You are on page 1of 7

Journal of Cerebral Blood Flow & Metabolism (2013) 33, 19761982

& 2013 ISCBFM All rights reserved 0271-678X/13 $32.00


www.jcbfm.com

ORIGINAL ARTICLE
Inhibition of microRNA-181 reduces forebrain ischemia-induced
neuronal loss
Jeong-mi Moon1,2, Lijun Xu1 and Rona G Giffard1

MicroRNA (miRNA), miR-181a, is enriched in the brain, and inhibition of miR-181a reduced astrocyte death in vitro and infarct
volume after stroke in vivo. This study investigated the role of miR-181a in neuronal injury in vitro and hippocampal neuronal loss
in vivo after forebrain ischemia. miR-181a levels were altered by transfection with mimic or antagomir. N2a cells subjected to serum
deprivation and oxidative stress showed less cell death when miR-181a was reduced and increased death when miR-181a
increased; protection was associated with increased Bcl-2 protein. In contrast, transfected primary neurons did not show altered
levels of cell death when miR-181a levels changed. Naive male rats and rats stereotactically infused with miR-181a antagomir or
control were subjected to forebrain ischemia and cornus ammonis (CA)1 neuronal survival and protein levels were assessed.
Forebrain ischemia increased miR-181a expression and decreased Bcl-2 protein in the hippocampal CA1 region. miR-181a
antagomir reduced miR-181a levels, reduced CA1 neuronal loss, increased Bcl-2 protein, and signicantly prevented the decrease of
glutamate transporter 1. Thus, miR-181a antagomir reduced evidence of astrocyte dysfunction and increased CA1 neuronal survival.
miR-181a inhibition is thus a potential target in the setting of forebrain or global cerebral ischemia as well as focal ischemia.

Journal of Cerebral Blood Flow & Metabolism (2013) 33, 19761982; doi:10.1038/jcbfm.2013.157; published online 4 September 2013
Keywords: apoptosis; astrocytes; brain ischemia; global ischemia; glutamate; hippocampus

INTRODUCTION miR-181a, a miRNA that is highly conserved across most


Several clinical situations such as cardiac arrest and resuscitation vertebrates, is enriched in the brain15,16 and increases during
or severe hypotension can lead to transient global cerebral maturation of hippocampal neurons.17 The overexpression of miR-
ischemia. Global ischemia causes selective, delayed death 181a induces drug addiction-related synaptic changes in the
of pyramidal neurons in the hippocampal cornus ammonis hippocampus17 and increases astrocyte death because of glucose
(CA)1 while sparing the neighboring dentate gyrus in rodents1 deprivation.18 miR-181a is upregulated in the infarct core and
and humans, resulting in persistent severe memory decits.2,3 downregulated in the penumbra after focal ischemia14,17 but it has
However, despite increasing knowledge of the pathologic not yet been studied in forebrain ischemia. In the present study,
mechanisms of global ischemia, the only known clinically we test the effect of reducing miR-181a on forebrain ischemia,
effective treatment is hypothermia.4,5 Neuronal death in CA1 and, because of the importance of apoptosis in this setting,
is not histologically detectable until 3 to 4 days after global/ characterize the effects on levels of Bcl-2, a target of miR-181a. We
forebrain ischemia, and this signicant delay between injury also examined the effect of miR-181a in N2a cells and primary
and the onset of neuronal death holds promise for the neurons subjected to stress.
development of future therapies, including ones targeting
apoptosis.68
MicroRNAs (miRNAs) are small regulatory noncoding RNAs MATERIALS AND METHODS
B22 bp in length that modulate protein synthesis by post- Cell Culture and Transfection
transcriptional silencing of gene expression via the recognition of Mouse neuroblastoma (N2a) cells (a kind gift from Drs Kurt Lucin and Tony
specic sequences in target messenger RNAs (mRNAs). Many Wyss-Coray at Stanford University) were grown in high-glucose Dulbeccos
miRNAs are highly expressed in the adult nervous system in a modied Eagles medium (Invitrogen, Carlsbad, CA, USA) supplemented
regulated manner, and pathologic conditions including focal and with 8% fetal bovine serum (Hyclone, Logan, UT, USA) and 1% antibiotics
global ischemia lead to altered regulation of brain miRNAs.911 (50 U/mL penicillin and 50 mg/mL streptomycin; Invitrogen) in a humidied
These reports suggested that miRNAs could have a critical role in atmosphere containing 5% CO2 at 37 1C. N2a cells were transfected with
molecular signaling associated with ischemic pathologic events. control miRNA, miR-181a mimic, or miR-181a inhibitor (Thermo Scientic
Dharmacon, Chicago, IL, USA) using Lipofectamine 2000 (Invitrogen)
Indeed, several laboratories have demonstrated that manipulation
according to the manufacturers protocol.
of miRNAs can attenuate infarct volume in a focal ischemia Primary mouse neuronal cultures were prepared from 16-day-old
model.1214 However, in contrast to focal ischemia, it still remains embryos as previously described19 in accordance with a protocol
unknown whether modulation of miRNAs can protect against approved by the Stanford University Animal Care and Use Committee
global ischemia. and after the NIH Guide for the Care and Use of Laboratory Animals. Isolated

