Skip to main content

REVIEW article

Front. Physiol., 01 April 2022
Sec. Clinical and Translational Physiology
This article is part of the Research Topic Targeting Innate and Adaptive Immunity for Improvement of Cardiovascular Disease View all 9 articles

Targeting Cell-Specific Molecular Mechanisms of Innate Immunity in Atherosclerosis

  • 1Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
  • 2Department of Cardiology, University Heart Center Luebeck, University Hospital, Luebeck, Germany
  • 3DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany

Mechanisms of innate immunity contribute to inflammation, one of the major underlying causes of atherogenesis and progression of atherosclerotic vessel disease. How immune cells exactly contribute to atherosclerosis and interact with molecules of cholesterol homeostasis is still a matter of intense research. Recent evidence has proposed a potential role of previously underappreciated cell types in this chronic disease including platelets and dendritic cells (DCs). The pathophysiology of atherosclerosis is studied in models with dysfunctional lipid homeostasis and several druggable molecular targets are derived from these models. Specific therapeutic approaches focussing on these immune mechanisms, however, have not been successfully introduced into everyday clinical practice, yet. This review highlights molecular insights into immune processes related to atherosclerosis and potential future translational approaches targeting these molecular mechanisms.

Introduction

Atherosclerosis is an inflammatory disease, features a dysbalance in metabolism and together with its sequelay still has the largest contribution to deaths in developed countries—accounting for an estimated 17.9 million deaths every year (Collaborators, 2018; WHO CVD Risk Chart Working Group, 2019). The process of atherosclerotic plaque growth is initiated when cholesterol-containing low-density lipoproteins accumulate in the intima and activate the endothelium and subendothelial cells and structures (Libby et al., 2019). Subsequently, chemokines, endothelial and leukocyte adhesion molecules trigger recruitment of “classical” inflammatory cells related to atherosclerosis—monocytes and T cells (Gencer et al., 2021). Transformation of monocytes into macrophages is paralleled by internalization of lipoproteins, and foam cells develop (Bobryshev, 2006). Further release of cytokines, proteases, and vasoactive molecules from macrophages and foam cells is triggered by inflammatory signaling, lesional T-cells recognize local antigens and a T helper-1 response is generated with secretion of pro-inflammatory cytokines, which contributes to local inflammation and growth of the plaque (Frostegård et al., 1999; Ait-Oufella et al., 2011). This process requires co-stimulation for example, by the CD40-CD40L axis. At later stages, the plaque becomes vulnerable featuring local proteolysis, plaque rupture and finally thrombus formation with serious and potentially fatal events caused by ischemia and tissue infarction. Besides this classical picture, recently other cells have been centrally implicated in the process of atherogenesis (Figure 1) such as the classical antigen presenting cells of our body dendritic cells (DCs) or platelets, which are cells of the coagulation system.

FIGURE 1
www.frontiersin.org

FIGURE 1. Different cell types contribute to atheroprogression through various (dysregulated) processes.

Immunological memory was assumed to be an exclusive feature of the adaptive immune response for decades (Netea et al., 2020). But it was recently demonstrated that the innate immune system is able to generate non-specific memory responses, too—a phenomenon referred to as “trained immunity”. Innate immune cells were found to trigger a robust response to subsequent inflammatory challenges after initial activation by certain stimuli, for example, fungal-derived agents or vaccines (Penkov et al., 2019; Mitroulis I. et al., 2021). Surprisingly, macrophages can persistently switch their phenotype to be pro-inflammatory (M1-like) after a short stimulation with pathogens. These trained, pro-inflammatory macrophages show an pro-atherogenic phenotype and were detected in patients with established atherosclerosis as well as in humans after experimental infection for up to 3 months (Leentjens et al., 2018).

In this review, we focus on the influence of cell-cell interactions on atherogenesis and discuss the roles of mainly DCs and platelets in the context of the disease.

Previously Underappreciated Immune Cell Types Contributing to Atherogenesis

Cells of both innate and adaptive immunity are known key factors for the initiation as well as the progression of atherosclerotic disease (Libby, 2002; Wolf and Ley, 2019a). One of the most investigated cell types involved in atherosclerosis are monocytes/macrophages, which can be classified into different phenotypic subsets (M1 and M2 Macrophages) that play oppossing roles in the progression of the disease (Murray and Wynn, 2011; Murray et al., 2014) by acting pro- (M1 macrophages) or anti-inflammatory (M2 Macrophages). During the last few years, it has been shown that vascular smooth muscle cells are able to differentiate into macrophage-like cells within atherosclerotic plaques (Rong et al., 2003; Feil et al., 2014; Shankman et al., 2015; Vengrenyuk et al., 2015). Macrophages develop to foam cells by lipid accumulation during the development of an atherosclerotic plaque (Randolph, 2014). These foam cells are thought to constitute the core of the atherosclerotic plaque and contribute to plaque destabilization. However, in recent years also other cells of innate immunity have entered the stage of research and gained increased attention as potential important contributors to the disease, like T-cells and dendritic cells (DCs).

T cells represent a group of cells of different phenotypes, which also play different roles in the disease process of atherosclerosis. For example, T helper1 cells (Th1) have a pro-atherosclerotic effect, while regulatory T cells (Tregs) play a more atheroprotective role. Tregs, however, can also “turn bad” and become pro-atherogenic (Wolf and Ley, 2019b; Saigusa et al., 2020).

DCs have been identified some years back as the classical antigen presenting cells of our body (Banchereau and Steinman, 1998). Interestingly, DCs were detected by several groups in high abundance within the vascular wall of humans and mice (Bobryshev and Lord, 1995; Choi et al., 2009). The frequency of their location within the vessel wall changes depending on the shear stress the area is exposed to (Lord and Bobryshev, 1999; Yilmaz et al., 2004; Jongstra-Bilen et al., 2006). In the atherosclerotic ApoE knockout mouse model, accumulation of cholesterol in the cell membrane of DCs resulted in enhanced MHC-II-dependent antigen presentation and CD4+ T-cell activation (Bonacina et al., 2018). Whether DCs are protective or contribute to the extension of atherosclerotic plaques has been addressed in several studies, has surprisingly produced conflicting results and is, thus, still a matter of debate. A study by Gautier et al. using transgenic animals with BcL-2 expression under a CD11c promoter, which causes in vivo expansion of DCs, showed reduction of atherosclerosis, which suggests that DCs are protective (Gautier et al., 2009). Another study, however, which depleted DCs in vivo using the CD11c-diphteria toxin (DT) receptor model, has produced results with reduced atherosclerosis as well (Paulson et al., 2010). An explanation may be that repeated injections of DT is lethal for the animals (Jung et al., 2002; Probst et al., 2005).

Research on DCs in atherosclerosis carried out over the last decade reveals pro-as well as anti-atherogenic roles for DC subsets at all stages of atherogenesis explained by their numerous functions including lipid uptake, antigen presentation, efferocytosis, and inflammation resolution (Subramanian and Tabas, 2014; Zhao et al., 2021; Langer, 2022). Meanwhile, it is well established, that tolerogenic DCs, characterized by expression of CD103, act atheroprotective via induction of regulatory T cells (Hermansson et al., 2011). It has moreover been shown recently, that CD11c+ cells derived from murine bone marrow are able to efflux cholesterol and secrete ApoE which mediates an atheroprotective effect of these cells (Sauter et al., 2021).

When talking about DCs in atherosclerosis, it has to be considered that DCs represent a large group of cells whose precise definition is difficult because there is no specific, unique surface marker. CD11c, which is used predominantly as a “DC marker”, can also be expressed by macrophage subsets. Therefore, it is necessary to identify DCs by using additionally different cell-surface markers in parallel. DCs can be considered as CD11chigh, MHC-IIhigh expressing cells that can be CD11b±, F4/80±, CD103±, DEC-205±, Clec9a±, and Mertklo/. Other markers, namely Flt3, c-Kit, CD272, and CD26, which are expressed by classical DCs have been added in defining DC sub-populations, not only by classical flow-cytometry but also by “histo-cytometry”, in the last years (Gerner et al., 2012; Miller et al., 2012; Subramanian and Tabas, 2014).

