Skip to main content
American Journal of Translational Research logoLink to American Journal of Translational Research
. 2022 Sep 15;14(9):6424–6444.

Cytokines associated with immune response in atherosclerosis

Jiqing Ma 1, Jianhua Luo 2, Yudong Sun 3, Zhiqing Zhao 1
PMCID: PMC9556506  PMID: 36247305

Abstract

Inflammation is an essential mechanism of immune response that involves a large number of different immune cells. Atherosclerosis is essentially an inflammatory disease caused by inappropriate activities of immune cells. During this process, various cytokines activate immune cells, regulate and transmit immune cell signals, and stimulate a local inflammatory environment. In this study, we reviewed the cytokines associated with immune activity in atherosclerosis, including their roles in immune cell activation and mediating immune cell chemotaxis. The findings give important insights into inflammatory immune microenvironment, including basic mechanisms and interactions, providing new ideas and options for clinical detection and treatment of this disease.

Keywords: Atherosclerosis, cytokine, immunity

Introduction

Atherosclerosis, a vascular disease strongly associated with high lipid levels, was first identified by Rudolf Virchow in the 1850s. As our understanding of its pathogenesis improved, it was established that atherosclerosis is not only due to lipid accumulation within the arterial wall, but also inappropriate body response to vascular damage. The disease involves a sequence of pathological events. First, substantial fibrous and lipid masses accumulate in the subendothelial layer of the artery, wrapping around the circulating cells to form plaques. This narrows or even occludes the blood vessels, obstructing blood flow and hypoxia, which may progress and develop myocardial infarction and stroke.

Several studies have shown that specific cytokines participate in different stages of immune cell activation, such as chemotaxis, differentiation, recruitment, and infiltration. Cytokines also regulate internal and external lipid flow and are essential chemical mediators in various pathophysiological processes, such as intercellular signal transduction. Experimental studies based on animal and patient samples have implicated cytokines in the development of atherosclerosis. In the past two decades, monoclonal antibodies against cytokines have become a standard treatment for chronic inflammatory diseases such as rheumatoid arthritis. Therefore, since atherosclerosis, is also inflammatory disease, similar treatment approaches are currently being explored as novel therapeutics for this disease. More than 20 clinical trials on the treatment of atherosclerosis by targeting immune-associated cytokines were included in ClinicalTrial.gov (Tables 1 and 2).

Table 1.

Summary of clinical trials of drugs targeted cytokines involved in immune cell activation

Target Drug Disease Phase Outcome NCT number Status
TNF-α Infliximab Psoriasis Unknown No results posted NCT01356758 Completed [65]
Atherosclerosis
Adalimumab Psoriasis IV Modest increase in vascular inflammation in carotids NCT01722214 Completed [66]
Vascular Inflammation
Coronary Atherosclerosis
Adalimumab Psoriasis IV Reduce vascular inflammation in patients with moderate to severe psoriasis NCT00940862 Completed [67]
Vascular Inflammation
Coronary Atherosclerosis
Etanercept Atherosclerosis in Psoriasis Patients Study Unknown No results posted NCT01522742 Terminated
IL-1β Canakinumab Atherosclerosis III Decreased hsCRP level and incidence of the primary endpoint NCT01327846 Completed [68]
IL-12/IL-23 Ustekinumab Psoriasis Unknown No effect on MACE NCT01356758 Completed
Atopic Dermatitis
Atherosclerosis
Multiple Methotrexate Coronary Artery Disease II CRP, IL-6 levels ↓ NCT02366091 Completed
Colchicine Coronary Artery Disease III MACE ↓ NCT02551094 Completed
Myocardial Infarction
Coronary Artery Disease IV Attenuated the increase in interleukin-6 and hsCRP concentrations but did not lower the risk of PCI-related myocardial injury NCT01709981 Active, not recruiting
Atherosclerotic Vascular Disease II No results posted NCT02162303 Completed

Table 2.

Summary of clinical trials of drugs targeted cytokines mediating immune cell chemotaxis

Target Drug Type Disease Phase Outcome Status NCT number
CCR2 MLN1202 humanized monoclonal antibody atherosclerosis II CRP level ↓ Completed [129] NCT00715169
CCR5 Maraviroc Small-molecule receptor antagonist STROKE II No results posted Not yet recruiting NCT04789616
Maraviroc Small-molecule receptor antagonist atherosclerosis IV significant improvements in several markers for cardiovascular risk, endothelial dysfunction, arterial stiffness, and early carotid atherosclerosis Completed [130] NCT03402815
CCL2 Bindarit Selective inhibitor Coronary restenosis II in-stent late loss↓ Completed [131] NCT01269242
CXCL12 JVS-100 nonviral DNA plasmid (transient CXCL12 expression) Ischemic heart failure II Failed to demonstrate its primary endpoint of improved composite score at 4 months after treatment Completed [132] NCT01643590
JVS-100 nonviral DNA plasmid (transient CXCL12 expression) Ischemic heart failure I/II No results posted Unknown NCT01961726
JVS-100 nonviral DNA plasmid (transient CXCL12 expression) Critical limb ischemia II No results posted Completed NCT01410331
JVS-100 nonviral DNA plasmid (transient CXCL12 expression) Peripheral arterial disease II Failed to improve outcomes in CLTI at 6 months Completed [133] NCT02544204
ACRX-100 nonviral DNA plasmid (transient CXCL12 expression) heart failure I No results posted Completed NCT01082094
CXCR2 AZD5069 Small-molecule receptor antagonist Coronary heart disease II No results posted Ongoing EudraCT 2016-000775-24
CXCR4 POL6326 Peptidic receptor antagonist Large reperfused ST-elevation myocardial infarction II No results posted Completed NCT01905475
PF-06747143 CXCR4 IgG1 antibody Acute Myeloid Leukemia I No results posted Terminated NCT02954653
BMS-936564 CXCR4 antagonist chronic lymphocytic leukemia (CLL) I No results posted Completed NCT01359657
MIF BAX69 MIF Antibody Metastatic Adenocarcinoma of the Colon or Rectum Malignant Solid Tumors I Safety evaluation Completed [99] NCT01765790

This review will summarize the different cytokines involved in the immune response during atherosclerosis, focusing on their mechanisms and interactions, and updating recent advances in targeted drug research.

Cytokines is involved in immune cell activation

Atherosclerosis is mainly caused by endothelial damage and high lipid levels in the arteries, which activate multiple immune cells that promote lesion formation. Increased infiltration of immune cells such as monocytes, macrophages, T lymphocytes (T cells), B lymphocytes (B cells), and dendritic cells (DCs), in lesion sites, especially the plaque. These cells are part of the body’s self-defense system, but play a role in atherosclerosis development. Some pro-inflammatory cytokines regulate genes that promote inflammation and activate immune cells and disrupt this self-defense system. Partial activities and interactions of these cytokines are represented in Figure 1.

Figure 1.

Figure 1

Schematic overview of cytokines involved in immune cell activation during atherosclerosis. Cytokines can be expressed in almost all types of cells in this environment, especially macrophages. Some of them, like TNF-α and IFN-γ, act as critical roles in this network, promoting the expression of other cytokines including IL-6, IL-8, CCL2, CXCL16, etc. IL-18 drives T cell polarization and induces MMP expression in vascular smooth muscle cells. IL-23 is mainly expressed by macrophages, causing subsequent inflammatory factors reaction. IL-1β has multiple pro-inflammatory functions, other than inducing MMPs and other cytokines, it can also affect the proliferation and migration of vascular smooth muscle cells. IL-6, also known as a key cytokine with diverse functions, can promote low-density lipoprotein uptake in macrophages and stimulate endothelial cells to secret adhesion molecules. More details are offered in the text. IL: interleukin; IFN-γ: interferon-γ; CCL2: C-C motif chemokine ligand 2; CXCL16: C-X-C motif chemokine ligand 16; MIP-1α: macrophage inflammatory protein-1α; CAMs: cell adhesion molecules; LPS: lipopolysaccharide; TMAO: trimetlylamine oxide; TNF-α: tumor necrosis factor-α; MMPs: matrix metalloproteinases. Figure was created using BioRender.com.

Tumor necrosis factor-α (TNF-α)

TNF-α, which mainly secreted by monocytes/macrophages, is one of the most important cytokines in atherosclerosis. TNF-α promotes the expression of multiple pro-inflammatory genes. In atherosclerosis, TNF-α produced by immune cells or endothelial cells increase expression levels of several key genes involved in inflammation and cell proliferation by activating nuclear factor-κb (NF-κB), p38 mitogen-activated protein kinase (MAPK), janus kinase (JAK), and other signaling pathways. The target proteins include different pro-inflammatory cytokines, cell adhesion molecules (CAMs) and chemokines such as interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8), C-C motif chemokine ligand 2 (CCL2). Increased TNF-α self-expression recruits more T cells and macrophages to the lesion site, accelerates the inflammatory cascade response, contributing to disease progression [1]. In addition, TNF-α causes increased leukocyte infiltration into blood vessels, which is an essential first step in plaque formation [2]. TNF-α chronically stimulates macrophages through a MAPK-dependent pathway, downregulates scavenger receptor gene expression, and reduces the effect on the reverse cholesterol pathway, which exacerbates atherosclerosis [3]. In addition to regulating the activation and recruitment of various immune cells, TNF-α has a pro-inflammatory effect on vascular smooth muscle cells. It stimulates the production of matrix metalloproteinases (MMPs), thrombogenic proteins and tissue factor, causing reduced plaque stability and even rupture [4]. TNF-α also regulates phenotypic transition where contractile vascular smooth muscle cells progress to a secretory function, facilitating monocyte migration [5] and contributing to atherosclerosis development.

More than 50% reduction in atherosclerotic lesion area and increased plaque necrosis and apoptosis have been found in TNF-α-/-Apoe-/- double knockout mice [6]. In a study of patients with psoriatic arthritis, the use of TNF-α inhibitors slowed the progression of atherosclerosis and improved vascular inflammation [7]. Therefore, it might be concluded that TNF-α is essential for atherosclerosis. It activates multiple pathways and recruits various immune cells with polydirectional pro-inflammatory effects, hence an ideal potential target for the treatment of atherosclerosis. Studies have also established that TNF-α level is significantly correlated with early carotid atherosclerosis [8]. This suggests that TNF-α can be used as an effective clinical marker for early atherosclerosis.

However, TNF-α as a potential therapeutic target for atherosclerosis has been well studied clinically. This may be due to the negative effects it has shown in some clinical trials, such as exacerbated heart failure and changes in lipidogram, which requires further safety tests [9].

Interestingly, a study showed that loss of p55, a TNF-α receptor, also known as TNF-α R1, appeared to promote the atherosclerosis process [10]. However, the opposite outcomes have been reported in recent studies: it has been found that TNF-α R1 promoted atherosclerosis in low-density lipoprotein receptor knock-out mice [11]. Brusatol was confirmed to inhibit the development of atherosclerosis by suppressing TNF-α R1 [12]. It seems the proatherogenic role of TNF-α R1 has been generally revealed.

Interleukin-1β (IL-1β)

IL-1β is a pro-inflammatory cytokine that is expressed mostly in macrophages, endothelial cells and vascular smooth muscle cells. It is induced by TNF-α and subsequently acts as a local paracrine and autocrine stimulator. Accordingly, IL-1β stimulates the secretion of multiple cytokines and CAMs, leading to immune cell extravasation and persistent local inflammation [13]. IL-1β also promotes the proliferation and migration of vascular smooth muscle cells and induces MMPs to accelerate degradation of atherosclerotic plaque fibrous skeleton [14]. This remodels and transforms the extracellular matrix, affecting plaque stability [15].

In animal models, IL-1β suppression can effectively slow down the development of atherosclerosis. Injection of IL-1β-induced receptors in Apoe-/- mice reduced the fatty streak area in arteries [16]. Under similar conditions, IL-1β-/-Apoe-/- double knockout mice had 30% less lesion area than the control group [17].

In the CANTOS (Canakinuub Anti-inflammatory Thrombosis Outcomes Study) study, patients treated with Canakinnub (a monoclonal antibody to IL-1β) had a significantly lower incidence of clinical outcomes such as atherosclerosis-related myocardial infarction and stroke than the placebo group [18]. The CANTOS trial also confirms the inflammatory hypothesis of atherosclerosis and provides further evidence that targeting inflammation offers an independent pathway for the atherosclerosis treatment. Additionally, the study lays the foundation for the development of additional inflammation-targeted drugs.