1
Department of Anesthesia, Stanford University School of Medicine, Stanford, California, USA and 2Department of Emergency Medicine, Chonnam National University School of
Medicine, Gwangju, South Korea. Correspondence: Dr RG Giffard, Stanford University School of Medicine, 300 Pasteur Drive, Grant Building S272A, Stanford, CA 94305-5117, USA.
E-mail: rona.giffard@stanford.edu
This work was supported in part by NIH Grants NS053898 and GM49831 to RGG.
Received 18 March 2013; revised 31 July 2013; accepted 2 August 2013; published online 4 September 2013
MiR-181 reduction protects from forebrain ischemia
J-m Moon et al
1977
cortical neurons were seeded on poly-D-lysine (Sigma, St Louis, MO, USA) temperature was 370.5 1C controlled by a homeothermic blanket
and laminin (Sigma)-coated 24- or 6-well plates in minimal essential (Harvard Apparatus, Holliston, MA, USA). Respiratory rate, oxygen
medium supplemented with 5% horse serum (Hyclone), 5% fetal bovine saturation, and heart rate were monitored with a small animal oximeter
serum (Hyclone), and 2 mM of L-glutamine (Sigma). On day 2, 60% of the (STARR Life Sciences, Allison Park, PA, USA). After forebrain ischemia, rats
medium was replaced by glial-conditioned medium supplemented with were killed at 5 or 24 hours after ischemia for protein and RNA isolation.
3% B-27 serum-free supplement (Invitrogen) and 3 mM cytosine Control rats injected with antagomir or mismatch but not subjected to
arabinoside. On day 4, primary neurons were transfected using Effectene ischemia were killed 1 and 3 days after injection to assess protein and
(Qiagen, Valencia, CA, USA) according to the manufacturers protocol and miRNA levels. For assessment of CA1 neuronal survival by cresyl violet
on day 5, they were exposed to injury. staining or assessment of protein by immunoblotting, rats were killed at 7
days after ischemia under deep anesthesia with isourane. Brains were
collected after perfusion with cold saline for protein or RNA, or saline
Injury Paradigms followed by ice-cold 4% phosphate-buffered paraformaldehyde for cresyl
Both N2a cells and primary neurons were subjected to injury 24 hours after violet staining. After 48 hours after xation in 4% paraformaldehyde, 50 mm
transfection. coronal sections were made with a vibratome (Leica VT1000S, Heidelberg,
N2a cells were exposed to 900 mM of H2O2 in Dulbeccos modied Eagles Germany). The investigator making the injections was masked to the
medium supplemented with 1% antibiotics for 24 hours. Primary neurons treatment and the surgeon performing forebrain ischemia was masked to
were deprived of serum for 48 hours by changing medium to a balanced the treatment group.
salt solution containing 5.5 mM glucose at pH 7.4, 116 mM NaCl, 1.8 mM
CaCl2, 0.8 mM MgSO4, 5.4 mM KCl, 1 mM NaH2PO4, 14.7 mM NaHCO3, 10 mM
N-(2-hydroxyethyl) piperazine-N-ethanesulfonic acid, and phenol red 10 mg. Assessment of CA1 Hippocampal Injury
Coronal sections were mounted on gelatin-coated slides and then
Quantitation of Cell Death incubated in 1% cresyl violet acetate (Sigma) for 5 minutes followed by
The viability of N2a cells was quantied by measuring the concentration of dehydration. The density of cresyl violet staining was determined in a xed
intracellular lactate dehydrogenase (with LDH kit from Sigma) released area of CA1 on each micrograph using the histogram function of Adobe
from dead cells into the media.20 The results were expressed as the Photoshop CS3 and normalized to sham by a researcher masked to
percentage of lactate dehydrogenase release compared with lactate treatment, as previously described.21
dehydrogenase release after freeze thaw to disrupt all cells.
The viability of primary neurons was assessed by cell counting after Immunoblotting
Hoechst 33342 (5 mmol/L; Sigma) and propidium iodide (5 mmol/L; Sigma)
staining. Hoechst stains all cells blue; propidium iodide stains only dead Immunoblotting was performed as previously described.22 Homogenates
cells red. Eight nonoverlapping uorescence images per well were prepared from either cell cultures or subdissected CA1 and dentate gyrus
acquired at  20 magnication with a Zeiss Axiovert 200M and Axiovision regions of the hippocampus from brain were studied. Briey, 20 to 40 mg of
software (Carl Zeiss Microscopy, Thornwood, NY, USA). The percentage of protein were separated on a 4 to 12% polyacrylamide gel (Invitrogen) and
dead cells was calculated using NIH Image J (US National Institutes of electrotransferred to an Immobilon polyvinylidene uoride membrane
Health, Bethesda, MD, USA). (Millipore, Billerica, MA, USA). Membranes were blocked in phosphate-
buffered saline containing 0.1% tween 20 and 5% non-fat milk for 1 hour
at room temperature, and then incubated overnight with primary antibody
Imaging-Activated Caspases
N2a cells were stained with Magic Red (Immunochemistry, Bloomington,
MN, USA) to identify cells with activated caspases 3 and 7 and
counterstained with Hoechst 33342 (5 mmol/L; Sigma) after 4 hours of
exposure to injury. The uorescence images were taken at  20
magnication. Caspase activity was calculated by dividing the uorescence
intensity of Magic Red by total cell number in each eld using Adobe
Photoshop CS3, and then normalized to the uorescence per cell for
normal N2a cells without injury.

