当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 动脉硬化血栓血管生物学 > 2005年 > 第9期 > 正文
编号:11257790
Genetic Deletion or Antibody Blockade of 1?1 Integrin Induces a Stable Plaque Phenotype in ApoE–/– Mice
     From the Department of Pathology (K.S., E.L., A.R., M.D., S.H.), Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; and Biogen Idec (A.d.F., A.S., H.G., V.K.) and Alnylam Pharmaceuticals (A.d.F., A.S., V.K.), Cambridge, Mass.

    Correspondence to S. Heeneman, PhD, Department of Pathology, P. Debeyelaan 25, 6229 HX Maastricht, The Netherlands. E-mail sheen@lpat.azm.nl

    Abstract

    Objective— Adhesive interactions between cells and the extracellular matrix play an important role in inflammatory diseases like atherosclerosis. We investigated the role of the collagen-binding integrin 1?1 in atherosclerosis.

    Methods and Results— ApoE–/– mice were 1-deficient or received early or delayed anti-1 antibody treatment. Deficiency in 1 integrin reduced the area of atherosclerotic plaques and altered plaque composition by reducing inflammation and increasing extracellular matrix. In advanced plaques, 1-deficient mice had a reduced macrophage and CD3+ cell content, collagen and smooth muscle cell content increased, lipid core sizes decreased, and cartilaginous metaplasia occurred. Anti-1 antibody treatment reduced the macrophage content in initial plaques after early and delayed treatment, decreased the CD3+ cell content in advanced plaques after delayed treatment, and increased the collagen content in initial and advanced plaques after delayed treatment. Migration assays performed on 1-deficient macrophages on collagen I and IV substrata revealed that 1-deficient cells can migrate on collagen I, but not IV. Anti-1 antibody treatment of ApoE–/– macrophages also inhibited migration of cells on collagen IV.

    Conclusions— Our results suggest that 1?1 integrin is involved in atherosclerosis by mediating the migration of leukocytes to lesions by adhesion to collagen IV. Blocking this integrin reduces atherosclerosis and induces a stable plaque phenotype.

    The role of 1?1 integrin was investigated in atherosclerosis. 1?1 integrin is involved in atherosclerosis by mediating the migration of leukocytes to lesions via adhesion to collagen IV. Blocking this integrin reduces atherosclerosis and induces a more stable plaque phenotype.

    Key Words: atherosclerosis ? macrophages ? extracellular matrix ? collagen ? integrin

    Introduction

    Inflammatory responses, as in atherosclerosis, involve adhesive interactions between cells and the extracellular matrix (ECM) that are necessary for cell attachment, extravasation and migration into tissues, proliferation, and differentiation. Major receptors for extracellular matrix ligands are integrins, which are transmembrane heterodimers consisting of an and ? subunit. The role of several integrins has previously been studied in atherosclerotic development. Integrin 4?1 has been shown to be expressed on smooth muscle cells (SMC) during pathological neointimal thickening and is involved in SMC differentiation.1 Furthermore, 4?1 integrin mediates adhesion of monocytes to the vascular endothelium, and anti-4 antibody treatment decreases leukocyte entry in mice fed an atherogenic diet2 as well as attenuates neointimal growth following carotid injury in ApoE–/– mice.3 Agonists against V?3 integrin were also shown to reduce neointima formation after injury,4 and it has been suggested that this integrin regulates the maturation of macrophages into foam cells.5 Yet another model of endothelial injury has revealed that 2?1 integrin mediates the adhesion of platelets to collagen in the vessel wall, and 2?1-deficient mice exhibit delayed thrombus formation after injury to the carotid artery.6

    The role of 1?1 integrin in atherosclerosis has not yet been investigated but has been shown to mediate inflammation in several other illnesses. Integrin 1?1 is a major collagen receptor that is highly expressed on activated monocytes7 and T-lymphocytes, including those found in atherosclerotic plaques.8 The role of 1?1 integrin has been studied in various models of inflammatory diseases including hypersensitivity and arthritis,9 kidney fibrosis,10 and colitis,11 in which it has been shown that a deficiency or blocking of this integrin using anti-1 antibodies attenuates the inflammatory response as seen by a reduced leukocyte infiltrate. These studies suggest a role for 1?1 integrin in the migration of leukocytes to sites of inflammation.

    See cover

    Besides its expression on immune cells, 1?1 integrin is also expressed on mesenchymal cells, most notably SMCs and fibroblasts. Human aortic SMCs have been reported to express 1?1 integrin and is involved in SMC differentiation.12 It has been demonstrated that 1-deficient fibroblasts cannot adhere to or migrate on collagen IV13 and are deficient in collagen-dependent proliferation.14 A major function attributed to 1?1 integrin is the regulation of feedback inhibition of collagen synthesis. This was shown by enhanced collagen-dependent downregulation of collagen synthesis on receptor stimulation,15 and a later study reported an increase in collagen synthesis in the dermis of 1-deficient mice.16

    Given that 1?1 integrin regulates collagen synthesis and mediates the migration of activated leukocytes during inflammation, we wanted to investigate its role in atherosclerosis. To examine the role of 1?1 integrin in plaque inflammation and development, we used a murine model of atherosclerosis in which 1 integrin was either knocked out or mice were treated with an 1-blocking antibody. Here we report that a deficiency or blocking of 1 integrin attenuates atherosclerosis and induces a stable plaque phenotype.

