Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2022

Open Access 01-12-2022 | Septicemia | Research

TNFα-mediated necroptosis in brain endothelial cells as a potential mechanism of increased seizure susceptibility in mice following systemic inflammation

Authors: Wan-Yu Huang, Yen-Ling Lai, Ko-Hung Liu, Shankung Lin, Hsuan-Ying Chen, Chih-Hung Liang, Hung-Ming Wu, Kuei-Sen Hsu

Published in: Journal of Neuroinflammation | Issue 1/2022

Login to get access

Abstract

Background

Systemic inflammation is a potent contributor to increased seizure susceptibility. However, information regarding the effects of systemic inflammation on cerebral vascular integrity that influence neuron excitability is scarce. Necroptosis is closely associated with inflammation in various neurological diseases. In this study, necroptosis was hypothesized to be involved in the mechanism underlying sepsis-associated neuronal excitability in the cerebrovascular components (e.g., endothelia cells).

Methods

Lipopolysaccharide (LPS) was used to induce systemic inflammation. Kainic acid intraperitoneal injection was used to measure the susceptibility of the mice to seizure. The pharmacological inhibitors C87 and GSK872 were used to block the signaling of TNFα receptors and necroptosis. In order to determine the features of the sepsis-associated response in the cerebral vasculature and CNS, brain tissues of mice were obtained for assays of the necroptosis-related protein expression, and for immunofluorescence staining to identify morphological changes in the endothelia and glia. In addition, microdialysis assay was used to assess the changes in extracellular potassium and glutamate levels in the brain.

Results

Some noteworthy findings, such as increased seizure susceptibility and brain endothelial necroptosis, Kir4.1 dysfunction, and microglia activation were observed in mice following LPS injection. C87 treatment, a TNFα receptor inhibitor, showed considerable attenuation of increased kainic acid-induced seizure susceptibility, endothelial cell necroptosis, microglia activation and restoration of Kir4.1 protein expression in LPS-treated mice. Treatment with GSK872, a RIP3 inhibitor, such as C87, showed similar effects on these changes following LPS injection.