Animal Groups
Male SpragueDawley rats (Charles River, Wilmington, MA, USA) weighing
280 to 320 g were divided into the following groups: sham (n 8),
forebrain ischemia without treatment (n 6), ischemia with miR-181a
antagomir (n 29), ischemia with mismatch miR-181a antagomir (n 29),
ischemia with vehicle injection (n 3), injection of miR-181a antagomir,
and mismatch antagomir without ischemia (n 11, 11).

Transfection of Rat Hippocampus In Vivo


Rats were anesthetized with isourane and placed in a stereotaxic frame
with a rat head holder. Three picomoles per gram of miR-181a antagomir
or mismatch miR-81a antagomir (Thermo Scientic Dharmacon), mixed
with the cationic lipid DOTAP 1:3 (Roche Applied Science, San Francisco,
CA, USA), were infused stereotactically just outside the left hippocampus
(from bregma  3.8 mm, M-L 2.0 mm, deep 2.5 mm) at 1 mL/minute,
maximal total volume 16 mL via a burr hole.21 After injection the bone
wound was closed with bone wax.

Transient Forebrain Ischemia Figure 1. The effect of mimic and inhibitor on miR-181a level and
Forebrain ischemia was induced 24 hours after transfection22 according to Bcl-2 protein expression in N2a cells. (A) miR-181a levels after
an animal protocol approved by the Stanford University Animal Care and transfection with 10 pmol of miR-181a mimic (Mimic) and 20 pmol of
Use Committee. Briey, hypotension (mean blood pressure o40 mm Hg) inhibitor (Inh) in N2a cells. n 6. (B) Effect of increased or decreased
was induced by removing blood into heparinized sterile tubing with miR-181a level on Bcl-2 protein expression, normalized to the
continuous femoral arterial blood pressure monitoring (PuritanBennett transfection control (Ctrl). n 8 to 11. Inset shows representative
Corporation, Wilmington, MA, USA). Both carotid arteries were clamped for western blots. *Po0.05, **Po0.01, ***Po0.001 compared with the
10 minutes, then unclamped and the shed blood reinfused. Rectal transfection control.

& 2013 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2013), 1976 1982
MiR-181 reduction protects from forebrain ischemia
J-m Moon et al
1978

Figure 2. Decreasing miR-181a levels reduces serum deprivation and oxidative stress-induced N2a cell death and caspases 3 and 7 activation.
(A) Cells were deprived of serum and incubated in the presence of added H2O2 for 24 hours, then cell death was assessed by lactate
dehydrogenase release. The extent of cell death in all injury conditions was significantly different from wash control (Wash). n 12. (Mean cell
death valuess.e. were Wash 9.00.71, Trans 33.01.04, Mimic 47.51.14, Inh 28.50.70.) (B, C) Caspase 3 and 7 activity after exposure to
injury for 4 hours was determined by Magic Red staining and normalized to wash control. Cultures were stained for activated caspases 3 and 7
(red) and nuclei were counterstained with Hoechst dye (blue) after exposure to injury for 4 hours. n 18. SD, serum deprivation;
Mimic 10 pmol of miR-181a mimic; Inh 20 pmol of miR-181a inhibitor. *Po0.05, **Po0.01, ***Po0.001 compared with transfection control
injury. Bar is 50 mm.

