Summary
Although it is a member of the Interleukin (IL)-1 family, IL-37 is unique in that it has wide-ranging anti-inflammatory characteristics. It was originally thought to prevent IL-18-mediated inflammation by binding to the IL-18-binding protein. However, upon discovery that it binds to the orphan receptor, IL-1R8, further studies have revealed an expanded role of IL-37 to include several intracellular and extracellular pathways that affect various aspects of inflammation. Its potential role specifically in cardiovascular diseases (CVD) stemmed initially from the discovery of elevated plasma IL-37 levels in human patients with acute coronary syndrome and atrial fibrillation. Other studies using mouse models of ischemia/reperfusion injury, vascular calcification and myocardial infarction have revealed that IL-37 can have a beneficial role in these conditions. This review will explore recent research on the effects of IL-37 on the pathogenesis of CVD.
Introduction
It is well-accepted that inflammation is a major driver of cardiovascular diseases (CVD), as well as many of its accompanying comorbidities. Although CVD is the number one cause of death and disability in world, there are surprisingly few novel treatments addressing this global burden. Targeting inflammation has been a primary focus in recent years, and much has been discovered in recent decades that provide hope for promising future therapeutic options. One exciting, and relatively new cytokine currently being studied is Interleukin (IL)-37. One of the very few anti-inflammatory cytokines of the IL-1 family, it was first discovered in the early 2000s as an IL-18 receptor binding protein.1 Later it was discovered to be a key inhibitor of inflammation, especially the innate immune response.2
Over the past two decades, dozens of publications have come out investigating associations of IL-37 with various disease states, and hypothesis-driven research has revealed a significant anti-inflammatory effect of the cytokine in many biological systems and cell types. Studies of human patients presenting with various CVD pathologies have often revealed a positive correlation between IL-37 expression and disease severity. Additional studies using mouse models to directly investigate the mechanisms of cardiovascular pathology allows this correlation to be investigated more in depth. Hypothesis-driven research has shown that IL-37 plays a protective role in quelling inflammatory pathways known to drive the development of CVD. In this review, we focus on the role of IL-37 in CVD, with particular attention to the human and animal studies that have been conducted within the last few years. Additionally, we discuss the connection between IL-37 and various CVD comorbidities that have an indirect effect on CVD risk and disease progression.
IL-37 and human clinical CVD studies
Genetic predisposition to cardiovascular disease is a significant risk factor in human populations. Gene variants associated with changes in inflammatory and lipid metabolism pathways have been identified using genome-wide association studies (GWAS). An IL-37 polymorphism previously associated with Ankylosing spondylitis, now identified as rs3811047, was recently found to also confer significant risk for coronary artery disease (CAD).3 In a novel study, researchers performed genetic analysis for association of rs3811047 with incidence of CAD. RT-qPCR analysis demonstrated that the polymorphism was significantly correlated with CAD and decreased mRNA expression of IL-37.3 It was concluded that rs3811047 polymorphism establishes IL-37 as a susceptibility gene for CAD. Common haplotypes of the IL-37 gene were investigated and revealed varied susceptibility to degradation,4 implicating genetic variability as an important factor in the stability of the protein, and thus its ability to suppress inflammation. Furthermore, a recent study of patients with gout revealed rare genetic variants of the IL-37 gene associated with earlier onset of gout and higher incidence of inflammation-related comorbidities.5 Interestingly, each of the 4 IL-37 sequence variations were found in exon 5, encoding an important functional domain of the protein, and the resulting IL-37 proteins were shown to have reduced anti-inflammatory activity in vitro.
IL-37 is an effective regulator of innate immune inflammation, especially concerning monocyte/macrophage activation. Monocytes are known to play a significant role in atherosclerosis development, with macrophage foam cells often comprising a large percentage of the plaque volume, and driving a vicious cycle of inflammation and plaque growth. Peripheral blood mononuclear cells (PBMC) from patients with coronary heart disease (CHD) or healthy controls were cultured and stimulated in vitro with recombinant IL-37 protein. Expression of inflammatory cytokines was elevated in PBMC of CHD patients compared to healthy controls, as expected. However, in the presence of recombinant IL-37, production of various potent inflammatory cytokines was significantly reduced.6 Lymphocytes, especially T-cells, can also contribute to the inflammation driving CVD. IL-37 has recently been implicated in enhancing regulatory T-cell (Treg) abundance, and production of anti-inflammatory cytokines in experiments using leukocytes from human atherosclerosis and acute coronary syndrome patients.7,8 rIL-37 treatment of dendritic cells (DCs) or T-cells isolated from patients led to induction of a tolerogenic dendritic cell phenotype, and increased IL-10 and TGF-β secretion by T-cells, two potent anti-inflammatory mediators.
