Open access

Dendritic Cells in Atherogenesis: From Immune Shapers to Therapeutic Targets

Written By

Ilse Van Brussel, Hidde Bult, Wim Martinet, Guido R.Y. De Meyer and Dorien M. Schrijvers

Submitted: 24 August 2012 Published: 27 February 2013

DOI: 10.5772/52900

From the Edited Volume

Current Trends in Atherogenesis

Edited by Rita Rezzani

Chapter metrics overview

2,280 Chapter Downloads

View Full Metrics

1. Introduction

Atherosclerosis has been formerly considered as a lipid-mediated disease. It has long been assumed that atherogenesis could be simply explained by lipid accumulation in the vessel wall leading to endothelial dysfunction with adverse vascular wall remodelling. However, over the last decade, a number of studies have clearly demonstrated that lipids are not the whole story in the pathogenesis of atherosclerosis. Accumulating evidence has shown that inflammation and the immune system play a major role in the initiation, progression and destabilization of atheromata [1,2,3,4]. Mainly innate immunity pathways have long been believed to contribute to atherogenesis, and special attention has been given to macrophages, because these effector cells are important for intracellular lipid accumulation and foam cell formation [5]. Yet, although macrophages constitute the largest cell population, other immune cell subsets, namely dendritic cells (DCs) and T cells, can also be found within atherosclerotic plaques and seem to participate in immune responses during atherogenesis.

DCs are the pacemakers of the immune system. These professional antigen-presenting cells play a key role in inducing adaptive immune responses on the one hand, and are critically involved in promoting and maintaining immune tolerance on the other [6]. They originate from hematopoietic stem cells in the bone marrow and circulate as precursors in the blood stream, taking residence in target tissues at sites of potential antigen entry. Within blood vessels [7] and other tissues, they give rise to immature interstitial DCs that act as sentinels, which continuously and efficiently sample the antigenic content of their microenvironment. In the steady state, immature DCs capture harmless self-antigens in the absence of inflammatory signals. They might enter the regional lymph nodes to present the self-antigen to naïve or resting T cells, which will be deleted by apoptosis, silenced by the induction of anergy or primed to become regulatory T cells [8]. In contrast, when infection and tissue damage occur, immature DCs take up antigens in the presence of inflammatory signals, which evokes activation and functional transformation into mature DCs. Meanwhile, they exit the non-lymphoid tissues to migrate via afferent lymph vessels to lymphoid tissues, where they completely mature. Mature DCs present short peptide fragments, which are bound to the surface molecules CD1 or major histocompatibility complex (MHC)-I or MHC-II. Consequently, they activate (naïve) T and B lymphocytes that recognize the presented antigen [9]. Morphological changes occur as well during the DC life cycle: DC precursors are often small, round-shaped cells that turn into larger cells with an irregular (star-like) shape and cytoplasmic protrusions (dendrites) as the cell matures, while migrating DCs are also called veiled cells, as they possess large cytoplasmic 'veils' rather than dendrites [10].

Following the first observation of DCs in human arteries in 1995 [11], numerous studies suggest that these cells presumably play a crucial role in directing innate or adaptive immunity against altered self-antigens present in atherosclerosis. Localization of DCs nearby vasa vasorum allows monitoring of the major access pathways to the vessel wall and screening of the tissue environment for the appearance of exogenous and endogenous stressors [12]. Once sufficiently activated, DCs in the arterial wall might present the (modified auto-) antigens, such as oxidized epitopes on apoptotic cells, oxidized low density lipoproteins (oxLDL) or heat shock proteins (Hsp) to T cells and initiate inflammatory responses.

Advertisement

2. (Auto-)antigens implicated in atherogenesis and their effects on DCs

Many (auto-)antigens are involved in atherogenesis, both endogenous and exogenous. Here, we summarize some of the best-studied endogenous self-antigens in relation to DC function.

2.1. Oxidized low density lipoprotein (oxLDL)

OxLDL is one of the best-studied antigens in atherogenesis. It is considered as a ‘neoantigen’, i.e. a self-antigen that has the potential to provoke an auto-immune response upon modification, but that is tolerated by the immune system in its normal (unmodified) form [13]. It has already been shown that oxLDL can induce differentiation of monocytes into phenotypically abnormal cells, when it is added to monocytes during the early stages of differentiation [14]. These cells have functional characteristics of DCs, such as decreased endocytosis capacity, increased ability to stimulate T cell proliferation and secretion of IL-12, but not IL-10. These findings were consistent with our own study (unpublished data), which showed that monocytes differentiated (at least partly) into DCs, when they were incubated with oxLDL. This was evidenced by a pronounced decrease in the expression of CD14, a typical monocyte/macrophage marker, and increased expression of CD1a, which is mainly expressed on cortical thymocytes and DCs, and CCR-6, a receptor for CCL20 that is expressed by resting T cells and DCs (figure 1).

Figure 1.

Effects of oxLDL on monocyte differentiation. Expression of CD14, CCR-6 and CD1a after 24h incubation of monocytes with 10 µg/mL oxLDL or 50 µg/mL oxLDL points to differentiation to a phenotype with characteristics of DCs (N=3). ***P<0.001, *P<0.05 versus control, Repeated Measures ANOVA and Dunnett’s post-hoc test.

Apart from the induction of monocyte differentiation into DCs, oxLDL can also activate DCs, as demonstrated by several in vitro studies. After 24h incubation with high concentrations of oxLDL (50 µg/mL), expression of activation markers CD40, CD80 and CD83 was significantly upregulated (figure 2), and endocytotic capacity was significantly reduced (figure 3; own unpublished data).

Figure 2.

Effects of oxLDL on maturation of monocyte-derived DCs. Expression of maturation markers CD40, CD80 and CD83 after 24h incubation of immature monocyte-derived DCs with 10 µg/mL or 50 µg/mL oxLDL (N=4). Black bars represent the positive control for DC maturation, monocyte-derived DCs stimulated with lipopolysaccharide (LPS; 0.1 µg/mL). ***P<0.001, **P<0.01, *P<0.05 versus control, Repeated Measures ANOVA and Dunnett’s post-hoc test.

Figure 3.

Effects of oxLDL on endocytotic capacity of monocyte-derived DCs. Decreased endocytotic capacity of monocyte-derived DCs 24h after stimulation with oxLDL (10 µg/mL or 50 µg/mL) or the positive control LPS (0.1 µg/mL) provides functional evidence of DC maturation (N=5). *P<0.05, Repeated Measures ANOVA, Dunnett’s post-hoc test.

Cell morphology pointed to DC maturation as well: oxLDL-stimulated monocyte-derived DCs became more elongated and were arranged in clusters, when compared to unstimulated monocyte-derived DCs. The arrangement in clusters was also more pronounced when cells were stimulated with 50 µg/mL oxLDL as compared to cells stimulated with the lower concentration of oxLDL (10 µg/mL) (figure 4; own unpublished data). Alderman et al. [15] compared the effects of mildly, moderately and highly oxidized LDL and reported a significant upregulation of DC activation markers, including HLA-DR, CD40 and CD86 when cells were incubated with highly oxidized LDL. Furthermore, highly oxidized LDL increased DC-induced T cell proliferation. However, high concentrations of highly oxidized LDL (100 µg/mL) inhibited DC function through increased DC apoptosis [15]. In contrast, another study demonstrated that oxLDL did not trigger maturation of immature DCs [14]. This seems to be a discrepancy, but can easily be explained by a concentration-dependent effect of oxLDL. Perrin-Cocon and colleagues [14] varied the oxLDL concentrations between 2.5-10.0 µg/mL, which could have been insufficient to obtain monocyte-derived DC maturation. Also Zaguri et al. [16] observed no effect of 10 µg/mL oxLDL on CD86, CD83, and CCR-7 expression on DCs, whereas all those activation markers were upregulated with higher concentrations of oxLDL (50-100 µg/mL). Finally, Nickel et al. [17] reported maturation and differentiation of DCs by 10 µg/mL, but he investigated other phenotypic outcomes, such as the expression of scavenger receptors LOX1 and CD36, the mannose receptor CD205 and the activation of the nuclear factor kappa B (NF-κB) pathway.

Figure 4.

Effects of oxLDL on morphology of monocyte-derived DCs. Representative micrographs of immature, monocyte-derived DC cultures after 24h incubation with medium (A), 0.1 µg/mL lipopolysaccharide (LPS; positive control for DC maturation; B), 10 µg/mL oxLDL (C), or 50 µg/mL oxLDL (D). Phase contrast light microscopy, magnification: 10x (A, D), 20x (B, C).

