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8期 > 正文
编号:11257671
Conditional Knockout of Macrophage PPARIncreases Atherosclerosis in C57BL/6 and Low-Density Lipoprotein Receptor–Deficient Mice
     From the Departments of Medicine (V.R.B., P.G.Y., S.V.R., S.F., M.F.L.), Pharmacology (M.F.L.), Pathology (S.F.), Nephrology (V.K., M.D.B.), and Molecular Physiology & Biophysics (M.A.M.), Vanderbilt University Medical Center, Nashville, Tenn.

    Correspondence to Vladimir Babaev, Sergio Fazio, or MacRae F. Linton, Department of Cardiovascular Medicine, Vanderbilt University School of Medicine, 312 PRB, Nashville, TN 37232-6300. E-mail vladimir.babaev@vanderbilt.edu

    Abstract

    Objective— Peroxisome proliferator-activated receptor gamma (PPAR) is highly expressed in macrophage-derived foam cells of atherosclerotic lesions, and its expression may have a dramatic impact on atherosclerosis.

    Methods and Results— To investigate the contribution of macrophage PPAR expression on atherogenesis in vivo, we generated macrophage-specific PPAR knockout (MacPPARKO) mice. C57BL/6 and low-density lipoprotein (LDL) receptor–deficient (LDLR–/–) mice were reconstituted with MacPPARKO or wild-type marrow and challenged with an atherogenic diet. No differences were found in serum lipids between recipients reconstituted with MacPPARKO and wild-type marrow. In contrast, both C57BL/6 and LDLR–/– mice transplanted with MacPPARKO marrow had significantly larger atherosclerotic lesions than control recipients. In addition, MacPPARKOLDLR–/– mice had higher numbers of macrophages in atherosclerotic lesions compared with controls. Peritoneal macrophages isolated from the MacPPARKO mice had decreased uptake of oxidized but not acetylated LDL and showed no changes in either cholesterol efflux or inflammatory cytokine expression. Macrophages from MacPPARKO mice had increased levels of migration and CC chemokine receptor 2 (CCR2) expression compared with wild-type macrophages.

    Conclusion— Thus, macrophage PPAR deficiency increases atherosclerosis under conditions of mild and severe hypercholesterolemia, indicating an antiatherogenic role for PPAR, which may be caused, at least in part, by modulation of CCR2 expression and monocyte recruitment.

    To investigate the contribution of macrophage PPAR on atherogenesis, we generated macrophage-specific PPAR knockout (MacPPARKO) mice. C57BL/6 and low-density lipoprotein (LDL) receptor–deficient mice were reconstituted with MacPPARKO marrow. These mice had significantly larger atherosclerotic lesions than control recipients. MacPPARKO macrophages had decreased uptake of oxidized LDL and increased CCR2 expression levels.

    Key Words: ABCA1 ? atherosclerosis ? CCR2 expression ? cholesterol efflux ? macrophages ? scavenger receptor CD36

    Introduction

    Peroxisome proliferator-activated receptor gamma (PPAR) is a nuclear transcription factor that regulates a large number of genes important in lipid metabolism and inflammation.1 The receptor is highly expressed in macrophages and macrophage-derived foam cells of atherosclerotic lesions,2–4 and its expression may critically affect macrophage functions that impact atherosclerosis, including activation, cytokine production, recruitment, and transformation into foam cells.

    Several studies have shown that the administration of PPAR agonists inhibits the development of atherosclerosis in low-density lipoprotein (LDL) receptor–deficient (LDLR–/–)5,6 and apolipoprotein E-deficient (apoE–/–) mice.7 Consistent with this, mice transplanted with bone marrow from a PPAR–/– chimera mouse exhibit a significant increase in atherosclerosis.8 These data all support an antiatherogenic role for macrophage PPAR in atherosclerotic lesion development.

    It has been assumed that the antiatherogenic effects of macrophage PPAR expression may derive from activation of genes responsible for cholesterol efflux, thus shifting the balance from lipid loading to lipid efflux.8 Recent studies, however, have not confirmed the role of PPAR ligands in cholesterol efflux by macrophages.9,10 PPAR may also exert antiinflammatory effects in macrophages directly,11 or through LXR12 by negatively interfering with the AP-1, NFB, and STAT signaling pathways,13 or by reducing tumor necrosis factor-, IL-1, and IL-6 secretion.14 However, the loss of PPAR expression in macrophages derived from embryonic stem cells does not appear to alter basal or stimulated levels of cytokine secretion.15,16

    In vitro studies have demonstrated that PPAR expression by human and murine monocytes directly inhibits CC chemokine receptor 2 (CCR2) expression and suppresses MCP-1–mediated chemotaxis.17 In addition, pretreatment of monocytes with PPAR agonists reduced their adhesion to vascular endothelium18 and their transendothelial migration.6 Based on these data, it appears that at least some of the antiatherogenic effects of macrophage PPAR may be attributed to inhibition of macrophage recruitment and migration; however, the physiological relevance of the inhibitory processes in vivo remains unclear.