While immune cells are considered major players in CVD, other vascular cells also play an important role in disease development. Endothelial cells are considered to be an initiating site of injury or inflammation in atherosclerosis development, and preventing such inflammation is a viable therapeutic option. IL-37 transfection of human coronary artery endothelial cells resulted in reduced intracellular adhesion molecule (ICAM)-1 expression compared to control following TLR2 stimulation.9 This was supported by another study in which cardiac microvascular endothelial cells isolated from IL-37tg mice show decreased NF-κB as well as MCP-1 protein levels compared to control cells following LPS stimulation in vitro.10 Reducing inflammation at the endothelial monolayer provides strong evidence that a systemic IL-37 treatment could have the potential to reduced vascular inflammatory diseases.
In addition to inflammation, the deposition of calcium and subsequent stiffening of the vasculature, including the aortic valve, during atherosclerosis development is a significant cause of morbidity. Coronary artery calcification (CAC) has been established as a predictor of CVD-related events and plaque burden.11 The role of IL-37 in this aspect of CVD remains unclear, and recent studies provide data indicating that IL-37 may promote aortic calcification, while others indicate the opposite. Multiple studies have established a positive correlation between aortic calcification and IL-37 serum levels observed in patients.12,13 Interestingly, patients with increased levels of osteoprotergerin (OPG), which inhibits vascular calcification, showed a positive correlation with IL-37 serum levels.13 Previous work utilizing the ApoE null atherosclerosis mouse model revealed that long-term treatment with rIL-37 was also associated with increased serum OPG levels, and resulted in decreased vascular calcification of the aortic root.14 The authors also showed that this benefit was dependent on OPG elevation, as addition of an anti-OPG antibody reversed the protective effects of IL-37 in vivo. Interestingly, histological analysis of calcified human aortic valves revealed a greater abundance of M1, pro-inflammatory macrophages, and diminished IL-37 expression when compared to control samples,15 which is opposite to the findings of elevated IL-37 levels found in the serum of CVD patients. This indicates that although circulating levels of IL-37 are elevated in the context of CVD, the localized expression of IL-37 within the plaque microenvironment does not necessarily correlate with serum concentration. These two studies indicate a protective role for both IL-37 and OPG in preventing vascular calcification, however, evidence of a direct causal relationship between the two proteins needs further investigation. Interestingly, it was recently discovered that rIL-37 treatment of bone marrow stem cells in vitro promoted osteogenic differentiation and calcium deposition through activation of the Akt/PI3K pathway.16 This study would suggest that IL-37 might promote atherosclerotic calcification, but would have a protective role in bone healing by promoting osteogenesis. The effects of IL-37 on the Akt/PI3K pathway, especially as it relates to apoptosis and autophagy, have also been studied in a hepatocellular carcinoma cell line. Li et al. showed that IL-37 inhibited proliferation, and induced autophagy as well as apoptosis though inhibition of the PI3K/Akt/mTOR pathway.17 Taken together, there is much to be discovered concerning the effects of IL-37 on these pathways, and the exact outcome may be unique to the cell type and microenvironment involved.
IL-37 has consistently been found at elevated levels in patients with CVD. Patients with atherosclerosis following an acute ischemic stroke (AIS), were found to have higher levels of IL-37 compared to healthy controls,18 which correlated with an unfavorable prognosis three months following the event. In another study, chronic heart failure (CHF) patients were studied during a 1-year follow-up, and levels of serum cytokines and number of major adverse cardiac events (MACE) were recorded. Results from this clinical study showed that plasma IL-37 levels were significantly higher in those patients experiencing CHF than in healthy controls. Patients with very high levels of IL-37 were more likely to have a MACE, and also to show reduced left ventricular ejection function.19 This association agrees with previous correlation studies that report a positive correlation between heart disease, inflammation and IL-37 serum levels. While causation cannot be determined in this type of study, the authors argue that elevated IL-37 levels may be a strong predictor of a poor patient prognosis. Similarly, elevated IL-37 levels in acute myocardial infarction patients were associated with worse clinical outcomes.20 Interestingly, patients undergoing elective coronary artery bypass surgery showed reduced levels of IL-37 compared to healthy controls,21 which increased during the physical therapy phase following discharge, implicating a beneficial role of increasing IL-37 levels during recovery. In lieu of human clinical trials manipulating IL-37 levels directly, further hypothesis-driven research will be required to determine the specific role of IL-37 in cases of chronic heart failure, myocardial infarction and other advanced cardiovascular clinical outcomes. While there are no currently approved clinical treatments using IL-37 protein, mouse models have been instrumental in elucidating the role of IL-37 in the context of CVD.
