You are on page 1of 8

Biochemical and Biophysical Research Communications 345 (2006) 1446–1453

BBRC
www.elsevier.com/locate/ybbrc

Central role of endogenous Toll-like receptor-2 activation in regulating


inflammation, reactive oxygen species production, and subsequent
neointimal formation after vascular injury
Tetsuro Shishido a,b,*, Naoki Nozaki a, Hiroki Takahashi a, Takanori Arimoto a,
Takeshi Niizeki a, Yo Koyama a, Jun-ichi Abe b, Yasuchika Takeishi a, Isao Kubota a

a
From Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
b
Center for Cardiovascular Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA

Received 25 April 2006


Available online 16 May 2006

Abstract

Background: It is now evident that inflammation after vascular injury has significant impact on the restenosis after revascularization
procedures such as angioplasty, stenting, and bypass grafting. However, the mechanisms that regulate inflammation and repair after vas-
cular injury are incompletely understood. Here, we report that vascular injury-mediated cytokine expression, reactive oxygen species
(ROS) production, as well as subsequent neointimal formation requires Toll-like receptor-2 (TLR-2) mediated signaling pathway in vivo.
Methods and results: Vascular injury was induced by cuff-placement around the femoral artery in non-transgenic littermates (NLC)
and TLR-2 knockout (TLR-2KO) mice. After cuff-placement in NLC mice, expression of TLR-2 was significantly increased in both
smooth muscle medial layer and adventitia. Interestingly, we found that inflammatory genes expression such as tumor necrosis fac-
tor-a, interleukin-1b (IL-1b), IL-6, and monocyte chemoattractant protein-1 were markedly decreased in TLR-2KO mice compared with
NLC mice. In addition, ROS production after vascular injury was attenuated in TLR-2KO mice compared with NLC mice. Since we
observed the significant role of endogenous TLR-2 activation in regulating inflammatory responses and ROS production after vascular
injury, we determined whether inhibition of endogenous TLR-2 activation can inhibit neointimal proliferation after vascular injury.
Neointimal hyperplasia was markedly suppressed in TLR-2KO mice compared with WT mice at both 2 and 4 weeks after vascular injury.
Conclusions: These findings suggested that endogenous TLR-2 activation might play a central role in the regulation of vascular
inflammation as well as subsequent neointimal formation in injured vessels.
 2006 Elsevier Inc. All rights reserved.

Keywords: Immune system; Inflammation; ROS; Toll-like receptor; Vascular injury

There is increasing evidence that inflammation of blood TNF-a, IL-1, IL-6, and MCP-1 remains unclear. Especial-
vessel plays a critical role during the initiation and mainte- ly, the ‘‘complexity’’ of inflammatory process in cardiovas-
nance of vascular diseases such as atherosclerosis forma- cular disease received attention after the data that
tion and restenosis after stent implantation and randomized and double-blind clinical study of infliximab
angioplasty. A number of inflammatory factors were sug- and etanercept, which are antagonists of TNF-a, worsened
gested to play a critical role in the regulation of the process the heart failure dose-dependently, even though many ani-
of injury and repair after vascular injury, but the exact role mal studies have shown the benefits in heart. While, the
and function of each inflammatory molecules including benefits of antagonizing TNF-a on neointimal formation
after vascular injury [1] or vascular smooth muscle prolifer-
*
Corresponding author. Fax: +1 585 273 3156.
ation in vein graft culture have also been reported [2]. In
E-mail address: Tetsuro_Shishido@URMC.Rochester.edu (T. Shishi- contrast, another report showed that TNF-a increased
do). inflammatory responses, but had no effect on neointimal

0006-291X/$ - see front matter  2006 Elsevier Inc. All rights reserved.
doi:10.1016/j.bbrc.2006.05.056
T. Shishido et al. / Biochemical and Biophysical Research Communications 345 (2006) 1446–1453 1447

