Elsevier

Journal of Neuroimmunology

Volume 313, 15 December 2017, Pages 82-88
Journal of Neuroimmunology

Brain expression of inflammatory mediators in Mesial Temporal Lobe Epilepsy patients

https://doi.org/10.1016/j.jneuroim.2017.10.014Get rights and content

Highlights

  • Activated microglia is present in hippocampus and anterior cortex of MTLE-HS.

  • IL-1β and TLR4 are upregulated in hippocampus and cortex of MTLE-HS patients.

  • Cortical and hippocampal upregulation of anti-inflammatory cytokine IL-10 was observed in MTLE-HS patients.

  • Inflammatory response associated with seizure propagation.

Abstract

Neuroinflammation may be central in epileptogenesis. In this study we analysed inflammatory reaction markers in brain tissue of Mesial Temporal Lobe Epilepsy with Hippocampal Sclerosis (MTLE-HS) patients. TLR4, IL-1β and IL-10 gene expression as well as the presence of activated HLA-DR + microglia was evaluated in 23 patients and 10 cadaveric controls. Inflammation characterized by the presence of HLA-DR+ microglia and TLR4, IL-1β overexpression was evident in hippocampus and anterior temporal cortex of MTLE-HS patients. Anti-inflammatory IL-10 was also overexpressed in MTLE-HS patients. Our results show that hippocampal neuroinflammation extends beyond lesional limits, as far as the anterior temporal cortex.

Introduction

Active inflammation has been documented not only in traditionally assumed inflammatory epilepsies but also in patients with pharmacoresistant epilepsy of diverse causes (Vezzani et al., 2011a, Vezzani and Ruegg, 2011). MTLE-HS is the more frequent partial epilepsy in adulthood. It is usually refractory with over 80% patients presenting a poor response to conventional anti-seizure drugs (ASDs). Refractory patients are often subjected to surgical resection of the hippocampus and amygdala in order to control seizures. This is one of the most successful epilepsy surgeries. Nevertheless, it is reported a seizure recurrence of 38% at 18 years of follow-up after surgery (Hemb et al., 2013). For MTLE-HS patients the efficient resolution of seizures is still an unmet clinical need. Understanding the epileptogenic process is fundamental for the development of new ASDs, but the mechanisms leading to MTLE-HS remain largely unknown.

Retrospective studies show that MTLE-HS patients often have a history of initial precipitating injury such as febrile seizures (FS), central nervous system infection and head trauma or hypoxia peripartum (Fisher et al., 1998). Among these factors FS is the most common with up to 80% of MTLE-HS patients reporting a history of complex FS (Fisher et al., 1998, Hesdorffer et al., 2016). It has been hypothesized that after the initial insult there is an abnormal cascade of damage repair, with the maintenance of chronic inflammation, leading to atrophy and hippocampal sclerosis (Fisher et al., 1998, Gallentine et al., 2017, Lewis et al., 2014). In fact, imaging studies have shown that prolonged and lateralized FS can produce acute hippocampal injury with oedema that resolves within 5 days (Scott et al., 2003). The follow-up of these children showed changes in hippocampal symmetry consistent with injury and neuronal loss (Scott et al., 2003, Shinnar et al., 2012). So, it is believed that FS initiate the abnormal network reorganization that will lead to the development of an epileptogenic structure. Alternatively, the asymmetry could represent a return (post-acute oedema) to a pre-existing hippocampal abnormality (Fernandez et al., 1998, Lewis et al., 2014).

In normal physiological conditions pro-inflammatory cytokines, such as TNF-α, IL-1, and IL-6, and their receptors are constitutively expressed at low levels in different brain regions by astrocytes, microglia cells, neurons and endothelial cells (Vezzani and Viviani, 2015). These proteins are claimed to be important in neuronal development, controlling neurite outgrowth, neurogenesis and cell survival (Vezzani and Viviani, 2015). These pro-inflammatory cytokines can also modulate voltage-gated and receptor-coupled ionic channels (Viviani et al., 2007) as well as neurotransmitter's receptors (Balosso et al., 2009, Roseti et al., 2015, Stellwagen et al., 2005) controlling synaptic pruning, transmission and plasticity in the adult brain (Marin and Kipnis, 2013). In fact, IL-1β and IL-6 seem to have a general inhibitory action on CNS voltage-gated channels inhibiting Ca2 +, Na+ and K+ currents (for review see (Vezzani and Viviani, 2015)). In ligand-gated ion channels, IL-1β is observed to be mainly excitatory increasing NMDA-mediated Ca2 + influx (Viviani et al., 2003). Additionally, IL-1β can promote excitability through the downmodulation of the astrocytic glutamate transporter (GLT-1) (Hu et al., 2000) while promoting the release of excitatory neurotransmitters such as Glutamate or ATP (Devinsky et al., 2013). In fact, neuroinflammation appears to be an important component in epileptogenesis, reflecting complex cross-talks between microglia, astrocytes and neurons (Aronica et al., 2012, Devinsky et al., 2013).

