Skip to main content
Erschienen in: Current Atherosclerosis Reports 3/2012

Open Access 01.06.2012 | Vascular Biology (RS Rosenson, Section Editor)

Molecular Pathways Regulating Macrophage Polarization: Implications for Atherosclerosis

verfasst von: Marten A. Hoeksema, J. Lauran Stöger, Menno P. J. de Winther

Erschienen in: Current Atherosclerosis Reports | Ausgabe 3/2012

Abstract

Recent years have seen a tremendous development of our insight into the biology of atherosclerosis and its acute thrombotic manifestations. Inflammation now takes center stage among traditional risk factors as a decisive factor in cardiovascular risk. Consequently, its assessment and modulation have become key to clinical care and fundamental research alike. Plaque macrophages orchestrate many of the inflammatory processes that occur throughout atherogenesis. These cells are characteristically heterogeneous and adopt diverse activation states in response to micro-environmental triggers. In this review, macrophage-mediated inflammation in atherosclerosis sets the scene for a discussion of the gene regulatory mechanisms that facilitate and shape polarized macrophage phenotypes. When applicable, we consider these factors within the context of atherosclerosis and reflect on opportunities for future application.

Introduction

Myocardial infarction and stroke remain among the leading causes of death and disease worldwide [1, 2]. To mitigate risk of these atherosclerotic complications, primary and secondary prevention strategies seek to correct aberrant blood cholesterol levels. Actively reducing low-density lipoprotein (LDL) cholesterol through lipid-modifying therapy (eg, statins) yields a proportional decrease in cardiovascular disease (CVD) risk [3]. However, there exists a considerable burden of residual risk, as current treatment strategies cannot prevent 75 % of major coronary events from occurring [4, 5]. Moreover, individuals afflicted by CVD are often times free of traditional risk factors [6], suggesting other dynamics contribute to plaque complication.
In this context, macrophage-mediated inflammation is paramount, contributing to atherosclerotic plaque initiation and progression through a variety of mechanisms [7]. We are developing a better understanding of the processes that regulate the induction and function of distinct macrophage subsets and their potential relevance in atherosclerosis. This review serves to highlight the cellular mediators that convert environmental cues to a heterogeneous array of functional macrophage phenotypes, thereby shaping inflammatory responses in health and disease.

Inflammation and Atherosclerosis

Over the past two decades, the inflammatory hypothesis of atherothrombosis has gained an increasingly strong footing through multiple lines of supportive evidence. Overall, an increased systemic burden of inflammation prompts a higher CVD incidence, as is the case in chronic inflammatory conditions such as rheumatic arthritis [8] and systemic lupus erythematosus [9]. Various soluble mediators of the inflammatory response have been found to predict future cardiovascular risk in atherosclerotic patients (well-described in [10]). High-sensitivity C-reactive protein (hsCRP) has formed a focus point in this respect, as systemic concentrations of this acute-phase protein compared favorably with LDL cholesterol and blood pressure as CVD risk factors [11], and were specifically associated to plaque vulnerability [12, 13]. Building on post hoc analyses from several other large-scale studies (eg, CARE, PROVE-IT TIMI 22, AFCAPS/TexCAPS trials [1416]), the JUPITER trial prospectively consolidated the correlation of hsCRP and cardiovascular outcome in a primary prevention setting [17]. Researchers observed that the clinical benefits of statin therapy were greatest when both LDL and hsCRP values were reduced, thus connecting both dyslipidemia and inflammation at the interface of CVD pathogenesis. Intriguingly, even with pre-existent LDL levels below the clinical cut-off point for treatment, persistent inflammation as measured by increased hsCRP levels puts patients at a higher than anticipated risk of CVD. In the AFCAPS/TexCAPS trial, these subjects responded strongly to treatment [16], indicating LDL burden is not a prerequisite to successful therapy. Apart from providing clinicians with valuable information for risk assessment, this finding proposes that an enhanced inflammatory state might in itself justify targeted therapy. Indeed, US and Canadian prevention guidelines have since embraced hsCRP measurements in the considerations for patients at intermediate risk. Moreover, a number of new trials, using either low-dose methotrexate (CIRT) or anti-IL-1β monoclonal antibodies (CANTOS) as anti-inflammatory treatment, are underway to address and possibly validate the hypothesis of inflammatory causality [18•, 19•]. These translational efforts could provide a major argument towards a more systematic implementation of anti-inflammatory therapy in our continuing battle to diminish residual cardiovascular risk.
Substantial experimental evidence complements the broad clinical involvement of inflammation in CVD outlined above. Now most agree that systemic risk factors interact with many cell types (both those intrinsic to the vasculature and immune cells attracted from the circulation) to drive plaque development. Particularly, monocyte-derived macrophages are considered critical participants in the atherogenic process, as they secrete pro-inflammatory cytokines and other mediators that affect lesion progression and stability. Consequently, many experimental studies have successfully targeted the abundance of monocytes/macrophages and their soluble repertoire in atherosclerosis as a means of prevention. For instance, atherosclerotic plaque formation was virtually abolished in hyperlipidemic mice lacking the macrophage-colony stimulating factor (M-CSF) gene, which exhibit impaired monocyte development and subsequent differentiation to macrophages [20, 21]. Other scientific efforts involved the abrogation of chemokine-dependent monocyte recruitment to the plaque [22], in addition to a wealth of studies addressing the various cytokines produced by macrophages and other cells (reviewed in [23]). Although not cell-specific, these data still offer valuable insight into how macrophages contribute to nascent lesions. Macrophage apoptosis is another important feature seen during atherosclerosis development. In early lesions, macrophage apoptosis and plaque size exist in an inverse relationship [24], whereas in later stages this process contributes to the plaque’s lipid core [25]. This ambiguity appears to be mediated by a process termed “efferocytosis” [26]. Combined with proof linking plaque macrophages to matrix metalloproteinase (MMP)-dependent collagen breakdown [27, 28], it is evident that these cells modulate inflammatory mechanisms to determine plaque susceptibility to rupture and clinical atherothrombosis. Associative imaging studies lend weight to this notion by demonstrating that vascular uptake of fluorodeoxyglucose (18F-FDG) correlates to plaque macrophage load and general inflammatory burden and can thereby assist in the prediction of cardiovascular events [29, 30].

Phenotypic Differences Between Macrophage Subsets

Cultured and tissue macrophages both exhibit pronounced heterogeneity, as was recognized early on [31]. Skewing of macrophages toward distinct polarization programs occurs in response to various environmental cues and has inspired extensive research into their significance in pathophysiology. Reflecting the Th1 and Th2 nomenclature in T-cells, polarized macrophage subsets were originally referred to as classically (M1) or alternatively activated (M2) [32]. The latter group was subsequently divided into M2a, M2b, M2c and tumor-associated macrophages (TAMs) to distinguish between inducing stimuli [33]. Later on, Mosser and Edwards [34] reassigned these macrophage subsets to three different classes: classically activated macrophages (CAMs, corresponding to M1), alternative activated macrophages (AAMs, also referred to as wound healing macrophages and analogous to M2a), and regulatory macrophages (RMs, consistent with M2b/c). These classes are best considered a continuum of functional states that encompasses a broad range of macrophage phenotypes with interchangeable characteristics.
CAMs are typically induced by the Th1-cytokine interferon-γ (IFNγ), possibly followed by activation with a Toll-like Receptor (TLR) ligand, such as lipopolysaccharides (LPS). IFNγ (originally termed macrophage-activating factor) prepares macrophages for a pro-inflammatory environment, giving rise to a potent inflammatory response upon microbial challenge. The resulting phenotype characteristically displays high interleukin-12 (IL-12) and low IL-10 production, combined with enhanced microbicidal effector functions through the induction of NADPH-oxidases and inducible nitric oxide synthase (iNOS). Therefore, these macrophages become very efficient in killing bacteria, viruses, parasites, and fungi. Their continuous induction and sustained activation, however, will cause tissue damage [35].
The Th2 cytokines IL-4 and IL-13 are produced by granulocytes, mast cells, and Th2 cells during injury and infection and generate AAMs [36, 37]. Opposite to CAMs, AAMs dampen inflammatory responses through an IL-12low and IL-10high expression profile. Furthermore, AAMs promote tissue repair and fibrosis through increased arginase-1-dependent production of the collagen precursors ornithine and proline [38]. Besides arginase-1, other markers for AAMs include Ym1, Fizz1, and the mannose receptor (MR).
Finally, regulatory macrophages (RMs) are induced in response to a wide range of stimuli, including immune complexes, prostaglandins, G-protein coupled receptor ligands, glucocorticoids, and uptake of apoptotic cells. However, a key cytokine in the induction of RMs is the anti-inflammatory, atheroprotective cytokine IL-10. The main task of RMs is to suppress and control immune responses by producing high levels of IL-10 and thereby contribute to the resolution of inflammatory responses [34, 39].

