Open Access

Inflammatory response in epilepsy is mediated by glial cell gap junction pathway (Review)

  • Authors:
    • Guangliang Wang
    • Jiangtao Wang
    • Cuijuan Xin
    • Jinyu Xiao
    • Jianmin Liang
    • Xuemei Wu
  • View Affiliations

  • Published online on: May 5, 2021     https://doi.org/10.3892/mmr.2021.12132
  • Article Number: 493
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epilepsy is a common neurological disease that affects more than 50 million people worldwide. Neuro­inflammation plays an important role in epilepsy. Activation of the immune system and an excessive inflammatory response can increase the frequency of seizures and increase the susceptibility to epilepsy. Therefore, anti-inflammatory therapies may have antiepileptic effects. Connexin 43 (Cx43) is a major component of astroglial hemichannels and gap junctions. Gap junctions are important for the direct exchange of substances and information between cells, as well as regulating the neuroinflammatory response, changing neuronal excitability, neuronal apoptosis, and synaptic remodeling. Cx43-mediated gap junction pathway can be crucial in epilepsy-induced neuroinflammatory cascades. Further, pro-inflammatory cytokines may in turn directly affect the expression of the Cx43 protein in astrocytes. Therefore, examining the association between neuroinflammation and epilepsy can be instrumental in uncovering the pathogenesis of epilepsy, which can lead to the development of novel and more effective antiepileptic drugs.

Introduction

Epilepsy is a common neurological disease that affects >50 million people worldwide. Approximately 30% of epileptic patients are resistant to various antiepileptic drugs and eventually develop refractory epilepsy (1,2). Although novel treatment strategies have been developed over the past few years, certain cases of refractory epilepsy cannot be controlled yet (3). In addition, most available treatments are focused on decreasing seizures by inhibiting the excitability of the nervous system (4). Therefore, the development of new antiepileptogenic therapies that can resolve seizures is imminent (5). Recent studies reported that neuroinflammation plays an important role in the development of epilepsy and abnormal neural circuits (611). Further, anti-inflammatory therapies were reported to have possible antiepileptogenic potential (12,13). Therefore, uncovering the role of inflammatory reactions in epilepsy can lead to the development of more effective and novel antiepileptic drugs which can decrease the disability rate and improve the quality of life in epileptic patients.

Inflammatory reaction in the central nervous system

The central nervous system can be termed as an ‘immunity exemption zone’ because the blood-brain barrier restricts the entry of various substances in the blood circulation (1417). However, accumulating evidence indicates that the immune exemption of the central nervous system is relative (18,19). Engelhardt et al (20) reported that although immature T cells cannot pass through the blood-brain barrier, activated T cells can directly attach to the surface of the cerebral vascular endothelium and pass through the blood-brain barrier in the direction of blood flow. After crossing the blood-brain barrier, T cells can mediate inflammation in the central nervous system (20).

The central nervous system has a special ‘immune defense line’. Cells involved in the brain's inflammatory reaction include the microglia, astrocytes and endothelial cells. Microglia are the immune sentinels of the central nervous system; they can be activated by injury stimuli and induce the corresponding inflammatory reaction, which maintains homeostasis in the central nervous system (21). The number of microglia is relatively small, accounting for ~5% of the glial population (2224). Following infection or damage to the central nervous system, the resting microglia acquire antigenicity, their shape becomes extended, and they are activated into macrophages to participate in the inflammatory reaction together with T cells inoculated within the blood circulation (25). However, hyperactivated microglia are important sources of pro-inflammatory factors as well as oxidative stress, and can cause neurotoxicity.

Astrocytes are also important contributors to the inflammatory reaction in the nervous system (26,27). Astrocytes can play a role in neuronal migration movement, maintain the potassium concentration in the central nervous system, regulate neuronal excitability, and present antigens to autoreactive T cells.

Microglia are activated leading to the production of inflammatory factors following trauma or other injuries. Activated microglia can release cytotoxic substances and cytokines (28). Further, tissue injury could lead to the infiltration of circulating immune cells (29). Additionally, brain tissues are also exposed to systemic inflammatory response, which further aggravates the immune response and leads to secondary neuronal damage (30).

