Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2012

Open Access 01-12-2012 | Research

JNK signaling is the shared pathway linking neuroinflammation, blood–brain barrier disruption, and oligodendroglial apoptosis in the white matter injury of the immature brain

Authors: Lan-Wan Wang, Yi-Fang Tu, Chao-Ching Huang, Chien-Jung Ho

Published in: Journal of Neuroinflammation | Issue 1/2012

Login to get access

Abstract

Background

White matter injury is the major form of brain damage in very preterm infants. Selective white matter injury in the immature brain can be induced by lipopolysaccharide (LPS)-sensitized hypoxic-ischemia (HI) in the postpartum (P) day 2 rat pups whose brain maturation status is equivalent to that in preterm infants less than 30 weeks of gestation. Neuroinflammation, blood–brain barrier (BBB) damage and oligodendrocyte progenitor apoptosis may affect the susceptibility of LPS-sensitized HI in white matter injury. c-Jun N-terminal kinases (JNK) are important stress-responsive kinases in various forms of insults. We hypothesized that LPS-sensitized HI causes white matter injury through JNK activation-mediated neuroinflammation, BBB leakage and oligodendroglial apoptosis in the white matter of P2 rat pups.

Methods

P2 pups received LPS (0.05 mg/kg) or normal saline injection followed by 90-min HI. Immunohistochemistry and immunoblotting were used to determine microglia activation, TNF-α, BBB damage, cleaved caspase-3, JNK and phospho-JNK (p-JNK), myelin basic protein (MBP), and glial fibrillary acidic protein (GFAP) expression. Immunofluorescence was performed to determine the cellular distribution of p-JNK. Pharmacological and genetic approaches were used to inhibit JNK activity.

Results

P2 pups had selective white matter injury associated with upregulation of activated microglia, TNF-α, IgG extravasation and oligodendroglial progenitor apoptosis after LPS-sensitized HI. Immunohistochemical analyses showed early and sustained JNK activation in the white matter at 6 and 24 h post-insult. Immunofluorescence demonstrated upregulation of p-JNK in activated microglia, vascular endothelial cells and oligodendrocyte progenitors, and also showed perivascular aggregation of p-JNK-positive cells around the vessels 24 h post-insult. JNK inhibition by AS601245 or by antisense oligodeoxynucleotides (ODN) significantly reduced microglial activation, TNF-α immunoreactivity, IgG extravasation, and cleaved caspase-3 in the endothelial cells and oligodendrocyte progenitors, and also attenuated perivascular aggregation of p-JNK-positive cells 24 h post-insult. The AS601245 or JNK antisense ODN group had significantly increased MBP and decreased GFAP expression in the white matter on P11 than the vehicle or scrambled ODN group.

