The Immune Response In

Embed Size (px)

Citation preview

  • 8/12/2019 The Immune Response In

    1/12

    Atherosclerosis is an inflammatory disease characterizedby intense immunological activity, which increasinglythreatens human health worldwide1. Atherosclerosisinvolves the formation in the arteries of lesions thatare characterized by inflammation, lipid accumula-tion, cell death and fibrosis. Over time, these lesions,which are known as atherosclerotic plaques, mature andgain new characteristics. Although clinical complica-tions of atherosclerosis can arise from plaques causingflow-limiting stenoses, the most severe clinical eventsfollow the rupture of a plaque, which exposes the pro-thrombotic material in the plaque to the blood andcauses sudden thrombotic occlusion of the artery atthe site of disruption. In the heart, atherosclerosis canlead to myocardial infarctionand heart failure; whereas inthe arteries that perfuse the brain, it can cause ischaemicstrokeand transient ischaemic attacks. If atherosclero-sis affects other arterial branches, it can result in renalimpairment, hypertension, abdominal aortic aneurysms

    and critical limb ischaemia. As our knowledge of thisdisease increases, we increasingly recognize that there isno simple answer to the question of whether the immuneresponse promotes or retards atherogenesis. Indeed, thetwo arms of the immune response can either promoteor attenuate aspects of atherosclerosis and its complica-tions. This Review summarizes our current understand-ing of the role of adaptive immunity in atherosclerosisand, in particular, weighs the evidence regarding theyin and yang of the immune response at various placesand times in the evolution of this lengthy and complexdisease. We do not discuss the arteriosclerosis of allo-grafted transplants, which is a distinct disease with a

    unique pathogenesis, although it might represent anextreme case of immune-driven arteriopathy.

    Immunological features of atherosclerosis

    In humans, atherosclerotic plaques contain blood-borneinflammatory and immune cells (mainly macrophagesand T cells), as well as vascular endothelial cells, smoothmuscle cells, extracellular matrix, lipids and acellularlipid-rich debris2. These lesions typically present asasymmetrical focal thickenings of the intima, which isthe innermost layer of the artery (FIG. 1). Accumulationof immune cells and lipid droplets in the intima occursduring the f irst stage of plaque formation. Lipid-ladenmacrophages, known as foam cells, outnumber othercells in early plaques (which are known as fatty streaks),but these nascent plaques also contain T cells. Fattystreaks are prevalent in young individuals, never causesymptoms, and can progress into mature atheroscleroticplaques or disappear with time.

    Mature plaques (also known as atheromas) have amore complex structure than fatty streaks (FIG. 1). Inthe centre of a plaque, foam cells and extracellular lipiddroplets form a core region that is surrounded by a capof smooth muscle cells and a collagen-rich matrix2.Other cell types present in plaques include dendritic cells(DCs)3, mast cells4, a few B cells2and probably naturalkiller T (NKT) cells. The shoulder region of the plaque,which is where it grows, and the interface between thecap and the core have particularly abundant accumu-lations of T cells and macrophages2. Many of theseimmune cells show signs of activation and produce pro-inflammatory cytokines such as interferon-(IFN) and

    *Center for Molecular

    Medicine, Department

    of Medicine, Karolinska

    University Hospital,

    Karolinska Institute,

    Stockholm, SE-17176,

    Sweden.Leducq Transatlantic

    Network of Excellence in

    Cardiovascular Research,

    Brigham and Womens

    Hospital and Harvard

    Medical School, Boston,Massachusetts, USA.Donald W. Reynolds

    Cardiovascular Clinical

    Research Center, Department

    of Medicine, Brigham

    and Womens Hospital and

    Harvard Medical School,

    Boston, Massachusetts

    02115, USA.

    Correspondence to P.L.

    e-mail:

    [email protected]

    doi:10.1038/nri1882

    Published online

    16 June 2006

    Plaque

    An atherosclerotic lesion

    consisting of a fibrotic cap

    surrounding a lipid-rich core.

    The lesion is the site of

    inflammation, lipid

    accumulation and cell death.Also known as an atheroma.

    The immune response inatherosclerosis: a double-edged swordGran K. Hansson*and Peter Libby

    Abstract | Immune responses participate in every phase of atherosclerosis. There is

    increasing evidence that both adaptive and innate immunity tightly regulate atherogenesis.

    Although improved treatment of hyperlipidaemia reduces the risk for cardiac and cerebral

    complications of atherosclerosis, these remain among the most prevalent of diseases

    and will probably become the most common cause of death globally within 15 years.This Review focuses on the role of immune mechanisms in the formation and activation of

    atherosclerotic plaques, and also includes a discussion of the use of inflammatory markers

    for predicting cardiovascular events. We also outline possible future targets for prevention,

    diagnosis and treatment of atherosclerosis.

    R E V I E W S

    508 |JULY 2006 |VOLUME 6 www.nature.com/reviews/immunol

    2006Nature Publishing Group

  • 8/12/2019 The Immune Response In

    2/12

    Cellular debrisand cholesterol

    Shoulder

    Media

    Blood-vessel lumen

    Endothelial cell

    Normal artery

    Elastic laminaIntima

    Media

    Foam cell T cellMacrophage Mast cell MonocyteCholesterol Dendritic cellDead cellSmoothmuscle cell

    Myocardial infarction

    An episode of acute cardiac

    ischaemia that leads to death

    of heart muscle cells. It is

    usually caused by a thrombotic

    atherosclerotic plaque.

    Ischaemic stroke

    An episode of acute regional

    ischaemia in the brain leading

    to nerve-cell death. It is usually

    caused by thrombi or emboli

    from atherosclerotic plaques.

    Aneurysm

    The local dilatation of an artery

    caused by weakening of the

    artery wall. Some, but not all,

    aneurysms are caused by

    atherosclerosis.

    Intima

    The innermost layer of anartery, which consists of loose

    connective tissue and is

    covered by a monolayer of

    endothelium. Atherosclerotic

    plaques form in the intima.

    Fibrous cap

    A structure composed of a

    dense collagen-rich

    extracellular matrix with

    occasional smooth muscle

    cells, macrophages and T cells

    that typically overlies the

    characteristic central lipid core

    of plaques.

    tumour-necrosis factor (TNF)5. With time, the plaquecan progress into an even more complex lesion, thelipid core of which has become a paucicellular pool ofcholesterol deposits surrounded by a fibrous capof vary-ing thickness. The fibrous cap prevents contact betweenthe blood and the pro-thrombotic material in the lesion(FIG. 1). Disruption of the cap can lead to thrombosis andmany of the adverse clinical outcomes associated withatherosclerosis.

    Models of atherogenesis in mutant mice

    Direct analysis of the early phases of human atherosclerosispresents obvious obstacles. Therefore, systematic inves-tigation of the mechanisms that initiate atherosclerosisrelies on animal models of the disease. The availableobservations indicate that there is substantial overlapbetween disease development in these animal models andthe human disease. Two strains of genetically altered micehave been particularly fruitful in this regard. Apoe/

    mice lack apolipoprotein E (APOE; which is a keycomponent in cholesterol metabolism), and developspontaneous hypercholesterolaemia and atheroscleroticdisease (which is exacerbated by an atherogenic diet)that progresses to myocardial infarction and stroke 6,7.Low-density-lipoprotein receptor (LDLR)-deficient micerespond to being fed with fat by developing hypercholes-terolaemia and atherosclerotic plaques8. The crossbreed-ing of these mice with mice that carry deletions in genesencoding crucial components of the immune systemhas provided important information on the role of theimmune system in the pathogenesis of atherosclerosis. Inaddition, bone-marrow transplantation of, and spleen-cell

    transfer to,Apoe/or Ldlr/mice has offered insights intothe role of specific populations of bone-marrow-derivedcells in disease development.

    Immune-cell recruitment initiates atherosclerotic-plaque formation.In experimental animals, endothelialcells in the arteries express leukocyte adhesion mol-ecules, in particular vascular cell-adhesion molecule 1(VCAM1), as part of the initial vascular response tocholesterol accumulation in the intima9(FIG. 2a). Thepatchy distribution of adhesion-molecule expressioncorresponds to the subsequent position at which fattystreaks form10. This patchy pattern of expression prob-ably reflects haemodynamic factors, because the shearstresses and disturbed fluid flows vary over the arterialbed in a similar way to the predilection sites for athero-sclerosis. Interestingly, exposing cultured endothelialcells to oscillatory shear stress that mimics arterialblood flow increases the expression of several leukocyte

    adhesion molecules11.Shortly after VCAM1 induction, monocytes and

    T cells enter the arterial intima (FIG. 2a). Under the influ-ence of macrophage colony-stimulating factor (M-CSF)produced by endothelial cells and smooth muscle cells12,the monocytes differentiate into macrophages13(FIG. 2b)and T cells can undergo antigen-dependent activation(FIG. 2c;see later). Interestingly, VCAM1 expression bythe endothelium ceases after a few weeks, but smoothmuscle cells begin to express this adhesion molecule14.Expression of VCAM1 and other adhesion molecules bysmooth muscle cells might promote the recruitment andretention of mononuclear cells in the arterial intima.

