You are on page 1of 14

Neuropharmacology 181 (2020) 108357

Contents lists available at ScienceDirect

Neuropharmacology
journal homepage: www.elsevier.com/locate/neuropharm

Lithium enhances post-stroke blood-brain barrier integrity, activates the


MAPK/ERK1/2 pathway and alters immune cell migration in mice
Matteo Haupt a, Bozena Zechmeister a, Bert Bosche b, c, d, Simone Lieschke a, Xuan Zheng a,
Lin Zhang a, Vivek Venkataramani e, Fengyan Jin f, Katharina Hein a, Martin S. Weber a, g,
Dirk M. Hermann c, Mathias Bähr a, Thorsten R. Doeppner a, *
a
University Medical Center Goettingen, Department of Neurology, Goettingen, Germany
b
Department of Neurocritical Care, First Stage Rehabilitation and Weaning, Reichshof-Eckenhagen, Germany
c
University of Duisburg-Essen Medical School, Department of Neurology, Essen, Germany
d
Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
e
University Medical Center Goettingen, Institute of Pathology, Goettingen, Germany
f
The First Hospital of Jilin University, Department of Hematology, Cancer Center, Changchun, Jilin, China
g
University Medical Center Goettingen, Institute of Neuropathology, Goettingen, Germany

A R T I C L E I N F O A B S T R A C T

Keywords: Lithium induces neuroprotection against cerebral ischemia, although the underlying mechanisms remain elusive.
Blood-brain barrier We have previously suggested a role for lithium in calcium regulation and (extra)cerebral vessel relaxation under
Neurovascular unit non-ischemic conditions. Herein, we aimed to investigate whether or not lithium contributes to post-stroke
Cerebral ischemia
stabilization of the blood-brain barrier (BBB) in mice. Using an oxygen-glucose-deprivation (OGD) model, we
Immune response
first analyzed the impact of lithium treatment on endothelial cells (EC) in vitro. Indeed, such treatment of EC
Lithium
Neuroprotection exposed to OGD resulted in increased cell survival as well as in enhanced expression of tight junction proteins
and P-glycoprotein. Additional in vivo studies demonstrated an increased stabilization of the BBB upon lithium
treatment in stroke mice, as shown by a reduced Evans blue extravasation and an elevation of tight junction
protein expression. Furthermore, stabilization of the BBB as a consequence of lithium treatment was associated
with an inhibition of matrix metalloproteinase-9 activity, independent of calveolin-1 regulation. In line with this,
flow cytometry analysis revealed that lithium treatment led to a decreased neutrophil invasion and an increased
T cell extravasation from the blood compartment towards the brain parenchyma. We finally identified the pro-
survival MAPK/ERK1/2 pathway as the key regulator of the impact of lithium on the BBB. In conclusion, we
demonstrate for the first time that lithium is able to enhance post-stroke BBB integrity. Importantly, our work
delivers novel insights into the exact mechanism of lithium-induced acute neuroprotection, providing critical
information for future clinical trials involving lithium treatment in stroke patients.

1. Introduction outcome in distinct neurobehavioral tests over a period of 56 days after


cerebral ischemia (Doeppner et al., 2017). These preclinical studies gave
Lithium has been successfully used for the treatment of bipolar dis­ rise to a clinical trial showing an improved motor recovery after lithium
orders for decades (Manji and Lenox, 1998). Recent evidence in rodents, treatment in patients with cortical ischemic stroke (Abdollahi et al.,
moreover, suggests that lithium has protective effects on neurons after 2014). However, none of these studies explained the mechanisms by
cerebral ischemia. In detail, lithium treatment yielded a reduction of which lithium induces its acute neuroprotective effects.
infarct volume, inhibition of apoptotic cell death and suppression of Lithium is a major modulator of the glycogen synthase kinase 3 beta
neuroinflammation (Chuang et al., 2011; Doeppner et al., 2017; Fan (GSK3-β), which in turn can regulate a large number of downstream
et al., 2015; Ren et al., 2003; Xu et al., 2003). Furthermore, lithium targets (Li X, Bijur G, 2007). Targets of GSK3-β include NFκB, Bcl-2, and
treatment enhanced neurological recovery, as observed by an improved members of the Wnt signaling pathway, all of which play critical roles in

* Corresponding author. Department of Neurology, University Medical Center Goettingen, Robert-Koch-Str. 40, 37075, Goettingen, Germany.
E-mail address: thorsten.doeppner@med.uni-goettingen.de (T.R. Doeppner).

https://doi.org/10.1016/j.neuropharm.2020.108357
Received 3 March 2020; Received in revised form 10 October 2020; Accepted 12 October 2020
Available online 13 October 2020
0028-3908/© 2020 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
M. Haupt et al. Neuropharmacology 181 (2020) 108357

the regulation of signal transduction and cell survival (Maurer et al., Collection, Manassas, Virginia, USA) were seeded in TC-plates (Sarstedt,
2014). Interestingly, we have previously shown that lithium-regulated Nuembrecht, Germany) and cultured under confluent conditions at a
signaling pathways involved in the pathophysiology of cerebral density of 6 × 104 cells/cm2. Cells were incubated with 10% fetal bovine
ischemia are pleiotropic and not solely dependent on GSK3-β (Doeppner serum-containing medium (Dulbecco’s Modified Eagle Medium/
et al., 2017). We found that post-stroke lithium treatment also modu­ Nutrient Mixture F-12; Thermo Fisher Scientific, Waltham, Massachu­
lated miR-124 expression, RE1-silencing transcription factor abundance setts, USA) which was enriched with lithium chloride (Sigma-Aldrich,
and protein deubiquitination in mice (Doeppner et al., 2017). Although St. Louis, Missouri, USA; subsequently addressed as lithium only) at a
these effects contributed to a better neurological recovery in these mice, final concentration of 1.25 mM. Controls were incubated in medium
the precise downstream targets of lithium during acute stage of the without lithium. The cells were placed for 14 h under OGD 24 h after
disease remained elusive at the time. seeding. For OGD, the cells were incubated in deoxygenated glucose-free
Among the complex pathophysiology of cerebral ischemia, the salt solution (BSS0) containing 10 x BSS (1.16 M NaCl, 54 mM KCl, 8 mM
opening of the blood-brain barrier (BBB) during the first hours following MgSO4 and 10 mM NaH2PO4H2O), 1 M NaHCO3, 1 M HEPES, 30 mM
stroke induction is a critical mechanism for cell injury development glycine and 1 M CaCl2. For lithium-treated cells, 1.25 mM lithium was
(Dirnagl et al., 1999). In addition, the BBB breakdown becomes clini­ added to the BSS0 solution. OGD atmosphere in the hypoxia chamber
cally more relevant for patients during the subacute phase of ischemic contained 0.2% O2, 5% CO2, and 65% humidity and were confirmed by
stroke, when vasogenic edema formation and subsequent the built-in sensor of the chamber (Toepffer Lab Systems, Goeppingen,
space-occupying brain swelling predominantly occurs (Bosche et al., Germany). After removing the BSS0 solution, the cells were incubated in
2003; Wijdicks et al., 2014; Liebeskind et al., 2019). In the endothelial the aforementioned cell culture medium with or without 1.25 mM
cells (EC) of the BBB, energy deficits during hypoxia lead to an increase lithium for 24 h in the incubator at 37 ◦ C. For Western blot analysis, the
of cytosolic Ca2+, contributing to irreversible cell injury (Schäfer et al., cells were lysed by shock freezing at − 80 ◦ C followed by ultrasound
2001). The latter triggers the dysregulation of the endothelial barrier treatment. After 10 min of centrifugation at 12,000 rpm, the supernatant
function and leads to hyperpermeability (Noll et al., 1995). In vitro ex­ was collected and the protein concentration was photometrically
periments suggested that lithium prevents this early Ca2+ overload by determined (Pierce™ BCA Protein Assay Kit; Thermo Fisher Scientific).
inhibiting the inositol-3-phosphate-sensitive Ca2+ release from the The samples were aliquoted and stored at − 80 ◦ C until the Western blot
endoplasmic reticulum of EC (Bosche et al., 2013). Further in vitro ex­ analysis were performed.
periments under non-hypoxic conditions suggest a complex
lithium-induced stabilization of the endothelium through the decrease 2.2. Measurement of cell viability
in myosin light chain phosphorylation in vessel grafts from de-nerved
murine aortas and porcine middle cerebral arteries (Bosche et al., A commercial live dead assay kit (ab115347; Abcam, Cambridge,
2016b, 2016a). In addition, Li and colleagues observed a protection of UK) was used to analyze cell viability according to the manufacturer’s
lithium-treated EC against oxygen-glucose-deprivation (OGD) in vitro protocol. Briefly, cells were incubated after their respective treatments
and a preservation of BBB function in vivo following lithium pretreat­ with the fluorescence dyes for 10 min at room temperature. Subse­
ment in an ischemia–reperfusion model in rats (Li et al., 2014). quently, cells labeled either green (live) or red (dead) were viewed and
Apart from the intracellular processes in EC, activation of matrix counted under the Axioplan 2 fluorescence microscope (Carl Zeiss, Jena,
metalloproteinases (MMP) play a key role in the pathophysiology of Germany). For quantification, we counted the number of live/dead cells
cerebral ischemia (Bosche et al., 2006; Gasche et al., 1999; Kurzepa within two sectors per well (at 10x magnification). The percentage of
et al., 2014; Yang et al., 2007). MMPs such as MMP-9 lead to the live cells in relation to the total number of cells were calculated and
degradation of extracellular endothelial tight junction proteins and presented in the figures.
hence to a more permeable BBB. MMPs are present as pro-forms, which MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bro­
become activated through cleavage after stroke (Medsker et al., 2016). mide; Thiazolyl blue) reagent (Sigma-Aldrich) was used in a colori­
Opening of the BBB does not only directly impair neural cell survival but metric assay to assess cell metabolism. The MTT assay was performed on
also facilitates peripheral immune cell migration into the central ner­ cells 24 h after OGD or without OGD. Therefore, the MTT was directly
vous system (Chamorro et al., 2012; Dirnagl et al., 1999). Resident added to culture wells and incubated for 3 h at 37 ◦ C. Following removal
phagocytes (microglia) activation, chemotaxis and expression of cell of the medium, the dye was solubilized for 5 min in DMSO (dimethyl
adhesion molecules play a pivotal role in this context (Xiong et al., 2016; sulfoxide; Merck, Darmstadt, Germany) under constant shaking. The
Yanamadala and Friedlander, 2010). The importance of these mecha­ absorbance was photometrically measured at 595 nm using a 96-well
nisms is illustrated by the fact that MMP-9 inhibition leads to neuro­ absorbance microplate reader (Tecan, Maennedorf, Switzerland). To
protection under experimental stroke conditions (Chaturvedi and normalize the data, metabolic activity of mouse brain EC without OGD
Kaczmarek, 2014). Indeed, lithium treatment has been shown to reduce was measured and set as 100%.
MMP-9 expression, subsequently leading to enhanced BBB integrity in
mice 3 days post-induction of traumatic brain injury (Yu et al., 2011). 2.3. Induction of focal cerebral ischemia
Nevertheless, the role of lithium in the context of BBB disruption
after cerebral ischemia and all types of stroke remains largely elusive but All animals were kept under circadian rhythm and had free access to
is highly relevant especially for further studies in humans (Bosche and food and water. Experiments were performed according to local au­
Macdonald, 2015; Bosche et al., 2020). In our study, we investigated thorities (LAVES, Lower Saxony, Germany) following the ARRIVE
whether or not lithium elicits its neuroprotective effects through guidelines.
impacting the BBB in adult stroke mice. More specifically, we focus on The induction of transient focal cerebral ischemia was obtained
immune cell migration, tight junction protein expression, cell survival using the middle cerebral artery (MCA) occlusion model as previously
signaling cascades, and the activation of MMP-9 upon in vitro OGD of described (Weise et al., 2006). In short, under anesthesia with 2% iso­
cultured EC as well as in stroke mice. flurane and 0.8 l/min O2, the right common carotid artery was isolated
and a 6-0 nylon monofilament (Doccol Corporation, Sharon, Massa­
2. Materials and methods chusetts, USA) inserted. The filament was gently pushed forward to­
wards the MCA where it was placed for 45 min. After filament removal,
2.1. Cell culture, treatment and lysis the wounds were carefully sutured. The occlusion and the reperfusion
were monitored under constant laser doppler flow. For pain treatment,
Mouse brain EC (bEnd.3, CRL-2299™; American Type Culture 0.1 mg/kg buprenorphine was injected subcutaneously (s.c.) before

