Anda di halaman 1dari 7

Mechanisms of cerebral edema in traumatic brain injury:

therapeutic developments
James J. Donkina and Robert Vinkb
a
Department of Pathology and Laboratory Medicine, Purpose of review
University of British Columbia, Vancouver BC, Canada
and bThe Discipline of Anatomy and Pathology,
Although a number of factors contribute to the high mortality and morbidity associated with
University of Adelaide, Adelaide SA, Australia traumatic brain injury (TBI), the development of cerebral edema with brain swelling remains
Correspondence to James Donkin, PhD, Department the most significant predictor of outcome. The present review summarizes the most recent
of Pathology and Laboratory Medicine, University of advances in the understanding of mechanisms associated with development of
British Columbia, Child and Family Research Institute,
950 West 28th Avenue, Vancouver, BC V5Z4H4, posttraumatic cerebral edema, and highlights areas of therapeutic promise.
Canada Recent findings
Tel: +1 604 875 2345x7146; fax: +1 604 875 3120;
e-mail: jdonkin@cmmt.ubc.ca Despite the predominance of cytotoxic (or cellular) edema in the first week after
traumatic brain injury, brain swelling can only occur with addition of water to the cranial
Current Opinion in Neurology 2010, 23:293–299
vault from the vasculature. As such, regulation of blood–brain barrier permeability has
become a focus of recent research seeking to manage brain edema. Aquaporins, matrix
metalloproteinases and vasoactive inflammatory agents have emerged as potential
mediators of cerebral edema following traumatic brain injury. In particular, kinins
(bradykinins) and tachykinins (substance P) seem to play an active physiological role in
modulating blood–brain barrier permeability after trauma. Substance P neurokinin-1
receptor antagonists show particular promise as novel therapeutic agents.
Summary
Attenuating blood–brain barrier permeability has become a promising approach to
managing brain edema and associated swelling given that increases in cranial water
content can only be derived from the vasculature. Inflammation, both classical and
neurogenic, offers a number of attractive targets.

Keywords
aquaporins, cytotoxic edema, neurogenic inflammation, neurotrauma, neurovascular
unit, trauma, vasogenic edema

Curr Opin Neurol 23:293–299


ß 2010 Wolters Kluwer Health | Lippincott Williams & Wilkins
1350-7540

brain centres such as those involved in respiration and


Introduction cardiac function.
The mechanisms associated with the development of
tissue damage following traumatic brain injury (TBI) Although interventions targeting brain edema and swel-
have been extensively studied over the past few decades ling have existed for some time, therapies have not
and it has become increasingly evident that the for- changed in over 50 years, largely because the mechanisms
mation of cerebral edema is one of the major factors associated with edema development are incompletely
leading to the high mortality and morbidity in affected understood. Accordingly, treatments have focussed on
individuals. Indeed, some studies have reported that management of the symptoms rather than control of the
cerebral edema may account for up to half of the mechanisms. Recently, significant progress has been
mortality in all victims of TBI [1], and in younger victims made toward identifying factors that mechanistically
of TBI, up to half of all mortality and morbidity [2]. contribute to edema formation after TBI. The present
Edema is harmful because it causes cell swelling, swel- review will summarize current understanding of edema
ling that alters cellular metabolite concentration and formation following TBI before considering recently
therefore cellular physiology, biochemistry and function. identified factors that contribute to the process of
When the swelling involves not only the cells themselves edema development.
but also the tissue parenchyma, there is a rapid increase
in intracranial pressure (ICP), which results in com-
pression of blood vessels, reduced tissue blood flow, Classification of edema
reduced oxygenation and eventually shifts tissue down It has long been established that cerebral edema can be
pressure gradients (herniations) that may crush vital classified into two main categories, namely cytotoxic (also
1350-7540 ß 2010 Wolters Kluwer Health | Lippincott Williams & Wilkins DOI:10.1097/WCO.0b013e328337f451

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
294 Inflammatory diseases and infection

known as cellular) edema or vasogenic edema [3]. Cyto- Figure 1 Schematic demonstrating cytotoxic and vasogenic
toxic edema is characterized by an increase in water cerebral edema
content within the intracellular compartment in response
to an osmotic gradient. It is usually associated with a
failure of the ATP-dependent Naþ/Kþ-pumps under
conditions of energy failure typically observed in cerebral
ischemia, anoxic-ischemic encephalopathy and severe
TBI. This leads to an increase in cellular ionic content,
an overall increase in cell osmolality and the influx of
water into the cells. It is essentially a compartment shift
of water in the skull, with water shifting from the extra-
cellular to the intracellular compartment (Fig. 1). As such,
cytotoxic edema of itself does not result in an increase in
brain water content or brain swelling, and no rise in ICP.
It does however, adversely impact on cellular function by
altering intracellular metabolite concentration.