Apart from IL-1β, NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammatory vesicles upstream of IL-1β is another possible target. Drugs, such as colchicine in the LODOCO (low-dose colchicine) study, have been shown to reduce IL-1β production by inhibiting NLRP3 inflammatory vesicle activity, with a lower risk of adverse cardiovascular events [19]. This finding was confirmed by the more comprehensive LODOCO2 study [20]. A new NLRP3 inhibitor, MCC950, which is currently under trial [21], might serve as a potentially effective treatment for atherosclerosis.

On the other hand, IL-1β facilitated the formation of fibrous cap and increased plaque stability in the late stages of lesion development. Conversely, plaque stability decreased in atherosclerotic mice knocked out of IL-1 receptors or treated with IL-1β antibodies. This suggests that plaque stability and subsequent possible cardiovascular events should be considered when administering IL-1β-related drugs, especially to patients with advanced atherosclerosis [22].

Interleukin-6 (IL-6)

IL-6 is mostly secreted by macrophages as well as other cell types including fibroblasts and endothelial cells. It is a multifunctional cytokine, which plays an important role in the inflammatory response of atherosclerosis. IL-6 promoted leukocyte recruitment by increasing the production of C-reactive protein (CRP) from liver, resulting in endothelial dysfunction [23]. It can promote low density lipoprotein (LDL) uptake and cytokines expression in macrophages [24]. Activate endothelial cells can express adhesion molecules and chemokines, which stimulated migration and proliferation of smooth muscle cells [25]. A recent study showed that age-associated mitochondrial dysfunction induced by IL-6 contributed to atherosclerosis formation [26].

In mice atherosclerosis models, exogenous IL-6 enhanced the development of early atherosclerosis lesions [27] and destabilized atherosclerosis plaques [28]. However, another study has shown that Apoe-/-IL-6-/- mice had the tendency to gain atherosclerosis more easily, which suggested the dual-modulatory function of IL-6 [29].

IL-6 is known to be involved in several signaling pathways. It can bind to the membrane-bound IL-6 receptor (IL-6R) on leucocytes and endothelial cells, or bind to gp130 with a compound of IL-6 and soluble IL-6R, then activate intracellular signaling in cells that can’t express IL-6R. The third way was trans-presentation through interaction between dendritic cells and receiver T cells [30]. The therapeutic targets for IL-6 pathways usually included IL-6, IL-6R, gp130 and downstream molecules of the janus kinase-signal transducer and activators of transcription pathway (JAK-STAT pathway). Now multiple antibody drugs for some inflammatory diseases targeting IL-6 related pathways have been studied in some clinical trials [31,32]. However, only Sarilumab was under recruitment for its phase IV clinical trial (NCT04350216). Notably, in the CANTOS study, the effect of canakinumab was significantly associated with the decreased level of IL-6 [33], suggesting the synergism of IL-1β and IL-6. Additionally, its role in predicting atherosclerosis was also observed in another study [34]. Therefore, IL-6 may work as a marker of atherosclerosis in the clinical setting.

Interleukin-18 (IL-18)

IL-18 was originally known as an interferon-γ (IFN-γ)-inducible factor because it induces IFN-γ expression. However, IL-18 is now known to be a multifunctional cytokine in various cells, including macrophages and endothelial cells, where its inactive precursors promote signaling through NF-κB pathway [35]. Its receptors occur on macrophages, endothelial cells and vascular smooth muscle cells and mediate interaction between immune cells and blood vessels [36]. It polarizes T cells to Th1 cells [37], the “war hawk” of helper T cells that promotes the development of inflammation. In addition, it amplifies MMPs in monocytes and vascular endothelial cells, which affects plaque stability [36]. IL-18 is a member of the IL-1 cytokine superfamily that also includes IL-1β, which is activated and released downstream of NLRP3 inflammatory vesicles to promote the development of atherosclerosis [38].

In one study, serum IL-18 was elevated in patients with coronary artery disease whereas IL-18 and its receptors were overexpressed in several immune cells, including macrophages, T cells, endothelial cells, and vascular smooth muscle cells in atherosclerotic plaques [39]. This suggested an association between IL-18 and atherosclerotic lesions.

A lower incidence of atherosclerosis was found in IL-18-/-Apoe-/- double knockout mice than in the control group [40]. Treatment with IL-18 inhibitors not only prevented plaque formation, but also transformed it into a more stable plaque phenotype [41]. Apoe-/- mice injected with IL-18 exhibited increased plaque burden [42]. Notably, IFN-γ-/-Apoe-/- double knockout mice were less lesioned than Apoe-/- mice injected with recombinant IL-18, suggesting a synergistic relationship between IL-18 and IFN-γ [40].

IL-18 is an important node in the inflammatory network. It synergizes with many cytokines involved in atherogenesis, such as IL-6, IL-12, and IFN-γ [43], amplifying inflammatory response in the lesion. A study found that IL-18 was related to substantial residual inflammatory risk among the patients who took canakinumab (IL-1β inhibitor) therapy [44]. Therefore, block IL-18 in drugs such as IL-18Bpa (an IL-18 neutralizing antibody), or upstream caspase-1 inhibitors may inhibit multiple pro-inflammatory cascades to attenuate lesion development. However, further research in this area is needed. Inhibition of upstream NLRP3 inflammatory vesicles may also inhibit IL-18 release, as described in section IL-1β above.

Interleukin-23 (IL-23)

Macrophages express both IL-23 and IL-23 receptors, which induces various cells to express Interleukin-17 (IL-17), Interleukin-22 (IL-22), and TNF-α pro-inflammatory factors [45]. The inactivation of IL-23-IL-22 axis signaling causes the intestinal barrier deterioration and ecological dysregulation, increasing systemic pro-atherogenic metabolites such as lipopolysaccharide (LPS) and oxidized trimethylamine and causing atherosclerosis progression [46].

IL-23 has been detected in both mice and human atherosclerotic plaques. Plasma levels of IL-23 were significantly higher in patients with atherosclerosis compared to healthy controls. Follow-up data showed that high plasma levels of IL-23 were correlated with mortality risk [47]. Notably, IL-23 and IL-23 receptor genes were highly expressed in carotid plaques compared to healthy vessels. Levels of IL-17 and TNF-α secreted were higher in monocytes from patients with carotid atherosclerosis treated with IL-23/LPS combination than in monocytes from healthy controls [47].

Briakinumab and ustekinumab, antibodies that target IL-23 subunit p40, have been shown to increase major adverse cardiovascular events (MACE) to different degrees in several clinical trials [48,49]. Other studies did not show exacerbated MACE rates, but this risk cannot be ignored. In addition, monoclonal antibodies Guselkumab, Tildrakizumab, and Risankizumab, which selectively inhibit IL-23 subunit p19, have been studied in clinical trials for psoriasis treatment, but the sample sizes were not sufficient to describe the effects of these drugs on atherosclerosis and subsequent cardiovascular events [50].

Interferon-γ (IFN-γ)

IFN-γ belongs to type II interferon family and is expressed by multiple immune cells, including natural killer cells (NK cells), T cells, and macrophages. It is a widely studied cytokine that regulates multiple human genes mainly through the JAK-STAT pathway [51]. It has a potent pro-lipidogenic effect on atherosclerosis: it induces macrophages to further secrete pro-inflammatory factors [52]. IFN-γ also induces the release of chemokines that attract monocytes and T lymphocytes, such as monocyte chemotactic protein-1, CXC (C-X-C motif) ligand 16 (CXCL16), and macrophage inflammatory protein 1α (MIP-1) and promotes monocyte differentiation into macrophages [53]. In addition, IFN-γ promotes uptake of oxidized low-density lipoprotein (oxLDL) by macrophages and vascular smooth muscle cells, reduces cholesterol efflux, and contributes to the development of foam cells [54], which lay the foundation for plaque formation.

Injecting IFN-γ into Apoe-/- mice increased plaque deposition and reduced vascular smooth muscle proliferation and collagen deposits in the plaque cap, suggesting that IFN-γ may also impair plaque stability [55]. In contrast, in IFN-γ-/-Apoe-/- double knockout mice, plaque shrinkage was observed [56]. IFN-γ is essential in all stages of atherosclerosis progression, from immune cell recruitment, LDL accumulation, to plaque development and stabilization.

Some lipid-lowering drugs such as statins and PCSK-9 inhibitors decrease IFN-γ [57,58] level in addition to their cholesterol lowering effect. Currently, new therapies targeting IFN-γ are being investigated. Neutralizing IFN-γ antibodies were used to reduce atherosclerosis in the grafted vessels and aorta in Apoe-/- mice undergoing heart transplantation [59]. Bioinformatics data analysis supported the ability of specific long-stranded non-coding RNAs (lncRNAs) to promote atherosclerosis by affecting the IFN-γ pathway [60]. Another study showed that microRNA miR-155, which is highly expressed in atherosclerotic plaques, also induces IFN-γ expression [61,62]. In systemic lupus erythematosus patients, using type I anifrolumab could reduce neutrophil extracellular trap formation and interleukin-10 (IL-10) levels [63]. However, it is important to note that restricted expression of IFN-γ may lead to immunosuppression and increase the incidence of infection [64]. Therefore, it is important to treat opportunistic infections when administering lipid-lowering drugs in the long term.

Cytokines mediate immune cell chemotaxis

Chemokines were originally named after their function of directing white blood cells to inflammation sites. However, recent advances in research have led to the discovery that they perform other functions besides immune cell recruitment, including keeping cellular homeostasis and activating different cell functions. Chemokines are highly active in inflammation sites and regulate various inflammatory cellular processes. As an inflammatory disease, atherosclerosis development and progression is driven by chemokines. Therefore, understanding the mechanism of atherosclerosis-related chemokines will inform the development of effective treatments to control atherosclerotic lessions.

Chemokines are a family of structurally similar cytokines. Most chemokines are secreted proteins with a molecular weight of about 10 kda. Each chemokine consists of a carboxy-terminal alpha helix structure that preferentially binds proteoglycans and extracellular matrix proteins on vascular endothelial cells. It also includes four cysteines at highly conserved positions. Based on the distribution of cysteine N-terminal residues, chemokines are classified into four subclasses: CC, CXC, CX3C, and XC. Chemokines bind to G protein-coupled receptors, initiate the dissociation of G protein subunits α, β, and γ, subsequently activate MAPK, phosphatidylinositol 3-kinase (PI3K) and phospholipase C (PLC) pathways. In addition, such binding increases intracellular calcium levels, causing cell polarization, adhesion and migration. G protein-coupled receptors are also known as conventional chemokine receptors (CKRs). Another type of receptors, the atypical chemokine receptors (ACKRs), are mainly considered as scavenger receptors. They act independently from the G protein signaling pathway, indirectly control the interaction between chemokines and CKRs by regulating the localization and function of chemokines. Usually, many chemokines from the same family bind to several different receptors and a specific receptor may have multiple chemokine ligands. Therefore, chemokines and their receptors together form a large network with complex interactions that need further mechanistic exploration. Partial activities and interactions of these chemokines are represented in Figure 2.

Figure 2.

Figure 2

Schematic overview of cytokines mediating immune cell chemotaxis during atherosclerosis. CCL2 can attract monocytes to the lesion and drive them to differentiate into macrophages. CCL2 further promotes macrophages polarization to M1. CX3CL1 was released by apoptotic cells and then recruits macrophages to form foam cells. MIF is a multipotent atypical chemokine, it selectively recruits T cells, monocytes and leucocytes through different receptors. It also promotes the expression of other cytokines, VCAM-1 and ICAM-1. Platelets are the factory of several chemokines including CCL5, CXCL4 and CXCL16. CCL5 can stop leucocytes from moving through the CCR1 and CCR5 receptors and migrate leucocytes to the endothelium. CXCL4 can also bind to CCR1 and form a complex with CCL5, performing chemotaxis. CXCL16 promotes oxLDL uptake of macrophages other than its chemotactic function. CXCL12 exerts diverse effects including recruitment and promoting adhesion by binding to different receptors. More details are offered in the text. CCL2: C-C motif chemokine ligand 2; CCL5: C-C motif chemokine ligand 5; CCR1: C-C motif chemokine receptor 1; CCR2: C-C motif chemokine receptor 2; CCR5: C-C Motif Chemokine Receptor 5; CXCL4: C-X-C motif chemokine ligand 4; CXCL12: C-X-C motif chemokine ligand 12; CXCL16: C-X-C motif chemokine ligand 16; CXCR2: C-X-C motif chemokine receptor 2; CXCR6: C-X-C motif chemokine receptor 6; CXCR7: C-X-C motif chemokine receptor 7; CX3CL1: C-X3-C motif chemokine ligand 1; CX3CR1: C-X3-C motif chemokine receptor 1; TNF-α: tumor necrosis factor-α; IL: interleukin; MIF: macrophage migration inhibitory factor; iNOS: inducible nitric oxide synthase; NO: nitric oxide; LDL: low-density lipoprotein; oxLDL: oxidized low-density lipoprotein. Figure was created using BioRender.com.