2
M. Haupt et al. Neuropharmacology 181 (2020) 108357

surgery and 4 mg/kg carprofen after surgery. This was followed by daily performed.
s.c. injections of 0.1 mg/kg buprenorphine in the morning and 4 mg/kg
carprofen in evening. For the sham group, animals underwent the very 2.7. Western blot analysis
same procedure except for insertion of the nylon filament.
For Western blot analysis, reducing sample buffer (Carl Roth,
2.4. Animal groups and drug treatment Karlsruhe, Germany) was added, and the tissue or cell culture samples
were heated for 5 min at 95 ◦ C. Thereafter, 35 μg of protein were
7-week-old male C57BL6 mice (Charles River, Wilmington, Massa­ separated using SDS-PAGE and transferred onto nitrocellulose mem­
chusetts, USA) were randomly divided in groups with survival times of branes (Bio-Rad, Hercules, California, USA). Following transfer, the
24 h or 72 h after MCA occlusion. Inclusion criteria include the detection membranes were blocked using 5% milk in PBS (skim milk powder;
of stroke lesions within the striatum, whereas exclusion criteria include Sigma-Aldrich). The membranes were then incubated with the primary
huge cortical infarcts, development of seizures, and spinning of animals antibodies occludin (1:1000; ab167161; Abcam), ZO-1 (1:1000;
when picked up by the tail. The mice received phosphate-buffered saline ab96587; Abcam), caveolin-1 (1:1000; ab2910; Abcam), c-Raf (1:1000;
(PBS) as solvent control, Lithium was dissolved in PBS whereas the #9422; Cell Signaling Technology), p-c-Raf (1:1000; #9421; Cell
MEK1/2 inhibitor U0126 (Cell Signaling Technology, Danvers, Massa­ Signaling Technology), MEK1/2 (1:1000; #9122; Cell Signaling Tech­
chusetts, USA) was dissolved in DMSO depending on their group allo­ nology), p-MEK1/2 (1:1000; ab194754; Abcam), ERK1/2 (1:2000; sc-
cation. Mice with 24 h survival were further divided into the following 93-G; Santa Cruz Biotechnology, Dallas, Texas, USA) p-ERK1/2
groups: sham, control, lithium, lithium + U0126 and U0126. For sham, (1:1000; #9101; Cell Signaling Technology), Bcl-2 (1:1000; sc-7382;
control and U0126 groups, PBS was given at the beginning of reperfu­ Santa Cruz Biotechnology), ICAM1 (1:1000; sc-8439; Santa Cruz
sion and at 6 h after MCA occlusion. For lithium and lithium + U0126 Biotechnology), VCAM-1 (1:1000; ab134047; Abcam), P-gp (1:1000;
groups, lithium was given at the beginning of reperfusion and at 6 h after ab170904; Abcam) and horseradish peroxidase coupled secondary anti-
MCA occlusion. The lithium dosage for the first injection was 1 mmol/kg mouse-antibody (1:10,000; ab97023; Abcam) and anti-rabbit-antibody
and for the second injection 2 mmol/kg, based on our previous study (1:10,000; ab6721; Abcam). As loading controls, anti-Tubulin
(Doeppner et al., 2017). The injection volume for the first injection was (1:10,000; sc-8035; Santa Cruz Biotechnology) or anti-GAPDH
around 50 μl and for the second around 100 μl depending on the indi­ (1:10,000; GTX627408; Irvine, GeneTex, California, USA) were used,
vidual bodyweight. The lithium + U0126 and U0126 groups addition­ after first visualization of the specific protein of interests. All antibodies
ally received intravenous injections of 0.5 mg/kg of U0126 (Cell used were diluted in 5% milk in PBS. The membranes were bathed in
Signaling Technology) at the beginning of the reperfusion. Control mice ECL reagent (Cell Signaling Technology) and developed with the
for the experiments with U0126 received the same volume of DMSO ChemiDoc™ XRS+ (Bio-Rad) imaging system. For quantification, the
intravenously. Mice with 72 h survival were further divided into a density of the protein of interest and the housekeeping protein tubulin or
control and a lithium group. Both groups received the same treatment as GAPDH was measured using the Image Lab software (Bio-Rad). The
the control and the lithium groups with 24 h survival and two additional results are presented as the ratio between protein of interest and
intraperitoneal injections with 2 mmol/kg of lithium each at 24 h and at housekeeping protein in percentage. Separate membranes were used for
48 h after MCA occlusion. Overview tables showing all treatment groups each antibody incubation and detection. Representative and the com­
and animal numbers are provided in Tables S1 and S2. For each plete, uncropped membranes for each experimental condition are shown
experiment conducted (i.e., Evans Blue extravasation, Western blot in the supplementary section of the manuscript (Figs. S1–S6).
analysis, immunohistochemical analyses, zymography and flow cytom­
etry), new experimental groups were generated with the 2.8. Immunohistochemical analyses
above-described treatment paradigms.
For immunohistochemical analyses, the mice were transcardially
2.5. Evans Blue extravasation perfused with ice cold PBS before killing. The paraffin embedding the
brain sections (diameter: 4 μm) were deparaffined and boiled in citrate
Evans Blue extravasation was performed as previously described puffer (pH 6.0) and washed in PBS. After blocking with buffer containing
(Doeppner et al., 2011). Briefly, 100 μl of 4% Evans Blue dye (Sig­ tris buffered saline (pH 7.4), 2% BSA, 10% donkey serum, 0.25% Triton
ma-Aldrich) was administered using a retrobulbar access route 2 h X-100, the sections were incubated overnight with the following pri­
before killing. Subsequently, the mice were sedated and cardiac perfu­ mary antibodies: Iba-1 (1:1000; 019–19741; Wako Inc., Osaka, Japan),
sion with PBS was performed. The hemispheres were weighed and lysed MMP-9 (1:500; sc-393859; Santa Cruz Biotechnology), claudin-1
in 50% trichloroacetic acid. After centrifugation for 20 min at 10,000 (1:1000; sc-137121M; Santa Cruz Biotechnology), ZO-1 (1:500;
rpm the supernatant was diluted with ethanol and the absorbance at ab96587; Abcam), caveolin-1 (1:800; ab2910; Abcam), NeuN (1:1000;
620 nm was measured photometrically. The measured Evans Blue con­ ab104225; Abcam), GFAP (1:1000; AB5541; Merck), CD31 (1:500;
centration was calculated based on a standard curve. The standard curve ab7388; Abcam) and 4‘,6-Diamidin-2-phenylindol (1:10,000; A1001;
was plotted by measuring the absorbance versus optical density of AppliChem, Darmstadt, Germany). Thereafter, the sections were incu­
samples with defined concentrations of Evans Blue (0.25–20 μg/ml). The bated for 1 h with the following secondary antibodies: AlexaFlour488-
final concentration was then calculated as (μg) Evans Blue per (g) tissue. anti-rabbit (1:10,000; 711-545-152; Jackson ImmunoResearch, Ely,
UK), Cy3-anti-mouse (1:10,000; 715-165-150; Jackson ImmunoR­
2.6. Tissue preparation for Western blot analysis esearch), Cy3-anti-rabbit (1:10,000; 711-165-152; Jackson ImmunoR­
esearch). For double staining using antibodies from the same host, the
The brain tissue was lysed in a buffer containing 50 mM Tris, 1% sections were blocked for 4 h with a Fab fragment antibody (1:50; 711-
Triton-X 100, 131 mM sodium chloride, 1 mM sodium diphosphate, 1 007-003, Jackson ImmunoResearch) at room temperature before incu­
mM sodium fluoride, 1 mM EDTA, protease inhibitors (cOmplete™; bation with the second primary antibody. Five pictures per hemisphere
Sigma-Aldrich) and phosphatase inhibitors (PhosSTOP™; Sigma- were taken in our regions of interest (ROI) with the Axioplan 2 fluo­
Aldrich) using a homogenisator for 10 min followed by centrifugation rescence microscope (Carl Zeiss) and the average intensity was calcu­
at 4 ◦ C with 16,000 rpm for 10 min. The supernatant was collected and lated or positive cells were counted. The subventricular zone (SVZ) and
the quantification of the protein concentration photometrically accom­ the basal ganglia were defined as our ROIs. Stereotactic coordinates for
plished (Pierce™ BCA Protein Assay Kit; Thermo Fisher Scientific). The the SVZ were 0.14 mm anterior, 2–3 mm ventral and 1–1.25 mm lateral
samples were aliquoted and stored at − 80 ◦ C until Western blot analysis from bregma and for basal ganglia 0.14 mm anterior, 2.5–3.25 mm