The inability of cytotoxic edema to cause brain swelling


is not always readily apparent and is perhaps best illus-
trated using an example as originally described by Simard
et al. [4]. If a piece of tissue is excised from a live brain, it
will show all the typical signs of cytotoxic edema such as
shifts in ionic and water content between the extracellu-
lar and intracellular compartments. However, over time,
that excised piece of tissue will not gain ionic content,
will not gain water content and will not swell. There
simply is no source for the ions and water; these can only
come from the vasculature. Swelling, and any associated
increase in ICP, therefore requires a vascular contribution
and active blood flow.

A vascular contribution is the hallmark of vasogenic


edema. By definition, vasogenic edema is the result of
the movement of water from the vasculature to the
extracellular space in response to an osmotic gradient
generated by the leakage of vascular components into
the brain parenchyma (Fig. 1). It is characterized by an
open blood–brain barrier (BBB) typically observed in
conditions such as TBI, brain tumours, infection, intra-
cerebral hemorrhage and inflammation. Given that
vasogenic edema results in an increase in brain water Cytotoxic edema is essentially a water compartment shift with no change
content, tissue swelling and an increase in ICP will in tissue water content or volume. In contrast, vasogenic edema
be observed. increases tissue water content, leading to swelling. Tissue swelling thus
requires a vascular contribution if it is to occur.

Variants of these two major forms of edema have also


been described for specific situations. For example, of water from the vasculature to the brain interstitial fluid
transependymal edema describes an increase in periven- in the absence of gross disruption of the BBB. Despite
tricular interstitial fluid due to a failure of the ependymal these distinct classifications of cerebral edema, in most
lining of the ventricular wall, common in obstructive or clinical situations there is a combination of different
communicating hydrocephalus. Hydrostatic edema is a types of edema depending on the disorder and time
variant of vasogenic edema that occurs when cerebral course of the disease [5].
perfusion pressure increases to a level at which autore-
gulatory mechanisms break down. This type of edema is
observed in hypertensive encephalopathies. Finally, Edema in traumatic brain injury
osmotic (or ionic) edema occurs when plasma osmolality Debate over which type of edema predominates in TBI
falls below brain osmolality and there is a net movement has persisted for a number of decades. Early studies

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Mechanisms of cerebral edema in TBI Donkin and Vink 295