C-C motif chemokine ligand 2 (CCL2)

CCL2 is the best known CC chemokine that was first recognized as a monocyte chemotactic factor. Studies have shown that the CCL2-CCR2 axis is required for monocytes to migrate from bone marrow to peripheral circulation [69]. However, CCL2 has recently been found to to promote tumor progression and immune surveillance. CCR2 is a CCL2 receptor that is expressed on the surface of monocytes after exposure to inflammatory stimuli. CCL2 is typically expressed in endothelial cells, monocytes/macrophages, smooth muscle cells, and T cells. It causes monocytes to aggregate due to inflammation or injury through downstream signaling pathways such as JAK-STAT pathway, MAPK pathway, and PI3K pathway [70]. Its expression is induced mainly by cytokines such as IL-1, interleukin-4 (IL-4), TNF-α, and IFN-γ, various growth factors, lipopolysaccharides, reactive oxygen species (ROS), oxLDLs and immune complexes [71]. CCL2 is a monocyte efflux signal that activates G protein-coupled receptors to mediate cellular trafficking, which subsequently directs monocytes along a chemokine gradient to the site of injury. It is associated with various diseases, including rheumatoid arthritis, atherosclerosis, diabetes, certain cancers (breast cancer, prostate cancer, pancreatic cancer, etc.). This article focuses on monocyte migration in atherosclerosis.

In addition to chemotaxis, CCL2 is important in cell polarization and survival. Its ability to direct macrophage polarization toward M1 via granulocyte-macrophage colony stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF) has been demonstrated in CCL2 knockout mice and CCR2-deficient mice [72]. In addition, CCL2 activates CCR2 and mediates inflammatory response to atherosclerosis via ERK-dependent downstream signaling of leukotriene liposomes in foam cells [73]. All these processes largely contribute to the development of atherosclerosis.

Studies in mice have yielded indisputable results. CCR2-/- knockout mice had significantly lowatherosclerosis but normal blood lipid and lipoprotein levels, demonstrating an association between CCL2 and atherosclerosis [74]. Further, treatment targeting CCL2-CCR2 axis effectively reduced lesion development and progression [75]. Clinical studies have also shown a significant correlation between CCL2 levels and atherosclerotic stroke in human patients [76].

Some anti-inflammatory and lipid-lowering drugs such as glucocorticoids [77] and statins [78] have been shown to have a non-selective inhibitory effect on CCL2. A recent study found the new effects of Colchicine to lower CCL2 levels in patients with acute coronary syndrome (ACS) [79]. Various cancer drugs targeting CCL2-CCR2 axis have been clinically tested with only a few of them showing positive results [80,81]. This outcome is probably due to the complex non-unilinear function of chemokines. When one of the pathways is blocked, its function is maintained by the compensatory effect of other pathways. In addition, inhibiting the chemotactic effect of CCL2 may affect the organism itself, including causing abnormalities in damage response to inflammation, which are important side effects of CCL2.

C-X3-C motif chemokine ligand 1 (CX3CL1)

CX3CL1 is involved in the initiation step of atherosclerotic plaque formation. Its toxicity damages vascular endothelial cells, causing vascular injury, which later set offcascade reactions. Compared to other chemokines, CX3CL1 is unique because it is both soluble and membrane-adhesive [82]. It is also specific to CX3CR1 receptors. CX3CR1 is typically expressed on leukocytes and binds to membrane-bound CX3CL1 of endothelial cells, activating lymphocytes and the release of lysis granules that destroy vascular endothelium [83]. Apoptotic cells also release CX3CL1 to recruit macrophages that remove apoptotic debris [84]. This clearance may be useful in early stages of lesion development. However, in advanced stage, CX3CL1-CX3CR1 axis signaling exacerbates the formation of foam cells [85], contributing to lesion progression. In addition, smooth muscle cells in atherosclerotic plaques also express CX3CR1 [86], which moves and converges CX3CL1 in near the lesion.

Platelets are actively involved in plaque formation, CX3CL1 promotes lesion development by activating platelets and through its adhesion to the endothelium [87]. Both the expression of CX3CR1 on platelets and its binding to CX3CL1 increase after hyperlipidemia, promoting platelet aggregation and monocyte recruitment [88]. The hemostatic and thrombogenic functions of platelets are highly correlated with atherosclerosis and the probability of subsequent plaque rupture. Platelet levels depend heavily on the regulation of chemokines, mainly CX3CL1, C-X3-C motif chemokine ligand 16 (CXCL16), C-X3-C motif chemokine ligand 12 (CXCL12), C-C motif chemokine ligand 12 (CCL12), and C-C motif chemokine ligand 22 (CCL22) [89].

In animal experiments, Apoe-/- and Ldlr-/- atherosclerotic mice were treated with F1, an amino-terminal modified CX3CR1 ligand with CX3CR1 antagonist activity. The results revealed that macrophages accumulated in the arteries, fewer monocytes were recruited, and atherosclerotic lesions were ameliorated [90]. This indicates that antagonizing CX3CR1 is a promising strategy for slowing the progression of atherosclerosis, but it needs to be tested clinically. Now clinical trials of specific CX3CL1 inhibitor is still blank, but colchicine showed a positive effect in inhibiting CX3CL1 in ACS patients [79].

Macrophage migration inhibitory factor (MIF)

MIF is a multipotent immunomodulatory cytokine with a unique structure. It was one of the first cytokines identified by Bloom and Bennett in 1966 when studying delayed hypersensitivity reaction.Its primary function is to inhibit random migration of macrophages from capillaries. Because it lacks the characteristic N-terminal cysteine of classical chemokines but exhibits chemokine-like functions and binds to classical chemokine receptors, MIF is classified as a novel atypical chemokine (ACKs). The expression level of MIF is low in normal vessels, but significantly high in inflammatory states. Two receptors bind to MIF: CXCR2 and CXCR4, which are predominantly expressed on the surface of monocytes and T cells, respectively. Both CXCR2 and CXCR4 are expressed on the surface of leukocytes, and by binding to both receptors, MIF promotes the recruitment of monocytes and T cells [91]. It also increases vascular cell adhesion molecule-1 (VCAM-1)/intercelluar adhesion molecule-1 (ICAM-1) expression and promotes leukocyte adhesion to vascular endothelium, CCL2 expression and macrophage activation [92]. MIF in plaques also promotes the release of other cytokines such as TNF-α and IL-1β, which exacerbate the local inflammatory environment. It promotes foam cell formation by stimulating enhanced oxLDL uptake and increases plaque instability by inducing matrix degradation through MMPs [93]. In additional experiments, MIF was found to affect plaque stability by inhibiting VSMC proliferation and regulating proteolytic activity and elastin and collagen breakdown [94]. It also inhibited p53 function, causing inhibition of apoptosis and promotion of inflammatory response, which contributed to lesion development [95].

In the MIF-deficient murine model, chronic inflammation developed later and more slowly in mice compared to controls, and plaque lesions were reduced [96]. Apoe-/- mice treated with MIF-neutralizing antibodies had significantly reduced inflammatory indices and aortic plaque area compared to the control group [97].

Drugs targeting MIF have been recently approved for use, such as Ibudilast for treating multiple sclerosis [98]. Other drugs like Imalumab [99] and IPG-1094 are under clinical trial, but the target diseases are still mainly limited to various types of tumors.

C-C motif chemokine ligand 5 (CCL5)

CCL5, also known as RANTES, is one of the chemokines that is highly expressed and released when platelets are activated at the lesion site. It is carried by platelets to activated endothelial cells and binds to CCR1 and CCR5, causing leukocytes to migrate to arterial intima [100]. It also promotes the recruitment of other platelets and immune cells [101]. The interaction between CCR5 and CCL5 mediates CD4+ T cell homing. Scientists have identified a specific subtype of CCR5+CD4+ T cells [102] capable of secreting IFN-γ, various interleukins and TNF-α. Some of these cytokines are pro-inflammatory cytokines that promote the development of atherosclerosis. CCL5 is known to form complexes with other chemokines such as C-X-C motif chemokine ligand 4 (CXCL4) and C-C motif chemokine Ligand17 (CCL17), which act in combination and carry each other to recruit immune cells that promote atherosclerosis [103].

CCL5 is highly expressed in atherosclerotic plaques. Mice treated with the CCL5 antagonist Met-RANTES had significantly low leukocyte infiltration levels and atherosclerotic lesions [104]. Atherosclerosis was also reduced in CCR5 knockout Apoe-/- mice compared to controls [105]. These experimental results validate that the crucial role of CCL5-CCR1-CCR5 axis in plaque formation. In samples of atherosclerotic patients, elevated circulating levels of RANTES were statistically associated with progression of acute coronary syndromes [106].

In addition, the role of CCL5-CCR5 axis has been studied in various diseases, including cancer, some inflammatory diseases, and AIDS. The drug Maraviroc (MVC), which targets CCR5 and is currently used as an antiviral drug for HIV treatment. MVC has been found to reduce the risk of atherosclerosis and alleviate advanced plaque progression in a mouse model [107]. MVC also affects carotid intima-media thickness and atherosclerotic plaques in HCV/HIV co-infected patients. Inhibition of CCR5 prevents the development of atherosclerosis in HCV/HIV co-infected patients, especially in the non-calcified phase [108]. A novel dual antagonist of CCR5 and CCR2, Cenicriviroc (CVC), which inhibits monocyte chemotaxis by reducing E-selectin expression, is a promising treatment for atherosclerosis [109]. However, further animal experiments and clinical trials are needed to identify suitable drug targets for atherosclerosis.

C-X-C motif chemokine ligand 4 (CXCL4)

CXCL4, also known as platelet factor 4 (PF4), exerts an anti-apoptotic effect on monocytes and stimulates their differentiation into macrophages [110]. Activated platelets synergistically act with chemokines to exacerbate the pathogenesis of atherosclerosis. Platelets secrete CXCL4, and the structural properties of its receptor CCR1 allow CXCL4 and CCL5 to interact, forming a complex that causes monocyte arrest on the endothelium at the site of inflammatory injury and consequently atherosclerotic lesions [103]. Also, immunohistochemical analysis of human carotid atherosclerotic lesion samples reveals co-localization of CXCL4 with ox-LDL. This confirms the hypothesis that CXCL4 binding to oxLDL subsequently mediates macrophage uptake and esterification, thereby promoting the formation of foam cells [111]. Additionally, a non-allelic variant isoform of CXCL4, CXCL4L1, exists, which has a distinct effect on monocyte, inhibiting chemotactic recruitment and angiogenesis as well as causing endothelial cell migration [112].

In vitro cell experiments indicate that CXCL4 promotes atherosclerosis by limiting apoptosis of neutrophils and monocytes under pro-inflammatory conditions and mediating T cell-platelet interactions with platelets [113]. Cell experiments on macrophages revealed that PF4 causes macrophage differentiation, resulting in the downregulation of the CD163 atherosclerotic protective receptor. Besides, analysis of human atherosclerotic plaque samples upregulated PF4 and downregulated CD163 expression [114]. In animal experiments, atherosclerotic plaque burden was reduced in both C57BL/6PF4-/- and Apoe-/- PF4-/- mice [115], which is similar to the effect of CX3CL1. Immunohistochemical analysis of atherosclerotic plaque samples from human carotid arteries identified the presence of PF4 in the endothelium and macrophages of the lesioned fraction, and the levels positively correlated with the severity of atherosclerosis [116].

Although the CXCL4-CCL5 complex affects atherosclerosis, targeting CCL5 alone caused a systemic immune response. Therefore, stable peptide inhibitors targeting the CCL5-CXCL4 complex structure have been designed to suppress atherosclerosis by reducing monocyte recruitment in mice models. For example, MKEY (a specifically designed compound to block CCL5-CXCR4 interaction) has demonstrated therapeutic benefit by inhibiting specific chemokines crucial for the development of atherosclerosis in mice [117].

C-X-C motif chemokine ligand 16 (CXCL16)

CXCL16 is a functionally diverse chemokine found in both membrane-binding type and secretory forms; On one hand, it protects against atherosclerosis and promotes it on the other hand. CXCL16 is expressed by dendritic cells, macrophages, B cells, T cells, smooth muscle cells, and endothelial cells. Membrane-binding CXCL16 act as an adhesion molecule for cells expressing the receptor CXCR6, promoting leukocyte aggregation and adhesion to the damaged vascular endothelium [118]. Nonetheless, macrophages in CXCL16-/- mice exhibit a reduced capacity to internalize LDL. CXCL16 also acts as a scavenger receptor for oxLDL, helping macrophages and smooth muscle cells absorb oxLDL, which protects against atherosclerosis in the early stage and promotes foam cell formation [119].

