25.03.2013 Views

CX3CR1 Deficiency Confers Protection From Intimal Hyperplasia ...

CX3CR1 Deficiency Confers Protection From Intimal Hyperplasia ...

CX3CR1 Deficiency Confers Protection From Intimal Hyperplasia ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>CX3CR1</strong><br />

<strong>Deficiency</strong> <strong>Confers</strong> <strong>Protection</strong> <strong>From</strong> <strong>Intimal</strong> <strong>Hyperplasia</strong> After Arterial Injury<br />

Peng Liu, Sarita Patil, Mauricio Rojas, Alan M. Fong, Susan S. Smyth and Dhavalkumar D.<br />

Patel<br />

Arterioscler Thromb Vasc Biol. 2006;26:2056-2062; originally published online June 29, 2006;<br />

doi: 10.1161/01.ATV.0000234947.47788.8c<br />

Arteriosclerosis, Thrombosis, and Vascular Biology is published by the American Heart Association, 7272<br />

Greenville Avenue, Dallas, TX 75231<br />

Copyright © 2006 American Heart Association, Inc. All rights reserved.<br />

Print ISSN: 1079-5642. Online ISSN: 1524-4636<br />

The online version of this article, along with updated information and services, is located on the<br />

World Wide Web at:<br />

http://atvb.ahajournals.org/content/26/9/2056<br />

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published<br />

in Arteriosclerosis, Thrombosis, and Vascular Biology can be obtained via RightsLink, a service of the<br />

Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for<br />

which permission is being requested is located, click Request Permissions in the middle column of the Web<br />

page under Services. Further information about this process is available in the Permissions and Rights<br />

Question and Answer document.<br />

Reprints: Information about reprints can be found online at:<br />

http://www.lww.com/reprints<br />

Subscriptions: Information about subscribing to Arteriosclerosis, Thrombosis, and Vascular Biology is online<br />

at:<br />

http://atvb.ahajournals.org//subscriptions/<br />

Downloaded from<br />

http://atvb.ahajournals.org/ by guest on March 25, 2013


CX 3CR1 <strong>Deficiency</strong> <strong>Confers</strong> <strong>Protection</strong> <strong>From</strong> <strong>Intimal</strong><br />

<strong>Hyperplasia</strong> After Arterial Injury<br />

Peng Liu, Sarita Patil, Mauricio Rojas, Alan M. Fong, Susan S. Smyth, Dhavalkumar D. Patel<br />

Objective—A functional polymorphism in the chemokine receptor CX 3CR1 is associated with protection from vascular<br />

diseases including coronary artery disease and internal carotid artery occlusive disease. We investigated the mechanisms<br />

by which CX 3CR1 may be involved by evaluating the inflammatory response to arterial injury in<br />

CX 3CR1-deficient animals.<br />

Methods and Results—Femoral arteries of CX 3CR1 / and wild-type (WT) mice were injured with an angioplasty guide<br />

wire. After 1, 5, 14, and 28 days, arteries were harvested and evaluated by histology, morphometry, and<br />

immunohistochemistry. Arterial injury upregulated the CX 3CR1 ligand CX 3CL1. In CX 3CR1 / compared with WT<br />

animals, the incidence of neointima formation was 58% lower (P0.0017), accompanied by no difference in the area<br />

of platelet accumulation at day 1 (P0.48) but a significant decrease in intimal monocyte infiltration at day 5<br />

(P0.006), vascular smooth muscle cell (VSMC) proliferation at days 5 and 14, and intimal area at day 28 (P0.009).<br />

Conclusions—In an endothelial denudation injury model, CX 3CR1 deficiency protects animals from developing intimal<br />

hyperplasia as a result of decreased monocyte trafficking to the lesion. CX 3CR1 deficiency decreases VSMC<br />

proliferation and intimal accumulation either directly or indirectly as a result of defective monocyte infiltration.<br />

(Arterioscler Thromb Vasc Biol. 2006;26:2056-2062.)<br />

Key Words: <strong>CX3CR1</strong> monocytes smooth muscle cells intimal hyperplasia vascular biology<br />