proposed that vasogenic edema from BBB opening was cellular compartment would then indirectly drive the
the main contributor following injury [6,7], although this entry of more ions and water from the vasculature, with
conclusion was largely based on a simplistic, cryogenic this entry being facilitated by the BBB being permeable
model of traumatic injury that is limited in terms of to ions and small molecules, albeit not to the larger
replicating many of the features associated with clinical plasma proteins commonly used to measure BBB per-
TBI. It is now recognized that TBI is a complex and meability. Thus, the ‘pure’ vasogenic phase would be
heterogenous injury and more recent experimental replaced by a mixed cytotoxic/vasogenic phase that
models attempt to reproduce as many of the features would be dominated by the cytotoxic, or cellular, com-
of clinical TBI as possible, including associated second- ponent as more cells become dysfunctional and die.
ary conditions such as arterial hypotension, hypoxia or Nonetheless, the driving force for the increased brain
ischemia. Using these more recent experimental models, water content, brain swelling and increased ICP, would
a biphasic profile encompassing both vasogenic and cyto- be the vascular contribution. Thus, interventions that
toxic components has emerged. With the aid of novel target the vascular contribution to edema, even if the
MRI techniques, vasogenic edema, as indicated by an dominant edema is cellular, may be particularly effective
increased water diffusion distance, was demonstrated to in the management of brain swelling.
occur in the first few hours after TBI [8,9], followed by
cytotoxic edema that developed more slowly over the
next few days and persisted for up to 2 weeks [8]. These Mediators of brain edema
observations, based on water diffusion distance, were A number of mediators have been identified that play a
confirmed by BBB permeability measurements demon- role in edema formation after TBI. Arguably, the most
strating that the barrier was open to large plasma proteins exciting recent developments include the identification
for only a few hours after TBI [10,11]. However, the BBB of aquaporin water channels as critical participants in the
does not simply close after this initial opening, with development of edema, and the focus on agents that
recent evidence suggesting that the BBB closes gradu- affect the BBB, and therefore the vascular contribution to
ally, with the smaller vascular components being per- brain swelling. These aspects are summarized below.
meable for up to 7 days after TBI [12]. What accounts for
this persistent permeability is unclear, although posttrau- Aquaporins
matic alterations to the endothelial cytoskeleton promot- The identification of the water-channel proteins, aqua-
ing endothelial barrier opening have been implicated porins (AQPs), as a key player in the development and
[13]. Thus, the BBB is maximally permeable at 4–6 h resolution of cerebral edema has highlighted their poten-
after TBI, before commencing to close and becoming tial as a therapeutic target to prevent brain swelling [16–
differentially permeable to smaller molecules over a 7- 18]. AQPs are integral membrane proteins belonging to a
day period. family that form pores in the membranes of mammalian
cells [19]. Of the 13 AQPs known to exist in mammals,
Given the critical role that the vascular contribution plays AQP1, AQP4 and AQP9 are highly expressed in brain
in brain water content and ICP changes, it is clear that an [20]. AQP4 is predominately expressed in the astrocytic
understanding of BBB changes following TBI, and their end foot processes in close proximity to intracerebral
contribution to edema, is essential to develop potential vessels and at the ventricular interface. AQP9 is co-
interventions. Considerable evidence now exists support- expressed with AQP4 in astrocytic foot processes,
ing that brain water content after TBI is maximal at 2–3 whereas AQP1 is expressed in the choroid plexus epi-
days after trauma [1], which is also the point at which ICP thelium and in ganglionar sensory neurons. Other AQPs
usually peaks. For brain water content and swelling to be have also been identified in brain, but these are expressed
maximal at this time point, there must still be an active at much lower concentrations [20].
vascular contribution despite the BBB being closed to
large plasma molecules after 6 h. Although a second A number of studies have now shown that AQP4 expres-
opening of the BBB has been mooted [14], such an event sion is markedly altered in both experimental and clinical
is not essential given the gradual closing of the barrier to brain injury [5,17,21,22], and that genetic variation of the
smaller vascular molecules over time. We therefore pro- channels may influence degree of edema [23]. Similar
pose that the initial transient opening of the BBB is increases in AQP1 and AQP9 after experimental TBI
associated with a brief period of ‘pure’ vasogenic edema, have been reported [24,25]. Initial studies suggested that
the presence of which would be permissive for the upregulation of AQPs after brain injury promoted edema
development of any subsequent cytotoxic edema [15]. formation [16] and it was accordingly postulated that
Cytotoxic edema would indeed develop with the gradual therapeutic inhibition of AQP4 would be beneficial in
development of cellular injury over time, and would edema control [18]. However, it subsequently became
become more prominent as more cells were affected. evident that the alterations in AQP4 expression are
The intracellular shift of ions and water from the extra- regionally distinct and dependent on the type of edema

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
296 Inflammatory diseases and infection

[26–28]. For example, in a model of rat cerebral ischemia, temporally and spatially associated with BBB disruption
inhibition of AQP4 expression was associated with and edema formation [41,42]. Consistent with this, mice
reduction in edema, infarct area and an improvement lacking the MMP-9 gene have been shown to be pro-
in functional outcome [29,30]. Rat cerebral ischemia tected in both focal and global ischemia as well as TBI
typically results in cytotoxic edema. In contrast, vaso- [41,43,44], with gene knockout mice having reduced
genic edema induced by cold lesion injury was exacer- BBB disruption and edema, a reduced inflammatory
bated in AQP4 knockout animals, suggesting that AQP4 response, improved integrity of white matter components
is essential for clearance of vasogenic edema [27]. plus improved functional outcome.