Furthermore, CXCL16 is secreted by platelets and also activates platelets by binding to CXCR6 on platelets, promoting platelet aggregation on the endothelium [120]. Its expression on platelets is associated with disease severity; platelets from patients with ACS exhibit enhanced CXCL16 expression than platelets from those with coronary artery disease [121].

HUNT study found that baseline levels of circulating CXCL16 were linked to a higher risk of death in patients with acute coronary syndromes [122]. A follow-up study also confirmed that CXCL16 is still useful for predicting atherosclerosis and subsequent cardiovascular events, either in plaque stability or in acute coronary syndromes, after excluding other contributing factors [123].

C-X-C motif chemokine ligand 12 (CXCL12)

The production of CXCL12, also known as stromal cell-derived factor 1 (SDF-1), is triggered by the endothelial cell-derived apoptotic vesicles via micro-126 during lesions [124]. Its ligands, including CXCR4 and CXCR7, and CXCL12, play different roles when bound to other ligands. CXCR7 acts as a negative regulator of CXCL12, internalizing CXCL12 and transmitting it to lysosomes for degradation, thereby regulating CXCL12/CXCR4 signaling. Besides, CXCR7 (also known as ACKR3) is involved in monocyte adhesion and survival [125]. Regardless of the receptor it binds, CXCL12 promotes macrophage differentiation, facilitating platelet phagocytosis, thereby causing foam cell formation [126].

In Apoe-/- mice, CXCL12 promotes lesion stabilization without affecting vessel diameter via smooth muscle cell mobilization, increased collagen content, and fibrous cap thickening [127], beneficial in advanced atherosclerosis.

CXCL12 antagonist LIT-927 in immunodeficient mice prone to lupus regulates the correction of immune changes, attenuates lymphocyte activity, and hence regulates inflammation. Its effect is better than that of CXCR4/CXCR7 antagonist AMD3100 [128]. This also suggests that controlling disease progression by antagonizing chemokines in the early stages of atherosclerosis yields a proactive preventive effect.

Anti-inflammatory cytokines in atherosclerosis

In addition to the pro-inflammatory cytokines mentioned above, anti-inflammatory cytokines should not be disregarded. IL-10 and transforming growth factor β (TGF-β) are the most representative anti-inflammatory cytokines. IL-10 was mainly produced by macrophages in plaque. The atheroprotective role of IL-10 was exemplified by lowering the expression of MMP and some pro-inflammatory cytokines (e.g. IL-1β, TNF-α, IL-8), promoting macrophages polarization towards the M2 phenotype and further inhibited the progress of atherosclerosis [134]. Recent studies showed that using exosome-mediated IL-10 mRNA can effectively control atherosclerosis in Apoe-/- mice [135]. For rheumatoid arthritis treatments, antibody fragment F8-mediated IL-10 has been studied in clinical trials (NCT02076659, NCT02270632), which taking hope to IL-10 therapy in atherosclerosis. TGF-β was expressed in various cells such as leukocytes, macrophages, and VSMCs. It showed anti-inflammatory properties by inhibiting inflammatory cytokines including TNF-α, IL-1β, etc, subsequently suppressed the adhesion and activation of inflammatory cells [136]. Several experiments conducted in mice confirmed the effects on regulating TGF-β [137-139]. Notably, regulatory T cells (Tregs) were the common source of both IL-10 and TGF-β. Clinical data also indicated the relevance between Treg level and coronary artery disease (CAD) [140]. Thus, targeting Tregs may also be potential access, such as activating protective immunity of Tregs by administering antigens [141]. Patients who had influenza vaccination injection after myocardial infarction with a lower risk of all cause death and cardiovascular death [142], which added evidence to this possible treatment.

It has been noted that IL-2 was not normally regarded as an anti-inflammatory cytokine. However, it had a positive function in atheroprotection. Mice experiments showed that IL-2 could alleviate atherosclerosis by promoting Treg expansion [143,144].

Aside from targeting pro-inflammatory cytokines, we needed to pay attention to anti-inflammatory cytokines and their potential avenues including increasing the level of anti-inflammatory cytokines or strengthening the cells which produce them. These studies and clinical trials of anti-inflammatory cytokines were relatively rare, probably due to the limited effects of enhancing anti-inflammatory function. Future treatment of atherosclerosis using potent anti-inflammatory agents or in combination with anti-inflammatory agents with pro-inflammatory cytokines inhibitors might be feasible.

Conclusion

Chemotaxis and activation of immune cells by cytokines are crucial mechanisms for the pathogenesis of atherosclerosis. Thus, investigating the role of cytokines is critical to unraveling the pathogenic mechanism of atherosclerosis and possible therapeutic interventions. The progression of atherosclerotic lesions can be effectively combatted by pinpointing key targets and making a global observation, as well as regulating the dynamic changes at the lesion site or even the inflammatory state of the whole organism. At present, research on the treatment of atherosclerosis by targeting cytokines is still ongoing, and some clinical effects are still uncertain. Combination of drug regimens have been proposed to address this, i.e., different cytokine inhibitors are combined to inhibit multiple inflammatory pathways; besides, cytokine inhibitors combined with lipid-lowering drugs are utilized to act on both cholesterol and inflammatory pathways. For instance, PCSK9 inhibitors (targeted cholesterol-lowering) alone can be used to treat atherosclerosis [145]. However, this also increases the risk of inflammation [146]. PCSK9 and cytokine antibodies might yield a 1+1>2 effect. Importantly, understanding the underlying mechanisms of the above cytokines can disclose the nature of the cytokine action network. To further combat atherosclerosis progression, critical nodes in the cytokine network can be targeted to limit immune cell chemotaxis, regulate immune cell activation, block signaling pathways including NF-κB and reduce the secretion of pro-inflammatory cytokines. This will facilitate efficient management of pathogenesis and mitigates or even prevents its development. Specifically, research should concentrate on multidirectional interactions and crosstalk of different cytokines and their receptors, their effects on normal tissues, and distinct or even opposite effects of a specific cytokine at each lesion stage so as to identify novel targets for therapeutic interventions.

Acknowledgements

This study was financed by the National Natural Science Foundation of China (NSFC) (grant nos. 81770482, 82000464).

Disclosure of conflict of interest

None.

