Skip to main content
Erschienen in: Journal of Inflammation 1/2011

Open Access 01.12.2011 | Review

Innate immunity and monocyte-macrophage activation in atherosclerosis

verfasst von: Joseph Shalhoub, Mika A Falck-Hansen, Alun H Davies, Claudia Monaco

Erschienen in: Journal of Inflammation | Ausgabe 1/2011

Abstract

Innate inflammation is a hallmark of both experimental and human atherosclerosis. The predominant innate immune cell in the atherosclerotic plaque is the monocyte-macrophage. The behaviour of this cell type within the plaque is heterogeneous and depends on the recruitment of diverse monocyte subsets. Furthermore, the plaque microenvironment offers polarisation and activation signals which impact on phenotype. Microenvironmental signals are sensed through pattern recognition receptors, including toll-like and NOD-like receptors thus dictating macrophage behaviour and outcome in atherosclerosis. Recently cholesterol crystals and modified lipoproteins have been recognised as able to directly engage these pattern recognition receptors. The convergent role of such pathways in terms of macrophage activation is discussed in this review.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1476-9255-8-9) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

All authors were involved directly in the drafting and critical revision of this review. CM is the senior author with overall responsibility for this work, giving the final approval for publication. All authors have read and approved the final manuscript.

Introduction

Atherothrombotic vascular disease is quickly becoming the leading cause of mortality worldwide, accounting for a fifth of all deaths [1]. The manifestations of the disease are often sudden and dramatic, including myocardial infarction and sudden death. Cerebrovascular atherothrombosis is responsible for ischaemic stroke, a major source of disability and dependence, and represents a rising health-economic burden [2].
Progress has been made in refining our understanding of the process of inflammation which underlies atherosclerosis since the early descriptions by Rudolf Virchow during the 19th century [3, 4] and subsequently Russell Ross in the late 1990s [58]. The development of an atherosclerotic plaque begins with the recruitment of blood-borne inflammatory cells at sites of lipid deposition [9] or arterial injury [5]. Local rheological factors, such as low and oscillatory (with vortices) blood-to-wall shear stress dictate the location of atherosclerotic plaques to characteristic points along the vasculature [10, 11].
Atherosclerosis shares features with diseases caused by chronic inflammation [7]. Inflammation is intrinsically linked with disease activity, as the numbers of monocyte-macrophages infiltrating the plaque [12] and their location at plaque rupture-sensitive sites (such as the fibrous cap and areas of erosion [13, 14]) is related to plaque vulnerability. Moreover, lymphocyte abundance and their activation markers relate to plaque activity [13]. Macrophage differentiation is acknowledged as critical for the development of atherosclerosis [15]. The intimate relationship between atherosclerosis and inflammation is further exemplified by the involvement of cytokines and chemokines at all stages of the process of atherosclerosis (reviewed in detail by [16]). The extent of the inflammatory infiltrates and their strategic location within the protective fibrous cap is associated with plaque rupture and/or thrombosis [17]. Adventitial inflammation has also been described [18], and is linked with an expansion of the adventitial vasa vasorum in unstable atherosclerosis [19]. The inflammatory nature of atherosclerosis is supported by the association between circulating plasma inflammatory markers, particularly C-reactive protein, with cardiovascular outcomes, even in the absence of dyslipidaemia [6]. Further evidence for a link between systemic inflammation and cardiovascular disease is the increased incidence of cardiovascular events in chronic inflammatory conditions, such as inflammatory arthritis and systemic lupus erythematosus [7, 8]. The expanding knowledge base regarding inflammation in atherosclerosis has resulted in a keen interest in targeted therapeutics and functional imaging tools for the high-risk atherosclerotic plaque [20].

Innate immunity is a key player in atherosclerosis

How is inflammation established and maintained within an atherosclerotic plaque? Inflammation in physiological conditions is a self-limiting ancient protective mechanism that defends the host from invading pathogens. It relies on two arms: innate immunity and adaptive immunity. Innate immunity is activated immediately upon encounter with the pathogen and is executed primarily by myeloid cells with the participation of some "innate" lymphocyte sub-populations. Adaptive immunity is a second line of defence that is based upon the generation of antigen-specific recognition apparatus at cellular (T cell receptor) and humoral (antibody) levels.
In the past decade it has become apparent that the innate arm of the immune inflammatory response is not merely a concoction of non-specific responses and phagocytosis. Rather it is the main orchestrator of the subsequent adaptive responses and is able to sense pathogen associated molecular patterns (PAMPs) with a specificity which was previously unsuspected. In inflammatory conditions, including atherosclerosis, the immune inflammatory apparatus is chronically activated, either due to the persistence of pro-inflammatory stimuli or due to the failure of regulatory mechanisms that should facilitate resolution. Significant progress has been made in the field linking innate immune sensors to the recognition of cholesterol [21] and modified lipoproteins [2224]. Thus diverse innate immune signalling pathways have been seen to cooperate to induce and maintain inflammation upon exposure to exogenous and, importantly, endogenous molecular patterns [21, 25].
The most abundant cell types within the atherosclerotic plaque are innate immune cells, such as monocyte-macrophages, dendritic cells (DCs) and mast cells. Monocytes-macrophages came to the forefront of research owing to new awareness that they may represent a more heterogeneous and phenotypically plastic population than previously anticipated. In this review we focus on the role of macrophage activation and phenotypic polarisation in lesion formation and vulnerability.

Macrophage heterogeneity in atherosclerosis

Macrophages are a heterogeneous population of cells that adapt in response to a variety of micro-environmental signals; their phenotype is very much a function of environmental cues [26, 27]. In a nomenclature mirroring Th1 and Th2 polarisation, macrophages are usually defined as M1 or M2 [28]. Classically activated (M1) macrophages were the first to be defined [29, 30] as pro-inflammatory. Alternatively activated (M2) macrophages have been originally characterised in the context of Th2-type immune responses [29]. Subsets of M2-like macrophages have been later found to contribute to wound healing and regulation of inflammatory processes [31]. Characteristic cytokine and chemokine signatures pertaining to human monocyte-to-macrophage differentiation and M1/M2 macrophage polarisation (Table 1) have been described [28, 32].
Table 1
Cytokine and chemokine genes, and those of receptors (in italics), known to be differentially transcribed in human M1 and M2 macrophage in vitro polarisation (Adapted from [28] and [27]).
M1 > M2
M2 > M1
CXCL11
Insulin-like growth factor 1
CCL19
CCL23
CXCL10
CCL18
Tumour necrosis factor ligand superfamily, member 2
CCL13
CCL15
Bone morphogenic protein 2
Interleukin 12B
Hepatocyte growth factor
Interleukin 15
Fibroblast growth factor 13
Tumour necrosis factor ligand superfamily, member 10
CXCL1
Interleukin 6
Transforming growth factor β receptor II
CCL20
CXCR4
Visfatin
Mannose receptor C type 1 (CD206)
Endothelial cell growth factor
 
CCL1
 
CCL17
 
CCL22
 
CCL13
 
Transforming growth factor β2
 
CCR7
 
Interleukin 2 receptor α chain
 
Interleukin 15 receptor α chain
 
Interleukin 7 receptor
 
CCL2 was upregulated in M-CSF differentiated macrophages in one study [27], whilst relatively increased by GM-CSF in another [28].
Macrophage phenotypic polarisation may have a role in the fate of an atherosclerotic plaque. The plaque is an environment with a strong skew towards Th1 lymphocytic responses, resulting in high levels of IFNγ [33, 34] which could in theory privilege M1-type macrophage polarisation. However, studies thus far have demonstrated macrophage heterogeneity within atherosclerosis, supporting that both M1 and M2 macrophages are present in human and murine atherosclerotic lesions. In an ApoE-/- murine model of atherosclerosis, early lesions were seen to be infiltrated by M2 (arginase I+) macrophages [35]. As lesions progressed a phenotypic switch was observed, with an eventual predominance of M1 (arginase II+) macrophages. Upon exposure to the oxidised phospholipid 1-palmitoyl-2-arachidonoyl-sn-3-phosphorylcholine (oxPAPC), murine macrophages adopted a previously undescribed phenotype (Figure 1) [36]. A reduction in the expression of genes characteristic of both M1 and M2, coupled with an up-regulation of a unique redox gene signature that includes haemoxygenase 1, was observed. This population, termed Mox macrophages, are nuclear factor erythroid 2-like 2 (Nrf2)-dependent and have been shown to comprise approximately 30% of all macrophages in advanced atherosclerotic lesions of LDLR-/- mice [36]. A variety of subtypes have been described which are considered to fall under the umbrella of alternatively activated M2 macrophages (reviewed in [31, 37]). An example of this occurs with administration of IL33 (which is functionally atheroprotective [38]) to genetically obese diabetic (ob/ob) mice, resulting in increased production of Th2 cytokines and polarisation of adipose tissue macrophages to a CD206+ M2 phenotype [39].
In human lesions different macrophage phenotypes exist, and do so in different plaque locations. M2 (CD68+ CD206+) macrophages were seen to reside in areas more stable zones of the plaque distant from the lipid core, with their M1 (CD68+ CCL2+) counterparts displaying a distinct tissue localisation pattern [40]. Subsequent work has confirmed this, finding CD68+ CD206+ cells far from the lipid core [41]. CD68+ CD206+ macrophages were also seen to contain smaller lipid droplets compared to CD68+ CD206-[41]. A subset of M2 macrophages has recently been detected in association with intraplaque haemorrhage in coronary atheromata [42]. These macrophages express high levels of CD163 (a scavenger receptor that binds to haemoglobin-haptoglobin (HbHp) complexes). They also express low levels of MHC Class II and display low release of the reactive oxidative species hydrogen peroxide. Expression of CD163 by peripheral blood monocytes was not shown to be different between the CD14+ CD16+ and CD14++ CD16- subsets. However, when monocytes were differentiated into macrophages in the presence of HbHp complexes for 8 days, they matured into a CD163high HLA-DRlow phenotype similar to the haemorrhage-associated macrophages within coronary plaques [42]. Differentiation into this macrophage subtype was dependent on the expression of CD163 and IL10 during in vitro blockade experiments. Interestingly, this polarisation was prevented by the incubation with specific inhibitors of endolysosomal acidification, such as chloroquine which is known to interfere with endosomal TLR signalling [42].
Lesion development and stability are not only determined by the influx and differentiation of inflammatory cell subsets, but also their ability to act on vascular extracellular matrix. Importantly, the macrophage subtypes display a differential expression of matrix metalloproteinase (MMP) and tissue inhibitor of metalloproteinase (TIMP) [43]. In particular, a subset of lesional foam cell macrophages characterised by a high expression of MMP14 (membrane type 1 MMP) and a low expression of TIMP3 were highly invasive and catabolic [44]. Moreover, such expression pattern of MMP14 and TIMP 3 was associated with markers of M1 polarisation [44], whilst expression of MMP12 was associated with an M2-typical down-regulation of arginase I [45]. Thus MMP expression by macrophage subsets is also heterogeneous, further highlighting the different functionalities of these cells.
The heterogeneity of macrophage phenotypes in the various studies is an important feature of our current view of atherosclerosis. Studies assessing multiple markers in human and murine lesions are needed to map such degree of heterogeneity. How is such heterogeneity generated? It is likely to be the result of recruitment of different monocytes subsets, or stimuli provided by the plaque microenvironment. Gordon and Martinez have proposed a four-stage paradigm of macrophage activation, where differentiation through exposure to growth factors is the first stage [46]. This stage is followed by priming (through cytokines, particularly IFNγ and IL4), activation (by TLR or similar), and finally resolution and repair (mediated by IL10, transforming growth factor (TGF)-β, nucleotides, glucocorticoids or lipotoxins) [46]. This review will explore the potential mechanisms leading to macrophage activation and polarisation in atherosclerosis.

Recruitment of monocyte subsets to atherosclerotic plaques

In both mice and humans, monocytes comprise 5 to 10% of peripheral blood leukocytes [25]. Two major circulating monocyte subsets have been described in humans and mice alike, the distinction made on the basis of size, granularity, and the differential expression of chemokine receptors and adhesion molecules [47]. The two mouse monocyte sub-populations are represented approximately equally in murine blood; they are distinguished based upon their expression of CCR2, CX3CR1 and Ly6C [48]) [49]. CCR2+ CX3CR1low Ly6C+ monocytes are termed 'inflammatory' monocytes, and CCR2- CX3CR1high Ly6C- are referred to as 'resident' monocytes [31, 47, 50].
Similarly to mouse monocytes, human monocytes can be separated into two groups based upon cell surface CD14 - a toll-like receptor (TLR) co-receptor sensing exogenous molecular patterns such as lipopolysaccharide (LPS) - and CD16 - a member of the family of Fc (Fragment, crystallisable) receptors FcγRIII. In humans, about 90% of monocytes are CD14++ CD16- and termed 'classical' monocytes [50, 51]. CD14+ CD16+ monocytes, which constitute the remaining minority, are referred to as 'non-classical' [5255] (Table 2).
Table 2
A comparison of human and murine monocyte subsets, highlighting differences in surface receptor phenotypes.
 
Human
Mouse
Classical/Inflammatory
CD14++ CD16- [195, 196]
(>90%)
Ly6C+ CCR2+ CD62L+ CX3CR1low [47, 59] (~50%)
Non-Classical/Resident
CD14+ CD16+ [195, 196]
(<10%)
Ly6C- CCR2- CD62L- CX3CR1high [47, 59] (~50%)
The approximate abundance in peripheral blood is shown in brackets, however this may not reflect the proportions in other sites such as the spleen.
To date, monocyte phenotype data has centred largely on the murine system [29]. Similarities between mice and humans may be accounted for, at least in part, by the expression of surface receptors. For instance, chemokine receptors CCR1 and CCR2 are highly expressed on both CD16- human and Ly6C+ murine monocytes, and CX3CR1 is increased on CD16+ human and Ly6C- mouse monocytes [47, 56, 57] (reviewed in [58]). More than 130 of these gene expression differences were conserved between mouse and human monocyte subsets, with many of these differences also confirmed at the protein level [59]. A notable difference among these was the high expression of peroxisome proliferator-activated receptor γ (PPARγ, discussed in greater detail below) in Ly6C- mouse monocytes, but not the proposed CD16+ counterpart [59]. As such, the differences between mouse and human monocyte subsets may be greater than had been expected and may be difficult to reconcile.
Two groups independently reported in 2007 that the Ly6C+ inflammatory monocyte subset increases its representation dramatically in the peripheral blood of the hypercholesterolemic apolipoprotein E (ApoE) deficient mouse on a high-fat diet [56, 60]. Conversely, hypercholesterolemia did not affect Ly6C- monocytes and also discouraged the conversion of Ly6C+ into Ly6C- monocytes. Other mechanisms proposed for this increase in Ly6C+ monocytes during hypercholesterolemia include increased proliferation and reduced apoptosis [61]. Ly6C+ monocytes are recruited to activated endothelium and are thought to represent the majority of infiltrating macrophages within atherosclerotic plaques [60]. Conversely, Ly6C- enter the atherosclerotic plaque in lower numbers and preferentially express CD11c upon entry [56]. This differential recruitment based upon Ly6C expression may condition the macrophage phenotype within the plaque, with reports that Ly6C+ monocytes differentiate into cells that resemble M1 macrophages and that cells derived from Ly6C- monocytes exhibit M2 characteristics [6265].
Chemokine receptors are necessary for monocytes to traverse the endothelium [56, 66] (reviewed in [16]). CX3CR1-/- (fractalkine receptor) [67, 68], CX3CL1-/- (fractalkine) [69] and CCR2-/-[70, 71] mice (in the context of low density lipoprotein receptor (LDLR) or ApoE deficiency) exhibited a reduction in - but not elimination of - atherosclerosis. Furthermore, deficiency of CCR5 (the receptor for CCL5, a chemokine also known as RANTES) in ApoE-/- mice does not appear to be protective in the early stages of atherosclerosis [72]. Subsequently, in a wire injury study also using the ApoE-/- mouse model, the authors found a significant reduction in the area neo-intima formation with concurrent CCR5 deficiency, but not with concurrent absence of the alternative CCL5 receptor CCR1 [73]. More recently, a multiple knockout model has reaffirmed the thinking that CCL2 (MCP1), CCR5 and CX3CR1 play independent and additive roles in atherogenesis [74]. Combined inhibition of CCL2, CCR5 and CX3CR1 in ApoE-/- mice results in a 90% reduction in atherosclerosis, which is related to progressive monocytopaenia [66, 74]. However, chemokine receptor utilisation during recruitment to atherosclerotic plaques differentiates Ly6C+ and Ly6C- monocytes. Ly6C+ monocytes are recruited to mouse atherosclerosis via CCR2, CCR5 and CX3CR1 [61]. Conversely, Ly6C- monocytes are recruited less frequently and through CCR5.
In human atherosclerosis, patients with coronary artery disease have increased numbers of circulating CD14+ CD16+ monocytes compared to controls [75]. Furthermore, these patients have raised levels of serum TNFα [76]. There is, however, data to the contrary with the finding that inflammatory genes and surface markers were down-regulated in monocytes of patients with coronary atherosclerosis [77]. Of relevance, CD14+ CD16+ monocytes have also been shown to exhibit pro-inflammatory and pro-atherosclerotic activity in a population of elderly human subjects. These activated monocytes exhibited increased interaction with endothelium and had higher expression of chemokine receptors [78]. Other studies have suggested that the bone marrow is the source of these monocytes [79, 80].

Macrophage differentiation in atherosclerosis

Early work relating to the effect of the colony stimulating factors (CSFs) on macrophage phenotype was undertaken by Hamilton and colleagues [81, 82]. A variety of groups have generated data using monocytes differentiated in vitro, via exposure to either M-CSF or GM-CSF [82, 83]. In vitro differentiation with M-CSF results in a macrophage phenotype close to that of M2 [28]. GM-CSF plays a role in the induction of a pro-inflammatory macrophage phenotype that resembles M1 polarisation, proficiently producing inflammatory cytokines such as TNFα and IL6, and being involved in tissue destruction [28].
In further murine studies, both M-CSF and GM-CSF have been shown to be important in plaque development. Smith et al studied ApoE-/- mice crossbred with the osteopetrotic mutation of the M-CSF gene. These mice were fed a low-fat chow diet with the double mutants exhibiting significantly smaller proximal aortic lesions, at an earlier stage of progression and with fewer macrophages as compared with their control ApoE-/- littermates [84]. The production of GM-CSF from smooth muscle cells leads to the activation of monocytes during atherogenesis [85]. In another study using the hypercholesterolaemic ApoE-/- mouse, animals on a high-fat diet were injected with doses of 10 μg/kg GM-CSF or G-CSF daily for 5 days on alternating weeks for a total of 20 doses during an 8 week period, finding that both G-CSF and GM-CSF treatment resulted in increased atherosclerotic lesion extent [86]. LDLR-null mice have been employed in a study which combined 5-bromo-2'-deoxyuridine pulse labelling with en face immunoconfocal microscopy to demonstrate that systemic injection of GM-CSF markedly increased intimal cell proliferation, whilst functional GM-CSF blockade inhibited proliferation [87].
In a key study, Waldo and colleagues examined human macrophages differentiated in vitro for 7 days with either M-CSF or GM-CSF [27]. They characterised gene expression, surface phenotype, cytokine production and lipid handling in these two macrophage groups. With regards to gene expression, they demonstrated differential expression of genes of inflammation (Table 1) and cholesterol homeostasis between the two groups, including that GM-CSF macrophages exhibited a ten-fold increased gene expression of PPARγ. M-CSF differentiated macrophages spontaneously accumulated cholesterol when incubated with unmodified low density lipoprotein (LDL), whilst GM-CSF differentiated macrophages took up similar levels only when exposed to protein kinase C. Macrophages differentiated with M-CSF were shown by immunofluoresence to express CD14 (CD68+ CD14+), whilst GM-CSF differentiated macrophages were CD68+ CD14-. Interestingly, human coronary plaque samples were shown to contain predominantly CD68+ CD14+[27].

Priming of macrophages in the atherosclerotic plaque

Macrophages are M1-primed by exposure to interferon (IFN)-γ [37]. The key role of IFNγ [88] has been confirmed in experimental atherosclerosis whereby ApoE-/- IFNγ receptor-/- mice displayed a substantial reduction in lesion size compared to ApoE-/-[89]. This reduction was manifest alongside a reduced level of macrophages and T lymphocytes within the lesions. Furthermore, murine cardiac allografts sited in IFNγ-/- recipients had reduced transplant atherosclerosis [90].
Alternative M2 polarisation has originally been described as the result of exposure to interleukin (IL4) [28, 40, 58, 91]. M2 macrophages have a notable role in catabasis, the process inflammation resolution which when fails results in progression of atherosclerosis [92].
Wound healing macrophages, concerned primarily with tissue repair, are similar to the alternatively activated (M2) macrophages which have been described above. Wound healing macrophages establish their phenotype upon exposure to IL4 and/or IL13 from Th2 cells and granulocytes. IL4 is an early innate signal released during tissue injury, stimulating macrophage arginase to convert arginine to ornithine which is a step in extra-cellular matrix collagen production [93]. This ornithine is a precursor for polyamines which have an effect on cytokine production, affording wound healing macrophages regulatory capabilities [94].
Regulatory macrophages, with anti-inflammatory activity, are most reliably defined and identified through IL10 levels or IL10/IL12 ratio (as they also downregulate IL12 [95]). These develop in response to a large number of stimuli, including IL10 produced by regulatory T cells, TGFβ [96], and glucocorticoids. The latter attenuate macrophage-mediated inflammation through inhibition of pro-inflammatory cytokine gene transcription [97], nonetheless capacity for phagocytosis does not appear to be impaired by glucocorticoids [98]. Unlike wound-healing macrophages, regulatory macrophages do not contribute to the production of extracellular matrix.

Macrophage activation pathways in atherosclerosis

Following the priming stage, activation of macrophages is reliant upon ligation of pattern recognition receptors (PRR) [29, 99], namely nucleotide-binding oligomerisation domain (NOD)-like receptors (NLRs) and TLRs.

Toll-like receptor signalling

TLRs are the most well-characterised PRRs, of which at least ten have been identified in humans [100]. TLRs may be found on the cell surface, as in the case of TLRs 1, 2, 4, 5 and 6, or reside intracellularly [101, 102]. TLRs are key activators of monocytes and macrophages.
Upon exposure to ligand, TLRs couple to signalling adaptors to induces two major downstream signalling pathways: the nuclear factor kappa B (NFκB) (Figure 2) and the interferon response factor (IRF) pathways. MyD88 is a universal adapter protein that carries signalling through all TLRs, except TLR3, leading to the activation of NFκB. MyD88-dependent signalling relies on recruitment of Mal (MyD88-adaptor like), which leads to the recruitment of the IL1 receptor-associated kinase (IRAK). Phosphorylation of IRAK signals to tumour-necrosis-factor-receptor-associated factor 6 (TRAF6). The subsequent nuclear translocation of NFκB and translation of inflammatory cytokines is driven by phosphorylation of the IκB kinase (IKK) complex upon activation of TRAF6. MyD88-independent signalling is via TRAM (TRIF-related adaptor molecule) and TRIF (TIR-domain-containing adaptor protein inducing IFNβ), and can activate both NFκB and IRF, inducing interferon synthesis. The importance of IL1/TLR signalling in atherosclerosis has been further highlighted by work implicating IRAK4 kinase in modified LDL-medicated experimental atherosclerosis [103].
The most characterised recognition system is the one sensing LPS. Serum LPS-binding protein (LBP) transfers LPS to CD14, which delivers it to the co-receptor MD2 [104, 105]. The availability of all members of the complex dictates the sensitivity of recognition of endotoxin at extremely low concentrations. Cells that do not express CD14, such as endothelial cells, are relatively unresponsive compared to CD14+ monocytes [104, 105]. CD14 acts as a co-receptor (along with TLR4 and MD2) for the detection of bacterial LPS. CD14, however, can only bind LPS in the presence of LBP. TLR2 may also be activated via scavenger co-receptors, including CD36 [106].

Toll-like receptor agonists

Initially, ligands binding to PRRs such as TLRs on/in innate immune cells were believed to be of a pathogenic aetiology; molecules or small molecular motifs derived from, conserved within or associated with groups of microorganisms (such as bacterial LPS). These have been nominated pathogen associated molecular patterns (PAMPs). More recently, such ligands have been classified as danger associated molecular patterns (DAMPs) encompassing a wider definition which embodies the existence of endogenous danger signals. The concept that oxidation reactions involving lipids, proteins and DNA produce non-microbial 'oxidation-specific epitopes' has emerged [107]. Of particular interest is that host-derived oxidation-specific epitopes represent endogenous DAMPs, are recognised by PRRs and are capable of driving the inflammation seen in atherosclerosis [107].
DAMPs that may bind TLRs are numerous, some of which have been proposed as endogenous culprits in atherosclerosis. Examples of endogenous ligands to TLR2 include necrotic cell products [108], apolipoprotein CIII [109], serum amyloid A [110], versican [111]. Furthermore, oxidised phospholipids, saturated fatty acids, and lipoprotein A have been shown to trigger macrophage apoptosis, under conditions of thapsigargin-induced endoplasmic reticulum stress, via a mechanism requiring both CD36 and TLR2 [112].
Hyaluronan fragment [113], biglycan [114], oxLDL [115, 116] and heat shock proteins [117] have been shown to act through both TLR2 and TLR4. Long surfactant protein A [118], tenascin C [119], fibrinogen [120], fibronectin EDA [121], heparan sulphate [122], β-defensin 2 [123], amyloid β peptide [24] and minimally modified LDL (mmLDL) [23] act via TLR4 alone. TLR3 detects mRNA [124, 125], whilst TLR7 and TLR9 detect nucleic acid-containing immune complexes [126, 127]. TLRs 5, 6 and 8 are yet to have endogenous ligands allocated to them [25].
Although both mmLDL and oxLDL are seen as ligands to TLR4, the pathways by which recognition occurs differ. The recognition of mmLDL is similar to that of LPS and involves CD14 and MD2 [22], whilst oxLDL initiates inflammatory responses through a TLR4/TLR6 heterodimer in association with CD36 but independently of CD14 [128]. A lipidic component of LDL, namely oxPAPC, has been shown as capable of inducing IL8 transcription via TLR4 in a manner which is independent of both CD14 and CD36 [129]. Further work, however, has seen oxPAPC inhibiting TLR4-dependent IL8 induction, along with inhibition of E-selectin and CCL2, whilst IL1β and TNFα signalling remained unhindered [130]. Downstream of TLR4/MD2/CD14, intracellular signalling in response to mmLDL stimulation has been investigated and, in addition to the canonical MyD88 pathway, an alternative pathway via sequential activation of spleen tyrosine kinase (Syk), phospholipase Cγ1, protein kinase C, and NADPH oxidase 2 (gp91phox/Nox2) has been proposed in the stimulation of pro-inflammatory cytokine production and the effects thereof [131].

Toll-like receptor expression in atherosclerosis

TLRs are differentially expressed by the various cell types in atherosclerosis, with TLR2 and TLR4 found on monocytes, macrophages, foam cells and myeloid DCs, as well as smooth muscle cells and B lymphocytes (reviewed by [25]). Human and mouse atherosclerosis is characterised by an increased expression of TLR1, TLR2 and TLR4 (and to some extent TLR5), mainly by macrophages and endothelial cells [116, 132]. In mouse atherosclerosis, TLR4 expression is exclusively by macrophages [116]. There has been shown to be co-localisation of p65 (an NFκB family member) with both TLR2 and TLR4 in macrophages in atherosclerosis [132].
The differential expression of the various TLRs by monocyte subsets and macrophage subtypes remains largely unknown at present, however there is some data to support the relative transcription of TLR5 being higher in M2 polarised human macrophages as compared with M1 [28]. The circulating monocytes of ApoE-/- mice with advanced atherosclerosis have increased TLR2 and TLR4 expression [133]. This is also the case for monocytes from patients with arterial disease when comparison is made with controls subjects [134137]. Interestingly, enhanced TLR signalling is restricted to patients with acute coronary syndromes [138140].

Role of Toll-like receptors in atherosclerosis

When recognising ligands, the majority of TLRs associate the signalling adaptor MyD88 to initiate an intracellular signalling cascade. More specifically, removing the MyD88 pathway led to a reduction in aortic atherosclerosis (by approximately 60%) and a decrease in macrophage recruitment to the artery wall (by approximately 75%), associated with reduced chemokine levels [141, 142]. In a functional human atherosclerosis study, a significant reduction of pro-inflammatory cytokines and MMPs was found after MyD88 inhibition [143].
The role of TLR2 and TLR4 has been extensively studied in models of atherosclerosis. The first indication of a role for TLR4 in atherosclerosis came from the finding that C3H/HeJ mice - that hold a missense mutation of TLR4's cytoplasmic component - are resistant to atherosclerosis [144, 145]. In accordance, specific deletion of TLR4 in ApoE-/- mice resulted in a 24% reduction in whole aortic atherosclerotic lesion area and significantly attenuated macrophage infiltration within these lesions [141]. TLR2 deletion in LDLR-/- mice limits lesion area by between a third and two-thirds [141, 146148], reducing intra-lesion inflammation as evidenced by a reduction in total infiltrating macrophage numbers [147, 148], and attenuates macrophage to smooth muscle cell ratio and extent of apoptosis [147].
Both TLR2 and TLR4 are known to be important in post-vascular injury neo-intimal lesion formation [149, 150]. In a hypercholesterolaemic rabbit model of atherosclerosis, carotid artery liposomal transfection of TLR2 and TLR4 cDNA revealed that upregulation of either TLR alone did not significantly affect carotid atherosclerosis. Interestingly, transfection of both TLR2 and TLR4 together resulted in a synergistic acceleration of atherosclerosis [151]. Recently, LDLR-/- mice transplanted with TLR2-/- TLR4-/- bone marrow displayed a reduction in both macrophage apoptosis and atherosclerotic plaque necrosis as compared with LDLR-/- mice transplanted with wild-type bone marrow, supporting an additive effect of TLR2 and TLR4 in murine atherosclerosis [112].
A different picture came from bone marrow chimera studies. Bone marrow transplantation from TLR2-/- to LDLR-/- mice was unable to prevent diet-induced atherosclerotic lesions [146]. Bone marrow transfer from C3H/HeJ to ApoE knockouts did not alter atherosclerosis susceptibility [152]. Synthetic TLR2 ligand administered dramatically increases atherosclerosis in LDLR-/- mice, with TLR2 deficient bone marrow transfer into this model preventing TLR2 ligand-induced atheroma [146]. Such studies raise the question of whether TLR2 signalling in myeloid cells is relevant in atherosclerosis, as compared with TLR2 expression by cells resident in the arterial wall. Importantly, it supports the role of endogenous TLR2 ligand action on myeloid cells in atherosclerosis, with exogenous agonists activating TLR2 on cells of a non-myeloid lineage.
What are the mechanisms through which TLR exert proatherogenic actions? Importantly, TLR2, TLR4 and TLR9 ligands promote lipid uptake by macrophages and, hence, foam cell formation [111, 153155]. Differentiated macrophages exhibit macropinocytosis (fluid phase uptake of lipids) which is dependent upon TLR4 [156]. However, the effect of TLR signalling are not limited to foam cell formation but have a direct effect on inflammation and matrix degradation.
Functional studies on human carotid endarterectomy specimens have shown sustained TLR2 activation in cells isolated from human atheromata [143]. TLR2 and MyD88 play a key role in NFκB activation, and in the production of inflammatory mediators CCL2, IL6, IL8, MMPs 1, 2, 3 and 9 [143]. Conversely TLR4, and its downstream signalling adaptor TRAM, were shown not to be rate-limiting for cytokine production in this context. This adds weight to the role of some (but not all) TLRs in plaque vulnerability.
Furthermore, and as alluded to above, TLR ligation may influence atherosclerosis through alterations in MMP and TIMP expression. The effect of LPS on human blood monocytes has been investigated and MMP3 is upregulated [157], whilst MMPs 1, 2, 7, 10 and 14 and TIMPs 1, 2 and 3 are not upregulated by LPS [157, 158]. Controversially, two separate studies have found upregulation [159] and no upregulation [157] of MMP9 in human blood monocytes stimulated with LPS. In human macrophages (from various sites) meanwhile, MMPs 2, 3, 8, 9 and 14, and TIMP1 have all been upregulated by LPS [158, 160163].
Using both human and murine models of atherosclerosis, we have investigated the consequence of endosomal TLRs in atherosclerosis and arterial injury. Deficiency of TLR3 accelerates the onset of atherosclerosis in ApoE-/- mice. Moreover, genetic deletion of TLR3 dramatically enhanced the development of elastic lamina breakages after collar-induced injury. The systemic (intraperitoneal) administration of double-stranded RNA (dsRNA) - a TLR3 agonist - decreased neointima formation upon arterial injury. Genetic deletion of TLR3 was associated with the presence of large interruptions of the elastic lamina after the placement of a perivascular collar for arterial injury development. Finally, lesion development in both human and mice was associated in an increase of expression of TLR3 and TLR3-associated responses, in particular in smooth muscle cells pointing to this cell type as the carrier of the protective effect. This data shows for the first time that while extracellular TLRs may be detrimental to atherosclerosis, intracellular TLRs may offer protection against hypercholesterolemia and injury-induced lesions. The mechanism of TLR3-induced protection is currently unknown. IFNβ production - that is a consequence of TLR3 dependent signalling - has been associated with a reduction in inflammasome activation and IL1 signalling, as well as with induction of IL10 [164]. However, it is uncertain whether the vasculoprotective effect of TLR3 may be mediated via IFNβ. Although IFNβ has been shown to be effective in an arterial injury model [165], a more recent report showed a potential deleterious role in atherosclerosis induced by hyperlipidemia [166]. It is also uncertain whether synthetic dsRNA is safe as therapeutic tool, as its administration elicits both pro-inflammatory and anti-inflammatory mediators [124]. Moreover, a recent study showed that dsRNA intravenous administration at high doses may lead to endothelial cell apoptosis and increased vascular lesion formation [167]. Further studies are needed to elucidate the mechanisms of vasculoprotection elicited by TLR3. TLR3 activation has been shown to elicit the production in the vasculature of IL10 [124] and of the B7 family members programmed cell death ligands PDL1 and PDL2, which are known to contribute to vascular protection [168, 169].
It is also unknown what endogenous agonists of TLR3 may be involved in protection, as the genetic removal of TLR3 accelerates atherosclerosis and elastic lamina damage. Interestingly, stathmin, a protein that participates in microtubule assembly and is upregulated in brain injury, has been described as a candidate TLR3 agonist and has been linked to the induction of a neuroprotective gene profile [170].

NOD-like receptors and inflammasomes and atherogenesis

NLRs are PRRs that sense intra-cellular microbial and non-microbial signals, in a similar fashion to the extra-cellular detection of these entities by most TLRs. NLRs have the capacity to form large cytoplasmic complexes known as "inflammasomes" (reviewed in [171]) through the assembly of NLRs, caspase and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). ASC acts to link the NLR and caspase, the latter of which are usually caspase 1 and 11 [172]. The inflammasome acts as a scaffold for the activation of caspase 1 as its central effector molecule [173]. Upon activation, inflammasome caspase 1 proteolytically activates pro-inflammatory cytokines, notably the conversion of pro-IL1β and pro-IL18 to IL1β and IL18, respectively.
It is largely agreed that inflammasome activation resulting in active IL1β release requires two separate signals [174]. A priming signal may be triggered by TLR activation, with resultant NFκB production leading to pro-IL1β synthesis, as well as inflammasome components such as caspase 11 [173]. Recognition of peptidoglycan by NOD1 and NOD2 can also trigger activation of NFκB signal transduction through Rip2 kinase [100]. The second signal, which activates the caspase 1 of a complete inflammasome, allowing the conversion of available pro-IL1β to IL1β includes activation by ATP of the P2X7 purinergic receptor with potassium efflux. The second signal may also be achieved by PAMPs such as bacterial toxins and viral DNA, or other DAMPs including oxidative stress, large particles and ultraviolet light [171].
Inflammasomes have been described in a number of inflammatory conditions [171] and evidence for their role in atherosclerosis is emerging. The NLRP3 inflammasome is currently the most characterised inflammasome (Figure 2). Recent work has shown that cholesterol crystals activate the NLRP3 inflammasome, which in turn results in cleavage and secretion of IL1 family cytokines [21]. Furthermore, LDLR-deficient mice transplanted with NLRP3-deficient bone marrow and fed a high-cholesterol diet had markedly decreased early atherosclerosis and inflammasome-dependent IL18 levels [21]. LDLR-/- mice bone-marrow transplanted with ASC-deficient or IL-1α/β-deficient bone marrow and fed on a high-cholesterol diet had consistent and marked reductions in both atherosclerosis and IL18 production [21]. Furthermore, ASC deficiency also attenuates neointimal formation after vascular injury via reduced expression of IL1β and IL18, with ASC-/- bone marrow chimeras also exhibiting significantly reduced neointimal formation [175]. These findings taken together suggest that crystalline cholesterol acts as an endogenous danger signal, its deposition in arteries being an early cause rather than a late consequence of inflammation.
Both IL1 and IL18 signal through MyD88, and their absence in experimental mouse atherosclerosis also has the effect of limiting atherosclerosis development [176, 177]. Devlin et al showed that IL1ra knockout mice on a cholesterol/chocolate diet, exhibited a 3-fold decrease in non-high-density lipoprotein (HDL) cholesterol and a trend toward increased foam cell lesion area compared to controls [178]. Complementing this experiment they showed, conversely, that increased IL1ra expression (using an IL1ra transgenic/LDLR-/- mouse on a cholesterol-saturated fat diet) resulted in a 40% increase in non-HDL cholesterol levels. Thus concluding that under certain conditions, chronic IL1ra depletion or over-expression could have an important effect on lipid metabolism.
This was also verified in human atherosclerotic arteries [179], although more recently, IL1ra administration has been shown to have lesser effect on inflammatory molecule production when compared to TLR inhibition in the context of human atherosclerosis [143].

Macrophage deactivation pathways in atherosclerosis

PPARγ has recently been highlighted as an important determinant of macrophage phenotype and function (Figure 3), which may explain the favourable effect of PPARγ modulation in experimental atherosclerosis [180, 181]. PPARγ is a ligand-activated nuclear receptor involved in reverse cholesterol transport and other metabolic cellular activities [46]. Its anti-inflammatory properties occur through negative interference with nuclear factor κB (NFκB), signal transducer and activator of transcription (STAT), and activating protein 1 (AP1) pathways [182]. PPARγ is strongly induced by IL4 [40, 183]. PPARγ upregulation may also be stimulated by oxidised LDL, with PPARγ being highly expressed in the foam cells of atherosclerotic lesions, and ligand activation of PPARγ promoting oxidised LDL uptake and foam cell formation [184].
The functional relationship between PPARγ and the wound healing M2-type macrophage phenotype [185, 186] has been proposed through the positive correlation between PPARγ expression levels and the M2 markers CD206 [187], CD36 scavenger receptor [184], IL10 [188] and alternative activated macrophage associated CC-chemokine 1 (AMAC1; CCL18) [40]. Primary human monocytes differentiated in vitro with IL4 in the presence of PPARγ agonist (termed M2γ macrophages) resulted in increased CD206 and reduced CD163 expression, above and beyond that which was seen with IL4 alone [40] (Figure 3). M2γ culture supernatant exerted a greater anti-inflammatory effect on M1 macrophages as compared with M2 culture supernatant [40]. Subsequent work has shown that M2γ macrophages have down-regulation of the nuclear liver × receptor α with resultant enhanced phagocytosis but reduced cholesterol handling [41]. PPARγ also limits MMP9 through inhibition of NFκB activation [189].
However, in the clinical arena, PPARγ agnonists have been shown to have complex and opposing effects on circulating levels of pro- and anti-inflammatory molecules [190193]. Furthermore, macrophages have been observed adhering to areas of intimal thickening in PPARγ-dependent manner [194].

Conclusions

Macrophages have been shown to exert a number of diverse functions in atherosclerosis, including inflammation, lipid metabolism and matrix degradation Recent studies have highlighted significant heterogeneity in macrophage behaviour and activation within atherosclerotic plaque. This heterogeneity is derived both from the heterogeneity of originating monocytes, and the inflammatory and lipidic stimuli available in the plaque. It is known that signalling pathways related to innate immunity are strong determinants for macrophage activation and there is growing evidence that they have a significant effect in plaque development and the complications thereof. Innate immune pathways may be activated by both infectious pathogens and endogenous danger signals. An example of the latter is the recognition by innate immune receptors of a growing number of lipoprotein components that are vital to the development of atherosclerosis. Oxidised LDL is seen to signal through TLR [2224], cholesterol crystals signalling through NLR [21], and oxPAPC signalling via NRF2 [36]. The convergence of these pathways gives rise to the activation of resident monocyte-macrophages leading to cytokine and chemokine production. Moreover, TLR activation might have a role in biasing macrophage polarisation towards an M1 phenotype, together with Th1 lymphocytes present in the plaque. These exciting new findings highlight a wealth of novel potential therapeutic and diagnostic targets that may be exploited in the future treatment of cardiovascular disease.

Acknowledgements

Mr Joseph Shalhoub's research is supported by the Circulation Foundation Mary Davies Research Fellowship, the Royal College of Surgeons of England/Rosetrees Trust Research Fellowship, the Graham-Dixon Charitable Trust and the Peel Medical Research Trust.
Dr Claudia Monaco has received funding from: the British Heart Foundation; European Commission under the 6th Framework Programme through the SME call for "Life sciences, genomics and biotechnology for health" (Contract number: LSHM-CT-2006-037400); European Collaborative Project on Inflammation and Vascular Wall Remodelling in Atherosclerosis Acronym: AtheroRemo; EU - HEALTH-2007-2.4.2-1. 2008; the Graham-Dixon Charitable Trust; the Kennedy Trustees. The Kennedy Institute of Rheumatology is funded by the Arthritis Research Campaign UK.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

All authors were involved directly in the drafting and critical revision of this review. CM is the senior author with overall responsibility for this work, giving the final approval for publication. All authors have read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Lopez AD, Mathers CD, Ezzati M, Jamison DT, Murray CJ: Global and regional burden of disease and risk factors, 2001: systematic analysis of population health data. Lancet. 2006, 367 (9524): 1747-57. 10.1016/S0140-6736(06)68770-9.PubMed Lopez AD, Mathers CD, Ezzati M, Jamison DT, Murray CJ: Global and regional burden of disease and risk factors, 2001: systematic analysis of population health data. Lancet. 2006, 367 (9524): 1747-57. 10.1016/S0140-6736(06)68770-9.PubMed
2.
Zurück zum Zitat Global Burden of Disease Study Operations Manual. 2009, Harvard University, University of Washington, Johns Hopkins University, University of Queensland, World Health Organization Global Burden of Disease Study Operations Manual. 2009, Harvard University, University of Washington, Johns Hopkins University, University of Queensland, World Health Organization
3.
Zurück zum Zitat Mayerl C, Lukasser M, Sedivy R, Niederegger H: Atherosclerosis research from past to present--on the track of two pathologists with opposing views, Carl von Rokitansky and Rudolf Virchow. Virchows Archiv. 2006 Mayerl C, Lukasser M, Sedivy R, Niederegger H: Atherosclerosis research from past to present--on the track of two pathologists with opposing views, Carl von Rokitansky and Rudolf Virchow. Virchows Archiv. 2006
4.
Zurück zum Zitat Methe H, Weis M: Atherogenesis and inflammation--was Virchow right?. Nephrology Dialysis Transplantation. 2007 Methe H, Weis M: Atherogenesis and inflammation--was Virchow right?. Nephrology Dialysis Transplantation. 2007
5.
Zurück zum Zitat Ross R: Atherosclerosis--an inflammatory disease. N Engl J Med. 1999, 340 (2): 115-26. 10.1056/NEJM199901143400207.PubMed Ross R: Atherosclerosis--an inflammatory disease. N Engl J Med. 1999, 340 (2): 115-26. 10.1056/NEJM199901143400207.PubMed
6.
Zurück zum Zitat Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Kastelein JJ, Koenig W, Libby P, Lorenzatti AJ, MacFadyen JG, Nordestgaard BG, Shepherd J, Willerson JT, Glynn RJ: Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008, 359 (21): 2195-207. 10.1056/NEJMoa0807646.PubMed Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Kastelein JJ, Koenig W, Libby P, Lorenzatti AJ, MacFadyen JG, Nordestgaard BG, Shepherd J, Willerson JT, Glynn RJ: Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008, 359 (21): 2195-207. 10.1056/NEJMoa0807646.PubMed
7.
Zurück zum Zitat Full L, Ruisanchez C, Monaco C: The inextricable link between atherosclerosis and prototypical inflammatory diseases rheumatoid arthritis and systemic lupus erythematosus. Arthritis Res Ther. 2009, 11 (2): 217-10.1186/ar2631.PubMedPubMedCentral Full L, Ruisanchez C, Monaco C: The inextricable link between atherosclerosis and prototypical inflammatory diseases rheumatoid arthritis and systemic lupus erythematosus. Arthritis Res Ther. 2009, 11 (2): 217-10.1186/ar2631.PubMedPubMedCentral
8.
Zurück zum Zitat Haque S, Mirjafari H, Bruce IN: Atherosclerosis in rheumatoid arthritis and systemic lupus erythematosus. Curr Opin Lipidol. 2008, 19 (4): 338-43. 10.1097/MOL.0b013e328304b65f.PubMed Haque S, Mirjafari H, Bruce IN: Atherosclerosis in rheumatoid arthritis and systemic lupus erythematosus. Curr Opin Lipidol. 2008, 19 (4): 338-43. 10.1097/MOL.0b013e328304b65f.PubMed
9.
Zurück zum Zitat Glass CK, Witztum JL: Atherosclerosis. the road ahead. Cell. 2001, 104 (4): 503-16. 10.1016/S0092-8674(01)00238-0.PubMed Glass CK, Witztum JL: Atherosclerosis. the road ahead. Cell. 2001, 104 (4): 503-16. 10.1016/S0092-8674(01)00238-0.PubMed
10.
Zurück zum Zitat Caro CG: Discovery of the role of wall shear in atherosclerosis. Arterioscler Thromb Vasc Biol. 2009, 29 (2): 158-61. 10.1161/ATVBAHA.108.166736.PubMed Caro CG: Discovery of the role of wall shear in atherosclerosis. Arterioscler Thromb Vasc Biol. 2009, 29 (2): 158-61. 10.1161/ATVBAHA.108.166736.PubMed
11.
Zurück zum Zitat Cheng C, Tempel D, van Haperen R, van der Baan A, Grosveld F, Daemen MJ, Krams R, de Crom R: Atherosclerotic lesion size and vulnerability are determined by patterns of fluid shear stress. Circulation. 2006, 113 (23): 2744-53. 10.1161/CIRCULATIONAHA.105.590018.PubMed Cheng C, Tempel D, van Haperen R, van der Baan A, Grosveld F, Daemen MJ, Krams R, de Crom R: Atherosclerotic lesion size and vulnerability are determined by patterns of fluid shear stress. Circulation. 2006, 113 (23): 2744-53. 10.1161/CIRCULATIONAHA.105.590018.PubMed
12.
Zurück zum Zitat Davies MJ, Richardson PD, Woolf N, Katz DR, Mann J: Risk of thrombosis in human atherosclerotic plaques: role of extracellular lipid, macrophage, and smooth muscle cell content. Br Heart J. 1993, 69 (5): 377-81. 10.1136/hrt.69.5.377.PubMedPubMedCentral Davies MJ, Richardson PD, Woolf N, Katz DR, Mann J: Risk of thrombosis in human atherosclerotic plaques: role of extracellular lipid, macrophage, and smooth muscle cell content. Br Heart J. 1993, 69 (5): 377-81. 10.1136/hrt.69.5.377.PubMedPubMedCentral
13.
Zurück zum Zitat van der Wal AC, Becker AE, van der Loos CM, Das PK: Site of intimal rupture or erosion of thrombosed coronary atherosclerotic plaques is characterized by an inflammatory process irrespective of the dominant plaque morphology. Circulation. 1994, 89 (1): 36-44.PubMed van der Wal AC, Becker AE, van der Loos CM, Das PK: Site of intimal rupture or erosion of thrombosed coronary atherosclerotic plaques is characterized by an inflammatory process irrespective of the dominant plaque morphology. Circulation. 1994, 89 (1): 36-44.PubMed
14.
Zurück zum Zitat Virmani R, Burke AP, Kolodgie FD, Farb A: Pathology of the thin-cap fibroatheroma: a type of vulnerable plaque. J Interv Cardiol. 2003, 16 (3): 267-72. 10.1034/j.1600-0854.2003.8042.x.PubMed Virmani R, Burke AP, Kolodgie FD, Farb A: Pathology of the thin-cap fibroatheroma: a type of vulnerable plaque. J Interv Cardiol. 2003, 16 (3): 267-72. 10.1034/j.1600-0854.2003.8042.x.PubMed
15.
Zurück zum Zitat Gleissner CA, Shaked I, Little KM, Ley K: CXC chemokine ligand 4 induces a unique transcriptome in monocyte-derived macrophages. J Immunol. 2010, 184 (9): 4810-8. 10.4049/jimmunol.0901368.PubMedPubMedCentral Gleissner CA, Shaked I, Little KM, Ley K: CXC chemokine ligand 4 induces a unique transcriptome in monocyte-derived macrophages. J Immunol. 2010, 184 (9): 4810-8. 10.4049/jimmunol.0901368.PubMedPubMedCentral
16.
Zurück zum Zitat Tedgui A, Mallat Z: Cytokines in atherosclerosis: pathogenic and regulatory pathways. Physiol Rev. 2006, 86 (2): 515-81. 10.1152/physrev.00024.2005.PubMed Tedgui A, Mallat Z: Cytokines in atherosclerosis: pathogenic and regulatory pathways. Physiol Rev. 2006, 86 (2): 515-81. 10.1152/physrev.00024.2005.PubMed
17.
Zurück zum Zitat Mauriello A, Sangiorgi GM, Virmani R, Trimarchi S, Holmes DR, Kolodgie FD, Piepgras DG, Piperno G, Liotti D, Narula J, Righini P, Ippoliti A, Spagnoli LG: A pathobiologic link between risk factors profile and morphological markers of carotid instability. Atherosclerosis. 2010, 208 (2): 572-80. 10.1016/j.atherosclerosis.2009.07.048.PubMed Mauriello A, Sangiorgi GM, Virmani R, Trimarchi S, Holmes DR, Kolodgie FD, Piepgras DG, Piperno G, Liotti D, Narula J, Righini P, Ippoliti A, Spagnoli LG: A pathobiologic link between risk factors profile and morphological markers of carotid instability. Atherosclerosis. 2010, 208 (2): 572-80. 10.1016/j.atherosclerosis.2009.07.048.PubMed
18.
Zurück zum Zitat Grabner R, Lotzer K, Dopping S, Hildner M, Radke D, Beer M, Spanbroek R, Lippert B, Reardon CA, Getz GS, Fu YX, Hehlgans T, Mebius RE, van der Wall M, Kruspe D, Englert C, Lovas A, Hu D, Randolph GJ, Weih F, Habenicht AJ: Lymphotoxin beta receptor signaling promotes tertiary lymphoid organogenesis in the aorta adventitia of aged ApoE-/- mice. J Exp Med. 2009, 206 (1): 233-48. 10.1084/jem.20080752.PubMedPubMedCentral Grabner R, Lotzer K, Dopping S, Hildner M, Radke D, Beer M, Spanbroek R, Lippert B, Reardon CA, Getz GS, Fu YX, Hehlgans T, Mebius RE, van der Wall M, Kruspe D, Englert C, Lovas A, Hu D, Randolph GJ, Weih F, Habenicht AJ: Lymphotoxin beta receptor signaling promotes tertiary lymphoid organogenesis in the aorta adventitia of aged ApoE-/- mice. J Exp Med. 2009, 206 (1): 233-48. 10.1084/jem.20080752.PubMedPubMedCentral
19.
Zurück zum Zitat Dunmore BJ, McCarthy MJ, Naylor AR, Brindle NP: Carotid plaque instability and ischemic symptoms are linked to immaturity of microvessels within plaques. J Vasc Surg. 2007, 45 (1): 155-9. 10.1016/j.jvs.2006.08.072.PubMed Dunmore BJ, McCarthy MJ, Naylor AR, Brindle NP: Carotid plaque instability and ischemic symptoms are linked to immaturity of microvessels within plaques. J Vasc Surg. 2007, 45 (1): 155-9. 10.1016/j.jvs.2006.08.072.PubMed
20.
Zurück zum Zitat Narula J, Strauss HW: The popcorn plaques. Nat Med. 2007, 13 (5): 532-4. 10.1038/nm0507-532.PubMed Narula J, Strauss HW: The popcorn plaques. Nat Med. 2007, 13 (5): 532-4. 10.1038/nm0507-532.PubMed
21.
Zurück zum Zitat Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, Abela GS, Franchi L, Nunez G, Schnurr M, Espevik T, Lien E, Fitzgerald KA, Rock KL, Moore KJ, Wright SD, Hornung V, Latz E: NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010, 464 (7293): 1357-61. 10.1038/nature08938.PubMedPubMedCentral Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, Abela GS, Franchi L, Nunez G, Schnurr M, Espevik T, Lien E, Fitzgerald KA, Rock KL, Moore KJ, Wright SD, Hornung V, Latz E: NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature. 2010, 464 (7293): 1357-61. 10.1038/nature08938.PubMedPubMedCentral
22.
Zurück zum Zitat Miller YI, Viriyakosol S, Binder CJ, Feramisco JR, Kirkland TN, Witztum JL: Minimally modified LDL binds to CD14, induces macrophage spreading via TLR4/MD-2, and inhibits phagocytosis of apoptotic cells. J Biol Chem. 2003, 278 (3): 1561-8. 10.1074/jbc.M209634200.PubMed Miller YI, Viriyakosol S, Binder CJ, Feramisco JR, Kirkland TN, Witztum JL: Minimally modified LDL binds to CD14, induces macrophage spreading via TLR4/MD-2, and inhibits phagocytosis of apoptotic cells. J Biol Chem. 2003, 278 (3): 1561-8. 10.1074/jbc.M209634200.PubMed
23.
Zurück zum Zitat Miller YI, Viriyakosol S, Worrall DS, Boullier A, Butler S, Witztum JL: Toll-like receptor 4-dependent and -independent cytokine secretion induced by minimally oxidized low-density lipoprotein in macrophages. Arteriosclerosis, Thrombosis, and Vascular Biology. 2005, 25 (6): 1213-9. 10.1161/01.ATV.0000159891.73193.31.PubMed Miller YI, Viriyakosol S, Worrall DS, Boullier A, Butler S, Witztum JL: Toll-like receptor 4-dependent and -independent cytokine secretion induced by minimally oxidized low-density lipoprotein in macrophages. Arteriosclerosis, Thrombosis, and Vascular Biology. 2005, 25 (6): 1213-9. 10.1161/01.ATV.0000159891.73193.31.PubMed
24.
Zurück zum Zitat Stewart CR, Stuart LM, Wilkinson K, van Gils JM, Deng J, Halle A, Rayner KJ, Boyer L, Zhong R, Frazier WA, Lacy-Hulbert A, Khoury JE, Golenbock DT, Moore KJ: CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat Immunol. 2010, 11 (2): 155-61. 10.1038/ni.1836.PubMedPubMedCentral Stewart CR, Stuart LM, Wilkinson K, van Gils JM, Deng J, Halle A, Rayner KJ, Boyer L, Zhong R, Frazier WA, Lacy-Hulbert A, Khoury JE, Golenbock DT, Moore KJ: CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat Immunol. 2010, 11 (2): 155-61. 10.1038/ni.1836.PubMedPubMedCentral
25.
Zurück zum Zitat Cole JE, Georgiou E, Monaco C: The Expression and Functions of Toll-Like Receptors in Atherosclerosis. Mediators of Inflammation. 2010 Cole JE, Georgiou E, Monaco C: The Expression and Functions of Toll-Like Receptors in Atherosclerosis. Mediators of Inflammation. 2010
26.
Zurück zum Zitat Van Ginderachter JA, Movahedi K, Hassanzadeh Ghassabeh G, Meerschaut S, Beschin A, Raes G, De Baetselier P: Classical and alternative activation of mononuclear phagocytes: picking the best of both worlds for tumor promotion. Immunobiology. 2006, 211 (6-8): 487-501. 10.1016/j.imbio.2006.06.002.PubMed Van Ginderachter JA, Movahedi K, Hassanzadeh Ghassabeh G, Meerschaut S, Beschin A, Raes G, De Baetselier P: Classical and alternative activation of mononuclear phagocytes: picking the best of both worlds for tumor promotion. Immunobiology. 2006, 211 (6-8): 487-501. 10.1016/j.imbio.2006.06.002.PubMed
27.
Zurück zum Zitat Waldo SW, Li Y, Buono C, Zhao B, Billings EM, Chang J, Kruth HS: Heterogeneity of human macrophages in culture and in atherosclerotic plaques. Am J Pathol. 2008, 172 (4): 1112-26. 10.2353/ajpath.2008.070513.PubMedPubMedCentral Waldo SW, Li Y, Buono C, Zhao B, Billings EM, Chang J, Kruth HS: Heterogeneity of human macrophages in culture and in atherosclerotic plaques. Am J Pathol. 2008, 172 (4): 1112-26. 10.2353/ajpath.2008.070513.PubMedPubMedCentral
28.
Zurück zum Zitat Martinez FO, Gordon S, Locati M, Mantovani A: Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J Immunol. 2006, 177 (10): 7303-11.PubMed Martinez FO, Gordon S, Locati M, Mantovani A: Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J Immunol. 2006, 177 (10): 7303-11.PubMed
29.
Zurück zum Zitat Gordon S: Alternative activation of macrophages. Nat Rev Immunol. 2003, 3 (1): 23-35. 10.1038/nri978.PubMed Gordon S: Alternative activation of macrophages. Nat Rev Immunol. 2003, 3 (1): 23-35. 10.1038/nri978.PubMed
30.
Zurück zum Zitat Gordon S: The macrophage: past, present and future. Eur J Immunol. 2007, 37 (Suppl 1): S9-17.PubMed Gordon S: The macrophage: past, present and future. Eur J Immunol. 2007, 37 (Suppl 1): S9-17.PubMed
31.
Zurück zum Zitat Mosser DM, Edwards JP: Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008, 8 (12): 958-69. 10.1038/nri2448.PubMedPubMedCentral Mosser DM, Edwards JP: Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008, 8 (12): 958-69. 10.1038/nri2448.PubMedPubMedCentral
32.
Zurück zum Zitat Mantovani A, Garlanda C, Locati M: Macrophage diversity and polarization in atherosclerosis: a question of balance. Arterioscler Thromb Vasc Biol. 2009, 29 (10): 1419-23. 10.1161/ATVBAHA.108.180497.PubMed Mantovani A, Garlanda C, Locati M: Macrophage diversity and polarization in atherosclerosis: a question of balance. Arterioscler Thromb Vasc Biol. 2009, 29 (10): 1419-23. 10.1161/ATVBAHA.108.180497.PubMed
33.
Zurück zum Zitat Hansson GK: Atherosclerosis An immune disease: The Anitschkov Lecture 2007. Atherosclerosis. 2008, 1-9. Hansson GK: Atherosclerosis An immune disease: The Anitschkov Lecture 2007. Atherosclerosis. 2008, 1-9.
34.
Zurück zum Zitat Mallat Z, Taleb S, Ait-Oufella H, Tedgui A: The role of adaptive T cell immunity in atherosclerosis. J Lipid Res. 2009, 50 (Suppl): S364-9.PubMedPubMedCentral Mallat Z, Taleb S, Ait-Oufella H, Tedgui A: The role of adaptive T cell immunity in atherosclerosis. J Lipid Res. 2009, 50 (Suppl): S364-9.PubMedPubMedCentral
35.
Zurück zum Zitat Khallou-Laschet J, Varthaman A, Fornasa G, Compain C, Gaston AT, Clement M, Dussiot M, Levillain O, Graff-Dubois S, Nicoletti A, Caligiuri G: Macrophage plasticity in experimental atherosclerosis. PLoS One. 2010, 5 (1): e8852-10.1371/journal.pone.0008852.PubMedPubMedCentral Khallou-Laschet J, Varthaman A, Fornasa G, Compain C, Gaston AT, Clement M, Dussiot M, Levillain O, Graff-Dubois S, Nicoletti A, Caligiuri G: Macrophage plasticity in experimental atherosclerosis. PLoS One. 2010, 5 (1): e8852-10.1371/journal.pone.0008852.PubMedPubMedCentral
36.
Zurück zum Zitat Kadl A, Meher AK, Sharma PR, Lee MY, Doran AC, Johnstone SR, Elliott MR, Gruber F, Han J, Chen W, Kensler T, Ravichandran KS, Isakson BE, Wamhoff BR, Leitinger N: Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circ Res. 2010, 107 (6): 737-46. 10.1161/CIRCRESAHA.109.215715.PubMedPubMedCentral Kadl A, Meher AK, Sharma PR, Lee MY, Doran AC, Johnstone SR, Elliott MR, Gruber F, Han J, Chen W, Kensler T, Ravichandran KS, Isakson BE, Wamhoff BR, Leitinger N: Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circ Res. 2010, 107 (6): 737-46. 10.1161/CIRCRESAHA.109.215715.PubMedPubMedCentral
37.
Zurück zum Zitat Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M: The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25 (12): 677-86. 10.1016/j.it.2004.09.015.PubMed Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M: The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25 (12): 677-86. 10.1016/j.it.2004.09.015.PubMed
38.
Zurück zum Zitat Miller AM, Xu D, Asquith DL, Denby L, Li Y, Sattar N, Baker AH, McInnes IB, Liew FY: IL-33 reduces the development of atherosclerosis. J Exp Med. 2008, 205 (2): 339-46. 10.1084/jem.20071868.PubMedPubMedCentral Miller AM, Xu D, Asquith DL, Denby L, Li Y, Sattar N, Baker AH, McInnes IB, Liew FY: IL-33 reduces the development of atherosclerosis. J Exp Med. 2008, 205 (2): 339-46. 10.1084/jem.20071868.PubMedPubMedCentral
39.
Zurück zum Zitat Miller AM, Asquith DL, Hueber AJ, Anderson LA, Holmes WM, McKenzie AN, Xu D, Sattar N, McInnes IB, Liew FY: Interleukin-33 induces protective effects in adipose tissue inflammation during obesity in mice. Circ Res. 2010, 107 (5): 650-8. 10.1161/CIRCRESAHA.110.218867.PubMedPubMedCentral Miller AM, Asquith DL, Hueber AJ, Anderson LA, Holmes WM, McKenzie AN, Xu D, Sattar N, McInnes IB, Liew FY: Interleukin-33 induces protective effects in adipose tissue inflammation during obesity in mice. Circ Res. 2010, 107 (5): 650-8. 10.1161/CIRCRESAHA.110.218867.PubMedPubMedCentral
40.
Zurück zum Zitat Bouhlel MA, Derudas B, Rigamonti E, Dievart R, Brozek J, Haulon S, Zawadzki C, Jude B, Torpier G, Marx N, Staels B, Chinetti-Gbaguidi G: PPARgamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab. 2007, 6 (2): 137-43. 10.1016/j.cmet.2007.06.010.PubMed Bouhlel MA, Derudas B, Rigamonti E, Dievart R, Brozek J, Haulon S, Zawadzki C, Jude B, Torpier G, Marx N, Staels B, Chinetti-Gbaguidi G: PPARgamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab. 2007, 6 (2): 137-43. 10.1016/j.cmet.2007.06.010.PubMed
41.
Zurück zum Zitat Chinetti-Gbaguidi G, Baron M, Bouhlel MA, Vanhoutte J, Copin C, Sebti Y, Derudas B, Mayi T, Bories G, Tailleux A, Haulon S, Zawadzki C, Jude B, Staels B: Human Atherosclerotic Plaque Alternative Macrophages Display Low Cholesterol Handling but High Phagocytosis Because of Distinct Activities of the PPAR{gamma} and LXR&alpha; Pathways. Circ Res. 2011 Chinetti-Gbaguidi G, Baron M, Bouhlel MA, Vanhoutte J, Copin C, Sebti Y, Derudas B, Mayi T, Bories G, Tailleux A, Haulon S, Zawadzki C, Jude B, Staels B: Human Atherosclerotic Plaque Alternative Macrophages Display Low Cholesterol Handling but High Phagocytosis Because of Distinct Activities of the PPAR{gamma} and LXR&alpha; Pathways. Circ Res. 2011
42.
Zurück zum Zitat Boyle JJ, Harrington HA, Piper E, Elderfield K, Stark J, Landis RC, Haskard DO: Coronary intraplaque hemorrhage evokes a novel atheroprotective macrophage phenotype. Am J Pathol. 2009, 174 (3): 1097-108. 10.2353/ajpath.2009.080431.PubMedPubMedCentral Boyle JJ, Harrington HA, Piper E, Elderfield K, Stark J, Landis RC, Haskard DO: Coronary intraplaque hemorrhage evokes a novel atheroprotective macrophage phenotype. Am J Pathol. 2009, 174 (3): 1097-108. 10.2353/ajpath.2009.080431.PubMedPubMedCentral
43.
Zurück zum Zitat Chase AJ, Bond M, Crook MF, Newby AC: Role of nuclear factor-kappa B activation in metalloproteinase-1, -3, and -9 secretion by human macrophages in vitro and rabbit foam cells produced in vivo. Arterioscler Thromb Vasc Biol. 2002, 22 (5): 765-71. 10.1161/01.ATV.0000015078.09208.92.PubMed Chase AJ, Bond M, Crook MF, Newby AC: Role of nuclear factor-kappa B activation in metalloproteinase-1, -3, and -9 secretion by human macrophages in vitro and rabbit foam cells produced in vivo. Arterioscler Thromb Vasc Biol. 2002, 22 (5): 765-71. 10.1161/01.ATV.0000015078.09208.92.PubMed
44.
Zurück zum Zitat Johnson JL, Sala-Newby GB, Ismail Y, Aguilera CM, Newby AC: Low tissue inhibitor of metalloproteinases 3 and high matrix metalloproteinase 14 levels defines a subpopulation of highly invasive foam-cell macrophages. Arterioscler Thromb Vasc Biol. 2008, 28 (9): 1647-53. 10.1161/ATVBAHA.108.170548.PubMedPubMedCentral Johnson JL, Sala-Newby GB, Ismail Y, Aguilera CM, Newby AC: Low tissue inhibitor of metalloproteinases 3 and high matrix metalloproteinase 14 levels defines a subpopulation of highly invasive foam-cell macrophages. Arterioscler Thromb Vasc Biol. 2008, 28 (9): 1647-53. 10.1161/ATVBAHA.108.170548.PubMedPubMedCentral
45.
Zurück zum Zitat Thomas AC, Sala-Newby GB, Ismail Y, Johnson JL, Pasterkamp G, Newby AC: Genomics of foam cells and nonfoamy macrophages from rabbits identifies arginase-I as a differential regulator of nitric oxide production. Arterioscler Thromb Vasc Biol. 2007, 27 (3): 571-7. 10.1161/01.ATV.0000256470.23842.94.PubMed Thomas AC, Sala-Newby GB, Ismail Y, Johnson JL, Pasterkamp G, Newby AC: Genomics of foam cells and nonfoamy macrophages from rabbits identifies arginase-I as a differential regulator of nitric oxide production. Arterioscler Thromb Vasc Biol. 2007, 27 (3): 571-7. 10.1161/01.ATV.0000256470.23842.94.PubMed
46.
Zurück zum Zitat Gordon S, Martinez FO: Alternative activation of macrophages: mechanism and functions. Immunity. 2010, 32 (5): 593-604. 10.1016/j.immuni.2010.05.007.PubMed Gordon S, Martinez FO: Alternative activation of macrophages: mechanism and functions. Immunity. 2010, 32 (5): 593-604. 10.1016/j.immuni.2010.05.007.PubMed
47.
Zurück zum Zitat Geissmann F, Jung S, Littman DR: Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity. 2003, 19 (1): 71-82. 10.1016/S1074-7613(03)00174-2.PubMed Geissmann F, Jung S, Littman DR: Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity. 2003, 19 (1): 71-82. 10.1016/S1074-7613(03)00174-2.PubMed
48.
Zurück zum Zitat Fleming TJ, O'HUigin C, Malek TR: Characterization of two novel Ly-6 genes. Protein sequence and potential structural similarity to alpha-bungarotoxin and other neurotoxins. J Immunol. 1993, 150 (12): 5379-90.PubMed Fleming TJ, O'HUigin C, Malek TR: Characterization of two novel Ly-6 genes. Protein sequence and potential structural similarity to alpha-bungarotoxin and other neurotoxins. J Immunol. 1993, 150 (12): 5379-90.PubMed
49.
Zurück zum Zitat Strauss-Ayali D, Conrad SM, Mosser DM: Monocyte subpopulations and their differentiation patterns during infection. J Leukoc Biol. 2007, 82 (2): 244-52. 10.1189/jlb.0307191.PubMed Strauss-Ayali D, Conrad SM, Mosser DM: Monocyte subpopulations and their differentiation patterns during infection. J Leukoc Biol. 2007, 82 (2): 244-52. 10.1189/jlb.0307191.PubMed
50.
Zurück zum Zitat Gautier EL, Jakubzick C, Randolph GJ: Regulation of the migration and survival of monocyte subsets by chemokine receptors and its relevance to atherosclerosis. Arterioscler Thromb Vasc Biol. 2009, 29 (10): 1412-8. 10.1161/ATVBAHA.108.180505.PubMedPubMedCentral Gautier EL, Jakubzick C, Randolph GJ: Regulation of the migration and survival of monocyte subsets by chemokine receptors and its relevance to atherosclerosis. Arterioscler Thromb Vasc Biol. 2009, 29 (10): 1412-8. 10.1161/ATVBAHA.108.180505.PubMedPubMedCentral
51.
Zurück zum Zitat Steinman RM, Idoyaga J: Features of the dendritic cell lineage. Immunol Rev. 2010, 234 (1): 5-17. 10.1111/j.0105-2896.2009.00888.x.PubMed Steinman RM, Idoyaga J: Features of the dendritic cell lineage. Immunol Rev. 2010, 234 (1): 5-17. 10.1111/j.0105-2896.2009.00888.x.PubMed
52.
Zurück zum Zitat Passlick B, Flieger D, Ziegler-Heitbrock HW: Identification and characterization of a novel monocyte subpopulation in human peripheral blood. Blood. 1989, 74 (7): 2527-34.PubMed Passlick B, Flieger D, Ziegler-Heitbrock HW: Identification and characterization of a novel monocyte subpopulation in human peripheral blood. Blood. 1989, 74 (7): 2527-34.PubMed
53.
Zurück zum Zitat Ziegler-Heitbrock HW, Fingerle G, Strobel M, Schraut W, Stelter F, Schutt C, Passlick B, Pforte A: The novel subset of CD14+/CD16+ blood monocytes exhibits features of tissue macrophages. Eur J Immunol. 1993, 23 (9): 2053-8. 10.1002/eji.1830230902.PubMed Ziegler-Heitbrock HW, Fingerle G, Strobel M, Schraut W, Stelter F, Schutt C, Passlick B, Pforte A: The novel subset of CD14+/CD16+ blood monocytes exhibits features of tissue macrophages. Eur J Immunol. 1993, 23 (9): 2053-8. 10.1002/eji.1830230902.PubMed
54.
Zurück zum Zitat Ancuta P, Rao R, Moses A, Mehle A, Shaw S, Luscinskas F, Gabuzda D: Fractalkine preferentially mediates arrest and migration of CD16+ monocytes. J Exp Med. 2003, 197 (12): 1701-7. 10.1084/jem.20022156.PubMedPubMedCentral Ancuta P, Rao R, Moses A, Mehle A, Shaw S, Luscinskas F, Gabuzda D: Fractalkine preferentially mediates arrest and migration of CD16+ monocytes. J Exp Med. 2003, 197 (12): 1701-7. 10.1084/jem.20022156.PubMedPubMedCentral
55.
Zurück zum Zitat Weber C, Belge KU, von Hundelshausen P, Draude G, Steppich B, Mack M, Frankenberger M, Weber KS, Ziegler-Heitbrock HW: Differential chemokine receptor expression and function in human monocyte subpopulations. J Leukoc Biol. 2000, 67 (5): 699-704.PubMed Weber C, Belge KU, von Hundelshausen P, Draude G, Steppich B, Mack M, Frankenberger M, Weber KS, Ziegler-Heitbrock HW: Differential chemokine receptor expression and function in human monocyte subpopulations. J Leukoc Biol. 2000, 67 (5): 699-704.PubMed
56.
Zurück zum Zitat Tacke F, Alvarez D, Kaplan TJ, Jakubzick C, Spanbroek R, Llodra J, Garin A, Liu J, Mack M, van Rooijen N, Lira SA, Habenicht AJ, Randolph GJ: Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest. 2007, 117 (1): 185-94. 10.1172/JCI28549.PubMedPubMedCentral Tacke F, Alvarez D, Kaplan TJ, Jakubzick C, Spanbroek R, Llodra J, Garin A, Liu J, Mack M, van Rooijen N, Lira SA, Habenicht AJ, Randolph GJ: Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest. 2007, 117 (1): 185-94. 10.1172/JCI28549.PubMedPubMedCentral
57.
Zurück zum Zitat Palframan RT, Jung S, Cheng G, Weninger W, Luo Y, Dorf M, Littman DR, Rollins BJ, Zweerink H, Rot A, von Andrian UH: Inflammatory chemokine transport and presentation in HEV: a remote control mechanism for monocyte recruitment to lymph nodes in inflamed tissues. J Exp Med. 2001, 194 (9): 1361-73. 10.1084/jem.194.9.1361.PubMedPubMedCentral Palframan RT, Jung S, Cheng G, Weninger W, Luo Y, Dorf M, Littman DR, Rollins BJ, Zweerink H, Rot A, von Andrian UH: Inflammatory chemokine transport and presentation in HEV: a remote control mechanism for monocyte recruitment to lymph nodes in inflamed tissues. J Exp Med. 2001, 194 (9): 1361-73. 10.1084/jem.194.9.1361.PubMedPubMedCentral
58.
Zurück zum Zitat Gordon S, Taylor PR: Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005, 5 (12): 953-64. 10.1038/nri1733.PubMed Gordon S, Taylor PR: Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005, 5 (12): 953-64. 10.1038/nri1733.PubMed
59.
Zurück zum Zitat Ingersoll MA, Spanbroek R, Lottaz C, Gautier EL, Frankenberger M, Hoffmann R, Lang R, Haniffa M, Collin M, Tacke F, Habenicht AJ, Ziegler-Heitbrock L, Randolph GJ: Comparison of gene expression profiles between human and mouse monocyte subsets. Blood. 2010, 115 (3): e10-9. 10.1182/blood-2009-07-235028.PubMedPubMedCentral Ingersoll MA, Spanbroek R, Lottaz C, Gautier EL, Frankenberger M, Hoffmann R, Lang R, Haniffa M, Collin M, Tacke F, Habenicht AJ, Ziegler-Heitbrock L, Randolph GJ: Comparison of gene expression profiles between human and mouse monocyte subsets. Blood. 2010, 115 (3): e10-9. 10.1182/blood-2009-07-235028.PubMedPubMedCentral
60.
Zurück zum Zitat Swirski F, Libby P, Aikawa E, Alcaide P, Luscinskas F, Weissleder R, Pittet M: Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J Clin Invest. 2007, 117 (1): 195-205. 10.1172/JCI29950.PubMedPubMedCentral Swirski F, Libby P, Aikawa E, Alcaide P, Luscinskas F, Weissleder R, Pittet M: Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J Clin Invest. 2007, 117 (1): 195-205. 10.1172/JCI29950.PubMedPubMedCentral
61.
Zurück zum Zitat Swirski FK, Weissleder R, Pittet MJ: Heterogeneous in vivo behavior of monocyte subsets in atherosclerosis. Arterioscler Thromb Vasc Biol. 2009, 29 (10): 1424-32. 10.1161/ATVBAHA.108.180521.PubMedPubMedCentral Swirski FK, Weissleder R, Pittet MJ: Heterogeneous in vivo behavior of monocyte subsets in atherosclerosis. Arterioscler Thromb Vasc Biol. 2009, 29 (10): 1424-32. 10.1161/ATVBAHA.108.180521.PubMedPubMedCentral
62.
Zurück zum Zitat Auffray C, Sieweke MH, Geissmann F: Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol. 2009, 27: 669-92. 10.1146/annurev.immunol.021908.132557.PubMed Auffray C, Sieweke MH, Geissmann F: Blood monocytes: development, heterogeneity, and relationship with dendritic cells. Annu Rev Immunol. 2009, 27: 669-92. 10.1146/annurev.immunol.021908.132557.PubMed
63.
Zurück zum Zitat Geissmann F, Gordon S, Hume DA, Mowat AM, Randolph GJ: Unravelling mononuclear phagocyte heterogeneity. Nat Rev Immunol. 2010, 10 (6): 453-60. 10.1038/nri2784.PubMedPubMedCentral Geissmann F, Gordon S, Hume DA, Mowat AM, Randolph GJ: Unravelling mononuclear phagocyte heterogeneity. Nat Rev Immunol. 2010, 10 (6): 453-60. 10.1038/nri2784.PubMedPubMedCentral
64.
Zurück zum Zitat Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K: Development of monocytes, macrophages, and dendritic cells. Science. 2010, 327 (5966): 656-61. 10.1126/science.1178331.PubMedPubMedCentral Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K: Development of monocytes, macrophages, and dendritic cells. Science. 2010, 327 (5966): 656-61. 10.1126/science.1178331.PubMedPubMedCentral
65.
Zurück zum Zitat Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo JL, Libby P, Weissleder R, Pittet MJ: The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med. 2007, 204 (12): 3037-47. 10.1084/jem.20070885.PubMedPubMedCentral Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo JL, Libby P, Weissleder R, Pittet MJ: The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med. 2007, 204 (12): 3037-47. 10.1084/jem.20070885.PubMedPubMedCentral
66.
Zurück zum Zitat Saederup N, Chan L, Lira SA, Charo IF: Fractalkine deficiency markedly reduces macrophage accumulation and atherosclerotic lesion formation in CCR2-/- mice: evidence for independent chemokine functions in atherogenesis. Circulation. 2008, 117 (13): 1642-8. 10.1161/CIRCULATIONAHA.107.743872.PubMedPubMedCentral Saederup N, Chan L, Lira SA, Charo IF: Fractalkine deficiency markedly reduces macrophage accumulation and atherosclerotic lesion formation in CCR2-/- mice: evidence for independent chemokine functions in atherogenesis. Circulation. 2008, 117 (13): 1642-8. 10.1161/CIRCULATIONAHA.107.743872.PubMedPubMedCentral
67.
Zurück zum Zitat Combadiere C, Potteaux S, Gao JL, Esposito B, Casanova S, Lee EJ, Debre P, Tedgui A, Murphy PM, Mallat Z: Decreased atherosclerotic lesion formation in CX3CR1/apolipoprotein E double knockout mice. Circulation. 2003, 107 (7): 1009-16. 10.1161/01.CIR.0000057548.68243.42.PubMed Combadiere C, Potteaux S, Gao JL, Esposito B, Casanova S, Lee EJ, Debre P, Tedgui A, Murphy PM, Mallat Z: Decreased atherosclerotic lesion formation in CX3CR1/apolipoprotein E double knockout mice. Circulation. 2003, 107 (7): 1009-16. 10.1161/01.CIR.0000057548.68243.42.PubMed
68.
Zurück zum Zitat Lesnik P, Haskell CA, Charo IF: Decreased atherosclerosis in CX3CR1-/- mice reveals a role for fractalkine in atherogenesis. J Clin Invest. 2003, 111 (3): 333-40.PubMedPubMedCentral Lesnik P, Haskell CA, Charo IF: Decreased atherosclerosis in CX3CR1-/- mice reveals a role for fractalkine in atherogenesis. J Clin Invest. 2003, 111 (3): 333-40.PubMedPubMedCentral
69.
Zurück zum Zitat Teupser D, Pavlides S, Tan M, Gutierrez-Ramos JC, Kolbeck R, Breslow JL: Major reduction of atherosclerosis in fractalkine (CX3CL1)-deficient mice is at the brachiocephalic artery, not the aortic root. Proc Natl Acad Sci USA. 2004, 101 (51): 17795-800. 10.1073/pnas.0408096101.PubMedPubMedCentral Teupser D, Pavlides S, Tan M, Gutierrez-Ramos JC, Kolbeck R, Breslow JL: Major reduction of atherosclerosis in fractalkine (CX3CL1)-deficient mice is at the brachiocephalic artery, not the aortic root. Proc Natl Acad Sci USA. 2004, 101 (51): 17795-800. 10.1073/pnas.0408096101.PubMedPubMedCentral
70.
Zurück zum Zitat 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 (6696): 894-7. 10.1038/29788.PubMed 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 (6696): 894-7. 10.1038/29788.PubMed
71.
Zurück zum Zitat Dawson TC, Kuziel WA, Osahar TA, Maeda N: Absence of CC chemokine receptor-2 reduces atherosclerosis in apolipoprotein E-deficient mice. Atherosclerosis. 1999, 143 (1): 205-11. 10.1016/S0021-9150(98)00318-9.PubMed Dawson TC, Kuziel WA, Osahar TA, Maeda N: Absence of CC chemokine receptor-2 reduces atherosclerosis in apolipoprotein E-deficient mice. Atherosclerosis. 1999, 143 (1): 205-11. 10.1016/S0021-9150(98)00318-9.PubMed
72.
Zurück zum Zitat Kuziel WA, Dawson TC, Quinones M, Garavito E, Chenaux G, Ahuja SS, RL Reddick, Maeda N: CCR5 deficiency is not protective in the early stages of atherogenesis in apoE knockout mice. Atherosclerosis. 2003, 167 (1): 25-32. 10.1016/S0021-9150(02)00382-9.PubMed Kuziel WA, Dawson TC, Quinones M, Garavito E, Chenaux G, Ahuja SS, RL Reddick, Maeda N: CCR5 deficiency is not protective in the early stages of atherogenesis in apoE knockout mice. Atherosclerosis. 2003, 167 (1): 25-32. 10.1016/S0021-9150(02)00382-9.PubMed
73.
Zurück zum Zitat Zernecke A, Liehn EA, Gao JL, Kuziel WA, Murphy PM, Weber C: Deficiency in CCR5 but not CCR1 protects against neointima formation in atherosclerosis-prone mice: involvement of IL-10. Blood. 2006, 107 (11): 4240-3. 10.1182/blood-2005-09-3922.PubMed Zernecke A, Liehn EA, Gao JL, Kuziel WA, Murphy PM, Weber C: Deficiency in CCR5 but not CCR1 protects against neointima formation in atherosclerosis-prone mice: involvement of IL-10. Blood. 2006, 107 (11): 4240-3. 10.1182/blood-2005-09-3922.PubMed
74.
Zurück zum Zitat Combadiere C, Potteaux S, Rodero M, Simon T, Pezard A, Esposito B, Merval R, Proudfoot A, Tedgui A, Mallat Z: Combined inhibition of CCL2, CX3CR1, and CCR5 abrogates Ly6C(hi) and Ly6C(lo) monocytosis and almost abolishes atherosclerosis in hypercholesterolemic mice. Circulation. 2008, 117 (13): 1649-57. 10.1161/CIRCULATIONAHA.107.745091.PubMed Combadiere C, Potteaux S, Rodero M, Simon T, Pezard A, Esposito B, Merval R, Proudfoot A, Tedgui A, Mallat Z: Combined inhibition of CCL2, CX3CR1, and CCR5 abrogates Ly6C(hi) and Ly6C(lo) monocytosis and almost abolishes atherosclerosis in hypercholesterolemic mice. Circulation. 2008, 117 (13): 1649-57. 10.1161/CIRCULATIONAHA.107.745091.PubMed
75.
Zurück zum Zitat Wildgruber M, Lee H, Chudnovskiy A, Yoon TJ, Etzrodt M, Pittet MJ, Nahrendorf M, Croce K, Libby P, Weissleder R, Swirski FK: Monocyte subset dynamics in human atherosclerosis can be profiled with magnetic nano-sensors. PLoS One. 2009, 4 (5): e5663-10.1371/journal.pone.0005663.PubMedPubMedCentral Wildgruber M, Lee H, Chudnovskiy A, Yoon TJ, Etzrodt M, Pittet MJ, Nahrendorf M, Croce K, Libby P, Weissleder R, Swirski FK: Monocyte subset dynamics in human atherosclerosis can be profiled with magnetic nano-sensors. PLoS One. 2009, 4 (5): e5663-10.1371/journal.pone.0005663.PubMedPubMedCentral
76.
Zurück zum Zitat Schlitt A, Heine GH, Blankenberg S, Espinola-Klein C, Dopheide JF, Bickel C, Lackner KJ, Iz M, Meyer J, Darius H, Rupprecht HJ: CD14+CD16+ monocytes in coronary artery disease and their relationship to serum TNF-alpha levels. Thromb Haemost. 2004, 92 (2): 419-24.PubMed Schlitt A, Heine GH, Blankenberg S, Espinola-Klein C, Dopheide JF, Bickel C, Lackner KJ, Iz M, Meyer J, Darius H, Rupprecht HJ: CD14+CD16+ monocytes in coronary artery disease and their relationship to serum TNF-alpha levels. Thromb Haemost. 2004, 92 (2): 419-24.PubMed
77.
Zurück zum Zitat Schirmer SH, Fledderus JO, van der Laan AM, van der Pouw-Kraan TC, Moerland PD, Volger OL, Baggen JM, Bohm M, Piek JJ, Horrevoets AJ, van Royen N: Suppression of inflammatory signaling in monocytes from patients with coronary artery disease. J Mol Cell Cardiol. 2009, 46 (2): 177-85. 10.1016/j.yjmcc.2008.10.029.PubMed Schirmer SH, Fledderus JO, van der Laan AM, van der Pouw-Kraan TC, Moerland PD, Volger OL, Baggen JM, Bohm M, Piek JJ, Horrevoets AJ, van Royen N: Suppression of inflammatory signaling in monocytes from patients with coronary artery disease. J Mol Cell Cardiol. 2009, 46 (2): 177-85. 10.1016/j.yjmcc.2008.10.029.PubMed
78.
Zurück zum Zitat Merino A, Buendia P, Martin-Malo A, Aljama P, Ramirez R, Carracedo J: Senescent CD14+CD16+ Monocytes Exhibit Proinflammatory and Proatherosclerotic Activity. J Immunol. 2011, 186 (3): 1809-15. 10.4049/jimmunol.1001866.PubMed Merino A, Buendia P, Martin-Malo A, Aljama P, Ramirez R, Carracedo J: Senescent CD14+CD16+ Monocytes Exhibit Proinflammatory and Proatherosclerotic Activity. J Immunol. 2011, 186 (3): 1809-15. 10.4049/jimmunol.1001866.PubMed
79.
Zurück zum Zitat Kuwana M, Okazaki Y, Kodama H, Izumi K, Yasuoka H, Ogawa Y, Kawakami Y, Ikeda Y: Human circulating CD14+ monocytes as a source of progenitors that exhibit mesenchymal cell differentiation. J Leukoc Biol. 2003, 74 (5): 833-45. 10.1189/jlb.0403170.PubMed Kuwana M, Okazaki Y, Kodama H, Izumi K, Yasuoka H, Ogawa Y, Kawakami Y, Ikeda Y: Human circulating CD14+ monocytes as a source of progenitors that exhibit mesenchymal cell differentiation. J Leukoc Biol. 2003, 74 (5): 833-45. 10.1189/jlb.0403170.PubMed
80.
Zurück zum Zitat Kamari Y, Shaish A, Shemesh S, Vax E, Grosskopf I, Dotan S, White M, Voronov E, Dinarello CA, Apte RN, Harats D: Reduced atherosclerosis and inflammatory cytokines in apolipoprotein-E-deficient mice lacking bone marrow-derived interleukin-1alpha. Biochem Biophys Res Commun. 2011, 405 (2): 197-203. 10.1016/j.bbrc.2011.01.008.PubMed Kamari Y, Shaish A, Shemesh S, Vax E, Grosskopf I, Dotan S, White M, Voronov E, Dinarello CA, Apte RN, Harats D: Reduced atherosclerosis and inflammatory cytokines in apolipoprotein-E-deficient mice lacking bone marrow-derived interleukin-1alpha. Biochem Biophys Res Commun. 2011, 405 (2): 197-203. 10.1016/j.bbrc.2011.01.008.PubMed
81.
Zurück zum Zitat Hamilton JA: Rheumatoid arthritis: opposing actions of haemopoietic growth factors and slow-acting anti-rheumatic drugs. Lancet. 1993, 342 (8870): 536-9. 10.1016/0140-6736(93)91653-4.PubMed Hamilton JA: Rheumatoid arthritis: opposing actions of haemopoietic growth factors and slow-acting anti-rheumatic drugs. Lancet. 1993, 342 (8870): 536-9. 10.1016/0140-6736(93)91653-4.PubMed
82.
Zurück zum Zitat Hamilton JA: Colony-stimulating factors in inflammation and autoimmunity. Nat Rev Immunol. 2008, 8 (7): 533-44. 10.1038/nri2356.PubMed Hamilton JA: Colony-stimulating factors in inflammation and autoimmunity. Nat Rev Immunol. 2008, 8 (7): 533-44. 10.1038/nri2356.PubMed
83.
Zurück zum Zitat Hashimoto S, Suzuki T, Dong HY, Yamazaki N, Matsushima K: Serial analysis of gene expression in human monocytes and macrophages. Blood. 1999, 94 (3): 837-44.PubMed Hashimoto S, Suzuki T, Dong HY, Yamazaki N, Matsushima K: Serial analysis of gene expression in human monocytes and macrophages. Blood. 1999, 94 (3): 837-44.PubMed
84.
Zurück zum Zitat Smith JD, Trogan E, Ginsberg M, Grigaux C, Tian J, Miyata M: Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and apolipoprotein E. Proc Natl Acad Sci USA. 1995, 92 (18): 8264-8. 10.1073/pnas.92.18.8264.PubMedPubMedCentral Smith JD, Trogan E, Ginsberg M, Grigaux C, Tian J, Miyata M: Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and apolipoprotein E. Proc Natl Acad Sci USA. 1995, 92 (18): 8264-8. 10.1073/pnas.92.18.8264.PubMedPubMedCentral
85.
Zurück zum Zitat Stojakovic M, Krzesz R, Wagner AH, Hecker M: CD154-stimulated GM-CSF release by vascular smooth muscle cells elicits monocyte activation--role in atherogenesis. J Mol Med. 2007, 85 (11): 1229-38. 10.1007/s00109-007-0225-y.PubMed Stojakovic M, Krzesz R, Wagner AH, Hecker M: CD154-stimulated GM-CSF release by vascular smooth muscle cells elicits monocyte activation--role in atherogenesis. J Mol Med. 2007, 85 (11): 1229-38. 10.1007/s00109-007-0225-y.PubMed
86.
Zurück zum Zitat Haghighat A, Weiss D, Whalin MK, Cowan DP, Taylor WR: Granulocyte colony-stimulating factor and granulocyte macrophage colony-stimulating factor exacerbate atherosclerosis in apolipoprotein E-deficient mice. Circulation. 2007, 115 (15): 2049-54. 10.1161/CIRCULATIONAHA.106.665570.PubMed Haghighat A, Weiss D, Whalin MK, Cowan DP, Taylor WR: Granulocyte colony-stimulating factor and granulocyte macrophage colony-stimulating factor exacerbate atherosclerosis in apolipoprotein E-deficient mice. Circulation. 2007, 115 (15): 2049-54. 10.1161/CIRCULATIONAHA.106.665570.PubMed
87.
Zurück zum Zitat Zhu SN, Chen M, Jongstra-Bilen J, Cybulsky MI: GM-CSF regulates intimal cell proliferation in nascent atherosclerotic lesions. J Exp Med. 2009, 206 (10): 2141-9. 10.1084/jem.20090866.PubMedPubMedCentral Zhu SN, Chen M, Jongstra-Bilen J, Cybulsky MI: GM-CSF regulates intimal cell proliferation in nascent atherosclerotic lesions. J Exp Med. 2009, 206 (10): 2141-9. 10.1084/jem.20090866.PubMedPubMedCentral
88.
Zurück zum Zitat McLaren JE, Ramji DP: Interferon gamma: a master regulator of atherosclerosis. Cytokine Growth Factor Rev. 2009, 20 (2): 125-35. 10.1016/j.cytogfr.2008.11.003.PubMed McLaren JE, Ramji DP: Interferon gamma: a master regulator of atherosclerosis. Cytokine Growth Factor Rev. 2009, 20 (2): 125-35. 10.1016/j.cytogfr.2008.11.003.PubMed
89.
Zurück zum Zitat Gupta S, Pablo AM, Jiang X, Wang N, Tall AR, Schindler C: IFN-gamma potentiates atherosclerosis in ApoE knock-out mice. J Clin Invest. 1997, 99 (11): 2752-61. 10.1172/JCI119465.PubMedPubMedCentral Gupta S, Pablo AM, Jiang X, Wang N, Tall AR, Schindler C: IFN-gamma potentiates atherosclerosis in ApoE knock-out mice. J Clin Invest. 1997, 99 (11): 2752-61. 10.1172/JCI119465.PubMedPubMedCentral
90.
Zurück zum Zitat Raisanen-Sokolowski A, Glysing-Jensen T, Koglin J, Russell ME: Reduced transplant arteriosclerosis in murine cardiac allografts placed in interferon-gamma knockout recipients. Am J Pathol. 1998, 152 (2): 359-65.PubMedPubMedCentral Raisanen-Sokolowski A, Glysing-Jensen T, Koglin J, Russell ME: Reduced transplant arteriosclerosis in murine cardiac allografts placed in interferon-gamma knockout recipients. Am J Pathol. 1998, 152 (2): 359-65.PubMedPubMedCentral
91.
Zurück zum Zitat Loke P, Nair MG, Parkinson J, Guiliano D, Blaxter M, Allen JE: IL-4 dependent alternatively-activated macrophages have a distinctive in vivo gene expression phenotype. BMC Immunol. 2002, 3: 7-10.1186/1471-2172-3-7.PubMedPubMedCentral Loke P, Nair MG, Parkinson J, Guiliano D, Blaxter M, Allen JE: IL-4 dependent alternatively-activated macrophages have a distinctive in vivo gene expression phenotype. BMC Immunol. 2002, 3: 7-10.1186/1471-2172-3-7.PubMedPubMedCentral
92.
Zurück zum Zitat Tabas I: Macrophage death and defective inflammation resolution in atherosclerosis. Nat Rev Immunol. 2010, 10 (1): 36-46.PubMedPubMedCentral Tabas I: Macrophage death and defective inflammation resolution in atherosclerosis. Nat Rev Immunol. 2010, 10 (1): 36-46.PubMedPubMedCentral
93.
Zurück zum Zitat Kreider T, Anthony RM, Urban JF, Gause WC: Alternatively activated macrophages in helminth infections. Curr Opin Immunol. 2007, 19 (4): 448-53. 10.1016/j.coi.2007.07.002.PubMedPubMedCentral Kreider T, Anthony RM, Urban JF, Gause WC: Alternatively activated macrophages in helminth infections. Curr Opin Immunol. 2007, 19 (4): 448-53. 10.1016/j.coi.2007.07.002.PubMedPubMedCentral
94.
Zurück zum Zitat Cordeiro-da-Silva A, Tavares J, Araujo N, Cerqueira F, Tomas A, Kong Thoo Lin P, Ouaissi A: Immunological alterations induced by polyamine derivatives on murine splenocytes and human mononuclear cells. Int Immunopharmacol. 2004, 4 (4): 547-56. 10.1016/j.intimp.2004.02.009.PubMed Cordeiro-da-Silva A, Tavares J, Araujo N, Cerqueira F, Tomas A, Kong Thoo Lin P, Ouaissi A: Immunological alterations induced by polyamine derivatives on murine splenocytes and human mononuclear cells. Int Immunopharmacol. 2004, 4 (4): 547-56. 10.1016/j.intimp.2004.02.009.PubMed
95.
Zurück zum Zitat Gerber JS, Mosser DM: Reversing lipopolysaccharide toxicity by ligating the macrophage Fc gamma receptors. J Immunol. 2001, 166 (11): 6861-8.PubMed Gerber JS, Mosser DM: Reversing lipopolysaccharide toxicity by ligating the macrophage Fc gamma receptors. J Immunol. 2001, 166 (11): 6861-8.PubMed
96.
Zurück zum Zitat Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM: Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest. 1998, 101 (4): 890-8. 10.1172/JCI1112.PubMedPubMedCentral Fadok VA, Bratton DL, Konowal A, Freed PW, Westcott JY, Henson PM: Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest. 1998, 101 (4): 890-8. 10.1172/JCI1112.PubMedPubMedCentral
97.
Zurück zum Zitat Sternberg EM: Neural regulation of innate immunity: a coordinated nonspecific host response to pathogens. Nat Rev Immunol. 2006, 6 (4): 318-28. 10.1038/nri1810.PubMedPubMedCentral Sternberg EM: Neural regulation of innate immunity: a coordinated nonspecific host response to pathogens. Nat Rev Immunol. 2006, 6 (4): 318-28. 10.1038/nri1810.PubMedPubMedCentral
98.
Zurück zum Zitat Liu Y, Cousin JM, Hughes J, Van Damme J, Seckl JR, Haslett C, Dransfield I, Savill J, Rossi AG: Glucocorticoids promote nonphlogistic phagocytosis of apoptotic leukocytes. J Immunol. 1999, 162 (6): 3639-46.PubMed Liu Y, Cousin JM, Hughes J, Van Damme J, Seckl JR, Haslett C, Dransfield I, Savill J, Rossi AG: Glucocorticoids promote nonphlogistic phagocytosis of apoptotic leukocytes. J Immunol. 1999, 162 (6): 3639-46.PubMed
99.
Zurück zum Zitat Mackaness GB: The Immunological Basis of Acquired Cellular Resistance. J Exp Med. 1964, 120: 105-20. 10.1084/jem.120.1.105.PubMedPubMedCentral Mackaness GB: The Immunological Basis of Acquired Cellular Resistance. J Exp Med. 1964, 120: 105-20. 10.1084/jem.120.1.105.PubMedPubMedCentral
100.
Zurück zum Zitat Yan ZQ, Hansson GK: Innate immunity, macrophage activation, and atherosclerosis. Immunol Rev. 2007, 219: 187-203. 10.1111/j.1600-065X.2007.00554.x.PubMed Yan ZQ, Hansson GK: Innate immunity, macrophage activation, and atherosclerosis. Immunol Rev. 2007, 219: 187-203. 10.1111/j.1600-065X.2007.00554.x.PubMed
101.
Zurück zum Zitat Medzhitov R: Toll-like receptors and innate immunity. Nat Rev Immunol. 2001, 1 (2): 135-45. 10.1038/35100529.PubMed Medzhitov R: Toll-like receptors and innate immunity. Nat Rev Immunol. 2001, 1 (2): 135-45. 10.1038/35100529.PubMed
102.
Zurück zum Zitat Medzhitov R, Preston-Hurlburt P, Janeway CA: A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997, 388 (6640): 394-7. 10.1038/41131.PubMed Medzhitov R, Preston-Hurlburt P, Janeway CA: A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997, 388 (6640): 394-7. 10.1038/41131.PubMed
103.
Zurück zum Zitat Kim TW, Febbraio M, Robinet P, Dugar B, Greene D, Cerny A, Latz E, Gilmour R, Staschke K, Chisolm G, Fox PL, Dicorleto PE, Smith JD, Li X: The Critical Role of IL-1 Receptor-Associated Kinase 4-Mediated NF-{kappa}B Activation in Modified Low-Density Lipoprotein-Induced Inflammatory Gene Expression and Atherosclerosis. J Immunol. 2011, 186 (5): 2871-80. 10.4049/jimmunol.1002242.PubMedPubMedCentral Kim TW, Febbraio M, Robinet P, Dugar B, Greene D, Cerny A, Latz E, Gilmour R, Staschke K, Chisolm G, Fox PL, Dicorleto PE, Smith JD, Li X: The Critical Role of IL-1 Receptor-Associated Kinase 4-Mediated NF-{kappa}B Activation in Modified Low-Density Lipoprotein-Induced Inflammatory Gene Expression and Atherosclerosis. J Immunol. 2011, 186 (5): 2871-80. 10.4049/jimmunol.1002242.PubMedPubMedCentral
104.
Zurück zum Zitat Wright SD, Ramos RA, Tobias PS, Ulevitch RJ, Mathison JC: CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science. 1990, 249 (4975): 1431-3. 10.1126/science.1698311.PubMed Wright SD, Ramos RA, Tobias PS, Ulevitch RJ, Mathison JC: CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science. 1990, 249 (4975): 1431-3. 10.1126/science.1698311.PubMed
105.
Zurück zum Zitat Tobias PS, Soldau K, Kline L, Lee JD, Kato K, Martin TP, Ulevitch RJ: Cross-linking of lipopolysaccharide (LPS) to CD14 on THP-1 cells mediated by LPS-binding protein. J Immunol. 1993, 150 (7): 3011-21.PubMed Tobias PS, Soldau K, Kline L, Lee JD, Kato K, Martin TP, Ulevitch RJ: Cross-linking of lipopolysaccharide (LPS) to CD14 on THP-1 cells mediated by LPS-binding protein. J Immunol. 1993, 150 (7): 3011-21.PubMed
106.
Zurück zum Zitat Hoebe K, Georgel P, Rutschmann S, Du X, Mudd S, Crozat K, Sovath S, Shamel L, Hartung T, Zähringer U, Beutler B: CD36 is a sensor of diacylglycerides. Nature. 2005, 433 (7025): 523-7. 10.1038/nature03253.PubMed Hoebe K, Georgel P, Rutschmann S, Du X, Mudd S, Crozat K, Sovath S, Shamel L, Hartung T, Zähringer U, Beutler B: CD36 is a sensor of diacylglycerides. Nature. 2005, 433 (7025): 523-7. 10.1038/nature03253.PubMed
107.
Zurück zum Zitat Miller YI, Choi SH, Wiesner P, Fang L, Harkewicz R, Hartvigsen K, Boullier A, Gonen A, Diehl CJ, Que X, Montano E, Shaw PX, Tsimikas S, Binder CJ, Witztum JL: Oxidation-specific epitopes are danger-associated molecular patterns recognized by pattern recognition receptors of innate immunity. Circ Res. 2011, 108 (2): 235-48. 10.1161/CIRCRESAHA.110.223875.PubMedPubMedCentral Miller YI, Choi SH, Wiesner P, Fang L, Harkewicz R, Hartvigsen K, Boullier A, Gonen A, Diehl CJ, Que X, Montano E, Shaw PX, Tsimikas S, Binder CJ, Witztum JL: Oxidation-specific epitopes are danger-associated molecular patterns recognized by pattern recognition receptors of innate immunity. Circ Res. 2011, 108 (2): 235-48. 10.1161/CIRCRESAHA.110.223875.PubMedPubMedCentral
108.
Zurück zum Zitat Li M, Carpio DF, Zheng Y, Bruzzo P, Singh V, Ouaaz F, Medzhitov RM, Beg AA: An essential role of the NF-kappa B/Toll-like receptor pathway in induction of inflammatory and tissue-repair gene expression by necrotic cells. J Immunol. 2001, 166 (12): 7128-35.PubMed Li M, Carpio DF, Zheng Y, Bruzzo P, Singh V, Ouaaz F, Medzhitov RM, Beg AA: An essential role of the NF-kappa B/Toll-like receptor pathway in induction of inflammatory and tissue-repair gene expression by necrotic cells. J Immunol. 2001, 166 (12): 7128-35.PubMed
109.
Zurück zum Zitat Kawakami A, Osaka M, Aikawa M, Uematsu S, Akira S, Libby P, Shimokado K, Sacks FM, Yoshida M: Toll-like receptor 2 mediates apolipoprotein CIII-induced monocyte activation. Circ Res. 2008, 103 (12): 1402-9. 10.1161/CIRCRESAHA.108.178426.PubMedPubMedCentral Kawakami A, Osaka M, Aikawa M, Uematsu S, Akira S, Libby P, Shimokado K, Sacks FM, Yoshida M: Toll-like receptor 2 mediates apolipoprotein CIII-induced monocyte activation. Circ Res. 2008, 103 (12): 1402-9. 10.1161/CIRCRESAHA.108.178426.PubMedPubMedCentral
110.
Zurück zum Zitat Cheng N, He R, Tian J, Ye P, Ye R: Cutting edge: TLR2 is a functional receptor for acute-phase serum amyloid A. J Immunol. 2008, 181 (1): 22-6.PubMedPubMedCentral Cheng N, He R, Tian J, Ye P, Ye R: Cutting edge: TLR2 is a functional receptor for acute-phase serum amyloid A. J Immunol. 2008, 181 (1): 22-6.PubMedPubMedCentral
111.
Zurück zum Zitat Kim S, Takahashi H, Lin WW, Descargues P, Grivennikov S, Kim Y, Luo JL, Karin M: Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature. 2009, 457 (7225): 102-6. 10.1038/nature07623.PubMedPubMedCentral Kim S, Takahashi H, Lin WW, Descargues P, Grivennikov S, Kim Y, Luo JL, Karin M: Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature. 2009, 457 (7225): 102-6. 10.1038/nature07623.PubMedPubMedCentral
112.
Zurück zum Zitat Seimon TA, Nadolski MJ, Liao X, Magallon J, Nguyen M, Feric NT, Koschinsky ML, Harkewicz R, Witztum JL, Tsimikas S, Golenbock D, Moore KJ, Tabas I: Atherogenic lipids and lipoproteins trigger CD36-TLR2-dependent apoptosis in macrophages undergoing endoplasmic reticulum stress. Cell Metab. 2010, 12 (5): 467-82. 10.1016/j.cmet.2010.09.010.PubMedPubMedCentral Seimon TA, Nadolski MJ, Liao X, Magallon J, Nguyen M, Feric NT, Koschinsky ML, Harkewicz R, Witztum JL, Tsimikas S, Golenbock D, Moore KJ, Tabas I: Atherogenic lipids and lipoproteins trigger CD36-TLR2-dependent apoptosis in macrophages undergoing endoplasmic reticulum stress. Cell Metab. 2010, 12 (5): 467-82. 10.1016/j.cmet.2010.09.010.PubMedPubMedCentral
113.
Zurück zum Zitat Scheibner K, Lutz M, Boodoo S, Fenton M, Powell J, Horton M: Hyaluronan fragments act as an endogenous danger signal by engaging TLR2. J Immunol. 2006, 177 (2): 1272-81.PubMed Scheibner K, Lutz M, Boodoo S, Fenton M, Powell J, Horton M: Hyaluronan fragments act as an endogenous danger signal by engaging TLR2. J Immunol. 2006, 177 (2): 1272-81.PubMed
114.
Zurück zum Zitat Schaefer L, Babelova A, Kiss E, Hausser HJ, Baliova M, Krzyzankova M, Marsche G, Young MF, Mihalik D, Gotte M, Malle E, Schaefer RM, Grone HJ: The matrix component biglycan is proinflammatory and signals through Toll-like receptors 4 and 2 in macrophages. J Clin Invest. 2005, 115 (8): 2223-33. 10.1172/JCI23755.PubMedPubMedCentral Schaefer L, Babelova A, Kiss E, Hausser HJ, Baliova M, Krzyzankova M, Marsche G, Young MF, Mihalik D, Gotte M, Malle E, Schaefer RM, Grone HJ: The matrix component biglycan is proinflammatory and signals through Toll-like receptors 4 and 2 in macrophages. J Clin Invest. 2005, 115 (8): 2223-33. 10.1172/JCI23755.PubMedPubMedCentral
115.
Zurück zum Zitat Holvoet P, Davey PC, De Keyzer D, Doukouré M, Deridder E, Bochaton-Piallat M-L, Gabbiani G, Beaufort E, Bishay K, Andrieux N, Benhabilès N, Marguerie G: Oxidized low-density lipoprotein correlates positively with toll-like receptor 2 and interferon regulatory factor-1 and inversely with superoxide dismutase-1 expression: studies in hypercholesterolemic swine and THP-1 cells. Arterioscler Thromb Vasc Biol. 2006, 26 (7): 1558-65. 10.1161/01.ATV.0000226553.01555.02.PubMed Holvoet P, Davey PC, De Keyzer D, Doukouré M, Deridder E, Bochaton-Piallat M-L, Gabbiani G, Beaufort E, Bishay K, Andrieux N, Benhabilès N, Marguerie G: Oxidized low-density lipoprotein correlates positively with toll-like receptor 2 and interferon regulatory factor-1 and inversely with superoxide dismutase-1 expression: studies in hypercholesterolemic swine and THP-1 cells. Arterioscler Thromb Vasc Biol. 2006, 26 (7): 1558-65. 10.1161/01.ATV.0000226553.01555.02.PubMed
116.
Zurück zum Zitat Xu XH, Shah PK, Faure E, Equils O, Thomas L, Fishbein MC, Luthringer D, Xu XP, Rajavashisth TB, Yano J, Kaul S, Arditi M: Toll-like receptor-4 is expressed by macrophages in murine and human lipid-rich atherosclerotic plaques and upregulated by oxidized LDL. Circulation. 2001, 104 (25): 3103-8. 10.1161/hc5001.100631.PubMed Xu XH, Shah PK, Faure E, Equils O, Thomas L, Fishbein MC, Luthringer D, Xu XP, Rajavashisth TB, Yano J, Kaul S, Arditi M: Toll-like receptor-4 is expressed by macrophages in murine and human lipid-rich atherosclerotic plaques and upregulated by oxidized LDL. Circulation. 2001, 104 (25): 3103-8. 10.1161/hc5001.100631.PubMed
117.
Zurück zum Zitat Asea A, Rehli M, Kabingu E, Boch JA, Bare O, Auron PE, Stevenson MA, Calderwood SK: Novel signal transduction pathway utilized by extracellular HSP70: role of toll-like receptor (TLR) 2 and TLR4. J Biol Chem. 2002, 277 (17): 15028-34. 10.1074/jbc.M200497200.PubMed Asea A, Rehli M, Kabingu E, Boch JA, Bare O, Auron PE, Stevenson MA, Calderwood SK: Novel signal transduction pathway utilized by extracellular HSP70: role of toll-like receptor (TLR) 2 and TLR4. J Biol Chem. 2002, 277 (17): 15028-34. 10.1074/jbc.M200497200.PubMed
118.
Zurück zum Zitat Guillot L, Balloy V, McCormack FX, Golenbock DT, Chignard M, Si-Tahar M: Cutting edge: the immunostimulatory activity of the lung surfactant protein-A involves Toll-like receptor 4. J Immunol. 2002, 168 (12): 5989-92.PubMed Guillot L, Balloy V, McCormack FX, Golenbock DT, Chignard M, Si-Tahar M: Cutting edge: the immunostimulatory activity of the lung surfactant protein-A involves Toll-like receptor 4. J Immunol. 2002, 168 (12): 5989-92.PubMed
119.
Zurück zum Zitat Midwood K, Sacre S, Piccinini AM, Inglis J, Trebaul A, Chan E, Drexler S, Sofat N, Kashiwagi M, Orend G, Brennan F, Foxwell B: Tenascin-C is an endogenous activator of Toll-like receptor 4 that is essential for maintaining inflammation in arthritic joint disease. Nat Med. 2009, 15 (7): 774-80. 10.1038/nm.1987.PubMed Midwood K, Sacre S, Piccinini AM, Inglis J, Trebaul A, Chan E, Drexler S, Sofat N, Kashiwagi M, Orend G, Brennan F, Foxwell B: Tenascin-C is an endogenous activator of Toll-like receptor 4 that is essential for maintaining inflammation in arthritic joint disease. Nat Med. 2009, 15 (7): 774-80. 10.1038/nm.1987.PubMed
120.
Zurück zum Zitat Smiley ST, King JA, Hancock WW: Fibrinogen stimulates macrophage chemokine secretion through toll-like receptor 4. J Immunol. 2001, 167 (5): 2887-94.PubMed Smiley ST, King JA, Hancock WW: Fibrinogen stimulates macrophage chemokine secretion through toll-like receptor 4. J Immunol. 2001, 167 (5): 2887-94.PubMed
121.
Zurück zum Zitat Okamura Y, Watari M, Jerud ES, Young DW, Ishizaka ST, Rose J, Chow JC, Strauss JF: The extra domain A of fibronectin activates Toll-like receptor 4. J Biol Chem. 2001, 276 (13): 10229-33. 10.1074/jbc.M100099200.PubMed Okamura Y, Watari M, Jerud ES, Young DW, Ishizaka ST, Rose J, Chow JC, Strauss JF: The extra domain A of fibronectin activates Toll-like receptor 4. J Biol Chem. 2001, 276 (13): 10229-33. 10.1074/jbc.M100099200.PubMed
122.
Zurück zum Zitat Kodaira Y, Nair SK, Wrenshall LE, Gilboa E, Platt JL: Phenotypic and functional maturation of dendritic cells mediated by heparan sulfate. J Immunol. 2000, 165 (3): 1599-604.PubMed Kodaira Y, Nair SK, Wrenshall LE, Gilboa E, Platt JL: Phenotypic and functional maturation of dendritic cells mediated by heparan sulfate. J Immunol. 2000, 165 (3): 1599-604.PubMed
123.
Zurück zum Zitat Biragyn A, Ruffini PA, Leifer CA, Klyushnenkova E, Shakhov A, Chertov O, Shirakawa AK, Farber JM, Segal DM, Oppenheim JJ, Kwak LW: Toll-like receptor 4-dependent activation of dendritic cells by beta-defensin 2. Science. 2002, 298 (5595): 1025-9. 10.1126/science.1075565.PubMed Biragyn A, Ruffini PA, Leifer CA, Klyushnenkova E, Shakhov A, Chertov O, Shirakawa AK, Farber JM, Segal DM, Oppenheim JJ, Kwak LW: Toll-like receptor 4-dependent activation of dendritic cells by beta-defensin 2. Science. 2002, 298 (5595): 1025-9. 10.1126/science.1075565.PubMed
124.
Zurück zum Zitat Cole JE, Navin TJ, Cross AJ, Goddard ME, Alexopoulou L, Mitra AT, Davies AH, Flavell RA, Feldmann M, Monaco C: From the Cover: Unexpected protective role for Toll-like receptor 3 in the arterial wall. Proc Natl Acad Sci USA. 2011, 108 (6): 2372-7. 10.1073/pnas.1018515108.PubMedPubMedCentral Cole JE, Navin TJ, Cross AJ, Goddard ME, Alexopoulou L, Mitra AT, Davies AH, Flavell RA, Feldmann M, Monaco C: From the Cover: Unexpected protective role for Toll-like receptor 3 in the arterial wall. Proc Natl Acad Sci USA. 2011, 108 (6): 2372-7. 10.1073/pnas.1018515108.PubMedPubMedCentral
125.
Zurück zum Zitat H. Ni, Capodici J, Lamphier M, Weissman D: mRNA is an endogenous ligand for Toll-like receptor 3. J Biol Chem. 2004, 279 (13): 12542-50. H. Ni, Capodici J, Lamphier M, Weissman D: mRNA is an endogenous ligand for Toll-like receptor 3. J Biol Chem. 2004, 279 (13): 12542-50.
126.
Zurück zum Zitat Boule MW, Broughton C, Mackay F, Akira S, Marshak-Rothstein A, Rifkin IR: Toll-like receptor 9-dependent and -independent dendritic cell activation by chromatin-immunoglobulin G complexes. J Exp Med. 2004, 199 (12): 1631-40. 10.1084/jem.20031942.PubMedPubMedCentral Boule MW, Broughton C, Mackay F, Akira S, Marshak-Rothstein A, Rifkin IR: Toll-like receptor 9-dependent and -independent dendritic cell activation by chromatin-immunoglobulin G complexes. J Exp Med. 2004, 199 (12): 1631-40. 10.1084/jem.20031942.PubMedPubMedCentral
127.
Zurück zum Zitat Leadbetter E, Rifkin I, Hohlbaum A, Beaudette B, Shlomchik M, Marshak-Rothstein A: Chromatin-IgG complexes activate B cells by dual engagement of IgM and Toll-like receptors. Nature. 2002, 416 (6881): 603-7. 10.1038/416603a.PubMed Leadbetter E, Rifkin I, Hohlbaum A, Beaudette B, Shlomchik M, Marshak-Rothstein A: Chromatin-IgG complexes activate B cells by dual engagement of IgM and Toll-like receptors. Nature. 2002, 416 (6881): 603-7. 10.1038/416603a.PubMed
128.
Zurück zum Zitat Stewart CR, Stuart LM, Wilkinson K, van Gils JM, Deng J, Halle A, Rayner KJ, Boyer L, Zhong R, Frazier WA, Lacy-Hulbert A, El Khoury J, Golenbock DT, Moore KJ: CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat Immunol. 2010, 11 (2): 155-61. 10.1038/ni.1836.PubMedPubMedCentral Stewart CR, Stuart LM, Wilkinson K, van Gils JM, Deng J, Halle A, Rayner KJ, Boyer L, Zhong R, Frazier WA, Lacy-Hulbert A, El Khoury J, Golenbock DT, Moore KJ: CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat Immunol. 2010, 11 (2): 155-61. 10.1038/ni.1836.PubMedPubMedCentral
129.
Zurück zum Zitat Walton KA, Hsieh X, Gharavi N, Wang S, Wang G, Yeh M, Cole AL, Berliner JA: Receptors involved in the oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine-mediated synthesis of interleukin-8. A role for Toll-like receptor 4 and a glycosylphosphatidylinositol-anchored protein. J Biol Chem. 2003, 278 (32): 29661-6. 10.1074/jbc.M300738200.PubMed Walton KA, Hsieh X, Gharavi N, Wang S, Wang G, Yeh M, Cole AL, Berliner JA: Receptors involved in the oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine-mediated synthesis of interleukin-8. A role for Toll-like receptor 4 and a glycosylphosphatidylinositol-anchored protein. J Biol Chem. 2003, 278 (32): 29661-6. 10.1074/jbc.M300738200.PubMed
130.
Zurück zum Zitat Walton KA, Cole AL, Yeh M, Subbanagounder G, Krutzik SR, Modlin RL, Lucas RM, Nakai J, Smart EJ, Vora DK, Berliner JA: Specific phospholipid oxidation products inhibit ligand activation of toll-like receptors 4 and 2. Arterioscler Thromb Vasc Biol. 2003, 23 (7): 1197-203. 10.1161/01.ATV.0000079340.80744.B8.PubMed Walton KA, Cole AL, Yeh M, Subbanagounder G, Krutzik SR, Modlin RL, Lucas RM, Nakai J, Smart EJ, Vora DK, Berliner JA: Specific phospholipid oxidation products inhibit ligand activation of toll-like receptors 4 and 2. Arterioscler Thromb Vasc Biol. 2003, 23 (7): 1197-203. 10.1161/01.ATV.0000079340.80744.B8.PubMed
131.
Zurück zum Zitat Bae YS, Lee JH, Choi SH, Kim S, Almazan F, Witztum JL, Miller YI: Macrophages generate reactive oxygen species in response to minimally oxidized low-density lipoprotein: toll-like receptor 4- and spleen tyrosine kinase-dependent activation of NADPH oxidase 2. Circ Res. 2009, 104 (2): 210-8. 10.1161/CIRCRESAHA.108.181040. 21p following 218PubMedPubMedCentral Bae YS, Lee JH, Choi SH, Kim S, Almazan F, Witztum JL, Miller YI: Macrophages generate reactive oxygen species in response to minimally oxidized low-density lipoprotein: toll-like receptor 4- and spleen tyrosine kinase-dependent activation of NADPH oxidase 2. Circ Res. 2009, 104 (2): 210-8. 10.1161/CIRCRESAHA.108.181040. 21p following 218PubMedPubMedCentral
132.
Zurück zum Zitat Edfeldt K, Swedenborg J, Hansson G, Yan Z: Expression of toll-like receptors in human atherosclerotic lesions: a possible pathway for plaque activation. Circulation. 2002, 105 (10): 1158-61.PubMed Edfeldt K, Swedenborg J, Hansson G, Yan Z: Expression of toll-like receptors in human atherosclerotic lesions: a possible pathway for plaque activation. Circulation. 2002, 105 (10): 1158-61.PubMed
133.
Zurück zum Zitat Schoneveld A, Hoefer I, Sluijter J, Laman J, de Kleijn D, Pasterkamp G: Atherosclerotic lesion development and Toll like receptor 2 and 4 responsiveness. Atherosclerosis. 2008, 197 (1): 95-104. 10.1016/j.atherosclerosis.2007.08.004.PubMed Schoneveld A, Hoefer I, Sluijter J, Laman J, de Kleijn D, Pasterkamp G: Atherosclerotic lesion development and Toll like receptor 2 and 4 responsiveness. Atherosclerosis. 2008, 197 (1): 95-104. 10.1016/j.atherosclerosis.2007.08.004.PubMed
134.
Zurück zum Zitat Geng H, Lu H, Zhang L, Zhang H, Zhou L, Wang H, Zhong R: Increased expression of Toll like receptor 4 on peripheral-blood mononuclear cells in patients with coronary arteriosclerosis disease. Clin Exp Immunol. 2006, 143 (2): 269-73. 10.1111/j.1365-2249.2005.02982.x.PubMedPubMedCentral Geng H, Lu H, Zhang L, Zhang H, Zhou L, Wang H, Zhong R: Increased expression of Toll like receptor 4 on peripheral-blood mononuclear cells in patients with coronary arteriosclerosis disease. Clin Exp Immunol. 2006, 143 (2): 269-73. 10.1111/j.1365-2249.2005.02982.x.PubMedPubMedCentral
135.
Zurück zum Zitat Methe H, Kim J, Kofler S, Weis M, Nabauer M, Koglin J: Expansion of circulating Toll-like receptor 4-positive monocytes in patients with acute coronary syndrome. Circulation. 2005, 111 (20): 2654-61. 10.1161/CIRCULATIONAHA.104.498865.PubMed Methe H, Kim J, Kofler S, Weis M, Nabauer M, Koglin J: Expansion of circulating Toll-like receptor 4-positive monocytes in patients with acute coronary syndrome. Circulation. 2005, 111 (20): 2654-61. 10.1161/CIRCULATIONAHA.104.498865.PubMed
136.
Zurück zum Zitat Shiraki R, Inoue N, Kobayashi S, Ejiri J, Otsui K, Honjo T, Takahashi M, Hirata K, Yokoyama M, Kawashima S: Toll-like receptor 4 expressions on peripheral blood monocytes were enhanced in coronary artery disease even in patients with low C-reactive protein. Life Sci. 2006, 80 (1): 59-66. 10.1016/j.lfs.2006.08.027.PubMed Shiraki R, Inoue N, Kobayashi S, Ejiri J, Otsui K, Honjo T, Takahashi M, Hirata K, Yokoyama M, Kawashima S: Toll-like receptor 4 expressions on peripheral blood monocytes were enhanced in coronary artery disease even in patients with low C-reactive protein. Life Sci. 2006, 80 (1): 59-66. 10.1016/j.lfs.2006.08.027.PubMed
137.
Zurück zum Zitat Kuwahata S, Fujita S, Orihara K, Hamasaki S, Oba R, Hirai H, Nagata K, Ishida S, Kataoka T, Oketani N, Ichiki H, Iriki Y, Saihara K, Okui H, Ninomiya Y, Tei C: High expression level of Toll-like receptor 2 on monocytes is an important risk factor for arteriosclerotic disease. Atherosclerosis. 2009 Kuwahata S, Fujita S, Orihara K, Hamasaki S, Oba R, Hirai H, Nagata K, Ishida S, Kataoka T, Oketani N, Ichiki H, Iriki Y, Saihara K, Okui H, Ninomiya Y, Tei C: High expression level of Toll-like receptor 2 on monocytes is an important risk factor for arteriosclerotic disease. Atherosclerosis. 2009
138.
Zurück zum Zitat Ashida K, Miyazaki K, Takayama E, Tsujimoto H, Ayaori M, Yakushiji T, Iwamoto N, Yonemura A, Isoda K, Mochizuki H, Hiraide H, Kusuhara M, Ohsuzu F: Characterization of the expression of TLR2 (toll-like receptor 2) and TLR4 on circulating monocytes in coronary artery disease. J Atheroscler Thromb. 2005, 12 (1): 53-60.PubMed Ashida K, Miyazaki K, Takayama E, Tsujimoto H, Ayaori M, Yakushiji T, Iwamoto N, Yonemura A, Isoda K, Mochizuki H, Hiraide H, Kusuhara M, Ohsuzu F: Characterization of the expression of TLR2 (toll-like receptor 2) and TLR4 on circulating monocytes in coronary artery disease. J Atheroscler Thromb. 2005, 12 (1): 53-60.PubMed
139.
Zurück zum Zitat Liuzzo G, Angiolillo DJ, Buffon A, Rizzello V, Colizzi C, Ginnetti F, Biasucci LM, Maseri A: Enhanced response of blood monocytes to in vitro lipopolysaccharide-challenge in patients with recurrent unstable angina. Circulation. 2001, 103 (18): 2236-41.PubMed Liuzzo G, Angiolillo DJ, Buffon A, Rizzello V, Colizzi C, Ginnetti F, Biasucci LM, Maseri A: Enhanced response of blood monocytes to in vitro lipopolysaccharide-challenge in patients with recurrent unstable angina. Circulation. 2001, 103 (18): 2236-41.PubMed
140.
Zurück zum Zitat Versteeg D, Hoefer IE, Schoneveld AH, De Kleijn DPV, Busser E, Strijder C, Emons M, Stella PR, Doevendans PA, Pasterkamp G: Monocyte toll-like receptor 2 and 4 responses and expression following percutaneous coronary intervention: association with lesion stenosis and fractional flow reserve. Heart. 2008, 94 (6): 770-776. 10.1136/hrt.2007.117259.PubMed Versteeg D, Hoefer IE, Schoneveld AH, De Kleijn DPV, Busser E, Strijder C, Emons M, Stella PR, Doevendans PA, Pasterkamp G: Monocyte toll-like receptor 2 and 4 responses and expression following percutaneous coronary intervention: association with lesion stenosis and fractional flow reserve. Heart. 2008, 94 (6): 770-776. 10.1136/hrt.2007.117259.PubMed
141.
Zurück zum Zitat Michelsen KS, Wong MH, Shah PK, Zhang W, Yano J, Doherty TM, Akira S, Rajavashisth TB, Arditi M: 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 USA. 2004, 101 (29): 10679-84. 10.1073/pnas.0403249101.PubMedPubMedCentral Michelsen KS, Wong MH, Shah PK, Zhang W, Yano J, Doherty TM, Akira S, Rajavashisth TB, Arditi M: 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 USA. 2004, 101 (29): 10679-84. 10.1073/pnas.0403249101.PubMedPubMedCentral
142.
Zurück zum Zitat Bjorkbacka H, Kunjathoor VV, Moore KJ, Koehn S, Ordija CM, Lee MA, Means T, Halmen K, Luster AD, Golenbock DT, Freeman MW: Reduced atherosclerosis in MyD88-null mice links elevated serum cholesterol levels to activation of innate immunity signaling pathways. Nat Med. 2004, 10 (4): 416-21. 10.1038/nm1008.PubMed Bjorkbacka H, Kunjathoor VV, Moore KJ, Koehn S, Ordija CM, Lee MA, Means T, Halmen K, Luster AD, Golenbock DT, Freeman MW: Reduced atherosclerosis in MyD88-null mice links elevated serum cholesterol levels to activation of innate immunity signaling pathways. Nat Med. 2004, 10 (4): 416-21. 10.1038/nm1008.PubMed
143.
Zurück zum Zitat Monaco C, Gregan SM, Navin TJ, Foxwell BM, Davies AH, Feldmann M: Toll-like receptor-2 mediates inflammation and matrix degradation in human atherosclerosis. Circulation. 2009, 120 (24): 2462-9. 10.1161/CIRCULATIONAHA.109.851881.PubMed Monaco C, Gregan SM, Navin TJ, Foxwell BM, Davies AH, Feldmann M: Toll-like receptor-2 mediates inflammation and matrix degradation in human atherosclerosis. Circulation. 2009, 120 (24): 2462-9. 10.1161/CIRCULATIONAHA.109.851881.PubMed
144.
Zurück zum Zitat Ishida BY, Blanche PJ, Nichols AV, Yashar M, Paigen B: Effects of atherogenic diet consumption on lipoproteins in mouse strains C57BL/6 and C3H. J Lipid Res. 1991, 32 (4): 559-68.PubMed Ishida BY, Blanche PJ, Nichols AV, Yashar M, Paigen B: Effects of atherogenic diet consumption on lipoproteins in mouse strains C57BL/6 and C3H. J Lipid Res. 1991, 32 (4): 559-68.PubMed
145.
Zurück zum Zitat Nishina P, Wang J, Toyofuku W, Kuypers F, Ishida B, Paigen B: Atherosclerosis and plasma and liver lipids in nine inbred strains of mice. Lipids. 1993, 28 (7): 599-605. 10.1007/BF02536053.PubMed Nishina P, Wang J, Toyofuku W, Kuypers F, Ishida B, Paigen B: Atherosclerosis and plasma and liver lipids in nine inbred strains of mice. Lipids. 1993, 28 (7): 599-605. 10.1007/BF02536053.PubMed
146.
Zurück zum Zitat Mullick AE, Tobias PS, Curtiss LK: Modulation of atherosclerosis in mice by Toll-like receptor 2. J Clin Invest. 2005, 115 (11): 3149-56. 10.1172/JCI25482.PubMedPubMedCentral Mullick AE, Tobias PS, Curtiss LK: Modulation of atherosclerosis in mice by Toll-like receptor 2. J Clin Invest. 2005, 115 (11): 3149-56. 10.1172/JCI25482.PubMedPubMedCentral
147.
Zurück zum Zitat Madan M, Amar S: Toll-like receptor-2 mediates diet and/or pathogen associated atherosclerosis: proteomic findings. PLoS ONE. 2008, 3 (9): e3204-10.1371/journal.pone.0003204.PubMedPubMedCentral Madan M, Amar S: Toll-like receptor-2 mediates diet and/or pathogen associated atherosclerosis: proteomic findings. PLoS ONE. 2008, 3 (9): e3204-10.1371/journal.pone.0003204.PubMedPubMedCentral
148.
Zurück zum Zitat Liu X, Ukai T, Yumoto H, Davey M, Goswami S, Gibson FC, Genco CA: Toll-like receptor 2 plays a critical role in the progression of atherosclerosis that is independent of dietary lipids. Atherosclerosis. 2008, 196 (1): 146-54. 10.1016/j.atherosclerosis.2007.03.025.PubMedPubMedCentral Liu X, Ukai T, Yumoto H, Davey M, Goswami S, Gibson FC, Genco CA: Toll-like receptor 2 plays a critical role in the progression of atherosclerosis that is independent of dietary lipids. Atherosclerosis. 2008, 196 (1): 146-54. 10.1016/j.atherosclerosis.2007.03.025.PubMedPubMedCentral
149.
Zurück zum Zitat Vink A, Schoneveld AH, van der Meer JJ, van Middelaar BJ, Sluijter JP, Smeets MB, Quax PH, Lim SK, Borst C, Pasterkamp G, de Kleijn DP: In vivo evidence for a role of toll-like receptor 4 in the development of intimal lesions. Circulation. 2002, 106 (15): 1985-90. 10.1161/01.CIR.0000032146.75113.EE.PubMed Vink A, Schoneveld AH, van der Meer JJ, van Middelaar BJ, Sluijter JP, Smeets MB, Quax PH, Lim SK, Borst C, Pasterkamp G, de Kleijn DP: In vivo evidence for a role of toll-like receptor 4 in the development of intimal lesions. Circulation. 2002, 106 (15): 1985-90. 10.1161/01.CIR.0000032146.75113.EE.PubMed
150.
Zurück zum Zitat Schoneveld AH, Oude Nijhuis MM, van Middelaar B, Laman JD, de Kleijn DP, Pasterkamp G: Toll-like receptor 2 stimulation induces intimal hyperplasia and atherosclerotic lesion development. Cardiovasc Res. 2005, 66 (1): 162-9. 10.1016/j.cardiores.2004.12.016.PubMed Schoneveld AH, Oude Nijhuis MM, van Middelaar B, Laman JD, de Kleijn DP, Pasterkamp G: Toll-like receptor 2 stimulation induces intimal hyperplasia and atherosclerotic lesion development. Cardiovasc Res. 2005, 66 (1): 162-9. 10.1016/j.cardiores.2004.12.016.PubMed
151.
Zurück zum Zitat Shinohara M, Hirata K, Yamashita T, Takaya T, Sasaki N, Shiraki R, Ueyama T, Emoto N, Inoue N, Yokoyama M, Kawashima S: Local overexpression of toll-like receptors at the vessel wall induces atherosclerotic lesion formation: synergism of TLR2 and TLR4. Arterioscler Thromb Vasc Biol. 2007, 27 (11): 2384-91. 10.1161/ATVBAHA.106.139253.PubMed Shinohara M, Hirata K, Yamashita T, Takaya T, Sasaki N, Shiraki R, Ueyama T, Emoto N, Inoue N, Yokoyama M, Kawashima S: Local overexpression of toll-like receptors at the vessel wall induces atherosclerotic lesion formation: synergism of TLR2 and TLR4. Arterioscler Thromb Vasc Biol. 2007, 27 (11): 2384-91. 10.1161/ATVBAHA.106.139253.PubMed
152.
Zurück zum Zitat Shi W, Wang NJ, Shih DM, Sun VZ, Wang X, Lusis AJ: Determinants of atherosclerosis susceptibility in the C3H and C57BL/6 mouse model: evidence for involvement of endothelial cells but not blood cells or cholesterol metabolism. Circ Res. 2000, 86 (10): 1078-84.PubMed Shi W, Wang NJ, Shih DM, Sun VZ, Wang X, Lusis AJ: Determinants of atherosclerosis susceptibility in the C3H and C57BL/6 mouse model: evidence for involvement of endothelial cells but not blood cells or cholesterol metabolism. Circ Res. 2000, 86 (10): 1078-84.PubMed
153.
Zurück zum Zitat Funk J, Feingold K, Moser A, Grunfeld C: Lipopolysaccharide stimulation of RAW 264.7 macrophages induces lipid accumulation and foam cell formation. Atherosclerosis. 1993, 98 (1): 67-82. 10.1016/0021-9150(93)90224-I.PubMed Funk J, Feingold K, Moser A, Grunfeld C: Lipopolysaccharide stimulation of RAW 264.7 macrophages induces lipid accumulation and foam cell formation. Atherosclerosis. 1993, 98 (1): 67-82. 10.1016/0021-9150(93)90224-I.PubMed
154.
Zurück zum Zitat Lee JG, Lim EJ, Park DW, Lee SH, Kim JR, Baek SH: A combination of Lox-1 and Nox1 regulates TLR9-mediated foam cell formation. Cell Signal. 2008, 20 (12): 2266-75. 10.1016/j.cellsig.2008.08.022.PubMed Lee JG, Lim EJ, Park DW, Lee SH, Kim JR, Baek SH: A combination of Lox-1 and Nox1 regulates TLR9-mediated foam cell formation. Cell Signal. 2008, 20 (12): 2266-75. 10.1016/j.cellsig.2008.08.022.PubMed
155.
Zurück zum Zitat Oiknine J, Aviram M: Increased susceptibility to activation and increased uptake of low density lipoprotein by cholesterol-loaded macrophages. Arterioscler Thromb. 1992, 12 (6): 745-53.PubMed Oiknine J, Aviram M: Increased susceptibility to activation and increased uptake of low density lipoprotein by cholesterol-loaded macrophages. Arterioscler Thromb. 1992, 12 (6): 745-53.PubMed
156.
Zurück zum Zitat S Choi, Harkewicz R, Lee J, Boullier A, Almazan F, Li A, Witztum J, Bae Y, Miller Y: Lipoprotein accumulation in macrophages via toll-like receptor-4-dependent fluid phase uptake. Circ Res. 2009, 104 (12): 1355-63. 10.1161/CIRCRESAHA.108.192880. S Choi, Harkewicz R, Lee J, Boullier A, Almazan F, Li A, Witztum J, Bae Y, Miller Y: Lipoprotein accumulation in macrophages via toll-like receptor-4-dependent fluid phase uptake. Circ Res. 2009, 104 (12): 1355-63. 10.1161/CIRCRESAHA.108.192880.
157.
Zurück zum Zitat Bar-Or A, Nuttall RK, Duddy M, Alter A, Kim HJ, Ifergan I, Pennington CJ, Bourgoin P, Edwards DR, Yong VW: Analyses of all matrix metalloproteinase members in leukocytes emphasize monocytes as major inflammatory mediators in multiple sclerosis. Brain. 2003, 126 (Pt 12): 2738-49.PubMed Bar-Or A, Nuttall RK, Duddy M, Alter A, Kim HJ, Ifergan I, Pennington CJ, Bourgoin P, Edwards DR, Yong VW: Analyses of all matrix metalloproteinase members in leukocytes emphasize monocytes as major inflammatory mediators in multiple sclerosis. Brain. 2003, 126 (Pt 12): 2738-49.PubMed
158.
Zurück zum Zitat Welgus HG, Campbell EJ, Cury JD, Eisen AZ, Senior RM, Wilhelm SM, Goldberg GI: Neutral metalloproteinases produced by human mononuclear phagocytes. Enzyme profile, regulation, and expression during cellular development. J Clin Invest. 1990, 86 (5): 1496-502. 10.1172/JCI114867.PubMedPubMedCentral Welgus HG, Campbell EJ, Cury JD, Eisen AZ, Senior RM, Wilhelm SM, Goldberg GI: Neutral metalloproteinases produced by human mononuclear phagocytes. Enzyme profile, regulation, and expression during cellular development. J Clin Invest. 1990, 86 (5): 1496-502. 10.1172/JCI114867.PubMedPubMedCentral
159.
Zurück zum Zitat Ardans JA, Economou AP, Martinson JM, Zhou M, Wahl LM: Oxidized low-density and high-density lipoproteins regulate the production of matrix metalloproteinase-1 and -9 by activated monocytes. J Leukoc Biol. 2002, 71 (6): 1012-8.PubMed Ardans JA, Economou AP, Martinson JM, Zhou M, Wahl LM: Oxidized low-density and high-density lipoproteins regulate the production of matrix metalloproteinase-1 and -9 by activated monocytes. J Leukoc Biol. 2002, 71 (6): 1012-8.PubMed
160.
Zurück zum Zitat Saren P, Welgus HG, Kovanen PT: TNF-alpha and IL-1beta selectively induce expression of 92-kDa gelatinase by human macrophages. J Immunol. 1996, 157 (9): 4159-65.PubMed Saren P, Welgus HG, Kovanen PT: TNF-alpha and IL-1beta selectively induce expression of 92-kDa gelatinase by human macrophages. J Immunol. 1996, 157 (9): 4159-65.PubMed
161.
Zurück zum Zitat Herman MP, Sukhova GK, Libby P, Gerdes N, Tang N, Horton DB, Kilbride M, Breitbart RE, Chun M, Schonbeck U: Expression of neutrophil collagenase (matrix metalloproteinase-8) in human atheroma: a novel collagenolytic pathway suggested by transcriptional profiling. Circulation. 2001, 104 (16): 1899-904. 10.1161/hc4101.097419.PubMed Herman MP, Sukhova GK, Libby P, Gerdes N, Tang N, Horton DB, Kilbride M, Breitbart RE, Chun M, Schonbeck U: Expression of neutrophil collagenase (matrix metalloproteinase-8) in human atheroma: a novel collagenolytic pathway suggested by transcriptional profiling. Circulation. 2001, 104 (16): 1899-904. 10.1161/hc4101.097419.PubMed
162.
Zurück zum Zitat Shapiro SD, Campbell EJ, Kobayashi DK, Welgus HG: Immune modulation of metalloproteinase production in human macrophages. Selective pretranslational suppression of interstitial collagenase and stromelysin biosynthesis by interferon-gamma. J Clin Invest. 1990, 86 (4): 1204-10. 10.1172/JCI114826.PubMedPubMedCentral Shapiro SD, Campbell EJ, Kobayashi DK, Welgus HG: Immune modulation of metalloproteinase production in human macrophages. Selective pretranslational suppression of interstitial collagenase and stromelysin biosynthesis by interferon-gamma. J Clin Invest. 1990, 86 (4): 1204-10. 10.1172/JCI114826.PubMedPubMedCentral
163.
Zurück zum Zitat Fabunmi RP, Sukhova GK, Sugiyama S, Libby P: Expression of tissue inhibitor of metalloproteinases-3 in human atheroma and regulation in lesion-associated cells: a potential protective mechanism in plaque stability. Circ Res. 1998, 83 (3): 270-8.PubMed Fabunmi RP, Sukhova GK, Sugiyama S, Libby P: Expression of tissue inhibitor of metalloproteinases-3 in human atheroma and regulation in lesion-associated cells: a potential protective mechanism in plaque stability. Circ Res. 1998, 83 (3): 270-8.PubMed
164.
Zurück zum Zitat Guarda G, Braun M, Staehli F, Tardivel A, Mattmann C, Forster I, Farlik M, Decker T, Du Pasquier RA, Romero P, Tschopp J: Type I interferon inhibits interleukin-1 production and inflammasome activation. Immunity. 2011, 34 (2): 213-23. 10.1016/j.immuni.2011.02.006.PubMed Guarda G, Braun M, Staehli F, Tardivel A, Mattmann C, Forster I, Farlik M, Decker T, Du Pasquier RA, Romero P, Tschopp J: Type I interferon inhibits interleukin-1 production and inflammasome activation. Immunity. 2011, 34 (2): 213-23. 10.1016/j.immuni.2011.02.006.PubMed
165.
Zurück zum Zitat Zhang LN, Velichko S, Vincelette J, Fitch RM, Vergona R, Sullivan ME, Croze E, Wang YX: Interferon-beta attenuates angiotensin II-accelerated atherosclerosis and vascular remodeling in apolipoprotein E deficient mice. Atherosclerosis. 2008, 197 (1): 204-11. 10.1016/j.atherosclerosis.2007.03.019.PubMed Zhang LN, Velichko S, Vincelette J, Fitch RM, Vergona R, Sullivan ME, Croze E, Wang YX: Interferon-beta attenuates angiotensin II-accelerated atherosclerosis and vascular remodeling in apolipoprotein E deficient mice. Atherosclerosis. 2008, 197 (1): 204-11. 10.1016/j.atherosclerosis.2007.03.019.PubMed
166.
Zurück zum Zitat Goossens P, Gijbels MJ, Zernecke A, Eijgelaar W, Vergouwe MN, van der Made I, Vanderlocht J, Beckers L, Buurman WA, Daemen MJ, Kalinke U, Weber C, Lutgens E, de Winther MP: Myeloid type I interferon signaling promotes atherosclerosis by stimulating macrophage recruitment to lesions. Cell Metab. 2010, 12 (2): 142-53. 10.1016/j.cmet.2010.06.008.PubMed Goossens P, Gijbels MJ, Zernecke A, Eijgelaar W, Vergouwe MN, van der Made I, Vanderlocht J, Beckers L, Buurman WA, Daemen MJ, Kalinke U, Weber C, Lutgens E, de Winther MP: Myeloid type I interferon signaling promotes atherosclerosis by stimulating macrophage recruitment to lesions. Cell Metab. 2010, 12 (2): 142-53. 10.1016/j.cmet.2010.06.008.PubMed
167.
Zurück zum Zitat Zimmer S, Steinmetz M, Asdonk T, Motz I, Coch C, Hartmann E, Barchet W, Wassmann S, Hartmann G, Nickenig G: Activation of Endothelial Toll-Like Receptor 3 Impairs Endothelial Function. Circ Res. 2011 Zimmer S, Steinmetz M, Asdonk T, Motz I, Coch C, Hartmann E, Barchet W, Wassmann S, Hartmann G, Nickenig G: Activation of Endothelial Toll-Like Receptor 3 Impairs Endothelial Function. Circ Res. 2011
168.
Zurück zum Zitat Koga N, Suzuki J, Kosuge H, Haraguchi G, Onai Y, Futamatsu H, Maejima Y, Gotoh R, Saiki H, Tsushima F, Azuma M, Isobe M: Blockade of the interaction between PD-1 and PD-L1 accelerates graft arterial disease in cardiac allografts. Arterioscler Thromb Vasc Biol. 2004, 24 (11): 2057-62. 10.1161/01.ATV.0000145015.23656.e4.PubMed Koga N, Suzuki J, Kosuge H, Haraguchi G, Onai Y, Futamatsu H, Maejima Y, Gotoh R, Saiki H, Tsushima F, Azuma M, Isobe M: Blockade of the interaction between PD-1 and PD-L1 accelerates graft arterial disease in cardiac allografts. Arterioscler Thromb Vasc Biol. 2004, 24 (11): 2057-62. 10.1161/01.ATV.0000145015.23656.e4.PubMed
169.
Zurück zum Zitat Groschel S, Piggott KD, Vaglio A, Ma-Krupa W, Singh K, Goronzy JJ, Weyand CM: TLR-mediated induction of negative regulatory ligands on dendritic cells. J Mol Med. 2008, 86 (4): 443-55. 10.1007/s00109-008-0310-x.PubMedPubMedCentral Groschel S, Piggott KD, Vaglio A, Ma-Krupa W, Singh K, Goronzy JJ, Weyand CM: TLR-mediated induction of negative regulatory ligands on dendritic cells. J Mol Med. 2008, 86 (4): 443-55. 10.1007/s00109-008-0310-x.PubMedPubMedCentral
170.
Zurück zum Zitat Bsibsi M, Bajramovic JJ, Vogt MH, van Duijvenvoorden E, Baghat A, Persoon-Deen C, Tielen F, Verbeek R, Huitinga I, Ryffel B, Kros A, Gerritsen WH, Amor S, van Noort JM: The microtubule regulator stathmin is an endogenous protein agonist for TLR3. J Immunol. 2010, 184 (12): 6929-37. 10.4049/jimmunol.0902419.PubMed Bsibsi M, Bajramovic JJ, Vogt MH, van Duijvenvoorden E, Baghat A, Persoon-Deen C, Tielen F, Verbeek R, Huitinga I, Ryffel B, Kros A, Gerritsen WH, Amor S, van Noort JM: The microtubule regulator stathmin is an endogenous protein agonist for TLR3. J Immunol. 2010, 184 (12): 6929-37. 10.4049/jimmunol.0902419.PubMed
171.
Zurück zum Zitat Martinon F, Mayor A, Tschopp J: The inflammasomes: guardians of the body. Annu Rev Immunol. 2009, 27: 229-65. 10.1146/annurev.immunol.021908.132715.PubMed Martinon F, Mayor A, Tschopp J: The inflammasomes: guardians of the body. Annu Rev Immunol. 2009, 27: 229-65. 10.1146/annurev.immunol.021908.132715.PubMed
172.
Zurück zum Zitat Wang S, Miura M, Jung YK, Zhu H, Li E, Yuan J: Murine caspase-11, an ICE-interacting protease, is essential for the activation of ICE. Cell. 1998, 92 (4): 501-9. 10.1016/S0092-8674(00)80943-5.PubMed Wang S, Miura M, Jung YK, Zhu H, Li E, Yuan J: Murine caspase-11, an ICE-interacting protease, is essential for the activation of ICE. Cell. 1998, 92 (4): 501-9. 10.1016/S0092-8674(00)80943-5.PubMed
173.
Zurück zum Zitat Mariathasan S, Monack DM: Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation. Nat Rev Immunol. 2007, 7 (1): 31-40. 10.1038/nri1997.PubMed Mariathasan S, Monack DM: Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation. Nat Rev Immunol. 2007, 7 (1): 31-40. 10.1038/nri1997.PubMed
174.
Zurück zum Zitat Burns K, Martinon F, Tschopp J: New insights into the mechanism of IL-1beta maturation. Curr Opin Immunol. 2003, 15 (1): 26-30. 10.1016/S0952-7915(02)00017-1.PubMed Burns K, Martinon F, Tschopp J: New insights into the mechanism of IL-1beta maturation. Curr Opin Immunol. 2003, 15 (1): 26-30. 10.1016/S0952-7915(02)00017-1.PubMed
175.
Zurück zum Zitat Yajima N, Takahashi M, Morimoto H, Shiba Y, Takahashi Y, Masumoto J, Ise H, Sagara J, Nakayama J, Taniguchi S, Ikeda U: Critical role of bone marrow apoptosis-associated speck-like protein, an inflammasome adaptor molecule, in neointimal formation after vascular injury in mice. Circulation. 2008, 117 (24): 3079-87. 10.1161/CIRCULATIONAHA.107.746453.PubMed Yajima N, Takahashi M, Morimoto H, Shiba Y, Takahashi Y, Masumoto J, Ise H, Sagara J, Nakayama J, Taniguchi S, Ikeda U: Critical role of bone marrow apoptosis-associated speck-like protein, an inflammasome adaptor molecule, in neointimal formation after vascular injury in mice. Circulation. 2008, 117 (24): 3079-87. 10.1161/CIRCULATIONAHA.107.746453.PubMed
176.
Zurück zum Zitat Kirii H, Niwa T, Yamada Y, Wada H, Saito K, Iwakura Y, Asano M, Moriwaki H, Seishima M: Lack of interleukin-1beta decreases the severity of atherosclerosis in ApoE-deficient mice. Arterioscler Thromb Vasc Biol. 2003, 23 (4): 656-60. 10.1161/01.ATV.0000064374.15232.C3.PubMed Kirii H, Niwa T, Yamada Y, Wada H, Saito K, Iwakura Y, Asano M, Moriwaki H, Seishima M: Lack of interleukin-1beta decreases the severity of atherosclerosis in ApoE-deficient mice. Arterioscler Thromb Vasc Biol. 2003, 23 (4): 656-60. 10.1161/01.ATV.0000064374.15232.C3.PubMed
177.
Zurück zum Zitat Elhage R, Jawien J, Rudling M, Ljunggren HG, Takeda K, Akira S, Bayard F, Hansson GK: Reduced atherosclerosis in interleukin-18 deficient apolipoprotein E-knockout mice. Cardiovasc Res. 2003, 59 (1): 234-40. 10.1016/S0008-6363(03)00343-2.PubMed Elhage R, Jawien J, Rudling M, Ljunggren HG, Takeda K, Akira S, Bayard F, Hansson GK: Reduced atherosclerosis in interleukin-18 deficient apolipoprotein E-knockout mice. Cardiovasc Res. 2003, 59 (1): 234-40. 10.1016/S0008-6363(03)00343-2.PubMed
178.
Zurück zum Zitat Devlin CM, Kuriakose G, Hirsch E, Tabas I: Genetic alterations of IL-1 receptor antagonist in mice affect plasma cholesterol level and foam cell lesion size. Proc Natl Acad Sci USA. 2002, 99 (9): 6280-5. 10.1073/pnas.092324399.PubMedPubMedCentral Devlin CM, Kuriakose G, Hirsch E, Tabas I: Genetic alterations of IL-1 receptor antagonist in mice affect plasma cholesterol level and foam cell lesion size. Proc Natl Acad Sci USA. 2002, 99 (9): 6280-5. 10.1073/pnas.092324399.PubMedPubMedCentral
179.
Zurück zum Zitat Dewberry R, Holden H, Crossman D, Francis S: Interleukin-1 receptor antagonist expression in human endothelial cells and atherosclerosis. Arterioscler Thromb Vasc Biol. 2000, 20 (11): 2394-400.PubMed Dewberry R, Holden H, Crossman D, Francis S: Interleukin-1 receptor antagonist expression in human endothelial cells and atherosclerosis. Arterioscler Thromb Vasc Biol. 2000, 20 (11): 2394-400.PubMed
180.
Zurück zum Zitat Ishii N, Matsumura T, Kinoshita H, Fukuda K, Motoshima H, Senokuchi T, Nakao S, Tsutsumi A, Kim-Mitsuyama S, Kawada T, Takeya M, Miyamura N, Nishikawa T, Araki E: Nifedipine induces peroxisome proliferator-activated receptor-gamma activation in macrophages and suppresses the progression of atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2010, 30 (8): 1598-605. 10.1161/ATVBAHA.109.202309.PubMed Ishii N, Matsumura T, Kinoshita H, Fukuda K, Motoshima H, Senokuchi T, Nakao S, Tsutsumi A, Kim-Mitsuyama S, Kawada T, Takeya M, Miyamura N, Nishikawa T, Araki E: Nifedipine induces peroxisome proliferator-activated receptor-gamma activation in macrophages and suppresses the progression of atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2010, 30 (8): 1598-605. 10.1161/ATVBAHA.109.202309.PubMed
181.
Zurück zum Zitat Nakaya H, Summers BD, Nicholson AC, Gotto AM, Hajjar DP, Han J: Atherosclerosis in LDLR-knockout mice is inhibited, but not reversed, by the PPARgamma ligand pioglitazone. Am J Pathol. 2009, 174 (6): 2007-14. 10.2353/ajpath.2009.080611.PubMedPubMedCentral Nakaya H, Summers BD, Nicholson AC, Gotto AM, Hajjar DP, Han J: Atherosclerosis in LDLR-knockout mice is inhibited, but not reversed, by the PPARgamma ligand pioglitazone. Am J Pathol. 2009, 174 (6): 2007-14. 10.2353/ajpath.2009.080611.PubMedPubMedCentral
182.
Zurück zum Zitat Chinetti G, Fruchart JC, Staels B: Peroxisome proliferator-activated receptors (PPARs): nuclear receptors at the crossroads between lipid metabolism and inflammation. Inflamm Res. 2000, 49 (10): 497-505. 10.1007/s000110050622.PubMed Chinetti G, Fruchart JC, Staels B: Peroxisome proliferator-activated receptors (PPARs): nuclear receptors at the crossroads between lipid metabolism and inflammation. Inflamm Res. 2000, 49 (10): 497-505. 10.1007/s000110050622.PubMed
183.
Zurück zum Zitat Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, Subramanian V, Mukundan L, Red Eagle A, Vats D, Brombacher F, Ferrante AW, Chawla A: Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature. 2007, 447 (7148): 1116-20. 10.1038/nature05894.PubMedPubMedCentral Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, Subramanian V, Mukundan L, Red Eagle A, Vats D, Brombacher F, Ferrante AW, Chawla A: Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature. 2007, 447 (7148): 1116-20. 10.1038/nature05894.PubMedPubMedCentral
184.
Zurück zum Zitat Tontonoz P, Nagy L, Alvarez JG, Thomazy VA, Evans RM: PPARgamma promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell. 1998, 93 (2): 241-52. 10.1016/S0092-8674(00)81575-5.PubMed Tontonoz P, Nagy L, Alvarez JG, Thomazy VA, Evans RM: PPARgamma promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell. 1998, 93 (2): 241-52. 10.1016/S0092-8674(00)81575-5.PubMed
185.
Zurück zum Zitat Straus DS, Glass CK: Anti-inflammatory actions of PPAR ligands: new insights on cellular and molecular mechanisms. Trends Immunol. 2007, 28 (12): 551-8. 10.1016/j.it.2007.09.003.PubMed Straus DS, Glass CK: Anti-inflammatory actions of PPAR ligands: new insights on cellular and molecular mechanisms. Trends Immunol. 2007, 28 (12): 551-8. 10.1016/j.it.2007.09.003.PubMed
186.
Zurück zum Zitat Heilbronn LK, Campbell LV: Adipose tissue macrophages, low grade inflammation and insulin resistance in human obesity. Curr Pharm Des. 2008, 14 (12): 1225-30. 10.2174/138161208784246153.PubMed Heilbronn LK, Campbell LV: Adipose tissue macrophages, low grade inflammation and insulin resistance in human obesity. Curr Pharm Des. 2008, 14 (12): 1225-30. 10.2174/138161208784246153.PubMed
187.
Zurück zum Zitat Coste A, Dubourdeau M, Linas MD, Cassaing S, Lepert JC, Balard P, Chalmeton S, Bernad J, Orfila C, Seguela JP, Pipy B: PPARgamma promotes mannose receptor gene expression in murine macrophages and contributes to the induction of this receptor by IL-13. Immunity. 2003, 19 (3): 329-39. 10.1016/S1074-7613(03)00229-2.PubMed Coste A, Dubourdeau M, Linas MD, Cassaing S, Lepert JC, Balard P, Chalmeton S, Bernad J, Orfila C, Seguela JP, Pipy B: PPARgamma promotes mannose receptor gene expression in murine macrophages and contributes to the induction of this receptor by IL-13. Immunity. 2003, 19 (3): 329-39. 10.1016/S1074-7613(03)00229-2.PubMed
188.
Zurück zum Zitat Kim SR, Lee KS, Park HS, Park SJ, Min KH, Jin SM, Lee YC: Involvement of IL-10 in peroxisome proliferator-activated receptor gamma-mediated anti-inflammatory response in asthma. Mol Pharmacol. 2005, 68 (6): 1568-75.PubMed Kim SR, Lee KS, Park HS, Park SJ, Min KH, Jin SM, Lee YC: Involvement of IL-10 in peroxisome proliferator-activated receptor gamma-mediated anti-inflammatory response in asthma. Mol Pharmacol. 2005, 68 (6): 1568-75.PubMed
189.
Zurück zum Zitat Hetzel M, Walcher D, Grub M, Bach H, Hombach V, Marx N: Inhibition of MMP-9 expression by PPARgamma activators in human bronchial epithelial cells. Thorax. 2003, 58 (9): 778-83. 10.1136/thorax.58.9.778.PubMedPubMedCentral Hetzel M, Walcher D, Grub M, Bach H, Hombach V, Marx N: Inhibition of MMP-9 expression by PPARgamma activators in human bronchial epithelial cells. Thorax. 2003, 58 (9): 778-83. 10.1136/thorax.58.9.778.PubMedPubMedCentral
190.
Zurück zum Zitat Marx N, Imhof A, Froehlich J, Siam L, Ittner J, Wierse G, Schmidt A, Maerz W, Hombach V, Koenig W: Effect of rosiglitazone treatment on soluble CD40L in patients with type 2 diabetes and coronary artery disease. Circulation. 2003, 107 (15): 1954-7. 10.1161/01.CIR.0000069272.06194.91.PubMed Marx N, Imhof A, Froehlich J, Siam L, Ittner J, Wierse G, Schmidt A, Maerz W, Hombach V, Koenig W: Effect of rosiglitazone treatment on soluble CD40L in patients with type 2 diabetes and coronary artery disease. Circulation. 2003, 107 (15): 1954-7. 10.1161/01.CIR.0000069272.06194.91.PubMed
191.
Zurück zum Zitat Glatz T, Stock I, Nguyen-Ngoc M, Gohlke P, Herdegen T, Culman J, Zhao Y: Peroxisome-proliferator-activated receptors gamma and peroxisome-proliferator-activated receptors beta/delta and the regulation of interleukin 1 receptor antagonist expression by pioglitazone in ischaemic brain. J Hypertens. 2010, 28 (7): 1488-97. 10.1097/HJH.0b013e3283396e4e.PubMed Glatz T, Stock I, Nguyen-Ngoc M, Gohlke P, Herdegen T, Culman J, Zhao Y: Peroxisome-proliferator-activated receptors gamma and peroxisome-proliferator-activated receptors beta/delta and the regulation of interleukin 1 receptor antagonist expression by pioglitazone in ischaemic brain. J Hypertens. 2010, 28 (7): 1488-97. 10.1097/HJH.0b013e3283396e4e.PubMed
192.
Zurück zum Zitat Moulin D, Bianchi A, Boyault S, Sebillaud S, Koufany M, Francois M, Netter P, Jouzeau JY, Terlain B: Rosiglitazone induces interleukin-1 receptor antagonist in interleukin-1beta-stimulated rat synovial fibroblasts via a peroxisome proliferator-activated receptor beta/delta-dependent mechanism. Arthritis Rheum. 2005, 52 (3): 759-69. 10.1002/art.20868.PubMed Moulin D, Bianchi A, Boyault S, Sebillaud S, Koufany M, Francois M, Netter P, Jouzeau JY, Terlain B: Rosiglitazone induces interleukin-1 receptor antagonist in interleukin-1beta-stimulated rat synovial fibroblasts via a peroxisome proliferator-activated receptor beta/delta-dependent mechanism. Arthritis Rheum. 2005, 52 (3): 759-69. 10.1002/art.20868.PubMed
193.
Zurück zum Zitat Halvorsen B, Heggen E, Ueland T, Smith C, Sandberg WJ, Damas JK, Otterdal K, Tonstad S, Aukrust P: Treatment with the PPARgamma agonist rosiglitazone downregulates interleukin-1 receptor antagonist in individuals with metabolic syndrome. Eur J Endocrinol. 2010, 162 (2): 267-73. 10.1530/EJE-09-0706.PubMed Halvorsen B, Heggen E, Ueland T, Smith C, Sandberg WJ, Damas JK, Otterdal K, Tonstad S, Aukrust P: Treatment with the PPARgamma agonist rosiglitazone downregulates interleukin-1 receptor antagonist in individuals with metabolic syndrome. Eur J Endocrinol. 2010, 162 (2): 267-73. 10.1530/EJE-09-0706.PubMed
194.
Zurück zum Zitat Fernandez AZ: Peroxisome proliferator-activated receptors in the modulation of the immune/inflammatory response in atherosclerosis. PPAR Res. 2008, 2008: 285842-PubMedPubMedCentral Fernandez AZ: Peroxisome proliferator-activated receptors in the modulation of the immune/inflammatory response in atherosclerosis. PPAR Res. 2008, 2008: 285842-PubMedPubMedCentral
195.
Zurück zum Zitat Ziegler-Heitbrock L: The CD14+ CD16+ blood monocytes: their role in infection and inflammation. J Leukoc Biol. 2007, 81 (3): 584-92.PubMed Ziegler-Heitbrock L: The CD14+ CD16+ blood monocytes: their role in infection and inflammation. J Leukoc Biol. 2007, 81 (3): 584-92.PubMed
196.
Zurück zum Zitat Yona S, Jung S: Monocytes: subsets, origins, fates and functions. Curr Opin Hematol. 2010, 17 (1): 53-9. 10.1097/MOH.0b013e3283324f80.PubMed Yona S, Jung S: Monocytes: subsets, origins, fates and functions. Curr Opin Hematol. 2010, 17 (1): 53-9. 10.1097/MOH.0b013e3283324f80.PubMed
Metadaten
Titel
Innate immunity and monocyte-macrophage activation in atherosclerosis
verfasst von
Joseph Shalhoub
Mika A Falck-Hansen
Alun H Davies
Claudia Monaco
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
Journal of Inflammation / Ausgabe 1/2011
Elektronische ISSN: 1476-9255
DOI
https://doi.org/10.1186/1476-9255-8-9

Weitere Artikel der Ausgabe 1/2011

Journal of Inflammation 1/2011 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.