You are on page 1of 15

Review

Cerebrovasc Dis 2016;42:155–169 Received: November 23, 2015


Accepted: March 1, 2016
DOI: 10.1159/000445170
Published online: April 26, 2016

Mechanism and Therapy of Brain Edema


after Intracerebral Hemorrhage
Haiping Zheng Chunli Chen Jie Zhang Zhiping Hu
Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China

Key Words Introduction


Brain edema · Hematoma · Intracerebral hemorrhage ·
Inflammation Intracerebral hemorrhage (ICH) accounts for about
10–15% of all strokes with high mortality [1], 1-month
mortality of ICH is approximate 40% and it increases
Abstract with age [2]. In the last 2 decades, the high mortality and
Background: Intracerebral hemorrhage (ICH) is a subtype of morbidity of ICH does not decrease over time. In the first
stroke with a severe high mortality and disability rate and few hours after ICH onset, primary brain injury by ICH
accounts for about 10–15% of all strokes. The oppression is mainly caused by the oppression and destruction to the
and destruction by hematoma to brain tissue cause the pri- near tissue by hematoma formation. And hematoma
mary brain injury. The inflammation and coagulation re- could also increase the intracranial pressure; it can even
sponse after ICH would accelerate the formation of brain induce brain herniation. The inflammation, thrombin ac-
edema around hematoma, resulting in a more severe and tivation, and erythrocyte lysis caused by primary injury
durable injury. Currently, treatments for ICH are focusing on could promote the formation of brain edema, which is
the primary injury including reducing intracranial hyperten- associated with poor outcome, and could cause more se-
sion, blood pressure control, and rehabilitation. There is a vere and durable injury [3]. Brain edema is a pathological
short-of-effective medical treatment for secondary inflam- phenomenon that water and brain tissue volume increase.
mation and reducing brain edema in ICH patients. So, it is Brain edema after ICH can be divided into perihemato-
very important to study on the relationship between brain mal edema (PHE) and intrahematomal edema. Most pa-
edema and ICH. Summary: Many molecular and cellular tients could survive the initial injury of smaller hemor-
mechanisms contribute to the formation and progress of rhage, but the secondary injury may result in severe neu-
brain edema after ICH; inhibition of brain edema provides rological deficits and even death [4]. The mechanism of
favorable outcome of ICH. Key Messages: This review main- brain edema is complicated; vasogenic factors, thrombin
ly discusses the pathology and mechanism of brain edema, formation, erythrocyte lysis and hemoglobin (Hb) toxic-
the effects of brain edema on ICH, and the methods of treat- ity [5] have been proved to be related to brain edema
ing brain edema after ICH. © 2016 S. Karger AG, Basel growth [6]. Currently, therapy for ICH focuses on the pri-
mary injury and preventing bleeding again, including de-
79.110.25.176 - 10/11/2016 11:15:41 PM

© 2016 S. Karger AG, Basel Dr. Zhiping Hu


1015–9770/16/0424–0155$39.50/0 Department of Neurology
The Second Xiangya Hospital of Central South University
Downloaded by:

E-Mail karger@karger.com
Renmin Road 139, Changsha, Hunan 410011 (China)
www.karger.com/ced
E-Mail huzhiping2014 @ 163.com
Color version available online
hydration and hemostasis. But, all these treatments fail to
provide a promising functional outcome or decrease ICH
mortality. In recent years, many studies focus on the
mechanism of secondary inflammation that can cause
brain edema and this may provide new therapy targets for
ICH [7]. Animal experiments demonstrated that fingoli- Plasma Clot
proteins retraction
mod could reduce edema, cell apoptosis and cerebra at- extravasation
rophy and show neuroprotective function in ICH rats [8].
A similar result was observed in a clinical study [9]. Like-
wise, dexamethasone can reduce cerebral cell apoptosis
and inhibit inflammation [10], and deferoxamine (DFX) Perihematoma Perihematoma
provides new therapy target [11]. But before these immu- osmotic pressure hydrostatic
increase pressure decrease
nomodulators or other anti-inflammatory agents are
used clinically, more experiments are needed.

Mechanism of Edema Vasogenic


edema

Brain edema increases in the first 24 h progressively


and increases rapidly 3 days after onset, reaches its initial
Fig. 1. Mechanism of vasogenic edema.
peak at the 4th or the 5th day and remains elevated slow-
ly until 9–14 days and then decreases [12]. PHE develops
in response to clot retraction and hydrostatic pressure
change [13], mass effect, thrombin formation, erythro- side the hematoma increases and the infiltration of peri-
cyte lysis, Hb toxicity, complement activation, plasma hematoma water and plasma increases the tension inside
proteins leakage and blood-brain barrier (BBB) disrup- the capsule progressively. Additionally, blood may leak
tion [5]. All of inflammation, thrombin activation and repeatedly from the abundant capillaries contained in the
red blood cell (RBC) lysis production contribute to BBB granulation tissue resulting in hematoma enlarging [15].
disruption resulting in edema formation, and it can be
diversified into 3 phases: (1)clot retraction could force the Vasogenic Factors
serum into the perihematomal space to form vasogenic Numerous studies show that during the early stage of
edema (1 h after ICH; fig. 1), (2) inflammation (fig. 2) and ICH, vasogenic factors can cause edema. Vasogenic fac-
thrombin activation (fig. 3)–related cytotoxic brain ede- tors mainly include clot formation and contraction [16],
ma through clotting cascade (peaking at 1–2 days), and decrease in hydrostatic pressure around hematoma space
(3) erythrocyte lysis and Hb toxicity-related injury (de- [17] and extravasation of plasma proteins [13]. MRI done
layed edema formation at about day 3; fig. 4) [14]. Both 2 h after onset of ICH shows edema in the area surround-
elevated oncotic pressure of perihematoma space due to ing the hematoma [18].
the infiltration of blood components from hematoma and Through MRI perfusion, weighted imaging and diffu-
BBB disruption caused by inflammation, thrombin cas- sion-weighted imaging, some experts found that water
cade and erythrocyte lysis products can aggravate vaso- diffusion in the perihematoma edema increases signifi-
genic edema. However, oxidative stress induced by vaso- cantly and is related to brain water volume independent-
genic edema, and release of cytotoxic substance could in- ly, and they think that most of the brain water comes from
duce cytotoxic edema. So, vasogenic edema and plasma [19]. Injecting autologous blood (control group)
cytotoxic edema interact with each other and lead to a or heparinized autologous blood (heparinized group)
vicious circle. Intrahematomal edema is mainly caused by into brain parenchyma of pigs showed that edema in-
tension hematoma. Tension hematoma is related to the creases significantly 1 h after ICH and lasts longer in the
formation of capsule-like granulation tissue during the control group, while ther heparinized group showed no
absorption of a hematoma. The capsule-like granulation obvious edema. Meanwhile, they also experimented on
can limit the absorption of liquified hematoma and cyto- rats by injecting thrombin or heparinized thrombin into
toxic substance. Subsequently, the oncotic pressure in- cerebral parenchyma; the results of these experiments
79.110.25.176 - 10/11/2016 11:15:41 PM

156 Cerebrovasc Dis 2016;42:155–169 Zheng/Chen/Zhang/Hu


DOI: 10.1159/000445170
Downloaded by:
Color version available online
ICH

Thrombin

Microglial RBCs
Inflammatory cells infiltration
activation
CD36

Astrocyte
activation TLR4 Heme

ROS TNF-į Other cytotoxins MMPs

NFNJB

Inflammatory
Inflammation gene expression ଭ

Cell swelling BBB disruption

Brain edema

Fig. 2. Mechanism of inflammation induces edema.

demonstrated that the heparinized group showed signifi- hydrostatic pressure. Plasma can also induce oxidative
cantly less edema. So, they concluded that the formation stress and release of inflammatory mediators, which will
and existence of clot was important for the formation and promote the secondary brain injury besides vasogenic
progress of brain edema [16]. Compared with spontane- edema [22].
ous ICHs (SICHs), thrombolysis-related ICHs have both
lower absolute and relative edema observed in clinical Inflammation
test, indicating that intrahematomal blood clotting is a To clarify the cause of edema, Gong et al. [23] con-
reasonable factor for the formation of hyperacute PHE ducted an immunocytochemistry experiment on rats. It
[20]. Animal experiment showed that plasma proteins demonstrated that inflammatory response took place in
can be detected in the tissue around hematoma after 1 h and around the hematoma after ICH with the infiltration
of ICH when BBB is not destructed, and so the leakage of of neutrophils and macrophages and activation of mi-
plasma proteins and related edema is not associated with croglia. Inflammation can cause cell swelling and BBB
BBB destruction in the hyperacute stage of ICH [13]. Clot disruption, and then causes brain edema. Activated mi-
contraction can decrease the perihematoma hydrostatic croglia and infiltrating leukocytes release cytotoxic me-
pressure; then it forces the plasma components into peri- diators contributing to secondary injury. Clinical studies
hematomal space, which leads to an increase in perihe- have proved that RBCs infiltrate into cerebral immedi-
matoma oncotic pressure close to the plasma level and ately, along with leukocyte from peripheral blood, mac-
water infiltration into the perihematomal space from rophages and plasma proteins [24], and animal experi-
blood followed [16, 21]. Furthermore, destruction by he- ments had shown that CD4+ T lymphocytes were the
matoma can induce a low metabolism of the perihema- main cause of brain leukocyte infiltration [25]. But with-
toma tissue, cell death and brain atrophy, which results in in 12 h after onset of ICH, inflammatory macrophages
the enlargement of perihematomal space and decrease in and dendritic cells comprise a majority of infiltrating leu-
79.110.25.176 - 10/11/2016 11:15:41 PM