    Figure 1 | Cellular composition of atherosclerotic plaques. The atherosclerotic plaque has a core containing lipids

    (which include esterified cholesterol and cholesterol crystals) and debris from dead cells. Surrounding it, a fibrous cap

    containing smooth muscle cells and collagen fibres stabilizes the plaque. Immune cells including macrophages, T cells

    and mast cells populate the plaque, and are frequently in an activated state. They produce cytokines, proteases, pro-

    thrombotic molecules and vasoactive substances, all of which can affect plaque inflammation and vascular function.

    Until complications occur, an intact endothelium covers the plaque.

    R E V I E W S

    NATURE REVIEWS |IMMUNOLOGY VOLUME 6 |JULY 2006 |509

    2006Nature Publishing Group

  • 8/12/2019 The Immune Response In

    3/12

    Endothelialcell

    MHC class II

    TCR

    APC

    Scavengerreceptor

    VLA4VCAM1

    Blood-vessellumen

    LDL

    oxLDL

    Monocyte

    MacrophageMonocyte

    Foam cell

    Chemokinereceptor Chemokine

    M-CSF

    T cell

    TLR T cell Endothelial cell

    LPS, HSP60or oxLDL

    Pro-inflammatory cytokinesProteasesProcoagulantsPro-apoptotic factors

    Increased adhesion moleculesIncreased permeabilityIncreased propensity forthrombus formation

    Decreased collagen productionDecreased proliferation

    CD4+T cell

    TH1 cell

    IL-12IL-15IL-18

    b

    c

    oxLDL

    TH1 cell

    Smoothmuscle cell

    MHC class IITCR

    IFNTNF

    CD40L CD40

    Macrophage

    ProteasesPro-inflammatorymediators

    Decreasedinflammation

    TH1 cell

    TH2 cell orregulatoryT cell

    Smoothmuscle cell

    TGF

    TGF

    IL-10

    e

    Macrophage

    a d

    T cell Endothelial cellT cell Endothelial cell

    T cell Endothelial cell

    Figure 2 | Recruitment and activation of immune cells in atherosclerotic plaques. a |Low-density lipoprotein (LDL)

    diffuses from the blood into the innermost layer of the artery, where LDL particles can associate with proteoglycans of the

    extracellular matrix. The LDL of this extracellular pool is modified by enzymes and oxygen radicals to form molecules such

    as oxidized LDL (oxLDL). Biologically active lipids are released and induce endothelial cells to express leukocyte adhesion

    molecules, such as vascular cell-adhesion molecule 1 (VCAM1). Monocytes and T cells bind to VCAM1-expressing

    endothelial cells through very late antigen 4 (VLA4) and respond to locally produced chemokines by migrating into the

    arterial tissue. b | Monocytes differentiate into macrophages in response to local macrophage colony-stimulating factor

    (M-CSF) and other stimuli. Expression of many pattern-recognition receptors increases, including scavenger receptors

    and Toll-like receptors (TLRs). Scavenger receptors mediate macrophage uptake of oxLDL particles, which leads tointracellular cholesterol accumulation and the formation of foam cells. TLRs bind lipopolysaccharide (LPS), heat-shock

    protein 60 (HSP60), oxLDL and other ligands, which instigates the production of many pro-inflammatory molecules by

    macrophages.c | T cells undergo activation after interacting with antigen-presenting cells (APCs), such as macrophages

    or dendritic cells, both of which process and present local antigens including oxLDL, HSP60 and possibly components

    of local microorganisms. A T helper 1 (TH1)-cell-dominated response ensues, possibly owing to the local production of

    interleukin-12 (IL-12), IL-18 and other cytokines. Antigen presentation and TH1-cell differentiation might also occur in

    regional lymph nodes. d | TH1 cells produce inflammatory cytokines including interferon-(IFN ) and tumour-necrosis

    factor (TNF) and express CD40 ligand (CD40L). These messengers prompt macrophage activation, production of

    proteases and other pro-inflammatory mediators, activate endothelial cells, increase adhesion-molecule expression

    and the propensity for thrombus formation, and inhibit smooth-muscle-cell proliferation and collagen production.

    e | Plaque inflammation might be attenuated in response to the anti-inflammatory cytokines IL-10 and transforming

    growth factor-(TGF), which are produced by several cell types including regulatory T cells, macrophages, and for TGF,also vascular cells and platelets. TCR, T-cell receptor.

    R E V I E W S

    510 |JULY 2006 |VOLUME 6 www.nature.com/reviews/immunol

    2006Nature Publishing Group

  • 8/12/2019 The Immune Response In

    4/12

  • 8/12/2019 The Immune Response In

    5/12

    Vasa vasorum

    Small nutrient vessels in the

    normal adventitia and outer

    media of the artery wall,

    which can also give rise to

    microvessels in the plaque.

    cells2. Small populations of mast cells, B cells and DCsoccur in plaques and, together with T cells, monocytesand macrophages, might traffic between the blood inthe arterial lumina, the lesioned artery wall, the vasavasorum microvessels that penetrate the artery andthe regional lymph nodes.

    The ratio of CD4+ to CD8+T cells in advancedplaques resembles that found in peripheral blood2. MostT cells are T cells,but there is also a small propor-tion of T cells. Lesions of early stages of experimentalatherosclerosis contain oligoclonal expansions of CD4+cells expressing an T-cell receptor (TCR)37, indi-cating activation in response to a limited set of localantigens (FIG. 2c). The CD4+T cells that are isolatedfrom human plaques are mostly CD45RO-expressingmemory and/or effector T cells38. An initial round ofT-cell activation in response to athero-antigens mightoccur in the regional lymph nodes, possibly after anti-gen presentation by DCs trafficking from the plaqueto the lymph node39. After entering the blood, previ-ously activated memory and/or effector T cells bind

    cell-surface adhesion molecules that are expressed byendothelial cells at the plaque surface and/or in the vasa

    vasorum, and then enter the plaque. Macrophages inthe plaque expressing MHC class II molecules mightthen present antigen to these T cells, leading to furtherrounds of activation.

    Very recently,Ath1, which is an atherosclerosis sus-ceptibility locus on mouse chromosome 1, was mappedto the gene encoding OX40 ligand (OX40L; also knownas TNFSF4), which is a co-stimulatory factor for T-cellactivation40. Reduced expression of this protein was asso-ciated with reduced atherosclerosis in inbred strains thatdiffer in their Tnfsf4 alleles and also in mice carrying atargeted deletion of this gene40. Polymorphisms in humanTNFSF4were found to be associated with coronaryatherosclerosis and with an increased risk for myocar-dial infarction in a human genetic epidemiology study 40.These data re-emphasize the importance ofimmuneactivation in atherosclerosis and its complications41.

    Plaque antigens activate local cellular adaptive immu-

    nity.Cloning T cells from surgically removed humanplaques has identified several cell-mediated, local adap-tive immune reactions. CD4+T-cell clones derived fromplaques recognize oxLDL, with other clones recognizingHSP60 or other antigens derived from certain pathogenicmicroorganisms, such as Chlamydia pneumoniae 42,43

    (FIG. 2c). In all of these cases, antigen recognition wasrestricted by HLA-DR and involved TCR+CD4+T cells42,44,45.

    Antigen-presenting cells selectively internalizeoxLDL particles through the scavenger-receptor path-way. After proteolytic processing, fragments of theprotein component of LDL, APOB, bind nascent MHCclass II molecules and traffic to the cell surface. Indeed,APOB fragments are among the peptides displayed mostfrequently by HLA-DR molecules in cultured humanlymphoblastoid cells46. Therefore, receptor-mediatedendocytosis and the antigen-presentation pathway facili-tate MHC class II presentation of LDL-derived peptides

    to CD4+T cells. As expected, no T cells react with nativeLDL components. However, oxidative modificationof LDL breaks tolerance and oxLDL-reactive T cells local-ize in plaques, lymph nodes, and in the blood of patientswith atherosclerosis and experimental animals42,47.OxLDL-reactive CD4+ T cells probably recognizeAPOB-derived oligopeptides carrying adducts formedduring oxidation48; whereas oxLDL-specific antibodiesreact with oxidized phospholipids such as phosphoryl-choline49,50, as well as aldehyde-peptide epitopes includingmalondialdehyde-lysine42,44,45.

    Most oxLDL-reactive CD4+T cells have a T helper 1(T

    H1)-cell phenotype42,47. Because T

    H1 cytokines (such as

    IFN) generally stimulate pro-atherosclerotic processes(see later), these T cells probably promote atherogenesis,a conclusion supported by adoptive-transfer studies insevere combined immunodeficient (SCID) mice lack-ing APOE51. As expected, these mice show substantiallyreduced atherosclerotic plaques compared with immuno-competentApoe/mice. Transfer of CD4+T cells fromimmunocompetentApoe/mice to SCID mice lacking

    ApoE increases the atherogenesis found in immuno-deficient mice, to almost the same level as that foundin fully immunocompetentApoe/mice. Therefore, thenet effect of CD4+T cells is to increase atherogenesis inmice susceptible to atherosclerotic disease. Obviously,this finding does not preclude the existence of T-cellsubsets that might mitigate disease.

