Skip to main content
Top
Published in: Journal of Inflammation 1/2015

Open Access 01-12-2015 | Research

Interleukin-18 expression increases in response to neurovascular damage following soman-induced status epilepticus in rats

Authors: Erik A. Johnson, Michelle A. Guignet, Thuy L. Dao, Tracey A. Hamilton, Robert K. Kan

Published in: Journal of Inflammation | Issue 1/2015

Login to get access

Abstract

Background

Status epilepticus (SE) can cause neuronal cell death and impaired behavioral function. Acute exposure to potent acetylcholinesterase inhibitors such as soman (GD) can cause prolonged SE activity, micro-hemorrhage and cell death in the hippocampus, thalamus and piriform cortex. Neuroinflammation is a prominent feature of brain injury with upregulation of multiple pro-inflammatory cytokines including those of the IL-1 family. The highly pleiotropic pro-inflammatory cytokine interleukin-18 (IL-18) belongs to the IL-1 family of cytokines and can propagate neuroinflammation by promoting immune cell infiltration, leukocyte and lymphocyte activation, and angiogenesis and helps facilitate the transition from the innate to the adaptive immune response. The purpose of this study is to characterize the regional and temporal expression of IL −18 and related factors in the brain following SE in a rat GD seizure model followed by localization of IL-18 to specific cell types.

Methods

The protein levels of IL-18, vascular endothelial growth factor and interferon gamma was quantified in the lysates of injured brain regions up to 72 h following GD-induced SE onset using bead multiplex immunoassays. IL-18 was localized to various cell types using immunohistochemistry and transmission electron microscopy. In addition, macrophage appearance scoring and T-cell quantification was determined using immunohistochemistry. Micro-hemorrhages were identified using hematoxylin and eosin staining of brain sections.

Results

Significant increases in IL-18 occurred in the piriform cortex, hippocampus and thalamus following SE. IL-18 was primarily expressed by endothelial cells and astrocytes associated with the damaged neurovascular unit. The increase in IL-18 was not related to macrophage accumulation, neutrophil infiltration or T-cell appearance in the injured tissue.

