Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2019

Open Access 01-12-2019 | Research

Experimental necrotizing enterocolitis induces neuroinflammation in the neonatal brain

Authors: George Biouss, Lina Antounians, Bo Li, Joshua S. O’Connell, Shogo Seo, Vincenzo D. Catania, Jennifer Guadagno, Abidur Rahman, Elke Zani-Ruttenstock, Nataliia Svergun, Agostino Pierro, Augusto Zani

Published in: Journal of Neuroinflammation | Issue 1/2019

Login to get access

Abstract

Background

Necrotizing enterocolitis (NEC) is an inflammatory gastrointestinal disease primarily affecting preterm neonates. Neonates with NEC suffer from a degree of neurodevelopmental delay that is not explained by prematurity alone. There is a need to understand the pathogenesis of neurodevelopmental delay in NEC. In this study, we assessed the macroscopic and microscopic changes that occur to brain cell populations in specific brain regions in a neonatal mouse model of NEC. Moreover, we investigated the role of intestinal inflammation as part of the mechanism responsible for the changes observed in the brain of pups with NEC.

Methods

Brains of mice were assessed for gross morphology and cerebral cortex thickness (using histology). Markers for mature neurons, oligodendrocytes, neural progenitor cells, microglia, and astrocytes were used to quantify their cell populations in different regions of the brain. Levels of cell apoptosis in the brain were measured by Western blotting and immunohistochemistry. Endoplasmic reticulum (ER) stress markers and levels of pro-inflammatory cytokines (in the ileum and brain) were measured by RT-qPCR and Western blotting. A Pearson test was used to correlate the levels of cytokines (ELISA) in the brain and ileum and to correlate activated microglia and astrocyte populations to the severity of NEC.

Results

NEC pups had smaller brain weights, higher brain-to-body weight ratios, and thinner cortices compared to control pups. NEC pups had increased levels of apoptosis and ER stress. In addition, NEC was associated with a reduction in the number of neurons, oligodendrocytes, and neural progenitors in specific regions of the brain. Levels of pro-inflammatory cytokines and the density of activated microglia and astrocytes were increased in the brain and positively correlated with the increase in the levels pro-inflammatory cytokines in the gut and the severity of NEC damage respectively.