2.2. Beta2-Glycoprotein I

Beta2-glycoprotein I (β2-GPI) is a plasma protein involved in the haemostatic system that has been detected in carotid atherosclerotic lesions [18]. A previous study in mice showed that the transfer of lymphocytes obtained from β2-GPI-immunized LDLr-/- mice into syngeneic mice resulted in larger fatty streaks within the recipients compared with mice that received lymphocytes from control mice [19]. From that study, it appeared that T cells specific for β2-GPI are able to increase atherosclerosis, suggesting that β2-GPI is a target auto-antigen in atherosclerosis [19].

In vitro studies have demonstrated that oxidative modification of β2-GPI, either spontaneously or induced by treatment with hydrogen peroxide, rendered the self-antigen able to induce an autoimmune response. Oxidized β2-GPI caused DC maturation, indicated by increased expression of CD80, CD86, CD83 and HLA-DR [20]. In addition, the interaction between oxidized β2-GPI and DCs led to enhanced secretion of IL-12, IL-1β, IL-6, IL-8, TNFα and IL-10. DCs stimulated with oxidized β2-GPI showed increased allostimulatory ability and induced T-helper (Th)1 polarization [20]. Also, glucose-modified β2-GPI caused phenotypic and functional maturation of iDCs, by activation of the p38 MAPK, ERK and NF-κB pathways. However, DCs stimulated with glucose-modified β2-GPI primed naïve T cells toward a Th2 polarization [21].

2.3. Heat shock proteins

Another category of auto-antigens that have been implicated in atherosclerosis are the stress-induced heat shock proteins (HSPs) [22]. HSPs are responsible for the repair or degradation of denatured proteins and, by maintaining protein conformation, they enhance the cell’s ability to survive under conditions of metabolic or oxidative stress [23]. The mRNA expression level of several HSPs, including HSP40 and HSP70, has been shown to be significantly increased in carotid endarterectomy specimens as compared to healthy arteries [24]. HSP70 seems to be homogenously distributed throughout the intima and media in healthy aortas, and a strong increase in its immunostaining intensity is observed in aortic atherosclerotic plaques [25]. They appear to stimulate an immune response leading to the development and progression of atherosclerosis [26]. A number of studies indicate that HSPs are associated with DC function and might trigger DC activation and maturation. DCs seem to overexpress HSP70 in atherosclerotic plaques and the latter protein is presumably an important trigger for DC activation [27]. Gp96 (of the HSP90 family) and HSP70 have indeed been shown to stimulate bone marrow-derived DCs in vitro to secrete cytokines [28] and to express antigen-presenting (MHC II) and costimulatory molecules (B7.2) [29]. However, Todryk and colleagues [30] reported that HSP70 targets immature DCs to make them significantly more able to capture antigens. The presence of HSP70 inhibited DC maturation induced by tumour cell lysates from parental B16 cells and maintained the DC precursor population in a more poorly differentiated phenotype. Thus, there is still controversy on whether HSPs activate DCs or keep them in an immature state, and data are lacking to robustly support a conclusion.

Advertisement

3. Survival of DCs in oxidative stress environments

Atherosclerosis is a disease that is associated with strong oxidative stress, and the creation of neo-epitopes is one of the consequences of this situation. As mentioned in section 2, the presence of reactive oxygen species (ROS) in atherosclerotic plaques may lead to the formation of oxLDL and oxidized β2-GPI, which might affect DC phenotype and function. Indeed, oxidative stress has been shown to alter the capacity of antigen-presenting cells to process antigens and to initiate a primary T-cell response. In this respect, it is interesting to unravel whether DCs show phenotypic adaptations in order to function under oxidative stress situations. In a recent study, we demonstrated that DCs appear to be resistant to the detrimental effects of oxidative stress. We showed by confocal live cell imaging that monocyte-derived DCs, which were generated as described earlier [31], were better capable of neutralizing ROS induced by tertiary-butylhydroperoxide (tert-BHP) in comparison to their precursor monocytes [31]. Tert-BHP was selected to induce ROS because it acutely evokes oxidative stress, resulting in cell toxicity [32]. Decomposition of tert-BHP to alkoxyl or peroxyl radicals accelerates lipid peroxidation chain reactions [33]. By means of a neutral red viability assay, we observed that tert-BHP induced significant and rapid cell death in both monocytes and DCs. Yet, monocyte-derived DCs were more resistant to tert-BHP-induced cell death than their precursor cells [31]. A PCR profiler array specific for oxidative stress and antioxidant-related pathways revealed an upregulation of several important antioxidant genes during differentiation of monocytes into DCs, including catalase, peroxiredoxin 2 (PRDX2) and glutathione peroxidase 3 (GPX3). Catalase encodes the enzyme that catalyses the decomposition of hydrogen peroxide to water and oxygen. GPX3 and PRDX2 are genes encoding enzymes that can detoxify hydrogen peroxide and lipid hydroperoxides [34,35]. However, PRDX2 is more efficient in neutralizing hydrogen peroxide than catalase or GPXs [36,37]. Immunoblotting or immunohistochemistry showed that the upregulated transcription of PRDX2 and GPX3 was translated in a significant increase at the protein level. Especially PRDX2 appears to be an important factor in the neutralization of ROS induced by tert-BHP [31]. Previously, and in accordance with our recent findings, two studies that used different detection methods reported high expression of antioxidant enzymes in monocyte-derived DCs. A functional study indicated indirectly that monocyte-derived DCs might show enhanced activity of catalase [38]. A proteomic analysis showed higher expression of superoxide dismutase (SOD)2, PRDX1 and PRDX2 in monocyte-derived DCs when compared to precursor monocytes [39]. The latter study also stated that DCs were more resistant than monocytes to apoptosis induced by high amounts of oxLDL [39]. It is conceivable that the good survival skills of monocyte-derived DCs in oxidative stress environments are crucial in atherosclerotic plaques, enabling these professional antigen-presenting cells to exert their function(s).

Advertisement

4. DC subtypes in mice and men

As discussed above, DCs process and present self and foreign antigens to T cells and are therefore important inducers of adaptive immune responses. However, ‘the’ DC does not exist, as DCs comprise a network of subsets that are phenotypically, functionally, and developmentally distinct [40,41]. It is essential to understand the diversity in DC subtypes to target DCs for immunomodulating therapies. Most studies on DC subsets have been performed in mice, because lymphoid tissue is easier to obtain from mice than from humans. Mature mouse DCs are identified based on their expression of the integrin alpha X chain CD11c, the costimulatory molecules CD40, CD80 and CD86, and high surface levels of the antigen-presenting molecule MHC II [42,43,40]. The T cell markers CD4 and CD8 (in the form of a αα-homodimer) are also expressed on mouse DCs, and can be used to distinguish different subtypes [44]. In general, three DC subsets can be characterized in mouse lymphoid tissue (table 1): 1) CD8α+ CD4- DCs; 2) CD8α- CD4+ DCs; and 3) CD8α- CD4- DCs [44]. The CD8α+ CD4- DCs are mainly localized in the T cell areas of lymphoid organs, whereas the CD8α- CD4+ DCs are found in the marginal zones. Yet, upon stimulation by microbial products, such as lipopolysaccharide, the latter can also migrate to the T cell zones [45,46]. Other markers that can be used to further subdivide mouse DC subsets include the integrin alpha M chain CD11b and the endocytosis receptor CD205 (DEC205). The CD8α+ CD4- DCs are also CD205+ CD11b- and they are mainly present in the thymus, and at moderate levels in lymph nodes [40]. Lymph nodes further contain, in contrast to spleen, CD8α- CD4- CD11b+ CD205mid DCs which are considered as the mature form of tissue interstitial DCs [40,42,43] (table 1). Another DC subtype, which is langerinhigh CD11b+ CD8αlow CD205high, is only found in skin-draining lymph nodes and considered as the mature form of epidermal Langerhans cells. These cells are also positive for MHC II and CD40, CD80 and CD86, suggesting that they are fully activated [42].

The numerous DC subtypes in mouse lymphoid organs are all able to present antigens to T cells, however, they differ in other aspects of DC-T cell communication [40]. CD8α+ DCs mainly induce Th1/Th17-polarizing cytokine responses in CD4+ effector T cells, whereas CD8α- DCs are able to induce Th2-biased cytokine responses [47,48,49,50]. CD8α+ DCs also seem to be specialized for the uptake and cross-presentation of exogenous antigens on MHC I and consequently stimulate CD8+ cytotoxic T cells, whereas CD8α- DCs mainly stimulate CD4+ T helper cells [51,52].