Conclusions

The findings of this study showed that TNFα-mediated necroptosis induced cerebrovascular endothelial damage, neuroinflammation and astrocyte Kir4.1 dysregulation, which may coalesce to contribute to the increased seizure susceptibility in LPS-treated mice. Pharmacologic inhibition targeting this necroptosis pathway may provide a promising therapeutic approach to the reduction of sepsis-associated brain endothelia cell injury, astrocyte ion channel dysfunction, and subsequent neuronal excitability.
Appendix
Available only for authorised users
Literature
1.
go back to reference Sonneville R, Verdonk F, Rauturier C, Klein IF, Wolff M, Annane D, Chretien F, Sharshar T. Understanding brain dysfunction in sepsis. Ann Intensive Care. 2013;3:15.PubMedPubMedCentral Sonneville R, Verdonk F, Rauturier C, Klein IF, Wolff M, Annane D, Chretien F, Sharshar T. Understanding brain dysfunction in sepsis. Ann Intensive Care. 2013;3:15.PubMedPubMedCentral
2.
go back to reference Idro R, Gwer S, Kahindi M, Gatakaa H, Kazungu T, Ndiritu M, Maitland K, Neville BG, Kager PA, Newton CR. The incidence, aetiology and outcome of acute seizures in children admitted to a rural Kenyan district hospital. BMC Pediatr. 2008;8:5.PubMedPubMedCentral Idro R, Gwer S, Kahindi M, Gatakaa H, Kazungu T, Ndiritu M, Maitland K, Neville BG, Kager PA, Newton CR. The incidence, aetiology and outcome of acute seizures in children admitted to a rural Kenyan district hospital. BMC Pediatr. 2008;8:5.PubMedPubMedCentral
4.
go back to reference Riazi K, Galic MA, Pittman QJ. Contributions of peripheral inflammation to seizure susceptibility: cytokines and brain excitability. Epilepsy Res. 2010;89:34–42.PubMed Riazi K, Galic MA, Pittman QJ. Contributions of peripheral inflammation to seizure susceptibility: cytokines and brain excitability. Epilepsy Res. 2010;89:34–42.PubMed
5.
go back to reference Cerri C, Genovesi S, Allegra M, Pistillo F, Puntener U, Guglielmotti A, Perry VH, Bozzi Y, Caleo M. The chemokine CCL2 mediates the seizure-enhancing effects of systemic inflammation. J Neurosci. 2016;36:3777–88.PubMedPubMedCentral Cerri C, Genovesi S, Allegra M, Pistillo F, Puntener U, Guglielmotti A, Perry VH, Bozzi Y, Caleo M. The chemokine CCL2 mediates the seizure-enhancing effects of systemic inflammation. J Neurosci. 2016;36:3777–88.PubMedPubMedCentral
6.
go back to reference Galic MA, Riazi K, Heida JG, Mouihate A, Fournier NM, Spencer SJ, Kalynchuk LE, Teskey GC, Pittman QJ. Postnatal inflammation increases seizure susceptibility in adult rats. J Neurosci. 2008;28:6904–13.PubMedPubMedCentral Galic MA, Riazi K, Heida JG, Mouihate A, Fournier NM, Spencer SJ, Kalynchuk LE, Teskey GC, Pittman QJ. Postnatal inflammation increases seizure susceptibility in adult rats. J Neurosci. 2008;28:6904–13.PubMedPubMedCentral
7.
go back to reference Huang WY, Lin S, Chen HY, Chen YP, Chen TY, Hsu KS, Wu HM. NADPH oxidases as potential pharmacological targets against increased seizure susceptibility after systemic inflammation. J Neuroinflammation. 2018;15:140.PubMedPubMedCentral Huang WY, Lin S, Chen HY, Chen YP, Chen TY, Hsu KS, Wu HM. NADPH oxidases as potential pharmacological targets against increased seizure susceptibility after systemic inflammation. J Neuroinflammation. 2018;15:140.PubMedPubMedCentral
8.
go back to reference Harre EM, Galic MA, Mouihate A, Noorbakhsh F, Pittman QJ. Neonatal inflammation produces selective behavioural deficits and alters N-methyl-D-aspartate receptor subunit mRNA in the adult rat brain. Eur J Neurosci. 2008;27:644–53.PubMedPubMedCentral Harre EM, Galic MA, Mouihate A, Noorbakhsh F, Pittman QJ. Neonatal inflammation produces selective behavioural deficits and alters N-methyl-D-aspartate receptor subunit mRNA in the adult rat brain. Eur J Neurosci. 2008;27:644–53.PubMedPubMedCentral