Conclusions

NEC is associated with severe changes in brain morphology, a pro-inflammatory response in the brain that alters cell homeostasis and density of brain cell populations in specific cerebral regions. We show that the severity of neuroinflammation is associated with the severity of NEC. Our findings suggest that early intervention during NEC may reduce the chance of acute neuroinflammation and cerebral damage.
Appendix
Available only for authorised users
Literature
1.
go back to reference Zani A, Pierro A. Necrotizing enterocolitis: controversies and challenges. F1000Research. 2015;4 pii: F1000 Faculty Rev-1373. Zani A, Pierro A. Necrotizing enterocolitis: controversies and challenges. F1000Research. 2015;4 pii: F1000 Faculty Rev-1373.
2.
go back to reference Singh R, Shah B, Allred EN, Grzybowski M, Martin CR, Leviton A. The antecedents and correlates of necrotizing enterocolitis and spontaneous intestinal perforation among infants born before the 28th week of gestation. J Neonatal Perinatal Med. 2016;9:159–70.CrossRef Singh R, Shah B, Allred EN, Grzybowski M, Martin CR, Leviton A. The antecedents and correlates of necrotizing enterocolitis and spontaneous intestinal perforation among infants born before the 28th week of gestation. J Neonatal Perinatal Med. 2016;9:159–70.CrossRef
3.
go back to reference Hall NJ, Eaton S, Pierro A. Necrotizing enterocolitis: prevention, treatment, and outcome. J Pediatr Surg. 2013;48:2359–67.CrossRef Hall NJ, Eaton S, Pierro A. Necrotizing enterocolitis: prevention, treatment, and outcome. J Pediatr Surg. 2013;48:2359–67.CrossRef
4.
go back to reference Mutanen A, Pierro A, Zani A. Perioperative complications following surgery for necrotizing enterocolitis. Eur J Pediatr Surg. 2018;28:148–51.CrossRef Mutanen A, Pierro A, Zani A. Perioperative complications following surgery for necrotizing enterocolitis. Eur J Pediatr Surg. 2018;28:148–51.CrossRef
5.
go back to reference Rees CM, Pierro A, Eaton S. Neurodevelopmental outcomes of neonates with medically and surgically treated necrotizing enterocolitis. Arch Dis Child Fetal Neonatal Ed. 2007;92:F193–8.CrossRef Rees CM, Pierro A, Eaton S. Neurodevelopmental outcomes of neonates with medically and surgically treated necrotizing enterocolitis. Arch Dis Child Fetal Neonatal Ed. 2007;92:F193–8.CrossRef
6.
go back to reference Hintz SR, Kendrick DE, Stoll BJ, Vohr BR, Fanaroff AA, Donovan EF, et al. Neurodevelopmental and growth outcomes of extremely low birth weight infants after necrotizing enterocolitis. Pediatrics. 2005;115:696–703.CrossRef Hintz SR, Kendrick DE, Stoll BJ, Vohr BR, Fanaroff AA, Donovan EF, et al. Neurodevelopmental and growth outcomes of extremely low birth weight infants after necrotizing enterocolitis. Pediatrics. 2005;115:696–703.CrossRef
7.
go back to reference Regev RH, Arnon S, Litmanovitz I, Bauer-Rusek S, Boyko V, Lerner-Geva L, et al. Association between neonatal morbidities and head growth from birth until discharge in very-low-birthweight infants born preterm: a population-based study. Dev Med Child Neurol. 2016;58:1159–66.CrossRef Regev RH, Arnon S, Litmanovitz I, Bauer-Rusek S, Boyko V, Lerner-Geva L, et al. Association between neonatal morbidities and head growth from birth until discharge in very-low-birthweight infants born preterm: a population-based study. Dev Med Child Neurol. 2016;58:1159–66.CrossRef
8.
go back to reference Shah DK, Doyle LW, Anderson PJ, Bear M, Daley AJ, Hunt RW, et al. Adverse neurodevelopment in preterm infants with postnatal sepsis or necrotizing enterocolitis is mediated by white matter abnormalities on magnetic resonance imaging at term. J Pediatr. 2008;153:170–5.CrossRef Shah DK, Doyle LW, Anderson PJ, Bear M, Daley AJ, Hunt RW, et al. Adverse neurodevelopment in preterm infants with postnatal sepsis or necrotizing enterocolitis is mediated by white matter abnormalities on magnetic resonance imaging at term. J Pediatr. 2008;153:170–5.CrossRef
9.
go back to reference Merhar SL, Ramos Y, Meinzen-Derr J, Kline-Fath BM. Brain magnetic resonance imaging in infants with surgical necrotizing enterocolitis or spontaneous intestinal perforation versus medical necrotizing enterocolitis. J Pediatr. 2014;164:410–2.CrossRef Merhar SL, Ramos Y, Meinzen-Derr J, Kline-Fath BM. Brain magnetic resonance imaging in infants with surgical necrotizing enterocolitis or spontaneous intestinal perforation versus medical necrotizing enterocolitis. J Pediatr. 2014;164:410–2.CrossRef
10.
