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

Open Access 01-12-2015 | Research

Neuronal self-injury mediated by IL-1β and MMP-9 in a cerebral palsy model of severe neonatal encephalopathy induced by immune activation plus hypoxia-ischemia

Authors: Alexandre Savard, Marie-Elsa Brochu, Mathilde Chevin, Clémence Guiraut, Djordje Grbic, Guillaume Sébire

Published in: Journal of Neuroinflammation | Issue 1/2015

Login to get access

Abstract

Background

Inflammation due to remote pathogen exposure combined to hypoxia/ischemia (HI) is one of the most common causes of neonatal encephalopathy affecting at-term or near-term human newborn, which will consequently develop cerebral palsy. Within term-equivalent rat brains exposed to systemic lipopolysaccharide (LPS) plus HI, it was previously showed that neurons produce IL-1β earlier than do glial cells, and that blocking IL-1 was neuroprotective. To further define the mechanisms whereby IL-1 exerts its neurotoxic effect, we hypothesize that IL-1β plays a pivotal role in a direct and/or indirect mechanistic loop of neuronal self-injury through matrix metalloproteinase (MMP)-9.

Methods

An established preclinical rat model of LPS+HI-induced neonatal encephalopathy was used. In situ hybridization, ELISA, and immunolabeling techniques were employed. Selective blocking compounds allowed addressing the respective roles of IL-1 and MMP-9.

Results

In LPS+HI-exposed forebrains, neuronal IL-1β was first detected in infarcted neocortical and striatal areas and later in glial cells of the adjacent white matter. Neuronal IL-1β played a key role: (i) in the early post-HI exacerbation of neuroinflammation and (ii) in generating both core and penumbral infarcted cerebral areas. Systemically administered IL-1 receptor antagonist (IL-1Ra) reached the brain and bound to the neocortical and deep gray neuronal membranes. Then, IL-1Ra down-regulated IL-1β mRNA and MMP-9 neuronal synthesis. Immediately post-HI, neuronal IL-1β up-regulated cytokine-induced neutrophil chemoattractant (CINC-1), monocyte chemoattractant protein-1 (MCP-1), and inducible nitric oxide synthase. MMP-9 would disrupt the blood–brain barrier, which, combined to CINC-1 up-regulation, would play a role in polymorphonuclear cell (PMN) infiltration into the LPS+HI-exposed brain. IL-1β blockade prevented PMN infiltration and oriented the phenotype of macrophagic/microglial cells towards anti-inflammatory and neurotrophic M2 profile. IL-1β increased the expression of activated caspase-3 and of receptor-interacting-protein (RIP)-3 within infarcted forebrain area. Such apoptotic and necroptotic pathway activations were prevented by IL-1Ra, as well as ensuing cerebral palsy-like brain damage and motor impairment.