Platelets are the most important cells of our body for wound sealing and initiation of the healing process after injury. Studies during the last two decades have moved platelets also more into the centre of inflammatory reactions (Koupenova et al., 2018). Some authors even suggest platelets to be part of the innate immune system (Croce and Libby, 2007). Interestingly, recent studies demonstrated the presence of platelets within the tissue after injury and their contribution to tissue remodeling processes such as apoptosis (Schleicher et al., 2015). In inflammation, platelets present receptors and release factors, which are important for the aggravation of the inflammatory response (Jenne et al., 2013). For development of atherosclerosis, the pontential significance of platelets has increased as studies some years ago demonstrated that platelet adhesion receptors or paracrine effectors can contribute significantly to plaque initiation and atherosclerotic inflammation (Massberg et al., 2002; Huo et al., 2003; Gawaz et al., 2005). Furthermore, at later stages of atherosclerosis, platelets are obviously crucial cells for the development of vessel occluding thrombi for example, in stroke or myocardial infarction patients, and targeting platelet associated mechanisms, which promote thrombus formation, is an approved approach to treat these patients (Nording et al., 2020).

Another immune cell type that is getting attention regarding its contribution to atheroprogression in the last years are neutrophil granulocytes (Döring et al., 2020). It has been shown before, that blood neutrophil counts in hypercholesterolaemic mice correlate with atherosclerotic lesion size (Drechsler et al., 2010). Neutrophils are able to form Neutrophil extracellular traps (NETs)—released decondensed chromatin that is decorated with granular proteins, forming a network of extracellular fibers (Brinkmann et al., 2004). NETs create a physical barrier that prevents the spread of pathogens and facilitates killing microbes by high concentrations of antimicrobial proteins and phagocytosis by other phagocytes (Brinkmann et al., 2004). NETs also promote thrombus formation (Laridan et al., 2019) and have been found in atherosclerotic plaques (Megens et al., 2012). The crosstalk of neutrophils with DCs can be realized via interactions between Mac-1 and DC-SIGN (van Gisbergen et al., 2005). Additionally, NETs and circulating immune complexes containing self-DNA and antimicrobial peptides were shown to trigger pDC activation via TLR9 (Lövgren et al., 2004; Means et al., 2005; Garcia-Romo et al., 2011; Lande et al., 2011). It was also reported that NETs in atherosclerotic lesions may stimulate a pDC-driven pathway of autoimmune activation and the generation of anti–double-stranded-DNA antibodies which may critically aggravate atherosclerosis lesion formation (Döring et al., 2012).

An interaction between neutrophils and platelets in atherosclerosis has been established. Activated platelets present HMGB1 to neutrophils, which causes them to perform autophagy and NET generation. This interaction may contribute to thromboinflammatory lesions (Maugeri et al., 2014). NETs are supposed to be involved at an early stage during the formation of coronary thrombus and lytic changes (Qi et al., 2017). A correlation between NET burden and DNase activity in ST-elevation acute coronary syndrome has been shown to be predictors of ST-segment resolution and infarct size (Mangold et al., 2015).

The role of dendritic cells (DCs) in atherosclerosis is controversial. DCs can present antigens to T cells, and may also shape plaque development by taking part in inflammatory processes as well as by influencing lipid metabolism. Platelets can interact directly with the endothelium and secrete different pro-atherogenic factors, which leads to dysfunction of the endothelium and recruitment of leukocytes. They furthermore play a significant role in immunity, express pro-inflammatory receptors (e.g., complement receptors) on their surface and contribute to atherosclerosis not only in their thrombus-forming function, but also via tissue remodelling, apoptosis and angiogenesis. Furthremore, mast cells take part in the plaque formation process and plaque growth by many different factors as indicated. Monocytes are the “progenitors” of macrophages. Macrophages turn into foam cells within plaques, the most prominent cell type within atheroclerotic lesions. Macrophages can be classified into a pro-inflammatory (regulatory, M2, Ly6C low expressing in mice), and anti-inflammatory cell (M1, Ly6C high expressing in mice), and express different cytokines or factors/mediators contributing to either inflammation (M1 macrophages) or tissue repair (M2 macrophages).

Implication of Dendritic Cells in Atherogenesis

DCs are cellular components of the body`’s immune response connecting the innate and the adaptive immune system. First, immature DCs sample antigens from their surrounding environment, which is in case of vessels the blood stream or the vascular wall. Interestingly, in mice regions of arterial curvature and branch points exposed to disturbed blood flow show a relative abundance of DCs (Lord and Bobryshev, 1999), implying that they are present in vascular regions being predisposed for atherosclerosis. Similarly, at later stages of atherosclerotic lesion development in patients, high numbers of mature DCs can be detected (Yilmaz et al., 2004). If one looks at patients with stable or unstable angina pectoris or myocardial infarction, the number of DCs in the atherosclerotic plaque increases, and their prevalence was found to correlate with a concomitant decrease in the number of circulating DCs and DC precursors (Yilmaz et al., 2006b). It has to be mentioned, however, that the clear identification of DCs is complex, particularly as they have overlapping phenotypes and share surface receptors with other immune cells (Schraml and Reis e Sousa, 2015). Therefore, further studies are needed using additional markers, which have been meanwhile identified. Compared to DCs in healthy patients, those found in patients with unstable angina produce higher levels of the proinflammatory mediators TNF-alpha and IL-1beta (Ranjit et al., 2004), and hallmarks of cardiovascular inflammation including CRP and IL- 6 correlate inversely with the number of circulating DCs (Kretzschmar et al., 2012). This could implicate a direct asscociation of the inflammatory response mediated by DCs with these acute phase proteins or vice versa. When DCs are activated by foreign antigens, for example, in the skin, they migrate to regional lymph nodes and present the ingested and processed antigens via the major histocompatibility complex (MHC) on their surface together with supporting signalling receptors such as CD80, CD86, and CD40 (Fujii et al., 2004). This process then causes activation of helper T-cells and killer T-cells targeted against the presented antigen (Ni and O'Neill, 1997). For the vascular wall, these processes are not as well characterized and the role of (auto-)antigens for atherosclerosis is albeit very likely, not profoundly proven nor fully understood. There are, however, interesting new concepts and very promising approaches using for example, auto-antigens for “vaccination” against atherosclerosis such as MDA-LDL, oxLDL or native LDL (George et al., 1998; Asgary et al., 2007). This and the potential of autoantigens such as oxidized LDL or heat shock proteins to induce some kind of autoimmunity during the process of atheroprogression underlines the involvement of DCs in atherogenesis (Kurien and Scofield, 2008; Rahman et al., 2017). Vaccines for infectious diseases and cancer are designed to boost the pro-inflammatory and lytic T cell response. A vaccine for atherosclerosis treatment in contrast has to induce immunological tolerance and/or functional neutralization to support the inflammatory response. To achieve this, there are two main strategies: inducing B cell-dependent production of neutralizing antibodies (e.g., anti-PCSK9 or anti-oxLDL antibodies) or inducing a durable Treg or Th1 response. At this point, it has to be mentioned that the idea of a vaccination against atherosclerosis still leaves us with important questions (e.g., vaccine formulation, route of delivery, schedule and durability of vaccination, proper patient selection for testing, and monitoring of efficacy endpoints or safety issues), which will have to be answered in extensive preclinical and clinical studies (Chyu and Shah, 2018; Roy et al., 2020).

How Metabolic Dysregulation Affects the Function of Innate Immune Cells Contributing to Atherosclerosis

The immune and metabolic response systems are linked to each other. They have evolved from common ancestral structures as shown by many evolutionary references (Leclerc and Reichhart, 2004). Obesity in humans leads to a dysregulation of the metabolic system resulting in a condition known as the “metabolic syndrome”. The metabolic syndrome causes a variety of pro-atherosclerotic effects on the arterial wall. Production of small dense LDL particles and decreased HDL levels increase vascular infiltration by lipids and the production of oxidiced LDL (ox-LDL). Ox-LDL delivers a danger signal to macrophages triggering development of foam cells, which in turn produce cytokines and growth factors for the growing atherosclerotic plaque. In addition, elevated blood levels of cytokines and of adipokines contribute to aggravation of the inflammatory reaction (Mathieu et al., 2006). In obese patients, a shift in macrophages towards a M1-like phenotype can be observed, which is caused by increased levels of free fatty acids (FFAs), cholesterol, LPS, and hypoxia (Kim et al., 2010), thereby further contributing to adipose tissue (AT) inflammation and inhibition of insulin signaling, whereas in lean individuums the anti-inflammatory M2-like macrophage phenotype prevails (Lumeng et al., 2008; Sun et al., 2021a). From a clinical prespective, it is undoubted and well established for decades that diabetes and atherosclerosis are linked, as the cardiovascular disease (CVD) risk is increased two-to-four-fold in diabetic persons compared to non-diabetic persons (Kannel and McGee, 1979). Importance of a metabolism—immune crosstalk involving DCs is documented by the fact that application of statins leads to a lower number of dendritic cells and less mature DCs in atherosclerotic plaques, representing one of the potential pleotropic anti-atherosclerotic effects of these clinically established substances (Yilmaz et al., 2006a). Interesintgly, statins not only reduce the risk of CV events by having an impact on cytokines such as CRP or IL-6 (Arnaud et al., 2005), but directly influence DC invasion (Kofler et al., 2008).