Conclusions

These data show that IL-18 is significantly upregulated following GD-induced SE and localized primarily to endothelial cells in damaged brain vasculature. IL-18 upregulation occurred following leukocyte/lymphocyte infiltration and in the absence of other IL-18-related cytokines, suggesting another function, potentially for angiogenesis related to GD-induced micro-hemorrhage formation. Further studies at more chronic time points may help to elucidate this function.
Literature
1.
go back to reference Bataillard A et al. Cardiovascular consequences of organophosphorus poisoning and of antidotes in conscious unrestrained rats. Pharmacol Toxicol. 1990;67(1):27–35.PubMedCrossRef Bataillard A et al. Cardiovascular consequences of organophosphorus poisoning and of antidotes in conscious unrestrained rats. Pharmacol Toxicol. 1990;67(1):27–35.PubMedCrossRef
2.
go back to reference Letienne R et al. Soman-induced hypertension in conscious rats is mediated by prolonged central muscarinic stimulation. Fundam Clin Pharmacol. 1999;13(4):468–74.PubMedCrossRef Letienne R et al. Soman-induced hypertension in conscious rats is mediated by prolonged central muscarinic stimulation. Fundam Clin Pharmacol. 1999;13(4):468–74.PubMedCrossRef
3.
go back to reference Shih TM, Scremin OU. Cerebral blood flow and metabolism in soman-induced convulsions. Brain Res Bull. 1992;28(5):735–42.PubMedCrossRef Shih TM, Scremin OU. Cerebral blood flow and metabolism in soman-induced convulsions. Brain Res Bull. 1992;28(5):735–42.PubMedCrossRef
4.
go back to reference McDonough Jr JH et al. Protection against nerve agent-induced neuropathology, but not cardiac pathology, is associated with the anticonvulsant action of drug treatment. Neurotoxicology. 1995;16(1):123–32.PubMed McDonough Jr JH et al. Protection against nerve agent-induced neuropathology, but not cardiac pathology, is associated with the anticonvulsant action of drug treatment. Neurotoxicology. 1995;16(1):123–32.PubMed
5.
go back to reference Zimmer LA, Ennis M, Shipley MT. Soman-induced seizures rapidly activate astrocytes and microglia in discrete brain regions. J Comp Neurol. 1997;378(4):482–92.PubMedCrossRef Zimmer LA, Ennis M, Shipley MT. Soman-induced seizures rapidly activate astrocytes and microglia in discrete brain regions. J Comp Neurol. 1997;378(4):482–92.PubMedCrossRef
6.
go back to reference Baille V et al. Soman-induced convulsions: the neuropathology revisited. Toxicology. 2005;215(1–2):1–24.PubMedCrossRef Baille V et al. Soman-induced convulsions: the neuropathology revisited. Toxicology. 2005;215(1–2):1–24.PubMedCrossRef
7.
go back to reference McLeod Jr CG, Singer AW, Harrington DG. Acute neuropathology in soman poisoned rats. Neurotoxicology. 1984;5(2):53–7.PubMed McLeod Jr CG, Singer AW, Harrington DG. Acute neuropathology in soman poisoned rats. Neurotoxicology. 1984;5(2):53–7.PubMed
8.
go back to reference Johnson EA, Kan RK. The acute phase response and soman-induced status epilepticus: temporal, regional and cellular changes in rat brain cytokine concentrations. J Neuroinflammation. 2010;7:40.PubMedCentralPubMedCrossRef Johnson EA, Kan RK. The acute phase response and soman-induced status epilepticus: temporal, regional and cellular changes in rat brain cytokine concentrations. J Neuroinflammation. 2010;7:40.PubMedCentralPubMedCrossRef
9.
go back to reference Johnson EA et al. Increased expression of the chemokines CXCL1 and MIP-1alpha by resident brain cells precedes neutrophil infiltration in the brain following prolonged soman-induced status epilepticus in rats. J Neuroinflammation. 2011;8:41.PubMedCentralPubMedCrossRef Johnson EA et al. Increased expression of the chemokines CXCL1 and MIP-1alpha by resident brain cells precedes neutrophil infiltration in the brain following prolonged soman-induced status epilepticus in rats. J Neuroinflammation. 2011;8:41.PubMedCentralPubMedCrossRef
10.
go back to reference Arend WP, Palmer G, Gabay C. IL-1, IL-18, and IL-33 families of cytokines. Immunol Rev. 2008;223:20–38.PubMedCrossRef Arend WP, Palmer G, Gabay C. IL-1, IL-18, and IL-33 families of cytokines. Immunol Rev. 2008;223:20–38.PubMedCrossRef
11.
go back to reference Prinz M, Hanisch UK. Murine microglial cells produce and respond to interleukin-18. J Neurochem. 1999;72(5):2215–8.PubMedCrossRef Prinz M, Hanisch UK. Murine microglial cells produce and respond to interleukin-18. J Neurochem. 1999;72(5):2215–8.PubMedCrossRef
12.
13.
go back to reference Wheeler RD et al. Detection of the interleukin 18 family in rat brain by RT-PCR. Brain Res Mol Brain Res. 2000;77(2):290–3.PubMedCrossRef Wheeler RD et al. Detection of the interleukin 18 family in rat brain by RT-PCR. Brain Res Mol Brain Res. 2000;77(2):290–3.PubMedCrossRef
14.
go back to reference Ryu HJ et al. The protective effects of interleukin-18 and interferon-gamma on neuronal damages in the rat hippocampus following status epilepticus. Neuroscience. 2010;170(3):711–21.PubMedCrossRef Ryu HJ et al. The protective effects of interleukin-18 and interferon-gamma on neuronal damages in the rat hippocampus following status epilepticus. Neuroscience. 2010;170(3):711–21.PubMedCrossRef
15.
go back to reference Gerdes N et al. Expression of interleukin (IL)-18 and functional IL-18 receptor on human vascular endothelial cells, smooth muscle cells, and macrophages: implications for atherogenesis. J Exp Med. 2002;195(2):245–57.PubMedCentralPubMedCrossRef Gerdes N et al. Expression of interleukin (IL)-18 and functional IL-18 receptor on human vascular endothelial cells, smooth muscle cells, and macrophages: implications for atherogenesis. J Exp Med. 2002;195(2):245–57.PubMedCentralPubMedCrossRef
16.
go back to reference Conti B et al. Cultures of astrocytes and microglia express interleukin 18. Brain Res Mol Brain Res. 1999;67(1):46–52.PubMedCrossRef Conti B et al. Cultures of astrocytes and microglia express interleukin 18. Brain Res Mol Brain Res. 1999;67(1):46–52.PubMedCrossRef
18.
go back to reference Szabo SJ et al. Molecular mechanisms regulating Th1 immune responses. Annu Rev Immunol. 2003;21:713–58.PubMedCrossRef Szabo SJ et al. Molecular mechanisms regulating Th1 immune responses. Annu Rev Immunol. 2003;21:713–58.PubMedCrossRef
19.
go back to reference Dai SM et al. Interleukin-18 enhances monocyte tumor necrosis factor alpha and interleukin-1beta production induced by direct contact with T lymphocytes: implications in rheumatoid arthritis. Arthritis Rheum. 2004;50(2):432–43.PubMedCrossRef Dai SM et al. Interleukin-18 enhances monocyte tumor necrosis factor alpha and interleukin-1beta production induced by direct contact with T lymphocytes: implications in rheumatoid arthritis. Arthritis Rheum. 2004;50(2):432–43.PubMedCrossRef
20.
go back to reference Jeon GS et al. Glial expression of interleukin-18 and its receptor after excitotoxic damage in the mouse hippocampus. Neurochem Res. 2008;33(1):179–84.PubMedCrossRef Jeon GS et al. Glial expression of interleukin-18 and its receptor after excitotoxic damage in the mouse hippocampus. Neurochem Res. 2008;33(1):179–84.PubMedCrossRef
21.
go back to reference Zhang XM et al. IL-18 deficiency aggravates kainic acid-induced hippocampal neurodegeneration in C57BL/6 mice due to an overcompensation by IL-12. Exp Neurol. 2007;205(1):64–73.PubMedCrossRef Zhang XM et al. IL-18 deficiency aggravates kainic acid-induced hippocampal neurodegeneration in C57BL/6 mice due to an overcompensation by IL-12. Exp Neurol. 2007;205(1):64–73.PubMedCrossRef
22.
go back to reference Morel JC et al. A novel role for interleukin-18 in adhesion molecule induction through NF kappa B and phosphatidylinositol (PI) 3-kinase-dependent signal transduction pathways. J Biol Chem. 2001;276(40):37069–75.PubMedCrossRef Morel JC et al. A novel role for interleukin-18 in adhesion molecule induction through NF kappa B and phosphatidylinositol (PI) 3-kinase-dependent signal transduction pathways. J Biol Chem. 2001;276(40):37069–75.PubMedCrossRef
23.
25.
go back to reference Ariel A et al. IL-12 and IL-18 induce MAP kinase-dependent adhesion of T cells to extracellular matrix components. J Leukoc Biol. 2002;72(1):192–8.PubMed Ariel A et al. IL-12 and IL-18 induce MAP kinase-dependent adhesion of T cells to extracellular matrix components. J Leukoc Biol. 2002;72(1):192–8.PubMed
26.
go back to reference Morel JC et al. Signal transduction pathways involved in rheumatoid arthritis synovial fibroblast interleukin-18-induced vascular cell adhesion molecule-1 expression. J Biol Chem. 2002;277(38):34679–91.PubMedCrossRef Morel JC et al. Signal transduction pathways involved in rheumatoid arthritis synovial fibroblast interleukin-18-induced vascular cell adhesion molecule-1 expression. J Biol Chem. 2002;277(38):34679–91.PubMedCrossRef
27.
28.
go back to reference Ogura T et al. Interleukin-18 stimulates hematopoietic cytokine and growth factor formation and augments circulating granulocytes in mice. Blood. 2001;98(7):2101–7.PubMedCrossRef Ogura T et al. Interleukin-18 stimulates hematopoietic cytokine and growth factor formation and augments circulating granulocytes in mice. Blood. 2001;98(7):2101–7.PubMedCrossRef