Conclusions

LPS-sensitized HI causes white matter injury through JNK activation-mediated upregulation of neuroinflammation, BBB leakage and oligodendrocyte progenitor apoptosis in the immature brain.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Volpe JJ: Neurology of the Newborn. 5th edition. W.B. Saunders Co, Philadelphia; 2008. Volpe JJ: Neurology of the Newborn. 5th edition. W.B. Saunders Co, Philadelphia; 2008.
3.
go back to reference Vincer MJ, Allen AC, Joseph KS, Stinson DA, Scot H, Wood E: Increasing prevalence of cerebral palsy among very preterm infants: a population-based study. Pediatrics 2006, 118:e1621-e1626.CrossRefPubMed Vincer MJ, Allen AC, Joseph KS, Stinson DA, Scot H, Wood E: Increasing prevalence of cerebral palsy among very preterm infants: a population-based study. Pediatrics 2006, 118:e1621-e1626.CrossRefPubMed
4.
go back to reference McElrath TF, Allred EN, Boggess KA, Kuban K, O’Shea TM, Paneth N, Leviton A: Maternal antenatal complications and the risk of neonatal cerebral white matter damage and later cerebral palsy in children born at an extremely low gestational age. Am J Epidemiol 2009, 170:819–828.CrossRefPubMedPubMedCentral McElrath TF, Allred EN, Boggess KA, Kuban K, O’Shea TM, Paneth N, Leviton A: Maternal antenatal complications and the risk of neonatal cerebral white matter damage and later cerebral palsy in children born at an extremely low gestational age. Am J Epidemiol 2009, 170:819–828.CrossRefPubMedPubMedCentral
5.
go back to reference Stoll BJ, Hansen NI, Adams-Chapman I, Fanaroff AA, Hintz SR, Vohr B, Higgins RD: Neurodevelopmental and growth impairment among extremely low-birth-weight infants with neonatal infection. JAMA 2004, 292:2357–2365.CrossRefPubMed Stoll BJ, Hansen NI, Adams-Chapman I, Fanaroff AA, Hintz SR, Vohr B, Higgins RD: Neurodevelopmental and growth impairment among extremely low-birth-weight infants with neonatal infection. JAMA 2004, 292:2357–2365.CrossRefPubMed
6.
go back to reference Yanowitz TD, Jordan JA, Gilmour CH, Towbin R, Bowen A, Roberts JM, Brozanski BS: Hemodynamic disturbances in premature infants born after chorioamnionitis: association with cord blood cytokine concentrations. Pediatr Res 2002, 51:310–316.CrossRefPubMed Yanowitz TD, Jordan JA, Gilmour CH, Towbin R, Bowen A, Roberts JM, Brozanski BS: Hemodynamic disturbances in premature infants born after chorioamnionitis: association with cord blood cytokine concentrations. Pediatr Res 2002, 51:310–316.CrossRefPubMed
7.
go back to reference Tsuji M, Saul JP, Plessis A, Eichenwald E, Sobh J, Crocker R, Volpe JJ: Cerebral intravascular oxygenation correlates with mean arterial pressure in critically ill premature infants. Pediatrics 2000, 106:625–632.CrossRefPubMed Tsuji M, Saul JP, Plessis A, Eichenwald E, Sobh J, Crocker R, Volpe JJ: Cerebral intravascular oxygenation correlates with mean arterial pressure in critically ill premature infants. Pediatrics 2000, 106:625–632.CrossRefPubMed
8.
go back to reference Kaukola T, Herva R, Perhomma M, Paakko E, Kingsmore S, Vainionpaa L, Hallman M: Population cohort associating chorioamnionitis, cord inflammatory cytokines and neurological outcome in very preterm, extremely low birth weight infants. Pediatr Res 2006, 59:478–483.CrossRefPubMed Kaukola T, Herva R, Perhomma M, Paakko E, Kingsmore S, Vainionpaa L, Hallman M: Population cohort associating chorioamnionitis, cord inflammatory cytokines and neurological outcome in very preterm, extremely low birth weight infants. Pediatr Res 2006, 59:478–483.CrossRefPubMed
9.
go back to reference Lehnardt S, Massillon L, Follett P, Jensen FE, Ratan R, Rosenberg PA, Volpe JJ, Vartanian T: Activation of innate immunity in the CNS triggers neurodegeneration through a Toll-like receptor 4-dependent pathway. PNAS 2003, 100:8514–8519.CrossRefPubMedPubMedCentral Lehnardt S, Massillon L, Follett P, Jensen FE, Ratan R, Rosenberg PA, Volpe JJ, Vartanian T: Activation of innate immunity in the CNS triggers neurodegeneration through a Toll-like receptor 4-dependent pathway. PNAS 2003, 100:8514–8519.CrossRefPubMedPubMedCentral