9.
go back to reference Kovacs R, Heinemann U, Steinhauser C. Mechanisms underlying blood-brain barrier dysfunction in brain pathology and epileptogenesis: role of astroglia. Epilepsia. 2012;53(Suppl 6):53–9.PubMed Kovacs R, Heinemann U, Steinhauser C. Mechanisms underlying blood-brain barrier dysfunction in brain pathology and epileptogenesis: role of astroglia. Epilepsia. 2012;53(Suppl 6):53–9.PubMed
10.
go back to reference Baruah J, Vasudevan A, Kohling R. Vascular integrity and signaling determining brain development, network excitability, and epileptogenesis. Front Physiol. 2019;10:1583.PubMed Baruah J, Vasudevan A, Kohling R. Vascular integrity and signaling determining brain development, network excitability, and epileptogenesis. Front Physiol. 2019;10:1583.PubMed
11.
go back to reference Lecuyer MA, Kebir H, Prat A. Glial influences on BBB functions and molecular players in immune cell trafficking. Biochim Biophys Acta. 2016;1862:472–82.PubMed Lecuyer MA, Kebir H, Prat A. Glial influences on BBB functions and molecular players in immune cell trafficking. Biochim Biophys Acta. 2016;1862:472–82.PubMed
12.
go back to reference Elwood E, Lim Z, Naveed H, Galea I. The effect of systemic inflammation on human brain barrier function. Brain Behav Immun. 2017;62:35–40.PubMedPubMedCentral Elwood E, Lim Z, Naveed H, Galea I. The effect of systemic inflammation on human brain barrier function. Brain Behav Immun. 2017;62:35–40.PubMedPubMedCentral
13.
go back to reference Rochfort KD, Cummins PM. The blood–brain barrier endothelium: a target for pro-inflammatory cytokines. Biochem Soc Trans. 2015;43:702–6.PubMed Rochfort KD, Cummins PM. The blood–brain barrier endothelium: a target for pro-inflammatory cytokines. Biochem Soc Trans. 2015;43:702–6.PubMed
14.
go back to reference Varatharaj A, Galea I. The blood-brain barrier in systemic inflammation. Brain Behav Immun. 2017;60:1–12.PubMed Varatharaj A, Galea I. The blood-brain barrier in systemic inflammation. Brain Behav Immun. 2017;60:1–12.PubMed
15.
go back to reference Ohno Y, Tokudome K, Kunisawa N, Iha HA, Kinboshi M, Mukai T, Serikawa T, Shimizu S. Role of astroglial Kir4.1 channels in the pathogenesis and treatment of epilepsy. Ther Targets Neurol Dis. 2015;2: e476. Ohno Y, Tokudome K, Kunisawa N, Iha HA, Kinboshi M, Mukai T, Serikawa T, Shimizu S. Role of astroglial Kir4.1 channels in the pathogenesis and treatment of epilepsy. Ther Targets Neurol Dis. 2015;2: e476.
16.
go back to reference Olsen ML, Sontheimer H. Functional implications for Kir4.1 channels in glial biology: from K+ buffering to cell differentiation. J Neurochem. 2008;107:589–601.PubMedPubMedCentral Olsen ML, Sontheimer H. Functional implications for Kir4.1 channels in glial biology: from K+ buffering to cell differentiation. J Neurochem. 2008;107:589–601.PubMedPubMedCentral
17.
go back to reference Newton K, Manning G. Necroptosis and inflammation. Annu Rev Biochem. 2016;85:743–63.PubMed Newton K, Manning G. Necroptosis and inflammation. Annu Rev Biochem. 2016;85:743–63.PubMed
18.
20.
go back to reference Chen AQ, Fang Z, Chen XL, Yang S, Zhou YF, Mao L, Xia YP, Jin HJ, Li YN, You MF, et al. Microglia-derived TNF-alpha mediates endothelial necroptosis aggravating blood brain-barrier disruption after ischemic stroke. Cell Death Dis. 2019;10:487.PubMedPubMedCentral Chen AQ, Fang Z, Chen XL, Yang S, Zhou YF, Mao L, Xia YP, Jin HJ, Li YN, You MF, et al. Microglia-derived TNF-alpha mediates endothelial necroptosis aggravating blood brain-barrier disruption after ischemic stroke. Cell Death Dis. 2019;10:487.PubMedPubMedCentral
