Literature
首页医源资料库在线期刊动脉硬化血栓血管生物学杂志2006年第26卷第9期

CX 3 CR1 Deficiency Confers Protection From Intimal Hyperplasia After Arterial Injury

来源:《动脉硬化血栓血管生物学杂志》
摘要:【摘要】Objective-AfunctionalpolymorphisminthechemokinereceptorCX3CR1isassociatedwithprotectionfromvasculardiseasesincludingcoronaryarterydiseaseandinternalcarotidarteryocclusivedisease。WeinvestigatedthemechanismsbywhichCX3CR1maybeinvolvedbyevaluati......

点击显示 收起

【摘要】  Objective- A functional polymorphism in the chemokine receptor CX 3 CR1 is associated with protection from vascular diseases including coronary artery disease and internal carotid artery occlusive disease. We investigated the mechanisms by which CX 3 CR1 may be involved by evaluating the inflammatory response to arterial injury in CX 3 CR1-deficient animals.

Methods and Results- Femoral arteries of CX 3 CR1 -/- and wild-type (WT) mice were injured with an angioplasty guide wire. After 1, 5, 14, and 28 days, arteries were harvested and evaluated by histology, morphometry, and immunohistochemistry. Arterial injury upregulated the CX 3 CR1 ligand CX 3 CL1. In CX 3 CR1 -/- compared with WT animals, the incidence of neointima formation was 58% lower ( P =0.0017), accompanied by no difference in the area of platelet accumulation at day 1 ( P =0.48) but a significant decrease in intimal monocyte infiltration at day 5 ( P =0.006), vascular smooth muscle cell (VSMC) proliferation at days 5 and 14, and intimal area at day 28 ( P =0.009).

Conclusions- In an endothelial denudation injury model, CX 3 CR1 deficiency protects animals from developing intimal hyperplasia as a result of decreased monocyte trafficking to the lesion. CX 3 CR1 deficiency decreases VSMC proliferation and intimal accumulation either directly or indirectly as a result of defective monocyte infiltration.

The chemokine receptor CX 3 CR1 has been implicated in the pathogenesis of vascular diseases. In a guidewire-induced femoral artery injury model, the CX 3 CR1 ligand (CX 3 CL1) was induced by arterial injury. CX 3 CR1 deficiency protected mice from developing intimal hyperplasia as a result of decreased monocyte trafficking to the lesion and VSMC proliferation.

【关键词】  CXCR monocytes smooth muscle cells intimal hyperplasia vascular biology


Introduction


Arterial injury elicits vessel wall inflammation by triggering platelet adhesion, leukocyte recruitment, and vascular smooth muscle cell (VSMC) proliferation and migration. 1-3 These stereotypic cellular responses serve as the basis for many vascular diseases, including atherosclerosis, restenosis, and vasculitis. The molecular mechanisms by which these responses are regulated are of tremendous interest.


Both animal models and human genetic association studies have implicated an important role for the chemokine receptor CX 3 CR1 and its ligand fractalkine (CX 3 CL1) in vascular disease. Mice that lack both CX 3 CR1 and apolipoprotein E (apoE) exhibit a reduction in atherosclerotic lesion formation in the aorta and aortic root compared with apoE-deficient mice. 4,5 CX 3 CL1-deficient mice have a reduction in atherosclerotic plaque burden in the innominate artery. 6 In humans, the V249I/T280M variant of CX 3 CR1 is associated with protection from coronary artery disease 7-9 and internal carotid artery occlusive disease. 10


While CX 3 CR1 and CX 3 CL1 are emerging as important mediators of vascular disease, the specific mechanisms regulating their involvement remain unclear. CX 3 CL1 is a transmembrane chemokine on activated endothelium that also exists in a soluble form. 11-13 Soluble CX 3 CL1 is a potent chemoattractant, and membrane-tethered CX 3 CL1 promotes cell adhesion by supporting the capture and firm adhesion of circulating CX 3 CR1-expressing cells. 14,15 CX 3 CR1 is expressed on monocytes, 16 VSMCs, 17,18 and platelets. 19 Thus, each of these cell types is a candidate to mediate the effects of CX 3 CR1 on vascular inflammatory responses.


The prevailing hypothesis is that the mechanism for CX 3 CR1 in the development of vascular diseases such as atherosclerosis is the adherence of CX 3 CR1-expressing monocytes to inflamed endothelium. 5,14 However, recent evidence that smooth muscle cells (SMCs) found in human atherosclerotic plaques also express CX 3 CR1 suggests an alternative mechanism. 18 Stimulation of aortic smooth muscle cells with soluble CX 3 CL1 leads to an increase in cell survival and proliferation. 20 Monocyte adhesion and arrest on neointimal SMC is dependent on CX 3 CL1, 21 and VSMCs undergo chemotaxis toward CX 3 CL1. 17 Thus, CX 3 CL1 may act not only on monocytes, but also may function to stimulate SMC proliferation and recruitment into the neointima.