    Recent studies have identified transcripts encodingV14J281-containing TCR -chains in plaques ofhypercholesterolaemic mice, indicating the presenceof NKT cells52. The abundance of CD1 molecules inplaques53indicates that CD1-mediated NKT-cell acti-

    vation takes place, but the absence of specific markersfor NKT cells has hampered direct immunohistologicaldemonstration of NKT cells in plaques. However, admin-istration of ligands that specifically activate NKT cells toApoe/mice shows that NKT-cell activation increasesearly atherosclerotic plaque development concomitantlywith increased local expression of pro-inflammatorycytokines, whereas abrogation of CD1-mediated antigenpresentation reduces disease52,54. Therefore, NKT cellscontribute to atherosclerosis, probably by antigen-specific activation in response to lipid antigens presentin plaques.

    A role for TH

    1/TH

    2-cytokine balance?Analyses ofcell-surface-marker expression and cytokine secretion

    indicate activation of a remarkably large proportionof T cells in plaques38. T

    H1-type cytokines dominate

    in mouse models of atherosclerosis and in humanplaques. For example, human plaques contain cellsproducing IFN, interleukin-12 (IL-12), IL-15, IL-18and TNF, but few cells producing the T

    H2-type cytokine

    IL-4 (REFS 42,55,56). Together with the histopatho-logical features of accumulation of macrophages andT cells, the predominance of T

    H1-type cytokines indi-

    cates that atherosclerosis is a TH

    1-cell-driven disease(FIG. 2d). This hypothesis is supported by studies ingenetically altered mice that show that there is reducedatherosclerosis in hypercholesterolaemic mice lacking

    R E V I E W S

    512 |JULY 2006 |VOLUME 6 www.nature.com/reviews/immunol

    2006Nature Publishing Group

  • 8/12/2019 The Immune Response In

    6/12

    Tissue factor

    A procoagulant that stimulates

    thrombus formation, when in

    contact with blood, by

    accelerating the action of

    factors VIIa and Xa.

    IFNor its receptor57,58, IL-12 (REF. 59), IL-18 (REF. 60),TNF61or the T

    H1-cell-inducing transcription factor

    T-bet62. Administration of recombinant IFN63orthe T

    H1-cell-inhibiting drug pentoxifyllin64to hyper-

    cholesterolaemic mice led to increased and decreasedatherosclerosis, respectively, lending further supportto this hypothesis64.

    If TH

    1 cytokines stimulate plaque formation andT

    H2 cytokines inhibit T

    H1-cell responses, can T

    H2-cell

    responses protect against atherosclerosis? In supportof this proposition, C57BL/6 mice (which are proneto T

    H1-type immune responses) develop fatty streaks

    if fed a high-cholesterol diet, whereas BALB/c mice(which are prone to T

    H2-type immune responses)

    are resistant to atherogenesis65,66. Targeted deletionof the gene encoding signal transducer and activa-tor of transcription 6 (STAT6), a transcription factorthat is essential for the differentiation of T

    H2 cells,

    renders BALB/c mice susceptible to atherogenesis andthis occurs in parallel with a switch from T

    H2-cell to

    TH1-cell responses66.

    Although these studies of the development offatty streaks indicate opposing roles for T

    H1 and T

    H2

    cells in disease development, mice that develop moreadvanced plaques show a more complicated picture.Pharmacological inhibition of T

    H1 cells using pentoxi-

    fyllin or IL-18-binding protein inhibits atherosclerosisinApoe/mice64,67, and administration of recombinantT

    H1 cytokines (recombinant IL-18 and IFN) exacer-

    bates disease63,68. However, data for IL-4, which isthe prototypic T

    H2 cytokine, are inconclusive. Some

    studies have shown that IL-4 has a protective effect,whereas others found reduced disease in the absenceof IL-4 (REFS 59,69). These divergent findings, underdifferent experimental conditions, might reflect thecomplex range of biological activities found for IL-4,including stimulation of scavenger-receptor expres-sion and the induction of elastin degrading MMP12,which can lead to aneurysm formation70. Defining therole of T

    H2 cells in atherosclerosis, therefore, requires

    further study.

    Pro-atherosclerotic action of TH1 cells.How can T

    H1

    cells promote disease development? IL-12 and IL-18,which are produced by macrophages and smooth mus-cle cells in plaques, can indirectly affect the develop-ment of plaques by promoting T

    H1-cell differentiation.

    By contrast, IFNand TNF directly accelerate disease

    through their actions on macrophages and vascularcells (FIG. 2c,d). IFNactivates macrophages, therebyincreasing their production of nitric oxide, pro-inflam-matory cytokines, and pro-thrombotic and vasoactivemediators. Additionally,IFNinhibits endothelial-cellproliferation71, the proliferation and differentiationof vascular smooth muscle cells72, and also decreasescollagen production by these smooth muscle cells73.Decreasing the cell and collagen content of the fibrouscap might reduce the stability of the plaque. Therefore,the combined effects of IFNon cells of the formingplaque promote inflammation and extracellular-matrixdestabilization.

    The pro-inflammatory cytokine TNF triggers vascu-lar inflammation through the NF-B pathway, inducingthe production of reactive oxygen and nitrogen species,proteolytic enzymes and pro-thrombotic tissue factorbyendothelial cells, and modulates the fibrinolytic capacityof the cells7476. TNF also has profound metabolic effectsthat include the suppression of lipoprotein lipase, whichleads to the accumulation of triglyceride-rich lipopro-teins in the blood. Such lipoproteins, and the TNF levels,have been associated with heart disease in clinical stud-ies7779. Genetic loss-of-function studies also support theidea that TNF has a pro-atherogenic role61.

    CD40 and CD40L: a co-stimulatory dyad with pro-atherogenic action.The cell-surface proteins CD40andCD40 ligand (CD40L; also known as CD154) have sev-eral similarities to soluble pro-inflammatory cytokines.CD40 ligation on cells found in plaques triggers aninflammatory response similar to that elicited by TNF,that is, secretion of other cytokines and MMPs, andexpression of adhesion molecules80. Importantly, CD40

    ligation causes expression of the procoagulant tissuefactor by human macrophages, something that solublepro-inflammatory cytokines do not do. Macrophages andT cells express CD40 and CD40L, as do vascular endothe-lial cells, smooth muscle cells and platelets81,82. Therefore,CD40 ligation propagates inflammatory activation in allthe main cell types involved in atherogenesis. Inhibitionof CD40 ligation andinactivation of the gene encodingCD40L reduces atherosclerotic plaques in hypercholes-terolaemic mice83,84. Unfortunately, CD40 blockade inhumans can promote platelet aggregation and thrombosis,which is an obstacle to its clinical application.

    Anti-atherogenic immunity

    Anti-inflammatory cytokines.Although local cellularimmunity predominantly promotes atherosclerosisthrough the action of cell-surface molecules (such asCD40CD40L) and cytokines (such as IFNand TNF),counterbalancing factors can function to dampen dis-ease activity (FIG. 2e). Two anti-inflammatory cytokines,IL-10 and transforming growth factor-(TGF), provideparticularly important atheroprotective signals.

    Two groups have reported previously that IL-10-deficient C57BL/6 mice that consume a fatty diet developan increased quantity of fatty streaks compared withwild-type mice85,86. By contrast, Il10transgenic C57BL/6mice do not develop fatty streaks, thereby providing evi-

    dence of a protective role for IL-10 in atherosclerosis85,86.The mouse model used in these early studies mimickedthe initial stage of atherogenesis, but the mice didnot develop lesions similar to human clinical disease.However, subsequent experiments usingApoe/mice,which develop atherosclerotic lesions that are more simi-lar to those found in humans, also show an atheroprotec-tive role for IL-10 (REF. 87). Interestingly, IL-10 promotesarteriopathy in transplanted hearts, indicating a morecomplex picture.

    The pluripotent cytokine TGFhas many effects on adiverse range of cell types and can inhibit atherosclerosisat least as well as IL-10. For example, TGFpromotes

    R E V I E W S

    NATURE REVIEWS |IMMUNOLOGY VOLUME 6 |JULY 2006 |513

    2006Nature Publishing Group

  • 8/12/2019 The Immune Response In

    7/12

    collagen production, which could increase plaquestability. Treatment with tamoxifen, which is a TGF-stimulating oestrogen-receptor agonist, reduces theformation of fatty streaks in C57BL/6 mice fed withfat88, whereas administration of TGF-specific block-ing antibodies or decoy receptors for TGFreducesatherosclerotic plaque formation in Ldlr /mice89,90.However, these studies did not identify the mechanismof action of TGF.

    Two more-recent studies show that TGFexerts itsatheroprotective effects by modulating T-cell activation.In the first study, crossbreeding mice carrying dominant-negative TGFreceptors (that were expressed underthe control of the Cd4 promoter) with Apoe/miceled to a fivefold increase in plaque size and advancedplaques were found in the proximal aorta of 12-week-old crossbred mice91. Notably, the plaques showed signsof increased inflammation and had fewer interstitialcollagen fibres, a characteristic of human plaques thatcause thrombosis (see later). In the second study, bonemarrow from mice that expressed a dominant-negative

    form of the type II TGFreceptor (that was expressedunder the control of the Cd2promoter) was transplantedto irradiated Ldlr /mice92. Again, plaques showed signsof substantial inflammation and a poorly developedcollagenous matrix. These studies show the importantatheroprotective effects of TGFthat occur through thedampening of T-cell activity.

