Abe 2018

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Received: 27 February 2018 Revised: 18 May 2018 Accepted: 22 May 2018

DOI: 10.1002/glia.23469

RESEARCH ARTICLE

Comparison of the detrimental features of microglia


and infiltrated macrophages in traumatic brain injury:
A study using a hypnotic bromovalerylurea
Naoki Abe1,2 | Mohammed E. Choudhury1 | Minori Watanabe1 | Shun Kawasaki1,3 |
Tasuku Nishihara2 | Hajime Yano1 | Shirabe Matsumoto4 | Takehiro Kunieda4 |
Yoshiaki Kumon4 | Toshihiro Yorozuya2 | Junya Tanaka1

1
Department of Molecular and Cellular
Physiology, Graduate School of Medicine, Abstract
Ehime University, Toon, Ehime, Japan Microglia and blood-borne macrophages in injured or diseased brains are difficult to distinguish
2
Department of Anesthesia and Perioperative because they share many common characteristics. However, the identification of microglia-
Medicine, Graduate School of Medicine, Ehime
specific markers and the use of flow cytometry have recently made it easy to discriminate these
University, Toon, Ehime, Japan
3
types of cells. In this study, we analyzed the features of blood-borne macrophages, and activated
Department of Surgery and Palliative
Medicine, Fujita Health University School of and resting microglia in a rat traumatic brain injury (TBI) model. Oxidative injury was indicated in
Medicine, Toyoake, Aichi, Japan macrophages and neurons in TBI lesions by the presence of 8-hydroxy-2’-deoxyguanosine
4
Department of Neurosurgery, Graduate (8-OHdG). Generation of mitochondrial reactive oxygen species (ROS) was markedly observed in
School of Medicine, Ehime University, Toon, granulocytes and macrophages, but not in activated or resting microglia. Dihydroethidium staining
Ehime, Japan
supported microglia not being the major source of ROS in TBI lesions. Furthermore, macrophages
Correspondence
Junya Tanaka, Department of Molecular and
expressed NADPH oxidase 2, interleukin-1β (IL-1β), and CD68 at higher levels than microglia. In
Cellular Physiology, Graduate School of contrast, microglia expressed transforming growth factor β1 (TGFβ1), interleukin-6 (IL-6), and
Medicine, Ehime University, Ehime 791-0295, tumor necrosis factor α at higher levels than macrophages. A hypnotic, bromovalerylurea (BU),
Japan.
which has anti-inflammatory effects, reduced both glycolysis and mitochondrial oxygen consump-
Email: jtanaka@m.ehime-u.ac.jp
tion. BU administration inhibited chemokine CCL2 expression, accumulation of monocytes/mac-
Funding information
Ministry of Education; Ehime University; Ehime rophages, 8-OHdG generation, mitochondrial ROS generation, and proinflammatory cytokine
University expression, and markedly ameliorated the outcome of the TBI model. Yet, BU did not inhibit
microglial activation or expression of TGFβ1 and insulin-like growth factor 1 (IGF-1). These results
indicate that macrophages are the major aggravating cell type in TBI lesions, in particular during
the acute phase. Activated microglia may even play favorable roles. Reduction of cellular energy
metabolism in macrophages and suppression of CCL2 expression in injured tissue may lead to
amelioration of TBI.

KEYWORDS

CCL2, FACS, glycolysis, mitochondria, oxidative stress, ROS

1 | I N T RO D UC T I O N chronic traumatic encephalopathy and Alzheimer’s disease. Although


the primary injury in TBI is initiated by mechanical force, the second-
Traumatic brain injury (TBI) is a leading cause of incurable neurological ary injury of subsequent neuroinflammatory events strongly aggra-
disability that affects several million people in the world every year vates the outcomes of TBI, in which infiltrated leukocytes and
(Corps, Roth, & McGavern, 2015; Gyoneva & Ransohoff, 2015; Nell & resident microglia play critical roles (Gyoneva & Ransohoff, 2015;
Brown, 1991). Most TBI patients suffer from long-lasting neurological McKee & Lukens, 2016; Webster et al., 2017).
problems, such as personality changes, deficits in learning and mem- Shortly after TBI, brain parenchymal cells around the lesion start
ory, and an increased risk of neurodegenerative changes, including to degenerate and then leukocytes infiltrate the lesion while exerting

Glia. 2018;1–16. wileyonlinelibrary.com/journal/glia © 2018 Wiley Periodicals, Inc. 1


2 ABE ET AL.

deleterious effects (Gyoneva & Ransohoff, 2015; Matsumoto et al., of Medicine. Male Wistar rats (7- to 8-weeks old, body weight
2007; Semple, Bye, Rancan, Ziebell, & Morganti-Kossmann, 2010). 250–270 g) were housed under standard laboratory conditions.
Furthermore, resident microglia that rapidly become activated in
response to brain injury are considered to play aggravating roles 2.2 | TBI and BU administration
(Bachstetter et al., 2013; Zhang et al., 2012). However, some studies
Stab wound injuries were made in the rat brain as described else-
demonstrate that infiltrated macrophages (Nishihara et al., 2011) and
where (Yokoyama, Sakamoto, Kameda, Imai, & Tanaka, 2006). In brief,
activated microglia (Act-MG) (Roth et al., 2014) have beneficial effects
rats were anesthetized with isoflurane, and a 15 mm longitudinal inci-
on damaged tissues and cells. Because of the conflicting data, the
sion was made in the skin to expose the skull. Two holes were made
term “double-edged sword” (Patel, Ritzel, McCullough, & Liu, 2013)
through the skull over the right hemisphere at approximately 2.5 and
has been used for the roles of these cells in injured brains.
4 mm right of the midline and 1 mm posterior to the bregma. Through
Act-MG are difficult to distinguish from infiltrated macrophages
each hole, a 26-gauge needle was inserted to a depth of approxi-
because of common functions and markers and similar morphological
mately 7 mm from the surface of the skull and the needle was moved
characteristics (Bennett et al., 2016). Therefore, the notion that micro-
in a fan like manner from the anterior then to the posterior in parallel
glia or macrophages have both ameliorative and destructive actions
with the midline. Thereafter, the needle was withdrawn, and the skin
might just reflect a confusion between the two types of cells.
incision was closed with quick-drying glue (Aron-Alpha; Toagosei,
Recently, microglia-specific proteins have been identified: transmem-
Tokyo, Japan). One hour after wounding, BU (1 mg/ml) dissolved in
brane protein 119 (TMEM119) (Bennett et al., 2016; Satoh et al.,
propylene glycol was subcutaneously injected at a dose of
2016) and Siglec-H (Konishi et al., 2017). These specific markers for
30 mg/kg. For control rats, propylene glycol only was injected.
microglia will bring about marked progress in understanding the roles
Wounded rats were then allowed to freely drink. Control rats were
of microglia in brain pathology.
given water and BU-treated rats were given water containing BU
Another method has recently been employed to distinguish macro-
(500 mg/l) for 7 days. Rats drank approximately 25 ml water per day;
phages and microglia from the brain; the combination of brain tissue
therefore, the daily BU dose was 50 mg/kg, which was comparable to
dissociation and flow cytometry (Hattori et al., 2017). Microglia express
the maximum clinically applicable dose. The Stroke Therapy Academic
CD45 at lower levels than macrophages, which is in contrast to
Industry Roundtable (STAIR) preclinical recommendations state that
CD11b, which is expressed at similar levels by both cell types. By the
multiple endpoints should be determined for anatomical and behav-
simultaneous use of antibodies to CD11b and CD45 after dissociation
ioral outcome at least 2–3 weeks or longer after onset of stroke to
of brain tissues into individual cells, flow cytometry can effectively dis-
determine the sustained benefit of experimental drugs (Fisher et al.,
criminate the two cell types from the brain. Furthermore, individually
2009). We, therefore, pursued the course of TBI for 15 weeks, using
sorted cells can be analyzed by quantitative real-time RT-PCR (qPCR).
magnetic resonance imaging (MRI) to measure cavity volumes and by
We have previously found marked anti-inflammatory effects of an
performing behavioral tests and anatomical evaluation at the endpoint
established hypnotic/sedative, bromovalerylurea (BU; C6H11BrN2O2,
as described below.
CAS: 496-67-3) (Higaki et al., 2016; Kikuchi et al., 2015). BU inhibits
CCL2 expression by lipopolysaccharide (LPS)-treated peritoneal macro-
phages (Kikuchi et al., 2015) and proinflammatory activation of microglia 2.3 | Measurement of lesion-induced cavity size
(Higaki et al., 2016). The antiinflammatory effects were at least partly
The stab wound injury caused extensive tissue loss. The resultant cav-
attributable to its suppressive effects on mitochondrial ATP production
ity volume was measured using the following two methods.
(Kawasaki et al., 2017). In this study using a rat stab-wound TBI model,
BU produced marked long-term amelioration of neurological deficits 2.3.1 | Magnetic resonance imaging
while reducing brain tissue loss. Furthermore, BU reduced the mitochon-
As described previously in Nishioka et al. (2016), MR images of
drial oxygen consumption rate (OCR) and glycolysis activity.
wounded rat heads were acquired with a 1.5 T MRminiSA (DS Pharma
By employing flow cytometry to discriminate microglia from mac-
Biomedical, Osaka, Japan) at 5 and 10 weeks after injury. A
rophages and by exploiting the marked ameliorative effects of BU, we
T1-weighted MR sequence was used with 1-mm slice thickness (total
differentially analyzed the contribution of blood-borne macrophages
13 slices). The lesion cavity area was calculated from the MR images
and microglia to the pathogenesis of TBI. Although microglia undergo
by marking low intensity areas, and the cavity volume was calculated
degeneration during the acute phase of TBI (Matsumoto et al., 2007),
by taking into account the slice thickness.
they may be ameliorating rather than aggravating compared with infil-
trated macrophages. 2.3.2 | Slices of fixed brains
Wounded rat brains were dissected out 15 weeks after TBI and
immersed in phosphate buffered saline (PBS) containing 4% parafor-
2 | MATERIALS AND METHODS
maldehyde and 2 mM MgCl2 for 7 days. Then, seven 2-mm thick
slices, centered on the injury site, were cut from each brain and
2.1 | Animals photographed. The images were processed to black (cavity) and white
All animal experiments were carried out in accordance with the Guide- (remaining brain tissue) images using Adobe Photoshop CS5 Extended
lines for Animal Experimentation of Ehime University Graduate School (Adobe Systems, San Jose, CA). The remaining brain tissue volume in
ABE ET AL. 3

the right hemisphere was calculated by multiplying the area by 2 mm areas of injured tissue around the stab wounds were dissected
(thickness of each slice). The percentage of lost volume was obtained (Figure 2b). Total RNA was extracted from the tissue homogenates
by dividing the right remaining brain tissue volume by the volume of using an RNeasy Lipid Tissue Mini Kit (Qiagen, Hilden, Germany). The
the left hemisphere. quality and quantity of purified RNA was examined using a NanoDrop
ND-2000 spectrophotometer (Thermo Fisher Scientific, Wilmington,