Dyslipidemia is one, but not the only epigenetic regulator that influences the action of antigen-presenting cells in the course of atherogenesis. Additionally, there exists a “response-to-injury hypothesis of atherosclerosis” (Glagov et al., 1987), which initially proposed that endothelial denudation is the first step in atherosclerosis and where activation and damage of the endothelial monolayer caused, for example ,by previous severe conditions including trauma, are supposed to effect a pool of endothelial progenitor cells and mono-nuclear progenitors in tissue reparation, triggering the development of the lesions (Du et al., 2012). It has been furthermore shown that metabolic changes can influence the phenotype and function of DCs (Sun et al., 2021b). Studies with patients suffering from diabetes mellitus type 1 and type 2 revealed that previous episodes of hyperglycemia can have a long-standing impact on the subsequent development of cardiovascular diseases—a phenomenon known as “metabolic memory” (Control et al., 2003; Holman et al., 2008). It has been postulated that epigenetic mechanisms may participate in conferring this metabolic memory (El-Osta et al., 2008). In vitro studies with aortic endothelial cells showed that transient incubation in high glucose followed by subsequent return of these cells to a normoglycemic environment was associated with increased gene expression of the p65 subunit of NF-κB, NF-κB activation, and expression of NF-κB–dependent proteins, including MCP-1 and VCAM-1(El-Osta et al., 2008).

DCs, Tregs and Atherosclerosis

DCs contribute to vascular inflammation by presentation of costimulatory molecules and subsequent signalling in immune cells, their activity may however be also controlled by T regulatory cells (TRegs (Lutgens et al., 2010). The main functions of Treg cells, which originate from CD4+ T cells, are the control of autoimmunity and the maintenance of self-tolerance. By interacting with DCs or their inhibition, Tregs are able to suppress the activation of effector T cells. This regulates priming and execution of T-effector responses (Ait-Oufella et al., 2014). Various cDC subtypes modulate Treg cell homeostasis in different ways (Roy et al., 2021), and Tregs are also involved in the activation, maturation, and function of DCs(Steinman and Banchereau, 2007; Chistiakov et al., 2013). T cell activation by DCs is dependent on toll-like receptor (TLR)-mediated maturation of DCs (Roy et al., 2021). In one study by Subramanian et al., bone marrow was transplanted from mice with CD11c+ DCs deficient in the TLR adaptor MYD88 into LDL receptor knockout mice, which led to decreased recruitment of both effector T cells and Treg cells to atherosclerotic lesions. The transplanted mice showed significantly larger atherosclerotic lesion sizes, propably caused by increased production of the monocyte-recruiting chemokine CCL2 due to the loss of Treg cells (Subramanian et al., 2013). Other studies have shown that the chemokines CCL17 and CCL22, secreted by tolerogenic CDs, can lead Tregs to atherosclerotic lesions. There, Tregs could suppress immunomodulatory properties of proinflammatory DCs and prevent differentiation of immature DCs to inflammatory subsets of mature DCs (Iellem et al., 2001; Weber et al., 2011; Chistiakov et al., 2014).

Platelets—Atherosclerosis

Platelet mediated thromboinflammation is an important cornerstone of atherosclerosis. Recently, an expert Consensus Document from the Third Maastricht Consensus Conference on Thrombosis was published, which focused both on the cellular and soluble parts of coagulation and their impact on inflammation in cardiovascular disease (d'Alessandro et al., 2020). Platelets are connected to vascular inflammation by adhesion receptors. They have two main classes of agonist receptors, G protein-coupled receptors (GPCRs) and immunoreceptor tyrosine-based activation motif (ITAM)-containing receptors, which sense changes in the environment and thereby initiate intracellular signaling required for integrin activation, which in turn is the major mechanism for attachment during thrombus formation (Bergmeier and Stefanini, 2018). Particularly, GPCRs form a “G protein highway to integrin activation”, which is crucial for classical hemostasis, as evidenced by severe bleeding phenotypes after genetic disruption or pharmacological intervention with individual components of this pathway. In contrast, ITAM receptors play a less important role during classical hemostasis but are considered to be more important for thromboinflammation, when platelet aggregation under flow does not happen (d'Alessandro et al., 2020).

The platelet collagen receptor GPVI has attracted attention, as clinical trials targeting this receptor were recently initiated or completed (Voors-Pette et al., 2019). Inhibition of this receptor using Revacept®, a lesion-directed antithrombotic drug binding to exposed collagen of the atherosclerotic plaque, was analyzed in a phase 2 randomized clinical trial with patients undergoing elective percutaneous coronary intervention (PCI) for stable ischemic heart disease (Mayer et al., 2021). As expected, bleeding was not increased and, the authors analyzed a short-term endpoint of a composite of death or myocardial injury, whereby Revacept® had no significant effect.

Another important mechanism, how platelets contribute to ahterosclerosis is their ability to form platelet–neutrophil hetero-aggregates, which are mediated through adhesion molecules such as P-selectin and P-selectin glycoprotein ligand 1 as well as glycoprotein Ib and macrophage-1 antigen (Evangelista et al., 1999; Zarbock et al., 2007; Sreeramkumar et al., 2014b), which then activates intracellular signaling. As a consequence, soluble mediators are released and also direct signaling between platelets and neutrophils is initiated, which leads to their reciprocal activation and neutrophil release of extracellular traps (NETs). These Nets are scaffolds of condensed chromatin, which are exremely prothrombotic and, thus, mediate atherothrombosis (Döring et al., 2017).

Complement—a central part of innate immunity—plays an important role in the progression of atherosclerosis (Haskard et al., 2008). Platelets have been shown to express receptors for complement factors on their surface. For instance, it was demonstrated that the expression of anaphylatoxin-receptors (C3aR and C5aR) and activation markers (i.e., P-selectin) on platelets correlate in CVD patients (Patzelt et al., 2015). It has been shown furthermore, that the complement anaphylatoxin C5a inversely correlates with platelet bound oxLDL (Nording et al., 2016). Complement receptor expression on platelets has recently been shown to play important roles in thrombosis (Sauter et al., 2018) and neovascularization (Nording et al., 2021). Furthermore, C3 can activate platelets independent of formation of the terminal complement complex, which provides evidence for the contribution of complement-dependent membrane perturbations in vivo to prothrombotic TF activation on myeloid cells (Subramaniam et al., 2017). Together, complement factors may present a promising target for therapeutic approaches in the prevention and treatment of atherosclerosis which will have to be addressed in future studies.

Platelets, DCs—Atherosclerosis

The interaction of immune cells and platelets has been suggested to be of importance in atherogenesis and atheroprogression (Figure 2) (Massberg et al., 2002; Nording et al., 2015). DCs are able to recruit platelets via CD11b/CD18 (Mac-1) and platelet JAM-C, which leads to DC activation and platelet phagocytosis—a process that may be important for the progression of atherosclerotic lesions (Langer et al., 2007). On the other hand, platelets are able to activate Mac-1 on leukocytes (Sreeramkumar et al., 2014a). Recently, we discovered a further functional crosstalk of platelets with DCs mediated by GPIbα, which causes activation of beta2 integrins on the immune cells, involving a PSGL1 dependent mechanism (unpublished data). Another study by Verschoor et al. showed, that platelets and CD8α+ DCs are also able to interact via GP1b and complement component C3 (Verschoor et al., 2011). Additionally, platelets induce the secretion of pro-inflammatory cytokines from leukoctyes by providing neutrophil and endothelial activators, and thereby drive the process of acute inflammation (Zarbock et al., 2007).

FIGURE 2
www.frontiersin.org

FIGURE 2. The contribution of DCs and platelets to atherosclerosis.

Platelets shape atherosclerosis by their adhesion to the endothelium, secretion of pro-atherogenic factors such as PF-4, SDF-1, RANTES, CD40L, IL-1β as well as by interaction with other celltypes and leukocyte recruitment which leads to inflammation and thrombus formation. Dendritic cells differentiate into activated DCs in an atherosclerotic environment and accumulate in the growing plaque. There, accumulating evidence suggests that they shape atherosclerosis via parakrine effectors, intracellular signalling and interaction with other celltypes. The net effect of DCs on atherosclerosis still is a matter of debate.

A receptor, which belongs to the tumor necrosis factor family of ligands and is interesting for the topic of this review, as it is expressed on both platelets and T cells is CD40 ligand (CD40L), which was described to be relevant for atheroprogression in a cell specific manner. CD40L is a costimulatory molecule and together with CD40 its corresponding receptor on DCs is responsible for T-cell priming during antigen presentation of infectious agents (Grewal and Flavell, 1996). Not only T cells, but also monocytes, macrophages and endothelial cells express CD40L indicatíng that this protein has a broader function in vivo. The fact that CD40L is upregulated on the surface of activated platelets has attracted attention, as platelet derived CD40L can activate endothelium and therey contribute directly to the inflammatory response at the vessel wall for example, during the development of atherosclerosis (Lievens et al., 2010). For thrombus formation, CD40L was shown to stabilize in a beta3 integrin--dependent manner in vivo and to induce platelet spreading and aggregation (Yacoub et al., 2010). As T-cells and platelets are crucial already for early steps of atherosclerosis, the predominant cell-specific role of CD40L was recently addressed generating platelet- and T cell specific CD40L knockout mice. With this approach it could be demonstrated that platelet CD40L mainly is important for atherothrombosis, wheras T cell CD40L has a major function for the development of atherosclerotic plaques, while platelet CD40L did not affect this aspect (Lacy et al., 2021).

Potential Therapeutic Applications

Lipid-lowering statin therapy is the standard therapy for primary and secondary prevention of cardiovascular diseases. But atherosclerosis is considered to be an inflammatory disease—therefore, anti-inflammatory approaches are promising targets in fighting atheroprogression.

One therapeutic aim is to reduce inflammation without affecting lipid levels, which was supposed to reduce the risk of cardiovascular disease: It has been shown that an anti-inflammatory therapy with canakinumab targeting the interleukin-1β innate immunity pathway could be promising, which led to a significantly lower rate of recurrent cardiovascular events than placebo, independent of lipid-level lowering (Ridker et al., 2017). Another milestone that supports the role of inflammation as a key mediator in the development of cardiovascular disease was a study with colchicine, where in patients with chronic coronary disease the risk of cardiovascular events was significantly lower with colchicine compared to those who received placebo (Nidorf et al., 2020). The discovery of trained immunity mentioned in the beginning of this review may be an attractive target for future therapeutic interventions or even vaccine strategies as it shows common mechanistic pathways with cardiometabolic disease, including epigenetic changes or macrophage activation in response to cytokines (Davis and Gallagher, 2019; Netea et al., 2020; Mitroulis i. et al., 2021). The activation of Tregs by tolerogenic DCs could also provide translational potential, because this interaction inhibits the inflammatory reaction within the lesions. To this end, focus should be put on the expansion of this DC subtype, as vaccination studies already showed promising effects (Hermansson et al., 2011, Nettersheim et al., 2020).

Conclusion

The role of macrophages, which develop into foam cells contributing to plaque build-up is well established. Recent insights highlight the role of previously underappreciated cells in this context including platelets or dendritic cells as well as their interaction for the pathogenesis of atherosclerosis. Cellular, paracrine mechanisms and crosstalk with other important pathophysiological principles such as metabolic dysregulation have been uncovered and further underline the translational potential of and urgent need for characterizing new players and underlying mechanisms in atheroprogression.

Author Contributions

MS and HFL wrote the manuscript.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Ait-oufella h., sage a. P., mallat z., tedgui a. (2014). Adaptive (T and B Cells) Immunity and Control by Dendritic Cells in Atherosclerosis. Circ. Res. 114, 1640–1660. doi:10.1161/circresaha.114.302761

PubMed Abstract | CrossRef Full Text | Google Scholar

Ait-oufella h., taleb s., mallat z., tedgui a. (2011). Recent Advances on the Role of Cytokines in Atherosclerosis. Atvb 31, 969–979. doi:10.1161/atvbaha.110.207415

PubMed Abstract | CrossRef Full Text | Google Scholar

Arnaud c., burger f., steffens s., veillard n. R., nguyen t. H., trono d., et al. (2005). Statins Reduce Interleukin-6-Induced C-Reactive Protein in Human Hepatocytes. Atvb 25, 1231–1236. doi:10.1161/01.atv.0000163840.63685.0c

PubMed Abstract | CrossRef Full Text | Google Scholar

Asgary s., saberi s.-a., azampanah s. (2007). Effect of Immunization against Ox-Ldl with Two Different Antigens on Formation and Development of Atherosclerosis. Lipids Health Dis. 6, 32. doi:10.1186/1476-511x-6-32

PubMed Abstract | CrossRef Full Text | Google Scholar

Banchereau j., steinman r. M. (1998). Dendritic Cells and the Control of Immunity. Nature 392, 245–252. doi:10.1038/32588

PubMed Abstract | CrossRef Full Text | Google Scholar

Bergmeier w., stefanini l. (2018). Platelets at the Vascular Interface. Res. Pract. Thromb. Haemost. 2, 27–33. doi:10.1002/rth2.12061

PubMed Abstract | CrossRef Full Text | Google Scholar

Bobryshev y. V., lord r. S. (1995). S-100 Positive Cells in Human Arterial Intima and in Atherosclerotic Lesions. Cardiovasc. Res. 29, 689–696. doi:10.1016/s0008-6363(96)88642-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Bobryshev y. V. (2006). Monocyte Recruitment and Foam Cell Formation in Atherosclerosis. Micron 37, 208–222. doi:10.1016/j.micron.2005.10.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Bonacina f., coe d., wang g., longhi m. P., baragetti a., moregola a., et al. (2018). Myeloid apolipoprotein e controls dendritic cell antigen presentation and t cell activation. Nat. Commun. 9, 3083. doi:10.1038/s41467-018-05322-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Brinkmann v., reichard u., goosmann c., fauler b., uhlemann y., weiss d. S., et al. (2004). Neutrophil Extracellular Traps Kill Bacteria. Science 303, 1532–1535. doi:10.1126/science.1092385

PubMed Abstract | CrossRef Full Text | Google Scholar

Chistiakov D. A., sobenin I. A., orekhov A. N., bobryshev Y. V. (2014). Dendritic Cells in Atherosclerotic Inflammation: the Complexity of Functions and the Peculiarities of Pathophysiological Effects. Front. Physiol. 5, 196. doi:10.3389/fphys.2014.00196

PubMed Abstract | CrossRef Full Text | Google Scholar

Chistiakov d. A., sobenin i. A., orekhov a. N. (2013). Regulatory T Cells in Atherosclerosis and Strategies to Induce the Endogenous Atheroprotective Immune Response. Immunol. Lett. 151, 10–22. doi:10.1016/j.imlet.2013.01.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Choi j.-h., do y., cheong c., koh h., boscardin s. B., oh y.-s., et al. (2009). Identification of Antigen-Presenting Dendritic Cells in Mouse Aorta and Cardiac Valves. J. Exp. Med. 206, 497–505. doi:10.1084/jem.20082129

PubMed Abstract | CrossRef Full Text | Google Scholar

Chyu k.-y., shah p. K. (2018). In Pursuit of an Atherosclerosis Vaccine. Circ. Res. 123, 1121–1123. doi:10.1161/circresaha.118.313842

PubMed Abstract | CrossRef Full Text | Google Scholar

Collaborators g. C. O. D. (2018). Global, Regional, and National Age-sex-specific Mortality for 282 Causes of Death in 195 Countries and Territories, 1980-2017: a Systematic Analysis for the Global burden of Disease Study 2017. Lancet 392, 1736–1788. doi:10.1016/S0140-6736(18)32203-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Control w. T. F. T. D., interventions c. T. E. O. D., group c. R. (2003). Sustained Effect of Intensive Treatment of Type 1 Diabetes Mellitus on Development and Progression of Diabetic Nephropathy: the Epidemiology of Diabetes Interventions and Complications (EDIC) Study. Jama 290, 2159–2167. doi:10.1001/jama.290.16.2159

PubMed Abstract | CrossRef Full Text | Google Scholar

Croce K., libby P. (2007). Intertwining of Thrombosis and Inflammation in Atherosclerosis. Curr. Opin. Hematol. 14, 55–61. doi:10.1097/00062752-200701000-00011

PubMed Abstract | CrossRef Full Text | Google Scholar

D'alessandro E., becker C., bergmeier W., bode C., bourne J. H., brown H., et al. (2020). Thrombo-inflammation in Cardiovascular Disease: an Expert Consensus Document from the Third Maastricht Consensus Conference on Thrombosis. Thromb. Haemost. 120, 538–564. doi:10.1055/s-0040-1708035

PubMed Abstract | CrossRef Full Text | Google Scholar

Davis f. M., gallagher k. A. (2019). Epigenetic Mechanisms in Monocytes/macrophages Regulate Inflammation in Cardiometabolic and Vascular Disease. Atvb 39, 623–634. doi:10.1161/atvbaha.118.312135

PubMed Abstract | CrossRef Full Text | Google Scholar

Döring y., libby p., soehnlein o. (2020). Neutrophil Extracellular Traps Participate in Cardiovascular Diseases. Circ. Res. 126, 1228–1241. doi:10.1161/circresaha.120.315931

PubMed Abstract | CrossRef Full Text | Google Scholar

Döring y., manthey h. D., drechsler m., lievens d., megens r. T. A., soehnlein o., et al. (2012). Auto-antigenic Protein-Dna Complexes Stimulate Plasmacytoid Dendritic Cells to Promote Atherosclerosis. Circulation 125, 1673–1683. doi:10.1161/circulationaha.111.046755

PubMed Abstract | CrossRef Full Text | Google Scholar

Döring y., soehnlein o., weber c. (2017). Neutrophil Extracellular Traps in Atherosclerosis and Atherothrombosis. Circ. Res. 120, 736–743. doi:10.1161/circresaha.116.309692

PubMed Abstract | CrossRef Full Text | Google Scholar

Drechsler m., megens R. T. A., van zandvoort m., weber c., soehnlein o. (2010). Hyperlipidemia-triggered Neutrophilia Promotes Early Atherosclerosis. Circulation 122, 1837–1845. doi:10.1161/circulationaha.110.961714

PubMed Abstract | CrossRef Full Text | Google Scholar

Du f., zhou j., gong r., huang x., pansuria m., virtue a., et al. (2012). Endothelial Progenitor Cells in Atherosclerosis. Front. Biosci. 17, 2327–2349. doi:10.2741/4055

CrossRef Full Text | Google Scholar

El-osta a., brasacchio d., yao d., pocai a., jones p. L., roeder r. G., et al. (2008). Transient High Glucose Causes Persistent Epigenetic Changes and Altered Gene Expression during Subsequent Normoglycemia. J. Exp. Med. 205, 2409–2417. doi:10.1084/jem.20081188

PubMed Abstract | CrossRef Full Text | Google Scholar

Evangelista v., manarini s., sideri r., rotondo s., martelli n., piccoli a., et al. (1999). Platelet/polymorphonuclear Leukocyte Interaction: P-Selectin Triggers Protein-Tyrosine Phosphorylation-dependent Cd11b/cd18 Adhesion: Role of Psgl-1 as a Signaling Molecule. Blood 93, 876–885. doi:10.1182/blood.v93.3.876

PubMed Abstract | CrossRef Full Text | Google Scholar

Feil s., fehrenbacher b., lukowski r., essmann f., schulze-osthoff k., schaller m., et al. (2014). Transdifferentiation of Vascular Smooth Muscle Cells to Macrophage-like Cells during Atherogenesis. Circ. Res. 115, 662–667. doi:10.1161/circresaha.115.304634

PubMed Abstract | CrossRef Full Text | Google Scholar

Frostegård j., ulfgren A.-K., nyberg p., hedin u., swedenborg j., andersson u., et al. (1999). Cytokine Expression in Advanced Human Atherosclerotic Plaques: Dominance of Pro-inflammatory (Th1) and Macrophage-Stimulating Cytokines. Atherosclerosis 145, 33–43. doi:10.1016/s0021-9150(99)00011-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Fujii s.-i., liu k., smith c., bonito a. J., steinman r. M. (2004). The Linkage of Innate to Adaptive Immunity via Maturing Dendritic Cells In Vivo Requires Cd40 Ligation in Addition to Antigen Presentation and Cd80/86 Costimulation. J. Exp. Med. 199, 1607–1618. doi:10.1084/jem.20040317

PubMed Abstract | CrossRef Full Text | Google Scholar

Garcia-romo g. S., caielli s., vega b., connolly j., allantaz f., xu z., et al. (2011). Netting Neutrophils Are Major Inducers of Type I Ifn Production in Pediatric Systemic Lupus Erythematosus. Sci. Transl. Med. 3, 3001201. doi:10.1126/scitranslmed.3001201

PubMed Abstract | CrossRef Full Text | Google Scholar

Gautier e. L., huby t., saint-charles f., ouzilleau b., pirault j., deswaerte v., et al. (2009). Conventional Dendritic Cells at the Crossroads between Immunity and Cholesterol Homeostasis in Atherosclerosis. Circulation 119, 2367–2375. doi:10.1161/circulationaha.108.807537

PubMed Abstract | CrossRef Full Text | Google Scholar

Gawaz m., langer h., may a. E. (2005). Platelets in Inflammation and Atherogenesis. J. Clin. Invest. 115, 3378–3384. doi:10.1172/jci27196

PubMed Abstract | CrossRef Full Text | Google Scholar

Gencer s., evans b. R., van der vorst e. P. C., Döring y., weber c. (2021). Inflammatory Chemokines in Atherosclerosis. Cells 10, 226. doi:10.3390/cells10020226

PubMed Abstract | CrossRef Full Text | Google Scholar

George j., afek a., gilburd b., levkovitz h., shaish a., goldberg i., et al. (1998). Hyperimmunization of Apo-E-Deficient Mice with Homologous Malondialdehyde Low-Density Lipoprotein Suppresses Early Atherogenesis. Atherosclerosis 138, 147–152. doi:10.1016/s0021-9150(98)00015-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Gerner m. Y., kastenmuller w., ifrim i., kabat j., germain r. N. (2012). Histo-cytometry: a Method for Highly Multiplex Quantitative Tissue Imaging Analysis Applied to Dendritic Cell Subset Microanatomy in Lymph Nodes. Immunity 37, 364–376. doi:10.1016/j.immuni.2012.07.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Glagov s., weisenberg e., zarins c. K., stankunavicius r., kolettis g. J. (1987). Compensatory Enlargement of Human Atherosclerotic Coronary Arteries. N. Engl. J Med. 316, 1371–1375. doi:10.1056/nejm198705283162204

PubMed Abstract | CrossRef Full Text | Google Scholar

Grewal i. S., flavell r. A. (1996). The Role of Cd40 Ligand in Costimulation and T-Cell Activation. Immunol. Rev. 153, 85–106. doi:10.1111/j.1600-065x.1996.tb00921.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Haskard d. O., boyle j. J., mason j. C. (2008). The Role of Complement in Atherosclerosis. Curr. Opin. Lipidol. 19, 478–482. doi:10.1097/mol.0b013e32830f4a06

PubMed Abstract | CrossRef Full Text | Google Scholar

Hermansson a., johansson d. K., ketelhuth D. F. J., andersson j., zhou x., hansson g. K. (2011). Immunotherapy with Tolerogenic Apolipoprotein B-100-Loaded Dendritic Cells Attenuates Atherosclerosis in Hypercholesterolemic Mice. Circulation 123, 1083–1091. doi:10.1161/circulationaha.110.973222

PubMed Abstract | CrossRef Full Text | Google Scholar

Holman r. R., paul s. K., bethel m. A., matthews d. R., neil h. A. W. (2008). 10-year Follow-Up of Intensive Glucose Control in Type 2 Diabetes. N. Engl. J. Med. 359, 1577–1589. doi:10.1056/nejmoa0806470

PubMed Abstract | CrossRef Full Text | Google Scholar

Huo y., schober a., forlow s. B., smith d. F., hyman m. C., jung s., et al. (2003). Circulating activated platelets exacerbate atherosclerosis in mice deficient in apolipoprotein e. Nat. Med. 9, 61–67. doi:10.1038/nm810

PubMed Abstract | CrossRef Full Text | Google Scholar

Iellem a., mariani m., lang r., recalde h., panina-bordignon p., sinigaglia f., et al. (2001). Unique Chemotactic Response Profile and Specific Expression of Chemokine Receptors Ccr4 and Ccr8 by Cd4+cd25+ Regulatory T Cells. J. Exp. Med. 194, 847–854. doi:10.1084/jem.194.6.847

PubMed Abstract | CrossRef Full Text | Google Scholar

Jenne c. N., urrutia r., kubes p. (2013). Platelets: Bridging Hemostasis, Inflammation, and Immunity. Int. Jnl. Lab. Hem. 35, 254–261. doi:10.1111/ijlh.12084

PubMed Abstract | CrossRef Full Text | Google Scholar

Jongstra-bilen j., haidari m., zhu S.-N., chen m., guha d., cybulsky m. I. (2006). Low-grade Chronic Inflammation in Regions of the normal Mouse Arterial Intima Predisposed to Atherosclerosis. J. Exp. Med. 203, 2073–2083. doi:10.1084/jem.20060245

PubMed Abstract | CrossRef Full Text | Google Scholar

Jung s., unutmaz d., wong p., sano G.-I., de los santos k., sparwasser t., et al. (2002). In Vivo depletion of Cd11c+ Dendritic Cells Abrogates Priming of Cd8+ T Cells by Exogenous Cell-Associated Antigens. Immunity 17, 211–220. doi:10.1016/s1074-7613(02)00365-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Kannel w. B., mcgee d. L. (1979). Diabetes and Cardiovascular Disease. Jama 241, 2035–2038. doi:10.1001/jama.1979.03290450033020

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim s. Y., choi y. J., joung s. M., lee b. H., jung Y.-S., lee j. Y. (2010). Hypoxic Stress Up-Regulates the Expression of Toll-like Receptor 4 in Macrophages via Hypoxia-Inducible Factor. Immunology 129, 516–524. doi:10.1111/j.1365-2567.2009.03203.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Kofler s., schlichting c., jankl s., nickel t., weis m. (2008). Dual Mode of Hmg-Coa Reductase Inhibition on Dendritic Cell Invasion. Atherosclerosis 197, 105–110. doi:10.1016/j.atherosclerosis.2007.08.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Koupenova m., clancy l., corkrey h. A., freedman j. E. (2018). Circulating Platelets as Mediators of Immunity, Inflammation, and Thrombosis. Circ. Res. 122, 337–351. doi:10.1161/circresaha.117.310795

PubMed Abstract | CrossRef Full Text | Google Scholar

Kretzschmar d., betge s., windisch a., pistulli r., rohm i., fritzenwanger m., et al. (2012). Recruitment of Circulating Dendritic Cell Precursors into the Infarcted Myocardium and Pro-inflammatory Response in Acute Myocardial Infarction. Clin. Sci. 123, 387–398. doi:10.1042/cs20110561

CrossRef Full Text | Google Scholar

Kurien b. T., scofield r. H. (2008). Autoimmunity and Oxidatively Modified Autoantigens. Autoimmun. Rev. 7, 567–573. doi:10.1016/j.autrev.2008.04.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Lacy m., Bürger c., shami a., ahmadsei m., winkels h., nitz k., et al. (2021). Cell-specific and Divergent Roles of the Cd40l-Cd40 axis in Atherosclerotic Vascular Disease. Nat. Commun. 12, 3754. doi:10.1038/s41467-021-23909-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Lande r., ganguly d., facchinetti v., frasca l., conrad c., gregorio j., et al. (2011). Neutrophils Activate Plasmacytoid Dendritic Cells by Releasing Self-Dna-Peptide Complexes in Systemic Lupus Erythematosus. Sci. Transl. Med. 3, 3001180. doi:10.1126/scitranslmed.3001180

PubMed Abstract | CrossRef Full Text | Google Scholar

Langer h. F. (2022). Chronic Inflammation in Atherosclerosis-The Cd40l/cd40 axis Belongs to Dendritic Cells and T Cells, Not Platelets. J. Thromb. Haemost 20, 3–5. doi:10.1111/jth.15591

PubMed Abstract | CrossRef Full Text | Google Scholar

Langer h. F., daub k., braun g., Schönberger t., may a. E., schaller m., et al. (2007). Platelets Recruit Human Dendritic Cells via Mac-1/jam-C Interaction and Modulate Dendritic Cell Function In Vitro. Atvb 27, 1463–1470. doi:10.1161/atvbaha.107.141515

CrossRef Full Text | Google Scholar

Laridan E., martinod K., de meyer S. F. (2019). Neutrophil Extracellular Traps in Arterial and Venous Thrombosis. Semin. Thromb. Hemost. 45, 86–93. doi:10.1055/s-0038-1677040

PubMed Abstract | CrossRef Full Text | Google Scholar

Leclerc v., reichhart J.-M. (2004). The Immune Response of drosophila Melanogaster. Immunol. Rev. 198, 59–71. doi:10.1111/j.0105-2896.2004.0130.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Leentjens j., bekkering s., joosten l. A. B., netea m. G., burgner d. P., riksen n. P. (2018). Trained Innate Immunity as a Novel Mechanism Linking Infection and the Development of Atherosclerosis. Circ. Res. 122, 664–669. doi:10.1161/circresaha.117.312465

PubMed Abstract | CrossRef Full Text | Google Scholar

Libby p., buring j. E., badimon l., hansson g. K., deanfield j., bittencourt m. S., et al. (2019). Atherosclerosis. Nat. Rev. Dis. Primers 5, 56. doi:10.1038/s41572-019-0106-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Libby p. (2002). Inflammation in Atherosclerosis. Nature 420, 868–874. doi:10.1038/nature01323

PubMed Abstract | CrossRef Full Text | Google Scholar

Lievens d., zernecke a., seijkens t., soehnlein o., beckers l., munnix i. C. A., et al. (2010). Platelet Cd40l Mediates Thrombotic and Inflammatory Processes in Atherosclerosis. Blood 116, 4317–4327. doi:10.1182/blood-2010-01-261206

PubMed Abstract | CrossRef Full Text | Google Scholar

Lord R. S., bobryshev Y. V. (1999). Clustering of Dendritic Cells in Athero-Prone Areas of the Aorta. Atherosclerosis 146, 197–198. doi:10.1016/s0021-9150(99)00119-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Lövgren t., eloranta m.-l., Båve u., alm g. V., Rönnblom l. (2004). Induction of Interferon-α Production in Plasmacytoid Dendritic Cells by Immune Complexes Containing Nucleic Acid Released by Necrotic or Late Apoptotic Cells and Lupus IgG. Arthritis Rheum. 50, 1861–1872. doi:10.1002/art.20254

PubMed Abstract | CrossRef Full Text | Google Scholar

Lumeng c. N., delproposto j. B., westcott d. J., saltiel a. R. (2008). Phenotypic Switching of Adipose Tissue Macrophages with Obesity Is Generated by Spatiotemporal Differences in Macrophage Subtypes. Diabetes 57, 3239–3246. doi:10.2337/db08-0872

PubMed Abstract | CrossRef Full Text | Google Scholar

Lutgens e., lievens d., beckers l., wijnands e., soehnlein o., zernecke a., et al. (2010). Deficient Cd40-Traf6 Signaling in Leukocytes Prevents Atherosclerosis by Skewing the Immune Response toward an Antiinflammatory Profile. J. Exp. Med. 207, 391–404. doi:10.1084/jem.20091293

PubMed Abstract | CrossRef Full Text | Google Scholar

Mangold a., alias s., scherz t., hofbauer T. M., jakowitsch j., Panzenböck a., et al. (2015). Coronary Neutrophil Extracellular Trap burden and Deoxyribonuclease Activity in St-Elevation Acute Coronary Syndrome Are Predictors of St-Segment Resolution and Infarct Size. Circ. Res. 116, 1182–1192. doi:10.1161/circresaha.116.304944

PubMed Abstract | CrossRef Full Text | Google Scholar

Massberg s., brand k., Grüner s., page s., Müller e., Müller i., et al. (2002). A Critical Role of Platelet Adhesion in the Initiation of Atherosclerotic Lesion Formation. J. Exp. Med. 196, 887–896. doi:10.1084/jem.20012044

PubMed Abstract | CrossRef Full Text | Google Scholar

Mathieu p., pibarot p., després j.-p. (2006). Metabolic Syndrome: the Danger Signal in Atherosclerosis. Vasc. Health Risk Manag. 2, 285–302. doi:10.2147/vhrm.2006.2.3.285

PubMed Abstract | CrossRef Full Text | Google Scholar

Maugeri n., campana l., gavina m., covino c., de metrio m., panciroli c., et al. (2014). Activated Platelets Present High Mobility Group Box 1 to Neutrophils, Inducing Autophagy and Promoting the Extrusion of Neutrophil Extracellular Traps. J. Thromb. Haemost. 12, 2074–2088. doi:10.1111/jth.12710

PubMed Abstract | CrossRef Full Text | Google Scholar

Mayer k., hein-rothweiler r., Schüpke s., janisch m., bernlochner i., ndrepepa g., et al. (2021). Efficacy and Safety of Revacept, a Novel Lesion-Directed Competitive Antagonist to Platelet Glycoprotein VI, in Patients Undergoing Elective Percutaneous Coronary Intervention for Stable Ischemic Heart Disease. JAMA Cardiol. 6, 753–761. doi:10.1001/jamacardio.2021.0475

PubMed Abstract | CrossRef Full Text | Google Scholar

Means t. K., latz e., hayashi f., murali m. R., golenbock d. T., luster a. D. (2005). Human Lupus Autoantibody-DNA Complexes Activate DCs through Cooperation of CD32 and TLR9. J. Clin. Invest. 115, 407–417. doi:10.1172/jci23025

