Introduction
Atherosclerosis, a chronic inflammatory disease of the arterial wall, is the primary pathology underlying cardiovascular disease (CVD), including stroke and myocardial infarction, which are major causes of disability and mortality in industrialized countries [
1]. The initiation and progression of atherosclerosis involves a complex interplay of various cell types and signaling pathways, leading to plaque formation and subsequent narrowing of the arteries [
2,
3,
4]. Activation of the innate immune pathway, mediated by concerted action of pattern recognition receptors (PRRs), is central in all stages of atherosclerosis [
3]. These receptors are expressed on both immune and non-immune cells in the arterial wall, such as macrophages, dendritic cells, endothelial cells, and smooth muscle cells. They recognize specific molecular patterns associated with pathogens (PAMPs) and danger signals from damaged or infected cells (DAMPs) [
5,
6,
7]. Among the well-known PRRs involved in atherosclerosis are the Toll-like receptors (TLRs), which can recognize various ligands, including lipopolysaccharides (LPS) from gram-negative bacteria, as well as endogenous ligands like oxidized low-density lipoprotein (oxLDL) and heat shock proteins (HSPs) [
8,
9,
10]. TLR signaling leads to the production of pro-inflammatory cytokines, chemokines, and adhesion molecules, promoting the recruitment and activation of immune cells in the arterial wall [
11,
12]. A pivotal inflammatory pathway in atherosclerosis downstream of TLR signaling is the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome pathway. NLRP3 is activated by various DAMPs, such as cholesterol crystals, oxLDL, and HSPs. Upon activation, NLRP3 triggers the release of pro-inflammatory cytokines, including interleukin-1β (IL-1β) and IL-18, which further contribute to atherogenesis by inducing the expression of adhesion molecules and chemokines in endothelial cells and enhancing the recruitment and activation of immune cells, such as macrophages [
13,
14,
15].
In addition to TLRs and NOD-like receptors (NLRs), other PRRs, including scavenger receptors and c-type lectin receptors, have been implicated in atherosclerosis [
16,
17,
18]. These receptors recognize a variety of ligands, including modified lipids, glycated proteins, and viral RNA, and activate innate immune signaling pathways that contribute to the pathogenesis of atherosclerosis. Scavenger receptors are critical in foam cell formation and the ensuing inflammatory response. They are involved in the uptake and transport of modified lipoproteins across the atherogenic vascular endothelium and their processing by macrophages, playing a central role in both atherogenesis and atheroprogression [
17]. C-type lectin receptor-mediated signaling pathways that control immune responses have been reviewed in [
19]. In atherogenesis, the collective involvement of innate immune pathways is crucial as they play a key role in promoting the recruitment and activation of immune cells in the arterial wall. Additionally, these pathways induce the production of pro-inflammatory cytokines and chemokines highlighting their potential for therapeutic approaches to curb vascular inflammation. Hence, a deeper understanding of their precise molecular mechanisms could pave the way for innovative immunotherapeutic interventions targeting atherosclerosis. In this Review, we summarize cellular immune responses in atherosclerosis and discuss innate immune pathways driving atherogenesis and atheroprogression focusing on TLR and NLR-dependent mechanisms and potential crosstalk with scavenger receptors. We also highlight how these pathways contribute to long-term reprogramming of the innate immune system in atherosclerosis.