formation after vascular injury [3]. These data support the reactive oxygen species (ROS) in injured vessels. Finally,
idea that TNF-a may not be the ‘‘solo’’ factor to regulate although we did not treat the vessels with TLR-2 ligand
inflammatory responses in the vessels and suggest the par- in our injury model, we observed marked decrease of
ticipation of other critical factor to regulate this vascular neointimal formation in TLR-2 knockout mice compared
injury process. with NLC, which is opposite to the lung injury model as
During the search of critical and responsible inflamma- previously described [7]. To our knowledge, this is the
tory response and signaling in vascular injury, we became first report to show the critical role of ‘‘endogenous’’
interested in Toll-like receptors. Toll-like receptors (TLRs) TLR-2 activation in injured vessel to regulate cytokine
have IL-1 receptor-like intracellular signaling pathways production, ROS production as well as subsequent neo-
and are widely accepted as a key receptor for immune intimal formation. Based on our current data, TLR-2
response. Activation of TLR pathway leads to nuclear could regulate variety of cytokines production including
localization of NF-jB/Rel-type transcription factor and TNF-a, IL-1b, IL-6, and MCP-1, and also ROS produc-
gene expressions of pro-inflammatory cytokine [4]. Until tion after vascular injury. These data suggest the central
now thirteen receptors have been identified with different role of endogenous TLR-2 activation after vascular inju-
or overlapping ligands. TLR-1 and 2 heterodimers can rec- ry, and inhibition of TLR-2 may provide a novel and
ognize bacterial lipoproteins, TLR-4 recognizes LPS from stronger therapeutic approach to inhibit vascular injury
Gram-negative bacteria, and TLR-9 recognizes bacterial than antagonizing TNF-a.
DNA (unmethylated CG motifs). In addition, other studies
have demonstrated that TLR-mediated pathways are also Materials and methods
activated by endogenous ligands, such as heat-shock pro-
tein and oxidative stress [5,6]. Interestingly, recent studies Animal treatment. The TLR-2 knockout (KO) mice were kindly sup-
showed that TLR-2 and 4 signal in response to the extracel- plied from Drs. Shizuo Akira and Osamu Takeuchi (Osaka University,
lular matrix component hyaluronan, which fragments Osaka, Japan) [10]. Male TLR-2 KO mice and normal wild-type (WT)
when lungs are injured [7]. TLR-2 and 4 drive inflamma- mice with same C57BL/6 background were used. Mice were housed in a
facility with a 12/12 light and dark cycle, and were given free access to
tion in response to the fragment form of hyaluronan. How- water and standard rodent chow. The room was kept specific pathogen-
ever, although TLR-2 and 4 double knock out mice free. The animals were handled according to the animal welfare regula-
markedly inhibited inflammatory response in bleomycin- tions of Yamagata University, and the study protocol was approved by
mediated lung injury, lung injury of interstitial thickness the Animal Subjects Committee of Yamagata University. The investiga-
tion conforms to the Guide for the Care and Use of Laboratory Animals
in lung tissue and protein accumulation in bronchoalveolar
published by the US National Institute of Health. The mice were anes-
lavage fluid in double knockout mice were significantly thetized with ketamine (70 mg/kg) and xylazine (7 mg/kg) by intraperi-
increased. These data suggest that inflammation is not a toneal injection. The femoral artery of the mouse was isolated from the
necessary ‘‘bad’’ process, and also possesses a ‘‘good’’ side. surrounding tissues, and a polyethylene cuff tube (2 mm long PE-90; inner
For example, inflammation counteracts infection and has a diameter, 0.86 mm; outer diameter, 1.27 mm; Becton–Dickinson, USA)
role to repair tissue injury. Therefore, the balance between was loosely placed around the artery and closed with suture as described
previously [11].
injury and repair during inflammation may decide the Physiological analysis. Body weight, blood pressure, and heart rate
‘‘fate’’ of the process of the disease [8]. were measured before and 4 weeks after cuff-placement. Blood pressure
Previously, Schoneveld et al. have reported that ‘‘exog- and heart rate were recorded by photoelectric oscillometric method (UR-
enous’’ ligand stimulation of TLR-2 induced neointimal 5000; Ueda Electric Works, Tokyo, Japan) in an awake state [12].
formation after vascular injury and atherosclerotic forma- Extraction of total RNAs and reverse transcriptase-polymerase chain
reaction (RT-PCR). RNA extraction and RT-PCR study were performed
tion, and these effects were reduced in TLR-2 knockout as described previously [13,14]. PCR primers for TLR-2 were 5 0 -GTC
mice [9]. Of note, the role of endogenous TLR-2 activation TCT GCG ACC TAG AAG TGG A-3 0 (forward) and 5 0 -CGG AGG
without the treatment of exogenous TLR-2 ligand in the GAA TAG AGG TGA AAG A-3 0 (reverse) (GenBank Accession No.
process of neointimal formation after vascular injury AF185284), primers for TNF-a were 5 0 -CAG CCT CTT CTC ATT CCT
GCT TGT G-3 0 (forward) and 5 0 -CTG GAA GAC TCC TCC CAG GTA
remains largely unknown. Since TLR-2 and 4 double
TAT-3 0 (reverse) (GenBank Accession No. X02611), primers for IL-1b
knockout mice enhanced lung injury, it is possible that were 5 0 -ATG GCA ACT GTT CCT GAA-3 0 (forward) and 5 0 -GTA CAA
the endogenous TLR-2 activation may have a protective AGC TCA TGG AGA-3 0 (reverse) (GenBank Accession No. BC011437),
effect and inhibit neointimal formation after vascular primers for IL-6 were 5 0 -TTC CCT ACT TCA CAA GTC-3 0 (forward)
injury. and 5 0 -GGT TTG CCG AGT AGA TCT-3 0 (reverse) (GenBank Acces-
To determine the role of endogenous TLR-2 activa- sion No. X54542), primers for MCP-1 were 5 0 - ACT GAA GCC AGC
TCT CTC TTC CTC-3 0 (forward) and 5 0 -TTC CTT CTT GGG GTC
tion after vascular injury, in this study we have created AGC ACA GAC-3 0 (reverse) (GenBank Accession No. J04467) and
vascular injury by cuff-placement onto the femoral artery primers for GAPDH were 5 0 -ACT CCA CTC ACG GCA AAT TCA
in non-transgenic littermate control (NLC) and TLR-2 ACG G-3 0 (forward) and 5 0 -AGG GGC GGA GAT GAT GAC CC-3 0
knockout mice. First we found that TLR-2 expression (reverse) (GenBank Accession No. M32599). The PCR products were
fractionated on a 1–2% agarose gel and visualized by ethidium bromide
was significantly increased after vascular injury. Next,
staining. The intensities of the bands were normalized for GAPDH and
we demonstrated that TLR-2 deficiency resulted in were expressed as fold increase over WT mice.
suppressed production of pro-inflammatory cytokines Immunohistochemistry. The frozen sections (4 lm thickness) were
including TNF-a, IL-1b, IL-6, and MCP-1, as well as immunohistochemically stained by the streptavidin–biotin–peroxidase
1448 T. Shishido et al. / Biochemical and Biophysical Research Communications 345 (2006) 1446–1453