Cytokines can also influence the strength of synaptic transmission as they can modulate NMDA, AMPA and GABA(A) receptor expression and their sub-unit composition. In accordance, several studies demonstrated that a fine-tuned cytokine production is necessary for learning and cognition and that a dysregulation may lead to excitotoxicity (McAfoose and Baune, 2009, Yirmiya and Goshen, 2011).

Seizure-induced cell death leads to the release of endogenous molecules (DAMPs) such as HMGB1 that are recognized by TLRs expressed in glial cells and neurons (Bianchi, 2007, Mazarati et al., 2011). The engagement of TLRs leads to the activation of innate immunity with the production of pro-inflammatory mediators. In fact, it has been demonstrated in animal models that several inflammatory cytokines (such as IL-1β, TNFα and Il-6) as well as TLRs (for example TLR4 and TLR9) are rapidly induced by seizures in activated astrocytes and microglia (De Simoni et al., 2000, Maroso et al., 2010, Ravizza et al., 2005, Tan et al., 2015, Vezzani et al., 1999). Also, microarray studies in rodent models of TLE showed that inflammation is one of the most upregulated biological processes during epileptogenesis (Gorter et al., 2006). Inflammatory molecules contribute to decrease seizure threshold by direct effects on neuronal excitability (Vezzani and Baram, 2007) and may also activate transcription of genes involved in neurogenesis, cell death, and synaptic plasticity (Vezzani et al., 2008b, Widera et al., 2006). In this way, inflammatory molecules can also participate in glial scar formation contributing to seizure-related hippocampal changes, such as neuronal loss, reactive gliosis and mossy fiber sprouting.

These experimental results are corroborated by studies in human brain tissue from drug-resistant epilepsies. In these patients activation of hippocampal microglia with concomitant over-expression of HLA-DR (Beach et al., 1995) and upregulation of diverse inflammatory mediators, such as complement proteins or pro-inflammatory cytokines, has been evidenced (Aronica et al., 2007, Choi et al., 2009, Crespel et al., 2002, Jamali et al., 2006, Kan et al., 2012, Omran et al., 2012, Ravizza et al., 2008). Upregulation of several inflammatory players has also been observed in cerebrospinal fluid and serum of epileptic patients (de Vries et al., 2016). Moreover, it is known that some conventional ASDs have an anti-inflammatory action (Matoth et al., 2000) and that administration of anti-inflammatory drugs can also have anti-convulsant effects with reduction of seizures (Hancock et al., 2013, Radu et al., 2017, Vezzani, 2015). On the other hand, anti-inflammatory cytokines such as IL-10 may protect against seizures (Ishizaki et al., 2009).

Studies using surgically-removed anterior cortical region are scarce. This region is thought to contribute to seizure propagation in MTLE-HS patients (Bartolomei et al., 2008). Thus, the aim of this study was to characterize the expression of inflammatory mediators namely, IL-1β, TLR4, and IL-10, both in the hippocampus and in the anterior temporal cortex.

Section snippets

Population

Resected fresh human tissue obtained from 23 MTLE-HS patients (13F, 10 M, see Table 1) who underwent epilepsy surgical treatment (selective amygdalohippocampectomy or anterior temporal lobectomy) at Neurosurgery Department of Hospital Santo António – Centro Hospitalar e Universitário do Porto (HSA – CHUP) has been analysed. The decision for surgery was taken by HSA multidisciplinary epilepsy team incorporating neurologists, neurosurgeons, neuroradiologists, neurophysiologists and

Results

In order to assess inflammatory response in MTLE-HS patients, we analysed the presence of activated HLA-DR + microglia and quantified the expression of inflammatory markers – TLR4, IL-1β and IL-10 - in the hippocampus and anterior temporal cortex of MTLE-HS patients and cadaveric controls.

