Elsevier

Neuropharmacology

Volume 107, August 2016, Pages 68-78
Neuropharmacology

Fluoxetine protects against IL-1β-induced neuronal apoptosis via downregulation of p53

https://doi.org/10.1016/j.neuropharm.2016.03.019Get rights and content

Highlights

  • Fluoxetine alleviated neuronal injury and p53 increase in a mouse model of tMCAO.

  • Fluoxetine attenuated IL-1β-induced apoptosis and p53 elevation in cortical neurons.

  • Downregulation of p53 was required for the neuroprotective effects of fluoxetine.

  • Fluoxetine down-regulated p53 expression via a p38-dependent pathway.

Abstract

Fluoxetine, a selective serotonin reuptake inhibitor, exerts neuroprotective effects in a variety of neurological diseases including stroke, but the underlying mechanism remains obscure. In the present study, we addressed the molecular events in fluoxetine against ischemia/reperfusion-induced acute neuronal injury and inflammation-induced neuronal apoptosis. We showed that treatment of fluoxetine (40 mg/kg, i.p.) with twice injections at 1 h and 12 h after transient middle cerebral artery occlusion (tMCAO) respectively alleviated neurological deficits and neuronal apoptosis in a mouse ischemic stroke model, accompanied by inhibiting interleukin-1β (IL-1β), Bax and p53 expression and upregulating anti-apoptotic protein Bcl-2 level. We next mimicked neuroinflammation in ischemic stroke with IL-1β in primary cultured cortical neurons and found that pretreatment with fluoxetine (1 μM) prevented IL-1β-induced neuronal apoptosis and upregulation of p53 expression. Furthermore, we demonstrated that p53 overexpression in N2a cell line abolished the anti-apoptotic effect of fluoxetine, indicating that p53 downregulation is required for the protective role of fluoxetine in IL-1β-induced neuronal apoptosis. Fluoxetine downregulating p53 expression could be mimicked by SB203580, a specific inhibitor of p38, but blocked by anisomycin, a p38 activator. Collectively, our findings have revealed that fluoxetine protects against IL-1β-induced neuronal apoptosis via p38-p53 dependent pathway, which give us an insight into the potential of fluoxetine in terms of opening up novel therapeutic avenues for neurological diseases including stroke.

Introduction

Previous studies have demonstrated that neuroinflammation is one of the key factors responsible for the pathogenesis of many neurological disorders, including ischemic stroke (Kim et al., 2015a), neurodegenerative diseases (Tuon et al., 2015), and depression (Alcocer-Gomez et al., 2015). There are accumulative evidences that several inflammatory cytokines, such as interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6), contribute directly to diverse forms of neuronal injury (Rothwell, 2003). Inhibiting inflammation has currently being considered as a promising neuroprotective strategy for treatment of stroke (Murray et al., 2015), epilepsy (Tao et al., 2015) and Alzheimer's disease (AD) (Sheng et al., 1996). Therefore, seeking potential therapeutic targets and effective neuroprotectants against inflammation-induced neuronal injury is crucial for the treatment of neurological diseases.

Fluoxetine, a classical selective serotonin reuptake inhibitor (SSRI), plays a crucial role in anti-inflammation, anti-tumor and neuroprotection apart from the effect on serotonin (5-HT) (Ghosh et al., 2015). Fluoxetine is widely used as the treatment of a wide variety of psychiatric disorders, including depression (Sakr et al., 2015), bipolar disorder (Detke et al., 2015), anxiety disorders (Gupta et al., 2015). Recently, fluoxetine has been demonstrated several beneficial effects on ischemic stroke patients (Chollet et al., 2014) as well as several animal models of stroke (Lim et al., 2009, Ng et al., 2015, Schaz et al., 2011, Singh and Chopra, 2014). For example, fluoxetine reduces infarct volume and neurological deficits in a rat acute ischemic stroke (Lim et al., 2009). It also reduces TNF-α, IL-1β, and nuclear factor-κB (NF-κB) levels, and protects the loss of biogenic amines in brain tissue of ischemic rats (Singh and Chopra, 2014). Therefore, the protection of fluoxetine against cerebral ischemic injury may be associated with it's anti-inflammatory role. However, the exact mechanism for fluoxetine inhibiting neuroinflammation-induced neuronal death remains unknown.