Conclusions

This work uncovered a new paradigm of neuronal self-injury orchestrated by neuronal synthesis of IL-1β and MMP-9. In addition, it reinforced the translational neuroprotective potential of IL-1 blockers to alleviate human perinatal brain injuries.
Literature
1.
go back to reference Grether JK, Nelson KB. Maternal infection and cerebral palsy in infants of normal birth weight. JAMA. 1997;278:207–11.CrossRefPubMed Grether JK, Nelson KB. Maternal infection and cerebral palsy in infants of normal birth weight. JAMA. 1997;278:207–11.CrossRefPubMed
2.
go back to reference Wintermark P, Boyd T, Gregas MC, Labrecque M, Hansen A. Placental pathology in asphyxiated newborns meeting the criteria for therapeutic hypothermia. Am J Obstet Gynecol. 2010;203:579. e571–579.CrossRefPubMed Wintermark P, Boyd T, Gregas MC, Labrecque M, Hansen A. Placental pathology in asphyxiated newborns meeting the criteria for therapeutic hypothermia. Am J Obstet Gynecol. 2010;203:579. e571–579.CrossRefPubMed
3.
go back to reference Chau V, Poskitt KJ, McFadden DE, Bowen-Roberts T, Synnes A, Brant R, et al. Effect of chorioamnionitis on brain development and injury in premature newborns. Ann Neurol. 2009;66:155–64.CrossRefPubMed Chau V, Poskitt KJ, McFadden DE, Bowen-Roberts T, Synnes A, Brant R, et al. Effect of chorioamnionitis on brain development and injury in premature newborns. Ann Neurol. 2009;66:155–64.CrossRefPubMed
4.
go back to reference Patel SD, Pierce L, Ciardiello AJ, Vannucci SJ. Neonatal encephalopathy: pre-clinical studies in neuroprotection. Biochem Soc Trans. 2014;42:564–8.CrossRefPubMed Patel SD, Pierce L, Ciardiello AJ, Vannucci SJ. Neonatal encephalopathy: pre-clinical studies in neuroprotection. Biochem Soc Trans. 2014;42:564–8.CrossRefPubMed
5.
go back to reference Girard S, Kadhim H, Beaudet N, Sarret P, Sebire G. Developmental motor deficits induced by combined fetal exposure to lipopolysaccharide and early neonatal hypoxia/ischemia: a novel animal model for cerebral palsy in very premature infants. Neuroscience. 2009;158:673–82.CrossRefPubMed Girard S, Kadhim H, Beaudet N, Sarret P, Sebire G. Developmental motor deficits induced by combined fetal exposure to lipopolysaccharide and early neonatal hypoxia/ischemia: a novel animal model for cerebral palsy in very premature infants. Neuroscience. 2009;158:673–82.CrossRefPubMed
8.
go back to reference Aden U, Favrais G, Plaisant F, Winerdal M, Felderhoff-Mueser U, Lampa J, et al. Systemic inflammation sensitizes the neonatal brain to excitotoxicity through a pro-/anti-inflammatory imbalance: key role of TNFalpha pathway and protection by etanercept. Brain Behav Immun. 2010;24:747–58.CrossRefPubMed Aden U, Favrais G, Plaisant F, Winerdal M, Felderhoff-Mueser U, Lampa J, et al. Systemic inflammation sensitizes the neonatal brain to excitotoxicity through a pro-/anti-inflammatory imbalance: key role of TNFalpha pathway and protection by etanercept. Brain Behav Immun. 2010;24:747–58.CrossRefPubMed
9.
go back to reference Eklind S, Mallard C, Leverin AL, Gilland E, Blomgren K, Mattsby-Baltzer I, et al. Bacterial endotoxin sensitizes the immature brain to hypoxic–ischaemic injury. Eur J Neurosci. 2001;13:1101–6.CrossRefPubMed Eklind S, Mallard C, Leverin AL, Gilland E, Blomgren K, Mattsby-Baltzer I, et al. Bacterial endotoxin sensitizes the immature brain to hypoxic–ischaemic injury. Eur J Neurosci. 2001;13:1101–6.CrossRefPubMed
10.
go back to reference Coumans AB, Middelanis JS, Garnier Y, Vaihinger HM, Leib SL, Von Duering MU, et al. Intracisternal application of endotoxin enhances the susceptibility to subsequent hypoxic-ischemic brain damage in neonatal rats. Pediatr Res. 2003;53:770–5.CrossRefPubMed Coumans AB, Middelanis JS, Garnier Y, Vaihinger HM, Leib SL, Von Duering MU, et al. Intracisternal application of endotoxin enhances the susceptibility to subsequent hypoxic-ischemic brain damage in neonatal rats. Pediatr Res. 2003;53:770–5.CrossRefPubMed
11.
go back to reference Wang X, Stridh L, Li W, Dean J, Elmgren A, Gan L, et al. Lipopolysaccharide sensitizes neonatal hypoxic-ischemic brain injury in a MyD88-dependent manner. J Immunol. 2009;183:7471–7.CrossRefPubMed Wang X, Stridh L, Li W, Dean J, Elmgren A, Gan L, et al. Lipopolysaccharide sensitizes neonatal hypoxic-ischemic brain injury in a MyD88-dependent manner. J Immunol. 2009;183:7471–7.CrossRefPubMed
12.
