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

Open Access 01-12-2015 | Review

New means to assess neonatal inflammatory brain injury

Authors: Chen Jin, Irene Londono, Carina Mallard, Gregory A. Lodygensky

Published in: Journal of Neuroinflammation | Issue 1/2015

Login to get access

Abstract

Preterm infants are especially vulnerable to infection-induced white matter injury, associated with cerebral palsy, cognitive and psychomotor impairment, and other adverse neurological outcomes. The etiology of such lesions is complex and multifactorial. Furthermore, timing and length of exposure to infection also influence neurodevelopmental outcomes. Different mechanisms have been posited to mediate the observed brain injury including microglial activation followed by subsequent release of pro-inflammatory species, glutamate-induced excitotoxicity, and vulnerability of developing oligodendrocytes to cerebral insults. The prevalence of such neurological impairments requires an urgent need for early detection and effective neuroprotective strategies. Accordingly, noninvasive methods of monitoring disease progression and therapy effectiveness are essential. While diagnostic tools using biomarkers from bodily fluids may provide useful information regarding potential risks of developing neurological diseases, the use of magnetic resonance imaging/spectroscopy has emerged as a promising candidate for such purpose. Various pharmacological agents have demonstrated protective effects in the immature brain in animal models; however, few studies have progressed to clinical trials with promising results.
Literature
1.
go back to reference Virchow R. Congenitale encephalitis und myelitis. Archiv f pathol Anat. 1867;38:129–38.CrossRef Virchow R. Congenitale encephalitis und myelitis. Archiv f pathol Anat. 1867;38:129–38.CrossRef
2.
3.
go back to reference Volpe JJ. Encephalopathy of prematurity includes neuronal abnormalities. Pediatrics. 2005;116:221–5.PubMedCrossRef Volpe JJ. Encephalopathy of prematurity includes neuronal abnormalities. Pediatrics. 2005;116:221–5.PubMedCrossRef
4.
go back to reference Volpe JJ. The encephalopathy of prematurity—brain injury and impaired brain development inextricably intertwined. Sem Pediatr Neurol. 2009;16:167–78.CrossRef Volpe JJ. The encephalopathy of prematurity—brain injury and impaired brain development inextricably intertwined. Sem Pediatr Neurol. 2009;16:167–78.CrossRef
5.
go back to reference Wu YW, Colford JM. Chorioamnionitis as a risk factor for cerebral palsy: a meta-analysis. JAMA Pediatr. 2000;284:1417–24.CrossRef Wu YW, Colford JM. Chorioamnionitis as a risk factor for cerebral palsy: a meta-analysis. JAMA Pediatr. 2000;284:1417–24.CrossRef
6.
go back to reference Costantine MM, How HY, Coppage K, Maxwell RA, Sibai BM. Does peripartum infection increase the incidence of cerebral palsy in extremely low birthweight infants? Am J Obstetr Gynecol. 2007;196:e6–8.CrossRef Costantine MM, How HY, Coppage K, Maxwell RA, Sibai BM. Does peripartum infection increase the incidence of cerebral palsy in extremely low birthweight infants? Am J Obstetr Gynecol. 2007;196:e6–8.CrossRef
7.
go back to reference Zanardo V, Vedovato S, Suppiej A, Trevisanuto D, Migliore M, Di Venosa B, et al. Histological inflammatory responses in the placenta and early neonatal brain injury. Pediatr Dev Pathol. 2008;11:350–4.PubMedCrossRef Zanardo V, Vedovato S, Suppiej A, Trevisanuto D, Migliore M, Di Venosa B, et al. Histological inflammatory responses in the placenta and early neonatal brain injury. Pediatr Dev Pathol. 2008;11:350–4.PubMedCrossRef
8.
go back to reference Hansen-Pupp I, Hallin A-L, Hellström-Westas L, Cilio C, Berg A-C, Stjernqvist K, et al. Inflammation at birth is associated with subnormal development in very preterm infants. Pediatr Res. 2008;64:183–8.PubMedCrossRef Hansen-Pupp I, Hallin A-L, Hellström-Westas L, Cilio C, Berg A-C, Stjernqvist K, et al. Inflammation at birth is associated with subnormal development in very preterm infants. Pediatr Res. 2008;64:183–8.PubMedCrossRef
9.
go back to reference Suppiej A, Franzoi M, Vedovato S, Marucco A, Chiarelli S, Zanardo V. Neurodevelopmental outcome in preterm histological chorioamnionitis. Early Hum Dev. 2009;85:187–9.PubMedCrossRef Suppiej A, Franzoi M, Vedovato S, Marucco A, Chiarelli S, Zanardo V. Neurodevelopmental outcome in preterm histological chorioamnionitis. Early Hum Dev. 2009;85:187–9.PubMedCrossRef
10.
go back to reference Leviton A, Allred EN, Kuban KCK, Hecht JL, Onderdonk AB, O’Shea TM, et al. Microbiologic and histologic characteristics of the extremely preterm infant’s placenta predict white matter damage and later cerebral palsy. The ELGAN study. Pediatr Res. 2010;67:95–101.PubMedCentralPubMedCrossRef Leviton A, Allred EN, Kuban KCK, Hecht JL, Onderdonk AB, O’Shea TM, et al. Microbiologic and histologic characteristics of the extremely preterm infant’s placenta predict white matter damage and later cerebral palsy. The ELGAN study. Pediatr Res. 2010;67:95–101.PubMedCentralPubMedCrossRef
11.
go back to reference Andrews WW, Cliver SP, Biasini F, Peralta-Carcelen AM, Rector R, Alriksson-Schmidt AI, et al. Early preterm birth: association between in utero exposure to acute inflammation and severe neurodevelopmental disability at 6 years of age. Am J Obstetr Gynecol. 2008;198:466. e1–466.e11.CrossRef Andrews WW, Cliver SP, Biasini F, Peralta-Carcelen AM, Rector R, Alriksson-Schmidt AI, et al. Early preterm birth: association between in utero exposure to acute inflammation and severe neurodevelopmental disability at 6 years of age. Am J Obstetr Gynecol. 2008;198:466. e1–466.e11.CrossRef
12.
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.PubMedCrossRef 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.PubMedCrossRef
13.
go back to reference Reiman M, Kujari H, Maunu J, Parkkola R, Rikalainen H, Lapinleimu H, et al. Does placental inflammation relate to brain lesions and volume in preterm infants? J Pediatr. 2008;152:642–7. e2.PubMedCrossRef Reiman M, Kujari H, Maunu J, Parkkola R, Rikalainen H, Lapinleimu H, et al. Does placental inflammation relate to brain lesions and volume in preterm infants? J Pediatr. 2008;152:642–7. e2.PubMedCrossRef
14.
go back to reference Nelson KB, Grether JK, Dambrosia JM, Walsh E, Kohler S, Satyanarayana G, et al. Neonatal cytokines and cerebral palsy in very preterm infants. Pediatr Res. 2003;53:600–7.PubMedCrossRef Nelson KB, Grether JK, Dambrosia JM, Walsh E, Kohler S, Satyanarayana G, et al. Neonatal cytokines and cerebral palsy in very preterm infants. Pediatr Res. 2003;53:600–7.PubMedCrossRef
15.
go back to reference Lahra MM, Jeffery HE. A fetal response to chorioamnionitis is associated with early survival after preterm birth. Am J Obstetr Gynecol. 2004;190:147–51.CrossRef Lahra MM, Jeffery HE. A fetal response to chorioamnionitis is associated with early survival after preterm birth. Am J Obstetr Gynecol. 2004;190:147–51.CrossRef
16.
go back to reference Hendson L, Russell L, Robertson CMT, Liang Y, Chen Y, Abdalla A, et al. Neonatal and neurodevelopmental outcomes of very low birth weight infants with histologic chorioamnionitis. J Pediatr. 2011;158:397–402.PubMedCrossRef Hendson L, Russell L, Robertson CMT, Liang Y, Chen Y, Abdalla A, et al. Neonatal and neurodevelopmental outcomes of very low birth weight infants with histologic chorioamnionitis. J Pediatr. 2011;158:397–402.PubMedCrossRef
17.
go back to reference Yoon BH, Kim CJ, Romero R, Jun JK, Park KH, Choi ST, et al. Experimentally induced intrauterine infection causes fetal brain white matter lesions in rabbits. Am J Obstetr Gynecol. 1997;177:797–802.CrossRef Yoon BH, Kim CJ, Romero R, Jun JK, Park KH, Choi ST, et al. Experimentally induced intrauterine infection causes fetal brain white matter lesions in rabbits. Am J Obstetr Gynecol. 1997;177:797–802.CrossRef
18.
go back to reference Pang Y, Rodts-Palenik S, Cai Z, Bennett WA, Rhodes PG. Suppression of glial activation is involved in the protection of IL-10 on maternal E. coli induced neonatal white matter injury. Brain Res Dev Brain Res. 2005;157:141–9.PubMedCrossRef Pang Y, Rodts-Palenik S, Cai Z, Bennett WA, Rhodes PG. Suppression of glial activation is involved in the protection of IL-10 on maternal E. coli induced neonatal white matter injury. Brain Res Dev Brain Res. 2005;157:141–9.PubMedCrossRef
19.
go back to reference Poggi SH, Park J, Toso L, Abebe D, Roberson R, Woodard JE, et al. No phenotype associated with established lipopolysaccharide model for cerebral palsy. Am J Obstetr Gynecol. 2005;192:727–33.CrossRef Poggi SH, Park J, Toso L, Abebe D, Roberson R, Woodard JE, et al. No phenotype associated with established lipopolysaccharide model for cerebral palsy. Am J Obstetr Gynecol. 2005;192:727–33.CrossRef
20.
go back to reference Nitsos I, Rees SM, Duncan J, Kramer BW, Harding R, Newnham JP, et al. Chronic exposure to intra-amniotic lipopolysaccharide affects the ovine fetal brain. J Soc Gynecol Investig. 2006;13:239–47.PubMedCrossRef Nitsos I, Rees SM, Duncan J, Kramer BW, Harding R, Newnham JP, et al. Chronic exposure to intra-amniotic lipopolysaccharide affects the ovine fetal brain. J Soc Gynecol Investig. 2006;13:239–47.PubMedCrossRef
21.
go back to reference Cai Z, Pan ZL, Pang Y, Evans OB, Rhodes PG. Cytokine induction in fetal rat brains and brain injury in neonatal rats after maternal lipopolysaccharide administration. Pediatr Res. 2000;47:64–72.PubMedCrossRef Cai Z, Pan ZL, Pang Y, Evans OB, Rhodes PG. Cytokine induction in fetal rat brains and brain injury in neonatal rats after maternal lipopolysaccharide administration. Pediatr Res. 2000;47:64–72.PubMedCrossRef
22.
