Elsevier

Neuropharmacology

Volume 105, June 2016, Pages 175-185
Neuropharmacology

P2X7 receptor antagonism reduces the severity of spontaneous seizures in a chronic model of temporal lobe epilepsy

https://doi.org/10.1016/j.neuropharm.2016.01.018Get rights and content

Highlights

  • P2X7 receptor antagonism caused a shift in the severity of chronic seizures.

  • A decrease in severe seizures was visible after one week of treatment.

  • P2X7 receptor antagonism was unsuccessful in decreasing the number of seizures.

  • P2X7 receptor antagonism did not alter microglial activation or astrogliosis.

Abstract

Background

The available pharmacotherapy for patients with epilepsy primarily address the symptoms and are ineffective in about 40% of patients. Brain inflammation gained support as potential target for developing new therapies, especially the P2X7 receptor (P2X7R), involved in processing of IL-1β, might be an interesting candidate. This study was designed to investigate the effect of a novel P2X7R antagonist on the severity and on the number of chronic spontaneous recurrent seizures (SRS), which was unexplored until now.

Methods

After one-week of vehicle treatment (20% HP-β-cyclodextrin), JNJ-42253432 was administered subcutaneously for another week under continuous video-electroencephalography monitoring (n = 17) in Sprague Dawley rats 3 months after kainic acid-induced status epilepticus. The proportion of different seizure classes, as well as the number of SRS/day were calculated for the vehicle and treatment period. In addition, post-mortem microglial activation and astrogliosis were assessed.

Results

A significant decrease of the proportion of type 4–5 SRS (p < 0.05), while an increase of type 1–3 was demonstrated (p < 0.05) from the vehicle to the treatment period. There was no effect of the P2X7R antagonist on the number of SRS/day or the glial markers.

Conclusions

The P2X7R antagonist gave rise to a less severe profile of the chronic seizure burden without suppressing the SRS frequency. More studies are needed to unravel the underlying mechanisms of the beneficial effect on seizure severity and whether the administration of the compound during early epileptogenesis could induce long-term disease-modifying effects.

Introduction

Epilepsy is a chronic neurological disorder characterised by the occurrence of spontaneous recurrent seizures (SRS) due to aberrant neuronal discharges. Currently available anti-epileptic drugs (AEDs) are merely symptomatic treatments and cannot prevent or cure epilepsy. There are more than 20 AEDs in clinical use and newer generations of AEDs have been developed with a better side effect profile. However, the number of patients being medically intractable has not changed, remaining at 30–40% (Kwan and Brodie, 2000, Wiebe and Jette, 2012). Inadequate control of seizures and the exposure to inefficient therapy regimens concomitantly leads to significant comorbidities such as intellectual disability and psychiatric problems, poor quality of life and even sudden unexpected death in epilepsy (Pugliatti et al., 2007, Weaver and Pohlmann-Eden, 2013). Therefore, there remains a tremendous need for the development of medicinal therapies targeting alternative mechanisms important in disease ontogenesis.

Epileptogenesis – the process in which a normal neuronal network develops into a hyperexcitable one – is characterised by different processes such as excitotoxicity, neurodegeneration, gliosis and metabolic changes (Goffin et al., 2008). A plethora of evidence supports an important contribution of brain inflammation in the disease ontogenesis. It has been shown that different cytokines, including IL-1β and TNF-α, can cause hyperexcitability (Hu et al., 2000, Dube et al., 2005, Stellwagen et al., 2005, Ravizza et al., 2006, Galic et al., 2012). Only recently, Bogdanovic et al. (2014) showed a positive relationship between hippocampal brain inflammation and seizure frequency in an epilepsy rat model by means of [11C]-PK11195 translocator protein (TSPO) positron emission tomography imaging. The P2X7 receptor (P2X7R), an ATP-gated, non-selective cation channel, has gained a lot of interest as a target to modulate brain inflammation owing to its important role in the processing and release of IL-1β (Solle et al., 2001), which in turn is a potent pro-convulsive mediator (Ravizza et al., 2006). In the brain, this receptor is preferably, but not solely, expressed on the cell-surface of immune-competent cells such as microglia. An upregulation of the P2X7R has been shown during different stages of epileptogenesis in several epilepsy models in the limbic regions, including hippocampus, amygdala and piriform cortex, as well as in the neocortex (Vianna et al., 2002, Rappold et al., 2006, Avignone et al., 2008, Dona et al., 2009, Kim et al., 2009, Jimenez-Pacheco et al., 2013). An increased expression has also been shown in the neocortex in surgical specimen of patients with temporal lobe epilepsy (TLE), which is the most common focal form of acquired epilepsy (Jimenez-Pacheco et al., 2013). All together, these facts make the P2X7R an attractive and potential alternative drug target for epilepsy (Henshall and Engel, 2015).