method as described previously [11,15]. Briefly, endogenous peroxidase used cuff-placement as a vascular injury model, because
and the non-specific binding of the antibody were blocked with 0.3% the neointimal formation induced by this model is
hydrogen peroxide in methanol and 2% goat serum in PBS, respectively.
The antibody to TLR-2 or non-specific IgG (Santa Cruze Biotechnology,
very reliable and reproducible, especially in mice. Again,
Inc., Santa Cruz, CA, USA) in 1% BSA in PBS was applied to the sections we did not treat the vessels with lipopolysaccharide
and incubated for 16–24 h at 4 C. After washing, biotinylated secondary (TLR-4 ligand) or synthetic TLR-2 ligand, Pam3Cys-
antibody and then streptavidin conjugate were applied to each tissue SK4, which is different from the previous reports [9].
sections. Color development was performed with 3,3 0 -diaminobenzidine, Although we did not add TLR-2 ligand, expression of
and slides were counterstained with hematoxylin [15].
Measurement of superoxide generation by DHE staining. The oxi-
TLR-2 mRNA was significantly increased after one week
dative fluorescent dye dihydroethidium (DHE; Sigma–Aldrich, St. of cuff-mediated vascular injury, and maintained its
Louis, MO, USA) was used to measure in situ production of super- increase until four weeks after injury as shown in Fig. 1.
oxide. Fresh unfixed frozen 30 mm thick sections obtained from the To investigate the localization of TLR-2 expression after
cuffed femoral artery of mice were placed on a glass slide. DHE cuff-mediated vascular injury, we performed immunohisto-
(20 mmol/L) was applied topically to each tissue section, and the sec-
tions were then incubated at 37 C for 30 min in a humidified chamber
chemical staining in vessels after 1 week of vascular injury
protected from light. In the presence of O2  , DHE is converted to the with anti-TLR-2 polyclonal antibody and compared the
fluorescent molecule ethidium bromide, which binds to DNA in the TLR-2 expression with non-injured vessels as shown in
nucleus and fluoresces red. Slides were briefly washed in PBS and Fig. 2. In non-injured vessel we could not detect TLR-2
coverslips were placed over the sections. For quantification of vascular expression in both smooth muscle medial layer and
ethidium fluorescence, fluorescence (intensity · area) was measured only
on the vascular area including endothelial, intimal, and medial
adventitial cells. In contrast, although we did not observe
area [16]. significant neointimal formation after 1 week of injury
Tissue preparation and histology. At 2 and 4 weeks after cuff-placement,
the mice were killed by an overdose of the anesthesia, and thoracic aortas
were perfused with phosphate buffered saline (PBS) and subsequently with A Cuff +
4% formaldehyde. Paraffin sections of the femoral artery were stained with
either elastica-Gorldner or hematoxylin and eosin. With the use of com- Cuff - 1 week 4 weeks
puterized morphometric analysis, the intimal cross-sectional area between
the lumen and the internal elastic lamina and the medial area between the
internal and external elastic lamina were measured as previously reported
TLR-2
[11,17] using Scion image for windowsPlus software (Image, Scion Cor-
poration, MD, USA).
Statistics analysis. All values are expressed as means ± SEM. To
compare histological data and gene expression of TLR-2, one-way GAPDH
ANOVA followed by post hoc procedure was performed. The other data
were compared with Student’s t-test. Statistical significance was accepted
at a value of P < 0.05.
B 3 *
*
Results
TLR-2 expressions
(fold increase)

