Skip to main content

EDITORIAL article

Front. Cell Dev. Biol., 07 November 2022
Sec. Signaling
This article is part of the Research Topic Neuroinflammation in Acquired Epilepsy View all 10 articles

Editorial: Neuroinflammation in acquired epilepsy

  • 1Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States
  • 2Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
  • 3Department of Pharmacology, Medical School of Southeast University, Nanjing, China

Editorial on the Research Topic
Neuroinflammation in acquired epilepsy

In spite of astonishing advances in epilepsy research and treatment over the past few decades, epilepsy remains one of the most common and devastating brain diseases and still affects approximately 65 million people globally (Devinsky et al., 2018). In addition to their wide-ranging side effects, antiseizure drugs (ASDs) are not effective in controlling seizures in more than 30% of patients who have pharmacoresistant epilepsy (Janmohamed et al., 2020). It is rather unfortunate that current medications provide merely symptomatic relief and have not been demonstrated to prevent epilepsy in people at risk or modify the disease progression (Galanopoulou et al., 2021). Therefore, there remains an urgent need for alternative antiepileptic treatments, despite the rapid expansion of modern ASDs that emerged during the first 2 decades of this century (Varvel et al., 2015; Löscher and Klein, 2020; Yu et al., 2022).

Mounting lines of evidence support an essential role for proinflammatory mediators in the brain in acquired epileptogenesis, a pathogenic process that is proposed to transform a normal brain to one generating seizures following various brain insults, such as de novo status epilepticus (SE), brain infections, traumatic brain injuries, brain tumors, and strokes (Yu et al., 2019; Korgaonkar et al., 2020; Terrone et al., 2020). It has been widely proposed that modulating key proinflammatory mediators might disrupt the epileptogenic processes and lead to modification and/or even prevention of epilepsy. In the current Research Topic, we bring together a diverse collection of primary research and review articles that highlight the roles of brain inflammation in epilepsy of various etiologies and pathogeneses.

Both inflammation and oxidative stress are well known for their pathophysiological roles in the epileptic brain. However, they are often studied as separate entities despite the evidence that the redox-based signaling cascades and inflammatory reactions have extensive crosstalk (Fabisiak and Patel). Recent studies have uncovered a variety of mechanisms whereby oxidative stress and neuroinflammation greatly influence each other in the context of epilepsy. For instance, neuroinflammation can be regulated by transcription factors such as NF-κB and nrf2 that are activated by reactive oxygen species (ROS). Neuroinflammation in turn can induce the expression and activity of NADPH oxidase (NOX), fostering a highly oxidative environment. Moreover, the oxidative and proinflammatory mediators can moulate distinct intracellular pathways expressed in different cell-types, exemplified by NOX-2 dependent increase in ROS in neurons and astrocytes triggered by SE, and myeloid differentiation primary response 88 (MyD88) dependent glial activation through Toll-like receptors (TLRs) (Almeida et al.). The reviews presented in this collection highlight how signaling crosstalk between neuroinflammation and oxidative stress and their cell type specific roles may be leveraged for novel therapeutic strategies for epilepsy.

Neuroinflammatory processes triggered by acute brain insults such as SE are highly regulated and show time- and age-dependency. Using a rat model of kainate-induced SE, Erisken et al. demonstrate prolonged induction of many key inflammatory genes, particularly those associated with stress-activated protein kinases, p38 and JNK signaling pathways, uniquely in adult brains. In contrast, many of the same genes show relatively transient expression in developing brains under similar experimental conditions, suggesting that the immature brains might be more resistant to SE-induced cell death and neuropathology. In line with findings in adult animals, hippocampal tissues from mesial temporal lobe epilepsy patients showed upregulation of inflammation-related genes. These results highlight the association between uncontrolled neuroinflammation and epileptogenesis and suggest that epileptic seizures might result from prolonged activation of neuroimmune processes beyond the homeostatic threshold.

Brain infection is a leading cause of epilepsy, but the underlying molecular mechanisms are poorly understood. Patel et al. use Theiler’s murine encephalomyelitis virus (TMEV) infection to generate acute brain inflammation and the subsequent spontaneous seizures in mice. They show TMEV infection-induced seizures likely due to impaired GABAergic inhibition, secondary to alterations in neuronal intracellular chloride regulation. Their results further suggest that the brain-derived neurotrophic factor (BDNF) might contribute to the development of brain infection-triggered seizures by reducing the expression of K+/Cl cotransporter 2 (KCC2). This has the potential to enhance accumulation of intracellular chloride and increases excitability by rendering GABA depolarizing instead of hyperpolarizing as observed in chemoconvulsant models of SE (Pathak et al., 2007; Yu et al., 2013). Notably, the upregulation of brain BDNF observed in TMEV-infected mice has also been found in chemoconvulsant models of SE (Zhu et al., 2012; Thomas et al., 20162016; Yu and Jiang 2020), and is believed to contribute to acquired epileptogenesis by acting on its high-affinity receptor, the tropomyosin related kinase B (TrkB) (Lin et al., 2020).