3
M. Haupt et al. Neuropharmacology 181 (2020) 108357

ventral and 1.5–2.25 mm lateral from bregma. The software ImageJ 2.11. Statistical analysis
(National Institutes of Health, Bethesda, Maryland, USA) were used for
cell counting and intensity quantification. For all experimental analyses, the experimenter was blinded towards
the treatment paradigm, and animals were randomly allocated to either
treatment paradigms. All data were normally distributed as indicated by
2.9. Zymographic measurement of MMP-2 and MMP-9 activity
the Kolmogorov-Smirnov test. Accordingly, parametric tests were
applied. These tests included the Student’s t-test (comparison between
The gelatin zymography measurement were performed with affinity-
two groups) and a one-way analysis of variance (ANOVA) followed by
support purification as previously described (Zhang and Gottschall,
Sidak’s multiple comparisons test (comparison between multiple
1997). Briefly, the mice were transcardially perfused with ice cold PBS
groups). Data were presented as means ± standard deviations (SD). p
before killing. After collecting the brains, hemispheres were lysed in a
values < 0.05 were regarded as statistically significant.
non-reducing lysis buffer containing 50 mmol/l Tris-HCl (pH 7.6), 150
mmol/l NaCl, 5 mmol/l CaCl2, 0.05% BRIJ-35, 0.02% NaN3 and 1%
3. Results
Triton X-100, and afterwards centrifuged for 5 min at 12,000 rpm. The
supernatant was collected, the protein concentration measured
Lithium increases EC viability, enhances P-gp expression, and in­
(Pierce™ BCA Protein Assay Kit; Thermo Fisher Scientific) and incu­
creases the abundance of tight junction proteins after OGD injury.
bated with 1:10 volume of sepharose 4B (Sigma-Aldrich) for 60 min.
In order to examine a putative impact of lithium on cerebral EC in
After incubation and centrifugation, the purified pellet was resuspended
vitro, cultured EC were exposed to OGD with subsequent re-cultivation
in lysis buffer containing additional 10% DMSO. Equal volumes were
under standard cell culture conditions. Thereafter, cell viability,
then incubated with non-reducing sample buffer (Carl Roth) and loaded
expression of P-glycoprotein (P-gp; synonyms: multidrug resistance
on a zymogram gel (Novex™) containing 0.1% gelatin. As standards,
protein 1 (MDR1), ATP-binding cassette sub-family B member 1
0.1 ng pro-MMP-2 (Merck) and pro-MMP9 (Merck) and 0.01 ng of active
(ABCB1), cluster of differentiation 243 (CD243)), and the abundance of
MMP-2 (Merck) and active MMP-9 (Merck) were used. After electro­
selected tight junction proteins were quantitatively measured 38 h after
phoresis, the gel was washed twice with 2.5% Triton X-100 and incu­
induction of OGD. The cell viability assay revealed a significant
bated for 96 h in developing buffer (Novex™). After incubation, the gel
enhancement in the ratio of live cells in the lithium-treated group in
was stained with SimplyBlue staining solution (Novex™) over night and
comparison to the control group (F(2,26) = 161.1, p = <0.0001)
then destained in deionized water. After destaining, white bands remain
(Fig. 1A). However, the ratio of live cells in the lithium-treated group
before a dark background. Following this, gels were scanned using a
was still significantly lower compared to the non-OGD group (F(2,26) =
ChemiDoc™ XRS+ (Bio-Rad) imaging system and densitometrically
161.1, p = <0.0001) (Fig. 1A). Using the MTT assay, we observed a
analyzed using the software ImageJ (National Institutes of Health).
significant increase (t(35) = 3.5, p = 0.0015) in cellular metabolism of the
lithium-treated group vs the control (Fig. 1B).
2.10. Analysis of brain infiltrating leukocyte by flow cytometry Quantitative Western blot analysis also revealed a significantly
higher expression of zonula occludens-1 (ZO-1) (t(26) = 3.786, p =
Infiltrating leukocytes and the subset of T cells, B cells, neutrophils, 0.0008) and occludin (t(18) = 2.771, p = 0.0126) in the lithium group
macrophages and monocytes were determined by flow cytometry with a compared to the control group (Fig. 1E–FE). In addition, P-gp abundance
fluorescence-activated cell sorter as described (Pösel et al., 2016). was significantly increased after treatment with lithium when EC were
Briefly, the right ischemic hemispheres were broken down with a pestle exposed to OGD (t(16) = 2.875, p = 0.011) (Fig. 1C). These results are in
by hand in lysis buffer (0.5% BSA, 5% glucose, DNase (10 mg/ml), PBS) favor of lithium enhancing both the cell-cell-integrity and modulating P-
and centrifuged at 1600 rpm for 10 min. Thereafter, the pellet was gp expression of cultivated ECs after OGD in vitro.
solved in 30% Percoll solution (GE Healthcare, Chicago, Illinois, USA)
and loaded on the Percoll gradient containing 45% and 70% Percoll 3.1. Lithium reduces post-ischemic brain extravasation of Evans Blue
phases. Following centrifugation, the leukocytes between the phases
were aspirated and dissolved in working solution (3% fetal calf serum in Next, the Evans Blue extravasation was used to investigate the post-
PBS). After washing, the cells were incubated with Zombie dye (1:500; stroke integrity of the BBB after lithium administration in mice. Evans
BioLegend, San Diego, California, USA) for 10 min as a viability control. Blue concentrations in brain tissue of the right ischemic hemisphere of
After adding anti-CD16/32 (1:100; BioLegend) for 10 min, the cells were both stroke mice and non-ischemic sham mice were analyzed 24 h after
centrifuged and washed again. Subsequently, the cells were incubated MCA occlusion. Spectrophotometric analysis revealed a significant in­
with anti-CD3 (1:50; Becton Dickinson, Franklin Lakes, New Jersey, crease in Evans Blue concentration in the brain parenchyma of controls
USA), anti-CD45 (1:100; BioLegend), anti-Ly6G (1:50; BioLegend), compared to the sham group (F(2,21) = 61.51, p = <0.0001) (Fig. 2A).
anti-CD19 (1:50; BioLegend) and anti-CD11b (1:50; BioLegend) anti­ However, the Evans Blue concentration in the lithium group was
bodies overnight. Gating and quantification were obtained with the significantly lower compared to the control group (F(2,9) = 37.15, p =
software FlowJo v. 10.5.3 (Becton Dickinson). The gating pattern was <0.0001) (Fig. 2A), suggesting an enhanced level of BBB stability due to
based on the study by Pösel and colleagues (Pösel et al., 2016) (Fig. 8H). lithium treatment.
In detail, leukocytes are defined as CD45high cells and microglia as
CD45int cells out of single cells. For the latter, it is not possible to 3.2. Lithium minimizes post-ischemic loss of tight junction proteins ZO-1,
distinguish between activated and inactivated cells. The CD45low frac­ occludin and claudin-1 in vivo
tion includes neuronal cells, astroglia, ependymal and endothelial cells
and were not analyzed in the present study. Neutrophils are defined as Since lithium led to an enhanced expression of tight junction proteins
Ly6G+CD3− cells and T cells are defined as Ly6G-CD3+ cells out of all of EC exposed to OGD, we next wanted to investigate if this also
leukocytes. Ly6G-CD3− cells were further divided into B cells and mon­ occurred in vivo. Using immunofluorescence and Western blotting, the
ocytes/macrophages. Herein, B cells are defined as CD11b-CD19+ cells expression of ZO-1, occludin, and claudin-1 was measured in ischemic
out of CD45highLy6G-CD3− cells and monocytes/macrophages are hemispheres both 24 h and 72 h after MCA occlusion.
defined as CD11b+CD19− cells out of CD45highLy6G-CD3− cells. The The immunofluorescence staining showed a significantly higher
three parental gates of single cells, CD45high cells and CD45high expression of claudin-1 in the lithium group compared to the control
Ly6G-CD3− cells were always set as 100% for further analysis in the group 24 h after MCA occlusion (t(15) = 8.654, p = 0.0001) (Fig. 2B).
subsequent gates. Further analysis revealed claudin-1 positive cells to co-localize with

4
­
M. Haupt et al. Neuropharmacology 181 (2020) 108357

Fig. 1. In vitro lithium treatment increases endothelial cell (EC) viability and enhances both P-glycoprotein (P-gp) and tight junction protein abundance.
EC underwent 14 h of oxygen glucose deprivation (OGD) followed by subsequent treatment with medium or medium containing 1.25 mM lithium chloride (OGD +
Li) for 24 h. (A) Analysis of live (green) and dead (red) cells using a commercial cell viability kit. n = 10 for non-OGD and OGD, n = 9 for Li + OGD. Scale bars: 50
μm. (B) Analysis of cell viability using the 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The metabolic activity of non-OGD group
were taken as 100%. n = 16 per group. (C) Quantitative measurement of P-gp receptor protein expression using Western blot analysis normalized with the
housekeeping protein glycerol-3-phosphate dehydrogenase (GAPDH). n = 8 for OGD, n = 10 for Li + OGD. (D) Non-quantitative immunofluorescence staining of ZO-
1, depicting representative staining areas. Scale bars: 20 μm. (E) Quantitative measurement of tight junction protein ZO-1 expression using Western blot analysis
normalized with the housekeeping protein Tubulin. n = 14 per group. (F) Quantitative measurement of tight junction proteins occludin expression using Western blot
analysis normalized with the housekeeping protein GAPDH. n = 6 for OGD, n = 14 for Li + OGD. Data are expressed as mean ± SD. *p < 0.05, **p < 0.01, ***p <
0.001, ****p < 0.0001.f

CD31 positive cells (Fig. S7). The quantitative Western blot analysis 0.0057) and sham group (F(2,9) = 10.69, p = <0.0001) (Fig. 3A). This is
revealed higher expression of occludin (t(14) = 3.108, p = 0.0077) and in line with our zymography analysis, which revealed a significantly
ZO-1 (t(14) = 3.493, p = 0.0036) in the lithium group compared to the enhanced pro-MMP-9 activity in control mice when compared to both
control group 24 h after MCA occlusion (Fig. 2C). After 72 h of reper­ sham mice (F(2,27) = 10.12, p = 0.0004) or mice treated with lithium
fusion, the quantitative Western blot analysis showed a higher expres­ (F(2,27) = 10.12, p = 0.0196). Moreover, the activity of activated MMP-9
sion of ZO-1 (t(10) = 2.468, p = 0.0332) in the lithium group compared to was significantly elevated in control mice compared to sham mice
the control group, with no significant impact of lithium on occludin (F(2,27) = 24.69, p = <0.0001) and lithium treated mice (F(2,27) = 24.69,
expression (t(10) = 0.7897, p = 0.448) at this time point (Fig. 2D). These p = 0.0007). However, the quantity of activated MMP-9 was still
results suggest that delivery of lithium minimizes the loss of tight significantly enhanced in lithium treated mice compared to sham mice
junction proteins 24 h after MCA occlusion. (F(2,27) = 24.69, p = 0.0332) (Fig. 3B). Interestingly, significant
increased pro-MMP-2 activity in control mice compared to sham mice
were observed (F(2,27) = 7.118, p = 0.0024), but no significant difference
3.3. Lithium reduces post-ischemic activity of MMP-9 by caveolin-1 between the control and the lithium group was seen (F(2,27) = 7.118, p =
independent pathways 0.2125) (Fig. 3B). Active MMP-2 activity was not detectable in all three
groups. As caveolin-1 has been shown to regulate the activity of MMPs,
We next evaluated possible mechanisms that might be involved in we proceeded to investigate the expression of caveolin-1 in our system.
the process of stabilization of the BBB. Therefore, the expression and Both our quantitative Western blot analysis (F(2,19) = 8.932, p = 0.9997)
activity of MMP-9 as well as the expression of caveolin-1 was measured and immunofluorescence staining (F(2,22) = 0.6207, p = 0.8275) showed
using immunofluorescence staining and zymography. The immunoflu­ no significant difference in caveolin-1 expression between the control
orescence staining showed a significantly higher levels of MMP-9 in the and the lithium group (Fig. 3C–D). However, our Western blot analysis
control group in comparison to the lithium group (F(2,21) = 16.53, p =