In the present study, we employed an endoluminal arterial injury model to investigate the role of CX 3 CR1 in the pathogenesis of vascular injury. This model elicits a stereotypic response that allows for assessment of the functional roles of platelets, monocytes, and VSMCs. Our data show enhanced expression of CX 3 CL1 after injury and a role for CX 3 CR1 in the development of intimal hyperplasia. A deficiency in CX 3 CR1 does not affect platelet function, but does significantly influence monocyte recruitment. CX 3 CR1 deficiency also plays a role in regulating SMC activities.


Materials and Methods


Animal Care


CX 3 CR1 -/- (KO) mice (a gift from Dr Philip Murphy ) were backcrossed onto the C57BL/6J background for 12 generations, and age-matched CX 3 CR1 +/+ (WT) mice generated from littermates were used as controls. All mice were bred and maintained in the barrier facility at the University of North Carolina and fed Prolab RMH-3000 (PMI Nutrition International, Richmond, Ind), a normal rodent chow diet. All experimental protocols were in compliance with Institutional Animal Care and Use Committee (IACUC) guidelines and were approved by the IACUC at the University of North Carolina at Chapel Hill.


Femoral Artery Injury


Femoral arteries of 8- to 12-week-old male CX 3 CR1 -/- and WT mice were injured by endoluminal passage of an angioplasty guide wire as previously described. 22 Arteries on one side were not injured and served as negative controls. Briefly, mice were anesthetized with inhaled isoflurane and femoral arteries were exposed by a longitudinal groin incision and viewed under a surgical microscope. The distal portion of the artery was encircled with an 8-0 nylon suture, a vascular clamp was placed proximally at the level of the inguinal ligament, and a 0.01-inch diameter guide wire (CrossIT-200XT; Guidant Corporation, Indianapolis, Ind) was introduced into the arterial lumen through an arteriotomy made in the distal perforating branch. After release of the clamp, the guidewire was advanced to the level of the aortic bifurcation and immediately pulled back 3 times to denude the endothelium. After removal of the wire, the arteriotomy site was ligated and skin was closed. Animals were routinely monitored after surgery.


Tissue Preparation, Histology, and Morphometry


Mice were euthanized at days 1 (n=20), 5 (n=16), 14 (n=8), and 28 (n=26) after arterial injury. Animals were perfused with phosphate-buffered saline for 5 minutes, followed by 4% paraformaldehyde for 20 more minutes at 100 cm H 2 O via cannulation of the left ventricle. The hind limbs were then harvested en bloc, fixed in 4% paraformaldehyde overnight, and decalcified in formic acid bone decalcifier (Immunocal; Decal Corporation, Tallman, NY) for 24 hours. Tissues containing the femoral artery were embedded in paraffin and cut into 5-µm sections for further analysis.


Six to 10 sections per femoral artery at 100-µm intervals were screened with H&E staining, and sections from the area with maximal injury response were further evaluated by staining with the Combined Masson?s elastin (CME) stain to visualize the arterial wall layers or processed for immunohistochemistry. The intima and media areas were measured by computerized morphometry (Image J, National Institutes of Health). Intimal hyperplasia was defined as the formation of a neointimal layer within the internal elastic lamina (IEL). Media area was calculated as the area encircled by the external elastic lamina (EEL) minus the area encircled by IEL. The intima-to-media (I/M) ratio was calculated as the intimal area divided by the media area. Arteries with a broken IEL or thrombosis by CME stain were excluded from the study.


Immunohistochemistry


For characterizing the molecular and cellular composition of arteries, immunohistochemical analysis was used to identify monocytes by mAb F4/80 (Serotec Inc, Raleigh, NC), VSMCs by alkaline phosphatase conjugated anti- -actin (Sigma, St. Louis, Mo), platelets by anti-thrombocyte (Inter-Cell Technologies, Princeton, NJ), endothelial cells by anti-von Willebrand factor (vWF) (DakoCytomation Inc, Carpinteria, Calif), and CX 3 CL1 by anti-fractalkine (R&D Systems, Minneapolis, Minn). Recombinant mouse fractalkine/CX 3 CL1 (R&D Systems) was used to validate the specificity of CX 3 CL1 staining. Antigen retrieval was performed for F4/80 staining with trypsin digestion and for CX 3 CL1 and vWF staining by steaming in a citrate buffer (0.01 mol/L, pH 6.0) for 40 minutes. Endogenous peroxide activity was quenched using 3% H 2 O 2 in methanol for 10 minutes at room temperature, and sections were blocked using 4% serum (goat, rabbit, or rat) for 10 to 60 minutes at room temperature. Primary antibodies and their respective controls were incubated overnight at 4°C. After washing, sections were incubated with a species-specific biotinylated secondary antibody (anti-mouse, anti-rabbit, anti-rat, and anti-goat, 1:200; Vector Laboratories, Burlingame, Calif) for 60 minutes at room temperature, followed by washes, and a subsequent 60-minute incubation at room temperature with streptavidin-horseradish peroxidase (Peroxidase Vectastain ABC Kit) to amplify antibody signal. After another wash, 3, 3'-diaminobenzidine (DAB) (Sigma, St. Louis, Mo) was used as a substrate for the peroxidase. Alkaline phosphatase staining by -actin antibody was visualized with the Vector Red Alkaline Phosphatase Substrate Kit (Vector Laboratories).