The protective role of IL-37 in animal models of CVD
The role of IL-37 in vivo has been well-studied using mouse models. To study the effects of IL-37 on atherosclerosis development, transduced hematopoietic stem cells that overexpress IL-37 in macrophages were transplanted into irradiated mice lacking the LDL receptor (LDLr−/−). After 10 weeks of high-fat diet, the overexpression of macrophage IL-37 led to decreased atherosclerotic plaque development compared to controls.22 Similarly, in a study where exogenous IL-37 was administered to atherosclerotic ApoE−/− mice, numbers of macrophages, T-lymphocytes, vascular smooth muscle cells (VSMC), as well as collagen in atherosclerotic plaques, were significantly diminished. This was accompanied by an increase in regulatory T cells (Treg) that inhibited dendritic cell maturation in vivo and in vitro.23 Similar results were observed using a transgenic IL-37 model constitutively expressing the cytokine, crossed to the ApoE−/− atherosclerosis mouse model. Plaque burden was significantly decreased compared to control mice lacking IL-37. Plaque stability was increased in the IL-37 transgenic group by inhibiting collagen degradation driven by matrix metalloproteinase (MMP)-2/13, well as preventing VSMC apoptosis.24 Another study using the same IL-37tg model on the ApoE−/− background found that atherosclerosis was inhibited, which the authors attribute to reduced dendritic cell maturation via the IL-1R8-TLR4-NF-κB pathway.25
Stabilizing atherosclerotic plaque leads to reduced risk for thrombosis and potential subsequent myocardial infarction or stroke. The inflammatory cascade that occurs in the brain following ischemic stroke has been well-established in murine models. Inflammation in the brain begins with activated resident microglia, followed by increased pro-inflammatory and chemotactic cytokines. In agreement with results observed in human patients, IL-37 was found to be elevated in mice following cerebral ischemia-reperfusion. However, it was observed that transgenic mice overexpressing IL-37 (IL-37tg) had less cerebral tissue damage and displayed improved locomotor function compared to wild type animals after ischemia-reperfusion.26 Additionally, in a mouse model of myocardial infarction where induced cardiospheres (iCS) were injected therapeutically to facilitate cardiac repair. iCS expressing IL-37 were shown to have reduced infarct size, as well as decreased circulating inflammatory cytokines.27 An earlier study investigated the role of IL-37-treated dendritic cells (DC) following myocardial infarction (MI). IL-37-treated DCs showed a less activated phenotype, and after adoptive transfer into mice post-MI, led to an increase in the number of regulatory T-cells, improved cardiac function, and reduced fibrosis and infiltrated inflammatory cells in the infarcted tissue.28 In support of this result, aged, endotoxemic mice with transgenic expression of IL-37 were found to be protected against myocardial inflammation, and showed improved cardiac function compared to wild type control mice.10 The protective effects directly attributable to IL-37 expression or treatment in these mouse models strongly implicates a therapeutic potential for IL-37 in the context of human inflammatory disease.
IL-37 in CVD comorbidities
Since many of the comorbidities of CVD are rooted in inflammation, it is worth mentioning the effect of IL-37 on other inflammatory diseases linked to CVD pathogenesis. Metabolic syndrome, which is often a precursor to more severe pathologies such as diabetes, and CVD, often involves chronic, low-grade inflammation. Using a mouse model, transgenic expression of IL-37 led to improved insulin-sensitivity, and reduced production of inflammatory cytokines in adipose tissue compared to control mice lacking IL-37.29 Transgenic IL-37 expression in mice has also been shown to reduce weight gain via reduced food intake following high fat diet administration.30 In the case of diabetes, IL-37 may also have a protective effect on various tissues and cell types. Recently, IL-37 has been shown to be downregulated in gestational diabetes mellitus (GDM) placental tissue compared to control samples.31 In addition, the microRNA miR-657, has been shown to directly target and reduce IL-37 expression in GDM macrophages, leading to increased inflammatory cytokine production.32 Furthermore, a cohort of diabetes mellitus type 2 patients receiving sitagliptin with vitamin D3 in addition to regular treatment, showed a significant downregulation of inflammatory mediators such as IL-17, IL-21 and INF-γ, and an upregulation of anti-inflammatory mediators, including the transcription factor FOXP3 and IL-37.33 Maintenance of adequate IL-37 levels in vivo appears to be important in prevention of metabolic syndrome and development of diabetes, while low levels of IL-37 may prove to be detrimental, as indicated by the correlation studies above. Hypothesis-driven experimental clinical research would be needed to fully elucidate the role of circulating and tissue specific IL-37 in vivo.
Another pathology closely related to CVD involves fibrosis. Investigation into the effect of IL-37 in this context has revealed a protective role for the cytokine in preventing lung and liver fibrosis via reduction of inflammation, in addition to increased autophagy and cell survival.34–36 This expands on the protective role of IL-37 in prevention of cardiac fibrosis and scaring following MI described above.