5
M. Haupt et al. Neuropharmacology 181 (2020) 108357

Fig. 2. Lithium preserves blood-brain


barrier (BBB) integrity and expres­
sion of tight junction proteins
claudin-1, zonula occludens-1 (ZO-1)
and occludin after cerebral ischemia
in mice. Mice were exposed to 45 min of
middle cerebral artery (MCA) occlusion
followed by 24 h or 72 h survival with
reperfusion. Sham group mice under­
went the surgery procedure without
MCA occlusion. Mice were treated with
phosphate buffered saline (PBS, control)
or intraperitoneally treated at the
beginning of reperfusion with 1 mmol/
kg lithium chloride and 6 h later with
another 2 mmol/kg of lithium chloride.
Mice with 72 h survival got two addi­
tional injections of PBS (control) or 2
mmol/kg lithium chloride 24 h and 48 h
after MCA occlusion. (A) Analysis of the
BBB integrity using the Evans Blue
extravasation assay of the right ischemic
hemisphere. n = 8 per group. (B)
Quantitative analysis of claudin-1
expression by immunofluorescence
staining within the right ischemic stria­
tum. Scale bars: 20 μm. n = 9 for control,
n = 8 for lithium. (C) Quantitative
analysis of ZO-1 and occludin expression
24 h after MCA occlusion using Western
blot analysis of the right ischemic
hemisphere. Normalized with house­
keeping proteins tubulin or glycerol-3-
phosphate dehydrogenase (GAPDH). n
= 6 for control, n = 10 for lithium. (D)
Quantitative analysis of ZO-1 and
occludin expression 72 h after MCA oc­
clusion using Western blot analysis of
the right ischemic hemisphere. Normal­
ized with housekeeping proteins tubulin
or GAPDH. n = 6 per group. Data are
expressed as mean ± SD. *p < 0.05, **p
< 0.01, ***p < 0.001, ****p < 0.0001.

revealed a significant induction of caveolin-1 expression in the control the ischemic hemisphere (Fig. 4A,C,E). However, we observed through
(F(2,19) = 8.932, p = 0.0064) and in the lithium group (F(2,19) = 8.932, p Western blot analysis that the phosphorylation of c-Raf (p-c-Raf) (F(2,19)
= 0.003) in comparison to the sham group. Immunohistochemical = 5.823, p = 0.0282, p = 0.0165), MEK1/2 (p-MEK1/2) (F(2,19) = 13.18,
analysis revealed caveolin-1 to be expressed in ECs, astroglial cells, and p = 0.0002, p = 0.0129) and ERK1/2 (p-ERK1/2) (F(2,19) = 13.19, p =
neurons (Fig. S8). Taken together, lithium significantly reduces MMP-9 0.0003, p = 0.0048), which marks these kinases as active were signifi­
activity through a caveolin-1 independent pathway. cantly increased in the lithium group compared to the sham (first p
value) and control (second p value) (Fig. 4B,D,F). In addition, Western
blot analysis revealed significantly higher levels of Bcl-2 in the sham
3.4. Lithium activates the MAPK/ERK1/2 pathway by elevated c-Raf, (F(2,19) = 6.041, p = 0.0409) and lithium group (F(2,19) = 6.041, p =
MEK1/2, and ERK1/2 phosphorylation 0.0466) both compared to control (Fig. 4G).

Evidence suggests that ERK1/2 affects the BBB integrity by regu­


lating ZO-1 and occludin expression (Liu et al., 2014). In view of these 3.5. Inhibition of the MAPK/ERK1/2 pathway reverses lithium-induced
findings, we analyzed the MAPK/ERK1/2 signaling pathway. Western effects on MMP-9 and BBB integrity
blot analysis of absolute c-Raf, MEK1/2 and ERK1/2 expression revealed
no difference between the sham, control and lithium group after 24 h in To further analyze whether the MAPK/ERK1/2 pathway is a key

6
M. Haupt et al. Neuropharmacology 181 (2020) 108357

Fig. 3. Lithium decreases matrix metallopeptidase- 9 (MMP-9) activity independent of caveolin-1 expression. Mice were exposed to 45 min of middle cerebral
artery (MCA) occlusion followed by 24 h survival. Sham group mice underwent the surgery procedure without MCA occlusion. Mice were treated with phosphate
buffered saline (PBS, control and sham) or intraperitoneally treated at the beginning of reperfusion with 1 mmol/kg lithium chloride and 6 h later with another 2
mmol/kg of lithium chloride. (A) Determination of MMP-9 quantity by immunofluorescence staining and measurement of fluorescence intensity of the ischemic and
sham striatum normalized with negative control. n = 8 per group. (B) Analysis of MMP-9 and MMP-2 activity using gelatine zymography of the right ischemic
hemisphere or right sham hemisphere. Commercially available pro-MPP-2/MMP-9 and active MMP-2/MMP-9 were used as standards (Std). n = 10 per group. (C)
Analysis of caveolin-1 expression using immunofluorescence staining and measurement of fluorescence intensity in the ischemic and sham striatum. Scale bars: 20
μm. n = 8 for sham and lithium, n = 9 for control. (D) Quantitative analysis of caveolin-1 expression by Western blot analysis of the right ischemic hemispheres and
sham hemispheres. Western blot was normalized with the housekeeping protein tubulin. n = 6 for sham and control, n = 10 for lithium. Data are expressed as mean
± SD. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

7
M. Haupt et al. Neuropharmacology 181 (2020) 108357

Fig. 4. Lithium modulates the MAPK/ERK1/2 pathway. Mice were exposed to 45 min of middle cerebral artery (MCA) occlusion followed by 24 h survival. Sham
group mice underwent the surgery procedure without MCA occlusion. Mice were treated with phosphate buffered saline (PBS, control) or intraperitoneally treated at
the beginning of reperfusion with 1 mmol/kg lithium chloride and 6 h later with another 2 mmol/kg of lithium chloride. (A–G) Quantitative analysis of c-Raf, p-c-
Raf, MEK1/2, p-MEK1/2, ERK1/2, p-ERK1/2, and Bcl-2 expression by Western blot analysis of the right ischemic hemisphere and sham hemispheres. Normalized
with the housekeeping proteins tubulin or glycerol-3-phosphate dehydrogenase (GAPDH). n = 6 for sham and control, n = 10 for lithium. Data are expressed as mean
± SD. *p < 0.05, **p < 0.01, ***p < 0.001.

player in lithium-induced enhancement of post-stroke BBB integrity, the phagocytes and thereby a modulated neuroinflammatory response by
MEK1/2 inhibitor U0126 was applied in order to inhibit the pathway. lithium.
Expression levels of MEK1/2, p-MEK1/2, ERK1/2, and p-ERK1/2 after
treatment with U0126 were determined using Western blot analysis at 3.7. Lithium modulates the expression of the endothelial surface proteins
24 h after MCA occlusion. Whereas absolute expression levels of MEK1/ ICAM-1 and P-gp after stroke in vivo
2 did not differ between the treatment groups (F(2,20) = 0.04393, p =
0.9571), the levels of p-MEK1/2 were significantly decreased in the Besides BBB opening, the expression of the intercellular adhesion
U0126 group in comparison to the control group (F(2,20) = 3.842, p = molecule 1 (ICAM-1) and vascular cell adhesion protein 1 (VCAM-1) also
0.0387) (Fig. 5A–B). Likewise, total levels of ERK1/2 were not affected play important roles in leukocyte infiltration into the brain parenchyma
by the various treatment schemes (F(2,20) = 0.2932, p = 0.7490), while (Yanamadala and Friedlander, 2010). Indeed, Western blot analysis of
the levels of p-ERK1/2 were significantly decreased in the U0126 group the ischemic hemispheres showed a significantly lower expression of
(F(2,20) = 6.577, p = 0.0037) compared to control (Fig. 5C–D), further ICAM-1 in lithium-treated mice when compared to controls (t(14) =
confirming inhibition of the MAPK/ERK1/2 pathway by U0126. The 3.659, p = 0.0026) (Fig. 7A). However, no difference in VCAM-1
subsequent zymography analysis showed no difference in pro-MMP-9 expression was observed (Fig. 7B). Since lithium showed an effect on
(F(2,21) = 0.7083, p = 0.5039), active-MMP-9 (F(2,21) = 0.001703, p = P-gp expression in ECs, we analyzed P-gp expression levels in mice
0.9983) and pro-MMP-2 (F(2,21) = 0.3667, p = 0.6974) activity between exposed to cerebral ischemia. Likewise, P-gp expression levels were
the controls and mice treated with either lithium + U0126 or U0126 significantly increased in lithium-treated mice when compared to con­
alone (Fig. 5E). In addition, spectrophotometric analysis revealed no trols (t(14) = 2.371, p = 0.0326) (Fig. 7C).
significant difference in Evans Blue concentrations between controls and
mice treated with either lithium + U0126 or U0126 alone (F(2,20) = 1.16, 3.8. Lithium modulates the early migration of immune cells into the
p = 0.3337) (Fig. 5F). These data show that activation of the MAPK/ infarct area
ERK1/2 pathway and subsequent inhibition of MMP-9 activity is critical
in lithium-induced stability of the post-stroke BBB. Next, we analyzed the impact of lithium on very early infiltrating
leukocytes. Herein, we focused on neutrophil, T cell, B cell, monocyte
3.6. Lithium reduces post-ischemic activation of phagocytes and macrophage markers. The brain tissue of the ischemic hemisphere
was analyzed by flow cytometry 24 h after MCA occlusion. The subsets
Neuroinflammation and BBB opening after hypoxia are closely of leukocytes (CD45high), resident microglia (CD45int), T cells
linked (Jiang et al., 2018). To evaluate whether lithium modulates (CD45highCD3+), neutrophils (CD45highLy6G+), B cells
high -
phagocyte activation 24 h after MCA occlusion, immunofluorescence (CD45 CD3 Ly6G CD19 ) and macrophages and monocytes
− +

staining for the calcium-binding adapter molecule 1 (Iba-1) of the right (CD45highCD3-Ly6G− CD11b+) were analyzed (Fig. 8H). The analysis
ischemic hemispheres was performed. The immunofluorescence staining showed a significantly higher proportion of leukocytes in the control
showed a significant decrease in morphologically activated phagocytes group vs both the lithium group (F(2,12) = 28.65, p = <0.0001) and sham
in the lithium group compared to the control group (t(14) = 5.702, p = group (F(2,12) = 28.65, p = 0.0002) (Fig. 8A). No difference in the subset
0.0001) (Fig. 6). These data suggest a very early effect on activation of of resident microglia was observed (Fig. 8B). Since extracranial