2
TLR-2 mRNA expression was markedly increased after
cuff-mediated vascular injury
1

There are contradictory data on TLR-2 expression in


atherosclerotic formation [13,14]. In contrast, TLR-ex- 0
pression after vascular injury remains unclear. Therefore,
ks
-

k
ee
ff

ee
Cu

first we investigated the time-dependent TLR-2 mRNA


1w

4w

expression after cuff-mediated vascular injury. At base-


line, body weight, heart rate, and blood pressure were Fig. 1. Effect of cuff-placement on expression of Toll-like receptor (TLR)-
similar between NLC and TLR-2 KO mice (data not 2 in femoral artery. (A) RT-PCR band of TLR-2 and GAPDH in wild-
type (WT) mice. (B) Results were quantified by densitometry and
shown) as previously reported [15,18]. At 4 weeks after
expressed as means ± SEM of six separate experiments. Each sample
cuff-placement, there were no significant differences in was obtained from 4–6 femoral arteries. Gene expressions of TLR-2 were
the body weight, heart rate, or blood pressure between increased 1 week after cuff-placement and subsequently slightly decreased
NLC and TLR-2 KO mice as shown in Table 1. We in cuff-injured artery in WT mice. *P < 0.05 vs. cuff.

Table 1
Physiological findings at 4 weeks after cuff-placement
Body weight (g) Heart rate (bpm) SBP (mmHg) DBP (mmHg)
WT mice 31.0 ± 0.4 556 ± 25 119 + 2 53 ± 4
TLR-2 KO mice 32.3 ± 0.9 564 ± 21 116 ± 1 52 ± 3
SBP and DBP indicate blood pressure in systolic and diastolic, respectively. All data are expressed as means ± SEM from six experiments.
T. Shishido et al. / Biochemical and Biophysical Research Communications 345 (2006) 1446–1453 1449

TLR-2 Control IgG

Fig. 2. Representative immunohistochemical photomicrographs of cross-sections of femoral arteries stained with antibody to TLR-2 (original
magnification 400·). After cuff-placement, TLR-2 immunoreactivity (brown staining) is markedly increased in cuffed-artery obtained from WT mice (left).
Right figure represents a negative control. Arrow and arrowhead indicate external elastic lamina and endothelium, respectively.

yet, we did observe significant increase of TLR-2 expres- superoxide production was markedly increased in injured
sion in both medial layer of smooth muscle cells and adven- vessels and adventitia (Fig. 4B). However, in TLR-2
titial cells. These data suggest that not only adventitial knockout mice superoxide production after one week of
fibroblasts, but also smooth muscle cells, could express a
functional TLR-2, which is different from TLR-4 expres- A Cuff 1W
sion [9].
TNF-α
Critical role of endogenous TLR-2 activation on
inflammatory genes expression after vascular injury
IL-1α
We examined inflammatory gene expressions of TNF-a,
IL-1b, IL-6, and MCP-1 after vascular injury. We selected
IL-6
one week after vascular injury to investigate these genes
expression, because (1) we and other group found that
growth rate of neointimal formation is maximum at one MCP-1
week after injury [11,19,20] the difference of cell compo-
nents in the injured vessels (smooth muscle vs. adventitial
GAPDH
cells) should not affect the genes expression results because
of limited amount of neointimal formation until one week
O
O
R- T

T
2K
2K
TL W

after vascular injury. After one week of cuff-placement,


R-
TL

TNF-a, IL-1b, IL-6, and MCP-1 mRNA expressions were


significantly increased in NLC mice, although we did not B TNF-α IL-1α IL-6 MCP-1
observe marked neointimal formation at this moment. 1.2
(Fold increase)

Interestingly, we found that all cytokine expressions, which 1.0


* * ** *
we investigated in this study, were significantly diminished 0.8
in TLR-2 knockout mice (Fig. 3). We did not treat any 0.6
0.4
exogenous TLR-2 ligand in the vessels, which is different
0.2
from the previous report [9], suggesting the central role
0
of endogenous TLR-2 activation on regulating cytokine
O
O

O
T

T
T

T
2K

2K

2K

2K
W

W
W

expressions after vascular injury.