VSMC Proliferation Analysis


Four days after arterial injury, mice were injected intraperitoneally with three doses of bromodeoxyuridine (BrdU) (Roche Diagnostics, Basel, Switzerland) of 30 mg/kg at 8-hour intervals before euthanasia. Arteries were harvested on day 5 after injury, and proliferating cells were identified by immunostaining with an anti-BrdU antibody (Roche Diagnostics). For all times other than day 5 after injury, an anti-PCNA antibody (Santa Cruz Laboratories, Santa Cruz, Calif) was used to identify proliferating cells. Proliferating VSMCs were defined as -actin-positive cells that were also positive for BrdU or PCNA staining in series sections. The proliferation index was calculated as the percentage of BrdU-stained or PCNA-stained nuclei of the total number of nuclei in the indicated area. VSMC proliferation was also determined in vitro utilizing primary cultures of VSMC isolated from aortas of CX 3 CR1 -/- and WT mice with a colorimetric assay based on the uptake of MTT by viable cells (Cell Proliferation Kit; Roche Applied Science, Indianapolis, Ind).


Platelet Function


Platelet function was tested in vitro with the Cone and Plate(let) Analyzer (CPA) system using a DiaMed Impact-R machine (DiaMed Israel Ltd) according to the manufacture?s instructions. Briefly, fresh samples of sodium citrated anticoagulated blood (130 µL) from CX 3 CR1 -/- and WT mice were placed in polystyrene wells and subjected to defined shear (1800/second) for 2 minutes. The samples were then thoroughly washed with phosphate-buffered saline, stained with May-Grünwald stain, and quantitated with an image analyzer. Platelet deposition on the polystyrene surface was evaluated by examining: (1) the percentage of total area covered with platelets designated as surface coverage; and (2) average size in µm 2 of surface-bound platelet thrombi.


Monocyte Adhesion


Monocyte adhesion was evaluated as previously described. 14 Briefly, spleens from CX 3 CR1 -/- and WT mice were homogenized in RPMI-1640 with 10 mmol/L HEPES using a manual tissue homogenizer. Red blood cells were lysed with red blood cell lysis buffer (0.14 mol/L NH 4 CI, 0.017 mol/L Tris-HCl pH7.5, adjust to pH7.2). Cells were washed with RPMI-1640 with 10% fetal bovine serum (FBS), passed through a 70-µm nylon filter, and suspended in RPMI-1640 with 10% FBS. Adhesion of the splenocytes to fractalkine under physiological flow conditions was then tested by the parallel plate flow chamber assay with recombinant fractalkine-secreted alkaline phosphatase fusion proteins immobilized on a glass coverslip. After the run, adherent cells were stained with MOMA-fluorescein isothiocyanate (FITC) antibody (Beckman Coulter, Inc, Fullerton, Calif) and the number of monocytes bound was quantified by counting MOMA+ cells.


Statistical Analysis


Numerical data presented in text and figure are expressed as mean±SEM. All sections were analyzed by 2 investigators, 1 blinded and 1 unblinded, with an inter-rater reliability of 0.95 to 0.99. Fisher?s exact test was utilized to determine incidence. Student unpaired t test was used to compare average numbers of cells or percentages between experimental groups. In all cases, P 0.05 was considered significant.


Results


Characterization of Intimal Hyperplasia After Injury


Wire injury resulted in endothelial denudation as evidenced by an absence of endothelial cells in femoral arteries from WT mice 24 hours after injury. The contralateral, uninjured control vessels retained their normal histology. Platelet adhesion to the injured vessel wall at day 1 was followed by monocyte recruitment to the intima by 5 days after injury ( Figure 1 ). At 14 days, the neointima was a mixture of monocytes and VSMCs, and by 28 days, the neointima was primarily composed of VSMC ( Figure 1 ). Taken together, intimal hyperplasia was readily apparent by 5 days and it was continuously detected between 5 and 28 days after injury. Monocyte recruitment to the intima occurred earlier than VSMC accumulation, which was the primary cellular component of neointima in the late stage (day 28). Eighty-six percent of WT mice developed intimal hyperplasia with an average Intima/Media (I/M) ratio of 0.8.


Figure 1. Cellular composition of injured arteries. Shown are immunohistochemical analyses of WT arteries at 0, 1, 5, 14, and 28 days after arterial injury. Platelets (brown), monocytes (brown), and VSMCs (red) were identified by anti-thrombocyte, anti-F4/80, and anti- -actin antibodies, respectively.


CX 3 CL1 Expression Is Induced in Injured Arteries


To define the role of CX 3 CR1 in the response to arterial injury, we first examined the expression pattern of its ligand, CX 3 CL1, by immunohistochemistry. CX 3 CL1 expression was undetectable in non-injured, control arteries at any stage of the injury response. In injured arteries, CX 3 CL1 was not detected at day 1. At day 5, CX 3 CL1 was expressed in endothelial cells and in a subset of the intimal SMC in WT and CX 3 CR1 -/- arteries ( Figure 2 ). This pattern of expression continued through the injury response. These data provide the evidence that CX 3 CL1 may be involved in the pathogenesis of guide wire induced vascular injury.