It has to be noticed that the association between mouse and human DC subsets remains elusive, making translation of the above-mentioned findings difficult. One of the major barriers in comparing mouse and human DC subsets is the lack of CD8α expression on human DCs [53]. As a result, it remains unclear which subtype represents the human equivalent of mature mouse CD8α+ DCs. Another important barrier is that most human studies are performed on blood, due to the limited availability of human spleen tissue. Moreover, human blood DCs are mainly immature and heterogeneous in their expression of a range of markers. It might be that part of the heterogeneity reflects differences in the maturation or activation state of DCs, rather than that they all represent separate sub-lineages. Yet, one subtype that is similar to its mouse counterpart is the human Langerhans cell, which expresses CD1a and langerin and is characterized by the presence of Birbeck granules [40].

DC subtype Subdivision according Phenotype DC subsets
to localization MOUSE HUMAN
cDCs lymphoid organ-resident cDC CD8α+ CD4- CD205+ CD11b- lineage- HLA-DR+ CD11c+ CD1b/c+
CD8α- CD4+ lineage- HLA-DR+ CD11c+ CD141+
CD8α- CD4- CD205mid CD11b+ lineage- HLA-DR+ CD11c+ CD16+
circulating cDC CD8α- CD11b+ CD11chigh CD1c+ CD11c+
CD8α+ CD205+ CD11c+ CD141+ CD11c+ XCR1+
pDCs lymphoid organ-resident pDC CD11c- CD304+
circulating pDC PDCA-1+ CD11c+ CD11b- CD303+ CD304+ CD123+
DC activation status Markers
Activated (mature) DCs Costimulatory molecules CD40 CD80 CD86
Activation molecules CD83

Table 1.

Markers used for characterization of DC subtypes in mice and men

cDC = conventional dendritic cell, pDC = plasmacytoid dendritic cell


lineage = cocktail of CD3, CD14, CD16, CD19, CD20, CD56; CD1c = BDCA-1; CD303 = BDCA-2; CD141 = BDCA-3; CD304 = BDCA-4


BDCA = blood dendritic cell antigen


In human blood, the first made classification is often the distinction between plasmacytoid (p)DCs, and myeloid or conventional DCs (cDCs) (table 1). Freshly isolated pDCs resemble plasma cells and have a morphology typical of that of large, round cells with a diffuse nucleus and few dendrites. These type I IFN-producing cells (IPCs) are specialized in innate antiviral immune responses by producing copious amounts of type I interferons. pDCs express CD303 (blood dendritic cell antigen (BDCA) 2), CD304 (BDCA 4) and CD123 (IL 3Rα), whereas cDCs are characterized by their expression of CD1c (BDCA 1) and CD11c [54] (table 1). In addition, pDCs and cDCs also express different sets of Toll-like receptors (TLRs). In brief, pDCs express mainly TLR7 and TLR9, whereas cDCs exhibit strong expression of TLR1, TLR2, TLR3, TLR4, and TLR8. Accordingly, pDCs mainly recognize viral components with subsequent production of a large amount of IFN-α. In contrast cDCs recognize bacterial components and produce pro-inflammatory cytokines such as IL-12p70, TNF-α, and IL-6 [54,7].

Furthermore, cDCs and pDCs also differ in migration behaviour. Generally it is assumed that myeloid (m)DCs are the conventional DCs that infiltrate peripheral tissues, while pDCs migrate directly from the blood into lymphoid organs [54]. Finally, a small third population of blood DCs expressing CD11c and BDCA-3 (CD141) but not BDCA-1, CD123 or BDCA-2 can be distinguished (table 1). Of particular importance is their superior antigen cross-presentation capacity and expression of the XC chemokine receptor 1 (XCR1), suggesting that they represent the human counterpart of mouse CD8α+ DCs. They emerge as a distinctive myeloid DC subset that is characterized by high expression of TLR3, production of IL-12 and IFN-β, and a superior capacity to induce T helper-1 cell responses, when compared to BDCA-1+ mDCs [54,7].

Only in a few recent studies, human DCs have been isolated from lymphoid tissues, which allow direct comparison with mouse DC subtypes. Mittag and colleagues [41] identified four DC subsets in human spleen that resemble DCs found in human blood. These include three cDC subtypes and one pDC subtype (table 1). The cDCs are all negative for lineage markers and positive for HLA-DR and CD11c, and they differ in their expression of CD1b/c (= BDCA-1), CD141 (= BDCA-3) and CD16. The pDCs express high levels of CD304 (= BDCA-4), but not CD11c [41]. Moreover, the hallmark functions of mouse CD8α+ DC subsets, which include IL-12p70 secretion and cross-presentation, appeared to be not restricted to the equivalent human CD141+ cDCs as thought earlier, but shared by CD1b/c+ and CD16+ DC subsets [41].

Advertisement

5. Discriminating between DCs and macrophages

It has become clear that DCs, especially DCs from myeloid origin, are very heterogeneous, representing several subtypes with a common origin, but different anatomical locations (lymphoid organs vs. non-lymphoid organs), function and phenotype. Moreover, there is also a very close relationship between myeloid DCs and macrophages (figure 5).

The distinction of the differences between macrophages and the heterogeneous family of DCs is notoriously difficult and complicated by the plasticity of both cell types [55]. Monocytes that exit the blood and enter tissues under inflammatory conditions can differentiate to macrophages, but also to DCs that share several phenotypic features and functions, making it difficult to unambiguously define macrophages and DCs as individual entities [56]. In addition, resting peripheral monocytes, obtained from mouse peritoneal cavity lavage, represent an immature population, capable of further differentiation along either the dendritic or the macrophage pathway, depending on the type of stimuli (cytokines, growth factors) they receive [10]. Furthermore, many DC subsets are not clearly defined and it is absolutely necessary to bear in mind that different groups use different methods to identify and characterize DCs [57]. Often, the starting populations are preselected based on randomly defined expression levels of markers that were believed to be specific for either DCs or macrophages, but are in fact expressed by both [58] (figure 5).

Figure 5.

Functional characteristics and surface markers of DCs and macrophages. Increasing evidence demonstrates an enormous overlap between what is considered a 'macrophage' and a ‘DC'. Abbreviations: MHC II, major histocompatibility complex class II; BDCA-1, blood dendritic-cell antigen-1; DC-SIGN, dendritic cell-specific ICAM-3-grabbing non-integrin.

Consequently, confusion in distinguishing between macrophages and DCs has been – at least in part – caused by the use of nonspecific cell surface markers, such as CD11c. In addition, the number of DC and macrophage subpopulations that can be defined is an exponential function of the number of markers that has been examined [59]. Moreover, since each gene/protein has its own intrinsic expression level, the heterogeneity is really unlimited [60]. CD11c, a commonly used DC marker, was already known to be expressed by most tissue macrophages before the use of CD11c-reporter transgenes as markers of DCs, and of CD11c-DTR mice to ‘selectively’ deplete them [59,61]. Other markers that have been used to track macrophages and DCs in mice include F4/80, CD11b and MHC II, but they have also turned out to be nonspecific [57]. Too little attention has been paid to the expression of antimicrobial effector molecules, such as lysozyme, which is highly secreted by monocytes and macrophages, but only weakly expressed, if at all, by DCs [62]. Part of the confusion may also result from the flexibility and plasticity of macrophages and from the presence of resident and migratory activated DCs in the same organ [63].

The confusion could be possibly resolved if the appropriate reflections are considered [57]. For example, the correctness of CD11c to identify DCs depends on the anatomical site in question. In the spleen and lymph nodes, mononuclear phagocytes with high expression levels of CD11c – though not those with low or intermediate CD11c – appear to be DCs rather than macrophages. Accumulating evidence confirms that spleen and lymph node DCs are functionally different from macrophages, do not originate from differentiating monocytes, and share fewer characteristics with monocytes than macrophages [64,65,66]. However, in the lung, high levels of CD11c are expressed on macrophages [67,68], and there are many other anatomical locations apart from the lymphoid organs where macrophages are CD11c-positive. It has been proposed many times that the same set of markers that allows us to discriminate between DCs and macrophages in lymphoid organs, can also be used in non-lymphoid organs, but it has become clear that this assumption is not correct.

Recent in vivo experiments in mice have increased our understanding of the development and functions of DC and macrophage subsets [69,70,71]. However, despite this progress in mice, corresponding human subsets are yet to be characterized. Until now, there is no morphologic or protein marker of macrophages or DCs which is unambiguous. Moreover, a single set of markers cannot be assumed to apply to all stages of cell differentiation and activation. In conclusion, there is insufficient knowledge to make definitive claims about any marker combination, particularly in non-lymphoid compartments.

If the distinction between DCs and macrophages cannot be made based on morphological features, can it be based on function? Several criteria to define DCs include the property of DCs to localize in the T cell zone of lymphoid organs where they can stimulate T cells, as well as their ability to migrate and carry antigen [72,73]. In contrast, macrophages are best defined by their phagocytic activity and are generally considered as tissue-resident cells. However, recent studies show that macrophages can also migrate and that Langerhans cells (i.e. DCs from the skin and mucosa that carry large Birbeck granules) are not important for T cell priming [74]. In addition, some macrophage subtypes, such as microglia, show only poor phagocytic capacity [57]. Taken together, there is no good functional criterion to define macrophages and monocyte-derived DCs (figure 5), since they represent not just two different cell populations, but various cell subtypes. As they are derived from a common precursor, it is really hard to fully identify macrophages and DCs as two separate entities.

Advertisement

6. Pro-and anti-atherogenic properties of various DC subtypes

We and others discovered a profoundly altered circulating DC compartment in patients with coronary artery disease (CAD), the clinical manifestation of atherosclerosis, as compared to healthy donors [75,76,77,78,79,80]. In 2006, we reported for the first time a decrease in circulating DC precursors (BDCA-1+ mDCs, BDCA-2+ pDCs) in CAD patients by flow cytometry. CAD was determined by angiography and defined as more than 50% stenosis in one or more coronary arteries [77]. In parallel, Yilmaz et al. [79] found a marked reduction in mDC precursors in CAD patients, though the decline in pDCs was less pronounced. Next, we studied whether the lower blood DC counts in CAD patients were related to the extent of atherosclerosis (one- versus three-vessel disease) or type (stable versus unstable angina pectoris) of CAD. Again, we observed significantly lower relative and absolute numbers of pDCs and mDCs in patients with coronary atherosclerosis [78]. Interestingly, the overall lineage-negative HLA-DR-positive blood DCs, which also include other blood DCs (such as BDCA-3+) or more mature blood DCs, confirmed the decline of BDCA+ DC precursors. However, the counts of circulating DCs dropped to the same extent in three groups of CAD patients, irrespective of the number (one or three) of affected arteries or the type (stable or unstable) of angina [78]. Consistent with our results, Yilmaz and colleagues [79] reported no differences between clinically stable or unstable CAD. Yet, in a later and more extended study with a cohort of 290 patients, in which a more refined ‘CAD score’ was used to classify patients, they found that the numbers of pDCs, mDCs, and total DCs decreased when the extent of coronary atherosclerosis increased [80].

Besides flow cytometric studies, we performed immunohistochemical analyses demonstrating increased intimal DC counts with evolving plaque stages, in close relationship with lesional T cells [81]. These findings strongly suggest that blood DCs migrate from the circulation to the atherosclerotic lesion, possibly attracted by chemokines produced by the inflammatory infiltrate in the plaque, and subsequently stimulate T cell proliferation [7]. However, it is unlikely that accumulation of DC into a single tissue site is responsible for the major changes in the number of circulating DCs in CAD [12]. Possibly, DCs may leave the blood to migrate into lymphoid tissues in response to systemic inflammatory activation, which redirects trafficking and compartmentalization of antigen-presenting DCs as well as lymphocytes. Indeed, it has been mentioned that DC numbers of lymph nodes attached to atherosclerotic wall segments exceed those in lymph nodes attached to non-atherosclerotic arteries [7]. The declined circulating DC numbers in atherosclerosis might also be the result of impaired differentiation from bone marrow progenitors. Interestingly, we recently showed that plasma Flt3 ligand (Flt3L) concentrations were reduced in CAD patients [75]. Flt3L is a major cytokine involved in both pDC and mDC development from haematopoietic stem cells and their release from the bone marrow [82,83,84]. As plasma Flt3L correlated with blood DC counts, the reduced blood DCs in CAD might be caused by impaired DC differentiation from bone marrow progenitors. Until now, it remains unclear why plasma Flt3L levels are lowered in CAD. Other possible explanations for the decrease of circulating DC subsets in CAD patients include DC activation resulting in enhanced migration or in loss of subset markers, drug-induced changes, or increased DC turnover, and are reviewed elsewhere [7].

The finding that blood DCs are decreased in CAD patients and that atherosclerotic arteries display a marked increase in the number of DCs suggest the involvement of DCs in the pathogenesis of atherosclerosis. Yet, the exact role of DCs in atherogenesis has not been fully clarified. Moreover, increasing evidence points to different behaviour of DC subsets in the initiation and progression of the disease. We have recently demonstrated in vitro that mDCs in CAD operate in a normal way, whereas pDCs from CAD patients are not only reduced in number, but also seem to be functionally impaired [75].

Most evidence points to a proatherogenic role for mDCs. Apolipoprotein E (ApoE)/IL-12 double knockout mice develop smaller atherosclerotic lesions than ApoE deficient (ApoE-/-) mice, illustrating the proatherogenic effect of IL-12, which is the main cytokine secreted by mDCs [85]. Moreover, daily IL-12 administration promotes atherosclerosis in ApoE-/- mice [86]. Because mDCs from CAD patients are still able to mature [75], it is plausible that the blood mDCs that are activated by atherosclerosis-favouring factors in the circulation migrate to the atherosclerotic plaque or the lymph nodes attached to the atherosclerotic wall segments. Once arrived, they might initiate and maintain the inflammatory response by continuous T-cell stimulation. Nevertheless, DCs are not only implicated in the immune response in atherosclerosis, they are also involved in cholesterol homeostasis. A recent study using a mouse model in which the receptor for diphtheria toxin was expressed under the CD11c promoter (CD11c-DTR) showed that (transient) depletion of CD11c+ cDCs resulted in enhanced cholesterolaemia [87]. The latter indicates that DCs are important in regulating the accumulation of lipids during the earliest stages of plaque formation. In contrast, enhancement of the life span and immunogenicity of DCs by specific overexpression of the anti-apoptotic gene hBcl-2 under the control of the CD11c promoter was associated with an atheroprotective decrease in plasma cholesterol levels, neutralizing the proatherogenic signature of enhanced T cell activation, a Th1 and Th17 cytokine expression profile, and elevated production of T-helper 1–driven IgG2c autoantibodies directed against oxidation-specific epitopes. -As a net result, there was no acceleration of atherosclerotic plaque progression [87].

It is not yet clear whether pDCs are proatherogenic or atheroprotective. PDCs might be involved in plaque destabilization, as they have the unique ability of producing large amounts of type I IFNs. This cytokine exerts strong antiviral effects, but more importantly, it induces marked upregulation of tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) on CD4+ T cells, which might lead to killing of plaque-resident cells, potentially weakening the scaffold of the lesion and rendering the plaque vulnerable [88]. In addition, nucleotides released from necrotic or apoptotic cells can induce IFN-α production by pDCs in the presence of antimicrobial peptides released from inflammatory cells [89]. Plaque-residing pDCs have also been shown to respond to CpGs (containing motifs typically found in microbial DNA) leading to enhanced IFN-α expression. This process amplifies inflammatory TLR-4, TNF-α, and IL-12 expression by mDCs, and correlates with plaque instability [90]. A recent study in ApoE-/- mice reported that administration of a plasmacytoid dendritic cell antigen-1 (PDCA-1) antibody to deplete pDCs protected from lesion formation [91], demonstrating that pDCs indeed exert proatherogenic functions during early lesion formation. In contrast, pDC depletion by administration of the 120G8 monoclonal antibody promoted plaque T-cell accumulation and exacerbated lesion development and progression in LDLr⁻/⁻ mice [92]. PDC depletion was accompanied by increased CD4⁺ T-cell proliferation, IFN-γ expression by splenic T cells, and plasma IFN-γ levels, pointing to a protective role for pDCs in atherosclerosis. Thus, the exact role of pDCs in atherosclerosis remains to be further unravelled.

Advertisement

7. DCs as therapeutic targets

Until now, it is impossible to fully inhibit the formation or progression of atherosclerotic lesions in the clinic. Current therapies for atherosclerosis (e.g. statins, stent placement) focus on relieving symptoms, and consequently many patients remain at high risk for future acute coronary events. A very effective strategy in other immune-related pathologies is vaccination, where the culprit protein or the weakened/dead version of the micro-organism is injected to the body in order to create a highly specific primary humoral immune-response [93]. New vaccines have recently been developed that deliver relevant antigens and adjuvants to redirect the immune system for the individual’s benefit [94]. Because DCs are the most effective antigen presenting cells that initiate and regulate the immune response, they seem extremely suitable as vaccine basis. On the one hand, they can activate T cells, on the other hand, they can specifically silence unwanted immune reactions by inducing tolerance [95]. They might function as natural adjuvants for the induction of antigen-specific T-cell responses. Approaches using DCs in atherosclerosis immunotherapy may be comparable to those already used for cancer immunotherapy [96,97,98], although a different immune response is required. One approach that is already intensively studied is the immunization with autologous, monocyte-derived DCs from the patient that are loaded with appropriate antigens ex vivo [96]. Such ex vivo generated and antigen-loaded DCs have nowadays been used as vaccines to improve immunity in patients with cancer [99] and chronic human immunodeficiency virus (HIV) infection [100,101], providing a “proof of principle” that DC vaccines can work.

In the context of atherosclerosis, immunization of hypercholesterolemic animals with oxLDL or specific epitopes of ApoB100 has already been shown to inhibit atherosclerosis [102,103,104,105,106]. When LDL receptor-deficient (LDLr-/-) rabbits were immunized with malondialdehyde modified LDL (MDA-LDL), a reduction in the extent of atherosclerotic lesions was observed in the aortic tree [102]. These observations were confirmed in LDLr-/- and apolipoprotein E deficient (ApoE-/-) mice [103,104]. Also hypercholesterolemic rabbits that were immunized with oxLDL showed reduced atherosclerotic lesions in the proximal aorta [107]. Possibly, oxLDL-pulsed DCs or DCs pulsed with immunogenic components of oxLDL could be used for vaccination as well, thereby avoiding the side effects of direct vaccination with oxLDL [108]. A series of studies have already used pulsed DCs as an immunotherapy for atherosclerosis in mice, however, results were not always consistent. Repeated injection of LDLr-/- mice with oxLDL-pulsed mature DCs resulted in attenuation of lesion development with a decreased amount of macrophages and increased collagen content, contributing to a more stable plaque phenotype [109]. Moreover, a similar approach was carried out using mice expressing the full-length human ApoB100 in the liver and humanized lipoprotein profiles [110]. Those mice were repeatedly injected with mature DCs that were incubated with IL-10 and ApoB100, prior to the initiation of a Western diet. The immunosuppressive cytokine IL-10 was used to induce tolerogenic DCs [110]. This approach resulted in attenuation of atherosclerotic lesion development in the aorta, which was associated with decreased cellular immunity to ApoB100. Also, decreased Th1 and Th2 responses most likely due to enhanced regulatory T cell (Treg) expansion were observed [110]. In contrast, subcutaneous injection of DCs that were simultaneously pulsed with LPS and MDA-LDL into ApoE-/- mice at frequent intervals during lesion formation caused a significant increase in lesion size in the aortic root [111]. These differential effects may be due to different forms of antigen presentation leading to qualitatively different immune responses. Apart from oxLDL, DCs might also be pulsed ex vivo by cultivating them with a total extract or suspension of atherosclerotic plaque tissue, for example, from patients undergoing carotid endarterectomy [95,108] (figure 6). A major advantage of such a therapy, where a patient is vaccinated with its own DCs pulsed by its own antigens is the efficiency, because it would imitate events as they occur in plaques in situ in the patient.

Figure 6.

Promising areas for further research to treat immune-mediated diseases, such as atherosclerosis. Immunization of patients with autologous, monocyte-derived DCs that are loaded with appropriate antigens ex vivo. This approach has already been proven successful in cancer and HIV patients.

Another promising area for further research is the development of tolerogenic vaccines for immune-mediated diseases (figure 7). Both foreign and self-antigens can be targets of tolerogenic processes. DCs can be converted to ‘tolerogenic DCs’ by addition of various immunomodulating agents, including IL-10, transforming growth factor-beta (TGF-β) and 1,25-dihydroxyvitamin D3 [8], or they can be generated by using small interfering RNA (siRNA) that specifically targets IL-12p35 gene [112] (figure 7). Tolerogenic DC-based immunotherapy has recently been tested in mice as a possible novel approach to induce immunological tolerance for prevention or treatment of atherosclerosis [110]. Hermansson et al. [110] used IL-10 to induce tolerogenic DCs. Another group showed that oral administration of calcitriol, the active form of vitamin D3, induced the generation of tolerogenic DCs as well as a significant increase in Foxp3+ Tregs in the lymph nodes, spleen, and atherosclerotic lesions of ApoE-/- mice, which resulted in an inhibition of atherosclerosis [113]. This was associated with increased IL-10 and decreased IL-12 mRNA expression. Furthermore, DCs from the calcitriol group showed reduced CD80 and CD86 expression and decreased proliferative activity of T lymphocytes, indicating that tolerogenic or maturation-resistant DCs show some similarities with immature DCs [113]. Hussain and colleagues [114] hypothesized that aspirin may also induce tolerogenic DCs and CD4+ CD25+ FoxP3+ Treg cells activity/augmentation in experimental models of autoimmune atherosclerosis. Aspirin-induced tolerogenic DCs initiated regulatory activity in responder T cells as they showed a decreased expression of costimulatory molecules and an increased expression of immunoglobulin-like transcript 3 (ILT-3), which is a co-inhibitor of T cell activation required to induce Tregs [114,115,116]. Indeed, the presentation of antigen complexes to T cells in the absence of costimulatory signals could lead to anergy or apoptosis of T cells, or the induction of Treg. Therefore, it might also be useful to adjust the expression of costimulatory molecules on pulsed DCs ex vivo prior to the vaccination [96,98,97,94,117,118].

Figure 7.

Generation of tolerogenic DCs to develop tolerogenic vaccines. Tolerogenic DC-based immunotherapy has recently been successfully tested in mice as a possible novel approach to induce immunological tolerance for prevention or treatment of atherosclerosis.

A completely different strategy that might be used in therapeutic intervention implicates the use of DCs to deplete specific immune cells, such as the detrimental Th1 or Th17 cells, in atherosclerosis. The opposite approach has been shown to work in a mouse model of atherosclerosis. Van Es et al. [119] used DCs to deplete atheroprotective Tregs by vaccinating LDLr-/- mice with DCs which were transfected with Foxp3 encoding mRNA. This approach resulted in a cytotoxic T lymphocyte (CTL) response against Foxp3 and a subsequent depletion of Foxp3+ Tregs. Vaccination against Foxp3 aggravated atherosclerosis, it resulted in a reduction of Foxp3+ regulatory T cells in spleen, lymph nodes and circulation, and in an increase in initial atherosclerotic lesion formation. Besides an increase in lesion size, vaccination against Foxp3 also induced a 30% increase in cellularity of the initial lesions, which may indicate an increase in inflammation within the lesions [119].

Another approach for therapeutic intervention against atherosclerosis might involve the direct targeting of DCs in vivo by manipulating the functions of different DC subsets [95]. Based on the hypothesis that cDCs act rather proatherogenic, whereas pDCs might be atheroprotective (see section 4), suppression of the myeloid DC subset and activation of the lymphoid subset might enable immune reactions in atherosclerosis to be regulated [95]. For future studies, it would be very useful to isolate DCs resident in plaques to be able to identify a unique antigen(s) on their surface. That would possibly lead to new strategies where plaque DCs can be targeted to deliver biologically active substances to atherosclerotic lesions. The challenge is to selectively identify regulatory molecules and novel therapies in order to inhibit DC migration and function during atherogenesis without affecting normal DC function under physiological conditions.

Advertisement

8. Conclusion

As it is now well accepted that atherosclerosis is an immune-mediated disease, the targeting of its cellular components might open possibilities for new therapeutic strategies to attenuate the progression of the disease. DCs seem to initiate and regulate immune responses in atherosclerosis and they are also involved in controlling cholesterol homeostasis by yet unknown mechanisms. It would be important to identify the pathway(s) through which CD11c+ cells may modulate the levels of plasma cholesterol. One should take into account that DCs represent a very heterogeneous population, with many subsets that have different phenotypes, functions, origin and anatomical distribution. So far, it is unclear if all DCs have equal antigen-presenting capacities, and very little is known about a preferential DC subset that is responsible for T cell-induced inflammation in the vessel wall. Moreover, there is a close relationship between DCs and macrophages, and the distinction between both cell types is even further complicated by their plasticity. Future studies are essential to determine which DC subtypes exert pro- or anti-atherogenic effects. It is crucial to understand the diversity in DC subsets to target DCs for immunomodulation therapies. Furthermore, functional differences between phenotypically similar mouse and human DC subtypes should also be studied. Nevertheless, DC-based vaccination strategies have been proven successful and animal studies provide some promising data for the treatment of atherosclerosis as well. Yet, several issues, such as the most appropriate antigen(s) for loading DCs and the optimal type of DC used for vaccination remain to be further investigated.

Advertisement

Acknowledgements

This work was supported by the University of Antwerp [GOA-BOF 2407 and TOP-GOA 3018].

References

  1. 1. Wick G, Perschinka H, and Millonig G. Atherosclerosis as an autoimmune disease: an update. Trends Immunol 2001;22(12) 665-669.
  2. 2. Libby P, Ridker PM, and Maseri A. Inflammation and atherosclerosis. Circulation 2002;105(9) 1135-1143.
  3. 3. Hansson GK, Robertson AK, and Soderberg-Naucler C. Inflammation and atherosclerosis. Annu Rev Pathol 2006;1(297-329.
  4. 4. Hansson GK and Libby P. The immune response in atherosclerosis: a double-edged sword. Nat Rev Immunol 2006;6(7) 508-519.
  5. 5. Takahashi K, Takeya M, and Sakashita N. Multifunctional roles of macrophages in the development and progression of atherosclerosis in humans and experimental animals. Med Electron Microsc 2002;35(4) 179-203.
  6. 6. Banchereau J, Briere F, Caux C et al. Immunobiology of dendritic cells. Annu Rev Immunol 2000;18(767-811.
  7. 7. Van Vré EA, Van Brussel I, Bosmans JM et al. Dendritic cells in human atherosclerosis: from circulation to atherosclerotic plaques. Mediators Inflamm 2011;2011(941396-
  8. 8. Steinman RM, Hawiger D, and Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol 2003;21(685-711.
  9. 9. Rossi M and Young JW. Human dendritic cells: potent antigen-presenting cells at the crossroads of innate and adaptive immunity. J Immunol 2005;175(3) 1373-1381.
  10. 10. Rezzani R, Rodella L, Zauli G et al. Mouse peritoneal cells as a reservoir of late dendritic cell progenitors. Br J Haematol 1999;104(1) 111-118.
  11. 11. Bobryshev YV and Lord RS. Ultrastructural recognition of cells with dendritic cell morphology in human aortic intima. Contacting interactions of Vascular Dendritic Cells in athero-resistant and athero-prone areas of the normal aorta. Arch Histol Cytol 1995;58(3) 307-322.
  12. 12. Niessner A and Weyand CM. Dendritic cells in atherosclerotic disease. Clin Immunol 2010;134(1) 25-32.
  13. 13. Milioti N, Bermudez-Fajardo A, Penichet ML et al. Antigen-induced immunomodulation in the pathogenesis of atherosclerosis. Clin Dev Immunol 2008;2008(723539-
  14. 14. Perrin-Cocon L, Coutant F, Agaugue S et al. Oxidized low-density lipoprotein promotes mature dendritic cell transition from differentiating monocyte. J Immunol 2001;167(7) 3785-3791.
  15. 15. Alderman CJ, Bunyard PR, Chain BM et al. Effects of oxidised low density lipoprotein on dendritic cells: a possible immunoregulatory component of the atherogenic micro-environment? Cardiovasc Res 2002;55(4) 806-819.
  16. 16. Zaguri R, Verbovetski I, Atallah M et al. 'Danger' effect of low-density lipoprotein (LDL) and oxidized LDL on human immature dendritic cells. Clin Exp Immunol 2007;149(3) 543-552.
  17. 17. Nickel T, Schmauss D, Hanssen H et al. oxLDL uptake by dendritic cells induces upregulation of scavenger-receptors, maturation and differentiation. Atherosclerosis 2009;205(2) 442-450.
  18. 18. Profumo E, Buttari B, and Rigano R. Oxidative stress in cardiovascular inflammation: its involvement in autoimmune responses. Int J Inflam 2011;2011(295705-
  19. 19. George J, Harats D, Gilburd B et al. Adoptive transfer of beta(2)-glycoprotein I-reactive lymphocytes enhances early atherosclerosis in LDL receptor-deficient mice. Circulation 2000;102(15) 1822-1827.
  20. 20. Buttari B, Profumo E, Mattei V et al. Oxidized beta2-glycoprotein I induces human dendritic cell maturation and promotes a T helper type 1 response. Blood 2005;106(12) 3880-3887.
  21. 21. Buttari B, Profumo E, Capozzi A et al. Advanced glycation end products of human beta(2) glycoprotein I modulate the maturation and function of DCs. Blood 2011;117(23) 6152-6161.
  22. 22. Wick G, Knoflach M, and Xu Q. Autoimmune and inflammatory mechanisms in atherosclerosis. Annu Rev Immunol 2004;22(361-403.
  23. 23. Benjamin IJ and McMillan DR. Stress (heat shock) proteins: molecular chaperones in cardiovascular biology and disease. Circ Res 1998;83(2) 117-132.
  24. 24. Nguyen TQ, Jaramillo A, Thompson RW et al. Increased expression of HDJ-2 (hsp40) in carotid artery atherosclerosis: a novel heat shock protein associated with luminal stenosis and plaque ulceration. J Vasc Surg 2001;33(5) 1065-1071.
  25. 25. Berberian PA, Myers W, Tytell M et al. Immunohistochemical localization of heat shock protein-70 in normal-appearing and atherosclerotic specimens of human arteries. Am J Pathol 1990;136(1) 71-80.
  26. 26. Pockley AG. Heat shock proteins, inflammation, and cardiovascular disease. Circulation 2002;105(8) 1012-1017.
  27. 27. Bobryshev YV and Lord RS. Expression of heat shock protein-70 by dendritic cells in the arterial intima and its potential significance in atherogenesis. J Vasc Surg 2002;35(2) 368-375.
  28. 28. Basu S, Suto R, Binder RJ et al. Heat shock proteins as novel mediators of cytokine secretion by macrophages. Cell Stress Chaperon 1998;3(11-
  29. 29. Basu S, Binder RJ, Suto R et al. Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-kappa B pathway. Int Immunol 2000;12(11) 1539-1546.
  30. 30. Todryk S, Melcher AA, Hardwick N et al. Heat shock protein 70 induced during tumor cell killing induces Th1 cytokines and targets immature dendritic cell precursors to enhance antigen uptake. J Immunol 1999;163(3) 1398-1408.
  31. 31. Van Brussel I, Schrijvers DM, Martinet W et al. Transcript and protein analysis reveals better survival skills of monocyte-derived dendritic cells compared to monocytes during oxidative stress. PLoS ONE 2012;7(8) e43357-
  32. 32. Prasad KD, Sai Ram M., Kumar R et al. Cytoprotective and antioxidant activity of Rhodiola imbricata against tert-butyl hydroperoxide induced oxidative injury in U-937 human macrophages. Mol Cell Biochem 2005;275(1-2) 1-6.
  33. 33. Martín C, Martínez R, Navarro R et al. tert-Butyl hydroperoxide-induced lipid signaling in hepatocytes: involvement of glutathione and free radicals. Biochem Pharmacol 2001;62(6) 705-712.
  34. 34. Schwaab V, Faure J, Dufaure JP et al. GPx3: the plasma-type glutathione peroxidase is expressed under androgenic control in the mouse epididymis and vas deferens. Mol Reprod Dev 1998;51(4) 362-372.
  35. 35. Manandhar G, Miranda-Vizuete A, Pedrajas JR et al. Peroxiredoxin 2 and peroxidase enzymatic activity of mammalian spermatozoa. Biol Reprod 2009;80(6) 1168-1177.
  36. 36. Berggren MI, Husbeck B, Samulitis B et al. Thioredoxin peroxidase-1 (peroxiredoxin-1) is increased in thioredoxin-1 transfected cells and results in enhanced protection against apoptosis caused by hydrogen peroxide but not by other agents including dexamethasone, etoposide, and doxorubicin. Arch Biochem Biophys 2001;392(1) 103-109.
  37. 37. Peskin AV, Low FM, Paton LN et al. The high reactivity of peroxiredoxin 2 with H(2)O(2) is not reflected in its reaction with other oxidants and thiol reagents. J Biol Chem 2007;282(16) 11885-11892.
  38. 38. Thorén FB, Betten A, Romero AI et al. Cutting edge: Antioxidative properties of myeloid dendritic cells: protection of T cells and NK cells from oxygen radical-induced inactivation and apoptosis. J Immunol 2007;179(1) 21-25.
  39. 39. Rivollier A, Perrin-Cocon L, Luche S et al. High expression of antioxidant proteins in dendritic cells: possible implications in atherosclerosis. Mol Cell Proteomics 2006;5(4) 726-736.