    Targeted disruption of the PPAR gene in mice causes early embryonic lethality19–21 and thus presents an obstacle to a systematic study of the gene’s role in atherogenesis. Therefore, we generated mice with a macrophage-specific PPAR knockout (MacPPARKO) using the Cre-loxP recombination system approach under the control of the murine M lysozyme promoter. C57BL6 and LDLR–/– mice were lethally irradiated, reconstituted with marrow from MacPPARKO or wild-type mice, and challenged with atherogenic diets. Mice reconstituted with MacPPARKO marrow exhibited significantly larger atherosclerotic lesions with increased numbers of macrophages. Macrophages from these mice also expressed higher levels of CCR2, suggesting that an increase in monocyte recruitment may be responsible for the accelerated atherosclerosis seen in these mice.

    Methods

    Animal Procedures

    Mice with the "floxed" PPAR (PPARfl/fl) gene,22 a mouse Cre line under the control of the murine M lysozyme promoter,23 and transgenic ROSA26R24 were at the sixth or more backcross into C57BL/6 background. ROSA26 mice,25 recipient C57BL/6, and LDLR–/– mice on C57BL/6 background were purchased from Jackson Laboratories Inc (Bar Harbor, Me). All mice were maintained in micro-isolator cages on a rodent chow diet containing 4.5% fat (PMI number 5010) and autoclaved acidified (pH 2.8) water. A butterfat diet contained 19.5% butterfat, 1.25% cholesterol, and 0.5% cholic acid (ICN, Avrora, Ohio). A Western diet consisted of 21% fat and 0.15% cholesterol (Teklad). Animal care and experimental procedures were performed according to the regulations of Vanderbilt University’s Animal Care Committee.

    Bone Marrow Transplantation

    Recipient mice were lethally irradiated (9 Gy) from a cesium gamma source, and 5x106 bone marrow cells were injected as described.26

    Flow Cytometry

    To examine lacZ gene expression, peritoneal macrophages were treated with ?-galactosidase-fluorescein di-?-D-galactopyranoside (Molecular Probes, Eugene, Ore) as described.26 To detect CCR2 expression, macrophages were incubated with nonspecific mouse IgG (Sigma), then with allophycocyanin (activated protein C)-conjugated mouse anti-human CCR2 (R&D Systems Inc, Minneapolis, Minn) and analyzed by flow cytometry (Becton Dickinson).

    Serum Lipids and Lipoprotein Profiles

    Mice were fasted for 4 hours, and the serum total cholesterol and triglycerides were determined as described.27 Fast performance liquid chromatography (fast protein liquid) was completed using a Superose 6 column (Pharmacia) on a high-performance liquid chromatography system model 600 (Waters).

    Analysis of Aortic Lesion

    The aorta was flushed through the left ventricle and dissected for en face preparation and image analysis. Cryosections of the proximal aorta were prepared and analyzed using an Imaging system KS 300 (Kontron Electronik GmbH) as described.28

    Modified LDL Uptake

    Thioglycolate-elicited peritoneal macrophages were cultured in DMEM with 10% fetal bovine serum for 2 days. Then cells were incubated with DiI-labeled human acetylated LDL (AcLDL) or oxidized LDL (Intracel Corp, Rockville, Md) at 37°C for 4 hours and analyzed under a fluorescent microscope or by fluorescence-activated cell sorter (FACS) flow cytometry.16

    Cholesterol Loading and Efflux

    Macrophages were cultured in DMEM containing 1% fetal bovine serum, 3 μCi/mL of 3H-cholesterol, and 70 μg/mL of human AcLDL for 48 hours. Cholesterol pools were equilibrated overnight in 0.1% bovine serum albumin–DMEM. Then cells were incubated with human apolipoprotein AI (20 μg/mL) or high-density lipoprotein (50 μg/mL) for up to 7 hours. For each time point, 3H-cholesterol was measured in aliquots of media. The cell lipids were extracted and used for measurement of 3H cholesterol.

    Ligand Treatment and Real-Time Polymerase Chain Reaction

    Macrophages were cultured in DMEM media supplemented with 5% lipoprotein-deficient fetal bovine serum (Intracel) with or without 10 μmol/L per mL rosiglitazone for 24 hours. Total RNA was isolated from peritoneal macrophages using the Trizol reagent (Invitrogen, Carlsbad, Calif) purified by RNA Easy kit (Qiagen). Relative quantitation of the target mRNA were performed on the ABI Prism 7000 Sequence Detection System (Applied Biosystems) and normalized to ?-actin or 18S ribosomal RNA. Probes for PPAR, CD36, apoE, ABCA1, MCP-1, Gro1, and CCR2 were provided by Applied Biosystems.