go back to reference Hava G, Vered L, Yael M, Mordechai H, Mahoud H. Alterations in behavior in adult offspring mice following maternal inflammation during pregnancy. Dev Psychobiol. 2006;48:162–8.PubMedCrossRef Hava G, Vered L, Yael M, Mordechai H, Mahoud H. Alterations in behavior in adult offspring mice following maternal inflammation during pregnancy. Dev Psychobiol. 2006;48:162–8.PubMedCrossRef
23.
go back to reference Mallard C, Welin A-K, Peebles D, Hagberg H, Kjellmer I. White matter injury following systemic endotoxemia or asphyxia in the fetal sheep. Neurochem Res. 2003;28:215–23.PubMedCrossRef Mallard C, Welin A-K, Peebles D, Hagberg H, Kjellmer I. White matter injury following systemic endotoxemia or asphyxia in the fetal sheep. Neurochem Res. 2003;28:215–23.PubMedCrossRef
24.
go back to reference Lodygensky GA, Kunz N, Perroud E, Somm E, Mlynarik V, Hüppi PS, et al. Definition and quantification of acute inflammatory white matter injury in the immature brain by MRI/MRS at high magnetic field. Pediatr Res. 2013;75:415–23.PubMedCrossRef Lodygensky GA, Kunz N, Perroud E, Somm E, Mlynarik V, Hüppi PS, et al. Definition and quantification of acute inflammatory white matter injury in the immature brain by MRI/MRS at high magnetic field. Pediatr Res. 2013;75:415–23.PubMedCrossRef
25.
go back to reference Cai Z, Pang Y, Lin S, Rhodes PG. Differential roles of tumor necrosis factor-α and interleukin-1 β in lipopolysaccharide-induced brain injury in the neonatal rat. Brain Res. 2003;975:37–47.PubMedCrossRef Cai Z, Pang Y, Lin S, Rhodes PG. Differential roles of tumor necrosis factor-α and interleukin-1 β in lipopolysaccharide-induced brain injury in the neonatal rat. Brain Res. 2003;975:37–47.PubMedCrossRef
26.
go back to reference Tissières P, Ochoda A, Dunn-Siegrist I, Drifte G, Morales M, Pfister R, et al. Innate immune deficiency of extremely premature neonates can be reversed by interferon-γ. PLoS One. 2012;7:e32863.PubMedCentralPubMedCrossRef Tissières P, Ochoda A, Dunn-Siegrist I, Drifte G, Morales M, Pfister R, et al. Innate immune deficiency of extremely premature neonates can be reversed by interferon-γ. PLoS One. 2012;7:e32863.PubMedCentralPubMedCrossRef
28.
go back to reference Shane AL, Stoll BJ. Neonatal sepsis: progress towards improved outcomes. J Infect. 2014;68 Suppl 1:S24–32.PubMedCrossRef Shane AL, Stoll BJ. Neonatal sepsis: progress towards improved outcomes. J Infect. 2014;68 Suppl 1:S24–32.PubMedCrossRef
29.
go back to reference Lavoie PM, Huang Q, Jolette E, Whalen M, Nuyt AM, Audibert F, et al. Profound lack of interleukin (IL)-12/IL-23p40 in neonates born early in gestation is associated with an increased risk of sepsis. J Infect Dis. 2010;202:1754–63.PubMedCentralPubMedCrossRef Lavoie PM, Huang Q, Jolette E, Whalen M, Nuyt AM, Audibert F, et al. Profound lack of interleukin (IL)-12/IL-23p40 in neonates born early in gestation is associated with an increased risk of sepsis. J Infect Dis. 2010;202:1754–63.PubMedCentralPubMedCrossRef
30.
go back to reference Strunk T, Currie A, Richmond P, Simmer K, Burgner D. Innate immunity in human newborn infants: prematurity means more than immaturity. J Matern Fetal Neonatal Med. 2011;24:25–31.PubMedCrossRef Strunk T, Currie A, Richmond P, Simmer K, Burgner D. Innate immunity in human newborn infants: prematurity means more than immaturity. J Matern Fetal Neonatal Med. 2011;24:25–31.PubMedCrossRef
31.
32.
go back to reference Strunk T, Prosser A, Levy O, Philbin V, Simmer K, Doherty D, et al. Responsiveness of human monocytes to the commensal bacterium Staphylococcus epidermidis develops late in gestation. Pediatr Res. 2012;72:10–8.PubMedCrossRef Strunk T, Prosser A, Levy O, Philbin V, Simmer K, Doherty D, et al. Responsiveness of human monocytes to the commensal bacterium Staphylococcus epidermidis develops late in gestation. Pediatr Res. 2012;72:10–8.PubMedCrossRef
33.
go back to reference Shen C-M, Lin S-C, Niu D-M, Kou YR. Development of monocyte Toll-like receptor 2 and Toll-like receptor 4 in preterm newborns during the first few months of life. Pediatr Res. 2013;73:685–91.PubMedCrossRef Shen C-M, Lin S-C, Niu D-M, Kou YR. Development of monocyte Toll-like receptor 2 and Toll-like receptor 4 in preterm newborns during the first few months of life. Pediatr Res. 2013;73:685–91.PubMedCrossRef
34.
go back to reference Schultz C, Rott C, Temming P, Schlenke P, Möller JC, Bucsky P. Enhanced interleukin-6 and interleukin-8 synthesis in term and preterm infants. Pediatr Res. 2002;51:317–22.PubMedCrossRef Schultz C, Rott C, Temming P, Schlenke P, Möller JC, Bucsky P. Enhanced interleukin-6 and interleukin-8 synthesis in term and preterm infants. Pediatr Res. 2002;51:317–22.PubMedCrossRef
35.
go back to reference Dammann O, Phillips TM, Allred EN, O’Shea TM, Paneth N, Van Marter LJ, et al. Mediators of fetal inflammation in extremely low gestational age newborns. Cytokine. 2001;13:234–9.PubMedCrossRef Dammann O, Phillips TM, Allred EN, O’Shea TM, Paneth N, Van Marter LJ, et al. Mediators of fetal inflammation in extremely low gestational age newborns. Cytokine. 2001;13:234–9.PubMedCrossRef
36.
go back to reference Nanthakumar N, Meng D, Goldstein AM, Zhu W, Lu L, Uauy R, et al. The mechanism of excessive intestinal inflammation in necrotizing enterocolitis: an immature innate immune response. PLoS One. 2011;6:e17776.PubMedCentralPubMedCrossRef Nanthakumar N, Meng D, Goldstein AM, Zhu W, Lu L, Uauy R, et al. The mechanism of excessive intestinal inflammation in necrotizing enterocolitis: an immature innate immune response. PLoS One. 2011;6:e17776.PubMedCentralPubMedCrossRef
37.
go back to reference Adib-Conquy M, Cavaillon J-M. Compensatory anti-inflammatory response syndrome. Thromb Haemost. 2009;101:36–47.PubMed Adib-Conquy M, Cavaillon J-M. Compensatory anti-inflammatory response syndrome. Thromb Haemost. 2009;101:36–47.PubMed
38.
39.
go back to reference Maheshwari A, Schelonka RL, Dimmitt RA, Carlo WA, Munoz-Hernandez B, Das A, et al. Cytokines associated with necrotizing enterocolitis in extremely-low-birth-weight infants. Pediatr Res. 2014;76:100–8.PubMedCentralPubMedCrossRef Maheshwari A, Schelonka RL, Dimmitt RA, Carlo WA, Munoz-Hernandez B, Das A, et al. Cytokines associated with necrotizing enterocolitis in extremely-low-birth-weight infants. Pediatr Res. 2014;76:100–8.PubMedCentralPubMedCrossRef
40.
go back to reference Chalak LF, Sánchez PJ, Adams-Huet B, Laptook AR, Heyne RJ, Rosenfeld CR. Biomarkers for severity of neonatal hypoxic-ischemic encephalopathy and outcomes in newborns receiving hypothermia therapy. J Pediatr. 2014;164:468–74. e1.PubMedCentralPubMedCrossRef Chalak LF, Sánchez PJ, Adams-Huet B, Laptook AR, Heyne RJ, Rosenfeld CR. Biomarkers for severity of neonatal hypoxic-ischemic encephalopathy and outcomes in newborns receiving hypothermia therapy. J Pediatr. 2014;164:468–74. e1.PubMedCentralPubMedCrossRef
42.
go back to reference Gutierrez EG, Banks WA, Kastin AJ. Murine tumor necrosis factor alpha is transported from blood to brain in the mouse. J Neuroimmunol. 1993;47:169–76.PubMedCrossRef Gutierrez EG, Banks WA, Kastin AJ. Murine tumor necrosis factor alpha is transported from blood to brain in the mouse. J Neuroimmunol. 1993;47:169–76.PubMedCrossRef
43.
go back to reference Banks WA, Kastin AJ, Gutierrez EG. Penetration of interleukin-6 across the murine blood–brain barrier. Neurosci Lett. 1994;179:53–6.PubMedCrossRef Banks WA, Kastin AJ, Gutierrez EG. Penetration of interleukin-6 across the murine blood–brain barrier. Neurosci Lett. 1994;179:53–6.PubMedCrossRef
44.
go back to reference Leviton A. Preterm birth and cerebral palsy: is tumor necrosis factor the missing link? Dev Med Child Neurol. 1993;35:553–8.PubMedCrossRef Leviton A. Preterm birth and cerebral palsy: is tumor necrosis factor the missing link? Dev Med Child Neurol. 1993;35:553–8.PubMedCrossRef
45.
go back to reference Banks WA, Ortiz L, Plotkin SR, Kastin AJ. Human interleukin (IL) 1 alpha, murine IL-1 alpha and murine IL-1 beta are transported from blood to brain in the mouse by a shared saturable mechanism. J Pharmacol Exp Ther. 1991;259:988–96.PubMed Banks WA, Ortiz L, Plotkin SR, Kastin AJ. Human interleukin (IL) 1 alpha, murine IL-1 alpha and murine IL-1 beta are transported from blood to brain in the mouse by a shared saturable mechanism. J Pharmacol Exp Ther. 1991;259:988–96.PubMed
46.
go back to reference Smith PLP, Hagberg H, Naylor AS, Mallard C. Neonatal peripheral immune challenge activates microglia and inhibits neurogenesis in the developing murine hippocampus. Dev Neurosci. 2014;36:119–31.PubMedCrossRef Smith PLP, Hagberg H, Naylor AS, Mallard C. Neonatal peripheral immune challenge activates microglia and inhibits neurogenesis in the developing murine hippocampus. Dev Neurosci. 2014;36:119–31.PubMedCrossRef
47.
48.
go back to reference Singh AK, Jiang Y. How does peripheral lipopolysaccharide induce gene expression in the brain of rats? Toxicology. 2004;201:197–207.PubMedCrossRef Singh AK, Jiang Y. How does peripheral lipopolysaccharide induce gene expression in the brain of rats? Toxicology. 2004;201:197–207.PubMedCrossRef
49.
go back to reference Tao-Cheng JH, Nagy Z, Brightman MW. Tight junctions of brain endothelium in vitro are enhanced by astroglia. J Neurosci. 1987;7:3293–9.PubMed Tao-Cheng JH, Nagy Z, Brightman MW. Tight junctions of brain endothelium in vitro are enhanced by astroglia. J Neurosci. 1987;7:3293–9.PubMed
50.
go back to reference Janzer RC, Raff MC. Astrocytes induce blood–brain barrier properties in endothelial cells. Nature. 1987;325:253–7.PubMedCrossRef Janzer RC, Raff MC. Astrocytes induce blood–brain barrier properties in endothelial cells. Nature. 1987;325:253–7.PubMedCrossRef
51.
52.
go back to reference Ek CJ, Dziegielewska KM, Habgood MD, Saunders NR. Barriers in the developing brain and neurotoxicology. Neurotoxicology. 2012;33:586–604.PubMedCrossRef Ek CJ, Dziegielewska KM, Habgood MD, Saunders NR. Barriers in the developing brain and neurotoxicology. Neurotoxicology. 2012;33:586–604.PubMedCrossRef
53.
go back to reference Stolp HB, Dziegielewska KM. Review: role of developmental inflammation and blood–brain barrier dysfunction in neurodevelopmental and neurodegenerative diseases. Neuropathol Appl Neurobiol. 2009;35:132–46.PubMedCrossRef Stolp HB, Dziegielewska KM. Review: role of developmental inflammation and blood–brain barrier dysfunction in neurodevelopmental and neurodegenerative diseases. Neuropathol Appl Neurobiol. 2009;35:132–46.PubMedCrossRef
54.
go back to reference Nagyőszi P, Wilhelm I, Farkas AE, Fazakas C, Dung NTK, Haskó J, et al. Expression and regulation of toll-like receptors in cerebral endothelial cells. Neurochem Int. 2010;57:556–64.PubMedCrossRef Nagyőszi P, Wilhelm I, Farkas AE, Fazakas C, Dung NTK, Haskó J, et al. Expression and regulation of toll-like receptors in cerebral endothelial cells. Neurochem Int. 2010;57:556–64.PubMedCrossRef
55.
go back to reference Chakravarty S, Herkenham M. Toll-like receptor 4 on nonhematopoietic cells sustains CNS inflammation during endotoxemia, independent of systemic cytokines. J Neurosci. 2005;25:1788–96.PubMedCrossRef Chakravarty S, Herkenham M. Toll-like receptor 4 on nonhematopoietic cells sustains CNS inflammation during endotoxemia, independent of systemic cytokines. J Neurosci. 2005;25:1788–96.PubMedCrossRef
56.
go back to reference Gosselin D, RIVEST S. MyD88 signaling in brain endothelial cells is essential for the neuronal activity and glucocorticoid release during systemic inflammation. Mol Psychiatry. 2008;13:480–97.PubMedCrossRef Gosselin D, RIVEST S. MyD88 signaling in brain endothelial cells is essential for the neuronal activity and glucocorticoid release during systemic inflammation. Mol Psychiatry. 2008;13:480–97.PubMedCrossRef
57.
go back to reference Ganong WF. Circumventricular organs: definition and role in the regulation of endocrine and autonomic function. Clin Exp Pharmacol Physiol. 2000;27:422–7.PubMedCrossRef Ganong WF. Circumventricular organs: definition and role in the regulation of endocrine and autonomic function. Clin Exp Pharmacol Physiol. 2000;27:422–7.PubMedCrossRef
58.
go back to reference LAFLAMME N, RIVEST S. Toll-like receptor 4: the missing link of the cerebral innate immune response triggered by circulating gram-negative bacterial cell wall components. FASEB J. 2001;15:155–63.PubMedCrossRef LAFLAMME N, RIVEST S. Toll-like receptor 4: the missing link of the cerebral innate immune response triggered by circulating gram-negative bacterial cell wall components. FASEB J. 2001;15:155–63.PubMedCrossRef
59.
go back to reference Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci. 2008;9:46–56.PubMedCentralPubMedCrossRef Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci. 2008;9:46–56.PubMedCentralPubMedCrossRef
61.
go back to reference Stridh L, Ek CJ, Wang X, Nilsson H, Mallard C. Regulation of toll-like receptors in the choroid plexus in the immature brain after systemic inflammatory stimuli. Transl Stroke Res. 2013;4:220–7.PubMedCentralPubMedCrossRef Stridh L, Ek CJ, Wang X, Nilsson H, Mallard C. Regulation of toll-like receptors in the choroid plexus in the immature brain after systemic inflammatory stimuli. Transl Stroke Res. 2013;4:220–7.PubMedCentralPubMedCrossRef
62.
go back to reference D’angelo B, Ek CJ, Sandberg M, Mallard C. Expression of the Nrf2-system at the blood-CSF barrier is modulated by neonatal inflammation and hypoxia-ischemia. J Inherit Metab Dis. 2013;36:479–90.PubMedCentralPubMedCrossRef D’angelo B, Ek CJ, Sandberg M, Mallard C. Expression of the Nrf2-system at the blood-CSF barrier is modulated by neonatal inflammation and hypoxia-ischemia. J Inherit Metab Dis. 2013;36:479–90.PubMedCentralPubMedCrossRef
63.
go back to reference Verney C, Pogledic I, Biran V, Adle-Biassette H, Fallet-Bianco C, Gressens P. Microglial reaction in axonal crossroads is a hallmark of noncystic periventricular white matter injury in very preterm infants. J Neuropathol Exp Neurol. 2012;71:251–64.PubMedCrossRef Verney C, Pogledic I, Biran V, Adle-Biassette H, Fallet-Bianco C, Gressens P. Microglial reaction in axonal crossroads is a hallmark of noncystic periventricular white matter injury in very preterm infants. J Neuropathol Exp Neurol. 2012;71:251–64.PubMedCrossRef
65.
go back to reference Thornton C, Rousset CI, Kichev A, Miyakuni Y, Vontell R, Baburamani AA, et al. Molecular mechanisms of neonatal brain injury. Neurol Res Int. 2012;2012:506320–16.PubMedCentralPubMedCrossRef Thornton C, Rousset CI, Kichev A, Miyakuni Y, Vontell R, Baburamani AA, et al. Molecular mechanisms of neonatal brain injury. Neurol Res Int. 2012;2012:506320–16.PubMedCentralPubMedCrossRef
66.
go back to reference Perry VH, Cunningham C, Holmes C. Systemic infections and inflammation affect chronic neurodegeneration. Nat Rev Immunol. 2007;7:161–7.PubMedCrossRef Perry VH, Cunningham C, Holmes C. Systemic infections and inflammation affect chronic neurodegeneration. Nat Rev Immunol. 2007;7:161–7.PubMedCrossRef
67.
68.
go back to reference Yao L, Kan EM, Lu J, Hao A, Dheen ST, Kaur C, et al. Toll-like receptor 4 mediates microglial activation and production of inflammatory mediators in neonatal rat brain following hypoxia: role of TLR4 in hypoxic microglia. J Neuroinflammation. 2013;10:23.PubMedCentralPubMedCrossRef Yao L, Kan EM, Lu J, Hao A, Dheen ST, Kaur C, et al. Toll-like receptor 4 mediates microglial activation and production of inflammatory mediators in neonatal rat brain following hypoxia: role of TLR4 in hypoxic microglia. J Neuroinflammation. 2013;10:23.PubMedCentralPubMedCrossRef
69.
go back to reference Ivacko JA, Sun R, Silverstein FS. Hypoxic-ischemic brain injury induces an acute microglial reaction in perinatal rats. Pediatr Res. 1996;39:39–47.PubMedCrossRef Ivacko JA, Sun R, Silverstein FS. Hypoxic-ischemic brain injury induces an acute microglial reaction in perinatal rats. Pediatr Res. 1996;39:39–47.PubMedCrossRef
70.
go back to reference Dommergues M-A, Plaisant F, Verney C, Gressens P. Early microglial activation following neonatal excitotoxic brain damage in mice: a potential target for neuroprotection. Neuroscience. 2003;121:619–28.PubMedCrossRef Dommergues M-A, Plaisant F, Verney C, Gressens P. Early microglial activation following neonatal excitotoxic brain damage in mice: a potential target for neuroprotection. Neuroscience. 2003;121:619–28.PubMedCrossRef
71.
go back to reference Eklind S, Mallard C, Arvidsson P, Hagberg H. Lipopolysaccharide induces both a primary and a secondary phase of sensitization in the developing rat brain. Pediatr Res. 2005;58:112–6.PubMedCrossRef Eklind S, Mallard C, Arvidsson P, Hagberg H. Lipopolysaccharide induces both a primary and a secondary phase of sensitization in the developing rat brain. Pediatr Res. 2005;58:112–6.PubMedCrossRef
72.
go back to reference Hickey E, Shi H, Van Arsdell G, Askalan R. Lipopolysaccharide-induced preconditioning against ischemic injury is associated with changes in toll-like receptor 4 expression in the rat developing brain. Pediatr Res. 2011;70:10–4.PubMedCrossRef Hickey E, Shi H, Van Arsdell G, Askalan R. Lipopolysaccharide-induced preconditioning against ischemic injury is associated with changes in toll-like receptor 4 expression in the rat developing brain. Pediatr Res. 2011;70:10–4.PubMedCrossRef
73.
go back to reference Destot-Wong K-D, Liang K, Gupta SK, Favrais G, Schwendimann L, Pansiot J, et al. The AMPA receptor positive allosteric modulator, S18986, is neuroprotective against neonatal excitotoxic and inflammatory brain damage through BDNF synthesis. Neuropharmacology. 2009;57:277–86.PubMedCrossRef Destot-Wong K-D, Liang K, Gupta SK, Favrais G, Schwendimann L, Pansiot J, et al. The AMPA receptor positive allosteric modulator, S18986, is neuroprotective against neonatal excitotoxic and inflammatory brain damage through BDNF synthesis. Neuropharmacology. 2009;57:277–86.PubMedCrossRef
74.
go back to reference Wu X, Zhu D, Jiang X, Okagaki P, Mearow K, Zhu G, et al. AMPA protects cultured neurons against glutamate excitotoxicity through a phosphatidylinositol 3-kinase-dependent activation in extracellular signal-regulated kinase to upregulate BDNF gene expression. J Neurochem. 2004;90:807–18.PubMedCrossRef Wu X, Zhu D, Jiang X, Okagaki P, Mearow K, Zhu G, et al. AMPA protects cultured neurons against glutamate excitotoxicity through a phosphatidylinositol 3-kinase-dependent activation in extracellular signal-regulated kinase to upregulate BDNF gene expression. J Neurochem. 2004;90:807–18.PubMedCrossRef
75.
go back to reference Manning SM, Talos DM, Zhou C, Selip DB, Park H-K, Park C-J, et al. NMDA receptor blockade with memantine attenuates white matter injury in a rat model of periventricular leukomalacia. J Neurosci. 2008;28:6670–8.PubMedCentralPubMedCrossRef Manning SM, Talos DM, Zhou C, Selip DB, Park H-K, Park C-J, et al. NMDA receptor blockade with memantine attenuates white matter injury in a rat model of periventricular leukomalacia. J Neurosci. 2008;28:6670–8.PubMedCentralPubMedCrossRef
77.
go back to reference Segovia KN, McClure M, Moravec M, Luo NL, Wan Y, Gong X, et al. Arrested oligodendrocyte lineage maturation in chronic perinatal white matter injury. Ann Neurol. 2008;63:520–30.PubMedCentralPubMedCrossRef Segovia KN, McClure M, Moravec M, Luo NL, Wan Y, Gong X, et al. Arrested oligodendrocyte lineage maturation in chronic perinatal white matter injury. Ann Neurol. 2008;63:520–30.PubMedCentralPubMedCrossRef
78.
go back to reference Riddle A, Dean J, Buser JR, Gong X, Maire J, Chen K, et al. Histopathological correlates of magnetic resonance imaging–defined chronic perinatal white matter injury. Ann Neurol. 2011;70:493–507.PubMedCentralPubMedCrossRef Riddle A, Dean J, Buser JR, Gong X, Maire J, Chen K, et al. Histopathological correlates of magnetic resonance imaging–defined chronic perinatal white matter injury. Ann Neurol. 2011;70:493–507.PubMedCentralPubMedCrossRef
79.
go back to reference Buser JR, Maire J, Riddle A, Gong X, Nguyen T, Nelson K, et al. Arrested preoligodendrocyte maturation contributes to myelination failure in premature infants. Ann Neurol. 2012;71:93–109.PubMedCentralPubMedCrossRef Buser JR, Maire J, Riddle A, Gong X, Nguyen T, Nelson K, et al. Arrested preoligodendrocyte maturation contributes to myelination failure in premature infants. Ann Neurol. 2012;71:93–109.PubMedCentralPubMedCrossRef
80.
go back to reference Goldenberg RL, Hauth JC, Andrews WW. Intrauterine infection and preterm delivery. N Engl J Med. 2000;342:1500–7.PubMedCrossRef Goldenberg RL, Hauth JC, Andrews WW. Intrauterine infection and preterm delivery. N Engl J Med. 2000;342:1500–7.PubMedCrossRef
81.
go back to reference Yoon BH, Jun JK, Romero R, Park KH, Gomez R, Choi JH, et al. Amniotic fluid inflammatory cytokines (interleukin-6, interleukin-1beta, and tumor necrosis factor-alpha), neonatal brain white matter lesions, and cerebral palsy. Am J Obstetr Gynecol. 1997;177:19–26.CrossRef Yoon BH, Jun JK, Romero R, Park KH, Gomez R, Choi JH, et al. Amniotic fluid inflammatory cytokines (interleukin-6, interleukin-1beta, and tumor necrosis factor-alpha), neonatal brain white matter lesions, and cerebral palsy. Am J Obstetr Gynecol. 1997;177:19–26.CrossRef
82.
go back to reference Duggan PJ, Maalouf EF, Watts TL, Sullivan MH, Counsell SJ, Allsop J, et al. Intrauterine T-cell activation and increased proinflammatory cytokine concentrations in preterm infants with cerebral lesions. Lancet. 2001;358:1699–700.PubMedCrossRef Duggan PJ, Maalouf EF, Watts TL, Sullivan MH, Counsell SJ, Allsop J, et al. Intrauterine T-cell activation and increased proinflammatory cytokine concentrations in preterm infants with cerebral lesions. Lancet. 2001;358:1699–700.PubMedCrossRef
83.
go back to reference Yoon BH, Romero R, Park JS, Kim CJ, Kim SH, Choi JH, et al. Fetal exposure to an intra-amniotic inflammation and the development of cerebral palsy at the age of three years. Am J Obstetr Gynecol. 2000;182:675–81.CrossRef Yoon BH, Romero R, Park JS, Kim CJ, Kim SH, Choi JH, et al. Fetal exposure to an intra-amniotic inflammation and the development of cerebral palsy at the age of three years. Am J Obstetr Gynecol. 2000;182:675–81.CrossRef
84.
go back to reference Satar M, Turhan E, Yapicioglu H, Narli N, Ozgunen FT, Cetiner S. Cord blood cytokine levels in neonates born to mothers with prolonged premature rupture of membranes and its relationship with morbidity and mortality. Eur Cytokine Netw. 2008;19:37–41.PubMed Satar M, Turhan E, Yapicioglu H, Narli N, Ozgunen FT, Cetiner S. Cord blood cytokine levels in neonates born to mothers with prolonged premature rupture of membranes and its relationship with morbidity and mortality. Eur Cytokine Netw. 2008;19:37–41.PubMed
85.
go back to reference Rocha G, Proença E, Guedes A, Carvalho C, Areias A, Ramos JP, et al. Cord blood levels of IL-6, IL-8 and IL-10 may be early predictors of bronchopulmonary dysplasia in preterm newborns small for gestational age. Dis Markers. 2012;33:51–60.PubMedCentralPubMedCrossRef Rocha G, Proença E, Guedes A, Carvalho C, Areias A, Ramos JP, et al. Cord blood levels of IL-6, IL-8 and IL-10 may be early predictors of bronchopulmonary dysplasia in preterm newborns small for gestational age. Dis Markers. 2012;33:51–60.PubMedCentralPubMedCrossRef
86.
go back to reference An H, Nishimaki S, Ohyama M, Haruki A, Naruto T, Kobayashi N, et al. Interleukin-6, interleukin-8, and soluble tumor necrosis factor receptor-I in the cord blood as predictors of chronic lung disease in premature infants. Am J Obstetr Gynecol. 2004;191:1649–54.CrossRef An H, Nishimaki S, Ohyama M, Haruki A, Naruto T, Kobayashi N, et al. Interleukin-6, interleukin-8, and soluble tumor necrosis factor receptor-I in the cord blood as predictors of chronic lung disease in premature infants. Am J Obstetr Gynecol. 2004;191:1649–54.CrossRef
87.
go back to reference Takao D, Ibara S, Tokuhisa T, Ishihara C, Maede Y, Matsui T, et al. Predicting onset of chronic lung disease using cord blood cytokines. Pediatr Int. 2014;56:566–70.PubMedCrossRef Takao D, Ibara S, Tokuhisa T, Ishihara C, Maede Y, Matsui T, et al. Predicting onset of chronic lung disease using cord blood cytokines. Pediatr Int. 2014;56:566–70.PubMedCrossRef
88.
go back to reference Hecht JL, Fichorova RN, Tang VF, Allred EN, McElrath TF, Leviton A, et al. Relationship between neonatal blood protein concentrations and placenta histologic characteristics in extremely low GA newborns. Pediatr Res. 2011;69:68–73.PubMedCentralPubMedCrossRef Hecht JL, Fichorova RN, Tang VF, Allred EN, McElrath TF, Leviton A, et al. Relationship between neonatal blood protein concentrations and placenta histologic characteristics in extremely low GA newborns. Pediatr Res. 2011;69:68–73.PubMedCentralPubMedCrossRef
89.
go back to reference Kuban KCK, O’Shea TM, Allred EN, Paneth N, Hirtz D, Fichorova RN, et al. Systemic inflammation and cerebral palsy risk in extremely preterm infants. J Child Neurol. 2014;29:1692–8.PubMedPubMedCentralCrossRef Kuban KCK, O’Shea TM, Allred EN, Paneth N, Hirtz D, Fichorova RN, et al. Systemic inflammation and cerebral palsy risk in extremely preterm infants. J Child Neurol. 2014;29:1692–8.PubMedPubMedCentralCrossRef
90.
go back to reference Ellison VJ, Mocatta TJ, Winterbourn CC, Darlow BA, Volpe JJ, Inder TE. The relationship of CSF and plasma cytokine levels to cerebral white matter injury in the premature newborn. Pediatr Res. 2005;57:282–6.PubMedCrossRef Ellison VJ, Mocatta TJ, Winterbourn CC, Darlow BA, Volpe JJ, Inder TE. The relationship of CSF and plasma cytokine levels to cerebral white matter injury in the premature newborn. Pediatr Res. 2005;57:282–6.PubMedCrossRef
91.
go back to reference Panigrahy A, Wisnowski JL, Furtado A, Lepore N, Paquette L, Bluml S. Neuroimaging biomarkers of preterm brain injury: toward developing the preterm connectome. Pediatr Radiol. 2012;42:33–61.CrossRef Panigrahy A, Wisnowski JL, Furtado A, Lepore N, Paquette L, Bluml S. Neuroimaging biomarkers of preterm brain injury: toward developing the preterm connectome. Pediatr Radiol. 2012;42:33–61.CrossRef
92.
go back to reference Maalouf EF, Duggan PJ, Counsell SJ, Rutherford MA, Cowan F, Azzopardi D, et al. Comparison of findings on cranial ultrasound and magnetic resonance imaging in preterm infants. Pediatrics. 2001;107:719–27.PubMedCrossRef Maalouf EF, Duggan PJ, Counsell SJ, Rutherford MA, Cowan F, Azzopardi D, et al. Comparison of findings on cranial ultrasound and magnetic resonance imaging in preterm infants. Pediatrics. 2001;107:719–27.PubMedCrossRef
93.
go back to reference O’Shea TM, Counsell SJ, Bartels DB, Dammann O. Magnetic resonance and ultrasound brain imaging in preterm infants. Early Hum Dev. 2005;81:263–71.PubMedCrossRef O’Shea TM, Counsell SJ, Bartels DB, Dammann O. Magnetic resonance and ultrasound brain imaging in preterm infants. Early Hum Dev. 2005;81:263–71.PubMedCrossRef
94.
go back to reference van Wezel-Meijler G, Steggerda SJ, Leijser LM. Cranial ultrasonography in neonates: role and limitations. Semin Perinatol. 2010;34:28.PubMedCrossRef van Wezel-Meijler G, Steggerda SJ, Leijser LM. Cranial ultrasonography in neonates: role and limitations. Semin Perinatol. 2010;34:28.PubMedCrossRef
95.
go back to reference Miller SP, Cozzio CC, Goldstein RB, Ferriero DM, Partridge JC, Vigneron DB, et al. Comparing the diagnosis of white matter injury in premature newborns with serial MR Imaging and transfontanel ultrasonography findings. AJNR Am J Neuroradiol. 2003;24:1661–9.PubMed Miller SP, Cozzio CC, Goldstein RB, Ferriero DM, Partridge JC, Vigneron DB, et al. Comparing the diagnosis of white matter injury in premature newborns with serial MR Imaging and transfontanel ultrasonography findings. AJNR Am J Neuroradiol. 2003;24:1661–9.PubMed
96.
go back to reference Inder TE, Anderson NJ, Spencer C, Wells S, Volpe JJ. White matter injury in the premature infant: a comparison between serial cranial sonographic and MR findings at term. AJNR Am J Neuroradiol. 2003;24:805–9.PubMed Inder TE, Anderson NJ, Spencer C, Wells S, Volpe JJ. White matter injury in the premature infant: a comparison between serial cranial sonographic and MR findings at term. AJNR Am J Neuroradiol. 2003;24:805–9.PubMed
97.
go back to reference Ciambra G, Arachi S, Protano C, Cellitti R, Caoci S, Di Biasi C, et al. Accuracy of transcranial ultrasound in the detection of mild white matter lesions in newborns. Neuroradiol J. 2013;26:284–9.PubMedCrossRef Ciambra G, Arachi S, Protano C, Cellitti R, Caoci S, Di Biasi C, et al. Accuracy of transcranial ultrasound in the detection of mild white matter lesions in newborns. Neuroradiol J. 2013;26:284–9.PubMedCrossRef
98.
go back to reference Mirmiran M, Barnes PD, Keller K, Constantinou JC, Fleisher BE, Hintz SR, et al. Neonatal brain magnetic resonance imaging before discharge is better than serial cranial ultrasound in predicting cerebral palsy in very low birth weight preterm infants. Pediatrics. 2004;114:992–8.PubMedCrossRef Mirmiran M, Barnes PD, Keller K, Constantinou JC, Fleisher BE, Hintz SR, et al. Neonatal brain magnetic resonance imaging before discharge is better than serial cranial ultrasound in predicting cerebral palsy in very low birth weight preterm infants. Pediatrics. 2004;114:992–8.PubMedCrossRef
99.
go back to reference Woodward LJ, Anderson PJ, Austin NC, Howard K, Inder TE. Neonatal MRI to predict neurodevelopmental outcomes in preterm infants. N Engl J Med. 2006;355:685–94.PubMedCrossRef Woodward LJ, Anderson PJ, Austin NC, Howard K, Inder TE. Neonatal MRI to predict neurodevelopmental outcomes in preterm infants. N Engl J Med. 2006;355:685–94.PubMedCrossRef
100.
go back to reference Giustetto P, Filippi M, Castano M, Terreno E. Non-invasive parenchymal, vascular and metabolic high-frequency ultrasound and photoacoustic rat deep brain imaging. J Vis Exp 2015. doi: 10.3791/52162 Giustetto P, Filippi M, Castano M, Terreno E. Non-invasive parenchymal, vascular and metabolic high-frequency ultrasound and photoacoustic rat deep brain imaging. J Vis Exp 2015. doi: 10.3791/52162
101.
go back to reference Guevara E, Berti R, Londono I, Xie N, Bellec P, Lesage F, et al. Imaging of an inflammatory injury in the newborn rat brain with photoacoustic tomography. PLoS One. 2013;8:e83045.PubMedCentralPubMedCrossRef Guevara E, Berti R, Londono I, Xie N, Bellec P, Lesage F, et al. Imaging of an inflammatory injury in the newborn rat brain with photoacoustic tomography. PLoS One. 2013;8:e83045.PubMedCentralPubMedCrossRef
102.
go back to reference Mento G, Bisiacchi PS. Neurocognitive development in preterm infants: insights from different approaches. Neurosci Biobehav Rev. 2012;36:536–55.PubMedCrossRef Mento G, Bisiacchi PS. Neurocognitive development in preterm infants: insights from different approaches. Neurosci Biobehav Rev. 2012;36:536–55.PubMedCrossRef
103.
go back to reference Shany E, Berger I. Neonatal electroencephalography: review of a practical approach. J Child Neurol. 2011;26:341–55.PubMedCrossRef Shany E, Berger I. Neonatal electroencephalography: review of a practical approach. J Child Neurol. 2011;26:341–55.PubMedCrossRef
104.
go back to reference El-Dib M, Chang T, Tsuchida TN, Clancy RR. Amplitude-integrated electroencephalography in neonates. Pediatr Neurol. 2009;41:315–26.PubMedCrossRef El-Dib M, Chang T, Tsuchida TN, Clancy RR. Amplitude-integrated electroencephalography in neonates. Pediatr Neurol. 2009;41:315–26.PubMedCrossRef
105.
go back to reference Dean JM, van de Looij Y, Sizonenko SV, Lodygensky GA, Lazeyras F, Bolouri H, et al. Delayed cortical impairment following lipopolysaccharide exposure in preterm fetal sheep. Ann Neurol. 2011;70:846–56.PubMedCrossRef Dean JM, van de Looij Y, Sizonenko SV, Lodygensky GA, Lazeyras F, Bolouri H, et al. Delayed cortical impairment following lipopolysaccharide exposure in preterm fetal sheep. Ann Neurol. 2011;70:846–56.PubMedCrossRef
106.
go back to reference Keogh MJ, Bennet L, Drury PP, Booth LC, Mathai S, Naylor AS, et al. Subclinical exposure to low-dose endotoxin impairs EEG maturation in preterm fetal sheep. Am J Physiol Regul Integr Comp Physiol. 2012;303:R270–8.PubMedCrossRef Keogh MJ, Bennet L, Drury PP, Booth LC, Mathai S, Naylor AS, et al. Subclinical exposure to low-dose endotoxin impairs EEG maturation in preterm fetal sheep. Am J Physiol Regul Integr Comp Physiol. 2012;303:R270–8.PubMedCrossRef
107.
go back to reference Watanabe K, Hayakawa F, Okumura A. Neonatal EEG: a powerful tool in the assessment of brain damage in preterm infants. Brain and Development. 1999;21:361–72.PubMedCrossRef Watanabe K, Hayakawa F, Okumura A. Neonatal EEG: a powerful tool in the assessment of brain damage in preterm infants. Brain and Development. 1999;21:361–72.PubMedCrossRef
108.
go back to reference Maruyama K, Okumura A, Hayakawa F, Kato T. Prognostic value of EEG depression in preterm infants for later development of cerebral palsy. Neuropediatrics. 2002;33:133.PubMedCrossRef Maruyama K, Okumura A, Hayakawa F, Kato T. Prognostic value of EEG depression in preterm infants for later development of cerebral palsy. Neuropediatrics. 2002;33:133.PubMedCrossRef
109.
go back to reference Baud O, d’Allest A-M, Lacaze-Masmonteil T, Zupan V, Nedelcoux H, Boithias C, et al. The early diagnosis of periventricular leukomalacia in premature infants with positive rolandic sharp waves on serial electroencephalography. The J Pediatr. 1998;132:813–7.PubMedCrossRef Baud O, d’Allest A-M, Lacaze-Masmonteil T, Zupan V, Nedelcoux H, Boithias C, et al. The early diagnosis of periventricular leukomalacia in premature infants with positive rolandic sharp waves on serial electroencephalography. The J Pediatr. 1998;132:813–7.PubMedCrossRef
110.
go back to reference Okumura A, Hayakawa F, Kato T, Maruyama K, Kubota T, Suzuki M, et al. Abnormal sharp transients on electroencephalograms in preterm infants with periventricular leukomalacia. The J Pediatr. 2003;143:26–30.PubMedCrossRef Okumura A, Hayakawa F, Kato T, Maruyama K, Kubota T, Suzuki M, et al. Abnormal sharp transients on electroencephalograms in preterm infants with periventricular leukomalacia. The J Pediatr. 2003;143:26–30.PubMedCrossRef
111.
go back to reference Shah DK, de Vries LS, Hellström-Westas L, Toet MC, Inder TE. Amplitude-integrated electroencephalography in the newborn: a valuable tool. Pediatrics. 2008;122:863–5.PubMedCrossRef Shah DK, de Vries LS, Hellström-Westas L, Toet MC, Inder TE. Amplitude-integrated electroencephalography in the newborn: a valuable tool. Pediatrics. 2008;122:863–5.PubMedCrossRef
112.
go back to reference Wikström S, Ley D, Hansen-Pupp I, Rosén I, Hellström-Westas L. Early amplitude-integrated EEG correlates with cord TNF-α and brain injury in very preterm infants. Acta Paediatr. 2008;97:915–9.PubMedCrossRef Wikström S, Ley D, Hansen-Pupp I, Rosén I, Hellström-Westas L. Early amplitude-integrated EEG correlates with cord TNF-α and brain injury in very preterm infants. Acta Paediatr. 2008;97:915–9.PubMedCrossRef
113.
go back to reference Lodygensky GA, Vasung L, Sizonenko SV, Hüppi PS. Neuroimaging of cortical development and brain connectivity in human newborns and animal models. J Anat. 2010;217:418–28.PubMedCentralPubMedCrossRef Lodygensky GA, Vasung L, Sizonenko SV, Hüppi PS. Neuroimaging of cortical development and brain connectivity in human newborns and animal models. J Anat. 2010;217:418–28.PubMedCentralPubMedCrossRef
114.
go back to reference Lodygensky GA, West T, Stump M, Holtzman DM, Inder TE, Neil JJ. In vivo MRI analysis of an inflammatory injury in the developing brain. Brain Behav Immun. 2010;24:759–67.PubMedCentralPubMedCrossRef Lodygensky GA, West T, Stump M, Holtzman DM, Inder TE, Neil JJ. In vivo MRI analysis of an inflammatory injury in the developing brain. Brain Behav Immun. 2010;24:759–67.PubMedCentralPubMedCrossRef
115.
go back to reference Nanba Y, Matsui K, Aida N, Sato Y, Toyoshima K, Kawataki M, et al. Magnetic resonance imaging regional T1 abnormalities at term accurately predict motor outcome in preterm infants. Pediatrics. 2007;120:e10–9.PubMedCrossRef Nanba Y, Matsui K, Aida N, Sato Y, Toyoshima K, Kawataki M, et al. Magnetic resonance imaging regional T1 abnormalities at term accurately predict motor outcome in preterm infants. Pediatrics. 2007;120:e10–9.PubMedCrossRef
116.
go back to reference Miller SP, Ferriero DM, Leonard C, Piecuch R, Glidden DV, Partridge JC, et al. Early brain injury in premature newborns detected with magnetic resonance imaging is associated with adverse early neurodevelopmental outcome. The J Pediatr. 2005;147:609–16.PubMedCrossRef Miller SP, Ferriero DM, Leonard C, Piecuch R, Glidden DV, Partridge JC, et al. Early brain injury in premature newborns detected with magnetic resonance imaging is associated with adverse early neurodevelopmental outcome. The J Pediatr. 2005;147:609–16.PubMedCrossRef
117.
go back to reference Chau V, Brant R, Poskitt KJ, Tam EWY, Synnes A, Miller SP. Postnatal infection is associated with widespread abnormalities of brain development in premature newborns. Pediatr Res. 2012;71:274–9.PubMedCentralPubMedCrossRef Chau V, Brant R, Poskitt KJ, Tam EWY, Synnes A, Miller SP. Postnatal infection is associated with widespread abnormalities of brain development in premature newborns. Pediatr Res. 2012;71:274–9.PubMedCentralPubMedCrossRef
118.
go back to reference Norris DG, Niendorf T, Hoehn-Berlage M, Kohno K, Schneider EJ, Hainz P, et al. Incidence of apparent restricted diffusion in three different models of cerebral infarction. Magn Reson Imaging. 1994;12:1175–82.PubMedCrossRef Norris DG, Niendorf T, Hoehn-Berlage M, Kohno K, Schneider EJ, Hainz P, et al. Incidence of apparent restricted diffusion in three different models of cerebral infarction. Magn Reson Imaging. 1994;12:1175–82.PubMedCrossRef
119.
go back to reference Tuor UI, Kozlowski P, Del Bigio MR, Ramjiawan B, Su S, Malisza K, et al. Diffusion- and T2-weighted increases in magnetic resonance images of immature brain during hypoxia-ischemia: transient reversal posthypoxia. Exp Neurol. 1998;150:321–8.PubMedCrossRef Tuor UI, Kozlowski P, Del Bigio MR, Ramjiawan B, Su S, Malisza K, et al. Diffusion- and T2-weighted increases in magnetic resonance images of immature brain during hypoxia-ischemia: transient reversal posthypoxia. Exp Neurol. 1998;150:321–8.PubMedCrossRef
120.
go back to reference Nedelcu J, Klein MA, Aguzzi A, Boesiger P, Martin E. Biphasic edema after hypoxic-ischemic brain injury in neonatal rats reflects early neuronal and late glial damage. Pediatr Res. 1999;46:297–304.PubMedCrossRef Nedelcu J, Klein MA, Aguzzi A, Boesiger P, Martin E. Biphasic edema after hypoxic-ischemic brain injury in neonatal rats reflects early neuronal and late glial damage. Pediatr Res. 1999;46:297–304.PubMedCrossRef
121.
go back to reference McKinstry RC, Miller JH, Snyder AZ, Mathur A, Schefft GL, Almli CR, et al. A prospective, longitudinal diffusion tensor imaging study of brain injury in newborns. Neurology. 2002;59:824–33.PubMedCrossRef McKinstry RC, Miller JH, Snyder AZ, Mathur A, Schefft GL, Almli CR, et al. A prospective, longitudinal diffusion tensor imaging study of brain injury in newborns. Neurology. 2002;59:824–33.PubMedCrossRef
122.
go back to reference Yang F, Sun X, Beech W, Teter B, Wu S, Sigel J, et al. Antibody to caspase-cleaved actin detects apoptosis in differentiated neuroblastoma and plaque-associated neurons and microglia in Alzheimer’s disease. Am J Pathol. 1998;152:379–89.PubMedCentralPubMed Yang F, Sun X, Beech W, Teter B, Wu S, Sigel J, et al. Antibody to caspase-cleaved actin detects apoptosis in differentiated neuroblastoma and plaque-associated neurons and microglia in Alzheimer’s disease. Am J Pathol. 1998;152:379–89.PubMedCentralPubMed
123.
go back to reference Hendrickson ML, Ling C, Kalil RE. Degeneration of axotomized projection neurons in the rat dLGN: temporal progression of events and their mitigation by a single administration of FGF2. PLoS One. 2012;7:e46918.PubMedCentralPubMedCrossRef Hendrickson ML, Ling C, Kalil RE. Degeneration of axotomized projection neurons in the rat dLGN: temporal progression of events and their mitigation by a single administration of FGF2. PLoS One. 2012;7:e46918.PubMedCentralPubMedCrossRef
124.
go back to reference Lodygensky GA, Menache C, Hüppi PS. Magnetic resonance imaging’s role in the care of the infant at risk for brain injury. In: Perlman J, editor. Neurology: neonatology questions and controversies. Philadelphia: Elsevier Health Sciences; 2011. p. 285–324. Lodygensky GA, Menache C, Hüppi PS. Magnetic resonance imaging’s role in the care of the infant at risk for brain injury. In: Perlman J, editor. Neurology: neonatology questions and controversies. Philadelphia: Elsevier Health Sciences; 2011. p. 285–324.
125.
go back to reference Inder T, Hüppi PS, Zientara GP, Maier SE, Jolesz FA, di Salvo D, et al. Early detection of periventricular leukomalacia by diffusion-weighted magnetic resonance imaging techniques. J Pediatr. 1999;134:631–4.PubMedCrossRef Inder T, Hüppi PS, Zientara GP, Maier SE, Jolesz FA, di Salvo D, et al. Early detection of periventricular leukomalacia by diffusion-weighted magnetic resonance imaging techniques. J Pediatr. 1999;134:631–4.PubMedCrossRef
126.
go back to reference Lodygensky GA, Marques JP, Maddage R, Perroud E, Sizonenko SV, Hüppi PS, et al. In vivo assessment of myelination by phase imaging at high magnetic field. Neuroimage. 2012;59:1979–87.PubMedCrossRef Lodygensky GA, Marques JP, Maddage R, Perroud E, Sizonenko SV, Hüppi PS, et al. In vivo assessment of myelination by phase imaging at high magnetic field. Neuroimage. 2012;59:1979–87.PubMedCrossRef
127.
go back to reference Song AW. Diffusion modulation of the fMRI signal: early investigations on the origin of the BOLD signal. Neuroimage. 2012;62:949–52.PubMedCrossRef Song AW. Diffusion modulation of the fMRI signal: early investigations on the origin of the BOLD signal. Neuroimage. 2012;62:949–52.PubMedCrossRef
128.
go back to reference Favrais G, van de Looij Y, Fleiss B, Ramanantsoa N, Bonnin P, Stoltenburg Didinger G, et al. Systemic inflammation disrupts the developmental program of white matter. Ann Neurol. 2011;70:550–65.PubMedCrossRef Favrais G, van de Looij Y, Fleiss B, Ramanantsoa N, Bonnin P, Stoltenburg Didinger G, et al. Systemic inflammation disrupts the developmental program of white matter. Ann Neurol. 2011;70:550–65.PubMedCrossRef
129.
go back to reference Counsell SJ, Allsop JM, Harrison MC, Larkman DJ, Kennea NL, Kapellou O, et al. Diffusion-weighted imaging of the brain in preterm infants with focal and diffuse white matter abnormality. Pediatrics. 2003;112:1–7.PubMedCrossRef Counsell SJ, Allsop JM, Harrison MC, Larkman DJ, Kennea NL, Kapellou O, et al. Diffusion-weighted imaging of the brain in preterm infants with focal and diffuse white matter abnormality. Pediatrics. 2003;112:1–7.PubMedCrossRef
130.
go back to reference Maalouf EF, Duggan PJ, Rutherford MA, Counsell SJ, Fletcher AM, Battin M, et al. Magnetic resonance imaging of the brain in a cohort of extremely preterm infants. J Pediatr. 1999;135:351–7.PubMedCrossRef Maalouf EF, Duggan PJ, Rutherford MA, Counsell SJ, Fletcher AM, Battin M, et al. Magnetic resonance imaging of the brain in a cohort of extremely preterm infants. J Pediatr. 1999;135:351–7.PubMedCrossRef
131.
go back to reference Tkác I, Rao R, Georgieff MK, Gruetter R. Developmental and regional changes in the neurochemical profile of the rat brain determined by in vivo 1H NMR spectroscopy. Magn Reson Med. 2003;50:24–32.PubMedCrossRef Tkác I, Rao R, Georgieff MK, Gruetter R. Developmental and regional changes in the neurochemical profile of the rat brain determined by in vivo 1H NMR spectroscopy. Magn Reson Med. 2003;50:24–32.PubMedCrossRef
132.
go back to reference Lodygensky GA, Inder TE, Neil JJ. Application of magnetic resonance imaging in animal models of perinatal hypoxic-ischemic cerebral injury. Int J Dev Neurosci. 2008;26:13–25.PubMedCentralPubMedCrossRef Lodygensky GA, Inder TE, Neil JJ. Application of magnetic resonance imaging in animal models of perinatal hypoxic-ischemic cerebral injury. Int J Dev Neurosci. 2008;26:13–25.PubMedCentralPubMedCrossRef
133.
go back to reference López-Villegas D, Lenkinski RE, Wehrli SL, Ho WZ, Douglas SD. Lactate production by human monocytes/macrophages determined by proton MR spectroscopy. Magn Reson Med. 1995;34:32–8.PubMedCrossRef López-Villegas D, Lenkinski RE, Wehrli SL, Ho WZ, Douglas SD. Lactate production by human monocytes/macrophages determined by proton MR spectroscopy. Magn Reson Med. 1995;34:32–8.PubMedCrossRef
134.
go back to reference Groenendaal F, Veenhoven RH, van der Grond J, Jansen GH, Witkamp TD, de Vries LS. Cerebral lactate and N-acetyl-aspartate/choline ratios in asphyxiated full-term neonates demonstrated in vivo using proton magnetic resonance spectroscopy. Pediatr Res. 1994;35:148–51.PubMedCrossRef Groenendaal F, Veenhoven RH, van der Grond J, Jansen GH, Witkamp TD, de Vries LS. Cerebral lactate and N-acetyl-aspartate/choline ratios in asphyxiated full-term neonates demonstrated in vivo using proton magnetic resonance spectroscopy. Pediatr Res. 1994;35:148–51.PubMedCrossRef
135.
go back to reference Barkovich AJ, Baranski K, Vigneron D, Partridge JC, Hallam DK, Hajnal BL, et al. Proton MR spectroscopy for the evaluation of brain injury in asphyxiated, term neonates. AJNR Am J Neuroradiol. 1999;20:1399–405.PubMed Barkovich AJ, Baranski K, Vigneron D, Partridge JC, Hallam DK, Hajnal BL, et al. Proton MR spectroscopy for the evaluation of brain injury in asphyxiated, term neonates. AJNR Am J Neuroradiol. 1999;20:1399–405.PubMed
136.
go back to reference Girard S, Tremblay L, Lepage M, Sébire G. Early detection of placental inflammation by MRI enabling protection by clinically relevant IL-1Ra administration. Am J Obstetr Gynecol. 2012;206:358. e1–9.CrossRef Girard S, Tremblay L, Lepage M, Sébire G. Early detection of placental inflammation by MRI enabling protection by clinically relevant IL-1Ra administration. Am J Obstetr Gynecol. 2012;206:358. e1–9.CrossRef
137.
go back to reference Drobyshevsky A, Prasad PV. Placental perfusion in uterine ischemia model as evaluated by dynamic contrast enhanced MRI. J Magn Reson Imaging. 2015:n/a–n/a Drobyshevsky A, Prasad PV. Placental perfusion in uterine ischemia model as evaluated by dynamic contrast enhanced MRI. J Magn Reson Imaging. 2015:n/a–n/a
138.
go back to reference Linduska N, Dekan S, Messerschmidt A, Kasprian G, Brugger PC, Chalubinski K, et al. Placental pathologies in fetal MRI with pathohistological correlation. Placenta. 2009;30:555–9.PubMedCrossRef Linduska N, Dekan S, Messerschmidt A, Kasprian G, Brugger PC, Chalubinski K, et al. Placental pathologies in fetal MRI with pathohistological correlation. Placenta. 2009;30:555–9.PubMedCrossRef
139.
go back to reference Sohlberg S, Mulic-Lutvica A, Olovsson M, Weis J, Axelsson O, Wikström J, Wikström A-K. MRI estimated placental perfusion in fetal growth assessment. Ultrasound Obstet Gynecol. 2015:n/a–n/a. Sohlberg S, Mulic-Lutvica A, Olovsson M, Weis J, Axelsson O, Wikström J, Wikström A-K. MRI estimated placental perfusion in fetal growth assessment. Ultrasound Obstet Gynecol. 2015:n/a–n/a.
140.
go back to reference Cagnin A, Kassiou M, Meikle SR, Banati RB. Positron emission tomography imaging of neuroinflammation. Neurotherapeutics. 2007;4:443–52.PubMedCrossRef Cagnin A, Kassiou M, Meikle SR, Banati RB. Positron emission tomography imaging of neuroinflammation. Neurotherapeutics. 2007;4:443–52.PubMedCrossRef
141.
go back to reference Hannestad J, Gallezot J-D, Schafbauer T, Lim K, Kloczynski T, Morris ED, et al. Endotoxin-induced systemic inflammation activates microglia: [11C]PBR28 positron emission tomography in nonhuman primates. Neuroimage. 2012;63:232–9.PubMedCentralPubMedCrossRef Hannestad J, Gallezot J-D, Schafbauer T, Lim K, Kloczynski T, Morris ED, et al. Endotoxin-induced systemic inflammation activates microglia: [11C]PBR28 positron emission tomography in nonhuman primates. Neuroimage. 2012;63:232–9.PubMedCentralPubMedCrossRef
142.
go back to reference Girard S, Sébire H, Brochu M-E, Briota S, Sarret P, Sébire G. Postnatal administration of IL-1Ra exerts neuroprotective effects following perinatal inflammation and/or hypoxic-ischemic injuries. Brain Behav Immun. 2012;26:1331–9.PubMedCrossRef Girard S, Sébire H, Brochu M-E, Briota S, Sarret P, Sébire G. Postnatal administration of IL-1Ra exerts neuroprotective effects following perinatal inflammation and/or hypoxic-ischemic injuries. Brain Behav Immun. 2012;26:1331–9.PubMedCrossRef
143.
go back to reference Opal SM, Fisher CJ, Dhainaut J-FA, Vincent J-L, Brase R, Lowry SF, et al. Confirmatory interleukin-1 receptor antagonist trial in severe sepsis: a phase III, randomized, doubleblind, placebo-controlled, multicenter trial. Crit Care Med. 1997;25:1115.PubMedCrossRef Opal SM, Fisher CJ, Dhainaut J-FA, Vincent J-L, Brase R, Lowry SF, et al. Confirmatory interleukin-1 receptor antagonist trial in severe sepsis: a phase III, randomized, doubleblind, placebo-controlled, multicenter trial. Crit Care Med. 1997;25:1115.PubMedCrossRef
144.
go back to reference Li S-J, Liu W, Wang J-L, Zhang Y, Zhao D-J, Wang T-J, et al. The role of TNF-α, IL-6, IL-10, and GDNF in neuronal apoptosis in neonatal rat with hypoxic-ischemic encephalopathy. Eur Rev Med Pharmacol Sci. 2014;18:905–9.PubMed Li S-J, Liu W, Wang J-L, Zhang Y, Zhao D-J, Wang T-J, et al. The role of TNF-α, IL-6, IL-10, and GDNF in neuronal apoptosis in neonatal rat with hypoxic-ischemic encephalopathy. Eur Rev Med Pharmacol Sci. 2014;18:905–9.PubMed
145.
go back to reference Gonzalez P, Burgaya F, Acarin L, Peluffo H, Castellano B, Gonzalez B. Interleukin-10 and interleukin-10 receptor-I are upregulated in glial cells after an excitotoxic injury to the postnatal rat brain. J Neuropathol Exp Neurol. 2009;68:391–403.PubMedCrossRef Gonzalez P, Burgaya F, Acarin L, Peluffo H, Castellano B, Gonzalez B. Interleukin-10 and interleukin-10 receptor-I are upregulated in glial cells after an excitotoxic injury to the postnatal rat brain. J Neuropathol Exp Neurol. 2009;68:391–403.PubMedCrossRef
146.
go back to reference Wallace KL, Lopez J, Shaffery JP, Wells A, Paul IA, Bennett WA. Interleukin-10/Ceftriaxone prevents E. coli-induced delays in sensorimotor task learning and spatial memory in neonatal and adult Sprague–Dawley rats. Brain Res Bull. 2010;81:141–8.PubMedCentralPubMedCrossRef Wallace KL, Lopez J, Shaffery JP, Wells A, Paul IA, Bennett WA. Interleukin-10/Ceftriaxone prevents E. coli-induced delays in sensorimotor task learning and spatial memory in neonatal and adult Sprague–Dawley rats. Brain Res Bull. 2010;81:141–8.PubMedCentralPubMedCrossRef
147.
go back to reference Mittal R, Gonzalez-Gomez I, Panigrahy A, Goth K, Bonnet R, Prasadarao NV. IL-10 administration reduces PGE-2 levels and promotes CR3-mediated clearance of Escherichia coli K1 by phagocytes in meningitis. J Exp Med. 2010;207:1307–19.PubMedCentralPubMedCrossRef Mittal R, Gonzalez-Gomez I, Panigrahy A, Goth K, Bonnet R, Prasadarao NV. IL-10 administration reduces PGE-2 levels and promotes CR3-mediated clearance of Escherichia coli K1 by phagocytes in meningitis. J Exp Med. 2010;207:1307–19.PubMedCentralPubMedCrossRef
148.
go back to reference Mesples B, Plaisant F, Gressens P. Effects of interleukin-10 on neonatal excitotoxic brain lesions in mice. Brain Res Dev Brain Res. 2003;141:25–32.PubMedCrossRef Mesples B, Plaisant F, Gressens P. Effects of interleukin-10 on neonatal excitotoxic brain lesions in mice. Brain Res Dev Brain Res. 2003;141:25–32.PubMedCrossRef
149.
go back to reference Rodts-Palenik S, Wyatt-Ashmead J, Pang Y, Thigpen B, Cai Z, Rhodes P, et al. Maternal infection-induced white matter injury is reduced by treatment with interleukin-10. Am J Obstetr Gynecol. 2004;191:1387–92.CrossRef Rodts-Palenik S, Wyatt-Ashmead J, Pang Y, Thigpen B, Cai Z, Rhodes P, et al. Maternal infection-induced white matter injury is reduced by treatment with interleukin-10. Am J Obstetr Gynecol. 2004;191:1387–92.CrossRef
150.
go back to reference Lechpammer M, Manning SM, Samonte F, Nelligan J, Sabo E, Talos DM, et al. Minocycline treatment following hypoxic/ischaemic injury attenuates white matter injury in a rodent model of periventricular leucomalacia. Neuropathol Appl Neurobiol. 2008;34:379–93.PubMedCentralPubMedCrossRef Lechpammer M, Manning SM, Samonte F, Nelligan J, Sabo E, Talos DM, et al. Minocycline treatment following hypoxic/ischaemic injury attenuates white matter injury in a rodent model of periventricular leucomalacia. Neuropathol Appl Neurobiol. 2008;34:379–93.PubMedCentralPubMedCrossRef
151.
go back to reference Cai Z, Lin S, Fan L-W, Pang Y, Rhodes PG. Minocycline alleviates hypoxic–ischemic injury to developing oligodendrocytes in the neonatal rat brain. Neuroscience. 2006;137:425–35.PubMedCrossRef Cai Z, Lin S, Fan L-W, Pang Y, Rhodes PG. Minocycline alleviates hypoxic–ischemic injury to developing oligodendrocytes in the neonatal rat brain. Neuroscience. 2006;137:425–35.PubMedCrossRef
152.
go back to reference Fan L-W, Pang Y, Lin S, Rhodes PG, Cai Z. Minocycline attenuates lipopolysaccharide-induced white matter injury in the neonatal rat brain. Neuroscience. 2005;133:159–68.PubMedCrossRef Fan L-W, Pang Y, Lin S, Rhodes PG, Cai Z. Minocycline attenuates lipopolysaccharide-induced white matter injury in the neonatal rat brain. Neuroscience. 2005;133:159–68.PubMedCrossRef
153.
go back to reference Zhu F, Zheng Y, Ding Y-Q, Liu Y, Zhang X, Wu R, et al. Minocycline and risperidone prevent microglia activation and rescue behavioral deficits induced by neonatal intrahippocampal injection of lipopolysaccharide in rats. PLoS One. 2014;9:e93966.PubMedCentralPubMedCrossRef Zhu F, Zheng Y, Ding Y-Q, Liu Y, Zhang X, Wu R, et al. Minocycline and risperidone prevent microglia activation and rescue behavioral deficits induced by neonatal intrahippocampal injection of lipopolysaccharide in rats. PLoS One. 2014;9:e93966.PubMedCentralPubMedCrossRef
154.
go back to reference Fan LW, Pang Y, Lin S, Tien LT, Ma T, Rhodes PG, et al. Minocycline reduces lipopolysaccharide-induced neurological dysfunction and brain injury in the neonatal rat. J Neurosci Res. 2005;82:71–82.PubMedCrossRef Fan LW, Pang Y, Lin S, Tien LT, Ma T, Rhodes PG, et al. Minocycline reduces lipopolysaccharide-induced neurological dysfunction and brain injury in the neonatal rat. J Neurosci Res. 2005;82:71–82.PubMedCrossRef
155.
go back to reference Filipovic R, Zecevic N. Neuroprotective role of minocycline in co-cultures of human fetal neurons and microglia. Exp Neurol. 2008;211:41–51.PubMedCrossRef Filipovic R, Zecevic N. Neuroprotective role of minocycline in co-cultures of human fetal neurons and microglia. Exp Neurol. 2008;211:41–51.PubMedCrossRef
156.
go back to reference Robertson NJ, Faulkner S, Fleiss B, Bainbridge A, Andorka C, Price D, et al. Melatonin augments hypothermic neuroprotection in a perinatal asphyxia model. Brain. 2013;136:90–105.PubMedCrossRef Robertson NJ, Faulkner S, Fleiss B, Bainbridge A, Andorka C, Price D, et al. Melatonin augments hypothermic neuroprotection in a perinatal asphyxia model. Brain. 2013;136:90–105.PubMedCrossRef
157.
go back to reference Wong C-S, Jow G-M, Kaizaki A, Fan L-W, Tien L-T. Melatonin ameliorates brain injury induced by systemic lipopolysaccharide in neonatal rats. Neuroscience. 2014;267:147–56.PubMedCentralPubMedCrossRef Wong C-S, Jow G-M, Kaizaki A, Fan L-W, Tien L-T. Melatonin ameliorates brain injury induced by systemic lipopolysaccharide in neonatal rats. Neuroscience. 2014;267:147–56.PubMedCentralPubMedCrossRef
158.
go back to reference Gressens P, Schwendimann L, Husson I, Sarkozy G, Mocaer E, Vamecq J, et al. Agomelatine, a melatonin receptor agonist with 5-HT(2C) receptor antagonist properties, protects the developing murine white matter against excitotoxicity. Eur J Pharmacol. 2008;588:58–63.PubMedCrossRef Gressens P, Schwendimann L, Husson I, Sarkozy G, Mocaer E, Vamecq J, et al. Agomelatine, a melatonin receptor agonist with 5-HT(2C) receptor antagonist properties, protects the developing murine white matter against excitotoxicity. Eur J Pharmacol. 2008;588:58–63.PubMedCrossRef
159.
go back to reference Balduini W, Carloni S, Perrone S, Bertrando S, Tataranno ML, Negro S, et al. The use of melatonin in hypoxic-ischemic brain damage: an experimental study. J Matern Fetal Neonatal Med. 2012;25 Suppl 1:119–24.PubMedCrossRef Balduini W, Carloni S, Perrone S, Bertrando S, Tataranno ML, Negro S, et al. The use of melatonin in hypoxic-ischemic brain damage: an experimental study. J Matern Fetal Neonatal Med. 2012;25 Suppl 1:119–24.PubMedCrossRef
160.
go back to reference Guven A, Uysal B, Gundogdu G, Oztas E, Ozturk H, Korkmaz A. Melatonin ameliorates necrotizing enterocolitis in a neonatal rat model. J Pediatr Surg. 2011;46:2101–7.PubMedCrossRef Guven A, Uysal B, Gundogdu G, Oztas E, Ozturk H, Korkmaz A. Melatonin ameliorates necrotizing enterocolitis in a neonatal rat model. J Pediatr Surg. 2011;46:2101–7.PubMedCrossRef
161.
go back to reference Cekmez F, Cetinkaya M, Tayman C, Canpolat FE, Kafa IM, Uysal S, et al. Evaluation of melatonin and prostaglandin E1 combination on necrotizing enterocolitis model in neonatal rats. Regul Pept. 2013;184:121–5.PubMedCrossRef Cekmez F, Cetinkaya M, Tayman C, Canpolat FE, Kafa IM, Uysal S, et al. Evaluation of melatonin and prostaglandin E1 combination on necrotizing enterocolitis model in neonatal rats. Regul Pept. 2013;184:121–5.PubMedCrossRef
162.
go back to reference Xiong T, Qu Y, Mu D, Ferriero D. Erythropoietin for neonatal brain injury: opportunity and challenge. Int J Dev Neurosci. 2011;29:583–91.PubMedCrossRef Xiong T, Qu Y, Mu D, Ferriero D. Erythropoietin for neonatal brain injury: opportunity and challenge. Int J Dev Neurosci. 2011;29:583–91.PubMedCrossRef
163.
go back to reference Mohamad O, Chen D, Zhang L, Hofmann C, Wei L, Yu SP. Erythropoietin reduces neuronal cell death and hyperalgesia induced by peripheral inflammatory pain in neonatal rats. Mol Pain. 2011;7:51.PubMedCentralPubMedCrossRef Mohamad O, Chen D, Zhang L, Hofmann C, Wei L, Yu SP. Erythropoietin reduces neuronal cell death and hyperalgesia induced by peripheral inflammatory pain in neonatal rats. Mol Pain. 2011;7:51.PubMedCentralPubMedCrossRef
164.
go back to reference Liu W, Shen Y, Plane JM, Pleasure DE, Deng W. Neuroprotective potential of erythropoietin and its derivative carbamylated erythropoietin in periventricular leukomalacia. Exp Neurol. 2011;230:227–39.PubMedCentralPubMedCrossRef Liu W, Shen Y, Plane JM, Pleasure DE, Deng W. Neuroprotective potential of erythropoietin and its derivative carbamylated erythropoietin in periventricular leukomalacia. Exp Neurol. 2011;230:227–39.PubMedCentralPubMedCrossRef
165.
go back to reference Juul S. Neuroprotective role of erythropoietin in neonates. J Matern Fetal Neonatal Med. 2012;25 Suppl 4:105–7.PubMed Juul S. Neuroprotective role of erythropoietin in neonates. J Matern Fetal Neonatal Med. 2012;25 Suppl 4:105–7.PubMed
166.
go back to reference Gitto E, Karbownik M, Reiter RJ, Tan DX, Cuzzocrea S, Chiurazzi P, et al. Effects of melatonin treatment in septic newborns. Pediatr Res. 2001;50:756–60.PubMedCrossRef Gitto E, Karbownik M, Reiter RJ, Tan DX, Cuzzocrea S, Chiurazzi P, et al. Effects of melatonin treatment in septic newborns. Pediatr Res. 2001;50:756–60.PubMedCrossRef
168.
go back to reference Merchant NM, Azzopardi DV, Counsell S, Gressens P, Dierl A, Gozar I et al. O-057 Melatonin As A Novel Neuroprotectant In Preterm Infants – A Double Blinded Randomised Controlled Trial (mint Study). Arch Dis Child. 2014;99:A43. Merchant NM, Azzopardi DV, Counsell S, Gressens P, Dierl A, Gozar I et al. O-057 Melatonin As A Novel Neuroprotectant In Preterm Infants – A Double Blinded Randomised Controlled Trial (mint Study). Arch Dis Child. 2014;99:A43.
169.
go back to reference Leuchter RH-V, Gui L, Poncet A, Hagmann C, Lodygensky GA, Martin E, et al. Association between early administration of high-dose erythropoietin in preterm infants and brain MRI abnormality at term-equivalent age. JAMA. 2014;312:817–24.PubMedCrossRef Leuchter RH-V, Gui L, Poncet A, Hagmann C, Lodygensky GA, Martin E, et al. Association between early administration of high-dose erythropoietin in preterm infants and brain MRI abnormality at term-equivalent age. JAMA. 2014;312:817–24.PubMedCrossRef
170.
go back to reference O’Gorman RL, Bucher HU, Held U, Koller BM, Hüppi PS, Hagmann CF, et al. Tract-based spatial statistics to assess the neuroprotective effect of early erythropoietin on white matter development in preterm infants. Brain. 2015;138:388–97.PubMedCrossRef O’Gorman RL, Bucher HU, Held U, Koller BM, Hüppi PS, Hagmann CF, et al. Tract-based spatial statistics to assess the neuroprotective effect of early erythropoietin on white matter development in preterm infants. Brain. 2015;138:388–97.PubMedCrossRef
171.
go back to reference Barrington KJ. The adverse neuro-developmental effects of postnatal steroids in the preterm infant: a systematic review of RCTs. BMC Pediatr. 2001;1:1.PubMedCentralPubMedCrossRef Barrington KJ. The adverse neuro-developmental effects of postnatal steroids in the preterm infant: a systematic review of RCTs. BMC Pediatr. 2001;1:1.PubMedCentralPubMedCrossRef
172.
go back to reference Shinwell ES, Karplus M, Reich D, Weintraub Z, Blazer S, Bader D, et al. Early postnatal dexamethasone treatment and increased incidence of cerebral palsy. Arch Dis Child Fetal Neonatal Ed. 2000;83:F177–81.PubMedCentralPubMedCrossRef Shinwell ES, Karplus M, Reich D, Weintraub Z, Blazer S, Bader D, et al. Early postnatal dexamethasone treatment and increased incidence of cerebral palsy. Arch Dis Child Fetal Neonatal Ed. 2000;83:F177–81.PubMedCentralPubMedCrossRef
173.
go back to reference Murphy BP, Inder TE, Huppi PS, Warfield S, Zientara GP, Kikinis R, et al. Impaired cerebral cortical gray matter growth after treatment with dexamethasone for neonatal chronic lung disease. Pediatrics. 2001;107:217–21.PubMedCrossRef Murphy BP, Inder TE, Huppi PS, Warfield S, Zientara GP, Kikinis R, et al. Impaired cerebral cortical gray matter growth after treatment with dexamethasone for neonatal chronic lung disease. Pediatrics. 2001;107:217–21.PubMedCrossRef
174.
go back to reference Wilson-Costello D, Walsh MC, Langer JC, Guillet R, Laptook AR, Stoll BJ, et al. Impact of postnatal corticosteroid use on neurodevelopment at 18 to 22 months’ adjusted age: effects of dose, timing, and risk of bronchopulmonary dysplasia in extremely low birth weight infants. Pediatrics. 2009;123:e430–7.PubMedCentralPubMedCrossRef Wilson-Costello D, Walsh MC, Langer JC, Guillet R, Laptook AR, Stoll BJ, et al. Impact of postnatal corticosteroid use on neurodevelopment at 18 to 22 months’ adjusted age: effects of dose, timing, and risk of bronchopulmonary dysplasia in extremely low birth weight infants. Pediatrics. 2009;123:e430–7.PubMedCentralPubMedCrossRef
175.
go back to reference Lodygensky GA, Rademaker K, Zimine S, Gex-Fabry M, Lieftink AF, Lazeyras F, et al. Structural and functional brain development after hydrocortisone treatment for neonatal chronic lung disease. Pediatrics. 2005;116:1–7.PubMedCrossRef Lodygensky GA, Rademaker K, Zimine S, Gex-Fabry M, Lieftink AF, Lazeyras F, et al. Structural and functional brain development after hydrocortisone treatment for neonatal chronic lung disease. Pediatrics. 2005;116:1–7.PubMedCrossRef
176.
go back to reference Benders MJNL, Groenendaal F, van Bel F, Ha Vinh R, Dubois J, Lazeyras F, et al. Brain development of the preterm neonate after neonatal hydrocortisone treatment for chronic lung disease. Pediatr Res. 2009;66:555–9.PubMedCentralPubMedCrossRef Benders MJNL, Groenendaal F, van Bel F, Ha Vinh R, Dubois J, Lazeyras F, et al. Brain development of the preterm neonate after neonatal hydrocortisone treatment for chronic lung disease. Pediatr Res. 2009;66:555–9.PubMedCentralPubMedCrossRef
177.
go back to reference Watterberg KL, Shaffer ML, Mishefske MJ, Leach CL, Mammel MC, Couser RJ, et al. Growth and neurodevelopmental outcomes after early low-dose hydrocortisone treatment in extremely low birth weight infants. Pediatrics. 2007;120:40–8.PubMedCrossRef Watterberg KL, Shaffer ML, Mishefske MJ, Leach CL, Mammel MC, Couser RJ, et al. Growth and neurodevelopmental outcomes after early low-dose hydrocortisone treatment in extremely low birth weight infants. Pediatrics. 2007;120:40–8.PubMedCrossRef
Metadata
Title
New means to assess neonatal inflammatory brain injury
Authors
Chen Jin
Irene Londono
Carina Mallard
Gregory A. Lodygensky
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/s12974-015-0397-2

Other articles of this Issue 1/2015

Journal of Neuroinflammation 1/2015 Go to the issue