Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-nmvwc Total loading time: 0 Render date: 2024-06-22T19:27:46.225Z Has data issue: false hasContentIssue false

Chapter 4 - The Pathogenesis of Preterm Brain Injury

from Section 1 - Epidemiology, Pathophysiology, and Pathogenesis of Fetal and Neonatal Brain Injury

Published online by Cambridge University Press:  13 December 2017

David K. Stevenson
Affiliation:
Stanford University, California
William E. Benitz
Affiliation:
Stanford University, California
Philip Sunshine
Affiliation:
Stanford University, California
Susan R. Hintz
Affiliation:
Stanford University, California
Maurice L. Druzin
Affiliation:
Stanford University, California
Get access

Summary

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2017

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Wilson-Costello, D, Friedman, H, Minich, N, et al. Improved survival rates with increased neurodevelopmental disability for extremely low birth weight infants in the 1990s. Pediatrics 2005; 115: 9971003.CrossRefGoogle ScholarPubMed
Moore, T, Hennessy, EM, Myles, J, et al. Neurological and developmental outcome in extremely preterm children born in England in 1995 and 2006: the EPICure studies. BMJ (Clinical Research Edition) 2012; 345: e7961.Google Scholar
Schlapbach, LJ, Adams, M, Proietti, E, et al. Outcome at two years of age in a Swiss national cohort of extremely preterm infants born between 2000 and 2008. BMC Pediatrics 2012; 12: 198.Google Scholar
Hintz, SR, Kendrick, DE, Wilson-Costello, DE, et al. Early-childhood neurodevelopmental outcomes are not improving for infants born at <25 weeks’ gestational age. Pediatrics 2011; 127: 6270.Google Scholar
Wilson-Costello, D, Friedman, H, Minich, N, et al. Improved neurodevelopmental outcomes for extremely low birth weight infants in 2000–2002. Pediatrics 2007; 119: 3745.Google Scholar
Hack, M. Young adult outcomes of very-low-birth-weight children. Semin Fetal Neonatal Med 2006; 11: 127–37.Google Scholar
Baron, IS, Erickson, K, Ahronovich, MD, et al. Cognitive deficit in preschoolers born late preterm. Early Hum Dev. 2011; 87: 115–9.Google Scholar
Committee on Understanding Premature Birth and Assuring Healthy Outcomes, Behrman, RE, Butler, AS, eds. Preterm Birth: Causes, Consequences, and Prevention. Washington, DC: Institute of Medicine of the National Academies, 2007. Available from http://books.nap.edu/openbook.php?record_id=11622&page=1.Google Scholar
Saigal, S, Doyle, LW. An overview of mortality and sequelae of preterm birth from infancy to adulthood. Lancet 2008; 371: 261–9.CrossRefGoogle ScholarPubMed
Perlman, JM. White matter injury in the preterm infant: an important determination of abnormal neurodevelopment outcome. Early Hum Dev 1998; 53: 99120.Google Scholar
Andiman, SE, Haynes, RL, Trachtenberg, FL, et al. The cerebral cortex overlying periventricular leukomalacia: analysis of pyramidal neurons. Brain Pathol 2010; 20: 803–14.Google Scholar
Reid, SM, Ditchfield, MR, Bracken, J, Reddihough, DS. Relationship between characteristics on magnetic resonance imaging and motor outcomes in children with cerebral palsy and white matter injury. Res Dev Disabil 2015; 45–6: 178–87.Google Scholar
Okumura, A, Kato, T, Kuno, K, et al. MRI findings in patients with spastic cerebral palsy. II. Correlation with type of cerebral palsy. Dev Med Child Neurol 1997; 39: 369–72.Google ScholarPubMed
Hamrick, SE, Miller, SP, Leonard, C, et al. Trends in severe brain injury and neurodevelopmental outcome in premature newborn infants: the role of cystic periventricular leukomalacia. J Pediatr 2004; 145: 593–9.Google Scholar
Buser, JR, Maire, J, Riddle, A, et al. Arrested preoligodendrocyte maturation contributes to myelination failure in premature infants. Ann Neurol 2012; 71: 93109.Google Scholar
Gano, D, Andersen, SK, Partridge, JC, et al. Diminished white matter injury over time in a cohort of premature newborns. J Pediatr 2015; 166: 3943.CrossRefGoogle Scholar
