Elsevier

Atherosclerosis

Volume 167, Issue 1, March 2003, Pages 25-32
Atherosclerosis

CCR5 deficiency is not protective in the early stages of atherogenesis in apoE knockout mice

https://doi.org/10.1016/S0021-9150(02)00382-9Get rights and content

Abstract

The accumulation of macrophages and T lymphocytes in vessel walls is a hallmark of atherogenesis. It has recently been demonstrated in mouse models of atherosclerosis that full disease potential is dependent on several regulators of leukocyte trafficking, including the chemokine monocyte chemotactic protein 1 (MCP-1) and the chemokine receptors CCR2 and CXCR2. A possible role for the chemokine receptor CCR5 in atherogenesis has been suggested by CCR5 expression on macrophages, T cells, coronary endothelial cells and aortic smooth muscle cells and by the presence of CCR5 ligands in atherosclerotic plaques. Moreover, individuals who are naturally deficient in CCR5 were reported to be at reduced risk for severe coronary artery disease (CAD) and early myocardial infarction (MI). To investigate whether CCR5 is pro-atherogenic in mice, we generated CCR5-deficient mice and crossed them with atherosclerosis-prone apoE-deficient mice. Although CCR5-deficient mice exhibit defects in induced macrophage trafficking, mean atherosclerotic lesion area did not differ significantly between apoE-deficient mice and apoE/CCR5-deficient mice after 16 weeks on a diet of normal chow. Ribonuclease protection assays (RPA) on RNA isolated from plaques from both apoE-deficient and apoE/CCR5-deficient animals showed strong signals for the macrophage marker F4/80 but no evidence for expression of prominent markers of T and B lymphocytes. These results indicate that the early stages of plaque formation in this model of lipid-mediated atherogenesis do not depend on CCR5.

Introduction

Atherosclerosis is a progressive vascular disease characterized by the early and persistent presence within arterial walls of macrophages, T lymphocytes and vascular dendritic cells [1], [2], [3], [4]. The increasing awareness that inflammatory mechanisms underlie atherogenesis has recently led to novel experimental strategies in mouse models to inhibit plaque initiation and progression. Interfering with macrophage and T cell adhesion to the endothelium resulted in large reductions in atherosclerotic fatty streak formation, an early event in plaque development [5], [6]. Also, atherosclerosis-prone mice with defective leukocyte trafficking because of disruptions in the genes for the chemokine monocyte chemotactic protein 1 (MCP-1) or the chemokine receptors CCR2 and CXCR2 have plaques that are smaller and less mature [7], [8], [9], [10]. CCR5 is another chemokine receptor that could potentially be involved in plaque development. CCR5 is expressed on macrophages, T cells, aortic smooth muscle cells and coronary endothelial cells [11], [12], [13], [14] and CCR5 ligands have been detected in plaques [15]. Furthermore, recent genetic screening studies showed that natural deficiency in CCR5 protects individuals from early myocardial infarction (MI) [16] and severe coronary artery disease (CAD) [17]. In the present report, we disrupted the mouse CCR5 gene to investigate whether the absence of CCR5 would have a measurable effect on early lesion formation in the apoE-deficient mouse model of hypercholesterolemia and spontaneous atherosclerotic plaque development [18].

Section snippets

Targeted deletion of the mouse CCR5 gene

The mouse CCR5 gene was isolated from a strain 129/Ola genomic DNA library by cross-hybridization to a radiolabeled human CCR2 coding region DNA fragment [19]. The entire CCR5 coding region, which occurs on a single exon, and flanking regions, were sequenced on both strands. The sequence has been deposited in GenBank under the accession number U68565. As described in Fig. 1, the entire CCR5 coding region was deleted from one chromosome and replaced with an expression cassette for neomycin

Generation of CCR5 knockout mice

As described in Fig. 1, embryonic stem (ES) cell clones harboring one disrupted CCR5 allele were injected into day 3.5 blastocysts from C57BL/6 females. Males with a high degree of chimerism were mated with C57BL/6 females to obtain germline transmission of the targeted CCR5 allele (Fig. 1B). Mating of male and female heterozygotes produced F2 CCR5−/− offspring at the expected Mendelian frequency. CCR5−/− mice breed well, regularly producing normal-size litters. As observed with CCR2−/− mice

Discussion

Previous studies have demonstrated that disruption of macrophage trafficking significantly retards lesion formation in atherosclerosis-prone mice [7], [8], [9]. Although CCR5-deficient mice exhibit defects in macrophage migration and function (Fig. 3 and [23], [24], [25], [26]), these defects do not lead to reductions in plaque area in apoE-deficient mice when measured at 16 weeks (Fig. 4) or to measurable differences in accumulation of RNA for the prominent macrophage marker F4/80. Others have

Acknowledgements

This work was supported by National Institutes of Health grant HL42630 (N. Maeda) and by a Beginning Grant-In-Aid from the American Heart Association, Texas Affiliate (W.A. Kuziel).

References (29)

  • T.J. Reape et al.

    Chemokines and atherosclerosis

    Atherosclerosis

    (1999)
  • Q. Nie et al.

    Inhibition of mononuclear cell recruitment in aortic intima by treatment with anti-ICAM-1 and anti-LFA-1 monoclonal antibodies in hypercholesterolemic rats: implications of the ICAM-1 and LFA-1 pathway in atherogenesis

    Lab. Invest.

    (1997)
  • M.F. Nageh et al.

    Deficiency of inflammatory cell adhesion molecules protects against atherosclerosis in mice

    Arterioscler. Thromb. Vasc. Biol.

    (1997)
  • L. Boring et al.

    Decreased lesion formation in CCR2−/− mice reveals a role for chemokines in the initiation of atherosclerosis

    Nature

    (1998)
  • Cited by (0)

    View full text