Next Article in Journal
Detection of Circulating Tumor Cells Using the Attune NxT
Next Article in Special Issue
Microarray Expression Profile of Myricetin-Treated THP-1 Macrophages Exhibits Alterations in Atherosclerosis-Related Regulator Molecules and LXR/RXR Pathway
Previous Article in Journal
Functionalization of Polylactide with Multiple Tetraphenyethane Inifer Groups to Form PLA Block Copolymers with Vinyl Monomers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The IL-1 Family and Its Role in Atherosclerosis

by
Leticia González
1,2,†,
Katherine Rivera
1,3,†,
Marcelo E. Andia
1,2 and
Gonzalo Martínez Rodriguez
4,*
1
Centro de Imágenes Biomédicas—Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
2
Instituto Milenio de Ingeniería e Inteligencia Artificial Para la Salud, iHEALTH, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
3
Programa de Doctorado en Ciencias Médicas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
4
División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(1), 17; https://doi.org/10.3390/ijms24010017
Submission received: 9 November 2022 / Revised: 9 December 2022 / Accepted: 15 December 2022 / Published: 20 December 2022

Abstract

:
The IL-1 superfamily of cytokines is a central regulator of immunity and inflammation. The family is composed of 11 cytokines (with agonist, antagonist, and anti-inflammatory properties) and 10 receptors, all tightly regulated through decoy receptor, receptor antagonists, and signaling inhibitors. Inflammation not only is an important physiological response against infection and injury but also plays a central role in atherosclerosis development. Several clinical association studies along with experimental studies have implicated the IL-1 superfamily of cytokines and its receptors in the pathogenesis of cardiovascular disease. Here, we summarize the key features of the IL-1 family, its role in immunity and disease, and how it helps shape the development of atherosclerosis.

1. Introduction

Though cardiovascular disease (CVD) remains one of the top causes of death worldwide, an epidemiological transition has been observed, where disease is no longer concentrated in industrialized countries, but increasingly affects individuals from less developed regions [1]. The classic assumption of the coronary patient—white middle-aged man with hypercholesterolemia and hypertension—is no longer the reality of the population currently affected. This stresses the importance of understanding the mechanisms governing the disease, to develop better-suited therapies and guidelines for the management of CVD burden.
The common underlying cause behind the manifestations of CVD disease is atherosclerosis. The chronic accumulation of lipids and immune cells in the vessel walls leads to the generation of the hallmark of the disease— the fibrolipidic plaque. The process starts with the accumulation of modified apolipoprotein B-containing lipoproteins in the intima layer of medium and large-sized arteries [2]. The triggered inflammatory process facilitates the expression of adhesion molecules on the endothelial cell lining of the vessels, leading to the recruitment of immune cells (primarily monocytes) from circulation. Upon infiltration, monocytes differentiate into macrophages, which express scavenger receptors and can engulf the modified lipoproteins, becoming foam cells [2]. The accumulation of intracellular cholesterol, due to constant influx, activates the stress-response machinery within foam cells, resulting in cell death [3]. Cell debris, foam cells, and extracellular lipids coalesce at the center of the plaque in the so-called necrotic core, which is kept contained by the formation of the fibrous cap—a structure composed of modified smooth muscle cells (SMC) and extracellular matrix proteins [4]. Neutrophils also significantly contribute to atherosclerosis progression, releasing inflammatory mediators and reactive oxygen species and contributing to plaque destabilization by releasing matrix proteinases [5].
The IL-1 family has proven to be one of the key drivers of the athero-inflammatory process, particularly through IL-1β and IL-18 activity. The aim of this review is to summarize the current understanding of this cytokine family and describe its role in atherosclerosis development, which might help explore new therapeutic avenues targeting these inflammatory mediators.

2. Systemic Inflammation and Cardiovascular Risk

The concept of traditional risk factors for atherosclerotic CVD has been established based on large-cohort studies, including the Framingham study [6]. However, recent research and changes in lifestyle have expanded the concept of what is currently considered a risk factor. The discovery of statins among other therapies has dramatically reduced the contribution of low-density lipoprotein (LDL) cholesterol to the burden of the disease [7]. Instead, triglyceride (TG) levels in circulation, often in the form of TG-rich lipoproteins, and low levels of high-density lipoprotein (HDL) are the main lipid abnormalities found in affected patients [8,9,10].
Despite the use of appropriate therapies to control risk factors, 42% of patients develop a recurrent ischemic event during the following 10 years after the first episode [11]. In the United States alone, 1 in 10 patients is hospitalized after an acute myocardial infarction (MI) within the following 30 days, with an estimated impact of USD 718 million per year [12]. This residual risk can be explained by multiple factors, including the inflammatory nature of atherosclerosis. Inflammatory biomarkers are currently used for risk stratification and therapy guidance in CVD patients [13]. High-sensitivity C-reactive protein (hsCRP) is an acute-phase reactant and a non-specific inflammatory marker that has been widely studied as a marker for systemic inflammation in risk stratification [13]. Several epidemiological studies have shown an association between hsCRP and future cardiovascular risk in “healthy” individuals [13,14,15]. Liuzzo et al. first reported the use of hsCRP, in combination with serum amyloid A, in predicting poor outcome in patients with severe unstable angina [16]. Later, Morrow et al. showed that elevated hsCRP in patients with unstable angina and non-ST elevation myocardial infarction (non-STEMI) correlated with increased 14-day mortality, alone and in combination with troponin T [17]. Furthermore, CRP was shown to be an independent predictor of cardiac risk and repeated coronary revascularization in a six-month follow-up study of unstable angina patients [18], and it was also associated with long-term risk of cardiac death, alone or in combination with troponin T [19]. Similar associations with long-term mortality were seen with CRP in non-STEMI patients undergoing aggressive revascularization strategies at early time points [20]. Finally, a significant association between hsCRP and future cardiovascular outcome has also been reported in patients with type 2 diabetes (T2D) and a recent acute coronary syndrome, despite treatment with statins and optimal glycemic control [21].

3. Athero-Inflammation, Cholesterol, and the NLRP3 Inflammasome

The important contribution of inflammation to atherosclerosis progression was already proposed by Rudolf Virchow in the 19th century; however, decades of research centered on the lipid hypothesis minimized this observation until recently [22]. Beyond the previously mentioned role of innate immunity (in the form of monocyte/macrophages and neutrophils), adaptive immunity has also been found to be involved in atherogenesis [23]. Indeed, the inflammatory process is involved in all stages of the disease: in the early phase through the infiltration of the vascular wall and the release of pro-inflammatory mediators that amplify the response, and at later stages, through the release of proteolytic enzymes by the immune cells, which contributes to plaque destabilization and the occurrence of acute coronary events [24]. The source of this low-grade vascular inflammation was early considered to be associated with infections, although no direct correlation between microbes and the development of atherosclerosis has been found [25]. Systemic inflammatory conditions—such as rheumatoid arthritis and psoriasis—are also associated with higher cardiovascular risk, indicating that systemic inflammation plays a crucial role in atherosclerosis development, independent of the source [26]. Wright et al. were the first to demonstrate that infectious agents are not required for the onset of atherosclerosis in germ-free apolipoprotein E mice, concluding that the high levels of cholesterol were sufficient to trigger the disease [27]. Instead, abundant research has shown that modified lipoproteins, as well as extracellular lipids in the form of cholesterol crystals (CCs), are known triggers of inflammation [28,29], establishing a connection between both the lipid and inflammatory hypothesis.
One of the ways by which CCs trigger inflammation is via activation of the nucleotide-binding oligomerization domain-like receptor, pyrin domain-containing 3 (NLRP3) inflammasome [30]. It has been suggested that, once phagocytosed, deficient clearance of CCs can induce lysosomal malfunction, resulting in the release of cathepsin B to the cytoplasm and the activation of the inflammasome [31,32]. Oxidized LDL has also been shown to activate the NLRP3 inflammasome through a similar mechanism, after interacting with the scavenger receptor CD36 [29].
Upon activation, a configurational change exposes the pyrin domain in NLRP3, allowing its interaction with the adaptor protein apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC) and the recruitment of procaspase-1, forming the inflammasome complex [33]. This interaction triggers the cleavage and activation of caspase-1 and then the processing of pro-IL-1β and pro-IL-18 into their active forms [34]. In the context of CVD, the activation of the NLRP3 inflammasome in macrophages, with the consequent release of IL-1β, has been shown to be implicated in the development of atherosclerosis and the clinical manifestations of the disease [30].

4. The IL-1 Family

4.1. History

The study of cytokines started in the 1940s with the active search for a fever mediator. Reports showed that supernatants of rabbit neutrophils were able to induce a rapid fever that was not due to contamination with endotoxin [35]. In 1974, Dinarello et al. described what they called “two distinct leukocytic pyrogens”, isolated from supernatants of neutrophil and monocyte cell cultures [36]. These two pyrogens differed in their molecular weight, isoelectric point, and capacity to induce sustained fever [36]. In 1977, the same group purified one of these pyrogens, showing that it had the ability to produce fever at a dose of 10 ng/kg [37]. The interleukin nomenclature was introduced in 1979, but the name IL-1β was not used until its cDNA was cloned in 1984 [38]. In the same year, a macrophage supernatant compound named “lymphocyte activating factor” was cloned in mice, revealing a second gene coding for IL-1 [39], and in the following year, human IL-1α was cloned [40].
In the early 1980s, there were reports of IL-1 inhibitory activity in the circulation of humans suffering from endotoxemia [41], which was then found in cultures of human monocytes [42] and the urine of patients with juvenile arthritis [43]. By the end of the decade, this inhibitory activity was attributed to a receptor antagonist of IL-1α and IL-1β [44]. In 1990, this component was identified as the IL-1 receptor antagonist (IL-1Ra) [45]. Several years later, the “IFNγ-inducing factor” was identified as IL-18 [46]. The remaining members of the IL-1 family were identified later, in the early 2000s [47].

4.2. Members of the IL-1 Family

4.2.1. Cytokines

There are 11 members in the IL-1 family of cytokines (Table 1), which could play either an agonistic or antagonistic role in relation to their receptors, with one—IL-37—considered to be anti-inflammatory [35]. They are divided in subfamilies according to the length of the precursor and the propiece for each precursor (Figure 1). The IL-1 subfamily is formed by IL-1α, IL-1β, and IL-33 and has the longest propieces. The IL-18 subfamily is formed by IL-18 and IL-37, with a smaller propiece. The IL-36 subfamily is formed by IL-36α, IL-36β, IL-36γ, and IL-36 receptor antagonist (IL-36Ra), with the shortest propiece [35]. Evolution analysis showed that these cytokines coevolved and are present in all vertebrates, appearing about 420 million years ago, underpinning their key functional role [48]. With the exception of IL-1Ra, the members of the IL-1 family are mostly intracellular, since they lack a signal peptide and need to be cleaved to generate a mature, active cytokine [49]. They are found primarily in the cytoplasm as precursors containing the conserved AXD amino acid sequence: A, an aliphatic amino acid; X, any amino acid; D, for aspartic acid [49]. This conserved AXD motif is important for the proper folding of the IL-1 family of cytokines into their three-dimensional structure [50]. Nine amino acids from the AXD sequence, the N-terminus cleavage site, were found, which allows the proper folding of the cytokines [50]. This is also the site where caspase-1 cleaves IL-1β [51].
This ancient family of cytokines is known for playing a key role in the orchestration of both innate and adaptive immune responses (Figure 2). They have been reported to act on a wide variety of immune cells, triggering type 1, 2, and 3 responses [49,52,64]. IL-1α and β have been involved in emergency hematopoiesis, in situations where an increased number of hematopoietic cells are needed, such as aging, chemotherapy, and inflammation [52]. IL-18 is a key factor in the regulation of type 1 immunity, acting on NK cells, group 1 innate lymphoid cells (ILC1), and Th1 cells [52]. IL-33 is central in the modulation of type 2 immunity, adaptive immunity, and inflammation [49]. It also plays a key role in models of influenza virus and allergic lung inflammation, and single nucleotide polymorphisms in the genes coding for IL-33 are associated with asthma [65]. IL-1α and β have also been reported to regulate IL-17 production, which is involved in protection against bacterial and fungal infections, particularly in mucosal tissue [64,66]. For more in-depth information regarding the role of the IL-1 family of cytokines in immunity, please refer to the following reviews [49,51,52,56].

4.2.2. Receptors

There are 10 members of the IL-1 family of receptors (Table 2) [67]. Like their ligand counterparts, they are found in all vertebrates and originate through gene duplication events, with some exceptions (e.g., IL-18R) [48]. They are characterized by an extracellular portion that presents Ig domains (usually three, with the exemption of IL-1R8 presenting only one) and the intracellular portion containing a Toll/IL-1 receptor (TIR) domain (except IL-1R2) [67]. The TIR domain allows for the interaction with specific TIR-containing adaptor proteins that are important for the activation of intracellular pathways, including: myeloid differentiation factor 88 (MyD88), TIR-domain-containing adapter-inducing interferon-β (TRIF), TRIF-related adaptor molecule (TRAM), sterile alpha- and armadillo-motif-containing protein (SARM), and TIR-domain-containing adapter protein (TIRAP/Mal) [68]. The active form of the receptors is a heterodimeric complex composed of a ligand-binding chain (IL-1R1, 2, 4, 5, and 6) and an accessory chain (IL1R 3, 3b, and 7) [67]. The proximity of the TIR domains to the two receptor chains initiates the intracellular signaling pathway. This signaling pathway most commonly involves the recruitment of MyD88 and IL-1R-associated kinase (IRAK), interaction with tumor necrosis factor (TNF)-α receptor-associated factor 6 (TRAF6), and finally, the activation of nuclear factor kappa B (NF-κB) [51].
Despite the shared use of MyD88 as an adaptor protein, the specificity of the IL-1 response is tightly controlled through the differential expression of both receptors and regulatory molecules. IL-1α and IL-1β are regulated by the expression of IL-1Ra and the decoy receptor IL-R2 [51]. A similar case is that of the IL-36 family, which is regulated by IL-36Ra [51]. IL-33 is regulated by cellular localization and by soluble receptors, while IL-18 activity is regulated by the IL-18 binding protein (IL-18BP) [67]. Another important regulatory molecule is IL-1R8 (also known as single Ig IL-1R-related molecule, SIGIRR) [70]. IL-1R8 is a decoy receptor mainly expressed by endothelial cells that can regulate the response to several IL-1 cytokines, including IL-1α, IL-1β, IL-33, and IL-18 [70]. IL-1R8 has also been reported to interfere with toll-like receptor signaling [70]. As key regulators of the host immunity, such a detailed system of regulation is required [72]. Failure to control processing and secretion of IL-1 cytokines results in a variety of syndromes characterized by fevers, rash, and other inflammatory manifestations, which will be discussed in the following sections.