Macrophage Differentiation and Polarization

The current framework of macrophage subsets is very well characterized in vitro but less so in in vivo settings. Here, a greater variety of external challenges elicit macrophage phenotypes that are considerably less adherent to the constraints of the existing paradigm. During monocyte-to-macrophage differentiation and subsequent macrophage activation, the collective imprint of these environmental factors shapes the macrophage phenotype. An intuitive overview of these sequences was recently described by Gordon and Martinez [40].
First, monocyte recruitment from the circulation will be followed by differentiation to mature tissue macrophages. This transition is primarily mediated by the growth factors M-CSF and granulocyte-macrophage colony-stimulating factor (GM-CSF) that influence the inflammatory potential of the resulting macrophage. Maturation with M-CSF was observed to lead to a more anti-inflammatory (IL-10highIL-12low) phenotype, whereas GM-CSF-induced differentiation gave rise to a macrophage population with pro-inflammatory (IL-10lowIL-12high) characteristics [41, 42].
The phenotype that ensues from the subsequent priming stage hinges on the local balance of cytokines and chemokines in the newly recruited macrophage’s environment. Priming with cytokine stimuli will affect the inflammatory potential of macrophages and their response to other stimuli. In addition to the previously described cytokine stimuli (IFNγ, IL-4/IL-13, and IL-10 for CAMs, AAMs, and RM, respectively), chemokines (eg, CXCL4) [43] and other plaque constituents (oxidized lipoproteins) [44] were also shown to induce unique macrophage phenotypes with distinctive characteristics.
Upon activation by Toll-like or analogous receptor stimuli, the macrophage will undergo functional maturation that results in a rapid induction of anti-microbial pathways for fast killing and clearance of pathogens. Finally, when the macrophage has fulfilled and survived its inflammatory task, it will undergo deactivation. In this final phase, the RM phenotype and its mediators transforming growth factor-β (TGF-β), IL-10, and lipoxins, are key contributors to the resolution of inflammation and tissue repair [40].

Macrophage Polarization and Disease

Macrophage subtypes have the capacity to switch from one phenotype to another as stimuli from the micro-environment change. Recently, such plasticity was elegantly demonstrated in a murine kidney ischemia-reperfusion model [45•]. Whereas initially pro-inflammatory CAMs were recruited to the kidney, later time points saw previously attracted macrophages switch to an MR+-AAM phenotype, signaling the resolution of inflammation and repair. Similar reports were made with regard to other disease states, as for instance in metabolic disease. Adipose tissue macrophages in lean mice resemble an AAM phenotype that supports adipocyte function and insulin sensitivity [46]. Obesity, however, switches the macrophage balance to a CAM phenotype as the result of inflammasome activation by various danger signals from adipocytes, thereby promoting inflammation and insulin resistance [47, 48]. In analogy to this dichotomy, differential macrophage function has been reported to influence the course of various other conditions, such as cancer and infectious disease [34, 49].
Likewise, in atherosclerosis, evidence implies that the balance of macrophage polarization is crucial in determining plaque outcome [4951]. Following the progression of atherosclerosis over time, it was shown that plaque macrophages in early lesions express arginase-1 (indicative of AAMs), whereas at later time points the expression of arginase-2 (referred to as a CAM marker) predominated in the plaque [52]. More recently, phenotypic switching in atherosclerosis was described upon induction of disease regression [53••]. Here, plaque macrophages restrained pro-inflammatory marker expression (eg, monocyte chemotactic protein-1 and tumor necrosis factor) and enhanced that of AAM markers such as arginase-1, MR, and CD163 upon normalization of the plasma lipid profile. However, the order of events in this model still requires clarification (ie, does phenotypic switching actively contribute to plaque regression or is it merely a consequence thereof). In human subjects too, it was shown that symptomatic atherosclerotic disease of the carotid arteries is associated with a CAM profile, characterized by high pro-inflammatory cytokine expression [54]. Moreover, enrichment of CD11c+-CAMs over MR+-AAMs characterizes the epicardial adipose tissue from patients with manifest coronary artery disease [55]. These reports are consistent with the current hypothesis that CAMs propagate plaque vulnerability through their inflammatory potential, whereas their AAM counterparts are likely to benefit plaque stability and regression through a repertoire of repair and fibrotic functions. From this perspective, Fig. 1 provides a schematic overview of the inducers, transducers, and effector functions of these macrophage populations in atherosclerosis development.
Although murine macrophage subtypes are well characterized, the translation to human pathophysiology is not straightforward. Where polarized macrophage subsets in mice can be easily distinguished based on their arginine-metabolizing enzyme expression (i.e. iNOS and arginase-1 for CAMs and AAMs respectively), in vitro polarized human macrophages do not express arginase-1 and fail to produce NO in amounts comparable to that of mouse macrophages [57]. In addition, markers like Ym1 and Fizz1, which are excellent for AAM detection in mice, are not expressed in humans [35]. Circumventing these issues through identification of generally more favorable markers of human tissue macrophages will prove an important step in translational atherosclerosis research.

Transcription Factors in Macrophage Subsets

To achieve expression of the aforementioned surface marker and cytokine profiles, different macrophage subsets are known to employ distinct signaling pathways (Fig. 1). These cascades usually activate certain transcription factors that effectively induce expression of archetypal genes, thereby accounting for the translation of external polarizing cues into a specific subtype [58]. Clearly, this designates transcription factors as important players in macrophage polarization that may represent an attractive alternative to surface markers for discerning macrophage populations in vivo. Moreover, these pathways may eventually yield suitable targets for the inhibition, depletion, or stimulation of a particular phenotype. An overview of transcription factors associated to macrophage heterogeneity and their functional relevance in experimental or human atherosclerotic lesions, as described below, can be found in Table 1.
Table 1
The role of transcription factors in macrophage polarization and atherosclerosis
Gene
Linked?
Model
Findings
Refs
Classically activated macrophage
STAT1
Yes
ApoE-/-+LDLR-/-; BMT
Hematopoietic STAT1 deficiency leads to decreased plaque sizes
[5961]
NF-κB p65
Yes
Human plaques
The presence of activated NF-κB p65 in human atherosclerotic plaques was demonstrated
[64]
IRF1
Yes
Human lymphocytes
IRF-1 expression was increased in lymphocytes from patients with acute coronary syndrome
[72, 74]
IRF5
No
N/A
N/A
[70, 71]
NFAT5
Yes
ApoE-/-
NFAT5 expression was highly expressed in murine atherosclerotic lesions
[82, 83]
SOCS1a
Yes
Human plaques
SOCS1 and SOCS3 are highly expressed by vascular smooth muscle cells and macrophages in inflammatory regions
[84, 85]
SOCS3
Yes
ApoE-/-
SOCS3 targeting oligonucleotides exacerbated the atherosclerotic process
[85, 86]
Regulatory macrophage
STAT3
Yes
LDLR-/-; adenovirus
Adenovirus expression of human STAT3 lowers aortic inflammatory cell infiltration
[87, 88]
Alternatively activated macrophage
STAT6
No
N/A
N/A
[62]
NF-κB p50
Yes
LDLR-/-; BMT
p50 deletion in bone marrow cells leads to more inflammatory and smaller lesions
[67, 68]
IRF3
No
N/A
N/A
[41]
IRF4
No
N/A
N/A
[69]
PPARγ
Yes
Human monocytes; IHC human plaques
PPARγ activation protects against atherosclerotic plaque formation in humans
[75, 76, 81•]
LXRα
Yes
LDLR-/-; lentivector and human plaques
Macrophage LXRα gene therapy ameliorates atherosclerosis in LDLR-/- mice and LXRα was found increased during regression. In contrast, human AAM plaque macrophages were shown to have decreased levels of LXRα.
[77, 79, 81•]
Cebpb
No
N/A
N/A
[89]
KLF2
Yes
KLF2+/- ApoE-/-
Hemizygous KLF2 deficiency increased atherosclerosis in apoE-/- mice
[90, 91]
KLF4
No
N/A
N/A
[92]
SOCS1a
Yes
Human plaques
SOCS1 and SOCS3 are highly expressed by vascular smooth muscle cells and macrophages in inflammatory regions
[84, 85]
a Involved in both AAM as CAM signaling
ApoE—apolipoprotein E; BMT—bone marrow transplantation; Cebpb—CCAAT/enhancer-binding protein beta; IHC—immunohistochemistry; IRF— Interferon regulatory factors; KLF—Krüppel-like factor; LDLR—low-density lipoprotein receptor; LXR—liver X receptor; N/A— not applicable; NFAT—nuclear factor of activated T cells; PPAR—peroxisome proliferator-activated receptor; SOCS—suppressor of cytokine signaling proteins; STAT— signal transducers and activators of transcription

STAT Signaling

Signal transducers and activators of transcription (STATs) are crucial transcription factors in the determination of macrophage phenotype. IFNγ signaling will lead to downstream phosphorylation and activation of STAT1. Macrophages lacking this mediator are unable to express a full CAM phenotype, as they fail to produce nitric oxide (NO) and have diminished expression of IL-12 and major histocompatibility complex (MHC) class II [59]. In murine atherosclerosis, macrophage STAT1 deficiency leads to attenuated atherosclerosis, resulting from a decrease in macrophage lipid accumulation, macrophage apoptosis, and plaque necrosis [60, 61]. IL-4 and IL-13, inducers of the AAM phenotype, also signal via JAK-STAT pathways. Both cytokines lead to the activation of STAT6. STAT6 signaling was shown to be necessary for the expression of many AAM markers, like arginase-1 [62], but has not been directly linked to atherosclerosis development.