Several preclinical studies have already demonstrated a seizure suppressive and neuroprotective effect of P2X7R antagonists on acutely provoked status epilepticus (SE) and the consequent pathology such as neuronal death (Engel et al., 2012, Jimenez-Pacheco et al., 2013, Mesuret et al., 2014). However, the effects of P2X7R antagonists on spontaneous seizures in the chronic epilepsy phase, remain unexplored. This study was designed to investigate the effects of the novel P2X7R antagonist JNJ-42253432 [2-methyl-N-([1-(4-phenylpiperazin-1-yl)cyclohexyl]methyl)-1,2,3,4-tetrahydroisoquinoline-5-carboxamide] (Lord et al., 2014) on the expression of SRS, in the established chronic epilepsy phase, after kainic acid-induced status epilepticus (KASE), a very common model for TLE. Most of the other available P2X7R antagonists have several limitations such as a lower affinity in rodents, lower specificity to the receptors and limited blood brain barrier permeability (Bartlett et al., 2014). On the contrary, the novel compound JNJ-42253432 has excellent pharmacokinetic and –dynamic properties, and has been demonstrated to be a potent P2X7R antagonist with a brain to plasma ratio equal to one (Lord et al., 2014).

With this study, we firstly aimed to investigate whether the P2X7R antagonist JNJ-42253432 is able to change the severity of SRS, using a modified scale of Racine (Racine, 1972), during the chronic phase by using a within group treatment paradigm. Secondly, we assessed the effect of JNJ-42253432 on the number of SRS. Finally, we determined whether JNJ-42253432 had an influence on the pathological hallmarks of the KASE model, more specifically microgliosis (OX-42 and TSPO) and astrogliosis (GFAP), during the chronic period.

Section snippets

Animals

Seven-week-old male Sprague Dawley rats (Harlan Laboratories B.V., The Netherlands) were individually housed in IVC cages on a 12:12 light/dark cycle with food (standard rat pellets) and water ad libitum under a temperature (22 ± 2 °C) and humidity (55 ± 10%) controlled environment. All animals were acclimatised for at least five days before the start of the experiments and all procedures were performed according to the European Ethics Committee (decree 86/609/CEE), the Animal Welfare Act (7

Treatment with the P2X7R antagonist JNJ-42253432 caused a shift from severe to milder SRS

The average concentration of JNJ-42253432 in plasma at the end of the treatment was 1364 ± 119 ng/ml. This concentration fell as expected within the therapeutic range indicating a sufficient P2X7R occupancy of 80% (Lord et al., 2014).

Classification of the SRS into different seizure types enabled to study the effect of the antagonist on the severity of the SRS during the chronic period. Interestingly, a significant effect was observed for the proportion of severe convulsive seizures (sum of S4

Discussion

For the first time, the effect of a P2X7R antagonist on SRS in rodents was evaluated. Promising results were obtained as we demonstrated an effect on the severity of the SRS, more particularly a shift from severe convulsive seizures (S4-5) to milder seizures (S1-3) after a treatment period of one week during the chronic epilepsy period. However, our results showed that JNJ-42253432 did not decrease the number of the SRS. The effects on the severity of seizures were not directly related to the

Conclusion

Overall, this study showed promising results regarding treatment of spontaneous seizures during the chronic period. A significant decrease of severe convulsive seizures was achieved after only one week of treatment, although the number of SRS was not suppressed by the P2X7R antagonist JNJ-42253432. Complementary studies are necessary to confirm and strengthen these findings using other P2X7R antagonists, and to exploit the potential effect of JNJ-42253432 given in combination with AEDs.

Funding

This work was supported by Janssen Pharmaceuticals Inc.; the Research Foundation Flanders (FWO) (grant numbers 1.5.110.14N, 1.5.144.12N, G.A009.13N); and the Queen Elisabeth Medical Foundation for Neurosciences [SD]. This work was also supported by the Bijzonder Onderzoeks Fonds of the University of Antwerp [HA and SD].

Statement of interest

None.

Acknowledgements

We would like to thank K Deseure and G Dézsi for their assistance regarding the mini-pump implantations. Our gratitude also goes to A De Vlam for her consistent support during the experiments.

References (57)

  • H. Amhaoul et al.

    Brain inflammation in a chronic epilepsy model: evolving pattern of the translocator protein during epileptogenesis

    Neurobiol. Dis.

    (2015)
  • J.N. Armstrong et al.

    Activation of presynaptic P2X7-like receptors depresses mossy fiber-CA3 synaptic transmission through p38 mitogen-activated protein kinase

    J. Neurosci. Off. J. Soc. Neurosci.

    (2002)
  • E. Avignone et al.

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

    J. Neurosci. Off. J. Soc. Neurosci.

    (2008)
  • R. Bartlett et al.

    The P2X7 receptor channel: recent developments and the use of P2X7 antagonists in models of disease

    Pharmacol. Rev.

    (2014)
  • J. Benicky et al.

    Angiotensin II AT1 receptor blockade ameliorates brain inflammation

    Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol.

    (2011)
  • L. Bernardino et al.

    Inflammatory events in hippocampal slice cultures prime neuronal susceptibility to excitotoxic injury: a crucial role of P2X7 receptor-mediated IL-1beta release

    J. Neurochem.

    (2008)
  • R.M. Bogdanovic et al.