4.3. Role in Disease

Autoimmune diseases are characterized by the dysregulation of the innate immune system, which becomes active in the absence of external threats. Many of these diseases are associated with episodes of systemic inflammation and fever, in which IL-1 cytokines play a role [53]. IL-1β has been implicated in the pathogenesis of rheumatoid arthritis (RA) [73], gout arthritis [74], ankylosing spondylitis (AS) [75], and adult-onset and systemic-onset juvenile idiopathic arthritis (Still’s disease) [47]. Treatment of these diseases with anakinra—an inhibitor of the IL-1R—or canakinumab—a monoclonal antibody against IL-1β—has been shown to effectively control inflammation, improving patients’ condition [53,76,77,78,79].
Excess IL-1 signaling is involved in some autoimmune hereditary syndromes such as Familial Mediterranean Fever (FMF), caused by a mutation in the MEFV gene, which codes for the pyrin protein [56]. Anakinra administration to FMF patients results in rapid symptom regression [56,80]. Colchicine, the standard treatment for FMF, is also a known blocker of NLRP3 inflammasome, which directly impacts IL-1β production [81]. Other syndromes where altered IL-1 signaling has been suggested to play a role, and in which anakinra treatment has resulted in positive results, are PAPA syndrome, hyperimmunoglobulinemia D periodic fever syndrome, TNF receptor-associated periodic syndrome, the non-hereditary Schnitzler syndrome [56], and cryopirinopathies [82]. A syndrome caused by a homozygous mutation in the gene coding for IL-1Ra has also been described, in which deficiency of IL-1Ra leads to severe inflammatory manifestations [83,84].
IL-1 cytokines are involved in maintaining the balance between tolerance to commensal microbiota and infectious agents [52]. High serum levels of IL-18 are associated with intestinal inflammation [52] and transgenic mice are more prone to colitis [85], while IL-18 inhibition is protective in models of inflammatory bowel disease (IBS) [86]. Inflammation and high levels of IL-18 have also been described in patients with Crohn’s disease [87,88]. IL-33 plays a role in IBS which varies depending on disease stage and inflammatory state [89]. In ulcerative colitis, blocking the IL-33 signaling pathway significantly reduced active disease [90]; however, IL-33-deficient mice are highly susceptible to colitis and colorectal cancer [91].
The IL-1 family of cytokines has been shown to be involved in neuroinflammation [53]. NLRP3 or caspase-1 deficient mice are protected from neuroinflammation and cognitive decline in models of Alzheimer’s disease [92]. In humans, IL1A allelic polymorphism leading to increased levels of IL-1α is associated with susceptibility to Alzheimer’s disease in some ethnic groups, due to promotion and secretion of the amyloid precursor protein [93]. Increased IL-1 also increases the risk of Parkinson’s, schizophrenia, some types of epilepsy, and febrile convulsions [94,95,96].
Finally, IL-1 cytokines have been implicated in the pathophysiology of CVD. As mentioned previously, inflammation is a key process in atherosclerosis development and is particularly critical in the transition to unstable plaques, which are associated with the clinical manifestations of the disease [2]. The role of the different subfamilies in atherosclerosis and CVD will be discussed in detail in the following sections.

5. IL-1 Subfamily

The IL-1 subfamily of cytokines is composed of IL-1α, IL-1β, IL-33, and IL-1Ra. IL-1α and IL-1β signal through IL-1R1, while IL-33 signals through IL-1R4 [47]. All three cytokines share IL-R3 as a co-receptor. The IL-1 subfamily is known to activate pro-inflammatory pathways, leading to the production of other cytokines and chemokines. IL-1Ra, on the contrary, specifically reduces the activity of both IL-1α and IL-1β [47]. IL-1R2 acts as a decoy receptor for both IL-1α and β, due to the similarities of the extracellular region of IL-1R1 [97]. The cytoplasmic region, however, is short and cannot signal. IL-1R2 can also interact with IL-1R3, allowing it to sequester the co-receptor as well [69]. A soluble, active form of IL-1R2 has also been described [98].

5.1. IL-1 Alpha

Evolutionary studies indicate that the IL-1α gene emerged from a duplication of the ancestral IL-1β gene [48]. The IL-1α gene consists of seven exons and six introns, with no TATA box in the promoter area, and generates a 31 kDa precursor protein [53]. The IL-1α precursor is constitutively expressed in epithelial cells, endothelial cells, and keratinocytes [53]. It can also be found in the surface of several cells, in particular monocytes and B cells, where it is called membrane IL-1α [99]. Membrane IL-1α can interact with and activate the IL-1R1 of adjacent cells, in a mechanism called juxtacrine [99].
IL-1α is considered a “dual-function” cytokine, since it can act both at the nucleus and cell membrane levels [51]. It contains a nuclear localization signal sequence in the precursor region, which allows it to induce the increased gene expression of other inflammatory agents, such as chemokine 8 [100]. The nuclear localization of IL-1α might also help control its pro-inflammatory properties. In the context of cell death, apoptotic signals trigger the localization of IL-1α to the nucleus, preventing inflammation [101]. In contrast, during necrosis, IL-1α moves from nucleus to cytosol, contributing to the highly inflammatory milieu of necrotic cell lysates [101]. Due to this property, IL-1α is considered a DAMP, activating several inflammatory reactions when interacting with its receptor [102]. Interestingly, the precursor of IL-1α is active, being able to engage its target receptor when released in the context of necrotic cell death, suggesting a key role in sterile inflammation [103]. This role of the IL-1α precursor has earned it the role of an “alarmin” [103].

5.2. IL-1 Beta

IL-1β is one of the most studied members of this family. The IL-1β gene has a structure similar to the one coding IL-1α, except for the presence of a TATA box near the promoter [53]. Contrary to IL-1α, IL-1β is not expressed in a constitutive manner in healthy individuals and it is only produced by a limited number of cell types including monocytes/macrophages and dendritic cells [53]. IL-1β is expressed as a 31 KDa inactive precursor, often in response to an insult (microbial products, other cytokines, and even IL-1α and IL-1β), in a process called priming [104]. In a second step, processing of the IL-1β precursor is triggered, rendering a fully matured 17 KDa protein able to engage the IL-1R [104]. Caspase-1 is required for this second step, which in turn is activated by the inflammasome [105]. The following release of mature IL-1β seems to involve externalization through vesicles [106], but secretion mediated by pyroptosis, a type of cell death associated with inflammation, has been described in monocytes/macrophages [107].
Production of mature IL-1β has also been described independent of caspase-1. Mature IL-1β has been detected in turpentine-induced necrosis in caspase-1 KO mice [108]. Neutrophil proteinase 3 (PR3) has been shown to process pro-IL-1β in a model of acute neutrophil-dominated arthritis lacking caspase-1 [109]. PR3 can also process extracellular IL-1β precursors, and similar activity has been described for granzyme, matrix metalloprotease 9, and neutrophil elastase [72]. Upon interacting with the IL-R1, IL-1β results in the activation of NK-κB, through the IRAK pathway, resulting in the production of downstream inflammatory mediators [105]. As such, IL-1β plays a central role in the local inflammatory response, mediating the production of IL-6, which can in turn act systemically, activating, among others, acute phase response proteins such as CRP [105].

5.3. IL-33

IL-33 was identified as a member of the IL-1 family in 2005 by Schmitz et al. [110]. Like IL-1α, it is considered a dual-function protein: it works in the nucleus regulating transcription and as an extracellular cytokine, upon release due to stress [52]. The gene contains seven exons, generating a 31 KDa protein with a nuclear localization sequence in the N-terminal domain and a receptor-binding sequence in the C-terminal [54]. IL-33 has been reported to be expressed in a constitutive manner in the nucleus of epithelial cells and the vascular endothelium, where it forms a complex with chromatin [55,111]. In conditions of necrotic cell death, the chromatin-IL-33 complex is disrupted, allowing for the release of IL-33 to the extracellular space as an alarmin signal [112]. Given this property, IL-33 has been shown to be involved in priming for allergic responses in basophils, mast cells, and eosinophils, among others [113]. It has also been shown to be prominent in the pathogenesis of several allergic conditions including asthma and atopic dermatitis [114,115].
IL-33 signals through the complex between IL-1R3 and its unique receptor suppressor of tumorigenesis 2 (ST2), also known as IL-1R4 [110,116]. ST2 is expressed in a transmembrane and a soluble form [110]. Both forms of this receptor are significantly expressed in aortic and coronary endothelial cells as well as immune cells [117,118]. ST2 activation by IL-33 involves the recruitment of MyD88, IRAK-1 and 4, and TRAF6, with the resulting activation of NF-κB and mitogen-activated protein kinases (MAPK) [119]. Several mechanisms have been described that regulate IL-33 bioavailability and activity. The interaction of IL-33 with soluble ST2 and the nuclear localization have both been described as involved in the neutralization of this cytokine [119]. Phosphorylation of transmembrane ST2 at S442, which results in the internalization and further degradation of the receptor by the proteasome, and inhibition of IL-33 signaling by the interaction of TIR domain of IL-1R8 with the IL-33 receptor complex have been reported as well [119,120].

5.4. IL-1 Receptor Antagonist

IL-1Ra was first described as a molecule with IL-1 inhibitory activity in the supernatant of human monocyte cultures in 1985 [56]. The IL-1RN gene can be expressed as four different protein forms, including IL-1Ra, due to alternative splicing [53]. IL-1Ra is a natural antagonist for IL-1α and IL-1β, since it competes with the IL-1 molecules for IL-1R1 [35]. Two forms of IL-1Ra have been described: secretory and intracellular [53]. The secretory form interacts with IL-1R1, limiting IL-1α and IL-1β activities. Even though IL-Ra has a similar affinity for IL-1R1 when compared with IL-1α and IL-1β, large molar quantities are required to block IL-1 activity, which might be explained by the high amount of IL-1R1 expressed in most cells [121]. The mechanism of action of the other three intracellular forms is less clear but it might involve intranuclear competition with IL-1α and a non-IL-1R1-dependent inhibition of intracellular signaling [72]. Dying cells have been shown to release intracellular IL-Ra, which can then bind IL-1R1 [56].
The important role of IL-1Ra has been demonstrated in KO animal models. IL-1Ra KO mice display increased inflammatory responses and develop joint and skin inflammation as well as vasculitis, according to their genetic background [56]. Severe clinical manifestations of inflammation were also reported in children with mutations in the IL-1RN gene, displaying pustular rash, joint swelling, and elevated CRP, among other manifestations [83,84].

5.5. Role of the IL-1 Family in Atherosclerosis

IL-1 cytokines are involved in the whole process of atherosclerosis development (Figure 3). Both IL-1α and IL-1β have been reported to be expressed in atherosclerotic plaque [122]. The principal source of these cytokines in the plaque are macrophages [122]. The local production of both cytokines can impair the normal function of endothelial cells, inducing oxidative stress, the production of procoagulant factors and impairing vasodilation, all of which can accelerate atherothrombosis [123]. Locally, several mediators of atherosclerosis pathogenesis have been shown to induce IL-1 cytokine expression and release. Oxidized LDL can interact with toll-like receptors and CD36, leading to the expression of IL-1α and IL-1β [29,124]. Necrosis and apoptosis, which occur at the height of the transition towards mature plaques, have also been reported to lead to IL-1 cytokine release [125].
The role of IL-1α and IL-1β has also been explored in animal models of atherogenesis. ApoE KO mice lacking IL-1β develop significantly smaller lesions compared to control mice [126]. Similar results were also seen when a neutralizing antibody against IL-1β was used in the same animal model [127]. Smaller lesions were also reported in apoE KO mice receiving either IL-1α or IL-1β-deficient bone marrow (BM) compared to controls, highlighting the importance of hematopoietic-derived IL-1 [128]. In a similar study, Kamari et al. showed that IL-1α deficiency seems to confer a higher degree of protection against atherosclerosis development [129]. Protection against fatty streak formation, the earliest step in lesion development, was also reported in C57BL6 mice lacking IL-1α [129]. Interestingly, combined deficiency of IL-1α and IL-1β does not result in additive protection [128]. Reduced expression of IL-1Ra in apoE KO mice resulted in larger lesions at early stages with significantly more macrophages, pointing to a role in modulating plaque composition [130]. Administration of recombinant IL-Ra [131] and overexpression of IL-Ra in LDL receptor (LDLR) KO mice resulted in reduced atheroma formation as well [132]. In the same line, IL-1R1 deficiency reduces atherosclerotic burden in the aortic root of apoE KO mice, reducing outward remodeling and increasing plaque stability [133]. Interestingly, atheroprotective effects of IL-1β have been described in later stages of disease development. Treatment of apoE KO mice with advanced lesions (long-term western-diet feeding) with an IL-1β antibody resulted in a significant reduction in SMC and collagen content, but increased the number of macrophages in the fibrous cap [134]. These changes did not affect lesion size but inhibited outward remodeling, which has been described as beneficial [134]. It is likely that the interplay between monocytes and SMC is a dynamic process, with different responses according to the stage of plaque development. Overall, it seems that NLRP3 inflammasome activation promotes a phenotypic change in SMC, fostering vulnerability and resulting in rupture-prone plaques [135].
CCs are another local mediator that can lead to IL-1 cytokine production in atherosclerotic plaque. CCs can activate the NLRP3 inflammasome by inducing the release of the lysosomal protease cathepsin to the cytoplasm, which in turn activates the NLRP3 inflammasome. LDLR KO mice transplanted with NLRP3, ASC, or IL-1β-deficient bone marrow developed smaller plaques when challenged with an atherogenic diet [28]. Similar results were observed with systemic or bone-marrow deficiency of caspase-1/11 in both apoE [136,137] and LDLR KO mice [138]. However, apoE KO mice lacking NLRP3, ASC, or caspase-1 did not show effects on atherosclerotic plaque size, plaque stability, or macrophage infiltration, stressing the importance of biological and experimental context [139]. Conversely, administration of a NLRP3 inflammasome inhibitor for 4 weeks resulted in a reduction in plaque burden in apoE KO mice [140]. Expression of the NLRP3 inflammasome has been reported in human plaques correlating with the severity of coronary artery disease (CAD) [141,142], and also in circulating monocytes of acute coronary syndrome (ACS) patients, compared to controls [143].
Clonal hematopoiesis, a key feature of aging, also involves IL-1 cytokines [52]. The expansion of the ten-eleven-translocation-2 (TET2) mutant hematopoietic clones is suggested to be an important driver of atherosclerosis [144]. Bone marrow transplantation of atherosclerosis-prone mice with TET2-deficient bone marrow resulted in an increase in plaque development, which was associated with increases in NLRP3-mediated IL-1β production in TET2-deficient macrophages [145]. In humans, carriers of clonal hematopoiesis of indeterminate potential (CHIP) experience a 2-fold increase in the risk of coronary artery disease and a 4-fold increase in early-onset myocardial infarction, compared to non-carriers [145].
In patients, genetic differences in the IL-1 gene cluster, which are associated with higher levels of pro-inflammatory cytokines, are associated with the presence of CAD, identified through invasive angiography [146]. It also increased the effect of oxidized phospholipids and lipoprotein(a) on cardiovascular events [146]. Canakinumab treatment of patients with stable CAD, with a history of previous MI and increased hsCRP, resulted in a reduction in cardiovascular events, highlighting the contribution of the IL-1 pathway to cardiovascular risk [147]. IL-1β concentrations have also been reported to be associated with an increased risk of death in patients with heart failure [148]. Similarly, in patients with acute MI, IL-1β levels at admission were independently associated with all-cause mortality, and high levels of IL-1β were associated with cardiovascular death and major adverse cardiovascular events (MACE) [149]. A meta-analysis carried out by Herder et al. showed that circulating levels of IL-1Ra were positively associated with CVD risk, based on the data of six population-based cohort studies [150]. This effect of IL-1Ra levels on CVD risk did not impact, however, overall risk prediction based on classical risk factors [150]. Treatment of STEMI patients with anakinra, on the other hand, did not have the effect of preventing recurrent ischemic events, but it might prevent new-onset heart failure [151]. Finally, a randomized, phase II trial looking at the effect of Xilonix—an anti-IL-1α antibody—on neointimal hyperplasia and post-procedural inflammation in the superficial femoral artery after angioplasty, showed a trend towards a reduction in restenosis [152].

6. IL-18 Subfamily

The IL-18 subfamily comprises IL-18 and IL-37, which have opposite effects (Figure 3). On one side, IL-18 is a potent pro-inflammatory cytokine originally described as an IFN-γ inducing factor by Th1 cells [46,153]. In contrast, IL-37 has been described as an anti-inflammatory cytokine due to its functions as a non-specific inhibitor of inflammation [154]. It was initially described as a cytokine that prevented IL-18-mediated inflammation [59]. Consequently, there is active research looking at the potential role of IL-37 in fighting several types of inflammatory diseases, including atherosclerosis [154,155].