IL-37 also is a potent inhibitor of allergic inflammatory response and various autoimmune disorders. As with the comorbidities already mentioned, these forms of inflammation also increase the risk of CVD. Various types of allergic inflammation, including atopic dermatitis,37 allergic rhinitis38 and allergic asthma,39 have been shown to be attenuated by IL-37 treatment in mice, primarily by reducing inflammatory response, among other mechanisms. Autoimmune diseases, such as systemic lupus erythematosus (SLE)40 and myasthenia gravis,41 have also proven to benefit from IL-37 treatment via reduced inflammation and autoantibody production (Figure 1).
Figure 1.
Summary of the inhibitors and promoters (left) of IL-37 and its effects discussed. Data originated from in vivo studies performed with patients (blue) or mice (red), or in vitro studies (green). Abbreviations: AIS, acute ischemic stroke; CHF, chronic heart failure; CAC, coronary artery calcification; CAD, coronary artery disease; DC, dendritic cell; EC, endothelial cell; ICAM-1, intracellular adhesion molecule 1; MCP-1, monocyte chemoattractant protein 1; OPG, osteoprotergerin; PBMCs, peripheral blood mononuclear cells.
Clinical therapeutic potential and considerations
Given the mounting evidence that IL-37 has potent anti-inflammatory and displays protective effects in a wide range of pathologies, harnessing its therapeutic potential for use in disease prevention is a logical step forward. Originally crystalized and shown to be stable in dimer form, it was recently discovered that monomeric IL-37 is up to 13 times more effective at preventing inflammation both in vitro and in vivo using a mouse model of endotoxic shock.42 This finding will be useful when developing a formulation of the cytokine for therapeutic use. There are many biologics currently developed for human use, and it is feasible that IL-37 may be added to that arsenal to combat any number of inflammatory diseases. While we have gained invaluable insight into the role of IL-37 in vivo using various mouse models, it should be noted that there is no known homolog of IL-37 in the mouse, even though recombinant IL-37 is clearly effective at preventing inflammation. The pathways studied in human vs. mouse may be slightly different concerning the effects of IL-37, or may not be targeted as dramatically as in the mouse, which has no endogenous IL-37 expression. Additionally, correlation studies in human patients comparing disease state to healthy controls often show a dramatic upregulation of IL-37, which some have interpreted as indication that IL-37 may contribute to the pathogenesis of disease. While, the opposite is more likely to be true given the experimental evidence showing IL-37 treatment effectively reducing inflammatory response in mouse models, it remains a possibility that IL-37 may not be beneficial in every case. Therapies developed around IL-37 treatment should take this into account and ensure that its use is effective each particular disease on a case by case basis (Table 1).
Table 1.
Compilation of recent studies of the role of IL-37 in CVD
References | Year | PubMed ID | Model | Injury/disease model | Tissue/cell type | IL-37 modification | Results |
---|---|---|---|---|---|---|---|
Chai et al.14 | 2015 | Mouse | Atherosclerosis | Arteries | IV injection | IL-37 decreased vascular calcification and inflammatory cytokines. | |
Yu et al.12 | 2016 | 27451144 | Human | Type 2 DM and CAC |
Tibial arterial wall Macrophages VSMCs Plasma |
— | Increased IL-37 concentrations are associated with arterial calcification. |
Xie et al.9 | 2016 | 27233003 |
Human In vitro |
Atherosclerosis | HCAECs | Transgenic | IL-37 decreased NF-κB and ICAM-1 expression after TLR2 activation. |
Zhu et al.28 | 2016 | 27919929 | Mouse | MI |
Myocardium Tregs DCs |
IP injection | IL-37 had a beneficial role in post-MI remodeling. |
Chai et al.13 | 2017 | 28630603 | Human | CAC | Plasma | — | IL-37, OPG, and hsCRP plasma levels are correlated with CAC. |
Shou et al.19 | 2017 | 28781417 | Human | Chronic heart failure | Plasma | — | IL-37 may be a predictor of major adverse cardiac events. |
Yan et al.4 | 2017 | 27665946 | Human | LPS stimulation | Macrophages |