Haynes, RL, Billiards, SS, Borenstein, NS, et al. Diffuse axonal injury in periventricular leukomalacia as determined by apoptotic marker fractin. Pediatr Res 2008; 63: 656–61.Google Scholar
Riddle, A, Dean, J, Buser, JR, et al. Histopathological correlates of magnetic resonance imaging-defined chronic perinatal white matter injury. Ann Neurol 2011; 70: 493507.Google Scholar
Riddle, A, Maire, J, Gong, X, et al. Differential susceptibility to axonopathy in necrotic and non-necrotic perinatal white matter injury. Stroke 2012; 43: 178–84.Google Scholar
Back, SA, Luo, NL, Mallinson, RA, et al. Selective vulnerability of preterm white matter to oxidative damage defined by F2-isoprostanes. Ann Neurol 2005; 58: 108–20.Google Scholar
Segovia, KN, McClure, M, Moravec, M, et al. Arrested oligodendrocyte lineage maturation in chronic perinatal white matter injury. Ann Neurol 2008; 63: 520–30.Google Scholar
Franklin, RJ, Gallo, V. The translational biology of remyelination: past, present, and future. Glia 2014; 62: 1905–15.Google Scholar
Billiards, SS, Haynes, RL, Folkerth, RD, et al. Myelin abnormalities without oligodendrocyte loss in periventricular leukomalacia. Brain Pathol 2008; 18: 153–63.Google Scholar
Verney, C, Pogledic, I, Biran, V, et al. 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.Google Scholar
Davidson, JO, Drury, PP, Green, CR, et al. Connexin hemichannel blockade is neuroprotective after asphyxia in preterm fetal sheep. PLoS One 2014; 9: e96558.Google Scholar
Ritter, J, Schmitz, T, Chew, LJ, et al. Neonatal hyperoxia exposure disrupts axon-oligodendrocyte integrity in the subcortical white matter. J Neurosci 2013; 33: 89909002.Google Scholar
Jablonska, B, Scafidi, J, Aguirre, A, et al. Oligodendrocyte regeneration after neonatal hypoxia requires Fox O1-mediated p27Kip1 expression. J Neurosci 2012; 32: 14775–93.Google Scholar
Yuen, TJ, Silbereis, JC, Griveau, A, et al. Oligodendrocyte-encoded HIF function couples postnatal myelination and white matter angiogenesis. Cell 2014; 158: 383–96.Google Scholar
Tolcos, M, Bateman, E, O’Dowd, R, et al. Intrauterine growth restriction affects the maturation of myelin. Exp Neurol 2011; 232: 5365.Google Scholar
Brehmer, F, Bendix, I, Prager, S, et al. Interaction of inflammation and hyperoxia in a rat model of neonatal white matter damage. PloS One 2012; 7: e49023.CrossRefGoogle Scholar
Favrais, G, van de Looij, Y, Fleiss, B, et al. Systemic inflammation disrupts the developmental program of white matter. Ann Neurol 2011; 70: 550–65.Google Scholar
Nobuta, H, Ghiani, CA, Paez, PM, et al. STAT3-mediated astrogliosis protects myelin development in neonatal brain injury. Ann Neurol 2012; 72: 750–65.Google Scholar
Woodward, LJ, Anderson, PJ, Austin, NC, et al. Neonatal MRI to predict neurodevelopmental outcomes in preterm infants. N Engl J Med 2006; 355: 685–94.Google Scholar
Miller, SP, Ferriero, DM, Leonard, C, et al. Early brain injury in premature newborns detected with magnetic resonance imaging is associated with adverse early neurodevelopmental outcome. J Pediatr 2005; 147: 609–16.Google Scholar
Pierson, CR, Folkerth, RD, Billiards, SS, et al. Gray matter injury associated with periventricular leukomalacia in the premature infant. Acta Neuropathol 2007; 114: 619–31.Google Scholar
Lin, Y, Okumura, A, Hayakawa, F, et al. Quantitative evaluation of thalami and basal ganglia in infants with periventricular leukomalacia. Dev Med Child Neurol 2001; 43: 481–5.Google Scholar
Peterson, BS, Vohr, B, Staib, LH, et al. Regional brain volume abnormalities and long-term cognitive outcome in preterm infants. JAMA 2000; 284: 1939–47.Google Scholar
Counsell, SJ, Dyet, LE, Larkman, DJ, et al. Thalamo-cortical connectivity in children born preterm mapped using probabilistic magnetic resonance tractography. Neuroimage 2007; 34: 896904.Google Scholar
Ball, G, Boardman, JP, Rueckert, D, et al. The effect of preterm birth on thalamic and cortical development. Cereb Cortex 2012; 22: 1016–24.Google Scholar
Smyser, CD, Snyder, AZ, Shimony, JS, et al. Effects of white matter injury on resting state fMRI measures in prematurely born infants. PloS One 2013; 8: e68098.Google Scholar
Pitcher, JB, Riley, AM, Doeltgen, SH, et al. Physiological evidence consistent with reduced neuroplasticity in human adolescents born preterm. J Neurosci 2012; 32: 16410–6.Google Scholar
Pitcher, JB, Schneider, LA, Burns, NR, et al. Reduced corticomotor excitability and motor skills development in children born preterm. J Physiol 2012; 590: 5827–44.Google Scholar
Ajayi-Obe, M, Saeed, N, Cowan, FM, et al. Reduced development of cerebral cortex in extremely preterm infants. Lancet 2000; 356: 1162–3.Google Scholar
Nosarti, C, Al-Asady, MH, Frangou, S, et al. Adolescents who were born very preterm have decreased brain volumes. Brain 2002; 125: 1616–23.Google Scholar
Martinussen, M, Fischl, B, Larsson, HB, et al. Cerebral cortex thickness in 15-year-old adolescents with low birth weight measured by an automated MRI-based method. Brain 2005; 128: 2588–96.Google Scholar
Isaacs, EB, Edmonds, CJ, Chong, WK, et al. Brain morphometry and IQ measurements in preterm children. Brain 2004; 127: 2595–607.Google Scholar
Abernethy, LJ, Cooke, RW, Foulder-Hughes, L. Caudate and hippocampal volumes, intelligence, and motor impairment in 7-year-old children who were born preterm. Pediatr Res 2004; 55: 884–93.Google Scholar
Gimenez, M, Junque, C, Narberhaus, A, et al. Hippocampal gray matter reduction associates with memory deficits in adolescents with history of prematurity. Neuroimage 2004; 23: 869–77.Google Scholar
Felderhoff-Mueser, U, Rutherford, MA, Squier, WV, et al. Relationship between MR imaging and histopathologic findings of the brain in extremely sick preterm infants. AJNR Am J Neuroradiol 1999; 20: 1349–57.Google Scholar
Takizawa, Y, Takashima, S, Itoh, M. A histopathological study of premature and mature infants with pontosubicular neuron necrosis: neuronal cell death in perinatal brain damage. Brain Res 2006; 1095: 200–6.Google Scholar
Bell, JE, Becher, JC, Wyatt, B, et al. Brain damage and axonal injury in a Scottish cohort of neonatal deaths. Brain 2005; 128: 1070–81.Google Scholar
Barkovich, AJ, Sargent, SK. Profound asphyxia in the premature infant: imaging findings. AJNR Am J Neuroradiol 1995; 16: 1837–46.Google Scholar
Geddes, R, Vannucci, RC, Vannucci, SJ. Delayed cerebral atrophy following moderate hypoxia-ischemia in the immature rat. Dev Neurosci 2001; 23: 180–5.Google Scholar
Goldberg, JL, Barres, BA. The relationship between neuronal survival and regeneration. Annu Rev Neurosci 2000; 23: 579612.CrossRefGoogle ScholarPubMed
Jacobson, MD, Weil, M, Raff, MC. Programmed cell death in animal development. Cell 1997; 88: 347–54.Google Scholar
Meng, SZ, Arai, Y, Deguchi, K, Takashima, S. Early detection of axonal and neuronal lesions in prenatal-onset periventricular leukomalacia. Brain Dev 1997; 19: 480–4.Google Scholar
Hirayama, A, Okoshi, Y, Hachiya, Y, et al. Early immunohistochemical detection of axonal damage and glial activation in extremely immature brains with periventricular leukomalacia. Clin Neuropathol 2001; 20: 8791.Google Scholar
de Graaf-Peters, VB, Hadders-Algra, M. Ontogeny of the human central nervous system: what is happening when? Early Hum Dev 2006; 82: 257–66.Google Scholar
Dean, JM, McClendon, E, Hansen, K, et al. Prenatal cerebral ischemia disrupts MRI-defined cortical microstructure through disturbances in neuronal arborization. Sci Transl Med 2013; 5: 168ra7.Google Scholar
Sizonenko, SV, Camm, EJ, Garbow, JR, et al. Developmental changes and injury induced disruption of the radial organization of the cortex in the immature rat brain revealed by in vivo diffusion tensor MRI. Cereb Cortex 2007; 17: 2609–17.Google Scholar
Back, SA, Riddle, A, Dean, J, Hohimer, AR. The instrumented fetal sheep as a model of cerebral white matter injury in the premature infant. Neurotherapeutics 2012; 9: 359–70.Google Scholar
Vinall, J, Grunau, RE, Brant, R, et al. Slower postnatal growth is associated with delayed cerebral cortical maturation in preterm newborns. Sci Transl Med 2013; 5: 168ra8.Google Scholar
Ball, G, Srinivasan, L, Aljabar, P, et al. Development of cortical microstructure in the preterm human brain. Proc Natl Acad Sci USA 2013; 110: 9541–6.Google Scholar
Constable, RT, Ment, LR, Vohr, BR, et al. Prematurely born children demonstrate white matter microstructural differences at 12 years of age, relative to term control subjects: an investigation of group and gender effects. Pediatrics 2008; 121: 306–16.Google Scholar
de Vries, LS, Eken, P, Groenendaal, F, et al. Antenatal onset of haemorrhagic and/or ischaemic lesions in preterm infants: prevalence and associated obstetric variables. Arch Dis Child Fetal Neonatal Ed 1998; 78: F51–6.Google Scholar
Hayakawa, F, Okumura, A, Kato, T, et al. Determination of timing of brain injury in preterm infants with periventricular leukomalacia with serial neonatal electroencephalography. Pediatrics 1999; 104: 1077–81.Google Scholar
Becher, JC, Bell, JE, Keeling, JW, et al. The Scottish perinatal neuropathology study: clinicopathological correlation in early neonatal deaths. Arch Dis Child Fetal Neonatal Ed 2004; 89: F399407.CrossRefGoogle ScholarPubMed
Kubota, T, Okumura, A, Hayakawa, F, et al. Combination of neonatal electroencephalography and ultrasonography: sensitive means of early diagnosis of periventricular leukomalacia. Brain Dev 2002; 24: 698702.Google Scholar
Weinberger, B, Anwar, M, Hegyi, T, et al. Antecedents and neonatal consequences of low Apgar scores in preterm newborns: a population study. Arch Pediatr Adolesc Med 2000; 154: 294300.Google Scholar
Osborn, DA, Evans, N, Kluckow, M. Hemodynamic and antecedent risk factors of early and late periventricular/intraventricular hemorrhage in premature infants. Pediatrics 2003; 112: 33–9.Google Scholar
Low, JA. Determining the contribution of asphyxia to brain damage in the neonate. J Obstet Gynaecol Res 2004; 30: 276–86.Google Scholar
Salhab, WA, Perlman, JM. Severe fetal acidemia and subsequent neonatal encephalopathy in the larger premature infant. Pediatr Neurol 2005; 32: 25–9.Google Scholar
George, S, Gunn, AJ, Westgate, JA, et al. Fetal heart rate variability and brainstem injury after asphyxia in preterm fetal sheep. Am J Physiol Regul Integr Comp Physiol 2004; 287: R925–33.Google Scholar
Dean, JM, George, SA, Wassink, G, et al. Suppression of post hypoxic-ischemic EEG transients with dizocilpine is associated with partial striatal protection in the preterm fetal sheep. Neuropharmacology 2006; 50: 491503.Google Scholar
Dean, JM, Gunn, AJ, Wassink, G, et al. Endogenous alpha[2]-adrenergic receptor-mediated neuroprotection after severe hypoxia in preterm fetal sheep. Neuroscience 2006; 142: 615–28.Google Scholar
Bennet, L, Roelfsema, V, George, S, et al. The effect of cerebral hypothermia on white and gray matter injury induced by severe hypoxia in preterm fetal sheep. J Physiol 2007; 578: 491506.Google Scholar
Gunn, AJ, Quaedackers, JS, Guan, J, et al. The premature fetus: not as defenseless as we thought, but still paradoxically vulnerable? Dev Neurosci 2001; 23: 175–9.Google Scholar
Volpe, JJ. Neurobiology of periventricular leukomalacia in the premature infant. Pediatr Res 2001; 50: 553–62.Google Scholar
Riddle, A, Luo, NL, Manese, M, et al. Spatial heterogeneity in oligodendrocyte lineage maturation and not cerebral blood flow predicts fetal ovine periventricular white matter injury. J Neurosci 2006; 26: 3045–55.Google Scholar
Osborn, DA, Evans, N, Kluckow, M. Clinical detection of low upper body blood flow in very premature infants using blood pressure, capillary refill time, and central-peripheral temperature difference. Arch Dis Child Fetal Neonatal Ed 2004; 89: F168–73.Google Scholar
Kluckow, M, Evans, N. Superior vena cava flow in newborn infants: a novel marker of systemic blood flow. Arch Dis Child Fetal Neonatal Ed 2000; 82: F182–7.Google Scholar
Meek, JH, Elwell, CE, McCormick, DC, et al. Abnormal cerebral haemodynamics in perinatally asphyxiated neonates related to outcome. Arch Dis Child Fetal Neonatal Ed 1999; 81: F110–15.Google Scholar
Hunt, RW, Evans, N, Rieger, I, Kluckow, M. Low superior vena cava flow and neurodevelopment at 3 years in very preterm infants. J Pediatr 2004; 145: 588–92.Google Scholar
Lou, HC, Lassen, NA, Tweed, WA, et al. Pressure passive cerebral blood flow and breakdown of the blood-brain barrier in experimental fetal asphyxia. Acta Paediatr Scand 1979; 68: 5763.Google Scholar
Soul, JS, Hammer, PE, Tsuji, M, et al. Fluctuating pressure-passivity is common in the cerebral circulation of sick premature infants. Pediatr Res 2007; 61: 467–73.Google Scholar
Martens, SE, Rijken, M, Stoelhorst, GM, et al. Is hypotension a major risk factor for neurological morbidity at term age in very preterm infants? Early Hum Dev 2003; 75: 7989.Google Scholar
Murphy, DJ, Hope, PL, Johnson, A. Neonatal risk factors for cerebral palsy in very preterm babies: case-control study. BMJ 1997; 314: 404–8.Google Scholar
Low, JA, Froese, AB, Galbraith, RS, et al. The association between preterm newborn hypotension and hypoxemia and outcome during the first year. Acta Paediatr 1993; 82: 433–7.Google Scholar
Trounce, JQ, Shaw, DE, Levene, MI, Rutter, N. Clinical risk factors and periventricular leucomalacia. Arch Dis Child. 1988; 63: 1722.CrossRefGoogle ScholarPubMed
Perlman, JM, Risser, R, Broyles, RS. Bilateral cystic periventricular leukomalacia in the premature infant: associated risk factors. Pediatrics 1996; 97: 822–7.Google Scholar
Dammann, O, Allred, EN, Kuban, KC, et al. Systemic hypotension and white-matter damage in preterm infants. Dev Med Child Neurol 2002; 44: 8290.Google Scholar
Cunningham, S, Symon, AG, Elton, RA, et al. Intra-arterial blood pressure reference ranges, death and morbidity in very low birth weight infants during the first seven days of life. Early Hum Dev 1999; 56: 151–65.Google Scholar
Limperopoulos, C, Bassan, H, Kalish, LA, et al. Current definitions of hypotension do not predict abnormal cranial ultrasound findings in preterm infants. Pediatrics 2007; 120: 966–77.Google Scholar
Bennet, L, Booth, L, Malpas, SC, et al. Acute systemic complications in the preterm fetus after asphyxia: the role of cardiovascular and blood flow responses. Clin Exp Pharmacol Physiol 2006; 33: 291–9.Google Scholar
Osborn, DA. Diagnosis and treatment of preterm transitional circulatory compromise. Early Hum Dev 2005; 81: 413–22.Google Scholar
Tan, WK, Williams, CE, During, MJ, et al. Accumulation of cytotoxins during the development of seizures and edema after hypoxic-ischemic injury in late gestation fetal sheep. Pediatr Res 1996; 39: 791–7.Google Scholar
Choi, DW. Excitotoxic cell death. J Neurobiol 1992; 23: 1261–76.Google Scholar
Mitani, A, Andou, Y, Kataoka, K. Selective vulnerability of hippocampal CA1 neurons cannot be explained in terms of an increase in glutamate concentration during ischemia in the gerbil: brain microdialysis study. Neuroscience 1992; 48: 307–13.Google Scholar
McDonald, JW, Johnston, MV, Young, AB. Differential ontogenic development of three receptors comprising the NMDA receptor/channel complex in the rat hippocampus. Exp Neurol 1990; 110: 237–47.Google Scholar
Fern, R, Moller, T. Rapid ischemic cell death in immature oligodendrocytes: a fatal glutamate release feedback loop. J Neurosci 2000; 20: 3442.Google Scholar
Follett, PL, Rosenberg, PA, Volpe, JJ, Jensen, FE. NBQX attenuates excitotoxic injury in developing white matter. J Neurosci 2000; 20: 9235–41.Google Scholar
Rossi, DJ, Oshima, T, Attwell, D. Glutamate release in severe brain ischaemia is mainly by reversed uptake. Nature 2000; 403: 316–21.Google Scholar
Desilva, TM, Kinney, HC, Borenstein, NS, et al. The glutamate transporter EAAT2 is transiently expressed in developing human cerebral white matter. J Comp Neurol 2007; 501: 879–90.Google Scholar
Dean, JM, Fraser, M, Shelling, AN, et al. Ontogeny of AMPA and NMDA receptor gene expression in the developing sheep white matter and cerebral cortex. Brain Res Mol Brain Res 2005; 139: 242–50.Google Scholar
Itoh, T, Beesley, J, Itoh, A, et al. AMPA glutamate receptor-mediated calcium signaling is transiently enhanced during development of oligodendrocytes. J Neurochem 2002; 81: 390402.Google Scholar
Follett, PL, Deng, W, Dai, W, et al. Glutamate receptor-mediated oligodendrocyte toxicity in periventricular leukomalacia: a protective role for topiramate. J Neurosci 2004; 24: 4412–20.Google Scholar
Back, SA, Riddle, A, McClure, MM. Maturation-dependent vulnerability of perinatal white matter in premature birth. Stroke 2007; 38: 724–30.Google Scholar
Fraser, M, Bennet, L, van Zijl, PL, et al. Extracellular amino acids and peroxidation products in the periventricular white matter during and after cerebral ischemia in preterm fetal sheep. J Neurochem 2008; 105: 2214–23.Google Scholar
Fraser, M, Bennet, L, Gunning, M, et al. Cortical electroencephalogram suppression is associated with post-ischemic cortical injury in 0.65 gestation fetal sheep. Brain Res Dev Brain Res 2005; 154: 4555.Google Scholar
Dohmen, C, Kumura, E, Rosner, G, et al. Extracellular correlates of glutamate toxicity in short-term cerebral ischemia and reperfusion: a direct in vivo comparison between white and gray matter. Brain Res 2005; 1037: 4351.Google Scholar
Henderson, JL, Reynolds, JD, Dexter, F, et al. Chronic hypoxemia causes extracellular glutamate concentration to increase in the cerebral cortex of the near-term fetal sheep. Brain Res Dev Brain Res 1998; 105: 287–93.Google Scholar
Loeliger, M, Watson, CS, Reynolds, JD, et al. Extracellular glutamate levels and neuropathology in cerebral white matter following repeated umbilical cord occlusion in the near term fetal sheep. Neuroscience 2003; 116: 705–14.Google Scholar
Kumura, E, Graf, R, Dohmen, C, et al. Breakdown of calcium homeostasis in relation to tissue depolarization: comparison between gray and white matter ischemia. J Cereb Blood Flow Metab 1999; 19: 788–93.Google Scholar
Back, SA, Gan, X, Li, Y, et al. Maturation-dependent vulnerability of oligodendrocytes to oxidative stress-induced death caused by glutathione depletion. J Neurosci 1998; 18: 6241–53.Google Scholar
Rosin, C, Bates, TE, Skaper, SD. Excitatory amino acid induced oligodendrocyte cell death in vitro: receptor-dependent and -independent mechanisms. J Neurochem 2004; 90: 1173–85.Google Scholar
Inder, T, Mocatta, T, Darlow, B, et al. Elevated free radical products in the cerebrospinal fluid of VLBW infants with cerebral white matter injury. Pediatr Res 2002; 52: 213–8.Google Scholar
Haynes, RL, Folkerth, RD, Keefe, RJ, et al. Nitrosative and oxidative injury to premyelinating oligodendrocytes in periventricular leukomalacia. J Neuropathol Exp Neurol 2003; 62: 441–50.Google Scholar
Welin, AK, Sandberg, M, Lindblom, A, et al. White matter injury following prolonged free radical formation in the 0.65 gestation fetal sheep brain. Pediatr Res 2005; 58: 100–5.Google Scholar
Deng, W, Rosenberg, PA, Volpe, JJ, Jensen, FE. Calcium-permeable AMPA/kainate receptors mediate toxicity and preconditioning by oxygen-glucose deprivation in oligodendrocyte precursors. Proc Natl Acad Sci USA 2003; 100: 6801–6.Google Scholar
Willoughby, RE Jr., Nelson, KB. Chorioamnionitis and brain injury. Clin Perinatol 2002; 29: 603–21.Google Scholar
Wang, X, Rousset, CI, Hagberg, H, Mallard, C. Lipopolysaccharide-induced inflammation and perinatal brain injury. Semin Fetal Neonatal Med 2006; 11: 343–53.Google Scholar
Yoon, BH, Park, CW, Chaiworapongsa, T. Intrauterine infection and the development of cerebral palsy. BJOG 2003; 110: 124–7.Google Scholar
Holcroft, CJ, Blakemore, KJ, Allen, M, Graham, EM. Association of prematurity and neonatal infection with neurologic morbidity in very low birth weight infants. Obstet Gynecol 2003; 101: 1249–53.Google Scholar
Ellison, VJ, Mocatta, TJ, Winterbourn, CC, et al. The relationship of CSF and plasma cytokine levels to cerebral white matter injury in the premature newborn. Pediatr Res 2005; 57: 282–6.Google Scholar
Duggan, PJ, Maalouf, EF, Watts, TL, et al. Intrauterine T-cell activation and increased proinflammatory cytokine concentrations in preterm infants with cerebral lesions. Lancet 2001; 358: 1699–700.Google Scholar
Kadhim, H, Tabarki, B, Verellen, G, et al. Inflammatory cytokines in the pathogenesis of periventricular leukomalacia. Neurology 2001; 56: 1278–84.Google Scholar
Flenady, V, Hawley, G, Stock, OM, et al. Prophylactic antibiotics for inhibiting preterm labour with intact membranes. Cochrane Database of Systematic Reviews 2013; 12: CD000246.Google Scholar
Keogh, MJ, Bennet, L, Drury, PP, 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.Google Scholar
Dean, JM, van de Looij, Y, Sizonenko, SV, et al. Delayed cortical impairment following lipopolysaccharide exposure in preterm fetal sheep. Ann Neurol 2011; 70: 846–56.Google Scholar
Mathai, S, Booth, LC, Davidson, JO, et al. Acute on chronic exposure to endotoxin in preterm fetal sheep. Am J Physiol Regul Integr Comp Physiol 2013; 304: R189–97.Google Scholar
Hellstrom, IC, Danik, M, Luheshi, GN, Williams, S. Chronic LPS exposure produces changes in intrinsic membrane properties and a sustained IL-β-dependent increase in GABAergic inhibition in hippocampal CA1 pyramidal neurons. Hippocampus 2005; 15: 656–64.Google Scholar
Luk, WP, Zhang, Y, White, TD, et al. Adenosine: a mediator of interleukin-1beta-induced hippocampal synaptic inhibition. J Neurosci 1999; 19: 4238–44.Google Scholar
Summers deLuca, L, Gommerman, JL. Fine-tuning of dendritic cell biology by the TNF superfamily. Nat Rev Immunol 2012; 12: 339–51.Google Scholar
Kim, IJ, Beck, HN, Lein, PJ, Higgins, D. Interferon gamma induces retrograde dendritic retraction and inhibits synapse formation. J Neurosci 2002; 22: 4530–9.Google Scholar
Duncan, JR, Cock, ML, Scheerlinck, JP, et al. White matter injury after repeated endotoxin exposure in the preterm ovine fetus. Pediatr Res 2002; 52: 941–9.Google Scholar
Nitsos, I, Rees, SM, Duncan, J, et al. Chronic exposure to intra-amniotic lipopolysaccharide affects the ovine fetal brain. J Soc Gynecol Invest 2006; 13: 239–47.Google Scholar
Duncan, JR, Cock, ML, Suzuki, K, et al. Chronic endotoxin exposure causes brain injury in the ovine fetus in the absence of hypoxemia. J Soc Gynecol Invest 2006; 13: 8796.Google Scholar
Dommergues, MA, Patkai, J, Renauld, JC, et al. Proinflammatory cytokines and interleukin-9 exacerbate excitotoxic lesions of the newborn murine neopallium. Ann Neurol 2000; 47: 5463.Google Scholar
Basu, A, Lazovic, J, Krady, JK, et al. Interleukin-1 and the interleukin-1 type 1 receptor are essential for the progressive neurodegeneration that ensues subsequent to a mild hypoxic/ischemic injury. J Cereb Blood Flow Metab 2005; 25: 1729.Google Scholar
Loddick, SA, Wong, ML, Bongiorno, PB, et al. Endogenous interleukin-1 receptor antagonist is neuroprotective. Biochem Biophys Res Commun 1997; 234: 211–5.Google Scholar
Kremlev, SG, Palmer, C. Interleukin-10 inhibits endotoxin-induced pro-inflammatory cytokines in microglial cell cultures. J Neuroimmunol 2005; 162: 7180.Google Scholar
Loddick, SA, Turnbull, AV, Rothwell, NJ. Cerebral interleukin-6 is neuroprotective during permanent focal cerebral ischemia in the rat. J Cereb Blood Flow Metab 1998; 18: 176–9.Google Scholar
Guan, J, Miller, OT, Waugh, KM, et al. TGF-β1 and neurological function after hypoxia-ischemia in adult rats. Neuroreport 2004; 15: 961–4.Google Scholar
Spera, PA, Ellison, JA, Feuerstein, GZ, Barone, FC. IL-10 reduces rat brain injury following focal stroke. Neurosci Lett 1998; 251: 189–92.Google Scholar
Back, SA, Luo, NL, Borenstein, NS, et al. Late oligodendrocyte progenitors coincide with the developmental window of vulnerability for human perinatal white matter injury. J Neurosci 2001; 21: 1302–12.Google Scholar
Cai, Z, Lin, S, Pang, Y, Rhodes, PG. Brain injury induced by intracerebral injection of interleukin-1β and tumor necrosis factor α in the neonatal rat. Pediatr Res 2004; 56: 377–84.Google Scholar
Allan, SM, Rothwell, NJ. Inflammation in central nervous system injury. Philos Trans R Soc Lond B 2003; 358: 1669–77.Google Scholar
Woiciechowsky, C, Schoning, B, Stoltenburg-Didinger, G, et al. Brain-IL-1β triggers astrogliosis through induction of IL-6: inhibition by propranolol and IL-10. Med Sci Monit 2004; 10:BR325–30.Google Scholar
Bal-Price, A, Brown, GC. Inflammatory neurodegeneration mediated by nitric oxide from activated glia-inhibiting neuronal respiration, causing glutamate release and excitotoxicity. J Neurosci 2001; 21: 6480–91.Google Scholar
van den Tweel, ER, Nijboer, C, Kavelaars, A, et al. Expression of nitric oxide synthase isoforms and nitrotyrosine formation after hypoxia-ischemia in the neonatal rat brain. J Neuroimmunol 2005; 167: 6471.Google Scholar
Raivich, G, Bohatschek, M, Kloss, CU, et al. Neuroglial activation repertoire in the injured brain: graded response, molecular mechanisms and cues to physiological function. Brain Res Rev 1999; 30: 77105.Google Scholar
Rock, RB, Gekker, G, Hu, S, et al. Role of microglia in central nervous system infections. Clin Microbiol Rev 2004; 17: 942–64.Google Scholar
Pang, Y, Cai, Z, Rhodes, PG. Effects of lipopolysaccharide on oligodendrocyte progenitor cells are mediated by astrocytes and microglia. J Neurosci Res 2000; 62: 510–20.Google Scholar
Yan, E, Castillo-Melendez, M, Nicholls, T, Hirst, J, Walker, D. Cerebrovascular responses in the fetal sheep brain to low-dose endotoxin. Pediatr Res 2004; 55: 855–63.Google Scholar
Folkerth, RD, Keefe, RJ, Haynes, RL, et al. Interferon-gamma expression in periventricular leukomalacia in the human brain. Brain Pathol 2004; 14: 265–74.Google Scholar
Tahraoui, SL, Marret, S, Bodenant, C, et al. Central role of microglia in neonatal excitotoxic lesions of the murine periventricular white matter. Brain Pathol 2001; 11: 5671.Google Scholar
Kinney, HC. Human myelination and perinatal white matter disorders. J Neurol Sci 2005; 228: 190–2.Google Scholar
Fan, LW, Pang, Y, Lin, S, et al. Minocycline attenuates lipopolysaccharide-induced white matter injury in the neonatal rat brain. Neuroscience 2005; 133: 159–68.CrossRefGoogle ScholarPubMed
Pang, Y, Rodts-Palenik, S, Cai, Z, et al. 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.Google Scholar
Gunn, AJ, Bennet, L. Is temperature important in delivery room resuscitation? Semin Neonatol 2001; 6: 241–9.Google Scholar
Yoneyama, Y, Sawa, R, Kubonoya, K, et al. Evidence for mechanisms of the acute-phase response to endotoxin in late-gestation fetal goats. Am J Obstet Gynecol 1998; 179: 750–5.Google Scholar
Mallard, C, Welin, AK, Peebles, D, et al. White matter injury following systemic endotoxemia or asphyxia in the fetal sheep. Neurochem Res 2003; 28: 215–23.Google Scholar
Peebles, DM, Miller, S, Newman, JP, et al. The effect of systemic administration of lipopolysaccharide on cerebral haemodynamics and oxygenation in the 0.65 gestation ovine fetus in utero. BJOG 2003; 110: 735–43.Google Scholar
Nitsos, I, Moss, TJ, Cock, ML, et al. Fetal responses to intra-amniotic endotoxin in sheep. J Soc Gynecol Invest 2002; 9: 80–5.Google Scholar
Dean, JM, Bennet, L, Back, SA, et al. What brakes the preterm brain? An arresting story. Pediatr Res 2014; 75: 227–33.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×