Mechanism and Therapy of Brain Edema Cerebrovasc Dis 2016;42:155–169 157


after ICH DOI: 10.1159/000445170
Downloaded by:
Color version available online
ICH
Coagulation cascade
Thrombin
activation

Activate BBB recovery

BMVEC and peri-vascular


Complements Microglial activation Cytokines PARs astrocyte proliferation

2–6 days
after ICH

Inflammation MMPs Endothelial cell Intercellular tight Src kinase


contraction junction open phosphorylation

1–2 days
after ICH

Basal membrane BMVEC and peri-vascular


degradation astrocyte injury

Brain edema BBB disruption

Fig. 3. Mechanism of thrombin induces edema.

kocytes [26]. Neutrophils or polymorphonuclear leuko- can express and release diverse toxic factors, which con-
cytes (PMNs) are the first leukocytes to infiltrate the ner- tribute to secondary injury, such as cytokines (interleukin
vous system within 4–5 h after ICH, and reach a peak (IL)-1β and others), chemokines, proteases, ROS, prosta-
value at 3 days [27]. PMNs may cause direct neurotoxic- glandins, cyclooxygenase-II and heme oxygenase (HO)
ity to ICH brain by release matrix metalloproteinases [30, 33]. TLRs are molecules that play a critical role in the
(MMPs), reactive oxygen species (ROS), and tumor ne- induction of innate and adaptive immunity, and TLR4 is
crosis factor-alpha (TNF-alpha) or other cytokines. Ani- expressed in microglia and some neurons 6 h through
mal experiments demonstrated that PMNs can induce in- 7 days after ICH [34]. Activated TLR4 can mediate mi-
flammation, but resting neutrophils could reduce the per- croglial autophagy and activation [35], and induce the
meability of BBB and activated neutrophils showed a dissociation of kappa-B (κB) with nuclear factor-κB (NF-
neutral effect [28]. So, neutrophils-induced inflamma- κB); then NF-κB activates and regulates the transcription
tion depends on MMPs, ROS and cytokines instead of of genes related to inflammation after it translates into the
reducing permeability of BBB. Leukocytes exist only for nucleus. Animal experiments showed that neutrophil,
about 2 days after infiltrating into hemorrhagic brain, but monocyte, inflammatory monocyte and microglia in-
it can cause further damages by stimulating microglia and creased significantly in the perihematomal brain com-
macrophages [14]. Microglia activation can also be medi- pared to TLR4-deficient mice, and the leukocytes infil-
ated by CD36 in erythrocyte [29], thrombin [30], and by trated from the peripheral blood [36]. Microglia reaches
heme via toll-like receptor 4 (TLR4) signaling pathways a peak at 72 h, begin to decrease by a week and return to
[31]. Activated microglia/macrophages appear in and basal levels by 21 days after ICH. Astrocytes can also ex-
around the injury tissue as early as 1 h after ICH [32]. The press MMPs together with activated microglia, and con-
primary role for resident microglia cell activation is to trolling microglia–astrocyte interactions may be a poten-
clear the hematoma, but excessive microglial activation tial way to minimize ICH-induced injury [7]. So, the ear-
79.110.25.176 - 10/11/2016 11:15:41 PM

158 Cerebrovasc Dis 2016;42:155–169 Zheng/Chen/Zhang/Hu


DOI: 10.1159/000445170
Downloaded by:
Color version available online
ICH

RBC lysis

CD36

Hb Heme Microglial activation

HO

Iron deposition TLR4

MyD88&TRIF
Regulate
Inflammatory gene
BBB disruption expression increases
AQP4 NF-NJB Cytokines

TLR2/TLR4
Brain edema

Inflammation

Fig. 4. Mechanism of RBC lysis production induces edema.

ly stage of the inflammatory response and brain edema degrade substrates including collagens IV, V, VII, and X,
after ICH is caused by microglia activation, infiltration of gelatin, laminin, and fibronectin, most of which are com-
inflammatory cells including leukocytes and macro- ponents of capillary basal membranes. So MMP-2 activa-
phages, and accumulation of proinflammatory mediators tion leads to basal membrane degradation that is a vital
released from these cells. part of BBB. And thrombin can trigger MMP-2 expres-
sion and membrane degradation by activating PARs [38].
Thrombin Thrombin also activates the phosphorylation of Src ki-
The coagulation cascade is activated as soon as blood nase (a proto-oncogene protein family with tyrosine pro-
flows into the brain tissue, and thrombin is an essential tein kinase activity), which leads to the injury of brain
component of the coagulation cascade [4, 5]. Thrombin microvascular endothelial cell (BMVEC) and peri-vascu-
induces disruption of BBB function and parenchymal cell lar astrocyte, resulting in the disruption of BBB and for-
death, and both of BBB disruption and cell toxicity by mation of edema through its PARs [40, 41]. However, Src
thrombin can trigger the formation of brain edema after kinase proto-oncogene members can stimulate the pro-
ICH [37]. Thrombin induces endothelial cell contraction liferation of newborn BMVECs and peri-vascular astro-
and the opening of intercellular tight junctions by activat- cytes after 2–6 days; it then promotes the resolution of
ing protease-activated receptors (PARs), which are edema and the recovery of BBB permeability. Thrombin
thrombin receptors [38]. MMPs protein is a zinc-con- also releases nitric oxide, TNF-α, IL-12, and IL-6, and
taining extracellular-matrix degrading protease and they thrombin-induced brain injury is partly mediated by
can degrade the entire extracellular matrix after being ac- complement [42]. Activation of microglia and mitogen-
tivated. MMP-9 can be an important biomarker of com- activated protein (MAP) kinases also play an important
plications and for selecting patients for trials investigat- role in causing thrombin-induced neurological injury af-
ing hemicraniectomy in ICH [39]. MMP-2 protein can ter ICH [43].
79.110.25.176 - 10/11/2016 11:15:41 PM

Mechanism and Therapy of Brain Edema Cerebrovasc Dis 2016;42:155–169 159


after ICH DOI: 10.1159/000445170
Downloaded by:
RBC Lysis Production formation of edema after ICH through reduced BBB per-
RBCs lysis occurs within 24 h of ICH and induces the meability or AQP4 levels, but the details of the mecha-
release of Hb and heme, which are taken up by microglia nism are still unknown [52]. An in vivo microdialysis
and neurons [14]; heme can be degraded by HO into bil- study shows that glutamate, taurine and asparagines ac-
iverdin, carbon monoxide and iron. All of Hb, heme and cumulate transiently in extracellular fluids in the perihe-
iron causes significant brain edema after the first 24 h, matoma region during the early period of ICH. These
which is related to a threefold increase of BBB permeabil- amino acids may play an important role in the pathology
ity [44]. Animal experiments showed that intracerebral of ICH, but the intricacies of the mechanism are not very
injection of packed RBCs does not produce significant clear [53]. A recent study shows that blood glutamate
edema on the first day but does at 3 days; so RBCs can grabbing cannot reduce the hematoma nor improve the
cause delayed brain edema, which results from cellular neurologic deficit in ICH but is a safe excitotoxic treat-
lysis and BBB disruption by RBCs [44, 45]. Brain iron is ment modality [54]. Animal experiments demonstrated
an erythrocyte degradation product; iron deposition is that ET-1 expression increased significantly at 6, 12, 18,
found in the perihematomal brain tissue during the 1st and 24 h after ICH, and they observed a positive correla-
day after ICH, peaking after 7 days and remaining at a tion between the number of ET-1-positive endothelial
high level for at least 2 weeks, and perihematoma water cells and BBB permeability [55]. New findings show that
content increases progressively over course of time [46]. ET-1 can lead to an increase in gene expression, including
Aquaporin-4 (AQP4) is involved in brain volume ho- genes associated with the inflammatory response, oxida-
meostasis and water balance; AQP4 is localized in perihe- tive stress, heme metabolism and iron homeostasis [56].
matomal region at 6 h post-ICH and AQP4 mRNA and So, ET-1 may be related with BBB disruption and it plays
AQP4 protein are increased post-ICH [47]. So, edema an important role in causing the secondary injury after
post-ICH is closely connected with AQP4. Upregulation ICH.
of AQP4 around hematoma brain is observed and reached
a peak at 3–7 days, and animal experiments showed that
the changes of brain water content are accompanied by Effects of Edema
an alteration of AQP4, and this expression is affected by
iron concentration [46]. Clinical study demonstrates that Studies have shown that brain edema is significantly
iron overload is associated with the progress of PHE in- associated with hematoma enlargement and increased
dicating a poor outcome and high in-hospital mortality midline shift, which lead to poorer functional outcome
[6]. And heme can promote microglial activation through of ICH [4]. Mass effect can contribute to the formation
TLR4; this in turn will induce NF-κB activation through of PHE [5]; it can also be a result of PHE [57]. Clinical
the myeloid differentiation primary response gene 88 observation shows that mass effect progresses within
(MyD88) or toll/IR-1 domain-containing adaptor pro- 2  days after ICH due to hematoma enlargement, and
tein-inducing interferon-beta (TRIF) signaling pathway, progresses again during the second and third weeks be-
and finally increase cytokine expression and inflamma- cause of increased brain edema [57]. So, we can see that
tory injury in ICH [31]. Hb can trigger inflammation after the interaction between mass effect and edema may lead
ICH through the assembly of TLR2/TLR4 heterodimers, to an unfavorable outcome. PHE may be a risk factor of
and MyD88 is required for ICH-induced TLR2/TLR4 acute progressing hemorrhagic stroke, and delayed PHE
heterodimerization [48]. may contribute to subacute progression of hemorrhagic
In addition to all the discussion mentioned earlier, ar- stroke, both these 2 types of stroke have worse clinical
ginine vasopressin (AVP) receptors, glutamate and endo- outcome than the nonprogressing types of hemorrhagic
thelin-1 (ET-1) may also be involved in the formation of stroke [15]. Multivariable logistic regression analysis
brain edema. Experiments showed that AVP can induce shows that absolute edema volume is not related to func-
the formation of brain edema by effects on astrocytes af- tion outcome nor mortality, but relative edema volume
ter ischemic stroke and the induction can be decreased by (absolute edema volume/hematoma volume) during the
AVP receptors antagonist [49, 50]. And, animal tests have first 24 h after ICH is an independent predictor of favor-
found that AVP receptors antagonist SR49059 signifi- able 12-week functional outcome in hyperacute SICH
cantly decreases cerebral edema at 24 and 72 h post-ICH without intraventricular extension [58]. Similarly, in a
injury and improved neurobehavioral deficits at 72 h prospective cohort study, the relative edema is proved to
[51]. So, AVP receptors may play an important role in the be associated with an improved outcome at 12 weeks but
79.110.25.176 - 10/11/2016 11:15:41 PM

