking, and climbing) is rising, and increased media attention towards these activities has also raised the profile of altitude related illness. Typical scenarios in which such illness might occur are a family trek to Everest base camp in Nepal (5360 m), a fund raising climb of Mount Kilimanjaro (5895 m), or a tourist visit to Machu Picchu (2430 m). Awareness of potential altitude related problems is important even for healthcare practitioners working at lower altitude, because patients may ask for advice about the safety of a proposed journey and how to prevent illness at altitude.
Article
Full-text available
The most common altitude-related symptom, high-altitude headache (HAH), has recently been suggested to originate from restricted cerebral venous drainage in the presence of increased inflow caused by hypoxia. In support of this novel hypothesis, retinal venous distension was shown to correlate with the degree of HAH. We quantified for the first time retinal vessel diameter changes at 4559 m using infrared fundus images obtained from a state of the art Spectralis™ HRA+OCT with a semiautomatic VesselMap 1® software. High-altitude exposure resulted in altered arterial and venous diameter changes at high altitude, however, independent of headache burden.
Article
Full-text available
Context.-Because of its onset in generally remote environments, high-altitude cerebral edema (HACE) has received little scientific attention. Understanding the pathophysiology might have implications for prevention and treatment of both this disorder and the much more common acute mountain sickness. Objectives.-To identify a clinical imaging correlate for HACE and determine whether the edema is primarily vasogenic or cytotoxic. Design,Case-comparison study. Setting,Community hospitals accessed by helicopter from mountains in Colorado and Alaska, Patients.-A consecutive sample of 9 men with MACE, between 18 and 35 years old, 8 of whom also had pulmonary edema, were studied after evacuation from high-altitude locations; 5 were mountain climbers and 4 were skiers. The control group, matched for age, sex, and altitude exposure, consisted of 3 subjects with high-altitude pulmonary edema only and 3 who had been entirely well at altitude. Four patients with HACE were available for follow-up imaging after complete recovery. Main Outcome Measures.-Magnetic resonance imaging (MRI) of the brain during acute, convalescent, and recovered phases of HACE, and once in controls, immediately after altitude exposure. Results.-Seven of the 9 patients with HACE showed intense T-2 Signal in white matter areas, especially the splenium of the corpus callosum, and no gray matter abnormalities. Control subjects demonstrated no such abnormalities. All patients completely recovered; in the 4 available for follow-up MRI, the changes had resolved entirely. Conclusions.-We conclude that HACE is characterized on MRI by reversible white matter edema, with a predilection for the splenium of the corpus callosum. This finding provides a clinical imaging correlate useful for diagnosis. It also suggests that the predominant mechanism is vasogenic (movement of fluid and protein out of the vascular compartment) and, thus, that the blood-brain barrier may be important in MACE.
Article
Extreme, expedition, and wilderness medicine are modern and rapidly evolving specialties that address the spirit of adventure and exploration. The relevance of and interest in these specialties are changing rapidly to match the underlying activities, which include global exploration, adventure travel, and military deployments. Extreme, expedition, and wilderness medicine share themes of providing best available medical care in the outdoors, especially in austere or remote settings. Early clinical and logistics decision making can often have important effects on subsequent outcomes. There are lessons to be learned from out-of-hospital care, military medicine, humanitarian medicine, and disaster medicine that can inform in-hospital medicine, and vice-versa. The future of extreme, expedition, and wilderness medicine will be defined by both recipients and practitioners, and empirical observations will be transformed by evidence-based practice.
Article
Objective: The study was undertaken to determine whether normobaric hypoxia causes elevated brain volume and intracranial pressure in individuals with symptoms consistent with acute mountain sickness (AMS). Methods: Thirteen males age = (26 (sd 6)) years were exposed to normobaric hypoxia (12% O2 ) and normoxia (21% O2 ). After 2 and 10 hours, AMS symptoms were assessed alongside ventricular and venous vessel volumes, cerebral blood flow, regional brain volumes, and intracranial pressure, using high-resolution magnetic resonance imaging. Results: In normoxia, neither lateral ventricular volume (R(2) = 0.07, p = 0.40) nor predominance of unilateral transverse venous sinus drainage (R(2) = 0.07, p = 0.45) was related to AMS symptoms. Furthermore, despite an increase in cerebral blood flow after 2 hours of hypoxia (hypoxia vs normoxia: Δ148ml/min(-1) , 95% confidence interval [CI] = 58 to 238), by 10 hours, when AMS symptoms had developed, cerebral blood flow was normal (Δ-51ml/min(-1) , 95% CI = -141 to 39). Conversely, at 10 hours brain volume was increased (Δ59ml, 95% CI = 8 to 110), predominantly due to an increase in gray matter volume (Δ73ml, 95% CI = 25 to 120). Therefore, cerebral spinal fluid volume was decreased (Δ-40ml, 95% CI = -67 to -14). The intracranial pressure response to hypoxia varied between individuals, and as hypothesized, the most AMS-symptomatic participants had the largest increases in intracranial pressure (AMS present, Δ7mmHg, 95% CI = -2.5 to 17.3; AMS not present, Δ-1mmHg, 95% CI = -3.3 to 0.5). Consequently, there was a significant relationship between the change in intracranial pressure and AMS symptom severity (R(2) = 0.71, p = 0.002). Interpretation: The data provide the strongest evidence to date to support the hypothesis that the "random" nature of AMS symptomology is explained by a variable intracranial pressure response to hypoxia.
Chapter
It should be noted that Dr. J.W. Severinghaus was the expert author of this chapter (Cerebral circulation at high altitude) in the First Edition of this textbook. Moreover, in updating this area, we also amalgamate from the First Edition the detailed chapter titled The High-Altitude Brain, which was authored by Drs. M.S. Raichle and T.F. Hornbein. Readers are directed to these original and elegant comprehensive reviews (Severinghaus: High altitude; exploration of human adaptation, New York, Basel; Raichle: High altitude; exploration of human adaptation, New York, Basel). Rather than reproduce this information here, and while summarizing some of this original material in the context of new findings within the last decade, we provide an update within the broad topic of the cerebral circulation and brain at high altitude. This chapter is comprised of seven sections. The introduction is followed by the major factors which regulate cerebral blood flow (CBF) are initially presented in order to emphasize the integrated mechanisms by which CBF is controlled. Next, detailed discussion is provided to examine the influence of exposure to high-altitude exposure on these aforementioned mechanisms which regulate CBF. We then briefly review recent advances in the understanding of neurological clinical syndromes that occur on exposure to high altitudes. The next two sections summarize the influence of high altitude on cognitive function and highlight other neurological-based symptoms and events that can occur upon exposure to high altitude. Finally, we suggest avenues for future research. © 2014 Springer Science+Business Media New York. All rights reserved.