against Bcl-2 (1:1000, Cell Signaling, Danvers, MA, USA), glutamate t-test if only two conditions, or one-way analysis of variance with the
transporter 1 (GLT-1; 1 : 500, Millipore, Temecula, CA, USA), and actin Bonferroni post-test. Po0.05 was considered signicant.
(1 : 5000, Sigma). After washing, membranes were incubated with
secondary antibody for 1 hour. Immunoreactive bands were visualized
using the LI-COR Odyssey infrared imaging system (LI-COR Biosciences,
Lincoln, NE, USA). Densitometric analysis of bands was performed using RESULTS
NIH image J and normalized to actin.
Altering miR-181a Levels Alters Bcl-2 Levels and Apoptotic Cell
Death of N2a cells
Real-Time Quantitative PCR To investigate the effect of altering miR-181a levels on N2a cells,
cells were transfected with miR-181a mimic, miR-181a inhibitor, or
Total RNA was extracted with TRIzol reagent (Invitrogen), and the
concentration and purity of the RNA samples were quantied by
control. Treatment with mimic increased miR-181a levels 4,700-
absorbance at 260 and 280 nm. Reverse transcription of miRNA was fold, whereas a decrease of 88% was seen with the inhibitor
performed using the TaqMan MicroRNA Reverse Transcription Kit (Applied (Figure 1A). As we previously found that Bcl-2 was a target of miR-
Biosystems, Carlsbad, CA, USA). Equal amounts of RNA (200 ng) were 181a in astrocytes, we therefore determined whether changing
reverse transcribed with 1.3 mM dNTPs (with dTTP), 50 U reverse levels of miR-181a would affect Bcl-2 levels in N2a cells. miR-181a
transcriptase, 10 U RNase inhibitor, and specic miRNA reverse trans- inhibitor treatment signicantly increased Bcl-2 protein levels,
criptase primers (Applied Biosystems) at 16 1C for 30 minutes, 42 1C for whereas mimic-treated cells had decreased levels relative to
30 minutes, and 851C for 5 minutes. PCR reactions were carried out using control (Figure 1B).
the TaqMan MicroRNA Assay Kit (Applied Biosystems) at 95 1C for N2a cells were next exposed to combined serum deprivation
10 minutes, followed by 40 cycles of 95 1C for 15 seconds and 60 1C for
1 minute. Predesigned primer/probes for miR-181a and mouse U6 were
and oxidative stress for 24 hours to induce apoptosis. Over-
also from Applied Biosystems. Relative change of miR-181a was calculated expression of miR-181a signicantly increased cell death, whereas
using the 2  DDCT method.23 Normalization was rst to U6, which has inhibitor effectively reduced cell death (Figure 2A). There was no
been shown to be stable under ischemic brain conditions11 and then to signicant difference in cell death between injury control without
control to give the relative fold change compared with the control (data not shown) vs with treatment with transfection control
sample. (Trans). To conrm that cell death was apoptotic, we stained the
N2a cells with Magic Red to detect active caspases 3 and 7.
Increasing miR-181a by treatment with mimic was associated with
Statistical Analysis many more cells showing activation of caspases 3 and 7, whereas
All cell culture data represent at least three independent experiments. All reduction of miR-181a by inhibitor treatment reduced the number
data reported are meanss.e.m. Statistical analysis was performed using of cells activating these enzymes (Figures 2B and C).

Journal of Cerebral Blood Flow & Metabolism (2013), 1976 1982 & 2013 ISCBFM
MiR-181 reduction protects from forebrain ischemia
J-m Moon et al
1979
Table 1. Levels of miR-181a and Bcl-2 protein in the dentate gyrus
region of hippocampus after forebrain ischemia

Reperfusion

Sham 5 hours 24 hours

miR-181a levels 1.020.14 0.520.06* 0.750.20


Bcl-2 protein 0.970.03 1.270.05** 1.290.04**
Values are means.e.m. n 3 for miR-181a levels, n 3 for Bcl-2 protein
levels. *P 0.054 for miR-181a level at 5 hours reperfusion compared with
sham. **Po0.01 compared with sham.

threshold cycle for U6 between the two cell types, but the
threshold cycle for miR-181a was signicantly lower in primary
neurons (20.090.03 vs 25.920.28 for N2a cells; n 5),
indicating greater abundance of miR-181a in primary neurons.
Transfection of primary neuronal cultures with miR-181a mimic or
inhibitor led to a dose-dependent change in miR-181a levels
(Figure 3A). Despite this, when transfected neurons were exposed
to serum deprivation, no change in the extent of injury was
observed (Figure 3B). After transfection but without injury, levels
of Bcl-2 protein were unaltered despite changes in miR-181a levels
(data not shown), in contrast to the results with N2a cells.

Forebrain Ischemia Changes miR-181a and Bcl-2 Protein Levels in


CA1 and Dentate Gyrus in the Hippocampus
CA1 is the subregion of the hippocampus most susceptible to
Figure 3. Altering miR-181a levels in primary neurons does not alter ischemia. We assessed levels of miR-181a 5 and 24 hours after
serum deprivation cell death. (A) miR-181a levels in primary neurons 10 minutes of forebrain ischemia. In CA1, miR-181a signicantly
after transfection with 10 or 50 pmol of miR-181a mimic, and 20 or increased 24 hours after ischemia (Figure 4A), whereas Bcl-2
40 pmol of miR-181a inhibitor. n 9. (B) Serum deprivation induced protein decreased (Figure 4B). In contrast, in the dentate gyrus
primary neuronal death assessed by cell counting. All injured groups miR-181a tended to decrease at 5 hours, whereas Bcl-2 protein
had significantly greater injury than wash control. There was no was increased at both 5 and 24 hours (Table 1).
significant difference in serum deprivation-induced cell death
between control transfected cultures (Trans) and not transfected
cultures (data not shown). n 10. *Po0.05, ***Po0.001 compared Reducing Levels of miR-181a Reduces Forebrain Ischemic Injury
with the transfection control.
Real-time quantitative PCR was performed 1 and 3 days after
injection to determine the extent to which miR-181a levels changed
after transfection with antagomir by stereotactic injection. miR-181a
antagomir, but not mismatched antagomir, signicantly reduced
miR-181a expression 1 and 3 days after a single treatment
(Figure 5A). Bcl-2 protein levels also changed with a signicant
increase on day 1 and a trend to increase on day 3 (Figures 5B and
5C). Therefore, we performed forebrain ischemia on rats 1 day after
stereotactic infusion of antagomir or control. Cresyl violet staining
performed 7 days after forebrain ischemia revealed that when
brains were transfected with miR-181a antagomir CA1, neuronal cell
death was signicantly reduced compared with rats treated with
mismatch miR-181a antagomir (Figures 5D and 5E).