2.4 | Behavioral assessments DE). cDNA samples were prepared from 0.5 μg RNA using ReverTra
Ace qPCR RT Master Mix with a gDNA remover kit (Toyobo, Osaka,
Learning and memory abilities after TBI were evaluated using Morris
Japan) and were then diluted three-fold. qPCR analysis was then per-
water maze and passive avoidance tests.
formed in triplicate using an MJ mini instrument (BioRad, Hercules,
CA) and Fast Start Universal SYBR Green Master (Roche Diagnostic
2.4.1 | Morris water maze test
Japan, Tokyo, Japan). All gene-specific mRNA measurements were
Spatial memory was assessed using a standard Morris water maze,
normalized to glyceraldehyde 3-phosphate dehydrogenase (GAPDH)
essentially as described elsewhere (Liu, Zhang, Wu, Wu, & Wang, 2013).
mRNA levels. All PCR primer sequences are listed in Supporting Infor-
The animals were tested in a 150-cm diameter × 45-cm tall circular pool
mation Table S1.
filled with water at 24  C. A 12-cm diameter circular transparent plat-
form was placed 2 cm below the waterline in the center of one quadrant.
BU-treated and non-treated TBI rats were tested. Four different signs 2.6 | Immunohistochemistry
were put on the walls of the quadrants (N, S, E, and W). On Day 0, the Under deep anesthesia, TBI rats at 1.5 or 7 dpi were subjected to per-
platform was indicated with a small flag. Rats were forced to swim freely fusion fixation using the fixative described in Section 2.3.2 and as
twice for 60 s from points S and E of the pool to find the platform with described elsewhere (Sugimoto et al., 2014). The brain region contain-
the flag. To memorize the location of the platform, rats were made to ing the injury was coronally cryosectioned at a thickness of 10 μm
stay on it for 10 s having successfully found the platform and for 15 s and subjected to immunohistochemical staining using antibodies listed
when the rat could not find the platform. On Days 1 and 2, the flag was in Supporting Information Table S2. Hoechst 33,342 (Sigma-Aldrich,
removed and rats were given one trial for 60 s to find the platform. The St. Louis, MO) was used for nuclear staining. To detect 8-hydroxy-20 -
number of rats accomplishing the task was recorded and the mean deoxyguanosine (8-OHdG), brain sections were incubated in boiling
latency to the platform, total distance swum, and the swim speed was 10 mM citrate buffer (pH 6.0) for 5 min followed by cooling in water
measured for each trial using a video-tracking system (Ethovision XT at room temperature, before incubation with an anti-8-OHdG anti-
7, Noldus Info. Tech., Wageningen, The Netherlands). body. Stained sections were observed with a Nikon A1 confocal laser
scanning microscope (Tokyo, Japan) or a scanning fluorescence micro-
2.4.2 | Passive avoidance test scope (BZ-9000; Keyence, Osaka, Japan).
Behavioral changes were also evaluated using a step-through passive
avoidance apparatus (Shuttle box; MPB-M001; Melquest, Toyama,
2.7 | Primary rat mixed glial and purified microglial
Japan) as described elsewhere (Lashgari, Motamedi, Zahedi Asl, Sha-
cell cultures
hidi, & Komaki, 2006). The apparatus consisted of an illuminated cham-
ber made of transparent plastic and a dark chamber made of dark Rat primary mixed glial and purified microglial cell cultures were pre-
plastic. The size of both chambers was the same (45 × 27 × 26 cm). pared as described previously (Tanaka et al., 1998). Whole forebrains
The floors of both chambers were made of stainless steel rods and the from neonatal rats were mechanically dissociated into individual cells.
dark chamber floor was equipped with an electric shock generator (SG- Mixed glial cultures were grown in Dulbecco’s modified Eagle’s medium
100; Melquest). A rectangular opening with an opaque guillotine door (DMEM) containing 10% fetal bovine serum in poly-L-lysine-coated
was located between the two chambers. First, individual rats were put 6-well culture plates for 11 days. Culture medium was then removed
in the light chamber and allowed to freely explore it for 30 s. Then, the and cells were incubated for 3 hr in serum-free E2 medium (DMEM
door was opened, and the latency for rats to completely enter the dark containing 10 mM HEPES, pH 7.3, Gibco, Grand Island, NY; 4.5 mg/ml
chamber was measured (up to 300 s). When the rats were habituated glucose, 5 μg/ml insulin, 5 nM sodium selenite, 5 μg/ml transferrin,
to enter the dark chamber within 20 s, the training was terminated. Gibco; and 0.2 mg/ml bovine serum albumin, Sigma-Aldrich) in the pres-
Then, rats were again placed in the light chamber, and the door was ence or absence of 1 μg/ml LPS (Sigma-Aldrich). Cells were then solubi-
opened 30 sec later. When the rats completely entered the dark cham- lized for qPCR using an RNeasy Lipid Tissue Mini Kit (Qiagen). Purified
ber, a 1 mA electric shock was applied for 3 s. Twenty-four hours later, microglial cells were prepared from the mixed glial culture and were
trained rats were again placed into the light chamber, and the latency used to determine cellular energy metabolism. BU (100 μg/ml; Higaki
to enter the dark chamber was measured (ceiling score 300 s). et al., 2016) was added to cultures 24 hr before collecting total RNA
for qPCR or measurement of cellular metabolism.

2.5 | Quantitative real time RT-PCR


At 1.5 or 7 days postinjury (dpi) TBI rats were decapitated under deep
2.8 | Flow cytometry
anesthesia, and their brains were dissected. The 2-mm thick coronal Under deep anesthesia, rats at 1.5 or 7 dpi were subjected to transcardial
sections centered on the wound holes were prepared. Rectangular perfusion with chilled PBS for 3 min to remove blood, followed by
4 ABE ET AL.

dissection of injured brains. Right brain hemispheres (~500 mg) of TBI 2.12 | Identification of cells generating superoxide
and normal rats were dissociated into single cells in a gentleMACS disso- anions using dihydroethidium
ciator using program 37C_ABDK_01 (Miltenyi Biotec, Tokyo, Japan) and
Dihydroethidium (DHE; Sigma-Aldrich) was used to monitor cells generat-
solutions provided with the adult brain dissociation kit (Miltenyi Biotec)
ing superoxide anions in TBI brain tissues (Choi et al., 2012). DHE
as previously described in Hattori et al. (2017). Nondissociated tissue
(10 mg/kg) was intraperitoneally administered to TBI model rats at 1.5 dpi
aggregates were removed using MACS SmartStrainers with 100-μm
and, 30 min later, the animals were euthanized and the brains dissected
pores (Miltenyi Biotec), and then debris and contaminating erythrocytes
out. Coronal slices were cut and fixed in 4% paraformaldehyde in PBS for
were removed from the cell suspensions using debris removal and red
30 min. After incubating in PBS containing 15% sucrose for 2 h, 10-μm
blood cell removal solutions provided with the adult brain dissociation kit.
thick cryosections were prepared. After immunohistochemical staining
The prepared cell suspensions were subjected to flow cytometry
using antibodies to Iba1 and CD45, the specimens were observed with a
analyses. The suspensions were diluted to 1 × 106 cells/100 μl with
Nikon A1 confocal laser scanning microscope (Tokyo, Japan).
PBS containing 2% fetal bovine serum and 2 mM ethylenediaminete-
traacetic acid. Cells were then incubated on ice with an anti-CD32
antibody for 20 min to block Fc receptors. Cells then were incubated
2.13 | Quantification of IGF-1 in brain tissues
with fluorescence-labeled antibodies (see Supporting Information IGF-1 protein in the right hemisphere of TBI rat brains administered
Table S3). To detect CD206+ cells, cells were incubated with an anti- vehicle or BU was quantified with an ELISA kit (R&D Systems). Before
CD206 antibody and then with an Alexa405-labeled anti-rabbit IgG the assay, brain tissues were homogenized in PBS and stored at
antibody (abcam) for 30 min on ice. To detect generated mitochon- −30  C overnight, followed by two freeze–thaw cycles to break cell
drial reactive oxygen species (ROS) (mitROS), cells isolated at 1.5 dpi membranes. Supernatants were then prepared and the ELISA assay
were suspended in E2 medium and incubated with MitoROS performed in triplicate.

520 (AAT Bioquest, Sunnyvale, CA) for 20 min at 37 C. The cells
labeled with antibodies for flow cytometry (see Supporting Informa- 2.14 | Determination of cell metabolic rates
tion Table S3) and/or MitoROS 520 were analyzed on a Gallios flow
Oxygen consumption rate (OCR) and extracellular acidification rate
cytometer (Beckman Coulter, Tokyo, Japan). Cell viability was ana-
(ECAR) were investigated to evaluate mitochondrial respiration and glycol-
lyzed using propidium iodide or Zombie NIR (BioLegend). Data were
ysis, respectively, in rat primary microglial cells and alveolar macrophages
analyzed using FlowJo Software (version.7.6.5, Treestar, Ashland, OR).
using XFp Extracellular Flux Analyzer (Agilent Seahorse Bioscience, Santa
Clara, CA) (Banh et al., 2016; Garaude et al., 2016). Rat alveolar macro-
2.9 | Cell sorting and qPCR phages, prepared as described elsewhere (Kikuchi et al., 2015), or micro-

mRNA levels in sorted cells were analyzed by qPCR. Before flow cyto- glial cells were seeded in E2 at 2.5 × 104 cells in eight-well plates provided

metry sorting, cells were resuspended with 5 volumes Cell Cover by the manufacturer. Two wells among the eight well were used as blank

(AL Anacyte Laboratories UG, Hamburg, Germany) at 4 C to stabilize wells. After overnight incubation in a CO2 incubator, the plates were

mRNA. Macrophages and microglial cells were collected on a BD FAC- placed in the XFp analyzer, which was operated according to the manufac-
turer’s instructions using oligomycin A (1 μM), carbonyl cyanide-p-
SAria flow cytometer (BD biosciences, Franklin Lakes, NJ). Total RNA
trifluoromethoxyphenylhydrazone (FCCP; 1 μM), rotenone (0.5 μM), and
was prepared from cells using an RNeasy micro kit (Qiagen) and then
antimycin A (0.5 μM), which were automatically and sequentially added to
reverse-transcribed to obtain cDNA. qPCR was performed as
cells to determine mitochondrial and glycolysis activities.
described earlier.

2.15 | Statistical analysis


2.10 | Quantification of 8-OHdG
Data expressed as means  SEM or SD were statistically analyzed using
To evaluate oxidative damage in injured brain tissues, 8-OHdG was
InStat3 software (GraphPad Software, La Jolla, CA). Data were sub-
quantified. Injured brain tissues were isolated at 1.5 dpi, and DNA
jected to two-tailed Student’s t test (unpaired) or ANOVA with Tukey’s
was extracted from the tissue (~150 mg) using a DNA Extractor TIS
post hoc test. Significance was set at p < .05 unless otherwise stated.
kit (Wako, Osaka, Japan) and hydrolyzed using a DNA hydrolysis kit
(Wako). The 8-OHdG levels were quantified using a highly sensitive
ELISA kit (Nikken SEIL, Shizuoka, Japan). 3 | RE SU LT S

2.11 | Quantification of interleukin-6 in 3.1 | Effects of BU on cell metabolism


cerebrospinal fluid
BU has marked antiinflammatory effects on microglia (Higaki et al.,
Cerebrospinal fluid (CSF) was taken at 1.5 dpi from the space over the 2016) and macrophages (Kikuchi et al., 2015), and the effects that are
midbrain via the small holes in the skull. Interleukin-6 (IL-6) concentra- at least partly attributable to suppressive effects on ATP synthesis
tion was determined using an ELISA kit (R&D Systems; Minneapo- (Kawasaki et al., 2017). In this study, the effects of BU on cell metabo-
lis, MN). lism were investigated in primary microglial cells (Figure 1) and
ABE ET AL. 5

alveolar macrophages (Supporting Information Figure S1) using the dysfunctions as revealed by Morris water maze and passive avoidance
Seahorse Mito-Stress Test, which assesses Oxygen consumption rate tests that were performed 15 weeks after TBI (Figure 2c–e). In the
(OCR) and extracellular acidification rate (ECAR) (Figure 1a) (Banh water maze test, BU reduced the duration to reach the platform and
et al., 2016; Garaude et al., 2016). By measuring OCR, basal increased the number of rats that accomplished the task (Figure 2d)
(Figure 1b) and maximal respiration (Figure 1 da), ATP production but did not affect swimming velocity (Figure 2dd). BU administration
(Figure 1c), spare respiration capacity (Figure 1db), and non- improved memory retention as revealed by the passive avoidance test
mitochondrial oxygen consumption (Figure 1e) were determined. (Figure 2e). Thus, BU brought about long-term amelioration in terms
ECAR reflects glycolysis activity. BU decreased all of the indices for of TBI-induced brain tissue loss and neurological dysfunctions.
glycolysis and mitochondrial metabolism that lead to reduced ATP
production. However, BU did not affect coupling efficiency 3.3 | CCL2 expression and proinflammatory
(Figure 1f ), which is calculated by dividing ATP production by basal
reactions
respiration, indicating that BU did not induce mitochondrial dysfunc-
tion but suppressed total cellular metabolism. BU suppresses CCL2 expression in LPS-treated peritoneal macro-
phages (Kikuchi et al., 2015). CCL2 is also expressed by astrocytes
and microglial cells (Berman, Guida, Warren, Amat, & Brosnan, 1996;
3.2 | Amelioration of TBI by BU
Liu et al., 2013; Tei et al., 2013). We, therefore, investigated whether
A penetrating stab injury was made in the right hemisphere of BU inhibited CCL2 expression in a mixed glial culture containing astro-
8-week-old male Wistar rats as a TBI model. BU was subcutaneously cytes, microglial cells and NG2 glia (oligodendrocyte progenitor cells).
administered 1 h after TBI. BU dissolved in water was then given to Unless stimulated with LPS, the mixed glial culture expressed CCL2
TBI rats for 7 days. Rats were then given normal water for 15 weeks. mRNA at a low level. Incubation with LPS markedly increased CCL2
Five and ten weeks after injury, the injured brains were monitored by mRNA levels, and BU significantly suppressed this upregulation
MRI (Figure 2a). At both time points, TBI-induced cavity or lost brain (Figure 3a).
tissue volumes were reduced in the BU administered rats. Rats were The injured brain regions were dissected at 1.5 dpi as shown in
euthanized 15 weeks after TBI and the cavity size in fixed brain slices Figure 3b. The dissected brain tissues were used for qPCR analyses.
was quantified [Figure 2b; also see Supporting information Figure S2 BU suppressed mRNA levels of CCL2 (Figure 3ca) and its receptor
displaying all rat brain slices examined (n = 12 for each group)]. No sig- CCR2 (Figure 3c[b]), the latter being mainly expressed by mono-
nificant differences in motor abilities were detected by open-field and cytes and macrophages (Gyoneva & Ransohoff, 2015; Semple,
rota-rod tests (data not shown); however, BU administration amelio- Kossmann, & Morganti-Kossmann, 2010). mRNAs for CD11b
rated spatial cognitive impairments and learning/memory (Figure 3da), a marker for myeloid leukocytes and microglia, Iba1

FIGURE 1 Effects of BU on energy metabolism in rat primary microglia as assessed by OCR and ECAR. (a) OCR (upper) and ECAR (lower) of cells treated
with vehicle or BU. OCR and ECAR values in the absence for measuring a basal respiration rate (b) or in the sequential presence of oligomycin for an ATP
production rate (c), FCCP for a maximal respiration rate (da) and spare respiratory capacity (db), and rotenone+antimycin a for nonmitochondrial OCR (e).
Coupling efficiency (f ) was calculated by dividing ATP production by basal respiration. Data (n = 3) are shown as means  SD and were statistically
analyzed with Student’s t test. *p < .05, ***p < .001 versus vehicle [Color figure can be viewed at wileyonlinelibrary.com]
6 ABE ET AL.

FIGURE 2 Amelioration of TBI in the rat forebrain by BU. (a) BU reduced the lesion cavity size 5 and 10 weeks after TBI as revealed by MRI.
Representative MR images of vehicle (aa) and BU-administered (ab) rats were taken 10 weeks after TBI. The measured cavity size or lost volume
(n = 12; mm3) based on MR images taken 5 (ac) or 10 (ad) weeks after TBI. (b) Brains were dissected, fixed and coronal slices were prepared
15 weeks after TBI. The volume of left (contralateral) and right (ipsilateral) hemispheres of brain slices were measured independently, and the
(left–right volume)/left volume values are shown. (c,d) BU administration ameliorated impaired learning ability as revealed by the Morris water
maze test. (c) Representative traces recorded on days 1 and 2 of rats administered with vehicle (water; ca) or BU (cb). (d) Duration to reach the
platform (da), distance moved to the platform (db), and the percentage of rats that reached the platform (dc) are shown. Swimming velocity was
not different between the two groups (dd). (e) Transfer latency (sec) in the passive avoidance test. Data are shown as means  SD and were
statistically analyzed with Student’s t test (n = 12). *p < .05, **p < .01, ***p < .001 versus vehicle [Color figure can be viewed at
wileyonlinelibrary.com]

(AIF1) (Figure 3db), a specific marker for microglia and monocytes/ IL-6 (Figure 3ed). BU also decreased the concentration of IL-6 in
macrophages (Matsumoto et al., 2007), and neutrophil elastase CSF (Figure 3f ). BU decreased mRNA levels of p40phox
(ELANE) (Figure 3dc) were all suppressed by BU administration. BU (Figure 3ga) and p67phox (Figure 3gb), subunits of NADPH oxidase
reduced mRNA levels of the proinflammatory cytokines, (NOX), which generates ROS (Ansari, Roberts, & Scheff, 2014).
interleukin-1β (IL-1β) (Figure 3ea), tumor necrosis factor α (TNFα; However, BU did not reduce mRNA levels for TGFβ1
Figure 3eb), inducible nitric oxide synthase (iNOS; Figure 3ec) and (Figure 3ha) or IGF-1 (Figure 3hb).
ABE ET AL. 7

FIGURE 3 Suppressive effects of BU on proinflammatory responses in vitro and in vivo. (a) BU suppressed LPS-induced CCL2 mRNA expression
in a mixed glial culture. Total RNA was collected 3 h after LPS addition. Data are shown as means  SEM (n = 4). **p < .01, *** p < .001 versus
control/LPS(−). ## p < .01, versus control/LPS(+). ANOVA and Tukey’s post hoc test. (b) the injured brain tissues denoted with dotted lines were
isolated at 1.5 dpi from brain slices for qPCR. (c) BU administration suppressed levels of CCL2 and CCR2 mRNA. (d) BU suppressed mRNA levels
of CD11b, Iba1, and neutrophil elastase. (e) mRNA levels of IL-1β, TNFα, iNOS and IL-6 were also suppressed by BU. (f) BU administration
reduced IL-6 protein levels in CSF at 1.5 dpi. (g) BU reduced mRNA levels of NOX subunits, p40 and p67phox. (h) BU did not affect mRNA levels
of TGFβ1 or IGF-1. * p < .05, **p < .01 versus vehicle. Data analyzed with Student’s t test are shown as means  SEM (n = 7 for qPCR and n =
4 for ELISA) [Color figure can be viewed at wileyonlinelibrary.com]

3.4 | Flow cytometry analyses of leukocytes and surface protein, TMEM119 (Bennett et al., 2016; Satoh et al., 2016)
microglia at 1.5 dpi (Figure 4d). TMEM119 expression in both resting and Act-MG fractions
was higher than that in the macrophage fraction.
Flow cytometry analyses using mixtures of antibodies to CD45 (clone
Resting and Act-MG were separated by forward (FS) and side scat-
OX1) and granulocytes (Figure 4a, see Supporting information
ter (SS) plots (Figure 4e). Microglia in normal brains were a very homog-
Figure S3 for gating strategy) or CD11b/c (clone OX42; Figure 4b)
enous population (Figure 4ea). We presupposed that 96% of microglial
revealed only a few macrophages and granulocytes in the normal brain
cells (96.0  1.40%; mean  SD; n = 5) are classified as resting microglia
(Figure 4aa, ba, and c). CD45hi macrophages and CD45+/granulocyte+ (Rest-MG) in normal rat brains, and the gating (dotted line in
cells (granulocytes or presumptive neutrophils) were abundantly pre- Figure 4ea) was set based on this estimation. Microglia excluded from
sent in vehicle-treated TBI brains, whereas the number of CD45lo the gating were defined as Act-MG, and are surrounded with a red line.
microglia was decreased (Figure 4ab, bb, and c). BU did not affect the The FS and SS values of the Act-MG were comparable with those of
numbers of accumulated granulocytes or microglia (Figure 4c). How- macrophages, which are denoted with blue dots surrounded with blue
ever, the number of macrophages in BU-treated brains was less than lines (Figure 4eb, ec). Although the total number of microglia decreased,
that in vehicle-treated brains. The discrimination of microglia from mac- the percentages of Act-MG were much increased in TBI brains regard-
rophages by flow cytometry based on the level of CD45 expression less of treatment (Figure 4f ). Act-MG characteristically had higher FS
was validated by determining the mRNA level for microglia-specific cell and SS values than Rest-MG (Supporting Information Figure S4). TBI
8 ABE ET AL.

increased FS and SS values of granulocytes, macrophages and Act-MG. that were granulocytes because of their segmented nuclei (yellow
Act-MG characteristically increased their CD11b/c expression com- arrows and the inset in Hc) and Iba1+/CD45+ macrophages (blue
pared with resting microglia, but their CD45 expression was not chan- arrows), which displayed round morphology, a feature that was dis-
ged in response to TBI. tinctive from the ramified microglial morphology (Sugimoto et al.,
Immunohistochemical analyses showed the presence of a few 2014). BU administration did not affect CD45 expression and mor-
Act-MG with weak CD45-immunofluorescence and ramified morphol- phology of microglia in the normal brain (Supporting Information
− +
ogy (Figure 4g,h; pink arrows). Also present were Iba1 /CD45 cells Figure S5).

FIGURE 4
ABE ET AL. 9

3.5 | ROS-generating cells and oxidative stress 3.6 | Characterization of sorted macrophages and
at 1.5 dpi activated and resting microglia by qPCR
ROS oxidizes DHE to produce red fluorescence from ethidium Macrophages and microglia accumulated in injured hemispheres were
(Choi et al., 2012). Cells emitting ethidium fluorescence were collected by cell sorting at 1.5 dpi, and cDNA prepared from the iso-
Iba1−/CD45+ granulocytes and Iba1+/CD45hi macrophages, but not lated cells was subjected to qPCR (Figure 6). NADPH-oxidase
+ lo/−
Iba1 /CD45 microglia with ramified processes (Figure 5a). Note 2 (NOX2; gp91phox) mRNA was found in macrophages at much higher
the absence of ramified microglia in the core lesion where abundant levels than in microglia (Figure 6a). Other NOX components, p40phox
granulocytes and macrophages are located as described elsewhere and p67phox, were expressed by microglia and macrophages at similar
(Matsumoto et al., 2007). BU did not affect CD45 expression and levels, and BU did not affect their expression (Supporting Information
also it did not cause ethidium fluorescence in the normal brains Figure S8). Glutathione peroxidase (GPx) mRNA was also expressed at
(Supporting Information Figure S6A). BU reduced the ethidium fluo- higher levels by macrophages than by microglia (Figure 6b). BU did
rescence in and around the lesion core (Supporting Information not affect the expression of these mRNAs. Higher mRNA levels of
Figure S6B). iNOS (Figure 6c) and IL-1β (Figure 6d) were expressed by macro-
Approximately 90% of ROS are generated by mitochondria (Gao, phages compared with microglia, and Act-MG expressed these
Laude, & Cai, 2008); therefore, mitROS generation by the individual mRNAs at higher levels than Rest-MG. BU reduced iNOS and IL-1β
cell types was evaluated using flow cytometry with a superoxide- mRNA levels in macrophages. Conversely, microglia expressed TNFα,
sensitive fluorescent molecule, MitoROS 520, which quickly enters IL-6, and TGFβ1 mRNAs at higher levels than macrophages. BU sup-
mitochondria and generates green fluorescence when oxidized by pressed TNFα and IL-6, but not TGFβ1 mRNA expression. Microglia
superoxide (Serrano-Nascimento et al., 2014) (Figure 5b). When com- and macrophages expressed IGF-1 mRNA at similar levels, and BU did
pared with granulocytes and macrophages, both activated and resting not affect its expression. Macrophages expressed mRNA encoding
microglia generated much lower levels of mitROS. TBI treatment CD68, a marker for mononuclear phagocytes (Song, Lee, & Schindler,
increased mitROS-generation in granulocytes, macrophages and rest- 2011), at higher levels than microglia (Figure 6i). The high expression
ing microglia but not in Act-MG. BU significantly reduced mitROS of CD68 in macrophages was confirmed by immunohistochemical
generation by macrophages and microglia. staining (Supporting Information Figure S9). Microglia in the close
To evaluate oxidative damage, 8-OHdG in DNA, a marker for vicinity of the lesion core displayed faint CD68-immunoreactvity.
oxidative stress (Valavanidis, Vlachogianni, & Fiotakis, 2009), was
immunohistochemically identified on cryosections of TBI brains at
3.7 | Effects of BU administration on TBI brains
1.5 dpi (Figure 5c). The majority of strongly 8-OHdG-positive cells
at 7 dpi
were macrophages (green arrowheads). A small number of NeuN+
neurons were weakly 8-OHdG-positive (pink arrowheads). In CD45+ cells in TBI brains at 7 dpi were analyzed by flow cytometry
vehicle- and BU-treated normal rat brains, no significant 8-OHdG- (Figure 7a–d; Supporting Information Figure S10). At this time point,
immunoreactivity was found (Supporting Information Figure S7a). the number of macrophages was less than that of total microglia in
Figure 5d shows that BU reduced 8-OHdG-immunoreactivity in and the injured hemisphere regardless of treatment. The number of Act-
around the lesion cores. 8-OHdG levels were quantified by ELISA. MG was much greater than that of Rest-MG. BU did not affect the
TBI increased 8-OHdG levels, and BU administration abolished this percentage of Act-MG or Rest-MG. BU decreased the number of mac-
increase to the control level (Figure 5e, also see Supporting Informa- rophages expressing CD86, a presumable M1 marker, and it weakly
tion Figure S7b for enlarged micrographs detecting 8-OHdG, Iba1, increased the number of CD206+ macrophages, which may be M2
and NeuN). cells (Figure 7c). NG2 chondroitin sulfate proteoglycan (NG2), a

FIGURE 4 Granulocytes, macrophages and microglia in normal, vehicle- and BU-administered TBI brains. (a) Flow cytometry analyses using
antibodies against granulocytes and CD45 distinguished granulocytes, macrophages (MΦ) and microglia (MG). Only a few granulocytes and
macrophages were present in normal brain (aa). The number of total microglia was reduced and the numbers of macrophages and granulocytes
were increased in TBI brains (ab, vehicle and ac, BU-administered). Representative results are shown. (b) Analyses using antibodies against
CD11b/c and CD45. Reduced numbers of microglia and increased numbers of macrophages are apparent in TBI brains (bb, vehicle and Bc, BU-
administered). (c) The frequency of each cell type with respect to total live cells is shown. Granulocyte and macrophage numbers were increased
and microglia numbers were reduced in TBI brains. BU administration significantly reduced the accumulation of macrophages (### indicates
significant reduction; p < .001). (d) Significantly higher levels of TMEM119 mRNA were found in sorted Act-MG and Rest-MG compared with
macrophages. (e) Act-MG and Rest-MG were distinguished by FS and SS plots. (ea) a 96% of microglial cells with small FS and SS values in normal
brains were defined as Rest-MG, denoted with the dotted line. A few Act-MG and macrophages were present in normal brain. The numbers of
Act-MG and macrophages in a TBI brain denoted with red or blue dots and lines, respectively, were increased after TBI (eb). BU decreased the
number of macrophages but not Act-MG (ec). (f ) TBI increased the percentage of Act-MG and reduced the percentage of Rest-MG, but BU did
not affect these percentages. Flow cytometry data analyzed with ANOVA and Tukey's post-hoc test are shown as means  SEM (n = 5). **p <
.01, *** p < .001 versus control of each group. $$$ p < .001, versus act-MG in each group. (g,h) Immunohistochemical staining of TBI brains at 1.5
dpi using antibodies against Iba1 and CD45. Nuclear staining was performed using Hoechst 33,342. (h) 3D-constructed images are shown in (g).
Blue arrows denote Iba1+/CD45+ macrophages, pink arrows Iba1+/CD45weak Act-MG, and yellow arrows Iba1−/CD45+ granulocytes. The inset in
(hc) shows a segmented nucleus of a granulocyte [Color figure can be viewed at wileyonlinelibrary.com]
10 ABE ET AL.

FIGURE 5 ROS generation by leukocytes and microglia in TBI brains and oxidative stress. (a) Immunohistochemical detection of ROS-generating
cells in TBI lesions at 1.5 dpi using DHE staining. Cells with strong DHE fluorescence were macrophages (blue arrows) and neutrophils (yellow)
with segmented nuclei. Microglial cells (pink) did not emit DHE fluorescence. (b) Granulocytes (ba), macrophages (bb), act-MG (bc) and rest-MG
(bd) were individually examined for mitROS generation by flow cytometry. Representative results (n = 5) are shown. Mean fluorescence intensity
for mitROS generation by granulocytes and macrophages in TBI brain was much stronger than that generated by microglia (be). BU significantly
suppressed macrophage and microglia-derived mitROS. Data were analyzed with ANOVA and Tukey’s post hoc test and are shown as means 
SEM (n = 5). ***p < .001 versus control of each group. ### p < .001, cells in vehicle-treated (Vehi) versus BU-treated TBI brains.
(c) Immunohistochemical staining of 8-OHdG in a vehicle-treated TBI brain. Iba1+ macrophages were strongly immunostained with anti8-OHdG
antibody (green arrowheads). NeuN+ degenerating neurons were weakly 8-OHdG-immunoreactive (pink arrowheads). (d) Immunohistochemical
staining of 8-OHdG in the right hemisphere of TBI brains. (dc) morphometric results show that BU treatment reduced 8-OHdG immunoreactivity.
Data were analyzed with Student's t-test and are shown as means  SEM *p < .05 versus vehicle (n = 3). (e) Quantification of 8-OHdG levels in
sham, vehicle- and BU-administered TBI brains by ELISA showed a TBI-induced increase of 8-OHdG levels and prevention of oxidative stress
effects by BU. data were analyzed with ANOVA and Tukey’s post hoc test and are shown as means  SEM (n = 4). *,# p < .05 versus sham and BU,
respectively [Color figure can be viewed at wileyonlinelibrary.com]
ABE ET AL. 11

FIGURE 6 Expression of mRNA by macrophages and microglia isolated from TBI brains by flow cytometry was analyzed by qPCR.
(a) Macrophages (MΦ) expressed NOX2 (a) and GPx (b) mRNA at higher levels than microglia. BU did not affect the expression. Macrophages
expressed iNOS (c) and IL-1β (d) mRNA at higher levels than microglia and BU suppressed this expression. Act-MG expressed iNOS mRNA at a
higher level than Rest-MG. Microglia expressed TNFα (e), IL-6 (f) and TGFβ1 (g) mRNAs at higher levels than macrophages. BU suppressed
expression of TNFα and IL-6 mRNAs but not that of TGFβ1. IGF-1 mRNA (h) was expressed at similar levels by all cells and BU did not affect its
expression. Macrophages expressed CD68 mRNA (i) at higher levels than microglia. BU did not affect CD68 expression. Data were analyzed with
ANOVA and Tukey's post-hoc test and are shown as means  SEM (n = 4). * $ p < .05, ** p < .01, ### p < .001. * versus vehicle-treated the same
cell type. $ versus vehicle-treated Act-MG. # versus vehicle-treated macrophages [Color figure can be viewed at wileyonlinelibrary.com]

marker for oligodendrocyte progenitor cells or NG2 glia, is expressed 4 | DI SCU SSION
by macrophages and microglia in injured brains that may be beneficial
cells for injured brains (Matsumoto et al., 2008; Smirkin et al., 2010).
4.1 | Strategy to determine the aggravating cell type
BU treatment did not affect the number of NG2+ macrophages.
in the TBI pathology
Figure 7e shows mRNA expression in TBI brain tissues at 7 dpi. BU
administration decreased levels of mRNAs encoding CCL2, CCR2, and It has been a focus of debate for decades whether Act-MG are benefi-
iNOS. Note that the level of CCL2 mRNA was much decreased com- cial or detrimental to injured brain tissues (Corps et al., 2015; Dud-
pared with that at 1.5 dpi. In contrast, BU increased levels of Iba1, varski Stankovic, Teodorczyk, Ploen, Zipp, & Schmidt, 2016; Karve,
GFAP, and IGF-1 mRNA. Proliferating cell nuclear antigen (PCNA) and Taylor, & Crack, 2016; Patel et al., 2013). Because Act-MG display
TGFβ1 mRNA levels were not affected by BU treatment. BU treat- similar phenotypes to those of infiltrated macrophages, they are diffi-
ment did not change the IGF-1 protein content in injured right hemi- cult to distinguish from each other. In this study, we took advantage
spheres (Figure 7k). Immunohistochemical observations showed that of flow cytometry to distinguish microglia from macrophages based
BU did not cause marked differences in Iba1, CD11b, NG2, and Ki67 on different levels of CD45 expression. We confirmed much higher
expression in the injured hemisphere at 7 dpi (Supporting Information expression of TMEM119 (Bennett et al., 2016; Satoh et al., 2016) in
Figure S11). However, dense accumulation of NG2+ macrophages isolated microglia compared with macrophages. Activated and resting
were often observed in the close vicinity of the lesions in BU-treated microglia were discriminated on the basis of FS and SS values. CD11b
brains (Supporting Information Figure S12). expression was also higher in Act-MG compared with Rest-MG.
12 ABE ET AL.

FIGURE 7 CD45+ cells from TBI brains were analyzed by flow cytometry (a–d) and effects of BU on mRNA and protein expression at 7 dpi were
analyzed (e–k). (a) Analyses with anti-CD11b and anti-CD45 antibodies. BU administration for 7 days did not affect the numbers of macrophages
or total microglia accumulated in injured hemispheres at 7 dpi. Representative flow cytometry plot data are shown in Supporting Information
Figure S10. (b) BU administration did not affect the ratio of Act-/Rest-microglia. Data (n = 4) are shown as means  SD. Statistical analyses using
ANOVA and Tukey’s post hoc test (a,b) showed significant differences (**, p < .01) compared with macrophage or Act-MG numbers in vehicle-
treated TBI brains, respectively. (c) CD86+ and CD206+ macrophages in the injured sites. BU administration weakly decreased the number of
CD86+ macrophages and increased the number of CD206+ macrophages. (d) BU administration did not affect the number of NG2+ macrophages.
BU-induced changes in mRNA levels and IGF-1 content in TBI brains at 7 dpi. (e) Changes in mRNA levels in injured tissues at 7 dpi as revealed
by qPCR. BU administration for 7 days reduced levels of mRNAs encoding CCL2, CCR2, and iNOS, and increased those encoding Iba1, NG2,
GFAP, and IGF-1. BU administration did not affect TGFβ1 or PCNA mRNA levels. (k) BU did not affect IGF-1 protein contents in injured brain
tissues. Data analyzed with Student’s t test (c–k) are shown as means  SEM (n = 4 for [c,d]; n = 5 for [e,k]. *, p < .05, ** p < .01, *** p < .001
versus vehicle [Color figure can be viewed at wileyonlinelibrary.com]

Macrophages, and Act-/Rest-MG were thus individually characterized BU are, therefore, likely to play aggravating roles in TBI. Based on
to evaluate their contribution to TBI pathogenesis. Total RNA was these proposals, we compared the pathophysiological contributions of
purified from sorted cells and mRNA level was standardized with microglia and macrophages to TBI.
GAPDH mRNA to evaluate the changes in the nature of cells but not
accumulated cell numbers.
Because BU had marked ameliorative actions on TBI brains, it
4.2 | Detrimental factors derived from macrophages
may be possible to infer the pathogenic mechanisms in TBI from the
and microglia
BU-induced changes. For example, BU reduced the generation of BU-mediated reduction in 8-OHdG generation in TBI lesions may be
8-OHdG in TBI brains; therefore, oxidative injury may play a deleteri- attributable to reduced macrophage accumulation as well as inhibi-
ous role in TBI, as has been proposed (Choi et al., 2012; Corps et al., tion of their mitROS generation. Rest- and Act-MG generated much
2015; Hall, Wang, & Miller, 2012; Zhang et al., 2012). Proinflamma- less amount of mitROS than macrophages. mitROS, which are 90%
tory mediators, whose expression and/or release were suppressed by of total cellular ROS (Gao et al., 2008), may be the main cause of
ABE ET AL. 13