NF-κB Signaling

Nuclear factor κB (NF-κB) is a key regulator of the initiation and resolution of inflammation [63]. NF-κB activation, more specifically that of its subunit p65, is a hallmark of classical macrophage activation and occurs in both endothelial cells, smooth muscle cells, and macrophages in human atherosclerotic lesions [64]. However, its activation is subject to multiple levels of (inhibitory) regulation that fine-tune macrophage function. Interestingly, attempts to inhibit NF-κB activity by deleting inhibitor of NF-κB kinase (IKK) 2, the kinase that activates NF-κB, yielded unexpected outcomes. IKK2 was shown to inhibit STAT1 signaling in macrophages, thereby affecting IL-12, iNOS, and MHC class II expression [65]. Therefore, IKK2 deletion did not diminish polarization towards the CAM phenotype, but instead encouraged certain CAM characteristics through enhanced STAT1 activity. As such, macrophage-specific deletion of IKK2 in atherosclerosis resulted in reduced NF-κB activity but an increased atherosclerotic plaque size [66].
Conversely, the p50 subunit of NF-κB was recently identified as a key regulator of the AAM phenotype both in vitro as in vivo [67]. In line, it was shown that targeted deletion of the p50 subunit in macrophages in atherosclerosis led to more inflammatory lesions. Surprisingly, these lesions were smaller in size, which was linked to a reduced uptake of oxidized LDL in activated p50-deficient macrophages [68].

IRF Signaling

Interferon regulatory factors (IRFs) are important mediators of macrophage polarization and several family members have been reported in relation to a specific phenotype. IRF4 was shown to be a key transcription factor controlling AAM polarization and its associated gene expression profile [69]. High expression of IRF5 was observed in CAMs and subsequently described as a central mediator in TLR signaling pathways. Here, it will directly activate the transcription of pro-inflammatory genes and repress anti-inflammatory IL-10 expression [70, 71]. Likewise, IRF1 was shown to cooperate with NF-κB in the induction of several pro-inflammatory cytokines [72]. Moreover, by antagonizing IRF4 function [73], IRF1 can also be classified as an important mediator in CAM signaling. Although the role of IRFs in atherosclerosis is still to be determined, lymphocytes from patients with acute coronary syndromes display increased IRF-1 expression, linking this transcription factor to cardiovascular risk [74].

PPAR and LXR Signaling

Another transcription factor found to be crucial for murine AAM polarization is peroxisome proliferator-activated receptor (PPAR) γ, a nuclear receptor [75]. Also in human monocytes, it was shown that PPARγ activation skews towards an AAM phenotype and conveys important functional differences [76]. A closely related nuclear receptor is liver X receptor (LXR) α, which was found to be of similar importance in AAM polarization. Besides regulating lipid metabolism and transport, LXRα enhances arginase-1 expression and suppresses inflammatory signaling in macrophages [77]. Both of these transcription factors fulfill additional protective roles in experimental atherosclerosis [7880], mainly by reducing intra-cellular accumulation of oxidized LDL [76, 81•]. Therefore, PPARγ and LXRα instill macrophages with powerful anti-inflammatory and metabolic functionalities.

Epigenetic Control of Macrophage Polarization Through Histone Modifications

In addition to transcriptional control, epigenetic regulation is essential for a properly directed expression of target genes. Without modifying the actual genetic code, epigenetic mechanisms affect DNA accessibility to the transcriptional apparatus to alter gene expression and even mRNA degradation by microRNAs. Remarkably, the resulting gene expression patterns can be passed down to daughter cells upon cell division or even trans-generationally. Epigenetic regulation can be achieved through DNA methylation (generally considered a relatively static process), as well as through applying certain modification to histones, such as acetylation or methylation. These marks can be changed very dynamically in response to many environmental stimuli.
In macrophages, several epigenetic mediators now are identified as key players in macrophage function and polarization [93]. For instance, Jmjd3, a JmjC family histone demethylase that erases lysine 27 trimethylation marks on histone 3, has been shown to specifically bind transcription start sites of promoters of NF-κB dependent genes. This would suggest that Jmjd3 provides additional control of NF-κB-dependent gene expression, although most pro-inflammatory genes were expressed independently of Jmjd3 [94, 95]. In addition, it has been described that Jmjd3 plays an essential role in polarization. In response to AAM polarization, Jmjd3 can induce the expression of IRF4, resulting in the transcription of key AAM marker genes [62, 69]. Recently, the histone deacetylase 3 (HDAC3) was also found to mediate macrophage polarization, as HDAC3-deficient macrophages were shown to be hyperresponsive to polarization with IL-4 [96]. Thus, alternative activation of macrophages can be controlled at an epigenetic level by targeting HDAC3 and Jmjd3, suggesting that epigenetic mediators might evolve to become promising immunomodulatory targets. This notion was recently substantiated by the finding that the bromodomain and extra terminal domain family of proteins-inhibitor (I-BET) is able to disrupt chromatin complexes responsible for the expression of key inflammatory genes during macrophage activation and could thereby protect against endotoxic shock and sepsis [97•]. This indeed very nicely demonstrates that synthetic compounds specifically targeting epigenetic mechanisms can act as excellent therapeutic agents for immunomodulation. As future studies further unravel the epigenomic landscape of macrophage subtypes, we will definitely see the development of other immunomodulatory compounds that target epigenetic regulation of macrophages. Moreover, in experimental atherosclerosis, it was shown that cell-specific histone methylation modifications and expression of accompanying lysine methyltransferases occur in carotid arteries. More specifically, differences in histone methylation modifications in vascular endothelial and smooth muscle cells were described between the offspring of hypercholesterolemic and normocholesterolemic mothers [98]. As histone modifications are readily altered in response to environmental stimuli, they can provide an attractive explanation how diet and lifestyle may contribute to atherosclerosis susceptibility [99].

Conclusions

Amongst a variety of atherogenic properties, macrophages direct and amplify the inflammatory response in atherosclerosis and thereby contribute to lesion initiation, progression, and clinical manifestation. The plaque macrophage phenotype is determined by a plethora of micro-environmental stimuli encountered during the successive phases of macrophage development. These factors can trigger activation of subtype-specific signaling pathways and downstream transcription factors, which are crucial mediators of macrophage polarization. Additionally, epigenetic modifiers comprise a relatively novel level of transcriptional regulation that has been shown to fine-tune the macrophage phenotype. Together, these molecular effectors may govern the critical balance between distinct macrophage subsets in atherosclerosis. Such modulation could in turn be expected to either propagate plaque progression or to confer atheroprotective mechanisms. It therefore remains crucial to improve our understanding of macrophage heterogeneity in atherosclerosis and identify the subsets most suitable for intervention.
Key areas for careful consideration in that light include the longstanding need for better, more specific markers that characterize human macrophage subsets. However, interspecies differences in atherosclerotic pathogenesis and macrophage marker expression currently prevent us from taking the next step in translational research [100]. Paradoxically, additional work with animal models will prove indispensable to addressing these challenges, because these approaches allow for specific deletion of transcriptional regulators involved in macrophage polarization in experimental atherosclerosis. These endeavors will provide us with great mechanical insight. Subsequently, by isolating those mediators important to murine polarization and ablating them in human macrophages, we can identify new polarization markers or even a polarization-specific gene profile for macrophage subsets. Alternatively, the activity of polarizing transcription factors could be used as a measure of the inflammatory state of atherosclerotic lesions. Thereby, gene regulation pathways may be valuable for diagnosis and individual risk assessment, when combined with lipid profiles and CRP-levels. In the future, one might even think of monitoring the activity of these pathways to evaluate the effects of therapeutic intervention.
As such, the exploration of transcription factors and epigenetic modifiers that uniquely shape the macrophage phenotype has the ability to advance our insight into the macrophage polarization paradigm considerably. This upholds its potential relevance for integration into the diagnostic and therapeutic algorithms of human atherosclerosis, as described above. Ultimately, we expect the greatest benefit from cell type-specific strategies that allow us to gently shift the plaque macrophage towards a desirable, atheroprotective phenotype, possibly by targeting the appropriate signaling pathways.

Acknowledgments

Due to publisher restrictions regarding the number of publications cited in this review, we were unable to reference the primary paper in some cases.

Disclosure

No conflicts of interest relevant to this article were reported.

Open Access

This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 2.0 International 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.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Lopez AD, Mathers CD, Ezzati M, et al. Global and regional burden of disease and risk factors, 2001: systematic analysis of population health data. Lancet. 2006;367(9524):1747–57.PubMedCrossRef Lopez AD, Mathers CD, Ezzati M, et al. Global and regional burden of disease and risk factors, 2001: systematic analysis of population health data. Lancet. 2006;367(9524):1747–57.PubMedCrossRef
2.
Zurück zum Zitat Lloyd-Jones D, Adams RJ, Brown TM, et al. Heart disease and stroke statistics–2010 update: a report from the American heart association. Circulation. 2010;121(7):e46–e215.PubMedCrossRef Lloyd-Jones D, Adams RJ, Brown TM, et al. Heart disease and stroke statistics–2010 update: a report from the American heart association. Circulation. 2010;121(7):e46–e215.PubMedCrossRef
3.
Zurück zum Zitat MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet. 2002;360(9326):7–22. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet. 2002;360(9326):7–22.
4.
Zurück zum Zitat Baigent C, Keech A, Kearney PM, et al. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet. 2005;366(9493):1267–78.PubMedCrossRef Baigent C, Keech A, Kearney PM, et al. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet. 2005;366(9493):1267–78.PubMedCrossRef
5.
Zurück zum Zitat Libby P. The forgotten majority: unfinished business in cardiovascular risk reduction. J Am Coll Cardiol. 2005;46(7):1225–8.PubMedCrossRef Libby P. The forgotten majority: unfinished business in cardiovascular risk reduction. J Am Coll Cardiol. 2005;46(7):1225–8.PubMedCrossRef
6.
Zurück zum Zitat Greenland P, Knoll MD, Stamler J, et al. Major risk factors as antecedents of fatal and nonfatal coronary heart disease events. JAMA. 2003;290(7):891–7.PubMedCrossRef Greenland P, Knoll MD, Stamler J, et al. Major risk factors as antecedents of fatal and nonfatal coronary heart disease events. JAMA. 2003;290(7):891–7.PubMedCrossRef
8.
Zurück zum Zitat Maradit-Kremers H, Nicola PJ, Crowson CS, et al. Cardiovascular death in rheumatoid arthritis: a population-based study. Arthritis Rheum. 2005;52(3):722–32.PubMedCrossRef Maradit-Kremers H, Nicola PJ, Crowson CS, et al. Cardiovascular death in rheumatoid arthritis: a population-based study. Arthritis Rheum. 2005;52(3):722–32.PubMedCrossRef
9.
Zurück zum Zitat Asanuma Y, Oeser A, Shintani AK, et al. Premature coronary-artery atherosclerosis in systemic lupus erythematosus. N Engl J Med. 2003;349(25):2407–15.PubMedCrossRef Asanuma Y, Oeser A, Shintani AK, et al. Premature coronary-artery atherosclerosis in systemic lupus erythematosus. N Engl J Med. 2003;349(25):2407–15.PubMedCrossRef
10.
Zurück zum Zitat Blake GJ, Ridker PM. Novel clinical markers of vascular wall inflammation. Circ Res. 2001;89(9):763–71.PubMedCrossRef Blake GJ, Ridker PM. Novel clinical markers of vascular wall inflammation. Circ Res. 2001;89(9):763–71.PubMedCrossRef
11.
Zurück zum Zitat Kaptoge S, Di Angelantonio E, Lowe G, et al. C-reactive protein concentration and risk of coronary heart disease, stroke, and mortality: an individual participant meta-analysis. Lancet. 2010;375(9709):132–40.PubMedCrossRef Kaptoge S, Di Angelantonio E, Lowe G, et al. C-reactive protein concentration and risk of coronary heart disease, stroke, and mortality: an individual participant meta-analysis. Lancet. 2010;375(9709):132–40.PubMedCrossRef
12.
Zurück zum Zitat Ridker PM. The time for cardiovascular inflammation reduction trials has arrived: how low to go for hsCRP? Arterioscler Thromb Vasc Biol. 2008;28(7):1222–4.PubMedCrossRef Ridker PM. The time for cardiovascular inflammation reduction trials has arrived: how low to go for hsCRP? Arterioscler Thromb Vasc Biol. 2008;28(7):1222–4.PubMedCrossRef
13.
Zurück zum Zitat Rubin J, Chang HJ, Nasir K, et al. Association between high-sensitivity C-reactive protein and coronary plaque subtypes assessed by 64-slice coronary computed tomography angiography in an asymptomatic population. Circ Cardiovasc Imaging. 2011;4(3):201–9.PubMedCrossRef Rubin J, Chang HJ, Nasir K, et al. Association between high-sensitivity C-reactive protein and coronary plaque subtypes assessed by 64-slice coronary computed tomography angiography in an asymptomatic population. Circ Cardiovasc Imaging. 2011;4(3):201–9.PubMedCrossRef
14.
Zurück zum Zitat Ridker PM, Rifai N, Pfeffer MA, et al. Inflammation, pravastatin, and the risk of coronary events after myocardial infarction in patients with average cholesterol levels. Cholesterol and Recurrent Events (CARE) investigators. Circulation. 1998;98(9):839–44.PubMed Ridker PM, Rifai N, Pfeffer MA, et al. Inflammation, pravastatin, and the risk of coronary events after myocardial infarction in patients with average cholesterol levels. Cholesterol and Recurrent Events (CARE) investigators. Circulation. 1998;98(9):839–44.PubMed
15.
Zurück zum Zitat Ridker PM, Cannon CP, Morrow D, et al. C-reactive protein levels and outcomes after statin therapy. N Engl J Med. 2005;352(1):20–8.PubMedCrossRef Ridker PM, Cannon CP, Morrow D, et al. C-reactive protein levels and outcomes after statin therapy. N Engl J Med. 2005;352(1):20–8.PubMedCrossRef
16.
Zurück zum Zitat Downs JR, Clearfield M, Weis S, et al. Primary prevention of acute coronary events with lovastatin in men and women with average cholesterol levels: results of AFCAPS/TexCAPS. Air Force/Texas coronary atherosclerosis prevention study. JAMA. 1998;279(20):1615–22.PubMedCrossRef Downs JR, Clearfield M, Weis S, et al. Primary prevention of acute coronary events with lovastatin in men and women with average cholesterol levels: results of AFCAPS/TexCAPS. Air Force/Texas coronary atherosclerosis prevention study. JAMA. 1998;279(20):1615–22.PubMedCrossRef
17.
Zurück zum Zitat Ridker PM, Danielson E, Fonseca FA, et al. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008;359(21):2195–207.PubMedCrossRef Ridker PM, Danielson E, Fonseca FA, et al. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med. 2008;359(21):2195–207.PubMedCrossRef
18.
Zurück zum Zitat • Ridker PM. Testing the inflammatory hypothesis of atherothrombosis: scientific rationale for the cardiovascular inflammation reduction trial (CIRT). J Thromb Haemost. 2009;7 Suppl 1:332–9. This prospective trial examined the impact of low-dose metothraxate on cardiovascular endpoints.PubMedCrossRef • Ridker PM. Testing the inflammatory hypothesis of atherothrombosis: scientific rationale for the cardiovascular inflammation reduction trial (CIRT). J Thromb Haemost. 2009;7 Suppl 1:332–9. This prospective trial examined the impact of low-dose metothraxate on cardiovascular endpoints.PubMedCrossRef
19.
Zurück zum Zitat • Ridker PM, Thuren T, Zalewski A, Libby P. Interleukin-1beta inhibition and the prevention of recurrent cardiovascular events: rationale and design of the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS). Am Heart J. 2011;162(4):597–605. This prospective trial studied the impact of canakinumab on cardiovascular endpoints.PubMedCrossRef • Ridker PM, Thuren T, Zalewski A, Libby P. Interleukin-1beta inhibition and the prevention of recurrent cardiovascular events: rationale and design of the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS). Am Heart J. 2011;162(4):597–605. This prospective trial studied the impact of canakinumab on cardiovascular endpoints.PubMedCrossRef
20.
Zurück zum Zitat Smith JD, Trogan E, Ginsberg M, et al. Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and apolipoprotein E. Proc Natl Acad Sci U S A. 1995;92(18):8264–8.PubMedCrossRef Smith JD, Trogan E, Ginsberg M, et al. Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and apolipoprotein E. Proc Natl Acad Sci U S A. 1995;92(18):8264–8.PubMedCrossRef
21.
Zurück zum Zitat Rajavashisth T, Qiao JH, Tripathi S, et al. Heterozygous osteopetrotic (op) mutation reduces atherosclerosis in LDL receptor- deficient mice. J Clin Invest. 1998;101(12):2702–10.PubMedCrossRef Rajavashisth T, Qiao JH, Tripathi S, et al. Heterozygous osteopetrotic (op) mutation reduces atherosclerosis in LDL receptor- deficient mice. J Clin Invest. 1998;101(12):2702–10.PubMedCrossRef
22.
Zurück zum Zitat Tacke F, Alvarez D, Kaplan TJ, et al. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest. 2007;117(1):185–94.PubMedCrossRef Tacke F, Alvarez D, Kaplan TJ, et al. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest. 2007;117(1):185–94.PubMedCrossRef
23.
Zurück zum Zitat Tedgui A, Mallat Z. Cytokines in atherosclerosis: pathogenic and regulatory pathways. Physiol Rev. 2006;86(2):515–81.PubMedCrossRef Tedgui A, Mallat Z. Cytokines in atherosclerosis: pathogenic and regulatory pathways. Physiol Rev. 2006;86(2):515–81.PubMedCrossRef
24.
Zurück zum Zitat van Vlijmen BJ, Gerritsen G, Franken AL, et al. Macrophage p53 deficiency leads to enhanced atherosclerosis in APOE*3-leiden transgenic mice. Circ Res. 2001;88(8):780–6.PubMedCrossRef van Vlijmen BJ, Gerritsen G, Franken AL, et al. Macrophage p53 deficiency leads to enhanced atherosclerosis in APOE*3-leiden transgenic mice. Circ Res. 2001;88(8):780–6.PubMedCrossRef
25.
Zurück zum Zitat Ball RY, Stowers EC, Burton JH, et al. Evidence that the death of macrophage foam cells contributes to the lipid core of atheroma. Atherosclerosis. 1995;114(1):45–54.PubMedCrossRef Ball RY, Stowers EC, Burton JH, et al. Evidence that the death of macrophage foam cells contributes to the lipid core of atheroma. Atherosclerosis. 1995;114(1):45–54.PubMedCrossRef
26.