Role of the inflammatory reaction in the development of epilepsy

Activation of the immune system and an excessive inflammatory response play crucial roles in the development of chronic seizures (3133). Moreover, neuronal inflammation associated with inflammatory cytokines signaling pathways may trigger epileptogenesis (34). Of note, patients diagnosed with relapsing remitting multiple sclerosis with epilepsy showed more extensive cortical inflammation compared with patients diagnosed with relapsing remitting multiple sclerosis without epilepsy (35). Therefore, examining the association between neuroinflammation and epilepsy can help uncover the pathogenesis. Clinical and animal studies suggested that the immune response is triggered during the pathophysiology of epilepsy and that the inflammatory reaction within the brain may be involved in the development of epilepsy (36,37). Further, the dysregulation of immunoinflammatory reactions during the pathological course of epilepsy was associated with seizure-induced plasticity (10). Rana and Musto (37) reported that neuronal inflammation generated by neural death and astrocytes proliferation was associated with the microglial activation in damaged areas such as the amygdala, piriform and hippocampus in a rat model of lithium-pilocarpine-induced epilepsy (38). This dysfunction can facilitate the occurrence of epileptic seizures or epilepsy-induced neuronal damage (39). Indeed, inflammatory reactions were found to increase the propensity for seizures, change neuronal excitability, damage the blood-brain barrier, and mediate neuronal apoptosis and synaptic remodeling by activating intracellular signaling pathways (40). Seizures can activate microglia and neurons in the brain, and produce a series of inflammatory reactions without additional exogenous stimuli (32,33). Consequently, microglia and neurons secrete large amounts of pro-inflammatory factors and prostaglandins, and activate the complement system, which ultimately promotes neuronal death and synapse regeneration, leading to chronic spontaneous seizures (10,39).

Noteworthy, adenosine triphosphate (ATP) was shown to activate the sterile inflammatory process through interactions with purinergic receptors (40). The ATP-gated ionotropic P2X7 receptor (P2X7R) can mediate the regulation of neuroinflammation and immune reactions in the central nervous system (41). Neuronal injury was reported to activate microglia and increase the expression of P2X7R (42,43). Monif et al (44) reported that the overexpression of P2X7R is sufficient to drive microglial activation. Moreover, the Fas ligand derived from microglia exacerbates P2X7-mediated microglial activation and triggers a vicious cycle of neuronal death (45,46). Low concentrations of ATP act as chemotactic agents for microglia recognition and migration to guide them to the site of injury (47). ATP-stimulated and P2X4R-mediated microglial activation might have an initial protective effect. Activated microglia can remove potentially necrotic cell debris and promote tissue repair, thereby contributing to neuroprotection. Further, activated microglia release neurotrophic factors through activated P2X4Rs and contribute to neuronal survival (48). P2X7R-activated microglia in neuron-microglia co-culture protect neurons from glutamate toxicity primarily by releasing tumor necrosis factor (TNF)-α. The depletion of microglia can lead to an increase in the levels of cytokines and chemokines such as interleukin (IL)-1β, TNF-α, cytokine-induced neutrophil chemoattractant 1 and monocyte chemoattractant protein-1 in the brain, which aggravates brain damage (49). In later stages of brain injury, ATP can stimulate the overexpression of microglia P2X7R, leading to microglia activation and proliferation, as well as cell death (50). Over-activated microglia upregulate the expression of surface immunomodulatory proteins and release of neurotoxic proinflammatory factors such as IL-1β, IL-6, and TNF-α, which can promote further activation of microglia. Long-term inflammatory reactions result in neuronal death that affects both healthy and damaged cells (51,52).

The expression of connexin 43 (Cx43) in astrocytes is affected by inflammatory cytokines (53). Treatment with IL-1β, for 24 h, inhibited the gap junction communication between the human embryonic astrocytes and decreased Cx43 mRNA and protein expression (54). IL-1β had a transient inhibitory effect on gap junction communication between primary astrocytes, and this inhibitory effect was produced through p38-dependent signaling pathway (55). Therefore, Cx43-mediated gap junction communication in astrocytes is closely correlated to the inflammatory response in the central nervous system. They regulate inflammation in the brain and selectively regulate the opening of gap junction communication by intracranial inflammatory factors (Fig. 1).

Regulation of astrocyte glial junction in epilepsy

Cx43 is a major component of astroglial hemichannels and gap junctions (56). Gap junctions play an important role in neuroinflammatory reactions (52). However, the impact of astrocyte Cx43, its hemichannel, and gap junctions on regulating the neuroinflammatory response in epilepsy is still unclear.

Astrocytes are the largest glial cell population in the central nervous system. Cx43 is a gap junction protein that is mainly expressed in astrocytes and mediates over 95% of the gap junction communication in the brain (57). Under physiological conditions, gap junctions allow the exchange of small molecules (<1.5 kDa) between cells. ATP mediates the migration of activated microglia to the injured area, especially in the initial phase of inflammation (41). Further, extracellular ATP induces the release of endogenous ATP from microglia and attracts distant microglia to the injury site, which leads to the promotion of the inflammatory cascade. ATP released by astrocyte hemichannels establishes an ATP gradient in the extracellular environment that can trigger microglial activation (58,59). Increased extracellular ATP concentration in the injury site mediates the activation of microglia around the lesion (60). Of note, the local injection of ATP mimicked the traumatic brain injury-induced microglial activation and the administration of the gap junction channel blocker, carbenic acid, significantly inhibited the microglial activation (61,62). Following injury, extracellular ATP is released from the open hemichannels and mediates a rapid reaction to microglial damage (63). Jesus et al (64) demonstrated that targeted knockout of astrocyte Cx43 expression decreased proinflammatory cytokine levels in the brain following lipopolysaccharide injection. In addition, hemichannel modulators like Cx43 mimetic peptide and Cx43 antisense oligonucleotides could inhibit the inflammatory response mediated by microglial activation following spinal cord injury (65). Taken together, it is plausible that astrocyte gap junction channel can act as a ‘switch’ in the inflammatory signaling cascade by promoting the release of ATP into the extracellular space. The inflammatory response can affect neuronal excitability, neuronal apoptosis and synaptic remodeling. These factors can lead to the development of abnormal neural excitability, which contributes to the pathogenesis of epilepsy. At the same time, pro-inflammatory cytokines can directly affect the expression of Cx43 protein in astrocytes (Fig. 1).

Conclusion

The occurrence, development and maintenance of epileptic seizures progress through a complicated process. The neuroinflammatory reaction can aggravate epilepsy and maintain recurrent episodes by increasing neuronal excitability, mediating neuronal apoptosis and remodeling the synapses. Therefore, controlling the neuroinflammatory reaction can mitigate the downstream cascade. The gap junction pathway mediated by astrocyte Cx43 can play a crucial role in controlling the epilepsy-induced neuroinflammatory cascade. Therefore, Cx43 can be a potential target for managing epileptic inflammatory reactions. Studies that examine the correlation between neuroinflammation and gap junctions will lead to a better understanding of epilepsy pathogenesis and can uncover new treatment targets.

Acknowledgements

Not applicable.

Funding

This study was funded by the National Natural Science Foundation of China (grant nos. 81801284, 81771396 and 31371125) and Jilin Provincial Ring-fenced Funding for Industrial Innovation Project (grant no. 2017C029-1).

Availability of data and materials

Not applicable.

Authors' contributions

CX and JX searched for literature; GW and JW wrote this review; JL and XW revised this review. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

Cx43

connexin 43

ATP

adenosine triphosphate

IL

interleukin

TNF

tumor necrosis factor

References

1 

Beghi E and Giussani G: Treatment of epilepsy in light of the most recent advances. Lancet Neurol. 18:7–8. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Brewer MK, Grossman TR, McKnight TR, Goldberg YP, Landy H and Gentry MS: The 4th International Lafora Epilepsy Workshop: Shifting paradigms, paths to treatment, and hope for patients. Epilepsy Behav. 90:284–286. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Conway CR, Udaiyar A and Schachter SC: Neurostimulation for depression in epilepsy. Epilepsy Behav. 88S:25–32. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Johnson EL: Seizures and epilepsy. Med Clin North Am. 103:309–324. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Klein P, Dingledine R, Aronica E, Bernard C, Blümcke I, Boison D, Brodie MJ, Brooks-Kayal AR, Engel J Jr, Forcelli PA, et al: Commonalities in epileptogenic processes from different acute brain insults: Do they translate? Epilepsia. 59:37–66. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Vezzani A, Balosso S and Ravizza T: Neuroinflammatory pathways as treatment targets and biomarkers in epilepsy. Nat Rev Neurol. 15:459–472. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Hodges SL and Lugo JN: Therapeutic role of targeting mTOR signaling and neuroinflammation in epilepsy. Epilepsy Res. 161:1062822020. View Article : Google Scholar : PubMed/NCBI

8 

Eastman CL, D'Ambrosio R and Ganesh T: Modulating neuroinflammation and oxidative stress to prevent epilepsy and improve outcomes after traumatic brain injury. Neuropharmacology. 172:1079072020. View Article : Google Scholar : PubMed/NCBI

9 

Terrone G, Balosso S, Pauletti A, Ravizza T and Vezzani A: Inflammation and reactive oxygen species as disease modifiers in epilepsy. Neuropharmacology. 167:1077422020. View Article : Google Scholar : PubMed/NCBI

10 

Sanz P and Garcia-Gimeno MA: Reactive glia inflammatory signaling pathways and epilepsy. Int J Mol Sci. 21:40962020. View Article : Google Scholar : PubMed/NCBI

11 

Mukherjee S, Arisi GM, Mims K, Hollingsworth G, O'Neil K and Shapiro LA: Neuroinflammatory mechanisms of post-traumatic epilepsy. J Neuroinflammation. 17:1932020. View Article : Google Scholar : PubMed/NCBI

12 

Korff CM and Dale RC: The immune system in pediatric seizures and epilepsies. Pediatrics. 140:e201635342017. View Article : Google Scholar : PubMed/NCBI

13 

Bauer J, Becker AJ, Elyaman W, Peltola J, Rüegg S, Titulaer MJ, Varley JA and Beghi E: Innate and adaptive immunity in human epilepsies. Epilepsia. 58 (Suppl 3):57–68. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Tamura R, Yoshida K and Toda M: Current understanding of lymphatic vessels in the central nervous system. Neurosurg Rev. 43:1055–1064. 2020. View Article : Google Scholar : PubMed/NCBI

15 

Kipnis J: Multifaceted interactions between adaptive immunity and the central nervous system. Science. 353:766–771. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Klein RS, Garber C and Howard N: Infectious immunity in the central nervous system and brain function. Nat Immunol. 18:132–141. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Wesselingh R, Butzkueven H, Buzzard K, Tarlinton D, O'Brien TJ and Monif M: Innate immunity in the central nervous system: A missing piece of the autoimmune encephalitis puzzle? Front Immunol. 10:20662019. View Article : Google Scholar : PubMed/NCBI

18 

Peralta Ramos JM, Iribarren P, Bousset L, Melki R, Baekelandt V and Van der Perren A: Peripheral inflammation regulates CNS immune surveillance through the recruitment of inflammatory monocytes upon systemic α-synuclein administration. Front Immunol. 10:802019. View Article : Google Scholar : PubMed/NCBI

19 

Prinz M and Priller J: The role of peripheral immune cells in the CNS in steady state and disease. Nat Neurosci. 20:136–144. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Engelhardt B, Vajkoczy P and Weller RO: The movers and shapers in immune privilege of the CNS. Nat Immunol. 18:123–131. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Greenhalgh AD, Zarruk JG, Healy LM, Baskar Jesudasan SJ, Jhelum P, Salmon CK, Formanek A, Russo MV, Antel JP, McGavern DB, et al: Peripherally derived macrophages modulate microglial function to reduce inflammation after CNS injury. PLoS Biol. 16:e20052642018. View Article : Google Scholar : PubMed/NCBI

22 

Bohlen CJ, Friedman BA, Dejanovic B and Sheng M: Microglia in brain development, homeostasis, and neurodegeneration. Annu Rev Genet. 53:263–288. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Tan YL, Yuan Y and Tian L: Microglial regional heterogeneity and its role in the brain. Mol Psychiatry. 25:351–367. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Illes P, Rubini P, Ulrich H, Zhao Y and Tang Y: Regulation of microglial functions by purinergic mechanisms in the healthy and diseased CNS. Cells. 9:11082020. View Article : Google Scholar : PubMed/NCBI

25 

Hsu M, Rayasam A, Kijak JA, Choi YH, Harding JS, Marcus SA, Karpus WJ, Sandor M and Fabry Z: Neuroinflammation-induced lymphangiogenesis near the cribriform plate contributes to drainage of CNS-derived antigens and immune cells. Nat Commun. 10:2292019. View Article : Google Scholar : PubMed/NCBI

26 

Wheeler MA, Jaronen M, Covacu R, Zandee SEJ, Scalisi G, Rothhammer V, Tjon EC, Chao CC, Kenison JE, Blain M, et al: Environmental control of astrocyte pathogenic activities in CNS inflammation. Cell. 176:581–596.e18. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Rothhammer V and Quintana FJ: Control of autoimmune CNS inflammation by astrocytes. Semin Immunopathol. 37:625–638. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Anderson WD, Greenhalgh AD, Takwale A, David S and Vadigepalli R: Novel influences of IL-10 on CNS inflammation revealed by integrated analyses of cytokine networks and microglial morphology. Front Cell Neurosci. 11:2332017. View Article : Google Scholar : PubMed/NCBI

29 

Chiang N and Serhan CN: Structural elucidation and physiologic functions of specialized pro-resolving mediators and their receptors. Mol Aspects Med. 58:114–129. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Dupuis N and Auvin S: Inflammation and epilepsy in the developing brain: Clinical and experimental evidence. CNS Neurosci Ther. 21:141–151. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Mukhtar I: Inflammatory and immune mechanisms underlying epileptogenesis and epilepsy: From pathogenesis to treatment target. Seizure. 82:65–79. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Wyatt-Johnson SK and Brewster AL: Emerging Roles for Microglial Phagocytic Signaling in Epilepsy. Epilepsy Curr. 20:33–38. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Hiragi T, Ikegaya Y and Koyama R: Microglia after seizures and in epilepsy. Cells. 7:262018. View Article : Google Scholar : PubMed/NCBI

34 

Paudel YN, Shaikh MF, Shah S, Kumari Y and Othman I: Role of inflammation in epilepsy and neurobehavioral comorbidities: Implication for therapy. Eur J Pharmacol. 837:145–155. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Calabrese M, De Stefano N, Atzori M, Bernardi V, Mattisi I, Barachino L, Rinaldi L, Morra A, McAuliffe MM, Perini P, et al: Extensive cortical inflammation is associated with epilepsy in multiple sclerosis. J Neurol. 255:581–586. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Drion CM, van Scheppingen J, Arena A, Geijtenbeek KW, Kooijman L, van Vliet EA, Aronica E and Gorter JA: Effects of rapamycin and curcumin on inflammation and oxidative stress in vitro and in vivo - in search of potential anti-epileptogenic strategies for temporal lobe epilepsy. J Neuroinflammation. 15:2122018. View Article : Google Scholar : PubMed/NCBI

37 

Rana A and Musto AE: The role of inflammation in the development of epilepsy. J Neuroinflammation. 15:1442018. View Article : Google Scholar : PubMed/NCBI

38 

Jung KH, Chu K, Lee ST, Kim JH, Kang KM, Song EC, Kim SJ, Park HK, Kim M, Lee SK, et al: Region-specific plasticity in the epileptic rat brain: A hippocampal and extrahippocampal analysis. Epilepsia. 50:537–549. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Vezzani A and Rüegg S: Introduction to the 2nd Meeting on Immunity and Inflammation in Epilepsy (IIE2016). Epilepsia. 58 (Suppl 3):7–10. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Mazarati AM, Lewis ML and Pittman QJ: Neurobehavioral comorbidities of epilepsy: Role of inflammation. Epilepsia. 58 (Suppl 3):48–56. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Allsopp RC, Dayl S, Schmid R and Evans RJ: Unique residues in the ATP gated human P2X7 receptor define a novel allosteric binding pocket for the selective antagonist AZ10606120. Sci Rep. 7:7252017. View Article : Google Scholar : PubMed/NCBI

42 

Nuka E, Ohnishi K, Terao J and Kawai Y: ATP/P2X7 receptor signaling as a potential anti-inflammatory target of natural polyphenols. PLoS One. 13:e02042292018. View Article : Google Scholar : PubMed/NCBI

43 

Zumerle S, Calì B, Munari F, Angioni R, Di Virgilio F, Molon B and Viola A: Intercellular calcium signaling induced by ATP potentiates macrophage phagocytosis. Cell Rep. 27:1–10.e4. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Monif M, Burnstock G and Williams DA: Microglia: Proliferation and activation driven by the P2X7 receptor. Int J Biochem Cell Biol. 42:1753–1756. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Li R, Shang Y, Hu X, Yu Y, Zhou T, Xiong W and Zou X: ATP/P2X7r axis mediates the pathological process of allergic asthma by inducing M2 polarization of alveolar macrophages. Exp Cell Res. 386:1117082020. View Article : Google Scholar : PubMed/NCBI

46 

Ryoden Y, Fujii T, Segawa K and Nagata S: Functional expression of the P2X7 ATP receptor requires eros. J Immunol. 204:559–568. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Burnstock G and Knight GE: The potential of P2X7 receptors as a therapeutic target, including inflammation and tumour progression. Purinergic Signal. 14:1–18. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Amadio S, Parisi C, Piras E, Fabbrizio P, Apolloni S, Montilli C, Luchetti S, Ruggieri S, Gasperini C, Laghi-Pasini F, et al: Modulation of P2X7 receptor during inflammation in multiple sclerosis. Front Immunol. 8:15292017. View Article : Google Scholar : PubMed/NCBI

49 

Wei L, Syed Mortadza SA, Yan J, Zhang L, Wang L, Yin Y, Li C, Chalon S, Emond P, Belzung C, et al: ATP-activated P2X7 receptor in the pathophysiology of mood disorders and as an emerging target for the development of novel antidepressant therapeutics. Neurosci Biobehav Rev. 87:192–205. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Beamer E, Fischer W and Engel T: The ATP-gated P2X7 receptor as a target for the treatment of drug-resistant epilepsy. Front Neurosci. 11:212017. View Article : Google Scholar : PubMed/NCBI

51 

Kaczmarek-Hajek K, Zhang J, Kopp R, Grosche A, Rissiek B, Saul A, Bruzzone S, Engel T, Jooss T, Krautloher A, et al: Re-evaluation of neuronal P2X7 expression using novel mouse models and a P2X7-specific nanobody. Elife. 7:e362172018. View Article : Google Scholar : PubMed/NCBI

52 

Kim Y, Davidson JO, Gunn KC, Phillips AR, Green CR and Gunn AJ: Role of hemichannels in CNS inflammation and the inflammasome pathway. Adv Protein Chem Struct Biol. 104:1–37. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Das A, Wallace GC IV, Holmes C, McDowell ML, Smith JA, Marshall JD, Bonilha L, Edwards JC, Glazier SS, Ray SK, et al: Hippocampal tissue of patients with refractory temporal lobe epilepsy is associated with astrocyte activation, inflammation, and altered expression of channels and receptors. Neuroscience. 220:237–246. 2012. View Article : Google Scholar : PubMed/NCBI

54 

John GR, Scemes E, Suadicani SO, Liu JS, Charles PC, Lee SC, Spray DC and Brosnan CF: IL-1beta differentially regulates calcium wave propagation between primary human fetal astrocytes via pathways involving P2 receptors and gap junction channels. Proc Natl Acad Sci USA. 96:11613–11618. 1999. View Article : Google Scholar : PubMed/NCBI

55 

Zvalova D, Cordier J, Mesnil M, Junier MP and Chneiweiss H: p38/SAPK2 controls gap junction closure in astrocytes. Glia. 46:323–333. 2004. View Article : Google Scholar : PubMed/NCBI

56 

De Bock M, Wang N, Decrock E, Bultynck G and Leybaert L: Intracellular cleavage of the Cx43 C-terminal domain by matrix-metalloproteases: A novel contributor to inflammation? Mediators Inflamm. 2015:2574712015. View Article : Google Scholar : PubMed/NCBI

57 

Jourdeuil K and Taneyhill LA: The gap junction protein connexin 43 controls multiple aspects of cranial neural crest cell development. J Cell Sci. 133:jcs2354402020. View Article : Google Scholar : PubMed/NCBI

58 

Nikolic L, Shen W, Nobili P, Virenque A, Ulmann L and Audinat E: Blocking TNFα-driven astrocyte purinergic signaling restores normal synaptic activity during epileptogenesis. Glia. 66:2673–2683. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Ghaemi A, Alizadeh L, Babaei S, Jafarian M, Khaleghi Ghadiri M, Meuth SG, Kovac S and Gorji A: Astrocyte-mediated inflammation in cortical spreading depression. Cephalalgia. 38:626–638. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Webster KM, Sun M, Crack P, O'Brien TJ, Shultz SR and Semple BD: Inflammation in epileptogenesis after traumatic brain injury. J Neuroinflammation. 14:102017. View Article : Google Scholar : PubMed/NCBI

61 

Illes P, Burnstock G and Tang Y: Astroglia-derived ATP modulates CNS neuronal circuits. Trends Neurosci. 42:885–898. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Nakamura Y, Park JH and Hayakawa K: Therapeutic use of extracellular mitochondria in CNS injury and disease. Exp Neurol. 324:1131142020. View Article : Google Scholar : PubMed/NCBI

63 

Li W, Li J, Sama AE and Wang H: Carbenoxolone blocks endotoxin-induced protein kinase R (PKR) activation and high mobility group box 1 (HMGB1) release. Mol Med. 19:203–211. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Jesus LB, Santos AB, Jesus EEV, Santos RGD, Grangeiro MS, Bispo-da-Silva A, Arruda MR, Argolo DS, Pinheiro AM, El-Bachá RS, et al: IDO, COX and iNOS have an important role in the proliferation of Neospora caninum in neuron/glia co-cultures. Vet Parasitol. 266:96–102. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Kwan T, Floyd CL, Kim S and King PH: RNA binding protein human antigen R is translocated in astrocytes following spinal cord injury and promotes the inflammatory response. J Neurotrauma. 34:1249–1259. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2021
Volume 24 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang G, Wang J, Xin C, Xiao J, Liang J and Wu X: Inflammatory response in epilepsy is mediated by glial cell gap junction pathway (Review). Mol Med Rep 24: 493, 2021
APA
Wang, G., Wang, J., Xin, C., Xiao, J., Liang, J., & Wu, X. (2021). Inflammatory response in epilepsy is mediated by glial cell gap junction pathway (Review). Molecular Medicine Reports, 24, 493. https://doi.org/10.3892/mmr.2021.12132
MLA
Wang, G., Wang, J., Xin, C., Xiao, J., Liang, J., Wu, X."Inflammatory response in epilepsy is mediated by glial cell gap junction pathway (Review)". Molecular Medicine Reports 24.1 (2021): 493.
Chicago
Wang, G., Wang, J., Xin, C., Xiao, J., Liang, J., Wu, X."Inflammatory response in epilepsy is mediated by glial cell gap junction pathway (Review)". Molecular Medicine Reports 24, no. 1 (2021): 493. https://doi.org/10.3892/mmr.2021.12132