References

  • 1.Parameswaran N, Patial S. Tumor necrosis factor-α signaling in macrophages. Crit Rev Eukaryot Gene Expr. 2010;20:87–103. doi: 10.1615/critreveukargeneexpr.v20.i2.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Dudeck J, Kotrba J, Immler R, Hoffmann A, Voss M, Alexaki VI, Morton L, Jahn SR, Katsoulis-Dimitriou K, Winzer S, Kollias G, Fischer T, Nedospasov SA, Dunay IR, Chavakis T, Müller AJ, Schraven B, Sperandio M, Dudeck A. Directional mast cell degranulation of tumor necrosis factor into blood vessels primes neutrophil extravasation. Immunity. 2021;54:468–483. e465. doi: 10.1016/j.immuni.2020.12.017. [DOI] [PubMed] [Google Scholar]
  • 3.Hsu HY, Twu YC. Tumor necrosis factor-alpha-mediated protein kinases in regulation of scavenger receptor and foam cell formation on macrophage. J Biol Chem. 2000;275:41035–41048. doi: 10.1074/jbc.M003464200. [DOI] [PubMed] [Google Scholar]
  • 4.Galis ZS, Muszynski M, Sukhova GK, Simon-Morrissey E, Libby P. Enhanced expression of vascular matrix metalloproteinases induced in vitro by cytokines and in regions of human atherosclerotic lesions. Ann N Y Acad Sci. 1995;748:501–507. doi: 10.1111/j.1749-6632.1994.tb17348.x. [DOI] [PubMed] [Google Scholar]
  • 5.Lamb FS, Choi H, Miller MR, Stark RJ. TNFα and reactive oxygen signaling in vascular smooth muscle cells in hypertension and atherosclerosis. Am J Hypertens. 2020;33:902–913. doi: 10.1093/ajh/hpaa089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Brånén L, Hovgaard L, Nitulescu M, Bengtsson E, Nilsson J, Jovinge S. Inhibition of tumor necrosis factor-alpha reduces atherosclerosis in apolipoprotein E knockout mice. Arterioscler Thromb Vasc Biol. 2004;24:2137–2142. doi: 10.1161/01.ATV.0000143933.20616.1b. [DOI] [PubMed] [Google Scholar]
  • 7.Eder L, Joshi AA, Dey AK, Cook R, Siegel EL, Gladman DD, Mehta NN. Association of tumor necrosis factor inhibitor treatment with reduced indices of subclinical atherosclerosis in patients with psoriatic disease. Arthritis Rheumatol. 2018;70:408–416. doi: 10.1002/art.40366. [DOI] [PubMed] [Google Scholar]
  • 8.Skoog T, Dichtl W, Boquist S, Skoglund-Andersson C, Karpe F, Tang R, Bond MG, de Faire U, Nilsson J, Eriksson P, Hamsten A. Plasma tumour necrosis factor-alpha and early carotid atherosclerosis in healthy middle-aged men. Eur Heart J. 2002;23:376–383. doi: 10.1053/euhj.2001.2805. [DOI] [PubMed] [Google Scholar]
  • 9.Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JT. Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-alpha, in patients with moderate-to-severe heart failure: results of the anti-TNF Therapy Against Congestive Heart Failure (ATTACH) trial. Circulation. 2003;107:3133–3140. doi: 10.1161/01.CIR.0000077913.60364.D2. [DOI] [PubMed] [Google Scholar]
  • 10.Schreyer SA, Peschon JJ, LeBoeuf RC. Accelerated atherosclerosis in mice lacking tumor necrosis factor receptor p55. J Biol Chem. 1996;271:26174–26178. doi: 10.1074/jbc.271.42.26174. [DOI] [PubMed] [Google Scholar]
  • 11.Xanthoulea S, Gijbels MJ, van der Made I, Mujcic H, Thelen M, Vergouwe MN, Ambagts MH, Hofker MH, de Winther MP. P55 tumour necrosis factor receptor in bone marrow-derived cells promotes atherosclerosis development in low-density lipoprotein receptor knock-out mice. Cardiovasc Res. 2008;80:309–318. doi: 10.1093/cvr/cvn193. [DOI] [PubMed] [Google Scholar]
  • 12.Kim CW, Oh ET, Park HJ. A strategy to prevent atherosclerosis via TNF receptor regulation. FASEB J. 2021;35:e21391. doi: 10.1096/fj.202000764R. [DOI] [PubMed] [Google Scholar]
  • 13.Wang X, Feuerstein GZ, Gu JL, Lysko PG, Yue TL. Interleukin-1 beta induces expression of adhesion molecules in human vascular smooth muscle cells and enhances adhesion of leukocytes to smooth muscle cells. Atherosclerosis. 1995;115:89–98. doi: 10.1016/0021-9150(94)05503-b. [DOI] [PubMed] [Google Scholar]
  • 14.Kaden JJ, Dempfle CE, Grobholz R, Tran HT, Kiliç R, Sarikoç A, Brueckmann M, Vahl C, Hagl S, Haase KK, Borggrefe M. Interleukin-1 beta promotes matrix metalloproteinase expression and cell proliferation in calcific aortic valve stenosis. Atherosclerosis. 2003;170:205–211. doi: 10.1016/s0021-9150(03)00284-3. [DOI] [PubMed] [Google Scholar]
  • 15.Eun SY, Ko YS, Park SW, Chang KC, Kim HJ. IL-1β enhances vascular smooth muscle cell proliferation and migration via P2Y2 receptor-mediated RAGE expression and HMGB1 release. Vascul Pharmacol. 2015;72:108–117. doi: 10.1016/j.vph.2015.04.013. [DOI] [PubMed] [Google Scholar]
  • 16.Elhage R, Maret A, Pieraggi MT, Thiers JC, Arnal JF, Bayard F. Differential effects of interleukin-1 receptor antagonist and tumor necrosis factor binding protein on fatty-streak formation in apolipoprotein E-deficient mice. Circulation. 1998;97:242–244. doi: 10.1161/01.cir.97.3.242. [DOI] [PubMed] [Google Scholar]
  • 17.Kamari Y, Werman-Venkert R, Shaish A, Werman A, Harari A, Gonen A, Voronov E, Grosskopf I, Sharabi Y, Grossman E, Iwakura Y, Dinarello CA, Apte RN, Harats D. Differential role and tissue specificity of interleukin-1alpha gene expression in atherogenesis and lipid metabolism. Atherosclerosis. 2007;195:31–38. doi: 10.1016/j.atherosclerosis.2006.11.026. [DOI] [PubMed] [Google Scholar]
  • 18.Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, Kastelein JJP, Cornel JH, Pais P, Pella D, Genest J, Cifkova R, Lorenzatti A, Forster T, Kobalava Z, Vida-Simiti L, Flather M, Shimokawa H, Ogawa H, Dellborg M, Rossi PRF, Troquay RPT, Libby P, Glynn RJ. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377:1119–1131. doi: 10.1056/NEJMoa1707914. [DOI] [PubMed] [Google Scholar]
  • 19.Nidorf SM, Eikelboom JW, Budgeon CA, Thompson PL. Low-dose colchicine for secondary prevention of cardiovascular disease. J Am Coll Cardiol. 2013;61:404–410. doi: 10.1016/j.jacc.2012.10.027. [DOI] [PubMed] [Google Scholar]
  • 20.Nidorf SM, Fiolet ATL, Mosterd A, Eikelboom JW, Schut A, Opstal TSJ, The SHK, Xu XF, Ireland MA, Lenderink T, Latchem D, Hoogslag P, Jerzewski A, Nierop P, Whelan A, Hendriks R, Swart H, Schaap J, Kuijper AFM, van Hessen MWJ, Saklani P, Tan I, Thompson AG, Morton A, Judkins C, Bax WA, Dirksen M, Alings M, Hankey GJ, Budgeon CA, Tijssen JGP, Cornel JH, Thompson PL LoDoCo2 Trial Investigators. Colchicine in patients with chronic coronary disease. N Engl J Med. 2020;383:1838–1847. doi: 10.1056/NEJMoa2021372. [DOI] [PubMed] [Google Scholar]
  • 21.Sharma A, Choi JSY, Stefanovic N, Al-Sharea A, Simpson DS, Mukhamedova N, Jandeleit-Dahm K, Murphy AJ, Sviridov D, Vince JE, Ritchie RH, de Haan JB. Specific NLRP3 inhibition protects against diabetes-associated atherosclerosis. Diabetes. 2021;70:772–787. doi: 10.2337/db20-0357. [DOI] [PubMed] [Google Scholar]
  • 22.Gomez D, Baylis RA, Durgin BG, Newman AAC, Alencar GF, Mahan S, St Hilaire C, Müller W, Waisman A, Francis SE, Pinteaux E, Randolph GJ, Gram H, Owens GK. Interleukin-1β has atheroprotective effects in advanced atherosclerotic lesions of mice. Nat Med. 2018;24:1418–1429. doi: 10.1038/s41591-018-0124-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Souza JR, Oliveira RT, Blotta MH, Coelho OR. Serum levels of interleukin-6 (Il-6), interleukin-18 (Il-18) and C-reactive protein (CRP) in patients with type-2 diabetes and acute coronary syndrome without ST-segment elevation. Arq Bras Cardiol. 2008;90:86–90. doi: 10.1590/s0066-782x2008000200004. [DOI] [PubMed] [Google Scholar]
  • 24.Schuett H, Luchtefeld M, Grothusen C, Grote K, Schieffer B. How much is too much? Interleukin-6 and its signalling in atherosclerosis. Thromb Haemost. 2009;102:215–222. doi: 10.1160/TH09-05-0297. [DOI] [PubMed] [Google Scholar]
  • 25.Su JH, Luo MY, Liang N, Gong SX, Chen W, Huang WQ, Tian Y, Wang AP. Interleukin-6: a novel target for cardio-cerebrovascular diseases. Front Pharmacol. 2021;12:745061. doi: 10.3389/fphar.2021.745061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Tyrrell DJ, Blin MG, Song J, Wood SC, Zhang M, Beard DA, Goldstein DR. Age-associated mitochondrial dysfunction accelerates atherogenesis. Circ Res. 2020;126:298–314. doi: 10.1161/CIRCRESAHA.119.315644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Huber SA, Sakkinen P, Conze D, Hardin N, Tracy R. Interleukin-6 exacerbates early atherosclerosis in mice. Arterioscler Thromb Vasc Biol. 1999;19:2364–2367. doi: 10.1161/01.atv.19.10.2364. [DOI] [PubMed] [Google Scholar]
  • 28.Zhang K, Huang XZ, Li XN, Feng M, Li L, Cai XJ, Zhang C, Liu XL, Zhang MX, Zhang Y, Wang XL, Zhang M. Interleukin 6 destabilizes atherosclerotic plaques by downregulating prolyl-4-hydroxylase α1 via a mitogen-activated protein kinase and c-Jun pathway. Arch Biochem Biophys. 2012;528:127–133. doi: 10.1016/j.abb.2012.09.007. [DOI] [PubMed] [Google Scholar]
  • 29.Schieffer B, Selle T, Hilfiker A, Hilfiker-Kleiner D, Grote K, Tietge UJ, Trautwein C, Luchtefeld M, Schmittkamp C, Heeneman S, Daemen MJ, Drexler H. Impact of interleukin-6 on plaque development and morphology in experimental atherosclerosis. Circulation. 2004;110:3493–3500. doi: 10.1161/01.CIR.0000148135.08582.97. [DOI] [PubMed] [Google Scholar]
  • 30.Heink S, Yogev N, Garbers C, Herwerth M, Aly L, Gasperi C, Husterer V, Croxford AL, Möller-Hackbarth K, Bartsch HS, Sotlar K, Krebs S, Regen T, Blum H, Hemmer B, Misgeld T, Wunderlich TF, Hidalgo J, Oukka M, Rose-John S, Schmidt-Supprian M, Waisman A, Korn T. Corrigendum: trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells. Nat Immunol. 2017;18:474. doi: 10.1038/ni0417-474b. [DOI] [PubMed] [Google Scholar]
  • 31.Takeuchi T, Yamanaka H, Harigai M, Tamamura R, Kato Y, Ukyo Y, Nakano T, Hsu B, Tanaka Y. Sirukumab in rheumatoid arthritis refractory to sulfasalazine or methotrexate: a randomized phase 3 safety and efficacy study in Japanese patients. Arthritis Res Ther. 2018;20:42. doi: 10.1186/s13075-018-1536-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Choy E, Caporali R, Xavier R, Fautrel B, Sanmarti R, Bao M, Devenport J, Pethö-Schramm A. Effects of concomitant glucocorticoids in TOZURA, a common-framework study programme of subcutaneous tocilizumab in rheumatoid arthritis. Rheumatology (Oxford) 2019;58:1056–1064. doi: 10.1093/rheumatology/key393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ridker PM, Libby P, MacFadyen JG, Thuren T, Ballantyne C, Fonseca F, Koenig W, Shimokawa H, Everett BM, Glynn RJ. Modulation of the interleukin-6 signalling pathway and incidence rates of atherosclerotic events and all-cause mortality: analyses from the Canakinumab Anti-Inflammatory Thrombosis Outcomes Study (CANTOS) Eur Heart J. 2018;39:3499–3507. doi: 10.1093/eurheartj/ehy310. [DOI] [PubMed] [Google Scholar]
  • 34.Eltoft A, Arntzen KA, Wilsgaard T, Mathiesen EB, Johnsen SH. Interleukin-6 is an independent predictor of progressive atherosclerosis in the carotid artery: the Tromsø study. Atherosclerosis. 2018;271:1–8. doi: 10.1016/j.atherosclerosis.2018.02.005. [DOI] [PubMed] [Google Scholar]
  • 35.Mantovani A, Dinarello CA, Molgora M, Garlanda C. Interleukin-1 and related cytokines in the regulation of inflammation and immunity. Immunity. 2019;50:778–795. doi: 10.1016/j.immuni.2019.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gerdes N, Sukhova GK, Libby P, Reynolds RS, Young JL, Schönbeck U. Expression of interleukin (IL)-18 and functional IL-18 receptor on human vascular endothelial cells, smooth muscle cells, and macrophages: implications for atherogenesis. J Exp Med. 2002;195:245–257. doi: 10.1084/jem.20011022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Akeda T, Yamanaka K, Tsuda K, Omoto Y, Gabazza EC, Mizutani H. CD8+ T cell granzyme B activates keratinocyte endogenous IL-18. Arch Dermatol Res. 2014;306:125–130. doi: 10.1007/s00403-013-1382-1. [DOI] [PubMed] [Google Scholar]