PubMed Abstract | CrossRef Full Text | Google Scholar

Megens R. T. A., vijayan s., lievens d., Döring y., van zandvoort M. A. M. J., grommes j., et al. (2012). Presence of Luminal Neutrophil Extracellular Traps in Atherosclerosis. Thromb. Haemost. 107 (3), 597–598. doi:10.1160/th11-09-0650

PubMed Abstract | CrossRef Full Text | Google Scholar

Miller j. C., brown b. D., Brown B. D., Shay T., Gautier E. L., Jojic V., et al. (2012). Deciphering the Transcriptional Network of the Dendritic Cell Lineage. Nat. Immunol. 13, 888–899. doi:10.1038/ni.2370

PubMed Abstract | CrossRef Full Text | Google Scholar

Mitroulis I., hajishengallis G., chavakis T. (2021b). Trained Immunity and Cardiometabolic Disease: the Role of Bone Marrow. Arterioscler Thromb. Vasc. Biol. 41, 48–54. doi:10.1161/ATVBAHA.120.314215

CrossRef Full Text | Google Scholar

Mitroulis i., hajishengallis g., chavakis t. (2021a). Trained Immunity and Cardiometabolic Disease: the Role of Bone Marrow. Arteriosclerosis, Thromb. Vasc. Biol. 41, 48–54.

Google Scholar

Murray p. J., allen j. E., biswas s. K., fisher e. A., gilroy d. W., goerdt s., et al. (2014). Macrophage Activation and Polarization: Nomenclature and Experimental Guidelines. Immunity 41, 14–20. doi:10.1016/j.immuni.2014.06.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Murray p. J., wynn t. A. (2011). Protective and Pathogenic Functions of Macrophage Subsets. Nat. Rev. Immunol. 11, 723–737. doi:10.1038/nri3073

PubMed Abstract | CrossRef Full Text | Google Scholar

Netea m. G., Domínguez-Andrés j., barreiro l. B., chavakis t., divangahi m., fuchs e., et al. (2020). Defining Trained Immunity and its Role in Health and Disease. Nat. Rev. Immunol. 20, 375–388. doi:10.1038/s41577-020-0285-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Ni k., o'neill H. (1997). The Role of Dendritic Cells in T Cell Activation. Immunol. Cell Biol. 75, 223–230. doi:10.1038/icb.1997.35

PubMed Abstract | CrossRef Full Text | Google Scholar

Nidorf s. M., fiolet a. T. L., mosterd a., eikelboom j. W., schut a., opstal t. S. J., et al. (2020). Colchicine in Patients with Chronic Coronary Disease. N. Engl. J. Med. 383, 1838–1847. doi:10.1056/nejmoa2021372

PubMed Abstract | CrossRef Full Text | Google Scholar

Nording h., baron l., haberthür d., emschermann f., mezger m., sauter m., et al. (2021). The C5a/c5a Receptor 1 axis Controls Tissue Neovascularization through Cxcl4 Release from Platelets. Nat. Commun. 12, 021–23499. doi:10.1038/s41467-021-23499-w

CrossRef Full Text | Google Scholar

Nording h., baron l., langer h. F. (2020). Platelets as Therapeutic Targets to Prevent Atherosclerosis. Atherosclerosis 307, 97–108. doi:10.1016/j.atherosclerosis.2020.05.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Nording h., giesser a., patzelt j., sauter r., emschermann f., stellos k., et al. (2016). Platelet Bound Oxldl Shows an Inverse Correlation with Plasma Anaphylatoxin C5a in Patients with Coronary Artery Disease. Platelets 27, 593–597. doi:10.3109/09537104.2016.1148807

PubMed Abstract | CrossRef Full Text | Google Scholar

Nording H. M., seizer P., langer H. F. (2015). Platelets in Inflammation and Atherogenesis. Front. Immunol. 6, 98. doi:10.3389/fimmu.2015.00098

PubMed Abstract | CrossRef Full Text | Google Scholar

Patzelt j., mueller K. A. L., breuning s., karathanos a., schleicher r., seizer p., et al. (2015). Expression of Anaphylatoxin Receptors on Platelets in Patients with Coronary Heart Disease. Atherosclerosis 238, 289–295. doi:10.1016/j.atherosclerosis.2014.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Paulson k. E., zhu S.-N., chen m., nurmohamed s., jongstra-bilen j., cybulsky m. I. (2010). Resident Intimal Dendritic Cells Accumulate Lipid and Contribute to the Initiation of Atherosclerosis. Circ. Res. 106, 383–390. doi:10.1161/circresaha.109.210781

PubMed Abstract | CrossRef Full Text | Google Scholar

Penkov s., mitroulis i., hajishengallis g., chavakis t. (2019). Immunometabolic Crosstalk: an Ancestral Principle of Trained Immunity? Trends Immunol. 40, 1–11. doi:10.1016/j.it.2018.11.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Probst h. C., tschannen k., odermatt b., schwendener r., zinkernagel r. M., van den broek m. (2005). Histological Analysis of Cd11c-Dtr/gfp Mice after In Vivo Depletion of Dendritic Cells. Clin. Exp. Immunol. 141, 398–404. doi:10.1111/j.1365-2249.2005.02868.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Qi h., yang s., zhang l. (2017). Neutrophil Extracellular Traps and Endothelial Dysfunction in Atherosclerosis and Thrombosis. Front. Immunol. 8, 928. doi:10.3389/fimmu.2017.00928

PubMed Abstract | CrossRef Full Text | Google Scholar

Rahman M., steuer J., gillgren P., hayderi A., liu A., Frostegård J. (2017). Induction of Dendritic Cell-Mediated Activation of T Cells from Atherosclerotic Plaques by Human Heat Shock Protein 60. J. Am. Heart Assoc. 6, e006778. doi:10.1161/JAHA.117.006778

PubMed Abstract | CrossRef Full Text | Google Scholar

Randolph G. J. (2014). Mechanisms that Regulate Macrophage burden in Atherosclerosis. Circ. Res. 114, 1757–1771. doi:10.1161/circresaha.114.301174

PubMed Abstract | CrossRef Full Text | Google Scholar

Ranjit s., dazhu l., qiutang z., yibo f., yushu l., xiang w., et al. (2004). Differentiation of Dendritic Cells in Monocyte Cultures Isolated from Patients with Unstable Angina. Int. J. Cardiol. 97, 551–555. doi:10.1016/j.ijcard.2004.05.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Ridker p. M., everett b. M., thuren t., macfadyen j. G., chang w. H., ballantyne c., et al. (2017). Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N. Engl. J. Med. 377, 1119–1131. doi:10.1056/nejmoa1707914

PubMed Abstract | CrossRef Full Text | Google Scholar

Rong j. X., shapiro m., trogan e., fisher e. A. (2003). Transdifferentiation of Mouse Aortic Smooth Muscle Cells to a Macrophage-like State after Cholesterol Loading. Proc. Natl. Acad. Sci. U.S.A. 100, 13531–13536. doi:10.1073/pnas.1735526100

PubMed Abstract | CrossRef Full Text | Google Scholar

Roy p., ali a. J., kobiyama k., ghosheh y., ley k. (2020). Opportunities for an Atherosclerosis Vaccine: from Mice to Humans. Vaccine 38, 4495–4506. doi:10.1016/j.vaccine.2019.12.039

PubMed Abstract | CrossRef Full Text | Google Scholar

Roy p., orecchioni m., ley k. (2021). How the Immune System Shapes Atherosclerosis: Roles of Innate and Adaptive Immunity. Nat. Rev. Immunol. doi:10.1038/s41577-021-00584-1

CrossRef Full Text | Google Scholar

Saigusa r., winkels h., ley k. (2020). T Cell Subsets and Functions in Atherosclerosis. Nat. Rev. Cardiol. 17, 387–401. doi:10.1038/s41569-020-0352-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Sauter m., sauter r. J., nording h., lin c., olbrich m., autenrieth s., et al. (2021). Apolipoprotein e derived from cd11c(+) cells ameliorates atherosclerosis. Iscience 25, 21.