    Migration Assay

    In vitro assays were performed in a 96-well modified Boyden chamber with a 3-μm filter pore size (Millipore). Cell solution (100 μL) was added to each well in the top filter plate portion of the assembly, and MCP-1 (0.1 μg/μL) or media was added to the bottom feeder wells. After 1 hour, the upper portion was removed and cell numbers were counted.

    Statistical Analysis

    The statistical differences in mean serum lipids and aortic lesion areas between the groups were determined using the SigmaStat V.2 software (SPSS Inc, Chicago, Ill) by Student t test and the Mann–Whitney rank sum test, respectively.

    Results

    Macrophage-Specific PPAR Knockout

    Using the Cre-loxP recombination system, we generated MacPPARKO mice on the C57BL/6 background. These mice were viable and fertile with no notable differences in body weight or plasma lipid levels when compared with PPARfl/fl littermates. The level of PPAR RNA was dramatically decreased in peritoneal macrophages from MacPPARKOmice compared with that of wild-type macrophages as analyzed by RT-PCR (Figure 1A) or quantitative real-time PCR normalized to 18S ribosomal RNA (Figure 1B). Deficiency in PPAR RNA was specific for macrophages and not observed in kidney, adipose, or liver tissue (data not shown).

    Figure 1. Macrophage-specific inactivation of PPAR analyzed by reverse-transcriptase polymerase chain reaction (PCR) (A), real-time PCR (B), and LacZ expression (C). Thioglycolate-elicited peritoneal macrophages were used for total RNA extraction and amplification with the primer sets specific for ?-actin and PPAR, resulting in 500-bp and 293-bp bands, respectively (lane 7, RNA only; lane 8, no template; lane 9, genomic DNA). PPAR expression in wild-type () and MacPPARKO () macrophage analyzed by real-time quantitative PCR. LacZ expression is detected by flow cytometry in peritoneal macrophages isolated from PPARfl/fl (black line), ROSA26 constituently expressing lacZ gene (blue line) and MacPPARKO/ROSA26R (red line) mice.

    To monitor the effectiveness of Cre recombinase, we crossed MacPPARKO mice with transgenic ROSA26R mice. This strain has a "floxed" STOP codon at the 5' end of the lacZ gene driven by the ?-actin promoter.24 In MacPPARKO/ROSA26R mice, Cre recombinase excised the STOP codon, releasing lacZ expression in the majority (93% to 96%) of peritoneal macrophages. The intensity of lacZ expression in these cells, as measured by a FACS assay, was significantly higher when compared with macrophages isolated from PPARfl/fl and ROSA26 mice ubiquitously expressing the lacZ gene (Figure 1C).

    Role of Macrophage PPAR in Atherosclerosis

    To examine the impact of MacPPARKO on atherosclerosis, 7-week-old female C57BL/6 and 8-week-old female LDLR–/– mice were lethally irradiated and transplanted with marrow from female MacPPARKO (n=16 for each experimental group) or PPARfl/fl mice (n=15 for each control group). Six or 4 weeks after transplantation, recipient mice were challenged with the butterfat or the Western diets for 16 or 8 weeks, respectively.

    Serum lipid levels did not differ significantly between the control and experimental groups of mice on either the chow or the atherogenic diets with exception that the triglycerides were higher in MacPPARKOC57BL/6 mice after 12 weeks of the butterfat diet (Tables I and II, available online at http://atvb.ahajournals.org). Similarly, serum lipoprotein profiles did not differ significantly between experimental and control groups of the recipient mice (Figure Ia and Ib, available online at http://atvb.ahajournals.org). In contrast, the extent of atherosclerotic lesions in the proximal aortas of C57BL/6 and LDLR–/– recipients reconstituted with MacPPARKO macrophages was significantly greater (48% and 84%) compared with PPARfl/flC57BL/6 (37 715±3010 versus 25 512±2660 μm2; P=0.005) and PPARfl/flLDLR–/– mice (125 120±9550 versus 67 948±6205 μm2; P<0.001), respectively (Figure 2A and 2B). MacPPARKOLDLR–/– recipients had larger (46%) lesion area in the distal aortas analyzed en face compared with PPARfl/flLDLR–/– mice (0.19± 0.01 versus 0.13±0.02%; P=0.031; Figure 2C). Thus, macrophage PPAR expression plays a protective role in atherosclerotic lesion formation.

    Figure 2. Quantification of atherosclerotic lesion area. The extent of atherosclerotic lesions was analyzed in the proximal aorta of recipient C57BL6 (A), LDLR–/– (B) transplanted with PPARfllfl (?), or MacPPARKO () marrow after 16 or 8 weeks on their atherogenic diets, respectively. The extent of atherosclerosis in "pinned out" aortas en face of LDLR–/– (C) mice.