The Effect of miR-181a Antagomir on GLT-1 in CA1 After Forebrain


Ischemia.
Figure 4. Temporal change of miR-181a and Bcl-2 protein in the CA1
region of hippocampus after forebrain ischemia. (A) miR-181a levels As we previously found that astrocyte impairment was a critical
at 5 and 24 hours of reperfusion (Rep) after forebrain ischemia. determinant of the extent of subsequent neuronal cell death in
n 3. (B) Bcl-2 protein at 5 and 24 hours of reperfusion after forebrain ischemia,22 we hypothesized that miR-181 antagomir
forebrain ischemia. n 3. *Po0.05 compared with sham, not might reduce astrocyte impairment. Earlier studies showed that
subjected to ischemia. reducing miR-181a levels in primary astrocyte cultures could
reduce oxidative stress,18 and oxidative stress contributes to
astrocyte impairment and reduced GLT-1 levels after forebrain
ischemia. We therefore determined the level of GLT-1 in the CA1
Effect of Altering miR-181a Levels in Primary Neurons subeld by immunoblot analysis. GLT-1 expression signicantly
We compared the expression of miR-181a in primary neurons and decreased in CA1 24 hours after forebrain ischemia compared
N2a cells by real-time quantitative PCR. There was no difference in with sham; however, treatment with miR-181a antagomir

& 2013 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2013), 1976 1982
MiR-181 reduction protects from forebrain ischemia
J-m Moon et al
1980

Figure 5. Pretreatment with miR-181a antagomir increases CA1 neuronal survival after forebrain ischemia. (A) miR-181a levels 1 and 3
days after injecting miR-181a antagomir or mismatch miR-181a antagomir just outside the hippocampus relative to sham. n 5 to 6.
*Po0.05 and **Po0.01 compared with sham. (B) Representative blots of Bcl-2 protein expression after transfection with indicated antagomir
in vivo. (C) Levels of Bcl-2 protein 1 and 3 days after injecting miR-181a antagomir or mismatch miR-181a antagomir relative to sham. n 3 to
6. *Po0.05 compared with sham. (D) Cresyl violet staining of hippocampi. Sham, which was not subjected to ischemia, shows a normal
hippocampus. After forebrain ischemia in a rat pretreated with mismatch miR-181a antagomir, selective neuronal loss of CA1 hippocampal
neurons (between white arrows) and reduced neuronal loss (between white arrows) in a rat treated with miR-181a antagomir outside the left
hippocampus is shown. (E) Quantitation of neuronal survival in CA1 of rats treated with miR-181a antagomir (Ant) or mismatch miR-181a
antagomir (Mis) relative to sham. n 15. ***P o0.001, compared with rats treated with mismatch miR-181a antagomir.

signicantly inhibited this decrease of GLT-1 compared with


vehicle or mismatch control antagomir (Figure 6).

DISCUSSION
Pretreatment with miR-181a antagomir reduces loss of CA1
pyramidal neurons 7 days after forebrain ischemia. This adds
forebrain ischemia to the settings in which reducing miR-181
provides neuroprotection. Treatment with antagomir inhibited the
changes seen in untreated animals, reducing miR-181a, increasing
Bcl-2 protein levels, and preserving neurons in CA1. Although
pretreatment was tested in this study, future studies of the effect
of changing miRNA levels after the onset of ischemia will be an
essential step in further demonstrating the likely relevance for
clinical treatment.
Overexpression of Bcl-2, the prototype anti-apoptotic member
of the eponymous family of apoptosis regulatory proteins, was Figure 6. Pretreatment with miR-181a antagomir preserves GLT-1
previously shown to be protective against both focal and global expression after forebrain ischemia. GLT-1 expression 24 hours after
reperfusion in the CA1 subfield of rats treated with miR-181a
cerebral ischemia in vivo and brain cell injury in vitro.2427 antagomir, mismatch miR-181a antagomir, vehicle (Isch), and sham
However, gene therapy in the setting of cardiac arrest is less rats, which were not subjected to ischemia. Inset shows representa-
likely to translate to clinical application than manipulation of a tive western blot. n 3. *Po0.05 compared with both Isch and
miRNA in this setting. We previously demonstrated that Bcl-2 is a mismatch.

Journal of Cerebral Blood Flow & Metabolism (2013), 1976 1982 & 2013 ISCBFM
MiR-181 reduction protects from forebrain ischemia
J-m Moon et al
1981
direct target of miR-181 by luciferase assay.18 Trials of mimic and glutamate receptor agonists35 or by overexpressing protective
antagomir miRNAs are already underway for several diseases, and proteins selectively in astrocytes21,22 reduces neuron cell
as the chemistry is the same only the sequence will differ. Thus, death. In this study, signicant decreases in GLT-1 protein were
extending this approach to cerebral ischemia has strong demonstrated 24 hours after ischemia, consistent with other
translational potential. studies in gerbils and rats,22,36 and antagomir treatment reduced
Although we demonstrate changes in one target of miR-181a, this loss.
a limitation of our study is that there are likely to be additional Despite the observed changes in GLT-1, miR-181a does not
targets of miR-181a that may have a role in the protection we directly target GLT-1. Immunoblotting did not show an increase of
observe, because miRNAs target multiple mRNAs. Our prior work GLT-1 protein in rats pretreated with miR-181a antagomir (data
showed that miR-181a can directly target Mcl-1 and Grp78/BIP in not shown), and no potential binding sites were identied in the
some cells,14,18 and a recent report showed that X-linked inhibitor 3-untranslated region of GLT-1 mRNA for miR-181a using the
of apoptosis is also a direct target, and that several antioxidant miR-Target analysis database (http://www.targetscan.org, http://
enzymes are likely targets.28 These targets, which are all pro- www.microRNA.org). Thus, the protection is likely indirect
survival proteins and can reduce oxidative stress, could also have a secondary to reduced oxidative stress in astrocytes. Reduced
role here. We think this is relevant because our earlier studies oxidative stress may reect increased levels of Bcl-2, Grp78, and
demonstrated a link between oxidative stress in astrocytes, Mcl-114,18 and possibly glutathione peroxidases 1 and 4, and
reduction in GLT-1, and CA1 neuronal cell death.21,22 miR-181 peroxiredoxin 2.28
was also shown to sensitize glioblastoma cells to apoptosis by Astrocytes support neurons and other surrounding cells
reducing Bcl-2.29 Future studies are needed to explore additional through multiple functions including regulation of blood ow,
potential targets in this setting. provision of substrates, metabolic and ionic homeostasis, and
One interesting observation is the difference in effect of miR- antioxidant defense.33,37,38 However, after ischemia or other
181a antagomir in different cell types. Reducing miR-181a central nervous system insults, astrocytes become reactive and
increased Bcl-2 in N2A cells and increased Bcl-2, Grp78, and Mcl- may lose some supportive functions, thereby contributing to
1 in primary astrocytes while increasing survival of both these cell neuronal death.39 Ischemic injury induces activation of astrocytes,
types. It failed to signicantly change levels of Bcl-2 in primary early loss of GLT-1 in the CA1 region, with recovery over days after
neurons, and it did not change outcome from serum deprivation- forebrain ischemia. Findings in forebrain ischemia are consistent
induced neuronal cell death. This observation is consistent with an with reports using the four vessel occlusion model of global
increasing body of literature that suggests that individual miRNAs ischemia.40,41 Thus, benecial effects of miR-181a on astrocytes
may have modest effects on their target mRNAs, and that often may lead to reduced or modulated activation and better
several miRNAs are required for larger, or in some cases, preserved protective function. Effects of miR-181a antagomir in
detectable alteration of target protein levels. In prostate cancer multiple cell types are probably important to the nal extent of
cells, miR-181a acts cooperatively with two other miRNAs to protection observed.
regulate Grp78 protein levels,30 in contrast to the response of
astrocytes in which increasing miR-181a alone effectively
downregulates Grp78.14,30 Further, cell lines are in general SUMMARY
dependent on inducing anti-apoptotic pathways for survival; Stereotactic delivery of miR-181a antagomir just outside the
hence, the N2A cells may be more sensitive to the ability of miR- hippocampus reduced neuronal cell death in rat forebrain
181 to reduce several anti-apoptotic proteins than are primary, ischemia, associated with increased Bcl-2 and preserved GLT-1
postmitotic neurons. Thus, the efciency of regulation of a target levels, consistent with protection that includes preserving
protein depends on the cell type and likely the physiologic setting; astrocyte function.
hence, the difference observed between N2A cells and primary
neurons may not be surprising.
Total hippocampal miR-181a was reported to be upregulated DISCLOSURE/CONFLICT OF INTEREST
30 minutes and 24 hours after 20 minutes of global ischemia,31 The authors declare no conict of interest.
and hippocampal Bcl-2 protein levels were previously found
to be decreased 24 hours after 10 minutes of global ischemia.32
Although these studies evaluated changes in whole hippocampus, ACKNOWLEDGEMENTS
we dissected out the selectively vulnerable CA1 region so as not to
The authors thank Robin White for assistance with microscopy, Yibing Ouyang for
average changes in the vulnerable region with changes in the help preparing gures, and William Magruder for assistance with preparing the
other hippocampal regions that survive. We found that miR-181a manuscript.
increased in CA1 but tended to decrease in dentate gyrus
(Table 1), whereas Bcl-2 protein levels decreased in CA1 but
increased in dentate gyrus. These results are consistent with the REFERENCES
pattern of neuronal cell death in CA1 and survival in dentate gyrus.
1 Kirino T, Tamura A, Sano K. Delayed neuronal death in the rat hippocampus
The signicant protection observed here against forebrain following transient forebrain ischemia. Acta Neuropathol 1984; 64: 139147.
ischemia was associated with increased Bcl-2 protein levels and 2 Zola-Morgan S, Squire L, Amaral D. Human amnesia and the medial temporal
preservation of the astrocyte glutamate transporter GLT-1. We region: enduring memory impairment following a bilateral lesion limited to eld
previously reported that inhibition of miR-181a in primary CA1 of the hippocampus. J Neurosci 1986; 6: 29502967.
astrocytes increased Bcl-2 protein and reduced oxidative stress 3 Rempel-Clower NL, Zola SM, Squire LR, Amaral DG. Three cases of enduring
during glucose deprivation.18 A close association was seen memory impairment after bilateral damage limited to the hippocampal
between oxidative stress, the early loss of astrocyte GLT-1, and formation. J Neurosci 1996; 16: 52335255.
delayed CA1 neuronal death in forebrain ischemia.21,22 Thus, the 4 Bernard SA, Gray TW, Buist MD, Jones BM, Silvester W, Gutteridge G et al.
Treatment of comatose survivors of out-of-hospital cardiac arrest with induced
preservation of GLT-1 observed here is consistent with protection
hypothermia. N Engl J Med 2002; 346: 557563.
of astrocyte function contributing to neuronal protection. 5 THACAS. Mild therapeutic hypothermia to improve the neurologic outcome after
Astrocytic glutamate uptake prevents or limits excitotoxic cardiac arrest. N Engl J Med 2002; 346: 549556.
neuronal injury.21,22,3335 In global and forebrain ischemia, 6 Padosch SA, Bottiger BW. Neuronal apoptosis following cerebral ischaemia:
reduced astrocytic glutamate uptake precedes neuronal loss. pathophysiology and possible therapeutic implications. Curr Opin Anesthesiol
Preventing this astrocyte dysfunction with metabotropic 2003; 16: 439445.

& 2013 ISCBFM Journal of Cerebral Blood Flow & Metabolism (2013), 1976 1982
MiR-181 reduction protects from forebrain ischemia
J-m Moon et al
1982
7 Petito CK, Feldmann E, Pulsinelli WA, Plum F. Delayed hippocampal damage in 25 Zhao H, Yenari MA, Cheng D, Barreto-Chang OL, Sapolsky RM, Steinberg GK. Bcl-2
humans following cardiorespiratory arrest. Neurology 1987; 37: 1281. Transfection via herpes simplex virus blocks apoptosis-inducing factor translo-
8 Teschendorf P, Padosch SA, Spohr F, Albertsmeier M, Schneider A, Vogel P et al. Time cation after focal ischemia in the rat. J Cereb Blood Flow Metab 2004; 24:
course of caspase activation in selectively vulnerable brain areas following global 681692.
cerebral ischemia due to cardiac arrest in rats. Neurosci Lett 2008; 448: 194199. 26 Martinou J-C, Dubois-Dauphin M, Staple JK, Rodriguez I, Frankowski H, Missotten
9 Jeyaseelan K, Lim KY, Armugam A. MicroRNA expression in the blood and brain of M et al. Overexpression of BCL-2 in transgenic mice protects neurons from
rats subjected to transient focal ischemia by middle cerebral artery occlusion. naturally occurring cell death and experimental ischemia. Neuron 1994; 13:
Stroke 2008; 39: 959966. 10171030.
10 Dharap A, Bowen K, Place R, Li L-C, Vemuganti R. Transient focal ischemia induces 27 Xu L, Lee JE, Giffard RG. Overexpression of bcl-2, bcl-xL or hsp70 in murine cortical
extensive temporal changes in rat cerebral MicroRNAome. J Cereb Blood Flow astrocytes reduces injury of co-cultured neurons. Neurosci Lett 1999; 277:
Metab 2009; 29: 675687. 193197.
11 Liu D-Z, Tian Y, Ander BP, Xu H, Stamova BS, Zhan X et al. Brain and blood 28 Hutchison ER, Kawamoto EM, Taub DD, Lal A, Abdelmohsen K, Zhang Y et al.
microRNA expression proling of ischemic stroke, intracerebral hemorrhage, and Evidence for miR-181 involvement in neuroinammatory responses of astrocytes.
kainate seizures. J Cereb Blood Flow Metab 2010; 30: 92101. Glia 2013; 61: 10181028.
12 Shi G, Liu Y, Liu T, Yan W, Liu X, Wang Y et al. Upregulated miR-29b promotes 29 Chen G, Zhu W, Shi D, Lv L, Zhang C, Liu P et al. MicroRNA-181a sensitizes human
neuronal cell death by inhibiting Bcl2L2 after ischemic brain injury. Exp Brain Res malignant glioma U87MG cells to radiation by targeting Bcl-2. Oncol Rep 2010; 23:
2012; 216: 225230. 9971003.
13 Yin K-J, Deng Z, Huang H, Hamblin M, Xie C, Zhang J et al. miR-497 regulates 30 Su SF, Chang YW, Andreu-Vieyra C, Fang JY, Yang Z, Han B et al. miR-30d,
neuronal death in mouse brain after transient focal cerebral ischemia. Neurobiol miR-181a and miR-199a-5p cooperatively suppress the endoplasmic reticulum
Dis 2010; 38: 1726. chaperone and signaling regulator GRP78 in cancer. Oncogene 2012.
14 Ouyang Y-B, Lu Y, Yue S, Xu L-J, Xiong X-X, White RE et al. miR-181 regulates 31 Yuan Y, Wang JY, Xu LY, Cai R, Chen Z, Luo BY. MicroRNA expression changes in
GRP78 and inuences outcome from cerebral ischemia in vitro and in vivo. the hippocampi of rats subjected to global ischemia. J Clin Neurosci 2010; 17:
Neurobiol Dis 2012; 45: 555563. 774778.
15 Chen C-Z, Li L, Lodish HF, Bartel DP. MicroRNAs modulate hematopoietic lineage 32 Zhang W, Wang B, Zhou S, Qiu Y. The effect of ischemic post-conditioning on
differentiation. Science 2004; 303: 8386. hippocampal cell apoptosis following global brain ischemia in rats. J Clin Neurosci