Google Scholar

Sauter r. J., sauter m., reis e. S., emschermann f. N., nording h., Ebenhöch s., et al. (2018). Functional Relevance of the Anaphylatoxin Receptor C3ar for Platelet Function and Arterial Thrombus Formation marks an Intersection point between Innate Immunity and Thrombosis. Circulation 138, 1720–1735. doi:10.1161/circulationaha.118.034600

PubMed Abstract | CrossRef Full Text | Google Scholar

Schleicher r. I., reichenbach f., kraft p., kumar a., lescan m., todt f., et al. (2015). Platelets Induce Apoptosis via Membrane-Bound Fasl. Blood 126, 1483–1493. doi:10.1182/blood-2013-12-544445

PubMed Abstract | CrossRef Full Text | Google Scholar

Schraml b. U., reis e sousa c. (2015). Defining Dendritic Cells. Curr. Opin. Immunol. 32, 13–20. doi:10.1016/j.coi.2014.11.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Shankman l. S., gomez d., cherepanova o. A., salmon m., alencar g. F., haskins r. M., et al. (2015). Klf4-dependent Phenotypic Modulation of Smooth Muscle Cells Has a Key Role in Atherosclerotic Plaque Pathogenesis. Nat. Med. 21, 628–637. doi:10.1038/nm.3866

PubMed Abstract | CrossRef Full Text | Google Scholar

Sreeramkumar v., adrover j. M., ballesteros i., cuartero m. I., rossaint j., bilbao i., et al. (2014a). Neutrophils Scan for Activated Platelets to Initiate Inflammation. Science 346, 1234–1238. doi:10.1126/science.1256478

PubMed Abstract | CrossRef Full Text | Google Scholar

Sreeramkumar v., adrover j. M., ballesteros i., cuartero m. I., rossaint j., bilbao i., et al. (2014b). Neutrophils Scan for Activated Platelets to Initiate Inflammation. Science 346, 1234–1238. doi:10.1126/science.1256478

PubMed Abstract | CrossRef Full Text | Google Scholar

Steinman r. M., banchereau j. (2007). Taking Dendritic Cells into Medicine. Nature 449, 419–426. doi:10.1038/nature06175

PubMed Abstract | CrossRef Full Text | Google Scholar

Subramaniam s., jurk k., hobohm l., Jäckel s., saffarzadeh m., schwierczek k., et al. (2017). Distinct Contributions of Complement Factors to Platelet Activation and Fibrin Formation in Venous Thrombus Development. Blood 129, 2291–2302. doi:10.1182/blood-2016-11-749879

PubMed Abstract | CrossRef Full Text | Google Scholar

Subramanian m., tabas i. (2014). Dendritic Cells in Atherosclerosis. Semin. Immunopathol 36, 93–102. doi:10.1007/s00281-013-0400-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Subramanian m., thorp e., hansson g. K., tabas i. (2013). Treg-mediated Suppression of Atherosclerosis Requires Myd88 Signaling in Dcs. J. Clin. Invest. 123, 179–188. doi:10.1172/jci64617

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun y., rawish e., nording h. M., langer h. F. (2021a). Inflammation in Metabolic and Cardiovascular Disorders-Role of Oxidative Stress. Life 11, 672. doi:10.3390/life11070672

CrossRef Full Text | Google Scholar

Sun y., zhou l., chen w., zhang l., zeng h., Sun Y., et al. (2021b). Immune Metabolism: a Bridge of Dendritic Cells Function. Int. Rev. Immunol. 1, 1–13. doi:10.1080/08830185.2021.1897124

CrossRef Full Text | Google Scholar

Van gisbergen K. P. J. M., sanchez-hernandez m., geijtenbeek T. B. H., van kooyk y. (2005). Neutrophils Mediate Immune Modulation of Dendritic Cells through Glycosylation-dependent Interactions between Mac-1 and Dc-Sign. J. Exp. Med. 201, 1281–1292. doi:10.1084/jem.20041276

PubMed Abstract | CrossRef Full Text | Google Scholar

Vengrenyuk y., nishi h., long x., ouimet m., savji n., martinez f. O., et al. (2015). Cholesterol Loading Reprograms the Microrna-143/145-Myocardin axis to Convert Aortic Smooth Muscle Cells to a Dysfunctional Macrophage-like Phenotype. Atvb 35, 535–546. doi:10.1161/atvbaha.114.304029

PubMed Abstract | CrossRef Full Text | Google Scholar

Verschoor a., neuenhahn m., navarini a. A., graef p., plaumann a., seidlmeier a., et al. (2011). A Platelet-Mediated System for Shuttling Blood-Borne Bacteria to CD8α+ Dendritic Cells Depends on Glycoprotein GPIb and Complement C3. Nat. Immunol. 12, 1194–1201. doi:10.1038/ni.2140

PubMed Abstract | CrossRef Full Text | Google Scholar

Voors-pette c., lebozec k., dogterom p., jullien l., billiald p., ferlan p., et al. (2019). Safety and Tolerability, Pharmacokinetics, and Pharmacodynamics of Act017, an Antiplatelet Gpvi (Glycoprotein Vi) Fab. Atvb 39, 956–964. doi:10.1161/atvbaha.118.312314

PubMed Abstract | CrossRef Full Text | Google Scholar

Weber c., meiler s., Döring y., koch m., drechsler m., megens r. T. A., et al. (2011). Ccl17-expressing Dendritic Cells Drive Atherosclerosis by Restraining Regulatory T Cell Homeostasis in Mice. J. Clin. Invest. 121, 2898–2910. doi:10.1172/jci44925

PubMed Abstract | CrossRef Full Text | Google Scholar

WHO CVD Risk Chart Working Group (2019). World Health Organization Cardiovascular Disease Risk Charts: Revised Models to Estimate Risk in 21 Global Regions. The Lancet. Global Health 7 (10), e1332–e1345. doi:10.1016/S2214-109X(19)30318-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Wolf d., ley k. (2019a). Immunity and Inflammation in Atherosclerosis. Circ. Res. 124, 315–327. doi:10.1161/circresaha.118.313591

PubMed Abstract | CrossRef Full Text | Google Scholar

Wolf d., ley k. (2019b). Immunity and Inflammation in Atherosclerosis. Circ. Res. 124, 315–327. doi:10.1161/circresaha.118.313591

PubMed Abstract | CrossRef Full Text | Google Scholar

Yacoub d., hachem a., Théorêt j.-f., gillis m.-a., mourad w., merhi y. (2010). Enhanced Levels of Soluble CD40 Ligand Exacerbate Platelet Aggregation and Thrombus Formation through a CD40-dependent Tumor Necrosis Factor Receptor-Associated Factor-2/Rac1/p38 Mitogen-Activated Protein Kinase Signaling Pathway. Atvb 30, 2424–2433. doi:10.1161/atvbaha.110.216143

CrossRef Full Text | Google Scholar

Yilmaz a., lochno m., traeg f., cicha i., reiss c., stumpf c., et al. (2004). Emergence of Dendritic Cells in Rupture-Prone Regions of Vulnerable Carotid Plaques. Atherosclerosis 176, 101–110. doi:10.1016/j.atherosclerosis.2004.04.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Yilmaz a., reiss c., weng a., cicha i., stumpf c., steinkasserer a., et al. (2006a). Differential Effects of Statins on Relevant Functions of Human Monocyte-Derived Dendritic Cells. J. Leukoc. Biol. 79, 529–538. doi:10.1189/jlb.0205064

PubMed Abstract | CrossRef Full Text | Google Scholar

Yilmaz a., weber j., cicha i., stumpf c., klein m., raithel d., et al. (2006b). Decrease in Circulating Myeloid Dendritic Cell Precursors in Coronary Artery Disease. J. Am. Coll. Cardiol. 48, 70–80. doi:10.1016/j.jacc.2006.01.078

CrossRef Full Text | Google Scholar

Zarbock a., polanowska-grabowska r. K., ley k. (2007). Platelet-neutrophil-interactions: Linking Hemostasis and Inflammation. Blood Rev. 21, 99–111. doi:10.1016/j.blre.2006.06.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao y., zhang j., zhang w., xu y. (2021). A Myriad of Roles of Dendritic Cells in Atherosclerosis. Clin. Exp. Immunol. 206, 12–27. doi:10.1111/cei.13634

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: atherosclerosis, inflammation, immune system, dendritic cell, platelets, thrombosis

Citation: Sauter M and Langer HF (2022) Targeting Cell-Specific Molecular Mechanisms of Innate Immunity in Atherosclerosis. Front. Physiol. 13:802990. doi: 10.3389/fphys.2022.802990

Received: 27 October 2021; Accepted: 07 March 2022;
Published: 01 April 2022.

Edited by:

Jan Larmann, Heidelberg University Hospital, Germany

Reviewed by:

Paul “Li-Hao” Huang, Fudan University, China
Alexander E. Berezin, Zaporizhia State Medical University, Ukraine
Cristina M. Sena, University of Coimbra, Portugal
Carmine Savoia, Sapienza University of Rome, Italy

Copyright © 2022 Sauter and Langer. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: H. F. Langer, harald.langer@uksh.de

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.