Zurück zum Zitat Seimon T, Tabas I. Mechanisms and consequences of macrophage apoptosis in atherosclerosis. J Lipid Res. 2009;50(Suppl):S382–7.PubMedCrossRef Seimon T, Tabas I. Mechanisms and consequences of macrophage apoptosis in atherosclerosis. J Lipid Res. 2009;50(Suppl):S382–7.PubMedCrossRef
27.
Zurück zum Zitat Aikawa M, Rabkin E, Okada Y, et al. Lipid lowering by diet reduces matrix metalloproteinase activity and increases collagen content of rabbit atheroma: a potential mechanism of lesion stabilization. Circulation. 1998;97(24):2433–44.PubMed Aikawa M, Rabkin E, Okada Y, et al. Lipid lowering by diet reduces matrix metalloproteinase activity and increases collagen content of rabbit atheroma: a potential mechanism of lesion stabilization. Circulation. 1998;97(24):2433–44.PubMed
28.
Zurück zum Zitat Johnson JL, Sala-Newby GB, Ismail Y, et al. 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.PubMedCrossRef Johnson JL, Sala-Newby GB, Ismail Y, et al. 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.PubMedCrossRef
29.
Zurück zum Zitat Pedersen SF, Graebe M, Fisker Hag AM, et al. Gene expression and 18FDG uptake in atherosclerotic carotid plaques. Nucl Med Commun. 2010;31(5):423–9.PubMed Pedersen SF, Graebe M, Fisker Hag AM, et al. Gene expression and 18FDG uptake in atherosclerotic carotid plaques. Nucl Med Commun. 2010;31(5):423–9.PubMed
30.
Zurück zum Zitat Rudd JH, Hyafil F, Fayad ZA. Inflammation imaging in atherosclerosis. Arterioscler Thromb Vasc Biol. 2009;29(7):1009–16.PubMedCrossRef Rudd JH, Hyafil F, Fayad ZA. Inflammation imaging in atherosclerosis. Arterioscler Thromb Vasc Biol. 2009;29(7):1009–16.PubMedCrossRef
31.
Zurück zum Zitat van der Wal AC, Das PK, Tigges AJ, Becker AE. Macrophage differentiation in atherosclerosis. An in situ immunohistochemical analysis in humans. Am J Pathol. 1992;141(1):161–8.PubMed van der Wal AC, Das PK, Tigges AJ, Becker AE. Macrophage differentiation in atherosclerosis. An in situ immunohistochemical analysis in humans. Am J Pathol. 1992;141(1):161–8.PubMed
32.
33.
Zurück zum Zitat Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453–61.PubMedCrossRef Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453–61.PubMedCrossRef
34.
Zurück zum Zitat Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8(12):958–69.PubMedCrossRef Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8(12):958–69.PubMedCrossRef
35.
Zurück zum Zitat Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011;11(11):723–37.PubMedCrossRef Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011;11(11):723–37.PubMedCrossRef
36.
Zurück zum Zitat Loke P, Gallagher I, Nair MG, et al. Alternative activation is an innate response to injury that requires CD4+ T cells to be sustained during chronic infection. J Immunol. 2007;179(6):3926–36.PubMed Loke P, Gallagher I, Nair MG, et al. Alternative activation is an innate response to injury that requires CD4+ T cells to be sustained during chronic infection. J Immunol. 2007;179(6):3926–36.PubMed
37.
Zurück zum Zitat Reese TA, Liang HE, Tager AM, et al. Chitin induces accumulation in tissue of innate immune cells associated with allergy. Nature. 2007;447(7140):92–6.PubMedCrossRef Reese TA, Liang HE, Tager AM, et al. Chitin induces accumulation in tissue of innate immune cells associated with allergy. Nature. 2007;447(7140):92–6.PubMedCrossRef
38.
Zurück zum Zitat Hesse M, Modolell M, La Flamme AC, et al. Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of L-arginine metabolism. J Immunol. 2001;167(11):6533–44.PubMed Hesse M, Modolell M, La Flamme AC, et al. Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of L-arginine metabolism. J Immunol. 2001;167(11):6533–44.PubMed
39.
Zurück zum Zitat Couper KN, Blount DG, Riley EM. IL-10: the master regulator of immunity to infection. J Immunol. 2008;180(9):5771–7.PubMed Couper KN, Blount DG, Riley EM. IL-10: the master regulator of immunity to infection. J Immunol. 2008;180(9):5771–7.PubMed
40.
Zurück zum Zitat Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32(5):593–604.PubMedCrossRef Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32(5):593–604.PubMedCrossRef
41.
Zurück zum Zitat Fleetwood AJ, Dinh H, Cook AD, et al. GM-CSF- and M-CSF-dependent macrophage phenotypes display differential dependence on type I interferon signaling. J Leukoc Biol. 2009;86(2):411–21.PubMedCrossRef Fleetwood AJ, Dinh H, Cook AD, et al. GM-CSF- and M-CSF-dependent macrophage phenotypes display differential dependence on type I interferon signaling. J Leukoc Biol. 2009;86(2):411–21.PubMedCrossRef
42.
Zurück zum Zitat Fleetwood AJ, Lawrence T, Hamilton JA, Cook AD. Granulocyte-macrophage colony-stimulating factor (CSF) and macrophage CSF-dependent macrophage phenotypes display differences in cytokine profiles and transcription factor activities: implications for CSF blockade in inflammation. J Immunol. 2007;178(8):5245–52.PubMed Fleetwood AJ, Lawrence T, Hamilton JA, Cook AD. Granulocyte-macrophage colony-stimulating factor (CSF) and macrophage CSF-dependent macrophage phenotypes display differences in cytokine profiles and transcription factor activities: implications for CSF blockade in inflammation. J Immunol. 2007;178(8):5245–52.PubMed
43.
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.PubMedCrossRef 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.PubMedCrossRef
44.
Zurück zum Zitat Kadl A, Meher AK, Sharma PR, et al. Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circ Res. 2010;107(6):737–46.PubMedCrossRef Kadl A, Meher AK, Sharma PR, et al. Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circ Res. 2010;107(6):737–46.PubMedCrossRef
45.
Zurück zum Zitat • Lee S, Huen S, Nishio H, et al. Distinct macrophage phenotypes contribute to kidney injury and repair. J Am Soc Nephrol. 2011;22(2):317–26. This article shows macrophage plasticity in a transient in vivo model of inflammation.PubMedCrossRef • Lee S, Huen S, Nishio H, et al. Distinct macrophage phenotypes contribute to kidney injury and repair. J Am Soc Nephrol. 2011;22(2):317–26. This article shows macrophage plasticity in a transient in vivo model of inflammation.PubMedCrossRef
46.
Zurück zum Zitat Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117(1):175–84.PubMedCrossRef Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117(1):175–84.PubMedCrossRef
47.
Zurück zum Zitat Stienstra R, van Diepen JA, Tack CJ, et al. Inflammasome is a central player in the induction of obesity and insulin resistance. Proc Natl Acad Sci U S A. 2011;108(37):15324–9.PubMedCrossRef Stienstra R, van Diepen JA, Tack CJ, et al. Inflammasome is a central player in the induction of obesity and insulin resistance. Proc Natl Acad Sci U S A. 2011;108(37):15324–9.PubMedCrossRef
48.
Zurück zum Zitat Chawla A, Nguyen KD, Goh YP. Macrophage-mediated inflammation in metabolic disease. Nat Rev Immunol. 2011;11(11):738–49.PubMedCrossRef Chawla A, Nguyen KD, Goh YP. Macrophage-mediated inflammation in metabolic disease. Nat Rev Immunol. 2011;11(11):738–49.PubMedCrossRef
49.
Zurück zum Zitat Stoger JL, Goossens P, de Winther MP. Macrophage heterogeneity: relevance and functional implications in atherosclerosis. Curr Vasc Pharmacol. 2010;8(2):233–48.PubMedCrossRef Stoger JL, Goossens P, de Winther MP. Macrophage heterogeneity: relevance and functional implications in atherosclerosis. Curr Vasc Pharmacol. 2010;8(2):233–48.PubMedCrossRef
50.
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.PubMedCrossRef 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.PubMedCrossRef
51.
Zurück zum Zitat Wolfs IM, Donners MM, de Winther MP. Differentiation factors and cytokines in the atherosclerotic plaque micro-environment as a trigger for macrophage polarisation. Thromb Haemost. 2011;106(5):763–71.PubMedCrossRef Wolfs IM, Donners MM, de Winther MP. Differentiation factors and cytokines in the atherosclerotic plaque micro-environment as a trigger for macrophage polarisation. Thromb Haemost. 2011;106(5):763–71.PubMedCrossRef
52.
Zurück zum Zitat Khallou-Laschet J, Varthaman A, Fornasa G, et al. Macrophage plasticity in experimental atherosclerosis. PLoS One. 2010;5(1):e8852.PubMedCrossRef Khallou-Laschet J, Varthaman A, Fornasa G, et al. Macrophage plasticity in experimental atherosclerosis. PLoS One. 2010;5(1):e8852.PubMedCrossRef
53.
Zurück zum Zitat •• Feig JE, Parathath S, Rong JX, et al. Reversal of hyperlipidemia with a genetic switch favorably affects the content and inflammatory state of macrophages in atherosclerotic plaques. Circulation. 2011;123(9):989–98. This article very elegantly shows that induction of atherosclerosis regression by reversal of hyperlipidemia leads to a shift from CAM macrophages to an AAM phenotype in atherosclerotic lesions.PubMedCrossRef •• Feig JE, Parathath S, Rong JX, et al. Reversal of hyperlipidemia with a genetic switch favorably affects the content and inflammatory state of macrophages in atherosclerotic plaques. Circulation. 2011;123(9):989–98. This article very elegantly shows that induction of atherosclerosis regression by reversal of hyperlipidemia leads to a shift from CAM macrophages to an AAM phenotype in atherosclerotic lesions.PubMedCrossRef
54.
Zurück zum Zitat Shalhoub J, Cross A, Allin D, et al. Cytokine profiling in culture reveals a predominance of M1 macrophage polarisation in symptomatic carotid plaques. Presented on the BSCR Spring Meeting. 2010;96(17):e23–e23. Shalhoub J, Cross A, Allin D, et al. Cytokine profiling in culture reveals a predominance of M1 macrophage polarisation in symptomatic carotid plaques. Presented on the BSCR Spring Meeting. 2010;96(17):e23–e23.
55.
Zurück zum Zitat Hirata Y, Tabata M, Kurobe H, et al. Coronary atherosclerosis is associated with macrophage polarization in epicardial adipose tissue. J Am Coll Cardiol. 2011;58(3):248–55.PubMedCrossRef Hirata Y, Tabata M, Kurobe H, et al. Coronary atherosclerosis is associated with macrophage polarization in epicardial adipose tissue. J Am Coll Cardiol. 2011;58(3):248–55.PubMedCrossRef
56.
Zurück zum Zitat Shalhoub J, Falck-Hansen MA, Davies AH, Monaco C. Innate immunity and monocyte-macrophage activation in atherosclerosis. J Inflamm (Lond). 2011;8:9.CrossRef Shalhoub J, Falck-Hansen MA, Davies AH, Monaco C. Innate immunity and monocyte-macrophage activation in atherosclerosis. J Inflamm (Lond). 2011;8:9.CrossRef
57.
Zurück zum Zitat Murray PJ, Wynn TA. Obstacles and opportunities for understanding macrophage polarization. J Leukoc Biol. 2011;89(4):557–63.PubMedCrossRef Murray PJ, Wynn TA. Obstacles and opportunities for understanding macrophage polarization. J Leukoc Biol. 2011;89(4):557–63.PubMedCrossRef
58.
Zurück zum Zitat Lawrence T, Natoli G. Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nat Rev Immunol. 2011;11(11):750–61.PubMedCrossRef Lawrence T, Natoli G. Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nat Rev Immunol. 2011;11(11):750–61.PubMedCrossRef
59.
Zurück zum Zitat Soler C, Felipe A, Garcia-Manteiga J, et al. Interferon-gamma regulates nucleoside transport systems in macrophages through signal transduction and activator of transduction factor 1 (STAT1)-dependent and -independent signalling pathways. Biochem J. 2003;375(Pt 3):777–83.PubMedCrossRef Soler C, Felipe A, Garcia-Manteiga J, et al. Interferon-gamma regulates nucleoside transport systems in macrophages through signal transduction and activator of transduction factor 1 (STAT1)-dependent and -independent signalling pathways. Biochem J. 2003;375(Pt 3):777–83.PubMedCrossRef
60.
Zurück zum Zitat Agrawal S, Febbraio M, Podrez E, et al. Signal transducer and activator of transcription 1 is required for optimal foam cell formation and atherosclerotic lesion development. Circulation. 2007;115(23):2939–47.PubMedCrossRef Agrawal S, Febbraio M, Podrez E, et al. Signal transducer and activator of transcription 1 is required for optimal foam cell formation and atherosclerotic lesion development. Circulation. 2007;115(23):2939–47.PubMedCrossRef
61.
Zurück zum Zitat Lim WS, Timmins JM, Seimon TA, et al. Signal transducer and activator of transcription-1 is critical for apoptosis in macrophages subjected to endoplasmic reticulum stress in vitro and in advanced atherosclerotic lesions in vivo. Circulation. 2008;117(7):940–51.PubMedCrossRef Lim WS, Timmins JM, Seimon TA, et al. Signal transducer and activator of transcription-1 is critical for apoptosis in macrophages subjected to endoplasmic reticulum stress in vitro and in advanced atherosclerotic lesions in vivo. Circulation. 2008;117(7):940–51.PubMedCrossRef
62.
Zurück zum Zitat Ishii M, Wen H, Corsa CA, et al. Epigenetic regulation of the alternatively activated macrophage phenotype. Blood. 2009;114(15):3244–54.PubMedCrossRef Ishii M, Wen H, Corsa CA, et al. Epigenetic regulation of the alternatively activated macrophage phenotype. Blood. 2009;114(15):3244–54.PubMedCrossRef
63.
Zurück zum Zitat Lawrence T, Gilroy DW, Colville-Nash PR, Willoughby DA. Possible new role for NF-kappaB in the resolution of inflammation. Nat Med. 2001;7(12):1291–7.PubMedCrossRef Lawrence T, Gilroy DW, Colville-Nash PR, Willoughby DA. Possible new role for NF-kappaB in the resolution of inflammation. Nat Med. 2001;7(12):1291–7.PubMedCrossRef
64.
Zurück zum Zitat Brand K, Page S, Rogler G, et al. Activated transcription factor nuclear factor-kappa B is present in the atherosclerotic lesion. J Clin Invest. 1996;97(7):1715–22.PubMedCrossRef Brand K, Page S, Rogler G, et al. Activated transcription factor nuclear factor-kappa B is present in the atherosclerotic lesion. J Clin Invest. 1996;97(7):1715–22.PubMedCrossRef
65.
Zurück zum Zitat Fong CH, Bebien M, Didierlaurent A, et al. An antiinflammatory role for IKKbeta through the inhibition of “classical” macrophage activation. J Exp Med. 2008;205(6):1269–76.PubMedCrossRef Fong CH, Bebien M, Didierlaurent A, et al. An antiinflammatory role for IKKbeta through the inhibition of “classical” macrophage activation. J Exp Med. 2008;205(6):1269–76.PubMedCrossRef
66.
Zurück zum Zitat Kanters E, Pasparakis M, Gijbels MJ, et al. Inhibition of NF-kappaB activation in macrophages increases atherosclerosis in LDL receptor-deficient mice. J Clin Invest. 2003;112(8):1176–85.PubMed Kanters E, Pasparakis M, Gijbels MJ, et al. Inhibition of NF-kappaB activation in macrophages increases atherosclerosis in LDL receptor-deficient mice. J Clin Invest. 2003;112(8):1176–85.PubMed
67.
Zurück zum Zitat Porta C, Rimoldi M, Raes G, et al. Tolerance and M2 (alternative) macrophage polarization are related processes orchestrated by p50 nuclear factor kappaB. Proc Natl Acad Sci U S A. 2009;106(35):14978–83.PubMedCrossRef Porta C, Rimoldi M, Raes G, et al. Tolerance and M2 (alternative) macrophage polarization are related processes orchestrated by p50 nuclear factor kappaB. Proc Natl Acad Sci U S A. 2009;106(35):14978–83.PubMedCrossRef
68.
Zurück zum Zitat Kanters E, Gijbels MJ, van der Made I, et al. Hematopoietic NF-kappaB1 deficiency results in small atherosclerotic lesions with an inflammatory phenotype. Blood. 2004;103(3):934–40.PubMedCrossRef Kanters E, Gijbels MJ, van der Made I, et al. Hematopoietic NF-kappaB1 deficiency results in small atherosclerotic lesions with an inflammatory phenotype. Blood. 2004;103(3):934–40.PubMedCrossRef
69.
Zurück zum Zitat Satoh T, Takeuchi O, Vandenbon A, et al. The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat Immunol. 2010;11(10):936–44.PubMedCrossRef Satoh T, Takeuchi O, Vandenbon A, et al. The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat Immunol. 2010;11(10):936–44.PubMedCrossRef
70.
Zurück zum Zitat Takaoka A, Yanai H, Kondo S, et al. Integral role of IRF-5 in the gene induction programme activated by toll-like receptors. Nature. 2005;434(7030):243–9.PubMedCrossRef Takaoka A, Yanai H, Kondo S, et al. Integral role of IRF-5 in the gene induction programme activated by toll-like receptors. Nature. 2005;434(7030):243–9.PubMedCrossRef
71.
Zurück zum Zitat Krausgruber T, Blazek K, Smallie T, et al. IRF5 Promotes inflammatory macrophage polarization and TH1-TH17 responses. Nat Immunol. 2011;12(3):231–8.PubMedCrossRef Krausgruber T, Blazek K, Smallie T, et al. IRF5 Promotes inflammatory macrophage polarization and TH1-TH17 responses. Nat Immunol. 2011;12(3):231–8.PubMedCrossRef
72.
Zurück zum Zitat Liu J, Cao S, Herman LM, Ma X. Differential regulation of interleukin (IL)-12 p35 and p40 gene expression and interferon (IFN)-gamma-primed IL-12 production by IFN regulatory factor 1. J Exp Med. 2003;198(8):1265–76.PubMedCrossRef Liu J, Cao S, Herman LM, Ma X. Differential regulation of interleukin (IL)-12 p35 and p40 gene expression and interferon (IFN)-gamma-primed IL-12 production by IFN regulatory factor 1. J Exp Med. 2003;198(8):1265–76.PubMedCrossRef
73.
Zurück zum Zitat Yoshida K, Yamamoto K, Kohno T, et al. Active repression of IFN regulatory factor-1-mediated transactivation by IFN regulatory factor-4. Int Immunol. 2005;17(11):1463–71.PubMedCrossRef Yoshida K, Yamamoto K, Kohno T, et al. Active repression of IFN regulatory factor-1-mediated transactivation by IFN regulatory factor-4. Int Immunol. 2005;17(11):1463–71.PubMedCrossRef
74.
Zurück zum Zitat Guo M, Mao X, Ji Q, et al. Inhibition of IFN regulatory factor-1 down-regulate Th1 cell function in patients with acute coronary syndrome. J Clin Immunol. 2010;30(2):241–52.PubMedCrossRef Guo M, Mao X, Ji Q, et al. Inhibition of IFN regulatory factor-1 down-regulate Th1 cell function in patients with acute coronary syndrome. J Clin Immunol. 2010;30(2):241–52.PubMedCrossRef
75.
Zurück zum Zitat Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, et al. Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature. 2007;447(7148):1116–20.PubMedCrossRef Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, et al. Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature. 2007;447(7148):1116–20.PubMedCrossRef
76.
Zurück zum Zitat Bouhlel MA, Derudas B, Rigamonti E, et al. PPARgamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab. 2007;6(2):137–43.PubMedCrossRef Bouhlel MA, Derudas B, Rigamonti E, et al. PPARgamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab. 2007;6(2):137–43.PubMedCrossRef
77.
Zurück zum Zitat Pourcet B, Feig JE, Vengrenyuk Y, et al. LXRalpha regulates macrophage arginase 1 through PU.1 and interferon regulatory factor 8. Circ Res. 2011;109(5):492–501.PubMedCrossRef Pourcet B, Feig JE, Vengrenyuk Y, et al. LXRalpha regulates macrophage arginase 1 through PU.1 and interferon regulatory factor 8. Circ Res. 2011;109(5):492–501.PubMedCrossRef
78.
Zurück zum Zitat Babaev VR, Yancey PG, Ryzhov SV, et al. Conditional knockout of macrophage PPARgamma increases atherosclerosis in C57BL/6 and low-density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol. 2005;25(8):1647–53.PubMedCrossRef Babaev VR, Yancey PG, Ryzhov SV, et al. Conditional knockout of macrophage PPARgamma increases atherosclerosis in C57BL/6 and low-density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol. 2005;25(8):1647–53.PubMedCrossRef
79.
Zurück zum Zitat Li G, Biju KC, Xu X, et al. Macrophage LXRalpha gene therapy ameliorates atherosclerosis as well as hypertriglyceridemia in LDLR(-/-) mice. Gene Ther. 2011;18(8):835–41.PubMedCrossRef Li G, Biju KC, Xu X, et al. Macrophage LXRalpha gene therapy ameliorates atherosclerosis as well as hypertriglyceridemia in LDLR(-/-) mice. Gene Ther. 2011;18(8):835–41.PubMedCrossRef