160 Cerebrovasc Dis 2016;42:155–169 Zheng/Chen/Zhang/Hu


DOI: 10.1159/000445170
Downloaded by:
Color version available online
ICH

Surgery Tension hematoma

Early clot removal Hematoma Capsule-like granulation

Clot retraction
Thrombin activation Inflammation RBC lysis and Hb toxicity
Hydrostatic pressure ଯ
Plasma proteins
Anti-PMNs
Microgilia inhibition
Immunomodulator

Osmotherapy HO-2
Src inhibitor Iron
TLR4 inhibitor
Completment inhibitor
PPARDž agonist

Iron chelator
Vasogenic edema (DFX)

Other medicines Reduce Cytotoxic edema Reduce Osmotherapy

Fig. 5. Treatments of brain edema after ICH.

is not related to mortality in supratentorial SICH with- decrease mortality of ICH. Recently, some experiments
out intraventricular extension [59]. And results demon- reviewed secondary inflammation, which contributes to
strate that hematoma volume is the strongest indepen- brain edema and the results of these experiments may
dent predictor of a poorer prognosis of supratentorial provide new ways of treatment for ICH [7]. Medical treat-
SICH. It seems that absolute edema volume is not re- ments of treatment on brain edema post-ICH include in-
lated to the outcome significantly, but it can contribute hibition of inflammation, thrombin activation and Hb
to hematoma expansion, which in turn independently degradation products–mediated toxicity [60], traditional
predicts mortality. They excluded infratentorial SICH dehydration and so on (fig. 5). A clinical study shows that
and those with intraventricular extension, as these 2 fac- blood pressure reduction (systolic blood reduction goals
tors have been found to be associated with poor progno- are 170–199, 140–169 and 110–139 mm Hg, respectively)
sis [58, 59]. can reduce hematoma expansion and brain edema ratio
after ICH with no significance between every group [61].
However, some large clinical studies show that intensive
Treatments of Brain Edema after ICH and continuous blood pressure reduction in the acute
phase of ICH can improve functional outcomes by de-
Currently, treatments for ICH mainly focus on the pri- creasing the hematoma expansion rate, and blood pres-
mary injury and on preventing bleeding again; focus is sure should be controlled to be under 140/90 mm Hg in
also directed toward primary supportive treatment, in- the first hour after onset or within 24 h [62–64]. And
cluding dehydration, hemostasis, blood pressure and in- these new therapeutic strategies focusing on multiple in-
tracranial pressure monitoring, and so on. All these ther- flammatory pathways seem to be more effective than
apies fail to provide a promising favorable outcome or those focusing on single pathways [65]. Besides these new
79.110.25.176 - 10/11/2016 11:15:41 PM

Mechanism and Therapy of Brain Edema Cerebrovasc Dis 2016;42:155–169 161


after ICH DOI: 10.1159/000445170
Downloaded by:
therapies, another effective way is surgical treatment to ized controlled study on 128 supratentorial ICH patients
remove hematoma, which will reduce mass effect and he- shows that mannitol (20%, 100 ml every 4 h for 5 days,
matoma-induced injury [60]. tapered in the next 2 days) failed to improve the outcome
at the end of 1 month and decrease mortality significant-
Traditional Dehydration Therapy ly [70]. Systematic reviews show similar results that there
Dehydration is often used to decrease intracranial is not enough evidence to support the routine use of man-
pressure during the acute phase of ICH, and hypertonic nitol in acute ICH patients without increased intracra-
dehydration of somotherapy is the most common treat- nial pressure, but intracranial pressure should be moni-
ment. Usual osmotherapy medicines are mannitol, glyc- tored closely [71]. When there exist clinical features or
erin fructose and albumin, other dehydrants include fu- image examination changes of intracranial hypertension,
rosemide, glucocorticoid, acetazolamide and so on, and especially brain herniation, mannitol should be used im-
mannitol is the most widely used dehydrant. We should mediately to decrease intracranial pressure. Hypertonic
choose and use dehydrants on the basis of the brain ede- saline (3 or 23.4%) can also be used as a dehydrant, which
ma type and evaluation of BBB integrity, as brain edema has a more lasting effect than mannitol without BBB dis-
mechanism differs from each other. ruption [72]. But hypertonic saline use has limitations, as
Animal experiments found that after an injection of it can cause disorders in water and salt metabolism. Ani-
mannitol through the internal carotid artery, BBB disrup- mal experiments have demonstrated that mannitol pro-
tion was observed and BBB permeability increased by 4–5 duces a dose-dependent increase in plasma osmolality
times at 5 min and then reversed within minutes, the and reduction of brain water content, and furosemide
mechanism may be that mannitol can induce the shrink- alone shows no effect on plasma osmotic pressure nor
age of cerebrovascular endothelial cells and vasodilata- brain water content [73]. Combination of mannitol and
tion [66, 67]. Hence, the use of mannitol may lead to the furosemide has a greater effect on plasma osmolality and
opening of BBB and aggravate brain edema before BBB is reduction of water content than mannitol alone in the rat
disrupted (within 6 h after ICH generally). Pharmacoki- ICH model [73]. Combination of furosemide and hyper-
netics study on mannitol for treatment of vasogenic ede- tonic saline has a better effect on reducing brain water
ma shows that about 84% of the infused mannitol is ex- content compared with hypertonic saline alone, and the
creted through urine. But mannitol accumulation is de- combination causes no more increase of osmolality and
tected in the cerebral tissue after multiple injections, even sodium concentration than that induced by hypertonic
exceeding the mannitol concentration in the plasma; this saline alone [74]. For subacute progressing, ICH exclud-
experiment observed a reversal of the osmotic concentra- ing rebleeding after CT, dehydrant and intracranial pres-
tion gradient between plasma and edematous brain, sure monitoring should be applied [75].
which can aggravate vasogenic brain edema. This study In conclusion, dehydration is not recommended for
demonstrates that a single dose of mannitol is unable to ICH patients without intracranial hypertension. When
reduce brain water content in edematous brain tissue or BBB is not disrupted (within 6 h after ICH generally) or
edema progression after 4 h of injection, but multiple plasma osmotic pressure is >320 mOsm, mannitol espe-
doses can increase brain water content in edematous tis- cially frequent mannitol using is not suitable, hypertonic
sue [68]. Mannitol accumulates in the injured brain tis- saline alone or with furosemide can be alternative op-
sue, which would increase osmotic pressure; then water tions. However, if there is intracranial hypertension espe-
in perihematoma flows into the hematoma; this results in cially brain herniation or progressing ICH excluding re-
hematoma expansion even midline shift. Mannitol should bleeding, dehydration should be used. In case the antici-
be used while being closely monitored for plasma osmot- pated dehydration target after multiple use of mannitol is
ic pressure; mannitol is not suitable to use when plasma not achieved, combination with furosemide or hyperton-
osmotic pressure is >320 mOsm because high plasma os- ic saline can be a substitute for adding mannitol dose, as
motic pressure leads to dehydration, but mannitol can multiple doses of mannitol can increase brain water con-
increase osmotic pressure of hematoma and reverse the tent in edematous tissue.
osmotic pressure gradient [69]. Meantime, the early use
of mannitol can shrink the normal brain and increase the Inhibition of Inflammation
pressure gradient between hematoma and normal brain. Inflammation plays an important role in brain edema
So, for those ICH patients who are still bleeding, early use after ICH and it can induce cytotoxic edema through
of mannitol may lead to ICH progress [69]. A random- multiple ways. So, inhibition of inflammation may be
79.110.25.176 - 10/11/2016 11:15:41 PM