6.1. IL-18

IL-18 is a pleiotropic pro-inflammatory cytokine generated because of proteolytic cleavage of pro-IL-18 by intracellular caspase-1 in the NLRP3 inflammasome [57,156]. The active form is secreted extracellularly, where it can exert several biological functions by binding to IL-18R, a heterodimeric complex consisting of the ligand-binding chain termed IL-1R5, and the co-receptor chain or signal-transducing chain termed IL-1R7 [58]. IL-18R is highly expressed in NK cells, driving its differentiation and activation. Nonetheless, several observations have also documented its expression in Th1 cells, B cells, CD4  +  NKT cells, mast cells, and basophils [157].
IL-18 activates intracellular signaling pathways by first recruiting MyD88 and activating NF-κB and MAPK pathways mainly to induce IFN-γ production for host defense against intracellular pathogen infection, which is augmented by IL-12 [46,158]. In fact, differentiation of naive T cells to the Th1 phenotype is synergistically induced by IL-12 and IL-18 [159]. In addition, there is a well-described role for IL-18 driving the Th2 response in the absence of IL-12 but in the presence of IL-2, involving the stimulation of NK cells, NKT cells, and even established Th1 cells to produce IL-3, IL-9, and IL-13 [160,161]. Thus, IL-18 stimulation can determine different responses that initiate and promote host defense and inflammation [57].
IL-18 is biologically and structurally related to IL-1β and is constitutively expressed in most cell types including human peripheral blood mononuclear cells (PBMCs) [162], macrophages [163], DCs [164], and vascular endothelial cells [165]. To prevent uncontrolled immune responses, IL-18 is regulated by the soluble IL-18BP, which can neutralize IL-18 activity due to a higher affinity for IL-18 compared to the IL-1R5, thus suppressing IFN-γ production and limiting cell responses [166,167,168]. Interestingly, recent evidence suggests IL-37 acts as a negative regulator or antagonist member of IL-18 by binding to IL-1R5 and blocking downstream signaling [169].

6.2. IL-37

IL-37 is a unique member of the IL-1 family that suppresses both innate and acquired immunity to maintain an anti-inflammatory state and can act as an anti-inflammatory alarmin [59,60,170]. The tertiary structure of the IL-37 precursor is closely related to the IL-18 precursor and the intron–exon borders of the IL-18 and IL-37 genes suggest a close association [171]. In fact, IL-37 is produced as a precursor, which is also cleaved by caspase-1 to generate its mature form [60]. Thus, IL-37 is mainly induced in an inflammatory context and IL-1β, IL-18, TNF-α, IFN-γ, and TGF-β can increase its synthesis [51], resulting in a compensatory inhibition of the production of pro-inflammatory cytokines [154]. There are five isoforms of IL-37, named from a to e—the IL-37b isoform is mainly detected in immune cells from peripheral blood [172]. The mechanism of action of IL-37 is based on the formation of a complex with IL-1R5 and IL-1R8 on the cell surface, which suppresses the phosphorylation of several inflammatory kinases such as mTOR [154,173]. In fact, IL-37 suppresses the production of IL-1β mediated by the NLRP3 and AIM2 inflammasomes, and the production of IL-18 by the NLRP3 inflammasome [174,175]. Moreover, IL-37 can also translocate to the nucleus and block Smad3 activation, resulting in anti-inflammatory effect due to the suppression of NF-κB and MAPK [51,174]. Interestingly, the anti-inflammatory properties of IL-18BP described above for IL-18 are lost when IL-37 levels increase, probably because IL-18BP also binds to IL-37 and consequently inhibits the anti-inflammatory effects of IL-37 itself, suggesting that the net IL-37 anti-inflammatory effect depends on both IL-18R and IL-18BP [154,169].
In humans, IL-37 is constitutively expressed in several different tissues and cells, such as blood monocytes, tissue macrophages, plasma cells, and T cells, which may help in the maintenance of the immune homeostasis [176]. While there is no known mouse homolog of human IL-37, transgenic mice expressing human IL-37b are protected from acute inflammation [177,178]. Importantly, altered IL-37 expression in the serum has been found in patients with different inflammatory diseases, suggesting that this cytokine is a potential tool for treating inflammation-driven diseases [154].

6.3. Role of the IL-18 Family in Atherosclerosis

The administration of exogenous IL-18 to athero-prone mice resulted in larger atherosclerotic lesions [179]. IFN-γ plays a key role in atherosclerotic immune cell recruitment, foam cell formation, and plaque progression and stability [180,181,182]. Accordingly, it is believed that the IL-18 pro-atherosclerotic effect is mediated by IFN-γ, which acts on lesion cells including macrophages, endothelial cells, and vascular SMC (VSMC) to accelerate plaque formation [165,179]. As expected, these effects are abolished in IFN-γ-deficient mice [179]. Interestingly, the proatherogenic effect of IL-18 can also occur in the absence of T cells—IFN-γ secreted by macrophages, NK cells, and VSMC has been shown to be sufficient for disease progression [183]. On the other hand, a recent study showed that induction of cell adhesion molecules by recombinant IL-18 via activation of NF-κB might be a crucial event in initiating atherosclerotic lesion development [184]. In fact, exogenous recombinant IL-18 administration significantly upregulates CD36 and NF-κB, while a NF-κB inhibitor blocked IL-18 signaling, restoring plaque stability [185,186]. This suggests a CD36 and NF-κB crosstalk involved in the progression and destabilization of atherosclerotic plaques. Paradoxically, IL-18-deficient mice have shown lipid abnormalities and increased lipid deposition in the arterial wall, as well as enhanced macrophage and Th17 infiltrate, IL-23-producing VSMC and macrophages, and a morphology resembling unstable plaques [187,188]. In this model, Th17 rather than Th1 produced IFN-γ, and IL-18 might not be needed for IFN-γ production in these lesions [188]. Interestingly, total cholesterol levels were positively regulated by IL-18, while HDL cholesterol may reduce IL-18BP levels [159]. This is relevant since it has been demonstrated that the inhibition of IL-18 activity by IL-18BP prevented fatty streak development and slowed down the progression of advanced atherosclerotic plaques in mice [189]. In addition, it was recently shown that both IL-18 and Na-Cl co-transporter (NCC) are necessary to coordinate the IL-18R cell signaling in atherosclerosis, rising a novel pathway for signaling in atherogenesis and plaque vulnerability involving the IL-18/IL-18R dyad [190].
Increased levels of the novel anti-inflammatory IL-37 have been reported in several human disease conditions [155]. Specifically, a previous study showed IL-37 expression in the foam-like cells of human atherosclerotic coronary and carotid artery plaques [191]. The imbalance of M1 and M2 macrophage polarization is directly associated with the progression of atherosclerotic disease, and a recent study showed a higher amount of M1 macrophages and less IL-37 expression in calcific aortic valves, compared to normal valves [192]. In addition, macrophage-specific IL-37 expression led to reduced plaque development and decreased systemic inflammation in atherosclerosis-prone mice [193]. These observations suggest that IL-37 could shift macrophage polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, which is particularly abundant in stable zones of plaque and asymptomatic lesions [194]. Interestingly, IL-37 has also been implicated in the regulation of cholesterol homeostasis and prevention of the initiation of atherosclerotic plaque development through decreased NF-κB signaling and reduced MCP-1 production in IL-37-Tg mice, suggesting a role in avoiding mononuclear cell infiltration [195,196].
A recent study showed that mice overexpressing IL-37 exhibited significant improvements in their atherosclerotic burden and plaque stability through differentiation of the T helper cell anti-inflammatory phenotype, decreasing matrix metalloproteinase (MMP)-2/13-mediated degradation of collagen and inhibition of VSMCs apoptosis [197]. Furthermore, it was recently shown that exogenous IL-37 resulted in a reduction in atherosclerotic plaque size through a decrease in Th1 and Th17 cells and an increase in Treg cells in mice [198]. In addition, it was shown that IL-37 may also attenuate atherosclerosis and atherosclerosis-related diseases through the inhibition of DC activation [199,200]. Moreover, recombinant IL-37 enhances vascular endothelial function by increasing the bioavailability of nitric oxide, and improves systemic insulin sensitivity and glucose tolerance, effects that can potentially prevent atherosclerosis development [201].
Blankenberg et al. were the first to show that serum levels of IL-18, in patients with documented CAD (stable and unstable angina), significantly predicted death by cardiovascular causes during follow-up [202]. Levels of IL-18 are also significantly elevated in patients with previous MI compared to controls, and they are significantly associated with coronary plaque area [203]. IL-18 levels have been shown to be prospectively predictive of cardiovascular events in healthy individuals, in nested case-control studies [204,205,206]. These results were further confirmed by a large prospective study in healthy men carried out by Jefferis et al., which also included a systematic review and meta-analysis of all available prospective data [204].
Taken together, these data strongly suggest that the IL-18 subfamily is a key mediator in the atherosclerotic pathogenesis as well as a potential biomarker.

7. IL-36 Subfamily

The IL-36 subfamily includes three proteins with agonistic activity—IL-36α, IL-36β, and IL-36γ-, one with antagonistic activity—IL-36Ra, and one with potential receptor antagonist activity—IL-38 (Figure 3) [51]. IL-36R is expressed in monocytes, myeloid dendritic cells (mDCs), and monocyte-derived dendritic cells (MDDCs) from normal mice to respond to IL-36, which is primarily secreted in the skin by keratinocytes, but also has been shown to be secreted in T cells, lung, and gut cells [207]. The IL-36 subfamily can be regulated by different cells and inflammatory components, such as IFN-γ, IL-1α, IL-22, IL-17A, and TNF-α [207]. In recent years, the IL-36 subfamily has generated great interest because of its dysregulation in inflammatory diseases [208].

7.1. IL-36 α, β, γ

IL-36α, IL-36β, and IL-36γ exert strong pro-inflammatory effects and share 21–37% sequence homology with IL-1 and IL-1Ra [61]. IL-36 cytokines are unlikely to be secreted by a caspase-1-dependent mechanism (as is the case for IL-1β and IL-18) because they lack a caspase cleavage site, suggesting that IL-36 cytokines adopt an alternative secretion mechanism, putatively, extracellularly [62,209]. Specifically, IL-36α needs both elastase and Cathepsin G for Lys3 and Ala4 cleavage and resulting activation [210]. In contrast, Cathepsin G cleaves IL-36β at residue Arg5, while Cathepsin S secreted by endothelial cells can cleave IL-36γ between Glut17 and Ser18 residues [210,211].
The active forms are obtained after removal of the nine amino acids preceding the AXD site, which allows them to bind IL-1R6, which dimerizes with IL-1R3 to activate NF-κB and MAPK transcription factors in a MyD88-dependent manner [50,51,62]. IL-36 cytokine signaling pathways appear to be tissue, cell, and context dependent [212]. For example, while primary human keratinocytes stimulated with IL-36α, IL-36β, or IL-36γ induced the expression of inflammatory cytokines [210], IL-36γ-treated endothelial cells result in IL-8, CCL2, CCL20, vascular cell adhesion molecule 1 (VCAM-1), and intercellular adhesion molecule (ICAM-1) upregulation [213]. Moreover, monocytes and DCs cultured with IL-36 resulted in an increased production of IL-6 and IL-12, suggesting that IL-36 plays an important role in the innate immune response [207,214].

7.2. IL-36Ra

IL-36Ra is an anti-inflammatory cytokine that shares 54% homology with IL-1Ra [61]. IL-36Ra is also synthesized as an inactive progenitor and its antagonistic function requires its cleavage at a methionine residue situated at the N-terminus by neutrophil-derived elastase to assume its active form [215]. IL-36Ra binds to IL-1R6 with a higher affinity than the agonists but does not lead to IL-1R3 recruitment and the consequent signaling, thereby competing and inhibiting the activity of IL-36α, IL-36β, and IL-36γ in a dose-dependent manner [50,216]. Specifically, it has been shown that IL-36Ra antagonizes IL-36α in mouse models that overexpress this agonist [217]. IL-36Ra is highly expressed in both immune and non-immune cells, and mice with IL-36Ra deficiency showed reduced weight gain and metabolic dysfunction [51,217].

7.3. IL-38

IL-38 is an IL-36 antagonist sharing similar characteristics with IL-36Ra and IL-1Ra [63,216]. Like IL-36 cytokines, IL-38 is secreted as a precursor and does not contain caspase-1 cleavage sites, yet its activation process is not completely understood [218]. The mechanism by which IL-38 exerts its anti-inflammatory actions also remains to be clarified; however, it was shown that the cleaved form of IL-38 can downregulate IL-6 expression by binding IL-1R9 (IL-1 receptor accessory protein l, IL-1RAPL1) and, subsequently, inhibiting the Jun N-terminal kinase (JNK) pathway [71]. It has also been proposed that IL-38 could instead bind IL-1R6 and IL-1R1, resulting in a reduction in IL-8 production [216]. Despite these data, further studies are needed to delineate the specific molecular mechanism of IL-38.

7.4. Role of the IL-36 Subfamily in Atherosclerosis

Experimental models have demonstrated that IL-36γ can exert atherosclerosis-promoting effects by augmenting macrophage foam cell formation and uptake of oxidized low-density lipoproteins through increased expression of the scavenger receptor CD36 in mice [219]. On the other hand, IL-36Ra inhibited the development of atherosclerosis by acting on the NLRP3 inflammasome [220]. From a clinical standpoint, it was recently shown that serum levels of IL-36 were significantly higher in patients with CAD compared to controls, and these levels correlated with the serum concentrations of TNF-α, IL-6, IL-32, and coronary artery stenosis [221]. Although these novel findings suggest a possible role for the IL-36 family in CAD, more data are eagerly awaited.

8. Conclusions

For several decades, the pathophysiology of atherosclerosis was thought to be mediated and driven exclusively by alterations in the metabolism and accumulation of lipids. However, current knowledge points to a strong influence of inflammation on atherosclerosis development, with both local and systemic effects. Moreover, there is a clinical need to address the residual inflammatory risk currently left untreated by available therapies. Among the different inflammatory mediators involved in atherosclerosis onset and progression, the IL-1 superfamily of cytokines plays a key role. Several research articles and clinical studies have provided insight into the contribution of the most well-known members of this superfamily to atherosclerosis development and its clinical manifestations (Table 3). Recent research has primed the potential benefit of non-specific (i.e., Colchicine [222]) and specific (i.e., Canakinumab [147]) IL-1 inhibitors to reduce cardiovascular events. However, the IL-1 superfamily also plays a critical role in host immunity, which might directly hamper the adoption of IL-1 blockers as potential treatments, as recent trials have raised concerns about the effects on non-cardiovascular events (such as infections or cancers) secondary to these drugs. Therefore, the future challenge is to optimize the net benefit between reducing athero-inflammation without affecting immune defenses. Targeting specific inflammatory pathways that are overrepresented in atherosclerosis or potentiating immunomodulatory cytokines—such as IL-37—might help accomplish this long-awaited goal.