go back to reference Larouche A, Roy M, Kadhim H, Tsanaclis AM, Fortin D, Sebire G. Neuronal injuries induced by perinatal hypoxic-ischemic insults are potentiated by prenatal exposure to lipopolysaccharide: animal model for perinatally acquired encephalopathy. Dev Neurosci. 2005;27:134–42.CrossRefPubMed Larouche A, Roy M, Kadhim H, Tsanaclis AM, Fortin D, Sebire G. Neuronal injuries induced by perinatal hypoxic-ischemic insults are potentiated by prenatal exposure to lipopolysaccharide: animal model for perinatally acquired encephalopathy. Dev Neurosci. 2005;27:134–42.CrossRefPubMed
13.
go back to reference Towfighi J, Mauger D, Vannucci RC, Vannucci SJ. Influence of age on the cerebral lesions in an immature rat model of cerebral hypoxia-ischemia: a light microscopic study. Brain Res Dev Brain Res. 1997;100:149–60.CrossRefPubMed Towfighi J, Mauger D, Vannucci RC, Vannucci SJ. Influence of age on the cerebral lesions in an immature rat model of cerebral hypoxia-ischemia: a light microscopic study. Brain Res Dev Brain Res. 1997;100:149–60.CrossRefPubMed
14.
go back to reference Brochu ME, Girard S, Lavoie K, Sebire G. Developmental regulation of the neuroinflammatory responses to LPS and/or hypoxia-ischemia between preterm and term neonates: an experimental study. J Neuroinflammation. 2011;8:55.CrossRefPubMedCentralPubMed Brochu ME, Girard S, Lavoie K, Sebire G. Developmental regulation of the neuroinflammatory responses to LPS and/or hypoxia-ischemia between preterm and term neonates: an experimental study. J Neuroinflammation. 2011;8:55.CrossRefPubMedCentralPubMed
15.
go back to reference Savard A, Lavoie K, Brochu ME, Grbic D, Lepage M, Gris D, et al. Involvement of neuronal IL-1beta in acquired brain lesions in a rat model of neonatal encephalopathy. J Neuroinflammation. 2013;10:110.CrossRefPubMedCentralPubMed Savard A, Lavoie K, Brochu ME, Grbic D, Lepage M, Gris D, et al. Involvement of neuronal IL-1beta in acquired brain lesions in a rat model of neonatal encephalopathy. J Neuroinflammation. 2013;10:110.CrossRefPubMedCentralPubMed
16.
go back to reference Relton JK, Martin D, Thompson RC, Russell DA. Peripheral administration of interleukin-1 receptor antagonist inhibits brain damage after focal cerebral ischemia in the rat. Exp Neurol. 1996;138:206–13.CrossRefPubMed Relton JK, Martin D, Thompson RC, Russell DA. Peripheral administration of interleukin-1 receptor antagonist inhibits brain damage after focal cerebral ischemia in the rat. Exp Neurol. 1996;138:206–13.CrossRefPubMed
17.
go back to reference Lalancette-Hebert M, Gowing G, Simard A, Weng YC, Kriz J. Selective ablation of proliferating microglial cells exacerbates ischemic injury in the brain. J Neurosci. 2007;27:2596–605.CrossRefPubMed Lalancette-Hebert M, Gowing G, Simard A, Weng YC, Kriz J. Selective ablation of proliferating microglial cells exacerbates ischemic injury in the brain. J Neurosci. 2007;27:2596–605.CrossRefPubMed
18.
go back to reference Borlongan CV, Cahill DW, Sanberg PR. Locomotor and passive avoidance deficits following occlusion of the middle cerebral artery. Physiol Behav. 1995;58:909–17.CrossRefPubMed Borlongan CV, Cahill DW, Sanberg PR. Locomotor and passive avoidance deficits following occlusion of the middle cerebral artery. Physiol Behav. 1995;58:909–17.CrossRefPubMed
19.
go back to reference Chu K, Kim M, Park KI, Jeong SW, Park HK, Jung KH, et al. Human neural stem cells improve sensorimotor deficits in the adult rat brain with experimental focal ischemia. Brain Res. 2004;1016:145–53.CrossRefPubMed Chu K, Kim M, Park KI, Jeong SW, Park HK, Jung KH, et al. Human neural stem cells improve sensorimotor deficits in the adult rat brain with experimental focal ischemia. Brain Res. 2004;1016:145–53.CrossRefPubMed
21.
go back to reference Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popovich PG. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci. 2009;29:13435–44.CrossRefPubMedCentralPubMed Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popovich PG. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci. 2009;29:13435–44.CrossRefPubMedCentralPubMed
23.
go back to reference Svedin P, Hagberg H, Savman K, Zhu C, Mallard C. Matrix metalloproteinase-9 gene knock-out protects the immature brain after cerebral hypoxia-ischemia. J Neurosci. 2007;27:1511–8.CrossRefPubMed Svedin P, Hagberg H, Savman K, Zhu C, Mallard C. Matrix metalloproteinase-9 gene knock-out protects the immature brain after cerebral hypoxia-ischemia. J Neurosci. 2007;27:1511–8.CrossRefPubMed
24.
go back to reference Jiang Z, Li C, Arrick DM, Yang S, Baluna AE, Sun H. Role of nitric oxide synthases in early blood–brain barrier disruption following transient focal cerebral ischemia. PLoS One. 2014;9, e93134.CrossRefPubMedCentralPubMed Jiang Z, Li C, Arrick DM, Yang S, Baluna AE, Sun H. Role of nitric oxide synthases in early blood–brain barrier disruption following transient focal cerebral ischemia. PLoS One. 2014;9, e93134.CrossRefPubMedCentralPubMed