Transgenic Exogenous in vitro stimulation |
Common genetic variants of IL-37 lead to different immune-inhibitory potencies. |
Yin et al.3 | 2017 | 28181534 | Human | CAD and MI | PBMCs | — | A single nucleotide polymorphism in the IL-37 gene (rs3811047) confers a significant risk of CAD. |
Yu et al.31 | 2017 | 28992508 | Human | GDM |
Placenta Umbilical tissue Serum |
— | IL-37 may have a protective role in the development of GDM. |
Ji et al.23 | 2017 | 28607385 |
Mouse Human biopsies |
Atherosclerosis |
Artery plasma |
IP injection | IL-37 reduced proinflammatory cell populations. |
Ellisdon et al.42 | 2017 | 28783685 |
Mouse In vitro |
Endotoxemia |
Plasma PBMCs |
IP injection | IL-37 monomer was more effective at suppressing proinflammatory events. |
Li et al.10,17 | 2017 | 28237874 |
Mouse In vitro |
Endotoxemia | Heart | Transgenic | IL-37 suppressed myocardial inflammation. |
McCurdy et al.22 | 2017 | 29030487 |
Mouse In vitro |
Atherosclerosis | Arteries | Transgenic | IL-37 reduced plaque growth, lipid intake, and pro-inflammatory cytokines by macrophages. |
Liu et al.20 | 2017 | 28237549 | Human | ACS | Plasma | — | IL-37 levels correlated with worse outcomes for myocardial infarction patients. |
Ballak et al.29 | 2018 | 30006351 | Mouse | High fat diet |
Adipose Liver Plasma |
IP injection | IL-37 reduced proinflammatory cytokines and improved glucose tolerance. |
Kuipers et al.30 | 2018 | 30072596 | Mouse | High fat diet | Plasma | Transgenic | IL-37 reduced food intake and body weight. |
Li et al.34 | 2018 | 29956068 | Mouse | Pulmonary fibrosis | Lungs | Transgenic | Il-37 is useful in preventing pulmonary fibrosis induced by bleomycin. |
Liu et al.24 | 2018 | 29439249 |
Mouse Human biopsies In vitro |
Atherosclerosis |
Vascular plaques T cells VSMCs |
Transgenic | Overexpression of IL-37 decreased atherosclerotic burden. |
Lv et al.39 | 2018 | 29319845 |
Mouse In vitro |
Asthma | Lungs | Intranasal spray | IL-37 attenuated HDM-induced asthma. |
Telikani et al.33 | 2019 | 30907193 | Human | Type 2 DM | PBMC | — | Sitagliptin plus vitamin D3 upregulated IL-37. |
Mao et al.8 | 2019 | 31686988 |
Human In vitro |
ACS | PBMC | Exogenous in vitro stimulation | IL-37 treated DCs reduced T-regs and expanded Th1 and Th17 cells. |
Wang et al.32 | 2019 | 30362558 |
Human In vitro |
GDM | Macrophages | — | miR-657 reduced IL-37 levels in gestational diabetes mellitus. |
Feng et al.36 | 2019 | 30735937 | Mouse | Hepatic damage | Liver | IV injection | IL-37 reduced proinflammatory cytokines and increased anti-inflammatory T-cells and macrophages. |
Zhang et al.26 | 2019 | 31061403 |
Mouse Human biopsies |
Ischemic stroke |
Brain Microglia |
Transgenic | IL-37 reduced severe locomotor deficits and cerebral infarct size. |
Kim et al.35 | 2019 | 31519861 |
Mouse Human biopsies In vitro |
Pulmonary fibrosis | Lungs |
IP injection Exogenous in vitro stimulation |
IL-37 reduced driving mechanisms of lung fibrosis. |
Liu et al.25 | 2019 | 31125703 |
Mouse In vitro |
Atherosclerosis | DCs | Transgenic | IL-37 inhibited DC maturation through the IL-1R8-TLR4-NF-κB pathway. |
Liu et al.41 | 2020 | 32111731 | Human | Myasthenia gravis | Plasma | — | IL-37 reduced proliferation and expression of proinflammatory cells. |
Lotfy et al.7 | 2020 | 32366176 | Human | Atherosclerosis |
Arterial tissue Blood T-regs |
Exogenous in vitro stimulation | IL-37 levels were increased in patients with chronic lower limb atherosclerotic ischemia suggesting an anti-inflammatory role for IL-37 involving T-regs. |
Racca et al.21 | 2020 | 32451455 | Human | Post-operative AF | Plasma | — | IL-37 correlated with rehabilitation post operation |
Zhang et al.18 | 2020 | 32801675 | Human | Ischemic stroke | Plasma | — | Plasma IL-37 levels were significantly increased after acute ischemic stroke and was associated with unfavorable prognoses. |
Zhou et al.15 | 2020 | 32117982 |
Human In vitro |
Inflammation | Macrophages | Exogenous in vitro stimulation | Il-37 inhibited M1 macrophage polarization via the inhibition of Notch1 and nuclear factor kappa B pathways. |
Li et al.6 | 2020 | 31746393 |
Human In vitro |
CHD | PBMC | Exogenous in vitro stimulation | IL-37 correlated with CHD and reduced proinflammatory cytokines in vitro. |