    Modified LDL Uptake and Cholesterol Efflux by Macrophages From MacPPARKO Mice

    In an effort to better understand the molecular basis of these effects, we first analyzed the impact of the macrophage PPAR gene deletion on uptake of modified LDL. Peritoneal macrophages were isolated and incubated with DiI-labeled oxidized and acetylated LDL. Microscopic analysis showed that PPAR–/– macrophages accumulated significantly less oxidized LDL than wild-type macrophages (Figure 3A). FACS analysis demonstrated that PPAR–/– macrophages had a reduced (43% to 57%) levels of oxidized LDL uptake but not AcLDL uptake, compared with wild-type macrophages (Figure 3B and 3C). To test cholesterol efflux, macrophages were loaded with human AcLDL cholesterol and incubated with high-density lipoprotein or apolipoprotein AI. No differences were noted in either high-density lipoprotein- or apolipoprotein AI-mediated cholesterol efflux from PPAR–/– and wild-type macrophages (Figure 4A and 4B). However, real-time PCR analysis revealed that wild-type macrophages treated with a PPAR ligand, rosiglitazone, expressed significantly higher levels of CD36 (269%) and ABCA1 (125%) but not apoE RNA compared with control nontreated cells (Figure 4C to 4F). In PPAR–/– macrophages, these stimulation effects for the CD36 and ABCA1 genes were lost.

    Figure 3. A, Visualization (A) and quantitation of DiI-oxidized LDL (B) or DiI-AcLDL (C) upatke by macrophages from PPARfl/fl () or MacPPARKO () mice. Peritoneal macrophages were incubated with (15 μg/mL) or increasing concentration of human DiI-oxidized LDL or DiI-AcLDL for 4 hours and examined by flow cytometry. The data are presented in relative units as an average of triplicate repeats (*P<0.05).

    Figure 4. Apolipoprotein AI (A) and high-density lipoprotein-mediated (B) cholesterol efflux, and CD36, ABCA1, or apolipoprotein E gene expression (C) by macrophages isolated from PPARfl/fl (black fill) or MacPPARKO (no fill) mice. Peritoneal macrophages were incubated for 24 hour in RPMI 1640 supplemented with 1% fetal bovine serum and44-cholesterol and then exposed with human apolipoprotein AI or high-density lipoprotein for up to 7.5 hours. Data are presented as a percentage (mean±SEM) of the total radioactivity in medium. For PPAR ligand experiments, macrophages were treated with or without rosiglitazone 10 μg/mL for 24 hours and the gene expression was measured by real-time quantitative PCR. The experiment was repeated twice with the same number of mice (n=4 for each group).

    Inflammatory Cytokine Gene Profiles and CCR2 Expression by Macrophages From MacPPARKO Mice

    To examine the antiinflammatory effects of macrophage PPAR, mRNA was isolated from lipopolysaccharide-activated macrophages and analyzed using an inflammatory response cytokines gene array kit. For both type of macrophages, expression levels for the majority of cytokines (IL1a, IL1b, IL6, IL12a, IL18, transforming growth factor-?, and tumor necrosis factor-) were not significantly different, with the exception of the Gro1 oncogene (Table III, available online at http://atvb.ahajournals.org). As confirmed by real-time PCR, PPAR–/– macrophages stimulated by lipopolysaccharide had increased levels of the Gro1 (1.7 fold) but not MCP-1 expression compared with wild-type macrophages (Figure IVa and IVb, available online at http://atvb.ahajournals.org).

    Finally, the impact of PPAR expression by macrophages on the CCR2/MCP-1 pathway was analyzed by real-time PCR. The level of mRNA CCR2 expression was significantly increased (1.7-fold) in PPAR–/– macrophages compared with wild-type macrophages (Figure 5A). In addition, PPAR–/– macrophages had increased levels CCR2 protein expression (107±12 versus 75±5; P<0.05) as detected by FACS (Figure 5B).

    Figure 5. CCR-2 gene (A) and protein (B) expression, and migration assay (C) using peritoneal macrophages isolated from PPARfl/fl () of or MacPPARKO () mice. Blood mononuclear cells were collected using Histopaque-1077 (Sigma) and seeded on a plastic for 30 minutes. After washing with phosphate-buffered saline, cells were used for RNA isolation to test CCR2 gene expression by real-time quantitative PCR (A) or stained with activated protein C-labeled CCR2 antibody and the fluorescent intensity was analyzed by flow cytometry (B). C, Thioglycolate-elicited peritoneal macrophages were analyzed by migration assay in the presence or absence of MCP-1. The experiments were performed twice with the same number of mice (n=4 per group).

    Given the pivotal role of the CCR2 pathway in monocyte recruitment, we performed a series of in vitro experiments to determine the ability of peritoneal macrophages to migrate. Macrophages from MacPPARKO mice migrated significantly faster in both nonstimulated and MCP-1–directed tests compared with macrophages from PPARfl/fl mice (Figure 5C). In addition, we stained sections from the proximal aorta of LDLR–/– recipients using a macrophage-specific antibody and DAPI (Figure 6). MacPPARKOLDLR–/– mice had a significantly increased (36%) number of macrophages per a section (195±14) compared with control PPARfl/flLDLR–/– mice (143±6; P<0.004).