    Several cell types can produce TGFand IL-10,including platelets, macrophages, endothelial cells,smooth muscle cells and regulatory T cells. Activationof regulatory T cells could therefore offer a means ofantigen-specific atheroprotection (FIG. 2e). A recent studysupports this idea by showing that the transfer of naturalCD4+CD25+regulatory T (T

    Reg

    ) cells reduces athero-sclerosis, whereas depletion of CD25+cells increases dis-ease inApoe/mice93. Depletion of CD25+cells in micelacking functional TGFreceptors on T cells did not alterplaque size, indicating that this cytokine mediates theatheroprotective effect of regulatory T cells93.

    Humoral immunity.In addition to innate immunityand T cells, antibodies with different specificities canparticipate in atherosclerosis. Humans and experimen-tal animals with disease have antibodies specific foroxLDL particles44. B-cell epitopes in oxLDL includeamino-acid residues of APOB that are modified bylipid peroxidation products, such as malondialdehyde

    and 4-hydroxynonenal. Although some clinical and epi-demiological studies have found positive correlationsbetween the presence of antibodies specific for oxLDLand the progression of atherosclerosis94,95, other studieshave not detected any correlation. Interestingly, anti-bodies specific for oxLDL, mainly of the IgM isotype,also circulate in asymptomatic humans96and crossreactwith apoptotic bodies49. These antibodies bind theoxidized phospholipids in oxLDL and also recognizephosphorylcholine in the cell wall of Streptococcuspneumoniae49. Phosphorylcholine-specific IgM con-sists of germline-encoded antibodies of the T15 typethat are produced by B1 cells49. Therefore, expansion

    of B-cell clones that produce T15-type antibodies, forexample during a pneumococcal infection, might affectthe development of plaques. Indeed, immunization ofLdlr /mice with a pneumococcal vaccine reduced theextent of atherosclerosis50.

    Molecular mimicry could explain the crossreactiv-ity between the humoral immune responses to oxLDL,apoptotic bodies and pneumococci. This mechanismmight also apply to HSP60, another antigen associatedwith atherosclerosis97. HSP60 is a chaperone moleculethat is involved in protein folding and can be detectedin plaques. Antibodies specific for HSP60 are found inexperimental animals that have atherosclerosis and havebeen correlated with disease progression in a humancohort study98. Present in prokaryotes and eukaryotes,HSP60 has shown remarkable sequence conservation dur-ing evolution. As antibodies specific for HSP60 crossreactbetween microbial and eukaryotic HSP60, antibodiesthat react to human HSP60 can be generated in responseto infection with microbes that express HSP60, such asC. pneumoniae99.

    Several further experimental, and some human, stud-ies show that humoral immunity can protect againstatherosclerosis. Splenectomy increases atherosclerosis inbothApoe/mice and humans100. InApoe/mice, trans-fer of splenic B cells from atherosclerotic animals intosplenectomized recipients protects against disease, pos-sibly because of the production of protective antibodiesby B cells100.

    Immunization experiments identify oxLDL andHSP60 as important antigens that can induce protective,as well as detrimental, immune responses (TABLE 1; seealso later). A tentative conclusion from these studies is thatT

    H1-type immune responses promote disease; whereas

    humoral immunity has protective effects, possibly byeliminating antigens before they reach plaques.

    Adaptive immunity disrupts plaques

    In general, the gravest clinical complications of athero-sclerosis result from the sudden thrombotic occlusion ofan artery101. The sudden onset of myocardial infarction,as well as many strokes and episodes of acute limb ischae-mia, is caused by thrombi that arise from atheroscleroticplaques that do not necessarily tightly narrow the artery.Therefore, many episodes of damage to the heart muscle,brain or lower extremities can occur without warning, alltoo often with devastating consequences.

    Physical disruption of a plaque is the most frequent

    cause of thrombotic occlusions. Indeed, the most fre-quent patho-anatomical substrate for sudden coronarythrombosis is rupture of the fibrous cap that overlies thelipid core of the plaque101(FIG. 3). Fibres of interstitial col-lagens (types I and III) normally confer biomechanicalstability on the fibrous cap of the plaque. As discussedearlier, the T

    H1-cytokine IFNstrongly inhibits the

    production of interstitial collagens by vascular smoothmuscle cells, which are the main source in the arterialwall of this extracellular-matrix macromolecule73. IFNcan also inhibit the proliferation of smooth musclecells, thereby reducing the stabilizing and collagen-synthesizing cellular component of the plaque72. Also,

    R E V I E W S

    514 |JULY 2006 |VOLUME 6 www.nature.com/reviews/immunol

    2006Nature Publishing Group

  • 8/12/2019 The Immune Response In

    8/12

    Angina pectoris

    A reversible attack of chest

    discomfort, usually caused by

    an imbalance between the

    oxygen demand of the working

    heart muscle and the

    insufficient supply through

    narrow, atheroscleroticcoronary arteries.

    Angioplasty

    A percutaneous catheter

    procedure that inflates a

    balloon in areas of narrowing

    (stenosis) in arteries.

    Statins

    A class of drugs that inhibit the

    rate-limiting enzyme (3-

    hydroxy-3-methylglutaryl

    coenzyme A reductase) in the

    pathway of cholesterol

    biosynthesis.

    proteases elaborated mainly from activated macrophagesin plaques can degrade collagen102,103. In addition, liga-tion of CD40 expressed by macrophages increases theproduction of matrix-degrading proteases that include

    the interstitial collagenases of the MMP family, MMP1,MMP8 and MMP13 (REF. 104). Therefore, T

    H1 cells

    probably have an essential role in regulating the func-tions of smooth muscle cells (collagen-fibre formation)and macrophages (collagen degradation) that cruciallyregulate the integrity of the fibrous cap of the plaqueand therefore its susceptibility to rupture and provokethrombosis.

    Once coagulation factors in the blood gain accessto the lipid core of the plaque following rupture of thefibrous cap, thrombosis commonly ensues. Tissue factor,the potent procoagulant expressed by a subpopulationof macrophages in the lipid core of the plaque, triggersthese thromboses101. As noted earlier, ligation of CD40expressed by macrophages strongly induces expressionof tissue factor80. Indeed, T cells expressing CD40L local-ize in the vicinity of macrophages that are expressing tis-sue factor in the lipid core of human plaques105. Becauseplatelets can also express CD40L82when activated, positivefeedback can amplify the local inflammatory response,once a thrombus begins to form, because of generation ofthe protease thrombin induced by tissue factor and plate-let activation induced by thrombin. Therefore, althoughT cells could orchestrate the pathophysiology of plaquedisruption, dysregulated antigen-nonspecific pathwaysprobablyamplify and sustain the formation of thrombi.Modulation of immunity in atherosclerosisImmunopharmacological intervention against sympto-matic atherosclerosis.Although thrombi cause most ofthe acute complications of atherosclerosis, the gradualformation of stenoses that impede blood flow causesmany of the chronic symptoms of atherosclerotic disease,such as angina pectoris (chest discomfort precipitatedtypically by physical or emotional stress). Recent decadeshave witnessed important advances in the ability of inter-

    ventions, particularly percutaneous procedures, to relievestenoses and reduce ischaemia. Until recently, however,the long-term success of mechanical procedures, suchas the deployment of arterial stents (metal scaffolds to

    hold arteries open) and balloon angioplasty (inflationof miniature balloons in blocked segments of arter-ies to expand the arterial lumen), has been limitedby re-growth of intimal tissue which is known as

    in-stent stenosis and restenosis, respectively. Thisfibro-proliferative response of the injured artery canre-occlude the lumen within months in a substantialminority of patients.

    Recently, the coating of stents with immunosup-pressive agents, for example sirolimus (Rapamycin),has shown striking effectiveness at reducing in-stentstenosis106. This advance has markedly improved clinicaloutcomes in patients undergoing percutaneous interven-tion. Early preclinical studies provided the experimentalbasis for this important therapeutic advance by show-ing that another immunosuppressant, cyclosporin,reduces intimal-cell proliferation in response to arterialinjury107.

    The use of statinshas shown striking clinical benefitin preventing atherosclerotic complications during thepast decade. Numerous clinical trials have establishedthat 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (drugs of the statin family)can reduce various atherosclerotic complications108.The lowering of LDL cholesterol concentrations in theblood doubtless accounts for much of this remarkableclinical benefit.However,recent data indicate that partof the clinical benefit of statins occurs because of ananti-inflammatory effect that is apparently not relatedto LDL reduction109. (See REF. 110for a detailed discus-sion of non-LDL-lowering effects of statins).

    By blocking HMG-CoA reductase, statins preventthe formation of lipids that control the function ofseveral intracellular proteins111. By acting on the MHCclass II transactivator (CIITA), statins can interfere withthe transcriptional induction of MHC class II mol-ecules, which would decrease immune activation in theplaque112. Statins can also limit the accelerated arterio-sclerosis (sclerosis of the arterial walls)that complicatessolid-organ transplantation, a disease that often occursin the absence of increased concentrations of LDL113.They also seem to reduce disease activity in patients withrheumatoid arthritis114and in mice with experimentalautoimmune encephalomyelitis115. All these results lend

    Table 1 | Immunization against atherosclerosis in experimental models

    Antigen Route Animal model Effect on atherosclerosis References

    MDA-LDL Subcutaneous WHHL rabbits Reduced 124

    oxLDL Subcutaneous Fat-fed NZW rabbits Reduced 125

    MDA-LDL Subcutaneous Apoe/mice Reduced 47,126

    MDA-LDL Subcutaneous Ldlr/mice Reduced 127

    APOB-peptides Subcutaneous Apoe/mice Reduced 95,128

    MDA-LDL Subcutaneous Cd4/Apoe/mice Reduced 129

    HSP65 Subcutaneous Ldlr/mice Increased 130

    HSP65 Peroral/nasal Ldlr/mice Reduced 131,132

    2-GPI Subcutaneous Ldlr/mice Increased 133

    APO, apolipoprotein; GPI, glycoprotein I; HSP65, heat-shock protein 65; LDL, low-density lipoprotein; LDLR, LDL receptor; MDA-LDL,malondialdehyde-modified LDL; NZW, New Zealand white; oxLDL, oxidized LDL; WHHL, Watanabe hereditably hyperlipidaemic.

