Elsevier

Cytokine

Volume 95, July 2017, Pages 55-63
Cytokine

Interleukin-37 suppresses the inflammatory response to protect cardiac function in old endotoxemic mice

https://doi.org/10.1016/j.cyto.2017.02.008Get rights and content

Highlights

  • IL-37 protects against cardiac dysfunction in old mice during endotoxemia.

  • Endotoxemic old IL-37tg mice have reduced myocardial inflammation.

  • IL-37 inhibits NF-κB activation in cardiac cells exposed to endotoxin.

  • IL-37 blocks endotoxin-induced inflammatory responses in cardiac cells.

Abstract

Myocardial inflammatory responses to endotoxemia are enhanced in old mice, which results in worse cardiac dysfunction. Anti-inflammatory cytokine interleukin (IL)-37 has a broad effect on innate immunoresponses. We hypothesized that IL-37 suppresses myocardial inflammatory responses to protect cardiac function during endotoxemia in old mice. Old (20–24 month) wild-type (WT), and IL-37 transgenic (IL-37tg) mice were treated with lipopolysaccharide (LPS, 0.5 mg/kg, iv) or normal saline (0.1 ml/mouse, iv). Six hours later, left ventricle (LV) function was assessed using a pressure-volume microcatheter. Levels of monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6 in plasma and myocardial tissue, as well as mononuclear cell density in the myocardium, were examined. Cardiac microvascular endothelial cells isolated from WT and IL-37tg mice were treated with LPS (0.2 µg/ml) for 0.5–24 h. Nuclear factor-kappa B (NF-κB) p65 phosphorylation was examined by immunoblotting, and MCP-1 levels in cell culture supernatant was determined using enzyme-linked immunosorbent assay. LV dysfunction in old WT endotoxemic mice was accompanied by up-regulated MCP-1, myocardial accumulation of mononuclear cells and production of TNF-α, IL-1β and IL-6. Expression of IL-37 suppressed myocardial inflammatory responses to endotoxemia in old mice, resulting in improved LV function. Treatment of old WT endotoxemic mice with recombinant IL-37 also improved LV function. In vitro experiments revealed that cardiac microvascular endothelial cells from IL-37tg mice had attenuated NF-κB activation and MCP-1 production following LPS stimulation. In conclusion, IL-37 is potent to suppress myocardial inflammation and protects against cardiac dysfunction during endotoxemia in old mice.

Introduction

Major surgery and trauma can cause endotoxemia [1]. Excessive production of pro-inflammatory mediators caused by bacterial lipopolysaccharide (LPS) frequently leads to cardiac dysfunction. We and others have observed that LPS induces cardiac contractile depression through up-regulation of myocardial production of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6 [2], [3], [4]. Importantly, morbidity and mortality due to the systemic inflammatory response syndrome associated with trauma or sepsis are significantly increased in the elderly [5], [6].

Old animals are found to display enhanced systemic and myocardial inflammatory responses to LPS [4], [7]. We previously reported that enhanced monocyte chemoattractant protein-1 (MCP-1) production in old endotoxemic mice has a critical role in mononuclear cell accumulation and cytokine production in the myocardium and contributes to the mechanism of exaggerated cardiac contractile depression. It appears that the MCP-1-mediated myocardial inflammation, i.e. mononuclear cell infiltration and production of cardiodepressant cytokines, plays an important role in cardiac contractile depression in old endotoxemic animals, and suppression of MCP-1 production and/or the MCP-1-mediated myocardial inflammation may have therapeutic potential for preservation of cardiac function in the elderly during endotoxemia.

Nuclear factor-kappa B (NF-κB) is a master transcription factor that mediates cellular inflammatory responses, including the expression of chemokines and cytokines [8]. Toll-like receptor (TLR) 4 on cell surfaces is activated in response to LPS and interacts with intracellular adaptors to activate NF-κB. Activated NF-κB translocates to the nucleus to initiate the transcription of pro-inflammatory genes [9]. Thus, NF-κB is critical in mediating LPS-induced cytokine production and cardiac dysfunction [10], [11], [12].