    (R)-[C]PK11195 brain uptake as a biomarker of inflammation and antiepileptic drug resistance: evaluation in a rat epilepsy model

    Neuropharmacology

    (2014)
  • E.A. Cavalheiro et al.

    Spontaneous recurrent seizures in rats: amino acid and monoamine determination in the hippocampus

    Epilepsia

    (1994)
  • H.K. Choi et al.

    The roles of P2X7 receptor in regional-specific microglial responses in the rat brain following status epilepticus

    Neurological Sci. Off. J. Italian Neurological. Soc. Italian Soc. Clin. Neurophysiol.

    (2012)
  • S. Dedeurwaerdere et al.

    PET imaging of brain inflammation during early epileptogenesis in a rat model of temporal lobe epilepsy

    EJNMMI Res.

    (2012)
  • F. Dona et al.

    Alteration of purinergic P2X4 and P2X7 receptor expression in rats with temporal-lobe epilepsy induced by pilocarpine

    Epilepsy Res.

    (2009)
  • C. Dube et al.

    Interleukin-1beta contributes to the generation of experimental febrile seizures

    Ann. Neurol.

    (2005)
  • T. Engel et al.

    Seizure suppression and neuroprotection by targeting the purinergic P2X7 receptor during status epilepticus in mice

    FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol.

    (2012)
  • D. Ferrari et al.

    Extracellular ATP triggers IL-1 beta release by activating the purinergic P2Z receptor of human macrophages

    J. Immunol.

    (1997)
  • M.A. Galic et al.

    Cytokines and brain excitability

    Front. Neuroendocrinol.

    (2012)
  • K. Goffin et al.

    Neuronuclear assessment of patients with epilepsy

    Seminars Nucl. Med.

    (2008)
  • J.L. Hellier et al.

    Recurrent spontaneous motor seizures after repeated low-dose systemic treatment with kainate: assessment of a rat model of temporal lobe epilepsy

    Epilepsy Res.

    (1998)
  • D.C. Henshall et al.

    P2X purinoceptors as a link between hyperexcitability and neuroinflammation in status epilepticus

    Epilepsy Behav. E B

    (2015)
  • S. Hu et al.

    Cytokine effects on glutamate uptake by human astrocytes

    Neuroimmunomodulation

    (2000)
  • B. Ji et al.

    Imaging of peripheral benzodiazepine receptor expression as biomarkers of detrimental versus beneficial glial responses in mouse models of Alzheimer's and other CNS pathologies

    J. Neurosci. Off. J. Soc. Neurosci.

    (2008)
  • A. Jimenez-Pacheco et al.

    Increased neocortical expression of the P2X7 receptor after status epilepticus and anticonvulsant effect of P2X7 receptor antagonist A-438079

    Epilepsia

    (2013)
  • J.E. Kim et al.

    The P2X7 receptor-pannexin-1 complex decreases muscarinic acetylcholine receptor-mediated seizure susceptibility in mice

    J. Clin. Investig.

    (2011)
  • J.E. Kim et al.

    Blockade of P2X receptor prevents astroglial death in the dentate gyrus following pilocarpine-induced status epilepticus

    Neurol. Res.

    (2009)
  • S. Kumar-Singh et al.

    Mean age-of-onset of familial alzheimer disease caused by presenilin mutations correlates with both increased Abeta42 and decreased Abeta40

    Hum. Mutat.

    (2006)
  • P. Kwan et al.

    Early identification of refractory epilepsy

    N. Engl. J. Med.

    (2000)
  • V.F. Labrousse et al.

    Impaired interleukin-1beta and c-Fos expression in the hippocampus is associated with a spatial memory deficit in P2X(7) receptor-deficient mice

    PloS One

    (2009)
  • X. Langlois et al.

    Use of the beta-imager for rapid ex vivo autoradiography exemplified with central nervous system penetrating neurokinin 3 antagonists

    J. Pharmacol. Exp. Ther.

    (2001)
  • S. Locovei et al.

    Pannexin1 is part of the pore forming unit of the P2X(7) receptor death complex

    Febs Lett.

    (2007)
  • Cited by (54)

    • More than a drug target: Purinergic signalling as a source for diagnostic tools in epilepsy

      2023, Neuropharmacology
      Citation Excerpt :

      In line with this, a recent study by us has shown that increased P2X7R expression contributed to the unresponsiveness towards ASMs during status epilepticus, possibly via pro-inflammatory processes (Beamer et al., 2022). More consistent results have been obtained during epilepsy development and once epilepsy was established, where it has been shown that P2X7R antagonisms reduce seizure severity and frequency in several models, including the PTZ kindling and KA-induced status epilepticus model (Amhaoul et al., 2016; Fischer et al., 2016; Jamali-Raeufy et al., 2020; Jimenez-Pacheco et al., 2016; Soni et al., 2015). Notably, seizure-suppression was still evident one week following drug-withdrawal suggesting disease-modifying potential (Jimenez-Pacheco et al., 2016).

    • The role of inflammatory signaling in comorbid depression and epilepsy

      2021, The Neuroscience of Depression: Genetics, Cell Biology, Neurology, Behavior, and Diet
    View all citing articles on Scopus
    View full text