  • 38.Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci. 2019;20:3328. doi: 10.3390/ijms20133328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Blankenberg S, Tiret L, Bickel C, Peetz D, Cambien F, Meyer J, Rupprecht HJ. Interleukin-18 is a strong predictor of cardiovascular death in stable and unstable angina. Circulation. 2002;106:24–30. doi: 10.1161/01.cir.0000020546.30940.92. [DOI] [PubMed] [Google Scholar]
  • 40.Elhage R, Jawien J, Rudling M, Ljunggren HG, Takeda K, Akira S, Bayard F, Hansson GK. Reduced atherosclerosis in interleukin-18 deficient apolipoprotein E-knockout mice. Cardiovasc Res. 2003;59:234–240. doi: 10.1016/s0008-6363(03)00343-2. [DOI] [PubMed] [Google Scholar]
  • 41.Mallat Z, Corbaz A, Scoazec A, Graber P, Alouani S, Esposito B, Humbert Y, Chvatchko Y, Tedgui A. Interleukin-18/interleukin-18 binding protein signaling modulates atherosclerotic lesion development and stability. Circ Res. 2001;89:E41–45. doi: 10.1161/hh1901.098735. [DOI] [PubMed] [Google Scholar]
  • 42.Whitman SC, Ravisankar P, Daugherty A. Interleukin-18 enhances atherosclerosis in apolipoprotein E(-/-) mice through release of interferon-gamma. Circ Res. 2002;90:E34–E38. doi: 10.1161/hh0202.105292. [DOI] [PubMed] [Google Scholar]
  • 43.Bahrami A, Sathyapalan T, Sahebkar A. The role of interleukin-18 in the development and progression of atherosclerosis. Curr Med Chem. 2021;28:1757–1774. doi: 10.2174/0929867327666200427095830. [DOI] [PubMed] [Google Scholar]
  • 44.Ridker PM, MacFadyen JG, Thuren T, Libby P. Residual inflammatory risk associated with interleukin-18 and interleukin-6 after successful interleukin-1β inhibition with canakinumab: further rationale for the development of targeted anti-cytokine therapies for the treatment of atherothrombosis. Eur Heart J. 2020;41:2153–2163. doi: 10.1093/eurheartj/ehz542. [DOI] [PubMed] [Google Scholar]
  • 45.Duvallet E, Semerano L, Assier E, Falgarone G, Boissier MC. Interleukin-23: a key cytokine in inflammatory diseases. Ann Med. 2011;43:503–511. doi: 10.3109/07853890.2011.577093. [DOI] [PubMed] [Google Scholar]
  • 46.Fatkhullina AR, Peshkova IO, Dzutsev A, Aghayev T, McCulloch JA, Thovarai V, Badger JH, Vats R, Sundd P, Tang HY, Kossenkov AV, Hazen SL, Trinchieri G, Grivennikov SI, Koltsova EK. An interleukin-23-interleukin-22 axis regulates intestinal microbial homeostasis to protect from diet-induced atherosclerosis. Immunity. 2018;49:943–957. e949. doi: 10.1016/j.immuni.2018.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Abbas A, Gregersen I, Holm S, Daissormont I, Bjerkeli V, Krohg-Sørensen K, Skagen KR, Dahl TB, Russell D, Almås T, Bundgaard D, Alteheld LH, Rashidi A, Dahl CP, Michelsen AE, Biessen EA, Aukrust P, Halvorsen B, Skjelland M. Interleukin 23 levels are increased in carotid atherosclerosis: possible role for the interleukin 23/interleukin 17 axis. Stroke. 2015;46:793–799. doi: 10.1161/STROKEAHA.114.006516. [DOI] [PubMed] [Google Scholar]
  • 48.Langley RG, Papp K, Gottlieb AB, Krueger GG, Gordon KB, Williams D, Valdes J, Setze C, Strober B. Safety results from a pooled analysis of randomized, controlled phase II and III clinical trials and interim data from an open-label extension trial of the interleukin-12/23 monoclonal antibody, briakinumab, in moderate to severe psoriasis. J Eur Acad Dermatol Venereol. 2013;27:1252–1261. doi: 10.1111/j.1468-3083.2012.04705.x. [DOI] [PubMed] [Google Scholar]
  • 49.Poizeau F, Nowak E, Kerbrat S, Le Nautout B, Droitcourt C, Drici MD, Sbidian E, Guillot B, Bachelez H, Ait-Oufella H, Happe A, Oger E, Dupuy A. Association between early severe cardiovascular events and the initiation of treatment with the anti-interleukin 12/23p40 antibody ustekinumab. JAMA Dermatol. 2020;156:1208–1215. doi: 10.1001/jamadermatol.2020.2977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Papp KA, Blauvelt A, Bukhalo M, Gooderham M, Krueger JG, Lacour JP, Menter A, Philipp S, Sofen H, Tyring S, Berner BR, Visvanathan S, Pamulapati C, Bennett N, Flack M, Scholl P, Padula SJ. Risankizumab versus ustekinumab for moderate-to-severe plaque psoriasis. N Engl J Med. 2017;376:1551–1560. doi: 10.1056/NEJMoa1607017. [DOI] [PubMed] [Google Scholar]
  • 51.Elyasi A, Voloshyna I, Ahmed S, Kasselman LJ, Behbodikhah J, De Leon J, Reiss AB. The role of interferon-γ in cardiovascular disease: an update. Inflamm Res. 2020;69:975–988. doi: 10.1007/s00011-020-01382-6. [DOI] [PubMed] [Google Scholar]
  • 52.Mallat Z, Ait-Oufella H, Tedgui A. Regulatory T cell responses: potential role in the control of atherosclerosis. Curr Opin Lipidol. 2005;16:518–524. doi: 10.1097/01.mol.0000182532.11512.90. [DOI] [PubMed] [Google Scholar]
  • 53.McLaren JE, Ramji DP. Interferon gamma: a master regulator of atherosclerosis. Cytokine Growth Factor Rev. 2009;20:125–135. doi: 10.1016/j.cytogfr.2008.11.003. [DOI] [PubMed] [Google Scholar]
  • 54.Leon ML, Zuckerman SH. Gamma interferon: a central mediator in atherosclerosis. Inflamm Res. 2005;54:395–411. doi: 10.1007/s00011-005-1377-2. [DOI] [PubMed] [Google Scholar]
  • 55.Gupta S, Pablo AM, Jiang X, Wang N, Tall AR, Schindler C. IFN-gamma potentiates atherosclerosis in ApoE knock-out mice. J Clin Invest. 1997;99:2752–2761. doi: 10.1172/JCI119465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Whitman SC, Ravisankar P, Elam H, Daugherty A. Exogenous interferon-gamma enhances atherosclerosis in apolipoprotein E-/- mice. Am J Pathol. 2000;157:1819–1824. doi: 10.1016/s0002-9440(10)64820-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Karmaus PW, Shi M, Perl S, Biancotto A, Candia J, Cheung F, Kotliarov Y, Young N, Fessler MB. Effects of rosuvastatin on the immune system in healthy volunteers with normal serum cholesterol. JCI Insight. 2019;4:e131530. doi: 10.1172/jci.insight.131530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Momtazi-Borojeni AA, Jaafari MR, Badiee A, Banach M, Sahebkar A. Therapeutic effect of nanoliposomal PCSK9 vaccine in a mouse model of atherosclerosis. BMC Med. 2019;17:223. doi: 10.1186/s12916-019-1457-8. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 59.Zhou J, Qin L, Yi T, Ali R, Li Q, Jiao Y, Li G, Tobiasova Z, Huang Y, Zhang J, Yun JJ, Sadeghi MM, Giordano FJ, Pober JS, Tellides G. Interferon-γ-mediated allograft rejection exacerbates cardiovascular disease of hyperlipidemic murine transplant recipients. Circ Res. 2015;117:943–955. doi: 10.1161/CIRCRESAHA.115.306932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wang CH, Shi HH, Chen LH, Li XL, Cao GL, Hu XF. Identification of key lncRNAs associated with atherosclerosis progression based on public datasets. Front Genet. 2019;10:123. doi: 10.3389/fgene.2019.00123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Bruen R, Fitzsimons S, Belton O. miR-155 in the resolution of atherosclerosis. Front Pharmacol. 2019;10:463. doi: 10.3389/fphar.2019.00463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Trotta R, Chen L, Ciarlariello D, Josyula S, Mao C, Costinean S, Yu L, Butchar JP, Tridandapani S, Croce CM, Caligiuri MA. miR-155 regulates IFN-γ production in natural killer cells. Blood. 2012;119:3478–3485. doi: 10.1182/blood-2011-12-398099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Casey KA, Smith MA, Sinibaldi D, Seto NL, Playford MP, Wang X, Carlucci PM, Wang L, Illei G, Yu B, Wang S, Remaley AT, Mehta NN, Kaplan MJ, White WI. Modulation of cardiometabolic disease markers by type i interferon inhibition in systemic lupus erythematosus. Arthritis Rheumatol. 2021;73:459–471. doi: 10.1002/art.41518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Browne SK. Anticytokine autoantibody-associated immunodeficiency. Annu Rev Immunol. 2014;32:635–657. doi: 10.1146/annurev-immunol-032713-120222. [DOI] [PubMed] [Google Scholar]
  • 65.Hjuler KF, Böttcher M, Vestergaard C, Deleuran M, Raaby L, Bøtker HE, Iversen L, Kragballe K. Increased prevalence of coronary artery disease in severe psoriasis and severe atopic dermatitis. Am J Med. 2015;128:1325–1334. e1322. doi: 10.1016/j.amjmed.2015.05.041. [DOI] [PubMed] [Google Scholar]
  • 66.Bissonnette R, Harel F, Krueger JG, Guertin MC, Chabot-Blanchet M, Gonzalez J, Maari C, Delorme I, Lynde CW, Tardif JC. TNF-α antagonist and vascular inflammation in patients with psoriasis vulgaris: a randomized placebo-controlled study. J Invest Dermatol. 2017;137:1638–1645. doi: 10.1016/j.jid.2017.02.977. [DOI] [PubMed] [Google Scholar]
  • 67.Bissonnette R, Tardif JC, Harel F, Pressacco J, Bolduc C, Guertin MC. Effects of the tumor necrosis factor-α antagonist adalimumab on arterial inflammation assessed by positron emission tomography in patients with psoriasis: results of a randomized controlled trial. Circ Cardiovasc Imaging. 2013;6:83–90. doi: 10.1161/CIRCIMAGING.112.975730. [DOI] [PubMed] [Google Scholar]
  • 68.Ridker PM, Thuren T, Zalewski A, Libby P. Interleukin-1β inhibition and the prevention of recurrent cardiovascular events: rationale and design of the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS) Am Heart J. 2011;162:597–605. doi: 10.1016/j.ahj.2011.06.012. [DOI] [PubMed] [Google Scholar]
  • 69.Serbina NV, Pamer EG. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat Immunol. 2006;7:311–317. doi: 10.1038/ni1309. [DOI] [PubMed] [Google Scholar]
  • 70.Gschwandtner M, Derler R, Midwood KS. More than just attractive: how CCL2 influences myeloid cell behavior beyond chemotaxis. Front Immunol. 2019;10:2759. doi: 10.3389/fimmu.2019.02759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bianconi V, Sahebkar A, Atkin SL, Pirro M. The regulation and importance of monocyte chemoattractant protein-1. Curr Opin Hematol. 2018;25:44–51. doi: 10.1097/MOH.0000000000000389. [DOI] [PubMed] [Google Scholar]
  • 72.Sierra-Filardi E, Nieto C, Domínguez-Soto A, Barroso R, Sánchez-Mateos P, Puig-Kroger A, López-Bravo M, Joven J, Ardavín C, Rodríguez-Fernández JL, Sánchez-Torres C, Mellado M, Corbí AL. CCL2 shapes macrophage polarization by GM-CSF and M-CSF: identification of CCL2/CCR2-dependent gene expression profile. J Immunol. 2014;192:3858–3867. doi: 10.4049/jimmunol.1302821. [DOI] [PubMed] [Google Scholar]
  • 73.Silva AR, Pacheco P, Vieira-de-Abreu A, Maya-Monteiro CM, D’Alegria B, Magalhães KG, de Assis EF, Bandeira-Melo C, Castro-Faria-Neto HC, Bozza PT. Lipid bodies in oxidized LDL-induced foam cells are leukotriene-synthesizing organelles: a MCP-1/CCL2 regulated phenomenon. Biochim Biophys Acta. 2009;1791:1066–1075. doi: 10.1016/j.bbalip.2009.06.004. [DOI] [PubMed] [Google Scholar]
  • 74.Boring L, Gosling J, Cleary M, Charo IF. Decreased lesion formation in CCR2-/- mice reveals a role for chemokines in the initiation of atherosclerosis. Nature. 1998;394:894–897. doi: 10.1038/29788. [DOI] [PubMed] [Google Scholar]
  • 75.Winter C, Silvestre-Roig C, Ortega-Gomez A, Lemnitzer P, Poelman H, Schumski A, Winter J, Drechsler M, de Jong R, Immler R, Sperandio M, Hristov M, Zeller T, Nicolaes GAF, Weber C, Viola JR, Hidalgo A, Scheiermann C, Soehnlein O. Chrono-pharmacological targeting of the CCL2-CCR2 axis ameliorates atherosclerosis. Cell Metab. 2018;28:175–182. e175. doi: 10.1016/j.cmet.2018.05.002. [DOI] [PubMed] [Google Scholar]
  • 76.Georgakis MK, Gill D, Rannikmäe K, Traylor M, Anderson CD, Lee JM, Kamatani Y, Hopewell JC, Worrall BB, Bernhagen J, Sudlow CLM, Malik R, Dichgans M. Genetically determined levels of circulating cytokines and risk of stroke. Circulation. 2019;139:256–268. doi: 10.1161/CIRCULATIONAHA.118.035905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Dhawan L, Liu B, Blaxall BC, Taubman MB. A novel role for the glucocorticoid receptor in the regulation of monocyte chemoattractant protein-1 mRNA stability. J Biol Chem. 2007;282:10146–10152. doi: 10.1074/jbc.M605925200. [DOI] [PubMed] [Google Scholar]
