Elsevier

Toxicology Letters

Volume 333, 15 October 2020, Pages 150-158
Toxicology Letters

Methamphetamine induced neuroinflammation in mouse brain and microglial cell line BV2: Roles of the TLR4/TRIF/Peli1 signaling axis

https://doi.org/10.1016/j.toxlet.2020.07.028Get rights and content

Highlights

  • Downregulation of Peli1 strikingly suppresses Meth-induced neuroinflammation.

  • The TLR4-TRIP axis is important for Peli1 modulation in Meth treated microglial cells.

  • Cytokines and signaling are dramatically decreased after TLR4-TRIP-Peli1 inhibition.

  • The TLR4-TRIP-Peli1 axis is obviously activated in the Meth-treated mouse brain.

Abstract

Methamphetamine (Meth), a highly addictive drug, can induce irreversible neuronal damage and cause neuropsychiatric and cognitive disorders. Meth's effects on modulating microglial neuroimmune functions and eliciting neuroinflammation have attracted considerable attention in recent years. Recent evident of the effect of the non-dependent domain containing adaptor inducing interferon (TRIF)/Pellino1 (Peli1) signaling axis on pro-inflammatory cytokine production provides novel clues for inflammation. Therefore, our study investigated Meth-induced neurotoxicity from a neuropathological perspective by examining TLR4-TRIF-Peli1 axis signaling activation. Meth significantly activated microglia accompanied by marked increase of TLR4 and TRIF expression, NF-kB and MAPK pathways activation and the production of IL-1β, TNF-α and IL-6. Peli1 was involved in Meth-mediated neuroinflammation and knockdown of Peli1 strongly reversed NF-kB and MAPK pathways activation and pro-inflammatory cytokine excretion. Intriguingly, Peli1 upregulation induced by Meth was dependent on TRIF rather than the myloid differentiation factor 88 (MyD88) pathway, since the silencing of TRIF significantly suppressed Meth-induced Peli1 upregulation, while MyD88 knockdown had no obvious impact. Additionally, an in vivo study verified TLR4-TRIF-Peli1 axis activation and an enhanced level of downstream cytokine expression in the cortex after Meth treatment. Therefore, these findings provide new insight regarding the specific contributions of the TRIF-Peli1 pathway to Meth-mediated neuroinflammation.

Introduction

Methamphetamine (Meth) abuse has become a global public health problem, with more than 33 million people using the drug worldwide; this number continues to grow due to its availability, ease of use, low price, and high potential for addiction (Courtney and Ray, 2014). Mounting reports have revealed that Meth exposure can induce irreversible neuronal damage and cause neuropsychiatric and cognitive disorders in which neuroinflammation may be involved (Kaushal et al., 2013; Xu et al., 2017). However, the mechanism underlying Meth induced neuroinflammation has yet to be clarified.

As resident innate immune cells in the central nervous system (CNS) (Yang and Zhou, 2019), microglial cells are the most motile cells and serve as the first line of defense in the CNS. Microglial activation may occur as a consequence of exogenous or endogenous stimuli, accompanied by the excretion of cytokines, including IL-1β, TNF-α and IL-6. Neuroinflammation is through to underlie numerous types of CNS injury and neurodegenerative diseases, including depression, anxiety, Parkinson's disease and Alzheimer's disease (Xu et al., 2018; Yang and Zhou, 2019).

Meth exposure has been proposed to induce neural damage in a direct or indirect manner. Our previous study showed that Meth treatment contributes to Alzheimer's disease like changes, including p-tau and amyloid precursor protein (APP) enhancement, via the inhibition of insulin signaling, which has suppressive effects on inflammation (Xu et al., 2018; Chen et al., 2019). Increasing evidence has indicated that Meth induces disturbances in microglial homeostasis through microglial activation in the brain (Kaushal et al., 2013; Xu et al., 2017) in which the Toll-like receptors (TLRs) exert important roles.