p53, as a tumor suppressor protein, is involved in numerous signaling pathways including cell cycle arrest, apoptosis, senescence, and DNA repair (Wang et al., 2014a). Currently, p53 is recognized as an important pro-apoptotic factor in the progress of neurological disorders, such as stroke (Zhai et al., 2014), depression (Ruan et al., 2015), and epilepsy (Engel et al., 2013), neurodegenerative diseases (Singh and Pati, 2015). For example, p53 induces neuronal apoptosis via death-associated protein kinase 1 (DAPK1) pathway in ischemic brain injury (Wang et al., 2014b). Inflammatory cytokines, especially IL-1β, induce NF-κB nuclear translocation, increasing p53-upregulated modulator of apoptosis (PUMA). Activated PUMA liberates p53 from Bcl-XL, causing Bax activation, and leading to apoptosis eventually (Suzuki et al., 2009). Based on this, we made a hypothesis that p53 may be involved in the protection of fluoxetine against inflammation-induced neuronal apoptosis.

In the present study, we performed serial experiments in vivo and in vitro to prove this proposal. We found that treatment of fluoxetine reduced neuronal apoptosis and suppressed the upregulation of IL-1β and p53 expression in transient middle cerebral artery occlusion (tMCAO) model of mice. Consistently, in vitro studies showed that fluoxetine prevented IL-1β-induced apoptosis and p53 upregulation in primary cortical neurons. Furthermore, by use of Neuroblastoma N2a cell line, we demonstrated that p53 downregulation mediated the neuroprotective effects of fluoxetine and p38 phosphorylation contributed to the p53-dependent neuronal apoptosis. This finding has provided an insight into a novel anti-inflammatory mechanism of fluoxetine that may contribute to the use of fluoxetine therapy for ischemic stroke and other neuronal injured diseases.

Section snippets

Animals

C57BL/6J mice (3-month old) were obtained from Comparative Medicine Centre of Yangzhou University. All animal experimental procedures were carried out according to the guideline of the Institutional Animal Care and Use Committee of Nanjing Medical University.

Reagents

Fluoxetine and ketanserin was purchased from Sigma Aldrich. IL-1β was purchased from Peprotech. Anisomycin, SB203580 and cyanopindolol were purchased from Tocris. Cell culture reagents were purchased from Invitrogen. anti-p53 Ab (# 2524,

Fluoxetine alleviated mouse neuronal injury in tMCAO model

In the present study, we further addressed the effect of fluoxetine on neuronal apoptosis in a mouse model of tMCAO. As shown in Fig. 1A–C, infarct volume and neurological score were significantly lower in fluoxetine-treated mice compared with those in saline-treated mice after tMCAO. Moreover, fluoxetine markedly inhibited massive loss and apoptosis of neurons in the area penumbra of cerebral cortex (Fig. 1D–F). Compared to the sham group, the protein levels of Bax increased, while Bcl-2

Discussion

Fluoxetine has been recognized to function as a neuroprotectant in several animal models of neurological diseases, such as Parkinson's disease (PD) (Kohl et al., 2012) and AD (Qiao et al., 2016). Nevertheless, it remains unclear whether fluoxetine plays a crucial role in alleviating neuronal apoptosis in acute neuronal injury. In the present study, we show that fluoxetine exerts the neuroprotective effect against ischemic injury in mouse tMCAO model. Moreover, fluoxetine inhibits apoptosis

Conflicts of interest

The authors of this manuscript declare no conflict of interest.

Acknowledgments

The work reported herein was supported by the grants from the National Natural Science Foundation of China (No.81473196 and No.81573403), the Natural Science Foundation of Jiangsu Province (BK20130039) and the key project of Natural Science Foundation of the Higher Education Institutions of Jiangsu Province (No. 15KJA310002).

References (50)

  • D.P. Singh et al.

    Flavocoxid, dual inhibitor of cyclooxygenase-2 and 5-lipoxygenase, exhibits neuroprotection in rat model of ischaemic stroke

    Pharmacol. Biochem. Behav.