The presence of HLA-DR + cells was prominent in the hippocampus (Fig. 1a) and also in the anterior cortical region (Fig. 1c) of MTLE-HS patients. The presence of HLA-DR + cells was not detected in the hippocampus

Discussion

The main goal of this study was to evaluate the inflammatory response in brain tissue of MTLE-HS patients. We observed the presence of activated HLA-DR+ microglial cells in the hippocampus and anterior temporal cortex of MTLE-HS patients. Similar results have already been observed both in animal models (Avignone et al., 2008) and in the hippocampus of MTLE-HS patients (Beach et al., 1995).

Concomitant with microglia activation we have observed the overexpression of the pro-inflammatory cytokine,

Conclusion

The acute or long-term exposure to inflammatory cytokines can determine the modifications in neurotransmission with opposite effects. Overall, the net effect will depend on cytokine balance and target cell. Our study gives further support to previous data showing a persistent activation of the innate immune response in epilepsy. The upregulation of the IL-1β – TLR4 axis in epilepsy, was particularly evident in the anterior cortical area, thus suggesting that it might contribute to seizure

Ethical publication statement

We confirm that we have read the Journal position on issues involved in ethical publication and affirm that this report is consistent with those guidelines.

Disclosure conflict of interests

None of the authors has any conflict of interest to disclose.

Acknowledgements

This research was partial supported by a FCT grant PIC/IC/83297/2007. The funders had no role in study design, data collection and analysis or preparation of the manuscript. The authors acknowledge the collaboration of Aurora Barros-Barbosa, PhD, J.Miguel Cordeiro, PhD, Graça Lobo, PhD, nurses from the epilepsy outpatient clinic and nurses from the surgical room, in sample collection. The authors also acknowledge Ms. Maria Rebelo and Ms. Sandra Brás for technical assistance, and Prof. Pedro

References (89)

  • V. Iori et al.

    Blockade of the IL-1R1/TLR4 pathway mediates disease-modification therapeutic effects in a model of acquired epilepsy

    Neurobiol. Dis.

    (2017)
  • J.L. Jankowsky et al.

    The role of cytokines and growth factors in seizures and their sequelae

    Prog. Neurobiol.

    (2001)
  • M.A. Kauffman et al.

    Association study between interleukin 1 beta gene and epileptic disorders: a HuGe review and meta-analysis

    Genet. Med.

    (2008)
  • R. Klee et al.

    Network pharmacology for antiepileptogenesis: tolerability of multitargeted drug combinations in nonepileptic vs. post-status epilepticus mice

    Epilepsy Res.

    (2015)
  • M. Maroso et al.

    Interleukin-1beta biosynthesis inhibition reduces acute seizures and drug resistant chronic epileptic activity in mice

    Neurotherapeutics

    (2011)
  • I. Matoth et al.

    Inhibitory effect of carbamazepine on inflammatory mediators produced by stimulated glial cells

    Neurosci. Res.

    (2000)
  • A. Mazarati et al.

    High-mobility group box-1 impairs memory in mice through both toll-like receptor 4 and receptor for advanced glycation end products

    Exp. Neurol.

    (2011)
  • J. McAfoose et al.

    Evidence for a cytokine model of cognitive function

    Neurosci. Biobehav. Rev.

    (2009)
  • P.J. Murray

    Understanding and exploiting the endogenous interleukin-10/STAT3-mediated anti-inflammatory response

    Curr. Opin. Pharmacol.

    (2006)
  • F.M. Noe et al.

    Pharmacological blockade of IL-1beta/IL-1 receptor type 1 axis during epileptogenesis provides neuroprotection in two rat models of temporal lobe epilepsy

    Neurobiol. Dis.

    (2013)
  • K. Pernhorst et al.

    TLR4, ATF-3 and IL8 inflammation mediator expression correlates with seizure frequency in human epileptic brain tissue

    Seizure

    (2013)
  • B.M. Radu et al.

    Nonsteroidal anti-inflammatory drugs in clinical and experimental epilepsy

    Epilepsy Res.

    (2017)
  • T. Ravizza et al.

    Innate and adaptive immunity during epileptogenesis and spontaneous seizures: evidence from experimental models and human temporal lobe epilepsy

    Neurobiol. Dis.

    (2008)
  • C. Roseti et al.

    GABAA currents are decreased by IL-1beta in epileptogenic tissue of patients with temporal lobe epilepsy: implications for ictogenesis

    Neurobiol. Dis.

    (2015)
  • O. Tomkins et al.

    Blood-brain barrier disruption results in delayed functional and structural alterations in the rat neocortex

    Neurobiol. Dis.

    (2007)
  • A. Vezzani et al.

    Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability

    Neuropharmacology

    (2015)
  • A. Vezzani et al.