TLRs are highly expressed in glial cells and mediate microglia activation. Toll-like receptor 4 (TLR4) is one of the most important receptors in the TLR family because it is the only one that can activate the MyD88 and the TRIF pathway (Trudler et al., 2010; Shirjang et al., 2017). TRIF is a TLR adapter and is unique to TLR3 and TLR4-mediated signaling pathways (Ullah et al., 2016). It mediates signaling from TLR4 through the activation of the TRIF-related adaptor molecule (TRAM) followed by regulating interferon regulatory factor 3 (IRF3) and NF-κB expression, which may potentiate the production of inflammatory factors in the late stage of inflammatory response process (Lundberg et al., 2013; Zhu et al., 2019). Recent evidence has suggested that blocking the TLR4 receptor abolishes the inflammatory signaling response in microglial cells, indicating the critical importance of TLR4 in microglia-mediated neuroinflammation (Kerfoot et al., 2004). Regarding the potential roles of TLR4 in Meth-mediated neuroinflammation, our recent work revealed a pronounced increase in TLR4 expression as well as inflammatory factor release in microglial cells after Meth treatment (Wan et al., 2017), however, the activation process linking TLR4 and inflammatory factors, remains poorly understood.

Peli1, an E3 ubiquitin ligase, has been reported to be essential for the modulation of TLR signaling. Available evidence has shown that the knockdown of Peli1 markedly suppresses the activation of NF‑κB and the expression of pro-inflammatory genes (Chang et al., 2009; Jiang et al., 2003). In a mouse encephalomyelitis (EAE) model, Peli1 was proven to participate in TLR-induced pro-inflammatory and chemokine gene expression via the MyD88-induced activation of the ERK, JNK and p38 MAPK pathways. Of note, the Peli1 protein is highly expressed in the brain and predominantly in microglia, demonstrating the crucial role of Peli1 in neuroinflammation (Parikh et al., 2015). Peli1 was demonstrated to be upregulated after Meth treatment in our recent study; the potential mechanisms involving Peli1-mediated signaling induced by Meth, however, remain poorly understood.

Combining the critical roles of Peli1 in neuroinflammation and the effects of Meth on Peli1, the current study expanded and strengthened our understanding of the potential mechanisms underlying Meth induced neurotoxicity along the TLR4-MyD88/TRIF-Peli1 axis. Here, we provided the direct evidence that Peli1 was involved in Meth induced neuroinflammation, and the TLR4-TRIF axis rather than TLR4-MyD88 axis was participated in Peli1 modulation. Meanwhile, an in vivo study was also performed, with the significant activation of TLR4-TRIF-Peli1 axis in Meth exposure mouse model. Therefore, targeting the TLR4-TRIF-Peli1 axis may provide the novel strategies for Meth induced neuroinflammation.

Section snippets

Animals

Eight-week-old male C57BL/6 J mice were purchased from the Experimental Animal Center of Nanjing Medical University (China). All animals were housed in a pathogen-free environment [12 h light: dark cycle, room temperature (RT) of 21 ± 1 °C relative humidity 40∼70%] with sufficient food and water. All experiments were approved by the Animal Experiment Ethics Committee of Nanjing Medical University (permission number: IACUC-1801008). The mice were weight matched and randomly divided into the

Meth induces microglial cell activation and inflammatory cytokine expression

To assess Meth-induced neuroinflammation, BV2 cells were incubated with Meth (0,100, 300 and 900 μM) for 24 h, and CD11b was detected. Meth above 100 μM substantially enhanced the expression of CD11b, with a peak response at 900 μM (Fig. 1A). Considering the cell injury (Fig. S1A) and the concentrations of Meth levels (from ≤2 μM to 600 μM) found in the blood, urine and tissue of Meth users (Reynolds et al., 2007), 300 μM Meth was chosen for the subsequent experiments, this concentration of

Discussion

Previous studies have revealed that Meth exposure contributes to TLR4 upregulation and cytokine release which might be regulated by NF-κB and MAPK signaling (Murphy et al., 2015; Du et al., 2017). However, the potential link between TLR4 and inflammatory signaling mediated by Meth remains poorly understood. In this study, we validated the effects of Peli1 in Meth-induced neuroinflammation, and expanded our previous finding that the TRIF-Peli1 signaling axis is involved in the Meth-mediated

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

This work was supported by the National Natural Science Foundation of China (81673213, 81701050), the Natural Science Foundation of Jiangsu Province (BK20191349), the Key Disease of Jiangsu Province Science and Technology Department (BL2014088), the Jiangsu Planned Projects for Postdoctoral Research Funds (2018K075B) and the Six talent peaks project in Jiangsu Province (WSW-329).

References (34)

  • S. Shirjang et al.

    Toll-like receptors as a key regulator of mesenchymal stem cell function: an up-to-date review

    Cell Immunol

    (2017)
  • H. Xu et al.

    Involvement of insulin signalling pathway in methamphetamine-induced hyperphosphorylation of Tau

    Toxicology

    (2018)
  • M. Chang et al.

    Peli1 facilitates TRIF-dependent Toll-like receptor signaling and proinflammatory cytokine production

    Nat Immunol

    (2009)
  • M. Chang et al.

    The ubiquitin ligase Peli1 negatively regulates T cell activation and prevents autoimmunity

    Nat Immunol

    (2011)
  • C.J. Cummings et al.

    Expression and function of the chemokine receptors CXCR1 and CXCR2 in sepsis

    J Immunol

    (1999)
  • D. Dai et al.

    Peli1 controls the survival of dopaminergic neurons through modulating microglia-mediated neuroinflammation

    Sci Rep

    (2019)
  • S.H. Du et al.

    Toll-like receptor 4 mediates methamphetamine-induced neuroinflammation through caspase-11 signaling pathway in astrocytes

    Front Mol Neurosci

    (2017)
  • Cited by (25)

    • Alpha-pyrrolidinopentiothiophenone (α-PVT) activates the TLR–NF-κB–MAPK signaling pathway and proinflammatory cytokine production and induces behavioral sensitization in mice

      2022, Pharmacology Biochemistry and Behavior
      Citation Excerpt :

      For example, TLR4 is known to detect and respond to morphine and cocaine, triggering a proinflammatory central immune signaling cascade (Hutchinson et al., 2010; Northcutt et al., 2015). In addition, it has been reported that exposure to methamphetamine contributes to TLR4 upregulation and cytokine release, which might be regulated by NF-κB and MAPK signaling in mouse astrocytes or microglial cells (Du et al., 2017; Murphy et al., 2015; Wan et al., 2017; Yang et al., 2020). The NF-κB and MAPK signaling pathways—more specifically, ERK, p38, and JNK—have been known to be highly relevant in regulating inflammation and innate immune responses (Soares-Silva et al., 2016).

    • LPS-mediated activation of TLR4 controls Toxoplasma gondii growth in human trophoblast cell (BeWo) and human villous explants in a dependent-manner of TRIF, MyD88, NF-κB and cytokines

      2022, Tissue and Cell
      Citation Excerpt :

      In this sense, the present study is innovative and necessary, since it is the first study to demonstrate the influence of TLR4, MyD88 and TRIF in the control of T. gondii infection in human trophoblast cells (BeWo line) and human villous explants. After that, we investigated the role of NF-κB during T. gondii infection in both BeWo cells and villous explants, since this transcription factor is triggered by MyD88 (Botos et al., 2011; Melo et al., 2011; Dupont et al., 2012) and, alternatively, by TRIF, besides IRFs (Lundberg et al., 2013; Yang et al., 2020). When both BeWo cells and villous explants were inhibited to NF-κB, there was a significant increase in T. gondii proliferation, showing that NF-κB is important, together with MyD88 and TRIF, to control parasite infection in these experimental models of maternal-fetal interface.

    • Covid-19 interface with drug misuse and substance use disorders

      2021, Neuropharmacology
      Citation Excerpt :

      Although both TNF-α and IL-1β, cytokines that modulate the activity of serotonin, dopamine, and glutamate, are canonically increased during viral infections, such as HIV (Chivero et al., 2017; Vartak-Sharma et al., 2014) and Covid-19 (McElvaney et al., 2020; Zhang et al., 2020), substances of abuse can also regulate activity of these cyokines. For instance, inflammatory cytokines and chemokines are increased and decreased following exposure to cocaine (Atluri et al., 2016; Kovalevich et al., 2015; Montesinos et al., 2020; Souza et al., 2021), methamphetamine (Karimi-Haghighi et al., 2020; Kays and Yamamoto, 2019; Wang et al., 2018; Yang et al., 2020b) and/or alcohol (Barr et al., 2016; Crews et al., 2006) and dysregulated following opioid exposure (Cisneros and Cunningham, 2021; Ezeomah et al., 2020). For example, we demonstrated increased and decreased inflammation in the nucleus accumbens relative to the hippocampus in rats withdrawn from chronic self-administration of the opioid fentanyl; these outcomes positively correlated to expression of specific host pattern recognition receptors that trigger inflammatory cascades (Cisneros and Cunningham, 2021; Ezeomah et al., 2020).

    View all citing articles on Scopus
    1

    These authors contributed equally to this study.

    View full text