In experimental TBI, an increase in AQP4 expression in MMP inhibitors, such as minocycline or TIMP-1, have
the glia limitans was observed but a downregulation of also been shown to block BBB injury, cerebral edema and
perivascular AQP4 was noted during the early period cell death in a number of experimental animal models
when vasogenic edema would be present [26]. Notably, [35,45–47]. However, recent data suggest a more bipha-
pharmacological reduction of edema formation and sic role for MMPs in TBI [48], with MMPs reported to
improved functional outcome was associated with restor- play an important role in neurogenesis, neurovascular
ation of the AQP4 channels to their normal state. The remodeling and matrix-trophic signaling in the later
speed at which the reappearance of AQP4 channels on stages of recovery from TBI and stroke [39,48]. Inhibition
the perivascular glial endfeet occurred after treatment at these delayed time points may in fact worsen recovery.
suggests that there was a posttranslational modification, As such, the most challenging aspect with respect to
perhaps involving subunit aggregation, rather than MMP inhibitors is the timing of administration in an
enhanced protein synthesis. Similarly, in a rat cortical effort to coordinate their beneficial and detrimental
contusion model, exacerbation of injury by a secondary effects following TBI. This balance between positive
insult involving hypoxia and hypotension led to a wor- and detrimental effects has been recognized for some
sening of brain edema, which was associated with a time in inflammation [49].
reduction in the APQ4 expression [31]. The increased
brain water content can only be attributed to a vascular Vasoactive agents
component as a cytotoxic compartment shift would not It is well established that inflammatory, vasoactive agents
increase brain water content. Thus, AQP4 upregulation is can increase BBB permeability and lead to cerebral
associated with the development of cytotoxic edema edema [50]. Recent studies in TBI have focused not
whereas perivascular downregulation occurs in regions only on mediators related to classical inflammation, but
experiencing vasogenic edema. Generalized inhibition of also those derived from neurogenic inflammation. In
AQP4 channels may therefore not be beneficial in those terms of classical inflammation, the bradykinin family
conditions in which vasogenic edema plays a critical role. of kinins has been strongly implicated in the develop-
Indeed, AQP4 activators have the potential to facilitate ment of edema following acute brain injury [51]. The
the clearance of the vasogenic component of edema, bradykinins are formed from the cleavage of kininogen by
whereas AQP4 inhibitors have the potential to protect kallikreins, with the active peptides (bradykinins and
the brain in cytotoxic edema. kallidin) producing their effects through two subtypes
of bradykinin receptors known as B1 and B2 receptors.
Matrix metalloproteinases Following TBI in mice [52], bradykinin itself was maxi-
The ability of matrix metalloproteinases (MMPs) to mally increased at 2 h after trauma whereas both the B1
degrade many types of extracellular matrix proteins, and B2 receptors were significantly upregulated in the
including the neurovascular basal lamina and tight junc- first 24 h. Despite the increase in both receptors after
tion proteins of the BBB, has been the subject of a trauma, only B2 receptor knockout mice had significantly
number of recent TBI investigations [32–34]. MMPs less edema and better functional outcomes after TBI
are zinc-dependent endopeptidases involved in the pro- [52], implying that B2 receptor binding may play an
cess of tissue remodeling following various pathologic integral role in edema formation after trauma. Adminis-
conditions. The regulation of MMP expression and acti- tration of a B2 receptor antagonist was subsequently
vation is complex and tightly controlled, and loss of this shown to reduce ICP and contusion volume in a rodent
control has been identified as potentially playing a critical focal contusion model [53], which confirmed earlier find-
role in the pathophysiology of synaptic loss and BBB ings in other models of acute brain injury [51,54,55].
breakdown in TBI, stroke and neurodegeneration Despite these positive findings, no positive effects of
[35,36–38]. MMPs, and in particular MMP-2, MMP- the B2 antagonists have been noted in subsequent
3 and MMP-9, are upregulated following TBI [33,39,40] clinical trials of the compounds [56,57].
in which they cause acute disruption of the BBB, leading
to vasogenic edema and subsequent cell death. Indeed, The other distinct family of kinins is the tachykinins, a
the upregulation of MMP-9 in particular has been group of peptide mediators that have been implicated in

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Mechanisms of cerebral edema in TBI Donkin and Vink 297

neurogenic inflammation. Neurogenic inflammation is a of depletion. Nimmo et al. [59] used this approach to
process that encompasses vasodilation, plasma extravasa- demonstrate that neuropeptide depletion results in a
tion and neuronal hypersensitivity caused by the release marked attenuation of early posttraumatic BBB per-
of neuropeptides from sensory neurons [58]. Although meability and any subsequent edema formation. Remark-
several neuropeptides have been implicated in neuro- ing that their study validated the assumption that vaso-
genic inflammation, calcitonin gene-related peptide genic edema is permissive for cytotoxic edema formation
(CGRP) has been identified as being associated with [15], they concluded that early inhibition of neurogenic
the vasodilation whereas substance P is thought to inflammation may present a novel approach to the treat-
enhance plasma protein extravasation. ment of posttraumatic edema formation.