21.
go back to reference Zille M, Ikhsan M, Jiang Y, Lampe J, Wenzel J, Schwaninger M. The impact of endothelial cell death in the brain and its role after stroke: a systematic review. Cell Stress. 2019;3:330–47.PubMedPubMedCentral Zille M, Ikhsan M, Jiang Y, Lampe J, Wenzel J, Schwaninger M. The impact of endothelial cell death in the brain and its role after stroke: a systematic review. Cell Stress. 2019;3:330–47.PubMedPubMedCentral
22.
go back to reference Shrum B, Anantha RV, Xu SX, Donnelly M, Haeryfar SM, McCormick JK, Mele T. A robust scoring system to evaluate sepsis severity in an animal model. BMC Res Notes. 2014;7:233.PubMedPubMedCentral Shrum B, Anantha RV, Xu SX, Donnelly M, Haeryfar SM, McCormick JK, Mele T. A robust scoring system to evaluate sepsis severity in an animal model. BMC Res Notes. 2014;7:233.PubMedPubMedCentral
23.
go back to reference Morrison RS, Wenzel HJ, Kinoshita Y, Robbins CA, Donehower LA, Schwartzkroin PA. Loss of the p53 tumor suppressor gene protects neurons from kainate-induced cell death. J Neurosci. 1996;16:1337–45.PubMedPubMedCentral Morrison RS, Wenzel HJ, Kinoshita Y, Robbins CA, Donehower LA, Schwartzkroin PA. Loss of the p53 tumor suppressor gene protects neurons from kainate-induced cell death. J Neurosci. 1996;16:1337–45.PubMedPubMedCentral
24.
25.
go back to reference Huang WY, Liu KH, Lin S, Chen TY, Tseng CY, Chen HY, Wu HM, Hsu KS. NADPH oxidase 2 as a potential therapeutic target for protection against cognitive deficits following systemic inflammation in mice. Brain Behav Immun. 2020;84:242–52.PubMed Huang WY, Liu KH, Lin S, Chen TY, Tseng CY, Chen HY, Wu HM, Hsu KS. NADPH oxidase 2 as a potential therapeutic target for protection against cognitive deficits following systemic inflammation in mice. Brain Behav Immun. 2020;84:242–52.PubMed
26.
go back to reference Tao K, Cai Q, Zhang X, Zhu L, Liu Z, Li F, Wang Q, Liu L, Feng D. Astrocytic histone deacetylase 2 facilitates delayed depression and memory impairment after subarachnoid hemorrhage by negatively regulating glutamate transporter-1. Ann Transl Med. 2020;8:691.PubMedPubMedCentral Tao K, Cai Q, Zhang X, Zhu L, Liu Z, Li F, Wang Q, Liu L, Feng D. Astrocytic histone deacetylase 2 facilitates delayed depression and memory impairment after subarachnoid hemorrhage by negatively regulating glutamate transporter-1. Ann Transl Med. 2020;8:691.PubMedPubMedCentral
27.
go back to reference Chen HY, Cheng FC, Pan HC, Hsu JC, Wang MF. Magnesium enhances exercise performance via increasing glucose availability in the blood, muscle, and brain during exercise. PLoS ONE. 2014;9: e85486.PubMedPubMedCentral Chen HY, Cheng FC, Pan HC, Hsu JC, Wang MF. Magnesium enhances exercise performance via increasing glucose availability in the blood, muscle, and brain during exercise. PLoS ONE. 2014;9: e85486.PubMedPubMedCentral
28.
go back to reference Mao XY, Zhou HH, Jin WL. Redox-related neuronal death and crosstalk as drug targets: focus on epilepsy. Front Neurosci. 2019;13:512.PubMedPubMedCentral Mao XY, Zhou HH, Jin WL. Redox-related neuronal death and crosstalk as drug targets: focus on epilepsy. Front Neurosci. 2019;13:512.PubMedPubMedCentral
29.
go back to reference Devinsky O, Vezzani A, Najjar S, De Lanerolle NC, Rogawski MA. Glia and epilepsy: excitability and inflammation. Trends Neurosci. 2013;36:174–84.PubMed Devinsky O, Vezzani A, Najjar S, De Lanerolle NC, Rogawski MA. Glia and epilepsy: excitability and inflammation. Trends Neurosci. 2013;36:174–84.PubMed