10.
go back to reference Eklind S, Mallard C, Leverin AL, Gilland E, Blomgren K, Mattsby-Baltzer I, Hagberg H: Bacterial endotoxin sensitizes the immature brain to hypoxic-ischemic injury. Eur J Neurosci 2001, 13:1101–1106.CrossRefPubMed Eklind S, Mallard C, Leverin AL, Gilland E, Blomgren K, Mattsby-Baltzer I, Hagberg H: Bacterial endotoxin sensitizes the immature brain to hypoxic-ischemic injury. Eur J Neurosci 2001, 13:1101–1106.CrossRefPubMed
11.
go back to reference Wang X, Svedin P, Nie C, Lapatto R, Zhu C, Gustavsson M, Sandberg M, Karlsson JO, Romero R, Hagberg H, Mallard C: N-acetylcysteine reduces lipopolysaccharide-sensitized hypoxic-ischemic brain injury. Ann Neurol 2007, 61:263–271.CrossRefPubMed Wang X, Svedin P, Nie C, Lapatto R, Zhu C, Gustavsson M, Sandberg M, Karlsson JO, Romero R, Hagberg H, Mallard C: N-acetylcysteine reduces lipopolysaccharide-sensitized hypoxic-ischemic brain injury. Ann Neurol 2007, 61:263–271.CrossRefPubMed
12.
go back to reference Back SA, Luo NL, Borenstein NS, Levin JM, Volpe JJ, Kinney HC: Late oligodendrocyte progenitors coincide with the developmental window of vulnerability for human perinatal white matter injury. J Neurosci 2001, 21:1302–1312.PubMed Back SA, Luo NL, Borenstein NS, Levin JM, Volpe JJ, Kinney HC: Late oligodendrocyte progenitors coincide with the developmental window of vulnerability for human perinatal white matter injury. J Neurosci 2001, 21:1302–1312.PubMed
13.
go back to reference Craig A, Luo NL, Beardsley DJ, Wingate-Pearse N, Walker DW, Hohimer AR, Back SA: Quantitative analysis of perinatal rodent oligodendrocyte lineage progression and its correlation with human. Exp Neurol 2003, 181:231–240.CrossRefPubMed Craig A, Luo NL, Beardsley DJ, Wingate-Pearse N, Walker DW, Hohimer AR, Back SA: Quantitative analysis of perinatal rodent oligodendrocyte lineage progression and its correlation with human. Exp Neurol 2003, 181:231–240.CrossRefPubMed
14.
go back to reference Wang LW, Chang YC, Lin CY, Hong JS, Huang CC: Low-dose lipopolysaccharide selectively sensitizes hypoxia-ischemia-induced white matter injury in the immature brain. Pediatr Res 2010, 68:41–47.CrossRefPubMedPubMedCentral Wang LW, Chang YC, Lin CY, Hong JS, Huang CC: Low-dose lipopolysaccharide selectively sensitizes hypoxia-ischemia-induced white matter injury in the immature brain. Pediatr Res 2010, 68:41–47.CrossRefPubMedPubMedCentral
16.
go back to reference Chew LJ, Takanohashi A, Bell M: Microglia and inflammation: impact on developmental brain injuries. Ment Retard Dev Disabil Res Rev 2006, 12:105–112.CrossRefPubMed Chew LJ, Takanohashi A, Bell M: Microglia and inflammation: impact on developmental brain injuries. Ment Retard Dev Disabil Res Rev 2006, 12:105–112.CrossRefPubMed
17.
go back to reference Muramatsu K, Fukuda A, Togari H, Wada Y, Nishino H: Vulnerability to cerebral hypoxic-ischemic insult in neonatal but not in adult rats is in parallel with disruption of the blood–brain barrier. Stroke 1997, 28:2281–2288.CrossRefPubMed Muramatsu K, Fukuda A, Togari H, Wada Y, Nishino H: Vulnerability to cerebral hypoxic-ischemic insult in neonatal but not in adult rats is in parallel with disruption of the blood–brain barrier. Stroke 1997, 28:2281–2288.CrossRefPubMed
18.
go back to reference Tu YF, Tsai YS, Wang LW, Wu HC, Huang CC, Ho CJ: Overweight worsens apoptosis, neuroinflammation and blood–brain barrier damage after hypoxic ischemia in neonatal brain through JNK hyperactivation. J Neuroinflammation 2011, 8:40–54.CrossRefPubMedPubMedCentral Tu YF, Tsai YS, Wang LW, Wu HC, Huang CC, Ho CJ: Overweight worsens apoptosis, neuroinflammation and blood–brain barrier damage after hypoxic ischemia in neonatal brain through JNK hyperactivation. J Neuroinflammation 2011, 8:40–54.CrossRefPubMedPubMedCentral
19.
go back to reference Tu YF, Lu PJ, Huang CC: Moderate dietary restriction reduces p53-mediated neurovascular damage and microglia activation after hypoxic ischemia in neonatal brain. Stroke 2012, 43:491–498.CrossRefPubMed Tu YF, Lu PJ, Huang CC: Moderate dietary restriction reduces p53-mediated neurovascular damage and microglia activation after hypoxic ischemia in neonatal brain. Stroke 2012, 43:491–498.CrossRefPubMed