Lei et al.38 | 2020 | 32855679 | Mouse | Allergic rhinitis | Nasal tissue | IP injection | IL-37 alleviated OVA-induced allergic symptoms. |
Li et al.27 | 2020 | 32916625 | Mouse | Ischemia-reperfusion | Heart | Transgenic cardiospheres | IL-37 maintained LV function and reduced infarct size. |
Xu et al.40 | 2020 | 31604808 | Mouse | SLE |
Serum MSCs T cells |
Transgenic cell transplantation | Mice transplanted with IL-37 overexpressing cells displayed improved survival and showed reduced signs of SLE. |
Hou et al.37 | 2020 | 32411145 |
Mouse In vitro |
Atopic dermatitis |
Eosinophils Fibroblasts |
Transgenic Exogenous in vitro stimulation |
IL-37 treatment resulted in an overall decrease of inflammatory mechanisms. |
ACS, Acute coronary syndrome; AF, atrial fibrillation; CAC, coronary artery calcification; CAD, coronary artery disease; CHD, coronary heart disease; DC, dendritic cell; GDM, gestational diabetes mellitus; hsCRP, high sensitivity C-reactive protein; HDM, house dust mites; HCAEC, human coronary artery endothelial cells; IP, intraperitoneal; IV, intravenous; LV, left ventricle; MSC, mesenchymal stem cells; MI, myocardial infarction; OPG, osteoprotegerin; OVA, ovalbumin; PBMC, peripheral blood mononuclear cells; Treg, regulatory T cell; SLE, systemic lupus erythematosus; Type 2 DM, type 2 diabetes mellitus; VSMC, vascular smooth muscle cells.
Conclusion
As an anti-inflammatory cytokine with expanding role in the regulation of inflammation, IL-37 has the potential to play a prominent role in therapeutic treatment of CVD. However, in order for this to be realized, there needs to be a deeper understanding of its role in various cardiovascular pathologies as well as ways to manipulate and administer the cytokine as a potential therapeutic agent against CVD. Thus, although promising at this time whether IL-37 can be used one day as a therapeutic agent against CVD in humans will depend on more studies.
Acknowledgements
This work was performed within the Russian Government Program of Competitive Growth of Kazan Federal University.
Conflict of interest. None declared.
Contributor Information
S McCurdy, Department of Medicine, University of California San Diego, San Diego, CA, USA.
J Yap, Department of Medicine, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
J Irei, Department of Medicine, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
J Lozano, Department of Medicine, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
W A Boisvert, Department of Medicine, Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA; Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlevskaya Str., Kazan, 420008, Russia.
References
- 1. Kumar S, Hanning CR, Brigham-Burke MR, Rieman DJ, Lehr R, Khandekar S, et al. Interleukin-1F7B (IL-1H4/IL-1F7) is processed by caspase-1 and mature IL-1F7B binds to the IL-18 receptor but does not induce IFN-gamma production. Cytokine 2002; 18:61–71. [DOI] [PubMed] [Google Scholar]
- 2. Nold MF, Nold-Petry CA, Zepp JA, Palmer BE, Bufler P, Dinarello CA.. IL-37 is a fundamental inhibitor of innate immunity. Nat Immunol 2010; 11:1014–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Yin D, Naji DH, Xia Y, Li S, Bai Y, Jiang G, et al. Genomic variant in IL-37 confers a significant risk of coronary artery disease. Sci Rep 2017; 7:42175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Yan J, Zhang Y, Cheng S, Kang B, Peng J, Zhang X, et al. Common genetic heterogeneity of human interleukin-37 leads to functional variance. Cell Mol Immunol 2017; 14:783–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Klück V, van Deuren RC, Cavalli G, Shaukat A, Arts P, Cleophas MC, et al. Rare genetic variants in interleukin-37 link this anti-inflammatory cytokine to the pathogenesis and treatment of gout. Ann Rheum Dis 2020; 79:536–44. [DOI] [PubMed] [Google Scholar]
- 6. Li H, Shen C, Chen B, Du J, Peng B, Wang W, et al. Interleukin-37 is increased in peripheral blood mononuclear cells of coronary heart disease patients and inhibits the inflammatory reaction. Mol Med Rep 2020; 21:151–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Lotfy H, Moaaz M, Moaaz M.. The novel role of IL-37 to enhance the anti-inflammatory response of regulatory T cells in patients with peripheral atherosclerosis. Vascular 2020; 28:629–42. [DOI] [PubMed] [Google Scholar]
- 8. Mao X, Zhu R, Zhang F, Zhong Y, Yu K, Wei Y, et al. IL-37 plays a beneficial role in patients with acute coronary syndrome. Mediators Inflamm 2019; 2019:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Xie Y, Li Y, Cai X, Wang X, Li J.. Interleukin-37 suppresses ICAM-1 expression in parallel with NF-kappaB down-regulation following TLR2 activation of human coronary artery endothelial cells. Int Immunopharmacol 2016; 38:26–30. [DOI] [PubMed] [Google Scholar]
- 10. Li J, Zhai Y, Ao L, Hui H, Fullerton DA, Dinarello CA, et al. Interleukin-37 suppresses the inflammatory response to protect cardiac function in old endotoxemic mice. Cytokine 2017; 95:55–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Budoff MJ, Hokanson JE, Nasir K, Shaw LJ, Kinney GL, Chow D, et al. Progression of coronary artery calcium predicts all-cause mortality. JACC Cardiovasc Imaging 2010; 3:1229–36. [DOI] [PubMed] [Google Scholar]
- 12. Yu K, Min X, Lin Y, Huang Y, Huang S, Liu L, et al. Increased IL-37 concentrations in patients with arterial calcification. Clin Chim Acta 2016; 461:19–24. [DOI] [PubMed] [Google Scholar]
- 13. Chai M, Zhang HT, Zhou YJ, Ji QW, Yang Q, Liu YY, et al. Elevated IL-37 levels in the plasma of patients with severe coronary artery calcification. J Geriatr Cardiol 2017; 14:285–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Chai M, Ji Q, Zhang H, Zhou Y, Yang Q, Zhou Y, et al. The protective effect of interleukin-37 on vascular calcification and atherosclerosis in apolipoprotein E-deficient mice with diabetes. J Interferon Cytokine Res 2015; 35:530–9. [DOI] [PubMed] [Google Scholar]
- 15. Zhou P, Li Q, Su S, Dong W, Zong S, Ma Q, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Ye C, Zhang W, Hang K, Chen M, Hou W, Chen J, et al. Extracellular IL-37 promotes osteogenic differentiation of human bone marrow mesenchymal stem cells via activation of the PI3K/AKT signaling pathway. Cell Death Dis 2019; 10:753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Li TT, Zhu D, Mou T, Guo Z, Pu JL, Chen QS, et al. IL-37 induces autophagy in hepatocellular carcinoma cells by inhibiting the PI3K/AKT/mTOR pathway. Mol Immunol 2017; 87:132–40. [DOI] [PubMed] [Google Scholar]
- 18. Zhang F, Zhu T, Li H, He Y, Zhang Y, Huang N, et al. Plasma interleukin-37 is elevated in acute ischemic stroke patients and probably associated with 3-month functional prognosis. Clin Interv Aging 2020; 15:1285–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Shou X, Lin J, Xie C, Wang Y, Sun C.. Plasma IL-37 elevated in patients with chronic heart failure and predicted major adverse cardiac events: a 1-year follow-up study. Dis Markers 2017; 2017:1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Liu K, Tang Q, Zhu X, Yang X.. IL-37 increased in patients with acute coronary syndrome and associated with a worse clinical outcome after ST-segment elevation acute myocardial infarction. Clin Chim Acta 2017; 468:140–4. [DOI] [PubMed] [Google Scholar]
- 21. Racca V, Torri A, Grati P, Panzarino C, Marventano I, Saresella M, et al. Inflammatory cytokines during cardiac rehabilitation after heart surgery and their association to postoperative atrial fibrillation. Sci Rep 2020; 10:8618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. McCurdy S, Baumer Y, Toulmin E, Lee B-H, Boisvert WA.. Macrophage-specific expression of IL-37 in hyperlipidemic mice attenuates atherosclerosis. J Immunol 2017; 199:3604–13. [DOI] [PubMed] [Google Scholar]
- 23. Ji Q, Meng K, Yu K, Huang S, Huang Y, Min X, et al. Exogenous interleukin 37 ameliorates atherosclerosis via inducing the Treg response in ApoE-deficient mice. Sci Rep 2017; 7:3310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Liu J, Lin J, He S, Wu C, Wang B, Liu J, et al. Transgenic overexpression of IL-37 protects against atherosclerosis and strengthens plaque stability. Cell Physiol Biochem 2018; 45:1034–50. [DOI] [PubMed] [Google Scholar]
- 25. Liu T, Liu J, Lin Y, Que B, Chang C, Zhang J, et al. IL-37 inhibits the maturation of dendritic cells through the IL-1R8-TLR4-NF-kappaB pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1338–49. [DOI] [PubMed] [Google Scholar]
- 26. Zhang SR, Nold MF, Tang S-C, Bui CB, Nold CA, Arumugam TV, et al. IL-37 increases in patients after ischemic stroke and protects from inflammatory brain injury, motor impairment and lung infection in mice. Sci Rep 2019; 9:6922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Li J, Zhang WJ, Yao H, Li TM.. Therapeutic effects of interleukin-37 and induced cardiosphere on treating myocardial ischemia-reperfusion injury. Int Immunopharmacol 2020; 88:106719. [DOI] [PubMed] [Google Scholar]
- 28. Zhu R, Sun H, Yu K, Zhong Y, Shi H, Wei Y, et al. Interleukin-37 and dendritic cells treated with interleukin-37 plus troponin I ameliorate cardiac remodeling after myocardial infarction. J Am Heart Assoc 2016; 5:e004406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Ballak DB, Li S, Cavalli G, Stahl JL, Tengesdal IW, van Diepen JA, et al. Interleukin-37 treatment of mice with metabolic syndrome improves insulin sensitivity and reduces pro-inflammatory cytokine production in adipose tissue. J Biol Chem 2018; 293:14224–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Kuipers EN, van Dam AD, Ballak DB, de Wit EA, Dinarello CA, Stienstra R, et al. IL-37 expression reduces lean body mass in mice by reducing food intake. Int J Mol Sci 2018; 19:2264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Yu Z, Liu J, Zhang R, Huang X, Sun T, Wu Y, et al. IL-37 and 38 signalling in gestational diabetes. J Reprod Immunol 2017; 124:8–14. [DOI] [PubMed] [Google Scholar]
- 32. Wang P, Wang H, Li C, Zhang X, Xiu X, Teng P, et al. Dysregulation of microRNA-657 influences inflammatory response via targeting interleukin-37 in gestational diabetes mellitus. J Cell Physiol 2019; 234:7141–8. [DOI] [PubMed] [Google Scholar]
- 33. Telikani Z, Sheikh V, Zamani A, Borzouei S, Salehi I, Amirzargar MA, et al. Effects of sitagliptin and vitamin D3 on T helper cell transcription factors and cytokine production in clinical subgroups of type 2 diabetes mellitus: highlights upregulation of FOXP3 and IL-37. Immunopharmacol Immunotoxicol 2019; 41:299–311. [DOI] [PubMed] [Google Scholar]
- 34. Li Y, Gao Q, Xu K, Peng X, Yuan X, Jiang W, et al. Interleukin-37 attenuates bleomycin-induced pulmonary inflammation and fibrosis in mice. Inflammation 2018; 41:1772–9. [DOI] [PubMed] [Google Scholar]
- 35. Kim MS, Baek AR, Lee JH, Jang AS, Kim DJ, Chin SS, et al. IL-37 attenuates lung fibrosis by inducing autophagy and regulating TGF-beta1 production in mice. J Immunol 2019; 203:2265–75. [DOI] [PubMed] [Google Scholar]
- 36. Feng XX, Chi G, Wang H, Gao Y, Chen Q, Ru YX, et al. IL-37 suppresses the sustained hepatic IFN-gamma/TNF-alpha production and T cell-dependent liver injury. Int Immunopharmacol 2019; 69:184–93. [DOI] [PubMed] [Google Scholar]
- 37. Hou T, Sun X, Zhu J, Hon KL, Jiang P, Chu IM, et al. IL-37 ameliorating allergic inflammation in atopic dermatitis through regulating microbiota and AMPK-mTOR signaling pathway-modulated autophagy mechanism. Front Immunol 2020; 11:752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Lei H, Sun Y, Quan S.. IL-37 relieves allergic inflammation by inhibiting the CCL11 signaling pathway in a mouse model of allergic rhinitis. Exp Ther Med 2020; 20:3114–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Lv J, Xiong Y, Li W, Cui X, Cheng X, Leng Q, et al. IL-37 inhibits IL-4/IL-13-induced CCL11 production and lung eosinophilia in murine allergic asthma. Allergy 2018; 73:1642–52. [DOI] [PubMed] [Google Scholar]
- 40. Xu J, Chen J, Li W, Lian W, Huang J, Lai B, et al. Additive therapeutic effects of mesenchymal stem cells and IL-37 for systemic lupus erythematosus. J Am Soc Nephrol 2020; 31:54–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Liu Z, Zhu L, Lu Z, Chen H, Fan L, Xue Q, et al. IL-37 represses the autoimmunity in myasthenia gravis via directly targeting follicular Th and B cells. J Immunol 2020; 204:1736–45. [DOI] [PubMed] [Google Scholar]
- 42. Ellisdon AM, Nold-Petry CA, D'Andrea L, Cho SX, Lao JC, Rudloff I, et al. Homodimerization attenuates the anti-inflammatory activity of interleukin-37. Sci Immunol 2017; 2:eaaj1548. [DOI] [PubMed] [Google Scholar]