Author Contributions

G.M.R. designed the study. G.M.R., L.G. and K.R. screened the available literature. L.G. and K.R. wrote the original draft. G.M.R. and M.E.A. reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

G.M.R.: Fondo Nacional de Desarrollo Científico y Tecnológico (FONDECYT) de la Agencia Nacional de Investigación y Desarrollo (ANID)-Proyecto FONDECYT Regular 1210655. LG, MA: ANID, Programa Iniciativa Científica Milenio–ICN2021_004. KR: ANID-Doctorado Nacional 21201346.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

All figures were created with BioRender.com.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dai, H.; Much, A.A.; Maor, E.; Asher, E.; Younis, A.; Xu, Y.; Lu, Y.; Liu, X.; Shu, J.; Bragazzi, N.L. Global, regional, and national burden of ischaemic heart disease and its attributable risk factors, 1990–2017: Results from the Global Burden of Disease Study 2017. Eur. Heart J. Qual. Care Clin. Outcomes 2022, 8, 50–60. [Google Scholar] [CrossRef] [PubMed]
  2. Libby, P. Inflammation in atherosclerosis. Nature 2002, 420, 868–874. [Google Scholar] [CrossRef] [PubMed]
  3. Rayner, K.J. Cell Death in the Vessel Wall: The Good, the Bad, the Ugly. Arterioscler. Thromb. Vasc. Biol. 2017, 37, e75–e81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Libby, P.; Ridker, P.M.; Hansson, G.K. Progress and challenges in translating the biology of atherosclerosis. Nature 2011, 473, 317–325. [Google Scholar] [CrossRef]
  5. Doring, Y.; Drechsler, M.; Soehnlein, O.; Weber, C. Neutrophils in atherosclerosis: From mice to man. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 288–295. [Google Scholar] [CrossRef] [Green Version]
  6. Andersson, C.; Johnson, A.D.; Benjamin, E.J.; Levy, D.; Vasan, R.S. 70-year legacy of the Framingham Heart Study. Nat. Rev. Cardiol. 2019, 16, 687–698. [Google Scholar] [CrossRef]
  7. Hoogeveen, R.C.; Ballantyne, C.M. Residual Cardiovascular Risk at Low LDL: Remnants, Lipoprotein(a), and Inflammation. Clin. Chem. 2021, 67, 143–153. [Google Scholar] [CrossRef]
  8. Allard-Ratick, M.P.; Kindya, B.R.; Khambhati, J.; Engels, M.C.; Sandesara, P.B.; Rosenson, R.S.; Sperling, L.S. HDL: Fact, fiction, or function? HDL cholesterol and cardiovascular risk. Eur. J. Prev. Cardiol. 2021, 28, 166–173. [Google Scholar] [CrossRef]
  9. Farnier, M.; Zeller, M.; Masson, D.; Cottin, Y. Triglycerides and risk of atherosclerotic cardiovascular disease: An update. Arch. Cardiovasc. Dis. 2021, 114, 132–139. [Google Scholar] [CrossRef]
  10. Martinez, G.; Rigotti, A.; Acevedo, M.; Navarrete, C.; Rosales, J.; Giugliano, R.P.; Corbalan, R. Cholesterol levels and the association of statins with in-hospital mortality of myocardial infarction patients insights from a Chilean registry of myocardial infarction. Clin. Cardiol. 2013, 36, 305–311. [Google Scholar] [CrossRef]
  11. Huynh, T.; Montigny, M.; Iftikhar, U.; Gagnon, R.; Eisenberg, M.; Lauzon, C.; Mansour, S.; Rinfret, S.; Afilalo, M.; Nguyen, M.; et al. Recurrent Cardiovascular Events in Survivors of Myocardial Infarction With ST-Segment Elevation (from the AMI-QUEBEC Study). Am. J. Cardiol. 2018, 121, 897–902. [Google Scholar] [CrossRef] [PubMed]
  12. Kwok, C.S.; Capers, Q.t.; Savage, M.; Gulati, M.; Potts, J.; Mohamed, M.O.; Nagaraja, V.; Patwala, A.; Heatlie, G.; Kontopantelis, E.; et al. Unplanned hospital readmissions after acute myocardial infarction: A nationwide analysis of rates, trends, predictors and causes in the United States between 2010 and 2014. Coron. Artery Dis. 2020, 31, 354–364. [Google Scholar] [CrossRef] [PubMed]
  13. Blake, G.J.; Ridker, P.M. C-reactive protein and other inflammatory risk markers in acute coronary syndromes. J. Am. Coll. Cardiol. 2003, 41, 37S–42S. [Google Scholar] [CrossRef] [Green Version]
  14. Ridker, P.M.; Cushman, M.; Stampfer, M.J.; Tracy, R.P.; Hennekens, C.H. Inflammation, aspirin, and the risk of cardiovascular disease in apparently healthy men. N. Engl. J. Med. 1997, 336, 973–979. [Google Scholar] [CrossRef]
  15. Ridker, P.M.; Buring, J.E.; Shih, J.; Matias, M.; Hennekens, C.H. Prospective study of C-reactive protein and the risk of future cardiovascular events among apparently healthy women. Circulation 1998, 98, 731–733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Liuzzo, G.; Biasucci, L.M.; Gallimore, J.R.; Grillo, R.L.; Rebuzzi, A.G.; Pepys, M.B.; Maseri, A. The prognostic value of C-reactive protein and serum amyloid a protein in severe unstable angina. N. Engl. J. Med. 1994, 331, 417–424. [Google Scholar] [CrossRef]
  17. Morrow, D.A.; Rifai, N.; Antman, E.M.; Weiner, D.L.; McCabe, C.H.; Cannon, C.P.; Braunwald, E. C-reactive protein is a potent predictor of mortality independently of and in combination with troponin T in acute coronary syndromes: A TIMI 11A substudy. Thrombolysis in Myocardial Infarction. J. Am. Coll. Cardiol. 1998, 31, 1460–1465. [Google Scholar] [CrossRef] [Green Version]
  18. Heeschen, C.; Hamm, C.W.; Bruemmer, J.; Simoons, M.L. Predictive value of C-reactive protein and troponin T in patients with unstable angina: A comparative analysis. CAPTURE Investigators. Chimeric c7E3 AntiPlatelet Therapy in Unstable angina REfractory to standard treatment trial. J. Am. Coll. Cardiol. 2000, 35, 1535–1542. [Google Scholar] [CrossRef] [Green Version]
  19. Lindahl, B.; Toss, H.; Siegbahn, A.; Venge, P.; Wallentin, L.; FRISC Study Group. Markers of myocardial damage and inflammation in relation to long-term mortality in unstable coronary artery disease. N. Engl. J. Med. 2000, 343, 1139–1147. [Google Scholar] [CrossRef]
  20. Mueller, C.; Buettner, H.J.; Hodgson, J.M.; Marsch, S.; Perruchoud, A.P.; Roskamm, H.; Neumann, F.J. Inflammation and long-term mortality after non-ST elevation acute coronary syndrome treated with a very early invasive strategy in 1042 consecutive patients. Circulation 2002, 105, 1412–1415. [Google Scholar] [CrossRef]
  21. Hwang, Y.C.; Morrow, D.A.; Cannon, C.P.; Liu, Y.; Bergenstal, R.; Heller, S.; Mehta, C.; Cushman, W.; Bakris, G.L.; Zannad, F.; et al. High-sensitivity C-reactive protein, low-density lipoprotein cholesterol and cardiovascular outcomes in patients with type 2 diabetes in the EXAMINE (Examination of Cardiovascular Outcomes with Alogliptin versus Standard of Care) trial. Diabetes Obes. Metab. 2018, 20, 654–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Mayerl, C.; Lukasser, M.; Sedivy, R.; Niederegger, H.; Seiler, R.; Wick, G. Atherosclerosis research from past to present—on the track of two pathologists with opposing views, Carl von Rokitansky and Rudolf Virchow. Virchows Arch. 2006, 449, 96–103. [Google Scholar] [CrossRef] [PubMed]
  23. Hansson, G.K.; Libby, P.; Schonbeck, U.; Yan, Z.Q. Innate and adaptive immunity in the pathogenesis of atherosclerosis. Circ. Res. 2002, 91, 281–291. [Google Scholar] [CrossRef] [PubMed]
  24. Libby, P.; Hansson, G.K. From Focal Lipid Storage to Systemic Inflammation: JACC Review Topic of the Week. J. Am. Coll. Cardiol. 2019, 74, 1594–1607. [Google Scholar] [CrossRef]
  25. Pothineni, N.V.K.; Subramany, S.; Kuriakose, K.; Shirazi, L.F.; Romeo, F.; Shah, P.K.; Mehta, J.L. Infections, atherosclerosis, and coronary heart disease. Eur. Heart J. 2017, 38, 3195–3201. [Google Scholar] [CrossRef]
  26. Arida, A.; Protogerou, A.D.; Kitas, G.D.; Sfikakis, P.P. Systemic Inflammatory Response and Atherosclerosis: The Paradigm of Chronic Inflammatory Rheumatic Diseases. Int. J. Mol. Sci. 2018, 19, 1890. [Google Scholar] [CrossRef] [Green Version]
  27. Wright, S.D.; Burton, C.; Hernandez, M.; Hassing, H.; Montenegro, J.; Mundt, S.; Patel, S.; Card, D.J.; Hermanowski-Vosatka, A.; Bergstrom, J.D.; et al. Infectious agents are not necessary for murine atherogenesis. J. Exp. Med. 2000, 191, 1437–1442. [Google Scholar] [CrossRef] [Green Version]
  28. Duewell, P.; Kono, H.; Rayner, K.J.; Sirois, C.M.; Vladimer, G.; Bauernfeind, F.G.; Abela, G.S.; Franchi, L.; Nunez, G.; Schnurr, M.; et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010, 464, 1357–1361. [Google Scholar] [CrossRef] [Green Version]
  29. Sheedy, F.J.; Grebe, A.; Rayner, K.J.; Kalantari, P.; Ramkhelawon, B.; Carpenter, S.B.; Becker, C.E.; Ediriweera, H.N.; Mullick, A.E.; Golenbock, D.T.; et al. CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat. Immunol. 2013, 14, 812–820. [Google Scholar] [CrossRef] [Green Version]
  30. Grebe, A.; Latz, E. Cholesterol crystals and inflammation. Curr. Rheumatol. Rep. 2013, 15, 313. [Google Scholar] [CrossRef]
  31. Weber, K.; Schilling, J.D. Lysosomes integrate metabolic-inflammatory cross-talk in primary macrophage inflammasome activation. J. Biol. Chem. 2014, 289, 9158–9171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Hornung, V.; Bauernfeind, F.; Halle, A.; Samstad, E.O.; Kono, H.; Rock, K.L.; Fitzgerald, K.A.; Latz, E. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat. Immunol. 2008, 9, 847–856. [Google Scholar] [CrossRef] [PubMed]
  33. Shao, B.Z.; Xu, Z.Q.; Han, B.Z.; Su, D.F.; Liu, C. NLRP3 inflammasome and its inhibitors: A review. Front. Pharmacol. 2015, 6, 262. [Google Scholar] [CrossRef] [Green Version]
  34. Sharma, B.R.; Kanneganti, T.D. NLRP3 inflammasome in cancer and metabolic diseases. Nat. Immunol. 2021, 22, 550–559. [Google Scholar] [CrossRef] [PubMed]
  35. Dinarello, C.A. Overview of the interleukin-1 family of ligands and receptors. Semin. Immunol. 2013, 25, 389–393. [Google Scholar] [CrossRef] [PubMed]
  36. Dinarello, C.A.; Goldin, N.P.; Wolff, S.M. Demonstration and characterization of two distinct human leukocytic pyrogens. J. Exp. Med. 1974, 139, 1369–1381. [Google Scholar] [CrossRef] [PubMed]
  37. Dinarello, C.A.; Renfer, L.; Wolff, S.M. Human leukocytic pyrogen: Purification and development of a radioimmunoassay. Proc. Natl. Acad. Sci. USA 1977, 74, 4624–4627. [Google Scholar] [CrossRef] [Green Version]
  38. Auron, P.E.; Webb, A.C.; Rosenwasser, L.J.; Mucci, S.F.; Rich, A.; Wolff, S.M.; Dinarello, C.A. Nucleotide sequence of human monocyte interleukin 1 precursor cDNA. Proc. Natl. Acad. Sci. USA 1984, 81, 7907–7911. [Google Scholar] [CrossRef] [Green Version]
  39. Lomedico, P.T.; Gubler, U.; Hellmann, C.P.; Dukovich, M.; Giri, J.G.; Pan, Y.C.; Collier, K.; Semionow, R.; Chua, A.O.; Mizel, S.B. Cloning and expression of murine interleukin-1 cDNA in Escherichia coli. Nature 1984, 312, 458–462. [Google Scholar] [CrossRef]
  40. March, C.J.; Mosley, B.; Larsen, A.; Cerretti, D.P.; Braedt, G.; Price, V.; Gillis, S.; Henney, C.S.; Kronheim, S.R.; Grabstein, K.; et al. Cloning, sequence and expression of two distinct human interleukin-1 complementary DNAs. Nature 1985, 315, 641–647. [Google Scholar] [CrossRef]
  41. Dinarello, C.A.; Rosenwasser, L.J.; Wolff, S.M. Demonstration of a circulating suppressor factor of thymocyte proliferation during endotoxin fever in humans. J. Immunol. 1981, 127, 2517–2519. [Google Scholar] [PubMed]
  42. Arend, W.P.; Joslin, F.G.; Massoni, R.J. Effects of immune complexes on production by human monocytes of interleukin 1 or an interleukin 1 inhibitor. J. Immunol. 1985, 134, 3868–3875. [Google Scholar] [PubMed]
  43. Prieur, A.M.; Kaufmann, M.T.; Griscelli, C.; Dayer, J.M. Specific interleukin-1 inhibitor in serum and urine of children with systemic juvenile chronic arthritis. Lancet 1987, 2, 1240–1242. [Google Scholar] [CrossRef]
  44. Seckinger, P.; Williamson, K.; Balavoine, J.F.; Mach, B.; Mazzei, G.; Shaw, A.; Dayer, J.M. A urine inhibitor of interleukin 1 activity affects both interleukin 1 alpha and 1 beta but not tumor necrosis factor alpha. J. Immunol. 1987, 139, 1541–1545. [Google Scholar] [PubMed]
  45. Eisenberg, S.P.; Evans, R.J.; Arend, W.P.; Verderber, E.; Brewer, M.T.; Hannum, C.H.; Thompson, R.C. Primary structure and functional expression from complementary DNA of a human interleukin-1 receptor antagonist. Nature 1990, 343, 341–346. [Google Scholar] [CrossRef] [PubMed]
  46. Okamura, H.; Tsutsi, H.; Komatsu, T.; Yutsudo, M.; Hakura, A.; Tanimoto, T.; Torigoe, K.; Okura, T.; Nukada, Y.; Hattori, K.; et al. Cloning of a new cytokine that induces IFN-gamma production by T cells. Nature 1995, 378, 88–91. [Google Scholar] [CrossRef] [PubMed]
  47. Dinarello, C.A. The IL-1 family of cytokines and receptors in rheumatic diseases. Nat. Rev. Rheumatol. 2019, 15, 612–632. [Google Scholar] [CrossRef] [PubMed]
  48. Rivers-Auty, J.; Daniels, M.J.D.; Colliver, I.; Robertson, D.L.; Brough, D. Redefining the ancestral origins of the interleukin-1 superfamily. Nat. Commun. 2018, 9, 1156. [Google Scholar] [CrossRef] [Green Version]
  49. Garlanda, C.; Dinarello, C.A.; Mantovani, A. The interleukin-1 family: Back to the future. Immunity 2013, 39, 1003–1018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Towne, J.E.; Renshaw, B.R.; Douangpanya, J.; Lipsky, B.P.; Shen, M.; Gabel, C.A.; Sims, J.E. Interleukin-36 (IL-36) ligands require processing for full agonist (IL-36alpha, IL-36beta, and IL-36gamma) or antagonist (IL-36Ra) activity. J. Biol. Chem. 2011, 286, 42594–42602. [Google Scholar] [CrossRef]