21.
go back to reference Dammann O, Durums S, Leviton A: Do white cells matter in white matter damage? Trends Neurosci 2001, 24:320–324.CrossRefPubMed Dammann O, Durums S, Leviton A: Do white cells matter in white matter damage? Trends Neurosci 2001, 24:320–324.CrossRefPubMed
22.
go back to reference Manning AM, Davis RJ: Target JNK for therapeutic benefit: from Junk to gold? Nat Rev Drug Discov 2003, 2:554–565.CrossRefPubMed Manning AM, Davis RJ: Target JNK for therapeutic benefit: from Junk to gold? Nat Rev Drug Discov 2003, 2:554–565.CrossRefPubMed
23.
go back to reference Cao J, Semenova MM, Solovyan VT, Han J, Coffey ET, Courtney MJ: Distinct requirements for p38alpha and c-Jun N-terminal kinase stress-activated protein kinases in different forms of apoptotic neuronal death. J Biol Chem 2004, 279:35903–35913.CrossRefPubMed Cao J, Semenova MM, Solovyan VT, Han J, Coffey ET, Courtney MJ: Distinct requirements for p38alpha and c-Jun N-terminal kinase stress-activated protein kinases in different forms of apoptotic neuronal death. J Biol Chem 2004, 279:35903–35913.CrossRefPubMed
24.
25.
go back to reference Gao Y, Signore AP, Yin W, Cao G, Yin XM, Sun F, Luo Y, Graham SH, Chen J: Neuroprotection against focal ischemic brain injury by inhibition of c-Jun N-terminal kinase and attenuation of the mitochondrial apoptosis-signaling pathway. J Cereb Blood Flow Metab 2005, 25:694–712.CrossRefPubMed Gao Y, Signore AP, Yin W, Cao G, Yin XM, Sun F, Luo Y, Graham SH, Chen J: Neuroprotection against focal ischemic brain injury by inhibition of c-Jun N-terminal kinase and attenuation of the mitochondrial apoptosis-signaling pathway. J Cereb Blood Flow Metab 2005, 25:694–712.CrossRefPubMed
26.
go back to reference Kuan CY, Whitmarsh AJ, Yang DD, Liao G, Schloemer AJ, Dong C, Bao J, Banasiak KJ, Haddad GG, Flavell RA, Davis RJ, Rakic P: A critical role of neural-specific JNK3 for ischemic apoptosis. PNAS 2003, 100:15184–15189.CrossRefPubMedPubMedCentral Kuan CY, Whitmarsh AJ, Yang DD, Liao G, Schloemer AJ, Dong C, Bao J, Banasiak KJ, Haddad GG, Flavell RA, Davis RJ, Rakic P: A critical role of neural-specific JNK3 for ischemic apoptosis. PNAS 2003, 100:15184–15189.CrossRefPubMedPubMedCentral
27.
go back to reference Guan QH, Pei DS, Zong YY, Xu TL, Zhang GY: Neuroprotection against ischemic brain injury by a small peptide inhibitor of c-Jun N-terminal kinase via nuclear and non-nuclear pathways. Neurosci 2006, 139:609–627.CrossRef Guan QH, Pei DS, Zong YY, Xu TL, Zhang GY: Neuroprotection against ischemic brain injury by a small peptide inhibitor of c-Jun N-terminal kinase via nuclear and non-nuclear pathways. Neurosci 2006, 139:609–627.CrossRef
28.
go back to reference Guan QH, Pei DS, Liu XM, Wang XT, Xu TL, Zhang GY: Neuroprotection against ischemic brain injury by SP600125 via suppressing the extrinsic and intrinsic pathways of apoptosis. Brain Res 2006, 1092:36–46.CrossRefPubMed Guan QH, Pei DS, Liu XM, Wang XT, Xu TL, Zhang GY: Neuroprotection against ischemic brain injury by SP600125 via suppressing the extrinsic and intrinsic pathways of apoptosis. Brain Res 2006, 1092:36–46.CrossRefPubMed
29.
go back to reference Uesugi M, Nakajima K, Tohyama Y, Kohsaka S, Kurihara T: Nonparticipation of nuclear factor kappa B in the signaling cascade of c-Jun N-terminal kinases and p38 mitogen activated protein kinase-dependent tumor necrosis factor-alpha induction in lipopolysaccharide-stimulated microglia. Brain Res 2006, 1073:48–59.CrossRefPubMed Uesugi M, Nakajima K, Tohyama Y, Kohsaka S, Kurihara T: Nonparticipation of nuclear factor kappa B in the signaling cascade of c-Jun N-terminal kinases and p38 mitogen activated protein kinase-dependent tumor necrosis factor-alpha induction in lipopolysaccharide-stimulated microglia. Brain Res 2006, 1073:48–59.CrossRefPubMed
30.