8
M. Haupt et al. Neuropharmacology 181 (2020) 108357

Fig. 5. The MEK1/2 inhibitor U0126 reverses lithium-induced effects on MMP-9 and post-stroke blood-brain barrier (BBB) integrity. Mice were exposed to
45 min of middle cerebral artery (MCA) occlusion followed by 24 h of survival. Animals were treated with phosphate buffered saline (PBS, control) or with 0.5 mg/kg
U0126 at the beginning of the reperfusion. The combined treatment approach consisted of an intravenous injection of 0.5 mg/kg U0126 plus an intraperitoneal
injection of 1 mmol/kg lithium chloride at the beginning of the reperfusion followed by another intraperitoneal injection of 2 mmol/kg lithium chloride 6 h later.
(A–D) Quantitative analysis of MEK1/2, p-MEK1/2, ERK1/2 and p-ERK1/2 expression patterns by Western blot analysis of the right ischemic hemisphere. Western
blots were normalized with the housekeeping protein glycerol-3-phosphate dehydrogenase (GAPDH). n = 8 for control and lithium + U0126, n = 7 for U0126. (E)
Analysis of MMP-9 and MMP-2 activity using gelatine zymography of the right ischemic hemisphere. Commercially available pro-MPP-2/MMP-9 and active MMP-2/
MMP-9 were used as standards (Std). n = 8 per group. (F) Analysis of the BBB integrity using the Evans Blue extravasation assay of the right ischemic hemisphere. n
= 8 for control and U0126, n = 7 for lithium + U0126. Data are expressed as mean ± SD. *p < 0.05, **p < 0.01.

leukocytes usually do not invade the brain parenchyma under physio­ stage of stroke. For the first time, we were able to show that lithium
logical conditions, we only observed some leukocytes in the sham group. significantly increases the stability of the BBB upon experimental stroke
Consequently, a further analysis of different cellular subpopulations in in mice. Lithium acts through impairing the activity of MMP-9 and
sham animals was not performed. enhancing phosphorylation as well as activation of the pro-survival
Further analyzing of the cell subtypes in control and lithium group MAPK/ERK1/2 pathway, all of which resulting in reduced BBB
revealed a significant increase in the subset of T cells (t(8) = 3.44, p = leakage. Conversely, inhibition of the MAPK/ERK1/2 pathway reverses
0.0088), and a significant decrease in the subset of neutrophils (t(8) = the lithium-induced reduction of MMP-9 activity and abolished the ef­
2.34, p = 0.0474) in the lithium group compared to the control group fect on BBB permeability. In addition, lithium decreases ICAM-1
(Fig. 8C–D). Lithium did not affect the population of B cells (t(8) = expression which reduces neutrophil invasion and increases T cell
0.1148, p = 0.9114), monocytes and macrophages (t(8) = 0.8556, p = migration into the brain.
0.4171) (Fig. 8E–F). Taken together, these results indicate that lithium Previous studies have already reported a modulation of Ca2+ ho­
treatment decreases the infiltration of leukocytes and especially meostasis and the survival-promoting effects by lithium treatment in
neutrophil invasion into the brain parenchyma at 24 h post-stroke. On vitro under both ischemic and non-ischemic conditions (Bosche et al.,
the contrary, the ratio of T cells is elevated at this early stage upon 2013, 2016a; 2016b; Cimarosti et al., 2001; Silachev et al., 2016).
lithium treatment. However, the role of tight junction protein expression has not been
explored in such studies. Opening of the BBB is a key event during the
4. Discussion acute phase of cerebral ischemia, where the degradation of tight junc­
tion proteins such as claudin-1, ZO-1 and occludin appears to be critical
Lithium has been recently recognized to exert neuroprotection upon (Jiang et al., 2018; Jiao et al., 2011; Yadav and Shin, 2015). Through an
experimental induction of stroke. Although the precise mechanism of in vitro approach, we showed that lithium enhances the resistance of
action still remains elusive, results from a clinical study seemed prom­ cultivated EC against OGD, inhibiting the OGD-induced downregulation
ising, demonstrating a trend towards better neurological recovery in of tight junction proteins. In this context, Krueger and colleagues iden­
stroke patients treated with lithium (Abdollahi et al., 2014). While tified four different stages of BBB breakdown after cerebral ischemia,
additional clinical trials should be conducted to verify the significance of with the ultimate loss of endothelial cells occurring during the last stage,
lithium treatment in stroke patients, understanding of the downstream indicating the importance of the present findings (Krueger et al., 2015).
mechanisms by which lithium induces its neuroprotective effects upon Furthermore, our in vivo studies support this observation where we
stroke is necessary to identify stroke patients who will benefit from such found lithium treatment to increase expressions of claudin-1, ZO-1 and
a lithium treatment. The present study therefore aimed to analyze, if occludin 24 h post-MCA occlusion. Interestingly, 72 h after MCA oc­
lithium plays a role in the BBB opening, a key mechanism in the acute clusion resulted in only a significant difference in ZO-1 expression

9
M. Haupt et al. Neuropharmacology 181 (2020) 108357

Fig. 7. Lithium modulates expression of the endothelial surface protein


Fig. 6. Lithium reduces post ischemic phagocyte activation. Mice were intercellular adhesion molecule 1 (ICAM-1) and the P-glycoprotein (P-gp)
exposed to 45 min of middle cerebral artery (MCA) occlusion followed by 24 h Mice were exposed to 45 min of middle cerebral artery (MCA) occlusion fol­
survival. Mice were treated with phosphate buffered saline (PBS, control) or lowed by 24 h survival. Mice were treated with phosphate buffered saline (PBS,
intraperitoneally treated at the beginning of reperfusion with 1 mmol/kg control) or intraperitoneally treated at the beginning of reperfusion with 1
lithium chloride and 6 h later with another 2 mmol/kg of lithium chloride. mmol/kg lithium chloride and 6 h later with another 2 mmol/kg of lithium
Analysis of morphologically activated IBA-1 positive phagocytes using immu­ chloride. (A) Measurement of ICAM-1 expression using Western blot analysis of
nofluorescence staining of the right ischemic striatum. Representative immu­ the right ischemic hemisphere normalized with the housekeeping protein
nofluorescence stainings are shown with scale bars: 20 μm. n = 8 per group, tubulin. (B) Quantification of vascular cell adhesion protein 1 (VCAM-1)
data are expressed as mean ± SD. ****p < 0.0001. expression by Western blot analysis of the right ischemic hemisphere normal­
ized with the housekeeping protein glycerol-3-phosphate dehydrogenase
between the control and the lithium-treated group. This can be (GAPDH). (C) Measurement of P-gp expression using Western blot analysis of
explained by the fact that development of BBB leakage upon induction of the right ischemic hemisphere normalized with the housekeeping protein
cerebral ischemia occurs with temporal resolution. Indeed, Jiao and tubulin. n = 6 for control and n = 10 for lithium. Data are expressed as mean ±
SD. *p < 0.05, **p < 0.01.
colleagues have previously analyzed the time-dependent dynamics of
tight junction protein degradation and restoration following cerebral
ischemia in mice (Jiao et al., 2011). Whereas Jiao et al. suggest a induced by an upregulated expression and increased activity of MMPs
restoration of tight junction protein expression at 120 h post-stroke (Jiao such as MMP-2 and MMP-9 (Kurzepa et al., 2014). Based on this
et al., 2011), our results are in favor of a partial restoration of the BBB at observation, different studies have proposed an antagonization of
72 h, indicated by the increased expression of occludin in the control MMP-9 activity to be a potential therapeutic strategy against cerebral
group. However, this could be context-dependent and likely to be a ischemia and subsequent BBB leakage (Medsker et al., 2016). Indeed,
consequence of the short duration of cerebral ischemia chosen for our some MMP-9 inhibitors have shown promising neuroprotective effects
study. In our study, we focused mainly on EC and tight junction (Jiang et al., 2001; Murozono et al., 2009). In line with former reports,
expression as the predominant factor for BBB integrity. Although there is we were able to observe a stroke-induced increase of pro-MMP-9,
no doubt about the role of EC in maintaining BBB integrity, the inter­ pro-MMP-2 and activated MMP-9 (Park et al., 2009). Here, we showed
action of EC with mural cells, immune cells, glial cells, and neural cells is that lithium inhibits the post-ischemic increase of pro-MMP-9 expres­
also critical (Daneman and Prat, 2015). Indeed, Sliachev and colleagues sion together with a reduced quantity of activated MMP-9. Apart from its
demonstrated that lithium treatment protects astrocytes against OGD in expression, MMP-9 activity can also be regulated by other means such as
vitro, indicating that the protective effect of lithium is not limited to EC the MAPK pathway, which has been shown to negatively regulate the
(Silachev et al., 2016). Therefore, lithium appears to modulate the entire activity of MMP-9 (Curry et al., 2010). In fact, we demonstrated that
neurovascular unit. lithium treatment led to an activation of the MAPK pathway as seen by
Post-ischemic degradation of tight junction proteins is mainly the induction of phosphorylated cRaf, MEK1/2 and ERK1/2.