  51. Dinarello, C.A. Overview of the IL-1 family in innate inflammation and acquired immunity. Immunol. Rev. 2018, 281, 8–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Mantovani, A.; Dinarello, C.A.; Molgora, M.; Garlanda, C. Interleukin-1 and Related Cytokines in the Regulation of Inflammation and Immunity. Immunity 2019, 50, 778–795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Khazim, K.; Azulay, E.E.; Kristal, B.; Cohen, I. Interleukin 1 gene polymorphism and susceptibility to disease. Immunol. Rev. 2018, 281, 40–56. [Google Scholar] [CrossRef] [PubMed]
  54. Carriere, V.; Roussel, L.; Ortega, N.; Lacorre, D.A.; Americh, L.; Aguilar, L.; Bouche, G.; Girard, J.P. IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proc. Natl. Acad. Sci. USA 2007, 104, 282–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Moussion, C.; Ortega, N.; Girard, J.P. The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: A novel ‘alarmin’? PLoS ONE 2008, 3, e3331. [Google Scholar] [CrossRef] [Green Version]
  56. Gabay, C.; Lamacchia, C.; Palmer, G. IL-1 pathways in inflammation and human diseases. Nat. Rev. Rheumatol. 2010, 6, 232–241. [Google Scholar] [CrossRef]
  57. Yasuda, K.; Nakanishi, K.; Tsutsui, H. Interleukin-18 in Health and Disease. Int. J. Mol. Sci. 2019, 20, 649. [Google Scholar] [CrossRef] [Green Version]
  58. Rex, D.A.B.; Agarwal, N.; Prasad, T.S.K.; Kandasamy, R.K.; Subbannayya, Y.; Pinto, S.M. A comprehensive pathway map of IL-18-mediated signalling. J. Cell Commun. Signal. 2020, 14, 257–266. [Google Scholar] [CrossRef]
  59. Boraschi, D.; Lucchesi, D.; Hainzl, S.; Leitner, M.; Maier, E.; Mangelberger, D.; Oostingh, G.J.; Pfaller, T.; Pixner, C.; Posselt, G.; et al. IL-37: A new anti-inflammatory cytokine of the IL-1 family. Eur. Cytokine Netw. 2011, 22, 127–147. [Google Scholar] [CrossRef] [Green Version]
  60. Nold, M.F.; Nold-Petry, C.A.; Zepp, J.A.; Palmer, B.E.; Bufler, P.; Dinarello, C.A. IL-37 is a fundamental inhibitor of innate immunity. Nat. Immunol. 2010, 11, 1014–1022. [Google Scholar] [CrossRef]
  61. Nicklin, M.J.; Barton, J.L.; Nguyen, M.; FitzGerald, M.G.; Duff, G.W.; Kornman, K. A sequence-based map of the nine genes of the human interleukin-1 cluster. Genomics 2002, 79, 718–725. [Google Scholar] [CrossRef] [PubMed]
  62. Zhou, L.; Todorovic, V. Interleukin-36: Structure, Signaling and Function. Adv. Exp. Med. Biol. 2021, 21, 191–210. [Google Scholar] [CrossRef] [PubMed]
  63. Yuan, X.; Peng, X.; Li, Y.; Li, M. Role of IL-38 and its related cytokines in inflammation. Mediat. Inflamm. 2015, 2015, 807976. [Google Scholar] [CrossRef] [Green Version]
  64. Mills, K.H.; Dungan, L.S.; Jones, S.A.; Harris, J. The role of inflammasome-derived IL-1 in driving IL-17 responses. J. Leukoc. Biol. 2013, 93, 489–497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Moffatt, M.F.; Gut, I.G.; Demenais, F.; Strachan, D.P.; Bouzigon, E.; Heath, S.; von Mutius, E.; Farrall, M.; Lathrop, M.; Cookson, W.; et al. A large-scale, consortium-based genomewide association study of asthma. N. Engl. J. Med. 2010, 363, 1211–1221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Korn, T.; Bettelli, E.; Oukka, M.; Kuchroo, V.K. IL-17 and Th17 Cells. Annu. Rev. Immunol. 2009, 27, 485–517. [Google Scholar] [CrossRef] [PubMed]
  67. Boraschi, D.; Tagliabue, A. The interleukin-1 receptor family. Semin. Immunol. 2013, 25, 394–407. [Google Scholar] [CrossRef]
  68. Li, X.; Qin, J. Modulation of Toll-interleukin 1 receptor mediated signaling. J. Mol. Med. 2005, 83, 258–266. [Google Scholar] [CrossRef]
  69. Boraschi, D.; Italiani, P.; Weil, S.; Martin, M.U. The family of the interleukin-1 receptors. Immunol. Rev. 2018, 281, 197–232. [Google Scholar] [CrossRef]
  70. Garlanda, C.; Anders, H.J.; Mantovani, A. TIR8/SIGIRR: An IL-1R/TLR family member with regulatory functions in inflammation and T cell polarization. Trends Immunol. 2009, 30, 439–446. [Google Scholar] [CrossRef]
  71. Mora, J.; Schlemmer, A.; Wittig, I.; Richter, F.; Putyrski, M.; Frank, A.C.; Han, Y.; Jung, M.; Ernst, A.; Weigert, A.; et al. Interleukin-38 is released from apoptotic cells to limit inflammatory macrophage responses. J. Mol. Cell. Biol. 2016, 8, 426–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Dinarello, C.A. Immunological and inflammatory functions of the interleukin-1 family. Annu. Rev. Immunol. 2009, 27, 519–550. [Google Scholar] [CrossRef] [PubMed]
  73. Eastgate, J.A.; Symons, J.A.; Wood, N.C.; Grinlinton, F.M.; di Giovine, F.S.; Duff, G.W. Correlation of plasma interleukin 1 levels with disease activity in rheumatoid arthritis. Lancet 1988, 2, 706–709. [Google Scholar] [CrossRef]
  74. Dalbeth, N.; Gosling, A.L.; Gaffo, A.; Abhishek, A. Gout. Lancet 2021, 397, 1843–1855. [Google Scholar] [CrossRef]
  75. Vazquez-Del Mercado, M.; Garcia-Gonzalez, A.; Munoz-Valle, J.F.; Garcia-Iglesias, T.; Martinez-Bonilla, G.; Bernard-Medina, G.; Sanchez-Ortiz, A.; Ornelas-Aguirre, J.M.; Salazar-Paramo, M.; Gamez-Nava, J.I.; et al. Interleukin 1beta (IL-1beta), IL-10, tumor necrosis factor-alpha, and cellular proliferation index in peripheral blood mononuclear cells in patients with ankylosing spondylitis. J. Rheumatol. 2002, 29, 522–526. [Google Scholar] [PubMed]
  76. Pascual, V.; Allantaz, F.; Arce, E.; Punaro, M.; Banchereau, J. Role of interleukin-1 (IL-1) in the pathogenesis of systemic onset juvenile idiopathic arthritis and clinical response to IL-1 blockade. J. Exp. Med. 2005, 201, 1479–1486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Horneff, G.; Peitz, J.; Kekow, J.; Foell, D. Canakinumab for first line steroid-free treatment in a child with systemic-onset juvenile idiopathic arthritis. Scand. J. Rheumatol. 2017, 46, 500–501. [Google Scholar] [CrossRef] [PubMed]
  78. Cavalli, G.; Dinarello, C.A. Treating rheumatological diseases and co-morbidities with interleukin-1 blocking therapies. Rheumatology 2015, 54, 2134–2144. [Google Scholar] [CrossRef] [Green Version]
  79. So, A.K.; Martinon, F. Inflammation in gout: Mechanisms and therapeutic targets. Nat. Rev. Rheumatol. 2017, 13, 639–647. [Google Scholar] [CrossRef]
  80. Roldan, R.; Ruiz, A.M.; Miranda, M.D.; Collantes, E. Anakinra: New therapeutic approach in children with Familial Mediterranean Fever resistant to colchicine. Jt. Bone Spine 2008, 75, 504–505. [Google Scholar] [CrossRef]
  81. Gonzalez, L.; Bulnes, J.F.; Orellana, M.P.; Munoz Venturelli, P.; Martinez Rodriguez, G. The Role of Colchicine in Atherosclerosis: From Bench to Bedside. Pharmaceutics 2022, 14, 1395. [Google Scholar] [CrossRef] [PubMed]
  82. Hoffman, H.M.; Throne, M.L.; Amar, N.J.; Sebai, M.; Kivitz, A.J.; Kavanaugh, A.; Weinstein, S.P.; Belomestnov, P.; Yancopoulos, G.D.; Stahl, N.; et al. Efficacy and safety of rilonacept (interleukin-1 Trap) in patients with cryopyrin-associated periodic syndromes: Results from two sequential placebo-controlled studies. Arthritis Rheum. 2008, 58, 2443–2452. [Google Scholar] [CrossRef]
  83. Reddy, S.; Jia, S.; Geoffrey, R.; Lorier, R.; Suchi, M.; Broeckel, U.; Hessner, M.J.; Verbsky, J. An autoinflammatory disease due to homozygous deletion of the IL1RN locus. N. Engl. J. Med. 2009, 360, 2438–2444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Aksentijevich, I.; Masters, S.L.; Ferguson, P.J.; Dancey, P.; Frenkel, J.; van Royen-Kerkhoff, A.; Laxer, R.; Tedgard, U.; Cowen, E.W.; Pham, T.H.; et al. An autoinflammatory disease with deficiency of the interleukin-1-receptor antagonist. N. Engl. J. Med. 2009, 360, 2426–2437. [Google Scholar] [CrossRef] [Green Version]
  85. Ishikura, T.; Kanai, T.; Uraushihara, K.; Iiyama, R.; Makita, S.; Totsuka, T.; Yamazaki, M.; Sawada, T.; Nakamura, T.; Miyata, T.; et al. Interleukin-18 overproduction exacerbates the development of colitis with markedly infiltrated macrophages in interleukin-18 transgenic mice. J. Gastroenterol. Hepatol. 2003, 18, 960–969. [Google Scholar] [CrossRef]
  86. Sivakumar, P.V.; Westrich, G.M.; Kanaly, S.; Garka, K.; Born, T.L.; Derry, J.M.; Viney, J.L. Interleukin 18 is a primary mediator of the inflammation associated with dextran sulphate sodium induced colitis: Blocking interleukin 18 attenuates intestinal damage. Gut 2002, 50, 812–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Monteleone, G.; Trapasso, F.; Parrello, T.; Biancone, L.; Stella, A.; Iuliano, R.; Luzza, F.; Fusco, A.; Pallone, F. Bioactive IL-18 expression is up-regulated in Crohn’s disease. J. Immunol. 1999, 163, 143–147. [Google Scholar] [PubMed]
  88. Furuya, D.; Yagihashi, A.; Komatsu, M.; Masashi, N.; Tsuji, N.; Kobayashi, D.; Watanabe, N. Serum interleukin-18 concentrations in patients with inflammatory bowel disease. J. Immunother. 2002, 25 (Suppl. S1), S65–S67. [Google Scholar] [CrossRef]
  89. Williams, M.A.; O’Callaghan, A.; Corr, S.C. IL-33 and IL-18 in Inflammatory Bowel Disease Etiology and Microbial Interactions. Front. Immunol. 2019, 10, 1091. [Google Scholar] [CrossRef] [Green Version]
  90. Sedhom, M.A.; Pichery, M.; Murdoch, J.R.; Foligne, B.; Ortega, N.; Normand, S.; Mertz, K.; Sanmugalingam, D.; Brault, L.; Grandjean, T.; et al. Neutralisation of the interleukin-33/ST2 pathway ameliorates experimental colitis through enhancement of mucosal healing in mice. Gut 2013, 62, 1714–1723. [Google Scholar] [CrossRef]
  91. Malik, A.; Sharma, D.; Zhu, Q.; Karki, R.; Guy, C.S.; Vogel, P.; Kanneganti, T.D. IL-33 regulates the IgA-microbiota axis to restrain IL-1alpha-dependent colitis and tumorigenesis. J. Clin. Investig. 2016, 126, 4469–4481. [Google Scholar] [CrossRef]
  92. Heneka, M.T.; Kummer, M.P.; Stutz, A.; Delekate, A.; Schwartz, S.; Vieira-Saecker, A.; Griep, A.; Axt, D.; Remus, A.; Tzeng, T.C.; et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 2013, 493, 674–678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Du, Y.; Dodel, R.C.; Eastwood, B.J.; Bales, K.R.; Gao, F.; Lohmuller, F.; Muller, U.; Kurz, A.; Zimmer, R.; Evans, R.M.; et al. Association of an interleukin 1 alpha polymorphism with Alzheimer’s disease. Neurology 2000, 55, 480–483. [Google Scholar] [CrossRef] [PubMed]
  94. Mrak, R.E.; Griffinbc, W.S. The role of activated astrocytes and of the neurotrophic cytokine S100B in the pathogenesis of Alzheimer’s disease. Neurobiol. Aging 2001, 22, 915–922. [Google Scholar] [CrossRef] [PubMed]
  95. Piancone, F.; La Rosa, F.; Marventano, I.; Saresella, M.; Clerici, M. The Role of the Inflammasome in Neurodegenerative Diseases. Molecules 2021, 26, 953. [Google Scholar] [CrossRef]
  96. Vezzani, A.; Balosso, S.; Ravizza, T. Neuroinflammatory pathways as treatment targets and biomarkers in epilepsy. Nat. Rev. Neurol. 2019, 15, 459–472. [Google Scholar] [CrossRef]
  97. Colotta, F.; Re, F.; Muzio, M.; Bertini, R.; Polentarutti, N.; Sironi, M.; Giri, J.G.; Dower, S.K.; Sims, J.E.; Mantovani, A. Interleukin-1 type II receptor: A decoy target for IL-1 that is regulated by IL-4. Science 1993, 261, 472–475. [Google Scholar] [CrossRef]
  98. Smith, D.E.; Hanna, R.; Della, F.; Moore, H.; Chen, H.; Farese, A.M.; MacVittie, T.J.; Virca, G.D.; Sims, J.E. The soluble form of IL-1 receptor accessory protein enhances the ability of soluble type II IL-1 receptor to inhibit IL-1 action. Immunity 2003, 18, 87–96. [Google Scholar] [CrossRef] [Green Version]
  99. Kurt-Jones, E.A.; Beller, D.I.; Mizel, S.B.; Unanue, E.R. Identification of a membrane-associated interleukin 1 in macrophages. Proc. Natl. Acad. Sci. USA 1985, 82, 1204–1208. [Google Scholar] [CrossRef] [Green Version]
  100. Werman, A.; Werman-Venkert, R.; White, R.; Lee, J.K.; Werman, B.; Krelin, Y.; Voronov, E.; Dinarello, C.A.; Apte, R.N. The precursor form of IL-1alpha is an intracrine proinflammatory activator of transcription. Proc. Natl. Acad. Sci. USA 2004, 101, 2434–2439. [Google Scholar] [CrossRef]
  101. Cohen, I.; Rider, P.; Carmi, Y.; Braiman, A.; Dotan, S.; White, M.R.; Voronov, E.; Martin, M.U.; Dinarello, C.A.; Apte, R.N. Differential release of chromatin-bound IL-1alpha discriminates between necrotic and apoptotic cell death by the ability to induce sterile inflammation. Proc. Natl. Acad. Sci. USA 2010, 107, 2574–2579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Di Paolo, N.C.; Shayakhmetov, D.M. Interleukin 1alpha and the inflammatory process. Nat. Immunol. 2016, 17, 906–913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Kim, B.; Lee, Y.; Kim, E.; Kwak, A.; Ryoo, S.; Bae, S.H.; Azam, T.; Kim, S.; Dinarello, C.A. The Interleukin-1alpha Precursor is Biologically Active and is Likely a Key Alarmin in the IL-1 Family of Cytokines. Front. Immunol. 2013, 4, 391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Stutz, A.; Golenbock, D.T.; Latz, E. Inflammasomes: Too big to miss. J. Clin. Investig. 2009, 119, 3502–3511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Grebe, A.; Hoss, F.; Latz, E. NLRP3 Inflammasome and the IL-1 Pathway in Atherosclerosis. Circ. Res. 2018, 122, 1722–1740. [Google Scholar] [CrossRef]
  106. Wesche, H.; Resch, K.; Martin, M.U. Effects of IL-1 receptor accessory protein on IL-1 binding. FEBS Lett. 1998, 429, 303–306. [Google Scholar] [CrossRef] [Green Version]