30.
go back to reference Blaser H, Dostert C, Mak TW, Brenner D. TNF and ROS crosstalk in inflammation. Trends Cell Biol. 2016;26:249–61.PubMed Blaser H, Dostert C, Mak TW, Brenner D. TNF and ROS crosstalk in inflammation. Trends Cell Biol. 2016;26:249–61.PubMed
31.
go back to reference Higashi K, Fujita A, Inanobe A, Tanemoto M, Doi K, Kubo T, Kurachi Y. An inwardly rectifying K(+) channel, Kir4.1, expressed in astrocytes surrounds synapses and blood vessels in brain. Am J Physiol Cell Physiol. 2001;281:C922–31.PubMed Higashi K, Fujita A, Inanobe A, Tanemoto M, Doi K, Kubo T, Kurachi Y. An inwardly rectifying K(+) channel, Kir4.1, expressed in astrocytes surrounds synapses and blood vessels in brain. Am J Physiol Cell Physiol. 2001;281:C922–31.PubMed
32.
go back to reference Kucheryavykh YV, Kucheryavykh LY, Nichols CG, Maldonado HM, Baksi K, Reichenbach A, Skatchkov SN, Eaton MJ. Downregulation of Kir4.1 inward rectifying potassium channel subunits by RNAi impairs potassium transfer and glutamate uptake by cultured cortical astrocytes. Glia. 2007;55:274–81.PubMed Kucheryavykh YV, Kucheryavykh LY, Nichols CG, Maldonado HM, Baksi K, Reichenbach A, Skatchkov SN, Eaton MJ. Downregulation of Kir4.1 inward rectifying potassium channel subunits by RNAi impairs potassium transfer and glutamate uptake by cultured cortical astrocytes. Glia. 2007;55:274–81.PubMed
33.
go back to reference Djukic B, Casper KB, Philpot BD, Chin LS, McCarthy KD. Conditional knock-out of Kir4.1 leads to glial membrane depolarization, inhibition of potassium and glutamate uptake, and enhanced short-term synaptic potentiation. J Neurosci. 2007;27:11354–65.PubMedPubMedCentral Djukic B, Casper KB, Philpot BD, Chin LS, McCarthy KD. Conditional knock-out of Kir4.1 leads to glial membrane depolarization, inhibition of potassium and glutamate uptake, and enhanced short-term synaptic potentiation. J Neurosci. 2007;27:11354–65.PubMedPubMedCentral
34.
go back to reference Ohno Y. Astrocytic Kir4.1 potassium channels as a novel therapeutic target for epilepsy and mood disorders. Neural Regen Res. 2018;13:651–2.PubMedPubMedCentral Ohno Y. Astrocytic Kir4.1 potassium channels as a novel therapeutic target for epilepsy and mood disorders. Neural Regen Res. 2018;13:651–2.PubMedPubMedCentral
35.
go back to reference Inyushin M, Kucheryavykh LY, Kucheryavykh YV, Nichols CG, Buono RJ, Ferraro TN, Skatchkov SN, Eaton MJ. Potassium channel activity and glutamate uptake are impaired in astrocytes of seizure-susceptible DBA/2 mice. Epilepsia. 2010;51:1707–13.PubMedPubMedCentral Inyushin M, Kucheryavykh LY, Kucheryavykh YV, Nichols CG, Buono RJ, Ferraro TN, Skatchkov SN, Eaton MJ. Potassium channel activity and glutamate uptake are impaired in astrocytes of seizure-susceptible DBA/2 mice. Epilepsia. 2010;51:1707–13.PubMedPubMedCentral
36.
go back to reference Zurolo E, de Groot M, Iyer A, Anink J, van Vliet EA, Heimans JJ, Reijneveld JC, Gorter JA, Aronica E. Regulation of Kir4.1 expression in astrocytes and astrocytic tumors: a role for interleukin-1 beta. J Neuroinflammation. 2012;9:280.PubMedPubMedCentral Zurolo E, de Groot M, Iyer A, Anink J, van Vliet EA, Heimans JJ, Reijneveld JC, Gorter JA, Aronica E. Regulation of Kir4.1 expression in astrocytes and astrocytic tumors: a role for interleukin-1 beta. J Neuroinflammation. 2012;9:280.PubMedPubMedCentral
37.
