ArticlePDF Available

Standardized induction of subarachnoid hemorrhage in mice by intracranial pressure monitoring

Authors:

Abstract

Subarachnoid hemorrhage (SAH) is the subtype of stroke with the most unfavorable outcome but the least well investigated molecular pathophysiology. Among others, not sufficiently well standardized in vivo models suitable for the use with transgenic animals may be responsible for this situation. Therefore the aim of the current study was to detect suitable intra-operative parameters for the controlled and standardized induction of SAH in mice and to characterize the long-term functional and histopathological outcome of mice subjected to this procedure. Experimental study in mice using the intraluminal Circle of Willis perforation (CWp) model of SAH. SAH induced a sharp increase of intracranial pressure (ICP) from 5.1+/-1.2 to 78.5+/-9.3 mm Hg (mean+/-SD; p<0.05), a concomitant drop of cerebral blood flow (rCBF) by 81+/-4% (p<0.05), and a significant Cushing reflex response (p<0.05). rCBF measurements alone could not reliably detect SAH. SAH resulted in significant brain edema formation (brain water content increase at 72 h: 2.9+/-0.9%; p<0.05), loss of hippocampal neurons (CA1: -56%, CA2: -55%; CA3: -72%; 7 days; p<0.05), severe neurological dysfunction over 7 days, and a mortality of 30%. Our results indicate that CWp in mice can be standardized by intra-operative ICP monitoring. CWp leads to prolonged intracranial hypertension, selective neuronal cell death in the hippocampus, and severe neurological dysfunction. CWp in mice with ICP monitoring may therefore become a valuable tool for future investigations of the molecular pathophysiology of SAH.
Standardized induction of subarachnoid hemorrhage in mice by
intracranial pressure monitoring – Characterization of the model
Sergej Feiler*, Benjamin Friedrich*, Karsten Schöller, Serge C. Thal, and Nikolaus Plesnila
Institute for Surgical Research and Department of Neurosurgery, University of Munich
Medical Center - Grosshadern, Marchioninistr. 15, 81377 Munich, Germany
*both authors contributed equally to this work
Address of correspondence:
Prof. Nikolaus Plesnila
Royal College of Surgeons in Ireland (RCSI)
123 St. Stephen’s Green,
Dublin 2, Ireland,
Phone: +353 1 402 2794,
E-mail: nikolausplesnila@rcsi.ie
Feiler et al., Brain damage and ICP after SAH in mice
ABSTRACT
Background and Purpose: Subarachnoid hemorrhage (SAH) is the subtype of stroke with
the most unfavorable outcome but the least well investigated molecular pathophysiology.
Among others, not sufficiently well standardized in vivo models suitable for the use with
transgenic animals may be responsible for this situation. Therefore the aim of the current
study was to detect suitable intra-operative parameters for the controlled and standardized
induction of SAH in mice and to characterize the long-term functional and histopathological
outcome of mice subjected to this procedure.
Methods: Experimental study in mice using the intraluminal Circle of Willis perforation (CWp)
model of SAH.
Results: SAH induced a sharp increase of intracranial pressure (ICP) from 5.1 ± 1.2 to 78.5
± 9.3 mmHg (mean +/- SD; p<0.05), a concomitant drop of cerebral blood flow (rCBF) by 81
± 4% (p<0.05), and a significant Cushing reflex response (p<0.05). rCBF measurements
alone could not reliably detect SAH. SAH resulted in significant brain edema formation (brain
water content increase at 72 h: 2.9 ± 0.9%; p<0.05), loss of hippocampal neurons (CA1: -
56%, CA2: -55%; CA3:-72%; 7 days; p<0.05), severe neurological dysfunction over 7 days,
and a mortality of 30%.
Conclusions: Our results indicate that CWp in mice can be standardized by intra-operative
ICP monitoring. CWp leads to prolonged intracranial hypertension, selective neuronal cell
death in the hippocampus, and severe neurological dysfunction. CWp in mice with ICP
monitoring may therefore become a valuable tool for future investigations of the molecular
pathophysiology of SAH.
Keywords: SAH – mouse – intracranial pressure – neuronal cell death – functional outcome
2
Feiler et al., Brain damage and ICP after SAH in mice
1. Background
Cerebral hemorrhages cause only 20% of all strokes but are responsible for ~50% of the
entire stroke-related mortality, i.e. hemorrhagic stroke is 4.5 times more fatal than ischemic
stroke (Rosamond, Folsom et al., 1999). One of the most common and severe causes for
hemorrhagic stroke is subarachnoid hemorrhage (SAH), a bleeding from a large extra-
parenchymal cerebral vessel mostly caused by a ruptured congenital aneurysm. SAH has an
over all mortality of about 50% and over 30% of survivors remain severely disabled (van Gijn
and Rinkel, 2001). Despite these facts and some pathophysiological similarities, SAH is by
far less frequently investigated than ischemic stroke. Consequently, several important
aspects of the pathophysiology of SAH, especially on the molecular level, are not well
defined thereby hampering the development of novel therapeutic strategies (Cahill and
Zhang, 2009). One of the reasons responsible for this situation may be that murine models of
SAH which can be used with transgenic animals have so far not been fully characterized and
standardized (Strbian, Durukan et al., 2008). To date important clinical parameters such as
intracranial pressure and neuropathology have not been investigated following experimental
SAH in mice. In contrast to other well standardized murine models of acute brain injury, e.g.
ischemic stroke, successful induction and the severity of the insult are not monitored intra-
operatively leading to heterogeneous results which can only be compensated with group
sizes not easily compatible with difficult to generate transgenic mouse strains and by grading
the severity of experimental SAH only post-hoc (Gao, Wang et al., 2006;Sozen, Tsuchiyama
et al., 2009). This makes a randomized study design difficult to achieve and limits the value
of these otherwise clinically highly relevant animal models significantly (Strbian, Durukan et
al., 2008).
So far several research groups studied SAH in mice either by perforating the Circle of Willis
at the skull base using an intraluminal filament inserted through the external carotid artery
(Kamii, Kato et al., 1999;Sozen, Tsuchiyama et al., 2009;Ishikawa, Kusaka et al., 2009;Liu,
Tang et al., 2007;McGirt, Parra et al., 2002;Parra, McGirt et al., 2002;Saito, Kamii et al.,
3
Feiler et al., Brain damage and ICP after SAH in mice
2001;Gao, Wang et al., 2006;McGirt, Lynch et al., 2002), by directly injecting blood into the
cisterna magna (Lin, Calisaneller et al., 2003), or by perforating an intracisternal vein (Altay,
Smithason et al., 2009). Since perforation of an intracerebral vessel by a filament resembles
aneurysmal hemorrhage closer than intraventricular injection of blood, Circulus of Willis
perforation (CWp) became the most widely used SAH model in mice. Pioneering studies
using CWp demonstrated delayed cerebral vasospasm (Gao, Wang et al., 2006;Kamii, Kato
et al., 1999;McGirt, Parra et al., 2002;McGirt, Lynch et al., 2002;Parra, McGirt et al.,
2002;Saito, Kamii et al., 2001), neurological dysfunction (Gao, Wang et al., 2006;McGirt,
Parra et al., 2002;McGirt, Lynch et al., 2002;Parra, McGirt et al., 2002;Liu, Tang et al.,
2007;Sozen, Tsuchiyama et al., 2009), brain edema formation (Liu, Tang et al., 2007;Sozen,
Tsuchiyama et al., 2009), and a clinically relevant mortality of ~30% (Gao, Wang et al.,
2006;Liu, Tang et al., 2007;Sozen, Tsuchiyama et al., 2009). So far, however, it remains
unclear how the initial bleeding is linked to these rather late and severe pathophysiological
findings since it is not known if experimental SAH in mice causes intracranial hypertension,
reduces cerebral perfusion pressure, and causes subsequent ischemic brain damage.
Therefore, the aim of the current study was to characterize intracranial pressure (ICP),
cerebral perfusion pressure (CPP), cerebral blood flow (CBF), and neuronal cell death
following experimental SAH and to identify intra-operative monitoring parameters (ICP, CBF)
which may help to further standardize Circle of Willis perforation in mice.
4
Feiler et al., Brain damage and ICP after SAH in mice
2. Materials and methods
Male C57Bl6 mice (23 to 25 g body weight; Charles River Laboratory, Sulzfeld, Germany)
were used for experiments according to the guidelines of the Animal Care Committee of the
District Government of Upper Bavaria (Protocol Number Az 55.2-1-54-2531-118-05).
2.1. Animal preparation and monitoring
Animals had free access to food and water prior to and after surgery. Anesthesia was
induced by intraperitoneal injection of midazolam (5 mg/kg; Ratiopharm, Ulm, Germany),
fentanyl (0.05 mg/kg; CuraMed, Karlsruhe, Germany), and medetomidin (0.5 mg/kg; Pfizer,
Germany) and maintained by hourly injections of one quarter of the initial dose as previously
described (Thal and Plesnila, 2007). Mice were intubated and mechanically ventilated with
35-38% O2 in room air (Minivent, Hugo Sachs, Hugstetten, Germany). End-tidal pCO2 was
measured on-line with a microcapnometer (CI240, Columbus Instruments, Columbus, USA).