    R E V I E W S

    NATURE REVIEWS |IMMUNOLOGY VOLUME 6 |JULY 2006 |515

    2006Nature Publishing Group

  • 8/12/2019 The Immune Response In

    9/12

    Blood-vessel lumen

    Endothelial cell

    Elasticlamina

    Cellular debrisand cholesterol

    Rupture

    Thrombus

    Platelet

    Erythrocyte

    Fibrin

    Foam cell T cellMacrophage Mast cell MonocyteCholesterol Dendritic cellDead cellSmoothmuscle cell

    Peroxisome-proliferator-

    activated receptors

    Nuclear receptors that

    participate in the regulation

    of cellular metabolism and

    differentiation.

    Thiazolidinedione

    A class of medication, used

    to treat diabetes, that binds

    peroxisome-proliferator-

    activated receptor-.

    support to the idea that the immunomodulatory actionsof statins also contribute to their effects in patients withatherosclerosis.

    Recent studies have establishedthat another categoryof anti-atherosclerotic drugs, the ligands for a group ofnuclear transcription factors known as peroxisome-proliferator-activated receptors(PPARs), can inhibit T-cellactivation in vitro. Activators of both PPAR

    (members

    of the fibrate class of drugs) and PPAR(members ofthe thiazolidinedionefamily of drugs) can reduce T-cellactivation, as was shown by decreased production ofIFN, TNF and IL-2 (REF. 116). PPARagonists alsoinhibit inflammatory activation of vascular smooth

    muscle cells117. Therefore, activation of PPARor PPARmight also affect atherosclerosis in a beneficial mannerby blunting the adaptive and innate immune responses.

    Nonspecific anti-inflammatory therapies, such asnon-steroidal anti-inflammatory drugs (NSAIDs), havenot improved the cardiovascular outcome. Indeed, treat-ment with NSAIDs selective for cyclooxygenase-2 seemsto increase the risk of thrombotic complications118,119.Despite their marked anti-inflammatory properties,glucocorticosteroids themselves probably increase,rather than decrease, atherogenesis, as chronic admin-istration of these agents adversely affects plasma lipopro-teins, promotes insulin resistance and sodium retention,

    Figure 3 | Plaque activation, rupture and thrombosis. When activated, immune cells including macrophages, T cells

    and mast cells can release pro-inflammatory cytokines, which reduce collagen formation and induce the expression of

    tissue factor. Proteases that attack the collagenous cap are also released by activated immune cells. The weakened plaque

    might fissure when subjected to the forces of arterial blood pressure. Exposure of subendothelial structures and

    procoagulants such as tissue factor promotes platelet aggregation and thrombosis. A thrombus forms and might occlude

    the lumen of the artery, leading to acute ischaemia.

    R E V I E W S

    516 |JULY 2006 |VOLUME 6 www.nature.com/reviews/immunol

    2006Nature Publishing Group

  • 8/12/2019 The Immune Response In

    10/12

    C-reactive protein

    An acute-phase reactant

    protein, the plasma

    concentration of which

    increases in inflammatory

    states.

    and inhibits collagen and elastin formation. It thereforedoes not offer a reasonable therapeutic alternative in thechronic phases of atherosclerosis.

    Vaccination against atherosclerosis?Parenteral immu-nization with malondialdehyde-modified LDL (thatis, LDL with a defined oxidative modification) ormalondialdehyde-modified peptides derived from theLDL protein apolipoprotein inhibits atherosclerosis andthis occurs in parallel with increased titres of antibodyspecific for the immunogen (TABLE 1). Interestingly, pro-tection through this route does not require CD4+T-cellhelp120. Therefore, protection seems to depend mostlyon humoral immunity, at least in this model.

    By contrast, the outcomes after immunization withHSP60 or its mycobacterial homologue HSP65 arecomplex (TABLE 1). Parenteral immunization in C57BL/6mice fed with fat, as well as Ldlr /mice, aggravatesdisease, whereas oral or nasal immunization elicitsprotective immunity. Induction of mucosal immunityinvolves activation of regulatory T cells that produce

    anti-inflammatory cytokines and also high titres ofspecific antibodies. Therefore, the precise mechanismby which mucosal immunization leads to reducedatherosclerosis remains to be clarified.

    Although several questions remain, the immuniza-tion experiments with malondialdehyde-modified LDLand HSP60 indicate that it is possible that a vaccina-tion strategy might protect against atherosclerosis andits complications. Obviously, many obstacles remain,rendering the success of this approach unpredictable,particularly in humans.

    Conclusion

    The evidence reviewed in this article supports theinvolvement of the immune response in atherosclerosisfrom its initiation through to its thrombotic complica-tions. The concept that adaptive immunity pivotallyregulates atherogenesis has already been clinically useful.Markers of the acute-phase response, notably C-reactiveprotein(CRP), predict the prognosis of patients whohave already sustained a cardiovascular event121. Lesserelevations of CRP concentration, measured with a highlysensitive assay and previously considered in the normalrange, can be used to predict cardiovascular events inapparently well populations109,122,123. Markers of height-ened innate immune responses, such as CRP, correlatewith worse outcomes in individuals with acute coronarysyndromes121.

    Systemic administration of non-selective immuno-suppressive drugs will probably not be useful for thetreatment of atherosclerosis, at least during its longasymptomatic phase, because of the need for prolongedtherapy and the potential toxicities of such treatments.

    However, the immune system in its full complexity offersmuch more subtle targets for therapeutic manipulation.As more details emerge of the specific pathways involvedin the immune response and the inflammation thatoccur in atherosclerosis, more selective interventionsmight prove appropriate for long-term anti-atheroscle-rotic therapy. Also, as our ability to gauge the risk ofacute complications improves, we might be able to targetin a much more selective manner those therapies thatwould otherwise impair host defences if administeredon a long-term basis.

    1. Murray, C. J. & Lopez, A. D. Global mortality,

    disability, and the contribution of risk factors: global

    burden of disease study. Lancet349, 14361442

    (1997).

    2. Jonasson, L., Holm, J., Skalli, O., Bondjers, G. &

    Hansson, G. K. Regional accumulations of T cells,

    macrophages, and smooth muscle cells in the human

    atherosclerotic plaque.Arteriosclerosis6, 131138

    (1986).

    3. Bobryshev, Y. V. & Lord, R. S. A. S-100 positive cells

    in human arterial intima and in atherosclerotic lesions.

    Cardiovas. Res.29, 689696 (1995).

    4. Kovanen, P. T., Kaartinen, M. & Paavonen, T.

    Infiltrates of activated mast cells at the site of coronary

    atheromatous erosion or rupture in myocardial

    infarction. Circulation92, 10841088 (1995).

    5. Jonasson, L., Holm, J., Skalli, O., Gabbiani, G. &

    Hansson, G. K. Expression of class II transplantation

    antigen on vascular smooth muscle cells in human

    atherosclerosis.J. Clin. Invest. 76, 125131 (1985).6. Plump, A. S. et al. Severe hypercholesterolemia and

    atherosclerosis in apolipoprotein E-deficient mice

    created by homologous recombination in ES cells.

    Cell71, 343353 (1992).

    7. Piedrahita, J. A., Zhang, S. H., Hagaman, J. R.,

    Oliver, P. M. & Maeda, N. Generation of mice carrying

    a mutant apolipoprotein E gene inactivated by gene

    targeting in embryonic stem cells. Proc. Natl Acad.

    Sci. USA89, 44714475 (1992).8. Ishibashi, S., Goldstein, J. L., Brown, M. S.,

    Herz, J. & Burns, D. K. Massive xanthomatosis

    and atherosclerosis in cholesterol-fed low density

    lipoprotein receptor-negative mice.J. Clin. Invest.

    93, 18851893 (1994).

    9. Cybulsky, M. I. & Gimbrone, M. A. Endothelial

    expression of a mononuclear leukocyte adhesion

    molecule during atherosclerosis. Science251,

    788791 (1991).

    10. Nakashima, Y., Raines, E. W., Plump, A. S.,

    Breslow, J. L. & Ross, R. Upregulation of VCAM-1

    and ICAM-1 at atherosclerosis-prone sites on the

    endothelium in the apoE-deficient mouse.

    Arterioscler. Thromb. Vasc. Biol. 18, 842851

    (1998).

    11. Dai, G. et al. Distinct endothelial phenotypes

    evoked by arterial waveforms derived from

    atherosclerosis-susceptible and-resistant regions

    of human vasculature. Proc. Natl Acad. Sci. USA

    101, 1487114876 (2004).