go back to reference Nwaobi SE, Cuddapah VA, Patterson KC, Randolph AC, Olsen ML. The role of glial-specific Kir4.1 in normal and pathological states of the CNS. Acta Neuropathol. 2016;132:1–21.PubMedPubMedCentral Nwaobi SE, Cuddapah VA, Patterson KC, Randolph AC, Olsen ML. The role of glial-specific Kir4.1 in normal and pathological states of the CNS. Acta Neuropathol. 2016;132:1–21.PubMedPubMedCentral
38.
go back to reference van Vliet EA, da Costa AS, Redeker S, van Schaik R, Aronica E, Gorter JA. Blood–brain barrier leakage may lead to progression of temporal lobe epilepsy. Brain. 2007;130:521–34.PubMed van Vliet EA, da Costa AS, Redeker S, van Schaik R, Aronica E, Gorter JA. Blood–brain barrier leakage may lead to progression of temporal lobe epilepsy. Brain. 2007;130:521–34.PubMed
39.
go back to reference Marchi N, Tierney W, Alexopoulos AV, Puvenna V, Granata T, Janigro D. The etiological role of blood-brain barrier dysfunction in seizure disorders. Cardiovasc Psychiatry Neurol. 2011;2011: 482415.PubMedPubMedCentral Marchi N, Tierney W, Alexopoulos AV, Puvenna V, Granata T, Janigro D. The etiological role of blood-brain barrier dysfunction in seizure disorders. Cardiovasc Psychiatry Neurol. 2011;2011: 482415.PubMedPubMedCentral
40.
go back to reference Banks WA, Gray AM, Erickson MA, Salameh TS, Damodarasamy M, Sheibani N, Meabon JS, Wing EE, Morofuji Y, Cook DG, Reed MJ. Lipopolysaccharide-induced blood-brain barrier disruption: roles of cyclooxygenase, oxidative stress, neuroinflammation, and elements of the neurovascular unit. J Neuroinflammation. 2015;12:223.PubMedPubMedCentral Banks WA, Gray AM, Erickson MA, Salameh TS, Damodarasamy M, Sheibani N, Meabon JS, Wing EE, Morofuji Y, Cook DG, Reed MJ. Lipopolysaccharide-induced blood-brain barrier disruption: roles of cyclooxygenase, oxidative stress, neuroinflammation, and elements of the neurovascular unit. J Neuroinflammation. 2015;12:223.PubMedPubMedCentral
41.
go back to reference Li JH, Pober JS. The cathepsin B death pathway contributes to TNF plus IFN-gamma-mediated human endothelial injury. J Immunol. 2005;175:1858–66.PubMed Li JH, Pober JS. The cathepsin B death pathway contributes to TNF plus IFN-gamma-mediated human endothelial injury. J Immunol. 2005;175:1858–66.PubMed
42.
go back to reference Zelic M, Roderick JE, O’Donnell JA, Lehman J, Lim SE, Janardhan HP, Trivedi CM, Pasparakis M, Kelliher MA. RIP kinase 1-dependent endothelial necroptosis underlies systemic inflammatory response syndrome. J Clin Invest. 2018;128:2064–75.PubMedPubMedCentral Zelic M, Roderick JE, O’Donnell JA, Lehman J, Lim SE, Janardhan HP, Trivedi CM, Pasparakis M, Kelliher MA. RIP kinase 1-dependent endothelial necroptosis underlies systemic inflammatory response syndrome. J Clin Invest. 2018;128:2064–75.PubMedPubMedCentral
43.
go back to reference Clark PR, Kim RK, Pober JS, Kluger MS. Tumor necrosis factor disrupts claudin-5 endothelial tight junction barriers in two distinct NF-kappaB-dependent phases. PLoS ONE. 2015;10: e0120075.PubMedPubMedCentral Clark PR, Kim RK, Pober JS, Kluger MS. Tumor necrosis factor disrupts claudin-5 endothelial tight junction barriers in two distinct NF-kappaB-dependent phases. PLoS ONE. 2015;10: e0120075.PubMedPubMedCentral
44.
go back to reference Elahy M, Jackaman C, Mamo JC, Lam V, Dhaliwal SS, Giles C, Nelson D, Takechi R. Blood-brain barrier dysfunction developed during normal aging is associated with inflammation and loss of tight junctions but not with leukocyte recruitment. Immun Ageing. 2015;12:2.PubMedPubMedCentral Elahy M, Jackaman C, Mamo JC, Lam V, Dhaliwal SS, Giles C, Nelson D, Takechi R. Blood-brain barrier dysfunction developed during normal aging is associated with inflammation and loss of tight junctions but not with leukocyte recruitment. Immun Ageing. 2015;12:2.PubMedPubMedCentral