R-

R-

R-

R-
TL

TL

TL

TL

ROS production after vascular injury was abolished in Fig. 3. (A) RT-PCR analysis for TNF-a, IL-1b, IL-6, and MCP-1
TLR-2 knockout mice mRNA. Cuffed-arteries at 1 week after operation and control intact
arteries were harvested and frozen. Each sample was obtained from 4–6
Several studies have demonstrated that ROS produc- arteries. One representative result of four independent RT-PCR is
shown. (B) Group data for relative band intensity of TNF-a, IL-1b,
tion was increased in cuff-injured artery [21]. In this
IL-6, and MCP-1. Expressions of pro-atherosclerotic genes were
study, we used dihydroethidium (DHE), an oxidative suppressed in TLR-2 KO mice compared with WT mice. All data are
fluorescent dye, to detect ROS production in injured expressed as means ± SEM from six experiments. *P < 0.05 and
vessels. The chemiluminescent signal attributable to **P < 0.01 vs. WT.
1450 T. Shishido et al. / Biochemical and Biophysical Research Communications 345 (2006) 1446–1453

A TLR-2 KO-
WT-Sham WT-Cuff Cuff

B TLR-2 KO-
WT-Cuff Cuff

vessel wall

adventitia

C 1.2
(Relative intensity)

1.0
Vascular DHE
fluorescence

0.8
0.6
*
0.4
0.2
0
O
T
W

2K
R-
TL

Fig. 4. In situ detection of superoxide by dihydroethidium fluorescence. (A) Cross-section of non-cuffed femoral artery obtained from control WT mice
and of cuffed-femoral artery in WT and TLR-2 KO mice. (Original magnification 100·). Although fluorescent detection was slight in non-cuffed artery,
dihydroethidium fluorescence was increased after cuff-placement. (B) Boxed region in (A) at vascular and adventitial area. (Original magnification 400·).
Arrow and arrowhead indicate external elastic lamina and endothelium, respectively. (C) Quantification of reactive oxygen species production in vascular
wall. Superoxide production from cuffed-artery was attenuated in TLR-2 KO mice compared with WT mice. Data are expressed as means ± SEM.
*P < 0.05 vs. WT.

injury was markedly suppressed compared with NLC TLR-2 KO mice after vascular injury was observed
mice (Fig. 4B and C). (Fig. 6A), and the ratio of the neointimal/medial area was
significantly attenuated at 2 and 4 week after vascular injury
Neointimal formation was markedly suppressed in TLR-2 in TLR-2 KO mice as shown in Fig. 6C (P < 0.05 at 2 weeks
knock out mice and P < 0.01 at 4 weeks after injury). These data suggest that
‘‘endogenous’’ TLR-2 activation is critical for neointimal
Since we found that both cytokine expressions and ROS formation after vascular injury.
production were significantly decreased in TLR-2 knockout
mice, we examined whether the deficiency of TLR-2 could Discussion
affect the neointimal formation after vascular injury,
although we did not add TLR-2 ligand. There was no obvi- In the present study, we described the important role of
ous difference in the morphological appearance of the medial endogenous TLR-2 activation on cytokine expression and
area and adventitial area between NLC and TLR-2 KO mice ROS production as well as subsequent neointimal forma-
in non-injured vessels (data not shown). After vascular tion after vascular injury. TLR-2 expression was signifi-
injury, we found significant neointimal formation after cantly increased in both medial layer of smooth muscle
vascular injury in NLC mice (Fig. 5, left). In contrast, the and adventitial cells after vascular injury. Interestingly,
neointimal formation was markedly suppressed in TLR-2 we found that the deficiency of TLR-2 markedly decreased
KO mice compared to NLC mice at 2 weeks (P < 0.05) and all cytokine expression, which we investigated, and ROS
also at 4 weeks (P < 0.01) after vascular injury (Fig. 5, right). production. Furthermore, we found that neointimal forma-
No significant difference in medial area between NLC and tion was significantly decreased in TLR-2 knockout mice
T. Shishido et al. / Biochemical and Biophysical Research Communications 345 (2006) 1446–1453 1451

WT TLR-2 KO

2 weeks

neointima

WT TLR-2 KO

4 weeks

neointima

Fig. 5. Histology of WT and TLR-2 KO mice. Representative histological photomicrographs of cross-sections of femoral arteries stained with elastica-
Gorldner in WT and TLR-2 KO mice are shown. Although neointimal hyperplasia was occurred in both WT and TLR-2 KO mouse, the neointimal
formation was inhibited in TLR-2 KO mouse compared with WT mouse. Scale bars, 100 lm.

compared to NLC mice. To our knowledge, these data pro- that this positive feedback loop of TLR-2 and NF-jB
vided the first direct evidence of the involvement of endog- worsened the inflammatory responses after vascular injury
enous TLR-2 activation in inflammation as well as (Fig. 7). In addition, we found that ROS production after
subsequent neointimal formation after vascular injury. vascular injury was also regulated by TLR-2 activation.
Based on this work and previous investigator’s data, we ROS also affects vascular inflammation and involves in
proposed a working model as shown in Fig. 7. Although cytokine production. Thus, it is also intriguing to hypoth-
the exact mechanism remains unclear, vascular injury esize the existence of direct and/or indirect positive feed-
activates endogenous TLR-2 activation. As previously back loop between TLR-2 activation and inflammatory
reported [4,10], TLR-2 activation can significantly increase responses in injured vessels (Fig. 7). We believe that these
NF-jB activation and subsequent cytokine production. positive feedback loops of TLR-2 signaling pathways have
While it is also reported that TLR-2 expression is strongly a central role in regulating inflammatory responses in
regulated by NF-jB activation [22]. Therefore, it is possible injured vessels. Therefore, the specific deletion of TLR-2
1452 T. Shishido et al. / Biochemical and Biophysical Research Communications 345 (2006) 1446–1453

A (μm2) vascular injury


18000
+
Medial Area

12000
oxidative stress TLR-2

6000

NF- B activation
+
0
2W

4W
W

+
2

O-

O-
T-

T-
W

2K

2K
W

inflammatory cytokine
B (μm2)
Fig. 7. Schematic representation of the TLR-2 mediated vascular inflam-
8000
mation. After vascular injury, many stimuli including oxidative stress
Neo-intimal Area

induce TLR-2 mediated inflammatory response at the injured artery.


Increase of inflammatory cytokine causes NF-jB activation, oxidative
stress production, and TLR-2 expressions. Up-regulated TLR-2 expres-
4000 sions elicit vascular inflammation prominently.
* **
vascular injury remains unclear. TLR-2 is activated by bac-
0
terial lipoproteins such as components of Gram-positive
2W

4W
W
W

bacteria [23], mycobacteria [24], and Borelia [25]. Recently,


4
2

O-

O-
T-
T-

Jiang et al. have reported that the fragment form of the


W

2K

2K
W

extracellular matrix component of hyaluronan activates


C TLR-2 and TLR-4 in epithelial cells [25]. Hyaluronan is a
60 massive sugar polymer and a key ‘‘shock-absorbing’’ mate-
rial in the extracellular matrix. During inflammation, hyal-
Intima / Media
Volume Ratio

uronidase breaks down hyaluronan, and generates pro-


40 inflammatory fragments of hyaluronan, which activates
NF-jB activation [8], and Jiang et al. reported that hyalu-
20
* ** ronan-mediated NF-jB activation was completely abol-
ished in TLR-4 and TLR-2 double knockout mice.
Therefore, the fragmentation of hyaluronan may involve
0 in endogenous TLR-2 activation after vascular injury. Fur-
ther investigation of the involvement of hyaluronan
2W

4W
2W

4W

fragmentation in vascular injury will be required.


O-

O-
T-

T-
W

2K

2K
W

Previously, Schoneveld et al. have reported that


Fig. 6. Comparison of the lesion formation after cuff-placement. Cross- Pam(3)Cys-SK(4), which is one of the synthetic TLR-2
sectional areas of media (A) and intima (B) were evaluated at 2 and 4 ligand, directly progresses neointimal formation and ath-
weeks after cuff-placement. There was no significant difference between erosclerotic plaque [9]. They also showed that, in vitro
WT and TLR-2 KO mice in medial area, however, neointimal hyperplasia
study using cultured fibroblasts, TLR-2 ligand induced
was suppressed in TLR-2 KO mice compared with WT mice at 2 and 4
weeks after cuff-injury. Cuff-injury increased the intimal/medial area ratio inflammatory cytokines, such as IL-1a, IL1b, IL-6, and
(C) in both WT and TLR-2 KO mice, however, TLR-2 KO mice had less IL-8. In the present study, we showed that productions
increases in intimal/medial area ratio than WT mice. *P < 0.05 and of pro-inflammatory cytokines, such as TNF-a, IL-1b,
**P < 0.01 vs. WT in same time point. Open and black bar represents WT IL-6, during vascular inflammation were suppressed in
and TLR-2 KO mice, respectively (n = 9, means ± SEM).
TLR-2 KO mice than in NLC mice. Moreover, ROS pro-
duction in the injured vessels was attenuated in TLR-2
expression can also inhibit other cytokines expression and KO mice compared with NLC mice. These findings indicat-
show a marked decrease of neointimal formation after ed that TLR-2 signaling pathway forms a central role in
vascular injury. Further investigation is necessary to deter- regulating inflammatory responses as well as subsequent
mine the role of these positive feedback loops in vascular neointimal formation, and may provide a novel and
injury. stronger therapeutic strategy to inhibit inflammatory
Although this study clearly shows the involvement of responses and subsequent neointimal formation than other
endogenous TLR-2 activation in neointimal formation, anti-inflammatory therapies such as dexamethazone or
the exact mechanism of endogenous TLR-2 activation after antagonizing TNF-a.
T. Shishido et al. / Biochemical and Biophysical Research Communications 345 (2006) 1446–1453 1453