IL-37 is a member of the IL-1 family of cytokines. It is expressed in humans, and its expression has been identified in most cell types, including monocytes, dendritic cells, endothelial cells and epithelial cells, and acts as a natural regulator of the inflammatory responses [13]. IL-37 has repressive effects on LPS-stimulated cells, such as macrophages and endothelial cells, which implies that it may interfere with the TLR4 signaling pathway. In vitro studies show that expression of IL-37 in macrophages or epithelial cells dampens constitutive or induced production of pro-inflammatory cytokines, such as IL-1α, IL-1β, TNF-α, and macrophage inflammatory protein (MIP)-2 [14]. In vivo studies indicate that expression IL-37 in mice protects against chemical-induced colitis [15], and metabolic syndrome [16]. Further, mice that express IL-37 transgenic gene exhibit reduced lung, kidney and liver injury during endotoxemic shock [14]. However, it remains unclear whether IL-37 suppresses myocardial inflammatory responses in old animals subjected to endotoxemia. In addition, the effect of IL-37 on cardiac dysfunction caused by endotoxemia has not been determined.

In the present study, we tested the hypothesis that IL-37 suppresses the inflammatory responses in old endotoxemic mice to protect cardiac function against endotoxemic depression. We examined: (1) whether old IL-37 transgenic (IL-37tg) mice have better cardiac function during endotoxemia, (2) whether expression of IL-37 results in reduced MCP-1 production in old mice during endotoxemia and attenuated myocardial mononuclear cell accumulation and cytokine production, (3) whether IL-37 suppresses LPS-induced NF-κB activation and (4) whether recombinant IL-37 can protect old mice against endotoxemic cardiac dysfunction.

Section snippets

Animals and treatment

Adult (3–4 month old) male C57BL/6 (wild-type, WT) mice were obtained from Jackson Laboratory (Bar Harbor, Maine, USA). Male WT mice of 20–24 months old were obtained from the National Institute on Aging (Bethesda, Maryland, USA). Old (20–24 month) male IL-37tg mice were from our colonies at University of Colorado Denver Anschutz Medical Campus. IL-37tg mice are C57BL/6 background [14]. They are fertile and display normal growth, behavior and lifespan. No any unique abnormality in organ/tissue has

Old IL-37tg mice and old WT mice treated with rIL-37 have better cardiac function during endotoxemia

There was no significant difference in LV function between old WT mice and old IL-37tg mice following treatment with normal saline (Table 1 and Fig. 1). Whereas LPS caused markedly decreases in LV contractile indices in both WT and IL-37tg mice, old IL-37tg mice displayed significantly improved developed pressure, ejection fraction and cardiac output (Table 1). Administration of recombinant IL-37 following injection of LPS had similar beneficial effects on LV function (Table 1). Compared to WT

Discussion

In a previous study, we demonstrated that old mice have enhanced myocardial inflammatory responses to endotoxemia, which leads to exaggerated cardiac dysfunction [4]. Neutralizing MCP-1 improves cardiac function in old endotoxemic mice by reducing mononuclear cell accumulation in the myocardium and associated cytokine production. In the present study, we observed that old IL-37tg mice have better cardiac function during endotoxemia compared to old WT mice, and recombinant IL-37 administered to

Conclusions

Transgenic expression of IL-37 and administration of recombinant IL-37 protect against cardiac dysfunction during endotoxemia in old mice. Cardiac protection by IL-37 is associated with suppression of MCP-1 production and myocardial inflammation. In addition, expression of IL-37 inhibits NF-κB activation and suppresses MCP-1 production in cardiac microvascular endothelial cells exposed to LPS. Thus, IL-37 protects hearts of old mice against endotoxemic contractile depression through suppression

Grant support

This study was supported in part by the National Institutes on Aging grant AG039545 (to XM) and the National Natural Science Foundation of China grant 81300123 (to JL). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Conflict of interest

The authors declare that they have no competing interests.

Authors’ contributions

XM designed the study. JL, YZ, LA and HH conducted the experiments and obtained the data. JL, YZ, DF, CA and XM analyzed and interpreted the data. JL and YZ drafted the manuscript. XM, DF and CD revised the manuscript critically for important intellectual contents. All authors have approved the final version of manuscript for publication.