Cellular innate immune responses in atherosclerosis
In response to inflammation, immune cells are rapidly recruited to the site of injury or infection [
20]. This involves a sequential crosstalk between integrins and endothelial adhesion molecules, facilitating tethering and rolling, firm adhesion, and transendothelial migration under laminar shear stress [
21]. The extent of inflammation determines which subsets of immune cells are recruited to the inflamed tissue. During acute inflammation, granulocytes like neutrophils and eosinophils are quickly recruited to neutralize and eliminate potentially harmful agents. Later, mononuclear cells, including lymphocytes and monocytes/macrophages, accumulate to aid in tissue resolution and repair [
22]. However, failure to resolve the acute inflammatory condition or continuous exposure to inflammatory stimuli can lead to chronic inflammation [
23]. At various stages of atherosclerosis, specific subsets of immune cells with either pro-atherogenic or atheroprotective properties accumulate. Among these cells, macrophages are the most abundant in atherosclerotic plaques, originating from circulating monocytes infiltrating the plaque or proliferation of lineage-committed resident macrophages [
24,
25]. These macrophages constitute a diverse and heterogeneous population, with distinct origins and varying effects on atherogenesis and atheroprogression. Advances in technology, such as genetic fate mapping, cytometry by time of flight (CyTOF), and single-cell RNA sequencing (scRNA-seq), have revealed the complexity of these macrophages and their roles in atherosclerosis [
26,
27,
28,
29,
30]. Macrophages play a critical role in the initiation and progression of atherosclerosis by taking up oxLDL and releasing pro-inflammatory cytokines, which sustain the ongoing inflammation. Their activation is influenced by various stimuli, such as pathogens, tissue damage, and inflammatory cytokines, and their phenotype can adapt depending on the local microenvironment [
31]. The specific contributions of immune cells to the initiation and progression of atherosclerosis have been extensively discussed in a recent review [
3].
Macrophage heterogeneity as revealed by emerging techniques
Recent emergence of new technologies including genetic lineage tracing and fate mapping, CyTOF, and scRNA-seq have been instrumental in our understanding of macrophage origin, heterogeneity, and function. By analyzing single cells using these techniques, distinct subsets of aortic macrophages have been identified in the mouse [
40,
41]. The main macrophage population implicated in atherosclerosis, and not found in healthy arteries, is the circulating monocyte-derived inflammatory macrophage subset, which are characterized by increased expression of inflammasome components and transcripts of cytokines and chemokines [
40,
42]. A pro-inflammatory type I interferon (IFN)-inducible macrophages, also derived from monocytes, express high levels of the IFN-inducible genes Ifit3, Irf7 and Isg15 [
43,
44]. A subset of macrophages identified during initiation and progression of atherosclerosis including advanced atherosclerotic plaques are the TREM2-expressing cells, which are derived from either circulating monocytes or embryonic precursors [
40,
41]. TREM2hi macrophages might have protective effects in vascular inflammation and resemble CD11c–YFP+ macrophages [
45]. A recent study identified a subset of macrophages residing in the intima of mouse aorta, the aortic intima-resident macrophages (MacAIR), with pro-atherosclerotic effects and express IL-lβ mRNA [
24]. These macrophages are differentiated from circulating monocytes recruited into the intima and are maintained in the aorta by local proliferation. Embryonically derived CX3CR1+ precursors are present in both atherosclerotic and healthy arteries [
46]. They proliferate in the adventitia, express lymphatic vessel endothelial hyaluronan receptor 1 (LYVE1), MHC class II, and macrophage mannose receptor C type 1 (MRC1) and limit arterial stiffness by inhibiting collagen production [
47]. Collectively, the utilization of single-cell analyses and lineage tracing have yielded valuable insights into the diverse characteristics of plaque macrophages, surpassing the limitations of conventional immunophenotyping. Nevertheless, achieving a comprehensive understanding of the contribution of each subset of aortic macrophages to the progression of atherosclerosis will require determining their specific roles in functional studies. Furthermore, it is imperative to investigate thoroughly their interaction with other immune cells, such as neutrophils and dendritic cells as this will be instructive in the design of targeting strategies with minimal off-target effects.
Regardless of their phenotypic differences, tissue macrophages function to maintain or restore homeostasis after tissue damage [
48]. They navigate through a complex network of chemoattractants, including chemokines, to migrate within tissues. Cellular signaling involving PI3-kinase and Rho family of GTPases regulates macrophage migration and chemotaxis and has been extensively reviewed [
49]. The microenvironment in the atherosclerotic lesion presents challenges to both resident and emigrated macrophages. Cytokine milieu and factors like modified LDL can influence macrophage dynamics, inhibiting their migration and leading to their retention in the atherosclerotic lesion [
50]. The microenvironment in the atherosclerotic lesion poses many challenges to the resident and the emigrated macrophages. Besides the cytokine milieu that can skew macrophages from one state to another, there are factors like modified LDL which, when taken up by macrophages, can inhibit their migration potential leading to their retention in the atherosclerotic lesion. Of note, neutrophils release reactive oxygen species (ROS) that contribute to lipid modification, while macrophages ingest modified lipids through scavenger receptors, transforming into foam cells as a critical event in plaque formation. Foam cells secrete inflammatory chemokines and cytokines, perpetuating inflammation. Excessive lipid uptake induces macrophage proliferation, contributing to their accumulation during advanced stages of atherosclerosis [
25]. Continued scavenger receptor-mediated lipid uptake leads to the generation and intracellular accumulation of misfolded proteins, ultimately resulting in cell death. Efficient clearance of apoptotic cells ("efferocytosis") is necessary for tissue homeostasis, and defective clearance leads to chronic inflammation. In advanced atherosclerotic lesions, cell death and defective efferocytosis contribute to the expansion of the necrotic core, potentially triggering plaque rupture and acute thrombosis [
51].