go back to reference Lee I, Neil JJ, Huettner PC, Smyser CD, Rogers CE, Shimony JS, et al. The impact of prenatal and neonatal infection on neurodevelopmental outcomes in very preterm infants. J Perinatol. 2014;34:741–7.CrossRef Lee I, Neil JJ, Huettner PC, Smyser CD, Rogers CE, Shimony JS, et al. The impact of prenatal and neonatal infection on neurodevelopmental outcomes in very preterm infants. J Perinatol. 2014;34:741–7.CrossRef
11.
go back to reference Shin SH, Kim EK, Yoo H, Choi YH, Kim S, Lee BK, et al. Surgical necrotizing enterocolitis versus spontaneous intestinal perforation in white matter injury on brain magnetic resonance imaging. Neonatology. 2016;110:148–54.CrossRef Shin SH, Kim EK, Yoo H, Choi YH, Kim S, Lee BK, et al. Surgical necrotizing enterocolitis versus spontaneous intestinal perforation in white matter injury on brain magnetic resonance imaging. Neonatology. 2016;110:148–54.CrossRef
12.
go back to reference Sun J, Pan X, Christiansen LI, Yuan XL, Skovgaard K, Chatterton DEW, et al. Necrotizing enterocolitis is associated with acute brain responses in preterm pigs. J Neuroinflammation. 2018;15:180.CrossRef Sun J, Pan X, Christiansen LI, Yuan XL, Skovgaard K, Chatterton DEW, et al. Necrotizing enterocolitis is associated with acute brain responses in preterm pigs. J Neuroinflammation. 2018;15:180.CrossRef
13.
go back to reference Niño DF, Zhou Q, Yamaguchi Y, Martin LY, Wang S, Fulton WB, et al. Cognitive impairments induced by necrotizing enterocolitis can be prevented by inhibiting microglial activation in mouse brain. Sci Transl Med. 2018;10:237.CrossRef Niño DF, Zhou Q, Yamaguchi Y, Martin LY, Wang S, Fulton WB, et al. Cognitive impairments induced by necrotizing enterocolitis can be prevented by inhibiting microglial activation in mouse brain. Sci Transl Med. 2018;10:237.CrossRef
14.
go back to reference Zani A, Cordischi L, Cananzi M, De Coppi P, Smith VV, Eaton S, et al. Assessment of a neonatal rat model of necrotizing enterocolitis. Eur J Pediatr Surg. 2008;18:423–6.CrossRef Zani A, Cordischi L, Cananzi M, De Coppi P, Smith VV, Eaton S, et al. Assessment of a neonatal rat model of necrotizing enterocolitis. Eur J Pediatr Surg. 2008;18:423–6.CrossRef
15.
go back to reference Zani A, Zani-Ruttenstock E, Peyvandi F, Lee C, Li B, Pierro A. A spectrum of intestinal injury models in neonatal mice. Pediatr Surg Int. 2016;32:65–70.CrossRef Zani A, Zani-Ruttenstock E, Peyvandi F, Lee C, Li B, Pierro A. A spectrum of intestinal injury models in neonatal mice. Pediatr Surg Int. 2016;32:65–70.CrossRef
16.
go back to reference Konsman J-P. The mouse brain in stereotaxic coordinates. 2nd ed. New York: Academic Press; Psychoneuroendocrinology. 2003. Konsman J-P. The mouse brain in stereotaxic coordinates. 2nd ed. New York: Academic Press; Psychoneuroendocrinology. 2003.
17.
go back to reference Lucas MM, Lenck-Santini PP, Holmes GL, Scott RC. Impaired cognition in rats with cortical dysplasia: additional impact of early-life seizures. Brain. 2011;134:1684–93.CrossRef Lucas MM, Lenck-Santini PP, Holmes GL, Scott RC. Impaired cognition in rats with cortical dysplasia: additional impact of early-life seizures. Brain. 2011;134:1684–93.CrossRef
18.
go back to reference Jensen EC. Quantitative analysis of histological staining and fluorescence using ImageJ. Anat Rec. 2013;296:378–81.CrossRef Jensen EC. Quantitative analysis of histological staining and fluorescence using ImageJ. Anat Rec. 2013;296:378–81.CrossRef
19.
go back to reference Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci. 1996;19:312–8.CrossRef Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci. 1996;19:312–8.CrossRef
20.
go back to reference Hovens IB, Nyakas C, Schoemaker RG. A novel method for evaluating microglial activation using ionized calcium-binding adaptor protein-1 staining: cell body to cell size ratio. Neuroimmunol Neuroinflammation. 2014;1:82–8.CrossRef Hovens IB, Nyakas C, Schoemaker RG. A novel method for evaluating microglial activation using ionized calcium-binding adaptor protein-1 staining: cell body to cell size ratio. Neuroimmunol Neuroinflammation. 2014;1:82–8.CrossRef
21.
go back to reference Hernandez-Rabaza V, Agusti A, Cabrera-Pastor A, Fustero S, Delgado O, Taoro-Gonzalez L, et al. Sildenafil reduces neuroinflammation and restores spatial learning in rats with hepatic encephalopathy: underlying mechanisms. J Neuroinflammation. 2015;12:195.CrossRef Hernandez-Rabaza V, Agusti A, Cabrera-Pastor A, Fustero S, Delgado O, Taoro-Gonzalez L, et al. Sildenafil reduces neuroinflammation and restores spatial learning in rats with hepatic encephalopathy: underlying mechanisms. J Neuroinflammation. 2015;12:195.CrossRef
22.