Acknowledgments [12] H. Takahashi, Y. Takeishi, T. Miyamoto, T. Shishido, T. Arimoto, T.


Konta, T. Miyashita, M. Ito, I. Kubota, Protein kinase C and
extracellular signal regulated kinase are involved in cardiac hyper-
We thank Mr. Eiji Tsuchida, Ms. Shuku Takahashi, and trophy of rats with progressive renal injury, Eur. J. Clin. Invest. 34
Ms. Sachi Adachi for their excellent technical assistance. (2004) 85–93.
This study was supported in part by Grants-in-Aid for Sci- [13] K. Edfeldt, J. Swedenborg, G.K. Hansson, Z.Q. Yan, Expression
entific Research (Nos. 16590657 and 14570635) and for the of toll-like receptors in human atherosclerotic lesions: a possible
21st Century Center of Excellence (COE) program of the pathway for plaque activation, Circulation 105 (2002)
1158–1161.
Japan Society for the Promotion of Science (both from [14] F.C. Gibson 3rd, C. Hong, H.H. Chou, H. Yumoto, J. Chen, E. Lien,
the Ministry of Education, Science, Sports and Culture, J. Wong, C.A. Genco, Innate immune recognition of invasive bacteria
Japan) and grants from the Mochida Memorial Founda- accelerates atherosclerosis in apolipoprotein E-deficient mice, Circu-
tion and the Japan Heart Foundation. lation 109 (2004) 2801–2806.
[15] T. Shishido, N. Nozaki, S. Yamaguchi, Y. Shibata, J. Nitobe, T.
Miyamoto, H. Takahashi, T. Arimoto, K. Maeda, M. Yamakawa, O.
References Takeuchi, S. Akira, Y. Takeishi, I. Kubota, Toll-like receptor-2
modulates ventricular remodeling after myocardial infarction, Circu-
[1] A.M. Miller, A.R. McPhaden, A. Preston, R.M. Wadsworth, C.L. lation 108 (2003) 2905–2910.
Wainwright, TNFalpha increases the inflammatory response to [16] N.J. Alp, M.A. McAteer, J. Khoo, R.P. Choudhury, K.M. Channon,
vascular balloon injury without accelerating neointimal formation, Increased endothelial tetrahydrobiopterin synthesis by targeted
Atherosclerosis 179 (2005) 51–59. transgenic GTP-cyclohydrolase I overexpression reduces endothelial
[2] Q. Javed, N. Swanson, H. Vohra, H. Thurston, A.H. Gershlick, dysfunction and atherosclerosis in ApoE-knockout mice, Arterioscler.
Tumor necrosis factor-alpha antibody eluting stents reduce vascular Thromb. Vasc. Biol. 24 (2004) 445–450.
smooth muscle cell proliferation in saphenous vein organ culture, [17] A. Vink, A.H. Schoneveld, J.J. van der Meer, B.J. van
Exp. Mol. Pathol. 73 (2002) 104–111. Middelaar, J.P. Sluijter, M.B. Smeets, P.H. Quax, S.K. Lim, C.
[3] A. Niemann-Jonsson, M.P. Ares, Z.Q. Yan, D.X. Bu, G.N. Fredrik- Borst, G. Pasterkamp, D.P. de Kleijn, In vivo evidence for a role
son, L. Branen, I. Porn-Ares, A.H. Nilsson, J. Nilsson, Increased rate of toll-like receptor 4 in the development of intimal lesions,
of apoptosis in intimal arterial smooth muscle cells through endog- Circulation 106 (2002) 1985–1990.
enous activation of TNF receptors, Arterioscler. Thromb. Vasc. Biol. [18] N. Nozaki, T. Shishido, Y. Takeishi, I. Kubota, Modulation of
21 (2001) 1909–1914. doxorubicin-induced cardiac dysfunction in toll-like receptor-2-
[4] S. Akira, Toll-like receptor signaling, J. Biol. Chem. 278 (2003) knockout mice, Circulation 110 (2004) 2869–2874.
38105–38108. [19] H.W. Liu, M. Iwai, Y. Takeda-Matsubara, L. Wu, J.M. Li, M.