oxidative stress-induced brain injury (Chouchani et al., 2014). NOX2 phagocytic activities (Matsumoto et al., 2008; Ritzel et al., 2015), mac-
is another source for ROS (Zhang et al., 2012), and it was more rophages may have stronger phagocytic activity than microglia
highly expressed by macrophages than microglia. Therefore, macro- because of their higher expression of CD68. Phagocytic elimination of
phages may be the major cell type responsible for the oxidative degenerated materials in injured brains may enhance regeneration
stress in TBI. However, BU did not affect NOX2 or p40phox and (Neumann, Kotter, & Franklin, 2009). On the other hand, it has also
p67phox mRNA levels in macrophages, indicating that NOX2 may be been insisted that the phagocytes accelerate neuronal death by elimi-
less critical for the oxidative stress. Macrophages expressed iNOS nating viable neurons (Neher et al., 2013; Vilalta & Brown, 2017). BU
mRNA at higher levels than microglia. iNOS-derived NO may play a did not affect the expression of CD68 by macrophages and microglia,
neuroprotective role (Sinz et al., 1999), but it can still be deleterious suggesting that phagocytosis may have both beneficial and detrimen-
because it forms highly toxic peroxynitrite (ONOO−) by reacting with tal effects on injured brains.
superoxide (Hall et al., 2012).
IL-1β expression is strongly increased after TBI and plays a central
role in enhancement of proinflammatory responses by increasing 4.4 | Neutrophils in the acute and macrophages
blood brain barrier permeability, immune cell recruitment, and resident in the subacute phases
glial cell activation (McKee & Lukens, 2016). Macrophages expressed
BU did not significantly affect accumulation of granulocytes, almost all
IL-1β mRNA at a higher level than microglia. BU inhibited IL-1β mRNA
of which were presumably neutrophils. Furthermore, BU did not
expression in TBI lesions and macrophages. IL-1β is produced as an
inhibit neutrophil mitROS generation. Neutrophils massively accumu-
inactive form that requires cleavage before its secretion by caspase-1,
late in the core lesion within hours after the onset of TBI (Gyoneva &
whose activation is regulated by inflammasomes (Latz, 2010). Endoge-
Ransohoff, 2015; McKee & Lukens, 2016), causing blood brain barrier
nous Toll-like receptor (TLR) ligands, referred to as damage-associated
molecular patterns (DAMPs) (Shichita et al., 2012; Webster et al., breakdown and brain edema. However, blockade of neutrophil entry

2017) induce mitROS generation (West et al., 2011), which increases into TBI lesions or genetic deletion/pharmacological inhibition of neu-

IL-1β release by activation of the major inflammasome, the trophil elastase does not lead to marked amelioration of TBI (Semple,
nucleotide-binding oligomerization domain-like receptor family, pyrin Bye, Ziebell, & Morganti-Kossmann, 2010; Semple, Trivedi, Gimlin, &
domain-containing 3 inflammasome (Garaude et al., 2016; Heid et al., Noble-Haeusslein, 2015; Weaver, Branch, Hernandez, Miller, & Quat-
2013). BU reduced mitROS generation by macrophages. Therefore, trocchi, 2000). The weak effects of BU on neutrophils also suggest
BU presumably reduced IL-1β actions by two ways; inhibition of that neutrophils may not be critical cells for secondary neuroinflam-
DAMPs-mediated mRNA expression and reduced mitROS-induced matory aggravation of TBI.
secretion. These observations suggest that macrophages but not Act- The high-level of mitROS may cause 8-OHdG generation in mac-
MG play a central aggravating role in TBI. rophages and their subsequent oxidative stress-induced degeneration,
likely leading to the decreased number of macrophages at 7 dpi. Fur-

4.3 | Favorable factors derived from macrophages thermore, iNOS mRNA levels were also higher in macrophages, and

and microglia the generated NO may produce peroxynitrite (Hall et al., 2012), which
would affect their viability. Although macrophages expressed GPx
Act- and Rest-MG expressed TNFα, IL-6, and TGFβ1 mRNA at higher
mRNA at very high levels, the scavenging effect of GPx may not be
levels than macrophages. Microglia and macrophages expressed IGF-1
sufficient to protect macrophages. Different from those present at 1.5
mRNA at similar levels. Although both TNFα and IL-6 are classified as
dpi, macrophages at 7 dpi may be ameliorative, given their polarization
proinflammatory cytokines, they have also been reported to exert
toward the M2 phenotype and previous reports describing ameliora-
neuroprotective actions (Lenzlinger, Morganti-Kossmann, Laurer, &
tive macrophages in injured brains that express IGF-1 and TGFβ1
McIntosh, 2001; Toku et al., 1998; Woodcock & Morganti-Kossmann,
(Nishihara et al., 2011; Smirkin et al., 2010; Sugimoto et al., 2014;
2013). However, BU suppressed the expression of TNFα and IL-6,
Wattananit et al., 2016).
indicating that they have some detrimental roles. BU did not affect
The majority of studies that inhibit CCR2-CCL2 interactions in
the expression of TGFβ1, which has an ameliorative action in ischemic
TBI models to prevent the infiltration of macrophages have shown the
and hemorrhagic brains (Taylor et al., 2017; Wattananit et al., 2016).
detrimental effects of monocyte-derived macrophages (Gyoneva
TGFβ1 inhibits TLR ligands-induced phosphorylation of IκB kinase in a
et al., 2015; Hsieh et al., 2014; Morganti et al., 2015; Semple, Koss-
sustained manner, leading to the sustained inhibition of NFκB translo-
cation into the nucleus (Islam et al., 2018). This action may be corre- mann, & Morganti-Kossmann, 2010; Song et al., 2017). However,

lated with the induction of anti-inflammatory macrophage and accumulated macrophages may be ameliorative at the subacute phase

microglia phenotypes in the subacute phase of severely damaged as shown here and also elsewhere (Chu et al., 2015; Miro-Mur et al.,
brain tissues (Islam et al., 2018; Taylor et al., 2017). The secretion of 2016; Smirkin et al., 2010; Wattananit et al., 2016). Regarding this,
favorable cytokines together with much less ROS and IL-1β produc- there should be two possibilities; phenotypic change into ameliorative
tion should make microglia more neuroprotective than macrophages, cells of monocyte-derived macrophages belonging to a single lineage
even when they are activated in TBI lesions. and the presence of two different lineages. According to the litera-
Although both microglia and macrophages have been implicated tures, the phenotypic change may be more probable (Miro-Mur et al.,
in removal of cell and tissue debris in injured brains through their 2016; Wattananit et al., 2016).
14 ABE ET AL.

4.5 | The BU actions on cell metabolism the Ministry of Education, Culture, Sports, Science, and Technology,
Japan (16 K20014 to SM; 17 K10836 to YK). We thank Jeremy Allen,
BU suppressed mitochondrial activities (basal, maximal and spare res-
PhD, from Edanz Group (www.edanzediting.com/ac) for editing a
piration, and ATP production) but not coupling efficacy, indicating that
draft of this manuscript.
BU suppressed mitochondrial activities without causing dysfunction.
Furthermore, BU also reduced ECAR. Taken together, BU appeared to
reduce overall cellular energy consumption, while suppressing mitROS CONFLIC T OF INT E RE ST
generation. Elevation of brain extracellular lactate concentration in
Nothing to declare.
TBI patients is correlated with poor outcomes (Carpenter, Jalloh, &
Hutchinson, 2015). The BU-induced decrease in ECAR implies a
decrease in lactate production or a reduced glycolysis rate. This BU RE FE RE NC ES
effect is likely correlated with improved outcome of the TBI model Ansari, M. A., Roberts, K. N., & Scheff, S. W. (2014). A time course of
NADPH-oxidase up-regulation and endothelial nitric oxide synthase
rats. Non-mitochondrial OCR is an indicator of the activities of
activation in the hippocampus following neurotrauma. Free Radical
hypoxia-inducible factor (HIF) prolyl hydroxylases, which consume Biology & Medicine, 77, 21–29.
α-ketoglutarate and O2 as co-substrates causing HIF ubiquitination Bachstetter, A. D., Rowe, R. K., Kaneko, M., Goulding, D., Lifshitz, J., & Van
(Banh et al., 2016). Because BU reduced the nonmitochondrial OCR, it Eldik, L. J. (2013). The p38alpha MAPK regulates microglial responsive-
ness to diffuse traumatic brain injury. The Journal of Neuroscience, 33,
might increase the levels of HIFs, which is possibly correlated with the
6143–6153.
BU-induced metabolic changes. Although the ameliorative effects of Banh, R. S., Iorio, C., Marcotte, R., Xu, Y., Cojocari, D., Rahman, A. A., …
BU on TBI may mainly be attributable to its antiinflammatory effects, Neel, B. G. (2016). PTP1B controls non-mitochondrial oxygen con-
sumption by regulating RNF213 to promote tumour survival during
BU might also have direct neuroprotective effects in TBI brains by
hypoxia. Nature Cell Biology, 18, 803–813.
decreasing neuronal cell metabolism, because hypothermia and Bennett, M. L., Bennett, F. C., Liddelow, S. A., Ajami, B., Zamanian, J. L.,
hypothyroidism-induced hypometabolic states exert neuroprotective Fernhoff, N. B., … Barres, B. A. (2016). New tools for studying microglia
effects (Rastogi et al., 2006; Robertson, Scafidi, McKenna, & Fiskum, in the mouse and human CNS. Proceedings of the National Academy of
Sciences of the United States of America, 113, E1738–E1746.
2009). Therefore, it is necessary to determine whether BU has a sig- Berman, J. W., Guida, M. P., Warren, J., Amat, J., & Brosnan, C. F. (1996).
nificant suppressive effect on neuronal cell metabolism. Localization of monocyte chemoattractant peptide-1 expression in the
central nervous system in experimental autoimmune encephalomyelitis
and trauma in the rat. Journal of Immunology, 156, 3017–3023.
Carpenter, K. L., Jalloh, I., & Hutchinson, P. J. (2015). Glycolysis and the
5 | CO NC LUSIO N
significance of lactate in traumatic brain injury. Frontiers in Neurosci-
ence, 9, 112.
By employing a combination of flow cytometry analyses, qPCR of iso- Choi, B. Y., Jang, B. G., Kim, J. H., Lee, B. E., Sohn, M., Song, H. K., &
lated cells, and immunohistochemistry, macrophages but not Act-MG Suh, S. W. (2012). Prevention of traumatic brain injury-induced neuro-
nal death by inhibition of NADPH oxidase activation. Brain Research,
were found to be a major source of ROS and IL-1β. However, such
1481, 49–58.
detrimental macrophages may undergo degeneration due to their own Chouchani, E. T., Pell, V. R., Gaude, E., Aksentijevic, D., Sundier, S. Y.,
ROS generation during the acute phase. Later, they might turn into Robb, E. L., … Murphy, M. P. (2014). Ischaemic accumulation of succi-
ameliorating cells for brain tissues by producing neuroprotective fac- nate controls reperfusion injury through mitochondrial ROS. Nature,
515, 431–435.
tors. Granulocytes may play less significant roles in aggravating TBI
Chu, H. X., Broughton, B. R., Kim, H. A., Lee, S., Drummond, G. R., &
pathology. Resident microglia may be neuroprotective cells because Sobey, C. G. (2015). Evidence that Ly6C(hi) monocytes are protective
they produce less IL-1β, NOX2, and mitROS while expressing more in acute ischemic stroke by promoting M2 macrophage polarization.
Stroke, 46, 1929–1937.
TGFβ1 than macrophages and IGF-1 at a comparable level. These
Corps, K. N., Roth, T. L., & McGavern, D. B. (2015). Inflammation and neu-
results support the notion that prevention of CCL2-CCR2 interaction roprotection in traumatic brain injury. JAMA Neurology, 72, 355–362.
leads to amelioration of TBI by reducing macrophage accumulation in Dudvarski Stankovic, N., Teodorczyk, M., Ploen, R., Zipp, F., &
TBI brains in the acute phase. Reduction of energy metabolism in Schmidt, M. H. (2016). Microglia-blood vessel interactions: A
double-edged sword in brain pathologies. Acta Neuropathologica, 131,
macrophages and microglia likely leads to amelioration of TBI. BU is a 347–363.
strong candidate to achieve this. The BU dosage for in vivo study was Fisher, M., Feuerstein, G., Howells, D. W., Hurn, P. D., Kent, T. A.,
approximately 50 mg/kg/day, which is comparable to the maximum Savitz, S. I., & Lo, E. H. (2009). Update of the stroke therapy academic
industry roundtable preclinical recommendations. Stroke, 40,
clinical dose. BU administration in the acute phase would ameliorate
2244–2250.
the outcome of TBI by suppressing CCL2 expression and cellular Gao, L., Laude, K., & Cai, H. (2008). Mitochondrial pathophysiology, reac-
energy metabolism, resulting in reduced accumulation of macrophages tive oxygen species, and cardiovascular diseases. The Veterinary Clinics
and reduced oxidative and inflammatory injuries. of North America. Small Animal Practice, 38(137–155), vi.
Garaude, J., Acin-Perez, R., Martinez-Cano, S., Enamorado, M., Ugolini, M.,
Nistal-Villan, E., … Sancho, D. (2016). Mitochondrial respiratory-chain
adaptations in macrophages contribute to antibacterial host defense.
ACKNOWLEDGMENTS Nature Immunology, 17, 1037–1045.
Gyoneva, S., Kim, D., Katsumoto, A., Kokiko-Cochran, O. N., Lamb, B. T., &
We are grateful to the staff of the Animal Center, Ehime University,
Ransohoff, R. M. (2015). Ccr2 deletion dissociates cavity size and tau
for their careful and gentle handling of the animals. This study was pathology after mild traumatic brain injury. Journal of Neuroinflamma-
partly supported by grants from Ehime University (#0503003000) and tion, 12, 228.
ABE ET AL. 15

Gyoneva, S., & Ransohoff, R. M. (2015). Inflammatory reaction after trau- Morganti, J. M., Jopson, T. D., Liu, S., Riparip, L. K., Guandique, C. K.,
matic brain injury: Therapeutic potential of targeting cell-cell communi- Gupta, N., … Rosi, S. (2015). CCR2 antagonism alters brain macrophage
cation by chemokines. Trends in Pharmacological Sciences, 36, 471–480. polarization and ameliorates cognitive dysfunction induced by trau-
Hall, E. D., Wang, J. A., & Miller, D. M. (2012). Relationship of nitric oxide matic brain injury. The Journal of Neuroscience, 35, 748–760.
synthase induction to peroxynitrite-mediated oxidative damage during Neher, J. J., Emmrich, J. V., Fricker, M., Mander, P. K., Thery, C., &
the first week after experimental traumatic brain injury. Experimental Brown, G. C. (2013). Phagocytosis executes delayed neuronal death
Neurology, 238, 176–182. after focal brain ischemia. Proceedings of the National Academy of Sci-
Hattori, T., Kaji, M., Ishii, H., Jureepon, R., Takarada-Iemata, M., Minh ences of the United States of America, 110, E4098–E4107.
Ta, H., … Hori, O. (2017). CD38 positively regulates postnatal develop- Nell, V., & Brown, D. S. (1991). Epidemiology of traumatic brain injury in
ment of astrocytes cell-autonomously and oligodendrocytes Johannesburg--II. Morbidity, mortality and etiology. Social Science &
non-cell-autonomously. Glia, 65, 974–989. Medicine, 33, 289–296.
Heid, M. E., Keyel, P. A., Kamga, C., Shiva, S., Watkins, S. C., & Salter, R. D. Neumann, H., Kotter, M. R., & Franklin, R. J. (2009). Debris clearance by
(2013). Mitochondrial reactive oxygen species induces microglia: An essential link between degeneration and regeneration.
NLRP3-dependent lysosomal damage and inflammasome activation. Brain, 132, 288–295.
Journal of Immunology, 191, 5230–5238. Nishihara, T., Ochi, M., Sugimoto, K., Takahashi, H., Yano, H., Kumon, Y., …
Higaki, H., Choudhury, M. E., Kawamoto, C., Miyamoto, K., Islam, A., Tanaka, J. (2011). Subcutaneous injection containing IL-3 and GM-CSF
Ishii, Y., … Tanaka, J. (2016). The hypnotic bromovalerylurea amelio- ameliorates stab wound-induced brain injury in rats. Experimental Neu-
rates 6-hydroxydopamine-induced dopaminergic neuron loss while rology, 229, 507–516.
suppressing expression of interferon regulatory factors by microglia. Nishioka, R., Sugimoto, K., Aono, H., Mise, A., Choudhury, M. E.,
Neurochemistry International, 99, 158–168. Miyanishi, K., … Tanaka, J. (2016). Treadmill exercise ameliorates
Hsieh, C. L., Niemi, E. C., Wang, S. H., Lee, C. C., Bingham, D., Zhang, J., … ischemia-induced brain edema while suppressing Na(+)/H(+) exchanger
Nakamura, M. C. (2014). CCR2 deficiency impairs macrophage infiltra- 1 expression. Experimental Neurology, 277, 150–161.
tion and improves cognitive function after traumatic brain injury. Jour- Patel, A. R., Ritzel, R., McCullough, L. D., & Liu, F. (2013). Microglia and
nal of Neurotrauma, 31, 1677–1688. ischemic stroke: A double-edged sword. International Journal of Physiol-
Islam, A., Choudhury, M. E., Kigami, Y., Utsunomiya, R., Matsumoto, S., ogy, Pathophysiology, and Pharmacology, 5, 73–90.
Watanabe, H., … Tanaka, J. (2018). Sustained anti-inflammatory effects Rastogi, L., Godbole, M. M., Ray, M., Rathore, P., Rathore, P., Pradhan, S.,
of TGF-beta1 on microglia/macrophages. Biochimica et Biophysica Acta, … Pandey, C. M. (2006). Reduction in oxidative stress and cell death
1864, 721–734.
explains hypothyroidism induced neuroprotection subsequent to
Karve, I. P., Taylor, J. M., & Crack, P. J. (2016). The contribution of astro-
ischemia/reperfusion insult. Experimental Neurology, 200, 290–300.
cytes and microglia to traumatic brain injury. British Journal of Pharma-
Ritzel, R. M., Patel, A. R., Grenier, J. M., Crapser, J., Verma, R.,
cology, 173, 692–702.
Jellison, E. R., & McCullough, L. D. (2015). Functional differences
Kawasaki, S., Abe, N., Ohtake, F., Islam, A., Choudhury, M. E.,
between microglia and monocytes after ischemic stroke. Journal of
Utsunomiya, R., … Tanaka, J. (2017). Effects of hypnotic bromovalery-
Neuroinflammation, 12, 106.
lurea on microglial BV2 cells. Journal of Pharmacological Sciences, 134,
Robertson, C. L., Scafidi, S., McKenna, M. C., & Fiskum, G. (2009). Mito-
116–123.
chondrial mechanisms of cell death and neuroprotection in pediatric
Kikuchi, S., Nishihara, T., Kawasaki, S., Abe, N., Kuwabara, J.,
ischemic and traumatic brain injury. Experimental Neurology, 218,
Choudhury, M. E., … Tanaka, J. (2015). The ameliorative effects of a
371–380.
hypnotic bromvalerylurea in sepsis. Biochemical and Biophysical
Roth, T. L., Nayak, D., Atanasijevic, T., Koretsky, A. P., Latour, L. L., &
Research Communications, 459, 319–326.
McGavern, D. B. (2014). Transcranial amelioration of inflammation and
Konishi, H., Kobayashi, M., Kunisawa, T., Imai, K., Sayo, A., Malissen, B., …
cell death after brain injury. Nature, 505, 223–228.
Kiyama, H. (2017). Siglec-H is a microglia-specific marker that discrimi-
Satoh, J., Kino, Y., Asahina, N., Takitani, M., Miyoshi, J., Ishida, T., &
nates microglia from CNS-associated macrophages and
Saito, Y. (2016). TMEM119 marks a subset of microglia in the human
CNS-infiltrating monocytes. Glia, 65, 1927–1943.
brain. Neuropathology, 36, 39–49.
Lashgari, R., Motamedi, F., Zahedi Asl, S., Shahidi, S., & Komaki, A. (2006).
Semple, B. D., Bye, N., Rancan, M., Ziebell, J. M., &
Behavioral and electrophysiological studies of chronic oral administra-
Morganti-Kossmann, M. C. (2010). Role of CCL2 (MCP-1) in traumatic
tion of L-type calcium channel blocker verapamil on learning and mem-
ory in rats. Behavioural Brain Research, 171, 324–328. brain injury (TBI): Evidence from severe TBI patients and CCL2−/−
Latz, E. (2010). NOX-free inflammasome activation. Blood, 116, mice. Journal of Cerebral Blood Flow and Metabolism, 30, 769–782.
1393–1394. Semple, B. D., Bye, N., Ziebell, J. M., & Morganti-Kossmann, M. C. (2010).
Lenzlinger, P. M., Morganti-Kossmann, M. C., Laurer, H. L., & Deficiency of the chemokine receptor CXCR2 attenuates neutrophil
McIntosh, T. K. (2001). The duality of the inflammatory response to infiltration and cortical damage following closed head injury. Neurobiol-
traumatic brain injury. Molecular Neurobiology, 24, 169–181. ogy of Disease, 40, 394–403.
Liu, S., Zhang, L., Wu, Q., Wu, Q., & Wang, T. (2013). Chemokine CCL2 Semple, B. D., Kossmann, T., & Morganti-Kossmann, M. C. (2010). Role of
induces apoptosis in cortex following traumatic brain injury. Journal of chemokines in CNS health and pathology: A focus on the CCL2/CCR2
Molecular Neuroscience, 51, 1021–1029. and CXCL8/CXCR2 networks. Journal of Cerebral Blood Flow and
Matsumoto, H., Kumon, Y., Watanabe, H., Ohnishi, T., Shudou, M., Metabolism, 30, 459–473.
Chuai, M., … Tanaka, J. (2008). Accumulation of macrophage-like cells Semple, B. D., Trivedi, A., Gimlin, K., & Noble-Haeusslein, L. J. (2015). Neu-
expressing NG2 proteoglycan and Iba1 in ischemic core of rat brain trophil elastase mediates acute pathogenesis and is a determinant of
after transient middle cerebral artery occlusion. Journal of Cerebral long-term behavioral recovery after traumatic injury to the immature
Blood Flow and Metabolism, 28, 149–163. brain. Neurobiology of Disease, 74, 263–280.
Matsumoto, H., Kumon, Y., Watanabe, H., Ohnishi, T., Shudou, M., Ii, C., … Serrano-Nascimento, C., da Silva, T. S., Nicola, J. P., Nachbar, R. T.,
Tanaka, J. (2007). Antibodies to CD11b, CD68, and lectin label neutro- Masini-Repiso, A. M., & Nunes, M. T. (2014). The acute inhibitory
phils rather than microglia in traumatic and ischemic brain lesions. Jour- effect of iodide excess on sodium/iodide symporter expression and
nal of Neuroscience Research, 85, 994–1009. activity involves the PI3K/Akt signaling pathway. Endocrinology, 155,
McKee, C. A., & Lukens, J. R. (2016). Emerging roles for the immune sys- 1145–1156.
tem in traumatic brain injury. Frontiers in Immunology, 7, 556. Shichita, T., Hasegawa, E., Kimura, A., Morita, R., Sakaguchi, R., Takada, I.,
Miro-Mur, F., Perez-de-Puig, I., Ferrer-Ferrer, M., Urra, X., Justicia, C., … Yoshimura, A. (2012). Peroxiredoxin family proteins are key initiators
Chamorro, A., & Planas, A. M. (2016). Immature monocytes recruited of post-ischemic inflammation in the brain. Nature Medicine, 18,
to the ischemic mouse brain differentiate into macrophages with fea- 911–917.
tures of alternative activation. Brain, Behavior, and Immunity, 53, Sinz, E. H., Kochanek, P. M., Dixon, C. E., Clark, R. S., Carcillo, J. A.,
18–33. Schiding, J. K., … Billiar, T. R. (1999). Inducible nitric oxide synthase is
16 ABE ET AL.