    12. Rajavashisth, T. B. et al. Induction of endothelial

    cell expression of granulocyte and macrophage

    colony-stimulating factors by modified low-density

    lipoproteins. Nature344, 254257 (1990).13. Smith, J. D. et al. Decreased atherosclerosis in mice

    deficient in both macrophage colony-stimulating factor

    (op) and apolipoprotein E. Proc. Natl Acad. Sci. USA

    92, 82648268 (1995).

    14. Li, H., Cybulsky, M. I., Gimbrone, M. A. &Libby, P. Inducible expression of vascular cell

    adhesion molecule-1 by vascular smooth muscle cellsin vitroand within rabbit atheroma.Am. J. Pathol.

    143, 15511559 (1993).

    15. Dong, Z. M. et al. The combined role of P- and

    E-selectins in atherosclerosis.J. Clin. Invest.

    102, 145152 (1998).

    16. Cybulsky, M. I. et al.A major role for VCAM-1,

    but not ICAM-1, in early atherosclerosis.J. Clin. Invest. 107, 12551262 (2001).

    17. Boring, L., Gosling, J., Cleary, M. & Charo, I. F.

    Decreased lesion formation in CCR2/mice reveals a

    role for chemokines in the initiation of atherosclerosis.

    Nature394, 894897 (1998).

    18. Gu, L. et al.Absence of monocyte chemoattractant

    protein-1 reduces atherosclerosis in low density

    lipoprotein receptor-deficient mice. Mol. Cell

    2, 275281 (1998).

    19. Mach, F. et al.Differential expression of three

    T lymphocyte-activating CXC chemokines by

    human atheroma-associated cells.J. Clin. Invest.

    104, 10411050 (1999).

    20. Haley, K. J. et al. Overexpression of eotaxin and

    the CCR3 receptor in human atherosclerosis: using

    genomic technology to identify a potential novel

    pathway of vascular inflammation. Circulation

    102, 21852189 (2000).

    21. Minami, M. et al. Expression of SR-PSOX, a novel

    cell-surface scavenger receptor for phosphatidylserine

    and oxidized LDL in human atherosclerotic lesions.

    Arterioscler. Thromb. Vasc. Biol. 21, 17961800

    (2001).

    22. Veillard, N. R.et al.Antagonism of RANTES receptors

    reduces atherosclerotic plaque formation in mice.

    Circ. Res. 94, 253261 (2004).

    23. Combadiere, C. et al. Decreased atherosclerotic

    lesion formation in CX3CR1/apolipoprotein E double

    knockout mice. Circulation107, 10091016(2003).

    24. Lesnik, P., Haskell, C. A. & Charo, I. F. Decreased

    atherosclerosis in CX3CR1/mice reveals a role

    for fractalkine in atherogenesis.J. Clin. Invest.

    111, 333340 (2003).

    25. Steinberg, D. Low density lipoprotein oxidation

    and its pathobiological significance.J. Biol. Chem.

    272, 2096320966 (1997).

    26. Peiser, L., Mukhopadhyay, S. & Gordon, S. Scavenger

    receptors in innate immunity. Curr. Opin. Immunol.

    14, 123128 (2002).

    27. Nicoletti, A. et al. The macrophage scavenger

    receptor type A directs modified proteins to antigen

    presentation. Eur. J. Immunol. 29, 512521 (1999).

    28. Moore, K. J. et al. Loss of receptor-mediated lipid

    uptake via scavenger receptor A or CD36 pathways

    does not ameliorate atherosclerosis in hyperlipidemic

    mice.J. Clin. Invest.115, 21922201 (2005).

    R E V I E W S

    NATURE REVIEWS |IMMUNOLOGY VOLUME 6 |JULY 2006 |517

    2006Nature Publishing Group

  • 8/12/2019 The Immune Response In

    11/12

    29. Bodzioch, M. et al. The gene encoding ATP-binding

    cassette transporter 1 is mutated in Tangier disease.

    Nature Genet. 22, 347351 (1999).

    30. Janeway, C. A. Jr & Medzhitov, R. Innate immune

    recognition.Annu. Rev. Immunol. 20, 197216

    (2002).31. Edfeldt, K., Swedenborg, J., Hansson, G. K. &

    Yan, Z. Q. Expression of toll-like receptors in human

    atherosclerotic lesions: a possible pathway for plaque

    activation. Circulation105, 11581161 (2002).

    32. Kol, A., Lichtman, A. H., Finberg, R. W., Libby, P. &

    Kurt-Jones, E. A. Cutting edge: heat shock protein(HSP) 60 activates the innate immune response:

    CD14 is an essential receptor for HSP60 activation

    of mononuclear cells.J. Immunol. 164, 1317

    (2000).

    33. Xu, X. H. et al. Toll-like receptor-4 is expressed

    by macrophages in murine and human lipid-rich

    atherosclerotic plaques and upregulated by oxidized

    LDL. Circulation104, 31033108 (2001).34. Miller, Y. I. et al. Minimally modified LDL binds to

    CD14, induces macrophage spreading via TLR4/

    MD-2, and inhibits phagocytosis of apoptotic cells.

    J. Biol. Chem. 278, 15611568 (2003).

    35. Michelsen, K. S. et al. Lack of Toll-like receptor 4

    or myeloid differentiation factor 88 reduces

    atherosclerosis and alters plaque phenotype in mice

    deficient in apolipoprotein E. Proc. Natl Acad. Sci.

    USA101, 1067910684 (2004).

    36. Bjorkbacka, H. et al. Reduced atherosclerosis in

    MyD88-null mice links elevated serum cholesterol

    levels to activation of innate immunity signaling

    pathways. Nature Med. 10, 416421 (2004).

    37. Paulsson, G., Zhou, X., Trnquist, E. & Hansson, G. K.

    Oligoclonal T cell expansions in atherosclerotic lesions

    of apoE-deficient mice.Arterioscler. Thromb. Vasc.

    Biol. 20, 1017 (2000).38. Stemme, S., Holm, J. & Hansson, G. K. T lymphocytes

    in human atherosclerotic plaques are memory cells

    expressing CD45RO and the integrin VLA-1.

    Arterioscler. Thromb.12, 206211 (1992).

    39. Angeli, V.et al. Dyslipidemia associated with

    atherosclerotic disease systemically alters dendritic

    cell mobilization. Immunity21, 561574 (2004).40. Wang, X. et al. Positional identification of TNFSF4,

    encoding OX40 ligand, as a gene that influences

    atherosclerosis susceptibility. Nature Genet.

    37, 365372 (2005).

    41. Swanberg, M. et al. MHC2TA is associated

    with differential MHC molecule expression and

    susceptibility to rheumatoid arthritis, multiple

    sclerosis and myocardial infarction. Nature Genet.

    37, 486494 (2005).42. Stemme, S. et al. T lymphocytes from human

    atherosclerotic plaques recognize oxidized low

    density lipoprotein. Proc. Natl Acad. Sci. USA

    92, 38933897 (1995).

    43. de Boer, O. J. et al. Unstable atherosclerotic

    plaques contain T-cells that respond to Chlamydia

    pneumoniae. Cardiovasc. Res.48, 402408 (2000).

    44. Palinski, W. et al. Low density lipoprotein undergoes

    oxidative modification in vivo. Proc. Natl Acad. Sci.

    USA86, 13721376 (1989).

    45. Palinski, W. et al. Cloning of monoclonal

    autoantibodies to epitopes of oxidized lipoproteins

    from apolipoprotein E-deficient mice. Demonstration

    of epitopes of oxidized low density lipoprotein in

    human plasma.J. Clin. Invest. 98, 800814 (1996).46. Chicz, R. M. et al. Specificity and promiscuity among

    naturally processed peptides bound to HLA-DR alleles.

    J. Exp. Med. 178, 2747 (1993).

    47. Zhou, X., Caligiuri, G., Hamsten, A., Lefvert, A. K. &

    Hansson, G. K. LDL immunization induces T-cell-dependent antibody formation and protection against

    atherosclerosis.Arterioscler. Thromb. Vasc. Biol.

    21, 108114 (2001).

    48. Stemme, S. et al. T lymphocytes from human

    atherosclerotic plaques recognize oxidized LDL.Proc. Natl Acad. Sci. USA92, 38933897 (1995).

    49. Shaw, P. X. et al. Natural antibodies with the T15

    idiotype may act in atherosclerosis, apoptotic

    clearance, and protective immunity.J. Clin. Invest.

    105, 173117340 (2000).

    50. Binder, C. J. et al. Pneumococcal vaccination

    decreases atherosclerotic lesion formation: molecular

    mimicry between Streptococcus pneumoniaeand

    oxidized LDL. Nature Med. 9, 736743 (2003).

    51. Zhou, X., Nicoletti, A., Elhage, R. & Hansson, G. K.

    Transfer of CD4+T cells aggravates atherosclerosis

    in immunodeficient apolipoprotein E knockout mice.

    Circulation102, 29192922 (2000).

    52. Tupin, E. et al. CD1d-dependent activation of NKT

    cells aggravates atherosclerosis.J. Exp. Med.

    199, 417422 (2004).

    53. Melian, A., Geng, Y. J., Sukhova, G. K., Libby, P. &

    Porcelli, S. A. CD1 expression in human atherosclerosis.