10
M. Haupt et al. Neuropharmacology 181 (2020) 108357

Fig. 8. Lithium decreases post ischemic infiltration of neutrophils but increases T cell invasion into the ischemic hemisphere. Mice were exposed to 45 min
of middle cerebral artery (MCA) occlusion followed by 24 h survival. Sham group mice underwent the surgery procedure without MCA occlusion. Mice were treated
with phosphate buffered saline (PBS, control) or intraperitoneally treated at the beginning of reperfusion with 1 mmol/kg lithium chloride and 6 h later with another
2 mmol/kg lithium chloride. (A–F) Flow cytometry of brain tissue from sham, control and lithium group analysing leukocytes (CD45high), resident microglia
(CD45int), neutrophils (CD45highLy6G+), T cells (CD45highCD3+), macrophages/monocytes (CD45highCD3-Ly6G-CD11b+) and B cells (CD45highCD3-Ly6G-CD19+). (G)
Representative flow cytometry measurements of brain tissue from sham, control and lithium treated mice with three parent gates: single cells, CD45high and
CD45highCD3-Ly6G-. (H) Scheme of gating strategy for flow cytometry analysis. n = 5 per group, data are expressed as mean ± SD. *p < 0.05, ***p < 0.001, ****p
< 0.0001.

Importantly, the inhibition of the MAPK pathway using a MEK1/2 in­ prescribed drugs from the brain side to the blood side (Abbott et al.,
hibitor, abolished the effect of lithium on MMP-9 activity and inhibited 2002; Schinkel, 1999). A previous study has shown that P-gp expression
the maintenance of BBB integrity, suggesting the significance of MAP­ is upregulated in the capillary endothelium at 24 h after focal cerebral
K/ERK1/2 activation in this context. It is important to note that the ischemia (Spudich et al., 2006). Additionally, Ramos and colleagues
MAPK/ERK1/2 pathway can play a dual role in the pathophysiology of demonstrated that ischemic stress in rats induce the expression of P-gp
cerebral ischemia, where it can lead to both neuroprotective or detri­ in normally non-expressive cells such as astrocytes and neurons (Ramos
mental effects (Sawe et al., 2008). However, our study showed that et al., 2004). However, the role of P-gp after cerebral ischemia is not
lithium protects BBB integrity following stroke through activating the clear. A study in P-gp knockout mice suggests that the reduction of P-gp
MAPK pathway and subsequently inhibiting MMP-9 activity. In addi­ expression is neuroprotective (Murozono et al., 2009). On the other
tion, the level of Bcl-2, an anti-apoptotic protein downstream of the hand, P-gp has been shown to protect cells from intracellular accumu­
MAPK pathway, was increased after lithium treatment. This is in line lation of toxic components and was found to be elevated in
with previous studies reporting reduced apoptosis upon lithium treat­ hypoxia-induced apoptosis in cells (Johnstone et al., 1999; Lazarowski
ment after ischemic stroke in mice (Doeppner et al., 2017). et al., 2007; Robinson et al., 1997). Moreover, Kraya et al. suggest that
Apart from the MAPK pathway, the activity of MMP-9 can be regu­ P-gp activity positively affects tight junctional protein expression and
lated by multiple different signaling molecules, and caveolin-1 has also BBB resistance (Kraya et al., 2016). Therefore, further studies on the role
been shown to play a role in impairing MMP activity (Gu et al., 2011). In of P-gp in the context of cerebral ischemia are important to determine if
fact, expression of caveolin-1 is downregulated after cerebral ischemia the elevated P-gp expression observed upon lithium treatment in our
which leads to a post-ischemic increase of MMP-9 activity (Gu et al., system contributes to its neuroprotective properties.
2012). In this study, we found caveolin-1 to be ubiquitously expressed in The post-stroke immune cell response is associated with opening of
the ischemic brain parenchyma, but we did not observe any change of the BBB, and the immune system is known to be a key player in the
caveolin-1 expression patterns in the lithium group. Taken together, we pathophysiology of stroke (Dirnagl et al., 1999). Consistent with the
conclude that lithium modulates BBB integrity and MMP-9 activity in­ observed stabilization of the BBB by lithium, we found a decrease in
dependent of caveolin-1. early infiltration of leukocytes into the ischemic hemisphere following
Besides these effects on cell survival and tight junction protein lithium treatment. Of note, the proportion of neutrophils within the
expression, we also observed a lithium-induced increase in P-gp infiltrated leukocytes was significantly reduced. Neutrophils are among
expression in EC in vitro and in the ischemic hemisphere of mice 24 h the first hematogenous immune cells found in the brain after cerebral
after cerebral ischemia in vivo. Physiologically, P-gp is expressed on the ischemia (Anrather and Iadecola, 2016). Their extravasation is regu­
luminal side of EC of the BBB and it is an important gate-keeper of the lated by the activation of EC, release of chemokines and by the expres­
BBB (Schinkel, 1999). As a gate-keeper, P-gp plays a role in controlling sion of adhesion molecules such as ICAM1 and VCAM-1 (Dirnagl et al.,
the efflux of both small endogenous molecules and a large fraction of 1999). Previous studies suggest that neutrophils play an important role

11
M. Haupt et al. Neuropharmacology 181 (2020) 108357

in the exacerbation of ischemic brain injury (Garcia-Bonilla et al., 2014; Methodology. Dirk M. Hermann: Conceptualization, Writing - original
Herz et al., 2015). In fact, neutrophils can contribute to the destabili­ draft. Mathias Bähr: Conceptualization, Writing - original draft.
zation of the BBB by releasing active MMP-9 (Ludewig et al., 2013; Thorsten R. Doeppner: Conceptualization, Formal analysis, Writing -
Rosell et al., 2008). Moreover, inhibition of ICAM1 is associated with original draft, Writing - review & editing, Supervision.
decreased brain damage and improved outcome of experimental stroke
(Kitagawa et al., 1998; Vemuganti et al., 2004). In contrast, inhibition of Acknowledgment
VCAM-1 expression showed no effect on neutrophil invasion and infarct
size (Justicia et al., 2006). Interestingly, we found lithium to reduce the We thank both Irina Graf and Regine Kruse for excellent technical
expression of ICAM1, but not VCAM-1 expression. Taken together, the assistance.
reduced ICAM-1 expression and decreased neutrophil invasion might be
another important aspect of the lithium-induced decrease in MMP-9 Appendix A. Supplementary data
activity and subsequent maintenance of BBB integrity.
Apart from the reduction in neutrophil invasion, we found an Supplementary data to this article can be found online at https://doi.
increased proportion of T cells within the infiltrated leukocytes in org/10.1016/j.neuropharm.2020.108357.
ischemic hemispheres of the lithium-treated group. This could indicate
that lithium selectively decreases neutrophil invasion, which leads to an References
increase in the proportion of T cells. It is important to note that the role
of T cells however, remains controversial with some studies having re­ Abbott, N.J., Khan, E.U., Rollinson, C.M.S., Reichel, A., Janigro, D., Dombrowski, S.M.,
ported a detrimental effect of early T cell infiltration after cerebral Dobbie, M.S., Begley, D.J., 2002. Drug resistance in epilepsy: the role of the blood-
brain barrier. Novartis Found. Symp 243, 38–53. https://doi.org/10.1002/
ischemia (Hurn et al., 2007; Kleinschnitz et al., 2010). On the other 0470846356.ch4.
hand, Li and colleagues found that post-ischemic administration of Abdollahi, F., Moqaddam, M.M., Majdinasab, N., Sadr, F., Sajedi, S.A.,
regulatory T cells markedly reduced brain infarct size by stabilizing the Mohammadianinejad, S.E., 2014. The effect of lithium in post-stroke motor recovery.
Clin. Neuropharmacol 37, 73–78. https://doi.org/10.1097/
BBB through suppression of MMP-9 (Li et al., 2013). Notably, we have
wnf.0000000000000028.
not established, if lithium directly increases T cells invasion or if it Anrather, J., Iadecola, C., 2016. Inflammation and stroke: an overview.
indirectly enhances the proportion of T cells within the infiltrated Neurotherapeutics 13, 661–670. https://doi.org/10.1007/s13311-016-0483-x.
Bosche, B., Dohmen, C., Graf, R., Neveling, M., Staub, F., Kracht, L., Sobesky, J.,
leukocyte population by decreasing the population of neutrophils.
Lehnhardt, F.-G., Heiss, W.-D., 2003. Extracellular concentrations of non-transmitter
Therefore, further studies on the effect of lithium on immune cell amino acids in peri-infarct tissue of patients predict malignant middle cerebral artery
migration are warranted, including a more detailed analysis of the infarction. Stroke 34, 2908–2913. https://doi.org/10.1161/01.
subpopulation of infiltrated T cells involved after stroke induction. STR.0000100158.51986.EB.
Bosche, B., Hamann, G.F., Dohmen, C., Graf, R., 2006. There is more to it than: the
Finally, it is noteworthy that lithium is a substance with pleiotropic greater the infarction volume, the more probable is a malignant MCA infarction.
effects, which can modulate many different pathways after cerebral Stroke 37, 762–764. https://doi.org/10.1161/01.STR.0000204054.30458.e8.
ischemia. Our previous study indicated that lithium induced an increase Bosche, B., Macdonald, RL., 2015. Letter by Bosche and Macdonald regarding article,
“relevance of blood-brain barrier disruption after endovascular treatment of
in miR-124 expression that is involved in neuroprotection (Doeppner ischemic stroke: dual-energy computed tomographic study”. Stroke 46, e126–127.
et al., 2017). This suggests that lithium does not exclusively affect the https://doi.org/10.1161/STROKEAHA.115.009131.
MAPK/ERK1/2-pathway followed by stabilization of the BBB. We pro­ Bosche, B., Mergenthaler, P., Doeppner, TR., Hescheler, J., Molcanyi, M., 2020. Complex
clearance mechanisms after intraventricular hemorrhage and rt-PA treatment-a
pose that lithium affects different signaling pathways, depending on the review on clinical trials. Transl Stroke Res. 11, 337–344. https://doi.org/10.1007/
temporal resolution of the disease. s12975-019-00735-6.
Taken together, we showed for the first time that lithium stabilizes Bosche, B., Molcanyi, M., Noll, T., Rej, S., Zatschler, B., Doeppner, T.R., Hescheler, J.,
Müller, D.J., Macdonald, R.L., Härtel, F.V., 2016a. A differential impact of lithium on
the BBB after experimental stroke. Furthermore, our results provide
endothelium-dependent but not on endothelium-independent vessel relaxation.
novel mechanistic insights of lithium-induced neuroprotection in the Prog. Neuro-Psychopharmacology Biol. Psychiatry 67, 98–106. https://doi.org/
context of post-ischemic BBB opening in mice. Importantly, with only a 10.1016/j.pnpbp.2016.02.004.
Bosche, B., Molcanyi, M., Rej, S., Doeppner, T.R., Obermann, M., Müller, D.J., Das, A.,
single study conducted using lithium as a treatment for stroke patients,
Hescheler, J., Loch Macdonald, R., Noll, T., Härtel, F.V., 2016b. Low-dose lithium
both our in vitro and in vivo work provide substantial translational and stabilizes human endothelial barrier by decreasing MLC phosphorylation and
clinical relevance, opening up novel stroke treatment possibilities universally augments cholinergic vasorelaxation capacity in a direct manner. Front.
through targeting BBB stabilization. Physiol. 7 https://doi.org/10.3389/fphys.2016.00593.
Bosche, B., Schäfer, M., Graf, R., Härtel, F.V., Schäfer, U., Noll, T., 2013. Lithium
prevents early cytosolic calcium increase and secondary injurious calcium overload
Sources of funding in glycolytically inhibited endothelial cells. Biochem. Biophys. Res. Commun. 434,
268–272. https://doi.org/10.1016/j.bbrc.2013.03.047.
Chamorro, Á., Meisel, A., Planas, A.M., Urra, X., Van De Beek, D., Veltkamp, R., 2012.
The study was supported by grants of the Deutsche For­ The immunology of acute stroke. Nat. Rev. Neurol. https://doi.org/10.1038/
schungsgemeinschaft (DFG) to BB (BO 4229/1-1 and BO 4229/2-1). nrneurol.2012.98.
Chaturvedi, M., Kaczmarek, L., 2014. MMP-9 inhibition: a therapeutic strategy in
ischemic stroke. Mol. Neurobiol. 49, 563–573. https://doi.org/10.1007/s12035-
Declarations of interest 013-8538-z.
Chuang, D.-M., Wang, Z., Chiu, C.-T., 2011. GSK-3 as a target for lithium-induced
None. neuroprotection against excitotoxicity in neuronal cultures and animal models of
ischemic stroke. Front. Mol. Neurosci. 4 (15) https://doi.org/10.3389/
fnmol.2011.00015.
CRediT authorship contribution statement Cimarosti, H., Rodnight, R., Tavares, A., Paiva, R., Valentim, L., Rocha, E., Salbego, C.,
2001. An investigation of the neuroprotective effect of lithium in organotypic slice
cultures of rat hippocampus exposed to oxygen and glucose deprivation. Neurosci.
Matteo Haupt: Conceptualization, Investigation, Formal analysis, Lett. 315, 33–36. https://doi.org/10.1016/S0304-3940(01)02310-2.
Visualization, Writing - original draft, Writing - review & editing. Curry, A., Guo, M., Patel, R., Liebelt, B., Sprague, S., Lai, Q., Zwagerman, N., Cao, F.X.,
Bozena Zechmeister: Methodology, Writing - review & editing. Bert Jimenez, D., Ding, Y., 2010. Exercise pre-conditioning reduces brain inflammation in
stroke via tumor necrosis factor-α, extracellular signal-regulated kinase 1/2 and
Bosche: Investigation, Writing - review & editing. Simone Lieschke:
matrix metalloproteinase-9 activity. Neurol. Res. 32, 756–762. https://doi.org/
Investigation, Writing - original draft, Writing - review & editing. Xuan 10.1179/174313209x459101.
Zheng: Investigation. Lin Zhang: Investigation, Methodology. Vivek Daneman, R., Prat, A., 2015. The blood–brain barrier. Cold Spring Harb. Perspect. Biol. 7
Venkataramani: Investigation, Writing - review & editing. Fengyan https://doi.org/10.1101/cshperspect.a020412.
Dirnagl, U., Iadecola, C., Moskowitz, M., 1999. Pathobiology of ischaemic stroke: an
Jin: Conceptualization, Writing - original draft. Katharina Hein: integrated view. Trends Neurosci. 22, 391–397. https://doi.org/10.1016/S0166-
Visualization, Resources. Martin S. Weber: Investigation, 2236(99)01401-0.