Although few studies have suggested a role for neuro-


genic inflammation in edema formation following TBI Conclusion
[59,60], Donkin et al. [61] recently demonstrated that Although a number of factors contribute to the high
injury results in an elevated perivascular substance P mortality and morbidity associated with TBI, the devel-
immunoreactivity that is associated with enhanced vas- opment of cerebral edema with brain swelling remains the
cular permeability and edema formation in a rodent most significant predictor of outcome. Brain swelling can
model of moderate diffuse TBI. A similar increase in only occur with addition of water from the vasculature
substance P immunoreactivity was also reported follow- (vasogenic edema), as cytotoxic edema is essentially a
ing mild concussive head injury [62] and reperfusion compartment shift of water from the extracellular to intra-
injury [63]. Subsequent studies in human TBI by the cellular compartment. As such, attenuating BBB per-
same group also reported elevated substance P immu- meability has increasingly become a promising approach
noreactivity in human TBI [64]. Specifically, patients to managing brain edema and associated swelling. AQPs,
who had sustained traumatic head injuries, who had died MMPs and vasoactive inflammatory agents have emerged
within 1 week and who had undergone postmortem and as potential mediators of cerebral edema following TBI.
detailed neuropathological examination, demonstrated Inflammation, both classical and neurogenic, offers a num-
an elevation in substance P immunoreactivity in cortical ber of attractive targets, with the tachykinins (substance P)
microvasculature. Moreover, the localization to perivas- in particular seeming to play an active physiological role in
cular neurons suggested that injury to the neuron may modulating BBB permeability after trauma.
result in a localized perivascular release of neuropeptides,
with a resultant increase in BBB permeability and edema Acknowledgement
R.V. is supported by the Neurosurgical Research Foundation, Australia.
formation. Notably, substance P is stored and co-released J.J.D is supported by the Alzheimer’s Society of Canada Postdoctoral
with CGRP, a potent endogenous vasodilator that Fellowship. We thank Tavik Morgenstern for the medical illustration.
potentiates edema formation in the presence of mediators
of increased vascular permeability, such as substance P
[65]. Thus, their combined release during neurogenic References and recommended reading
Papers of particular interest, published within the annual period of review, have
inflammation would theoretically facilitate a profound been highlighted as:
edema response.  of special interest
 of outstanding interest
Additional references related to this topic can also be found in the Current
Given the increased perivascular substance P after acute World Literature section in this issue (pp. 337–338).
injury to the brain, Donkin et al. [61] subsequently 1 Marmarou A. Pathophysiology of traumatic brain edema: current concepts.
administered the substance P neurokinin-1 receptor Acta Neurochir Suppl 2003; 86:7–10.
antagonist N-acetyl-tryptophan after TBI and noted a 2 Feickert HJ, Drommer S, Heyer R. Severe head injury in children: impact of risk
factors. J Trauma 1999; 47:33–38.
marked attenuation of BBB permeability and subsequent
3 Klatzo I. Pathophysiological aspects of brain edema. Acta Neuropathol (Berl)
edema formation. Similar findings were noted in a reper- 1987; 72:236–239.
fusion model of transient ischemia [66], with highly 4 Simard JM, Kent TA, Chen M, et al. Brain oedema in focal ischaemia:
significant reductions in edema formation as measured molecular pathophysiology and theoretical implications. Lancet Neurol
2007; 6:258–268.
at 24 h after the induction of stroke. Although the results
5 Nag S, Manias JL, Stewart DJ. Pathology and new players in the pathogenesis
using neurokinin-1 antagonists were useful in establish- of brain edema. Acta Neuropathol 2009; 118:197–217.
ing a role for substance P in brain injury, an alternative 6 Marmarou A, Takagi H, Shulman K. Biomechanics of brain edema and effects
approach to establishing a more general role for neuro- on local cerebral blood flow. Adv Neurol 1980; 28:345–358.

genic inflammation in TBI is by inhibition of central 7 Reulen HJ, Graham R, Spatz M, Klatzo I. Role of pressure gradients and bulk
flow in dynamics of vasogenic brain edema. J Neurosurg 1977; 46:24–35.
neurogenic inflammation by neuropeptide depletion. 8 Barzo P, Marmarou A, Fatouros P, et al. Contribution of vasogenic and cellular
Neuropeptide depletion can be accomplished by chronic edema to traumatic brain swelling measured by diffusion-weighted imaging.
J Neurosurg 1997; 87:900–907.
preinjury administration of the vanilloid receptor agonist
9 Hanstock CC, Faden AI, Bendall MR, Vink R. Diffusion-weighted imaging
capsaicin, which stimulates the release of neuropeptides differentiates ischemic tissue from traumatized tissue. Stroke 1994; 25:843–
from the presynaptic sensory nerve terminals to the point 848.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
298 Inflammatory diseases and infection