    (2014)
  • D.B. Wang et al.

    p53 and mitochondrial function in neurons

    Biochim. Biophys. Acta

    (2014)
  • E. Alcocer-Gomez et al.

    Stress-induced depressive behaviors require a functional NLRP3 inflammasome

    Mol. Neurobiol.

    (2015)
  • J. Borycz et al.

    5-HT 1B receptor-mediated serotoninergic modulation of methylphenidate-induced locomotor activation in rats

    Neuropsychopharmacology

    (2008)
  • H. Boutin et al.

    Role of IL-1alpha and IL-1beta in ischemic brain damage

    J. Neurosci.

    (2001)
  • S. Ding et al.

    Robust presynaptic serotonin 5-HT(1B) receptor inhibition of the striatonigral output and its sensitization by chronic fluoxetine treatment

    J. Neurophysiol.

    (2015)
  • T. Engel et al.

    CHOP regulates the p53-MDM2 axis and is required for neuronal survival after seizures

    Brain

    (2013)
  • S. Gupta et al.

    Antidepressant regulatory warnings, prescription patterns, suicidality and other aggressive behaviors in major depressive disorder and anxiety disorders

    Psychiatr. Q.

    (2015)
  • K. Heyne et al.

    A novel mechanism of crosstalk between the p53 and NFkappaB pathways: MDM2 binds and inhibits p65RelA

    Cell Cycle

    (2013)
  • B. Hoesel et al.

    The complexity of NF-kappaB signaling in inflammation and cancer

    Mol. Cancer

    (2013)
  • R. Jin et al.

    Role of inflammation and its mediators in acute ischemic stroke

    J. Cardiovasc. Transl. Res.

    (2013)
  • E.H. Kim et al.

    Targeting CD36-mediated inflammation reduces acute brain injury in transient, but not permanent, ischemic stroke

    CNS Neurosci. Ther.

    (2015)
  • E.S. Kim et al.

    Reactivation of p53 via MDM2 inhibition

    Cell Death Dis.

    (2015)
  • J.Y. Kim et al.

    Pretreatment with lycopene attenuates oxidative stress-induced apoptosis in human mesenchymal stem cells

    Biomol. Ther. (Seoul)

    (2015)
  • Z. Kohl et al.

    Fluoxetine rescues impaired hippocampal neurogenesis in a transgenic A53T synuclein mouse model

    Eur. J. Neurosci.

    (2012)
  • Cited by (40)

    • Minocycline promotes functional recovery in ischemic stroke by modulating microglia polarization through STAT1/STAT6 pathways

      2021, Biochemical Pharmacology
      Citation Excerpt :

      At days in vitro (DIV) 10, microglia cells were seeded on 6-well plates and incubated with 100 ng/mL LPS plus 20 ng/mL IFN-γ followed by 50 μM minocycline or saline for 24 h. Primary cortical neurons were prepared from embryonic 13–15 day mouse embryos as previously described [27] and maintained in neurobasal medium containing 1 × B27 supplement, 1 mM L-glutamine, and 100 U/ml penicillin/streptomycin. After seven days, cortical neurons were used for the neuron‐microglia co‐culture test.

    • Fluoxetine mitigating late-stage cognition and neurobehavior impairment induced by cerebral ischemia reperfusion injury through inhibiting ERS-mediated neurons apoptosis in the hippocampus

      2019, Behavioural Brain Research
      Citation Excerpt :

      Fluoxetine, a selective serotonin-reuptake inhibitor (SSRI), also was demonstrated its neuroprotective effect in the field of cerebral ischemic diseases [5]. The mechanisms of fluoxetine neuroprotection have been addressed by many scholars, including regulating Bcl-2 (B-cell leukemia-2) and Bax (Bcl-2-associated X protein) expression, inhabiting expression of interleukin-1β (IL-1β), COX-2, TNF-alpha and NF-kappaB activity, downregulation of P53, inhibiting inflammation and cell apoptosis [5–7]. These studies presented exact proof that fluoxetine owned the theoretical evidence in defending the cognition function after cerebral ischemia reperfusion injury.

    View all citing articles on Scopus
    View full text