12
M. Haupt et al. Neuropharmacology 181 (2020) 108357

Doeppner, T.R., Kaltwasser, B., Elali, A., Zechariah, A., Hermann, D.M., Bähr, M., 2011. Li, P., Gan, Y., Sun, B.-L., Zhang, F., Lu, B., Gao, Y., Liang, W., Thomson, A.W., Chen, J.,
Acute hepatocyte growth factor treatment induces long-term neuroprotection and Hu, X., 2013. Adoptive regulatory T-cell therapy protects against cerebral ischemia.
stroke recovery via mechanisms involving neural precursor cell proliferation and Ann. Neurol. 74, 458–471. https://doi.org/10.1002/ana.23815.
differentiation. J. Cerebr. Blood Flow Metabol. 31, 1251–1262. https://doi.org/ Li, X., Bijur, G., Jope, R.S, 2007. Glycogen synthas kinase-3beta, mood stabilizers, and
10.1038/jcbfm.2010.211. neuroprotection 4, 137–144.
Doeppner, T.R., Kaltwasser, B., Sanchez-Mendoza, E.H., Caglayan, A.B., Bähr, M., Liebeskind, DS., Jüttler, E., Shapovalov, Y., Yegin, A., Landen, J., Jauch, EC., 2019.
Hermann, D.M., 2017. Lithium-induced neuroprotection in stroke involves increased Cerebral edema associated with large hemispheric infarction. Stroke 50, 2619–2625.
miR-124 expression, reduced RE1-silencing transcription factor abundance and https://doi.org/10.1161/STROKEAHA.118.024766.
decreased protein deubiquitination by GSK3β inhibition-independent pathways. Liu, W., Wang, P., Shang, C., Chen, L., Cai, H., Ma, J., Yao, Y., Shang, X., Xue, Y., 2014.
J. Cerebr. Blood Flow Metabol. 37, 914–926. https://doi.org/10.1177/ Endophilin-1 regulates blood–brain barrier permeability by controlling ZO-1 and
0271678X16647738. occludin expression via the EGFR–ERK1/2 pathway. Brain Res. 1573, 17–26.
Fan, M., Song, C., Wang, T., Li, L., Dong, Y., Jin, W., Lu, P., 2015. Protective effects of https://doi.org/10.1016/j.brainres.2014.05.022.
lithium chloride treatment on repeated cerebral ischemia–reperfusion injury in mice. Ludewig, P., Sedlacik, J., Gelderblom, M., Bernreuther, C., Korkusuz, Y., Wagener, C.,
Neurol. Sci. 36, 315–321. https://doi.org/10.1007/s10072-014-1943-x. Gerloff, C., Fiehler, J., Magnus, T., Horst, A.K., 2013. Carcinoembryonic antigen-
Garcia-Bonilla, L., Moore, J.M., Racchumi, G., Zhou, P., Butler, J.M., Iadecola, C., related cell adhesion molecule 1 inhibits MMP-9-mediated blood-brain-barrier
Anrather, J., 2014. Inducible nitric oxide synthase in neutrophils and endothelium breakdown in a mouse model for ischemic stroke. Circ. Res. 113, 1013–1022.
contributes to ischemic brain injury in mice. J. Immunol. 193, 2531–2537. https:// https://doi.org/10.1161/CIRCRESAHA.113.301207.
doi.org/10.4049/jimmunol.1400918. Manji, H.K., Lenox, R.H., 1998. Lithium: a molecular transducer of mood-stabilization in
Gasche, Y., Fujimura, M., Morita-Fujimura, Y., Copin, J.-C., Kawase, M., Massengale, J., the treatment of bipolar disorder. Neuropsychopharmacology 19, 161–166. https://
Chan, P.H., 1999. Early appearance of activated matrix metalloproteinase-9 after doi.org/10.1016/S0893-133X(98)00021-9.
focal cerebral ischemia in mice: a possible role in blood—brain barrier dysfunction. Maurer, U., Preiss, F., Brauns-Schubert, P., Schlicher, L., Charvet, C., 2014. GSK-3-at the
J. Cerebr. Blood Flow Metabol. 19, 1020–1028. https://doi.org/10.1097/00004647- crossroads of cell death and survival. J. Cell Sci. 127, 1369–1378. https://doi.org/
199909000-00010. 10.1242/jcs.138057.
Gu, Y., Dee, C.M., Shen, J., 2011. Interaction of free radicals, matrix metalloproteinases Medsker, B., Forno, E., Simhan, H., Juan, C., Sciences, R., 2016. Matrix
and caveolin-1 impacts blood-brain barrier permeability. In: Biosci (Schol (Ed.), metalloproteinases as therapeutic targets for stroke 70, 773–779. https://doi.org/
Front 3, 1216–1231. 10.1097/OGX.0000000000000256.Prenatal.
Gu, Y., Zheng, G., Xu, M., Li, Y., Chen, X., Zhu, W., Tong, Y., Chung, S.K., Liu, K.J., Murozono, M., Matsumoto, S., Okada, S., Nagaoka, D., Isshiki, A., Watanabe, Y., 2009.
Shen, J., 2012. Caveolin-1 regulates nitric oxide-mediated matrix metalloproteinases Reduction of brain infarction induced by a transient brain ischemia in mdr1a
activity and blood-brain barrier permeability in focal cerebral ischemia and knockout mice. Neurochem. Res. 34, 1555–1561. https://doi.org/10.1007/s11064-
reperfusion injury. J. Neurochem. 120, 147–156. https://doi.org/10.1111/j.1471- 009-9943-6.
4159.2011.07542.x. Noll, T., Muhs, A., Besselmann, M., Watanabe, H., Piper, H.M., 1995. Initiation of
Herz, J., Sabellek, P., Lane, T.E., Gunzer, M., Hermann, D.M., Doeppner, T.R., 2015. Role hyperpermeability in energy-depleted coronary endothelial monolayers. Am. J.
of neutrophils in exacerbation of brain injury after focal cerebral ischemia in Physiol. Circ. Physiol. 268, H1462–H1470. https://doi.org/10.1152/
hyperlipidemic mice. Stroke 46, 2916–2925. https://doi.org/10.1161/ ajpheart.1995.268.4.H1462.
STROKEAHA.115.010620. Park, K.P., Rosell, A., Foerch, C., Xing, C., Kim, W.J., Lee, S., Opdenakker, G., Furie, K.L.,
Hurn, P.D., Subramanian, S., Parker, S.M., Afentoulis, M.E., Kaler, L.J., Vandenbark, A. Lo, E.H., 2009. Plasma and brain matrix metalloproteinase-9 after acute focal
A., Offner, H., 2007. T- and B-cell-deficient mice with experimental stroke have cerebral ischemia in rats. Stroke 40, 2836–2842. https://doi.org/10.1161/
reduced lesion size and inflammation. J. Cerebr. Blood Flow Metabol. 27, STROKEAHA.109.554824.
1798–1805. https://doi.org/10.1038/sj.jcbfm.9600482. Pösel, C., Möller, K., Boltze, J., Wagner, D.C., Weise, G., 2016. Isolation and flow
Jiang, X., Andjelkovic, A.V., Zhu, L., Yang, T., Bennett, M.V.L., Chen, J., Keep, R.F., cytometric analysis of immune cells from the ischemic mouse brain. J. Vis. Exp 2016.
Shi, Y., 2018. Blood-brain barrier dysfunction and recovery after ischemic stroke. https://doi.org/10.3791/53658.
Prog. Neurobiol. https://doi.org/10.1016/j.pneurobio.2017.10.001. Ramos, A.J., Lazarowski, A., Villar, M.J., Brusco, A., 2004. Transient expression of MDR-
Jiang, X., Namura, S., Nagata, I., 2001. Matrix metalloproteinase inhibitor KB-R7785 1/P-Glycoprotein in a model of partial cortical devascularization. Cell. Mol.
attenuates brain damage resulting from permanent focal cerebral ischemia in mice. Neurobiol. 24, 101–107. https://doi.org/10.1023/B: CEMN.0000012728.19117.73.
Neurosci. Lett. 305, 41–44. Ren, M., Senatorov, V.V., Chen, R.-W., Chuang, D.-M., 2003. Postinsult treatment with
Jiao, H., Wang, Z., Liu, Y., Wang, P., Xue, Y., 2011. Specific role of tight junction proteins lithium reduces brain damage and facilitates neurological recovery in a rat
claudin-5, occludin, and ZO-1 of the blood-brain barrier in a focal cerebral ischemic ischemia/reperfusion model. Proc. Natl. Acad. Sci. 100, 6210–6215. https://doi.org/
insult. J. Mol. Neurosci. 44, 130–139. https://doi.org/10.1007/s12031-011-9496-4. 10.1073/pnas.0937423100.
Johnstone, R.W., Cretney, E., Smyth, M.J., 1999. P-glycoprotein protects leukemia cells Robinson, L.J., Roberts, W.K., Ling, T.T., Lamming, D., Sternberg, S.S., Roepe, P.D.,
against caspase-dependent, but not caspase-independent, cell death. Blood 93, 1997. Human MDR 1 protein overexpression delays the apoptotic cascade in Chinese