  • 78.Romano M, Diomede L, Sironi M, Massimiliano L, Sottocorno M, Polentarutti N, Guglielmotti A, Albani D, Bruno A, Fruscella P, Salmona M, Vecchi A, Pinza M, Mantovani A. Inhibition of monocyte chemotactic protein-1 synthesis by statins. Lab Invest. 2000;80:1095–1100. doi: 10.1038/labinvest.3780115. [DOI] [PubMed] [Google Scholar]
  • 79.Tucker B, Kurup R, Barraclough J, Henriquez R, Cartland S, Arnott C, Misra A, Martínez G, Kavurma M, Patel S. Colchicine as a novel therapy for suppressing chemokine production in patients with an acute coronary syndrome: a pilot study. Clin Ther. 2019;41:2172–2181. doi: 10.1016/j.clinthera.2019.07.015. [DOI] [PubMed] [Google Scholar]
  • 80.Pienta KJ, Machiels JP, Schrijvers D, Alekseev B, Shkolnik M, Crabb SJ, Li S, Seetharam S, Puchalski TA, Takimoto C, Elsayed Y, Dawkins F, de Bono JS. Phase 2 study of carlumab (CNTO 888), a human monoclonal antibody against CC-chemokine ligand 2 (CCL2), in metastatic castration-resistant prostate cancer. Invest New Drugs. 2013;31:760–768. doi: 10.1007/s10637-012-9869-8. [DOI] [PubMed] [Google Scholar]
  • 81.Brana I, Calles A, LoRusso PM, Yee LK, Puchalski TA, Seetharam S, Zhong B, de Boer CJ, Tabernero J, Calvo E. Carlumab, an anti-C-C chemokine ligand 2 monoclonal antibody, in combination with four chemotherapy regimens for the treatment of patients with solid tumors: an open-label, multicenter phase 1b study. Target Oncol. 2015;10:111–123. doi: 10.1007/s11523-014-0320-2. [DOI] [PubMed] [Google Scholar]
  • 82.White GE, Greaves DR. Fractalkine: a survivor’s guide: chemokines as antiapoptotic mediators. Arterioscler Thromb Vasc Biol. 2012;32:589–594. doi: 10.1161/ATVBAHA.111.237412. [DOI] [PubMed] [Google Scholar]
  • 83.Liu H, Jiang D. Fractalkine/CX3CR1 and atherosclerosis. Clin Chim Acta. 2011;412:1180–1186. doi: 10.1016/j.cca.2011.03.036. [DOI] [PubMed] [Google Scholar]
  • 84.Elliott MR, Koster KM, Murphy PS. Efferocytosis signaling in the regulation of macrophage inflammatory responses. J Immunol. 2017;198:1387–1394. doi: 10.4049/jimmunol.1601520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Landsman L, Bar-On L, Zernecke A, Kim KW, Krauthgamer R, Shagdarsuren E, Lira SA, Weissman IL, Weber C, Jung S. CX3CR1 is required for monocyte homeostasis and atherogenesis by promoting cell survival. Blood. 2009;113:963–972. doi: 10.1182/blood-2008-07-170787. [DOI] [PubMed] [Google Scholar]
  • 86.Lucas AD, Bursill C, Guzik TJ, Sadowski J, Channon KM, Greaves DR. Smooth muscle cells in human atherosclerotic plaques express the fractalkine receptor CX3CR1 and undergo chemotaxis to the CX3C chemokine fractalkine (CX3CL1) Circulation. 2003;108:2498–2504. doi: 10.1161/01.CIR.0000097119.57756.EF. [DOI] [PubMed] [Google Scholar]
  • 87.Noels H, Weber C, Koenen RR. Chemokines as therapeutic targets in cardiovascular disease. Arterioscler Thromb Vasc Biol. 2019;39:583–592. doi: 10.1161/ATVBAHA.118.312037. [DOI] [PubMed] [Google Scholar]
  • 88.Postea O, Vasina EM, Cauwenberghs S, Projahn D, Liehn EA, Lievens D, Theelen W, Kramp BK, Butoi ED, Soehnlein O, Heemskerk JW, Ludwig A, Weber C, Koenen RR. Contribution of platelet CX(3)CR1 to platelet-monocyte complex formation and vascular recruitment during hyperlipidemia. Arterioscler Thromb Vasc Biol. 2012;32:1186–1193. doi: 10.1161/ATVBAHA.111.243485. [DOI] [PubMed] [Google Scholar]
  • 89.Gleissner CA, von Hundelshausen P, Ley K. Platelet chemokines in vascular disease. Arterioscler Thromb Vasc Biol. 2008;28:1920–1927. doi: 10.1161/ATVBAHA.108.169417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Poupel L, Boissonnas A, Hermand P, Dorgham K, Guyon E, Auvynet C, Charles FS, Lesnik P, Deterre P, Combadiere C. Pharmacological inhibition of the chemokine receptor, CX3CR1, reduces atherosclerosis in mice. Arterioscler Thromb Vasc Biol. 2013;33:2297–2305. doi: 10.1161/ATVBAHA.112.300930. [DOI] [PubMed] [Google Scholar]
  • 91.Bernhagen J, Krohn R, Lue H, Gregory JL, Zernecke A, Koenen RR, Dewor M, Georgiev I, Schober A, Leng L, Kooistra T, Fingerle-Rowson G, Ghezzi P, Kleemann R, McColl SR, Bucala R, Hickey MJ, Weber C. MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med. 2007;13:587–596. doi: 10.1038/nm1567. [DOI] [PubMed] [Google Scholar]
  • 92.Amin MA, Haas CS, Zhu K, Mansfield PJ, Kim MJ, Lackowski NP, Koch AE. Migration inhibitory factor up-regulates vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 via Src, PI3 kinase, and NFkappaB. Blood. 2006;107:2252–2261. doi: 10.1182/blood-2005-05-2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sinitski D, Kontos C, Krammer C, Asare Y, Kapurniotu A, Bernhagen J. Macrophage migration inhibitory factor (MIF)-based therapeutic concepts in atherosclerosis and inflammation. Thromb Haemost. 2019;119:553–566. doi: 10.1055/s-0039-1677803. [DOI] [PubMed] [Google Scholar]
  • 94.Pan JH, Sukhova GK, Yang JT, Wang B, Xie T, Fu H, Zhang Y, Satoskar AR, David JR, Metz CN, Bucala R, Fang K, Simon DI, Chapman HA, Libby P, Shi GP. Macrophage migration inhibitory factor deficiency impairs atherosclerosis in low-density lipoprotein receptor-deficient mice. Circulation. 2004;109:3149–3153. doi: 10.1161/01.CIR.0000134704.84454.D2. [DOI] [PubMed] [Google Scholar]
  • 95.Mitchell RA, Liao H, Chesney J, Fingerle-Rowson G, Baugh J, David J, Bucala R. Macrophage migration inhibitory factor (MIF) sustains macrophage proinflammatory function by inhibiting p53: regulatory role in the innate immune response. Proc Natl Acad Sci U S A. 2002;99:345–350. doi: 10.1073/pnas.012511599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Verschuren L, Kooistra T, Bernhagen J, Voshol PJ, Ouwens DM, van Erk M, de Vries-van der Weij J, Leng L, van Bockel JH, van Dijk KW, Fingerle-Rowson G, Bucala R, Kleemann R. MIF deficiency reduces chronic inflammation in white adipose tissue and impairs the development of insulin resistance, glucose intolerance, and associated atherosclerotic disease. Circ Res. 2009;105:99–107. doi: 10.1161/CIRCRESAHA.109.199166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Burger-Kentischer A, Göbel H, Kleemann R, Zernecke A, Bucala R, Leng L, Finkelmeier D, Geiger G, Schaefer HE, Schober A, Weber C, Brunner H, Rütten H, Ihling C, Bernhagen J. Reduction of the aortic inflammatory response in spontaneous atherosclerosis by blockade of macrophage migration inhibitory factor (MIF) Atherosclerosis. 2006;184:28–38. doi: 10.1016/j.atherosclerosis.2005.03.028. [DOI] [PubMed] [Google Scholar]
  • 98.Fox RJ, Coffey CS, Conwit R, Cudkowicz ME, Gleason T, Goodman A, Klawiter EC, Matsuda K, McGovern M, Naismith RT, Ashokkumar A, Barnes J, Ecklund D, Klingner E, Koepp M, Long JD, Natarajan S, Thornell B, Yankey J, Bermel RA, Debbins JP, Huang X, Jagodnik P, Lowe MJ, Nakamura K, Narayanan S, Sakaie KE, Thoomukuntla B, Zhou X, Krieger S, Alvarez E, Apperson M, Bashir K, Cohen BA, Coyle PK, Delgado S, Dewitt LD, Flores A, Giesser BS, Goldman MD, Jubelt B, Lava N, Lynch SG, Moses H, Ontaneda D, Perumal JS, Racke M, Repovic P, Riley CS, Severson C, Shinnar S, Suski V, Weinstock-Guttman B, Yadav V, Zabeti A. Phase 2 trial of ibudilast in progressive multiple sclerosis. N Engl J Med. 2018;379:846–855. doi: 10.1056/NEJMoa1803583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Mahalingam D, Patel MR, Sachdev JC, Hart LL, Halama N, Ramanathan RK, Sarantopoulos J, Völkel D, Youssef A, de Jong FA, Tsimberidou AM. Phase I study of imalumab (BAX69), a fully human recombinant antioxidized macrophage migration inhibitory factor antibody in advanced solid tumours. Br J Clin Pharmacol. 2020;86:1836–1848. doi: 10.1111/bcp.14289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.von Hundelshausen P, Weber KS, Huo Y, Proudfoot AE, Nelson PJ, Ley K, Weber C. RANTES deposition by platelets triggers monocyte arrest on inflamed and atherosclerotic endothelium. Circulation. 2001;103:1772–1777. doi: 10.1161/01.cir.103.13.1772. [DOI] [PubMed] [Google Scholar]
  • 101.Zernecke A, Weber C. Chemokines in atherosclerosis: proceedings resumed. Arterioscler Thromb Vasc Biol. 2014;34:742–750. doi: 10.1161/ATVBAHA.113.301655. [DOI] [PubMed] [Google Scholar]
  • 102.Li J, McArdle S, Gholami A, Kimura T, Wolf D, Gerhardt T, Miller J, Weber C, Ley K. CCR5+T-bet+FoxP3+ effector CD4 T cells drive atherosclerosis. Circ Res. 2016;118:1540–1552. doi: 10.1161/CIRCRESAHA.116.308648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.von Hundelshausen P, Agten SM, Eckardt V, Blanchet X, Schmitt MM, Ippel H, Neideck C, Bidzhekov K, Leberzammer J, Wichapong K, Faussner A, Drechsler M, Grommes J, van Geffen JP, Li H, Ortega-Gomez A, Megens RT, Naumann R, Dijkgraaf I, Nicolaes GA, Döring Y, Soehnlein O, Lutgens E, Heemskerk JW, Koenen RR, Mayo KH, Hackeng TM, Weber C. Chemokine interactome mapping enables tailored intervention in acute and chronic inflammation. Sci Transl Med. 2017;9:eaah6650. doi: 10.1126/scitranslmed.aah6650. [DOI] [PubMed] [Google Scholar]
  • 104.Veillard NR, Kwak B, Pelli G, Mulhaupt F, James RW, Proudfoot AE, Mach F. Antagonism of RANTES receptors reduces atherosclerotic plaque formation in mice. Circ Res. 2004;94:253–261. doi: 10.1161/01.RES.0000109793.17591.4E. [DOI] [PubMed] [Google Scholar]
  • 105.Braunersreuther V, Zernecke A, Arnaud C, Liehn EA, Steffens S, Shagdarsuren E, Bidzhekov K, Burger F, Pelli G, Luckow B, Mach F, Weber C. Ccr5 but not Ccr1 deficiency reduces development of diet-induced atherosclerosis in mice. Arterioscler Thromb Vasc Biol. 2007;27:373–379. doi: 10.1161/01.ATV.0000253886.44609.ae. [DOI] [PubMed] [Google Scholar]
  • 106.Blanchet X, Cesarek K, Brandt J, Herwald H, Teupser D, Küchenhoff H, Karshovska E, Mause SF, Siess W, Wasmuth H, Soehnlein O, Koenen RR, Weber C, von Hundelshausen P. Inflammatory role and prognostic value of platelet chemokines in acute coronary syndrome. Thromb Haemost. 2014;112:1277–1287. doi: 10.1160/TH14-02-0139. [DOI] [PubMed] [Google Scholar]
  • 107.Cipriani S, Francisci D, Mencarelli A, Renga B, Schiaroli E, D’Amore C, Baldelli F, Fiorucci S. Efficacy of the CCR5 antagonist maraviroc in reducing early, ritonavir-induced atherogenesis and advanced plaque progression in mice. Circulation. 2013;127:2114–2124. doi: 10.1161/CIRCULATIONAHA.113.001278. [DOI] [PubMed] [Google Scholar]
  • 108.Maggi P, Bruno G, Perilli F, Saracino A, Volpe A, Santoro C, Ladisa N, Angarano G. Effects of therapy with maraviroc on the carotid intima media thickness in HIV-1/HCV co-infected patients. In Vivo. 2017;31:125–131. doi: 10.21873/invivo.11035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.D’Antoni ML, Mitchell BI, McCurdy S, Byron MM, Ogata-Arakaki D, Chow D, Mehta NN, Boisvert WA, Lefebvre E, Shikuma CM, Ndhlovu LC, Baumer Y. Cenicriviroc inhibits trans-endothelial passage of monocytes and is associated with impaired E-selectin expression. J Leukoc Biol. 2018;104:1241–1252. doi: 10.1002/JLB.5A0817-328RRR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Scheuerer B, Ernst M, Dürrbaum-Landmann I, Fleischer J, Grage-Griebenow E, Brandt E, Flad HD, Petersen F. The CXC-chemokine platelet factor 4 promotes monocyte survival and induces monocyte differentiation into macrophages. Blood. 2000;95:1158–1166. [PubMed] [Google Scholar]