  107. Brough, D.; Rothwell, N.J. Caspase-1-dependent processing of pro-interleukin-1beta is cytosolic and precedes cell death. J. Cell Sci. 2007, 120, 772–781. [Google Scholar] [CrossRef] [Green Version]
  108. Fantuzzi, G.; Ku, G.; Harding, M.W.; Livingston, D.J.; Sipe, J.D.; Kuida, K.; Flavell, R.A.; Dinarello, C.A. Response to local inflammation of IL-1 beta-converting enzyme- deficient mice. J. Immunol. 1997, 158, 1818–1824. [Google Scholar]
  109. Joosten, L.A.; Netea, M.G.; Fantuzzi, G.; Koenders, M.I.; Helsen, M.M.; Sparrer, H.; Pham, C.T.; van der Meer, J.W.; Dinarello, C.A.; van den Berg, W.B. Inflammatory arthritis in caspase 1 gene-deficient mice: Contribution of proteinase 3 to caspase 1-independent production of bioactive interleukin-1beta. Arthritis Rheum. 2009, 60, 3651–3662. [Google Scholar] [CrossRef] [Green Version]
  110. Schmitz, J.; Owyang, A.; Oldham, E.; Song, Y.; Murphy, E.; McClanahan, T.K.; Zurawski, G.; Moshrefi, M.; Qin, J.; Li, X.; et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity 2005, 23, 479–490. [Google Scholar] [CrossRef] [Green Version]
  111. Gautier, V.; Cayrol, C.; Farache, D.; Roga, S.; Monsarrat, B.; Burlet-Schiltz, O.; Gonzalez de Peredo, A.; Girard, J.P. Extracellular IL-33 cytokine, but not endogenous nuclear IL-33, regulates protein expression in endothelial cells. Sci. Rep. 2016, 6, 34255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Demyanets, S.; Stojkovic, S.; Huber, K.; Wojta, J. The Paradigm Change of IL-33 in Vascular Biology. Int. J. Mol. Sci. 2021, 22, 13288. [Google Scholar] [CrossRef] [PubMed]
  113. Bouffi, C.; Rochman, M.; Zust, C.B.; Stucke, E.M.; Kartashov, A.; Fulkerson, P.C.; Barski, A.; Rothenberg, M.E. IL-33 markedly activates murine eosinophils by an NF-kappaB-dependent mechanism differentially dependent upon an IL-4-driven autoinflammatory loop. J. Immunol. 2013, 191, 4317–4325. [Google Scholar] [CrossRef] [Green Version]
  114. Grotenboer, N.S.; Ketelaar, M.E.; Koppelman, G.H.; Nawijn, M.C. Decoding asthma: Translating genetic variation in IL33 and IL1RL1 into disease pathophysiology. J. Allergy Clin. Immunol. 2013, 131, 856–865. [Google Scholar] [CrossRef] [PubMed]
  115. Shimizu, M.; Matsuda, A.; Yanagisawa, K.; Hirota, T.; Akahoshi, M.; Inomata, N.; Ebe, K.; Tanaka, K.; Sugiura, H.; Nakashima, K.; et al. Functional SNPs in the distal promoter of the ST2 gene are associated with atopic dermatitis. Hum. Mol. Genet. 2005, 14, 2919–2927. [Google Scholar] [CrossRef] [Green Version]
  116. Chackerian, A.A.; Oldham, E.R.; Murphy, E.E.; Schmitz, J.; Pflanz, S.; Kastelein, R.A. IL-1 receptor accessory protein and ST2 comprise the IL-33 receptor complex. J. Immunol. 2007, 179, 2551–2555. [Google Scholar] [CrossRef] [Green Version]
  117. Demyanets, S.; Kaun, C.; Pentz, R.; Krychtiuk, K.A.; Rauscher, S.; Pfaffenberger, S.; Zuckermann, A.; Aliabadi, A.; Groger, M.; Maurer, G.; et al. Components of the interleukin-33/ST2 system are differentially expressed and regulated in human cardiac cells and in cells of the cardiac vasculature. J. Mol. Cell Cardiol. 2013, 60, 16–26. [Google Scholar] [CrossRef] [Green Version]
  118. Lu, J.; Kang, J.; Zhang, C.; Zhang, X. The role of IL-33/ST2L signals in the immune cells. Immunol. Lett. 2015, 164, 11–17. [Google Scholar] [CrossRef]
  119. Pinto, S.M.; Subbannayya, Y.; Rex, D.A.B.; Raju, R.; Chatterjee, O.; Advani, J.; Radhakrishnan, A.; Keshava Prasad, T.S.; Wani, M.R.; Pandey, A. A network map of IL-33 signaling pathway. J. Cell Commun. Signal. 2018, 12, 615–624. [Google Scholar] [CrossRef]
  120. Zhao, W.; Hu, Z. The enigmatic processing and secretion of interleukin-33. Cell Mol. Immunol. 2010, 7, 260–262. [Google Scholar] [CrossRef] [Green Version]
  121. Arend, W.P.; Welgus, H.G.; Thompson, R.C.; Eisenberg, S.P. Biological properties of recombinant human monocyte-derived interleukin 1 receptor antagonist. J. Clin. Investig. 1990, 85, 1694–1697. [Google Scholar] [CrossRef] [PubMed]
  122. Moyer, C.F.; Sajuthi, D.; Tulli, H.; Williams, J.K. Synthesis of IL-1 alpha and IL-1 beta by arterial cells in atherosclerosis. Am. J. Pathol. 1991, 138, 951–960. [Google Scholar] [PubMed]
  123. Abbate, A.; Toldo, S.; Marchetti, C.; Kron, J.; Van Tassell, B.W.; Dinarello, C.A. Interleukin-1 and the Inflammasome as Therapeutic Targets in Cardiovascular Disease. Circ. Res. 2020, 126, 1260–1280. [Google Scholar] [CrossRef] [PubMed]
  124. Lipton, B.A.; Parthasarathy, S.; Ord, V.A.; Clinton, S.K.; Libby, P.; Rosenfeld, M.E. Components of the protein fraction of oxidized low density lipoprotein stimulate interleukin-1 alpha production by rabbit arterial macrophage-derived foam cells. J. Lipid Res. 1995, 36, 2232–2242. [Google Scholar] [CrossRef] [PubMed]
  125. Clarke, M.C.; Talib, S.; Figg, N.L.; Bennett, M.R. Vascular smooth muscle cell apoptosis induces interleukin-1-directed inflammation: Effects of hyperlipidemia-mediated inhibition of phagocytosis. Circ. Res. 2010, 106, 363–372. [Google Scholar] [CrossRef] [Green Version]
  126. Kirii, H.; Niwa, T.; Yamada, Y.; Wada, H.; Saito, K.; Iwakura, Y.; Asano, M.; Moriwaki, H.; Seishima, M. Lack of interleukin-1beta decreases the severity of atherosclerosis in ApoE-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 656–660. [Google Scholar] [CrossRef] [Green Version]
  127. Bhaskar, V.; Yin, J.; Mirza, A.M.; Phan, D.; Vanegas, S.; Issafras, H.; Michelson, K.; Hunter, J.J.; Kantak, S.S. Monoclonal antibodies targeting IL-1 beta reduce biomarkers of atherosclerosis in vitro and inhibit atherosclerotic plaque formation in Apolipoprotein E-deficient mice. Atherosclerosis 2011, 216, 313–320. [Google Scholar] [CrossRef] [PubMed]
  128. Kamari, Y.; Shaish, A.; Shemesh, S.; Vax, E.; Grosskopf, I.; Dotan, S.; White, M.; Voronov, E.; Dinarello, C.A.; Apte, R.N.; et al. Reduced atherosclerosis and inflammatory cytokines in apolipoprotein-E-deficient mice lacking bone marrow-derived interleukin-1alpha. Biochem. Biophys. Res. Commun. 2011, 405, 197–203. [Google Scholar] [CrossRef]
  129. Kamari, Y.; Werman-Venkert, R.; Shaish, A.; Werman, A.; Harari, A.; Gonen, A.; Voronov, E.; Grosskopf, I.; Sharabi, Y.; Grossman, E.; et al. Differential role and tissue specificity of interleukin-1alpha gene expression in atherogenesis and lipid metabolism. Atherosclerosis 2007, 195, 31–38. [Google Scholar] [CrossRef]
  130. Isoda, K.; Sawada, S.; Ishigami, N.; Matsuki, T.; Miyazaki, K.; Kusuhara, M.; Iwakura, Y.; Ohsuzu, F. Lack of interleukin-1 receptor antagonist modulates plaque composition in apolipoprotein E-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 1068–1073. [Google Scholar] [CrossRef] [Green Version]
  131. Elhage, R.; Maret, A.; Pieraggi, M.T.; Thiers, J.C.; Arnal, J.F.; 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. [Google Scholar] [CrossRef] [PubMed]
  132. Devlin, C.M.; Kuriakose, G.; Hirsch, E.; Tabas, I. Genetic alterations of IL-1 receptor antagonist in mice affect plasma cholesterol level and foam cell lesion size. Proc. Natl. Acad. Sci. USA 2002, 99, 6280–6285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Alexander, M.R.; Moehle, C.W.; Johnson, J.L.; Yang, Z.; Lee, J.K.; Jackson, C.L.; Owens, G.K. Genetic inactivation of IL-1 signaling enhances atherosclerotic plaque instability and reduces outward vessel remodeling in advanced atherosclerosis in mice. J. Clin. Investig. 2012, 122, 70–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Gomez, D.; Baylis, R.A.; Durgin, B.G.; Newman, A.A.C.; Alencar, G.F.; Mahan, S.; St Hilaire, C.; Muller, W.; Waisman, A.; Francis, S.E.; et al. Interleukin-1beta has atheroprotective effects in advanced atherosclerotic lesions of mice. Nat. Med. 2018, 24, 1418–1429. [Google Scholar] [CrossRef] [PubMed]
  135. Burger, F.; Baptista, D.; Roth, A.; da Silva, R.F.; Montecucco, F.; Mach, F.; Brandt, K.J.; Miteva, K. NLRP3 Inflammasome Activation Controls Vascular Smooth Muscle Cells Phenotypic Switch in Atherosclerosis. Int. J. Mol. Sci. 2021, 23, 340. [Google Scholar] [CrossRef]
  136. Gage, J.; Hasu, M.; Thabet, M.; Whitman, S.C. Caspase-1 deficiency decreases atherosclerosis in apolipoprotein E-null mice. Can. J. Cardiol. 2012, 28, 222–229. [Google Scholar] [CrossRef]
  137. Usui, F.; Shirasuna, K.; Kimura, H.; Tatsumi, K.; Kawashima, A.; Karasawa, T.; Hida, S.; Sagara, J.; Taniguchi, S.; Takahashi, M. Critical role of caspase-1 in vascular inflammation and development of atherosclerosis in Western diet-fed apolipoprotein E-deficient mice. Biochem. Biophys. Res. Commun. 2012, 425, 162–168. [Google Scholar] [CrossRef]
  138. Hendrikx, T.; Jeurissen, M.L.; van Gorp, P.J.; Gijbels, M.J.; Walenbergh, S.M.; Houben, T.; van Gorp, R.; Pottgens, C.C.; Stienstra, R.; Netea, M.G.; et al. Bone marrow-specific caspase-1/11 deficiency inhibits atherosclerosis development in Ldlr(-/-) mice. FEBS J. 2015, 282, 2327–2338. [Google Scholar] [CrossRef]
  139. Menu, P.; Pellegrin, M.; Aubert, J.F.; Bouzourene, K.; Tardivel, A.; Mazzolai, L.; Tschopp, J. Atherosclerosis in ApoE-deficient mice progresses independently of the NLRP3 inflammasome. Cell Death Dis. 2011, 2, e137. [Google Scholar] [CrossRef] [Green Version]
  140. van der Heijden, T.; Kritikou, E.; Venema, W.; van Duijn, J.; van Santbrink, P.J.; Slutter, B.; Foks, A.C.; Bot, I.; Kuiper, J. NLRP3 Inflammasome Inhibition by MCC950 Reduces Atherosclerotic Lesion Development in Apolipoprotein E-Deficient Mice-Brief Report. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1457–1461. [Google Scholar] [CrossRef] [Green Version]
  141. Shi, X.; Xie, W.L.; Kong, W.W.; Chen, D.; Qu, P. Expression of the NLRP3 Inflammasome in Carotid Atherosclerosis. J. Stroke Cerebrovasc. Dis. 2015, 24, 2455–2466. [Google Scholar] [CrossRef] [PubMed]
  142. Paramel Varghese, G.; Folkersen, L.; Strawbridge, R.J.; Halvorsen, B.; Yndestad, A.; Ranheim, T.; Krohg-Sorensen, K.; Skjelland, M.; Espevik, T.; Aukrust, P.; et al. NLRP3 Inflammasome Expression and Activation in Human Atherosclerosis. J. Am. Heart Assoc. 2016, 5, e003031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Altaf, A.; Qu, P.; Zhao, Y.; Wang, H.; Lou, D.; Niu, N. NLRP3 inflammasome in peripheral blood monocytes of acute coronary syndrome patients and its relationship with statins. Coron. Artery Dis. 2015, 26, 409–421. [Google Scholar] [CrossRef] [PubMed]
  144. Jaiswal, S.; Natarajan, P.; Silver, A.J.; Gibson, C.J.; Bick, A.G.; Shvartz, E.; McConkey, M.; Gupta, N.; Gabriel, S.; Ardissino, D.; et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N. Engl. J. Med. 2017, 377, 111–121. [Google Scholar] [CrossRef]
  145. Fuster, J.J.; MacLauchlan, S.; Zuriaga, M.A.; Polackal, M.N.; Ostriker, A.C.; Chakraborty, R.; Wu, C.L.; Sano, S.; Muralidharan, S.; Rius, C.; et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science 2017, 355, 842–847. [Google Scholar] [CrossRef] [Green Version]
  146. Tsimikas, S.; Duff, G.W.; Berger, P.B.; Rogus, J.; Huttner, K.; Clopton, P.; Brilakis, E.; Kornman, K.S.; Witztum, J.L. Pro-inflammatory interleukin-1 genotypes potentiate the risk of coronary artery disease and cardiovascular events mediated by oxidized phospholipids and lipoprotein(a). J. Am. Coll. Cardiol. 2014, 63, 1724–1734. [Google Scholar] [CrossRef] [Green Version]
  147. Ridker, P.M.; Everett, B.M.; Thuren, T.; MacFadyen, J.G.; Chang, W.H.; Ballantyne, C.; Fonseca, F.; Nicolau, J.; Koenig, W.; Anker, S.D.; et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N. Engl. J. Med. 2017, 377, 1119–1131. [Google Scholar] [CrossRef]
  148. Pascual-Figal, D.A.; Bayes-Genis, A.; Asensio-Lopez, M.C.; Hernandez-Vicente, A.; Garrido-Bravo, I.; Pastor-Perez, F.; Diez, J.; Ibanez, B.; Lax, A. The Interleukin-1 Axis and Risk of Death in Patients with Acutely Decompensated Heart Failure. J. Am. Coll. Cardiol. 2019, 73, 1016–1025. [Google Scholar] [CrossRef]
  149. Silvain, J.; Kerneis, M.; Zeitouni, M.; Lattuca, B.; Galier, S.; Brugier, D.; Mertens, E.; Procopi, N.; Suc, G.; Salloum, T.; et al. Interleukin-1beta and Risk of Premature Death in Patients with Myocardial Infarction. J. Am. Coll. Cardiol. 2020, 76, 1763–1773. [Google Scholar] [CrossRef]
  150. Herder, C.; de Las Heras Gala, T.; Carstensen-Kirberg, M.; Huth, C.; Zierer, A.; Wahl, S.; Sudduth-Klinger, J.; Kuulasmaa, K.; Peretz, D.; Ligthart, S.; et al. Circulating Levels of Interleukin 1-Receptor Antagonist and Risk of Cardiovascular Disease: Meta-Analysis of Six Population-Based Cohorts. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1222–1227. [Google Scholar] [CrossRef] [Green Version]
  151. Abbate, A.; Kontos, M.C.; Abouzaki, N.A.; Melchior, R.D.; Thomas, C.; Van Tassell, B.W.; Oddi, C.; Carbone, S.; Trankle, C.R.; Roberts, C.S.; et al. Comparative safety of interleukin-1 blockade with anakinra in patients with ST-segment elevation acute myocardial infarction (from the VCU-ART and VCU-ART2 pilot studies). Am. J. Cardiol. 2015, 115, 288–292. [Google Scholar] [CrossRef]
