Abstract

Research on the antiepileptic effects of (endo-)cannabinoids has remarkably progressed in the years following the discovery of fundamental role of the endocannabinoid (eCB) system in controlling neural excitability. Moreover, an increasing number of well-documented cases of epilepsy patients exhibiting multi-drug resistance report beneficial effects of cannabis use. Pre-clinical and clinical research has increasingly focused on the antiepileptic effectiveness of exogenous administration of cannabinoids and/or pharmacologically induced increase of eCBs such as anandamide (also known as arachidonoylethanolamide [AEA]). Concomitant research has uncovered the contribution of neuroinflammatory processes and peripheral immunity to the onset and progression of epilepsy. Accordingly, modulation of inflammatory pathways such as cyclooxygenase-2 (COX-2) was pursued as alternative therapeutic strategy for epilepsy. Palmitoylethanolamide (PEA) is an endogenous fatty acid amide related to the centrally and peripherally present eCB AEA, and is a naturally occurring nutrient that has long been recognized for its analgesic and anti-inflammatory properties. Neuroprotective and anti-hyperalgesic properties of PEA were evidenced in neurodegenerative diseases, and antiepileptic effects in pentylenetetrazol (PTZ), maximal electroshock (MES) and amygdaloid kindling models of epileptic seizures. Moreover, numerous clinical trials in chronic pain revealed that PEA treatment is devoid of addiction potential, dose limiting side effects and psychoactive effects, rendering PEA an appealing candidate as antiepileptic compound or adjuvant. In the present study, we aimed at assessing antiepileptic properties of PEA in a mouse model of acute epileptic seizures induced by systemic administration of kainic acid (KA). KA-induced epilepsy in rodents is assumed to resemble to different extents human temporal lobe epilepsy (TLE) depending on the route of KA administration; intracerebral (i.c.) injection was recently shown to most closely mimic human TLE, while systemic KA administration causes more widespread pathological damage, both in brain and periphery. To explore the potential of PEA to exert therapeutic effects both in brain and periphery, acute and subchronic administration of PEA by intraperitoneal (i.p.) injection was assessed on mice with systemically administered KA. Specifically, we investigated: (i) neuroprotective and anticonvulsant properties of acute and subchronic PEA treatment in KA-induced seizure models, and (ii) temporal dynamics of eCB and eicosanoid (eiC) levels in hippocampus and plasma over 180 min post seizure induction in PEA-treated and non-treated KA-injected mice vs. vehicle injected mice. Finally, we compared the systemic PEA treatment with, and in combination with, pharmacological blockade of fatty acid amide hydrolase (FAAH) in brain and periphery, in terms of anticonvulsant properties and modulation of eCBs and eiCs. Here, we demonstrate that subchronic administration of PEA significantly alleviates seizure intensity, promotes neuroprotection and induces modulation of the plasma and hippocampal eCB and eiC levels in systemic KA-injected mice.

Highlights

  • Epilepsy is one of the most common neurological disorders worldwide with severe impact on the life quality of patients and often leading to long-term cognitive impairments (Xu et al, 2013)

  • In this study we focused on the analysis of AEA, 2-arachidonoyl glycerol (2-AG), PEA, arachidonic acid (AA), prostaglandin E2 (PGE2) and prostaglandin D2 (PGD2)

  • We focused our analysis on the PGE2 and PGD2 due to their prominent involvement in inflammatory processes in general, and arising from kainic acid (KA)-induced excitotoxicity, in particular

Read more

Summary

Introduction

Epilepsy is one of the most common neurological disorders worldwide with severe impact on the life quality of patients and often leading to long-term cognitive impairments (Xu et al, 2013). Multiple causes of epileptogenesis such as head trauma, genetic and metabolic factors, and infections, combined with the diversity of epileptic manifestation and types, and yet unclarified mechanism of epileptogenesis challenge the development of effective antiepileptic therapies (Narain, 2014; Amini et al, 2015). In this context, identification of molecular causes of and/or correlates with epilepsy in various animal models is essential to discover new drug targets and markers for follow-up monitoring. A large variability in patient response, psychoactive effects, possible long-term side effects in young patients, as well as the unpredictable risk of cannabinoid-receptor 1 (CB1) desensitization subsequently reducing antiepileptic effects or even aggravating seizures, remain main concerns for its clinical use (Lutz, 2004; Szaflarski and Bebin, 2014; Blair et al, 2015; Katona, 2015; Mechoulam, 2017)

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.