TLR and NLR signaling in arterial inflammation
The innate immune signaling field has been completely revolutionized by the discovery of the TLRs in the mid-1990s, an extremely important finding that was awarded the 2011 Nobel Prize in Physiology or Medicine. TLRs are a family of PRRs that play a key role in the innate immune system. They recognize conserved PAMPs from foreign pathogens or DAMPs from damaged tissue [
101] . Currently, 13 TLRs have been discovered in mammals, 10 in humans (TLR1-10) and 12 in mice (TLR1-9,11,12,13) [
12]. All TLRs are classified as type I transmembrane receptors and share a conserved structure including an extracellular leucine-rich region that mediates the recognition of PAMPs, transmembrane domains, and cytoplasmic Toll-interleukin 1 receptor (TIR) domains essential for signal transduction [
102] . The specificity of the downstream response depends on which TLR activated but also on the specific PAMP including lipids, nucleic acids, and proteins recognized by the receptors [
103]. TLR4 represents a peculiar example among the group since it can recognize exogenous but also endogenous ligands with very different structures and functions such as LPS, oxLDL, the plant diterpene paclitaxel, the fusion protein of respiratory syncytial virus (RSV), fibronectin and many others [
12,
103]. Upon PAMPs binding, TLRs homo- or heterodimerize leading to the activation of a MyD88 (myeloid differentiation primary response protein 88)-dependent pathway or a TRIF (TIR-domain-containing adaptor protein inducing IFNβ)-dependent pathway. Both MyD88 and TRIF pathways lead to the activation of the E3 ubiquitin ligase TNF receptor-associated factor 6 (TRAF6), which in turn recruits and activates the TGFβ-activated protein kinase 1 (TAK1; also known as MAP3K7) complex [
104,
105] (
Figure 2). On one hand, active TAK1 triggers the phosphorylation of the canonical IKK complex, ultimately leading to NF-kB (nuclear factor-κB) activation; on the other hand, TAK1 is also the starting point of the MAPK cascade activation. Specifically, TAK1 simultaneously phosphorylates MAPK kinases 3/6 (MKK p38 and JNKs respectively. The third member of the MAPK family, extracellular signal−regulated protein kinases 1 and 2 (ERK1/2), is activated by IKKβ, which activates MKK1/2 upstream of ERK1/2. This cascade of events leads to the phosphorylation and subsequent nuclear translocation of several transcription factors including cyclic AMP-responsive element-binding protein (CREB) and activator protein 1 (AP-1) that play an important role in the expression of cytokines, chemokines, and interferons.
Among the TLRs, TLR4 stands out as the founding member of the TLR family identified as the first human homologue of the
Drosophila Toll by Medzhitov et al. in 1997 [
106]. This receptor is majorly known to be the binding partner of LPS, a membrane component of Gram-negative bacteria [
102]. Its peculiarity is that it signals via both MyD88- and TRIF-dependent pathways. First, it recruits MyD88 to trigger the initial activation of NF-kB and MAPK. Subsequently, TLR4 is endocytosed and transported to the endosome, where it initiates the TRIF pathway that leads to the late activation of NF-kB and MAPK. Downstream consequences of these events include the expression of pro-inflammatory cytokines and chemokines such as IL-1, IL-6, TNFα, IL-8, and adhesion molecules like E-selectin, VCAM-1, and ICAM-1. Due to its involvement in innate immunity and inflammation, TLR4 has been described as a crucial mediator of atherosclerosis. In 2002, Edfeldt et al. showed that the expression of TLR4, as well as TLR1 and TLR2, is markedly augmented in human atherosclerotic lesions [
107]. Moreover,
Tlr4 deficiency attenuated aortic atherosclerosis, plaque lipid content, and macrophage burden when compared to
ApoE-/- controls[
108]. TLR4 has been demonstrated to play a role in both early and late stages of atherosclerosis [
109]. It is well established that in the early phase of the disease, endothelial cells are activated and this process is characterized by the expression of adhesion molecules. The upregulation of ICAM-1, VCAM, ELAM-1, but also IL-6 and IL-8 in human coronary artery endothelial cells depends on the LPS-TLR4 axis [
110]. Activated TLR4 plays a central role also in macrophages by inducing the expression of inflammatory cytokines and proteases and it contributes to early-stage intimal foam cell accumulation at lesion-prone aortic arch in
ApoE−/− TLR4−/− mice compared to
ApoE−/− controls [
111,
112].