45.
go back to reference Moriwaki K, Chan FK. The inflammatory signal adaptor RIPK3: functions beyond necroptosis. Int Rev Cell Mol Biol. 2017;328:253–75.PubMed Moriwaki K, Chan FK. The inflammatory signal adaptor RIPK3: functions beyond necroptosis. Int Rev Cell Mol Biol. 2017;328:253–75.PubMed
46.
go back to reference Erickson MA, Banks WA. Cytokine and chemokine responses in serum and brain after single and repeated injections of lipopolysaccharide: multiplex quantification with path analysis. Brain Behav Immun. 2011;25:1637–48.PubMedPubMedCentral Erickson MA, Banks WA. Cytokine and chemokine responses in serum and brain after single and repeated injections of lipopolysaccharide: multiplex quantification with path analysis. Brain Behav Immun. 2011;25:1637–48.PubMedPubMedCentral
47.
go back to reference Lehtimaki KA, Peltola J, Koskikallio E, Keranen T, Honkaniemi J. Expression of cytokines and cytokine receptors in the rat brain after kainic acid-induced seizures. Brain Res Mol Brain Res. 2003;110:253–60.PubMed Lehtimaki KA, Peltola J, Koskikallio E, Keranen T, Honkaniemi J. Expression of cytokines and cytokine receptors in the rat brain after kainic acid-induced seizures. Brain Res Mol Brain Res. 2003;110:253–60.PubMed
48.
go back to reference Ravizza T, Vezzani A. Pharmacological targeting of brain inflammation in epilepsy: therapeutic perspectives from experimental and clinical studies. Epilepsia Open. 2018;3:133–42.PubMedPubMedCentral Ravizza T, Vezzani A. Pharmacological targeting of brain inflammation in epilepsy: therapeutic perspectives from experimental and clinical studies. Epilepsia Open. 2018;3:133–42.PubMedPubMedCentral
49.
go back to reference Cai Q, Gan J, Luo R, Qu Y, Li S, Wan C, Mu D. The role of necroptosis in status epilepticus-induced brain injury in juvenile rats. Epilepsy Behav. 2017;75:134–42.PubMed Cai Q, Gan J, Luo R, Qu Y, Li S, Wan C, Mu D. The role of necroptosis in status epilepticus-induced brain injury in juvenile rats. Epilepsy Behav. 2017;75:134–42.PubMed
50.
go back to reference Liu Y, Liu T, Lei T, Zhang D, Du S, Girani L, Qi D, Lin C, Tong R, Wang Y. RIP1/RIP3-regulated necroptosis as a target for multifaceted disease therapy (Review). Int J Mol Med. 2019;44:771–86.PubMedPubMedCentral Liu Y, Liu T, Lei T, Zhang D, Du S, Girani L, Qi D, Lin C, Tong R, Wang Y. RIP1/RIP3-regulated necroptosis as a target for multifaceted disease therapy (Review). Int J Mol Med. 2019;44:771–86.PubMedPubMedCentral
52.
go back to reference Fauster A, Rebsamen M, Huber KV, Bigenzahn JW, Stukalov A, Lardeau CH, Scorzoni S, Bruckner M, Gridling M, Parapatics K, et al. A cellular screen identifies ponatinib and pazopanib as inhibitors of necroptosis. Cell Death Dis. 2015;6: e1767.PubMedPubMedCentral Fauster A, Rebsamen M, Huber KV, Bigenzahn JW, Stukalov A, Lardeau CH, Scorzoni S, Bruckner M, Gridling M, Parapatics K, et al. A cellular screen identifies ponatinib and pazopanib as inhibitors of necroptosis. Cell Death Dis. 2015;6: e1767.PubMedPubMedCentral
Metadata
Title
TNFα-mediated necroptosis in brain endothelial cells as a potential mechanism of increased seizure susceptibility in mice following systemic inflammation
Authors
Wan-Yu Huang
Yen-Ling Lai
Ko-Hung Liu
Shankung Lin
Hsuan-Ying Chen
Chih-Hung Liang
Hung-Ming Wu
Kuei-Sen Hsu
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2022
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-022-02406-0

Other articles of this Issue 1/2022

Journal of Neuroinflammation 1/2022 Go to the issue