80.
Zurück zum Zitat Qu A, Shah YM, Manna SK, Gonzalez FJ. Disruption of endothelial peroxisome proliferator-activated receptor gamma accelerates diet-induced atherogenesis in LDL receptor-null mice. Arterioscler Thromb Vasc Biol. 2012;32(1):65–73.PubMedCrossRef Qu A, Shah YM, Manna SK, Gonzalez FJ. Disruption of endothelial peroxisome proliferator-activated receptor gamma accelerates diet-induced atherogenesis in LDL receptor-null mice. Arterioscler Thromb Vasc Biol. 2012;32(1):65–73.PubMedCrossRef
81.
Zurück zum Zitat • Chinetti-Gbaguidi G, Baron M, Bouhlel MA, et al. Human atherosclerotic plaque alternative macrophages display low cholesterol handling but high phagocytosis because of distinct activities of the PPARgamma and LXRalpha pathways. Circ Res. 2011;108(8):985–95. This group (also reference 76) was the first to demonstrate CAM and AAM macrophages in human atherosclerosis and identified functional differences in macrophage lipid handling.PubMedCrossRef • Chinetti-Gbaguidi G, Baron M, Bouhlel MA, et al. Human atherosclerotic plaque alternative macrophages display low cholesterol handling but high phagocytosis because of distinct activities of the PPARgamma and LXRalpha pathways. Circ Res. 2011;108(8):985–95. This group (also reference 76) was the first to demonstrate CAM and AAM macrophages in human atherosclerosis and identified functional differences in macrophage lipid handling.PubMedCrossRef
82.
Zurück zum Zitat Buxadé M, Lunazzi G, Minguillón J, et al. Gene expression induced by Toll-like receptors in macrophages requires the transcription factor NFAT5. JEM. 2012. Buxadé M, Lunazzi G, Minguillón J, et al. Gene expression induced by Toll-like receptors in macrophages requires the transcription factor NFAT5. JEM. 2012.
83.
Zurück zum Zitat Halterman JA, Kwon HM, Zargham R, et al. Nuclear factor of activated T cells 5 regulates vascular smooth muscle cell phenotypic modulation. Arterioscler Thromb Vasc Biol. 2011;31(10):2287–96.PubMedCrossRef Halterman JA, Kwon HM, Zargham R, et al. Nuclear factor of activated T cells 5 regulates vascular smooth muscle cell phenotypic modulation. Arterioscler Thromb Vasc Biol. 2011;31(10):2287–96.PubMedCrossRef
84.
Zurück zum Zitat Whyte CS, Bishop ET, Ruckerl D, et al. Suppressor of cytokine signaling (SOCS)1 is a key determinant of differential macrophage activation and function. J Leukoc Biol. 2011;90(5):845–54.PubMedCrossRef Whyte CS, Bishop ET, Ruckerl D, et al. Suppressor of cytokine signaling (SOCS)1 is a key determinant of differential macrophage activation and function. J Leukoc Biol. 2011;90(5):845–54.PubMedCrossRef
85.
Zurück zum Zitat Ortiz-Munoz G, Martin-Ventura JL, Hernandez-Vargas P, et al. Suppressors of cytokine signaling modulate JAK/STAT-mediated cell responses during atherosclerosis. Arterioscler Thromb Vasc Biol. 2009;29(4):525–31.PubMedCrossRef Ortiz-Munoz G, Martin-Ventura JL, Hernandez-Vargas P, et al. Suppressors of cytokine signaling modulate JAK/STAT-mediated cell responses during atherosclerosis. Arterioscler Thromb Vasc Biol. 2009;29(4):525–31.PubMedCrossRef
86.
Zurück zum Zitat Liu Y, Stewart KN, Bishop E, et al. Unique expression of suppressor of cytokine signaling 3 is essential for classical macrophage activation in rodents in vitro and in vivo. J Immunol. 2008;180(9):6270–8.PubMed Liu Y, Stewart KN, Bishop E, et al. Unique expression of suppressor of cytokine signaling 3 is essential for classical macrophage activation in rodents in vitro and in vivo. J Immunol. 2008;180(9):6270–8.PubMed
87.
Zurück zum Zitat Williams L, Bradley L, Smith A, Foxwell B. Signal transducer and activator of transcription 3 is the dominant mediator of the anti-inflammatory effects of IL-10 in human macrophages. J Immunol. 2004;172(1):567–76.PubMed Williams L, Bradley L, Smith A, Foxwell B. Signal transducer and activator of transcription 3 is the dominant mediator of the anti-inflammatory effects of IL-10 in human macrophages. J Immunol. 2004;172(1):567–76.PubMed
88.
Zurück zum Zitat Khan JA, Cao M, Kang BY, et al. AAV/hSTAT3-gene delivery lowers aortic inflammatory cell infiltration in LDLR KO mice on high cholesterol. Atherosclerosis. 2010;213(1):59–66.PubMedCrossRef Khan JA, Cao M, Kang BY, et al. AAV/hSTAT3-gene delivery lowers aortic inflammatory cell infiltration in LDLR KO mice on high cholesterol. Atherosclerosis. 2010;213(1):59–66.PubMedCrossRef
89.
Zurück zum Zitat Ruffell D, Mourkioti F, Gambardella A, et al. A CREB-C/EBPbeta cascade induces M2 macrophage-specific gene expression and promotes muscle injury repair. Proc Natl Acad Sci U S A. 2009;106(41):17475–80.PubMedCrossRef Ruffell D, Mourkioti F, Gambardella A, et al. A CREB-C/EBPbeta cascade induces M2 macrophage-specific gene expression and promotes muscle injury repair. Proc Natl Acad Sci U S A. 2009;106(41):17475–80.PubMedCrossRef
90.
Zurück zum Zitat van Tits LJ, Stienstra R, van Lent PL, et al. Oxidized LDL enhances pro-inflammatory responses of alternatively activated M2 macrophages: a crucial role for kruppel-like factor 2. Atherosclerosis. 2011;214(2):345–9.PubMedCrossRef van Tits LJ, Stienstra R, van Lent PL, et al. Oxidized LDL enhances pro-inflammatory responses of alternatively activated M2 macrophages: a crucial role for kruppel-like factor 2. Atherosclerosis. 2011;214(2):345–9.PubMedCrossRef
91.
Zurück zum Zitat Atkins GB, Wang Y, Mahabeleshwar GH, et al. Hemizygous deficiency of kruppel-like factor 2 augments experimental atherosclerosis. Circ Res. 2008;103(7):690–3.PubMedCrossRef Atkins GB, Wang Y, Mahabeleshwar GH, et al. Hemizygous deficiency of kruppel-like factor 2 augments experimental atherosclerosis. Circ Res. 2008;103(7):690–3.PubMedCrossRef
92.
Zurück zum Zitat Liao X, Sharma N, Kapadia F, et al. Kruppel-like factor 4 regulates macrophage polarization. J Clin Invest. 2011;121(7):2736–49.PubMedCrossRef Liao X, Sharma N, Kapadia F, et al. Kruppel-like factor 4 regulates macrophage polarization. J Clin Invest. 2011;121(7):2736–49.PubMedCrossRef
93.
Zurück zum Zitat Takeuch O, Akira S. Epigenetic control of macrophage polarization. Eur J Immunol. 2011;41(9):2490–3.PubMedCrossRef Takeuch O, Akira S. Epigenetic control of macrophage polarization. Eur J Immunol. 2011;41(9):2490–3.PubMedCrossRef
94.
Zurück zum Zitat De Santa F, Totaro MG, Prosperini E, et al. The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing. Cell. 2007;130(6):1083–94.PubMedCrossRef De Santa F, Totaro MG, Prosperini E, et al. The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing. Cell. 2007;130(6):1083–94.PubMedCrossRef
95.
Zurück zum Zitat De Santa F, Narang V, Yap ZH, et al. Jmjd3 Contributes to the control of gene expression in LPS-activated macrophages. EMBO J. 2009;28(21):3341–52.PubMedCrossRef De Santa F, Narang V, Yap ZH, et al. Jmjd3 Contributes to the control of gene expression in LPS-activated macrophages. EMBO J. 2009;28(21):3341–52.PubMedCrossRef
96.
Zurück zum Zitat Mullican SE, Gaddis CA, Alenghat T, et al. Histone deacetylase 3 is an epigenomic brake in macrophage alternative activation. Genes Dev. 2011;25(23):2480–8.PubMedCrossRef Mullican SE, Gaddis CA, Alenghat T, et al. Histone deacetylase 3 is an epigenomic brake in macrophage alternative activation. Genes Dev. 2011;25(23):2480–8.PubMedCrossRef
97.
Zurück zum Zitat • Nicodeme E, Jeffrey KL, Schaefer U, et al. Suppression of inflammation by a synthetic histone mimic. Nature. 2010;468(7327):1119–23. This article is the first to demonstrate that pharmacologic disruption of chromatin complexes can be used to inhibit inflammatory responses.PubMedCrossRef • Nicodeme E, Jeffrey KL, Schaefer U, et al. Suppression of inflammation by a synthetic histone mimic. Nature. 2010;468(7327):1119–23. This article is the first to demonstrate that pharmacologic disruption of chromatin complexes can be used to inhibit inflammatory responses.PubMedCrossRef
98.
Zurück zum Zitat Alkemade FE, van Vliet P, Henneman P, et al. Prenatal exposure to apoE deficiency and postnatal hypercholesterolemia are associated with altered cell-specific lysine methyltransferase and histone methylation patterns in the vasculature. Am J Pathol. 2010;176(2):542–8.PubMedCrossRef Alkemade FE, van Vliet P, Henneman P, et al. Prenatal exposure to apoE deficiency and postnatal hypercholesterolemia are associated with altered cell-specific lysine methyltransferase and histone methylation patterns in the vasculature. Am J Pathol. 2010;176(2):542–8.PubMedCrossRef
99.
Zurück zum Zitat Wierda RJ, Geutskens SB, Jukema JW, et al. Epigenetics in atherosclerosis and inflammation. J Cell Mol Med. 2010;14(6A):1225–40.PubMedCrossRef Wierda RJ, Geutskens SB, Jukema JW, et al. Epigenetics in atherosclerosis and inflammation. J Cell Mol Med. 2010;14(6A):1225–40.PubMedCrossRef
100.
Zurück zum Zitat Weber C, Zernecke A, Libby P. The multifaceted contributions of leukocyte subsets to atherosclerosis: lessons from mouse models. Nat Rev Immunol. 2008;8(10):802–15.PubMedCrossRef Weber C, Zernecke A, Libby P. The multifaceted contributions of leukocyte subsets to atherosclerosis: lessons from mouse models. Nat Rev Immunol. 2008;8(10):802–15.PubMedCrossRef
Metadaten
Titel
Molecular Pathways Regulating Macrophage Polarization: Implications for Atherosclerosis
verfasst von
Marten A. Hoeksema
J. Lauran Stöger
Menno P. J. de Winther
Publikationsdatum
01.06.2012
Verlag
Current Science Inc.
Erschienen in
Current Atherosclerosis Reports / Ausgabe 3/2012
Print ISSN: 1523-3804
Elektronische ISSN: 1534-6242
DOI
https://doi.org/10.1007/s11883-012-0240-5

Weitere Artikel der Ausgabe 3/2012

Current Atherosclerosis Reports 3/2012 Zur Ausgabe

Vascular Biology (RS Rosenson, Section Editor)

Myeloperoxidase Production by Macrophage and Risk of Atherosclerosis

Genetics (AJ Marian, Section Editor)

Genetics of Thoracic Aortic Aneurysms

Genetics (AJ Marian, Section Editor)

The Complex Genetic Basis of Plasma Triglycerides

Vascular Biology (RS Rosenson, Section Editor)

Mitochondrial Reactive Oxygen Species and Risk of Atherosclerosis

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Eingreifen von Umstehenden rettet vor Erstickungstod!

15.05.2024 Fremdkörperaspiration Nachrichten

Wer sich an einem Essensrest verschluckt und um Luft ringt, benötigt vor allem rasche Hilfe. Dass Umstehende nur in jedem zweiten Erstickungsnotfall bereit waren, diese zu leisten, ist das ernüchternde Ergebnis einer Beobachtungsstudie aus Japan. Doch es gibt auch eine gute Nachricht.

Neue S3-Leitlinie zur unkomplizierten Zystitis: Auf Antibiotika verzichten?

15.05.2024 Harnwegsinfektionen Nachrichten

Welche Antibiotika darf man bei unkomplizierter Zystitis verwenden und wovon sollte man die Finger lassen? Welche pflanzlichen Präparate können helfen? Was taugt der zugelassene Impfstoff? Antworten vom Koordinator der frisch überarbeiteten S3-Leitlinie, Prof. Florian Wagenlehner.

Schadet Ärger den Gefäßen?

14.05.2024 Arteriosklerose Nachrichten

In einer Studie aus New York wirkte sich Ärger kurzfristig deutlich negativ auf die Endothelfunktion gesunder Probanden aus. Möglicherweise hat dies Einfluss auf die kardiovaskuläre Gesundheit.

Update Innere Medizin

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