[5] R.M. Vabulas, P. Ahmad-Nejad, C. da Costa, T. Miethke, C.J. Okumura, T.X. Cui, M. Horiuchi, Effect of estrogen and AT1
Kirschning, H. Hacker, H. Wagner, Endocytosed HSP60s use toll-like receptor blocker on neointima formation, Hypertension 40 (2002)
receptor 2 (TLR2) and TLR4 to activate the toll/interleukin-1 451–457, discussion 448–450.
receptor signaling pathway in innate immune cells, J. Biol. Chem. [20] J. Suzuki, M. Iwai, H. Nakagami, L. Wu, R. Chen, T. Sugaya, M.
276 (2001) 31332–31339. Hamada, K. Hiwada, M. Horiuchi, Role of angiotensin II-regulated
[6] S. Frantz, R.A. Kelly, T. Bourcier, Role of TLR-2 in the activation of apoptosis through distinct AT1 and AT2 receptors in neointimal
nuclear factor kappaB by oxidative stress in cardiac myocytes, J. Biol. formation, Circulation 106 (2002) 847–853.
Chem. 276 (2001) 5197–5203. [21] J. Kawai, K. Ando, A. Tojo, T. Shimosawa, K. Takahashi, M.L.
[7] D. Jiang, J. Liang, J. Fan, S. Yu, S. Chen, Y. Luo, G.D. Prestwich, Onozato, M. Yamasaki, T. Ogita, T. Nakaoka, T. Fujita, Endoge-
M.M. Mascarenhas, H.G. Garg, D.A. Quinn, R.J. Homer, D.R. nous adrenomedullin protects against vascular response to injury in
Goldstein, R. Bucala, P.J. Lee, R. Medzhitov, P.W. Noble, Regula- mice, Circulation 109 (2004) 1147–1153.
tion of lung injury and repair by Toll-like receptors and hyaluronan, [22] S.J. Kammanadiminti, B.J. Mann, L. Dutil, K. Chadee, Regulation
Nat. Med. 11 (2005) 1173–1179. of Toll-like receptor-2 expression by the Gal-lectin of Entamoeba
[8] L.A. O’Neill, TLRs play good cop, bad cop in the lung, Nat. Med. 11 histolytica, FASEB J. 18 (2004) 155–157.
(2005) 1161–1162. [23] R. Schwandner, R. Dziarski, H. Wesche, M. Rothe, C.J. Kirsch-
[9] A.H. Schoneveld, M.M. Oude Nijhuis, B. van Middelaar, J.D. ning, Peptidoglycan- and lipoteichoic acid-induced cell activation is
Laman, D.P. de Kleijn, G. Pasterkamp, Toll-like receptor 2 stimu- mediated by toll-like receptor 2, J. Biol. Chem. 274 (1999)
lation induces intimal hyperplasia and atherosclerotic lesion devel- 17406–17409.
opment, Cardiovasc. Res. 66 (2005) 162–169. [24] H.D. Brightbill, D.H. Libraty, S.R. Krutzik, R.B. Yang, J.T. Belisle,
[10] O. Takeuchi, K. Hoshino, T. Kawai, H. Sanjo, H. Takada, T. Ogawa, J.R. Bleharski, M. Maitland, M.V. Norgard, S.E. Plevy, S.T. Smale,
K. Takeda, S. Akira, Differential roles of TLR2 and TLR4 in P.J. Brennan, B.R. Bloom, P.J. Godowski, R.L. Modlin, Host defense
recognition of Gram-negative and Gram-positive bacterial cell wall mechanisms triggered by microbial lipoproteins through toll-like
components, Immunity 11 (1999) 443–451. receptors, Science 285 (1999) 732–736.
[11] M. Akishita, M. Horiuchi, H. Yamada, L. Zhang, G. Shirakami, K. [25] M. Hirschfeld, C.J. Kirschning, R. Schwandner, H. Wesche, J.H.
Tamura, Y. Ouchi, V.J. Dzau, Inflammation influences vascular Weis, R.M. Wooten, J.J. Weis, Cutting edge: inflammatory signaling
remodeling through AT2 receptor expression and signaling, Physiol. by Borrelia burgdorferi lipoproteins is mediated by toll-like receptor
Genomics 2 (2000) 13–20. 2, J. Immunol. 163 (1999) 2382–2386.

You might also like