    The role of cytokines in the pathophysiology of epilepsy

    Brain Behav. Immun.

    (2008)
  • A. Vezzani et al.

    IL-1 receptor/Toll-like receptor signaling in infection, inflammation, stress and neurodegeneration couples hyperexcitability and seizures

    Brain Behav. Immun.

    (2011)
  • B. Viviani et al.

    Cytokines and neuronal ion channels in health and disease

    Int. Rev. Neurobiol.

    (2007)
  • E.E. de Vries et al.

    Inflammatory mediators in human epilepsy: a systematic review and meta-analysis

    Neurosci. Biobehav. Rev.

    (2016)
  • I. Weissberg et al.

    Albumin induces excitatory synaptogenesis through astrocytic TGF-beta/ALK5 signaling in a model of acquired epilepsy following blood-brain barrier dysfunction

    Neurobiol. Dis.

    (2015)
  • R. Yirmiya et al.

    Immune modulation of learning, memory, neural plasticity and neurogenesis

    Brain Behav. Immun.

    (2011)
  • A.T. Argaw et al.

    IL-1beta regulates blood-brain barrier permeability via reactivation of the hypoxia-angiogenesis program

    J. Immunol.

    (2006)
  • E. Aronica et al.

    Astrocyte immune responses in epilepsy

    Glia

    (2012)
  • S. Auvin et al.

    Inflammation induced by LPS enhances epileptogenesis in immature rat and may be partially reversed by IL1RA

    Epilepsia

    (2010)
  • E. Avignone et al.

    Status epilepticus induces a particular microglial activation state characterized by enhanced purinergic signaling

    J. Neurosci.

    (2008)
  • F. Bartolomei et al.

    Epileptogenicity of brain structures in human temporal lobe epilepsy: a quantified study from intracerebral EEG

    Brain

    (2008)
  • M.E. DAMPs Bianchi

    PAMPs and alarmins: all we need to know about danger

    J. Leukoc. Biol.

    (2007)
  • O. Braganza et al.

    Albumin is taken up by hippocampal NG2 cells and astrocytes and decreases gap junction coupling

    Epilepsia

    (2012)
  • T. Butler et al.

    Transient and chronic seizure-induced inflammation in human focal epilepsy

    Epilepsia

    (2016)
  • L.P. Cacheaux et al.

    Transcriptome profiling reveals TGF-beta signaling involvement in epileptogenesis

    J. Neurosci.

    (2009)
  • S. Chakravarty et al.

    Toll-like receptor 4 on nonhematopoietic cells sustains CNS inflammation during endotoxemia, independent of systemic cytokines

    J. Neurosci.

    (2005)
  • J. Choi et al.

    Cellular injury and neuroinflammation in children with chronic intractable epilepsy

    J. Neuroinflammation

    (2009)
  • Y. David et al.

    Astrocytic dysfunction in epileptogenesis: consequence of altered potassium and glutamate homeostasis?

    J. Neurosci.

    (2009)
  • Cited by (47)

    • Chronic vagus nerve stimulation (VNS) altered IL-6, IL-1β, CXCL-1 and IL-13 levels in the hippocampus of rats with LiCl-pilocarpine-induced epilepsy

      2022, Brain Research
      Citation Excerpt :

      The proinflammatory cytokines IL-Iβ, IL-2, and IL-6, typically in low quantities within the brain, are observed at higher concentrations after seizures (Alyu and Dikmen, 2017; Gabriel and Arisi, 2015; Ho et al., 2015). Work in human epileptic tissue has shown that proinflammatory molecules, such as IL-Iβ and CXCL8, are upregulated in removed foci (Leal et al., 2017). Moreover, transgenic mice overexpressing IL-Iβ in astrocytes exhibited diminished seizure susceptibility (Auvin et al., 2010), while IL-1R1 knockout mice showed less potential for seizures (Feng et al., 2016).

    • Epilepsy progression is associated with cumulative DNA methylation changes in inflammatory genes

      2022, Progress in Neurobiology
      Citation Excerpt :

      Microglia are the inflammatory effector cells of the CNS (Schetters et al., 2017). In fact, enhanced MHC-II surface expression is a common marker of activated microglia, a hallmark of neurological pathology, including in MTLE-HS (Leal et al., 2017). In the neocortex, we described a similar but milder representation of inflammatory mechanisms showing progressive DNA methylation alterations, which is consistent with the aforementioned milder damaging profile in this tissue.

    View all citing articles on Scopus
    View full text