    Methods

    Mice

    To study the role of 1 integrin in atherosclerosis, 1–/–//ApoE–/– mice were used. ApoE–/– mice on a C57BL/6 background were obtained from The Jackson Laboratory (Bar Harbor, Me) and were backcrossed 10 times with 1–/– mice, originally on a Balb/c background13 to generate male and female 1–/–//Apo–/– (homozygous, n=13), 1+/–//ApoE–/– (heterozygous, n=8) and 1+/+//ApoE–/– (wild-type, WT, n=12) mice. Mice were fed normal chow for 27 weeks after which they were euthanized after an 8-hour fast. Approximately 0.5 to 1.0 mL of blood was drawn from the vena cava for lipoprotein analysis. To study effects of antibody blockade of 1 integrin, ApoE–/– mice on a C57BL/6 background were used, obtained from Iffa Credo S.A. (Charles River Co, Lyon, France) and fed normal chow. Mice were injected intraperitoneally twice a week with 200 μg9 of a murinized version of a hamster anti-rodent 1-blocking antibody (muHa31/8) or murine IgG isotype control antibody (MOPC21) (Biogen Idec, Cambridge, Mass). Mice received treatment for 12 weeks beginning at either 5 weeks of age when no atherosclerotic lesions are present in the aortic arch (early treatment: anti-1 Ab n=13, control n=14) or 17 weeks of age when advanced lesions begin to appear in the aortic arch (delayed treatment: anti-1 Ab n=11, control n=14). Animal experiments were approved by the institutional committee for the welfare of laboratory animals of the University of Maastricht.

    Antibody Infiltration

    To determine whether antibody infiltrated plaques, muHa31/8 monoclonal antibody (mAb) was labeled with Alexa488 using a protein labeling kit (Molecular Probes), and 200 μg were injected intraperitoneally into 53-week-old ApoE–/– mice 3 times in the period of one week. Control mice did not receive any antibody. After treatment, mice were euthanized and the arterial tree was briefly perfused with PBS. Aortic arches were snap frozen in Tissue Tek OCT compound for histological analysis, and carotid arteries and abdominal aortas were placed in PBS until analysis by two-photon laser scanning microscopy (TPLSM). Aortic arch cryosections were fixed in acetone, counterstained with hematoxylin and mounted with Prolong Anti-Fade (Molecular Probes), and viewed with a fluorescence microscope. For analysis by TPLSM, carotid arteries and abdominal aortas were placed in perfusion chambers and glass micro-pipettes were inserted into their ends so they could be infused with PBS and kept taut during imaging.

    Lipid Profiles

    Lipid profiles were assessed as described previously.17

    Histology and Morphometry

    Histological and morphometric analyses were performed as described previously.17 To confirm the presence of cartilaginous metaplasia observed in plaques of 1-deficient mice, aortic arch sections were stained with alcian blue and toluidine blue.

    Immunohistochemistry

    Aortic arch sections were immunostained with Mac3 rat mAb (1:30; Pharmingen) to detect macrophages, CD3 rabbit polyclonal antibody (1:200; Dako) to detect T-lymphocytes, and SMAFITC mAb (1:500; Sigma) as a marker for -smooth muscle actin–positive vascular SMCs. To determine 1 integrin expression in macrophages in atherosclerotic plaques of aortic arches, double immunohistochemistry was performed using Alexa488-conjugated Ha31/8 anti-1 mAb (Biogen Idec) and CD11b-PE (1:200; Pharmingen). To identify characteristics of cartilaginous metaplasia, sections were stained with antibodies against collagen II (goat polyclonal, 1:75; Santa Cruz Biotechnology, Inc), osteocalcin (OC) (rabbit polyclonal, 1:50; ANAWA Trading SA), osteonectin (ON) (rabbit polyclonal, 1:2000; Zymed Laboratories, Inc), osteopontin (OPN) (goat polyclonal, 1:25), bone morphogenetic protein 2 and 4 (BMP-2, mouse monoclonal, 1:20; Genetics Institute, Inc., BMP-4, goat polyclonal, 1:25; Santa Cruz), matrix GLA protein (MGP) (mouse monoclonal, 1:25), osteoprotegerin (OPG, 1:100) and osteoprotegerin ligand (OPGL, 1:75). To determine collagen IV expression in atherosclerotic plaques, mouse aortic arch and human carotid artery paraffin sections were stained with antibody against collagen IV (rabbit polyclonal anti-mouse 1:300; anti-human 1:50).

    Cell Isolation

    Bone marrow–derived macrophages were isolated from the femur and tibia of 1–/–//Apo–/– and 1+/+//Apo–/– mice. Cells were cultured in standard RPMI containing L-glutamine, HEPES, 10% fetal calf serum, 100 IU/mL penicillin/streptomycin, and 15% L929 cell conditioned medium.