Arterial injury elicits vessel wall inflammation by triggering<br />

platelet adhesion, leukocyte recruitment, and vascular<br />

smooth muscle cell (VSMC) proliferation and migration.<br />

1–3 These stereotypic cellular responses serve as the basis<br />

for many vascular diseases, including atherosclerosis, restenosis,<br />

and vasculitis. The molecular mechanisms by which<br />

these responses are regulated are of tremendous interest.<br />

Both animal models and human genetic association studies<br />

have implicated an important role for the chemokine receptor<br />

<strong>CX3CR1</strong> and its ligand fractalkine (CX3CL1) in vascular<br />

disease. Mice that lack both <strong>CX3CR1</strong> and apolipoprotein E<br />

(apoE) exhibit a reduction in atherosclerotic lesion formation<br />

in the aorta and aortic root compared with apoE-deficient<br />

mice. 4,5 CX3CL1-deficient mice have a reduction in atherosclerotic<br />

plaque burden in the innominate artery. 6 In humans,<br />

the V249I/T280M variant of <strong>CX3CR1</strong> is associated with<br />

protection from coronary artery disease7–9 and internal carotid<br />

artery occlusive disease. 10<br />

While <strong>CX3CR1</strong> and CX3CL1 are emerging as important<br />

mediators of vascular disease, the specific mechanisms regulating<br />

their involvement remain unclear. CX3CL1 is a<br />

transmembrane chemokine on activated endothelium that also<br />

exists in a soluble form. 11–13 Soluble CX3CL1 is a potent<br />

chemoattractant, and membrane-tethered CX3CL1 promotes<br />

cell adhesion by supporting the capture and firm adhesion of<br />

circulating CX 3CR1-expressing cells. 14,15 CX 3CR1 is expressed<br />

on monocytes, 16 VSMCs, 17,18 and platelets. 19 Thus,<br />

each of these cell types is a candidate to mediate the effects<br />

of CX 3CR1 on vascular inflammatory responses.<br />

The prevailing hypothesis is that the mechanism for<br />

CX 3CR1 in the development of vascular diseases such as<br />

atherosclerosis is the adherence of CX 3CR1-expressing<br />

monocytes to inflamed endothelium. 5,14 However, recent<br />

evidence that smooth muscle cells (SMCs) found in human<br />

atherosclerotic plaques also express CX 3CR1 suggests an<br />

alternative mechanism. 18 Stimulation of aortic smooth muscle<br />

cells with soluble CX 3CL1 leads to an increase in cell<br />

survival and proliferation. 20 Monocyte adhesion and arrest on<br />

neointimal SMC is dependent on CX 3CL1, 21 and VSMCs<br />

undergo chemotaxis toward CX 3CL1. 17 Thus, CX 3CL1 may<br />

act not only on monocytes, but also may function to stimulate<br />

SMC proliferation and recruitment into the neointima.<br />

In the present study, we employed an endoluminal arterial<br />

injury model to investigate the role of CX 3CR1 in the<br />

pathogenesis of vascular injury. This model elicits a stereotypic<br />

response that allows for assessment of the functional<br />

roles of platelets, monocytes, and VSMCs. Our data show<br />

enhanced expression of CX 3CL1 after injury and a role for<br />

Original received January 17, 2006; final version accepted June 19, 2006.<br />

<strong>From</strong> the Department of Medicine (P.L., M.R., A.M.F., S.S.S., D.D.P.), Thurston Arthritis Research Center (P.L., S.P., A.M.F., D.D.P.), and Carolina<br />

Cardiovascular Biology Center (M.R., S.S.S.), University of North Carolina at Chapel Hill, Chapel Hill, NC.<br />

Correspondence to Dhavalkumar D. Patel, Novartis Institutes for BioMedical Research, WSJ 386.11.25, CH-4002 Basel, Switzerland. E-mail<br />

dhavalkumar.patel@novartis.com<br />

© 2006 American Heart Association, Inc.<br />

Arterioscler Thromb Vasc Biol. is available at http://www.atvbaha.org DOI: 10.1161/01.ATV.0000234947.47788.8c<br />

Downloaded from<br />

http://atvb.ahajournals.org/ 2056 by guest on March 25, 2013


CX 3CR1 in the development of intimal hyperplasia. A deficiency<br />

in CX 3CR1 does not affect platelet function, but does<br />

significantly influence monocyte recruitment. CX 3CR1 deficiency<br />

also plays a role in regulating SMC activities.<br />

Materials and Methods<br />

Animal Care<br />

CX 3CR1 / (KO) mice (a gift from Dr Philip Murphy [NIH]) were<br />

backcrossed onto the C57BL/6J background for 12 generations, and<br />

age-matched CX 3CR1 / (WT) mice generated from littermates were<br />

used as controls. All mice were bred and maintained in the barrier<br />

facility at the University of North Carolina and fed Prolab RMH-<br />

3000 (PMI Nutrition International, Richmond, Ind), a normal rodent<br />

chow diet. All experimental protocols were in compliance with<br />

Institutional Animal Care and Use Committee (IACUC) guidelines<br />

and were approved by the IACUC at the University of North<br />

Carolina at Chapel Hill.<br />

Femoral Artery Injury<br />

Femoral arteries of 8- to 12-week-old male CX 3CR1 / and WT mice<br />

were injured by endoluminal passage of an angioplasty guide wire as<br />

previously described. 22 Arteries on one side were not injured and<br />

served as negative controls. Briefly, mice were anesthetized with<br />

inhaled isoflurane and femoral arteries were exposed by a longitudinal<br />

groin incision and viewed under a surgical microscope. The<br />

distal portion of the artery was encircled with an 8-0 nylon suture, a<br />

vascular clamp was placed proximally at the level of the inguinal<br />

ligament, and a 0.01-inch diameter guide wire (CrossIT-200XT;<br />

Guidant Corporation, Indianapolis, Ind) was introduced into the<br />

arterial lumen through an arteriotomy made in the distal perforating<br />

branch. After release of the clamp, the guidewire was advanced to<br />

the level of the aortic bifurcation and immediately pulled back 3<br />

times to denude the endothelium. After removal of the wire, the<br />

arteriotomy site was ligated and skin was closed. Animals were<br />

routinely monitored after surgery.<br />

Tissue Preparation, Histology, and Morphometry<br />

Mice were euthanized at days 1 (n20), 5 (n16), 14 (n8), and 28<br />

(n26) after arterial injury. Animals were perfused with phosphatebuffered<br />

saline for 5 minutes, followed by 4% paraformaldehyde for<br />

20 more minutes at 100 cm H 2O via cannulation of the left ventricle.<br />

The hind limbs were then harvested en bloc, fixed in 4% paraformaldehyde<br />

overnight, and decalcified in formic acid bone decalcifier<br />

(Immunocal; Decal Corporation, Tallman, NY) for 24 hours. Tissues<br />

containing the femoral artery were embedded in paraffin and cut into<br />

5-m sections for further analysis.<br />

Six to 10 sections per femoral artery at 100-m intervals were<br />

screened with H&E staining, and sections from the area with<br />

maximal injury response were further evaluated by staining with the<br />

Combined Masson’s elastin (CME) stain to visualize the arterial wall<br />

layers or processed for immunohistochemistry. The intima and<br />

media areas were measured by computerized morphometry (Image J,<br />

National Institutes of Health). <strong>Intimal</strong> hyperplasia was defined as the<br />

formation of a neointimal layer within the internal elastic lamina<br />

(IEL). Media area was calculated as the area encircled by the<br />

external elastic lamina (EEL) minus the area encircled by IEL. The<br />

intima-to-media (I/M) ratio was calculated as the intimal area<br />

divided by the media area. Arteries with a broken IEL or thrombosis<br />

by CME stain were excluded from the study.<br />

Immunohistochemistry<br />

For characterizing the molecular and cellular composition of arteries,<br />

immunohistochemical analysis was used to identify monocytes by<br />

mAb F4/80 (Serotec Inc, Raleigh, NC), VSMCs by alkaline phosphatase<br />

conjugated anti--actin (Sigma, St. Louis, Mo), platelets by<br />

anti-thrombocyte (Inter-Cell Technologies, Princeton, NJ), endothelial<br />

cells by anti-von Willebrand factor (vWF) (DakoCytomation Inc,<br />

Carpinteria, Calif), and CX 3CL1 by anti-fractalkine (R&D Systems,<br />

Liu et al CX 3CR1 <strong>Deficiency</strong> in Arterial Injury 2057<br />

Minneapolis, Minn). Recombinant mouse fractalkine/CX 3CL1<br />

(R&D Systems) was used to validate the specificity of CX 3CL1<br />

staining. Antigen retrieval was performed for F4/80 staining with<br />

trypsin digestion and for CX 3CL1 and vWF staining by steaming in<br />

a citrate buffer (0.01 mol/L, pH 6.0) for 40 minutes. Endogenous<br />

peroxide activity was quenched using 3% H 2O 2 in methanol for 10<br />

minutes at room temperature, and sections were blocked using 4%<br />

serum (goat, rabbit, or rat) for 10 to 60 minutes at room temperature.<br />

Primary antibodies and their respective controls were incubated<br />

overnight at 4°C. After washing, sections were incubated with a<br />

species-specific biotinylated secondary antibody (anti-mouse, antirabbit,<br />

anti-rat, and anti-goat, 1:200; Vector Laboratories, Burlingame,<br />

Calif) for 60 minutes at room temperature, followed by<br />

washes, and a subsequent 60-minute incubation at room temperature<br />

with streptavidin-horseradish peroxidase (Peroxidase Vectastain<br />

ABC Kit) to amplify antibody signal. After another wash, 3,<br />

3-diaminobenzidine (DAB) (Sigma, St. Louis, Mo) was used as a<br />

substrate for the peroxidase. Alkaline phosphatase staining by<br />

-actin antibody was visualized with the Vector Red Alkaline<br />

Phosphatase Substrate Kit (Vector Laboratories).<br />

VSMC Proliferation Analysis<br />

Four days after arterial injury, mice were injected intraperitoneally<br />

with three doses of bromodeoxyuridine (BrdU) (Roche Diagnostics,<br />

Basel, Switzerland) of 30 mg/kg at 8-hour intervals before euthanasia.<br />

Arteries were harvested on day 5 after injury, and proliferating<br />

cells were identified by immunostaining with an anti-BrdU antibody<br />

(Roche Diagnostics). For all times other than day 5 after injury, an<br />

anti-PCNA antibody (Santa Cruz Laboratories, Santa Cruz, Calif)<br />

was used to identify proliferating cells. Proliferating VSMCs were<br />

defined as -actin–positive cells that were also positive for BrdU or<br />

PCNA staining in series sections. The proliferation index was<br />

calculated as the percentage of BrdU-stained or PCNA-stained nuclei<br />

of the total number of nuclei in the indicated area. VSMC proliferation<br />

was also determined in vitro utilizing primary cultures of<br />

VSMC isolated from aortas of CX 3CR1 / and WT mice with a<br />

colorimetric assay based on the uptake of MTT by viable cells (Cell<br />

Proliferation Kit; Roche Applied Science, Indianapolis, Ind).<br />

Platelet Function<br />

Platelet function was tested in vitro with the Cone and Plate(let)<br />

Analyzer (CPA) system using a DiaMed Impact-R machine (DiaMed<br />

Israel Ltd) according to the manufacture’s instructions. Briefly, fresh<br />

samples of sodium citrated anticoagulated blood (130 L) from<br />

CX 3CR1 / and WT mice were placed in polystyrene wells and<br />

subjected to defined shear (1800/second) for 2 minutes. The samples<br />

were then thoroughly washed with phosphate-buffered saline,<br />

stained with May-Grünwald stain, and quantitated with an image<br />

analyzer. Platelet deposition on the polystyrene surface was evaluated<br />

by examining: (1) the percentage of total area covered with<br />

platelets designated as surface coverage; and (2) average size in m 2<br />

of surface-bound platelet thrombi.<br />

Monocyte Adhesion<br />

Monocyte adhesion was evaluated as previously described. 14 Briefly,<br />

spleens from CX 3CR1 / and WT mice were homogenized in<br />

RPMI-1640 with 10 mmol/L HEPES using a manual tissue homogenizer.<br />

Red blood cells were lysed with red blood cell lysis buffer<br />

(0.14 mol/L NH 4CI, 0.017 mol/L Tris-HCl pH7.5, adjust to pH7.2).<br />

Cells were washed with RPMI-1640 with 10% fetal bovine serum<br />

(FBS), passed through a 70-m nylon filter, and suspended in<br />

RPMI-1640 with 10% FBS. Adhesion of the splenocytes to fractalkine<br />

under physiological flow conditions was then tested by the<br />

parallel plate flow chamber assay with recombinant fractalkinesecreted<br />

alkaline phosphatase fusion proteins immobilized on a glass<br />

coverslip. After the run, adherent cells were stained with MOMAfluorescein<br />

isothiocyanate (FITC) antibody (Beckman Coulter, Inc,<br />

Fullerton, Calif) and the number of monocytes bound was quantified<br />

by counting MOMA cells.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ by guest on March 25, 2013


2058 Arterioscler Thromb Vasc Biol. September 2006<br />

Statistical Analysis<br />

Numerical data presented in text and figure are expressed as<br />

meanSEM. All sections were analyzed by 2 investigators, 1<br />

blinded and 1 unblinded, with an inter-rater reliability of 0.95 to<br />

0.99. Fisher’s exact test was utilized to determine incidence. Student<br />

unpaired t test was used to compare average numbers of cells or<br />

percentages between experimental groups. In all cases, P0.05 was<br />

considered significant.<br />

Results<br />

Characterization of <strong>Intimal</strong> <strong>Hyperplasia</strong><br />

After Injury<br />

Wire injury resulted in endothelial denudation as evidenced<br />

by an absence of endothelial cells in femoral arteries from<br />

WT mice 24 hours after injury. The contralateral, uninjured<br />

control vessels retained their normal histology. Platelet adhesion<br />

to the injured vessel wall at day 1 was followed by<br />

monocyte recruitment to the intima by 5 days after injury<br />

(Figure 1). At 14 days, the neointima was a mixture of<br />

monocytes and VSMCs, and by 28 days, the neointima was<br />

primarily composed of VSMC (Figure 1). Taken together,<br />

intimal hyperplasia was readily apparent by 5 days and it was<br />

continuously detected between 5 and 28 days after injury.<br />

Monocyte recruitment to the intima occurred earlier than<br />

VSMC accumulation, which was the primary cellular component<br />

of neointima in the late stage (day 28). Eighty-six<br />

percent of WT mice developed intimal hyperplasia with an<br />

average Intima/Media (I/M) ratio of 0.8.<br />

CX 3CL1 Expression Is Induced in Injured Arteries<br />

To define the role of CX 3CR1 in the response to arterial<br />

injury, we first examined the expression pattern of its ligand,<br />

CX 3CL1, by immunohistochemistry. CX 3CL1 expression was<br />

undetectable in non-injured, control arteries at any stage of<br />

the injury response. In injured arteries, CX 3CL1 was not<br />

detected at day 1. At day 5, CX 3CL1 was expressed in<br />

endothelial cells and in a subset of the intimal SMC in WT<br />

and CX 3CR1 / arteries (Figure 2). This pattern of expression<br />

continued through the injury response. These data provide the<br />

evidence that CX 3CL1 may be involved in the pathogenesis<br />

of guide wire induced vascular injury.<br />

CX 3CR1-Deficient Mice Are Protected <strong>From</strong><br />

<strong>Intimal</strong> <strong>Hyperplasia</strong><br />

To assess whether CX 3CR1 plays a role in mediating intimal<br />

hyperplasia after arterial injury, we measured the incidence<br />

and extent of neointima formation in CX 3CR1 / and WT<br />

mice. The overall incidence of intimal hyperplasia at 5, 14,<br />

and 28 days was decreased in CX 3CR1 / mice by 58% (8/21<br />

Figure 1. Cellular composition of injured<br />

arteries. Shown are immunohistochemical<br />

analyses of WT arteries at 0, 1, 5, 14,<br />

and 28 days after arterial injury. Platelets<br />

(brown), monocytes (brown), and VSMCs<br />

(red) were identified by anti-thrombocyte,<br />

anti-F4/80, and anti--actin antibodies,<br />

respectively.<br />

KO versus 26/29 WT, P0.0017). Compared with the<br />

average intimal area in WT mice, the average intimal area in<br />

CX 3CR1 / mice was reduced by 84%, 56%, and 74% at 5,<br />

14, and 28 days, respectively (Figure 3). In contrast, no<br />

significant differences in luminal and medial areas were<br />

detected between WT and CX 3CR1 / arteries (data not<br />

shown). These data indicate that CX 3CR1 deficient mice are<br />

protected from intimal hyperplasia after acute arterial injury,<br />

and that the arterial injury model is appropriate to study the<br />

functional roles of CX 3CR1.<br />

Normal Platelet Function in CX 3CR1 / Mice<br />

To define whether platelet CX 3CR1 is functionally relevant in<br />

guide wire induced injury, we examined platelet accumulation<br />

on the injured luminal surface by immunostaining. By<br />

morphometric analysis, there was no difference in the area of<br />

the platelet thrombus that accumulated along injured<br />

CX 3CR1 / and WT arteries (Figure 4A and 4B). We also<br />

examined platelet function under near-physiological conditions<br />

in vitro using blood collected from CX 3CR1 / and WT<br />

mice. As shown in Figure 4C and 4D, the absence of CX 3CR1<br />

did not affect shear-induced platelet thrombus formation.<br />

These data suggest that platelet CX 3CR1 may be not an<br />

essential mediator of platelet adhesion or aggregation in<br />

response to arterial injury or high shear.<br />

Defective Monocyte Recruitment in<br />

CX 3CR1 / Mice<br />

CX 3CR1 is believed to be critically important for monocyte<br />

recruitment to the inflamed or injured vessel. We tested this<br />

hypothesis by examining monocyte infiltration into the vascular<br />

wall by F4/80 antigen staining (Figure 5A). There was<br />

a 100% and an 87% decrease in monocyte accumulation in<br />

CX 3CR1 / mice at 5 and 14 days, respectively, compared<br />

with WT mice (Figure 5B). Monocyte adhesion to CX3CL1<br />

Figure 2. CX 3CL1 staining in injured vessels. Immunostaining of<br />

CX 3CL1 and endothelial cells (vWF) in WT mice is shown in control<br />

and injured arteries at 5 days after injury. Control arteries<br />

have no CX3CL1 expression and injured arteries show expression<br />

of CX3CL1 by vWF endothelial cells.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ by guest on March 25, 2013


under physiological flow conditions was also substantially<br />

lower with CX 3CR1 / compared with WT cells (Figure 5C).<br />

Taken together, these results provide strong evidence that<br />

CX 3CR1 plays a critical role in monocyte recruitment to the<br />

inflamed vessel wall.<br />

Role of CX 3CR1 in VSMC Response to<br />

Vascular Injury<br />

To assess whether CX 3CR1 is important for the function of<br />

VSMC in response to arterial injury, we took 3 approaches.<br />

First, we evaluated the numbers of VSMC in the vessel wall<br />

by immunohistochemistry for -actin. Second, we evaluated<br />

VSMC proliferation by BrdU incorporation and by immunohistochemistry<br />

for PCNA expression in -actin positive cells.<br />

Third, we evaluated the in vitro proliferation of primary<br />

VSMC isolated from CX 3CR1 / and WT mouse aortas.<br />

In injured arteries, substantial VSMC proliferation was<br />

observed at 5 days and 14 days (Figure 6). At 5 days, most<br />

proliferating cells were localized in the media. The proliferation<br />

index in CX 3CR1 / mice was decreased by 45%<br />

compared with WT mice (8.52.7% versus 15.33.4%;<br />

P0.07). As the lesion progressed at 14 days, an 85%<br />

decrease in proliferating cells was detected in the intima of<br />

Liu et al CX 3CR1 <strong>Deficiency</strong> in Arterial Injury 2059<br />

Figure 3. Injury-induced intimal hyperplasia<br />

in wild type (WT) and CX 3CR1 /<br />

(KO) mice. Unilateral wire injury was performed<br />

in femoral arteries of WT and<br />

CX 3CR1 / mice, with the contralateral<br />

uninjured side used as a negative control.<br />

A, CME stains of representative sections<br />

of control and injured femoral arteries<br />

at 28 days after injury. <strong>Intimal</strong><br />

hyperplasia is defined as the formation<br />

of a neointimal layer within the internal<br />

elastic lamina (arrows). B, Average intimal<br />

areas of injured WT and KO arteries<br />

at 5, 14, and 28 days after injury. Results<br />

are reported as meanSEM. **P0.05.<br />

CX 3CR1 / mice compared with WT animals (3.12.0%<br />

versus 206.0%; P0.03). In vitro, while WT aortic VSMC<br />

proliferated in response to CX3CL1 (P0.005), CX 3CR1 /<br />

aortic VSMC did not (PNS). These results suggest that<br />

CX 3CR1 deficiency diminishes VSMC proliferation in the<br />

early development of intimal hyperplasia in response to<br />

arterial injury.<br />

Discussion<br />

Inflammation is a driving force behind vascular diseases such<br />

as atherosclerosis and restenosis. Arterial injury is the initial<br />

stage of the pathohistological changes of these diseases. We<br />

found that guidewire-induced endothelial denudation of<br />

mouse femoral arteries elicits an inflammatory response with<br />

upregulation of CX 3CL1 expression. Therefore, we investigated<br />

the mechanisms of action of CX 3CR1 in vascular<br />

inflammation by testing the effects of CX 3CR1 deficiency in<br />

this injury model, which stimulates acute inflammatory responses<br />

similar to restenosis. Because CX 3CR1 is expressed<br />

on platelets, monocytes and VSMC, we focused on these cell<br />

types.<br />

CX 3CR1 does not appear to play a critical role in platelet<br />

accumulation along the denuded endothelial surface. In our<br />

Downloaded from<br />

http://atvb.ahajournals.org/ by guest on March 25, 2013<br />

Figure 4. CX 3CR1 effect on platelet<br />

adhesion. One day after injury, control<br />

and injured femoral arteries were immunostained<br />

for platelets (A, B). A, Platelets<br />

(brown) adhering to the luminal surface<br />

in vessels of injured WT and injured<br />

CX 3CR1 / (KO), but not control WT<br />

mice. B, Area of adherent platelets measured<br />

by morphometric analysis in<br />

injured CX 3CR1 / (n7) and WT arteries<br />

(n10). Results are expressed as<br />

meanSEM. Shear-induced platelet<br />

adhesion represented by surface coverage<br />

(C) and aggregation represented by<br />

average size (D) were measured in blood<br />

from WT (n4) and CX 3CR1 / (n4)<br />

mice using the CPA technology. Data are<br />

expressed as meanSEM.


2060 Arterioscler Thromb Vasc Biol. September 2006<br />

model, platelets and neutrophils cover the denuded luminal<br />

surface within 24 hours. 22–24 The area of platelet accumulation<br />

was unaffected by the absence of CX 3CR1. Likewise, no<br />

significant differences were observed in shear-induced platelet<br />

thrombus formation in blood from WT and CX 3CR1deficient<br />

mice. These results suggest that platelet CX 3CR1<br />

does not play a major role in the initial phases of platelet<br />

adhesion and aggregation stimulated by the vascular injury.<br />

Likewise, CX 3CL1 was not highly expressed within the first<br />

24 hours after injury. Whether CX 3CR1 contributes to other<br />

platelet responses cannot be determined from our study.<br />

Regenerated endothelial cells expressed high levels of<br />

CX 3CL1. Similar to previous reports, 22 we observed regeneration<br />

of endothelial cells within 5 days of arterial injury.<br />

The endothelium before injury did not express CX 3CL1, but<br />

the regenerated endothelium expressed high levels of<br />

CX 3CL1. In contrast to WT animals that had robust monocyte<br />

accumulation, CX 3CR1 / mice failed to recruit monocytes to<br />

the intima despite expression of CX 3CL1. Interestingly,<br />

monocytes accumulated in the adventitia of WT and<br />

CX 3CR1 / animals after injury, suggesting that monocyte<br />

recruitment to the adventitia may occur through mechanisms<br />

distinct from those required for intimal recruitment. A primary<br />

difference between these two sites is the presence of<br />

arterial shear forces, and monocyte capture and transmigration<br />

along the artery may require CX 3CR1-driven adhesion<br />

and/or migration to resist the shear. In contrast, recruitment of<br />

Figure 5. CX 3CR1 effect on monocyte<br />

adhesion. Monocyte recruitment to<br />

injured vessels in vivo was tested by<br />

immunohistochemical staining of F4/80<br />

antigen (A, B) and adhesion to CX3CL1<br />

in vitro was tested by the parallel plate<br />

flow chamber adhesion assay using<br />

splenocytes from WT and CX 3CR1 /<br />

mice (C). A, Monocyte staining (brown) in<br />

injured WT and CX 3CR1 / arteries at 5<br />

days after surgery. B, Average number of<br />

monocytes in the intima of WT and<br />

CX 3CR1 / (KO) arteries 5 days after<br />

injury (n16). Results are expressed as<br />

the meanSEM. **P0.05. C, Average<br />

number of Moma2-FITC monocytes<br />

bound to CX 3CL1. Results are expressed<br />

as the meanSEM. **P0.05.<br />

tissue macrophages or transvenous migration of monocyte to<br />

the adventitia may occur by CX 3CR1-independent mechanisms.<br />

While we cannot exclude the possibility that adventitial<br />

macrophages traffic and migrate toward soluble CX 3CL1<br />

through the vessel wall to the luminal surface, the data<br />

support a primary role for CX 3CR1 in the rapid capture and<br />

firm adhesion of monocytes under flow conditions. 14 Clinical<br />

cohorts have shown that 2 single nucleotide polymorphisms<br />

of CX 3CR1, V249I and T280M, are associated with reduced<br />

prevalence of atherosclerosis and coronary artery disease. 8,9<br />

In addition, the M280 allele is associated with a reduced risk<br />

of internal carotid artery (ICA) occlusive disease. 10 Our<br />

laboratory has shown that the protein encoded by the M280<br />

allele has impaired adhesive capacity, 7 suggesting that cell<br />

adhesion is an important mechanism by which CX 3CR1<br />

exerts its effect on recruiting cells during vascular<br />

inflammation.<br />

In addition to CX 3CR1, CCR2 appears to mediate the<br />

response to arterial injury as well. In the same mouse model<br />

of wire-induced femoral artery injury, CCR2 / mice had a<br />

phenotype comparable to CX 3CR1 / mice in terms of a<br />

reduced intimal hyperplasia and intima/media ratio 4 weeks<br />

after injury. 23 In the CCR2 study, no macrophage infiltration<br />

was seen by MOMA-2 or CD68 staining in either WT or<br />

CCR2 / animals and the authors concluded that CCR2 did<br />

not affect macrophage accumulation within the arterial wall<br />

after injury. Although both CX 3CR1 and CCR2 mediate<br />

Downloaded from<br />

http://atvb.ahajournals.org/ by guest on March 25, 2013<br />

Figure 6. VSMC proliferation in response to vascular<br />

injury. Shown is the proliferation of VSMCs in<br />

the intima and media of injured WT and <strong>CX3CR1</strong> /<br />

arteries at 5 and 14 days after injury. Proliferating<br />

VSMCs were defined as those -actin positive<br />

cells that were BrdU (d5) or PCNA (d14) positive.<br />

Results are represented as meanSEM. *P0.07,<br />

**P0.03.


monocyte chemotaxis and trafficking to sites of inflammation,<br />

CX 3CR1 mediates direct monocyte binding to its<br />

membrane-bound ligand, CX 3CL1, without the help of other<br />

adhesion molecules, such as integrins. 15 Thus, monocyte<br />

trafficking to damaged tissue during arterial injury may favor<br />

the CX 3CR1–CX 3CL1 system. Recently, Schober et al reported<br />

an alternative mechanism for CCR2 in acute vascular<br />

injury. 25 In their study, CCL2 was found to be co-localized<br />

with platelets on the denuded carotid artery surface of<br />

apoE / mice fed with a high-fat diet within 24 hours after<br />

wire injury. Even though platelets do not contain CCL2, 26<br />

low-affinity CCR2-dependent binding of CCL2 to human<br />

platelets is detected in vitro, 27 suggesting that adherent<br />

platelets with immobilized CCL2 mediate monocyte recruitment<br />

after mechanical injury. Monocyte adhesion to the<br />

luminal surface is not detected within 24 hours under our<br />

experimental conditions or in similar wire-induced arterial<br />

injury models under either normolipidemic 23 or hyperlipidemic<br />

25,28 conditions. Nevertheless, platelet deposition may<br />

play a vital role in the pathogenesis of restenosis, 29 and CCR2<br />

may be involved in that process.<br />

SMC accumulate in the intima in both atherosclerosis and<br />

restenosis. 30,31 This process is also seen in animals following<br />

guidewire-induced arterial injury. CX 3CR1 is expressed in<br />

human SMC cultured from coronary arteries and in atherosclerotic<br />

lesions, 17,18 suggesting that CX 3CR1 may play a role<br />

in regulating SMC function. In our study, intimal hyperplasia<br />

was significantly decreased in CX 3CR1 / arteries after<br />

injury. Concurrently, there were decreased numbers of<br />

VSMC in the neointima. VSMC proliferation in response to<br />

injury was also impaired in CX 3CR1 / mice as measured by<br />

both BrdU incorporation and PCNA immunohistochemistry.<br />

Whether the VSMC effects are primary or secondary cannot<br />

be addressed by this study. However, several data point to a<br />

primary effect of CX 3CR1 on VSMC function in this model.<br />

In a recent report of interleukin (IL)-15’s effect on CX 3CR1/<br />

CX 3CL1 expression and the response to arterial injury, 32<br />

IL-15 attenuated SMC proliferation and CX 3CR1/CX 3CL1<br />

mRNA expression on serum stimulation. Using a periadventitial<br />

injury model, the authors further demonstrated that<br />

IL-15 upregulation after injury reduced intimal thickening,<br />

and blockade of IL-15 increased CX 3CR1/CX 3CL1 expression.<br />

These results suggested that IL-15 up-regulation decreases<br />

neointimal formation in response to arterial injury via<br />

suppressing CX 3CR1 signaling in SMC. CX 3CR1 is a typical<br />

G protein-coupled receptor, and the binding of CX 3CL1 to<br />

CX 3CR1 initiates multiple signal transduction pathways that<br />

are associated with cell proliferation and survival, such as<br />

extracellular signal regulated kinase (ERK)/P38 MAPK pathways<br />

33 and the PI3K/Akt pathway. 34 In addition, tumor<br />

necrosis factor- stimulates CX 3CL1 expression in human<br />

aortic SMC, 35 and the induction of CX 3CR1 facilitates SMC<br />

proliferation via NF-B in aortic SMCs. 20 Additionally,<br />

CX 3CR1/CX 3CL1 may directly regulate SMC migration in<br />

response to vascular inflammation. Cultured human coronary<br />

artery SMC have been shown to express CX 3CR1 and<br />

undergo chemotaxis toward CX 3CL1. 17 US28, a viral receptor<br />

for CX 3CL1 can mediate SMC migration in vitro. 36 Although<br />

direct evidence for a role of CX 3CR1-mediated SMC migra-<br />

Liu et al CX 3CR1 <strong>Deficiency</strong> in Arterial Injury 2061<br />

tion in arterial injury needs to be further demonstrated in<br />

vivo, our findings of substantially reduced intimal hyperplasia<br />

and SMC proliferation in CX 3CR1 / mice after mouse<br />

femoral artery denudation strongly suggest that, in addition to<br />

mediating monocyte infiltration at early stages of the injury<br />

response, CX 3CR1 may also play an important role in<br />

vascular remodeling in the late stage of injury by regulating<br />

SMC functions.<br />

In summary, we demonstrate that CX 3CR1 plays a critical<br />

role in the regulation of cellular responses to acute vascular<br />

injury. CX 3CL1 expression is upregulated on endothelial and<br />

smooth muscle cells after injury. Mice lacking CX 3CR1 have<br />

lower incidence and degree of intimal hyperplasia after<br />

injury. The role of CX 3CR1 in regulating vascular inflammation<br />

involves monocyte accumulation in the vessel wall and<br />

VSMC proliferation and migration. Combined with data that<br />

CX 3CR1 polymorphisms associated with defective cell adhesion<br />

protect humans from atherosclerotic diseases, our findings<br />

suggest that CX 3CR1 could be an effective drug target<br />

for both acute and chronic vascular injuries, such as restenosis<br />

and atherosclerosis.<br />

Acknowledgments<br />

The authors sincerely thank Gail Grossman and Kirk McNaughton<br />

for their technical assistance in tissue processing and staining, Rishi<br />

Rampersad for animal husbandry and Drs Teresa Tarrant and Maya<br />

Jerath for proofreading the manuscript.<br />

Sources of Funding<br />

This study was supported by National Institute of Health R01s<br />

CA098110 (D.D.P.) and HL074219 (S.S.S.).<br />

None.<br />

Disclosures<br />

References<br />

1. Ross R. Atherosclerosis–an inflammatory disease. N Engl J Med. 1999;<br />

340:115–126.<br />

2. Schwartz SM, deBlois D, O’Brien ER. The intima. Soil for atherosclerosis<br />

and restenosis. Circ Res. 1995;77:445–465.<br />

3. Charo IF, Taubman MB. Chemokines in the pathogenesis of vascular<br />

disease. Circ Res. 2004;95:858–866.<br />

4. Combadiere C, Potteaux S, Gao JL, Esposito B, Casanova S, Lee EJ,<br />

Debre P, Tedgui A, Murphy PM, Mallat Z. Decreased atherosclerotic<br />

lesion formation in <strong>CX3CR1</strong>/apolipoprotein E double knockout mice.<br />

Circulation. 2003;107:1009–1016.<br />

5. Lesnik P, Haskell CA, Charo IF. Decreased atherosclerosis in <strong>CX3CR1</strong>-/mice<br />

reveals a role for fractalkine in atherogenesis. J Clin Invest. 2003;<br />

111:333–340.<br />

6. Teupser D, Pavlides S, Tan M, Gutierrez-Ramos JC, Kolbeck R, Breslow<br />

JL. Major reduction of atherosclerosis in fractalkine (CX3CL1)-deficient<br />

mice is at the brachiocephalic artery, not the aortic root. Proc Natl Acad<br />

Sci U S A. 2004;101:17795–17800.<br />

7. McDermott DH, Fong AM, Yang Q, Sechler JM, Cupples LA, Merrell<br />

MN, Wilson PW, D’Agostino RB, O’Donnell CJ, Patel DD, Murphy PM.<br />

Chemokine receptor mutant <strong>CX3CR1</strong>–M280 has impaired adhesive<br />

function and correlates with protection from cardiovascular disease in<br />

humans. J Clin Invest. 2003;111:1241–1250.<br />

8. McDermott DH, Halcox JP, Schenke WH, Waclawiw MA, Merrell MN,<br />

Epstein N, Quyyumi AA, Murphy PM. Association between polymorphism<br />

in the chemokine receptor <strong>CX3CR1</strong> and coronary vascular<br />

endothelial dysfunction and atherosclerosis. Circ Res. 2001;89:401–407.<br />

9. Moatti D, Faure S, Fumeron F, Amara Mel W, Seknadji P, McDermott<br />

DH, Debre P, Aumont MC, Murphy PM, de Prost D, Combadiere C.<br />

Polymorphism in the fractalkine receptor <strong>CX3CR1</strong> as a genetic risk factor<br />

for coronary artery disease. Blood. 2001;97:1925–1928.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ by guest on March 25, 2013


2062 Arterioscler Thromb Vasc Biol. September 2006<br />

10. Ghilardi G, Biondi ML, Turri O, Guagnellini E, Scorza R. Internal carotid<br />

artery occlusive disease and polymorphisms of fractalkine receptor<br />

<strong>CX3CR1</strong>: a genetic risk factor. Stroke. 2004;35:1276–1279.<br />

11. Harrison JK, Jiang Y, Wees EA, Salafranca MN, Liang HX, Feng L,<br />

Belardinelli L. Inflammatory agents regulate in vivo expression of fractalkine<br />

in endothelial cells of the rat heart. J Leukoc Biol. 1999;66:<br />

937–944.<br />

12. Garton KJ, Gough PJ, Blobel CP, Murphy G, Greaves DR, Dempsey PJ,<br />

Raines EW. Tumor necrosis factor-alpha-converting enzyme (ADAM17)<br />

mediates the cleavage and shedding of fractalkine (CX3CL1). J Biol<br />

Chem. 2001;276:37993–38001.<br />

13. Tsou CL, Haskell CA, Charo IF. Tumor necrosis factor-alpha-converting<br />

enzyme mediates the inducible cleavage of fractalkine. J Biol Chem.<br />

2001;276:44622–44626.<br />

14. Fong AM, Robinson LA, Steeber DA, Tedder TF, Yoshie O, Imai T, Patel<br />

DD. Fractalkine and <strong>CX3CR1</strong> mediate a novel mechanism of leukocyte<br />

capture, firm adhesion, and activation under physiologic flow. J Exp Med.<br />

1998;188:1413–1419.<br />

15. Haskell CA, Cleary MD, Charo IF. Molecular uncoupling of fractalkinemediated<br />

cell adhesion and signal transduction. Rapid flow arrest of<br />

<strong>CX3CR1</strong>-expressing cells is independent of G-protein activation. J Biol<br />

Chem. 1999;274:10053–10058.<br />

16. Imai T, Hieshima K, Haskell C, Baba M, Nagira M, Nishimura M,<br />

Kakizaki M, Takagi S, Nomiyama H, Schall TJ, Yoshie O. Identification<br />

and molecular characterization of fractalkine receptor <strong>CX3CR1</strong>, which<br />

mediates both leukocyte migration and adhesion. Cell. 1997;91:521–530.<br />

17. Lucas AD, Bursill C, Guzik TJ, Sadowski J, Channon KM, Greaves DR.<br />

Smooth muscle cells in human atherosclerotic plaques express the fractalkine<br />

receptor <strong>CX3CR1</strong> and undergo chemotaxis to the CX3C chemokine<br />

fractalkine (CX3CL1). Circulation. 2003;108:2498–2504.<br />

18. Wong BW, Wong D, McManus BM. Characterization of fractalkine<br />

(CX3CL1) and <strong>CX3CR1</strong> in human coronary arteries with native atherosclerosis,<br />

diabetes mellitus, and transplant vascular disease. Cardiovasc<br />

Pathol. 2002;11:332–338.<br />

19. Schafer A, Schulz C, Eigenthaler M, Fraccarollo D, Kobsar A, Gawaz M,<br />

Ertl G, Walter U, Bauersachs J. Novel role of the membrane-bound<br />

chemokine fractalkine in platelet activation and adhesion. Blood. 2004;<br />

103:407–412.<br />

20. Chandrasekar B, Mummidi S, Perla RP, Bysani S, Dulin NO, Liu F,<br />

Melby PC. Fractalkine (CX3CL1) stimulated by nuclear factor kappaB<br />

(NF-kappaB)-dependent inflammatory signals induces aortic smooth<br />

muscle cell proliferation through an autocrine pathway. Biochem J. 2003;<br />

373:547–558.<br />

21. Zeiffer U, Schober A, Lietz M, Liehn EA, Erl W, Emans N, Yan ZQ,<br />

Weber C. Neointimal smooth muscle cells display a proinflammatory<br />

phenotype resulting in increased leukocyte recruitment mediated by<br />

P-selectin and chemokines. Circ Res. 2004;94:776–784.<br />

22. Roque M, Fallon JT, Badimon JJ, Zhang WX, Taubman MB, Reis ED.<br />

Mouse model of femoral artery denudation injury associated with the<br />

rapid accumulation of adhesion molecules on the luminal surface and<br />

recruitment of neutrophils. Arterioscler Thromb Vasc Biol. 2000;20:<br />

335–342.<br />

23. Roque M, Kim WJ, Gazdoin M, Malik A, Reis ED, Fallon JT, Badimon<br />

JJ, Charo IF, Taubman MB. CCR2 deficiency decreases intimal hyperplasia<br />

after arterial injury. Arterioscler Thromb Vasc Biol. 2002;22:<br />

554–559.<br />

24. Smyth SS, Reis ED, Zhang W, Fallon JT, Gordon RE, Coller BS.<br />

Beta(3)-integrin-deficient mice but not P-selectin-deficient mice develop<br />

intimal hyperplasia after vascular injury: correlation with leukocyte<br />

recruitment to adherent platelets 1 hour after injury. Circulation. 2001;<br />

103:2501–2507.<br />

25. Schober A, Zernecke A, Liehn EA, von Hundelshausen P, Knarren S,<br />

Kuziel WA, Weber C. Crucial role of the CCL2/CCR2 axis in neointimal<br />

hyperplasia after arterial injury in hyperlipidemic mice involves early<br />

monocyte recruitment and CCL2 presentation on platelets. Circ Res.<br />

2004;95:1125–1133.<br />

26. Gear AR, Camerini D. Platelet chemokines and chemokine receptors:<br />

linking hemostasis, inflammation, and host defense. Microcirculation.<br />

2003;10:335–350.<br />

27. Clemetson KJ, Clemetson JM, Proudfoot AE, Power CA, Baggiolini M,<br />

Wells TN. Functional expression of CCR1, CCR3, CCR4, and CXCR4<br />

chemokine receptors on human platelets. Blood. 2000;96:4046–4054.<br />

28. Weingartner O, Kasper M, Reynen K, Bramke S, Marquetant R, Sedding<br />

DG, Braun-Dullaeus R, Strasser RH. Comparative morphometric and<br />

immunohistological assessment of the development of restenosis after<br />

arterial injury and a cholesterol-rich diet in apolipoprotein E -/-mice and<br />

C57BL/6 control mice. Coron Artery Dis. 2005;16:391–400.<br />

29. Farb A, Sangiorgi G, Carter AJ, Walley VM, Edwards WD, Schwartz RS,<br />

Virmani R. Pathology of acute and chronic coronary stenting in humans.<br />

Circulation. 1999;99:44–52.<br />

30. Reidy MA, Fingerle J, Lindner V Factors controlling the development of<br />

arterial lesions after injury. Circulation. 1992;86:III43–III46.<br />

31. Schwartz RS Pathophysiology of restenosis: interaction of thrombosis,<br />

hyperplasia, and/or remodeling. Am J Cardiol. 1998;81:14E–17E.<br />

32. Cercek M, Matsumoto M, Li H, Chyu KY, Peter A, Shah PK, Dimayuga<br />

PC. Autocrine role of vascular IL-15 in intimal thickening. Biochem<br />

Biophys Res Commun. 2006;339:618–623.<br />

33. Cambien B, Pomeranz M, Schmid-Antomarchi H, Millet MA,<br />

Breittmayer V, Rossi B, Schmid-Alliana A. Signal transduction pathways<br />

involved in soluble fractalkine-induced monocytic cell adhesion. Blood.<br />

2001;97:2031–2037.<br />

34. Shulby SA, Dolloff NG, Stearns ME, Meucci O, Fatatis A. <strong>CX3CR1</strong>fractalkine<br />

expression regulates cellular mechanisms involved in<br />

adhesion, migration, and survival of human prostate cancer cells. Cancer<br />

Res. 2004;64:4693–4698.<br />

35. Ollivier V, Faure S, Tarantino N, Chollet-Martin S, Deterre P, Combadiere<br />

C, de Prost D. Fractalkine/CX3CL1 production by human aortic<br />

smooth muscle cells impairs monocyte procoagulant and inflammatory<br />

responses. Cytokine. 2003;21:303–311.<br />

36. Streblow DN, Soderberg-Naucler C, Vieira J, Smith P, Wakabayashi E,<br />

Ruchti F, Mattison K, Altschuler Y, Nelson JA. The human cytomegalovirus<br />

chemokine receptor US28 mediates vascular smooth muscle cell<br />

migration. Cell. 1999;99:511–520.<br />

Downloaded from<br />

http://atvb.ahajournals.org/ by guest on March 25, 2013

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!