go back to reference Deng YY, Lu J, Sivakumar V, Ling EA, Kaur C: Amoeboid microglia in the periventricular white matter induce oligodendrocyte damage through expression of proinflammatory cytokines via MAP kinase signaling pathway in hypoxic neonatal rats. Brain Pathol 2008, 18:387–400.CrossRefPubMed Deng YY, Lu J, Sivakumar V, Ling EA, Kaur C: Amoeboid microglia in the periventricular white matter induce oligodendrocyte damage through expression of proinflammatory cytokines via MAP kinase signaling pathway in hypoxic neonatal rats. Brain Pathol 2008, 18:387–400.CrossRefPubMed
31.
go back to reference Yatsusshige H, Ostrowski RP, Tsubokawa T, Colohan A, Zhang JH: Role of c-Jun N-terminal Kinase in early brain injury after subarachnoid hemorrhage. J Neurosci Res 2007, 85:1436–1448.CrossRef Yatsusshige H, Ostrowski RP, Tsubokawa T, Colohan A, Zhang JH: Role of c-Jun N-terminal Kinase in early brain injury after subarachnoid hemorrhage. J Neurosci Res 2007, 85:1436–1448.CrossRef
32.
go back to reference Karahashi H, Michelsen KS, Arditi M: Lipopolysaccharide-induced apoptosis in transformed bovine brain endothelial cells and human dermal microvessel endothelial cells: the role of JNK. J Immunol 2009, 182:7280–7286.CrossRefPubMedPubMedCentral Karahashi H, Michelsen KS, Arditi M: Lipopolysaccharide-induced apoptosis in transformed bovine brain endothelial cells and human dermal microvessel endothelial cells: the role of JNK. J Immunol 2009, 182:7280–7286.CrossRefPubMedPubMedCentral
33.
go back to reference Pirianov G, Jesurasa A, Mehmet H: Developmentally regulated changes in c-Jun N-terminal kinase signaling determine the apoptotic response of oligodendrocyte lineage cells. Cell Death Differ 2006, 13:531–533.CrossRefPubMed Pirianov G, Jesurasa A, Mehmet H: Developmentally regulated changes in c-Jun N-terminal kinase signaling determine the apoptotic response of oligodendrocyte lineage cells. Cell Death Differ 2006, 13:531–533.CrossRefPubMed
34.
go back to reference Repici M, Centeno C, Tomasi S, Forloni G, Bonny C, Vercelli A, Borsello T: Time-course of c-Jun N-terminal kinase activation after cerebral ischemia and effect of D-JNKI1 on c-Jun and caspase-3 activation. Neuroscience 2007, 150:40–49.CrossRefPubMed Repici M, Centeno C, Tomasi S, Forloni G, Bonny C, Vercelli A, Borsello T: Time-course of c-Jun N-terminal kinase activation after cerebral ischemia and effect of D-JNKI1 on c-Jun and caspase-3 activation. Neuroscience 2007, 150:40–49.CrossRefPubMed
35.
go back to reference Herdegen T, Claret FX, Kallunki T, Martin-Villalba A, Winter C, Hunter T, Karin M: Lasting N-terminal phosphorylation of c-Jun and activation of c-Jun N-terminal kinases after neuronal injury. J Neurosci 1998, 18:5124–5135.PubMed Herdegen T, Claret FX, Kallunki T, Martin-Villalba A, Winter C, Hunter T, Karin M: Lasting N-terminal phosphorylation of c-Jun and activation of c-Jun N-terminal kinases after neuronal injury. J Neurosci 1998, 18:5124–5135.PubMed
36.
go back to reference Chang YC, Huang CC, Hung PL, Huang HM: Rolipram, a phosphodiesterase type IV inhibitor, exacerbates periventricular white matter lesions in rat pups. Pediatr Res 2008, 64:234–239.CrossRefPubMed Chang YC, Huang CC, Hung PL, Huang HM: Rolipram, a phosphodiesterase type IV inhibitor, exacerbates periventricular white matter lesions in rat pups. Pediatr Res 2008, 64:234–239.CrossRefPubMed
37.
go back to reference Carboni S, Hiver A, Szyndralewiez C, Gaillard P, Gotteland JP, Vitte PA: AS601245 (1,3-Benzothiazol-2-yl (2-{[2-(3-pyridinyl) ethyl] amino}-4 pyrimidinyl) Acetonitrile): a c-Jun NH2-terminal protein kinase inhibitor with neuroprotective properties. J Pharmacol Exp Ther 2004, 310:25–32.CrossRefPubMed Carboni S, Hiver A, Szyndralewiez C, Gaillard P, Gotteland JP, Vitte PA: AS601245 (1,3-Benzothiazol-2-yl (2-{[2-(3-pyridinyl) ethyl] amino}-4 pyrimidinyl) Acetonitrile): a c-Jun NH2-terminal protein kinase inhibitor with neuroprotective properties. J Pharmacol Exp Ther 2004, 310:25–32.CrossRefPubMed
38.