10 Barzo P, Marmarou A, Fatouros P, et al. Magnetic resonance imaging 34 Vajtr D, Benada O, Kukacka J, et al. Correlation of ultrastructural changes of
monitored acute blood–brain barrier changes in experimental traumatic brain endothelial cells and astrocytes occurring during blood brain barrier damage
injury. J Neurosurg 1996; 85:1113–1121. after traumatic brain injury with biochemical markers of BBB leakage and
inflammatory response. Physiol Res 2009; 58:263–268.
11 O’Connor CA, Cernak I, Vink R. The temporal profile of edema formation
differs between male and female rats following diffuse traumatic brain injury. 35 Candelario-Jalil E, Yang Y, Rosenberg GA. Diverse roles of matrix metallo-
Acta Neurochir Suppl 2006; 96:121–124.  proteinases and tissue inhibitors of metalloproteinases in neuroinflammation
and cerebral ischemia. Neuroscience 2009; 158:983–994.
12 Habgood MD, Bye N, Dziegielewska KM, et al. Changes in blood–brain
Excellent review summarizing the role of MMPs.
barrier permeability to large and small molecules following traumatic brain
injury in mice. Eur J Neurosci 2007; 25:231–238. 36 Ding JY, Kreipke CW, Schafer P, et al. Synapse loss regulated by matrix
metalloproteinases in traumatic brain injury is associated with hypoxia in-
13 Luh C, Kuhlmann CR, Ackermann B, et al. Inhibition of myosin light chain
ducible factor-1alpha expression. Brain Res 2009; 1268:125–134.
 kinase reduces brain edema formation after traumatic brain injury. J Neuro-
chem 2010; 112:1015–1025. 37 Rosenberg GA. Matrix metalloproteinases and their multiple roles in neuro-
Demonstrates that cytoskeletal rearrangement within the endothelial barrier at 24 h degenerative diseases. Lancet Neurol 2009; 8:205–216.
after TBI leads to increased BBB permeability and facilitation of vasogenic edema
38 Rosenberg GA, Yang Y. Vasogenic edema due to tight junction disruption by
formation.
matrix metalloproteinases in cerebral ischemia. Neurosurg Focus 2007;
14 Baskaya MK, Rao AM, Dogan A, et al. The biphasic opening of the blood– 22:E4.
brain barrier in the cortex and hippocampus after traumatic brain injury in rats.
39 Falo MC, Fillmore HL, Reeves TM, Phillips LL. Matrix metalloproteinase-3
Neurosci Lett 1997; 226:33–36.
expression profile differentiates adaptive and maladaptive synaptic plasti-
15 Beaumont A, Marmarou A, Hayasaki K, et al. The permissive nature of blood city induced by traumatic brain injury. J Neurosci Res 2006; 84:768–
brain barrier (BBB) opening in edema formation following traumatic brain 781.
injury. Acta Neurochir Suppl 2000; 76:125–129.
40 Vilalta A, Sahuquillo J, Rosell A, et al. Moderate and severe traumatic brain
16 Manley GT, Fujimura M, Ma T, et al. Aquaporin-4 deletion in mice reduces injury induce early overexpression of systemic and brain gelatinases. Intensive
brain edema after acute water intoxication and ischemic stroke. Nat Med Care Med 2008; 34:1384–1392.
2000; 6:159–163.
41 Asahi M, Wang X, Mori T, et al. Effects of matrix metalloproteinase-9 gene
17 Papadopoulos MC, Verkman AS. Potential utility of aquaporin modulators for knock-out on the proteolysis of blood–brain barrier and white matter com-
therapy of brain disorders. Prog Brain Res 2008; 170:589–601. ponents after cerebral ischemia. J Neurosci 2001; 21:7724–7732.
18 Taya K, Gulsen S, Okuno K, et al. Modulation of AQP4 expression by the 42 Gasche Y, Fujimura M, Morita-Fujimura Y, et al. Early appearance of activated
selective V1a receptor antagonist, SR49059, decreases trauma-induced matrix metalloproteinase-9 after focal cerebral ischemia in mice: a possible
brain edema. Acta Neurochir Suppl 2008; 102:425–429. role in blood–brain barrier dysfunction. J Cereb Blood Flow Metab 1999;
19:1020–1028.
19 Pasantes-Morales H, Cruz-Rangel S. Brain volume regulation: osmolytes and
 aquaporin perspectives. Neuroscience 2009 [Epub ahead of print]. 43 Gidday JM, Gasche YG, Copin JC, et al. Leukocyte-derived matrix metallo-
A review critically assessing the role of osmolytes and AQPs in brain volume proteinase-9 mediates blood–brain barrier breakdown and is proinflammatory
regulation. after transient focal cerebral ischemia. Am J Physiol Heart Circ Physiol 2005;
289:H558–H568.
20 Badaut J, Lasbennes F, Magistretti PJ, Regli L. Aquaporins in brain: distribu-
tion, physiology, and pathophysiology. J Cereb Blood Flow Metab 2002; 44 Wang X, Jung J, Asahi M, et al. Effects of matrix metalloproteinase-9 gene
22:367–378. knock-out on morphological and motor outcomes after traumatic brain injury.
J Neurosci 2000; 20:7037–7042.
21 Aoki K, Uchihara T, Tsuchiya K, et al. Enhanced expression of aquaporin 4 in