  40. 40. Shortman K and Liu YJ. Mouse and human dendritic cell subtypes. Nat Rev Immunol 2002;2(3) 151-161.
  41. 41. Mittag D, Proietto AI, Loudovaris T et al. Human dendritic cell subsets from spleen and blood are similar in phenotype and function but modified by donor health status. J Immunol 2011;186(11) 6207-6217.
  42. 42. Henri S, Vremec D, Kamath A et al. The dendritic cell populations of mouse lymph nodes. J Immunol 2001;167(2) 741-748.
  43. 43. Anjuere F, Martin P, Ferrero I et al. Definition of dendritic cell subpopulations present in the spleen, Peyer's patches, lymph nodes, and skin of the mouse. Blood 1999;93(2) 590-598.
  44. 44. Vremec D, Pooley J, Hochrein H et al. CD4 and CD8 expression by dendritic cell subtypes in mouse thymus and spleen. J Immunol 2000;164(6) 2978-2986.
  45. 45. De Smedt T, Pajak B, Muraille E et al. Regulation of dendritic cell numbers and maturation by lipopolysaccharide in vivo. J Exp Med 1996;184(4) 1413-1424.
  46. 46. Reis e Sousa, Hieny S, Scharton-Kersten T et al. In vivo microbial stimulation induces rapid CD40 ligand-independent production of interleukin 12 by dendritic cells and their redistribution to T cell areas. J Exp Med 1997;186(11) 1819-1829.
  47. 47. Pulendran B, Smith JL, Caspary G et al. Distinct dendritic cell subsets differentially regulate the class of immune response in vivo. Proc Natl Acad Sci U S A 1999;96(3) 1036-1041.
  48. 48. Maldonado-Lopez R, De ST, Pajak B et al. Role of CD8alpha+ and CD8alpha- dendritic cells in the induction of primary immune responses in vivo. J Leukoc Biol 1999;66(2) 242-246.
  49. 49. Maldonado-Lopez R, De ST, Michel P et al. CD8alpha+ and CD8alpha- subclasses of dendritic cells direct the development of distinct T helper cells in vivo. J Exp Med 1999;189(3) 587-592.
  50. 50. Moser M and Murphy KM. Dendritic cell regulation of TH1-TH2 development. Nat Immunol 2000;1(3) 199-205.
  51. 51. Pooley JL, Heath WR, and Shortman K. Cutting edge: intravenous soluble antigen is presented to CD4 T cells by CD8- dendritic cells, but cross-presented to CD8 T cells by CD8+ dendritic cells. J Immunol 2001;166(9) 5327-5330.
  52. 52. den Haan JM, Lehar SM, and Bevan MJ. CD8(+) but not CD8(-) dendritic cells cross-prime cytotoxic T cells in vivo. J Exp Med 2000;192(12) 1685-1696.
  53. 53. Dzionek A, Fuchs A, Schmidt P et al. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood. J Immunol 2000;165(11) 6037-6046.
  54. 54. Van Brussel I, Berneman ZN, and Cools N. Optimizing dendritic cell-based immunotherapy: tackling the complexity of different arms of the immune system. Mediators Inflamm 2012;2012(690643-
  55. 55. Manthey HD and Zernecke A. Dendritic cells in atherosclerosis: functions in immune regulation and beyond. Thromb Haemost 2011;106(5) 772-778.
  56. 56. Becker L, Liu NC, Averill MM et al. Unique proteomic signatures distinguish macrophages and dendritic cells. PLoS One 2012;7(3) e33297-
  57. 57. Geissmann F, Gordon S, Hume DA et al. Unravelling mononuclear phagocyte heterogeneity. Nat Rev Immunol 2010;10(6) 453-460.
  58. 58. Schulz O, Jaensson E, Persson EK et al. Intestinal CD103+, but not CX3CR1+, antigen sampling cells migrate in lymph and serve classical dendritic cell functions. J Exp Med 2009;206(13) 3101-3114.
  59. 59. Hume DA. Differentiation and heterogeneity in the mononuclear phagocyte system. Mucosal Immunol 2008;1(6) 432-441.
  60. 60. Hume DA. Probability in transcriptional regulation and its implications for leukocyte differentiation and inducible gene expression. Blood 2000;96(7) 2323-2328.
  61. 61. Hume DA. Macrophages as APC and the dendritic cell myth. J Immunol 2008;181(9) 5829-5835.
  62. 62. Keshav S, Chung P, Milon G et al. Lysozyme is an inducible marker of macrophage activation in murine tissues as demonstrated by in situ hybridization. J Exp Med 1991;174(5) 1049-1058.
  63. 63. Steinman RM. Dendritic cells: versatile controllers of the immune system. Nat Med 2007;13(10) 1155-1159.
  64. 64. Naik SH, Metcalf D, van NA et al. Intrasplenic steady-state dendritic cell precursors that are distinct from monocytes. Nat Immunol 2006;7(6) 663-671.
  65. 65. Varol C, Landsman L, Fogg DK et al. Monocytes give rise to mucosal, but not splenic, conventional dendritic cells. J Exp Med 2007;204(1) 171-180.
  66. 66. Jakubzick C, Bogunovic M, Bonito AJ et al. Lymph-migrating, tissue-derived dendritic cells are minor constituents within steady-state lymph nodes. J Exp Med 2008;205(12) 2839-2850.
  67. 67. Jakubzick C, Tacke F, Ginhoux F et al. Blood monocyte subsets differentially give rise to CD103+ and CD103- pulmonary dendritic cell populations. J Immunol 2008;180(5) 3019-3027.
  68. 68. Gonzalez-Juarrero M, Shim TS, Kipnis A et al. Dynamics of macrophage cell populations during murine pulmonary tuberculosis. J Immunol 2003;171(6) 3128-3135.
  69. 69. Onai N, Obata-Onai A, Schmid MA et al. Identification of clonogenic common Flt3+M-CSFR+ plasmacytoid and conventional dendritic cell progenitors in mouse bone marrow. Nat Immunol 2007;8(11) 1207-1216.
  70. 70. Fogg DK, Sibon C, Miled C et al. A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science 2006;311(5757) 83-87.
  71. 71. Naik SH, Sathe P, Park HY et al. Development of plasmacytoid and conventional dendritic cell subtypes from single precursor cells derived in vitro and in vivo. Nat Immunol 2007;8(11) 1217-1226.
  72. 72. Banchereau J and Steinman RM. Dendritic cells and the control of immunity. Nature 1998;392(6673) 245-252.
  73. 73. O'Doherty U, Peng M, Gezelter S et al. Human blood contains two subsets of dendritic cells, one immunologically mature and the other immature. Immunology 1994;82(3) 487-493.
  74. 74. Kissenpfennig A, Henri S, Dubois B et al. Dynamics and function of Langerhans cells in vivo: dermal dendritic cells colonize lymph node areas distinct from slower migrating Langerhans cells. Immunity 2005;22(5) 643-654.
  75. 75. Van Brussel I, Van Vré EA, De Meyer GR et al. Decreased numbers of peripheral blood dendritic cells in patients with coronary artery disease are associated with diminished plasma Flt3 ligand levels and impaired plasmacytoid dendritic cell function. Clin Sci (Lond) 2010;120(9) 415-426.
  76. 76. Van Brussel I, Van Vré EA, De Meyer GRY et al. Expression of dendritic cell markers CD11c/BDCA-1 and CD123/BDCA-2 in coronary artery disease upon activation in whole blood. J Immunol Methods 2010;362(1-2) 168-175.
  77. 77. Van Vré EA, Hoymans VY, Bult H et al. Decreased number of circulating plasmacytoid dendritic cells in patients with atherosclerotic coronary artery disease. Coron Artery Dis 2006;17(3) 243-248.
  78. 78. Van Vré EA, Van Brussel I, de Beeck KO et al. Changes in blood dendritic cell counts in relation to type of coronary artery disease and brachial endothelial cell function. Coron Artery Dis 2010;21(2) 87-96.
  79. 79. Yilmaz A, Weber J, Cicha I et al. Decrease in circulating myeloid dendritic cell precursors in coronary artery disease. J Am Coll Cardiol 2006;48(1) 70-80.
  80. 80. Yilmaz A, Schaller T, Cicha I et al. Predictive value of the decrease in circulating dendritic cell precursors in stable coronary artery disease. Clin Sci (Lond) 2009;116(4) 353-363.
  81. 81. Van Vré EA, Bosmans JM, Van Brussel I et al. Immunohistochemical characterisation of dendritic cells in human atherosclerotic lesions: possible pitfalls. Pathology 2011;43(3) 239-247.