go back to reference Fernández-Arjona MDM, Grondona JM, Fernández-Llebrez P, Granados-Durán P, López-Ávalos MD. Microglia morphological categorization in a rat model of neuroinflammation by hierarchical cluster and principal components analysis. Front Cell Neurosci. 2017;11:235.CrossRef Fernández-Arjona MDM, Grondona JM, Fernández-Llebrez P, Granados-Durán P, López-Ávalos MD. Microglia morphological categorization in a rat model of neuroinflammation by hierarchical cluster and principal components analysis. Front Cell Neurosci. 2017;11:235.CrossRef
23.
go back to reference Yang G, Bao P, Zhang L, Lyu Z, Zhou B, Chen K, et al. Critical role of myeloid differentiation factor 88 in necrotizing enterocolitis. Pediatr Res. 2014;75:707–15.CrossRef Yang G, Bao P, Zhang L, Lyu Z, Zhou B, Chen K, et al. Critical role of myeloid differentiation factor 88 in necrotizing enterocolitis. Pediatr Res. 2014;75:707–15.CrossRef
24.
go back to reference Cho SX, Berger PJ, Nold-Petry CA, Nold MF. The immunological landscape in necrotising enterocolitis. Expert Rev Mol Med. 2016;18:e12.CrossRef Cho SX, Berger PJ, Nold-Petry CA, Nold MF. The immunological landscape in necrotising enterocolitis. Expert Rev Mol Med. 2016;18:e12.CrossRef
25.
go back to reference Xu C, Bailly-Maitre B, Reed JC. Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest. 2005;115:2656–64.CrossRef Xu C, Bailly-Maitre B, Reed JC. Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest. 2005;115:2656–64.CrossRef
26.
go back to reference Parikh NA, Lasky RE, Kennedy KA, McDavid G, Tyson JE. Perinatal factors and regional brain volume abnormalities at term in a cohort of extremely low birth weight infants. PLoS One. 2013;8:e68204.CrossRef Parikh NA, Lasky RE, Kennedy KA, McDavid G, Tyson JE. Perinatal factors and regional brain volume abnormalities at term in a cohort of extremely low birth weight infants. PLoS One. 2013;8:e68204.CrossRef
27.
go back to reference Bonifacio SL, Glass HC, Chau V, Berman JI, Xu D, Brant R, et al. Extreme premature birth is not associated with impaired development of brain microstructure. J Pediatr. 2010;157:726–32.CrossRef Bonifacio SL, Glass HC, Chau V, Berman JI, Xu D, Brant R, et al. Extreme premature birth is not associated with impaired development of brain microstructure. J Pediatr. 2010;157:726–32.CrossRef
28.
go back to reference Shim SY, Jeong HJ, Son DW, Jeong JS, Oh SH, Park SY, et al. Altered microstructure of white matter except the corpus callosum is independent of prematurity. Neonatology. 2012;102:309–15.CrossRef Shim SY, Jeong HJ, Son DW, Jeong JS, Oh SH, Park SY, et al. Altered microstructure of white matter except the corpus callosum is independent of prematurity. Neonatology. 2012;102:309–15.CrossRef
29.
go back to reference Cao C, Yu X, Liao Z, Zhu N, Huo H, Wang M, et al. Hypertonic saline reduces lipopolysaccharide-induced mouse brain edema through inhibiting aquaporin 4 expression. Crit Care. 2012;16:R186.CrossRef Cao C, Yu X, Liao Z, Zhu N, Huo H, Wang M, et al. Hypertonic saline reduces lipopolysaccharide-induced mouse brain edema through inhibiting aquaporin 4 expression. Crit Care. 2012;16:R186.CrossRef
30.
go back to reference Cardoso FL, Herz J, Fernandes A, Rocha J, Sepodes B, Brito MA, et al. Systemic inflammation in early neonatal mice induces transient and lasting neurodegenerative effects. J Neuroinflammation. 2015;12:82.CrossRef Cardoso FL, Herz J, Fernandes A, Rocha J, Sepodes B, Brito MA, et al. Systemic inflammation in early neonatal mice induces transient and lasting neurodegenerative effects. J Neuroinflammation. 2015;12:82.CrossRef
31.
go back to reference Brunse A, Abbaspour A, Sangild PT. Brain barrier disruption and region-specific neuronal degeneration during necrotizing enterocolitis in preterm pigs. Dev Neurosci. 2018;40:198–208.CrossRef Brunse A, Abbaspour A, Sangild PT. Brain barrier disruption and region-specific neuronal degeneration during necrotizing enterocolitis in preterm pigs. Dev Neurosci. 2018;40:198–208.CrossRef
32.
go back to reference Dickerson BC, Fenstermacher E, Salat DH, Wolk DA, Maguire RP, Desikan R, et al. Detection of cortical thickness correlates of cognitive performance: reliability across MRI scan sessions, scanners, and field strengths. Neuroimage. 2008;39:10–8.CrossRef Dickerson BC, Fenstermacher E, Salat DH, Wolk DA, Maguire RP, Desikan R, et al. Detection of cortical thickness correlates of cognitive performance: reliability across MRI scan sessions, scanners, and field strengths. Neuroimage. 2008;39:10–8.CrossRef
33.
go back to reference Shaw P, Kabani NJ, Lerch JP, Eckstrand K, Lenroot R, Gogtay N, et al. Neurodevelopmental trajectories of the human cerebral cortex. J Neurosci. 2008;28:3586–94.CrossRef Shaw P, Kabani NJ, Lerch JP, Eckstrand K, Lenroot R, Gogtay N, et al. Neurodevelopmental trajectories of the human cerebral cortex. J Neurosci. 2008;28:3586–94.CrossRef
34.
go back to reference Peterson BS, Vohr B, Staib LH, Cannistraci CJ, Dolberg A, Schneider KC, et al. Regional brain volume abnormalities and long-term cognitive outcome in preterm infants. J Am Med Assoc. 2000;284:1939–47.CrossRef Peterson BS, Vohr B, Staib LH, Cannistraci CJ, Dolberg A, Schneider KC, et al. Regional brain volume abnormalities and long-term cognitive outcome in preterm infants. J Am Med Assoc. 2000;284:1939–47.CrossRef
35.
go back to reference Soria-Pastor S, Padilla N, Zubiaurre-Elorza L, Ibarretxe-Bilbao N, Botet F, Costas-Moragas C, et al. Decreased regional brain volume and cognitive impairment in preterm children at low risk. Pediatrics. 2009;124:e1161–70.CrossRef Soria-Pastor S, Padilla N, Zubiaurre-Elorza L, Ibarretxe-Bilbao N, Botet F, Costas-Moragas C, et al. Decreased regional brain volume and cognitive impairment in preterm children at low risk. Pediatrics. 2009;124:e1161–70.CrossRef
36.
go back to reference Li B, Zani A, Lee C, Zani-Ruttenstock E, Zhang Z, Li X, et al. Endoplasmic reticulum stress is involved in the colonic epithelium damage induced by maternal separation. J Pediatr Surg. 2016;51:1001–4.CrossRef Li B, Zani A, Lee C, Zani-Ruttenstock E, Zhang Z, Li X, et al. Endoplasmic reticulum stress is involved in the colonic epithelium damage induced by maternal separation. J Pediatr Surg. 2016;51:1001–4.CrossRef
37.
go back to reference Wang YW, Zhou Q, Zhang X, Qian QQ, Xu JW, Ni PF, et al. Mild endoplasmic reticulum stress ameliorates lipopolysaccharide-induced neuroinflammation and cognitive impairment via regulation of microglial polarization. J Neuroinflammation. 2017;14:233.CrossRef Wang YW, Zhou Q, Zhang X, Qian QQ, Xu JW, Ni PF, et al. Mild endoplasmic reticulum stress ameliorates lipopolysaccharide-induced neuroinflammation and cognitive impairment via regulation of microglial polarization. J Neuroinflammation. 2017;14:233.CrossRef
38.
go back to reference Sprenkle NT, Sims SG, Sánchez CL, Meares GP. Endoplasmic reticulum stress and inflammation in the central nervous system. Mol Neurodegener. 2017;12:42–60.CrossRef Sprenkle NT, Sims SG, Sánchez CL, Meares GP. Endoplasmic reticulum stress and inflammation in the central nervous system. Mol Neurodegener. 2017;12:42–60.CrossRef
39.
go back to reference Thornton C, Baburamani AA, Kichev A, Hagberg H. Oxidative stress and endoplasmic reticulum (ER) stress in the development of neonatal hypoxic-ischaemic brain injury. Biochem Soc Trans. 2017;45:1067–76.CrossRef Thornton C, Baburamani AA, Kichev A, Hagberg H. Oxidative stress and endoplasmic reticulum (ER) stress in the development of neonatal hypoxic-ischaemic brain injury. Biochem Soc Trans. 2017;45:1067–76.CrossRef
40.
go back to reference Herrero MT, Barcia C, Navarro JM. Functional anatomy of thalamus and basal ganglia. Childs Nerv Syst. 2002;18:386–404.CrossRef Herrero MT, Barcia C, Navarro JM. Functional anatomy of thalamus and basal ganglia. Childs Nerv Syst. 2002;18:386–404.CrossRef
41.
go back to reference Gilmore JH, Shi F, Woolson SL, Knickmeyer RC, Short SJ, Lin W, et al. Longitudinal development of cortical and subcortical gray matter from birth to 2 years. Cereb Cortex. 2012;22:2478–85.CrossRef Gilmore JH, Shi F, Woolson SL, Knickmeyer RC, Short SJ, Lin W, et al. Longitudinal development of cortical and subcortical gray matter from birth to 2 years. Cereb Cortex. 2012;22:2478–85.CrossRef
42.
go back to reference Eichenbaum H. Prefrontal-hippocampal interactions in episodic memory. Nat Rev Neurosci. 2017;18:547–58.CrossRef Eichenbaum H. Prefrontal-hippocampal interactions in episodic memory. Nat Rev Neurosci. 2017;18:547–58.CrossRef
43.
go back to reference Hagberg H, Mallard C, Ferriero DM, Vannucci SJ, Levison SW, Vexler ZS, et al. The role of inflammation in perinatal brain injury. Nat Rev Neurol. 2015;11:192–208.CrossRef Hagberg H, Mallard C, Ferriero DM, Vannucci SJ, Levison SW, Vexler ZS, et al. The role of inflammation in perinatal brain injury. Nat Rev Neurol. 2015;11:192–208.CrossRef
44.
go back to reference Qing L, Black SM. Neonatal hypoxic-ischemic brain injury: apoptotic and non-apoptotic cell death. J Neurol Neuromedicine. 2016;1:5–10.CrossRef Qing L, Black SM. Neonatal hypoxic-ischemic brain injury: apoptotic and non-apoptotic cell death. J Neurol Neuromedicine. 2016;1:5–10.CrossRef
45.
go back to reference Barkovich AJ. Concepts of myelin and myelination in neuroradiology. AJNR Am J Neuroradiol. 2000;21:1099–109.PubMed Barkovich AJ. Concepts of myelin and myelination in neuroradiology. AJNR Am J Neuroradiol. 2000;21:1099–109.PubMed
46.
go back to reference Jiang ZD, Wang C, Chen C. Neonatal necrotizing enterocolitis adversely affects neural conduction of the rostral brainstem in preterm babies. Clin Neurophysiol. 2014;125:2277–85.CrossRef Jiang ZD, Wang C, Chen C. Neonatal necrotizing enterocolitis adversely affects neural conduction of the rostral brainstem in preterm babies. Clin Neurophysiol. 2014;125:2277–85.CrossRef
47.
go back to reference Jurewicz A, Matysiak M, Tybor K, Kilianek L, Raine CS, Selmaj K. Tumour necrosis factor-induced death of adult human oligodendrocytes is mediated by apoptosis inducing factor. Brain. 2005;128:2675–88.CrossRef Jurewicz A, Matysiak M, Tybor K, Kilianek L, Raine CS, Selmaj K. Tumour necrosis factor-induced death of adult human oligodendrocytes is mediated by apoptosis inducing factor. Brain. 2005;128:2675–88.CrossRef
48.
go back to reference Zajicek JP, Wing M, Scolding NJ, Compston DA. Interactions between oligodendrocytes and microglia: a major role for complement and tumour necrosis factor in oligodendrocyte adherence and killing. Brain. 1992;115:1611–31.CrossRef Zajicek JP, Wing M, Scolding NJ, Compston DA. Interactions between oligodendrocytes and microglia: a major role for complement and tumour necrosis factor in oligodendrocyte adherence and killing. Brain. 1992;115:1611–31.CrossRef
49.
go back to reference Bradl M, Lassmann H. Oligodendrocytes: biology and pathology. Acta Neuropathol. 2010;119:37–53.CrossRef Bradl M, Lassmann H. Oligodendrocytes: biology and pathology. Acta Neuropathol. 2010;119:37–53.CrossRef
50.
go back to reference Graham V, Khudyakov J, Ellis P, Pevny L. SOX2 functions to maintain neural progenitor identity. Neuron. 2003;39:749–65.CrossRef Graham V, Khudyakov J, Ellis P, Pevny L. SOX2 functions to maintain neural progenitor identity. Neuron. 2003;39:749–65.CrossRef
51.
go back to reference Ellis P, Fagan BM, Magness ST, Hutton S, Taranova O, Hayashi S, et al. SOX2, a persistent marker for multipotential neural stem cells derived from embryonic stem cells, the embryo or the adult. Dev Neurosci. 2004;26:148–65.CrossRef Ellis P, Fagan BM, Magness ST, Hutton S, Taranova O, Hayashi S, et al. SOX2, a persistent marker for multipotential neural stem cells derived from embryonic stem cells, the embryo or the adult. Dev Neurosci. 2004;26:148–65.CrossRef
52.
go back to reference Bland ST, Beckley JT, Young S, Tsang V, Watkins LR, Maier SF, et al. Enduring consequences of early-life infection on glial and neural cell genesis within cognitive regions of the brain. Brain Behav Immun. 2010;24:329–38.CrossRef Bland ST, Beckley JT, Young S, Tsang V, Watkins LR, Maier SF, et al. Enduring consequences of early-life infection on glial and neural cell genesis within cognitive regions of the brain. Brain Behav Immun. 2010;24:329–38.CrossRef
53.
go back to reference Smith PLP, Hagberg H, Naylor AS, Mallard C. Neonatal peripheral immune challenge activates microglia and inhibits neurogenesis in the developing murine hippocampus. Dev Neurosci. 2014;36:119–31.CrossRef Smith PLP, Hagberg H, Naylor AS, Mallard C. Neonatal peripheral immune challenge activates microglia and inhibits neurogenesis in the developing murine hippocampus. Dev Neurosci. 2014;36:119–31.CrossRef
54.
go back to reference Bilbo SD, Levkoff LH, Mahoney JH, Watkins LR, Rudy JW, Maier SF. Neonatal infection induces memory impairments following an immune challenge in adulthood. Behav Neurosci. 2005;119:293–301.CrossRef Bilbo SD, Levkoff LH, Mahoney JH, Watkins LR, Rudy JW, Maier SF. Neonatal infection induces memory impairments following an immune challenge in adulthood. Behav Neurosci. 2005;119:293–301.CrossRef
55.
go back to reference Johnson S, Wolke D, Hennessy E, Marlow N. Educational outcomes in extremely preterm children: neuropsychological correlates and predictors of attainment. Dev Neuropsychol. 2011;36:74–95.CrossRef Johnson S, Wolke D, Hennessy E, Marlow N. Educational outcomes in extremely preterm children: neuropsychological correlates and predictors of attainment. Dev Neuropsychol. 2011;36:74–95.CrossRef
56.
go back to reference van der Ree M, Tanis JC, Van Braeckel KNJA, Bos AF, Roze E. Functional impairments at school age of preterm born children with late-onset sepsis. Early Hum Dev. 2011;87:821–6.CrossRef van der Ree M, Tanis JC, Van Braeckel KNJA, Bos AF, Roze E. Functional impairments at school age of preterm born children with late-onset sepsis. Early Hum Dev. 2011;87:821–6.CrossRef
57.
go back to reference Pike K, Brocklehurst P, Jones D, Kenyon S, Salt A, Taylor D, et al. Outcomes at 7 years for babies who developed neonatal necrotising enterocolitis: the ORACLE Children Study. Arch Dis Child Fetal Neonatal Ed. 2012;97:F318–22.CrossRef Pike K, Brocklehurst P, Jones D, Kenyon S, Salt A, Taylor D, et al. Outcomes at 7 years for babies who developed neonatal necrotising enterocolitis: the ORACLE Children Study. Arch Dis Child Fetal Neonatal Ed. 2012;97:F318–22.CrossRef
58.
go back to reference Arcuri C, Mecca C, Bianchi R, Giambanco I, Donato R. The pathophysiological role of microglia in dynamic surveillance, phagocytosis and structural remodeling of the developing CNS. Front Mol Neurosci. 2017;10:191.CrossRef Arcuri C, Mecca C, Bianchi R, Giambanco I, Donato R. The pathophysiological role of microglia in dynamic surveillance, phagocytosis and structural remodeling of the developing CNS. Front Mol Neurosci. 2017;10:191.CrossRef
59.
go back to reference Tahraoui S, Marret S, Bodenant C, Leroux P, Dommergues M, Evrard P, et al. Central role of microglia in neonatal excitotoxic lesions of the murine periventricular white matter. Brain Pathol. 2001;11:56–71.CrossRef Tahraoui S, Marret S, Bodenant C, Leroux P, Dommergues M, Evrard P, et al. Central role of microglia in neonatal excitotoxic lesions of the murine periventricular white matter. Brain Pathol. 2001;11:56–71.CrossRef
60.
go back to reference Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541:481–7.CrossRef Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541:481–7.CrossRef
61.
go back to reference Bi F, Huang C, Tong J, Qiu G, Huang B, Wu Q, et al. Reactive astrocytes secrete lcn2 to promote neuron death. Proc Natl Acad Sci. 2013;110:4069–74.CrossRef Bi F, Huang C, Tong J, Qiu G, Huang B, Wu Q, et al. Reactive astrocytes secrete lcn2 to promote neuron death. Proc Natl Acad Sci. 2013;110:4069–74.CrossRef
62.
go back to reference Selip DB, Jantzie LL, Chang M, Jackson MC, Fitzgerald EC, Boll G, et al. Regional differences in susceptibility to hypoxic-ischemic injury in the preterm brain: exploring the spectrum from white matter loss to selective grey matter injury in a rat model. Neurol Res Int. 2012;2012:1–11.CrossRef Selip DB, Jantzie LL, Chang M, Jackson MC, Fitzgerald EC, Boll G, et al. Regional differences in susceptibility to hypoxic-ischemic injury in the preterm brain: exploring the spectrum from white matter loss to selective grey matter injury in a rat model. Neurol Res Int. 2012;2012:1–11.CrossRef
63.
go back to reference Lechpammer M, Manning SM, Samonte F, Nelligan J, Sabot E, Talos DM, et al. Minocycline treatment following hypoxic/ischaemic injury attenuates white matter injury in a rodent model of periventricular leucomalacia. Neuropathol Appl Neurobiol. 2008;34:379–93.CrossRef Lechpammer M, Manning SM, Samonte F, Nelligan J, Sabot E, Talos DM, et al. Minocycline treatment following hypoxic/ischaemic injury attenuates white matter injury in a rodent model of periventricular leucomalacia. Neuropathol Appl Neurobiol. 2008;34:379–93.CrossRef
64.
go back to reference Billiards SS, Haynes RL, Folkerth RD, Trachtenberg FL, Liu LG, Volpe JJ, et al. Development of microglia in the cerebral white matter of the human fetus and infant. J Comp Neurol. 2006;497:199–208.CrossRef Billiards SS, Haynes RL, Folkerth RD, Trachtenberg FL, Liu LG, Volpe JJ, et al. Development of microglia in the cerebral white matter of the human fetus and infant. J Comp Neurol. 2006;497:199–208.CrossRef
65.
go back to reference Viscardi RM, Lyon NH, Sun CCJ, Richard Hebel J, Hasday JD. Inflammatory cytokine mRNAs in surgical specimens of necrotizing enterocolitis and normal newborn intestine. Pediatr Pathol Lab Med. 1997;17:547–59.CrossRef Viscardi RM, Lyon NH, Sun CCJ, Richard Hebel J, Hasday JD. Inflammatory cytokine mRNAs in surgical specimens of necrotizing enterocolitis and normal newborn intestine. Pediatr Pathol Lab Med. 1997;17:547–59.CrossRef
66.
go back to reference Wadhawan R, Oh W, Hintz SR, Blakely ML, Das A, Bell EF, et al. Neurodevelopmental outcomes of extremely low birth weight infants with spontaneous intestinal perforation or surgical necrotizing enterocolitis. J Perinatol. 2014;34:64–70.CrossRef Wadhawan R, Oh W, Hintz SR, Blakely ML, Das A, Bell EF, et al. Neurodevelopmental outcomes of extremely low birth weight infants with spontaneous intestinal perforation or surgical necrotizing enterocolitis. J Perinatol. 2014;34:64–70.CrossRef
67.
go back to reference Daulatzai MA. Chronic functional bowel syndrome enhances gut-brain axis dysfunction, neuroinflammation, cognitive impairment, and vulnerability to dementia. Neurochem Res. 2014;39:624–44.CrossRef Daulatzai MA. Chronic functional bowel syndrome enhances gut-brain axis dysfunction, neuroinflammation, cognitive impairment, and vulnerability to dementia. Neurochem Res. 2014;39:624–44.CrossRef
68.