A thermostatically regulated, feedback-controlled heating pad (FHC, Bowdoinham, USA) was
used to maintain rectal temperature at 37°C. Anesthesia was terminated by intraperitoneal
injection of atipamezol (2.5 mg/kg; Pfizer, Karlsruhe, Germany), naloxon (1.2 mg/kg; Inresa,
Freiburg, Germany), and flumazenil (0.5 mg/kg; Hoffmann-La-Roche, Grenzach-Wyhlen,
Germany). To prevent hypothermia animals were kept in an incubator at 33°C ambient
temperature for 24 hours after surgery.
2.2. ICP measurement and laser-Doppler flowmetry
ICP was continuously measured in the epidural space of the right hemisphere from 40
minutes before until 30 minutes after SAH using a Codman ICP microsensor (Johnson &
Johnson Medical Limited, Berkshire, UK). To assess ICP 24 and 72 hours after SAH animals
were re-anesthetized, intubated, and the ICP probe was placed for 10 minutes into the
epidural space as described above.
For continuous monitoring of regional cerebral blood flow (rCBF) a flexible laser-Doppler
probe (Periflux 4001 Master, Perimed, Stockholm, Sweden) was glued onto the skull above
the territory of the left middle cerebral artery (MCA). rCBF was measured at a sampling rate
5
Feiler et al., Brain damage and ICP after SAH in mice
of 10 Hz from 30 minutes before until 30 minutes after SAH. The LDF probe was removed at
the end of surgery.
2.3. Induction of SAH by endovascular filament perforation
SAH was induced by a modification of the endovascular filament perforation method that has
been described in rats and mice previously (Bederson, Germano et al., 1995;Kamii, Kato et
al., 1999;Parra, McGirt et al., 2002). Briefly, animals were anesthetized and placed in a
supine position. The neck was opened by a midline incision and the left carotid artery was
exposed. A 5-0 monofilament was advanced via the external carotid artery (ECA) into the
internal carotid artery (ICA) until the ipsilateral rCBF decreased to ensure the position of the
tip of the filament near the bifurcation of the ICA and the MCA. Then the filament was pushed
further until a sharp increase in ICP indicated the successful induction of SAH. Subsequently,
the suture was withdrawn into the ECA to allow full perfusion of the ICA. In sham operated
animals the same procedure was performed, i.e. the suture was introduced in the ICA,
without causing a rCBF decrease and SAH.
2.4. Neuroscore and body weight
Neurological deficits were assessed daily for 7 days by an investigator blinded towards the
treatment of the animals. Neurological deficits were quantified using a neuroscore as
previously described (Lehmberg, Beck et al., 2000) (Tab. 2). Mice with no neurological
deficits received a score of 0 points, animals with the most severe loss of neurological
function a score of 33 points. Body weight, a sensitive indicator for the overall well-being of
rodents, was assessed daily for 7 days using a balance with movement artifact correction
(Mettler-Toledo, Giessen, Germany).
2.5. Quantification of Cerebral Edema
Animals were sacrificed 24 or 72 hours after SAH and the wet weight (WW) of both
hemispheres was assessed. Thereafter, the hemispheres were dried for 24 h at 110°C and
their dry weight (DW) was determined. Hemispheric water content (%) was calculated using
the following formula ((WW-DW) / WW) x 100.
6
Feiler et al., Brain damage and ICP after SAH in mice
2.6. Quantification of Ischemic Brain damage
Animals were anesthetized and transcardially perfused with 2% paraformaldehyde (PFA) 7
days after SAH. Brains were removed, dehydrated, and embedded in paraffin. Coronal
sections (bregma -1.6 mm) were prepared and stained with Cresyl Violet. The number of
surviving neurons in the hippocampus was quantified in a region of interest of 0.4 x 0.4 mm
as previously described (Bermueller, Thal et al., 2006).
2.7. Experimental groups
In a first experimental series physiological parameters were assessed and the neurological
function was assessed for 7 days after SAH or sham surgery (n=6 each). In a parallel series
mortality was quantified in SAH and sham operated mice (n=10 per group). In a second
series ICP was measured 24 or 72 hours after SAH or in sham operated animals (n=6 each).
In a third series brain water content was measured 24 or 72 hours after SAH or in sham
operated animals (n=6 each). In a fourth series ischemic neuronal damage was assessed in
sham operated animals or 7 days after SAH (n=6 each). Accordingly, 80 animals were
included in the current analysis.
Animals that died during and after the operation were replaced until the final group size was
n=6. In the series where mortality was assessed animals were not replaced.
2.8. Statistical Analysis
Data are presented as means ± SEM if not indicated otherwise. Statistical analysis was
performed with the software package SigmaStat 3.1 (SPSS Science Inc., Chicago, IL, USA).
Data over time were analyzed with Kruskal-Wallis ANOVA on ranks. The Student-Newman-
Keuls Test was used for multiple comparisons. Mortality was compared between groups with
the Survival Log Rank Test. Statistical significance was assumed at p<0.05.
7
Feiler et al., Brain damage and ICP after SAH in mice
3. RESULTS
3.1. Physiological Parameters
SAH by filament perforation resulted in an immediate increase of ICP to a peak of 78.5 ± 9.3
mmHg (p<0.05 vs. baseline) followed by a plateau that was reached between 5 minutes
(39.6 ± 8.4 mmHg) and 10 minutes (20.0 ± 3.6 mmHg) after SAH. In sham-operated animals,
ICP remained around the physiological level of 5.0 ± 0.5 mmHg for the whole monitoring
period (Fig. 1A). The increase in ICP resulted in an immediate increase of mean arterial
blood pressure (p<0.05 vs. sham), the well known Cushing response (Fig. 1B), in a
significant drop in cerebral perfusion pressure (CPP) down to 25 mmHg (p<0.05 vs. sham;
Fig. 1C), and the subsequent reduction of cerebral blood flow (CBF) to ischemic levels (19 ±
4% of baseline; p<0.05 vs. sham; Fig. 1D). After ICP dropped to values around 20 mmHg
CCP normalized and CBF recovered to normal values of 80 ± 9.8% of baseline. Except the
Cushing response mice subjected to SAH or sham surgery did not differ concerning blood
pressure and blood gases (Tab. 1).
Vessel perforation resulted in a hematoma at the perforation site (Fig. 1E, dotted circle) and
in a significant extravasation of blood into the subarachnoid space of both hemispheres (Fig.
1E) indicating severe SAH. These changes were only observed when advancement of the
filament into the Circle of Willis was accompanied by a sharp increase in ICP. An isolated
drop in CBF, a method proposed to be useful for monitoring SAH induction in mice, did not
necessarily result in SAH as evidenced by simultaneous ICP measurements (Fig. 1F) and
subsequent visual inspection of blood extravasation. Therefore, ICP measurements are
mandatory in order to reliably monitor experimental SAH in mice.
3.2. Mortality
One animal of the filament perforation group died on postoperative day 1 and another 2
animals died on postoperative day 2 (30% mortality). There was no mortality in sham
operated animals (n=10 each; Fig. 2A).
8
Feiler et al., Brain damage and ICP after SAH in mice
3.3. Cerebral Edema
SAH by filament perforation resulted in a significant (p<0.05) and continuous increase of
brain water content in both hemispheres up to 72 hours after SAH (Fig. 2B).
3.4. Chronic ICP-Measurement
Brain edema formation resulted in significant intracranial hypertension 24 hours after SAH
(10.0 ± 0.7 mmHg; p<0.05; Fig. 2C). Three days after SAH ICP returned to normal values.
3.5. Brain Damage
SAH in mice did not result in infarction but in selective, bilateral neuronal loss in the
hippocampus (Fig. 3A). Quantification of the number of viable hippocampal neurons 7 days
after SAH revealed a significant loss (p<0.05) of neurons in all regions of the hippocampus
as compared to sham operated controls. The most severe loss was observed in the
ipsilateral CA3 region (Fig. 3B). No cell death was observed in the cerebral cortex or in the
striatum (Fig. 3C).
3.6. Neurological Evaluation and Body Weight
SAH resulted in severe motor and behavioral deficits (15 out of 33 points) during the first 3
days after SAH. Thereafter mice gradually but incompletely recovered until the end of the
observation period as compared to sham operated animals (p<0.05; Fig. 4A). In addition to
the neurological deficits mice of the SAH group exhibited a significant and long lasting loss of
body weight (- 20%) with only minimal recovery (p<0.001; Fig. 4B).
9
Feiler et al., Brain damage and ICP after SAH in mice
4. DISCUSSION
In the current study we characterized the pattern of brain damage after filament perforation-
induced SAH in mice and demonstrate that the measurement of intracranial pressure is the
only parameter able to reliably detect the induction of SAH by filament perforation.
So far most animals models of SAH were performed on rats, rabbits, cats, or primates (Barry,
Gogjian et al., 1979;Delgado, Brismar et al., 1985;Dorsch, Branston et al., 1989). Despite
multiple advantages, e.g. availability, size, or available standardized models, these species
have, in contrast to mice, the inherent disadvantage of not being amenable to genetic
manipulation. Therefore several groups developed murine SAH models by directly injecting
blood into the cisterna magna (Lin, Calisaneller et al., 2003), dissecting an intracisternal vein
(Altay, Smithason et al., 2009), or perforating an artery at the skull base by introducing a
filament into the cerebral circulation through an extra-cranial approach (Kamii, Kato et al.,
1999;Sozen, Tsuchiyama et al., 2009;Ishikawa, Kusaka et al., 2009;Liu, Tang et al.,
2007;McGirt, Parra et al., 2002;Parra, McGirt et al., 2002;Saito, Kamii et al., 2001;Gao,
Wang et al., 2006;McGirt, Lynch et al., 2002). The cisterna magna injection model and the
venous dissection model can be performed quickly, are highly reproducible due to the
controlled application of the blood, result in an acute increase of ICP and subsequent
decrease of CBF, and lead the development of brain edema and intracranial hypertension as
also observed in man (Broderick, Brott et al., 1994;Claassen, Carhuapoma et al., 2002).