162 Cerebrovasc Dis 2016;42:155–169 Zheng/Chen/Zhang/Hu


DOI: 10.1159/000445170
Downloaded by:
beneficial to ICH. In the early stages of ICH, inflamma- golimod can also reduce brain edema, apoptosis and
tion is mainly caused by PMNs infiltration and microglia brain atrophy [8]. Further study shows that fingolimod
activation. decreases not only the number of perihematomal T-
lymphocytes in brain but also the expressions of cyto-
Inhibition of PMNs Infiltration and Microglia kines (interferon-γ, and IL-17) [83]. And in this animal
Activation experiment, fingolimod had functions of reducing brain
Neutrophil depletion by using anti-PMN can reduce edema, improving short-term sensorimotor function
BBB permeation, MMP-9 expression, perihematomal ax- and long-term motor and coordination and cognitive
onal injury and the astrocytic and microglia/macrophage function in rats after experimental ICH [83]. Fu et al. [9]
responses and provide a better outcome of ICH [76, 77]. conducted a 2-arm proof-of-concept study including 23
Similarly, MMP inhibitor (GM6001), ROS scavengers or patients with primary supratentorial ICH with hemato-
TNF-alphaR neutralizing antibody can decrease the neu- mal volume of 5–30 ml. These 23 patients received stan-
rotoxicity caused by PMNs, and then protect neurons dard treatment alone (control group) or combined with
from injury [27]. fingolimod (0.5 mg, orally for 3 consecutive days). Re-
Activation of microglia can lead to the release of di- sults revealed that the fingolimod group has a signifi-
verse toxic factors (such as cytokines, proteases, ROS and cantly better outcome compared with that of the control
so on), which contribute to secondary injury after ICH. group. Patients treated with fingolimod have a higher
So, inhibition of microglia activation may be beneficial to Glasgow Coma Scale score, lower National Institutes of
ICH. It has been proved that Minocycline, an inhibitor of Health Stroke Scale score, improved neurologic func-
microglia activation, can reduce PHE, ICH-induced brain tion and fewer ICH-related lung infections without dif-
tissue loss and improve functional outcome in ICH rats ferences in the occurrence of adverse events when com-
[78]. And minocycline can also ameliorate the damage pared with the control group. Dexamethasone can pro-
after ICH by protecting BBB, decreasing TNF-alpha and mote the recovery of ICH injury by inhibiting the
MMPs expression, and reducing microvessel loss and ex- inflammatory response and reduce brain edema because
travasation of plasma proteins and the numbers of TNF- of its capacity to decrease apoptotic cell death and inhib-
alpha-positive cells and neutrophils [79]. In another rat iting the intercellular adhesion molecule-1 and MMP-9
experiment, it was found that coinjection of iron and mi- expression [10, 84].
nocycline significantly reduced brain edema, BBB leakage
and brain cell death caused by iron, compared with an Inhibition of Thrombin
injection of iron alone [80]. Tuftsin, another inhibitor of Thrombin inhibitor, Hirudin, can prevent the BBB
microglia activation, can reduce brain edema after ICH as disruption caused by thrombin, but it can also affect the
well [81]. Tuftsin fragment 1–3, a macrophage/microg- clotting hemostatic function; it may not be an assured
lial inhibitory factor, is a neuroprotective agent for ICH form of treatment [40]. Acute administration of the Src
treatment, as it can reduce stroke volume, edema, degen- inhibitor PP2 blocks the thrombin pathway, decreases
erating neurons, and neurobehavioral deficits [81]. TLR4 glucose hypermetabolism and cell death around ICH, and
can mediate the autophagy of microglia; treatment of au- reduces brain edema that occurs after ICH without affect-
tophagy inhibitor (3-methyladenine) decreases microg- ing coagulation [40, 41]. But delayed and chronic admin-
lial activation and inflammatory injury [35]. istration of PP2 prevents edema resolution and BBB re-
pair because chronic Src kinase activation can promote
Immunomodulator the BBB repair after ICH. N-acetylheparin is an inhibitor
Edema is significantly associated with hematoma en- of complement activation; it can attenuate the brain in-
largement and increased midline shift, resulting in jury induced by thrombin, and it may be a neuroprotec-
poorer functional outcome of ICH. Agents that can re- tive agent for ICH [85]. Both argatroban or cyclohexi-
duce PHE process provide protective effects for ICH. mide can prevent thrombin cytotoxicity and exhibit an
Fingolimod (FTY720), a sphingosine 1-phosphate re- obviously neuroprotective function against ICH-induced
ceptors analog, is an immune-modulating drug that can injury; MAP kinase inhibitor (PD98059) and a c-Jun N-
prevent the migration of lymphocytes from primary and terminal kinase inhibitor (SP600125) can also prevent
secondary lymphoid organs and may ameliorate cere- ICH-induced neuron loss [43]. However, these drugs
bral inflammation, it can improve neurobehavior and have no effect on hematoma volume or brain edema after
cognitive outcomes in experiment ICH mice [82]. Fin- ICH.
79.110.25.176 - 10/11/2016 11:15:41 PM

Mechanism and Therapy of Brain Edema Cerebrovasc Dis 2016;42:155–169 163


after ICH DOI: 10.1159/000445170
Downloaded by:
Inhibition of RBCs Lysis and Hb Toxicity nescent cells, including RBCs. Rosiglitazone is a peroxi-
RBC lysis releases Hb and heme; brain iron is a heme some proliferator-activated receptor γ (PPARγ) agonist.
degradation product; excess iron released is found to in- It can increase PPARγ-regulated gene (catalase and CD36)
duct secondary brain injury [46]. DFX, an iron chelator, expression, but reduce proinflammatory gene (TNF-α, IL-
has been proved to be neuroprotective in nature in many 1β, MMP-9, and so on) expression and neuronal damage
ICH animal experiments. By injection of autologous [29]. So, PPARγ agonist can promote the resolution of
blood into the pigs cerebral, Gu et al. [86] found that DFX hematoma and is a potential treatment of ICH.
can reduce perihematoma volume, through chelating
iron directly and may be also by inhibiting oxidative Other Medicines
stress-induced cell death. In another experiment, results In addition to all that has been discussed earlier, there
showed that DFX can reduce white matter edema after exist many other potential therapy targets for ICH. The
ICH in piglets, decrease levels of TNF-alpha and recep- 3-hydroxy-3-methylglutaryl-coenzyme A reductase in-
tor-interacting protein kinase 1 [87]. A rats experiment hibitors or statins have pleiotropic effects, and continued
showed that DFX can penetrate BBB rapidly and reduce statins use after ICH is associated to lower mortality with-
ICH-induced ventricle enlargement, brain edema and in 6 months and early neurological improvement because
brain atrophy without severe side effects [88]. DFX can of its anti-inflammation and neuroprotective function
reduce free iron in cerebrospinal fluid as well as ICH-in- [93]. Depletion of GR-1-positive (GR-1(+)) cells by the
duced neurological deficits and acute neuronal death in administration of anti-GR-1 can reduce circulating GR-
rat experiment. It can also inhibit the endogenous re- 1(+) cells and astrocyte immunoreactivity and decrease
sponse to ICH and the upregulation of ferritin and AQP4, brain neutrophils after ICH; it may be a neuroprotective
which are related to a poor outcome of ICH [46, 89, 90]. agent [94]. Nuclear factor erythroid 2-related factor 2
Pro-oxidant heme, which is released from Hb catabolism (Nrf2) is a key transcriptional factor for antioxidant re-
of heme, plays an important role in the resolution of he- sponse element-regulated genes and it can induce and
matoma. HO controls the degradation of pro-oxidant upregulate cytoprotective and antioxidant genes that at-
heme and HO-2 may provide protection for ICH-in- tenuate tissue injury. Nrf2 is neuroprotective by reducing
duced brain injury [91]. Heme activates microglial via leukocyte infiltration, excessive free radical oxidative in-
TRL4 signaling pathway, and TLR2 and TLR4 play an im- jury, DNA damage, and cytochrome c release during the
portant role in the secondary injury [31, 48]. So, inhibi- early phase of ICH [95]. Pyrroloquinoline quinone (PQQ)
tion of TLR2 and TLR4 may be beneficial for ICH and has roles of antioxidant properties, cofactor of dehydro-
some experiments have proved this hypothesis. An ICH genase, and amine oxidase is a neuroprotective agent in
mouse model demonstrates that TAK-242 (ethyl (6R)- experimental stroke and spinal cord injury models. Ani-
6-(N-(2-chloro-4-fluorophenyl)sulfamoyl)cyclohex- mal test demonstrates that PQQ can improve the locomo-
1-ene-1-carboxylate, Takeda), which is a TLR4 antago- tor functions, reduce the hematoma volumes and allevi-
nist, can reduce brain water content, neurological deficit ate the expansion of brain edema after ICH. Also, pre-
scores, and levels of inflammatory factors. TAK-242 also treated rats with PQQ significantly reduce the production
decreases the levels of deoxyribonucleic acid (DNA) of ROS after ICH, probably due to its antioxidant effects
damage, neuronal degeneration and the expression of [96]. SR49059, an AVP V(1a) receptor competitive an-
TLR4 downstream signaling molecules [92]. Specific tagonist is proved to have a function of reducing brain
TLR4 antibody (Mts510) and TLR4-knockout have a edema and neurobehavioral deficits after ICH by decreas-
similar function of reducing brain edema and neurologi- ing the BBB disruption and AQP4 levels [51, 52]. Animal
cal deficits [31]. experiments demonstrated that granulocyte-colony stim-
An increasing number of proinflammatory mediators ulating factor can protect ICH by reducing BBB perme-
and cytotoxins are released by blood components after ation, cell apoptosis and brain edema [97]. Erythropoie-
ICH. Lysis of extravasated erythrocytes in the hematoma tin can protect BBB and decrease BBB disruption and
releases cytotoxic Hb, heme and iron, resulting in second- brain edema post-ICH [98].
ary injury. Phagocytic cells, brain’s microglia and hema-
togenous macrophages can phagocytose and process ex- Surgical Treatment
travasated erythrocytes and subsequent toxicity occurs Due to lack of optimal treatment of ICH, clinicians
before lysis after ICH. CD36 and catalase are proteins that have devoted to the hematoma clearance especially for
can mediate phagocytosis of damaged, apoptotic, or se- patients with hematoma volume that is >30 ml or more,
79.110.25.176 - 10/11/2016 11:15:41 PM

164 Cerebrovasc Dis 2016;42:155–169 Zheng/Chen/Zhang/Hu


DOI: 10.1159/000445170
Downloaded by:
by surgical intervention or thrombolytic evacuation. The or brainstem compression resulting in aggravating con-
operation forms include craniotomy, decompressive cra- sciousness disturbance (Glasgow Coma Scale score ≤13),
niectomy, stereotactic or transcatheter aspiration, laparo- clot clearance should be applied before the condition of
scopic aspiration and so on. the patient deteriorates to relieve oppression and save
life. If cerebellar hematoma exceeds 3 cm in diameter but
Early Clot Removal the 4th ventricle is not totally obliterated, surgical clot
A clinical study demonstrated that hematoma remov- removal may not be required and conservative treatment
al by surgery can significantly reduce the volume of the or ventricular drainage can be a feasible treatment [106–
mass including brain edema and hematoma in putam- 108].
inal hemorrhage patients as compared to the conserva- In recent years, some trials have shown that MIS or
tive group [99]. Another set of clinical data showed that minimal-invasive hematoma evacuation in combination
surgical treatment of hypertensive putaminal hematoma with thrombolytics have encouraging outcome [109,
with 30 ml or more decreases mortality and provides bet- 110]. Animal experiments found that MIS can reduce the
ter outcome compared with the non-operation group glutamate content [111], neurological deficit scores, peri-
[100]. Burr hole craniectomy of hypertensive basal gan- hematomal ET-1 levels, BBB permeability and brain wa-
glia hemorrhage can decrease brain edema grades and ter content compared to the model control group, espe-
reduce secondary injury by coagulation end products– cially performing MIS at 6–12 h after ICH [112]. In a co-
activated inflammatory cascade [101]. And the experi- hort study, MIS combined with recombinant tissue-type
ment showed that gross-total removal of hematoma plasminogen activator significantly reduced the 30-day
group has a better outcome than the sub-total removal of mortality and without effects on early or delayed PHE
the hematoma group; this indicates that residue hema- formation [113]. MIS with local fibrinolysis decreased le-
toma can still promote secondary injury and may result thality from 35 to 21% among patients with parenchymal
in a comparatively worse result [101]. However, a paral- ICH and from 98 to 48% among patients with ventricular
lel-group trial about surgical trial in ICH (STICH) found hemotamponade compared to treatment without use of
that early surgery (within 24 h after onset) has no overall fibrinolytics, and the local use of fibrinolytics had no sys-
benefit compared with initial conservative treatment for tematic effect on the blood coagulation system [114]. Me-
spontaneous supratentorial ICH patients [102]. Meta- ta-analysis demonstrates that neuroendoscopic approach
analysis showed that early surgery before the patients de- with external ventricular drainage may be a better man-
teriorate would improve ICH outcome for spontaneous agement for intraventricular hemorrhage secondary to
supratentorial ICH patients without intraventricular spontaneous supratentorial hemorrhage than neuroen-
hemorrhage meeting the conditions: undertaken within doscopic with intraventricular fibrinolysis [115].
8 h of ictus, the volume of the hematoma was 20–50 ml, Early clot removal seems to have great benefit for ICH
the Glasgow Coma Score was between 9 and 12, or the patients, but the effect of surgery is still controversial and
patient was aged between 50 and 69 [103]. And large clin- depends on many factors, such as body condition, hema-
ical randomised trial (STICH II) also showed that early toma location, volume and so on. Clinicians should assess
surgery (within 12 h after onset) for spontaneous super- patients’ condition comprehensively before surgical
ficial ICH patients without intraventricular hemorrhage treatments.
would improve clinical functional outcome, and no in-
creasing death or disability rate was observed at 6 months Surgical Treatment for Intrahematomal Edema
[104]. Another meta-analysis about minimal invasive Intrahematoma edema is mainly caused by tension he-
surgery (MIS) demonstrated that MIS can reduce pri- matoma. Tension hematoma is related to the formation
mary and secondary brain injury for supratentorial ICH of capsule-like granulation, and conservative treatment
patients meeting the following conditions: age of 30–80 has no favorable effect. So, surgical treatment should be
with superficial hematoma, Glasgow Coma Scale score of applied if tension hematoma is diagnosed after imaging
≥9, hematoma volume between 25 and 40 ml, and within examination. A clinical test from China found that for
72 h after onset of symptoms [105]. For cerebellar hem- patients with tension hematoma, dehydration therapy
orrhage, urgent clot removal should be undertaken be- fails to ameliorate symptoms, and surgery can improve
fore deterioration if there exists totally the 4th ventricu- clinical conditions and decrease intracranial pressure.
lar obliterated [106]. When cerebellar hemorrhage oc- And intracranial hematoma puncture drainage may be
curs with blood collections exceeding 3 cm in diameter superior to craniotomy [116].
79.110.25.176 - 10/11/2016 11:15:41 PM

Mechanism and Therapy of Brain Edema Cerebrovasc Dis 2016;42:155–169 165


after ICH DOI: 10.1159/000445170
Downloaded by:
Conclusions Immunomodulator such as fingolimod or dexamethasone
are neuroprotective agents. However, to apply new treat-
ICH is a subtype of stroke with severe outcome and is ments of ICH in clinic needs much more randomized con-
short of efficient treatment. The secondary injury induced trolled trials, animal and clinical experiments.
by ICH is involved with numerous cellular and molecular
mechanisms. Edema after ICH plays an important role in
ICH-induced injury and is associated with poor outcome Disclosure Statement
of ICH. Vasogenic factors, inflammation, thrombin acti-
vation, RBC lysis and Hb toxicity contribute to the forma- Thanks to the financial funding of famous professor progress
tion of brain edema after ICH. Dehydration therapy and of Xiangya, the key built disciplines of the ministry, and the key
research and development program of Hunan Science and Tech-
new targets that block the inflammation cascade inhibit nology Agency (soft science research): analysis of clinical factors
the thrombin activation, prevent or alleviate the lysis of and gene polymorphism in progressing hemorrhagic stroke. We
RBC, and surgical treatment may be effective treatments. have no conflict of interest to claim.

References
1 Flower O, Smith M: The acute management 11 Selim M: Deferoxamine mesylate: a new hope cioli A, Duldner JE, Topol EJ, Broderick JP:
of intracerebral hemorrhage. Curr Opin Crit for intracerebral hemorrhage: from bench to Decreased perihematomal edema in throm-
Care 2011;17:106–114. clinical trials. Stroke 2009; 40(3 suppl):S90– bolysis-related intracerebral hemorrhage
2 van Asch CJ, Luitse MJ, Rinkel GJ, van der S91. compared with spontaneous intracerebral
Tweel I, Algra A, Klijn CJ: Incidence, case fa- 12 Venkatasubramanian C, Mlynash M, Finley- hemorrhage. Stroke 2000;31:596–600.
tality, and functional outcome of intracere- Caulfield A, Eyngorn I, Kalimuthu R, Snider 21 Kuroiwa T, Cahn R, Juhler M, Goping G,
bral haemorrhage over time, according to age, RW, Wijman CA: Natural history of perihe- Campbell G, Klatzo I: Role of extracellular
sex, and ethnic origin: a systematic review and matomal edema after intracerebral hemor- proteins in the dynamics of vasogenic brain
meta-analysis. Lancet Neurol 2010; 9: 167– rhage measured by serial magnetic resonance edema. Acta Neuropathol 1985;66:3–11.
176. imaging. Stroke 2011;42:73–80. 22 Wagner KR, Dean C, Beiler S, Bryan DW,
3 Keep RF, Hua Y, Xi G: Intracerebral haemor- 13 Wagner KR, Xi G, Hua Y, Kleinholz M, de Packard BA, Smulian AG, Linke MJ, de Cour-
rhage: mechanisms of injury and therapeutic Courten-Myers GM, Myers RE, Broderick JP, ten-Myers GM: Plasma infusions into porcine
targets. Lancet Neurol 2012;11:720–731. Brott TG: Lobar intracerebral hemorrhage cerebral white matter induce early edema, ox-
4 Xi G, Keep RF, Hoff JT: Mechanisms of brain model in pigs: rapid edema development in idative stress, pro-inflammatory cytokine
injury after intracerebral haemorrhage. Lan- perihematomal white matter. Stroke 1996;27: gene expression and DNA fragmentation: im-
cet Neurol 2006;5:53–63. 490–497. plications for white matter injury with in-
5 Hoff JT, Xi G: Brain edema from intracerebral 14 Ziai WC: Hematology and inflammatory sig- creased blood-brain-barrier permeability.
hemorrhage. Acta Neurochir Suppl 2003; 86: naling of intracerebral hemorrhage. Stroke Curr Neurovasc Res 2005;2:149–155.
11–15. 2013;44(6 suppl 1):S74–S78. 23 Gong C, Hoff JT, Keep RF: Acute inflamma-
6 Bakhshayesh B, Hosseininezhad M, Saadat 15 Chen S, Zeng L, Hu Z: Progressing haemor- tory reaction following experimental intrace-
SN, Ansar MM, Ramezani H, Saadat SM: Iron rhagic stroke: categories, causes, mechanisms rebral hemorrhage in rat. Brain Res 2000;871:
overload is associated with perihematoma and managements. J Neurol 2014; 261: 2061– 57–65.
edema growth following intracerebral hem- 2078. 24 Wang J: Preclinical and clinical research on
orrhage that may contribute to in-hospital 16 Xi G, Wagner KR, Keep RF, Hua Y, de Cour- inflammation after intracerebral hemor-
mortality and long-term functional outcome. ten-Myers GM, Broderick JP, Brott TG, Hoff rhage. Prog Neurobiol 2010;92:463–477.
Curr Neurovasc Res 2014;11:248–253. JT: Role of blood clot formation on early ede- 25 Mracsko E, Javidi E, Na SY, Kahn A, Liesz A,
7 Wang J, Doré S: Inflammation after intracere- ma development after experimental intrace- Veltkamp R: Leukocyte invasion of the brain
bral hemorrhage. J Cereb Blood Flow Metab rebral hemorrhage. Stroke 1998; 29: 2580– after experimental intracerebral hemorrhage
2007;27:894–908. 2586. in mice. Stroke 2014;45:2107–2114.
8 Lu L, Barfejani AH, Qin T, Dong Q, Ayata C, 17 Kuroiwa T, Shibutani M, Tajima T, Hirasawa 26 Hammond MD, Ai Y, Sansing LH: Gr1+ mac-
Waeber C: Fingolimod exerts neuroprotec- H, Okeda R: Hydrostatic pressure versus os- rophages and dendritic cells dominate the in-
tive effects in a mouse model of intracerebral motic pressure in the development of vaso- flammatory infiltrate 12 hours after experi-
hemorrhage. Brain Res 2014;1555:89–96. genic brain edema induced by cold injury. mental intracerebral hemorrhage. Transl
9 Fu Y, Hao J, Zhang N, Ren L, Sun N, Li YJ, Adv Neurol 1990;52:11–19. Stroke Res 2012;3:s125–s131.
Yan Y, Huang D, Yu C, Shi FD: Fingolimod 18 Linfante I, Llinas RH, Caplan LR, Warach S: 27 Nguyen HX, O’Barr TJ, Anderson AJ: Poly-
for the treatment of intracerebral hemor- MRI features of intracerebral hemorrhage morphonuclear leukocytes promote neuro-
rhage: a 2-arm proof-of-concept study. JAMA within 2 hours from symptom onset. Stroke toxicity through release of matrix metallopro-
Neurol 2014;71:1092–1101. 1999;30:2263–2267. teinases, reactive oxygen species, and TNF-
10 Lee IN, Cheng WC, Chung CY, Lee MH, Lin 19 Butcher KS, Baird T, MacGregor L, Desmond alpha. J Neurochem 2007;102:900–912.
MH, Kuo CH, Weng HH, Yang JT: Dexa- P, Tress B, Davis S: Perihematomal edema in 28 Joice SL, Mydeen F, Couraud PO, Weksler
methasone reduces brain cell apoptosis and primary intracerebral hemorrhage is plasma BB, Romero IA, Fraser PA, Easton AS: Modu-
inhibits inflammatory response in rats with derived. Stroke 2004;35:1879–1885. lation of blood-brain barrier permeability by
intracerebral hemorrhage. J Neurosci Res 20 Gebel JM, Brott TG, Sila CA, Tomsick TA, neutrophils: in vitro and in vivo studies. Brain
2015;93:178–188. Jauch E, Salisbury S, Khoury J, Miller R, Pan- Res 2009;1298:13–23.
79.110.25.176 - 10/11/2016 11:15:41 PM

166 Cerebrovasc Dis 2016;42:155–169 Zheng/Chen/Zhang/Hu


DOI: 10.1159/000445170
Downloaded by:
29 Zhao X, Sun G, Zhang J, Strong R, Song W, 43 Ohnishi M, Katsuki H, Fujimoto S, Takagi M, totoxic treatment modality. J Cereb Blood
Gonzales N, Grotta JC, Aronowski J: Hema- Kume T, Akaike A: Involvement of thrombin Flow Metab 2015;35:1206–1212.
toma resolution as a target for intracerebral and mitogen-activated protein kinase path- 55 Wang LK, Hong Z, Wu GF, Li C: Perihema-
hemorrhage treatment: role for peroxisome ways in hemorrhagic brain injury. Exp Neurol tomal endothelin-1 level is associated with an
proliferator-activated receptor gamma in mi- 2007;206:43–52. increase in blood-brain barrier permeability
croglia/macrophages. Ann Neurol 2007; 61: 44 Bhasin RR, Xi G, Hua Y, Keep RF, Hoff JT: in a rabbit model of intracerebral hematoma.
352–362. Experimental intracerebral hemorrhage: ef- Chin Med J (Engl) 2013;126:3433–3438.
30 van Rossum D, Hanisch UK: Microglia. fect of lysed erythrocytes on brain edema and 56 Bickford JS, Ali NF, Nick JA, Al-Yahia M,
Metab Brain Dis 2004;19:393–411. blood-brain barrier permeability. Acta Neu- Beachy DE, Doré S, Nick HS, Waters MF: En-
31 Lin S, Yin Q, Zhong Q, Lv FL, Zhou Y, Li JQ, rochir Suppl 2002;81:249–251. dothelin-1-mediated vasoconstriction alters
Wang JZ, Su BY, Yang QW: Heme activates 45 Shi BZ, Meng XF, Yang JX, Hao XW, Cui T, cerebral gene expression in iron homeostasis
TLR4-mediated inflammatory injury via Lu ZF, Zhang HR, Zhou JC, Liu JF: Effect of and eicosanoid metabolism. Brain Res 2014;
MyD88/TRIF signaling pathway in intracere- erythrocytes on brain water content and 1588:25–36.
bral hemorrhage. J Neuroinflammation 2012; haem oxygenase-1 expression in rats with 57 Zazulia AR, Diringer MN, Derdeyn CP, Pow-
9:46. traumatic intracerebral haemorrhage. Acta ers WJ: Progression of mass effect after intra-
32 Wang J, Doré S: Heme oxygenase-1 exacer- Neurochir (Wien) 2012; 154: 1081–1086; dis- cerebral hemorrhage. Stroke 1999; 30: 1167–
bates early brain injury after intracerebral cussion 1086. 1173.
haemorrhage. Brain 2007; 130(pt 6):1643– 46 Qing WG, Dong YQ, Ping TQ, Lai LG, Fang 58 Gebel JM Jr, Jauch EC, Brott TG, Khoury J,
1652. LD, Min HW, Xia L, Heng PY: Brain edema Sauerbeck L, Salisbury S, Spilker J, Tomsick
33 Taylor RA, Sansing LH: Microglial responses after intracerebral hemorrhage in rats: the TA, Duldner J, Broderick JP: Relative edema
after ischemic stroke and intracerebral hem- role of iron overload and aquaporin 4. J Neu- volume is a predictor of outcome in patients
orrhage. Clin Dev Immunol 2013; 2013: rosurg 2009;110:462–468. with hyperacute spontaneous intracerebral
746068. 47 Xu J, Qiu GP, Huang J, Zhang B, Sun SQ, Gan hemorrhage. Stroke 2002;33:2636–2641.
34 Teng W, Wang L, Xue W, Guan C: Activation SW, Lu WT, Wang KJ, Huang SQ, Zhu SJ: In- 59 Gupta M, Verma R, Parihar A, Garg RK,
of TLR4-mediated NFkappaB signaling in ternalization of aquaporin-4 after collage- Singh MK, Malhotra HS: Perihematomal ede-
hemorrhagic brain in rats. Mediators In- nase-induced intracerebral hemorrhage. ma as predictor of outcome in spontaneous
flamm 2009;2009:473276. Anat Rec (Hoboken) 2015;298:554–561. intracerebral hemorrhage. J Neurosci Rural
35 Yang Z, Liu B, Zhong L, Shen H, Lin C, Lin L, 48 Wang YC, Zhou Y, Fang H, Lin S, Wang PF, Pract 2014;5:48–54.
Zhang N, Yuan B: Toll-like receptor-4-medi- Xiong RP, Chen J, Xiong XY, Lv FL, Liang QL, 60 Sonni S, Lioutas VA, Selim MH: New avenues
ated autophagy contributes to microglial acti- Yang QW: Toll-like receptor 2/4 heterodimer for treatment of intracranial hemorrhage.
vation and inflammatory injury in mouse mediates inflammatory injury in intracerebral Curr Treat Options Cardiovasc Med 2014;16:
models of intracerebral haemorrhage. Neuro- hemorrhage. Ann Neurol 2014;75:876–889. 277.
pathol Appl Neurobiol 2015;41:e95–e106. 49 Hertz L, Xu J, Chen Y, Gibbs ME, Du T, et al: 61 Qureshi AI, Palesch YY, Martin R, Novitzke
36 Sansing LH, Harris TH, Welsh FA, Kasner SE, Antagonists of the vasopressin V1 receptor J, Cruz-Flores S, Ehtisham A, Ezzeddine MA,
Hunter CA, Kariko K: Toll-like receptor 4 and of the β(1)-adrenoceptor inhibit cytotox- Goldstein JN, Hussein HM, Suri MF, Tariq N:
contributes to poor outcome after intracere- ic brain edema in stroke by effects on astro- Effect of systolic blood pressure reduction on
bral hemorrhage. Ann Neurol 2011; 70: 646– cytes – but the mechanisms differ. Curr Neu- hematoma expansion, perihematomal ede-
656. ropharmacol 2014;12:308–323. ma, and 3-month outcome among patients
37 Lee KR, Kawai N, Kim S, Sagher O, Hoff JT: 50 Zhao XY, Wu CF, Yang J, Gao Y, Sun FJ, with intracerebral hemorrhage: results from
Mechanisms of edema formation after intra- Wang DX, Wang CH, Lin BC: Effect of argi- the antihypertensive treatment of acute cere-
cerebral hemorrhage: effects of thrombin on nine vasopressin on the cortex edema in the bral hemorrhage study. Arch Neurol 2010;67:
cerebral blood flow, blood-brain barrier per- ischemic stroke of Mongolian gerbils. Neuro- 570–576.
meability, and cell survival in a rat model. J peptides 2015;51:55–62. 62 Qureshi AI, Palesch YY, Martin R, Toyoda K,
Neurosurg 1997;86:272–278. 51 Manaenko A, Fathali N, Khatibi NH, Lekic T, Yamamoto H, Wang Y, Wang Y, Hsu CY,
38 Guan JX, Sun SG, Cao XB, Chen ZB, Tong ET: Shum KJ, Martin R, Zhang JH, Tang J: Post- Yoon BW, Steiner T, Butcher K, Hanley DF,
Effect of thrombin on blood brain barrier per- treatment with SR49059 improves outcomes Suarez JI: Interpretation and implementation
meability and its mechanism. Chin Med J following an intracerebral hemorrhagic of intensive blood pressure reduction in acute
(Engl) 2004;117:1677–1681. stroke in mice. Acta Neurochir Suppl 2011; cerebral hemorrhage trial (INTERACT II). J
39 Senn R, Elkind MS, Montaner J, Christ-Crain 111:191–196. Vasc Interv Neurol 2014;7:34–40.
M, Katan M: Potential role of blood biomark- 52 Manaenko A, Fathali N, Khatibi NH, Lekic T, 63 Qureshi AI, Palesch YY: Antihypertensive
ers in the management of nontraumatic intra- Hasegawa Y, Martin R, Tang J, Zhang JH: Ar- treatment of acute cerebral hemorrhage
cerebral hemorrhage. Cerebrovasc Dis 2014; ginine-vasopressin V1a receptor inhibition (ATACH) II: design, methods, and rationale.
38:395–409. improves neurologic outcomes following an Neurocrit Care 2011;15:559–576.
40 Liu DZ, Ander BP, Xu H, Shen Y, Kaur P, intracerebral hemorrhagic brain injury. Neu- 64 Kobayashi J, Koga M, Tanaka E, Okada Y,
Deng W, Sharp FR: Blood-brain barrier rochem Int 2011;58:542–548. Kimura K, Yamagami H, Okuda S, Hasegawa
breakdown and repair by Src after thrombin- 53 Qureshi AI, Ali Z, Suri MF, Shuaib A, Baker Y, Shiokawa Y, Furui E, Nakagawara J, Kario
induced injury. Ann Neurol 2010; 67: 526– G, Todd K, Guterman LR, Hopkins LN: Ex- K, Okata T, Arihiro S, Sato S, Nagatsuka K,
533. tracellular glutamate and other amino acids in Minematsu K, Toyoda K: Continuous antihy-
41 Liu DZ, Sharp FR: The dual role of SRC ki- experimental intracerebral hemorrhage: an in pertensive therapy throughout the initial 24
nases in intracerebral hemorrhage. Acta Neu- vivo microdialysis study. Crit Care Med 2003; hours of intracerebral hemorrhage: the stroke
rochir Suppl 2011;111:77–81. 31:1482–1489. acute management with urgent risk-factor as-
42 Babu R, Bagley JH, Di C, Friedman AH, Ad- 54 da Silva-Candal A, Vieites-Prado A, Gutiér- sessment and improvement-intracerebral
amson C: Thrombin and hemin as central fac- rez-Fernández M, Rey RI, Argibay B, Mirel- hemorrhage study. Stroke 2014;45:868–870.
tors in the mechanisms of intracerebral hem- man D, Sobrino T, Rodríguez-Frutos B, Cas- 65 Zhou Y, Wang Y, Wang J, Anne Stetler R,
orrhage-induced secondary brain injury and tillo J, Campos F: Blood glutamate grabbing Yang QW: Inflammation in intracerebral
as potential targets for intervention. Neuro- does not reduce the hematoma in an intrace- hemorrhage: from mechanisms to clinical
surg Focus 2012;32:E8. rebral hemorrhage model but it is a safe exci- translation. Prog Neurobiol 2014;115:25–44.
79.110.25.176 - 10/11/2016 11:15:41 PM

Mechanism and Therapy of Brain Edema Cerebrovasc Dis 2016;42:155–169 167


after ICH DOI: 10.1159/000445170
Downloaded by:
66 Joshi S, Singh-Moon R, Wang M, Bruce JN, edema following intracerebral hemorrhage in 94 Loftspring MC, Johnson HL, Johnson AJ,
Bigio IJ, Mayevsky A: Real-time hemodynam- the rat. Exp Neurol 2007;207:227–237. Clark JF: Depletion of GR-1-positive cells is
ic response and mitochondrial function 80 Zhao F, Hua Y, He Y, Keep RF, Xi G: Mino- associated with reduced neutrophil inflam-
changes with intracarotid mannitol injection. cycline-induced attenuation of iron overload mation and astrocyte reactivity after experi-
Brain Res 2014;1549:42–51. and brain injury after experimental intracere- mental intracerebral hemorrhage. Transl
67 Rapoport SI: Osmotic opening of the blood- bral hemorrhage. Stroke 2011;42:3587–3593. Stroke Res 2012;3(suppl 1):147–154.
brain barrier: principles, mechanism, and 81 Wang J, Tsirka SE: Tuftsin fragment 1–3 is 95 Wang J, Fields J, Zhao C, Langer J, Thimmu-
therapeutic applications. Cell Mol Neurobiol beneficial when delivered after the induction lappa RK, Kensler TW, Yamamoto M, Bisw-
2000;20:217–230. of intracerebral hemorrhage. Stroke 2005; 36: al S, Doré S: Role of Nrf2 in protection
68 Kaufmann AM, Cardoso ER: Aggravation of 613–618. against intracerebral hemorrhage injury in
vasogenic cerebral edema by multiple-dose 82 Rolland WB 2nd, Manaenko A, Lekic T, mice. Free Radic Biol Med 2007;43:408–414.
mannitol. J Neurosurg 1992;77:584–589. Hasegawa Y, Ostrowski R, Tang J, Zhang JH: 96 Lu H, Shen J, Song X, Ge J, Cai R, Dai A, Ji-
69 Diringer MN, Zazulia AR: Osmotic therapy: FTY720 is neuroprotective and improves ang Z: Protective effect of pyrroloquinoline
fact and fiction. Neurocrit Care 2004; 1: 219– functional outcomes after intracerebral hem- quinone (PQQ) in rat model of intracerebral
233. orrhage in mice. Acta Neurochir Suppl 2011; hemorrhage. Cell Mol Neurobiol 2015; 35:
70 Misra UK, Kalita J, Ranjan P, Mandal SK: 111:213–217. 921–930.
Mannitol in intracerebral hemorrhage: a ran- 83 Rolland WB, Lekic T, Krafft PR, Hasegawa Y, 97 Chu H, Tang Y, Dong Q: Protection of gran-
domized controlled study. J Neurol Sci 2005; Altay O, Hartman R, Ostrowski R, Manaenko ulocyte-colony stimulating factor to hemor-
234:41–45. A, Tang J, Zhang JH: Fingolimod reduces ce- rhagic brain injuries and its involved mecha-
71 Qureshi AI, Mendelow AD, Hanley DF: In- rebral lymphocyte infiltration in experimen- nisms: effects of vascular endothelial growth
tracerebral haemorrhage. Lancet 2009; 373: tal models of rodent intracerebral hemor- factor and aquaporin-4. Neuroscience 2014;
1632–1644. rhage. Exp Neurol 2013;241:45–55. 260:59–72.
72 Qureshi AI, Wilson DA, Traystman RJ: 84 Yang JT, Lee TH, Lee IN, Chung CY, Kuo CH, 98 Chu H, Ding H, Tang Y, Dong Q: Erythro-
Treatment of elevated intracranial pressure Weng HH: Dexamethasone inhibits ICAM-1 poietin protects against hemorrhagic blood-
in experimental intracerebral hemorrhage: and MMP-9 expression and reduces brain brain barrier disruption through the effects
comparison between mannitol and hyper- edema in intracerebral hemorrhagic rats. of aquaporin-4. Lab Invest 2014; 94: 1042–
tonic saline. Neurosurgery 1999; 44: 1055– Acta Neurochir (Wien) 2011;153:2197–2203. 1053.
1063; discussion 1063–1064. 85 Gong Y, Xi GH, Keep RF, Hoff JT, Hua Y: 99 Okuda M, Suzuki R, Moriya M, Fujimoto
73 Thenuwara K, Todd MM, Brian JE Jr: Effect Complement inhibition attenuates brain ede- M, Chang CW, Fujimoto T: The effect of he-
of mannitol and furosemide on plasma osmo- ma and neurological deficits induced by matoma removal for reducing the develop-
lality and brain water. Anesthesiology 2002; thrombin. Acta Neurochir Suppl 2005; 95: ment of brain edema in cases of putaminal
96:416–421. 389–392. hemorrhage. Acta Neurochir Suppl 2006;
74 Mayzler O, Leon A, Eilig I, Fuxman Y, Beni- 86 Gu Y, Hua Y, Keep RF, Morgenstern LB, Xi G: 96:74–77.
fla M, Freixo PC, Gurevich B, Agassi R, Artru Deferoxamine reduces intracerebral hemato- 100 Kaya RA, Türkmenoğlu O, Ziyal IM, Dalkiliç
AA, Shapria Y: The effect of hypertonic (3%) ma-induced iron accumulation and neuronal T, Sahin Y, Aydin Y: The effects on progno-
saline with and without furosemide on plas- death in piglets. Stroke 2009;40:2241–2243. sis of surgical treatment of hypertensive pu-
ma osmolality, sodium concentration, and 87 Xie Q, Gu Y, Hua Y, Liu W, Keep RF, Xi G: taminal hematomas through transsylvian
brain water content after closed head trauma Deferoxamine attenuates white matter injury transinsular approach. Surg Neurol 2003;59:
in rats. J Neurosurg Anesthesiol 2006; 18: 24– in a piglet intracerebral hemorrhage model. 176–183; discussion 183.
31. Stroke 2014;45:290–292. 101 Zuo Y, Cheng G, Gao DK, Zhang X, Zhen
75 Morgenstern LB, Hemphill JC 3rd, Anderson 88 Okauchi M, Hua Y, Keep RF, Morgenstern HN, Zhang W, Xiao SC: Gross-total hema-
C, Becker K, Broderick JP, Connolly ES Jr, LB, Xi G: Effects of deferoxamine on intrace- toma removal of hypertensive basal ganglia
Greenberg SM, Huang JN, MacDonald RL, rebral hemorrhage-induced brain injury in hemorrhages: a long-term follow-up. J Neu-
Messé SR, Mitchell PH, Selim M, Tamargo RJ: aged rats. Stroke 2009;40:1858–1863. rol Sci 2009;287:100–104.
Guidelines for the management of spontane- 89 Hatakeyama T, Okauchi M, Hua Y, Keep RF, 102 Mendelow AD, Gregson BA, Fernandes
ous intracerebral hemorrhage: a guideline for Xi G: Deferoxamine reduces neuronal death HM, Murray GD, Teasdale GM, Hope DT,
healthcare professionals from the American and hematoma lysis after intracerebral hem- Karimi A, Shaw MD, Barer DH: Early sur-
Heart Association/American Stroke Associa- orrhage in aged rats. Transl Stroke Res 2013; gery versus initial conservative treatment in
tion. Stroke 2010;41:2108–2129. 4:546–553. patients with spontaneous supratentorial in-
76 Sansing LH, Harris TH, Kasner SE, Hunter 90 Wan S, Hua Y, Keep RF, Hoff JT, Xi G: Def- tracerebral haematomas in the international
CA, Kariko K: Neutrophil depletion dimin- eroxamine reduces CSF free iron levels fol- surgical trial in intracerebral haemorrhage
ishes monocyte infiltration and improves lowing intracerebral hemorrhage. Acta Neu- (STICH): a randomised trial. Lancet 2005;
functional outcome after experimental intra- rochir Suppl 2006;96:199–202. 365:387–397.
cerebral hemorrhage. Acta Neurochir Suppl 91 Wang J, Zhuang H, Doré S: Heme oxygenase 103 Gregson BA, Broderick JP, Auer LM, Batjer
2011;111:173–178. 2 is neuroprotective against intracerebral H, Chen XC, Juvela S, Morgenstern LB, Pan-
77 Moxon-Emre I, Schlichter LC: Neutrophil de- hemorrhage. Neurobiol Dis 2006;22:473–476. tazis GC, Teernstra OP, Wang WZ, Zucca-
pletion reduces blood-brain barrier break- 92 Wang YC, Wang PF, Fang H, Chen J, Xiong rello M, Mendelow AD: Individual patient
down, axon injury, and inflammation after XY, Yang QW: Toll-like receptor 4 antago- data subgroup meta-analysis of surgery for
intracerebral hemorrhage. J Neuropathol Exp nist attenuates intracerebral hemorrhage-in- spontaneous supratentorial intracerebral
Neurol 2011;70:218–235. duced brain injury. Stroke 2013; 44: 2545– hemorrhage. Stroke 2012;43:1496–1504.
78 Wu J, Yang S, Hua Y, Liu W, Keep RF, Xi G: 2552. 104 Mendelow AD, Gregson BA, Rowan EN,
Minocycline attenuates brain edema, brain 93 Tapia-Pérez JH, Rupa R, Zilke R, Gehring S, Murray GD, Gholkar A, Mitchell PM: Early
atrophy and neurological deficits after intra- Voellger B, Schneider T: Continued statin surgery versus initial conservative treatment
cerebral hemorrhage. Acta Neurochir Suppl therapy could improve the outcome after in patients with spontaneous supratentorial
2010;106:147–150. spontaneous intracerebral hemorrhage. lobar intracerebral haematomas (STICH II):
79 Wasserman JK, Schlichter LC: Minocycline Neurosurg Rev 2013; 36: 279–287; discussion a randomised trial. Lancet 2013; 382: 397–
protects the blood-brain barrier and reduces 287. 408.
79.110.25.176 - 10/11/2016 11:15:41 PM

168 Cerebrovasc Dis 2016;42:155–169 Zheng/Chen/Zhang/Hu


DOI: 10.1159/000445170
Downloaded by:
105 Zhou X, Chen J, Li Q, Ren G, Yao G, Liu M, orrhage. Curr Opin Crit Care 2014; 20: 148– type plasminogen activator reduces perihe-
Dong Q, Guo J, Li L, Guo J, Xie P: Minimal- 152. matomal edema and mortality in patients
ly invasive surgery for spontaneous supra- 110 Dey M, Stadnik A, Awad IA: Thrombolytic with spontaneous intracerebral hemor-
tentorial intracerebral hemorrhage: a meta- evacuation of intracerebral and intraventric- rhage. J Huazhong Univ Sci Technolog Med
analysis of randomized controlled trials. ular hemorrhage. Curr Cardiol Rep 2012;14: Sci 2014;34:165–171.
Stroke 2012;43:2923–2930. 754–760. 114 Krylov VV, Burov SA, Dash’ian VG,
106 Kirollos RW, Tyagi AK, Ross SA, van Hille 111 Wu G, Li C, Wang L, Mao Y, Hong Z: Min- Galankina IE: [Local fibrinolysis in surgical
PT, Marks PV: Management of spontaneous imally invasive procedures for evacua- treatment of non-traumatic intracranial
cerebellar hematomas: a prospective treat- tion  of intracerebral hemorrhage reduces hemorrhages]. Vestn Ross Akad Med Nauk
ment protocol. Neurosurgery 2001; 49: perihematomal glutamate content, blood- 2013;7:24–31.
1378–1386; discussion 1386–1387. brain barrier permeability and brain ede- 115 Li Y, Zhang H, Wang X, She L, Yan Z, Zhang
107 Witsch J, Neugebauer H, Zweckberger K, ma in rabbits. Neurocrit Care 2011; 14: N, Du R, Yan K, Xu E, Pang L: Neuroendo-
Jüttler E: Primary cerebellar haemorrhage: 118–126. scopic surgery versus external ventricular
complications, treatment and outcome. Clin 112 Wang L, Wang F, Wu G, Shi J: Early-stage drainage alone or with intraventricular fibri-
Neurol Neurosurg 2013;115:863–869. minimally invasive procedures decrease nolysis for intraventricular hemorrhage sec-
108 Papacocea A, Papacocea T, Dănăilă L, Ion D, perihematomal endothelin-1 levels and im- ondary to spontaneous supratentorial hem-
Bădărău A, Papacocea R: [Primary intracer- prove neurological functioning in a rabbit orrhage: a systematic review and meta-anal-
ebellar hematomas: surgical indications, model of intracerebral hemorrhage. Neurol ysis. PLoS One 2013;8:e80599.
prognosis]. Chirurgia (Bucur) 2010; 105: Res 2015;37:320–327. 116 Liu J, Huang Y, Wang A: Diagnosis and
805–807. 113 Lian LF, Xu F, Tang ZP, Xue Z, Liang QM, treatment of intracerebral tension hemato-
109 Barnes B, Hanley DF, Carhuapoma JR: Mini- Hu Q, Zhu WH, Kang HC, Liu XY, Wang ma following cerebral hemorrhage. J Int
mally invasive surgery for intracerebral haem- FR, Zhu SQ: Intraclot recombinant tissue- Neurol Neurosurg 2009;5:400–402.

79.110.25.176 - 10/11/2016 11:15:41 PM

Mechanism and Therapy of Brain Edema Cerebrovasc Dis 2016;42:155–169 169


after ICH DOI: 10.1159/000445170
Downloaded by:

You might also like