go back to reference Lin HY, Wu CL, Huang CC: The Akt-endothelial nitric oxide synthase pathway in lipopolysaccharide preconditioning-induced hypoxic-ischemic tolerance in the neonatal rat brain. Stroke 2010, 41:1543–1551.CrossRefPubMed Lin HY, Wu CL, Huang CC: The Akt-endothelial nitric oxide synthase pathway in lipopolysaccharide preconditioning-induced hypoxic-ischemic tolerance in the neonatal rat brain. Stroke 2010, 41:1543–1551.CrossRefPubMed
39.
go back to reference Paxinos G, Watson C: The rat brain in stereotaxic coordinates. Academic, New York; 1986. Paxinos G, Watson C: The rat brain in stereotaxic coordinates. Academic, New York; 1986.
40.
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, 27:1511–1518.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, 27:1511–1518.CrossRefPubMed
41.
go back to reference Lin HY, Huang CC, Chang KF: Lipopolysaccharide preconditioning reduces neuroinflammation against hypoxic ischemia and provides long-term outcome of neuroprotection in neonatal rat. Pediatr Res 2009, 66:254–259.CrossRefPubMed Lin HY, Huang CC, Chang KF: Lipopolysaccharide preconditioning reduces neuroinflammation against hypoxic ischemia and provides long-term outcome of neuroprotection in neonatal rat. Pediatr Res 2009, 66:254–259.CrossRefPubMed
42.
go back to reference Eklind S, Hagberg H, Wang X, Savman K, Leverin AL, Hedtjarn M, Mallard C: Effect of lipopolysaccharide on global gene expression in the immature rat brain. Pediatr Res 2006, 60:161–168.CrossRefPubMed Eklind S, Hagberg H, Wang X, Savman K, Leverin AL, Hedtjarn M, Mallard C: Effect of lipopolysaccharide on global gene expression in the immature rat brain. Pediatr Res 2006, 60:161–168.CrossRefPubMed
43.
go back to reference Wang X, Stridh L, Li W, Dean J, Elmgren A, Gan L, Eriksson K, Hagberg H, Mallard C: Lipopolysaccharide sensitizes neonatal hypoxic-ischemic brain injury in a MyD88-dependent mammer. J Immunol 2009, 183:7471–7477.CrossRefPubMed Wang X, Stridh L, Li W, Dean J, Elmgren A, Gan L, Eriksson K, Hagberg H, Mallard C: Lipopolysaccharide sensitizes neonatal hypoxic-ischemic brain injury in a MyD88-dependent mammer. J Immunol 2009, 183:7471–7477.CrossRefPubMed
44.
go back to reference Fan LW, Mitchell HJ, Tien LT, Zheng B, Pang Y, Rhodes PG, Cai Z: α-phenyl-n-tert-butyl-nitrone reduces lipopolysaccharide-induced white matter injury in the neonatal rat brain. Dev Neurobiol 2008, 68:365–378.CrossRefPubMed Fan LW, Mitchell HJ, Tien LT, Zheng B, Pang Y, Rhodes PG, Cai Z: α-phenyl-n-tert-butyl-nitrone reduces lipopolysaccharide-induced white matter injury in the neonatal rat brain. Dev Neurobiol 2008, 68:365–378.CrossRefPubMed
45.
go back to reference Zacchigna S, Lambrechts D, Carmeliet P: Neurovascular signaling defects in neurodegeneration. Nat Rev Neurosci 2008, 9:169–181.CrossRefPubMed Zacchigna S, Lambrechts D, Carmeliet P: Neurovascular signaling defects in neurodegeneration. Nat Rev Neurosci 2008, 9:169–181.CrossRefPubMed
46.
go back to reference Back SA, Volpe JJ: Cellular and molecular pathogenesis of periventricular white matter injury. Ment Retard Dev Disabil Res Rev 1997, 3:96–107.CrossRef Back SA, Volpe JJ: Cellular and molecular pathogenesis of periventricular white matter injury. Ment Retard Dev Disabil Res Rev 1997, 3:96–107.CrossRef
47.
go back to reference Kuno R, Wang J, Kawanokuchi J, Takeuchi H, Mizuno T, Suzumura A: Autocrine activation of microglia by tumor necrosis factor-α. J Neuroimmunol 2005, 162:89–96.CrossRefPubMed Kuno R, Wang J, Kawanokuchi J, Takeuchi H, Mizuno T, Suzumura A: Autocrine activation of microglia by tumor necrosis factor-α. J Neuroimmunol 2005, 162:89–96.CrossRefPubMed
48.
go back to reference Baud V, Karin M: Signal transduction by tumor necrosis factor and its relatives. Trends Cell Biol 2001, 9:372–377.CrossRef Baud V, Karin M: Signal transduction by tumor necrosis factor and its relatives. Trends Cell Biol 2001, 9:372–377.CrossRef
49.
go back to reference Remmers M, Schmidt-Kastner R, Belayev L, Lin B, Busto R, Ginsberg MD: Protein extravasation and cellular uptake after high-dose human-albumin treatment of transient focal cerebral ischemia in rats. Brain Res 1999, 827:237–242.CrossRefPubMed Remmers M, Schmidt-Kastner R, Belayev L, Lin B, Busto R, Ginsberg MD: Protein extravasation and cellular uptake after high-dose human-albumin treatment of transient focal cerebral ischemia in rats. Brain Res 1999, 827:237–242.CrossRefPubMed
50.
go back to reference Del Bigio MR, Deck JHN, Davidson GS: Glial swelling with eosinophilia in human post-mortem brains: a change indicative of plasma extravasation. Acta Neuropathol 2000, 100:688–694.CrossRefPubMed Del Bigio MR, Deck JHN, Davidson GS: Glial swelling with eosinophilia in human post-mortem brains: a change indicative of plasma extravasation. Acta Neuropathol 2000, 100:688–694.CrossRefPubMed
51.
go back to reference Jensen MB, Finsen B, Zimmer J: Morphological and immunophenotypic microglial changes in the denervated fascia dentata of adult rats: Correlation with blood–brain barrier damage and astroglial reactions. Exp Neurol 1997, 143:103–116.CrossRefPubMed Jensen MB, Finsen B, Zimmer J: Morphological and immunophenotypic microglial changes in the denervated fascia dentata of adult rats: Correlation with blood–brain barrier damage and astroglial reactions. Exp Neurol 1997, 143:103–116.CrossRefPubMed
53.
go back to reference Lucas R, Garcia I, Donati YRA, Hribar M, Mandriota SJ, Giroud C, Buurman WA, Fransen L, Suter PM, Nunez G, Pepper MS, Grau GE: Both TNF receptors are required for direct TNF-mediated cytotoxicity in microvascular endothelial cells. Eur J Immunol 1998, 28:3577–3586.CrossRefPubMed Lucas R, Garcia I, Donati YRA, Hribar M, Mandriota SJ, Giroud C, Buurman WA, Fransen L, Suter PM, Nunez G, Pepper MS, Grau GE: Both TNF receptors are required for direct TNF-mediated cytotoxicity in microvascular endothelial cells. Eur J Immunol 1998, 28:3577–3586.CrossRefPubMed
54.
go back to reference De Boer AG, Breimer DD: Cytokines and blood–brain barrier permeability. Prog Brain Res 1998, 115:425–451.CrossRefPubMed De Boer AG, Breimer DD: Cytokines and blood–brain barrier permeability. Prog Brain Res 1998, 115:425–451.CrossRefPubMed
55.
go back to reference McColl BW, Rothwell NJ, Allan SM: Systemic inflammation alters the kinetics of cerebrovascular tight junction disruption after experimental stroke in mice. J Neurosci 2008, 28:9451–9462.CrossRefPubMed McColl BW, Rothwell NJ, Allan SM: Systemic inflammation alters the kinetics of cerebrovascular tight junction disruption after experimental stroke in mice. J Neurosci 2008, 28:9451–9462.CrossRefPubMed
56.
go back to reference D’Mello C, Le T, Swain MG: Cerebral microglia recruit monocytes into the brain in response to tumor necrosis factor-α signaling during peripheral organ inflammation. J Neurosci 2009, 29:2089–2102.CrossRefPubMed D’Mello C, Le T, Swain MG: Cerebral microglia recruit monocytes into the brain in response to tumor necrosis factor-α signaling during peripheral organ inflammation. J Neurosci 2009, 29:2089–2102.CrossRefPubMed
57.
go back to reference Volpe JJ: Systemic inflammation, oligodendroglial maturation and encephalopathy of prematurity. Ann Neurol 2011, 70:525–529.CrossRefPubMed Volpe JJ: Systemic inflammation, oligodendroglial maturation and encephalopathy of prematurity. Ann Neurol 2011, 70:525–529.CrossRefPubMed
58.
go back to reference Baud O, Li J, Zhang Y, Neve RL, Volpe JJ, Rosenberg PA: Nitric oxide-induced cell death in developing oligodendrocytes is associated with mitochondrial dysfunction and apoptosis-inducing factor translocation. Eur J Neurosci 2004, 20:1713–1726.CrossRefPubMed Baud O, Li J, Zhang Y, Neve RL, Volpe JJ, Rosenberg PA: Nitric oxide-induced cell death in developing oligodendrocytes is associated with mitochondrial dysfunction and apoptosis-inducing factor translocation. Eur J Neurosci 2004, 20:1713–1726.CrossRefPubMed
59.
go back to reference Li J, Baud O, Vartanian T, Volpe JJ, Rosenberg PA: Peroxynitrite generated by inducible nitric oxide synthase and NADPH oxidase mediates microglial toxicity to oligodendrocytes. Proc Natl Acad Sci USA 2005, 102:9936–9941.CrossRefPubMedPubMedCentral Li J, Baud O, Vartanian T, Volpe JJ, Rosenberg PA: Peroxynitrite generated by inducible nitric oxide synthase and NADPH oxidase mediates microglial toxicity to oligodendrocytes. Proc Natl Acad Sci USA 2005, 102:9936–9941.CrossRefPubMedPubMedCentral
60.
go back to reference Haynes RL, Folkerth RD, Keefe RJ, Sung I, Swzeda LI, Rosenberg PA, Volpe JJ, Kinney HC: Nitrosative and oxidative injury to premyelinating oligodendrocytes in periventricular leukomalacia. J Neuropathol Exp Neurol 2003, 62:441–450.CrossRefPubMed Haynes RL, Folkerth RD, Keefe RJ, Sung I, Swzeda LI, Rosenberg PA, Volpe JJ, Kinney HC: Nitrosative and oxidative injury to premyelinating oligodendrocytes in periventricular leukomalacia. J Neuropathol Exp Neurol 2003, 62:441–450.CrossRefPubMed
61.
go back to reference Back SA, Luo NL, Mallinson RA, O’Malley JP, Wallen LD, Frei B, Morrow JD, Petito CK, Roberts CT, Murdoch GH, Montine TJ: Selective vulnerability of preterm white matter to oxidative damage defined by F2-isoprostanes. Ann Neurol 2005, 58:108–120.CrossRefPubMed Back SA, Luo NL, Mallinson RA, O’Malley JP, Wallen LD, Frei B, Morrow JD, Petito CK, Roberts CT, Murdoch GH, Montine TJ: Selective vulnerability of preterm white matter to oxidative damage defined by F2-isoprostanes. Ann Neurol 2005, 58:108–120.CrossRefPubMed
62.
go back to reference Kamata H, Honda S, Maeda S, Chang L, Hirata H, Karin M: Reactive oxygen species promote TNF-α-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases. Cell 2005, 120:649–661.CrossRefPubMed Kamata H, Honda S, Maeda S, Chang L, Hirata H, Karin M: Reactive oxygen species promote TNF-α-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases. Cell 2005, 120:649–661.CrossRefPubMed
63.
go back to reference Shen HM, Liu ZG: JNK signaling is a key modulator in cell death mediated by reactive oxygen and nitrogen species. Free Radical Biol Med 2006, 40:928–939.CrossRef Shen HM, Liu ZG: JNK signaling is a key modulator in cell death mediated by reactive oxygen and nitrogen species. Free Radical Biol Med 2006, 40:928–939.CrossRef
64.
go back to reference Liu HN, Giasson BI, Mushynski WE, Almazan G: AMPA receptor-mediated toxicity in oligodendrocyte progenitors involves free radical generation and activation of JNK, calpain and caspase 3. J Neurochem 2002, 82:398–409.CrossRefPubMed Liu HN, Giasson BI, Mushynski WE, Almazan G: AMPA receptor-mediated toxicity in oligodendrocyte progenitors involves free radical generation and activation of JNK, calpain and caspase 3. J Neurochem 2002, 82:398–409.CrossRefPubMed
65.
go back to reference Borsello T, Clarke PGH, Hirt L, Vercelli A, Repici M, Schorderet DF, Bogousslavsky J, Bonny C: A peptide inhibitor of c-Jun N-terminal kinase protects against excitotoxicity and cerebral ischemia. Nat Med 2003, 9:1180–1186.CrossRefPubMed Borsello T, Clarke PGH, Hirt L, Vercelli A, Repici M, Schorderet DF, Bogousslavsky J, Bonny C: A peptide inhibitor of c-Jun N-terminal kinase protects against excitotoxicity and cerebral ischemia. Nat Med 2003, 9:1180–1186.CrossRefPubMed
66.
go back to reference Pirianov G, Brywe K, Mallard C, Edwards AD, Flavell RA, Hagberg H, Mehmet H: Deletion of the c-Jun N-terminal kinase 3 gene protects neonatal mice against cerebral hypoxic-ischemic injury. J Cereb Blood Flow Metab 2007, 27:1022–1032.PubMed Pirianov G, Brywe K, Mallard C, Edwards AD, Flavell RA, Hagberg H, Mehmet H: Deletion of the c-Jun N-terminal kinase 3 gene protects neonatal mice against cerebral hypoxic-ischemic injury. J Cereb Blood Flow Metab 2007, 27:1022–1032.PubMed
Metadata
Title
JNK signaling is the shared pathway linking neuroinflammation, blood–brain barrier disruption, and oligodendroglial apoptosis in the white matter injury of the immature brain
Authors
Lan-Wan Wang
Yi-Fang Tu
Chao-Ching Huang
Chien-Jung Ho
Publication date
01-12-2012
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2012
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/1742-2094-9-175

Other articles of this Issue 1/2012

Journal of Neuroinflammation 1/2012 Go to the issue