    Migration Assay

    Cell migration was assayed using 24-well Transwell migration chambers (Costar) with a pore size of 8 μm. Membranes were coated with 110 μg/mL of type I collagen (isolated as previously described)18 or 50 μg/mL of type IV collagen (BD Biosciences) and incubated at 37°C for 1 hour until complete gel formation. Membranes were allowed to air dry, then 106 cells suspended in serum-free medium were added to each chamber. Cells isolated from WT mice were incubated with 100 μg/mL of 1-blocking antibody or control antibody for 30 minutes before addition to chambers. Complete medium including 100 ng/mL MCP-1 (R&D Systems, Inc) was added to lower chambers, and migration was carried out at 37°C for 4 and 12 hours. Nonmigrated cells were removed from membranes, and migrated cells within the membrane were fixed with methanol and stained with toluidine blue. Membranes were cut out of inserts and mounted onto slides in immersion oil. The number of migrated cells was counted on 5 randomly chosen microscopic fields of each membrane.

    FACS Analysis

    Fluorescence-activated cell sorting (FACS) analysis (FACS Calibur, Becton Dickinson) was performed on cells isolated from peripheral blood, spleen, and lymph nodes as well as peritoneal macrophages from antibody-treated mice (anti-1 Ab n=6, control Ab n=6 of both early and delayed treatment groups). Cells were labeled with T lymphocyte–specific antibodies: CD3FITC, CD4Cy-Chrome, CD8PE and CD25PE, and antibodies to detect macrophages: Gr1FITC and Mac1PE (Pharmingen).

    Statistics

    Statistical analyses were performed using a nonparametric Mann–Whitney U test. Data are expressed as mean±SEM, and differences were considered statistically significant at P<0.05.

    Results

    1 Deficiency

    General

    Body weight did not differ in 1-deficient mice compared with controls and plasma total cholesterol, triglycerides, low-density lipoprotein (LDL), and high-density lipoprotein (HDL) levels did not differ between 1–/– and 1+/+ mice, but were elevated in 1+/– relative to 1+/+ mice (Table I, available online at http://atvb.ahajournals.org). Autopsy of organs revealed no abnormalities or pathologies.

    Expression of 1 Integrin

    Expression of 1 integrin was present on SMCs and slightly on T-lymphocytes (data not shown). Expression was also detected on macrophages in atherosclerotic plaques. Figure I (available online at http://atvb.ahajournals.org) shows CD11b+ macrophages in atherosclerotic plaques that stained positive for 1 integrin.

    Extent of Atherosclerosis

    Complete deletion of 1 integrin reduced total plaque area by 42% compared with WT mice (Figure 1). The decrease in plaque area was attributable to a 38% decrease in advanced atherosclerotic plaque area. Absence of one 1 allele was sufficient to significantly reduce the total plaque area as seen in heterozygous mice in which total plaque area was reduced by 37% and advanced plaque area was reduced by 16% compared with WT mice (P<0.05). Plaques representative of the three groups of mice are shown in Figure 1c, in which it can be seen that plaque area was significantly reduced in 1 homozygous and heterozygous mice compared with controls.

    Figure 1. Deficiency in 1 integrin reduced plaque area in aortic arches. A, Total plaque area of 1–/– and 1+/– mice was significantly smaller than those of 1+/+ mice. B, Average area of individual advanced plaques of 1–/– mice was smaller compared with those of 1+/+ mice. C, HE-stained sections of representative advanced atherosclerotic plaques of 1–/–, 1+/–, and 1+/+ mice showing relative area of plaques. *P<0.05.

    Atherosclerotic Plaque Composition

    To further characterize atherosclerosis, the composition of lesions was analyzed. Histological analysis of advanced plaques demonstrated that 1-deficiency resulted not only in a decreased size, but also a dramatic alteration in plaque composition. Lesions from 1-deficient mice were more fibrotic and less lipid-rich than those of WT mice, whereas plaques of 1 heterozygous mice were of an intermediate phenotype (Figure 2). Analysis of initial lesions in the aortic arch revealed that the relative number of CD3+ cells, macrophages, collagen, and SMA content did not differ in initial atherosclerotic plaques. Similarly, initial lesions in heterozygous mice were not altered compared with those in WT mice.

    Figure 2. Plaque composition of initial and advanced atherosclerotic plaques in aortic arches of 1-deficient mice. There were significantly less (A) macrophages (Mac3+, red stained cells) and (B) CD3+ cells (arrows) in advanced plaques of 1–/– compared with 1+/+ mice; there was a significantly higher (C) collagen content (SR+) and (D) SMC content (SMA+ cells, arrows) in advanced plaques of 1–/– compared with 1+/+ mice. E, Lipid cores were smaller in plaques of 1–/– compared with 1+/+ mice (arrows). *P<0.05.

    Differences in plaque composition were more pronounced in advanced lesions of homozygous and heterozygous mice. Inflammatory cell content significantly decreased in homozygous mice, reflected by a decrease in the percentage of macrophages and CD3+ cells. Furthermore, lipid cores of plaques were smaller whereas extracellular matrix as measured by collagen and SMA content increased. Atherosclerotic plaques of heterozygous mice showed an intermediate plaque phenotype with a significant decrease in CD3+ cells and a trend toward a decrease in macrophage content (P=0.07) and collagen content (P=0.06).

    The presence of cartilaginous metaplasia observed in advanced plaques of 1-deficient mice was confirmed with immunohistochemical staining against collagen II, which is the major matrix protein of cartilage tissue, as well as with alcian blue and toluidine blue, which stain chondroid tissue (Figure II, available online at http://atvb.ahajournals.org). To further identify characteristics of cartilaginous metaplasia, we performed immunohistochemistry using bone markers. The regulators of bone formation OC, ON, OPN, BMP-2, and BMP-4 were present in areas of cartilaginous metaplasia (Figure III, available online at http://atvb.ahajournals.org). MGP, another protein involved in bone formation, was only present in plaque macrophages. Two regulators of osteoclastogenesis, OPG and OPGL, did not show expression.

    Antibody Treatment

    General

    Average body weight of mice did not differ in the anti-1 antibody–treated group compared with controls in either of the early or delayed treatment groups. There were no differences in plasma total cholesterol, triglycerides, or LDL, however HDL was slightly elevated in anti-1 antibody–treated mice compared with controls in the early treatment group (Table I). Autopsy of organs revealed no abnormalities or pathologies.

    To ascertain possible systemic effects of antibody treatment on atherosclerotic lesion development, FACS analysis was performed on cells isolated from peripheral blood, spleen, and lymph nodes and on peritoneal macrophages of anti-1 antibody–treated and control mice. Analysis revealed no differences in the amount of Mac1+ macrophages between the two groups in either blood or peritoneum. Similarly, there were no differences in the amount of CD4+ (helper), CD8+ (cytotoxic) T-lymphocytes, nor in the activation status of T-lymphocytes (reflected by CD4+/CD8+ ratio and CD4+CD25+ cells) between the 2 groups in any of the tissues. There was, however, an increase in the amount of CD3+ T-lymphocytes (total T-lymphocytes) in peripheral blood of anti-1 antibody–treated mice compared with controls in both the early and delayed treatment groups (Figure IV, available online at http://atvb.ahajournals.org).

    Antibody Infiltration

    To ascertain whether anti-1 antibody infiltrated atherosclerotic plaques, we used TPLSM and fluorescence microscopy. Atherosclerotic plaques of mice injected with Alexa488-labeled anti-1 antibody displayed more fluorescence in plaques of aortic arches compared with control mice, which were not injected with antibody. Similarly, using TPLSM, which combines 3-D resolution and large penetration depth,19 showed a positive signal for antibody labeled with the green fluorescent label Alexa488 in atherosclerotic plaques in carotid arteries (Figure V, available online at http://atvb.ahajournals.org).

    Extent of Atherosclerosis

    There were no significant differences in the individual plaque area between anti-1 antibody–treated and control mice in either of the early (initial: anti-1 mAb 14 596±1455 versus control 15 761±1955, P=0.06; advanced: anti-1 mAb 49 593±8051 versus control 91 519±14468, P>0.05) or delayed treatment groups (initial: anti-1 mAb 45 184±9635 versus control 31 076±5729, P>0.05; advanced: anti-1 mAb 141 300±19 809 versus control 120 498±10 671, P>0.05).

    Atherosclerotic Plaque Composition

    Differences in plaque composition between anti-1 antibody–treated and control mice were less striking than in 1-deficient mice. Significant differences observed were a decreased macrophage content in initial atherosclerotic plaques of anti-1 antibody–treated mice compared with controls in both the early and delayed treatment groups (Figure 3a), a decreased CD3+ cell content in advanced plaques of anti-1 antibody–treated mice of the delayed treatment group compared with controls (Figure 3b), and an increased collagen content in initial and advanced plaques of anti-1 antibody–treated mice compared with controls of the delayed treatment group (Figure 3c). There were no differences in SMA content in any of the groups (early treatment, initial: anti-1 Ab 3.0±0.6 versus control 1.8±0.4, P>0.05; early treatment, advanced: anti-1 Ab 1.9±1.1 versus control 3.7±0.5, P>0.05; delayed treatment, initial: anti-1 Ab 0.7±0.5 versus control 2.8±1.1, P>0.05; delayed treatment, advanced: anti-1 Ab 3.4±0.6 versus control 4.5±0.4, P>0.05).

    Figure 3. Plaque composition of initial and advanced atherosclerotic plaques in aortic arches of antibody-treated mice. There were significantly less (A) macrophages (Mac3+, red stained cells) and (B) CD3+ cells (brown stained cells, arrows) and a higher (C) collagen content (SR+ areas shown in red) in plaques of anti-1 antibody-treated compared with control mice (Lu denotes lumen). *P<0.05.

    Cell Migration Assay

    Because we observed a reduced macrophage content in both 1-deficient and anti-1 antibody–treated mice, we wanted to test the ability of 1-deficient bone marrow–derived macrophages to migrate on collagen substrata. Cells were allowed to migrate on collagen I and IV monolayers as it has previously been reported that 1 integrin binds these types of collagen.13 After 4 and 12 hours of incubation, significantly less 1–/– cells had migrated on collagen IV compared with WT cells (Figure 4a). There was no significant difference in the number of 1-deficient cells migrating on collagen I compared with WT cells after 4 or 12 hours of migration (Figure 4b). Treatment of WT cells with 1-blocking antibody inhibited migration on collagen IV compared with control antibody after 4 and 12 hours of migration (Figure 4c).

    Figure 4. Cells lacking 1 integrin or receiving anti-1 antibody treatment are deficient in migrating on collagen IV, but not collagen I. A, Significantly less 1–/– macrophages migrated on collagen IV compared with 1+/+ after 4 and 12 hours. B, No significant difference in the number of 1–/– compared with 1+/+ macrophages migrated on collagen I after 4 and 12 hours. C, Significantly less anti-1 antibody-treated compared with control antibody-treated macrophages migrated on collagen IV after 4 and 12 hours. *P<0.05.

    Collagen IV Expression

    Because 1-deficient macrophages as well as macrophages treated with 1-blocking antibody were inhibited from migrating on collagen type IV, the expression of collagen IV was determined in atherosclerotic plaques. Collagen IV is strongly expressed in the region of endothelial cells and slightly in areas surrounding cells in both mouse and human atherosclerotic plaques (Figure VI, available online at http://atvb.ahajournals.org).

    Discussion

    To investigate the role of 1?1 integrin in atherosclerosis we have used 1 integrin, ApoE double knockout mice as well as administered an 1-blocking antibody to normal ApoE knockout mice in an early and delayed treatment setting. Deficiency in 1 integrin did not prevent the initiation of lesion formation, but did reduce the size of advanced atherosclerotic plaques and induced a more stable plaque phenotype as characterized by decreased inflammation and increased extracellular matrix content. In both the early and delayed anti-1 antibody treatment groups, macrophage content was decreased in initial plaques whereas collagen content increased in advanced plaques. Furthermore, the CD3+ cell content was decreased in advanced plaques after delayed treatment.

    Antibody treatment against 1 integrin was less effective in attenuating atherosclerosis compared with complete genetic deletion; however, the consequences of antibody intervention do not necessarily correlate with the phenotype of corresponding null animals. Knockout animals are deficient in a particular protein from birth, whereas antibody-treated mice receive treatment against the already existing protein beginning at a later time point for a limited period of time. Furthermore, in the present study we euthanized 1-deficient mice after 26 weeks of age, whereas in the antibody intervention study mice were euthanized after 17 or 29 weeks of age in the early and delayed treatment groups, respectively. Previous studies have also demonstrated differences between effects of using knockout mice and antibody intervention. For instance, complete genetic deletion of CD154 (CD40 ligand) in ApoE–/– mice resulted in smaller advanced plaques,20 whereas with anti-CD40 ligand antibody treatment, advanced plaque area was not reduced compared with controls.21

    Despite differences in the effects of 1-deficiency and anti-1 antibody treatment, plaque composition was altered in both experiments to a more stable phenotype in two important respects, by reducing the macrophage content and increasing the collagen content. Indeed, the two major functions of 1?1 integrin are the regulation of collagen synthesis and the mediation of migration of activated leukocytes into inflamed tissues.7,16 During the course of inflammation, leukocytes migrate through the subendothelial basement membrane,7 which is rich in collagen type IV.22 Central to the migration of cells into inflammatory sites are adhesive interactions between cells and extracellular matrix proteins that are widely mediated by the integrin family of adhesion molecules.23 1?1 is a major collagen-binding integrin with a preference for collagen IV and is expressed on activated leukocytes. It has been demonstrated in vitro that lipopolysaccharide (LPS)-activated monocytes highly express 1?1 integrin.24 Activated T-lymphocytes also express 1?1 and it has been shown in vivo to be involved in the migration and retention of leukocytes in tissues.25

    T-lymphocytes found in atherosclerotic plaques express 1?1 integrin,8 and we have demonstrated here that macrophages in normal mouse atherosclerotic plaques also express this integrin. However, the role of 1?1 integrin in lesion development has not yet been determined. Our study revealed a reduced inflammatory cell content particularly in lesions of 1-deficient, but also in anti-1 antibody–treated mice, suggesting a role for 1?1 integrin in the accumulation of leukocytes in atherosclerotic plaques. To shed light on the mechanism by which this occurs we performed migration assays, which demonstrated that 1-deficient and anti-1 antibody–treated macrophages are inhibited from migrating on collagen IV. The subendothelial basement membrane of vessel walls consists largely of collagen IV, through which leukocytes must migrate to enter the intima during atherogenesis.26 In the present study, we have also shown by immunohistochemical staining that collagen IV is expressed in the region of endothelial cells in both human and mouse atherosclerotic plaques. It appears, therefore, that 1?1 integrin is necessary for the infiltration of leukocytes during atherogenesis. Results of the migration assays also suggest that antibody treatment against 1?1 integrin was successful in blocking its function in vivo.

    Furthermore, there was an increase in the level of CD3+ T-lymphocytes in the peripheral blood of anti-1 antibody–treated mice as determined by FACS analysis as well as a significantly reduced CD3+ cell content in advanced plaques after delayed anti-1 antibody treatment. These findings are consistent with a role of 1?1 in the migration and/or retention of activated T-lymphocytes in collagen-rich tissues.27 By preventing migration and localization of 1?1-positive T-lymphocytes in tissues such as the vessel wall, antibody treatment against 1?1 results in an increased circulating T-lymphocyte population.

    Our findings are consistent with previous studies on 1?1 integrin in various models of inflammatory diseases. Monocyte accumulation and activation was found to be reduced in mouse models of colitis in which 1 integrin was deleted or blocked by antibody treatment.11 A decrease in macrophage accumulation was also observed in a model of kidney fibrosis in which 1 integrin was deleted.10 A deficiency in 1?1 integrin as well as anti-1 antibody treatment was reported to be protective against experimental murine arthritis, delayed-type hypersensitivity, and contact hypersensitivity as shown by a reduced leukocyte infiltrate.9 In addition, in vitro 1-blocking antibody treatment attenuated the proliferation of T-lymphocytes isolated from draining lymph nodes of arthritic rats28 and inhibited T-lymphocytes cultured from peripheral blood of arthritic patients from migrating on collagen IV.29

    Besides mediating activated leukocyte migration to sites of inflammation, 1?1 integrin regulates collagen synthesis by negative feedback inhibition. This was demonstrated in 1-deficient mice in which steady state collagen synthesis was observed in normal and wounded dermis.16 Furthermore, receptor stimulation has been shown to increase the downregulation of collagen synthesis.15 In our study we found an increased collagen content in atherosclerotic plaques of both 1-deficient and anti-1 antibody–treated mice. This was particularly evident in advanced plaques, which is to be expected given that initial plaques consist of little or no collagen. An increased collagen content in atherosclerotic lesions contributes to their stability as does a thick fibrous cap, which we also observed in 1-deficient mice. Atherosclerotic lesions consist primarily of collagen types I and III and although 1?1 prefers to bind collagen IV, it is also a receptor for collagen I. It has been reported that 1?1 integrin downregulates collagen I mRNA levels in cells suspended in collagen gels,30 and collagen I synthesis was increased in the dermis of 1-deficient mice because of elevated collagen I mRNA levels.16 This suggests that negative feedback inhibition of collagen synthesis mediated by 1?1 integrin occurs in atherosclerosis.

    In addition to an increased collagen content, we found an increase in SMC content as revealed by SMA-positive cells in advanced atherosclerotic plaques of 1-deficient mice. SMCs are extracellular matrix–producing cells that are responsible for the collagens present in atherosclerotic plaques and are particularly involved in the formation of fibrous caps. The abundance of SMCs in atherosclerotic plaques is therefore important to plaque stability, however the role of 1?1 integrin in SMC proliferation and accumulation in atherosclerotic plaques is unclear.

    Along with a change in plaque composition to a more stable plaque phenotype, cartilaginous metaplasia was observed in plaques of 1-deficient mice. Cartilaginous metaplasia may be a pathway by which calcification develops in atherosclerotic lesions,31 and increasing morphological and molecular evidence suggests that atherosclerotic calcification shares similarities with bone formation. Several proteins involved in osteogenesis have been identified in human atherosclerotic lesions such as OC, ON, OPN, BMP-2, BMP-4, and MGP and are associated with calcification.32 OC is an osteoblast-specific protein that can also be expressed by macrophages, whereas ON is a noncollagenous protein that accumulates in the ECM of bone tissue. BMP-2 and 4 are osteogenic factors that trigger osteoblast differentiation. OPG, a member of the tumor necrosis factor (TNF)- receptor superfamily, is expressed on osteoblast-like cells and inhibits their differentiation.

    In our study, we observed the expression of all of these bone markers to various degrees in areas of cartilaginous metaplasia, which suggests that the cartilaginous tissue found in these lesions may be a precursor to calcification. It is important to note that although cartilaginous metaplasia was more extensive in lesions of 1-deficient mice relative to controls, there was no gross difference in the expression pattern of cartilage and bone markers between the two groups. The role of 1?1 integrin in cartilaginous metaplasia is unknown; however, its occurrence in atherosclerotic plaques may be an indirect effect of 1-deficiency. It has been reported that two of the matrix components known to be important in bone formation, fibronectin and collagen I, are also important in promoting mineralization of vascular cells.33 Because collagen type I is abundantly present in atherosclerotic plaques and an increased collagen content was observed in plaques of 1-deficient mice, this may have led to the formation of cartilaginous metaplasia observed in these plaques.

    These results indicate a role of 1?1 integrin in atherosclerosis and emphasizes the importance of integrin-mediated adhesive interactions in this inflammatory disease. Complete deletion of 1?1 integrin not only reduced plaque area, but also altered plaque composition by reducing inflammation and increasing the ECM content, which are crucial features of a stable atherosclerotic plaque. Antibody intervention against 1 integrin, although less effective in attenuating atherosclerosis compared with complete genetic deletion, modulated plaque characteristics in 2 very important respects by decreasing the leukocyte content and increasing collagen content. It appears that 1?1 integrin is involved in the inflammatory process of atherosclerosis by mediating the migration of leukocytes to lesions.

    Acknowledgments

    E.L. is a post-doctoral fellow of the Dr E. Dekker program of the Dutch Heart Foundation (2000T41). The authors thank Linda Beckers, Anique Janssen, Ine Middendorp, Mat Rousch, and Marjan Smook for excellent technical assistance.

    References

    Duplaa C, Couffinhal T, Dufourcq P, Llanas B, Moreau C, Bonnet J. The integrin very late antigen-4 is expressed in human smooth muscle cell. Involvement of alpha 4 and vascular cell adhesion molecule-1 during smooth muscle cell differentiation. Circ Res. 1997; 80: 159–169.

    Shih PT, Brennan ML, Vora DK, Territo MC, Strahl D, Elices MJ, Lusis AJ, Berliner JA. Blocking very late antigen-4 integrin decreases leukocyte entry and fatty streak formation in mice fed an atherogenic diet. Circ Res. 1999; 84: 345–351.

    Barringhaus KG, Phillips JW, Thatte JS, Sanders JM, Czarnik AC, Bennett DK, Ley KF, Sarembock IJ. alpha(4)beta(1) integrin (VLA-4) blockade attenuates both early and late leukocyte recruitment and neointimal growth following carotid injury in apolipoprotein E (–/–) mice. J Vasc Res. 2004; 41: 252–260.

    Sajid M, Stouffer GA. The role of alpha(v)beta3 integrins in vascular healing. Thromb Haemost. 2002; 87: 187–193.

    Antonov AS, Kolodgie FD, Munn DH, Gerrity RG. Regulation of macrophage foam cell formation by alphaVbeta3 integrin: potential role in human atherosclerosis. Am J Pathol. 2004; 165: 247–258.

    He L, Pappan LK, Grenache DG, Li Z, Tollefsen DM, Santoro SA, Zutter MM. The contributions of the alpha 2 beta 1 integrin to vascular thrombosis in vivo. Blood. 2003; 102: 3652–3657.

    de Fougerolles AR, Koteliansky VE. Regulation of monocyte gene expression by the extracellular matrix and its functional implications. Immunol Rev. 2002; 186: 208–220.

    Stemme S, Holm J, Hansson GK. T lymphocytes in human atherosclerotic plaques are memory cells expressing CD45RO and the integrin VLA-1. Arterioscler Thromb. 1992; 12: 206–211.

    de Fougerolles AR, Sprague AG, Nickerson-Nutter CL, Chi-Rosso G, Rennert PD, Gardner H, Gotwals PJ, Lobb RR, Koteliansky VE. Regulation of inflammation by collagen-binding integrins alpha1beta1 and alpha2beta1 in models of hypersensitivity and arthritis. J Clin Invest. 2000; 105: 721–729.

    Sampson NS, Ryan ST, Enke DA, Cosgrove D, Koteliansky V, Gotwals P. Global gene expression analysis reveals a role for the alpha 1 integrin in renal pathogenesis. J Biol Chem. 2001; 276: 34182–34188.

    Krieglstein CF, Cerwinka WH, Sprague AG, Laroux FS, Grisham MB, Koteliansky VE, Senninger N, Granger DN, de Fougerolles AR. Collagen-binding integrin alpha1beta1 regulates intestinal inflammation in experimental colitis. J Clin Invest. 2002; 110: 1773–1782.

    Belkin VM, Belkin AM, Koteliansky VE. Human smooth muscle VLA-1 integrin: purification, substrate specificity, localization in aorta, and expression during development. J Cell Biol. 1990; 111: 2159–2170.

    Gardner H, Kreidberg J, Koteliansky V, Jaenisch R. Deletion of integrin alpha 1 by homologous recombination permits normal murine development but gives rise to a specific deficit in cell adhesion. Dev Biol. 1996; 175: 301–313.

    Pozzi A, Wary KK, Giancotti FG, Gardner HA. Integrin alpha1beta1 mediates a unique collagen-dependent proliferation pathway in vivo. J Cell Biol. 1998; 142: 587–594.

    Langholz O, Rockel D, Mauch C, Kozlowska E, Bank I, Krieg T, Eckes B. Collagen and collagenase gene expression in three-dimensional collagen lattices are differentially regulated by alpha 1 beta 1 and alpha 2 beta 1 integrins. J Cell Biol. 1995; 131: 1903–1915.

    Gardner H, Broberg A, Pozzi A, Laato M, Heino J. Absence of integrin alpha1beta1 in the mouse causes loss of feedback regulation of collagen synthesis in normal and wounded dermis. J Cell Sci. 1999; 112: 263–272.

    Lutgens E, Gijbels M, Smook M, Heeringa P, Gotwals P, Koteliansky VE, Daemen MJ. Transforming growth factor-beta mediates balance between inflammation and fibrosis during plaque progression. Arterioscler Thromb Vasc Biol. 2002; 22: 975–982.

    Havenith MG, Cleutjens JP, Beek C, vd Linden E, De Goeij AF, Bosman FT. Human specific anti-type IV collagen monoclonal antibodies, characterization and immunohistochemical application. Histochemistry. 1987; 87: 123–128.

    van Zandvoort M, Engels W, Douma K, Beckers L, Oude Egbrink M, Daemen M, Slaaf DW. Two-photon microscopy for imaging of the (atherosclerotic) vascular wall: a proof of concept study. J Vasc Res. 2004; 41: 54–63.

    Lutgens E, Gorelik L, Daemen MJ, de Muinck ED, Grewal IS, Koteliansky VE, Flavell RA. Requirement for CD154 in the progression of atherosclerosis. Nat Med. 1999; 5: 1313–1316.

    Lutgens E, Cleutjens KB, Heeneman S, Koteliansky VE, Burkly LC, Daemen MJ. Both early and delayed anti-CD40L antibody treatment induces a stable plaque phenotype. Proc Natl Acad Sci U S A. 2000; 97: 7464–7469.

    Sukhova GK, Zhang Y, Pan JH, Wada Y, Yamamoto T, Naito M, Kodama T, Tsimikas S, Witztum JL, Lu ML, Sakara Y, Chin MT, Libby P, Shi GP. Deficiency of cathepsin S reduces atherosclerosis in LDL receptor-deficient mice. J Clin Invest. 2003; 111: 897–906.

    Hemler ME. VLA proteins in the integrin family: structures, functions, and their role on leukocytes. Annu Rev Immunol. 1990; 8: 365–400.

    Rubio MA, Sotillos M, Jochems G, Alvarez V, Corbi AL. Monocyte activation: rapid induction of alpha 1/beta 1 (VLA-1) integrin expression by lipopolysaccharide and interferon-gamma. Eur J Immunol. 1995; 25: 2701–2705.

    Andreasen SO, Thomsen AR, Koteliansky VE, Novobrantseva TI, Sprague AG, de Fougerolles AR, Christensen JP. Expression and functional importance of collagen-binding integrins, alpha1beta1 and alpha2beta1, on virus-activated T cells. J Immunol. 2003; 171: 2804–2811.

    Newby AC. An overview of the vascular response to injury: a tribute to the late Russell Ross. Toxicol Lett. 2000; 112–113:519–529.

    Ray SJ, Franki SN, Pierce RH, Dimitrova S, Koteliansky V, Sprague AG, Doherty PC, de Fougerolles AR, Topham DJ. The collagen binding alpha1beta1 integrin VLA-1 regulates CD8 T cell-mediated immune protection against heterologous influenza infection. Immunity. 2004; 20: 167–179.

    Ianaro A, Cicala C, Calignano A, Koteliansky V, Gotwals P, Bucci M, Gerli R, Santucci L, Fiorucci S, Cirino G. Anti-very late antigen-1 monoclonal antibody modulates the development of secondary lesion and T-cell response in experimental arthritis. Lab Invest. 2000; 80: 73–80.

    Bank I, Koltakov A, Goldstein I, Chess L. Lymphocytes expressing alpha1beta1 integrin (very late antigen-1) in peripheral blood of patients with arthritis are a subset of CD45RO(+) T-cells primed for rapid adhesion to collagen IV. Clin Immunol. 2002; 105: 247–258.

    Riikonen T, Westermarck J, Koivisto L, Broberg A, Kahari VM, Heino J. Integrin alpha 2 beta 1 is a positive regulator of collagenase (MMP-1) and collagen alpha 1(I) gene expression. J Biol Chem. 1995; 270: 13548–13552.

    Qiao JH, Mertens RB, Fishbein MC, Geller SA. Cartilaginous metaplasia in calcified diabetic peripheral vascular disease: morphologic evidence of enchondral ossification. Hum Pathol. 2003; 34: 402–407.

    Dhore CR, Cleutjens JP, Lutgens E, Cleutjens KB, Geusens PP, Kitslaar PJ, Tordoir JH, Spronk HM, Vermeer C, Daemen MJ. Differential expression of bone matrix regulatory proteins in human atherosclerotic plaques. Arterioscler Thromb Vasc Biol. 2001; 21: 1998–2003.

    Watson KE, Parhami F, Shin V, Demer LL. Fibronectin and collagen I matrixes promote calcification of vascular cells in vitro, whereas collagen IV matrix is inhibitory. Arterioscler Thromb Vasc Biol. 1998; 18: 1964–1971.(Kitty Schapira; Esther Lu)