human brain with infarction. Acta Neuropathol 2003; 106:121–124. 45 Fujimoto M, Takagi Y, Aoki T, et al. Tissue inhibitor of metalloproteinases
protect blood–brain barrier disruption in focal cerebral ischemia. J Cereb
22 Papadopoulos MC, Verkman AS. Aquaporin-4 and brain edema. Pediatr
Blood Flow Metab 2008; 28:1674–1685.
Nephrol 2007; 22:778–784.
46 Homsi S, Federico F, Croci N, et al. Minocycline effects on cerebral edema:
23 Sorani MD, Zador Z, Hurowitz E, et al. Novel variants in human aquaporin-4
relations with inflammatory and oxidative stress markers following traumatic
reduce cellular water permeability. Hum Mol Genet 2008; 17:2379–2389.
brain injury in mice. Brain Research 2009; 1291:122–132.
24 Ding JY, Kreipke CW, Speirs SL, et al. Hypoxia-inducible factor-1alpha
47 Tejima E, Guo S, Murata Y, et al. Neuroprotective effects of over-expressing
signaling in aquaporin upregulation after traumatic brain injury. Neurosci Lett
tissue inhibitor of metalloproteinase TIMP-1. J Neurotrauma 2009; 26:1935–
2009; 453:68–72.
1941.
25 Tran N, Kim S, Vincent H, et al. Aquaporin-1-mediated cerebral edema
48 Zhao B-Q, Wang S, Kim H-Y, et al. Role of matrix metalloproteinases in
following traumatic brain injury: effects of acidosis and corticosteroid admin-
delayed cortical responses after stroke. Nat Med 2006; 12:441–445.
istration. J Neurosurg 2009 [Epub ahead of print].
49 Morganti-Kossmann MC, Rancan M, Stahel PF, Kossmann T. Inflammatory
26 Ghabriel MN, Thomas A, Vink R. Magnesium restores altered aquaporin-4
response in acute traumatic brain injury: a double-edged sword. Curr Opin
immunoreactivity following traumatic brain injury to a preinjury state. Acta
Crit Care 2002; 8:101–105.
Neurochir Suppl 2006; 96:402–406.
50 Abbott NJ. Inflammatory mediators and modulation of blood–brain barrier
27 Papadopoulos MC, Manley GT, Krishna S, Verkman AS. Aquaporin-4 facil-
permeability. Cell Mol Neurobiol 2000; 20:131–147.
itates reabsorption of excess fluid in vasogenic brain edema. FASEB J 2004;
18:1291–1293. 51 Plesnila N, Schulz J, Stoffel M, et al. Role of bradykinin B2 receptors in the
formation of vasogenic brain edema in rats. J Neurotrauma 2001; 18:1049–
28 Sun M-C, Honey CR, Berk C, et al. Regulation of aquaporin-4 in a traumatic
1058.
brain injury model in rats. J Neurosurg 2003; 98:565–569.
52 Trabold R, Eros C, Zweckberger K, et al. The role of bradykinin B(1) and B(2)
29 Fazzina G, Amorini AM, Marmarou C, et al. The PKC activator phorbol myristate
 receptors for secondary brain damage after traumatic brain injury in mice.
acetate decreases brain edema by AQP4 down-regulation following middle
J Cereb Blood Flow Metab 2010; 30:130–139.
cerebral artery occlusion in the rat. J Neurotrauma 2009 [Epub ahead of print].
Receptor knockout mice were used to demonstrate that the bradykinin B2
30 Kikuchi K, Tancharoen S, Matsuda F, et al. Edaravone attenuates cerebral receptors were linked to development of BBB permeability and edema formation
ischemic injury by suppressing aquaporin-4. Biochem Biophys Res Commun after TBI.
2009; 390:1121–1125.
53 Zweckberger K, Plesnila N. Anatibant, a selective nonpeptide bradykinin B2
31 Taya K, Marmarou C, Okuno K, et al. Effect of secondary insults upon receptor antagonist, reduces intracranial hypertension and histopathological
 aquaporin-4 water channels following experimental cortical contusion in rats. damage after experimental traumatic brain injury. Neurosci Lett 2009;
J Neurotrauma 2009; 27:229–239. 454:115–117.
Demonstrates that under conditions of secondary hypoxia or hypotension following
54 Su J, Cui M, Tang Y, et al. Blockade of bradykinin B2 receptor more effectively
TBI, blunting of the AQP4 response is deleterious to outcome.
reduces postischemic blood–brain barrier disruption and cytokines release
32 Grossetete M, Phelps J, Arko L, et al. Elevation of matrix metalloproteinases 3 than B1 receptor inhibition. Biochem Biophys Res Commun 2009; 388:205–
and 9 in cerebrospinal fluid and blood in patients with severe traumatic brain 211.
injury. Neurosurg 2009; 65:702–708.
55 Zausinger S, Lumenta DB, Pruneau D, et al. Therapeutical efficacy of a novel
33 Hayashi T, Kaneko Y, Yu S, et al. Quantitative analyses of matrix metallopro- nonpeptide bradykinin B2 receptor antagonist on brain edema formation and
teinase activity after traumatic brain injury in adult rats. Brain Res 2009; ischemic tissue damage in focal cerebral ischemia. Acta Neurochir Suppl
1280:172–177. 2003; 86:205–207.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Mechanisms of cerebral edema in TBI Donkin and Vink 299

