Interleukin-18 levels and mouse Leydig cell apoptosis during lipopolysaccharide-induced acute inflammatory conditions

https://doi.org/10.1016/j.jri.2020.103167Get rights and content

Highlights

  • IL-18 is an indispensable cytokine in the maintenance of homeostasis in the testis.

  • LPS increases TNF-α and IL-6 levels but not IL-18 levels in Leydig cells.

  • IL-18 levels are significantly increased in macrophages after LPS stimulation.

  • High-dose IL-18 induces Leydig cell apoptosis via a death-receptor-mediated pathway.

  • The overproduction of IL-18 is harmful to Leydig cells during acute inflammation.

Abstract

Interleukin (IL)-18 is an inflammasome-mediated cytokine produced by germ cells, Leydig cells, and resident macrophages that is indispensable in the maintenance of homeostasis in the testis. We previously demonstrated that endogenous IL-18 induces testicular germ cell apoptosis during acute inflammation when plasma IL-18 levels are very high. However, the impact of acute inflammation and IL-18 on Leydig cells remained unclear. TM3 cells, a mouse Leydig cell line, and RAW264.7 cells, a mouse macrophage cell line, were stimulated with lipopolysaccharide (LPS) or recombinant IL-18 (rIL-18). We assessed the expression of inflammatory cytokines, caspase cleavage, and markers of apoptotic pathways. In Leydig cells, caspase 3 cleavage was increased and death-receptor-mediated apoptotic pathways were activated after LPS stimulation. However, LPS stimulation did not increase IL-18 expression in the Leydig cell line. When high-dose rIL-18 was administered to the Leydig cell line to mimic levels seem after inflammation, rIL-18 upregulated Tnf-α mRNA, Fadd mRNA, and Fas protein, promoted cleavage of caspase-8 and caspase-3, and induced apoptosis. Low-dose rIL-18 did not stimulate apoptosis. To determine if the high level of IL-18 seen in the testes after inflammation was derived from immune cells, we examined IL-18 protein expression in a macrophage cell line, RAW264.7. In contrast to the TM3 cells, IL-18 was significantly increased in RAW264.7 cells after LPS stimulation. These results suggest that high-dose IL-18 derived from macrophages is harmful to Leydig cells. Reducing the overexpression of IL-18 could be a new therapeutic approach to prevent Leydig cell apoptosis as a result of acute inflammation.

Introduction

Post-intensive-care syndrome is defined as new or worsening impairments in physical, cognitive, or mental health status arising after a critical illness and persisting beyond acute care hospitalization (Elliott et al., 2014). Several physical manifestations of post-intensive-care syndrome, such as respiratory and neuromuscular problems, have been reported, but few reports have described reproductive dysfunction and gonad physiology after a severe illness (Herridge et al., 2003; Stevens et al., 2007). Every year 20–30 million people worldwide suffer from sepsis, a life-threatening complication of severe infections, and ∼20 % of adult male patients suffering sepsis are between the peak reproductive ages of 18–45 years old (Beale et al., 2009; Reinhart et al., 2013). Recently, long-term mortality and quality of life after sepsis have been examined (Winters et al., 2010). Understanding the effects of severe illnesses on the gonads and reproductive function is crucial to understanding how quality of life may be affected.

Interleukin (IL)-18 is an important cytokine to maintain the homeostasis of testicular cells. IL-18 is produced by germ cells, Leydig cells, and resident macrophages and may regulate testicular function via autocrine and paracrine signaling under physiologic conditions (Abu Elhija et al., 2008a,d; Komsky et al., 2012; Strand et al., 2005). IL-18 is also a known inflammasome-mediated proinflammatory cytokine, and IL-18 levels are increased in mouse testes by inflammatory stimulation, such as lipopolysaccharide (LPS) administration (Abu Elhija et al., 2008b,c). We previously showed using a knockout mouse model that endogenous IL-18 has both pro-apoptotic and anti-apoptotic effects on mouse testicular germ cells during endotoxemia depending on the inflammatory stage. Endogenous IL-18 induced germ cell apoptosis during the acute phase of inflammation and suppressed germ cell apoptosis during the recovery phase (Inoue et al., 2015). We could not elucidate the effect of IL-18 on Leydig cells using this mouse model.

The primary function of Leydig cells is to synthesize and secrete androgens (Shalet, 2009). Several in vivo studies have demonstrated that LPS causes dysfunctional testicular steroidogenesis, reduces the levels of steroidogenic acute regulatory protein and 3β-hydroxysteroid dehydrogenase in the testes and decreases serum testosterone (Allen et al., 2004; O’Bryan et al., 2000; Reddy et al., 2006; Wang et al., 2020). Shortages of androgen lead to male infertility (Jungwirth et al., 2012). Thus, we hypothesized that high-dose IL-18, which is induced by inflammatory stimuli or conditions such as sepsis, would induce Leydig cell apoptosis, affecting steroidogenesis and male fertility. In this study, we investigated the impact of IL-18 on Leydig cell apoptosis under inflammatory conditions in vitro.

Section snippets

Cell culture, LPS and rIL-18 stimulation, and cell viability

A Leydig cell line (TM3) and a macrophage cell line (RAW264.7) were purchased from the American Type Culture Collection (Manassas, VA, USA). TM3 cells and RAW264.7 cells that had undergone 4 passages were used for the experiments. The TM3 cells were cultured in Dulbecco's modified eagle medium (DMEM)/F12 medium (American Type Culture Collection) supplemented with penicillin (50 U/mL), streptomycin (50 μg/mL; MP Biomedicals, Illkirch, France), 5 % horse serum (American Type Culture Collection),

LPS induced Leydig cell apoptosis via a death- receptor-mediated pathway

To establish inflammatory conditions, we treated a Leydig cell line, TM3 cells, with LPS and examined apoptosis as indicated by cleaved caspase-3 protein expression. The amount of cleaved caspase-3 in TM3 cells increased after 12 h of LPS treatment and remained elevated after 48 h of treatment with either 200 ng/mL LPS or 1000 ng/mL LPS (Fig. 1A, B). These results suggest that endotoxins induce Leydig cell apoptosis via caspase-3 dependent pathways. We also examined cleaved caspase-8 levels, a

Discussion

This study demonstrated that both LPS and high-dose rIL-18 induce apoptosis in Leydig cells via the Fas/FasL/caspase-8-dependent apoptotic pathway. Additionally, they demonstrated that LPS can induce Leydig cell apoptosis via a TNF/TNFR/caspase-8-dependent pathway. Zhou et al. (2016) showed that Leydig cell apoptosis was induced by LPS via the mitochondrial pathway. To our knowledge, our study is the first to demonstrate that Leydig cell apoptosis is also induced by LPS via

Funding

This work was supported by JSPS KAKENHI Grant Number 16K20391.

Declaration of Competing Interest

The authors declare that there are no conflicts of interest that could be perceived as prejudicing the impartiality of the research reported.

Acknowledgements

We thank Shannon Wyszomierski, PhD for editing the manuscript.

References (29)

  • M. Abu Elhija et al.

    Constitutive expression of IL-18 binding protein in murine testicular tissues and cells

    Eur. Cytokine Netw.

    (2008)
  • J.A. Allen et al.

    Bacterial endotoxin lipopolysaccharide and reactive oxygen species inhibit Leydig cell steroidogenesis via perturbation of mitochondria

    Endocrine

    (2004)
  • R. Beale et al.

    Promoting Global Research Excellence in Severe Sepsis (PROGRESS): lessons from an international sepsis registry

    Infection

    (2009)
  • D. Elliott et al.

    Exploring the scope of post-intensive care syndrome therapy and care: engagement of non-critical care providers and survivors in a second stakeholders meeting

    Crit. Care Med.

    (2014)
  • Cited by (12)

    • Prenatal and postnatal exposure to polystyrene microplastics induces testis developmental disorder and affects male fertility in mice

      2023, Journal of Hazardous Materials
      Citation Excerpt :

      Our subsequent results of transcriptomic analysis suggested the alterations in the pathways of immune response upon PS-MP exposure, and these were verified by the measurement of testicular cytokines. Similar to a previous study (Hou et al., 2021), the concentrations of IL-6, a pro-inflammatory cytokine, were increased significantly in PS-MP-exposure groups; however, the acute-phase cytokines G-CSF and IL-18 (Aslankoc et al., 2020; Inoue et al., 2020), showed no significant changes. These findings implied that long-term MP exposure primarily induced a chronic inflammatory reaction in the testis, but not persistent acute inflammation.

    • Protective role of Cytoglobin and Neuroglobin against the Lipopolysaccharide (LPS)-induced inflammation in Leydig cells ex vivo

      2022, Reproductive Biology
      Citation Excerpt :

      In the current study, an LPS-induced inflammation model was established for Leydig cells to examine the potential protective roles of both CYGB and NGB in an ex vivo system for the first time. LPS is a mammalian cell endotoxin derived from the cellular membrane of gram-negative bacteria and widely used as an inflammation inducer to examine inflammation-dependent changes in testicular function or testosterone hormone levels [23,34]. We established an LPS-induced inflammation model in TM3 mouse Leydig cell line which was genetically modified to upregulate or downregulate CYGB and NGB genes.

    • IL-18R-mediated HSC quiescence and MLKL-dependent cell death limit hematopoiesis during infection-induced shock

      2021, Stem Cell Reports
      Citation Excerpt :

      Whereas IL-18R-driven quiescence may be detrimental during acute infection by limiting blood cell production, enforced quiescence may be beneficial in long-term recovery by preventing cell death. IL-18 has been shown to induce apoptosis of various cell types under inflammatory conditions (Finotto et al., 2004; Inoue et al., 2020; Marino and Cardier, 2003). Leydig cells treated with recombinant IL-18 had increased Fas receptor, and cleaved caspases-3 and -8 (Inoue et al., 2020).

    View all citing articles on Scopus
    View full text