  • 111.Nassar T, Sachais BS, Akkawi S, Kowalska MA, Bdeir K, Leitersdorf E, Hiss E, Ziporen L, Aviram M, Cines D, Poncz M, Higazi AA. Platelet factor 4 enhances the binding of oxidized low-density lipoprotein to vascular wall cells. J Biol Chem. 2003;278:6187–6193. doi: 10.1074/jbc.M208894200. [DOI] [PubMed] [Google Scholar]
  • 112.Sarabi A, Kramp BK, Drechsler M, Hackeng TM, Soehnlein O, Weber C, Koenen RR, Von Hundelshausen P. CXCL4L1 inhibits angiogenesis and induces undirected endothelial cell migration without affecting endothelial cell proliferation and monocyte recruitment. J Thromb Haemost. 2011;9:209–219. doi: 10.1111/j.1538-7836.2010.04119.x. [DOI] [PubMed] [Google Scholar]
  • 113.Li N. CD4+ T cells in atherosclerosis: regulation by platelets. Thromb Haemost. 2013;109:980–990. doi: 10.1160/TH12-11-0819. [DOI] [PubMed] [Google Scholar]
  • 114.Gleissner CA, Shaked I, Erbel C, Böckler D, Katus HA, Ley K. CXCL4 downregulates the atheroprotective hemoglobin receptor CD163 in human macrophages. Circ Res. 2010;106:203–211. doi: 10.1161/CIRCRESAHA.109.199505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Sachais BS, Turrentine T, Dawicki McKenna JM, Rux AH, Rader D, Kowalska MA. Elimination of platelet factor 4 (PF4) from platelets reduces atherosclerosis in C57Bl/6 and apoE-/- mice. Thromb Haemost. 2007;98:1108–1113. [PubMed] [Google Scholar]
  • 116.Pitsilos S, Hunt J, Mohler ER, Prabhakar AM, Poncz M, Dawicki J, Khalapyan TZ, Wolfe ML, Fairman R, Mitchell M, Carpenter J, Golden MA, Cines DB, Sachais BS. Platelet factor 4 localization in carotid atherosclerotic plaques: correlation with clinical parameters. Thromb Haemost. 2003;90:1112–1120. doi: 10.1160/TH03-02-0069. [DOI] [PubMed] [Google Scholar]
  • 117.Vajen T, Koenen RR, Werner I, Staudt M, Projahn D, Curaj A, Sönmez TT, Simsekyilmaz S, Schumacher D, Möllmann J, Hackeng TM, Hundelshausen PV, Weber C, Liehn EA. Blocking CCL5-CXCL4 heteromerization preserves heart function after myocardial infarction by attenuating leukocyte recruitment and NETosis. Sci Rep. 2018;8:10647. doi: 10.1038/s41598-018-29026-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Linke B, Meyer Dos Santos S, Picard-Willems B, Keese M, Harder S, Geisslinger G, Scholich K. CXCL16/CXCR6-mediated adhesion of human peripheral blood mononuclear cells to inflamed endothelium. Cytokine. 2019;122:154081. doi: 10.1016/j.cyto.2017.06.008. [DOI] [PubMed] [Google Scholar]
  • 119.Gutwein P, Abdel-Bakky MS, Schramme A, Doberstein K, Kämpfer-Kolb N, Amann K, Hauser IA, Obermüller N, Bartel C, Abdel-Aziz AA, El Sayed el SM, Pfeilschifter J. CXCL16 is expressed in podocytes and acts as a scavenger receptor for oxidized low-density lipoprotein. Am J Pathol. 2009;174:2061–2072. doi: 10.2353/ajpath.2009.080960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Meyer Dos Santos S, Blankenbach K, Scholich K, Dörr A, Monsefi N, Keese M, Linke B, Deckmyn H, Nelson K, Harder S. Platelets from flowing blood attach to the inflammatory chemokine CXCL16 expressed in the endothelium of the human vessel wall. Thromb Haemost. 2015;114:297–312. doi: 10.1160/TH14-11-0911. [DOI] [PubMed] [Google Scholar]
  • 121.Seizer P, Stellos K, Selhorst G, Krämer BF, Lang MR, Gawaz M, May AE. CXCL16 is a novel scavenger receptor on platelets and is associated with acute coronary syndrome. Thromb Haemost. 2011;105:1112–1114. doi: 10.1160/TH10-10-0689. [DOI] [PubMed] [Google Scholar]
  • 122.Jansson AM, Aukrust P, Ueland T, Smith C, Omland T, Hartford M, Caidahl K. Soluble CXCL16 predicts long-term mortality in acute coronary syndromes. Circulation. 2009;119:3181–3188. doi: 10.1161/CIRCULATIONAHA.108.806877. [DOI] [PubMed] [Google Scholar]
  • 123.Laugsand LE, Åsvold BO, Vatten LJ, Janszky I, Platou C, Michelsen AE, Arain F, Damås JK, Aukrust P, Ueland T. Soluble CXCL16 and risk of myocardial infarction: the HUNT study in Norway. Atherosclerosis. 2016;244:188–194. doi: 10.1016/j.atherosclerosis.2015.11.022. [DOI] [PubMed] [Google Scholar]
  • 124.Zernecke A, Bidzhekov K, Noels H, Shagdarsuren E, Gan L, Denecke B, Hristov M, Köppel T, Jahantigh MN, Lutgens E, Wang S, Olson EN, Schober A, Weber C. Delivery of microRNA-126 by apoptotic bodies induces CXCL12-dependent vascular protection. Sci Signal. 2009;2:ra81. doi: 10.1126/scisignal.2000610. [DOI] [PubMed] [Google Scholar]
  • 125.Koenen J, Bachelerie F, Balabanian K, Schlecht-Louf G, Gallego C. Atypical chemokine receptor 3 (ACKR3): a comprehensive overview of its expression and potential roles in the immune system. Mol Pharmacol. 2019;96:809–818. doi: 10.1124/mol.118.115329. [DOI] [PubMed] [Google Scholar]
  • 126.Chatterjee M, von Ungern-Sternberg SN, Seizer P, Schlegel F, Büttcher M, Sindhu NA, Müller S, Mack A, Gawaz M. Platelet-derived CXCL12 regulates monocyte function, survival, differentiation into macrophages and foam cells through differential involvement of CXCR4-CXCR7. Cell Death Dis. 2015;6:e1989. doi: 10.1038/cddis.2015.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Akhtar S, Gremse F, Kiessling F, Weber C, Schober A. CXCL12 promotes the stabilization of atherosclerotic lesions mediated by smooth muscle progenitor cells in Apoe-deficient mice. Arterioscler Thromb Vasc Biol. 2013;33:679–686. doi: 10.1161/ATVBAHA.112.301162. [DOI] [PubMed] [Google Scholar]
  • 128.Schall N, Daubeuf F, Marsol C, Gizzi P, Frossard N, Bonnet D, Galzi JL, Muller S. A selective neutraligand for CXCL12/SDF-1α with beneficial regulatory functions in MRL/Lpr lupus prone mice. Front Pharmacol. 2021;12:752194. doi: 10.3389/fphar.2021.752194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Gilbert J, Lekstrom-Himes J, Donaldson D, Lee Y, Hu M, Xu J, Wyant T, Davidson M. Effect of CC chemokine receptor 2 CCR2 blockade on serum C-reactive protein in individuals at atherosclerotic risk and with a single nucleotide polymorphism of the monocyte chemoattractant protein-1 promoter region. Am J Cardiol. 2011;107:906–911. doi: 10.1016/j.amjcard.2010.11.005. [DOI] [PubMed] [Google Scholar]
  • 130.Francisci D, Pirro M, Schiaroli E, Mannarino MR, Cipriani S, Bianconi V, Alunno A, Bagaglia F, Bistoni O, Falcinelli E, Bury L, Gerli R, Mannarino E, De Caterina R, Baldelli F. Maraviroc intensification modulates atherosclerotic progression in hiv-suppressed patients at high cardiovascular risk. A randomized, crossover pilot study. Open Forum Infect Dis. 2019;6:ofz112. doi: 10.1093/ofid/ofz112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Colombo A, Basavarajaiah S, Limbruno U, Picchi A, Lettieri C, Valgimigli M, Sciahbasi A, Prati F, Calabresi M, Pierucci D, Guglielmotti A. A double-blind randomised study to evaluate the efficacy and safety of bindarit in preventing coronary stent restenosis. EuroIntervention. 2016;12:e1385–e1394. doi: 10.4244/EIJY15M12_03. [DOI] [PubMed] [Google Scholar]
  • 132.Chung ES, Miller L, Patel AN, Anderson RD, Mendelsohn FO, Traverse J, Silver KH, Shin J, Ewald G, Farr MJ, Anwaruddin S, Plat F, Fisher SJ, AuWerter AT, Pastore JM, Aras R, Penn MS. Changes in ventricular remodelling and clinical status during the year following a single administration of stromal cell-derived factor-1 non-viral gene therapy in chronic ischaemic heart failure patients: the STOP-HF randomized phase II trial. Eur Heart J. 2015;36:2228–2238. doi: 10.1093/eurheartj/ehv254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Hammad TA, Rundback J, Bunte M, Miller L, Patel PD, Sadanandan S, Fitzgerald M, Pastore J, Kashyap V, Henry TD, Shishehbor MH. Stromal cell-derived factor-1 plasmid treatment for patients with peripheral artery disease (STOP-PAD) trial: six-month results. J Endovasc Ther. 2020;27:669–675. doi: 10.1177/1526602820919951. [DOI] [PubMed] [Google Scholar]
  • 134.Han X, Boisvert WA. Interleukin-10 protects against atherosclerosis by modulating multiple atherogenic macrophage function. Thromb Haemost. 2015;113:505–512. doi: 10.1160/TH14-06-0509. [DOI] [PubMed] [Google Scholar]
  • 135.Bu T, Li Z, Hou Y, Sun W, Zhang R, Zhao L, Wei M, Yang G, Yuan L. Exosome-mediated delivery of inflammation-responsive Il-10 mRNA for controlled atherosclerosis treatment. Theranostics. 2021;11:9988–10000. doi: 10.7150/thno.64229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Munjal A, Khandia R. Atherosclerosis: orchestrating cells and biomolecules involved in its activation and inhibition. Adv Protein Chem Struct Biol. 2020;120:85–122. doi: 10.1016/bs.apcsb.2019.11.002. [DOI] [PubMed] [Google Scholar]
  • 137.Statement of Retraction: 1,25(OH)2D3 improves blood lipid metabolism, liver function, and atherosclerosis by constraining the TGF-β/Smad signaling pathway in rats with hyperlipidemia. Cell Cycle. 2022;21:1437. doi: 10.1080/15384101.2022.2066295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Jiang H, Ruan Z, Wang Z, Wu B, Gao Z, Ye X, Lu X. Simvastatin reduces atherosclerotic plaques and endothelial inflammatory response in atherosclerosis rats through TGF-β/Smad pathway. Minerva Med. 2020;111:504–507. doi: 10.23736/S0026-4806.19.06119-6. [DOI] [PubMed] [Google Scholar]
  • 139.Liu D, Song J, Ji X, Liu Z, Li T, Hu B. PRDM16 upregulation induced by microRNA-448 inhibition alleviates atherosclerosis via the TGF-β signaling pathway inactivation. Front Physiol. 2020;11:846. doi: 10.3389/fphys.2020.00846. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 140.Potekhina AV, Pylaeva E, Provatorov S, Ruleva N, Masenko V, Noeva E, Krasnikova T, Arefieva T. Treg/Th17 balance in stable CAD patients with different stages of coronary atherosclerosis. Atherosclerosis. 2015;238:17–21. doi: 10.1016/j.atherosclerosis.2014.10.088. [DOI] [PubMed] [Google Scholar]
  • 141.Czerkinsky C, Holmgren J. Mucosal delivery routes for optimal immunization: targeting immunity to the right tissues. Curr Top Microbiol Immunol. 2012;354:1–18. doi: 10.1007/82_2010_112. [DOI] [PubMed] [Google Scholar]
  • 142.Fröbert O, Götberg M, Erlinge D, Akhtar Z, Christiansen EH, MacIntyre CR, Oldroyd KG, Motovska Z, Erglis A, Moer R, Hlinomaz O, Jakobsen L, Engstrøm T, Jensen LO, Fallesen CO, Jensen SE, Angerås O, Calais F, Kåregren A, Lauermann J, Mokhtari A, Nilsson J, Persson J, Stalby P, Islam A, Rahman A, Malik F, Choudhury S, Collier T, Pocock SJ, Pernow J. Influenza vaccination after myocardial infarction: a randomized, double-blind, placebo-controlled, multicenter trial. Circulation. 2021;144:1476–1484. doi: 10.1161/CIRCULATIONAHA.121.057042. [DOI] [PubMed] [Google Scholar]
  • 143.Dietrich T, Hucko T, Schneemann C, Neumann M, Menrad A, Willuda J, Atrott K, Stibenz D, Fleck E, Graf K, Menssen HD. Local delivery of IL-2 reduces atherosclerosis via expansion of regulatory T cells. Atherosclerosis. 2012;220:329–336. doi: 10.1016/j.atherosclerosis.2011.09.050. [DOI] [PubMed] [Google Scholar]
  • 144.Dinh TN, Kyaw TS, Kanellakis P, To K, Tipping P, Toh BH, Bobik A, Agrotis A. Cytokine therapy with interleukin-2/anti-interleukin-2 monoclonal antibody complexes expands CD4+CD25+Foxp3+ regulatory T cells and attenuates development and progression of atherosclerosis. Circulation. 2012;126:1256–1266. doi: 10.1161/CIRCULATIONAHA.112.099044. [DOI] [PubMed] [Google Scholar]
  • 145.Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, Kuder JF, Wang H, Liu T, Wasserman SM, Sever PS, Pedersen TR. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med. 2017;376:1713–1722. doi: 10.1056/NEJMoa1615664. [DOI] [PubMed] [Google Scholar]
  • 146.Pradhan AD, Aday AW, Rose LM, Ridker PM. Residual inflammatory risk on treatment with PCSK9 inhibition and statin therapy. Circulation. 2018;138:141–149. doi: 10.1161/CIRCULATIONAHA.118.034645. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Translational Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES