Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-x4r87 Total loading time: 0 Render date: 2024-04-27T22:16:46.935Z Has data issue: false hasContentIssue false

Section 3 - Diagnosis of the Infant with 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

References

Finer, NN, Robertson, CM, Peters, KL, et al. Factors affecting outcome in hypoxic-ischemic encephalopathy in term infants. Am J Dis Child 1983; 137: 21–5.Google Scholar
Finer, NN, Robertson, CM, Richards, RT, et al. Hypoxic–ischemic encephalopathy in term neonates: perinatal factors and outcome. J Pediatr 1981; 98: 112–17.CrossRefGoogle ScholarPubMed
Mizrahi, EM, Kellaway, P. Characterization and classification of neonatal seizures. Neurology 1987; 37: 1837–44.Google Scholar
Roland, EH, Hill, A. Clinical aspects of perinatal hypoxic-ischemic brain injury. Semin Pediatr Neurol 1995; 2:5771.CrossRefGoogle ScholarPubMed
Vannucci, R. Hypoxic-ischemic encephalopathy. Am J Perinatol 2000; 17: 113–20.Google Scholar
Blair, E, Stanley, FJ. Intrapartum asphyxia: a rare cause of cerebral palsy. J Pediatr 1988; 112: 515–19.Google Scholar
Nelson, KB, Ellenberg, JH. Antecedents of cerebral palsy: multivariate analysis of risk. N Engl J Med 1986; 315: 81–6.Google Scholar
Nelson, KB. What proportion of cerebral palsy is related to birth asphyxia? J Pediatr 1988; 112: 572–4.Google Scholar
Naeye, RL, Peters, EC. Antenatal hypoxia and low IQ values. Am J Dis Child 1987; 141: 50–4.Google Scholar
Cowan, F, Rutherford, M, Groenendaal, F, et al. Origin and timing of brain lesions in term infants with neonatal encephalopathy. Lancet 2003; 361: 736–42.Google Scholar
Levene, MI, Grindulis, H, Sands, C, et al. Comparison of two methods of predicting outcome in perinatal asphyxia. Lancet 1986; 1: 67–9.Google Scholar
Sarnat, HB, Sarnat, MS. Neonatal encephalopathy following fetal distress: a clinical and electroencephalographic study. Arch Neurol 1976; 33: 696705.Google Scholar
Shankaran, S, Laptook, AR, Ehrenkranz, RA, et al. Whole-body hypothermia for neonates with hypoxic–ischemic encephalopathy. N Engl J Med 2005; 353: 1574–84.Google Scholar
de Vries, L, Jongmans, MJ. Long-term outcome after neonatal hypoxic-ischaemic encephalopathy Arch Dis Child Fetal Neonatal Ed 2010; 95: F220–4.Google Scholar
Mwaniki, MK, Atieno, M, Lawn, JE, Newton, CRJ. Long-term neurodevelopmental outcomes after intrauterine and neonatal insults: a systematic review. Lancet 2012; 279: 445–52.Google Scholar
Shankaran, S, Laptook, AR, Tyson, JE, et al. Evolution of encephalopathy during whole body hypothermia for neonatal hypoxic-ischemic encephalopathy. J Pediatr 2012; 160: 567–72.e3Google Scholar
Merchant, N, Azzopardi, D. Early predictors of outcome in infants treated with hypothermia for hypoxic-ischemic encephalopathy. Dev Med Child Neurol 2015; 57(Suppl. 3): 816.Google Scholar
Ramasawmy, V, Horton, J, Vandermeer, B, et al. Systematic review of biomarkers of brain injury in term neonatal encephalopathy. Pediatr Neurol 2009; 40: 215–26.Google Scholar
Chalak, LF, Sánchez, PJ, Adams-Huet, B, et al. Biomarkers for severity of neonatal hypoxic-ischemic encephalopathy and outcomes in newborns receiving hypothermia therapy. J Pediatr 2014; 164: 468–74.Google Scholar
Takeuchi, T, Watanabe, K. The EEG evolution and neurological prognosis of neonates with perinatal hypoxia. Brain Dev 1989; 11: 115–20.CrossRefGoogle ScholarPubMed
Scher, MS, Painter, MJ, Bergman, I, et al. EEG diagnosis of neonatal seizures: clinical correlations and outcome. Pediatr Neurol 1989; 5: 1724.CrossRefGoogle ScholarPubMed
Toet, MC, Hellström-Westas, L, Groenendaal, F, et al. Amplitude integrated EEG 3 and 6 hours after birth in full term neonates with hypoxicischemic encephalopathy. Arch Dis Child Fetal Neonatal Ed 1999; 81: F1923.Google Scholar
Van Laerhoven, H, de Haan, TR, Offriga, M, et al. Prognostic tests in term neonates with hypoxic-ischemic encephalopathy: a systematic review. Pediatrics 2013 131: 8898.CrossRefGoogle ScholarPubMed
Scalais, E, François-Adant, A, Nuttin, C, et al. Multimodality evoked potentials as a prognostic tool in term asphyxiated newborns. Electroencephalogr Clin Neurophysiol 1998; 108: 199207Google Scholar
Kuenzle, C, Baenziger, O, Martin, E, et al. Prognostic value of early MR imaging in term infants with severe perinatal asphyxia. Neuropediatrics 1994; 25: 191200.Google Scholar
Baenziger, O, Martin, E, Steinlin, M, et al. Early pattern recognition in severe perinatal asphyxia: a prospective MRI study. Neuroradiology 1993; 35: 437–42.Google Scholar
Triulzi, F, Parazzini, C, Righini, A. Patterns of damage in the mature neonatal brain. Pediatr Radiol 2006; 36: 608–20.Google Scholar
Miller, SP, Ramaswamy, V, Michelson, D, et al. Patterns of brain injury in term neonatal encephalopathy. J Pediatr 2005; 146: 153–60.Google Scholar
Chau, V, Poskitt, KJ, Dunham, CP, et al. Magnetic resonance imaging in the encephalopathic term newborn. Curr Pediatr Rev 2014; 10: 2836.Google Scholar
Hüppi, PS, Murphy, B, Maier, SE, et al. Microstructural brain development after perinatal cerebral white matter injury assessed by diffusion tensor magnetic resonance imaging. Pediatrics 2001; 107: 455–60.CrossRefGoogle ScholarPubMed
Robertson, RL, Ben-Sira, L, Barnes, PD, et al. MR line-scan diffusion-weighted imaging of term neonates with perinatal brain ischemia. Am J Neuroradiol 1999; 20: 658–70.Google ScholarPubMed
Myer, JE. Uber die Lokalisation frühkindlicher Hirnshäden in arteriellen Grenzgebieten. Arch Psychiatr Zeitschr Neurol 1953; 190: 328–41.Google Scholar
Volpe, JJ, Herscovitch, P, Perlman, JM, et al. Positron emission tomography in the asphyxiated term newborn: parasagittal impairment of cerebral blood flow. Ann Neurol 1985; 17: 287–96.Google Scholar
Volpe, JJ, Pasternak, JF. Parasagittal cerebral injury in neonatal hypoxic-ischemic encephalopathy: clinical and neuroradiological features. J Pediatr 1979; 91: 472–6.Google Scholar
Friede, RL. Developmental Neuropathology, 2nd edn. New York: Springer-Verlag, 1989.Google Scholar
Voit, T, Lemburg, P, Neuen, E, et al. Damage of thalamus and basal ganglia in asphyxiated full-term neonates. Neuropediatrics 1987; 18: 176–81.Google Scholar
Johnston, MV, Hoon, AH. Possible mechanisms in infants for selective basal ganglia damage from asphyxia, kernicterus, or mitochondrial encephalopathies. J Child Neurol 2000; 15: 588–91.CrossRefGoogle ScholarPubMed
Malamud, N, Hirano, A. Atlas of Neuropathology, 2nd edn. Berkeley:University of California Press, 1974.Google Scholar
Gilles, FH. Hypotensive brain stem necrosis: selective symmetrical necrosis of tegmental neuronal aggregates following cardiac arrest. Arch Pathol 1969; 88: 3241.Google Scholar
Rorke, LB. Pathology of Perinatal Brain Injury. New York:Raven Press, 1982.Google Scholar
Roland, EH, Hill, A, Norman, MG, et al. Selective brainstem injury in an asphyxiated newborn. Ann Neurol 1988; 23: 8992.CrossRefGoogle Scholar
Pasternak, JF, Gorey, MT. The syndrome of acute near-total intrauterine asphyxia in the term infant. Pediatr Neurol 1998; 18: 391–8.Google Scholar
Natsume, J, Watanabe, K, Kuno, K, et al. Clinical, neurophysiologic, and neuropathological features of an infant with brain damage of total asphyxia type (Myers). Pediatr Neurol 1995; 13: 61–4.Google Scholar
Barmada, MA, Moossy, J, Shuman, RM. Cerebral infarcts with arterial occlusion in neonates. Ann Neurol 1979; 6: 495502.CrossRefGoogle ScholarPubMed
Banker, BQ. Cerebral vascular disease in infancy and childhood. I. Occlusive vascular disease. J Neuropathol Exp Neurol 1961; 20: 127–40.CrossRefGoogle Scholar
Koelfen, W, Freund, M, Varnholt, V. Neonatal stroke involving the middle cerebral artery in term infants: clinical presentation, EEG and imaging studies, and outcome. Dev Med Child Neurol 1995; 37: 204–12.Google Scholar
Levine, SC, Huttenlocher, P, Banich, MT, et al. Factors affecting cognitive function of hemiplegic children. Dev Med Child Neurol 1987; 29: 2735.Google Scholar
Rivkin, MJ, Anderson, ML, Kaye, EM. Neonatal idiopathic cerebral venous thrombosis: an unrecognized cause of transient seizures or lethargy. Ann Neurol 1992; 32: 51–6.Google Scholar
Wong, VK, LeMesurier, J, Franceschini, R, et al. Cerebral venous thrombosis as a cause of neonatal seizures. Pediatr Neurol 1987; 3: 235–7.Google Scholar
Banker, BQ, Larroche, JC. Periventricular leukomalacia of infancy. Arch Neurol 1962; 7: 386410.Google Scholar
Guzzetta, F, Shackleford, GD, Volpe, S, et al. Periventricular intraparenchymal echodensities in the premature newborn: critical determination of neurologic outcome. Pediatrics 1986; 78: 9951006.Google Scholar
Fawer, CL, Calame, A, Perentes, E, et al. Periventricular leukomalacia: a correlation study between real-time ultrasound and autopsy findings. Neuroradiology 1985; 27: 292300.Google Scholar
Trounce, JQ, Rutter, N, Levene, MI. Periventricular leucomalacia and intraventricular haemorrhage in the preterm neonate. Arch Dis Child 1986; 16: 1196–202.Google Scholar
Trounce, JQ, Shaw, DE, Leverne, MI, et al. Clinical risk factors and periventricular leucomalacia. Arch Dis Child 1988; 63: 1722.Google Scholar
Sarkar, S, Shankaran, S, Laptook, AR et al. Screening cranial imaging at multiple time points improves cystic periventricular leukomalacia detection. Am J Perinatol 2015; 32 (epub ahead of print).Google Scholar
De Vries, LS, Connell, JA, Dubowitz, LMS, et al. Neurological, electrophysiological and MRI abnormalities in infants with extensive cystic leukomalacia. Neuropediatrics 1987; 18: 61–6.CrossRefGoogle ScholarPubMed
Dolfin, T, Skidmore, MB, Fong, KW, et al. Incidence, severity, and timing of subependymal and intraventricula hemorrhages in preterm infants born in a perinatal unit as detected by serial real-time ultrasound. Pediatrics 1983; 71: 541–6.CrossRefGoogle Scholar
Enzmann, D, Murphy-Irwin, K, Stevenson, D, et al. The natural history of subependymal germinal matrix hemorrhage. Am J Perinatol 1985; 2: 123–33.Google Scholar
Scher, MS, Wright, FS, Lockman, LA, et al. Intraventricular haemorrhage in the full-term neonate. Arch Neurol 1982; 39: 769–72.Google Scholar
Volpe, JJ. Intracranial hemorrhage:subdural, primary subarachnoid, cerebellar, intraventricular (term infant), and miscellaneous. In Neurology of the Newborn, 5th edn. Philadelphia: Saunders. 2008: 483516.Google Scholar
Bouwer, AJ, Groenendaal, F, Benders, MJ, de Vries, LS. Early and late complications of germinal matrix-intraventricular haemorrhage in the preterm infant: what is new? Neonatology 2014; 106: 296303.Google Scholar

References

Guideline II: Minimum technical standards for pediatric electroencephalography, April 2016. Available at www.acns.org/practice/guidelines.Google Scholar
Shellhaas, RA, Chang, T, Tsuchida, T, et al. The American Clinical Neurophysiology Society’s guideline on continuous electroencephalography monitoring in neonates. J Clin Neurophysiol 2011; 28(6): 611–7.Google Scholar
Tsuchida, TN, Wusthoff, CJ, Shellhaas, RA, et al. American Clinical Neurophysiology Society standardized EEG terminology and categorization for the description of continuous EEG monitoring in neonates: report of the American Clinical Neurophysiology Society critical care monitoring committee. J Clin Neurophysiol 2013; 30(2): 161–73.Google Scholar
Neurodiagnostic Society. Skin safety during EEG procedures, April 2016. Available at www.aset.org/i4a/pages/index.cfm?pageid=4134.Google Scholar
Klem, GH, Lüders, HO, Jasper, HH, Elger, C. The ten-twenty electrode system of the International Federation. The International Federation of Clinical Neurophysiology. Electroencephalogr Clin Neurophysiol Suppl 1999; 52: 36.Google Scholar
World Health Organization (WHO). Guidelines on neonatal seizures, WHO Guidelines Approved by the Guidelines Review Committee, 2011. Available at www.ncbi.nlm.nih.gov/books/NBK304092/.Google Scholar
Murray, DM, Boylan, GB, Ali, I, et al. Defining the gap between electrographic seizure burden, clinical expression and staff recognition of neonatal seizures. Arch Dis Child Fetal Neonatal Ed 2008; 93(3): F187–91.Google Scholar
Clancy, RR, Legido, A, Lewis, D. Occult neonatal seizures. Epilepsia 1988; 29(3): 256–61.CrossRefGoogle ScholarPubMed
Glass, HC, Wusthoff, CJ, Shellhaas, RA, et al. Risk factors for EEG seizures in neonates treated with hypothermia: a multicenter cohort study. Neurology 2014; 82(14): 1239–44.Google Scholar
Wusthoff, CJ, Dlugos, DJ, Gutierrez-Colina, A, et al. Electrographic seizures during therapeutic hypothermia for neonatal hypoxic-ischemic encephalopathy. J Child Neurol. 2011; 26(6): 724–8.Google Scholar
Committee on Fetus and Newborn, Papile, L-A, Baley, JE, et al. Hypothermia and neonatal encephalopathy. Pediatrics 2014; 133(6): 1146–50.Google ScholarPubMed
Scher, MS, Alvin, J, Gaus, L, et al. Uncoupling of EEG-clinical neonatal seizures after antiepileptic drug use. Pediatr Neurol 2003; 28(4): 277–80.CrossRefGoogle ScholarPubMed
Tsuchida, TN. EEG background patterns and prognostication of neonatal encephalopathy in the era of hypothermia. J Clin Neurophysiol 2013; 30(2): 122–5.CrossRefGoogle ScholarPubMed
Bye, AM, Lee, D, Naidoo, D, Flanagan, D. The effects of morphine and midazolam on EEGs in neonates. J Clin Neurosci 1997; 4(2): 173–5.Google Scholar
Young, GB, da Silva, OP. Effects of morphine on the electroencephalograms of neonates: a prospective, observational study. Clin Neurophysiol 2000; 111(11): 1955–60.Google Scholar
Le Bihannic, A, Beauvais, K, Busnel, A, et al. Prognostic value of EEG in very premature newborns. Arch Dis Child Fetal Neonatal Ed 2012; 97(2): F106–9.CrossRefGoogle ScholarPubMed
Abend, NS, Wusthoff, CJ. Neonatal seizures and status epilepticus. J Clin Neurophysiol 2012; 29(5): 441–8.Google Scholar
Hrachovy, RA, Mizrahi, EM. Atlas of Neonatal Electroencephalography, 4th edn. New York: Demos Medical, 2015.Google Scholar
Clancy, R, Wusthoff, C. Brain Monitoring: Normal Neonatal EEG. Ambler, PA: Moberg Multimedia, 2011.Google Scholar
Hellström-Westas, L, Rosén, I, Vries, LS, Greisen, G. Amplitude-integrated EEG classification and interpretation in preterm and term infants. NeoReviews 2006; 7(2): e7687.Google Scholar
al Naqeeb, N, Edwards, AD, Cowan, FM, Azzopardi, D. Assessment of neonatal encephalopathy by amplitude-integrated electroencephalography. Pediatrics 1999; 103(6 Pt 1): 1263–71.Google Scholar
Bjerre, I, Hellström-Westas, L, Rosén, I, Svenningsen, N. Monitoring of cerebral function after severe asphyxia in infancy. Arch Dis Child 1983; 58(12): 9971002.Google Scholar
Hellström-Westas, L, Rosén, I, Svenningsen, NW. Predictive value of early continuous amplitude integrated EEG recordings on outcome after severe birth asphyxia in full term infants. Arch Dis Child Fetal Neonatal Ed 1995; 72(1): F34–8.Google Scholar
Hellström-Westas, L, Rosén, I, Swenningsen, NW. Silent seizures in sick infants in early life: diagnosis by continuous cerebral function monitoring. Acta Paediatr Scand 1985; 74(5): 741–8.Google Scholar
Bonifacio, SL, Glass, HC, Peloquin, S, Ferriero, DM. A new neurological focus in neonatal intensive care. Nat Rev Neurol 2011; 7(9): 485–94.Google Scholar
Toet, MC, van der Meij, W, de Vries, LS, et al. Comparison between simultaneously recorded amplitude integrated electroencephalogram (cerebral function monitor) and standard electroencephalogram in neonates. Pediatrics. 2002; 109(5): 772–9.Google Scholar
Shellhaas, RA, Soaita, AI, Clancy, RR. Sensitivity of amplitude-integrated electroencephalography for neonatal seizure detection. Pediatrics 2007; 120(4): 770–7.Google Scholar
Hellström-Westas, L, Rosén, I. Continuous brain-function monitoring: state of the art in clinical practice. Semin Fetal Neonatal Med 2006; 11(6): 503–11.CrossRefGoogle ScholarPubMed
Norman, E, Wikström, S, Rosén, I, et al. Premedication for intubation with morphine causes prolonged depression of electrocortical background activity in preterm infants. Pediatr Res 2013; 73(1): 8794.Google Scholar
Hellström-Westas, L, Bell, AH, Skov, L, et al. Cerebroelectrical depression following surfactant treatment in preterm neonates. Pediatrics 1992; 89(4 Pt 1): 643–7.Google Scholar
Merchant, N, Azzopardi, D. Early predictors of outcome in infants treated with hypothermia for hypoxic-ischaemic encephalopathy. Dev Med Child Neurol 2015; 57: 816.Google Scholar
Thoresen, M, Hellström-Westas, L, Liu, X, de Vries, LS. Effect of hypothermia on amplitude-integrated electroencephalogram in infants with asphyxia. Pediatrics 2010; 126(1): e131–9.Google Scholar
Gluckman, PD, Wyatt, JS, Azzopardi, D, et al. Selective head cooling with mild systemic hypothermia after neonatal encephalopathy: multicentre randomised trial. Lancet 2005; 365(9460): 663–70.Google Scholar
Azzopardi, DV, Strohm, B, Edwards, AD, et al. Moderate hypothermia to treat perinatal asphyxial encephalopathy. N Engl J Med 2009; 361(14): 1349–58.Google Scholar
Toet, MC, Hellström-Westas, L, Groenendaal, F, et al. Amplitude integrated EEG 3 and 6 hours after birth in full term neonates with hypoxic-ischaemic encephalopathy. Arch Dis Child Fetal Neonatal Ed 1999; 81(1): F1923.Google Scholar
Bonifacio, SL, deVries, LS, Groenendaal, F. Impact of hypothermia on predictors of poor outcome: how do we decide to redirect care? Semin Fetal Neonatal Med 2015; 20(2): 122–7.Google Scholar
Abend, NS, Wusthoff, CJ, Goldberg, EM, Dlugos, DJ. Electrographic seizures and status epilepticus in critically ill children and neonates with encephalopathy. Lancet Neurol. 2013; 12(12): 1170–9.Google Scholar
Nash, KB, Bonifacio, SL, Glass, HC, et al. Video-EEG monitoring in newborns with hypoxic-ischemic encephalopathy treated with hypothermia. Neurology 2011; 76(6): 556–62.Google Scholar
Shah, DK, Wusthoff, CJ, Clarke, P, et al. Electrographic seizures are associated with brain injury in newborns undergoing therapeutic hypothermia. Arch Dis Child Fetal Neonatal Ed 2014; 99(3): F219–24.Google Scholar
Rakshasbhuvankar, A, Paul, S, Nagarajan, L, et al. EEG for detection of neonatal seizures: a systematic review. Seizure. 2015; 33: 90–8.Google Scholar
Olischar, M, Shany, E, Aygün, C, et al. Amplitude-integrated electroencephalography in newborns with inborn errors of metabolism. Neonatology 2012; 102(3): 203–11.Google Scholar
Tekgunduz, , Caner, I, Eras, Z, et al. Prognostic value of amplitude-integrated electroencephalography in neonates with hypernatremic dehydration. J Matern-Fetal Neonatal Med 2014; 27(7): 658–63.CrossRefGoogle ScholarPubMed
ter Horst, HJ, van Olffen, M, Remmelts, HJ, et al. The prognostic value of amplitude integrated EEG in neonatal sepsis and/or meningitis. Acta Paediatr Oslo Nor 2010; 99(2): 194200.Google Scholar
Verboon-Maciolek, MA, Groenendaal, F, Hahn, CD, et al. Human parechovirus causes encephalitis with white matter injury in neonates. Ann Neurol 2008; 64(3): 266–73.Google Scholar
Kidokoro, H, Kubota, T, Hayashi, N, et al. Absent cyclicity on aEEG within the first 24 h is associated with brain damage in preterm infants. Neuropediatrics 2010; 41(6): 241–5.CrossRefGoogle ScholarPubMed
Wood, NS, Costeloe, K, Gibson, AT, et al. The EPICure study: associations and antecedents of neurological and developmental disability at 30 months of age following extremely preterm birth. Arch Dis Child Fetal Neonatal Ed 2005; 90(2): F134–40.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(1): 6270.Google Scholar
Hellström-Westas, L. Comparison between tape-recorded and amplitude-integrated EEG monitoring in sick newborn infants. Acta Paediatr Oslo Nor 1992; 81(10): 812–9.Google Scholar
Olischar, M, Klebermass, K, Kuhle, S, et al. Reference values for amplitude-integrated electroencephalographic activity in preterm infants younger than 30 weeks’ gestational age. Pediatrics 2004; 113(1 Pt 1):e61–6.Google Scholar
Selton, D, Andre, M, Hascoët, JM. Normal EEG in very premature infants: reference criteria. Clin Neurophysiol 2000; 111(12): 2116–24.Google Scholar
Hayakawa, M, Okumura, A, Hayakawa, F, et al. Background electroencephalographic (EEG) activities of very preterm infants born at less than 27 weeks gestation: a study on the degree of continuity. Arch Dis Child Fetal Neonatal Ed 2001; 84(3): F163–7.Google Scholar
Sisman, J, Campbell, DE, Brion, LP. Amplitude-integrated EEG in preterm infants: maturation of background pattern and amplitude voltage with postmenstrual age and gestational age. J Perinatol 2005; 25(6): 391–6.Google Scholar
Klebermass, K, Kuhle, S, Olischar, M, et al. Intra- and extrauterine maturation of amplitude-integrated electroencephalographic activity in preterm infants younger than 30 weeks of gestation. Biol Neonate 2006; 89(2): 120–5.Google Scholar
Burdjalov, VF, Baumgart, S, Spitzer, AR. Cerebral function monitoring: a new scoring system for the evaluation of brain maturation in neonates. Pediatrics 2003; 112(4): 855–61.Google Scholar
Kuhle, S, Klebermass, K, Olischar, M, et al. Sleep-wake cycles in preterm infants below 30 weeks of gestational age: preliminary results of a prospective amplitude-integrated EEG study. Wien Klin Wochenschr. 2001; 113(7–8): 219–23.Google Scholar
Hellström-Westas, L, Klette, H, Thorngren-Jerneck, K, Rosén, I. Early prediction of outcome with aEEG in preterm infants with large intraventricular hemorrhages. Neuropediatrics 2001; 32(6): 319–24.Google Scholar
Aso, K, Abdab-Barmada, M, Scher, MS. EEG and the neuropathology in premature neonates with intraventricular hemorrhage. J Clin Neurophysiol 1993; 10(3): 304–13.Google Scholar
Olischar, M, Klebermass, K, Waldhoer, T, et al. Background patterns and sleep-wake cycles on amplitude-integrated electroencephalography in preterms younger than 30 weeks gestational age with peri-/intraventricular haemorrhage. Acta Paediatr Oslo Nor 2007; 96(12): 1743–50.Google Scholar
Chalak, LF, Sikes, NC, Mason, MJ, Kaiser, JR. Low-voltage aEEG as predictor of intracranial hemorrhage in preterm infants. Pediatr Neurol 2011; 44(5): 364–9.Google Scholar
Olischar, M, Klebermass, K, Kuhle, S, et al. Progressive posthemorrhagic hydrocephalus leads to changes of amplitude-integrated EEG activity in preterm infants. Childs Nerv Syst 2004; 20(1): 41–5.Google Scholar
Olischar, M, Klebermass, K, Hengl, B, et al. Cerebrospinal fluid drainage in posthaemorrhagic ventricular dilatation leads to improvement in amplitude-integrated electroencephalographic activity. Acta Paediatr Oslo Nor 2009; 98(6): 1002–9.Google Scholar
Klebermass-Schrehof, K, Rona, Z, Waldhör, T, et al. Can neurophysiological assessment improve timing of intervention in posthaemorrhagic ventricular dilatation? Arch Dis Child Fetal Neonatal Ed 2013; 98(4): F291–7.Google Scholar
de Vries, LS, Liem, KD, van Dijk, K, et al. Early versus late treatment of posthaemorrhagic ventricular dilatation: results of a retrospective study from five neonatal intensive care units in the Netherlands. Acta Paediatr Oslo Nor 2002; 91(2): 212–7.Google Scholar
Song, J, Zhu, C, Xu, F, et al. Predictive value of early amplitude-integrated electroencephalography for later diagnosed cerebral white matter damage in preterm infants. Neuropediatrics 2014; 45(5): 314–20.Google ScholarPubMed
Song, J, Xu, F, Wang, L, et al. Early amplitude-integrated electroencephalography predicts brain injury and neurological outcome in very preterm infants. Sci Rep 2015; 5: 13810.CrossRefGoogle ScholarPubMed
Wikström, S, Pupp, IH, Rosén, I, et al. Early single-channel aEEG/EEG predicts outcome in very preterm infants. Acta Paediatr Oslo Nor 2012; 101(7): 719–26.Google Scholar
Klebermass, K, Olischar, M, Waldhoer, T, et al. Amplitude-integrated EEG pattern predicts further outcome in preterm infants. Pediatr Res 2011; 70(1): 102–8.Google Scholar
Welch, C, Helderman, J, Williamson, E, O’Shea, TM. Brain wave maturation and neurodevelopmental outcome in extremely low gestational age neonates. J Perinatol 2013; 33(11): 867–71.Google Scholar
Natalucci, G, Leuchter, RH-V, Bucher, HU, et al. Functional brain maturation assessed during early life correlates with anatomical brain maturation at term-equivalent age in preterm infants. Pediatr Res 2013; 74(1): 6874.Google Scholar
Greisen, G, Pryds, O. Low CBF, discontinuous EEG activity, and periventricular brain injury in ill, preterm neonates. Brain Dev 1989; 11(3): 164–8.Google Scholar
Van Os, S, Klaessens, J, Hopman, J, et al. Preservation of electrocortical brain activity during hypoxemia in preterm lambs. Exp Brain Res 2003; 151(1): 54–9.Google Scholar
West, CR, Groves, AM, Williams, CE, et al. Early low cardiac output is associated with compromised electroencephalographic activity in very preterm infants. Pediatr Res 2006; 59(4 Pt 1): 610–5.Google Scholar
Shah, D, Paradisis, M, Bowen, JR. Relationship between systemic blood flow, blood pressure, inotropes, and aEEG in the first 48 h of life in extremely preterm infants. Pediatr Res 2013; 74(3): 314–20.Google Scholar
Lemmers, PMA, Toet, MC, van Bel, F. Impact of patent ductus arteriosus and subsequent therapy with indomethacin on cerebral oxygenation in preterm infants. Pediatrics 2008; 121(1): 142–7.Google Scholar
Evans, N, Kluckow, M. Early ductal shunting and intraventricular haemorrhage in ventilated preterm infants. Arch Dis Child Fetal Neonatal Ed 1996; 75(3): F183–6.Google Scholar
Miller, SP, Mayer, EE, Clyman, RI, et al. Prolonged indomethacin exposure is associated with decreased white matter injury detected with magnetic resonance imaging in premature newborns at 24 to 28 weeks’ gestation at birth. Pediatrics 2006; 117(5): 1626–31.Google Scholar
Kabra, NS, Schmidt, B, Roberts, RS, et al. Neurosensory impairment after surgical closure of patent ductus arteriosus in extremely low birth weight infants: results from the Trial of Indomethacin Prophylaxis in Preterms. J Pediatr 2007; 150(3): 229–34, 234.e1.Google Scholar
Lemmers, PMA, Molenschot, MC, Evens, J, et al. Is cerebral oxygen supply compromised in preterm infants undergoing surgical closure for patent ductus arteriosus? Arch Dis Child Fetal Neonatal Ed 2010; 95(6): F429–34.Google Scholar
Leslie, ATFS, Jain, A, El-Khuffash, A, et al. Evaluation of cerebral electrical activity and cardiac output after patent ductus arteriosus ligation in preterm infants. J Perinatol 2013; 33(11): 861–6.Google Scholar
Helderman, JB, Welch, CD, Leng, X, O’Shea, TM. Sepsis-associated electroencephalographic changes in extremely low gestational age neonates. Early Hum Dev 2010; 86(8): 509–13.Google Scholar

References

Barnes, P. Pediatric brain imaging. In Blickman, J, Parker, B, Barnes, P, eds., Pediatric Radiology: The Requisites, 3rd edn. Philadelphia: Elsevier, 2009: 205–70.Google Scholar
Barnes, P. Neuroimaging in the evaluation of pattern and timing of fetal and neonatal brain abnormalities. In Stevenson, D, Benitz, W, Sunshine, P, eds., Fetal and Neonatal Brain Injury, 4th edn. New York: Cambridge University Press, 2009: 209–31.Google Scholar
Izbudak, I1, Grant, PE. MR imaging of the term and preterm neonate with diffuse brain injury. Magn Reson Imaging Clin North Am 2011; 19(4): 709–31.Google Scholar
Barkovich, AJ, Raybaud, C:Pediatric Neuroimaging, 5th edn. Philadelphia: Lippincott Williams & Wilkins, Wolters Kluwer, 2011.Google Scholar
Volpe, JJ. Neurology of the Newborn, 5th edn. Philadelphia: Saunders Elsevier, 2008.Google Scholar
Ment, L, Bada, H, Barnes, P, et al. Practice parameter: neuroimaging of the neonate. Neurology 2002; 58: 1726–38.Google Scholar
D’Alton, ME, Hankins, GDV, et al. Neonatal Encephalopathy and Cerebral Palsy: Defining the Pathogenesis and Pathophysiology, 1st edn. (Washington, DC: American College of Obstetricians and Gynecologists, 2003), table 5-2, 570; Neonatal Encephalopathy and Neurologic Outcome, 2nd edn. (2014), 149–72).Google Scholar
Barkovich, A. MR imaging of the neonatal brain. Neuroimag Clin North Am 2006; 16: 117–36.Google Scholar
Rutherford, M, Srinivasan, L, Dyet, L, et al. MRI in perinatal brain injury. Pediatr Radiol 2006; 36: 582–92.Google Scholar
Zimmerman, R, Bilaniuk, L. Neuroimaging evaluation of cerebral palsy. Clin Perinatol 2006; 33: 517–44.Google Scholar
Triulzi, F, Parazzini, C, Righini, A. Patterns of damage in the mature neonatal brain. Pediatr Radiol 2006; 36: 608–20.Google Scholar
Panigrahy, A, Blumel, S. Advances in MR neuroimaging techniques in the evaluation of neonatal encephalopathy. Top Magn Reson Imaging 2007; 18: 330.Google Scholar
Barnes, PD, Taylor, GA. Imaging of the neonatal central nervous system. Neurosurg Clin North Am 1998; 1: 1748.Google Scholar
Allison, JW, Seibert, JJ. Transcranial Doppler in the newborn with asphyxia. Neuroimag Clin North Am 1999; 9: 1116.Google Scholar
Epelman, M1, Daneman, A, Chauvin, N, Hirsch, W. Head Ultrasound and MR imaging in the evaluation of neonatal encephalopathy: competitive or complementary imaging studies? Magn Reson Imaging Clin North Am. 2012; 20(1): 93115.Google Scholar
Debillon, T, N’Guyen, S, Muet, A, et al. Limitations of ultrasonography for diagnosing white matter damage in preterm infants. Arch Dis Child Fetal Neonatal Ed 2003; 88: F275–9.Google Scholar
Laptook, A, O’Shea, M, Shankaran, S, et al. Adverse neurodevelopmental outcomes among extremely low birth weight infants with normal head ultrasound. Pediatrics 2005; 115: 673–80.Google Scholar
Pinto-Martin, JA, Riolo, S, Cnaan, A, et al. Cranial ultrasound prediction of disabling and nondisabling cerebral palsy at age two in a low birthweight population. Pediatrics 1995; 95: 249–54.Google Scholar
Maalouf, EF, Duggan, PJ, Counsell, SJ, et al. Comparison of findings on cranial ultrasound and magnetic resonance imaging in preterm neonates. Pediatrics 2001; 107: 719–27.Google Scholar
Sie, LT, van der Knapp, MD, van Wezel-Meijler, G, et al. Early MR features of hypoxic-ischemic brain injury in neonates with periventricular densities on sonograms. AJNR Am J Neuroradiol 2000; 21: 852–61.Google Scholar
Childs, AM, Cornette, L, Romenghi, LA, et al. Magnetic resonance and cranial ultrasound characteristics of periventricular white matter abnormalities in newborn infants. Clin Radiol 2001; 56: 647–55.Google Scholar
Roelants-van Rijn, AM, Groenendaal, F, Beek, FJ, et al. Parenchymal brain injury in the preterm infant: comparison of cranial ultrasound, MRI and neurodevelopmental outcome. Neuropediatrics 2001; 32: 80–9.Google Scholar
Inder, TE, Anderson, NJ, Spencer, C, et al. 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.Google Scholar
Miller, SP, Cozzio, C, Goldstein, RB, 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.Google Scholar
Hintz, SR, Barnes, PD, Bulas, D, et al. Neuroimaging and neurodevelopmental outcome in extremely preterm infants. Pediatrics. 2015; 135(1): e3242.Google Scholar
Mirmiran, M, Barnes, P, Keller, K, et al. Neonatal brain MRI before discharge is better than serial cranial ultrasound in predicting cerebral palsy in very low birth weight preterm infants. Pediatrics 2004; 114: 992–8.Google Scholar
Sanchez, R, Khong, PL, Ringertz, H. Radiologic protection in pediatric radiology: ICRP recommendations. Pediatr Radiol. 2013; 43(8): 920–1.Google Scholar
Vertinzksy, A, Barnes, P. Macrocephaly, increased intracranial pressure, and hydrocephalus in the infant and young child. Top Magn Reson Imaging 2007; 18: 3152.Google Scholar
Barnes, P. Imaging of NAI and the mimics: issues and controversies in the era of evidence-based medicine. Radiol Clin North Am 2011; 49: 205–29.Google Scholar
Mukherjee, P. Advanced pediatric imaging. Neuroimag Clin North Am 2006; 16: 1210.Google Scholar
Levine, D, Barnes, PD, Robertson, RR, et al. Fast MR imaging of fetal central nervous system abnormalities. Radiology 2003; 229: 5161.Google Scholar
Garel, C, Delezide, A, Elmaleh-Berges, M, et al. Contribution of fetal MRI in the evaluation of cerebral ischemic lesions. AJNR Am J Neuroradiol 2004; 25: 1563–8.Google Scholar
Levine, D, Barnes, P. MR imaging of fetal CNS abnormalities. In Levine, D, ed., Atlas of Fetal MRI. Boca Raton, FL: Taylor & Francis, 2005: 2572.Google Scholar
Griffiths, P, Widjaja, E, Paley, M, et al. Imaging of the fetal spine in utero: diagnostic accuracy and impact on management. Pediatr Radiol 2006; 36: 927–33.Google Scholar
Garel, C. New advances in fetal MR neuroimaging. Pediatr Radiol 2006; 36: 621–5.Google Scholar
Glenn, O, Barkovich, J. Magnetic resonance imaging of the fetal brain and spine, part 1. AJNR Am J Neuroradiol 2006; 27: 1604–11; Part 2. AJNR Am J Neuroradiol 2006; 27: 1807–14.Google Scholar
Barkovich, AJ, Baranski, K, Vigneron, D, et al. Proton MR spectroscopy in the evaluation of asphyxiated term neonates. AJNR Am J Neuroradiol 1999; 20: 1399–405.Google Scholar
Vigneron, DB. Magnetic resonance spectroscopic imaging of human brain development. Neuroimag Clin North Am 2006; 16; 7586.Google Scholar
Cecil, K. MR spectroscopy of metabolic disorders. Neuroimag Clin North Am 2006; 16: 87116.Google Scholar
Barkovich, A, Miller, S, Bartha, A. MR imaging, MR spectroscopy, and diffusion tensor imaging of sequential studies in neonates with encephalopathy. AJNR Am J Neuroradiol 2006; 27: 533–47.Google Scholar
Wang, J, Licht, D. Pediatric perfusion MRI using arterial spin labeling. Neuroimag Clin North Am 2006; 16: 149–68.Google Scholar
Huppi, PS, Maier, SE, Peled, S, et al. Microstructural development of human newborn cerebral white matter assessed in vivo by diffusion tensor magnetic resonance imaging. Pediatr Res 1998; 44: 584–90.Google Scholar
Neil, JJ, Shiran, SI, McKinstry, RC. Normal brain in human newborns: apparent diffusion coefficient and diffusion anisotropy measured by using diffusion tensor MR imaging. Radiology 1998; 209: 5766.Google Scholar
Johnson, AJ, Lee, BCP, Lin, W. Echoplanar diffusion-weighted imaging in neonates and infants with suspected hypoxic–ischemic injury. AJNR Am J Neuroradiol 1999; 172: 219–26.Google Scholar
Robertson, RL, Ben-Sira, L, Barnes, PD, et al. MR line scan diffusion imaging of term neonates with perinatal brain ischemia. AJNR Am J Neuroradiol 1999; 20: 1658–70.Google Scholar
Phillips, MD, Zimmerman, RA. Diffusion imaging in pediatric hypoxic-ischemic injury. Neuroimag Clin North Am 1999; 9: 4152.Google Scholar
Inder, T, Huppi, PS, Zientara, GP, et al. Early detection of periventricular leukomalacia by diffusion-weighted magnetic resonance imaging techniques. J Pediatr 1999; 134: 631–4.Google Scholar
Inder, TE, Huppi, PS, Warfield, S, et al. Periventricular white matter injury in the premature infant followed by reduced cerebral cortical gray matter volume at term. Ann Neurol 1999; 46: 755–60.Google Scholar
Huppi, PS, Murphy, B, Maier, SE, et al. Microstructural brain development after perinatal cerebral white matter injury assessed by diffusion tensor MR imaging. Pediatrics 2001; 107: 455–60.Google Scholar
Arzoumanian, Y, Mirmiran, M, Barnes, P, et al. Diffusion tensor brain imaging findings at term-equivalent age may predict neurologic abnormalities in low birth weight preterm infants. AJNR Am J Neuroradiol 2003; 24: 1646–53.Google Scholar
Counsell, S, Allsop, J, Harrison, M, et al. Diffusion-weighted imaging of the brain in preterm infants with focal and diffuse white matter abnormality. Pediatrics 2003; 112: 17.Google Scholar
Sagar, P, Grant, PE. Diffusion-weighted imaging: pediatric clinical applications. Neuroimag Clin North Am 2006; 16: 4574.Google Scholar
Mukherjee, P, McKinstry, R. Diffusion tensor imaging and tractography of human brain development. Neuroimag Clin North Am 2006; 16: 1944.Google Scholar
van der Knaap, M, Valk, J. Classification of congenital abnormalities of the CNS. AJNR Am J Neuroradiol 1988; 9: 315–26.Google Scholar
Barkovich, AJ, Truwit, CL. Brain damage from perinatal asphyxia:correlation of MR findings with gestational age. AJNR Am J Neuroradiol 1990; 11: 1087–96.Google Scholar
Aida, N, Nishimura, G, Hachiya, Y, et al. MR imaging of perinatal brain damage: comparison of clinical outcome with initial follow-up MR findings. AJNR Am J Neuroradiol 1992; 19: 1909–22.Google Scholar
Barkovich, A, Hallam, D. Neuroimaging in perinatal hypoxic–ischemic injury. Ment Retard Dev Disabil Res Rev 1997; 3: 2841.Google Scholar
Cowan, F, Rutherford, M, Groenendaal, F, et al. Origin and timing of brain lesions in term infants with neonatal encephalopathy. Lancet 2003; 361: 736–42.Google Scholar
Miller, S, Ramaswamy, V, Michelson, D, et al. Patterns of brain injury in term neonatal encephalopathy. J Pediatr 2005; 146: 453–60.Google Scholar
Lupton, BA, Hill, A, Roland, EH, et al. Brain swelling in the asphyxiated term newborn:pathogenesis and outcome. Pediatrics 1988; 82: 139–46.Google Scholar
Vannucci, RC, Christensen, MA, Jager, JY. Nature, time-course, and extent of cerebral edema in perinatal hypoxic–ischemic brain damage. Pediatr Neurol 1993; 9: 2934.Google Scholar
Pinto-Martin, JA, Riolo, S, Cnaan, A, et al. Cranial ultrasound prediction of disabling and nondisabling cerebral palsy at age two in a low birthweight population. Pediatrics 1995; 95: 249–54.Google Scholar
Goetz, MC, Gretebeck, RJ, Oh, KS, et al. Incidence, timing and follow-up of periventricular leukomalacia. Am J Perinatol 1995; 12: 325–7.Google Scholar
Valkama, AM, Pääkkö, ELE, Vainionpää, LK, et al. Magnetic resonance imaging at term and neuromotor outcome in preterm infants. Acta Paediatr 2000; 89: 348–55.Google Scholar
Perlman, JM, Rollins, N. Surveillance protocol for the detection of intracranial abnormalities in premature neonates. Arch Pediatr Adolesc Med 2000; 154: 822–6.Google Scholar
Volpe, JJ. Neurobiology of periventricular leukomalacia in the premature infant. Pediatr Res 2001; 50: 553–62.Google Scholar
Panigrahy, A, Barnes, PD, Robertson, RL, et al. Volumetric brain differences in children with periventricular T2-signal hyperintensities:a grouping by gestational age at birth. AJR Am J Roentgenol 2001; 177: 695702.Google Scholar
Miller, SP, Hoque, NN, Piecuch, RE, et al. The spectrum of cerebellar hemorrhage in premature newborns. Pediatr Res 2003; 53: 537A (abstract #3040).Google Scholar
Austin, NC, Woodward, L, Spencer, C, et al. Neurodevelopmental outcome at one year in a regional cohort of very low birth weight infants:correlation with MRI at term. Pediatr Res 2003; 53: 398A (abstract #2253).Google Scholar
Jenster, M, Bonifacio, S, Ruel, T, et al. Maternal or neonatal infection:association with neonatal encephalopathy outcomes. Pediatric Res 2014; 76: 93–9.Google Scholar
Inder, TE, Wells, SJ, Mogridge, NB, et al. Defining the nature of the cerebral abnormalities in the premature infant: a qualitative magnetic resonance imaging study. J Pediatr 2003; 143: 171–9.Google Scholar
Panigrahy, A, Barnes, PD, Robertson, RL, et al. Quantitative analysis of the corpus callosum in children with cerebral palsy and developmental delay: correlation with cerebral white matter volume. Pediatr Radiol 2005; 35: 1199–207.Google Scholar
Bodensteiner, J, Johnsen, S. MRI findings in children surviving extremely premature delivery and extremely low birthweight with cerebral palsy. J Child Neurol 2006; 21: 743–7.Google Scholar
Dyet, L, Kennea, N, Counsell, S, et al. Natural history of brain lesions in extremely preterm infants studied with serial MRI from birth and neurodevelopmental assessment Pediatrics 2006; 118: 536–48.Google Scholar
Khwaja, O, Volpe, J. Pathogenesis of cerebral white matter injury of prematurity. Arch Dis Child Fetal Neonatal Ed 2008; 93: F153–61.Google Scholar
Barkovich, AJ. MR and CT evaluation of profound neonatal and infantile asphyxia. AJNR Am J Neuroradiol 1992; 13: 959–72.Google Scholar
Barkovich, AJ, Sargent, SK. Profound asphyxia in the premature infant: imaging findings. AJNR Am J Neuroradiol 1995; 16: 1837–46.Google Scholar
Barkovich, AJ, Westmark, K, Partridge, C, et al. Perinatal asphyxia: MR findings in the first 10 days. AJNR Am J Neuroradiol 1995; 16: 427–39.Google Scholar
Roland, EH, Poskitt, K, et al. Perinatal hypoxic-ischemic thalamic injury: clinical features and neuroimaging. Ann Neurol 1998; 44: 161–6.Google Scholar
Sargent, M, Poskitt, K, Roland, E, et al. Cerebellar vermian atrophy after neonatal hypoxic–ischemic encephalopathy. AJNR Am J Neuroradiol 2004; 25: 1008–15.Google Scholar
Boichot, C, Walker, P, Durand, C, et al. Term neonate prognosis after perinatal asphyxia: contributions of MR imaging, MR spectroscopy, relaxation times, and ADCs. Radiology 2006; 239: 839–48.Google Scholar
Shankaran, S, Barnes, P, Hintz, S, et al. Brain injury following trial of hypothermia for neonatal hypoxic-ischemic encephalopathy. Arch Dis Child Fetal Neonatal Ed 2012; 97(6): F398404.Google Scholar
Bulas, D, Neonatal, Glass P. ECMO: neuroimaging and neurodevelopmental outcome. Semin Perinatol 2005; 29: 5865.Google Scholar
Zuerrer, M, Martin, E, Boltshauser, E. MRI of intracranial hemorrhage in neonates and infants at 2.35 tesla. Neuroradiology 1991; 33: 223–9.Google Scholar
DeVeber, G, Andrew, M, Adams, C, et al. Cerebral sinovenous thrombosis in children. N Engl J Med 2001; 345: 417–23.Google Scholar
Carvalho, K, Bodensteiner, J, Connolly, P, et al. Cerebral venous thrombosis in children. J Child Neurol 2001; 16: 574–85.Google Scholar
Lynch, J, Hirtz, D, deVeber, G, et al. Report of the National Institute of Neurologic Disorders and Stroke Workshop on Perinatal and Childhood Stroke. Pediatrics 2002; 109: 116–23.Google Scholar
DeVeber, G. Arterial ischemic strokes in infants and children: an overview of current approaches. Semin Thrombos Hemost 2003; 29: 567–73.Google Scholar
Sebire, G, Tabarki, B, Saunders, DE, et al. Cerebral venous thrombosis in children: risk factors, presentation, diagnosis, and outcome. Brain 2005; 128: 477–89.Google Scholar
Kirton, A, deVeber, G. Cerebral palsy secondary to perinatal ischemic stroke. Clin Perinatol 2006; 33: 367–86.Google Scholar
Fitzgerald, K, Williams, LS, Garg, BP, et al. Cerebral sinovenous thrombosis in the neonate. Arch Neurol 2006; 63: 405–9.Google Scholar
Barnes, C, deVeber, G. Prothrombotic abnormalities in childhood ischaemic stroke. Thromb Res 2006; 118: 6774.Google Scholar
Ehtisham, A, Stern, B. Cerebral venous thrombosis: a review. Neurologist 2006; 12: 32–8.Google Scholar
Huang, A, Robertson, R. Spontaneous superficial parenchymal and leptomeningeal hemorrhage in term neonates. AJNR Am J Neuroradiol 2004; 25: 469–75.Google Scholar
Govaert, P, Vanhaesebrouck, P, de Praeter, C. Traumatic neonatal intracranial bleeding and stroke. Arch Dis Child 1992; 67: 840–5.Google Scholar
Castillo, M, Fordham, LA. MR of neurologically symptomatic newborns after vacuum extraction delivery. AJNR Am J Neuroradiol 1995; 16: 816–18.Google Scholar
Heuser, C, Knight, S, Esplin, S, et al. Tachysystole in term labor:incidence, risk factors, outcomes, and effect on fetal heart tracings. Am J Obstet Gynecol 2013; 209: 32e16.Google Scholar
Keller, K, Barnes, P. Rickets vs. abuse: a national and internationsl epidemic. Pediatr Radiol 2008; 38: 1210–16.Google Scholar
Alexander, J, Leveno, K, Hauth, J, et al. Fetal injury associated with cesarean delivery. Obstet Gynecol 2006; 108: 885–90.Google Scholar
Doumouchtsis, S, Arulkumaran, S. Head trauma after instrumental births. Clin Perinatol 2008; 35: 6983.Google Scholar
Schneider, JF1, Hanquinet, S, Severino, M, Rossi, A. MR imaging of neonatal brain infections. Magn Reson Imaging Clin North Am. 2011; 19(4): 761–75.Google Scholar
Jenster, M, Bonifacio, S, Ruel, T, et al. Maternal or neonatal infection: association with neonatal encephalopathy outcomes. Pediatr Res 2014; 76: 93–9.Google Scholar
DeVries, L, Verboon-Maciolek, M, Cowan, F, et al. The role of cranial US and MRI in the diagnosis of infections of the central nervous system. Early Hum Dev 2006; 82: 819–25.Google Scholar
Hoon, AH. Neuroimaging in cerebral palsy:patterns of brain dysgenesis and injury. J Child Neurol 2005; 12: 936–9.Google Scholar
Rodriguez, D, Young Poussaint, T. Neuroimaging of the child with developmental delay. Top Magn Reson Imaging 2007; 18: 7592.Google Scholar
Valk, J, van der Knapp, MS. Toxic encephalopathy. AJNR Am J Neuroradiol 1992; 13: 747–60.Google Scholar
van der Knaap, MS, Valk, J. Magnetic Resonance of Myelination and Myelin Disorders, 3rd edn. New York: Springer, 2005.Google Scholar
Patay, Z. MR imaging workup of inborn errors of metabolism of early postnatal onset. Magn Reson Imaging Clin North Am. 2011; 19(4): 733–59;Google Scholar
Vanderver, A. Tools for diagnosis of leukodystrophies and other disorders presenting with white matter disease. Curr Neurol Neurosci Rep 2005; 5: 110–18.Google Scholar
Murakami, Y, Yamashita, Y, Matsuishi, T, et al. Cranial MRI of neurologically impaired children suffering from neonatal hypoglycemia. Pediatr Radiol 1999; 29: 23–7.Google Scholar
Alkalay, A, Flores-Sarnat, L, Sarnat, H, et al. Brain imaging findings in neonatal hypoglycemia. Clin Pediatr 2005; 44: 783–90.Google Scholar
Barkovich, A, Ali, F, Rowley, H, et al. Imaging patterns of neonatal hypoglycemia. AJNR Am J Neuroradiol 1998; 19: 523–8.Google Scholar
Govaert, P, Lequin, M, Swarte, R, et al. Changes in globus pallidus with (pre) term kernicterus. Pediatrics 2003; 112: 1256–63.Google Scholar
Dublin, A, Hald, J, Wootton-Gorges, S. Isolated sulfite oxidase deficiency: MR imaging features. AJNR Am J Neuroradiol 2002; 23: 484–5.Google Scholar

References

Jöbsis, FF. Non-invasive, infra-red monitoring of cerebral O2 sufficiency, blood volume, HbO2-Hb shifts and bloodflow. Acta Neurol Scand Suppl 1977; 64: 452–3.Google Scholar
Andropoulos, DB, Stayer, SA, Diaz, LK, Ramamoorthy, C. Neurological monitoring for congenital heart surgery. Anesth Analg 2004; 99(5): 1365–75.Google Scholar
Hoffman, GM. Neurologic monitoring on cardiopulmonary bypass: what are we obligated to do? Ann Thorac Surg 2006; 81(6): S2373–80.Google Scholar
Meek, JH, Elwell, CE, McCormick, DC, et al. Abnormal cerebral haemodynamics in perinatally asphyxiated neonates related to outcome. Arch Child Fetal Neonatal Ed 1999; 81(2): F110–5.Google Scholar
Lemmers, PMA, Toet, M, van Schelven, LJ, van Bel, F. Cerebral oxygenation and cerebral oxygen extraction in the preterm infant: the impact of respiratory distress syndrome. Exp Brain Res Exp 2006; 173(3): 458–67.Google Scholar
Benni, PB, Chen, B, Dykes, FD, et al. Validation of the CAS neonatal NIRS system by monitoring vv-ECMO patients: preliminary results. Adv Exp Med Biol 2005; 566: 195201.Google Scholar
Jain, V, Buckley, EM, Licht, DJ, et al. Cerebral oxygen metabolism in neonates with congenital heart disease quantified by MRI and optics. J Cereb Blood Flow Metab 2014; 34(3): 380–8.Google Scholar
Bassan, H, Gauvreau, K, Newburger, JW, et al. Identification of pressure passive cerebral perfusion and its mediators after infant cardiac surgery. Pediatr Res 2005; 57(1):3541.Google Scholar
Menke, J, Voss, U, Möller, G, Jorch, G. Reproducibility of cerebral near infrared spectroscopy in neonates. Biol Neonate 2003; 83(1): 611.Google Scholar
Schneider, A, Minnich, B, Hofstätter, E, et al. Comparison of four near-infrared spectroscopy devices shows that they are only suitable for monitoring cerebral oxygenation trends in preterm infants. Acta Paediatr Oslo Nor 2014; 103(9): 934–8.Google Scholar
Dix, LML, van Bel, F, Baerts, W, Lemmers, PMA. Comparing near-infrared spectroscopy devices and their sensors for monitoring regional cerebral oxygen saturation in the neonate. Pediatr Res 2013; 74(5): 557–63.Google Scholar
McNeill, S, Gatenby, JC, McElroy, S, Engelhardt, B. Normal cerebral, renal and abdominal regional oxygen saturations using near-infrared spectroscopy in preterm infants. J Perinatol 2011; 31(1): 51–7.Google Scholar
Alderliesten, T, Dix, L, Baerts, W, et al. Reference values of regional cerebral oxygen saturation during the first 3 days of life in preterm neonates. Pediatr Res 2016; 79: 5564.Google Scholar
Cohen, E, Baerts, W, Alderliesten, T, et al. Growth restriction and gender influence cerebral oxygenation in preterm neonates. Arch Dis Child Fetal Neonatal Ed 2016; 101(2): F156–61.Google Scholar
Lemmers, PM, van Bel, F. Left-to-right differences of regional cerebral oxygen saturation and oxygen extraction in preterm infants during the first days of life. Pediatr Res 2009; 65(2): 226–30.Google Scholar
Grossauer, K, Pichler, G, Schmölzer, G, et al. Comparison of peripheral and cerebral tissue oxygenation index in neonates. Arch Dis Child Fetal Neonatal Ed 2009; 94(2): F156.Google Scholar
Bernal, NP, Hoffman, GM, Ghanayem, NS, Arca, MJ. Cerebral and somatic near-infrared spectroscopy in normal newborns. J Pediatr Surg 2010; 45(6): 1306–10.Google Scholar
Hyttel-Sorensen, S, Pellicer, A, Alderliesten, T, et al. Cerebral near infrared spectroscopy oximetry in extremely preterm infants: phase II randomised clinical trial. BMJ 2015; 350: g7635.Google Scholar
Hou, X, Ding, H, Teng, Y, et al. Research on the relationship between brain anoxia at different regional oxygen saturations and brain damage using near-infrared spectroscopy. Physiol Meas 2007; 28(10): 1251–65.Google Scholar
Kurth, CD, Levy, WJ, McCann, J. Near-infrared spectroscopy cerebral oxygen saturation thresholds for hypoxia-ischemia in piglets. J Cereb Blood Flow Metab Off J Int Soc Cereb Blood Flow Metab 2002; 22(3): 335–41.CrossRefGoogle ScholarPubMed
Dent, CL, Spaeth, JP, Jones, BV, et al. Brain magnetic resonance imaging abnormalities after the Norwood procedure using regional cerebral perfusion. J Thorac Cardiovasc Surg 2005; 130(6): 1523–30.Google Scholar
Verhagen, EA, Van Braeckel, KNJA, van der Veere, CN, et al. Cerebral oxygenation is associated with neurodevelopmental outcome of preterm children at age 2 to 3 years. Dev Med Child Neurol 2015; 57(5): 449–55.Google Scholar
Fuchs, H, Lindner, W, Buschko, A, et al. Brain oxygenation monitoring during neonatal resuscitation of very low birth weight infants. J Perinatol Off J Calif Perinat Assoc 2012; 32(5): 356–62.Google Scholar
Binder, C, Urlesberger, B, Avian, A, et al. Cerebral and peripheral regional oxygen saturation during postnatal transition in preterm neonates. J Pediatr 2013; 163(2): 394–9.Google Scholar
Fuchs, H, Lindner, W, Buschko, A, et al. Cerebral oxygenation in very low birth weight infants supported with sustained lung inflations after birth. Pediatr Res 2011; 70(2): 176–80.CrossRefGoogle ScholarPubMed
Brady, KM, Mytar, JO, Lee, JK, et al. Monitoring cerebral blood flow pressure autoregulation in pediatric patients during cardiac surgery. Stroke J Cereb Circ 2010; 41(9): 1957–62.Google Scholar
Wong, FY, Leung, TS, Austin, T, et al. Impaired autoregulation in preterm infants identified by using spatially resolved spectroscopy. Pediatrics 2008; 121(3): e604–11.Google Scholar
Eriksen, VR, Hahn, GH, Greisen, G. Cerebral autoregulation in the preterm newborn using near-infrared spectroscopy: a comparison of time-domain and frequency-domain analyses. J Biomed Opt 2015; 20(3): 37009.Google Scholar
Tsuji, M, Saul, JP, du Plessis, A, et al. Cerebral intravascular oxygenation correlates with mean arterial pressure in critically ill premature infants. Pediatrics 2000; 106(4): 625–32.Google Scholar
O’Leary, H, Gregas, MC, Limperopoulos, C, et al. Elevated cerebral pressure passivity is associated with prematurity-related intracranial hemorrhage. Pediatrics 2009; 124(1): 302–9.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(4): 467–73.Google Scholar
Chock, VY, Ramamoorthy, C, Van Meurs, KP. Cerebral autoregulation in neonates with a hemodynamically significant patent ductus arteriosus. J Pediatr 2012; 160: 936–42.Google Scholar
Binder-Heschl, C, Urlesberger, B, Schwaberger, B, et al. Borderline hypotension: how does it influence cerebral regional tissue oxygenation in preterm infants? J Matern-Fetal Neonatal Med Off J Eur Assoc Perinat Med Fed Asia Ocean Perinat Soc Int Soc Perinat Obstet 2016; 29(14): 2341–6.Google Scholar
Garner, RS, Burchfield, DJ. Treatment of presumed hypotension in very low birthweight neonates: effects on regional cerebral oxygenation. Arch Dis Child Fetal Neonatal Ed 2013; 98(2): F117–21.Google Scholar
Verhagen, EA, Ter Horst, HJ, Keating, P, et al. Cerebral oxygenation in preterm infants with germinal matrix-intraventricular hemorrhages. Stroke 2010; 41(12): 2901–7.Google Scholar
Sorensen, LC, Maroun, LL, Borch, K, et al. Neonatal cerebral oxygenation is not linked to foetal vasculitis and predicts intraventricular haemorrhage in preterm infants. Acta Paediatr 2008; 97(11): 1539–34.Google Scholar
Alderliesten, T, Lemmers, PMA, Smarius, JJM, et al. Cerebral oxygenation, extraction, and autoregulation in very preterm infants who develop peri-intraventricular hemorrhage. J Pediatr 2013; 162(4):698704.e2.Google Scholar
Lemmers, PM, Toet, MC, van Bel, F. Impact of patent ductus arteriosus and subsequent therapy with indomethacin on cerebral oxygenation in preterm infants. Pediatrics 2008; 121(1): 142–7.Google Scholar
Chock, VY, Ramamoorthy, C, Van Meurs, KP. Cerebral oxygenation during different treatment strategies for a patent ductus arteriosus. Neonatology 2011; 100: 233–40.Google Scholar
Underwood, MA, Milstein, JM, Sherman, MP. Near-infrared spectroscopy as a screening tool for patent ductus arteriosus in extremely low birth weight infants. Neonatology 2007; 91(2): 134–9.Google Scholar
Vanderhaegen, J, De Smet, D, Meyns, B, et al. Surgical closure of the patent ductus arteriosus and its effect on the cerebral tissue oxygenation. Acta Paediatr 2008; 97(12): 1640–4.Google Scholar
Meier, SD, Eble, BK, Stapleton, GE, et al. Mesenteric oxyhemoglobin desaturation improves with patent ductus arteriosus ligation. J Perinatol 2006; 26(9): 562–4.Google Scholar
Zaramella, P, Freato, F, Quaresima, V, et al. Surgical closure of patent ductus arteriosus reduces the cerebral tissue oxygenation index in preterm infants: a near-infrared spectroscopy and Doppler study. Pediatr Int 2006; 48(3): 305–12.CrossRefGoogle ScholarPubMed
Lemmers, PM, Molenschot, MC, Evens, J, et al. Is cerebral oxygen supply compromised in preterm infants undergoing surgical closure for patent ductus arteriosus? Arch Child Fetal Neonatal Ed 2010; 95(6):F429–34.Google Scholar
Bailey, SM, Hendricks-Muñoz, KD, Wells, JT, Mally, P. Packed red blood cell transfusion increases regional cerebral and splanchnic tissue oxygen saturation in anemic symptomatic preterm infants. Am J Perinatol 2010; 27(6): 445–53.Google Scholar
Dani, C, Pratesi, S, Fontanelli, G, et al. Blood transfusions increase cerebral, splanchnic, and renal oxygenation in anemic preterm infants. Transfusion (Paris) 2010; 50(6): 1220–6.Google Scholar
Wardle, SP, Weindling, AM. Peripheral fractional oxygen extraction and other measures of tissue oxygenation to guide blood transfusions in preterm infants. Semin Perinatol 2001; 25(2): 60–4.Google Scholar
van Hoften, JC, Verhagen, EA, Keating, P, et al. Cerebral tissue oxygen saturation and extraction in preterm infants before and after blood transfusion. Arch Child Fetal Neonatal Ed 2011; 95(5): F352–8.Google Scholar
Toet, MC, Lemmers, PM, van Schelven, LJ, van Bel, F. Cerebral oxygenation and electrical activity after birth asphyxia: their relation to outcome. Pediatrics 2006; 117(2): 333–9.CrossRefGoogle ScholarPubMed
Ancora, G, Maranella, E, Grandi, S, et al. Early predictors of short term neurodevelopmental outcome in asphyxiated cooled infants: a combined brain amplitude integrated electroencephalography and near infrared spectroscopy study. Brain Dev 2013; 35(1): 2631.Google Scholar
Lemmers, PM, Zwanenburg, RJ, Benders, MJ, et al. Cerebral oxygenation and brain activity after perinatal asphyxia: does hypothermia change their prognostic value? Pediatr Res 2013; 74(2): 180–5.Google Scholar
Massaro, AN, Bouyssi-Kobar, M, Chang, T, et al. Brain perfusion in encephalopathic newborns after therapeutic hypothermia. AJNR Am J Neuroradiol 2013; 34(8): 1649–55.CrossRefGoogle ScholarPubMed
Wintermark, P, Hansen, A, Warfield, SK, et al. Near-infrared spectroscopy versus magnetic resonance imaging to study brain perfusion in newborns with hypoxic-ischemic encephalopathy treated with hypothermia. NeuroImage 2014; 85(1): 287–93.Google Scholar
Peng, S, Boudes, E, Tan, X, et al. Does near-infrared spectroscopy identify asphyxiated newborns at risk of developing brain injury during hypothermia treatment? Am J Perinatol 2015; 32(6): 555–64.Google Scholar
Massaro, AN, Govindan, RB, Vezina, G, et al. Impaired cerebral autoregulation and brain injury in newborns with hypoxic-ischemic encephalopathy treated with hypothermia. J Neurophysiol 2015; 114(2): 818–24.Google Scholar
Shellhaas, RA, Thelen, BJ, Bapuraj, JR, et al. Limited short-term prognostic utility of cerebral NIRS during neonatal therapeutic hypothermia. Neurology 2013; 81(3): 249–55.Google Scholar
Austin, EH 3rd, Edmonds, HLJ, Auden, SM, et al. Benefit of neurophysiologic monitoring for pediatric cardiac surgery. J Thorac Cardiovasc Surg 1997; 114(5): 707–15, 717; discussion 715–6.Google Scholar
Sood, ED, Benzaquen, JS, Davies, RR, et al. Predictive value of perioperative near-infrared spectroscopy for neurodevelopmental outcomes after cardiac surgery in infancy. J Thorac Cardiovasc Surg 2013; 145(2): 438–45.e1; discussion 444–5.Google Scholar
Hoffman, GM, Brosig, CL, Mussatto, KA, et al. Perioperative cerebral oxygen saturation in neonates with hypoplastic left heart syndrome and childhood neurodevelopmental outcome. J Thorac Cardiovasc Surg 2013; 146(5): 1153–64.Google Scholar
McQuillen, PS, Hamrick, SEG, Perez, MJ, et al. Balloon atrial septostomy is associated with preoperative stroke in neonates with transposition of the great arteries. Circulation 2006; 113(2): 280–5.Google Scholar
Toet, MC, Flinterman, A, Laar, I, et al. Cerebral oxygen saturation and electrical brain activity before, during, and up to 36 hours after arterial switch procedure in neonates without pre-existing brain damage: its relationship to neurodevelopmental outcome. Exp Brain Res 2005; 165(3): 343–50.Google Scholar
Johnson, BA, Hoffman, GM, Tweddell, JS, et al. Near-infrared spectroscopy in neonates before palliation of hypoplastic left heart syndrome. Ann Thorac Surg 2009; 87(2): 571–7.Google Scholar
Papademetriou, MD, Tachtsidis, I, Elliot, MJ, et al. Multichannel near infrared spectroscopy indicates regional variations in cerebral autoregulation in infants supported on extracorporeal membrane oxygenation. J Biomed Opt 2012; 17(6): 067008.Google Scholar
Rhondali, O, Juhel, S, Mathews, S, et al. Impact of sevoflurane anesthesia on brain oxygenation in children younger than 2 years. Paediatr Anaesth 2014; 24(7): 734–40.Google Scholar
Conforti, A, Giliberti, P, Mondi, V, et al. Near infrared spectroscopy: experience on esophageal atresia infants. J Pediatr Surg 2014; 49(7): 1064–8.Google Scholar
Kawamura, T, Kakogawa, J, Takeuchi, Y, et al. Measurement of placental oxygenation by transabdominal near-infrared spectroscopy. Am J Perinatol 2007; 24(3): 161–6.Google Scholar
Arimitsu, T, Uchida-Ota, M, Yagihashi, T, et al. Functional hemispheric specialization in processing phonemic and prosodic auditory changes in neonates. Front Psychol 2011; 2: 202.Google Scholar
Bartocci, M, Bergqvist, LL, Lagercrantz, H, Anand, KJS. Pain activates cortical areas in the preterm newborn brain. Pain 2006; 122(1–2): 109–17.Google Scholar
Benaron, DA, Parachikov, IH, Friedland, S, et al. Continuous, noninvasive, and localized microvascular tissue oximetry using visible light spectroscopy. Anesthesiology 2004; 100(6): 1469–75.Google Scholar
Heninger, C, Ramamoorthy, C, Amir, G, et al. Esophageal saturation during antegrade cerebral perfusion: a preliminary report using visible light spectroscopy. Paediatr Anaesth 2006; 16(11): 1133–7.Google Scholar

References

Lakshmanan, J, Ross, MG. Mechanism(s) of in utero meconium passage. J Perinatol 2008; 28(Suppl 3): S813.CrossRefGoogle ScholarPubMed
Miller, PW, Coen, RW, Benirschke, K. Dating the time interval from meconium passage to birth. Obstet Gynecol 1985; 66: 459–62.Google Scholar
Incerti, M, Locatelli, A, Consonni, S, et al. Can placental histology establish the timing of meconium passage during labor? Acta Obstet Gynecol Scand 2011; 90: 863–8.Google Scholar
Sepulveda, WH, Gonzalez, C, Cruz, MA, et al. Vasoconstrictive effect of bile acids on isolated human placental chorionic veins. Eur J Obstet Gynecol 1995; 173: 1075–8.Google Scholar
Holcberg, G, Huleihel, M, Katz, M, et al. Vasoconstrictive activity of meconium stained amniotic fluid in the human placental vasculature. Eur J Obstet Gynecol Reprod Biol 1999; 87: 147–50.CrossRefGoogle ScholarPubMed
King, EL, Redline, RW, Smith, SD, et al. Myocytes of chorionic vessels from placentas with meconium-associated vascular necrosis exhibit apoptotic markers. Hum Pathol 2004; 35: 412–17.Google Scholar
Holcberg, G, Sapir, O, Huleihel, M, et al. Vasoconstrictive activity of oxytocin in meconium impregnated human placentas. Eur J Obstet Gynecol Reprod Biol 2002; 101: 139–42.Google Scholar
Cimic, A, Baergen, R. Meconium-associated umbilical vascular myonecrosis: correlations with adverse outcome. Pediatr Dev Pathol 2016; 19(4): 315–9.Google Scholar
Altshuler, G, Hyde, S. Meconium-induced vasocontraction: a potential cause of cerebral and other fetal hypoperfusion and of poor pregnancy outcome. J Child Neurol 1989; 4: 137–42.Google Scholar
Altshuler, G, Arizawa, M, Molnar-Nadasy, G. Meconium-induced umbilical cord vascular necrosis and ulceration: a potential link between the placenta and poor outcome. Obstet Gynecol 1992; 79: 760–6.Google Scholar
Locatelli, A, Regalia, AL, Patregnani, C, et al. Prognostic value of change in amniotic fluid color curing labor. Fetal Diagn Ther 2005; 20: 59.Google Scholar
Sheiner, E, Hadar, A, Shoham-Vardi, I. The effect of meconium on perinatal outcome: a prospective analysis. J Matern Fetal Neonat Med 2002; 11: 54–9.Google Scholar
De Beaufort, AJ, Pelikan, DMV, Elferink, JGR, et al. Effect of interleukin 8 in meconium on in vitro neutrophil chemotaxis. Lancet 1998; 352: 102–5.Google Scholar
Burgess, AM, Hutchins, GM. Inflammation of the lungs, umbilical cord and placenta associated with meconium passage in utero. Pathol Res Pract 1996; 192: 1121–8.Google Scholar
Yamada, T, Minakami, H, Matsubara, S, et al. Meconium-stained amniotic fluid exhibits chemotactic activity for polymorphonuclear leukocytes in vitro. J Reprod Immunol 2000; 46: 2130.Google Scholar
Ferber, A, Minior, VK, Bornstein, E, et al. Fetal “nonreassuring status” is associated with elevation of nucleated red blood cell counts and interleukin-6. Am J Obstet Gynecol 2005; 192: 1427–9.Google Scholar
Dulay, AT, Buhimschi, IA, Zhao, G, et al. Nucleated red blood cells are a direct response to mediators of inflammation in newborns with early-onset neonatal sepsis. Am J Obstet Gynecol 2008; 198: 426.Google Scholar
Ferber, A, Minior, VK, Bornstein, E, et al. Fetal “nonreassuring status” is associated with elevation of nucleated red blood cell counts and interleukin-6. J Obstet Gynecol 2005; 192: 1427–9.Google Scholar
Ferber, A, Fridel, Z, Weissmann-Brenner, A, et al. Are elevated nucleated red blood cell counts an indirect reflection of enhanced erythropoietin activity? Am J Obstet Gynecol 2004; 190: 1473–5.Google Scholar
Maier, RF, Gunther, A, Vogel, M, et al. Umbilical venous erythropoietin and umbilical arterial pH in relation to morphologic placental abnormalities. Obstet Gynecol 1994; 84: 81–7.Google Scholar
Hermansen, MC. Nucleated red blood cells in the fetus and newborn. Arch Dis Child Fetal Neonatal Ed 2001; 84: F211–15.Google Scholar
Silva, AM, Smith, RN, Lehmann, CU, et al. Neonatal nucleated red blood cells and the prediction of cerebral white matter injury in preterm infants. Obstet Gynecol 2006; 107: 550–6.Google Scholar
Phelan, JP, Korst, LM, Ahn, MO, et al. Neonatal nucleated red blood cell and lymphocyte counts in fetal brain injury. Obstet Gynecol 1998; 91: 485–9.Google Scholar
Korst, LM, Phelan, JP, Ahn, MO, et al. Nucleated red blood cells: an update in the marker for fetal asphyxia. Am J Obstet Gynecol 1996; 175: 843–6.Google Scholar
Phelan, JP, Ahn, MO, Korst, LM, et al. Nucleated red blood cells: a marker for fetal asphyxia? Am J Obstet Gynecol 1995; 173: 1380–4.Google Scholar
Axt-Fliedner, R, Hendrik, HJ, Schmidt, W. Nucleated red blood cell counts in growth-restricted neonates with absent or reversed-end-diastolic umbilical artery velocity. Clin Exp Obstet Gynecol 2002; 29: 242–6.Google Scholar
McCarthy, JM, Capullari, T, Thompson, Z, et al. Umbilical nucleated red blood cell counts: normal values and the effect of labor. J Perinatol 2006; 26: 8992.Google Scholar
Redline, RW, O’Riordan, A. Placental lesions associated with cerebral palsy and neurologic impairment following term birth. Arch Pathol Lab Med 2000; 124: 1785–91.Google Scholar
Manegold, G, Meyer-Monard, S, Tichelli, A, et al. Cesarean section due to fetal distress increases the number of stem cells in umbilical cord blood. Transfusion 2008; 48: 871–6.CrossRefGoogle ScholarPubMed
Baschat, AA, Gungor, S, Kush, ML, et al. Nucleated red blood cell counts in the first week of life: a critical appraisal of relationships with perinatal outcome in preterm growth-restricted neonates. Am J Obstet Gynecol 2007; 197(286): e18.Google Scholar
Buonocore, G, Perrone, S, Gioia, D, et al. Nucleated red blood cell count at birth as an index of perinatal brain damage. Am J Obstet Gynecol 1999; 181: 1500–5.Google Scholar
Ghosh, B, Mittal, S, Kumar, S, et al. Prediction of perinatal asphyxia with nucleated red blood cells in cord blood of newborns. Int J Gynaecol Obstet 2003; 81: 267–71.Google Scholar
Hanion-Lundberg, KM, Kirby, RS, Gandhi, S, et al. Nucleated red blood cells in cord blood of singleton term neonates. Am J Obstet Gynecol 1997; 176: 1149–54.Google Scholar
Hebbar, S, Misha, M, Rai, L. Significance of maternal and cord blood nucleated red blood cell count in pregnancies complicated by preeclampsia. J Pregnancy 2014; 496416.Google Scholar
Hamrick, SE, Miller, SP, Newton, NR, et al. Neonatal red blood cell counts: not associated with brain injury or outcome. Pediatr Neurol 2003; 29: 278–83.Google Scholar
Li, J, Kobata, K, Kamei, Y, et al. Nucleated red blood cell counts: an early predictor of brain injury and 2-year outcome in neonates with hypoxic-ischemic encephalopathy in the era of cooling-based treatment. Brain Dev 2014; 36: 472–8.Google Scholar
Walsh, BH, Boylan, GB, Murray, DM. Nucleated red blood cells and early EEG: predicting Sarnat stage and two year outcome. Early Hum Dev 2011; 87: 335–9.Google Scholar
Christensen, RD, Henry, E, Andres, RL, et al. Reference ranges for blood concentrations of nucleated red blood cells in neonates. Neonatology 2011; 99: 289–94.Google Scholar
Phelan, JP, Ahn, MO, Korst, L, et al. Intrapartum asphyxial brain injury with absent multiorgan system dysfunction. J Matern Fetal Med 1998; 7: 1922.Google Scholar
Benirschke, K. The use of the placenta in the understanding of perinatal injury. In Donn, SM, Fisher, CW, eds., Risk Management Techniques in Perinatal and Neonatal Practice. Armonk, NY: Futura, 1996: 325–45.Google Scholar
Grafe, MR. The correlation of prenatal brain damage with placental pathology. J Neuropathol Exp Neurol 1994; 53: 407–15.Google Scholar
Baergen, RN. Manual of Benirschke and Kaufmann’s Pathology of the Human Placenta. New York: Springer, 2005.Google Scholar
Benirschke, K, Kaufmann, P, Baergen, RN. Pathology of the Human Placenta, 32nd edn. New York: Springer, 2006.Google Scholar
Miller, ME, Higginbottom, M, Smith, DA. Short umbilical cord: its origin and relevance. Pediatrics 1981; 67: 618–21.Google Scholar
Kumazaki, K, Nakayama, M, Sumida, Y, et al. Placental features in preterm infants with periventricular leukomalacia. Pediatrics 2002; 109: 650–5.Google Scholar
Chasen, ST, Baergen, RN. Necrotizing funisitis with intrapartum umbilical cord rupture. J Perinatol 1999; 19: 325–6.Google Scholar
Altshuler, G, Arizawa, M, Molnar-Nadasdy, G. Meconium induced umbilical cord vascular necrosis and ulceration: a potential link between the placenta and poor pregnancy outcome. Obstet Gynecol 1992; 79: 760–6.Google Scholar
Santamaria, M, Benirschke, K, Carpenter, PM, et al. Transplacental hemorrhage associated with placental neoplasms. Pediatr Pathol 1987; 7: 601–15.Google Scholar
Karsidag, ATK, Kars, B, Dansuk, R, et al. Brain damage to the survivor within 30 minutes of co-twin demise in monochorionic twins. Fetal Diagn Ther 2005; 20: 91–5.Google Scholar
Spellancy, WN, Gravem, H, Fisch, RO. The umbilical cord complications of true knots, nuchal coils and cords around the body: a report from the collaborative study of cerebral palsy. Am J Obstet Gynecol 1966; 94: 1136–42.Google Scholar
Baergen, RN, Malicki, D, Behling, C, et al. Morbidity, mortality, and placental pathology in excessively long umbilical cords: retrospective study. Pediatr Dev Pathol 2001; 4: 144–53.Google Scholar
Spinillo, A, Cesari, S, Bariselli, S, et al. Placental lesions associated with oligohydramnios in fetal growth restricted (FGR) pregnancies. Placenta 2015; 36: 538–44.Google Scholar
Machin, GA, Ackerman, J, Gilbert-Barness, E. Abnormal umbilical cord coiling is associated with adverse perinatal outcomes. Pediatr Dev Pathol 2000; 3: 462–71.Google Scholar
Redline, RW. Clinical and pathological umbilical cord abnormalities in fetal thrombotic vasculopathy. Hum Pathol 2004; 35: 1494–8.Google Scholar
Peng, HQ, Levitin-Smith, M, Rochelson, B, et al. Umbilical cord stricture and overcoiling are common causes of fetal demise. Pediatr Dev Pathol 2006; 9: 1419.CrossRefGoogle ScholarPubMed
Murphy, DJ, MacKenzie, IZ. The mortality and morbidity associated with umbilical cord prolapse. Br J Obstet Gynaecol 1995; 102: 826–30.Google Scholar
Ikeda, T, Murata, Y, Quilligan, EJ, et al. Physiologic and histologic changes in near-term fetal lambs exposed to asphyxia by partial umbilical cord occlusion. Am J Obstet Gynecol 1998; 178: 2432.Google Scholar
Kraus, FT, Acheen, VI. Fetal thrombotic vasculopathy in the placenta: cerebral thrombi and infarcts, coagulopathies and cerebral palsy. Hum Pathol 1999; 30: 759–69.Google Scholar
Heifetz, SA. Thrombosis of the umbilical cord: analysis of 52 cases and literature review. Pediatr Pathol 1988; 8: 3754.Google Scholar
Kalish, RB, Hunter, T, Sharma, G, et al. Clinical significance of the umbilical cord twist. Am J Obstet Gynecol 2003; 189: 736–9.Google Scholar
Strong, TH, Elliott, JP, Radin, TR. Non-coiled umbilical blood vessels: a new marker for the fetus at risk. Obstet Gynecol 1993; 81: 409–11.Google ScholarPubMed
Benirschke, K. Obstetrically important lesions of the umbilical cord. J Reprod Med 1994; 39: 6272.Google Scholar
de Laat, MWM, van Alderen, ED, Franx, A, et al. The umbilical coiling index in complicated pregnancy. Eur J Obstet Gynecol Reprod Biol 2007; 130: 6672.Google Scholar
Heifetz, SA. The umbilical cord: obstetrically important lesions. Clin Obstet Gynecol 1996; 39: 7187.Google Scholar
Hersh, J, Buchino, JJ. Umbilical cord torsion/constriction sequence. In Saul, RA, ed., Proceedings of the Greenwood Genetics Conference, vol. 7. Clinton, SC: Jacobs Press, 1988: 181–2.Google Scholar
Gardiner, JP. The umbilical cord: normal length; length in cord complications; etiology and frequency of coiling. Surg Gynecol Obstet 1922; 34: 252–6.Google Scholar
Snider, W. Placental pathology casebook. J Perinatol 1997; 17: 327–9.Google Scholar
Naeye, RL, Tafari, N. Noninfectious disorders of the placenta, fetal membranes and umbilical cord. In Naeye, RL, Tafari, N, eds., Risk Factors in Pregnancy and Disease of the Fetus and Newborn. Baltimore: Williams & Wilkins, 1983: 145–72.Google Scholar
Moessinger, AC, Blanc, WA, Marone, PA, et al. Umbilical cord length as an index of fetal activity: experimental study and clinical implications. Pediatr Res 1982; 16: 109–12.Google Scholar
Boué, DR, Stanley, C, Baergen, RN. Placental pathology casebook. J Perinatol 1995; 15: 429–31.Google Scholar
Faye-Petersen, O, Baergen, RN. Long umbilical cords and pre-viable fetal death. Pediatr Dev Pathol 2001; 4: 414.Google Scholar
Gilbert-Barness, E, Drut, RM, Drut, R, et al. Developmental abnormalities resulting in short umbilical cord. Birth Defects Original Article Series 1993; 29: 113–40.Google Scholar
Collins, JC. Umbilical cord accidents: human studies. Semin Perinatol 2002; 26: 7982.Google Scholar
Soernes, T. Umbilical cord encirclements and fetal growth restriction. Obstet Gynecol 1995; 86: 725–8.Google Scholar
Vanhaesebrouck, P, Vanneste, K, de Praeter, C, et al. Tight nuchal cord and neonatal hypovolemic shock. Arch Dis Child 1987; 62: 1276–7.Google Scholar
Nelson, KB, Grether, JK. Potentially asphyxiating conditions and spastic cerebral palsy in infants of normal birth weight. Am J Obstet Gynecol 1998; 179: 507–13.Google Scholar
Lin, MG. Umbilical cord prolapse. Obstet Gynecol Surv 2006; 61: 269–77.Google Scholar
Torrey, WE. Vasa previa. Am J Obstet Gynecol 1952; 63: 146–52.Google Scholar
Heinonen, S, Ryynänen, M, Kirkinen, P, et al. Perinatal diagnostic evaluation of velamentous umbilical cord insertion: clinical, Doppler, and ultrasonic findings. Obstet Gynecol 1996; 87: 112–17.Google Scholar
Cordero, DR, Helfgott, AW, Landy, HJ, et al. A non-hemorrhagic manifestation of vasa previa: a clinicopathologic case report. Obstet Gynecol 1993; 82: 698700.Google Scholar
Redline, RW, Ariel, I, Baergen, RN, et al. Fetal vascular obstructive lesions: nosology and reproducibility of placental reaction patterns. Pediatr Dev Pathol 2004; 7: 443–52.Google Scholar
Sander, CM, Gilliland, D, Akers, C, et al. Livebirths with placental hemorrhagic endovasculitis: interlesional relationships and perinatal outcomes. Arch Pathol Lab Med 2002; 126: 157–64.Google Scholar
Redline, RW. Severe fetal placental vascular lesions in term infants with neurologic impairment. Am J Obstet Gynecol 2005; 192: 452–7.Google Scholar
Bejar, RF, Wozniak, P, Allard, M, et al. Antenatal origin of neurologic damage in newborn infants. I. Preterm infants. Am J Obstet Gynecol 1988; 159: 357–63.Google Scholar
Redline, RW, Wilson-Costello, D, Borawski, E, et al. The relationship between placental and other perinatal risk factors for neurologic impairment in very low birth weight children. Pediatr Res 2000; 47: 721–6.Google Scholar
Redline, RW, Boyd, T, Campbell, V, et al. Maternal vascular malperfusion: nosology and reproducibility of placental reaction patterns. Pediatr Dev Pathol 2004; 7: 237–49.Google Scholar
Burke, CJ, Tannenberg, AE, Payton, DJ. Ischaemic cerebral injury, intrauterine growth retardation, and placental infarction. Dev Med Child Neurol 1997; 39: 726–30.Google Scholar
Redline, RW, Wilson-Costello, D, Borawski, E, et al. Placental lesions associated with neurologic impairment and cerebral palsy in very low-birth-weight infants. Arch Pathol Lab Med 1998; 122: 1091–8.Google Scholar
Parra-Saavedra, M, Simeone, S, Triunfo, S, et al. Correlation between histological signs of placental underperfusion and perinatal morbidity in late-onset small-for-gestational-age fetuses. Ultrasound Obstet Gynecol 2015; 45: 149–55.Google Scholar
Ananth, CV, Friedman, AM. Ischemic placental disease and risks of perinatal mortality and morbidity and neurodevelopmental outcomes. Semin Perinatol 2014; 38: 151–8.Google Scholar
Redline, RW. Villitis of unknown etiology: noninfectious chronic villitis in the placenta. Hum Pathol 2007; 38: 1439–46.Google Scholar
Bendon, RW, Hommel, AB. Maternal floor infarction in autoimmune disease: two cases. Pediatr Pathol Lab Med 1996; 16: 293–7.Google Scholar
Sebire, NJ, Backos, M, Goldin, RD, et al. Placental massive perivillous fibrin deposition associated with antiphospholipid syndrome. BJOG 2002; 109: 570–3.Google Scholar
Katzman, PJ, Genest, DR. Maternal floor infarction and massive previllous fibrin deposition: histological definitions, association with intrauterine fetal growth restriction, and risk of recurrence. Pediatr Dev Pathol 2002; 5: 159–64.Google Scholar
Chang, P, Miller, D, Tsang, P, et al. Intravenous immunoglobulin in antiphospholipid syndrome and maternal floor infarction when standard treatment fails: a case report. Am J Perinatol 2006; 23: 125–9.Google Scholar
Makino, A, Suzuki, Y, Yamamoto, T, et al. Use of aspirin and low-molecular-weight heparin to prevent recurrence of maternal floor infarction in women without evidence of antiphospholipid antibody syndrome. Fetal Diagn Ther 2004; 19: 261–5.Google Scholar
Chang, P, Millar, D, Tsang, P, et al. Intravenous immunoglobulin in antiphospholipid syndrome and maternal floor infarction when standard treatment fails. Am J Perinatol 2006; 23: 125–9.Google Scholar
Kim, CJ, Romero, R, Chaemsaithong, P, et al. Chronic inflammation of the placenta: definition, classification, pathogenesis, and clinical significance. Am J Obstet Gynecol 2015; 213(4 Suppl): S5369.Google Scholar
Rudzinski, E, Gilroy, M, Newbill, C, et al. Positive C4d immunostaining of placental villous syncytiotrophoblasts supports host-versus-graft rejection in villitis of unknown etiology. Pediatr Dev Pathol 2013; 16: 713.Google Scholar
Redline, RW, O’Riordan, MA. Placental lesions associated with cerebral palsy and neurologic impairment following term birth. Arch Pathol Lab Med 2000; 124: 1785–91.Google Scholar
Scher, MS, Trucco, GS, Beggarly, ME, et al. Neonates with electrically confirmed seizures and possible placental associations. Pediatr Neurol 1998; 19: 3741.Google Scholar
Mir, IN, Johnson-Welch, SF, Nelson, DB, et al. Placental pathology is associated with severity of neonatal encephalopathy and adverse developmental outcomes following hypothermia. Am J Obstet Gynecol 2015; 213:849.e17.Google Scholar
Lee, J, Romero, R, Chaiworapongsa, T, et al. Characterization of the fetal blood transcriptome and proteome in maternal anti-fetal rejection: evidence of a distinct and novel type of human fetal systemic inflammatory response. Am J Reprod Immunol 2013 Oct; 70: 265–84.Google Scholar
Adams-Chapman, I, Vaucher, YE, Bejar, RF, et al. Maternal floor infarction of the placenta: association with central nervous system injury and adverse neurodevelopmental outcome. J Perinatol 2002; 22: 236–41.Google Scholar
Andres, RL, Kuyper, W, Resnik, R, et al. The association of maternal floor infarction of the placenta with adverse perinatal outcome. Am J Obstet Gynecol 1990; 163: 935–8.Google Scholar
Heller, D, Tellier, R, Pabbaraju, K, et al. Placental massive perivillous fibrinoid deposition associated with coxsackievirus A16: report of a case and review of the literature. Pediatr Dev Pathol 2016; 19(5): 421–3.Google Scholar
Romero, R, Whitten, A, Korzeniewski, SJ, et al. Maternal floor infarction/massive perivillous fibrin deposition: a manifestation of maternal antifetal rejection? Am J Reprod Immunol 2013; 70: 285–98.Google Scholar
Linn, RL, Kiley, J, Minturn, L, et al. Recurrent massive perivillous fibrin deposition in the placenta associated with fetal renal tubular dysgenesis: case report and literature review. Pediatr Dev Pathol 2013; 16: 378–86.Google Scholar
Al-Adnani, M, Kiho, L, Scheimberg, I, et al. Recurrent placental massive perivillous fibrin deposition associated with polymyositis: a case report and review of the literature. Pediatr Dev Pathol 2008; 11: 226–9.Google Scholar
Griffin, AC, Strauss, AW, Bennett, MJ, et al. Mutations in long-chain 3-hydroxyacyl coenzyme a dehydrogenase are associated with placental maternal floor infarction/massive perivillous fibrin deposition. Pediatr Dev Pathol 2012; 15: 368–74.Google Scholar

References

Reid, SM, Carlin, JB, Reddihough, DS. Rates of cerebral palsy in Victoria, Australia, 1970 to 2004: has there been a change? Dev Med Child Neurol 2011; 53: 907–12.Google Scholar
Behrman, RE, Butler, AS, eds. Preterm Birth: Causes, Consequences, and Prevention. Washington, DC: Institute of Medicine of the National Academies, 2007 Available at http://books.nap.edu/openbook.php?record_id=11622&page=1.Google Scholar
McIntyre, S, Blair, E, Badawi, N, et al. Antecedents of cerebral palsy and perinatal death in term and late preterm singletons. Obstet Gynecol 2013; 122: 869–77.Google Scholar
Himmelmann, K, Hagberg, G, Beckung, E, et al. The changing panorama of cerebral palsy in Sweden. IX. Prevalence and origin in the birth-year period 1995–1998. Acta Paediatr 2005; 94: 287–94.Google Scholar
Centers for Disease Control and Prevention (CDC). Economic costs associated with mental retardation, cerebral palsy, hearing loss, and vision impairment–United States, 2003. MMWR Morb Mortal Wkly Rep 2004; 53: 57–9.Google Scholar
Kruse, M, Michelsen, SI, Flachs, EM, et al. Lifetime costs of cerebral palsy. Dev Med Child Neurol 2009; 51: 622–8.Google Scholar
Gunn, AJ, Bennet, L. Timing of injury in the fetus and neonate. Curr Opin Obstet Gynecol 2008; 20: 175–81.Google Scholar
Jacobs, SE, Berg, M, Hunt, R, et al. Cooling for newborns with hypoxic ischaemic encephalopathy. Cochrane Database Syst Rev 2013; 1: CD003311.Google Scholar
Gunn, AJ, Thoresen, M. Hypothermic neuroprotection. NeuroRx 2006; 3: 154–69.Google Scholar
Beilharz, EJ, Williams, CE, Dragunow, M, et al. Mechanisms of delayed cell death following hypoxic-ischemic injury in the immature rat: evidence for apoptosis during selective neuronal loss. Brain Res Mol Brain Res 1995; 29: 114.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
Roth, SC, Baudin, J, Cady, E, et al. Relation of deranged neonatal cerebral oxidative metabolism with neurodevelopmental outcome and head circumference at 4 years. Dev Med Child Neurol 1997; 39: 718–25.Google Scholar
Shankaran, S, Laptook, AR, Ehrenkranz, RA, et al. Whole-body hypothermia for neonates with hypoxic-ischemic encephalopathy. N Engl J Med 2005; 353: 1574–84.Google Scholar
Gluckman, PD, Wyatt, JS, Azzopardi, D, et al. Selective head cooling with mild systemic hypothermia after neonatal encephalopathy: multicentre randomised trial. Lancet 2005; 365: 663–70.Google Scholar
Westgate, JA, Wibbens, B, Bennet, L, et al. The intrapartum deceleration in center stage: a physiological approach to interpretation of fetal heart rate changes in labor. Am J Obstet Gynecol 2007; 197: e1e11.236.Google Scholar
de Haan, HH, Gunn, AJ, Williams, CE, Gluckman, PD. Brief repeated umbilical cord occlusions cause sustained cytotoxic cerebral edema and focal infarcts in near-term fetal lambs. Pediatr Res 1997; 41: 96104.Google Scholar
Graham, EM, Ruis, KA, Hartman, AL, et al. A systematic review of the role of intrapartum hypoxia-ischemia in the causation of neonatal encephalopathy. Am J Obstet Gynecol 2008; 199: 587–95.Google Scholar
MacLennan, A, International Cerebral Palsy Task Force, et al. A template for defining a causal relation between acute intrapartum events and cerebral palsy: international consensus statement. BMJ 1999; 319: 1054–9.Google Scholar
Cowan, F, Rutherford, M, Groenendaal, F, et al. Origin and timing of brain lesions in term infants with neonatal encephalopathy. Lancet 2003; 361: 736–42.Google Scholar
Iwata, O, Iwata, S, Bainbridge, A, et al. Supra- and sub-baseline phosphocreatine recovery in developing brain after transient hypoxia-ischaemia: relation to baseline energetics, insult severity and outcome. Brain 2008; 131: 2220–6.Google Scholar
Szeto, HH. Mitochondria-targeted cytoprotective peptides for ischemia-reperfusion injury. Antioxid Redox Signal 2008; 10: 601–19.Google Scholar
Edwards, AD, Brocklehurst, P, Gunn, AJ, et al. Neurological outcomes at 18 months of age after moderate hypothermia for perinatal hypoxic ischaemic encephalopathy: synthesis and meta-analysis of trial data. BMJ 2010; 340: c363.Google Scholar
Nelson, KB, Dambrosia, JM, Ting, TY, Grether, JK. Uncertain value of electronic fetal monitoring in predicting cerebral palsy. N Engl J Med 1996; 334: 613–8.Google Scholar
Murray, DM, O’Riordan, MN, Horgan, R, et al. Fetal heart rate patterns in neonatal hypoxic-ischemic encephalopathy: relationship with early cerebral activity and neurodevelopmental outcome. Am J Perinatol 2009; 26: 605–12.Google Scholar
Wayenberg, JL. Threshold of metabolic acidosis associated with neonatal encephalopathy in the term newborn. J Matern Fetal Neonatal Med 2005; 18: 381–5.Google Scholar
Low, JA, Lindsay, BG, Derrick, EJ. Threshold of metabolic acidosis associated with newborn complications. Am J Obstet Gynecol 1997; 177: 1391–4.Google Scholar
Shah, S, Tracy, M, Smyth, J. Postnatal lactate as an early predictor of short-term outcome after intrapartum asphyxia. J Perinatol 2004; 24: 1620.Google Scholar
Boardman, JP, Hawdon, JM. Hypoglycaemia and hypoxic-ischaemic encephalopathy. Dev Med Child Neurol 2015; 57: 2933.Google Scholar
Nadeem, M, Murray, DM, Boylan, GB, et al. Early blood glucose profile and neurodevelopmental outcome at two years in neonatal hypoxic-ischaemic encephalopathy. BMC Pediatr 2011; 11: 10.Google Scholar
Chouthai, NS, Sobczak, H, Khan, R, et al. Hyperglycemia is associated with poor outcome in newborn infants undergoing therapeutic hypothermia for hypoxic ischemic encephalopathy. J Neonatal Perinatal Med 2015; 8: 125–31.Google Scholar
Basu, SK, Kaiser, JR, Guffey, D, et al. Hypoglycaemia and hyperglycaemia are associated with unfavourable outcome in infants with hypoxic ischaemic encephalopathy: a post hoc analysis of the CoolCap Study. Arch Dis Child Fetal Neonatal Ed 2016; 101:F149–55.Google Scholar
Shelley, HJ. Glycogen reserves and their changes at birth and in anoxia. Br Med Bull 1961; 17: 137–43.Google Scholar
Dawes, GS, Mott, JC, Shelley, HJ. The importance of cardiac glycogen for the maintenance of life in foetal lambs and newborn animals during anoxia. J Physiol 1959; 146: 516–38.Google Scholar
Wassink, G, Bennet, L, Booth, LC, et al. The ontogeny of hemodynamic responses to prolonged umbilical cord occlusion in fetal sheep. J Appl Physiol 2007; 103: 1311–7.Google Scholar
Laptook, AR, Shankaran, S, Ambalavanan, N, et al. Outcome of term infants using Apgar scores at 10 minutes following hypoxic-ischemic encephalopathy. Pediatrics 2009; 124: 1619–26.Google Scholar
Natarajan, G, Shankaran, S, Laptook, AR, et al. Apgar scores at 10 min and outcomes at 6–7 years following hypoxic-ischaemic encephalopathy. Arch Dis Child Fetal Neonatal Ed 2013; 98: F473–9.Google Scholar
Bayram, F, Ozerkan, K, Cengiz, C, et al. Perinatal asphyxia is associated with the umbilical cord nucleated red blood cell count in pre-eclamptic pregnancies. J Obstet Gynaecol 2010; 30: 383–6.Google Scholar
Li, J, Kobata, K, Kamei, Y, et al. Nucleated red blood cell counts: an early predictor of brain injury and 2-year outcome in neonates with hypoxic-ischemic encephalopathy in the era of cooling-based treatment. Brain Dev 2014; 36: 472–8.Google Scholar
Christensen, RD, Lambert, DK, Richards, DS. Estimating the nucleated red blood cell ‘emergence time’ in neonates. J Perinatol 2014; 34: 116–9.Google Scholar
Ferber, A, Fridel, Z, Weissmann-Brenner, A, et al. Are elevated fetal nucleated red blood cell counts an indirect reflection of enhanced erythropoietin activity? Am J Obstet Gynecol 2004; 190: 1473–5.Google Scholar
Christensen, RD, Henry, E, Andres, RL, Bennett, ST. Reference ranges for blood concentrations of nucleated red blood cells in neonates. Neonatology 2011; 99: 289–94.CrossRefGoogle ScholarPubMed
Bennet, L, Booth, L, Gunn, AJ. Potential biomarkers for hypoxic-ischemic encephalopathy. Semin Fetal Neonatal Med 2010; 15: 253–60.Google Scholar
Gazzolo, D, Abella, R, Marinoni, E, et al. New markers of neonatal neurology. J Matern Fetal Neonatal Med 2009; 2(Suppl 3): 5761.Google Scholar
Ramaswamy, V, Horton, J, Vandermeer, B, et al. Systematic review of biomarkers of brain injury in term neonatal encephalopathy. Pediatr Neurol 2009; 40: 215–26.Google Scholar
Chalak, LF, Sanchez, PJ, Adams-Huet, B, et al. Biomarkers for severity of neonatal hypoxic-ischemic encephalopathy and outcomes in newborns receiving hypothermia therapy. J Pediatr 2014; 164: 468–74.Google Scholar
Loukovaara, M, Teramo, K, Alfthan, H, et al. Amniotic fluid S100B protein and erythropoietin in pregnancies at risk for fetal hypoxia. Eur J Obstet Gynecol Reprod Biol 2009; 142: 115–8.Google Scholar
Routsi, C, Stamataki, E, Nanas, S, et al. Increased levels of serum S100B protein in critically ill patients without brain injury. Shock 2006; 26: 20–4.Google Scholar
Garnier, Y, Frigiola, A, Li Volti, G, et al. Increased maternal/fetal blood S100B levels following systemic endotoxin administration and periventricular white matter injury in preterm fetal sheep. Reprod Sci 2009; 16: 758–66.Google Scholar
Friel, LA, Romero, R, Edwin, S, et al. The calcium binding protein, S100B, is increased in the amniotic fluid of women with intra-amniotic infection/inflammation and preterm labor with intact or ruptured membranes. J Perinat Med 2007; 35: 385–93.Google Scholar
Schulpis, KH, Margeli, A, Akalestos, A, et al. Effects of mode of delivery on maternal-neonatal plasma antioxidant status and on protein S100B serum concentrations. Scand J Clin Lab Invest 2006; 66: 733–42.Google Scholar
Bokesch, PM, Appachi, E, Cavaglia, M, et al. A glial-derived protein, S100B, in neonates and infants with congenital heart disease: evidence for preexisting neurologic injury. Anesth Analg 2002; 95: 889–92.Google Scholar
Bruschettini, M, van den Hove, DL, Gazzolo, D, et al. A single course of antenatal betamethasone reduces neurotrophic factor S100B concentration in the hippocampus and serum in the neonatal rat. Brain Res Dev Brain Res 2005; 159: 113–8.Google Scholar
Vicente, E, Tramontina, F, Leite, MC, et al. S100B levels in the cerebrospinal fluid of rats are sex and anaesthetic dependent. Clin Exp Pharmacol Physiol 2007; 34: 1126–30.Google Scholar
Gazzolo, D, Vinesi, P, Marinoni, E, et al. S100B protein concentrations in cord blood: correlations with gestational age in term and preterm deliveries. Clin Chem 2000; 46: 9981000.Google Scholar
Wijnberger, LD, Nikkels, PG, van Dongen, AJ, et al. Expression in the placenta of neuronal markers for perinatal brain damage. Pediatr Res 2002; 51: 492–6.Google Scholar
Zaigham, M, Lundberg, F, Hayes, R, et al. Umbilical cord blood concentrations of ubiquitin carboxy-terminal hydrolase L1 (UCH-L1) and glial fibrillary acidic protein (GFAP) in neonates developing hypoxic-ischemic encephalopathy. J Matern Fetal Neonatal Med 2015:1–7.Google Scholar
Looney, AM, Walsh, BH, Moloney, G, et al. Downregulation of umbilical cord blood levels of miR-374a in neonatal hypoxic ischemic encephalopathy. J Pediatr 2015; 167: 269–73.Google Scholar
Whitehead, CL, Teh, WT, Walker, SP, et al. Circulating microRNAs in maternal blood as potential biomarkers for fetal hypoxia in-utero. PloS One 2013; 8: e78487.Google Scholar
Qu, Y, Wu, J, Chen, D, et al. MiR-139-5p inhibits HGTD-P and regulates neuronal apoptosis induced by hypoxia-ischemia in neonatal rats. Neurobiol Dis 2014; 63: 184–93.Google Scholar
Sarnat, HB, Sarnat, MS. Neonatal encephalopathy following fetal distress: a clinical and electroencephalographic study. Arch Neurol 1976; 33: 696705.Google Scholar
Wyatt, JS, Gluckman, PD, Liu, PY, et al. Determinants of outcomes after head cooling for neonatal encephalopathy. Pediatrics 2007; 119: 912–21.Google Scholar
Nagdyman, N, Komen, W, Ko, HK, et al. Early biochemical indicators of hypoxic-ischemic encephalopathy after birth asphyxia. Pediatr Res 2001; 49: 502–6.Google Scholar
Nagdyman, N, Grimmer, I, Scholz, T, et al. Predictive value of brain-specific proteins in serum for neurodevelopmental outcome after birth asphyxia. Pediatr Res 2003; 54: 270–5.Google Scholar
Thorngren-Jerneck, K, Alling, C, Herbst, A, et al. S100 protein in serum as a prognostic marker for cerebral injury in term newborn infants with hypoxic ischemic encephalopathy. Pediatr Res 2004; 55: 406–12.Google Scholar
Martins, RO, Rotta, NT, Portela, LV, Souza, DO. S100B protein related neonatal hypoxia. Arq Neuropsiquiatr 2006; 64: 24–9.Google Scholar
Murabayashi, M, Minato, M, Okuhata, Y, et al. Kinetics of serum S100B in newborns with intracranial lesions. Pediatr Int 2008; 50: 1722.Google Scholar
Bashir, M, Frigiola, A, Iskander, I, et al. Urinary S100A1B and S100BB to predict hypoxic ischemic encephalopathy at term. Front Biosci (Elite Ed) 2009; 1: 560–7.Google Scholar
Giuseppe, D, Sergio, C, Pasqua, B, et al. Perinatal asphyxia in preterm neonates leads to serum changes in protein S-100 and neuron specific enolase. Curr Neurovasc Res 2009; 6: 110–6.Google Scholar
Giussani, DA, Thakor, AS, Frulio, R, Gazzolo, D. Acute hypoxia increases S100beta protein in association with blood flow redistribution away from peripheral circulations in fetal sheep. Pediatr Res 2005; 58: 179–84.Google Scholar
Fujii, EY, Kozuki, M, Mu, J, et al. Correlation of neuron-specific enolase and S100B with histological cerebral damage in fetal sheep after severe asphyxia. Brain Res 2004; 1018: 136–40.Google Scholar
Nylen, K, Ost, M, Csajbok, LZ, et al. Serum levels of S100B, S100A1B and S100BB are all related to outcome after severe traumatic brain injury. Acta Neurochir 2008; 150: 221–7.Google Scholar
Berger, RP, Adelson, PD, Pierce, MC, et al. Serum neuron-specific enolase, S100B, and myelin basic protein concentrations after inflicted and noninflicted traumatic brain injury in children. J Neurosurg 2005; 103: 61–8.Google Scholar
Denihan, NM, Boylan, GB, Murray, DM. Metabolomic profiling in perinatal asphyxia: a promising new field. Biomed Res Int 2015; 2015: 254076.Google Scholar
Shalak, LF, Laptook, AR, Velaphi, SC, Perlman, JM. Amplitude-integrated electroencephalography coupled with an early neurologic examination enhances prediction of term infants at risk for persistent encephalopathy. Pediatrics 2003; 111: 351–7.Google Scholar
van Rooij, LG, Toet, MC, Osredkar, D, et al. Recovery of amplitude integrated electroencephalographic background patterns within 24 hours of perinatal asphyxia. Arch Dis Child Fetal Neonatal Ed 2005; 90: F245–51.Google Scholar
Murray, DM, Boylan, GB, Ryan, CA, Connolly, S. Early EEG findings in hypoxic-ischemic encephalopathy predict outcomes at 2 years. Pediatrics 2009; 124: e459–67.Google Scholar
Pezzani, C, Radvanyi-Bouvet, MF, Relier, JP, Monod, N. Neonatal electroencephalography during the first twenty-four hours of life in full-term newborn infants. Neuropediatrics 1986; 17: 11–8.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
Gunn, AJ, Wyatt, JS, Whitelaw, A, et al. Therapeutic hypothermia changes the prognostic value of clinical evaluation of neonatal encephalopathy. J Pediatr 2008; 152: 55–8.Google Scholar
Azzopardi, D. Predictive value of the amplitude integrated EEG in infants with hypoxic ischaemic encephalopathy: data from a randomised trial of therapeutic hypothermia. Arch Dis Child Fetal Neonatal Ed 2014; 99: F80–2.Google Scholar
Cseko, AJ, Bango, M, Lakatos, P, et al. Accuracy of amplitude-integrated electroencephalography in the prediction of neurodevelopmental outcome in asphyxiated infants receiving hypothermia treatment. Acta Paediatr 2013; 102: 707–11.Google Scholar
Hallberg, B, Grossmann, K, Bartocci, M, Blennow, M. The prognostic value of early aEEG in asphyxiated infants undergoing systemic hypothermia treatment. Acta Paediatr 2010; 99: 531–6.Google Scholar
Thoresen, M, Hellstrom-Westas, L, Liu, X, de Vries, LS. Effect of hypothermia on amplitude-integrated electroencephalogram in infants with asphyxia. Pediatrics 2010; 126: e131–9.Google Scholar
Padden, B, Scheer, I, Brotschi, B, et al. Does amplitude-integrated electroencephalogram background pattern correlate with cerebral injury in neonates with hypoxic-ischaemic encephalopathy? J Paediatr Child Health 2015; 51: 180–5.Google Scholar
Wong, FY, Barfield, CP, Walker, AM. Power spectral analysis of two-channel EEG in hypoxic-ischaemic encephalopathy. Early Hum Dev 2007; 83: 379–83.Google Scholar
Doyle, OM, Greene, BR, Murray, DM, et al. The effect of frequency band on quantitative EEG measures in neonates with hypoxic-ischaemic encephalopathy. In Conference Proceedings of the IEEE Engineering in Medicine and Biology Society (Piscataway, NJ: IEEE, 2007): 717–21.Google Scholar
Jiang, D, Wu, W, Jia, X, et al. Scaling exponents of EEG are related to the temporal process of the therapeutic hypothermia following ischemic brain injury. In Conference Proceedings of the IEEE Engineering in Medicine and Biology Society, vol. 1 (Piscataway, NJ: IEEE, 2009): 2192–5.Google Scholar
Tichauer, KM, Elliott, JT, Hadway, JA, Lee, TY, St Lawrence, K. Cerebral metabolic rate of oxygen and amplitude-integrated electroencephalography during early reperfusion after hypoxia-ischemia in piglets. J Appl Physiol 2009; 106: 1506–12.Google Scholar
Bennet, L, Roelfsema, V, Pathipati, P, et al. Relationship between evolving epileptiform activity and delayed loss of mitochondrial activity after asphyxia measured by near-infrared spectroscopy in preterm fetal sheep. J Physiol 2006; 572: 141–54.Google Scholar
Bennet, L, Roelfsema, V, Dean, J, et al. Regulation of cytochrome oxidase redox state during umbilical cord occlusion in preterm fetal sheep. Am J Physiol Regul Integr Comp Physiol 2007; 292: R1569–76.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
Scher, MS. Controversies regarding neonatal seizure recognition. Epileptic Disord 2002; 4: 139–58.Google Scholar
Dean, JM, George, S, Naylor, AS, et al. Partial neuroprotection with low-dose infusion of the 2-adrenergic receptor agonist clonidine after severe hypoxia in preterm fetal sheep. Neuropharmacology 2008; 55: 166–74.Google Scholar
Bennet, L, Dean, JM, Wassink, G, Gunn, AJ. Differential effects of hypothermia on early and late epileptiform events after severe hypoxia in preterm fetal sheep. J Neurophysiol 2007; 97: 572–8.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
Dean, JM, Gunn, AJ, Wassink, G, Bennet, L. Transient NMDA receptor-mediated hypoperfusion following umbilical cord occlusion in preterm fetal sheep. Exp Physiol 2006; 91: 423–29.Google Scholar
Scher, MS, Bova, JM, Dokianakis, SG, Steppe, DA. Physiological significance of sharp wave transients on EEG recordings of healthy pre-term and full-term neonates. Electroencephalogr Clin Neurophysiol 1994; 90: 179–85.Google Scholar
Vecchierini-Blineau, MF, Nogues, B, Louvet, S, Desfontaines, O. Positive temporal sharp waves in electroencephalograms of the premature newborn. Neurophysiol Clin 1996; 26: 350–62.Google Scholar
Marics, G, Cseko, A, Vasarhelyi, B, et al. Prevalence and etiology of false normal aEEG recordings in neonatal hypoxic-ischaemic encephalopathy. BMC Pediatr 2013; 13: 194.Google Scholar
Lear, CA, Koome, MM, Davidson, JO, et al. The effects of dexamethasone on post-asphyxial cerebral oxygenation in the preterm fetal sheep. J Physiol 2014; 592: 5493–505.Google Scholar
Davidson, JO, Quaedackers, JS, George, SA, et al. Maternal dexamethasone and EEG hyperactivity in preterm fetal sheep. J Physiol 2011; 589: 3823–35.Google Scholar
Counsell, SJ, Ball, G, Edwards, AD. New imaging approaches to evaluate newborn brain injury and their role in predicting developmental disorders. Curr Opin Neurol 2014; 27: 168–75.Google Scholar
Robertson, NJ, Thayyil, S, Cady, EB, Raivich, G. Magnetic resonance spectroscopy biomarkers in term perinatal asphyxial encephalopathy: from neuropathological correlates to future clinical applications. Curr Pediatr Rev 2014; 10: 3747.Google Scholar
Martin, E, Buchli, R, Ritter, S, et al. Diagnostic and prognostic value of cerebral 31P magnetic resonance spectroscopy in neonates with perinatal asphyxia. Pediatr Res 1996; 40: 749–58.Google Scholar
Thayyil, S, Chandrasekaran, M, Taylor, A, et al. Cerebral magnetic resonance biomarkers in neonatal encephalopathy: a meta-analysis. Pediatrics 2010; 125: e382–95.Google Scholar
Vannucci, RC, Towfighi, J, Vannucci, SJ. Secondary energy failure after cerebral hypoxia-ischemia in the immature rat. J Cereb Blood Flow Metab 2004; 24: 1090–7.Google Scholar
Cady, EB, Iwata, O, Bainbridge, A, et al. Phosphorus magnetic resonance spectroscopy 2 h after perinatal cerebral hypoxia-ischemia prognosticates outcome in the newborn piglet. J Neurochem 2008; 107: 1027–35.Google Scholar
Alderliesten, T, de Vries, LS, Benders, MJ, et al. MR imaging and outcome of term neonates with perinatal asphyxia: value of diffusion-weighted MR imaging and 1H MR spectroscopy. Radiology 2011; 261: 235–42.Google Scholar
Alderliesten, T, Nikkels, PG, Benders, MJ, et al. Antemortem cranial MRI compared with postmortem histopathologic examination of the brain in term infants with neonatal encephalopathy following perinatal asphyxia. Arch Dis Child Fetal Neonatal Ed 2013; 98: F304–9.Google Scholar
Shankaran, S, McDonald, SA, Laptook, AR, et al. Neonatal magnetic resonance imaging pattern of brain injury as a biomarker of childhood outcomes following a trial of hypothermia for neonatal hypoxic-ischemic encephalopathy. J Pediatr 2015; 167: 987–93.Google Scholar
Wilkinson, D. MRI and withdrawal of life support from newborn infants with hypoxic-ischemic encephalopathy. Pediatrics 2010; 126: e451–8.Google Scholar
Wisnowski, JL, Wu, TW, Reitman, AJ, et al. The effects of therapeutic hypothermia on cerebral metabolism in neonates with hypoxic-ischemic encephalopathy: an in vivo 1H-MR spectroscopy study. J Cereb Blood Flow Metab 2016; 36:1075–86.Google Scholar
Liao, SM, Culver, JP. Near infrared optical technologies to illuminate the status of the neonatal brain. Current Pediatr Rev 2014; 10: 7386.Google Scholar
Liu, P, Chalak, LF, Lu, H. Non-invasive assessment of neonatal brain oxygen metabolism: a review of newly available techniques. Early Hum Dev 2014; 90: 695701.Google Scholar
Bale, G, Mitra, S, Meek, J, et al. A new broadband near-infrared spectroscopy system for in-vivo measurements of cerebral cytochrome-c-oxidase changes in neonatal brain injury. Biomed Optics Express 2014; 5: 3450–66.Google Scholar
Drury, PP, Bennet, L, Booth, LC, et al. Maturation of the mitochondrial redox response to profound asphyxia in fetal sheep. PLoS One 2012; 7: e39273.Google Scholar
van Bel, F, Lemmers, P, Naulaers, G. Monitoring neonatal regional cerebral oxygen saturation in clinical practice: value and pitfalls. Neonatology 2008; 94: 237–44.Google Scholar
Bainbridge, A, Tachtsidis, I, Faulkner, SD, et al. Brain mitochondrial oxidative metabolism during and after cerebral hypoxia-ischemia studied by simultaneous phosphorus magnetic-resonance and broadband near-infrared spectroscopy. Neuroimage 2014; 102: 173–83.Google Scholar
Matcher, SJ, Elwell, CE, Cooper, CE, et al. Performance comparison of several published tissue near-infrared spectroscopy algorithms. Anal Biochem 1995; 227: 5468.Google Scholar
Cooper, CE, Cope, M, Springett, R, et al. Use of mitochondrial inhibitors to demonstrate that cytochrome oxidase near-infrared spectroscopy can measure mitochondrial dysfunction noninvasively in the brain. J Cereb Blood Flow Metab 1999; 19: 2738.Google Scholar
Peeters-Scholte, C, van den Tweel, E, Groenendaal, F, van Bel, F. Redox state of near infrared spectroscopy-measured cytochrome aa(3) correlates with delayed cerebral energy failure following perinatal hypoxia-ischaemia in the newborn pig. Exp Brain Res 2004; 156: 20–6.Google Scholar
Jensen, EC, Bennet, L, Hunter, CJ, et al. Post-hypoxic hypoperfusion is associated with suppression of cerebral metabolism and increased tissue oxygenation in near-term fetal sheep. J Physiol 2006; 572: 131–9.Google Scholar
Winter, JD, Tichauer, KM, Gelman, N, et al. Changes in cerebral oxygen consumption and high-energy phosphates during early recovery in hypoxic-ischemic piglets: a combined near-infrared and magnetic resonance spectroscopy study. Pediatr Res 2009; 65: 181–7.Google Scholar
Kurth, CD, McCann, JC, Wu, J, et al. Cerebral oxygen saturation-time threshold for hypoxic-ischemic injury in piglets. Anesth Analg 2009; 108: 1268–77.Google Scholar
Perlman, JM. Summary proceedings from the neurology group on hypoxic-ischemic encephalopathy. Pediatrics 2006; 117: S2833.Google Scholar
Greisen, G. Cerebral blood flow and oxygenation in infants after birth asphyxia: clinically useful information? Early Hum Dev 2014; 90: 703–5.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
Pryds, O, Greisen, G, Lou, H, Friis-Hansen, B. Vasoparalysis associated with brain damage in asphyxiated term infants. J Pediatr 1990; 117: 119–25.Google Scholar
Marks, KA, Mallard, CE, Roberts, I, et al. Nitric oxide synthase inhibition attenuates delayed vasodilation and increases injury after cerebral ischemia in fetal sheep. Pediatr Res 1996; 40: 185–91.Google Scholar
Wintermark, P, Hansen, A, Warfield, SK, et al. Near-infrared spectroscopy versus magnetic resonance imaging to study brain perfusion in newborns with hypoxic-ischemic encephalopathy treated with hypothermia. Neuroimage 2014; 85: 287–93.Google Scholar
Shellhaas, RA, Thelen, BJ, Bapuraj, JR, et al. Limited short-term prognostic utility of cerebral NIRS during neonatal therapeutic hypothermia. Neurology 2013; 81: 249–55.Google Scholar
Ancora, G, Maranella, E, Locatelli, C, et al. Changes in cerebral hemodynamics and amplitude integrated EEG in an asphyxiated newborn during and after cool cap treatment. Brain Dev 2009; 31: 442–4.Google Scholar
Massaro, AN, Govindan, RB, Vezina, G, et al. Impaired cerebral autoregulation and brain injury in newborns with hypoxic-ischemic encephalopathy treated with hypothermia. J Neurophysiol 2015; 114: 818–24.Google Scholar
Burton, VJ, Gerner, G, Cristofalo, E, et al. A pilot cohort study of cerebral autoregulation and 2-year neurodevelopmental outcomes in neonates with hypoxic-ischemic encephalopathy who received therapeutic hypothermia. BMC Neurol 2015; 15: 209.Google Scholar
Wiberg-Itzel, E, Lipponer, C, Norman, M, et al. Determination of pH or lactate in fetal scalp blood in management of intrapartum fetal distress: randomized, controlled multicentre trial. BMJ 2008; 336: 1284–7.Google Scholar
Borruto, F, Comparetto, C, Treisser, A. Prevention of cerebral palsy during labour: role of foetal lactate. Arch Gynecol Obstet 2008; 278: 1722.Google Scholar
Ramanah, R, Martin, A, Clement, MC, et al. Fetal scalp lactate microsampling for non-reassuring fetal status during labor: a prospective observational study. Fetal Diagn Ther 2010; 27: 14–9.Google Scholar
Wiberg, N, Kallen, K, Herbst, A, Olofsson, P. Relation between umbilical cord blood pH, base deficit, lactate, 5-minute Apgar score and development of hypoxic ischemic encephalopathy. Acta Obstet Gynaecol Scand 2010; 89: 1263–9.Google Scholar
Hogan, L, Ingemarsson, I, Thorngren-Jerneck, K, Herbst, A. How often is a low 5-min Apgar score in term newborns due to asphyxia? Eur J Obstet Gynecol Reprod Biol 2007; 130: 169–75.Google Scholar
Thorngren-Jerneck, K, Herbst, A. Low 5-minute Apgar score: a population-based register study of 1 million term births. Obstet Gynecol 2001; 98: 6570.Google Scholar
Kovalak, EE, Dede, FS, Gelisen, O, et al. Nonreassuring fetal heart rate patterns and nucleated red blood cells in term neonates. Arch Gynecol Obstet 2011; 283: 1005–9.Google Scholar
Gazzolo, D, Marinoni, E, Di Iorio, R, et al. Urinary S100B protein measurements: a tool for the early identification of hypoxic-ischemic encephalopathy in asphyxiated full-term infants. Crit Care Med 2004; 32: 131–6.Google Scholar
Florio, P, Luisi, S, Moataza, B, et al. High urinary concentrations of activin A in asphyxiated full-term newborns with moderate or severe hypoxic ischemic encephalopathy. Clin Chem 2007; 53: 520–2.Google Scholar
Aly, H, Hassanein, S, Nada, A, et al. Vascular endothelial growth factor in neonates with perinatal asphyxia. Brain Dev 2009; 31: 600–4.Google Scholar
Reinke, SN, Walsh, BH, Boylan, GB, et al. 1H NMR derived metabolomic profile of neonatal asphyxia in umbilical cord serum: implications for hypoxic ischemic encephalopathy. J Proteome Res 2013; 12: 4230–9.Google Scholar
Thoresen, M. Patient selection and prognostication with hypothermia treatment. Semin Fetal Neonatal Med 2010; 15: 247–52.Google Scholar
Ambalavanan, N, Carlo, WA, Shankaran, S, et al. Predicting outcomes of neonates diagnosed with hypoxemic-ischemic encephalopathy. Pediatrics 2006; 118: 2084–93.Google Scholar
Haiju, Z, Suyuan, H, Xiufang, F, et al. The combined detection of umbilical cord nucleated red blood cells and lactate: early prediction of neonatal hypoxic ischemic encephalopathy. J Perinat Med 2008; 36: 240–7.Google Scholar
Bednarek, N, Svedin, P, Garnotel, R, et al. Increased MMP-9 and TIMP-1 in mouse neonatal brain and plasma and in human neonatal plasma after hypoxia-ischemia: a potential marker of neonatal encephalopathy. Pediatr Res 2012; 71: 6370.Google Scholar
Kirimi, E, Peker, E, Tuncer, O, et al. Increased serum malondialdehyde level in neonates with hypoxic-ischaemic encephalopathy: prediction of disease severity. J Int Med Res 2010; 38: 220–6.Google Scholar
van Laerhoven, H, de Haan, TR, Offringa, M, et al. Prognostic tests in term neonates with hypoxic-ischemic encephalopathy: a systematic review. Pediatrics 2013; 131: 8898.Google Scholar
Horn, AR, Swingler, GH, Myer, L, et al. Early clinical signs in neonates with hypoxic ischemic encephalopathy predict an abnormal amplitude-integrated electroencephalogram at age 6 hours. BMC Pediatr 2013; 13: 52.Google Scholar
Jose, A, Matthai, J, Paul, S. Correlation of EEG, CT, and MRI brain with neurological outcome at 12 months in term newborns with hypoxic ischemic encephalopathy. J Clin Neonatol 2013; 2: 125–30.Google Scholar
Ancora, G, Soffritti, S, Lodi, R, et al. A combined a-EEG and MR spectroscopy study in term newborns with hypoxic-ischemic encephalopathy. Brain Dev 2010; 32: 835–42.Google Scholar
Twomey, E, Twomey, A, Ryan, S, et al. MR imaging of term infants with hypoxic-ischaemic encephalopathy as a predictor of neurodevelopmental outcome and late MRI appearances. Pediatr Radiol 2010; 40: 1526–35.Google Scholar
Garcia-Alix, A, Cabanas, F, Pellicer, A, et al. Neuron-specific enolase and myelin basic protein: relationship of cerebrospinal fluid concentrations to the neurologic condition of asphyxiated full-term infants. Pediatrics 1994; 93: 234–40.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
×