Elsevier

Chemosphere

Volume 218, March 2019, Pages 589-598
Chemosphere

RIP1 and RIP3 contribute to Tributyltin-induced toxicity in vitro and in vivo

https://doi.org/10.1016/j.chemosphere.2018.11.140Get rights and content

Highlights

  • TBT-induced cell death was significantly downregulated by RIP1 inhibition or knockdown, or RIP3 deficiency.

  • The mortality induced by acute TBT exposure was markedly reduced by RIP1 inhibitor pretreatment or RIP3 deficiency.

  • The immunotoxic effects induced by TBT were attenuated in RIP3−/− mice or WT mice treated with RIP1 inhibitor.

Abstract

Tributyltin (TBT), a widely distributed environmental pollutant, is toxic to animals and human beings. Although its toxicity, especially the immunosuppressive effect, has been reported a lot, the underlying molecular mechanisms are still unclear. In this study, we investigated the mechanisms of TBT-induced cytotoxicity both in vitro and in vivo. TBT induced cell death in both J774A.1 macrophages and mouse bone marrow-derived macrophages (BMDMs) as measured by the LDH and Annexin V-FITC/PI dual staining assays. Pretreatment with RIP1 inhibitor Necrostatin-1 (Nec-1) or transfection with Rip1 siRNA significantly suppressed TBT-induced cytotoxicity in J774A.1 macrophages or human embryonic kidney cell line (HEK293 cells). TBT-induced cell death was also markedly inhibited in RIP3−/− BMDMs. In agreement with in vitro results, TBT-induced in vivo immunotoxic effects including leukocyte depletion and thymus atrophy were significantly attenuated in RIP3−/− mice or WT mice treated with Nec-1. Notably, the mortality rate induced by TBT was remarkably reduced in RIP3−/− mice (100% vs. 12.5% lethality) or Nec-1-treated mice (100% vs. 59.2% lethality) respectively. These results reveal a critical role of RIP1 and RIP3 in TBT-induced toxicity both in vitro and in vivo.

Introduction

Tributyltin (TBT), which is an organotin compound, has been reported to cause neurotoxicity, reproductive/developmental toxicity and immunotoxicity and hence the use of TBT is currently restricted in some fields (Boyer, 1989; Pagliarani et al., 2013). However, due to its previous widespread use in agriculture and industry (Boyer, 1989; Fent, 1996), the risk of human exposure to TBT through drinking water and contaminated fish products is still high (Sousa et al., 2017). TBT has been detected in human blood and liver with concentration up to 8.18 ng mL−1 (Kannan and Giesy, 1999). Therefore, the toxic effects of TBT and the underlying mechanisms should be comprehensively investigated.

High dose of TBT treatment causes the acute toxicity, and results in high mortality rate in animals (Takagi et al., 1992; Ohtaki et al., 2007). Additionally, the toxicity of TBT on immune system and its immunosuppressive effects have been extensively reported. TBT exposure can cause thymus atrophy, depletion of T-lymphocytes in spleen and lymph nodes, decreased macrophage and NK cell activity, and diminished T-helper (Th) 2 cell polarization (Snoeij et al., 1987; Boyer, 1989; Kergosien and Rice, 1998; Whalen et al., 2002; Kato et al., 2006). However, the molecular mechanisms by which TBT exerts these immunotoxic effects are still not clear. Previous studies proposed that TBT induced thymus atrophy through inhibiting cell proliferation or inducing apoptosis in immune cells (Raffray and Cohen, 1993; Gennari et al., 1997; Tomiyama et al., 2009). Regarding the underlying mechanism of TBT-induced apoptosis, although different mechanisms including intracellular Ca2+ overload, inhibition of ATP synthesis, caspase activation and release of cytochrome c have been detected and reported (Viviani et al., 1995; Stridh et al., 2001; Tomiyama et al., 2009), however, there is no direct genetic evidence confirming the mechanisms of cytotoxicity and accordingly the molecular targets by TBT have not been identified yet. Moreover, some studies have reported that high concentration of TBT also induced necrosis (Stridh et al., 1999b; Botelho et al., 2015; Khondee et al., 2016), suggesting that TBT-induced cytotoxicity is not limited to apoptosis, and other mode of cell death might also contribute to the immunotoxicity of TBT. And the studies which investigate the mechanism of TBT-induced necrosis are still lacking.

Necrosis, which has gained intensive investigations in recent years, represents another type of programmed cell death in addition to apoptosis. Programmed necrosis can be tightly regulated by two Receptor-interacting protein (RIP) family members, RIP1 and RIP3. Upon exposure to death stimuli, RIP1 interacts with RIP3, which leads to the assembly of necrosome complex and the induction of necrosis via downstream executor MLKL (mixed-lineage kinase domain-like) (Sun et al., 2012; Wu et al., 2013; Christofferson et al., 2014; Chan et al., 2015). Necrosis can be induced by various stimuli, including death cytokines, intracellular ATP depletion, mitochondrial depolarization, poly-(ADP-ribose) polymerase (PARP) activation, reactive oxygen species (ROS) production and certain viruses, etc (Vanlangenakker et al., 2012). To date, at least to our knowledge, there is still no report that a certain environmental pollutant can induce necrosis via RIP1 and RIP3.

In this study, we have investigated the cytotoxic effects of TBT and explored the underlying molecular mechanisms both in vitro and in vivo. Our results indicated that TBT induced the occurrence of necrotic cell death in macrophages through RIP1-RIP3 pathway. Inhibition or genetic deletion of RIP1-RIP3 pathway was able to rescue the cell death induced by TBT. Moreover, pretreatment with RIP1 inhibitor or RIP3 deficiency significantly protected the mice from TBT-induced mortality and alleviated the tissue damage as well as immunosuppressive effect induced by TBT, suggesting that RIP1 and RIP3 also contribute to the toxicity of TBT in vivo.

Section snippets

Reagents

Tri-n-butyltin chloride (TBT) (purity 99%) was purchased from J&K chemical Ltd. Necrostatin-1 (Nec-1) was purchased from Enzo Lifesciences. Dimethyl sulfoxide (DMSO) (purity≥ 99.5%) was provided by Sigma-Aldrich. Anti-RIP1 (mouse) mAb was from BD Transduction. Phospho-RIP1 (Ser166) rabbit mAb was purchased from Cell Signaling Technology. Anti-actin mAb was from Biosharp. Murine M-CSF was from Peprotech. Annexin V-FITC Apoptosis Detection kit was from Beyotime Company of Biotechnology (Shanghai,

TBT induces obvious cytotoxicity in mouse macrophages

In order to investigate the immunotoxicity of TBT, we detected the effect of TBT on macrophages, which are important immune cells involved in both innate and adaptive immune system. J774A.1 macrophages were exposed to different concentrations of TBT for 4 h and the cytotoxicity was evaluated by LDH release assay. As Fig. 1A shown, TBT induced cell death in J774A.1 macrophages in a dose-dependent manner, from 20% cell death induced by 0.4 μM TBT to 65% cell death by 2.5 μM TBT. In 0.6 μM and

Discussion

In the present study, we investigated the toxicity of TBT and the underlying molecular mechanisms both in vitro and in vivo. First, we explored the cytotoxic effects of TBT in macrophages, using both J774A.1 macrophage cell line and primary bone marrow-derived macrophages. Macrophages, as the first type of immune cells to fight against pathogens invasion, are important components of immune system and exist in almost every organ. Macrophages regulate both innate and adaptive immune responses and

Conclusion

In this study, our results reveal an obvious role of RIP1 and RIP3 in TBT-induced toxicity both in vitro and in vivo. The astonishing protective effect exerted by RIP1 inhibition or RIP3 genetic deletion suggests that RIP1 and RIP3-mediated necrosis pathway is critical for TBT-induced acute toxicity in animals. It might provide clues to investigate the underlying mechanisms of the toxicity induced by other organotin compounds.

Conflicts of interest

The authors declare no conflict of interest with the content of this article.

Acknowledgments

We would like to thank Dr. Jiahuai Han of Xiamen University for providing RIP3−/− mice. This study was supported by the National Natural Science Foundation of China under Grant 21677076 and 31500698, the Natural Science Foundation of Jiangsu Province of China under Grant BK20150772, the Innovative and Entrepreneurial Talent Cultivation (Shuangchuang) Program of Jiangsu Province, Postgraduate Research & Practice Innovation Program of Jiangsu Province, and the startup research grant at Nanjing

References (36)

  • H. Stridh et al.

    Cytochrome c release and caspase activation in hydrogen peroxide- and tributyltin-induced apoptosis

    FEBS Lett.

    (1998)
  • H. Stridh et al.

    Caspase involvement in the induction of apoptosis by the environmental toxicants tributyltin and triphenyltin

    Toxicol. Appl. Pharmacol.

    (1999)
  • L. Sun et al.

    Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase

    Cell

    (2012)
  • G.I. Vladimer et al.

    The NLRP12 inflammasome recognizes Yersinia pestis

    Immunity

    (2012)
  • M.M. Whalen et al.

    Brief butyltin exposure induces irreversible inhibition of the cytotoxic function on human natural killer cells, in vitro

    Environ. Res.

    (2002)
  • A. Aderem et al.

    Mechanisms of phagocytosis in macrophages

    Annu. Rev. Immunol.

    (1999)
  • F.K. Chan et al.

    Programmed necrosis in the cross talk of cell death and inflammation

    Annu. Rev. Immunol.

    (2015)
  • Y.W. Chen et al.

    Tributyltin exposure at noncytotoxic doses dysregulates pancreatic beta-cell function in vitro and in vivo

    Arch. Toxicol.

    (2017)
  • 1

    These authors contributed equally.

    View full text