Since TLRs are located at the cell surface or in endosomes, our body has developed another defense system able to recognize pathogens that have invaded the cytosol, the cytoplasmic PRRs. This family includes the RIG-I-like receptors (RLRs) and NLRs [
102,
113]. The structure of the NLRs includes three domains, a C-terminal leucine-rich repeat (LRR) domain, an intermediate nucleotide binding and oligomerization domain (NOD, also called NACHT domain), an N-terminal pyrin (PYD), and a caspase activation and recruitment domain (CARD) [
114]. NLRs consist of 22 members in humans which are known to initiate different pro-inflammatory responses downstream of the recognition of PAMPs and DAMPs. The best-known NLRs are the ones leading to inflammasome formation. Inflammasomes are multiprotein complexes containing a sensor that recruits an adaptor molecule to activate the effector caspase-1, which catalyzes the maturation of the pro-inflammatory cytokines pro-IL-1β and pro-IL-18 [
115]. Ten sensors have been discovered so far namely NLRP1, NLRP3, NLRC4, AIM2, pyrin, NLRP2, NLRP6, NLRP7, NLRP11, and NLRP12 [
113,
116]. These receptors become activated by very different molecules. For example, NLRP1 recognizes anthrax lethal factor released by the Gram-positive bacterium
Bacillus anthracis, NLRC4 is activated by NAIPs (NOD-like receptor family apoptosis inhibitory proteins), which bind to flagellin or constituents of the Type III secretion system of bacteria, AIM2 detects cytosolic dsDNA, and pyrin identifies bacterial toxin-induced modifications of Rho GTPases [
117]. In this regard, NLRP3 is exceptional since it is activated by a broad variety of stimuli related to bacterial, viral, and fungal infections, as well as inflammatory endogenous DAMPs [
118]. Upon recognition of PAMPs or DAMPs, all inflammasomes share a common activation process where they require a sensor, the adaptor ASC (apoptosis-associated speck-like protein containing a CARD), and the effector protein caspase-1 (
Figure 2). In particular, the sensor forms oligomers that recruit the common adaptor protein ASC, which in turn binds pro-caspase-1 [
118]. This binding enables proximity-driven autocatalytic caspase-1 maturation and cleaved caspase-1 forms an active heterotetramer able to process pro-IL-1β and pro-IL-18. The release of the mature IL-1β and IL-18 exacerbates a strong pro-inflammatory response [
114,
116]. Mature caspase-1 also mediates the cleavage of GSDMD (gasdermin D), a protein with a pore-forming activity that permeabilizes the membrane leading to pyroptosis, an inflammasome-induced cell death [
119]. It has also been demonstrated that GSDMD-dependent pyroptosis enables the release of IL-1β and IL-18, with GSDMD pores representing channels for the secretion of these molecules [
120].
NLRP3 inflammasome activation in the pathogenesis of atherosclerosis
Among the inflammasomes, NLRP3 and AIM2 stand out as key drivers of atherosclerosis with NLRP3 being the most extensively studied [
121,
122]. NLRP3 inflammasome is activated via two distinct steps
– the priming and the activation [
118]. The priming phase is triggered upon the recognition of PAMPs or DAMPs via receptors such as TLRs, NOD2 (nucleotide-binding oligomerization domain 2), or through the recognition of TNFα. This first step leads to the NF-kB-driven transcriptional upregulation of NLRP3 itself, pro-caspase-1 and pro-IL-1β, and posttranslational modification of NLRP3. The activation step requires the oligomerization of NLRP3, which together with ASC, pro-caspase 1, and NIMA-related kinase 7 (NEK7) assemble as the inflammasome. Accumulating data indicate the requirement of IKKβ for the rapid formation of NLRP3 inflammasome and subsequent induction of pro-inflammatory responses [
123,
124,
125].