go back to reference Sherman MP, Zaghouani H, Niklas V. Gut microbiota, the immune system, and diet influence the neonatal gut-brain axis. Pediatr Res. 2015;77:127–35.CrossRef Sherman MP, Zaghouani H, Niklas V. Gut microbiota, the immune system, and diet influence the neonatal gut-brain axis. Pediatr Res. 2015;77:127–35.CrossRef
69.
go back to reference Sochocka M, Diniz BS, Leszek J. Inflammatory response in the CNS: friend or foe? Mol Neurobiol. 2017;54:8071–89.CrossRef Sochocka M, Diniz BS, Leszek J. Inflammatory response in the CNS: friend or foe? Mol Neurobiol. 2017;54:8071–89.CrossRef
71.
go back to reference Erny D, Hrabě de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015;18:965–77.CrossRef Erny D, Hrabě de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015;18:965–77.CrossRef
72.
go back to reference Stilling RM, Dinan TG, Cryan JF. Microbial genes, brain & behaviour - epigenetic regulation of the gut-brain axis. Genes Brain Behav. 2014;13:69–86.CrossRef Stilling RM, Dinan TG, Cryan JF. Microbial genes, brain & behaviour - epigenetic regulation of the gut-brain axis. Genes Brain Behav. 2014;13:69–86.CrossRef
73.
go back to reference Moschopoulos C, Kratimenos P, Koutroulis I, Shah BV, Mowes A, Bhandari V. The neurodevelopmental perspective of surgical necrotizing enterocolitis: the role of the gut-brain axis. Mediat Inflamm. 2018;2018:1–8.CrossRef Moschopoulos C, Kratimenos P, Koutroulis I, Shah BV, Mowes A, Bhandari V. The neurodevelopmental perspective of surgical necrotizing enterocolitis: the role of the gut-brain axis. Mediat Inflamm. 2018;2018:1–8.CrossRef
74.
go back to reference Stefanutti G, Pierro A, Parkinson EJ, Smith VV, Eaton S. Moderate hypothermia as a rescue therapy against intestinal ischemia and reperfusion injury in the rat. Crit Care Med. 2008;36:1564–72.CrossRef Stefanutti G, Pierro A, Parkinson EJ, Smith VV, Eaton S. Moderate hypothermia as a rescue therapy against intestinal ischemia and reperfusion injury in the rat. Crit Care Med. 2008;36:1564–72.CrossRef
75.
go back to reference Hall NJ, Eaton S, Peters MJ, Hiorns MP, Alexander N, Azzopardi DV, et al. Mild controlled hypothermia in preterm neonates with advanced necrotizing enterocolitis. Pediatrics. 2010;125:e300–8.CrossRef Hall NJ, Eaton S, Peters MJ, Hiorns MP, Alexander N, Azzopardi DV, et al. Mild controlled hypothermia in preterm neonates with advanced necrotizing enterocolitis. Pediatrics. 2010;125:e300–8.CrossRef
76.
go back to reference Jenkins DD, Lee T, Chiuzan C, Perkel JK, Rollins LG, Wagner CL, et al. Altered circulating leukocytes and their chemokines in a clinical trial of therapeutic hypothermia for neonatal hypoxic ischemic encephalopathy. Pediatr Crit Care Med. 2013;14:786–95.CrossRef Jenkins DD, Lee T, Chiuzan C, Perkel JK, Rollins LG, Wagner CL, et al. Altered circulating leukocytes and their chemokines in a clinical trial of therapeutic hypothermia for neonatal hypoxic ischemic encephalopathy. Pediatr Crit Care Med. 2013;14:786–95.CrossRef
77.
go back to reference Jantzie LL, Todd KG. Doxycycline inhibits proinflammatory cytokines but not acute cerebral cytogenesis after hypoxia-ischemia in neonatal rats. J Psychiatry Neurosci. 2010;35:20–32.CrossRef Jantzie LL, Todd KG. Doxycycline inhibits proinflammatory cytokines but not acute cerebral cytogenesis after hypoxia-ischemia in neonatal rats. J Psychiatry Neurosci. 2010;35:20–32.CrossRef
78.
go back to reference Kannan S, Dai H, Navath RS, Balakrishnan B, Jyoti A, Janisse J, et al. Dendrimer-based postnatal therapy for neuroinflammation and cerebral palsy in a rabbit model. Sci Transl Med. 2012;4:130ra146.CrossRef Kannan S, Dai H, Navath RS, Balakrishnan B, Jyoti A, Janisse J, et al. Dendrimer-based postnatal therapy for neuroinflammation and cerebral palsy in a rabbit model. Sci Transl Med. 2012;4:130ra146.CrossRef
79.
go back to reference Nance E, Porambo M, Zhang F, Mishra MK, Buelow M, Getzenberg R, et al. Systemic dendrimer-drug treatment of ischemia-induced neonatal white matter injury. J Control Release. 2015;214:112–20.CrossRef Nance E, Porambo M, Zhang F, Mishra MK, Buelow M, Getzenberg R, et al. Systemic dendrimer-drug treatment of ischemia-induced neonatal white matter injury. J Control Release. 2015;214:112–20.CrossRef
Metadata
Title
Experimental necrotizing enterocolitis induces neuroinflammation in the neonatal brain
Authors
George Biouss
Lina Antounians
Bo Li
Joshua S. O’Connell
Shogo Seo
Vincenzo D. Catania
Jennifer Guadagno
Abidur Rahman
Elke Zani-Ruttenstock
Nataliia Svergun
Agostino Pierro
Augusto Zani
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2019
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-019-1481-9

Other articles of this Issue 1/2019

Journal of Neuroinflammation 1/2019 Go to the issue