an endogenous neuroprotectant after traumatic brain injury in rats and Wattananit, S., Tornero, D., Graubardt, N., Memanishvili, T., Monni, E.,
mice. The Journal of Clinical Investigation, 104, 647–656. Tatarishvili, J., … Kokaia, Z. (2016). Monocyte-derived macrophages
Smirkin, A., Matsumoto, H., Takahashi, H., Inoue, A., Tagawa, M., Ohue, S., contribute to spontaneous long-term functional recovery after stroke
… Tanaka, J. (2010). Iba1(+)/NG2(+) macrophage-like cells expressing a in mice. The Journal of Neuroscience, 36, 4182–4195.
variety of neuroprotective factors ameliorate ischemic damage of the Weaver, K. D., Branch, C. A., Hernandez, L., Miller, C. H., &
brain. Journal of Cerebral Blood Flow and Metabolism, 30, 603–615. Quattrocchi, K. B. (2000). Effect of leukocyte-endothelial adhesion
Song, L., Lee, C., & Schindler, C. (2011). Deletion of the murine scavenger antagonism on neutrophil migration and neurologic outcome after cor-
receptor CD68. Journal of Lipid Research, 52, 1542–1550. tical trauma. The Journal of Trauma, 48, 1081–1090.
Song, S., Kong, X., Acosta, S., Sava, V., Borlongan, C. V., & Sanchez-Ramos, J. Webster, K. M., Sun, M., Crack, P., O’Brien, T. J., Shultz, S. R., &
(2017). Effects of an inhibitor of monocyte recruitment on recovery from Semple, B. D. (2017). Inflammation in epileptogenesis after traumatic
traumatic brain injury in mice treated with granulocyte Colony-stimulating brain injury. Journal of Neuroinflammation, 14, 10.
factor. International Journal of Molecular Science, 18, E1418. West, A. P., Brodsky, I. E., Rahner, C., Woo, D. K., Erdjument-Bromage, H.,
Sugimoto, K., Nishioka, R., Ikeda, A., Mise, A., Takahashi, H., Yano, H., … Tempst, P., … Ghosh, S. (2011). TLR signalling augments macrophage
Tanaka, J. (2014). Activated microglia in a rat stroke model express bactericidal activity through mitochondrial ROS. Nature, 472,
NG2 proteoglycan in peri-infarct tissue through the involvement of 476–480.
TGF-beta1. Glia, 62, 185–198. Woodcock, T., & Morganti-Kossmann, M. C. (2013). The role of markers of
Tanaka, J., Toku, K., Matsuda, S., Sudo, S., Fujita, H., Sakanaka, M., … inflammation in traumatic brain injury. Frontiers in Neurology, 4, 18.
Maeda, N. (1998). Induction of resting microglia in culture medium Yokoyama, A., Sakamoto, A., Kameda, K., Imai, Y., & Tanaka, J. (2006). NG2
devoid of glycine and serine. Glia, 24, 198–215. proteoglycan-expressing microglia as multipotent neural progenitors in
Taylor, R. A., Chang, C. F., Goods, B. A., Hammond, M. D., Mac Grory, B., normal and pathologic brains. Glia, 53, 754–768.
Ai, Y., … Sansing, L. H. (2017). TGF-beta1 modulates microglial pheno- Zhang, Q. G., Laird, M. D., Han, D., Nguyen, K., Scott, E., Dong, Y., …
type and promotes recovery after intracerebral hemorrhage. The Jour- Brann, D. W. (2012). Critical role of NADPH oxidase in neuronal oxida-
nal of Clinical Investigation, 127, 280–292. tive damage and microglia activation following traumatic brain injury.
Tei, N., Tanaka, J., Sugimoto, K., Nishihara, T., Nishioka, R., Takahashi, H., PLoS One, 7, e34504.
… Ohnishi, T. (2013). Expression of MCP-1 and fractalkine on endothe-
SUPPOR TI NG I NFORMATION
lial cells and astrocytes may contribute to the invasion and migration
of brain macrophages in ischemic rat brain lesions. Journal of Neurosci- Additional supporting information may be found online in the Sup-
ence Research, 91, 681–693.
porting Information section at the end of the article.
Toku, K., Tanaka, J., Yano, H., Desaki, J., Zhang, B., Yang, L., … Maeda, N.
(1998). Microglial cells prevent nitric oxide-induced neuronal apoptosis
in vitro. Journal of Neuroscience Research, 53, 415–425.
Valavanidis, A., Vlachogianni, T., & Fiotakis, C. (2009). 8-hydroxy-2’
How to cite this article: Abe N, Choudhury ME,
-deoxyguanosine (8-OHdG): A critical biomarker of oxidative stress Watanabe M, et al. Comparison of the detrimental features of
and carcinogenesis. Journal of Environmental Science and Health. Part C, microglia and infiltrated macrophages in traumatic brain injury:
Environmental Carcinogenesis & Ecotoxicology Reviews, 27, 120–139.
A study using a hypnotic bromovalerylurea. Glia. 2018;1–16.
Vilalta, A., & Brown, G. C. (2017). Neurophagy, the phagocytosis of live
neurons and synapses by glia, contributes to brain development and https://doi.org/10.1002/glia.23469
disease. The FEBS Journal. https://doi.org/10.1111/febs.14323

You might also like