29.
go back to reference Yoo JK et al. IL-18 induces monocyte chemotactic protein-1 production in macrophages through the phosphatidylinositol 3-kinase/Akt and MEK/ERK1/2 pathways. J Immunol. 2005;175(12):8280–6.PubMedCrossRef Yoo JK et al. IL-18 induces monocyte chemotactic protein-1 production in macrophages through the phosphatidylinositol 3-kinase/Akt and MEK/ERK1/2 pathways. J Immunol. 2005;175(12):8280–6.PubMedCrossRef
30.
go back to reference Yoshimoto T et al. Interleukin 18 together with interleukin 12 inhibits IgE production by induction of interferon-gamma production from activated B cells. Proc Natl Acad Sci U S A. 1997;94(8):3948–53.PubMedCentralPubMedCrossRef Yoshimoto T et al. Interleukin 18 together with interleukin 12 inhibits IgE production by induction of interferon-gamma production from activated B cells. Proc Natl Acad Sci U S A. 1997;94(8):3948–53.PubMedCentralPubMedCrossRef
31.
go back to reference Micallef MJ et al. Interferon-gamma-inducing factor enhances T helper 1 cytokine production by stimulated human T cells: synergism with interleukin-12 for interferon-gamma production. Eur J Immunol. 1996;26(7):1647–51.PubMedCrossRef Micallef MJ et al. Interferon-gamma-inducing factor enhances T helper 1 cytokine production by stimulated human T cells: synergism with interleukin-12 for interferon-gamma production. Eur J Immunol. 1996;26(7):1647–51.PubMedCrossRef
32.
go back to reference Walker W et al. IL-18 and CD28 use distinct molecular mechanisms to enhance NK cell production of IL-12-induced IFN-gamma. J Immunol. 1999;162(10):5894–901.PubMed Walker W et al. IL-18 and CD28 use distinct molecular mechanisms to enhance NK cell production of IL-12-induced IFN-gamma. J Immunol. 1999;162(10):5894–901.PubMed
33.
go back to reference Dinarello CA et al. Overview of interleukin-18: more than an interferon-gamma inducing factor. J Leukoc Biol. 1998;63(6):658–64.PubMed Dinarello CA et al. Overview of interleukin-18: more than an interferon-gamma inducing factor. J Leukoc Biol. 1998;63(6):658–64.PubMed
34.
go back to reference Nakanishi K et al. Interleukin-18 regulates both Th1 and Th2 responses. Annu Rev Immunol. 2001;19:423–74.PubMedCrossRef Nakanishi K et al. Interleukin-18 regulates both Th1 and Th2 responses. Annu Rev Immunol. 2001;19:423–74.PubMedCrossRef
35.
go back to reference Watanabe-Fukunaga R et al. Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates apoptosis. Nature. 1992;356(6367):314–7.PubMedCrossRef Watanabe-Fukunaga R et al. Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates apoptosis. Nature. 1992;356(6367):314–7.PubMedCrossRef
36.
go back to reference Cho ML et al. Interleukin-18 induces the production of vascular endothelial growth factor (VEGF) in rheumatoid arthritis synovial fibroblasts via AP-1-dependent pathways. Immunol Lett. 2006;103(2):159–66.PubMedCrossRef Cho ML et al. Interleukin-18 induces the production of vascular endothelial growth factor (VEGF) in rheumatoid arthritis synovial fibroblasts via AP-1-dependent pathways. Immunol Lett. 2006;103(2):159–66.PubMedCrossRef
37.
38.
go back to reference Bernardini G et al. Analysis of the role of chemokines in angiogenesis. J Immunol Methods. 2003;273(1–2):83–101.PubMedCrossRef Bernardini G et al. Analysis of the role of chemokines in angiogenesis. J Immunol Methods. 2003;273(1–2):83–101.PubMedCrossRef
39.
go back to reference Shichita T et al. Pivotal role of cerebral interleukin-17-producing gammadeltaT cells in the delayed phase of ischemic brain injury. Nat Med. 2009;15(8):946–50.PubMedCrossRef Shichita T et al. Pivotal role of cerebral interleukin-17-producing gammadeltaT cells in the delayed phase of ischemic brain injury. Nat Med. 2009;15(8):946–50.PubMedCrossRef
40.
41.
go back to reference Shih TM, McDonough Jr JH. Neurochemical mechanisms in soman-induced seizures. J Appl Toxicol. 1997;17(4):255–64.PubMedCrossRef Shih TM, McDonough Jr JH. Neurochemical mechanisms in soman-induced seizures. J Appl Toxicol. 1997;17(4):255–64.PubMedCrossRef
42.
go back to reference Koplovitz I, Skvorak JP. Electrocorticographic changes during generalized convulsive status epilepticus in soman intoxicated rats. Epilepsy Res. 1998;30(2):159–64.PubMedCrossRef Koplovitz I, Skvorak JP. Electrocorticographic changes during generalized convulsive status epilepticus in soman intoxicated rats. Epilepsy Res. 1998;30(2):159–64.PubMedCrossRef
43.