    Figure 6. Detection of macrophages and DAPI-stained nuclei in the proximal aorta of PPARfllflLDLR–/– and MacPPARKOLDLR–/– mice. Sections from the proximal aorta were stained with monoclonal antibody to macrophages, MOMA-2 (upper), DAPI to detect nuclei (middle), and after merge of the images (lower). Note that macrophage-rich area of MacPPARKOLDLR–/– mice contains more nuclei. (10x).

    Discussion

    To examine the role of macrophage PPAR expression in atherosclerotic lesion formation, we generated mice with MacPPARKO using the Cre-LoxP recombination system under control of the murine M lysozyme promoter. Then, C57BL6 and LDLR–/– mice were reconstituted with MacPPARKO or wild-type macrophages and fed atherogenic diets. The serum lipid levels and lipoprotein profiles were similar between control and experimental groups of recipients. In contrast, mice reconstituted with MacPPARKO macrophages exhibited significantly larger atherosclerotic lesions compared with controls. Thus, local expression of macrophage PPAR in artery walls plays a protective antiatherogenic role under conditions of mild and severe hypercholesterolemia.

    In these studies, we have used a novel macrophage-specific knockout approach to examine the role of macrophage PPAR in atherogenesis. To our knowledge, this is the first report of the Lys-M-Cre approach to examine the effects of macrophage-specific gene expression in atherosclerosis in vivo. Although Akiyama et al10 previously developed a macrophage-specific knockout of PPAR driven by the MX1 promoter, they did not report the impact of PPAR deficiency on atherosclerosis. Furthermore, our approach has an advantage over the MX1 promoter approach in that the LysM-Cre mice do not need induction to develop PPAR deficiency. Chawla et al8 reported similar antiatherogenic effects of macrophage PPAR expression in LDLR–/– mice reconstituted with PPAR–/– marrow from a chimeric mouse. Consistent with the results of genetic deletion of macrophage PPAR, administration of PPAR agonists to LDLR–/– mice5,6 and apoE–/– mice,7 and in balloon injury experiments,29 have also demonstrated an antiatherogenic role for PPAR.

    To investigate possible mechanisms by which macrophage PPAR delivers its antiatherogenic effects, we first focused on the ability of macrophages to take-up modified lipoproteins. We found that MacPPARKO macrophages have decreased uptake of oxidized but not acetylated LDL and, unlike wild-type macrophages, did not show an increase in CD36 expression in response treatment with PPAR agonists. These findings are consistent with previous ex vivo studies demonstrating that PPAR has a critical role in the basal regulation of the CD36 gene in macrophages.4 Interestingly, Liang et al have recently reported that, in the setting of extreme insulin resistance caused by leptin deficiency found in ob/ob and ob/ob LDLR–/– mice, treatment with a thiazolidinedione results in reduced systemic insulin resistance leading to reduced macrophage CD36 protein, despite an increase in macrophage CD36 gene expression, caused by correction of defective insulin signaling in the macrophage.30 In contrast, in vivo thiazoladinedione treatment of LDLR–/– mice on a Western diet, a model associated with mild insulin resistance, resulted in a 3-fold increase in macrophage CD36 expression.30 Thus, the impact of PPAR agonists on macrophage CD36 protein may vary with the degree of insulin resistance. Given that PPAR agonists have been reported to reduce atherosclerosis in LDLR-deficient mice,5,6 the potentially proatherogenic effects of CD36 upregulation on foam cell formation15,31 are apparently outweighed by other antiatherogenic effects of PPAR agonists in this model.

    Next, we found that PPAR–/– and wild-type macrophages have similar basal levels of cholesterol efflux. However, the agonist treatment increased ABCAI gene expression levels in wild-type but not in PPAR–/– macrophages, consistent with previous studies suggesting that the PPAR-LXR-ABCA1 pathway may be important in modulating the development of atherosclerosis.8,32

    Macrophage PPAR mediates the activation of a large number of genes that are important in inflammation.33 To test how PPAR deficiency affects the macrophage’s ability to produce cytokines in response to lipopolysaccharide, inflammatory cytokine gene expression profiles were compared in macrophages from MacPPARKO and wild-type mice. The majority of cytokines had similar levels of expression for both groups of macrophages, indicating that at least some of the previously described effects of PPAR agonists on cytokine gene expression are independent of PPAR gene expression.15,16 At the same time, MacPPARKOLDLR–/– mice had increased macrophage numbers in atherosclerotic lesions, augmented macrophage CCR2 expression, and migration. These data suggest that PPAR modulates CCR2 expression and may affect monocyte recruitment.