16 Miska E, Alvarez Saavedra E, Townsend M, Yoshii A, Sestan N, Rakic P et al. 2012; 19: 570573.
Microarray analysis of microRNA expression in the developing mammalian brain. 33 Hamby M, Sofroniew M. Reactive astrocytes as therapeutic targets for CNS
Genome Biol 2004; 5: R68. disorders. Neurotherapeutics 2010; 7: 494506.
17 Saba R, Storchel PH, Aksoy-Aksel A, Kepura F, Lippi G, Plant TD et al. Dopamine- 34 Raghavendra Rao VL, Muralikrishna Rao A, Dogan A, Bowen KK, Hatcher J,
regulated microRNA MiR-181a controls GluA2 surface expression in hippocampal Rothstein JD et al. Glial glutamate transporter GLT-1 down-regulation precedes
neurons. Mol Cell Biol 2012; 32: 619632. delayed neuronal death in gerbil hippocampus following transient global cerebral
18 Ouyang Y-B, Lu Y, Yue S, Giffard RG. miR-181 targets multiple Bcl-2 family ischemia. Neurochem Int 2000; 36: 531537.
members and inuences apoptosis and mitochondrial function in astrocytes. 35 Liu Z, Huo W, Sun W, Lv M, Li F, Su Z. A sequential impairment of cortical
Mitochondrion 2012; 12: 213219. astrocytes and GABAergic neurons during ischemia is improved by mGluR(1,5)
19 Dugan L, Bruno V, Amagasu S, Giffard R. Glia modulate the response of murine activation. Neurol Sci 2013; 34: 11891195.
cortical neurons to excitotoxicity: glia exacerbate AMPA neurotoxicity. J Neurosci 36 Kim DS, Kwak SE, Kim JE, Jung JY, Won MH, Choi SY et al. Transient ischaemia
1995; 15: 45454555. affects plasma membrane glutamate transporter, not vesicular glutamate trans-
20 Xu L, Koumenis IL, Tilly JL, Giffard RG. Overexpression of bcl-xL protects astrocytes porter, expressions in the gerbil hippocampus. Anat Histol Embryol 2006; 35:
from glucose deprivation and is associated with higher glutathione, ferritin, and 265270.
iron levels. Anesthesiology 1999; 91: 1036. 37 Kettenmann H, Ransom BR (eds). Neuroglia. Oxford University Press: New York, NY,
21 Xu L, Emery JF, Ouyang Y-B, Voloboueva LA, Giffard RG. Astrocyte targeted USA, 2012.
overexpression of Hsp72 or SOD2 reduces neuronal vulnerability to forebrain 38 Nedergaard M, Ransom B, Goldman SA. New roles for astrocytes: redening the
ischemia. Glia 2010; 58: 10421049. functional architecture of the brain. Trends Neurosci 2003; 26: 523530.
22 Ouyang Y-B, Voloboueva LA, Xu L-J, Giffard RG. Selective dysfunction of hippo- 39 Takano T, Oberheim N, Cotrina ML, Nedergaard M. Astrocytes and ischemic injury.
campal CA1 astrocytes contributes to delayed neuronal damage after transient Stroke 2009; 40: S8S12.
forebrain ischemia. J Neurosci 2007; 27: 42534260. 40 Zhang M, Li W-B, Geng J-X, Li Q-J, Sun X-C, Xian X-H et al. The upregulation of glial
23 Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time glutamate transporter-1 participates in the induction of brain ischemic tolerance
quantitative PCR and the 2  DDCT method. Methods 2001; 25: 402408. in rats. J Cereb Blood Flow Metab 2007; 27: 13521368.
24 Linnik MD, Zahos P, Geschwind MD, Federoff HJ. Expression of bcl-2 from a 41 Zhang M, Li W-B, Liu Y-X, Liang C-J, Liu L-Z, Cui X et al. High expression of GLT-1 in
defective herpes simplex virus1 vector limits neuronal death in focal cerebral hippocampal CA3 and dentate gyrus subelds contributes to their inherent
ischemia. Stroke 1995; 26: 16701675. resistance to ischemia in rats. Neurochem Int 2011; 59: 10191028.

Journal of Cerebral Blood Flow & Metabolism (2013), 1976 1982 & 2013 ISCBFM

You might also like