56 Ker K, Blackhall K. Beta-2 receptor antagonists for acute traumatic brain 62 Donkin JJ, Cernak I, Rodgers KM, Vink R. Mild concussive head injury results
injury. Cochrane Database Syst Rev 2008:CD006686. in increased brain substance P immunoreactivity. In: Proceedings of the 7th
International Neurotrauma Symposium. Medimond International Proceedings;
57 Shakur H, Andrews P, Asser T, et al. The BRAIN TRIAL: a randomised,
2004. pp. 75–78.
 placebo controlled trial of a bradykinin B2 receptor antagonist (Anatibant) in
patients with traumatic brain injury. Trials 2009; 10:109.
Clinical trial demonstrating a lack of benefit or harm of the B2 antagonists in TBI. 63 Turner RJ, Blumbergs PC, Sims NR, et al. Increased substance P immuno-
58 Geppetti P, Bertrand C, Ricciardolo FL, Nadel JA. New aspects on the role of reactivity and edema formation following reversible ischemic stroke. Acta
kinins in neurogenic inflammation. Can J Physiol Pharmacol 1995; 73:843–847. Neurochir Suppl 2006; 96:263–266.

59 Nimmo AJ, Cernak I, Heath DL, et al. Neurogenic inflammation is associated


with development of edema and functional deficits following traumatic brain 64 Zacest AC, Vink R, Manavis J, et al. Substance P immunoreactivity increases
injury in rats. Neuropeptides 2004; 38:40–47. following human traumatic brain injury. Acta Neurochir Suppl 2010;
106:211–216.
60 Vink R, Young A, Bennett CJ, et al. Neuropeptide release influences brain
edema formation after diffuse traumatic brain injury. Acta Neurochir Suppl
2003; 86:257–260. 65 Brain SD, Williams TJ. Inflammatory oedema induced by synergism between
calcitonin gene-related peptide (CGRP) and mediators of increased vascular
61 Donkin JJ, Nimmo AJ, Cernak I, et al. Substance P is associated with the permeability. Br J Pharmacol 1985; 86:855–860.
 development of brain edema and functional deficits after traumatic brain injury.
J Cereb Blood Flow Metab 2009; 29:1388–1398.
The first demonstration that substance P plays a critical role in TBI with its 66 Turner R, Vink R. Inhibition of neurogenic inflammation as a novel treat-
neurokinin-1 antagonist reducing BBB permeability and edema, as well as im- ment for ischemic stroke. Drug News Perspect 2007; 20:221–
proving functional outcome. 226.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Anda mungkin juga menyukai