  82. 82. Blom B, Ho S, Antonenko S et al. Generation of interferon alpha-producing predendritic cell (Pre-DC)2 from human CD34(+) hematopoietic stem cells. J Exp Med 2000;192(12) 1785-1796.
  83. 83. Chen W, Antonenko S, Sederstrom JM et al. Thrombopoietin cooperates with FLT3-ligand in the generation of plasmacytoid dendritic cell precursors from human hematopoietic progenitors. Blood 2004;103(7) 2547-2553.
  84. 84. Colonna M, Trinchieri G, and Liu YJ. Plasmacytoid dendritic cells in immunity. Nat Immunol 2004;5(12) 1219-1226.
  85. 85. Davenport P and Tipping PG. The role of interleukin-4 and interleukin-12 in the progression of atherosclerosis in apolipoprotein E-deficient mice. Am J Pathol 2003;163(3) 1117-1125.
  86. 86. Lee TS, Yen HC, Pan CC et al. The role of interleukin 12 in the development of atherosclerosis in ApoE-deficient mice. Arterioscler Thromb Vasc Biol 1999;19(3) 734-742.
  87. 87. Paulson KE, Zhu SN, Chen M et al. Resident Intimal Dendritic Cells Accumulate Lipid and Contribute to the Initiation of Atherosclerosis. Circ Res 2010;106(2) 383-390.
  88. 88. Sato K, Niessner A, Kopecky SL et al. TRAIL-expressing T cells induce apoptosis of vascular smooth muscle cells in the atherosclerotic plaque. J Exp Med 2006;203(1) 239-250.
  89. 89. Marshak-Rothstein A, Busconi L, Rifkin IR et al. The stimulation of Toll-like receptors by nuclear antigens: a link between apoptosis and autoimmunity. Rheum Dis Clin North Am 2004;30(3) 559-74, ix.
  90. 90. Niessner A, Sato K, Chaikof EL et al. Pathogen-sensing plasmacytoid dendritic cells stimulate cytotoxic T-cell function in the atherosclerotic plaque through interferon-alpha. Circulation 2006;114(23) 2482-2489.
  91. 91. Döring Y, Manthey HD, Drechsler M et al. Auto-antigenic protein-DNA complexes stimulate plasmacytoid dendritic cells to promote atherosclerosis. Circulation 2012;125(13) 1673-1683.
  92. 92. Daissormont IT, Christ A, Temmerman L et al. Plasmacytoid Dendritic Cells Protect Against Atherosclerosis by Tuning T-Cell Proliferation and Activity. Circ Res 2011;
  93. 93. de Jager SC and Kuiper J. Vaccination strategies in atherosclerosis. Thromb Haemost 2011;106(5) 796-803.
  94. 94. Steinman RM. Dendritic cells in vivo: a key target for a new vaccine science. Immunity 2008;29(3) 319-324.
  95. 95. Bobryshev YV. Can dendritic cells be exploited for therapeutic intervention in atherosclerosis? Atherosclerosis 2001;154(2) 511-512.
  96. 96. Timmerman JM and Levy R. Dendritic cell vaccines for cancer immunotherapy. Annu Rev Med 1999;50(507-529.
  97. 97. Palucka AK, Ueno H, Fay JW et al. Taming cancer by inducing immunity via dendritic cells. Immunol Rev 2007;220(129-150.
  98. 98. Markiewicz MA and Kast WM. Progress in the development of immunotherapy of cancer using ex vivo-generated dendritic cells expressing multiple tumor antigen epitopes. Cancer Invest 2004;22(3) 417-434.
  99. 99. Davis ID, Jefford M, Parente P et al. Rational approaches to human cancer immunotherapy. J Leukoc Biol 2003;73(1) 3-29.
  100. 100. Lu W, Arraes LC, Ferreira WT et al. Therapeutic dendritic-cell vaccine for chronic HIV-1 infection. Nat Med 2004;10(12) 1359-1365.
  101. 101. Garcia F, Lejeune M, Climent N et al. Therapeutic immunization with dendritic cells loaded with heat-inactivated autologous HIV-1 in patients with chronic HIV-1 infection. J Infect Dis 2005;191(10) 1680-1685.
  102. 102. Palinski W, Miller E, and Witztum JL. Immunization of low density lipoprotein (LDL) receptor-deficient rabbits with homologous malondialdehyde-modified LDL reduces atherogenesis. Proc Natl Acad Sci U S A 1995;92(3) 821-825.
  103. 103. Freigang S, Horkko S, Miller E et al. Immunization of LDL receptor-deficient mice with homologous malondialdehyde-modified and native LDL reduces progression of atherosclerosis by mechanisms other than induction of high titers of antibodies to oxidative neoepitopes. Arterioscler Thromb Vasc Biol 1998;18(12) 1972-1982.
  104. 104. Zhou X, Caligiuri G, Hamsten A et al. LDL immunization induces T-cell-dependent antibody formation and protection against atherosclerosis. Arterioscler Thromb Vasc Biol 2001;21(1) 108-114.
  105. 105. Catanzaro DF, Zhou Y, Chen R et al. Potentially reduced exposure cigarettes accelerate atherosclerosis: evidence for the role of nicotine. Cardiovasc Toxicol 2007;7(3) 192-201.
  106. 106. Nilsson J, Nordin FG, Schiopu A et al. Oxidized LDL antibodies in treatment and risk assessment of atherosclerosis and associated cardiovascular disease. Curr Pharm Des 2007;13(10) 1021-1030.
  107. 107. Ameli S, Hultgardh-Nilsson A, Regnstrom J et al. Effect of immunization with homologous LDL and oxidized LDL on early atherosclerosis in hypercholesterolemic rabbits. Arterioscler Thromb Vasc Biol 1996;16(8) 1074-1079.
  108. 108. Bobryshev YV. Dendritic cells and their role in atherogenesis. Lab Invest 2010;90(7) 970-984.
  109. 109. Habets KL, van Puijvelde GH, van Duivenvoorde LM et al. Vaccination using oxidized low-density lipoprotein-pulsed dendritic cells reduces atherosclerosis in LDL receptor-deficient mice. Cardiovasc Res 2010;85(3) 622-630.
  110. 110. Hermansson A, Johansson DK, Ketelhuth DF et al. Immunotherapy with tolerogenic apolipoprotein B-100-loaded dendritic cells attenuates atherosclerosis in hypercholesterolemic mice. Circulation 2011;123(10) 1083-1091.
  111. 111. Hjerpe C, Johansson D, Hermansson A et al. Dendritic cells pulsed with malondialdehyde modified low density lipoprotein aggravate atherosclerosis in Apoe(-/-) mice. Atherosclerosis 2010;209(2) 436-441.
  112. 112. Hill JA, Ichim TE, Kusznieruk KP et al. Immune modulation by silencing IL-12 production in dendritic cells using small interfering RNA. J Immunol 2003;171(2) 691-696.
  113. 113. Takeda M, Yamashita T, Sasaki N et al. Oral administration of an active form of vitamin D3 (calcitriol) decreases atherosclerosis in mice by inducing regulatory T cells and immature dendritic cells with tolerogenic functions. Arterioscler Thromb Vasc Biol 2010;30(12) 2495-2503.
  114. 114. Hussain M, Javeed A, Ashraf M et al. Aspirin may do wonders by the induction of immunological self-tolerance against autoimmune atherosclerosis. Med Hypotheses 2012;78(1) 171-173.
  115. 115. Buckland M, Jago CB, Fazekasova H et al. Aspirin-treated human DCs up-regulate ILT-3 and induce hyporesponsiveness and regulatory activity in responder T cells. Am J Transplant 2006;6(9) 2046-2059.
  116. 116. Buckland M, Jago C, Fazekesova H et al. Aspirin modified dendritic cells are potent inducers of allo-specific regulatory T-cells. Int Immunopharmacol 2006;6(13-14) 1895-1901.
  117. 117. Benko S, Magyarics Z, Szabo A et al. Dendritic cell subtypes as primary targets of vaccines: the emerging role and cross-talk of pattern recognition receptors. Biol Chem 2008;389(5) 469-485.
  118. 118. Dubsky P, Ueno H, Piqueras B et al. Human dendritic cell subsets for vaccination. J Clin Immunol 2005;25(6) 551-572.
  119. 119. van Es T, van Puijvelde GH, Foks AC et al. Vaccination against Foxp3(+) regulatory T cells aggravates atherosclerosis. Atherosclerosis 2010;209(1) 74-80.

Written By

Ilse Van Brussel, Hidde Bult, Wim Martinet, Guido R.Y. De Meyer and Dorien M. Schrijvers

Submitted: 24 August 2012 Published: 27 February 2013