Exposure to diisopropylfluorophosphate (DFP), a structural analog of type G chemical warfare agents (e.g., sarin and soman), is well known to induce SE, gliosis, neuronal death, and eventually the development of spontaneous recurrent seizures in rodents. Gage et al. show that both male and female rats which experience DFP-induced SE develop unique regions of glial scarring in the piriform cortex and amygdala, but not in the hippocampus. DFP-induced cortical glial scars are characterized by a massive clustering of reactive microglia, with increase in Iba1-and CD68-positive cells, surrounded by hypertrophic astrocytes and a decrease in NeuN-positive neurons in the scar core. Although female rats have been shown to require a higher dose of DFP to induce SE when housed in a room with only females, Rao et al. demonstrate that when both sexes are housed in the same room and administered the same DFP solution, SE severity was not different between sexes. These results reinforce the importance of sex as a key biological variable in experimental design and suggest that housing animals of both sexes together and using the same batch of test reagents will reduce experimental variability.

The benefits of low-intensity physical exercise to the CNS have been shown in animal models and patients with neurological diseases, such as Alzheimer’s disease, Parkinson’s disease, stroke, epilepsy, multiple sclerosis, anxiety and depression (Allendorfer and Bamman, 2018). Jia et al. demonstrate that the conventional ASD, valproate, combined with low-intensity exercise can reduce seizures and associated co-morbidities in kainate-treated mice. The reduction in seizure burden appears to be correlated with the suppression of inflammatory cytokines (IL-1β, IL-6, and TNF-α) and the immune receptor TLR4 in the hippocampus. Given that TLR4 is involved in epileptogenesis of diverse etiologies (Maroso et al., 2010; Korgaonkar et al., 2020), these findings suggest that the non-pharmacological intervention like low-intensity exercise might reduce neuroinflammation and provide an adjunctive strategy to enhance efficacy of conventional ASDs to treat epilepsy.

Developing preventive treatment for epilepsy is challenging because it is currently impossible to identify individuals that will develop epilepsy after initial precipitating brain insults. Theoretically, biomarkers that identify “at risk” individuals would facilitate the development of potential antiepileptogenic treatment (Simonato et al., 2021). By reviewing data from 60 human patients with focal epilepsy of autoimmune etiology, Sakamoto et al. propose a diagnostic algorithm that might help to predict the underlying autoimmune etiology of epilepsy before antibody testing results become available. Over 30% of epilepsy patients suffer from pharmacoresistant seizures associated with cognitive and psychiatric co-morbidities. Analyzing a microarray dataset from the Gene Expression Omnibus database, Min et al. identify 25 genes differentially expressed in the peripheral blood of patients with valproate resistance in epilepsy and significantly enriched in T-cell receptor recognition. While the potential confound posed by the differential seizure burden between valproate sensitive and resistant groups needs to be considered, these findings suggest that the peripheral blood T-cells and the differentially expressed genes could serve as biomarkers for refractory epilepsy. Identification of reliable biomarkers for diverse types of epilepsy and pharmacoresistance could facilitate both early diagnosis and development of new therapies, needed to achieve the ultimate goals of “no seizures, no side effects, and no co-morbidities” in epilepsy treatment.

The series of articles presented here address diverse ways in which neuroinflammation could shape acquired epilepsy and offers insights into how these processes may be leveraged to inform mechanisms of epileptogenesis, identify biomarkers, and to develop novel strategies for disease modification and treatment in epilepsy.

Author contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Funding

JJ is supported by the NIH/NINDS grants (R01NS100947, R21NS109687, and R61NS124923). VS is supported by the NIH/NINDS grants (R01NS069861 and R01NS097750).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Allendorfer, J. B., and Bamman, M. M. (2018). Getting the brain into shape: Exercise in neurological disorders. Clin. Ther. 40, 6–7. doi:10.1016/j.clinthera.2017.12.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Devinsky, O., Vezzani, A., O'Brien, T. J., Jette, N., Scheffer, I. E., de Curtis, M., et al. (2018). Epilepsy. Nat. Rev. Dis. Prim. 4, 18024. doi:10.1038/nrdp.2018.24

CrossRef Full Text

Galanopoulou, A. S., Loscher, W., Lubbers, L., O'Brien, T. J., Staley, K., Vezzani, A., et al. (2021). Antiepileptogenesis and disease modification: Progress, challenges, and the path forward-Report of the Preclinical Working Group of the 2018 NINDS-sponsored antiepileptogenesis and disease modification workshop. Epilepsia Open 6, 276–296. doi:10.1002/epi4.12490

PubMed Abstract | CrossRef Full Text | Google Scholar

Janmohamed, M., Brodie, M. J., and Kwan, P. (2020). Pharmacoresistance - epidemiology, mechanisms, and impact on epilepsy treatment. Neuropharmacology 168, 107790. doi:10.1016/j.neuropharm.2019.107790

PubMed Abstract | CrossRef Full Text | Google Scholar

Korgaonkar, A. A., Li, Y., Sekhar, D., Subramanian, D., Guevarra, J., Swietek, B., et al. (2020). Toll-like receptor 4 signaling in neurons enhances calcium-permeable alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor currents and drives post-traumatic epileptogenesis. Ann. Neurol. 87, 497–515. doi:10.1002/ana.25698

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, T. W., Harward, S. C., Huang, Y. Z., and McNamara, J. O. (2020). Targeting BDNF/TrkB pathways for preventing or suppressing epilepsy. Neuropharmacology 167, 107734. doi:10.1016/j.neuropharm.2019.107734

PubMed Abstract | CrossRef Full Text | Google Scholar

Löscher, W., and Klein, P. (2020). The feast and famine: Epilepsy treatment and treatment gaps in early 21st century. Neuropharmacology 170, 108055. doi:10.1016/j.neuropharm.2020.108055

PubMed Abstract | CrossRef Full Text | Google Scholar

Maroso, M., Balosso, S., Ravizza, T., Liu, J., Aronica, E., Iyer, A. M., et al. (2010). Toll-like receptor 4 and high-mobility group box-1 are involved in ictogenesis and can be targeted to reduce seizures. Nat. Med. 16, 413–419. doi:10.1038/nm.2127

PubMed Abstract | CrossRef Full Text | Google Scholar

Pathak, H. R., Weissinger, F., Terunuma, M., Carlson, G. C., Hsu, F. C., Moss, S. J., et al. (2007). Disrupted dentate granule cell chloride regulation enhances synaptic excitability during development of temporal lobe epilepsy. J. Neurosci. 27, 14012–14022. doi:10.1523/JNEUROSCI.4390-07.2007

PubMed Abstract | CrossRef Full Text | Google Scholar

Simonato, M., Agoston, D. V., Brooks-Kayal, A., Dulla, C., Fureman, B., Henshall, D. C., et al. (2021). Identification of clinically relevant biomarkers of epileptogenesis - a strategic roadmap. Nat. Rev. Neurol. 17, 231–242. doi:10.1038/s41582-021-00461-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Terrone, G., Balosso, S., Pauletti, A., Ravizza, T., and Vezzani, A. (2020). Inflammation and reactive oxygen species as disease modifiers in epilepsy. Neuropharmacology 167, 107742. doi:10.1016/j.neuropharm.2019.107742

PubMed Abstract | CrossRef Full Text | Google Scholar

Thomas, A. X., Cruz Del Angel, Y., Gonzalez, M. I., Carrel, A. J., Carlsen, J., Lam, P. M., et al. (2016). Rapid increases in proBDNF after pilocarpine-induced status epilepticus in mice are associated with reduced proBDNF cleavage machinery. eNeuro 3, ENEURO.0020. doi:10.1523/ENEURO.0020-15.2016

PubMed Abstract | CrossRef Full Text | Google Scholar

Varvel, N. H., Jiang, J., and Dingledine, R. (2015). Candidate drug targets for prevention or modification of epilepsy. Annu. Rev. Pharmacol. Toxicol. 55, 229–247. doi:10.1146/annurev-pharmtox-010814-124607

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, J., Proddutur, A., Elgammal, F. S., Ito, T., and Santhakumar, V. (2013). Status epilepticus enhances tonic GABA currents and depolarizes GABA reversal potential in dentate fast-spiking basket cells. J. Neurophysiol. 109, 1746–1763. doi:10.1152/jn.00891.2012

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, Y., and Jiang, J. (2020). COX-2/PGE2 axis regulates hippocampal BDNF/TrkB signaling via EP2 receptor after prolonged seizures. Epilepsia Open 5, 418–431. doi:10.1002/epi4.12409

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, Y., Li, W., and Jiang, J. (2022). TRPC channels as emerging targets for seizure disorders. Trends Pharmacol. Sci. 43, 787–798. doi:10.1016/j.tips.2022.06.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, Y., Nguyen, D. T., and Jiang, J. (2019). G protein-coupled receptors in acquired epilepsy: Druggability and translatability. Prog. Neurobiol. 183, 101682. doi:10.1016/j.pneurobio.2019.101682

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, X., Han, X., Blendy, J. A., and Porter, B. E. (2012). Decreased CREB levels suppress epilepsy. Neurobiol. Dis. 45, 253–263. doi:10.1016/j.nbd.2011.08.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: epileptogenesis, BDNF, biomarker, brain infection, NADPH oxidase, oxidative stress, reactive oxygen species

Citation: Jiang J, Santhakumar V and Zhu X (2022) Editorial: Neuroinflammation in acquired epilepsy. Front. Cell Dev. Biol. 10:1074537. doi: 10.3389/fcell.2022.1074537

Received: 19 October 2022; Accepted: 25 October 2022;
Published: 07 November 2022.

Edited and reviewed by:

Ana Cuenda, Spanish National Research Council (CSIC), Spain

Copyright © 2022 Jiang, Santhakumar and Zhu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jianxiong Jiang, jjiang18@uthsc.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.