Inflammation drives atherosclerosis development and progression, and the crucial proof comes from the CANTOS (Canakinumab Anti-Inflammatory Thrombosis Outcomes Study) trial, which demonstrated that targeting IL-1β with the neutralizing antibody canakinumab leads to a significant decrease of cardiovascular events compared to patients treated with placebo [
126]. Accordingly, NLRP3 involvement in atherosclerosis has been extensively investigated for its prominent role in mediating the release of both IL-1β and IL-18, which contribute to exacerbating vascular inflammation. Paramel Varghese et al. demonstrated that NLRP3 mRNA together with caspase 1, ASC, IL-1β and IL-18 are significantly increased in human atherosclerotic plaques compared to healthy arteries [
127]. Interestingly, they also observed enhanced IL-1β protein release from freshly isolated human carotid plaques upon activation of TLR4 with LPS as a first signal and ATP as a second hit. Therefore, they provided strong evidence of the importance of TLR4-mediated NLRP3 inflammasome activation in atherosclerosis. The first evidence regarding the contribution of NLRP3 in diet-induced atherosclerosis in a mouse model was from bone marrow transplantations of
Ldlr−/− mice with bone marrow-derived from either wild-type,
Nlrp3−/−, Asc−/−, or
Il-1α−/−/Il-1β −/− mice [
121]. The deficiency of these single proteins in the bone marrow significantly reduced the development of atherosclerotic lesions compared to
Ldlr−/− mice transplanted with wild-type bone marrow. Recently, Christ et al. showed that
Ldlr−/−/Nlrp3−/− double knockout mice had significantly reduced atherosclerotic plaque sizes after 8 weeks of western type diet feeding compared to
Ldlr−/− mice [
122]. However, contrasting findings have been reported for the role of NLRP3 in atherosclerosis and it is important to acknowledge the remarkable influence of factors such as gender, age, specific diet, duration of atherogenic diet feeding, and environmental conditions on the phenotype of NLRP3-deficient mice [
15,
116]. These should be accounted for when designing therapeutic strategies to inhibit NLRP3 for atheroprotection.
Although therapeutic blocking of IL-1 signaling has proven to be beneficial, it comes at the expense of high rate of infections [
126]. Therefore, there is the need for more specific targeting strategies including inhibition of NLRP3. The most specific and potent NLRP3 inhibitor known to date is the diarylsulfonylurea compound MCC950, able to specifically inhibit NLRP3 activation in isolated mouse and human macrophages [
128]. Moreover, it does not affect the function of other inflammasomes. Importantly, this compound has been demonstrated to be therapeutically effective against several preclinical models including myocardial infarction [
129] and atherosclerosis [
130]. MCC950 inhibits IL-1β release in bone marrow-derived macrophages and dendritic cells stimulated with LPS and cholesterol crystals. Whereas
in vivo, it reduces atherosclerotic lesion development and the invasion of macrophages in the carotid artery plaques of
ApoE-/- mice [
130]. Together, these data reveal the critical role of the TLR-NLRP3 axis in the context of innate immune signaling and its involvement in several inflammation-related diseases such as atherosclerosis.
Crosstalk between Scavenger receptors and TLRs in chronic vascular inflammation
The crosstalk and synergy between different classes of innate pattern recognition receptors are essential for an effectively coordinated inflammatory response and host defense. While significant progress has been made in understanding the individual functions of these receptors, there is still limited knowledge regarding their collaborative interactions within the innate immune system. The growing body of evidence strongly suggests that SRs cooperate with TLRs as signaling partners. This cooperative network extends across various SRs, such as SR-A1, MARCO, CD36, and SCARF1, which engage with specific TLRs to modulate immune reactions. For example, it has been shown that SR-A1 and TLR4 cooperate in the phagocytosis of Escherichia coli, while SR-A1 and TLR2 cooperate to augment the phagocytosis of S. aureus. Additionally, by inducing the internalization of pathogens, SR-A1 promotes the inflammatory response mediated by endosomal TLRs, like TLR3. Moreover, it has been reported that MARCO interacts with TLR2 and CD14 to recognize Mycobacterium tuberculosis.