  152. El Sayed, H.; Kerensky, R.; Stecher, M.; Mohanty, P.; Davies, M. A randomized phase II study of Xilonix, a targeted therapy against interleukin 1alpha, for the prevention of superficial femoral artery restenosis after percutaneous revascularization. J. Vasc. Surg. 2016, 63, 133–141.e1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Micallef, M.J.; Ohtsuki, T.; Kohno, K.; Tanabe, F.; Ushio, S.; Namba, M.; Tanimoto, T.; Torigoe, K.; Fujii, M.; Ikeda, M.; et al. Interferon-gamma-inducing factor enhances T helper 1 cytokine production by stimulated human T cells: Synergism with interleukin-12 for interferon-gamma production. Eur. J. Immunol. 1996, 26, 1647–1651. [Google Scholar] [CrossRef] [PubMed]
  154. Su, Z.; Tao, X. Current Understanding of IL-37 in Human Health and Disease. Front. Immunol. 2021, 12, 696605. [Google Scholar] [CrossRef] [PubMed]
  155. McCurdy, S.; Liu, C.A.; Yap, J.; Boisvert, W.A. Potential role of IL-37 in atherosclerosis. Cytokine 2019, 122, 154169. [Google Scholar] [CrossRef] [PubMed]
  156. Tsutsumi, N.; Yokota, A.; Kimura, T.; Kato, Z.; Fukao, T.; Shirakawa, M.; Ohnishi, H.; Tochio, H. An innate interaction between IL-18 and the propeptide that inactivates its precursor form. Sci. Rep. 2019, 9, 6160. [Google Scholar] [CrossRef] [Green Version]
  157. Nakamura, S.; Otani, T.; Okura, R.; Ijiri, Y.; Motoda, R.; Kurimoto, M.; Orita, K. Expression and responsiveness of human interleukin-18 receptor (IL-18R) on hematopoietic cell lines. Leukemia 2000, 14, 1052–1059. [Google Scholar] [CrossRef] [Green Version]
  158. Yoshimoto, T.; Takeda, K.; Tanaka, T.; Ohkusu, K.; Kashiwamura, S.; Okamura, H.; Akira, S.; Nakanishi, K. IL-12 up-regulates IL-18 receptor expression on T cells, Th1 cells, and B cells: Synergism with IL-18 for IFN-gamma production. J. Immunol. 1998, 161, 3400–3407. [Google Scholar]
  159. Cimato, T.R.; Palka, B.A. Effects of statins on TH1 modulating cytokines in human subjects. PeerJ 2015, 3, e764. [Google Scholar] [CrossRef] [Green Version]
  160. Nakanishi, K. Unique Action of Interleukin-18 on T Cells and Other Immune Cells. Front. Immunol. 2018, 9, 763. [Google Scholar] [CrossRef] [Green Version]
  161. Nakanishi, K.; Yoshimoto, T.; Tsutsui, H.; Okamura, H. Interleukin-18 is a unique cytokine that stimulates both Th1 and Th2 responses depending on its cytokine milieu. Cytokine Growth Factor Rev. 2001, 12, 53–72. [Google Scholar] [CrossRef]
  162. Puren, A.J.; Fantuzzi, G.; Dinarello, C.A. Gene expression, synthesis, and secretion of interleukin 18 and interleukin 1beta are differentially regulated in human blood mononuclear cells and mouse spleen cells. Proc. Natl. Acad. Sci. USA 1999, 96, 2256–2261. [Google Scholar] [CrossRef]
  163. Akita, K.; Ohtsuki, T.; Nukada, Y.; Tanimoto, T.; Namba, M.; Okura, T.; Takakura-Yamamoto, R.; Torigoe, K.; Gu, Y.; Su, M.S.; et al. Involvement of caspase-1 and caspase-3 in the production and processing of mature human interleukin 18 in monocytic THP.1 cells. J. Biol. Chem. 1997, 272, 26595–26603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Gardella, S.; Andrei, C.; Costigliolo, S.; Poggi, A.; Zocchi, M.R.; Rubartelli, A. Interleukin-18 synthesis and secretion by dendritic cells are modulated by interaction with antigen-specific T cells. J. Leukoc. Biol. 1999, 66, 237–241. [Google Scholar] [CrossRef] [PubMed]
  165. Gerdes, N.; Sukhova, G.K.; Libby, P.; Reynolds, R.S.; Young, J.L.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Novick, D.; Kim, S.H.; Fantuzzi, G.; Reznikov, L.L.; Dinarello, C.A.; Rubinstein, M. Interleukin-18 binding protein: A novel modulator of the Th1 cytokine response. Immunity 1999, 10, 127–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Park, S.Y.; Hisham, Y.; Shin, H.M.; Yeom, S.C.; Kim, S. Interleukin-18 Binding Protein in Immune Regulation and Autoimmune Diseases. Biomedicines 2022, 10, 1750. [Google Scholar] [CrossRef] [PubMed]
  168. Yellayi, S.; Braun, S.; Domingues, H.G.; Kityatana, N.; Murali, R.; Johnson, R.P.; Mansfield, K.; Westmoreland, S.V.; O’Neil, S.P. Cloning and characterization of rhesus IL-18 binding protein, a natural antagonist to IL-18. Cytokine 2010, 51, 232–239. [Google Scholar] [CrossRef]
  169. Nold-Petry, C.A.; Lo, C.Y.; Rudloff, I.; Elgass, K.D.; Li, S.; Gantier, M.P.; Lotz-Havla, A.S.; Gersting, S.W.; Cho, S.X.; Lao, J.C.; et al. IL-37 requires the receptors IL-18Rα and IL-1R8 (SIGIRR) to carry out its multifaceted anti-inflammatory program upon innate signal transduction. Nat. Immunol. 2015, 16, 354–365. [Google Scholar] [CrossRef]
  170. Cavalli, G.; Dinarello, C.A. Suppression of inflammation and acquired immunity by IL-37. Immunol. Rev. 2018, 281, 179–190. [Google Scholar] [CrossRef] [PubMed]
  171. Fields, J.K.; Günther, S.; Sundberg, E.J. Structural Basis of IL-1 Family Cytokine Signaling. Front. Immunol. 2019, 10, 1412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Bulau, A.M.; Nold, M.F.; Li, S.; Nold-Petry, C.A.; Fink, M.; Mansell, A.; Schwerd, T.; Hong, J.; Rubartelli, A.; Dinarello, C.A.; et al. Role of caspase-1 in nuclear translocation of IL-37, release of the cytokine, and IL-37 inhibition of innate immune responses. Proc. Natl. Acad. Sci. USA 2014, 111, 2650–2655. [Google Scholar] [CrossRef]
  173. Wald, D.; Qin, J.; Zhao, Z.; Qian, Y.; Naramura, M.; Tian, L.; Towne, J.; Sims, J.E.; Stark, G.R.; Li, X. SIGIRR, a negative regulator of Toll-like receptor-interleukin 1 receptor signaling. Nat. Immunol. 2003, 4, 920–927. [Google Scholar] [CrossRef] [PubMed]
  174. Li, S.; Amo-Aparicio, J.; Neff, C.P.; Tengesdal, I.W.; Azam, T.; Palmer, B.E.; López-Vales, R.; Bufler, P.; Dinarello, C.A. Role for nuclear interleukin-37 in the suppression of innate immunity. Proc. Natl. Acad. Sci. USA 2019, 116, 4456–4461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Rudloff, I.; Ung, H.K.; Dowling, J.K.; Mansell, A.; D’Andrea, L.; Ellisdon, A.M.; Whisstock, J.C.; Berger, P.J.; Nold-Petry, C.A.; Nold, M.F. Parsing the IL-37-Mediated Suppression of Inflammasome Function. Cells 2020, 9, 178. [Google Scholar] [CrossRef] [Green Version]
  176. Rudloff, I.; Cho, S.X.; Lao, J.C.; Ngo, D.; McKenzie, M.; Nold-Petry, C.A.; Nold, M.F. Monocytes and dendritic cells are the primary sources of interleukin 37 in human immune cells. J. Leukoc. Biol. 2017, 101, 901–911. [Google Scholar] [CrossRef]
  177. Dinarello, C.A.; Nold-Petry, C.; Nold, M.; Fujita, M.; Li, S.; Kim, S.; Bufler, P. Suppression of innate inflammation and immunity by interleukin-37. Eur. J. Immunol. 2016, 46, 1067–1081. [Google Scholar] [CrossRef] [Green Version]
  178. McNamee, E.N.; Masterson, J.C.; Jedlicka, P.; McManus, M.; Grenz, A.; Collins, C.B.; Nold, M.F.; Nold-Petry, C.; Bufler, P.; Dinarello, C.A.; et al. Interleukin 37 expression protects mice from colitis. Proc. Natl. Acad. Sci. USA 2011, 108, 16711–16716. [Google Scholar] [CrossRef] [Green Version]
  179. Whitman, S.C.; Ravisankar, P.; Daugherty, A. Interleukin-18 enhances atherosclerosis in apolipoprotein E(-/-) mice through release of interferon-gamma. Circ. Res. 2002, 90, E34–E38. [Google Scholar] [CrossRef] [Green Version]
  180. Elyasi, A.; Voloshyna, I.; Ahmed, S.; Kasselman, L.J.; Behbodikhah, J.; De Leon, J.; Reiss, A.B. The role of interferon-γ in cardiovascular disease: An update. Inflamm. Res. 2020, 69, 975–988. [Google Scholar] [CrossRef]
  181. Moss, J.W.; Ramji, D.P. Interferon-γ: Promising therapeutic target in atherosclerosis. World J. Exp. Med. 2015, 5, 154–159. [Google Scholar] [CrossRef] [PubMed]
  182. Schindler, H.; Lutz, M.B.; Röllinghoff, M.; Bogdan, C. The production of IFN-gamma by IL-12/IL-18-activated macrophages requires STAT4 signaling and is inhibited by IL-4. J. Immunol. 2001, 166, 3075–3082. [Google Scholar] [CrossRef] [PubMed]
  183. Tenger, C.; Sundborger, A.; Jawien, J.; Zhou, X. IL-18 accelerates atherosclerosis accompanied by elevation of IFN-gamma and CXCL16 expression independently of T cells. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 791–796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Bhat, O.M.; Uday Kumar, P.; Harishankar, N.; Ravichandaran, L.; Bhatia, A.; Dhawan, V. Interleukin-18-induced cell adhesion molecule expression is associated with feedback regulation by PPAR-γ and NF-κB in Apo E-/- mice. Mol. Cell Biochem. 2017, 428, 119–128. [Google Scholar] [CrossRef]
  185. Bhat, O.M.; Kumar, P.U.; Giridharan, N.V.; Kaul, D.; Kumar, M.J.; Dhawan, V. Interleukin-18-induced atherosclerosis involves CD36 and NF-κB crosstalk in Apo E-/- mice. J. Cardiol. 2015, 66, 28–35. [Google Scholar] [CrossRef] [Green Version]
  186. Bhat, O.M.; Kumar, P.U.; Rao, K.R.; Ahmad, A.; Dhawan, V. Terminalia arjuna prevents Interleukin-18-induced atherosclerosis via modulation of NF-κB/PPAR-γ-mediated pathway in Apo E-/- mice. Inflammopharmacology 2018, 26, 583–598. [Google Scholar] [CrossRef] [PubMed]
  187. Netea, M.G.; Joosten, L.A.; Lewis, E.; Jensen, D.R.; Voshol, P.J.; Kullberg, B.J.; Tack, C.J.; van Krieken, H.; Kim, S.H.; Stalenhoef, A.F.; et al. Deficiency of interleukin-18 in mice leads to hyperphagia, obesity and insulin resistance. Nat. Med. 2006, 12, 650–656. [Google Scholar] [CrossRef]
  188. Pejnovic, N.; Vratimos, A.; Lee, S.H.; Popadic, D.; Takeda, K.; Akira, S.; Chan, W.L. Increased atherosclerotic lesions and Th17 in interleukin-18 deficient apolipoprotein E-knockout mice fed high-fat diet. Mol. Immunol. 2009, 47, 37–45. [Google Scholar] [CrossRef]
  189. Mallat, Z.; Corbaz, A.; Scoazec, A.; Besnard, S.; Lesèche, G.; Chvatchko, Y.; Tedgui, A. Expression of interleukin-18 in human atherosclerotic plaques and relation to plaque instability. Circulation 2001, 104, 1598–1603. [Google Scholar] [CrossRef] [Green Version]
  190. Wang, J.; Sun, C.; Gerdes, N.; Liu, C.; Liao, M.; Liu, J.; Shi, M.A.; He, A.; Zhou, Y.; Sukhova, G.K.; et al. Interleukin 18 function in atherosclerosis is mediated by the interleukin 18 receptor and the Na-Cl co-transporter. Nat. Med. 2015, 21, 820–826. [Google Scholar] [CrossRef] [Green Version]
  191. Ji, Q.; Zeng, Q.; Huang, Y.; Shi, Y.; Lin, Y.; Lu, Z.; Meng, K.; Wu, B.; Yu, K.; Chai, M.; et al. Elevated plasma IL-37, IL-18, and IL-18BP concentrations in patients with acute coronary syndrome. Mediat. Inflamm. 2014, 2014, 165742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Zhou, P.; Li, Q.; Su, S.; Dong, W.; Zong, S.; Ma, Q.; Yang, X.; Zuo, D.; Zheng, S.; Meng, X.; et al. Interleukin 37 Suppresses M1 Macrophage Polarization Through Inhibition of the Notch1 and Nuclear Factor Kappa B Pathways. Front. Cell Dev. Biol. 2020, 8, 56. [Google Scholar] [CrossRef] [PubMed]
  193. McCurdy, S.; Baumer, Y.; Toulmin, E.; Lee, B.H.; Boisvert, W.A. Macrophage-Specific Expression of IL-37 in Hyperlipidemic Mice Attenuates Atherosclerosis. J. Immunol. 2017, 199, 3604–3613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Chinetti-Gbaguidi, G.; Colin, S.; Staels, B. Macrophage subsets in atherosclerosis. Nat. Rev. Cardiol. 2015, 12, 10–17. [Google Scholar] [CrossRef]
  195. Ballak, D.B.; van Diepen, J.A.; Moschen, A.R.; Jansen, H.J.; Hijmans, A.; Groenhof, G.J.; Leenders, F.; Bufler, P.; Boekschoten, M.V.; Müller, M.; et al. IL-37 protects against obesity-induced inflammation and insulin resistance. Nat. Commun. 2014, 5, 4711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Li, J.; Zhai, Y.; Ao, L.; Hui, H.; Fullerton, D.A.; Dinarello, C.A.; Meng, X. Interleukin-37 suppresses the inflammatory response to protect cardiac function in old endotoxemic mice. Cytokine 2017, 95, 55–63. [Google Scholar] [CrossRef]
  197. Liu, J.; Lin, J.; He, S.; Wu, C.; Wang, B.; Liu, J.; Duan, Y.; Liu, T.; Shan, S.; Yang, K.; et al. Transgenic Overexpression of IL-37 Protects Against Atherosclerosis and Strengthens Plaque Stability. Cell Physiol. Biochem. 2018, 45, 1034–1050. [Google Scholar] [CrossRef] [Green Version]
  198. Ji, Q.; Meng, K.; Yu, K.; Huang, S.; Huang, Y.; Min, X.; Zhong, Y.; Wu, B.; Liu, Y.; Nie, S.; et al. Exogenous interleukin 37 ameliorates atherosclerosis via inducing the Treg response in ApoE-deficient mice. Sci. Rep. 2017, 7, 3310. [Google Scholar] [CrossRef] [Green Version]
  199. Hermansson, A.; Johansson, D.K.; Ketelhuth, D.F.; Andersson, J.; Zhou, X.; Hansson, G.K. Immunotherapy with tolerogenic apolipoprotein B-100-loaded dendritic cells attenuates atherosclerosis in hypercholesterolemic mice. Circulation 2011, 123, 1083–1091. [Google Scholar] [CrossRef] [Green Version]
  200. Zhu, R.; Zhang, F.; Pan, C.; Yu, K.; Zhong, Y.; Zeng, Q. Role of IL-37- and IL-37-Treated Dendritic Cells in Acute Coronary Syndrome. Oxid. Med. Cell. Longev. 2021, 2021, 6454177. [Google Scholar] [CrossRef]
  201. Ballak, D.B.; Brunt, V.E.; Sapinsley, Z.J.; Ziemba, B.P.; Richey, J.J.; Zigler, M.C.; Johnson, L.C.; Gioscia-Ryan, R.A.; Culp-Hill, R.; Eisenmesser, E.Z.; et al. Short-term interleukin-37 treatment improves vascular endothelial function, endurance exercise capacity, and whole-body glucose metabolism in old mice. Aging Cell 2020, 19, e13074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Blankenberg, S.; Tiret, L.; Bickel, C.; Peetz, D.; Cambien, F.; Meyer, J.; Rupprecht, H.J.; AtheroGene, I. Interleukin-18 is a strong predictor of cardiovascular death in stable and unstable angina. Circulation 2002, 106, 24–30. [Google Scholar] [CrossRef] [PubMed]