    Monocyte CCR2 expression is increased in hypercholesterolemic patients.34 Native LDL and oxidative stress increase CCR2 gene expression, whereas antioxidants rapidly inhibit it.35 Treatment with oxidized LDL activates PPAR expression and PPAR agonists markedly attenuate CCR2 expression in circulating monocytes.17 Recent in vivo studies have shown that PPAR activators suppress the recruitment of inflammatory cells via a PPAR-dependent mechanism in cases of experimental glomerulonephritis36 and myocardial infarction in rats.37 All these data support the concept that PPAR-modulated CCR2 expression may impact the development of atherosclerosis through an effect on monocyte recruitment.

    Mounting evidence suggests that the MCP-1/CCR2 pathway is important in atherogenesis. Targeted deletion of CCR2 or its ligand MCP-1 significantly decreased macrophage recruitment and atherosclerotic lesion size in apoE–/– mice.38–40 Clinical studies also demonstrated that coronary atherosclerosis is decreased in patients with a polymorphism of the CCR2 gene that reduces its function.41 In addition, PPAR agonists inhibit CCR2 expression in monocytes and atherosclerosis development in rats.42,43 Thus, PPAR-mediated CCR2 expression by macrophages may be an important pathway in atherogenesis and provides a novel therapeutic target for prevention or treatment of atherosclerosis.

    The role of macrophage PPAR in atherosclerosis is clearly complex and likely includes important effects on cholesterol homeostasis and inflammatory pathways, which may vary with lesion stage and metabolic factors such as insulin resistance.30 Our conditional macrophage-specific knockout of PPAR presents a new opportunity to study the role of macrophage PPAR gene expression in atherogenesis in vivo. C57BL6 and LDLR–/– mice reconstituted with PPAR–/– macrophages developed significantly larger atherosclerotic lesions compared with control mice in response to atherogenic diets. In the absence of any notable changes in serum lipids between control and experimental mice, the increase in atherosclerosis suggests that macrophage PPAR is crucial for these antiatherogenic effects. The increase in CCR2 expression by macrophages from MacPPARKO mice suggests a novel role for PPAR in monocyte recruitment and the development of atherosclerosis.

    Acknowledgments

    This work was supported by National Institutes of Health grants HL65405, HL53989, HL 57986, DK59637 (Lipid, Lipoprotein, and Atherosclerosis Core of the Vanderbilt Mouse Metabolic Phenotyping Centers). V.R.B. is supported by an American Heart Association grant (0160160B). The authors thank Lei Ding and Youmin Zhang for excellent technical expertise.

    References

    Duval C, Chinetti G, Trottein F, Fruchart JC, Staels B. The role of PPARs in atherosclerosis. Trends in Mol Med. 2002; 8: 422–430.

    Ricote M, Huang J, Fajas L, Li A, Welch J, Najib J, Witztum Jl, Auwerx J, Palinski W, Glass CK. Expression of the peroxisome proliferator-activated receptor gamma in human atherosclerosis and regulation in macrophages by colony stimulating factors and oxidized low density lipoprotein. Proc Natl Acad Sci U S A. 1998; 95: 7614–7619.

    Marx N, Schonbeck U, Lazar MA, Libby P, Plutzky J. Peroxisome proliferator-activated receptor gamma activators inhibit gene expression and migration in human vascular smooth muscle cells. Circ Res. 1998; 83: 1097–1103.

    Tontonoz P, Nagy L, Alvarez JGA, Thomazy VA, Evans RM. PPAR-Gamma Promotes Monocyte/Macrophage Differentiation and Uptake Of Oxidized LDL. Cell. 1998; 93: 241–252.

    Li AC, Brown KK, Silvestre MJ, Willson TM, Palinski W, Glass CK. Peroxisome proliferator-activated receptor gamma ligands inhibit development of atherosclerosis in LDL receptor-deficient mice. J Clin Invest. 2000; 106: 523–531.

    Collins AR, Meehan WP, Kintscher U, Jackson S, Wakino S, Noh G, Palinski W, Hsueh WA, Law RE. Troglitazone inhibits formation of early atherosclerotic lesions in diabetic and nondiabetic low density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol. 2001; 21: 365–371.

    Chen Z, Ishibashi S, Perrey S, Osuga J, Gotoda T, Kitamine T, Tamura Y, Okazaki H, Yahagi N, Iizuka Y, Shionoiri F, Ohashi K, Harada K, Shimano H, Nagai R, Yamada N. Troglitazone inhibits atherosclerosis in apolipoprotein E-knockout mice: pleiotropic effects on CD36 expression and HDL. Arterioscler Thromb Vasc Biol. 2001; 21: 372–377.

    Chawla A, Boisvert WA, Lee CH, Laffitte BA, Barak Y, Joseph SB, Liao D, Nagy L, Edwards PA, Curtiss LK, Evans RM, Tontonoz P. A PPAR gamma-LXR-ABCA1 pathway in macrophages is involved in cholesterol efflux and atherogenesis. Mol Cell. 2001; 7: 161–171.

    Claudel T, Leibowitz MD, Fievet C, Tailleux A, Wagner B, Repa JJ, Torpier G, Lobaccaro JM, Paterniti JR, Mangelsdorf DJ, Heyman RA, Auwerx J. Reduction of atherosclerosis in apolipoprotein E knockout mice by activation of the retinoid X receptor. Proc Natl Acad Sci U S A. 2001; 98: 2610–2615.

    Akiyama TE, Sakai S, Lambert G, Nicos CJ, Kimihiko N, Matsusue K, Pimprale S, Lee Y-H, Ricote M, Glass C, Brewer B, Gonzalez FJ. Conditional Disruption of the Peroxisome Proliferator-Activated Receptor g Gene in Mice Results in Lovered Expression of ABCA1, ABCG1, and apoE in Macrophages and Reduced Cholesterol Efflux. Mol Cell Biol. 2002; 22: 2607–2619.

    Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK. The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature. 1998; 391: 79–82.

    Joseph SB, Castrillo A, Laffitte BA, Mangelsdorf DJ, Tontonoz P. Reciprocal regulation of inflammation and lipid metabolism by liver X receptors. Nat Med. 2003; 9: 213–219.

    Chinetti G, Fruchart JC, Staels B. Peroxisome proliferator-activated receptors (PPARs): nuclear receptors at the crossroads between lipid metabolism and inflammation. Inflam Res. 2000; 49: 497–505.

    Jiang C, Ting AT, Seed B. PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature. 1998; 391: 82–86.

    Moore KJ, Rosen ED, Fitzgerald ML, Randow F, Andersson LP, Altshuler D, Milstone DS, Mortensen RM, Spiegelman BM, Freeman MW. The role of PPAR-gamma in macrophage differentiation and cholesterol uptake. Nat Med. 2001; 7: 41–47.

    Chawla A, Barak Y, Nagy L, Liao D, Tontonoz P, Evans RM. PPAR-gamma dependent and independent effects on macrophage-gene expression in lipiud metabolism and inflammation. Nat Med. 2001; 7: 48–52.

    Han KH, Chang MK, Boullier A, Green SR, Li A, Glass CK, Quehenberger O. Oxidized LDL reduces monocyte CCR2 expression through pathways involving peroxisome proliferator-activated receptor gamma. J Clin Invest. 2000; 106: 793–802.

    Toriumi Y, Hiraoka M, Watanabe M, Yoshida M. Pioglitazone reduces monocyte adhesion to vascular endothelium under flow by modulating RhoA GTPase and focal adhesion kinase. FEBS Lett. 2003; 553: 419–422.

    Barak Y, Nelson MC, Ong ES, Jones YZ, Ruiz-Lozano P, Chien KR, Koder A, Evans RM. PPAR Is Required for Placental, Cardiac, and Adipose Tissue Development. Molecular Cell. 1999; 4: 585–595.

    Kubota N, Terauchi Y, Miki H, Tamemoto H, Yamauchi T, Komeda K, Satoh S, Nakano R, Ishii C, Sugiyama T. PPAR Mediates High-Fat Diet-Induced Adipocyte Hypertrophy and Insulin Resistance. Mol Cell. 1999; 4: 597–609.

    Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, Spiegelman BM, Mortensen RM. PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro. Mol Cell. 1999; 4: 611–617.

    Jones JR, Shelton KD, Guan Y, Breyer MD, Magnuson MA. Generation and functional confirmation of a conditional null PPARgamma allele in mice. Genesis: J Gen Dev. 2002; 32: 134–137.

    Clausen BE, Burkhardt C, Reith W, Renkawitz R, Forster I. Conditional gene targeting in macrophages and granulocytes using LysMCre mice. Transgenic Res. 1999; 8: 265–277.

    Soriano P. Generalized lacZ expression with the ROSA26 Cre reporter strain. Nat Genet. 1999; 21: 70–71.

    Friedrich G, Soriano P. Promoter traps in embryonic stem cells: a genetic screen to identify and mutate developmental genes in mice. Genes & Development. 1991; 5: 1513–1523.

    Linton MF, Atkinson JB, Fazio S. Prevention of atherosclerosis in apoE deficient mice by bone marrow transplantation. Science. 1995; 267: 1034–1037.

    Fazio S, Babaev VR, Murray AB, Hasty AH, Carter KJ, Gleaves LA, Atkinson JB, Linton MF. Increased atherosclerosis in mice reconstituted with apolipoprotein E null macrophages. Proc Natl Acad Sci U S A. 1997; 94: 4647–4652.

    Babaev VR, Patel MB, Semenkovich CF, Fazio S, Linton MF. Macrophage lipoprotein lipase promotes foam cell formation and atherosclerosis in LDL receptor deficient mice. J Biol Chem. 2000; 275: 26293–26299.

    Law RE, Meehan WP, Xi XP, Graf K, Wuthrich DA, Coats W, Faxon D, Hsueh WA. Troglitazone inhibits vascular smooth muscle cell growth and intimal hyperplasia. J Clin Invest. 1996; 98: 1897–1905.

    Liang C-P, Han S, Okamoto H, Carnemolla R, Tabas I, Accili D, Tall AR. Increased CD36 protein as a response to defective insulin signaling in macrophages. J Clin Invest. 2004; 113: 764–773.

    Chinetti G, Lestavel S, Bocher V, Remaley AT, Neve B, Torra IP, Teissier E, Minnich A, Jaye M, Duverger N, Brewer HB, Fruchart JC, Clavey V, Staels B. PPAR-alpha and PPAR-gamma activators induce cholesterol removal from human macrophage foam cells through stimulation of the ABCA1 pathway. Nat Med. 2001; 7: 53–58.

    Li AC, Binder CJ, Gutierrez A, Brown KK, Plotkin CR, Pattison JW, Valledor AF, Davis RA, Willson TM, Witztum JL, Palinski W, Glass CK. Differential inhibition of macrophage foam-cell formation and atherosclerosis in mice by PPAR{alpha}, {beta}/{delta}, and {gamma}. J Clin Invest. 2004; 114: 1564–1576.

    Welch JS, Ricote M, Akiyama TE, Gonzalez FJ, Glass CK. PPAR{gamma} and PPAR{delta} negatively regulate specific subsets of lipopolysaccharide and IFN-{gamma} target genes in macrophages. Proc Natl Acad Sci U S A. 2003; 100: 6712–6717.

    Han KH, Han KO, Green SR, Quehenberger O. Expression of the monocyte chemoattractant protein-1 receptor CCR2 is increased in hypercholesterolemia. Differential effects of plasma lipoproteins on monocyte function. J Lipid Res. 1999; 40: 1053–1063.

    Saccani A, Saccani S, Orlando S, Sironi M, Bernasconi S, Ghezzi P, Mantovani A, Sica A. Redox regulation of chemokine receptor expression. Proc Natl Acad Sci U S A. 2000; 97: 2761–2766.

    Haraguchi K, Shimura H, Onaya T. Suppression of experimental crescentic glomerulonephritis by peroxisome proliferator-activated receptor (PPAR)gamma activators. Clin Exp Nephrol. 2003; 7: 27–32.

    Ito H, Nakano A, Kinoshita M, Matsumori A. Pioglitazone, a peroxisome proliferator-activated receptor-gamma agonist, attenuates myocardial ischemia/reperfusion injury in a rat model. Lab Invest. 2003; 83: 1715–1721.

    Boring L, Gosling J, Cleary M, Charo IF. Decreased lesion formation in CCR2–/– mice reveals a role for chemokines in the initiation of atherosclerosis. Nature. 1998; 394: 894–897.

    Dawson TC, Kuziel WA, Osahar TA, Maeda N. Absence of CC chemokine receptor-2 reduces atherosclerosis in apolipoprotein E-deficient mice. Atherosclerosis. 1999; 143: 205–211.

    Gosling J, Slaymaker S, Gu L, Tseng S, Zlot CH, Young SG, Rollins BJ, Charo IF. MCP-1 deficiency reduces susceptibility to atherosclerosis in mice that overexpress human apolipoprotein B. J Clin Invest. 1999; 103: 773–778.

    Valdes AM, Wolfe ML, O’Brien EJ, Spurr NK, Gefter W, Rut A, Groot PH, Rader DJ. Val64Ile polymorphism in the C-C chemokine receptor 2 is associated with reduced coronary artery calcification. Arterioscler Thromb Vasc Biol. 2002; 22: 1924–1928.

    Ishibashi M, Egashira K, Hiasa K, Inoue S, Ni W, Zhao Q, Usui M, Kitamoto S, Ichiki T, Takeshita A. Antiinflammatory and antiarteriosclerotic effects of pioglitazone. Hypertension. 2002; 40: 687–693.

    Kataoka C, Egashira K, Ishibashi M, Inoue S, Ni W, Hiasa K, Kitamoto S, Usui M, Takeshita A. Novel anti-inflammatory actions of amlodipine in a rat model of arteriosclerosis induced by long-term inhibition of nitric oxide synthesis. Am J Physiol: Heart Circ Physiol. 2004; 286: H768–H774.

    Kliewer SA, Lenhard JM, Willson TM, Patel I, Morris DC, Lehmann JM. A prostaglandin J2 metabolite binds peroxisome proliferator-activated receptor gamma and promotes adipocyte differentiation. Cell. 1995; 83: 813–819.(Vladimir R. Babaev; Patri)