Similarly, CD-36 by coupling to TLR4/6 heterodimer can trigger a sterile inflammatory response, as it induces expression of IL- 1β and TNFα through NF-κB activation when exposed to modified LDL [
131]. Moreover, it has been demonstrated that SCARF1 promoted LPS-TLR4-mediated signal transduction through NF-κB and MAP kinase pathways, leading to enhanced inflammatory cytokine release, such as IL-6, TNFα, and IFN-β [
132]. In the presence of LPS, it has been observed that SCARF1 induces TLR4 translocation to lipid microdomains on the cell surface. This translocation facilitates the initiation of signaling events. Subsequently, the complex formed by LPS, TLR4, and SCARF1 is internalized into intracellular endosomes [
133]. In addition, it was shown that TLR3 can interact with SCARF1 in the presence of TLR3 ligand. Specifically, upon treatment with PIC (poly I:C, dsRNA), TLR3 and SCARF1 were found to colocalize within endosomes in THP-1 monocytes. As a result, the formation of the SCARF1-TLR3-PIC complex led to higher activation of the NF-κB pathway, as well as increased phosphorylation of MAP kinases p38 and JNK. Additionally, this complex resulted in enhanced secretion of pro-inflammatory cytokines, such as IL-6 and IL-8. Thus, TLR3 acts as a coreceptor for SCARF1 and enhances its PIC-induced activation [
133].
Interestingly, it has been demonstrated that SR-A and MARCO compete with cell-surface TLR4 for ligand recognition, limiting its inflammatory response, but strikingly increase responses by intracellular pathogen sensors TLR3, NALP3 (NACHT domain-, leucine-rich repeat-, and pyrin domain-containing protein 3), and NOD2. SR-A/MARCO regulates this pathway by directly interacting with the TRAF-C domain of TRAF6, thereby preventing its dimerization or ubiquitylation, which are typically required for TLR4- NF-κB-mediated activation. SR-A/MARCO-induced rapid ligand internalization prevented detection by surface TLRs whilst enhancing ligand availability in intracellular compartments, thus enabling effective sensing and robust immune responses by intracellular sensors [
134]. These interactions between SRs and TLRs impact not only pathogen recognition and phagocytosis but also intracellular signal transduction and cytokine release, thus orchestrating a finely tuned immune response. As our understanding continues to evolve, unraveling the intricate interplay of these receptors promises to offer new avenues for therapeutic interventions and strategies for immune regulation.
Reprogramming of immune cells in atherosclerosis
It has been established that the innate immune system can develop a prolonged pro-inflammatory phenotype whereby innate immune cells including monocytes and macrophages acquire memory characteristics driven by epigenetic and metabolic reprogramming. This phenomenon is termed trained immunity and it is distinct from the adaptive immunity largely due to lack of specificity, whereas it can provide protection against similar but also unrelated stimuli [
135]. The growing body of evidence linking trained immunity to atherosclerosis highlights the potential of utilizing the concept of trained immunity for promising pharmacological interventions aimed at averting atheroprogression. The induction of trained immunity can be achieved through brief stimulation with β-glucan and Bacille Calmette-Guérin (BCG) vaccination for instance, or with endogenous atherogenic stimuli such as oxLDL and lipoprotein(a) [
136,
137,
138,
139]. This initial stimulation is then followed by a subsequent challenge with TLR agonists, resulting in augmented pro-inflammatory cytokine profile, enhanced foam cell formation, increased expression of scavenger receptors and decreased expression of cholesterol efflux transporters [
140].