References (40)

  • U. Grandel et al.

    Endotoxin-induced myocardial tumor necrosis factor-alpha synthesis depresses contractility of isolated rat hearts: evidence for a role of sphingosine and cyclooxygenase-2-derived thromboxane production

    Circulation

    (2000)
  • H. Slimani et al.

    Enhanced monocyte chemoattractant protein-1 production in aging mice exaggerates cardiac depression during endotoxemia

    Crit. Care

    (2014)
  • D.C. Angus et al.

    Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care

    Crit. Care Med.

    (2001)
  • M.E. Starr et al.

    Sepsis in old age: review of human and animal studies

    Aging Dis.

    (2014)
  • J.B. Kim et al.

    Inhibition of LPS-induced iNOS, COX-2 and cytokines expression by poncirin through the NF-kappa B inactivation in RAW 264.7 macrophage cells

    Biol. Pharm. Bull.

    (2007)
  • J.H. Boyd et al.

    Toll-like receptor stimulation in cardiomyocytes decreases contractility and initiates an NF-kappa B dependent inflammatory response

    Cardiovasc. Res.

    (2006)
  • S.B. Haudek et al.

    Overexpression of cardiac I-kBa prevents endotoxin-induced myocardial dysfunction

    Am. J. Physiol. Heart Circ. Physiol.

    (2001)
  • A. Matsumori et al.

    Suppression of cytokines and nitric oxide production, and protection against lethal endotoxemia and viral myocarditis by a new NF-kappa B inhibitor

    Eur. J. Heart Fail.

    (2004)
  • M.F. Nold et al.

    IL-37 is a fundamental inhibitor of innate immunity

    Nat. Immunol.

    (2010)
  • E.N. McNamee et al.

    Interleukin 37 expression protects mice from colitis

    Proc. Natl. Acad. Sci. USA

    (2011)
  • Cited by (27)

    • Interleukin-37 alleviates myocardial injury induced by coxsackievirus B3 via inhibiting neutrophil extracellular traps formation

      2022, International Immunopharmacology
      Citation Excerpt :

      When NFκb and IκBα bind in the cytoplasm, they remain inactive [33]. Previous studies have found that IL-37 inhibits many inflammatory responses related to the inhibition of NFκb/IκBα activation [34–36]. Moreover, in our previous study [37], we found that IL-37 may promote Treg response by significantly limiting Th17 response, thereby reducing the development of myocarditis.

    • The Role of the Interleukin 1 Family in Lung Disease

      2021, Encyclopedia of Respiratory Medicine, Second Edition
    • Interleukin-37 reduces inflammation and impairs phagocytosis of platelets in immune thrombocytopenia (ITP)

      2020, Cytokine
      Citation Excerpt :

      The increase of pro-inflammatory cytokines is usually followed by an increase of anti-inflammatory cytokines as a feedback loop; however, the increase of anti-inflammatory cytokines usually cannot fully compensate the effect of proinflammatory cytokines [34]. IL-37 was induced by inflammatory stimuli including pro-inflammatory cytokines, and was discovered to impinge on the regulation of inflammation and autoimmunity as a protective factor against the uncontrolled inflammatory responses [35–40]. Conversely, neutralizing monoclonal anti-IL-37 antibodies were able to increase inflammatory cytokines production in cultured PBMCs from healthy donors [21].

    • Potential role of IL-37 in atherosclerosis

      2019, Cytokine
      Citation Excerpt :

      Interestingly, while monocytes were responsible for a majority of the expression, DCs were the only cell type to show steady state IL-37 secretion without inflammatory stimulus such as LPS [63], indicating their role in constitutive immune suppression and potential for preventing cardiovascular disease complications. The protective effects of IL-37 on myocardium also extends to aged mice (20–24 months old), including both IL-37-Tg as well as mice treated with recombinant IL-37 [35]. Following challenge with LPS, mice were analyzed for cytokine levels in myocardial tissue, as well as secretion in the serum.

    View all citing articles on Scopus
    1

    Equal contribution.

    View full text