Figure 2. CX 3 CL1 staining in injured vessels. Immunostaining of CX 3 CL1 and endothelial cells (vWF) in WT mice is shown in control and injured arteries at 5 days after injury. Control arteries have no CX3CL1 expression and injured arteries show expression of CX3CL1 by vWF+ endothelial cells.


CX 3 CR1-Deficient Mice Are Protected From Intimal Hyperplasia


To assess whether CX 3 CR1 plays a role in mediating intimal hyperplasia after arterial injury, we measured the incidence and extent of neointima formation in CX 3 CR1 -/- and WT mice. The overall incidence of intimal hyperplasia at 5, 14, and 28 days was decreased in CX 3 CR1 -/- mice by 58% (8/21 KO versus 26/29 WT, P =0.0017). Compared with the average intimal area in WT mice, the average intimal area in CX 3 CR1 -/- mice was reduced by 84%, 56%, and 74% at 5, 14, and 28 days, respectively ( Figure 3 ). In contrast, no significant differences in luminal and medial areas were detected between WT and CX 3 CR1 -/- arteries (data not shown). These data indicate that CX 3 CR1 deficient mice are protected from intimal hyperplasia after acute arterial injury, and that the arterial injury model is appropriate to study the functional roles of CX 3 CR1.


Figure 3. Injury-induced intimal hyperplasia in wild type (WT) and CX 3 CR1 -/- (KO) mice. Unilateral wire injury was performed in femoral arteries of WT and CX 3 CR1 -/- mice, with the contralateral uninjured side used as a negative control. A, CME stains of representative sections of control and injured femoral arteries at 28 days after injury. Intimal hyperplasia is defined as the formation of a neointimal layer within the internal elastic lamina (arrows). B, Average intimal areas of injured WT and KO arteries at 5, 14, and 28 days after injury. Results are reported as mean±SEM. ** P <0.05.


Normal Platelet Function in CX 3 CR1 -/- Mice


To define whether platelet CX 3 CR1 is functionally relevant in guide wire induced injury, we examined platelet accumulation on the injured luminal surface by immunostaining. By morphometric analysis, there was no difference in the area of the platelet thrombus that accumulated along injured CX 3 CR1 -/- and WT arteries ( Figure 4A and 4 B). We also examined platelet function under near-physiological conditions in vitro using blood collected from CX 3 CR1 -/- and WT mice. As shown in Figure 4C and 4 D, the absence of CX 3 CR1 did not affect shear-induced platelet thrombus formation. These data suggest that platelet CX 3 CR1 may be not an essential mediator of platelet adhesion or aggregation in response to arterial injury or high shear.


Figure 4. CX 3 CR1 effect on platelet adhesion. One day after injury, control and injured femoral arteries were immunostained for platelets (A, B). A, Platelets (brown) adhering to the luminal surface in vessels of injured WT and injured CX 3 CR1 -/- (KO), but not control WT mice. B, Area of adherent platelets measured by morphometric analysis in injured CX 3 CR1 -/- (n=7) and WT arteries (n=10). Results are expressed as mean±SEM. Shear-induced platelet adhesion represented by surface coverage (C) and aggregation represented by average size (D) were measured in blood from WT (n=4) and CX 3 CR1 -/- (n=4) mice using the CPA technology. Data are expressed as mean±SEM.


Defective Monocyte Recruitment in CX 3 CR1 -/- Mice


CX 3 CR1 is believed to be critically important for monocyte recruitment to the inflamed or injured vessel. We tested this hypothesis by examining monocyte infiltration into the vascular wall by F4/80 antigen staining ( Figure 5 A). There was a 100% and an 87% decrease in monocyte accumulation in CX 3 CR1 -/- mice at 5 and 14 days, respectively, compared with WT mice ( Figure 5 B). Monocyte adhesion to CX3CL1 under physiological flow conditions was also substantially lower with CX 3 CR1 -/- compared with WT cells ( Figure 5 C). Taken together, these results provide strong evidence that CX 3 CR1 plays a critical role in monocyte recruitment to the inflamed vessel wall.


Figure 5. CX 3 CR1 effect on monocyte adhesion. Monocyte recruitment to injured vessels in vivo was tested by immunohistochemical staining of F4/80 antigen (A, B) and adhesion to CX3CL1 in vitro was tested by the parallel plate flow chamber adhesion assay using splenocytes from WT and CX 3 CR1 -/- mice (C). A, Monocyte staining (brown) in injured WT and CX 3 CR1 -/- arteries at 5 days after surgery. B, Average number of monocytes in the intima of WT and CX 3 CR1 -/- (KO) arteries 5 days after injury (n=16). Results are expressed as the mean±SEM. ** P <0.05. C, Average number of Moma2-FITC+ monocytes bound to CX 3 CL1. Results are expressed as the mean±SEM. ** P <0.05.


Role of CX 3 CR1 in VSMC Response to Vascular Injury


To assess whether CX 3 CR1 is important for the function of VSMC in response to arterial injury, we took 3 approaches. First, we evaluated the numbers of VSMC in the vessel wall by immunohistochemistry for -actin. Second, we evaluated VSMC proliferation by BrdU incorporation and by immunohistochemistry for PCNA expression in -actin positive cells. Third, we evaluated the in vitro proliferation of primary VSMC isolated from CX 3 CR1 -/- and WT mouse aortas.


In injured arteries, substantial VSMC proliferation was observed at 5 days and 14 days ( Figure 6 ). At 5 days, most proliferating cells were localized in the media. The proliferation index in CX 3 CR1 -/- mice was decreased by 45% compared with WT mice (8.5±2.7% versus 15.3±3.4%; P =0.07). As the lesion progressed at 14 days, an 85% decrease in proliferating cells was detected in the intima of CX 3 CR1 -/- mice compared with WT animals (3.1±2.0% versus 20±6.0%; P =0.03). In vitro, while WT aortic VSMC proliferated in response to CX3CL1 ( P =0.005), CX 3 CR1 -/- aortic VSMC did not ( P =NS). These results suggest that CX 3 CR1 deficiency diminishes VSMC proliferation in the early development of intimal hyperplasia in response to arterial injury.


Figure 6. VSMC proliferation in response to vascular injury. Shown is the proliferation of VSMCs in the intima and media of injured WT and CX 3 CR1 -/- arteries at 5 and 14 days after injury. Proliferating VSMCs were defined as those -actin positive cells that were BrdU (d5) or PCNA (d14) positive. Results are represented as mean±SEM. * P =0.07, ** P =0.03.


Discussion


Inflammation is a driving force behind vascular diseases such as atherosclerosis and restenosis. Arterial injury is the initial stage of the pathohistological changes of these diseases. We found that guidewire-induced endothelial denudation of mouse femoral arteries elicits an inflammatory response with upregulation of CX 3 CL1 expression. Therefore, we investigated the mechanisms of action of CX 3 CR1 in vascular inflammation by testing the effects of CX 3 CR1 deficiency in this injury model, which stimulates acute inflammatory responses similar to restenosis. Because CX 3 CR1 is expressed on platelets, monocytes and VSMC, we focused on these cell types.


CX 3 CR1 does not appear to play a critical role in platelet accumulation along the denuded endothelial surface. In our model, platelets and neutrophils cover the denuded luminal surface within 24 hours. 22-24 The area of platelet accumulation was unaffected by the absence of CX 3 CR1. Likewise, no significant differences were observed in shear-induced platelet thrombus formation in blood from WT and CX 3 CR1-deficient mice. These results suggest that platelet CX 3 CR1 does not play a major role in the initial phases of platelet adhesion and aggregation stimulated by the vascular injury. Likewise, CX 3 CL1 was not highly expressed within the first 24 hours after injury. Whether CX 3 CR1 contributes to other platelet responses cannot be determined from our study.


Regenerated endothelial cells expressed high levels of CX 3 CL1. Similar to previous reports, 22 we observed regeneration of endothelial cells within 5 days of arterial injury. The endothelium before injury did not express CX 3 CL1, but the regenerated endothelium expressed high levels of CX 3 CL1. In contrast to WT animals that had robust monocyte accumulation, CX 3 CR1 -/- mice failed to recruit monocytes to the intima despite expression of CX 3 CL1. Interestingly, monocytes accumulated in the adventitia of WT and CX 3 CR1 -/- animals after injury, suggesting that monocyte recruitment to the adventitia may occur through mechanisms distinct from those required for intimal recruitment. A primary difference between these two sites is the presence of arterial shear forces, and monocyte capture and transmigration along the artery may require CX 3 CR1-driven adhesion and/or migration to resist the shear. In contrast, recruitment of tissue macrophages or transvenous migration of monocyte to the adventitia may occur by CX 3 CR1-independent mechanisms. While we cannot exclude the possibility that adventitial macrophages traffic and migrate toward soluble CX 3 CL1 through the vessel wall to the luminal surface, the data support a primary role for CX 3 CR1 in the rapid capture and firm adhesion of monocytes under flow conditions. 14 Clinical cohorts have shown that 2 single nucleotide polymorphisms of CX 3 CR1, V249I and T280M, are associated with reduced prevalence of atherosclerosis and coronary artery disease. 8,9 In addition, the M280 allele is associated with a reduced risk of internal carotid artery (ICA) occlusive disease. 10 Our laboratory has shown that the protein encoded by the M280 allele has impaired adhesive capacity, 7 suggesting that cell adhesion is an important mechanism by which CX 3 CR1 exerts its effect on recruiting cells during vascular inflammation.


In addition to CX 3 CR1, CCR2 appears to mediate the response to arterial injury as well. In the same mouse model of wire-induced femoral artery injury, CCR2 -/- mice had a phenotype comparable to CX 3 CR1 -/- mice in terms of a reduced intimal hyperplasia and intima/media ratio 4 weeks after injury. 23 In the CCR2 study, no macrophage infiltration was seen by MOMA-2 or CD68 staining in either WT or CCR2 -/- animals and the authors concluded that CCR2 did not affect macrophage accumulation within the arterial wall after injury. Although both CX 3 CR1 and CCR2 mediate monocyte chemotaxis and trafficking to sites of inflammation, CX 3 CR1 mediates direct monocyte binding to its membrane-bound ligand, CX 3 CL1, without the help of other adhesion molecules, such as integrins. 15 Thus, monocyte trafficking to damaged tissue during arterial injury may favor the CX 3 CR1-CX 3 CL1 system. Recently, Schober et al reported an alternative mechanism for CCR2 in acute vascular injury. 25 In their study, CCL2 was found to be co-localized with platelets on the denuded carotid artery surface of apoE -/- mice fed with a high-fat diet within 24 hours after wire injury. Even though platelets do not contain CCL2, 26 low-affinity CCR2-dependent binding of CCL2 to human platelets is detected in vitro, 27 suggesting that adherent platelets with immobilized CCL2 mediate monocyte recruitment after mechanical injury. Monocyte adhesion to the luminal surface is not detected within 24 hours under our experimental conditions or in similar wire-induced arterial injury models under either normolipidemic 23 or hyperlipidemic 25,28 conditions. Nevertheless, platelet deposition may play a vital role in the pathogenesis of restenosis, 29 and CCR2 may be involved in that process.


SMC accumulate in the intima in both atherosclerosis and restenosis. 30,31 This process is also seen in animals following guidewire-induced arterial injury. CX 3 CR1 is expressed in human SMC cultured from coronary arteries and in atherosclerotic lesions, 17,18 suggesting that CX 3 CR1 may play a role in regulating SMC function. In our study, intimal hyperplasia was significantly decreased in CX 3 CR1 -/- arteries after injury. Concurrently, there were decreased numbers of VSMC in the neointima. VSMC proliferation in response to injury was also impaired in CX 3 CR1 -/- mice as measured by both BrdU incorporation and PCNA immunohistochemistry. Whether the VSMC effects are primary or secondary cannot be addressed by this study. However, several data point to a primary effect of CX 3 CR1 on VSMC function in this model. In a recent report of interleukin (IL)-15?s effect on CX 3 CR1/CX 3 CL1 expression and the response to arterial injury, 32 IL-15 attenuated SMC proliferation and CX 3 CR1/CX 3 CL1 mRNA expression on serum stimulation. Using a periadventitial injury model, the authors further demonstrated that IL-15 upregulation after injury reduced intimal thickening, and blockade of IL-15 increased CX 3 CR1/CX 3 CL1 expression. These results suggested that IL-15 up-regulation decreases neointimal formation in response to arterial injury via suppressing CX 3 CR1 signaling in SMC. CX 3 CR1 is a typical G protein-coupled receptor, and the binding of CX 3 CL1 to CX 3 CR1 initiates multiple signal transduction pathways that are associated with cell proliferation and survival, such as extracellular signal regulated kinase (ERK)/P38 MAPK pathways 33 and the PI3K/Akt pathway. 34 In addition, tumor necrosis factor- stimulates CX 3 CL1 expression in human aortic SMC, 35 and the induction of CX 3 CR1 facilitates SMC proliferation via NF- B in aortic SMCs. 20 Additionally, CX 3 CR1/CX 3 CL1 may directly regulate SMC migration in response to vascular inflammation. Cultured human coronary artery SMC have been shown to express CX 3 CR1 and undergo chemotaxis toward CX 3 CL1. 17 US28, a viral receptor for CX 3 CL1 can mediate SMC migration in vitro. 36 Although direct evidence for a role of CX 3 CR1-mediated SMC migration in arterial injury needs to be further demonstrated in vivo, our findings of substantially reduced intimal hyperplasia and SMC proliferation in CX 3 CR1 -/- mice after mouse femoral artery denudation strongly suggest that, in addition to mediating monocyte infiltration at early stages of the injury response, CX 3 CR1 may also play an important role in vascular remodeling in the late stage of injury by regulating SMC functions.


In summary, we demonstrate that CX 3 CR1 plays a critical role in the regulation of cellular responses to acute vascular injury. CX 3 CL1 expression is upregulated on endothelial and smooth muscle cells after injury. Mice lacking CX 3 CR1 have lower incidence and degree of intimal hyperplasia after injury. The role of CX 3 CR1 in regulating vascular inflammation involves monocyte accumulation in the vessel wall and VSMC proliferation and migration. Combined with data that CX 3 CR1 polymorphisms associated with defective cell adhesion protect humans from atherosclerotic diseases, our findings suggest that CX 3 CR1 could be an effective drug target for both acute and chronic vascular injuries, such as restenosis and atherosclerosis.


Acknowledgments


The authors sincerely thank Gail Grossman and Kirk McNaughton for their technical assistance in tissue processing and staining, Rishi Rampersad for animal husbandry and Drs Teresa Tarrant and Maya Jerath for proofreading the manuscript.


Sources of Funding


This study was supported by National Institute of Health R01s CA098110 (D.D.P.) and HL074219 (S.S.S.).


Disclosures


None.

【参考文献】
  Ross R. Atherosclerosis-an inflammatory disease. N Engl J Med. 1999; 340: 115-126.

Schwartz SM, deBlois D, O?Brien ER. The intima. Soil for atherosclerosis and restenosis. Circ Res. 1995; 77: 445-465.

Charo IF, Taubman MB. Chemokines in the pathogenesis of vascular disease. Circ Res. 2004; 95: 858-866.

Combadiere C, Potteaux S, Gao JL, Esposito B, Casanova S, Lee EJ, Debre P, Tedgui A, Murphy PM, Mallat Z. Decreased atherosclerotic lesion formation in CX3CR1/apolipoprotein E double knockout mice. Circulation. 2003; 107: 1009-1016.

Lesnik P, Haskell CA, Charo IF. Decreased atherosclerosis in CX3CR1-/- mice reveals a role for fractalkine in atherogenesis. J Clin Invest. 2003; 111: 333-340.

Teupser D, Pavlides S, Tan M, Gutierrez-Ramos JC, Kolbeck R, Breslow JL. Major reduction of atherosclerosis in fractalkine (CX3CL1)-deficient mice is at the brachiocephalic artery, not the aortic root. Proc Natl Acad Sci U S A. 2004; 101: 17795-17800.

McDermott DH, Fong AM, Yang Q, Sechler JM, Cupples LA, Merrell MN, Wilson PW, D?Agostino RB, O?Donnell CJ, Patel DD, Murphy PM. Chemokine receptor mutant CX3CR1-M280 has impaired adhesive function and correlates with protection from cardiovascular disease in humans. J Clin Invest. 2003; 111: 1241-1250.

McDermott DH, Halcox JP, Schenke WH, Waclawiw MA, Merrell MN, Epstein N, Quyyumi AA, Murphy PM. Association between polymorphism in the chemokine receptor CX3CR1 and coronary vascular endothelial dysfunction and atherosclerosis. Circ Res. 2001; 89: 401-407.

Moatti D, Faure S, Fumeron F, Amara Mel W, Seknadji P, McDermott DH, Debre P, Aumont MC, Murphy PM, de Prost D, Combadiere C. Polymorphism in the fractalkine receptor CX3CR1 as a genetic risk factor for coronary artery disease. Blood. 2001; 97: 1925-1928.

Ghilardi G, Biondi ML, Turri O, Guagnellini E, Scorza R. Internal carotid artery occlusive disease and polymorphisms of fractalkine receptor CX3CR1: a genetic risk factor. Stroke. 2004; 35: 1276-1279.

Harrison JK, Jiang Y, Wees EA, Salafranca MN, Liang HX, Feng L, Belardinelli L. Inflammatory agents regulate in vivo expression of fractalkine in endothelial cells of the rat heart. J Leukoc Biol. 1999; 66: 937-944.

Garton KJ, Gough PJ, Blobel CP, Murphy G, Greaves DR, Dempsey PJ, Raines EW. Tumor necrosis factor-alpha-converting enzyme (ADAM17) mediates the cleavage and shedding of fractalkine (CX3CL1). J Biol Chem. 2001; 276: 37993-38001.

Tsou CL, Haskell CA, Charo IF. Tumor necrosis factor-alpha-converting enzyme mediates the inducible cleavage of fractalkine. J Biol Chem. 2001; 276: 44622-44626.

Fong AM, Robinson LA, Steeber DA, Tedder TF, Yoshie O, Imai T, Patel DD. Fractalkine and CX3CR1 mediate a novel mechanism of leukocyte capture, firm adhesion, and activation under physiologic flow. J Exp Med. 1998; 188: 1413-1419.

Haskell CA, Cleary MD, Charo IF. Molecular uncoupling of fractalkine-mediated cell adhesion and signal transduction. Rapid flow arrest of CX3CR1-expressing cells is independent of G-protein activation. J Biol Chem. 1999; 274: 10053-10058.

Imai T, Hieshima K, Haskell C, Baba M, Nagira M, Nishimura M, Kakizaki M, Takagi S, Nomiyama H, Schall TJ, Yoshie O. Identification and molecular characterization of fractalkine receptor CX3CR1, which mediates both leukocyte migration and adhesion. Cell. 1997; 91: 521-530.

Lucas AD, Bursill C, Guzik TJ, Sadowski J, Channon KM, Greaves DR. Smooth muscle cells in human atherosclerotic plaques express the fractalkine receptor CX3CR1 and undergo chemotaxis to the CX3C chemokine fractalkine (CX3CL1). Circulation. 2003; 108: 2498-2504.

Wong BW, Wong D, McManus BM. Characterization of fractalkine (CX3CL1) and CX3CR1 in human coronary arteries with native atherosclerosis, diabetes mellitus, and transplant vascular disease. Cardiovasc Pathol. 2002; 11: 332-338.

Schafer A, Schulz C, Eigenthaler M, Fraccarollo D, Kobsar A, Gawaz M, Ertl G, Walter U, Bauersachs J. Novel role of the membrane-bound chemokine fractalkine in platelet activation and adhesion. Blood. 2004; 103: 407-412.

Chandrasekar B, Mummidi S, Perla RP, Bysani S, Dulin NO, Liu F, Melby PC. Fractalkine (CX3CL1) stimulated by nuclear factor kappaB (NF-kappaB)-dependent inflammatory signals induces aortic smooth muscle cell proliferation through an autocrine pathway. Biochem J. 2003; 373: 547-558.

Zeiffer U, Schober A, Lietz M, Liehn EA, Erl W, Emans N, Yan ZQ, Weber C. Neointimal smooth muscle cells display a proinflammatory phenotype resulting in increased leukocyte recruitment mediated by P-selectin and chemokines. Circ Res. 2004; 94: 776-784.

Roque M, Fallon JT, Badimon JJ, Zhang WX, Taubman MB, Reis ED. Mouse model of femoral artery denudation injury associated with the rapid accumulation of adhesion molecules on the luminal surface and recruitment of neutrophils. Arterioscler Thromb Vasc Biol. 2000; 20: 335-342.

Roque M, Kim WJ, Gazdoin M, Malik A, Reis ED, Fallon JT, Badimon JJ, Charo IF, Taubman MB. CCR2 deficiency decreases intimal hyperplasia after arterial injury. Arterioscler Thromb Vasc Biol. 2002; 22: 554-559.

Smyth SS, Reis ED, Zhang W, Fallon JT, Gordon RE, Coller BS. Beta(3)-integrin-deficient mice but not P-selectin-deficient mice develop intimal hyperplasia after vascular injury: correlation with leukocyte recruitment to adherent platelets 1 hour after injury. Circulation. 2001; 103: 2501-2507.

Schober A, Zernecke A, Liehn EA, von Hundelshausen P, Knarren S, Kuziel WA, Weber C. Crucial role of the CCL2/CCR2 axis in neointimal hyperplasia after arterial injury in hyperlipidemic mice involves early monocyte recruitment and CCL2 presentation on platelets. Circ Res. 2004; 95: 1125-1133.

Gear AR, Camerini D. Platelet chemokines and chemokine receptors: linking hemostasis, inflammation, and host defense. Microcirculation. 2003; 10: 335-350.

Clemetson KJ, Clemetson JM, Proudfoot AE, Power CA, Baggiolini M, Wells TN. Functional expression of CCR1, CCR3, CCR4, and CXCR4 chemokine receptors on human platelets. Blood. 2000; 96: 4046-4054.

Weingartner O, Kasper M, Reynen K, Bramke S, Marquetant R, Sedding DG, Braun-Dullaeus R, Strasser RH. Comparative morphometric and immunohistological assessment of the development of restenosis after arterial injury and a cholesterol-rich diet in apolipoprotein E -/-mice and C57BL/6 control mice. Coron Artery Dis. 2005; 16: 391-400.

Farb A, Sangiorgi G, Carter AJ, Walley VM, Edwards WD, Schwartz RS, Virmani R. Pathology of acute and chronic coronary stenting in humans. Circulation. 1999; 99: 44-52.

Reidy MA, Fingerle J, Lindner V Factors controlling the development of arterial lesions after injury. Circulation. 1992; 86: III43-III46.

Schwartz RS Pathophysiology of restenosis: interaction of thrombosis, hyperplasia, and/or remodeling. Am J Cardiol. 1998; 81: 14E-17E.

Cercek M, Matsumoto M, Li H, Chyu KY, Peter A, Shah PK, Dimayuga PC. Autocrine role of vascular IL-15 in intimal thickening. Biochem Biophys Res Commun. 2006; 339: 618-623.

Cambien B, Pomeranz M, Schmid-Antomarchi H, Millet MA, Breittmayer V, Rossi B, Schmid-Alliana A. Signal transduction pathways involved in soluble fractalkine-induced monocytic cell adhesion. Blood. 2001; 97: 2031-2037.

Shulby SA, Dolloff NG, Stearns ME, Meucci O, Fatatis A. CX3CR1-fractalkine expression regulates cellular mechanisms involved in adhesion, migration, and survival of human prostate cancer cells. Cancer Res. 2004; 64: 4693-4698.

Ollivier V, Faure S, Tarantino N, Chollet-Martin S, Deterre P, Combadiere C, de Prost D. Fractalkine/CX3CL1 production by human aortic smooth muscle cells impairs monocyte procoagulant and inflammatory responses. Cytokine. 2003; 21: 303-311.

Streblow DN, Soderberg-Naucler C, Vieira J, Smith P, Wakabayashi E, Ruchti F, Mattison K, Altschuler Y, Nelson JA. The human cytomegalovirus chemokine receptor US28 mediates vascular smooth muscle cell migration. Cell. 1999; 99: 511-520.


作者单位: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 Cardiovascular Biology Center (M.R., S.S.S.), University of North Carolina at Chapel Hill, Chapel Hill, NC.

作者: Peng Liu; Sarita Patil; Mauricio Rojas; Alan M. Fo
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具