1075–1085. https://doi.org/10.1182/blood.v93.3.1075.403k35_1075_1085. hamster ovary fibroblasts. Biochemistry 36, 11169–11178. https://doi.org/
Justicia, C., Martín, A., Rojas, S., Gironella, M., Cervera, Á., Panés, J., Chamorro, Á., 10.1021/bi9627830.
Planas, A.M., 2006. Anti-VCAM-1 antibodies did not protect against ischemic Rosell, A., Cuadrado, E., Ortega-Aznar, A., Hernández-Guillamon, M., Lo, E.H.,
damage either in rats or in mice. J. Cerebr. Blood Flow Metabol. 26, 421–432. Montaner, J., 2008. MMP-9–positive neutrophil infiltration is associated to
https://doi.org/10.1038/sj.jcbfm.9600198. blood–brain barrier breakdown and basal lamina type IV collagen degradation
Kitagawa, K., Matsumoto, M., Mabuchi, T., Yagita, Y., Ohtsuki, T., Hori, M., during hemorrhagic transformation after human ischemic stroke. Stroke 39,
Yanagihara, T., 1998. Deficiency of intercellular adhesion molecule 1 attenuates 1121–1126. https://doi.org/10.1161/STROKEAHA.107.500868.
microcirculatory disturbance and infarction size in focal cerebral ischemia. Sawe, N., Steinberg, G., Zhao, H., 2008. Dual roles of the MAPK/ERK1/2 cell signaling
J. Cerebr. Blood Flow Metabol. 18, 1336–1345. https://doi.org/10.1097/00004647- pathway after stroke. J. Neurosci. Res. https://doi.org/10.1002/jnr.21604.
199812000-00008. Schäfer, M., Bahde, D., Bosche, B., Ladilov, Y., Schäfer, C., Piper, H.M., Noll, T., 2001.
Kleinschnitz, C., Schwab, N., Kraft, P., Hagedorn, I., Dreykluft, A., Schwarz, T., Modulation of early [Ca2+]i rise in metabolically inhibited endothelial cells by
Austinat, M., Nieswandt, B., Wiendl, H., Stoll, G., 2010. Early detrimental T-cell xestospongin C. Am. J. Physiol. - hear. Circ. Physiol. 280 https://doi.org/10.1152/
effects in experimental cerebral ischemia are neither related to adaptive immunity ajpheart.2001.280.3.h1002.
nor thrombus formation. Blood 115, 3835–3842. https://doi.org/10.1182/blood- Schinkel, A.H., 1999. P-Glycoprotein, a gatekeeper in the blood-brain barrier. Adv. Drug
2009-10-249078. Deliv. Rev. https://doi.org/10.1016/S0169-409X(98)00085-4.
Kraya, R., Komin, A., Searson, P., 2016. On chip bioelectric impedance spectroscopy Silachev, D.N., Plotnikov, E.Y., Babenko, V.A., Savchenko, E.S., Zorova, L.D., Pevzner, I.
reveals the effect of P-Glycoprotein efflux üumps on the paracellular impedance of B., Gulyaev, M.V., Pirogov, Y.A., Sukhikh, G.T., Zorov, D.B., 2016. Protection of
tight junctions at the blood–brain barrier. IEEE Trans. NanoBioscience 15, 697–703. neurovascular unit cells with lithium chloride and sodium valproate prevents brain
https://doi.org/10.1109/TNB.2016.2604322. damage in neonatal ischemia/hypoxia. Bull. Exp. Biol. Med. 160, 313–318. https://
Krueger, M., Bechmann, I., Immig, K., Reichenbach, A., Härtig, W., Michalski, D., 2015. doi.org/10.1007/s10517-016-3159-y.
Blood-brain barrier breakdown involves four distinct stages of vascular damage in Spudich, A., Kilic, E., Xing, H., Kilic, Ü., Rentsch, K.M., Wunderli-Allenspach, H.,
various models of experimental focal cerebral ischemia. J. Cerebr. Blood Flow Bassetti, C.L., Hermann, D.M., 2006. Inhibition of multidrug resistance transporter-1
Metabol. 35, 292–303. https://doi.org/10.1038/jcbfm.2014.199. facilitates neuroprotective therapies after focal cerebral ischemia. Nat. Neurosci. 9,
Kurzepa, Jacek, Kurzepa, Joanna, Golab, P., Czerska, S., Bielewicz, J., 2014. The 487–488. https://doi.org/10.1038/nn1676.
significance of matrix metalloproteinase (MMP)-2 and MMP-9 in the ischemic stroke. Vemuganti, R., Dempsey, R.J., Bowen, K.K., 2004. Inhibition of intercellular adhesion
Int. J. Neurosci. 124, 707–716. https://doi.org/10.3109/00207454.2013.872102. molecule-1 protein expression by antisense oligonucleotides is neuroprotective after
Lazarowski, A., Caltana, L., Merelli, A., Rubio, M.D., Ramos, A.J., Brusco, A., 2007. transient middle cerebral artery occlusion in rat. Stroke 35, 179–184. https://doi.
Neuronal mdr-1 gene expression after experimental focal hypoxia: a new obstacle for org/10.1161/01.STR.0000106479.53235.3E.
neuroprotection? J. Neurol. Sci. 258, 84–92. https://doi.org/10.1016/j. Weise, J., Sandau, R., Schwarting, S., Crome, O., Wrede, A., Schulz-Schaeffer, W., Zerr, I.,
jns.2007.03.004. Bähr, M., 2006. Deletion of cellular prion protein results in reduced Akt activation,
Li, H., Gao, A., Feng, D., Wang, Y., Zhang, L., Cui, Y., Li, B., Wang, Z., Chen, G., 2014. enhanced postischemic caspase-3 activation, and exacerbation of ischemic brain
Evaluation of the protective potential of brain microvascular endothelial cell injury. Stroke 37, 1296–1300. https://doi.org/10.1161/01.STR.0000217262.03192.
autophagy on blood-brain barrier integrity during experimental cerebral ischemia- d4.
reperfusion injury. Transl. Stroke Res. 5, 618–626. https://doi.org/10.1007/s12975- Wijdicks, E.F.M., Sheth, K.N., Carter, B.S., Greer, D.M., Kasner, S.E., Kimberly, W.T.,
014-0354-x. Schwab, S., Smith, E.E., Tamargo, R.J., Wintermark, M., American Heart Association
Stroke Council, 2014. Recommendations for the management of cerebral and

13
M. Haupt et al. Neuropharmacology 181 (2020) 108357

cerebellar infarction with swelling: a statement for healthcare professionals from the Yanamadala, V., Friedlander, R.M., 2010. Complement in neuroprotection and
American Heart Association/American Stroke Association. Stroke 45, 1222–1238. neurodegeneration. Trends Mol. Med. 16, 69–76. https://doi.org/10.1016/j.
https://doi.org/10.1161/01.str.0000441965.15164.d6. molmed.2009.12.001.
Xiong, X.-Y., Liu, L., Yang, Q.-W., 2016. Functions and mechanisms of microglia/ Yang, Y., Estrada, E.Y., Thompson, J.F., Liu, W., Rosenberg, G.A., 2007. Matrix
macrophages in neuroinflammation and neurogenesis after stroke. Prog. Neurobiol. metalloproteinase-mediated disruption of tight junction proteins in cerebral vessels
142, 23–44. https://doi.org/10.1016/j.pneurobio.2016.05.001. is reversed by synthetic matrix metalloproteinase inhibitor in focal ischemia in rat.
Xu, J., Culman, J., Blume, A., Brecht, S., Gohlke, P., 2003. Chronic treatment with a low J. Cerebr. Blood Flow Metabol. 27, 697–709. https://doi.org/10.1038/sj.
dose of lithium protects the brain against ischemic injury by reducing apoptotic jcbfm.9600375.
death. Stroke 34, 1287–1292. https://doi.org/10.1161/01. Yu, F., Wang, Z., Tchantchou, F., Chiu, C.-T., Zhang, Y., Chuang, D.-M., 2011. Lithium
STR.0000066308.25088.64. ameliorates neurodegeneration, suppresses neuroinflammation, and improves
Yadav, B.K., Shin, B.-S., 2015. Single-nucleotide polymorphisms of tight junction behavioral performance in a mouse model of traumatic brain injury. J. Neurotrauma
component claudin-1 associated with leukoaraiosis. J. Stroke Cerebrovasc. Dis. 24, 29, 362–374. https://doi.org/10.1089/neu.2011.1942.
1662–1670. https://doi.org/10.1016/j.jstrokecerebrovasdis.2015.03.038. Zhang, J.W., Gottschall, P.E., 1997. Zymographic measurement of gelatinase activity in
brain tissue after detergent extraction and affinity-support purification. J. Neurosci.
Methods 76, 15–20. https://doi.org/10.1016/S0165-0270(97)00065-4.

14

You might also like