In the atherosclerotic lesion, the innate immune cells possess the capability to modify their chromatin structure by both histone methylation and acetylation. These epigenetic modifications enhance the accessibility of transcription factors regulating immune-related genes and ultimately govern the persistent pro-inflammatory phenotype [
141]. At the level of histone methylation, trained human monocytes by oxLDL showed enrichment of H3K4me3 on the promoter regions of pro-atherogenic genes compared to untrained controls [
138]. Additionally, isolated monocytes of patients with symptomatic atherosclerosis exhibited increased pro-inflammatory cytokine production upon ex vivo LPS stimulation compared to monocytes derived from asymptomatic atherosclerotic patients [
142]. This phenotype was associated with epigenetic rewiring specifically involving histone methylation. The H3K4me3 mark is also implicated in promoting foam cell formation, which is evident through the increased expression of scavenger receptors and decreased expression of ATP-binding cassette transporters involved in cholesterol efflux in oxLDL-trained monocytes [
138]. It has been shown that training of human monocytes by oxLDL relies on histone methylation, whereby the training can be completely abolished by employing methylthioadenosine, a pharmacological blocker of histone methyltransferases [
138]. Another study demonstrated that statins – a lipid-lowering therapy – can prevent the in vitro training induced by either oxLDL or β-glucan, resulting in downregulation of cytokine production and epigenetic reprogramming [
143]. However, a three-month statin treatment in familial hypercholesterolemia patients had no effect on epigenetic reprogramming and did not revert trained immunity [
144].
Another significant epigenetic alteration associated with trained immunity involves histone acetylation carried out by histone acetyltransferases. The activation of innate immune cells coincides with the addition of H3K27ac to distal regulatory regions, marking active promoters and enhancers [
145]. Additionally, it has been observed that the distal regulatory elements which acquire H3K27ac, typically also gain H3K4me1 mark. However, the loss of H3K27ac does not generally correlate with the loss of H3K4me1, indicating that H3K4me1 has an epigenetic memory function [
145]. As in the case of histone methylation, there are significant changes in histone acetylation between healthy vessels and atherosclerotic lesions. The analysis revealed an enrichment of acetylation at 2 positions -H3K9 and H3K27- in macrophages, and these significant changes were increased during the progression of atherosclerosis (early versus advanced stages) [
146]. Upon BCG vaccination, genes that are directly involved in inflammation and cytokine production were found to exhibit heightened levels of H3K27ac. Intriguingly, one of those genes is OLR1, which encodes the receptor of oxLDL and is considered a marker of atherosclerosis [
147].
It is well established that chromatin remodeling through histone modifications plays a pivotal role in regulating the polarization of macrophages towards M1 pro-inflammatory and M2 anti-inflammatory sub-types [
148,
149]. Studies have been performed to examine the impact of histone modifications on macrophage polarization to understand their role in controlling the expression of inflammatory genes. For example, the levels of H3K4me3 mark were elevated in M1 macrophages, particularly on the promoter regions of pro-inflammatory genes, when compared to M2 and naive macrophages [
150]. Another example is the enrichment of H3K4me1 mark on the IL-1β promoter that leads to enhanced production of IL-1β driving atherogenesis [
151]. Conversely, H3K4me1 together with H3K27ac were found to be enhanced by mediator 1 on M2 marker genes, exerting suppressive effects on the development of atherosclerosis [
152,
153]. However, in the context of trained immunity, an analysis of M1 pro-inflammatory and M2 anti-inflammatory markers in oxLDL-induced trained macrophages displayed no significant differences[
138]. Similarly, with β-glucan-trained monocytes, the activation markers for both M1 and M2 macrophages were elevated without a significant difference [
136]. This demonstrates that β-glucan did not skew macrophages towards a particular subtype, but rather likely triggered a global pan cell activation.
The effects of the long-lasting epigenetic reprogramming were reinforced by the recent groundbreaking study highlighting the necessity of the TLR-NLR axis for immune reprogramming under atherogenic conditions. Specifically, when
Ldlr-/- mice were exposed to a western diet, a broad systemic inflammation emerged; nonetheless, this inflammation became undetectable in the serum shortly after switching to chow diet. On the transcriptional level, the alterations in gene expression and epigenetic modifications within myeloid progenitor cells, induced by the western diet, led to an exacerbation in their rate of proliferation and an enhancement of their innate immune reactions. This demonstrates that although western diet provoked transient systemic inflammation, it resulted in enduring modifications to myeloid cell responses. Importantly, all these effects were found to be dependent on the NLRP3/IL-1β pathway, which has emerged as a central pathway in chronic vascular inflammation [
122].