go back to reference Johnson EA et al. Cell-specific DNA fragmentation may be attenuated by a survivin-dependent mechanism after traumatic brain injury in rats. Exp Brain Res. 2005;167(1):17–26.PubMedCrossRef Johnson EA et al. Cell-specific DNA fragmentation may be attenuated by a survivin-dependent mechanism after traumatic brain injury in rats. Exp Brain Res. 2005;167(1):17–26.PubMedCrossRef
44.
go back to reference Paxinos G, Watson C. The Rat Brain in Stereotaxic Coordinates. San Diego: Elsevier Academic Press; 2005. p. 205. Paxinos G, Watson C. The Rat Brain in Stereotaxic Coordinates. San Diego: Elsevier Academic Press; 2005. p. 205.
45.
go back to reference Hedtjarn M et al. Interleukin-18 involvement in hypoxic-ischemic brain injury. J Neurosci. 2002;22(14):5910–9.PubMed Hedtjarn M et al. Interleukin-18 involvement in hypoxic-ischemic brain injury. J Neurosci. 2002;22(14):5910–9.PubMed
46.
go back to reference Yatsiv I et al. Elevated intracranial IL-18 in humans and mice after traumatic brain injury and evidence of neuroprotective effects of IL-18-binding protein after experimental closed head injury. J Cereb Blood Flow Metab. 2002;22(8):971–8.PubMedCrossRef Yatsiv I et al. Elevated intracranial IL-18 in humans and mice after traumatic brain injury and evidence of neuroprotective effects of IL-18-binding protein after experimental closed head injury. J Cereb Blood Flow Metab. 2002;22(8):971–8.PubMedCrossRef
47.
go back to reference Andoh T et al. Protective effect of IL-18 on kainate- and IL-1 beta-induced cerebellar ataxia in mice. J Immunol. 2008;180(4):2322–8.PubMedCrossRef Andoh T et al. Protective effect of IL-18 on kainate- and IL-1 beta-induced cerebellar ataxia in mice. J Immunol. 2008;180(4):2322–8.PubMedCrossRef
48.
49.
go back to reference Mori I et al. Impaired microglial activation in the brain of IL-18-gene-disrupted mice after neurovirulent influenza A virus infection. Virology. 2001;287(1):163–70.PubMedCrossRef Mori I et al. Impaired microglial activation in the brain of IL-18-gene-disrupted mice after neurovirulent influenza A virus infection. Virology. 2001;287(1):163–70.PubMedCrossRef
50.
go back to reference Bouchaud C et al. The role of nitric oxide in the neuropathology in soman intoxication. Brain Res. 1994;660(2):249–54.PubMedCrossRef Bouchaud C et al. The role of nitric oxide in the neuropathology in soman intoxication. Brain Res. 1994;660(2):249–54.PubMedCrossRef
51.
go back to reference Lallement G et al. Acute soman poisoning in primates neither pretreated nor receiving immediate therapy: value of gacyclidine (GK-11) in delayed medical support. Arch Toxicol. 1999;73(2):115–22.PubMedCrossRef Lallement G et al. Acute soman poisoning in primates neither pretreated nor receiving immediate therapy: value of gacyclidine (GK-11) in delayed medical support. Arch Toxicol. 1999;73(2):115–22.PubMedCrossRef
52.
go back to reference Lemercier G et al. Histological and histochemical changes in the central nervous system of the rat poisoned by an irreversible anticholinesterase organophosphorus compound. Acta Neuropathol. 1983;61(2):123–9.PubMedCrossRef Lemercier G et al. Histological and histochemical changes in the central nervous system of the rat poisoned by an irreversible anticholinesterase organophosphorus compound. Acta Neuropathol. 1983;61(2):123–9.PubMedCrossRef
53.
go back to reference Collombet JM et al. Soman poisoning induces delayed astrogliotic scar and angiogenesis in damaged mouse brain areas. Neurotoxicology. 2007;28(1):38–48.PubMedCrossRef Collombet JM et al. Soman poisoning induces delayed astrogliotic scar and angiogenesis in damaged mouse brain areas. Neurotoxicology. 2007;28(1):38–48.PubMedCrossRef
54.
go back to reference Jander S, Schroeter M, Stoll G. Interleukin-18 expression after focal ischemia of the rat brain: association with the late-stage inflammatory response. J Cereb Blood Flow Metab. 2002;22(1):62–70.PubMedCrossRef Jander S, Schroeter M, Stoll G. Interleukin-18 expression after focal ischemia of the rat brain: association with the late-stage inflammatory response. J Cereb Blood Flow Metab. 2002;22(1):62–70.PubMedCrossRef
Metadata
Title
Interleukin-18 expression increases in response to neurovascular damage following soman-induced status epilepticus in rats
Authors
Erik A. Johnson
Michelle A. Guignet
Thuy L. Dao
Tracey A. Hamilton
Robert K. Kan
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Journal of Inflammation / Issue 1/2015
Electronic ISSN: 1476-9255
DOI
https://doi.org/10.1186/s12950-015-0089-9

Other articles of this Issue 1/2015

Journal of Inflammation 1/2015 Go to the issue
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discuss last year's major advances in heart failure and cardiomyopathies.