However, both models lack ruptured vessels and endothelial damage which are important
factors of the pathology of SAH (Sobey and Faraci, 1998). Therefore several groups, in
analogy to a model used in rats, developed a murine model where the Circulus of Willis
(CWp) is perforated by an intraluminal filament introduced through the carotid artery. Due to
the obvious replication of the human pathophysiology this model quickly became the most
frequently and widely used experimental SAH model in mice (Kamii, Kato et al., 1999;Sozen,
Tsuchiyama et al., 2009;Ishikawa, Kusaka et al., 2009;Liu, Tang et al., 2007;McGirt, Parra et
al., 2002;Parra, McGirt et al., 2002;Saito, Kamii et al., 2001;Gao, Wang et al., 2006;McGirt,
10
Feiler et al., Brain damage and ICP after SAH in mice
Lynch et al., 2002). CWp causes delayed cerebral vasospasm (Gao, Wang et al.,
2006;Kamii, Kato et al., 1999;McGirt, Parra et al., 2002;McGirt, Lynch et al., 2002;Parra,
McGirt et al., 2002;Saito, Kamii et al., 2001), neurological dysfunction (Gao, Wang et al.,
2006;McGirt, Parra et al., 2002;McGirt, Lynch et al., 2002;Parra, McGirt et al., 2002;Liu,
Tang et al., 2007;Sozen, Tsuchiyama et al., 2009), brain edema (Liu, Tang et al.,
2007;Sozen, Tsuchiyama et al., 2009), and a clinically relevant mortality of ~30% (Gao,
Wang et al., 2006;Liu, Tang et al., 2007;Sozen, Tsuchiyama et al., 2009). CWp has however,
also some significant disadvantages. So far clinically and pathophysiologically important
findings linking these late changes to the initial bleeding such as intracranial pressure and
neuronal damage were not investigated and CWp has the inherent problem that the volume
of intracranial blood can neither be controlled nor monitored. Accordingly, so far CWp falls
short fulfilling the first and second criteria of an ideal SAH model proposed by Schwartz et al.
which are 1) reproducible and consistent blood deposition in the subarachnoid space 2)
uniform, controlled degree of hemorrhage 3) mechanism of hemorrhage closely simulating
aneurysmal SAH 4) blood distribution correlating with aneurysmal SAH 5) ease of
performance and 6) reasonable costs (Schwartz, Masago et al., 2000). Our results clearly
show that monitoring cerebral blood flow is not sufficient to detect successful induction of
SAH since already the advancement of the filament into the internal carotid artery may cause
a drop of CBF without an increase of intracranial pressure (Fig. 1F). Accordingly, the
increase of intracranial pressure, which is a direct measure for the space occupying effect of
the nascent intracranial hematoma, is the only reliable parameter necessary to be recorded
in order to correctly detect induction of SAH in mice. Since the ICP increase and the absolute
ICP values following SAH are very uniform and show little scattering, we conclude that also
the amount of extravasated blood is quite uniform in mice. Hence, CWp exerts many, if not
all, criteria claimed to be important for experimental SAH (see above).
So far the pattern of SAH induced brain damage was not known in mice. Our results indicate
that CWp in mice causes mainly damage to the hippocampus while the cortex and the
11
Feiler et al., Brain damage and ICP after SAH in mice
striatum are not significantly affected. Interestingly, except of the ipsilateral CA3 region which
is most severely damaged, cell death affects both hippocampi equally. This indicates that
either cell death is caused by the initial global increase of ICP and subsequent global
ischemia or that the extravasated blood is distributed in a very uniform fashion within the
subarachnoid space thereby causing acute vasospasm and a subsequent reduction of
cerebral blood flow in both hemispheres. We favor the latter explanation due to the actually
demonstrated quite even distribution of blood in the subarachnoid space (Fig. 1E) and some
recent results in man indicating ICP-independent early ischemia after SAH (Schubert, Seiz et
al., 2009). However, only additional experiments measuring absolute levels of CBF with high
spatial resolution by 14C-iodoantipyrene autoradiography at different time points after CWp
and performing experiments on craniotomized animals which will not display the initial ICP
peak will finally prove this hypothesis.
5. Conclusion
Taken together, the current study characterizes the pattern of neuronal damage following the
intraluminal perforation SAH model in mice and demonstrates that the measurement of
intracranial pressure is the only parameter which reliable detects the induction of SAH. The
results of the current study further characterize and standardize the most widely used model
of murine SAH and help to pave the way for further investigations of the molecular
mechanisms of SAH-induced brain damage by using transgenic animals.
12
Feiler et al., Brain damage and ICP after SAH in mice
RERERENCES
Rosamond WD, Folsom AR, Chambless LE, Wang CH, McGovern PG, Howard G, Copper
LS, Shahar E. Stroke incidence and survival among middle-aged adults: 9-year follow-up of
the Atherosclerosis Risk in Communities (ARIC) cohort. Stroke., 1999; 30: 736-43.
van Gijn J, Rinkel GJ. Subarachnoid haemorrhage: diagnosis, causes and management.
Brain, 2001; 124: 249-78.
Cahill J, Zhang JH. Subarachnoid hemorrhage: is it time for a new direction? Stroke., 2009;
40: S86-S87.
Strbian D, Durukan A, Tatlisumak T. Rodent models of hemorrhagic stroke. Curr. Pharm.
Des., 2008; 14: 352-8.
Gao J, Wang H, Sheng H, Lynch JR, Warner DS, Durham L, Vitek MP, Laskowitz DT. A
novel apoE-derived therapeutic reduces vasospasm and improves outcome in a murine
model of subarachnoid hemorrhage. Neurocrit. Care., 2006; 4: 25-31.
Sozen T, Tsuchiyama R, Hasegawa Y, Suzuki H, Jadhav V, Nishizawa S, Zhang JH. Role of
interleukin-1beta in early brain injury after subarachnoid hemorrhage in mice. Stroke., 2009;
40: 2519-25.
Kamii H, Kato I, Kinouchi H, Chan PH, Epstein CJ, Akabane A, Okamoto H, Yoshimoto T.
Amelioration of vasospasm after subarachnoid hemorrhage in transgenic mice
overexpressing CuZn-superoxide dismutase. Stroke, 1999; 30: 867-71.
Sozen T, Tsuchiyama R, Hasegawa Y, Suzuki H, Jadhav V, Nishizawa S, Zhang JH. Role of
interleukin-1beta in early brain injury after subarachnoid hemorrhage in mice. Stroke., 2009;
40: 2519-25.
13
Feiler et al., Brain damage and ICP after SAH in mice
Ishikawa M, Kusaka G, Yamaguchi N, Sekizuka E, Nakadate H, Minamitani H, Shinoda S,
Watanabe E. Platelet and leukocyte adhesion in the microvasculature at the cerebral surface
immediately after subarachnoid hemorrhage. Neurosurgery., 2009; 64: 546-53.
Liu S, Tang J, Ostrowski RP, Titova E, Monroe C, Chen W, Lo W, Martin R, Zhang JH.
Oxidative stress after subarachnoid hemorrhage in gp91phox knockout mice. Can. J. Neurol.
Sci., 2007; 34: 356-61.
McGirt MJ, Parra A, Sheng H, Higuchi Y, Oury TD, Laskowitz DT, Pearlstein RD, Warner DS.
Attenuation of cerebral vasospasm after subarachnoid hemorrhage in mice overexpressing
extracellular superoxide dismutase. Stroke., 2002; 33: 2317-23.
Parra A, McGirt MJ, Sheng H, Laskowitz DT, Pearlstein RD, Warner DS. Mouse model of
subarachnoid hemorrhage associated cerebral vasospasm: methodological analysis. Neurol.
Res., 2002; 24: 510-6.
Saito A, Kamii H, Kato I, Takasawa S, Kondo T, Chan PH, Okamoto H, Yoshimoto T.
Transgenic CuZn-superoxide dismutase inhibits NO synthase induction in experimental
subarachnoid hemorrhage. Stroke., 2001; 32: 1652-7.
McGirt MJ, Lynch JR, Parra A, Sheng H, Pearlstein RD, Laskowitz DT, Pelligrino DA, Warner
DS. Simvastatin increases endothelial nitric oxide synthase and ameliorates cerebral
vasospasm resulting from subarachnoid hemorrhage. Stroke., 2002; 33: 2950-6.
Lin CL, Calisaneller T, Ukita N, Dumont AS, Kassell NF, Lee KS. A murine model of
subarachnoid hemorrhage-induced cerebral vasospasm. J. Neurosci. Methods, 2003; 123:
89-97.
Altay T, Smithason S, Volokh N, Rasmussen PA, Ransohoff RM, Provencio JJ. A novel
method for subarachnoid hemorrhage to induce vasospasm in mice. J. Neurosci. Methods.,
2009.
14
Feiler et al., Brain damage and ICP after SAH in mice
Kamii H, Kato I, Kinouchi H, Chan PH, Epstein CJ, Akabane A, Okamoto H, Yoshimoto T.
Amelioration of vasospasm after subarachnoid hemorrhage in transgenic mice
overexpressing CuZn-superoxide dismutase. Stroke, 1999; 30: 867-71.
McGirt MJ, Parra A, Sheng H, Higuchi Y, Oury TD, Laskowitz DT, Pearlstein RD, Warner DS.
Attenuation of cerebral vasospasm after subarachnoid hemorrhage in mice overexpressing
extracellular superoxide dismutase. Stroke., 2002; 33: 2317-23.
McGirt MJ, Lynch JR, Parra A, Sheng H, Pearlstein RD, Laskowitz DT, Pelligrino DA, Warner
DS. Simvastatin increases endothelial nitric oxide synthase and ameliorates cerebral
vasospasm resulting from subarachnoid hemorrhage. Stroke., 2002; 33: 2950-6.
Parra A, McGirt MJ, Sheng H, Laskowitz DT, Pearlstein RD, Warner DS. Mouse model of
subarachnoid hemorrhage associated cerebral vasospasm: methodological analysis. Neurol.
Res., 2002; 24: 510-6.
Saito A, Kamii H, Kato I, Takasawa S, Kondo T, Chan PH, Okamoto H, Yoshimoto T.
Transgenic CuZn-superoxide dismutase inhibits NO synthase induction in experimental
subarachnoid hemorrhage. Stroke., 2001; 32: 1652-7.
McGirt MJ, Parra A, Sheng H, Higuchi Y, Oury TD, Laskowitz DT, Pearlstein RD, Warner DS.
Attenuation of cerebral vasospasm after subarachnoid hemorrhage in mice overexpressing
extracellular superoxide dismutase. Stroke., 2002; 33: 2317-23.
McGirt MJ, Lynch JR, Parra A, Sheng H, Pearlstein RD, Laskowitz DT, Pelligrino DA, Warner
DS. Simvastatin increases endothelial nitric oxide synthase and ameliorates cerebral
vasospasm resulting from subarachnoid hemorrhage. Stroke., 2002; 33: 2950-6.
Parra A, McGirt MJ, Sheng H, Laskowitz DT, Pearlstein RD, Warner DS. Mouse model of
subarachnoid hemorrhage associated cerebral vasospasm: methodological analysis. Neurol.
Res., 2002; 24: 510-6.
15
Feiler et al., Brain damage and ICP after SAH in mice
Liu S, Tang J, Ostrowski RP, Titova E, Monroe C, Chen W, Lo W, Martin R, Zhang JH.
Oxidative stress after subarachnoid hemorrhage in gp91phox knockout mice. Can. J. Neurol.
Sci., 2007; 34: 356-61.
Sozen T, Tsuchiyama R, Hasegawa Y, Suzuki H, Jadhav V, Nishizawa S, Zhang JH. Role of
interleukin-1beta in early brain injury after subarachnoid hemorrhage in mice. Stroke., 2009;
40: 2519-25.
Liu S, Tang J, Ostrowski RP, Titova E, Monroe C, Chen W, Lo W, Martin R, Zhang JH.
Oxidative stress after subarachnoid hemorrhage in gp91phox knockout mice. Can. J. Neurol.
Sci., 2007; 34: 356-61.
Sozen T, Tsuchiyama R, Hasegawa Y, Suzuki H, Jadhav V, Nishizawa S, Zhang JH. Role of
interleukin-1beta in early brain injury after subarachnoid hemorrhage in mice. Stroke., 2009;
40: 2519-25.
Liu S, Tang J, Ostrowski RP, Titova E, Monroe C, Chen W, Lo W, Martin R, Zhang JH.
Oxidative stress after subarachnoid hemorrhage in gp91phox knockout mice. Can. J. Neurol.
Sci., 2007; 34: 356-61.
Sozen T, Tsuchiyama R, Hasegawa Y, Suzuki H, Jadhav V, Nishizawa S, Zhang JH. Role of
interleukin-1beta in early brain injury after subarachnoid hemorrhage in mice. Stroke., 2009;
40: 2519-25.
Thal SC, Plesnila N. Non-invasive intraoperative monitoring of blood pressure and arterial
pCO2 during surgical anesthesia in mice. J. Neurosci. Methods, 2007; 159: 261-7.
Bederson JB, Germano IM, Guarino L. Cortical blood flow and cerebral perfusion pressure in
a new noncraniotomy model of subarachnoid hemorrhage in the rat. Stroke, 1995; 26: 1086-
91.
16
Feiler et al., Brain damage and ICP after SAH in mice
Kamii H, Kato I, Kinouchi H, Chan PH, Epstein CJ, Akabane A, Okamoto H, Yoshimoto T.
Amelioration of vasospasm after subarachnoid hemorrhage in transgenic mice
overexpressing CuZn-superoxide dismutase. Stroke, 1999; 30: 867-71.
Parra A, McGirt MJ, Sheng H, Laskowitz DT, Pearlstein RD, Warner DS. Mouse model of
subarachnoid hemorrhage associated cerebral vasospasm: methodological analysis. Neurol.
Res., 2002; 24: 510-6.
Lehmberg J, Beck J, Baethmann A, Uhl E. Influence of the bradykinin B1/B2-receptor-
antagonist B 9430 on the cerebral microcirculation and outcome of gerbils from global
cerebral ischemia. Acta Neurochir. Suppl, 2000; 76: 39-41.
Bermueller C, Thal SC, Plesnila N, Schmid-Elsaesser R, Kreimeier U, Zausinger S.
Hypertonic fluid resuscitation from subarachnoid hemorrhage in rats: a comparison between
small volume resuscitation and mannitol. J. Neurol. Sci., 2006; 241: 73-82.
Barry KJ, Gogjian MA, Stein BM. Small animal model for investigation of subarachnoid
hemorrhage and cerebral vasospasm. Stroke, 1979; 10: 538-41.
Delgado TJ, Brismar J, Svendgaard NA. Subarachnoid haemorrhage in the rat: angiography
and fluorescence microscopy of the major cerebral arteries. Stroke, 1985; 16: 595-602.
Dorsch N, Branston NM, Symon L, Jakubowski J. Intracranial pressure changes following
primate subarachnoid haemorrhage. Neurol. Res., 1989; 11: 201-4.
Lin CL, Calisaneller T, Ukita N, Dumont AS, Kassell NF, Lee KS. A murine model of
subarachnoid hemorrhage-induced cerebral vasospasm. J. Neurosci. Methods, 2003; 123:
89-97.
Altay T, Smithason S, Volokh N, Rasmussen PA, Ransohoff RM, Provencio JJ. A novel
method for subarachnoid hemorrhage to induce vasospasm in mice. J. Neurosci. Methods.,
2009.
17
Feiler et al., Brain damage and ICP after SAH in mice
Kamii H, Kato I, Kinouchi H, Chan PH, Epstein CJ, Akabane A, Okamoto H, Yoshimoto T.
Amelioration of vasospasm after subarachnoid hemorrhage in transgenic mice
overexpressing CuZn-superoxide dismutase. Stroke, 1999; 30: 867-71.
Sozen T, Tsuchiyama R, Hasegawa Y, Suzuki H, Jadhav V, Nishizawa S, Zhang JH. Role of
interleukin-1beta in early brain injury after subarachnoid hemorrhage in mice. Stroke., 2009;
40: 2519-25.
Ishikawa M, Kusaka G, Yamaguchi N, Sekizuka E, Nakadate H, Minamitani H, Shinoda S,
Watanabe E. Platelet and leukocyte adhesion in the microvasculature at the cerebral surface
immediately after subarachnoid hemorrhage. Neurosurgery., 2009; 64: 546-53.
Liu S, Tang J, Ostrowski RP, Titova E, Monroe C, Chen W, Lo W, Martin R, Zhang JH.
Oxidative stress after subarachnoid hemorrhage in gp91phox knockout mice. Can. J. Neurol.
Sci., 2007; 34: 356-61.
McGirt MJ, Parra A, Sheng H, Higuchi Y, Oury TD, Laskowitz DT, Pearlstein RD, Warner DS.
Attenuation of cerebral vasospasm after subarachnoid hemorrhage in mice overexpressing
extracellular superoxide dismutase. Stroke., 2002; 33: 2317-23.
Parra A, McGirt MJ, Sheng H, Laskowitz DT, Pearlstein RD, Warner DS. Mouse model of
subarachnoid hemorrhage associated cerebral vasospasm: methodological analysis. Neurol.
Res., 2002; 24: 510-6.
Saito A, Kamii H, Kato I, Takasawa S, Kondo T, Chan PH, Okamoto H, Yoshimoto T.
Transgenic CuZn-superoxide dismutase inhibits NO synthase induction in experimental
subarachnoid hemorrhage. Stroke., 2001; 32: 1652-7.
McGirt MJ, Lynch JR, Parra A, Sheng H, Pearlstein RD, Laskowitz DT, Pelligrino DA, Warner
DS. Simvastatin increases endothelial nitric oxide synthase and ameliorates cerebral
vasospasm resulting from subarachnoid hemorrhage. Stroke., 2002; 33: 2950-6.
18
Feiler et al., Brain damage and ICP after SAH in mice
Broderick JP, Brott TG, Duldner JE, Tomsick T, Leach A. Initial and recurrent bleeding are
the major causes of death following subarachnoid hemorrhage. Stroke, 1994; 25: 1342-7.
Claassen J, Carhuapoma JR, Kreiter KT, Du EY, Connolly ES, Mayer SA. Global cerebral
edema after subarachnoid hemorrhage: frequency, predictors, and impact on outcome.
Stroke, 2002; 33: 1225-32.
Sobey CG, Faraci FM. Subarachnoid haemorrhage: what happens to the cerebral arteries?
Clin. Exp. Pharmacol. Physiol, 1998; 25: 867-76.
Kamii H, Kato I, Kinouchi H, Chan PH, Epstein CJ, Akabane A, Okamoto H, Yoshimoto T.
Amelioration of vasospasm after subarachnoid hemorrhage in transgenic mice
overexpressing CuZn-superoxide dismutase. Stroke, 1999; 30: 867-71.
Sozen T, Tsuchiyama R, Hasegawa Y, Suzuki H, Jadhav V, Nishizawa S, Zhang JH. Role of
interleukin-1beta in early brain injury after subarachnoid hemorrhage in mice. Stroke., 2009;
40: 2519-25.
Ishikawa M, Kusaka G, Yamaguchi N, Sekizuka E, Nakadate H, Minamitani H, Shinoda S,
Watanabe E. Platelet and leukocyte adhesion in the microvasculature at the cerebral surface
immediately after subarachnoid hemorrhage. Neurosurgery., 2009; 64: 546-53.
Liu S, Tang J, Ostrowski RP, Titova E, Monroe C, Chen W, Lo W, Martin R, Zhang JH.
Oxidative stress after subarachnoid hemorrhage in gp91phox knockout mice. Can. J. Neurol.
Sci., 2007; 34: 356-61.
McGirt MJ, Parra A, Sheng H, Higuchi Y, Oury TD, Laskowitz DT, Pearlstein RD, Warner DS.
Attenuation of cerebral vasospasm after subarachnoid hemorrhage in mice overexpressing
extracellular superoxide dismutase. Stroke., 2002; 33: 2317-23.
19
Feiler et al., Brain damage and ICP after SAH in mice
Parra A, McGirt MJ, Sheng H, Laskowitz DT, Pearlstein RD, Warner DS. Mouse model of
subarachnoid hemorrhage associated cerebral vasospasm: methodological analysis. Neurol.
Res., 2002; 24: 510-6.
Saito A, Kamii H, Kato I, Takasawa S, Kondo T, Chan PH, Okamoto H, Yoshimoto T.
Transgenic CuZn-superoxide dismutase inhibits NO synthase induction in experimental
subarachnoid hemorrhage. Stroke., 2001; 32: 1652-7.
McGirt MJ, Lynch JR, Parra A, Sheng H, Pearlstein RD, Laskowitz DT, Pelligrino DA, Warner
DS. Simvastatin increases endothelial nitric oxide synthase and ameliorates cerebral
vasospasm resulting from subarachnoid hemorrhage. Stroke., 2002; 33: 2950-6.
Kamii H, Kato I, Kinouchi H, Chan PH, Epstein CJ, Akabane A, Okamoto H, Yoshimoto T.
Amelioration of vasospasm after subarachnoid hemorrhage in transgenic mice
overexpressing CuZn-superoxide dismutase. Stroke, 1999; 30: 867-71.
McGirt MJ, Parra A, Sheng H, Higuchi Y, Oury TD, Laskowitz DT, Pearlstein RD, Warner DS.
Attenuation of cerebral vasospasm after subarachnoid hemorrhage in mice overexpressing
extracellular superoxide dismutase. Stroke., 2002; 33: 2317-23.
McGirt MJ, Lynch JR, Parra A, Sheng H, Pearlstein RD, Laskowitz DT, Pelligrino DA, Warner
DS. Simvastatin increases endothelial nitric oxide synthase and ameliorates cerebral
vasospasm resulting from subarachnoid hemorrhage. Stroke., 2002; 33: 2950-6.
Parra A, McGirt MJ, Sheng H, Laskowitz DT, Pearlstein RD, Warner DS. Mouse model of
subarachnoid hemorrhage associated cerebral vasospasm: methodological analysis. Neurol.
Res., 2002; 24: 510-6.
Saito A, Kamii H, Kato I, Takasawa S, Kondo T, Chan PH, Okamoto H, Yoshimoto T.
Transgenic CuZn-superoxide dismutase inhibits NO synthase induction in experimental
subarachnoid hemorrhage. Stroke., 2001; 32: 1652-7.
20
Feiler et al., Brain damage and ICP after SAH in mice
McGirt MJ, Parra A, Sheng H, Higuchi Y, Oury TD, Laskowitz DT, Pearlstein RD, Warner DS.
Attenuation of cerebral vasospasm after subarachnoid hemorrhage in mice overexpressing
extracellular superoxide dismutase. Stroke., 2002; 33: 2317-23.
McGirt MJ, Lynch JR, Parra A, Sheng H, Pearlstein RD, Laskowitz DT, Pelligrino DA, Warner
DS. Simvastatin increases endothelial nitric oxide synthase and ameliorates cerebral
vasospasm resulting from subarachnoid hemorrhage. Stroke., 2002; 33: 2950-6.
Parra A, McGirt MJ, Sheng H, Laskowitz DT, Pearlstein RD, Warner DS. Mouse model of
subarachnoid hemorrhage associated cerebral vasospasm: methodological analysis. Neurol.
Res., 2002; 24: 510-6.
Liu S, Tang J, Ostrowski RP, Titova E, Monroe C, Chen W, Lo W, Martin R, Zhang JH.
Oxidative stress after subarachnoid hemorrhage in gp91phox knockout mice. Can. J. Neurol.
Sci., 2007; 34: 356-61.
Sozen T, Tsuchiyama R, Hasegawa Y, Suzuki H, Jadhav V, Nishizawa S, Zhang JH. Role of
interleukin-1beta in early brain injury after subarachnoid hemorrhage in mice. Stroke., 2009;
40: 2519-25.
Liu S, Tang J, Ostrowski RP, Titova E, Monroe C, Chen W, Lo W, Martin R, Zhang JH.
Oxidative stress after subarachnoid hemorrhage in gp91phox knockout mice. Can. J. Neurol.
Sci., 2007; 34: 356-61.
Sozen T, Tsuchiyama R, Hasegawa Y, Suzuki H, Jadhav V, Nishizawa S, Zhang JH. Role of
interleukin-1beta in early brain injury after subarachnoid hemorrhage in mice. Stroke., 2009;
40: 2519-25.
Liu S, Tang J, Ostrowski RP, Titova E, Monroe C, Chen W, Lo W, Martin R, Zhang JH.
Oxidative stress after subarachnoid hemorrhage in gp91phox knockout mice. Can. J. Neurol.
Sci., 2007; 34: 356-61.
21
Feiler et al., Brain damage and ICP after SAH in mice
Sozen T, Tsuchiyama R, Hasegawa Y, Suzuki H, Jadhav V, Nishizawa S, Zhang JH. Role of
interleukin-1beta in early brain injury after subarachnoid hemorrhage in mice. Stroke., 2009;
40: 2519-25.
Schwartz AY, Masago A, Sehba FA, Bederson JB. Experimental models of subarachnoid
hemorrhage in the rat: a refinement of the endovascular filament model. J. Neurosci.
Methods, 2000; 96: 161-7.
Schubert GA, Seiz M, Hegewald AA, Manville J, Thome C. Acute hypoperfusion immediately
after subarachnoid hemorrhage: a xenon contrast-enhanced CT study. J. Neurotrauma,
2009; 26: 2225-31.
22
Feiler et al., Brain damage and ICP after SAH in mice
FIGURE LEGENDS
Figure 1
Time course of ICP (A), mean arterial blood pressure (B), cerebral perfusion pressure (C),
and rCBF (D) before and after induction of SAH in mice by perforation of the Circulus of
Willis by an intraluminal filament (CWp; n=8). CWp results in a uniform distribution of blood in
the subarachnoid space (E). Monitoring the induction of SAH by rCBF alone may result in
false positive results since advancement of the filament into the Circulus of Willis may result
in a reduction of rCBF without vessel perforation as indicated by the lack of ICP increase (F).
Figure 2
Mortality during the first 7 days after CWp (A; n=10). Brain water content (B) and intracranial
pressure (ICP; n=6; C) 24 and 72 hours after murine SAH.
Figure 3
Brain damage 7 days after SAH by CWp in mice. Neuronal cell death is observed bilaterally
in the hippocampus (A & B) while the cortex and the striatum are not significantly affected
(C).
Figure 4:
Functional outcome following SAH in mice by CWp (n=6). Neuroscore performance is
significantly altered during the first three days after SAH but recovers thereafter ( A), while
body weight reduces gradually during the same period of time and shows only little recovery
(B).
23
Feiler et al., Brain damage and ICP after SAH in mice
Table 1
24
pH pCO pO HCO3- Na+KCa2++ Cl
SAH 7.3 ±
0.02
±82.0 ±
10.7
±144.5 ± ± 1.3
±
0.03
112.7 ±
Sham 7.28
±
0.02
± ±
22.6
20.2 ±154.7 ±
2.2
4.9 ±
0.4
1.3 ±
0.01
113.1 ±
1.3
pH pCO2pO2Na K+Cl-
SAH 7.30
0.02
41.8 ±
3.5
±
10.7
20.2 ±
1.2
±
1.9
4.6 ±
0.2
1.3
±
0.03
±
1.1
Sham 7.28
±
0.02
43.8 ±
5.6
80.0 ±
22.6
±
2.1
± ± ±
0.01
±
Physiological parameters 30 min after SAH
Feiler et al., Brain damage and ICP after SAH in mice
Table 2
25
Neuroscore
Item Points Best Worse
Grasp Reflex (1) per paw, if
missing 0 4
Righting Reflexes
orienting the head (2) per side, if
missing 0 4
falling reflex (1) if missing 0 1
Mobility
spontaneous
(0) normal
(4) intermediate
(8) missing
0 8
circling (2) if present 0 2
Fur (0) smooth
(4) fuzzy 0 4
Nibbling
(0) normal
(4) intermediate
(8) missing
0 8
Flight (0) normal
(2) missing 0 2
Total 0 33
Item Points Best Worse
Grasp Reflex (1) per paw, if
missing 0 4
Righting Reflexes
orienting the head (2) per side, if
missing 0 4
falling reflex (1) if missing 0 1
Mobility
(0) normal
(4) intermediate
(8) missing
0 8
circling (2) if present 0 2
Fur (0) smooth
(4) fuzzy 0 4
Nibbling
(0) normal
intermediate
(8) missing
0 8
Flight 0 2
... Most of the blood can thereafter be found in the subarachnoid space, while amounts of blood vary (Hubschmann and Kornhauser 1980). The neurological deficits after endovascular perforation are severe (Feiler et al. 2010;Matsumura et al. 2019). Rolling and adhesion of leukocytes as sign of increased leukocyte-endothelial interaction can be observed early after endovascular perforation, while they are not present in the prechiasmatic injection model presented below. ...
... Beyond the aforementioned sufferings, SAH induces neuronal damage (Güresir et al. 2015). Sequelae of neuronal damage are neurological deficits, which, in case of endovascular perforation, are described as severe (Feiler et al. 2010). Examples of observable neurological restrictions are forelimb flexion, decreased resistance to lateral push, without and, as sign of deterioration, with circling. ...
Article
Full-text available
Subarachnoid hemorrhage is an acute life-threatening cerebrovascular disease with high socio-economic impact. The most frequent cause, the rupture of an intracerebral aneurysm, is accompanied by abrupt changes in intracerebral pressure, cerebral perfusion pressure and, consequently, cerebral blood flow. As aneurysms rupture spontaneously, monitoring of these parameters in patients is only possible with a time delay, upon hospitalization. To study alterations in cerebral perfusion immediately upon ictus, animal models are mandatory. This article addresses the points necessarily to be included in an animal project proposal according to EU directive 2010/63/EU for the protection of animals used for scientific purposes and herewith offers an insight into animal welfare aspects of using rodent models for the investigation of cerebral perfusion after subarachnoid hemorrhage. It compares surgeries, model characteristics, advantages, and drawbacks of the most-frequently used rodent models—the endovascular perforation model and the prechiasmatic and single or double cisterna magna injection model. The topics of discussing anesthesia, advice on peri- and postanesthetic handling of animals, assessing the severity of suffering the animals undergo during the procedure according to EU directive 2010/63/EU and weighing the use of these in vivo models for experimental research ethically are also presented. In conclusion, rodent models of subarachnoid hemorrhage display pathophysiological characteristics, including changes of cerebral perfusion similar to the clinical situation, rendering the models suited to study the sequelae of the bleeding. A current problem is low standardization of the models, wherefore reporting according to the ARRIVE guidelines is highly recommended. Graphical Abstract Animal welfare aspects of rodent models of subarachnoid hemorrhage. Rodent models for investigation of cerebral perfusion after subarachnoid hemorrhage are compared regarding surgeries and model characteristics, and 3R measures are suggested. Anesthesia is discussed, and advice given on peri- and postanesthetic handling. Severity of suffering according to 2010/63/EU is assessed and use of these in vivo models weighed ethically.
... Cerebral blood ow (CBF) and intracranial pressure (ICP) were monitored as previously described 22,23,24 using a laser Doppler probe (Perimed, Järfälla, Sweden) and microcatheter transducer (Millar, Houston, USA), respectively. SAH was induced using the endovascular perforation model as described previously. ...
Preprint
Full-text available
Background and Purpose Impaired cerebral circulation, induced by blood vessel constrictions and microthrombi, leads to delayed cerebral ischemia after subarachnoid hemorrhage (SAH). 12/15-Lipooxygenase (12/15-LOX) overexpression has been implicated in worsening early brain injury outcomes following SAH. However, it is unknown if 12/15-LOX is important in delayed pathophysiological events after SAH. Since 12/15-LOX produces metabolites that induce inflammation and vasoconstriction, we hypothesized that 12/15-LOX leads to microvessel constriction and microthrombi formation after SAH, and thus 12/15-LOX is an important target to prevent delayed cerebral ischemia. Methods SAH was induced in C57BL/6 and 12/15-LOX−/− mice of both sexes by endovascular perforation. Expression of 12/15-LOX was assessed in brain tissue slices and in vitro. C57BL/6 mice were administered either ML351 (12/15-LOX inhibitor) or vehicle. Mice were evaluated for daily neuroscore and euthanized on day five to assess cerebral 12/15-LOX expression, vessel constrictions, platelet activation, microthrombi, neurodegeneration, infarction, cortical perfusion, and for development of delayed deficits. Finally, the effect of 12/15-LOX inhibition on platelet activation was assessed in SAH patient samples using a platelet spreading assay. Results In SAH mice, 12/15-LOX was upregulated in brain vascular cells and there was an increase in 12-S-HETE. Inhibition of 12/15-LOX improved brain perfusion on days 4–5 and attenuated delayed pathophysiological events, including microvessel constrictions, microthrombi, neuronal degeneration, and infarction. Additionally, 12/15-LOX inhibition reduced platelet activation in human and mouse blood samples. Conclusions Cerebrovascular 12/15-LOX overexpression plays a major role in brain dysfunction after SAH by triggering microvessel constrictions and microthrombi formation, which reduces brain perfusion. Inhibiting 12/15-LOX may be a therapeutic target to improve outcomes after SAH.
... Intracranial pressure monitoring is helpful in guiding clinicians to accurately assess the degree of craniocerebral injury and early detection of intracranial space, which is conducive to reducing secondary brain injury and improving the prognosis of patients. The placement of intracranial intraventricular pressure probe is conducive to dynamic monitoring of intracranial pressure changes, effectively avoiding increased blood viscosity, renal function injury, and electrolyte disturbance caused by blind application of dehydrating agents, shortening the placement time of drainage tube and reducing the risk of intracranial infection [20]. Intracranial pressure monitoring can determine the drainage flow according to intracranial pressure, which can effectively avoid rebleeding caused by intracranial pressure fluctuation [21]. ...
... A further advancement was applied to ensure perforation of the wall at the bifurcation of the anterior and middle cerebral arteries. An intracranial pressure monitor was applied to confirm the success of the perforation 23 . Subsequently, the suture was retracted, and the ICA was re-perfused. ...
Article
Full-text available
The Circle of Willis perforation (cWp) mouse model is a key tool in subarachnoid hemorrhage (SAH) research; however, inconsistent bleeding volumes can challenge experimental reliability. To address this issue, we introduced the ROB Scoring System, a novel protocol integrating Rotarod Tests (RT), Open-field Tests (OT) video analysis, and daily Body Weight Loss (BWL) monitoring to precisely categorize SAH severity. Forty C57BL/6 mice underwent cWp SAH induction, categorized by ROB into severity subgroups (severe, moderate, mild). Validation compared ROB trends in subgroups, and ROB outcomes with autopsy results on postoperative days three and seven for acute and sub-acute evaluations. Mortality rates were analyzed via the survival log-rank test, revealing a significant difference among SAH subgroups (P < 0.05). Strong correlations between ROB grades and autopsy findings underscored its precision. Notably, the severe group exhibited 100% mortality within 4 days post SAH onset. Single parameters (RT, OT, BWL) were insufficient for distinguishing SAH severity levels. The ROB score represents a significant advancement, offering an objective method for precise categorization and addressing inherent bleeding variations in the cWp SAH model. This standardized protocol enhances the reliability and effectiveness of the SAH translational research, providing a valuable tool for future investigations into this critical area.
... A further advancement was applied to ensure perforation of the wall at the bifurcation of the anterior and middle cerebral arteries. An intracranial pressure monitor was applied to con rm the success of the perforation [12]. Subsequently, the suture was retracted, and the ICA was reperfused. ...
Preprint
Full-text available
The Circle of Willis perforation (cWp) mouse model is a key tool in subarachnoid hemorrhage (SAH) research; however, inconsistent bleeding volumes can challenge experimental reliability. To address this issue, we introduced the ROB Scoring System, a novel protocol integrating Rotarod Tests (RT), Open-field Tests (OT) video analysis, and daily Body Weight Loss (BWL) monitoring to precisely categorize SAH severity. Forty C57BL/6 mice underwent cWp induction, categorized by ROB into severity subgroups (severe, moderate, mild). Validation compared ROB outcomes with autopsy results on postoperative days 3 and 7 for acute and sub-acute evaluations. Mortality rates were analyzed via the survival log-rank test, revealing a significant difference among severity groups (P < 0.0001). Strong correlations between ROB grades and autopsy findings underscored its precision. Notably, the severe group exhibited 100% mortality within 4 days post-SAH onset. Individual parameters (RT, OT, BWL) were insufficient for distinguishing SAH severity levels. The ROB Scoring System represents a significant advancement, offering precise categorization and addressing inherent bleeding variations in the cWp mouse model. This standardized protocol enhances the reliability and effectiveness of SAH research, providing a valuable tool for future investigations into this critical.
... Induction of SAH was strictly carried out as Feiler instructed before [16]. Briefly, anesthesia was induced by a rodent ventilator supplementing air with 3% isoflurane in 70/30% medical air/oxygen. ...
Article
Full-text available
Subarachnoid hemorrhage refers to an uncommon but severe subtype of stroke leading to high mortality and disability rates. Electroacupuncture, a traditional Chinese medical therapy combined with modern technology, shows evident curative effects on cerebral vascular diseases. This study attempts to investigate the possible treatment effects and mechanisms of EA on early brain injury after SAH. Data were gathered among sham group, SAH-induced group, and EA-treated group of male SD rats, concerning mortality rates, weight loss, rotarod latencies, cerebral blood flow, cell apoptosis, pro-inflammatory cytokines releasing, apoptotic protein level, microglia activation and related signal pathway. All results were collected 24–72 h after SAH induction. EA treatment demonstrated significant improvement on motor function 24 h after SAH without significant changes in mortality rate, weight loss, and cerebral blood flow. Another important finding was that EA regulated Bax and Bcl-2 imbalance and reduced cleaved casepase-3 caused by SAH. Additionally, levels of TNF-α, IL-1β, IL-6 were suppressed. The neuron apoptosis was suppressed by EA. The M1 polarization of activated microglia decreased while M2 polarized phenotype increased after EA treatment. Furthermore, pSTAT3-NOX2 signal axis, the M1 phenotype related activation pathway, was depressed after EA treatment. These findings suggested that EA improved motor deficits and ameliorated early brain injury after SAH probably via decreasing neuron apoptosis and anti-inflammation, which may involve modulation of microglia polarization. Taken together, EA may be a potential therapy for SAH treatment.
Preprint
Full-text available
Background The occurrence of delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage is a main determinant for functional outcome. Despite two decades of animal studies investigating novel treatment strategies, the standard therapy for delayed cerebral ischemia has not changed. This translational gap raises the question to what extent experimental mouse models of subarachnoid hemorrhage can accurately mimic human delayed cerebral ischemia. The objective of this systematic review was to determine whether there are difference in the occurrence and timing of delayed cerebral ischemia between the various experimental mouse models of aneurysmal subarachnoid hemorrhage. Our aim was to identify which mouse model most closely mimics the human pathophysiology following aneurysmal subarachnoid hemorrhage. Methods This study was funded by ZonMw (project number 114024130). The review was preregistered at PROSPERO (protocol ID: CRD42020115578). A comprehensive search was performed in Medline via the PubMed interface and in EMBASE via the Ovid interface up to 2nd November 2018The following exclusion criteria were used in both the title and abstract and full text phase: 1) not an original, full-length research paper, 2) no English language version available, 3) published before 1999, 4) not an animal study, 5) not a mouse study, 6) use of transgenic mice, 7) no subarachnoid hemorrhage induction, 8) no delayed cerebral ischemia measured, 9) reported an observation time less than six hours, 10) cross-over study, or any study design without a control group. Data was extracted by one reviewer. The SYRCLE risk of bias tool was used in duplo by two independent reviewers to assess risks of bias in the included studies, with discrepancies being resolved through discussion. A narrative synthesis of the evidence was performed. Results The literature search retrieved a total of 1461 papers, of which 71 publications met the inclusion criteria. Most studies were assessed at an unclear risk for most types of bias. Mice models were highly standardized: the C57Bl/6 strain was used in 53 studies (74.6%), only male animals were used in 55 studies (77.5%). To model a subarachnoid hemorrhage, perforation of the anterior cerebral artery / internal carotid artery with a suture was performed in 43 studies (60.6%), while direct injection of blood was performed in 24 studies (33.8%). The presence of delayed cerebral ischemia was established through neurological outcomes (44 studies, 62.0%), ex-vivo histology (39 studies, 54.9%) or in-vivo imaging (10 studies, 14.1%) assessment. Incidence of delayed cerebral ischemia was similarly high for all outcome measures (between 85-87%) and the timing of delayed cerebral ischemia occurrence was similar, despite differences in subarachnoid hemorrhage induction method. Conclusion Our results show that established perforation and injection-models for experimental subarachnoid hemorrhage are highly standardized. Yet, there is a high inconsistency in definitions of delayed cerebral ischemia in experimental mouse models. Although perforation model results in a higher rate of delayed cerebral ischemia, there is no effect on the time of occurrence after ictus. It is unclear to what extent the delayed cerebral ischemia demonstrated in these animal models is comparable to the clinical situation.
Article
Full-text available
A common benchmark in the brain tissue mechanics literature is that the properties of acute brain slices should be measured within 8 h of the experimental animal being sacrificed. The core assumption is that—since there is no substantial protein degradation during this time—there will be no change to elastic modulus. This assumption overlooks the possibility of other effects (such as osmotic swelling) that may influence the mechanical properties of the tissue. To achieve consistent and accurate analysis of brain mechanics, it is important to account for or mitigate these effects. Using atomic force microscopy (AFM), tissue hydration and volume measurements, we find that acute brain slices in oxygenated artificial cerebrospinal fluid (aCSF) with a standard osmolarity of 300 mOsm/l experience rapid swelling, softening, and increases in hydration within the first 2 hours after slicing. Reductions in elastic modulus can be partly mitigated by addition of chondroitinase ABC enzyme (CHABC). Increasing aCSF osmolarity to 400 mOsm/l does not prevent softening but may hasten equilibration of samples to a point where measurements of relative elastic modulus are consistent across experiments.
Article
This chapter discusses the clinical aspects of subarachnoid hemorrhage (SAH). The clinical hallmark of SAH is a history of sudden, unusually severe headache. A period of unresponsiveness longer than one hour occurs in almost half the patients, and focal signs develop at the same time of the headache or soon afterward in one-third of patients. In patients in whom headache is the only symptom, it may be difficult to recognize the seriousness of the underlying condition. Another problem is that in the group of patients whose headache came on within a split second, innocuous forms of headache outnumber subarachnoid hemorrhage by 10 to 1. Vomiting occurs in 70% of patients with aneurysmal rupture but also in 43% of patients with innocuous thunderclap headache, and preceding bouts of similar headaches are recalled in 20% of patients with aneurysmal rupture and 15% of patients with innocuous thunderclap headache. The chapter discusses epidemiological aspects for aneurismal subarachnoid hemorrhage, causes of subarachnoid hemorrhage, nonaneurysmal perimesencephahc hemorrhage, and others. The discussion on practical management includes early rebleeding, prevention of rebleeding, prevention of secondary cerebral ischemia, intracerebral hematoma, and others.
Article
1. Subarachnoid haemorrhage (SAH) is a unique disorder and a major clinical problem that most commonly occurs when an aneurysm in a cerebral artery ruptures, leading to bleeding and clot formation. Subarachnoid haemorrhage results in death or severe disability of 50‐70% of victims and is the cause of up to 10% of all strokes. Delayed cerebral vasospasm, which is the most critical clinical complication that occurs after SAH, seems to be associated with both impaired dilator and increased constrictor mechanisms in cerebral arteries. Mechanisms contributing to development of vasospasm and abnormal reactivity of cerebral arteries after SAH have been intensively investigated in recent years. In the present review we focus on recent advances in our knowledge of the roles of nitric oxide (NO) and cGMP, endothelin (ET), protein kinase C (PKC) and potassium channels as they relate to SAH. 2. Nitric oxide is produced by the endothelium and is an important regulator of cerebral vascular tone by tonically maintaining the vasculature in a dilated state. Endothelial injury after SAH may interfere with NO production and lead to vasoconstriction and impaired responses to endothelium‐dependent vasodilators. Inactivation of NO by oxyhemoglobin or superoxide from erythrocytes may also occur in the subarachnoid space after SAH. 3. Nitric oxide stimulates activity of soluble guanylate cyclase in vascular muscle, leading to intracellular generation of cGMP and relaxation. Subarachnoid haemorrhage appears to cause impaired activity of soluble guanylate cyclase, resulting in reduced basal levels of cGMP in cerebral vessels and often decreased responsiveness of cerebral arteries to NO. 4. Endothelin is a potent, long‐lasting vasoconstrictor that may contribute to the spasm of cerebral arteries after SAH. Endothelin is present in increased levels in the cerebrospinal fluid of SAH patients. Pharmacological inhibition of ET synthesis or of ET receptors has been reported to attenuate cerebral vasospasm. Production of and vasoconstriction by ET may be due, in part, to the decreased activity of NO and formation ofcGMP. 5. Protein kinase C is an important enzyme involved in the contraction of vascular muscle in response to several agonists, including ET. Activity of PKC appears to be increased in cerebral arteries after SAH, indicating that PKC may be critical in the development of cerebral vasospasm. Recent evidence suggests that PKC activation may occur in cerebral arteries after SAH as a result of decreased negative feedback influence of NO/cGMP 6. Cerebral arteries are depolarized after SAH, possibly due to decreased activity of potassium channels in vascular muscle. Decreased basal activation of potassium channels may be due to several mechanisms, including impaired activity of NO (and/or cGMP) or increased activity of PKC. Vasodilator drugs that produce hyperpolarization, such as potassium channel openers, appear to be unusually effective in cerebral arteries after SAH. 7. Thus, endothelial damage and reduced activity of NO may contribute to cerebral vascular dysfunction after SAH. Potassium channels may represent an important therapeutic target for the treatment of cerebral vasospasm after SAH.
Article
The acute neurological deficit present immediately after subarachnoid hemorrhage (SAH) correlates with overall outcome. Only limited data are available to quantify changes in cerebral perfusion in this acute phase, and this study sought to characterize those changes within the first 12 h post-SAH. Xenon contrast-enhanced CT scanning was performed in 17 patients (Hunt and Hess grade [HH] 1-3, n = 9; HH 4-5, n = 8) within 12 h after SAH. Cerebral blood flow (CBF) was analyzed in all cortical and central vascular regions of interest (ROI), as well as infratentorial ROI. Hemodynamic stress distribution (central/cortical ROI) was also calculated. Asymptomatic patients without perfusion deficits served as controls (n = 5), and Glasgow Outcome Scale score (GOS) was determined 3 months after the event. Intracranial pressure (ICP) and cerebral perfusion pressure (CPP) were within normal limits in all patients. CBF was significantly reduced in all patients with SAH (34 mL/100 g x min) compared to controls (67 mL/100 g x min; p < 0.001). Patients in better clinical condition (HH 1-3) presented with significantly less reduction of CBF (41 mL/100 g x min) compared to patients with more severe hemorrhage (HH 4-5: 24 mL/100 g x min; p < 0.001), and had better outcomes. Changes in perfusion were more pronounced in supratentorial than in infratentorial ROI. Hemodynamic stress distribution was most pronounced in patients with higher HH grade (p < 0.05). The first 12 h after SAH are characterized by persistent, severe reduction of CBF, which in turn correlates with HH grade, but is independent of ICP or CPP. Acute peripheral vasospasm of the microvasculature, not detectable by conventional angiography, may account for this early phase of prolonged hypoperfusion.
Article
Mouse models take advantage of genetic manipulations that can be achieved in this species. There are currently two accepted mouse models of subarachnoid hemorrhage (SAH) and cerebral vasospasm (CVs). Both are technically demanding and labor intensive. In this study, we report a reproducible and technically feasible method to induce SAH, and subsequently CVs, in mice. We tested this model in multiple strains of mice that are commonly used for genetic manipulation. SAH was induced in C57BL/6NCr, FVB, 129S1, BalbC and SJL mice, weighing 28-32 g, by an intracisternal vessel transection technique. Animals were perfused with India ink at 24h postprocedure and vessel diameters were quantified. Brain slices were obtained for hematoxylin-eosin staining (H&E) to look for vascular changes consistent with CVs. There was no mortality during or after the procedure. Four of the five mouse strains showed significant CVs at 24 h postprocedure characterized by decreased vessel diameter of the middle cerebral artery close to the Circle of Willis. Histologically, the vessel wall displayed significant corrugation and thickening, consistent with CVs. A novel mouse model to induce SAH is described and tested in several mouse strains. Four of the five strains used in this study developed CVs after the induction of SAH. The procedure is brief, straightforward, reproducible with low mortality, and applicable to commonly used background strains for genetically engineered mice.
Article
The role of interleukin (IL)-1beta remains unknown in early brain injury (EBI) after subarachnoid hemorrhage (SAH), although IL-1beta has been repeatedly reported to increase in the brain and cerebrospinal fluid. The aim of this study is to examine the effects of IL-1beta inactivation on EBI after SAH in mice. The endovascular perforation model of SAH was produced and 112 mice were assigned to sham, SAH+ vehicle, and SAH+ N-Ac-Tyr-Val-Ala-Asp-chloromethyl ketone (Ac-YVAD-CMK, 6 and 10 mg/kg) groups. Ac-YVAD-CMK, a selective inhibitor of IL-1beta converting enzyme, or vehicle was administered intraperitoneally 1 hour post-SAH. EBI was assessed in terms of mortality within 24 hours, neurological scores, brain water content at 24 and 72 hours, Evans blue dye extravasation and Western blot for IL-1beta, c-Jun N-Terminal kinase (JNK), matrix metalloproteinase (MMP)-9, and zonula occludens (ZO)-1 at 24 hours after SAH. High-dose (10 mg/kg) but not low-dose (6 mg/kg) treatment group significantly improved neurological scores, mortality, brain water content, and Evans blue dye extravasation compared with the vehicle group. Although both dosages of Ac-YVAD-CMK attenuated the mature IL-1beta induction, only high-dose treatment group significantly inhibited the phosphorylation of JNK, MMP-9 induction, and ZO-1 degradation. IL-1beta activation may play an important role in the pathogenesis of EBI after SAH. The neurovascular protection of Ac-YVAD-CMK may be provided by the inhibition of JNK-mediated MMP-9 induction and the consequent preservation of tight junction protein ZO-1.
Article
Pathophysiology after subarachnoid hemorrhage (SAH) caused by aneurysmal rupture has not been well examined. The purpose of this study was to observe platelet-leukocyte-endothelial cell interactions as indexes of inflammatory and prothrombogenic responses in the acute phase of SAH, using an in vivo cranial window method. Subarachnoid hemorrhage was induced in C57Bl/6J mice by using the endovascular perforation method. Intravital microscopy was used to monitor the rolling and adhesion of platelets and leukocytes that were labeled with different fluorochromes. Regional cerebral blood flow was measured with laser Doppler flowmetry. The platelet-leukocyte-endothelial cell interactions were observed 30 minutes, 2 hours, and 8 hours after SAH. The effect of P-selectin antibody and apocynin, an inhibitor of nicotinamide adenine dinucleotide phosphate oxidase, on these responses was examined at 2 hours after SAH, and compared with a different SAH model in which autologous blood was injected into the foramen magna. SAH was accompanied by a 60% decrease in regional cerebral blood flow, whereas no changes in regional cerebral blood flow were observed on the contralateral side. SAH elicited time- and size-dependent increases in rolling and adherent platelets and leukocytes in cerebral venules. All of these interactions were attenuated by treatment with a P-selectin antibody or apocynin. There was no significant blood cell recruitment observed in the blood-injected SAH model. SAH at the skull base induced P-selectin- and oxygen radical-mediated platelet-leukocyte-endothelial cell interactions in venules at the cerebral surface. These early inflammatory and prothrombogenic responses may cause a whole-brain injury immediately after SAH.
Article
Background and purpose: Despite recent advances in the treatment of patients after subarachnoid hemorrhage, morbidity and mortality rates have failed to improve significantly. Although this was often blamed on vasospasm, is it time to consider alternative etiologies? Summary of Review- Early brain injury (EBI) is a recently described term that describes the immediate injury to the brain after subarachnoid hemorrhage. A number of pathways have been recognized as having a role in the etiology of EBI. This review provides a brief synopsis of EBI and its implications for the future. Conclusions: EBI may be responsible for the detrimental effects seen in patients after subarachnoid hemorrhage. Additional studies are needed to determine the pathophysiology of EBI and to explore potential therapeutic options.
Article
A method for induction of subarachnoid hemorrhage (SAH) in a rat model is described. Resolution of the hemorrhage was documented photographically and microscopically at intervals from 1 hr to 8 days. Photographs indicated that most of the hemorrhage was resorbed within 3 days, an observation confirmed microscopically by the amount of red blood cells in the subarachnoid space. Significant cerebral vasospasm was documented within the first 2 days after the induction of hemorrhage with the basilar artery returning to baseline values at an average of 3 days followed by moderate dilatation at 5 to 8 days. The suitability of the rat as an animal model for further investigation of subarachnoid hemorrhage is discussed.
Article
In 29 anaesthetized baboons avulsion of a small intracranial artery was used to produce a subarachnoid haemorrhage, in a closed-skull situation. Intracranial pressure was measured by extradural transducers, and arterial pressure was also measured continuously, with periodic measurements of cerebral blood flow. After haemorrhage there was an immediate fall in cerebral perfusion pressure in nearly all cases, reaching zero in 9 animals. In 18 there was a significant pressor response in the systemic circulation, but perfusion pressure usually remained low in spite of this response. Perfusion pressure recovered after a few minutes in most cases. In the 19 cases where intracranial pressure was measured on both sides, differences occurred in 11, with the higher pressure always on the same side as the haemorrhage. The difference was evident very soon after haemorrhage in 9 cases, and lasted over half an hour in 5 of them. The mechanism of arrest of bleeding was, in most of this series, not that of a zero perfusion pressure. Explanations for this and for the occurrence of differential pressures are discussed.
Article
A subarachnoid haemorrhage (SAH) in the rat was produced by the injection of blood via a previously implanted catheter connected to the cisterna magna. Repeated angiographical examinations of the vertebro-basilar arteries revealed a biphasic vasospasm with a maximal acute spasm at ten minutes and a maximal late spasm at two days after cisternal blood injection. Fluorescence microscopical examination of the major cerebral arteries at day two after the SAH revealed a reduction in the fluorescence intensity and in the number of histochemically visible sympathetic nerve terminals.