25.
go back to reference Allan SM, Tyrrell PJ, Rothwell NJ. Interleukin-1 and neuronal injury. Nat Rev Immunol. 2005;5:629–40.CrossRefPubMed Allan SM, Tyrrell PJ, Rothwell NJ. Interleukin-1 and neuronal injury. Nat Rev Immunol. 2005;5:629–40.CrossRefPubMed
26.
go back to reference Tchelingerian JL, Le Saux F, Jacque C. Identification and topography of neuronal cell populations expressing TNF alpha and IL-1 alpha in response to hippocampal lesion. J Neurosci Res. 1996;43:99–106.CrossRefPubMed Tchelingerian JL, Le Saux F, Jacque C. Identification and topography of neuronal cell populations expressing TNF alpha and IL-1 alpha in response to hippocampal lesion. J Neurosci Res. 1996;43:99–106.CrossRefPubMed
27.
go back to reference Kadhim H, Tabarki B, De Prez C, Sebire G. Cytokine immunoreactivity in cortical and subcortical neurons in periventricular leukomalacia: are cytokines implicated in neuronal dysfunction in cerebral palsy? Acta Neuropathol. 2003;105:209–16.PubMed Kadhim H, Tabarki B, De Prez C, Sebire G. Cytokine immunoreactivity in cortical and subcortical neurons in periventricular leukomalacia: are cytokines implicated in neuronal dysfunction in cerebral palsy? Acta Neuropathol. 2003;105:209–16.PubMed
28.
go back to reference Sairanen TR, Lindsberg PJ, Brenner M, Siren AL. Global forebrain ischemia results in differential cellular expression of interleukin-1beta (IL-1beta) and its receptor at mRNA and protein level. J Cereb Blood Flow Metab. 1997;17:1107–20.CrossRefPubMed Sairanen TR, Lindsberg PJ, Brenner M, Siren AL. Global forebrain ischemia results in differential cellular expression of interleukin-1beta (IL-1beta) and its receptor at mRNA and protein level. J Cereb Blood Flow Metab. 1997;17:1107–20.CrossRefPubMed
29.
go back to reference Degos V, Peineau S, Nijboer C, Kaindl AM, Sigaut S, Favrais G, et al. G protein-coupled receptor kinase 2 and group I metabotropic glutamate receptors mediate inflammation-induced sensitization to excitotoxic neurodegeneration. Ann Neurol. 2013;73:667–78.CrossRefPubMed Degos V, Peineau S, Nijboer C, Kaindl AM, Sigaut S, Favrais G, et al. G protein-coupled receptor kinase 2 and group I metabotropic glutamate receptors mediate inflammation-induced sensitization to excitotoxic neurodegeneration. Ann Neurol. 2013;73:667–78.CrossRefPubMed
30.
go back to reference Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453–61.CrossRefPubMed Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453–61.CrossRefPubMed
32.
go back to reference Danjo S, Ishihara Y, Watanabe M, Nakamura Y, Itoh K. Pentylentetrazole-induced loss of blood–brain barrier integrity involves excess nitric oxide generation by neuronal nitric oxide synthase. Brain Res. 2013;1530:44–53.CrossRefPubMed Danjo S, Ishihara Y, Watanabe M, Nakamura Y, Itoh K. Pentylentetrazole-induced loss of blood–brain barrier integrity involves excess nitric oxide generation by neuronal nitric oxide synthase. Brain Res. 2013;1530:44–53.CrossRefPubMed
33.
go back to reference Kaiser WJ, Upton JW, Mocarski ES. Viral modulation of programmed necrosis. Curr Opin Virol. 2013;3:296–306.CrossRefPubMed Kaiser WJ, Upton JW, Mocarski ES. Viral modulation of programmed necrosis. Curr Opin Virol. 2013;3:296–306.CrossRefPubMed
34.
go back to reference Yager J, Towfighi J, Vannucci RC. Influence of mild hypothermia on hypoxic-ischemic brain damage in the immature rat. Pediatr Res. 1993;34:525–9.CrossRefPubMed Yager J, Towfighi J, Vannucci RC. Influence of mild hypothermia on hypoxic-ischemic brain damage in the immature rat. Pediatr Res. 1993;34:525–9.CrossRefPubMed
35.
go back to reference Azzopardi D, Strohm B, Marlow N, Brocklehurst P, Deierl A, Eddama O, et al. Effects of hypothermia for perinatal asphyxia on childhood outcomes. N Engl J Med. 2014;371:140–9.CrossRefPubMed Azzopardi D, Strohm B, Marlow N, Brocklehurst P, Deierl A, Eddama O, et al. Effects of hypothermia for perinatal asphyxia on childhood outcomes. N Engl J Med. 2014;371:140–9.CrossRefPubMed
Metadata
Title
Neuronal self-injury mediated by IL-1β and MMP-9 in a cerebral palsy model of severe neonatal encephalopathy induced by immune activation plus hypoxia-ischemia
Authors
Alexandre Savard
Marie-Elsa Brochu
Mathilde Chevin
Clémence Guiraut
Djordje Grbic
Guillaume Sébire
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2015
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-015-0330-8

Other articles of this Issue 1/2015

Journal of Neuroinflammation 1/2015 Go to the issue