    A potential mechanism for T cell activation by foam

    cells.Am. J. Pathol.155, 775786 (1999).

    54. Nakai, Y. et al. Natural killer T cells accelerate

    atherogenesis in mice. Blood104, 20512059

    (2004).

    55. Uyemura, K. et al. Cross-regulatory roles of interleukin

    (IL)-12 and IL-10 in atherosclerosis.J. Clin. Invest.97, 21302138 (1996).56. Frostegard, J. et al. Cytokine expression in advanced

    human atherosclerotic plaques: dominance of

    pro-inflammatory (Th1) and macrophage-stimulating

    cytokines.Atherosclerosis145, 3343. (1999).

    57. Gupta, S. et al. IFN-potentiates atherosclerosis inApoE knock-out mice.J. Clin. Invest.99, 27522761

    (1997).58. Buono, C. et al. Influence of interferon-on the extent

    and phenotype of diet-induced atherosclerosis in the

    LDLR-deficient mouse.Arterioscler. Thromb. Vasc.

    Biol.23, 454460 (2003).

    59. Davenport, P. & Tipping, P. G. The role of interleukin-4

    and interleukin-12 in the progression of

    atherosclerosis in apolipoprotein E-deficient mice.

    Am. J. Pathol.163, 11171125 (2003).

    60. Elhage, R. et al. Reduced atherosclerosis in

    interleukin-18 deficient apolipoprotein E-knockout

    mice. Cardiovasc. Res.59, 234240 (2003).

    61. Branen, L. et al. Inhibition of tumor necrosis

    factor-reduces atherosclerosis in apolipoprotein Eknockout mice.Arterioscler. Thromb. Vasc. Biol.

    24, 21372142 (2004).

    62. Buono, C. et al. T-bet deficiency reduces

    atherosclerosis and alters plaque antigen-specific

    immune responses. Proc. Natl Acad. Sci. USA

    102,15961601 (2005).

    63. Whitman, S. C., Ravisankar, P., Elam, H. &

    Daugherty, A. Exogenous interferon-enhancesatherosclerosis in apolipoprotein E/mice.

    Am. J. Pathol.157, 18191824 (2000).

    64. Laurat, E. et al. In vivodownregulation of T helper

    cell 1 immune responses reduces atherogenesis

    in apolipoprotein E-knockout mice. Circulation

    104, 197202 (2001).

    65. Paigen, B., Morrow, A., Brandon, C., Mitchell, D. &

    Holmes, P. Variation in susceptibility to atherosclerosis

    among inbred strains of mice.Atherosclerosis57,

    6573 (1985).

    66. Huber, S. A., Sakkinen, P., David, C., Newell, M. K. &

    Tracy, R. P. T helper-cell phenotype regulatesatherosclerosis in mice under conditions of mild

    hypercholesterolemia. Circulation103, 26102616

    (2001).

    67. Mallat, Z. et al. Interleukin-18/interleukin-18 binding

    protein signaling modulates atherosclerotic lesion

    development and stability. Circ. Res. 89, E41E45

    (2001).

    68. Whitman, S. C., Ravisankar, P. & Daugherty, A.

    Interleukin-18 enhances atherosclerosis in

    apolipoprotein E/mice through release of

    interferon-. Circ. Res. 90, e34e38 (2002).69. King, V. L., Szilvassy, S. J. & Daugherty, A.

    Interleukin-4 deficiency decreases atherosclerotic

    lesion formation in a site-specific manner in female

    LDL receptor/mice.Arterioscler. Thromb. Vasc. Biol.

    22, 456461 (2002).

    70. Shimizu, K., Shichiri, M., Libby, P., Lee, R. T. & Mitchell,

    R. N. Th2-predominant inflammation and blockade of

    IFN-signaling induce aneurysms in allografted aortas.

    J. Clin. Invest.114, 300308 (2004).71. Friesel, R., Komoriya, A. & Maciag, T. Inhibition of

    endothelial cell proliferation by -interferon.J. CellBiol. 104, 689696 (1987).

    72. Hansson, G. K., Hellstrand, M., Rymo, L., Rubbia, L. &

    Gabbiani, G. Interferon inhibits both proliferationand expression of differentiation-specific -smoothmuscle actin in arterial smooth muscle cells.

    J. Exp. Med. 170, 15951608 (1989).

    73. Amento, E. P., Ehsani, N., Palmer, H. & Libby, P.

    Cytokines and growth factors positively and negatively

    regulate interstitial collagen gene expression in

    human vascular smooth muscle cells.Arterioscler.

    Thromb.11, 12231230 (1991).

    74. van Hinsbergh, V. W. M., van den Berg, E. A., Fiers, W.

    & Dooijewaard, G. Tumor necrosis factor induces the

    production of urokinase-type plasminogen activator

    by human endothelial cells. Blood75, 19911998

    (1990).

    75. Lee, E. et al. Regulation of matrix metalloproteinases

    and plasminogen activator inhibitor-1 synthesis by

    plasminogen in cultured human vascular smooth

    muscle cells. Circ. Res.78, 4449 (1996).

    76. Saren, P., Welgus, H. G. & Kovanen, P. T. TNF-and IL-1selectively induce expression of 92-kDagelatinase by human macrophages.J. Immunol.

    157, 41594165 (1996).

    77. Jovinge, S. et al. Evidence for a role of tumor necrosis

    factor in disturbances of triglyceride and glucosemetabolism predisposing to coronary heart disease.

    Metabolism47, 113118 (1998).78. Boquist, S. et al.Alimentary lipemia, postprandial

    triglyceride-rich lipoproteins, and common carotid

    intima-media thickness in healthy, middle-aged men.

    Circulation100, 723728 (1999).

    79. Beutler, B. & Cerami, A. Cachectin and tumour

    necrosis factor as two sided of the same biological

    coin. Nature320, 584588 (1986).

    80. Mach, F., Schoenbeck, U., Bonnefoy, J.-Y., Pober, J. &

    Libby, P. Activation of monocyte/macrophage functions

    related to acute atheroma complication by ligation

    of CD40. Induction of collagenase, stromelysin, and

    tissue factor. Circulation96, 396399 (1997).

    81. Mach, F. et al. Functional CD40 ligand is expressed on

    human vascular endothelial cells, smooth muscle cells,

    and macrophages- implications for CD40CD40

    ligand signaling in atherosclerosis. Proc. Natl Acad.

    Sci. USA94, 19311936 (1997).

    82. Henn, V. et al. CD40 ligand on activated platelets

    triggers an inflammatory reaction of endothelial cells.

    Nature391, 591594 (1998).

    83. Mach, F., Schnbeck, U., Sukhova, G. K., Atkinson, E.

    & Libby, P. Reduction of atherosclerosis in mice by

    inhibition of CD40 signalling. Nature394, 200203

    (1998).

    84. Lutgens, E. et al. Requirement for CD154 in

    the progression of atherosclerosis. Nature Med.

    5, 13131316 (1999).

    85. Mallat, Z. et al. Protective role of interleukin-10

    in atherosclerosis. Circ. Res.85, e17e24 (1999).

    86. Pinderski Oslund, L. J. et al. Interleukin-10 blocks

    atherosclerotic events in vitroand in vivo.Arterioscler.

    Thromb. Vasc. Biol.19, 284753 (1999).87. Caligiuri, G. et al. Interleukin-10 deficiency increases

    atherosclerosis, thrombosis, and low-density

    lipoproteins in apolipoprotein E knockout mice.

    Mol. Med. 9, 1017 (2003).

    88. Grainger, D. J. et al. The serum concentration of active

    transforming growth factor- is severely depressed inadvanced atherosclerosis. Nature Med. 1, 7479

    (1995).

    89. Mallat, Z. et al. Inhibition of transforming growthfactor-signaling accelerates atherosclerosis andinduces an unstable plaque phenotype in mice.

    Circ. Res.89, 930934 (2001).

    90. Lutgens, E. et al. Transforming growth factor-mediates balance between inflammation and fibrosis

    during plaque progression.Arterioscler. Thromb. Vasc.

    Biol.22, 975982 (2002).

    91. Robertson, A. K. et al. Disruption of TGF-signalingin T cells accelerates atherosclerosis.J. Clin. Invest.

    112, 13421350 (2003).

    92. Gojova, A. et al. Specific abrogation of transforming

    growth factor-signaling in T cells altersatherosclerotic lesion size and composition in mice.

    Blood102, 40524058 (2003).

    93. Ait-Oufella, H. et al., Natural regulatory T cells control

    the development of atherosclerosis in mice. Nature

    Med.12, 178180 (2006).

    94. Salonen, J. T. et al.Autoantibody against oxidised

    LDL and progression of carotid atherosclerosis.

    Lancet339, 883887 (1992).95. Fredrikson, G. N. et al. Identification of immune

    responses against aldehyde-modified peptide

    sequences in apoB associated with cardiovascular

    disease.Arterioscler. Thromb. Vasc. Biol.23, 872878

    (2003).

    96. Shaw, P. X. et al. Human-derived anti-oxidized LDL

    autoantibody blocks uptake of oxidized LDL

    by macrophages and localizes to atherosclerotic

    lesions in vivo.Arterioscler. Thromb. Vasc. Biol.

    21, 13331339 (2001).

    97. Kol, A., Sukhova, G. K., Lichtman, A. H. & Libby, P.

    Chlamydial heat shock protein 60 localizes in human

    atheroma and regulates macrophage tumor necrosis

    factor-and matrix metalloproteinase expression.Circulation98, 300307 (1998).

    98. Xu, Q. et al.Association of serum antibodies to

    heat-shock protein 65 with carotid atherosclerosis.

    Lancet341, 255259 (1993).

    R E V I E W S

    518 |JULY 2006 |VOLUME 6 www.nature.com/reviews/immunol

    2006Nature Publishing Group

  • 8/12/2019 The Immune Response In

    12/12

    99. Perschinka, H. et al. Cross-reactive B-cell epitopes

    of microbial and human heat shock protein 60/65

    in atherosclerosis.Arterioscler. Thromb. Vasc. Biol.

    23,10601065 (2003).

    100. Caligiuri, G., Nicoletti, A., Poirier, B. & Hansson, G. K.

    Protective immunity against atherosclerosis carried

    by B cells of hypercholesterolemic mice.J. Clin. Invest.

    109, 745753 (2002).

    101. Libby, P. & Aikawa, M. Stabilization of atherosclerotic

    plaques: new mechanisms and clinical targets.

    Nature Med. 8, 12571262 (2002).

    102. Henney, A. M. et al. Localization of stromelysingene expression in atherosclerotic plaques byin situ hybridization. Proc. Natl Acad. Sci. USA

    88, 81548158 (1991).

    103. Galis, Z. S., Sukhova, G. K., Lark, M. W. & Libby, P.

    Increased expression of matrix metalloproteinases

    and matrix degrading activity in vulnarable regions

    of human atherosclerotic plaques.J. Clin. Invest.

    94, 24932503 (1994).104. Dollery, C. & Libby, P. Atherosclerosis and

    proteinase activation. Cardiovasc. Res.69, 625635

    (2006).

    105. Buchner, K. et al. CD40 ligand is selectively expressed

    on CD4+T cells and platelets: implications for

    CD40CD40L signalling in atherosclerosis.J. Pathol.

    201, 288295 (2003).

    106. Poon, M., Badimon, J. J. & Fuster, V. Overcoming

    restenosis with sirolimus: from alphabet soup

    to clinical reality. Lancet359, 619622 (2002).107. Jonasson, L., Holm, J. & Hansson, G. K. Cyclosporin A

    inhibits smooth muscle proliferation in the vascular

    response to injury. Proc. Natl Acad. Sci. USA

    85, 23032306 (1988).

    108. Randomised trial of cholesterol lowering in

    4444 patients with coronary heart disease:

    the Scandinavian Simvastatin Survival Study (4S).Lancet344, 13831389 (1994).

    109. Ridker, P. M. et al. C-reactive protein levels and

    outcomes after statin therapy. N. Engl. J. Med.

    352, 2028 (2005).

    110. Greenwood, J., Steinman, L. & Zamvil, S.S. Statin

    therapy and autoimmune disease: from protein

    prenylation to immunomodulation. Nature Rev.

    Immunol.6, 358370

    111. Takemoto, M. & Liao, J. K. Pleiotropic effects of

    3-hydroxy-3-methylglutaryl coenzyme a reductase

    inhibitors.Arterioscler. Thromb. Vasc. Biol.

    21, 17121719 (2001).

    112. Kwak, B., Mulhaupt, F., Myit, S. & Mach, F. Statins

    as a newly recognized type of immunomodulator.

    Nature Med. 6, 13991402 (2000).

    113. Mehra, M. R. & Raval, N. Y. Metaanalysis of statinsand survival in de novocardiac transplantation.Transplant. Proc.36, 15391541 (2004).

    114. McCarey, D. W. et al. Trial of atorvastatin in rheumatoid

    arthritis (TARA): double-blind, randomised placebo-

    controlled trial. Lancet363, 20152021 (2004).

    115.Youssef, S. et al. The HMG-CoA reductase inhibitor,

    atorvastatin, promotes a Th2 bias and reverses

    paralysis in central nervous system autoimmune

    disease. Nature420, 7884 (2002).

    116. Marx, N. et al. PPAR activators as antiinflammatory

    mediators in human T lymphocytes: implications

    for atherosclerosis and transplantation-associated

    arteriosclerosis. Circ. Res. 90, 703710 (2002).

    117. Staels, B. et al.Activation of human aortic smooth-muscle cells is inhibited by PPARbut not by PPARactivators. Nature393, 790793 (1998).

    118. Fitzgerald, G. A. Coxibs and cardiovascular disease.

    N. Engl. J. Med. 351, 17091711 (2004).

    119. Solomon, S. D. et al. Cardiovascular risk associated

    with celecoxib in a clinical trial for colorectal adenoma

    prevention. N. Engl. J. Med. 352, 10711080 (2005).

    120. Mattner, J. et al. Exogenous and endogenous

    glycolipid antigens activate NKT cells during microbial

    infections. Nature434, 525529 (2005).

    121. Liuzzo, G. et al. The prognostic value of C-reactive

    protein and serum amyloid A protein in severe unstable

    angina. N. Engl. J. Med. 331, 417424 (1994).

    122. Ridker, P. M., Hennekens, C. H., Buring, J. E. &

    Rifai, N. C-reactive protein and other markers of

    inflammation in the prediction of cardiovascular

    disease in women. N. Engl. J. Med. 342, 836843

    (2000).123. Danesh, J. et al. C-reactive protein and other circulating

    markers of inflammation in the prediction of coronary

    heart disease. N. Engl. J. Med. 350, 13871397

    (2004).

    124. Palinski, W., Miller, E. & Witztum, J. L. Immunization

    of low density lipoprotein (LDL) receptor-deficient

    rabbits with homologous malondialdehyde-modified

    LDL reduces atherogenesis. Proc. Natl Acad. Sci. USA

    92, 821825 (1995).

    125.Ameli, S. et al. Effect of immunization with

    homologous LDL and oxidized LDL on early

    atherosclerosis in hypercholesterolemic rabbits.

    Arterioscler. Thromb. Vasc. Biol.16, 10741079

    (1996).126. George, J. et al. Hyperimmunization of apo-E-deficient

    mice with homologous malondialdehyde low-density

    lipoprotein suppresses early atherogenesis.

    Atherosclerosis138, 147152 (1998).

    127. Freigang, S., Hrkk, S., Miller, E., Witztum, J. L. &

    Palinski, W. Immunization of LDL receptor-deficient

    mice with homologous malondialdehyde-modified

    and native LDL reduces progression of atherosclerosis

    by mechanisms other than induction of high titers ofantibodies to oxidative neoepitopes.Arterioscler.

    Thromb. Vasc. Biol.18, 19721982 (1998).

    128. Fredrikson, G. N. et al. Inhibition of atherosclerosis

    in apoE-null mice by immunization with apoB-100

    peptide sequences.Arterioscler. Thromb. Vasc.

    Biol.23, 879884 (2003).

    129. Zhou, X., Robertson, A. K., Hjerpe, C. &

    Hansson, G. K. Adoptive transfer of CD4+T cells

    reactive to modified low-density lipoprotein

    aggravates atherosclerosis.Arterioscler. Thromb.

    Vasc. Biol.26, 864870 (2006).

    130.Afek, A. et al. Immunization of low-density

    lipoprotein receptor deficient (LDL-RD) mice with

    heat shock protein 65 (HSP-65) promotes earlyatherosclerosis.J. Autoimmun. 14, 115121

    (2000).

    131. Harats, D., Yacov, N., Gilburd, B., Shoenfeld, Y. &

    George, J. Oral tolerance with heat shock protein 65

    attenuates Mycobacterium tuberculosis-induced and

    high-fat-diet-driven atherosclerotic lesions.J. Am. Coll.

    Cardiol.40, 13331338 (2002).

    132. Maron, R. et al. Mucosal administration of heat shock

    protein-65 decreases atherosclerosis and

    inflammation in aortic arch of low-density lipoprotein

    receptor-deficient mice. Circulation106, 17081715

    (2002).

    133. George, J. et al. Induction of early atherosclerosis

    in LDL-receptor-deficient mice immunized with

    2-glycoprotein I. Circulation98, 11081115(1998).

    AcknowledgementsWe regret that we have not been able to cite many important

    papers owing to space limitations. Our research is supported

    by grants from the Swedish Research Council, Heart-Lung

    Foundation, European Community, US National Institutes of

    Health and Leducq Foundation.

    Competing interests statementThe authors declare no competing financial interests.

    DATABASESThe following terms in this article are linked online to:

    Entrez Gene:http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=gene

    APOE | CCL2 | CD14 | CD40 | CD40L | CD68 | CX3CR1 |

    HSP60 | IFN | IL-4 | LDLR | MMP1 | PPAR | PPAR | TLR4 | TNF |VCAM1

    FURTHER INFORMATIONPeter Libbys homepage:

    http://reynolds.brighamandwomens.org/faculty/libby.asp

    Gran K. Hanssons homepage:

    http://www.ki.se/medicin/medicine_ks/experimental_

    cardiovascular_research_unit/index_en.html

    Access to this links box is available online.

    R E V I E W S

    NATURE REVIEWS | IMMUNOLOGY VOLUME 6 | JULY 2006 | 519