  203. Hulthe, J.; McPheat, W.; Samnegard, A.; Tornvall, P.; Hamsten, A.; Eriksson, P. Plasma interleukin (IL)-18 concentrations is elevated in patients with previous myocardial infarction and related to severity of coronary atherosclerosis independently of C-reactive protein and IL-6. Atherosclerosis 2006, 188, 450–454. [Google Scholar] [CrossRef] [PubMed]
  204. Jefferis, B.J.; Papacosta, O.; Owen, C.G.; Wannamethee, S.G.; Humphries, S.E.; Woodward, M.; Lennon, L.T.; Thomson, A.; Welsh, P.; Rumley, A.; et al. Interleukin 18 and coronary heart disease: Prospective study and systematic review. Atherosclerosis 2011, 217, 227–233. [Google Scholar] [CrossRef] [Green Version]
  205. Everett, B.M.; Bansal, S.; Rifai, N.; Buring, J.E.; Ridker, P.M. Interleukin-18 and the risk of future cardiovascular disease among initially healthy women. Atherosclerosis 2009, 202, 282–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Thompson, S.R.; Novick, D.; Stock, C.J.; Sanders, J.; Brull, D.; Cooper, J.; Woo, P.; Miller, G.; Rubinstein, M.; Humphries, S.E. Free Interleukin (IL)-18 levels, and the impact of IL18 and IL18BP genetic variation, in CHD patients and healthy men. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 2743–2749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Foster, A.M.; Baliwag, J.; Chen, C.S.; Guzman, A.M.; Stoll, S.W.; Gudjonsson, J.E.; Ward, N.L.; Johnston, A. IL-36 promotes myeloid cell infiltration, activation, and inflammatory activity in skin. J. Immunol. 2014, 192, 6053–6061. [Google Scholar] [CrossRef] [Green Version]
  208. Yuan, Z.C.; Xu, W.D.; Liu, X.Y.; Liu, X.Y.; Huang, A.F.; Su, L.C. Biology of IL-36 Signaling and Its Role in Systemic Inflammatory Diseases. Front. Immunol. 2019, 10, 2532. [Google Scholar] [CrossRef] [Green Version]
  209. Clancy, D.M.; Henry, C.M.; Sullivan, G.P.; Martin, S.J. Neutrophil extracellular traps can serve as platforms for processing and activation of IL-1 family cytokines. FEBS J. 2017, 284, 1712–1725. [Google Scholar] [CrossRef] [Green Version]
  210. Henry, C.M.; Sullivan, G.P.; Clancy, D.M.; Afonina, I.S.; Kulms, D.; Martin, S.J. Neutrophil-Derived Proteases Escalate Inflammation through Activation of IL-36 Family Cytokines. Cell Rep. 2016, 14, 708–722. [Google Scholar] [CrossRef] [Green Version]
  211. Byrne, J.; Baker, K.; Houston, A.; Brint, E. IL-36 cytokines in inflammatory and malignant diseases: Not the new kid on the block anymore. Cell Mol. Life Sci. 2021, 78, 6215–6227. [Google Scholar] [CrossRef] [PubMed]
  212. Han, Y.; Huard, A.; Mora, J.; da Silva, P.; Brüne, B.; Weigert, A. IL-36 family cytokines in protective versus destructive inflammation. Cell Signal. 2020, 75, 109773. [Google Scholar] [CrossRef] [PubMed]
  213. Bridgewood, C.; Stacey, M.; Alase, A.; Lagos, D.; Graham, A.; Wittmann, M. IL-36γ has proinflammatory effects on human endothelial cells. Exp. Dermatol. 2017, 26, 402–408. [Google Scholar] [CrossRef]
  214. Vigne, S.; Palmer, G.; Lamacchia, C.; Martin, P.; Talabot-Ayer, D.; Rodriguez, E.; Ronchi, F.; Sallusto, F.; Dinh, H.; Sims, J.E.; et al. IL-36R ligands are potent regulators of dendritic and T cells. Blood 2011, 118, 5813–5823. [Google Scholar] [CrossRef]
  215. Macleod, T.; Doble, R.; McGonagle, D.; Wasson, C.W.; Alase, A.; Stacey, M.; Wittmann, M. Neutrophil Elastase-mediated proteolysis activates the anti-inflammatory cytokine IL-36 Receptor antagonist. Sci. Rep. 2016, 6, 24880. [Google Scholar] [CrossRef] [Green Version]
  216. van de Veerdonk, F.L.; Stoeckman, A.K.; Wu, G.; Boeckermann, A.N.; Azam, T.; Netea, M.G.; Joosten, L.A.; van der Meer, J.W.; Hao, R.; Kalabokis, V.; et al. IL-38 binds to the IL-36 receptor and has biological effects on immune cells similar to IL-36 receptor antagonist. Proc. Natl. Acad. Sci. USA 2012, 109, 3001–3005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Blumberg, H.; Dinh, H.; Trueblood, E.S.; Pretorius, J.; Kugler, D.; Weng, N.; Kanaly, S.T.; Towne, J.E.; Willis, C.R.; Kuechle, M.K.; et al. Opposing activities of two novel members of the IL-1 ligand family regulate skin inflammation. J. Exp. Med. 2007, 204, 2603–2614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Kumar, S.; McDonnell, P.C.; Lehr, R.; Tierney, L.; Tzimas, M.N.; Griswold, D.E.; Capper, E.A.; Tal-Singer, R.; Wells, G.I.; Doyle, M.L.; et al. Identification and initial characterization of four novel members of the interleukin-1 family. J. Biol. Chem. 2000, 275, 10308–10314. [Google Scholar] [CrossRef] [Green Version]
  219. Zhang, M.; Liu, J.; Gao, R.; Hu, Y.; Lu, L.; Liu, C.; Ai, L.; Pan, J.; Tian, L.; Fan, J. Interleukin-36γ aggravates macrophage foam cell formation and atherosclerosis progression in ApoE knockout mice. Cytokine 2021, 146, 155630. [Google Scholar] [CrossRef] [PubMed]
  220. Tian, Y.; Ling, X.Y.; Chen, D.L.; Zhang, X.Q.; Qiu, C.M. Interleukin-36 receptor antagonist attenuates atherosclerosis development by inhibiting NLRP3 inflammasome. J. Cell Physiol. 2020, 235, 9992–9996. [Google Scholar] [CrossRef]
  221. Kazemian, S.; Ahmadi, R.; Rafiei, A.; Azadegan-Dehkordi, F.; Khaledifar, A.; Abdollahpour-Alitappeh, M.; Bagheri, N. The Serum Levels of IL-36 in Patients with Coronary Artery Disease and Their Correlation with the Serum Levels of IL-32, IL-6, TNF-α, and Oxidative Stress. Int. Arch. Allergy Immunol. 2022, 183, 1–9. [Google Scholar] [CrossRef] [PubMed]
  222. Tardif, J.C.; Kouz, S.; Waters, D.D.; Bertrand, O.F.; Diaz, R.; Maggioni, A.P.; Pinto, F.J.; Ibrahim, R.; Gamra, H.; Kiwan, G.S.; et al. Efficacy and Safety of Low-Dose Colchicine after Myocardial Infarction. N. Engl. J. Med. 2019, 381, 2497–2505. [Google Scholar] [CrossRef] [PubMed]
Figure 1. IL-1 superfamily of cytokines. Schematic representation of the three subfamilies, processing enzymes, and main role.
Figure 1. IL-1 superfamily of cytokines. Schematic representation of the three subfamilies, processing enzymes, and main role.
Ijms 24 00017 g001
Figure 2. IL-1 superfamily in innate and adaptive immunity. IL-1 cytokines exert their effects on several cells of both the innate and adaptive immune system, triggering type 1, 2, and 3 immune responses. DCs = dendritic cells, NK = natural killer cells, Th1 = Th helper 1 cells, ILC = innate lymphoid cells, Th17 = Th helper 17 cells.
Figure 2. IL-1 superfamily in innate and adaptive immunity. IL-1 cytokines exert their effects on several cells of both the innate and adaptive immune system, triggering type 1, 2, and 3 immune responses. DCs = dendritic cells, NK = natural killer cells, Th1 = Th helper 1 cells, ILC = innate lymphoid cells, Th17 = Th helper 17 cells.
Ijms 24 00017 g002
Figure 3. Role of the IL-1 family in atherosclerosis. Common signaling pathway for IL-1 family cytokines, which bind to IL-1R family members, recruiting MyD88 and IRAK and resulting in the activation of NF-κB and MAPK and then promoting the transcription of several atherosclerotic pro-inflammatory genes. CCs: Cholesterol Crystals; NET: Neutrophil Extracellular Traps.
Figure 3. Role of the IL-1 family in atherosclerosis. Common signaling pathway for IL-1 family cytokines, which bind to IL-1R family members, recruiting MyD88 and IRAK and resulting in the activation of NF-κB and MAPK and then promoting the transcription of several atherosclerotic pro-inflammatory genes. CCs: Cholesterol Crystals; NET: Neutrophil Extracellular Traps.
Ijms 24 00017 g003
Table 1. IL-1 family of cytokines.
Table 1. IL-1 family of cytokines.
Cytokine NameAlternative NameSubfamilyMain RoleReferences
IL-1αIL-1F1IL-1Pro-inflammatory[52,53]
IL-1βIL-1F2IL-1Pro-inflammatory[52,53]
IL-33IL-1F11IL-1Pro-inflammatory, type 2 immunity[52,54,55]
IL-RaIL-1F3IL-1Antagonist of IL-1α and IL-1β[53,56]
IL-18IL-1F4IL-18Pro-inflammatory, Type 1 immunity[57,58]
IL-37IL-1F7IL-18Anti-inflammatory[59,60]
IL-36RαIL-1F5IL-36Antagonist of IL-36α, β, γ[61,62]
IL-36 α, β, γIL-1F6, 8, 9IL-36Pro-inflammatory[61,62]
IL-38IL-F10IL-36Anti-inflammatory[63]
Table 2. IL-1 family of receptors.
Table 2. IL-1 family of receptors.
ReceptorAlternative NameRoleLigandsReferences
IL-1R1IL-1RIIL-1 binding receptorIL-1 α, IL-1β, and
IL-1Ra
[49,67,69]
IL-1R2IL-1RIIIL-1 decoy receptorIL-1β[49,67,69]
IL-1R3IL-1RAcPIL-1 co-receptorIL-1 α, IL-1β, IL-33, IL-36[49,67,69]
IL-1R4ST2/T1IL-33 binding receptorIL-33[49,67,69]
IL-1R5IL-18Rα/IL-1Rrp1IL-18 binding receptorIL-18[49,67,69]
IL-1R6IL-36R/IL-1Rrp2IL-36 binding receptorIL-36[49,67,69]
IL-1R7IL-18RβIL-18 co-receptorIL-18[49,67,69]
IL-1R8TIR8/SIGIRRIL-37 co-receptor/decoy receptorIL-37[69,70]
IL-1R9APL/TIGIRR-2/IL-1RAPLIL-38 co-receptorIL-38[69,71]
IL-1R10TIGIRR, TIGIRR-1/IL-1RAPL2Orphan receptor-[69]
Table 3. IL-1 family of cytokines and its role in atherosclerosis.
Table 3. IL-1 family of cytokines and its role in atherosclerosis.
CytokineAnimal StudiesHuman StudiesReferences
IL-1α
(i)
Deficiency in mice confers protection against disease, even at early stages.
(ii)
Bone marrow deficiency in mice results in small lesions and reduction in inflammation.
(i)
Treatment with Xilonix results in a reduction in restenosis in the femoral artery after angioplasty.
[128,129,152]
IL-1β
(i)
Deficiency or neutralization in mice results in smaller lesions.
(ii)
Bone marrow deficiency in mice results in smaller lesions.
(iii)
In advanced lesions, it affects VSMC phenotype.
(iv)
Inhibition of outward remodeling and promotion of plaque instability.
(v)
Increased levels are produced by macrophages in a clonal hematopoiesis-influenced atherosclerotic model.
(i)
IL-1β concentration is associated with increased risk of death in HF patients.
(ii)
IL-1β levels at admission are independently associated with all-cause mortality in patients with acute MI.
(iii)
High levels are associated with cardiovascular death and MACE.
(iv)
Canakinumab administration to stable CAD patients results in reduction in cardiovascular events.
[126,127,128,134,135,145,147,148,149]
IL-1Ra
(i)
Reduced expression in mice results in larger lesions and increased macrophage content at early stages.
(ii)
Recombinant IL-1Ra administration or overexpression affects lipoprotein metabolism and foam cell formation.
(i)
Circulating levels are positively associated with CVD risk, without affecting overall risk prediction.
(ii)
Treatment of STEMI patients with anakinra might prevent new-onset HF.
[130,131,132,150,151]
IL-18
(i)
Exogenous administration to mice results in larger plaques.
(ii)
Recombinant IL-18 administration upregulates CD36 and modulates NF-κB, inducing atherosclerosis.
(iii)
Pro-atherogenic effect is mediated by IFN-γ expression.
(iv)
Deficiency results in lipid abnormalities, lipid deposition and changes in morphology associated with unstable plaques.
(v)
Inhibition by IL-18BP prevents fatty streak development and slows down disease progression.
(i)
Serum levels in CAD patients significantly predict CV death.
(ii)
Circulating levels are increased in patients with previous MI and are associated with coronary plaque area.
(iii)
IL-18 levels are prospectively predictive of CV events in healthy individuals.
[179,183,184,185,189,202,203,204,206]
IL-37
(i)
Macrophage expression leads to reduced plaque development and decreased systemic inflammation.
(ii)
Regulation of cholesterol homeostasis and prevention of atherosclerosis (IL-37 transgenic mice model).
(iii)
Overexpression reduces atherosclerosis burden and improves stability through decreased collagen degradation and VSMC apoptosis.
(iv)
Exogenous administration increases Treg cells and decreases Th1 and Th17 cells, reducing plaque size.
(v)
Inhibition of DC activation could also mediate anti-atherogenic effect.
(i)
Expression has been reported in human atherosclerotic plaques.
[191,193,195,196,197,198,199,200]
IL-36 subfamily
(i)
IL-36γ promotes foam cell formation by increasing CD36-mediated oxidized lipoprotein uptake in mice.
(ii)
IL-36Ra attenuates atherosclerosis development through inhibition of the NLRP3 inflammasome in mice.
(i)
IL-36 serum levels are increased in CAD patients, and they correlate with inflammatory cytokines and coronary artery stenosis.
[219,220,221]
CAD: Coronary Artery Disease; CV: Cardiovascular; CVD: Cardiovascular Disease; DC: Dendritic Cell; HF: Heart Failure; MACE: Major Adverse Cardiovascular Events; MI: Myocardial Infarction; NLRP3: Nucleotide-binding Oligomerization Domain-like Receptor, Pyrin Domain-containing 3; STEMI: ST elevation myocardial infarction; VSMC: Vascular Smooth Muscle Cells.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

González, L.; Rivera, K.; Andia, M.E.; Martínez Rodriguez, G. The IL-1 Family and Its Role in Atherosclerosis. Int. J. Mol. Sci. 2023, 24, 17. https://doi.org/10.3390/ijms24010017

AMA Style

González L, Rivera K, Andia ME, Martínez Rodriguez G. The IL-1 Family and Its Role in Atherosclerosis. International Journal of Molecular Sciences. 2023; 24(1):17. https://doi.org/10.3390/ijms24010017

Chicago/Turabian Style

González, Leticia, Katherine Rivera, Marcelo E. Andia, and Gonzalo Martínez Rodriguez. 2023. "The IL-1 Family and Its Role in Atherosclerosis" International Journal of Molecular Sciences 24, no. 1: 17. https://doi.org/10.3390/ijms24010017

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop