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

Open Access 01-12-2019 | Biomarkers | Research

Potential biomarkers for neuroinflammation and neurodegeneration at short and long term after neonatal hypoxic-ischemic insult in rat

Authors: Nozha Borjini, Sandra Sivilia, Alessandro Giuliani, Mercedes Fernandez, Luciana Giardino, Fabrizio Facchinetti, Laura Calzà

Published in: Journal of Neuroinflammation | Issue 1/2019

Login to get access

Abstract

Background

Hypoxic-ischemic (HI) encephalopathy causes life-long morbidity and premature mortality in term neonates. Therapies in addition to whole-body cooling are under development to treat the neonate at risk for HI encephalopathy, but are not a quickly measured serum inflammatory or neuronal biomarkers to rapidly and accurately identify brain injury in order to follow the efficacy of therapies.

Methods

In order to identify potential biomarkers for early inflammatory and neurodegenerative events after neonatal hypoxia-ischemia, both male and female Wistar rat pups at postnatal day 7 (P7) were used and had their right carotid artery permanently doubly occluded and exposed to 8% oxygen for 90 min. Sensory and cognitive parameters were assessed by open field, rotarod, CatWalk, and Morris water maze (MWM) test. Plasma and CSF biomarkers were investigated on the acute (24 h and 72 h) and chronic phase (4 weeks). Brains were assessed for gene expression analysis by quantitative RT-PCR Array.

Results

We found a delay of neurological reflex maturation in HI rats. We observed anxiolytic-like baseline behavior in males more than females following HI injury. HI rats held on the rotarod for a shorter time comparing to sham. HI injury impaired spatial learning ability on MWM test. The CatWalk assessment demonstrated a long-term deficit in gait parameters related to the hind paw. Proinflammatory biomarkers such as IL-6 in plasma and CCL2 and TNF-α in CSF showed an upregulation at 24 h after HI while other cytokines, such as IL-17A and CCL5, were upregulated after 72 h in CSF. At 24 h post-injury, we observed an increase of Edn1, Hif1-α, and Mmp9 mRNA levels in the ipsilateral vs the contralateral hemisphere of HI rats. An upregulation of genes involved with clotting and hematopoietic processes was observed 72 h post-injury.

Conclusions

Our work showed that, in the immature brain, the HI injury induced an early increased production of several proinflammatory mediators detectable in plasma and CSF, followed by tissue damage in the hypoxic hemisphere and short-term as well as long-lasting neurobehavioral deficits.
Appendix
Available only for authorised users
Literature
1.
go back to reference Bhalala US, Koehler RC, Kannan S. Neuroinflammation and neuroimmune dysregulation after acute hypoxic-ischemic injury of developing brain. Front Pediatr. 2014;2:144.PubMed Bhalala US, Koehler RC, Kannan S. Neuroinflammation and neuroimmune dysregulation after acute hypoxic-ischemic injury of developing brain. Front Pediatr. 2014;2:144.PubMed
2.
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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
3.
go back to reference Kharoshankaya L, Stevenson NJ, Livingstone V, Murray DM, Murphy BP, Ahearne CE, et al. Seizure burden and neurodevelopmental outcome in neonates with hypoxic-ischemic encephalopathy. Dev Med Child Neurol. 2016;58:1242–8.CrossRefPubMedPubMedCentral Kharoshankaya L, Stevenson NJ, Livingstone V, Murray DM, Murphy BP, Ahearne CE, et al. Seizure burden and neurodevelopmental outcome in neonates with hypoxic-ischemic encephalopathy. Dev Med Child Neurol. 2016;58:1242–8.CrossRefPubMedPubMedCentral
5.
go back to reference Laptook AR. Birth asphyxia and hypoxic-ischemic brain injury in the preterm infant. Clin Perinatol. 2016;43:529–45.CrossRefPubMed Laptook AR. Birth asphyxia and hypoxic-ischemic brain injury in the preterm infant. Clin Perinatol. 2016;43:529–45.CrossRefPubMed
6.
7.
go back to reference Vexler ZS, Ferriero DM. Molecular and biochemical mechanisms of perinatal brain injury. Semin Neonatol SN. 2001;6:99–108.CrossRefPubMed Vexler ZS, Ferriero DM. Molecular and biochemical mechanisms of perinatal brain injury. Semin Neonatol SN. 2001;6:99–108.CrossRefPubMed
8.
go back to reference Acharya J, Rajamohan AG, Skalski MR, Law M, Kim P, Gibbs W. CT angiography of the head in extracorporeal membrane oxygenation. AJNR Am J Neuroradiol. 2017;38:773–6.CrossRefPubMedPubMedCentral Acharya J, Rajamohan AG, Skalski MR, Law M, Kim P, Gibbs W. CT angiography of the head in extracorporeal membrane oxygenation. AJNR Am J Neuroradiol. 2017;38:773–6.CrossRefPubMedPubMedCentral
9.
go back to reference Jan S, Northington FJ, Parkinson CM, Stafstrom CE. EEG monitoring technique influences the management of hypoxic-ischemic seizures in neonates undergoing therapeutic hypothermia. Dev Neurosci. 2017;39:82–8.CrossRefPubMed Jan S, Northington FJ, Parkinson CM, Stafstrom CE. EEG monitoring technique influences the management of hypoxic-ischemic seizures in neonates undergoing therapeutic hypothermia. Dev Neurosci. 2017;39:82–8.CrossRefPubMed
10.
go back to reference Lv H, Wang Q, Wu S, Yang L, Ren P, Yang Y, et al. Neonatal hypoxic ischemic encephalopathy-related biomarkers in serum and cerebrospinal fluid. Clin Chim Acta Int J Clin Chem. 2015;450:282–97.CrossRef Lv H, Wang Q, Wu S, Yang L, Ren P, Yang Y, et al. Neonatal hypoxic ischemic encephalopathy-related biomarkers in serum and cerebrospinal fluid. Clin Chim Acta Int J Clin Chem. 2015;450:282–97.CrossRef
11.
go back to reference Thoresen M, Hellström-Westas L, Liu X, de Vries LS. Effect of hypothermia on amplitude-integrated electroencephalogram in infants with asphyxia. Pediatrics. 2010;126:e131–9.CrossRefPubMed Thoresen M, Hellström-Westas L, Liu X, de Vries LS. Effect of hypothermia on amplitude-integrated electroencephalogram in infants with asphyxia. Pediatrics. 2010;126:e131–9.CrossRefPubMed
12.
go back to reference Thornton C, Rousset CI, Kichev A, Miyakuni Y, Vontell R, Baburamani AA, et al. Molecular mechanisms of neonatal brain injury. Neurol Res Int. 2012;2012:e506320.CrossRef Thornton C, Rousset CI, Kichev A, Miyakuni Y, Vontell R, Baburamani AA, et al. Molecular mechanisms of neonatal brain injury. Neurol Res Int. 2012;2012:e506320.CrossRef
13.
go back to reference Disdier C, Zhang J, Fukunaga Y, Lim Y-P, Qiu J, Santoso A, et al. Alterations in inter-alpha inhibitor protein expression after hypoxic-ischemic brain injury in neonatal rats. Int J Dev Neurosci Off J Int Soc Dev Neurosci. 2018;65:54–60.CrossRef Disdier C, Zhang J, Fukunaga Y, Lim Y-P, Qiu J, Santoso A, et al. Alterations in inter-alpha inhibitor protein expression after hypoxic-ischemic brain injury in neonatal rats. Int J Dev Neurosci Off J Int Soc Dev Neurosci. 2018;65:54–60.CrossRef
14.
go back to reference Lai JCY, Rocha-Ferreira E, Ek CJ, Wang X, Hagberg H, Mallard C. Immune responses in perinatal brain injury. Brain Behav Immun. 2017;63:210–23.CrossRefPubMed Lai JCY, Rocha-Ferreira E, Ek CJ, Wang X, Hagberg H, Mallard C. Immune responses in perinatal brain injury. Brain Behav Immun. 2017;63:210–23.CrossRefPubMed
15.
go back to reference Saliba E, Henrot A. Inflammatory mediators and neonatal brain damage. Biol Neonate. 2001;79:224–7.CrossRefPubMed Saliba E, Henrot A. Inflammatory mediators and neonatal brain damage. Biol Neonate. 2001;79:224–7.CrossRefPubMed
16.
go back to reference Thorngren-Jerneck K, Alling C, Herbst A, Amer-Wahlin I, Marsal K. 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.CrossRefPubMed Thorngren-Jerneck K, Alling C, Herbst A, Amer-Wahlin I, Marsal K. 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.CrossRefPubMed
17.
go back to reference McRae A, Gilland E, Bona E, Hagberg H. Microglia activation after neonatal hypoxic-ischemia. Dev Brain Res. 1995;84:245–52.CrossRef McRae A, Gilland E, Bona E, Hagberg H. Microglia activation after neonatal hypoxic-ischemia. Dev Brain Res. 1995;84:245–52.CrossRef
18.
go back to reference Sawada M, Suzumura A, Yamamoto H, Marunouchi T. Activation and proliferation of the isolated microglia by colony stimulating factor-1 and possible involvement of protein kinase C. Brain Res. 1990;509:119–24.CrossRefPubMed Sawada M, Suzumura A, Yamamoto H, Marunouchi T. Activation and proliferation of the isolated microglia by colony stimulating factor-1 and possible involvement of protein kinase C. Brain Res. 1990;509:119–24.CrossRefPubMed
19.
go back to reference Hedtjärn M, Leverin A-L, Eriksson K, Blomgren K, Mallard C, Hagberg H. Interleukin-18 involvement in hypoxic-ischemic brain injury. J Neurosci. 2002;22:5910–9.CrossRefPubMedPubMedCentral Hedtjärn M, Leverin A-L, Eriksson K, Blomgren K, Mallard C, Hagberg H. Interleukin-18 involvement in hypoxic-ischemic brain injury. J Neurosci. 2002;22:5910–9.CrossRefPubMedPubMedCentral
20.
go back to reference Doverhag C, Hedtjärn M, Poirier F, Mallard C, Hagberg H, Karlsson A, et al. Galectin-3 contributes to neonatal hypoxic-ischemic brain injury. Neurobiol Dis. 2010;38:36–46.CrossRefPubMed Doverhag C, Hedtjärn M, Poirier F, Mallard C, Hagberg H, Karlsson A, et al. Galectin-3 contributes to neonatal hypoxic-ischemic brain injury. Neurobiol Dis. 2010;38:36–46.CrossRefPubMed
21.
go back to reference Svedin P, Hagberg H, Sävman K, Zhu C, Mallard C. Matrix metalloproteinase-9 gene knock-out protects the immature brain after cerebral hypoxia–ischemia. J Neurosci. 2007;27:1511–8.CrossRefPubMedPubMedCentral Svedin P, Hagberg H, Sävman K, Zhu C, Mallard C. Matrix metalloproteinase-9 gene knock-out protects the immature brain after cerebral hypoxia–ischemia. J Neurosci. 2007;27:1511–8.CrossRefPubMedPubMedCentral
22.
go back to reference Kaur C, Rathnasamy G, Ling E-A. Roles of activated microglia in hypoxia induced neuroinflammation in the developing brain and the retina. J Neuroimmune Pharmacol Off J Soc NeuroImmune Pharmacol. 2013;8:66–78.CrossRef Kaur C, Rathnasamy G, Ling E-A. Roles of activated microglia in hypoxia induced neuroinflammation in the developing brain and the retina. J Neuroimmune Pharmacol Off J Soc NeuroImmune Pharmacol. 2013;8:66–78.CrossRef
24.
go back to reference Riljak V, Kraf J, Daryanani A, Jiruška P, Otáhal J. Pathophysiology of perinatal hypoxic-ischemic encephalopathy - biomarkers, animal models and treatment perspectives. Physiol Res. 2016;65:S533–45.PubMed Riljak V, Kraf J, Daryanani A, Jiruška P, Otáhal J. Pathophysiology of perinatal hypoxic-ischemic encephalopathy - biomarkers, animal models and treatment perspectives. Physiol Res. 2016;65:S533–45.PubMed
26.
go back to reference Rice JE, Vannucci RC, Brierley JB. The influence of immaturity on hypoxic-ischemic brain damage in the rat. Ann Neurol. 1981;9:131–41.CrossRefPubMed Rice JE, Vannucci RC, Brierley JB. The influence of immaturity on hypoxic-ischemic brain damage in the rat. Ann Neurol. 1981;9:131–41.CrossRefPubMed
27.
go back to reference Jensen FE, Applegate CD, Holtzman D, Belin TR, Burchfiel JL. Epileptogenic effect of hypoxia in the immature rodent brain. Ann Neurol. 1991;29:629–37.CrossRefPubMed Jensen FE, Applegate CD, Holtzman D, Belin TR, Burchfiel JL. Epileptogenic effect of hypoxia in the immature rodent brain. Ann Neurol. 1991;29:629–37.CrossRefPubMed
28.
go back to reference Leonard AS, Hyder SN, Kolls BJ, Arehart E, KC WN, Veerapandiyan A, et al. Seizure predisposition after perinatal hypoxia: effects of subsequent age and of an epilepsy predisposing gene mutation. Epilepsia. 2013;54:1789–800.CrossRefPubMed Leonard AS, Hyder SN, Kolls BJ, Arehart E, KC WN, Veerapandiyan A, et al. Seizure predisposition after perinatal hypoxia: effects of subsequent age and of an epilepsy predisposing gene mutation. Epilepsia. 2013;54:1789–800.CrossRefPubMed
29.
go back to reference Owens J, Robbins CA, Wenzel HJ, Schwartzkroin PA. Acute and chronic effects of hypoxia on the developing hippocampus. Ann Neurol. 1997;41:187–99.CrossRefPubMed Owens J, Robbins CA, Wenzel HJ, Schwartzkroin PA. Acute and chronic effects of hypoxia on the developing hippocampus. Ann Neurol. 1997;41:187–99.CrossRefPubMed
30.
go back to reference Rakhade SN, Klein PM, Huynh T, Hilario-Gomez C, Kosaras B, Rotenberg A, et al. Development of later life spontaneous seizures in a rodent model of hypoxia-induced neonatal seizures. Epilepsia. 2011;52:753–65.CrossRefPubMedPubMedCentral Rakhade SN, Klein PM, Huynh T, Hilario-Gomez C, Kosaras B, Rotenberg A, et al. Development of later life spontaneous seizures in a rodent model of hypoxia-induced neonatal seizures. Epilepsia. 2011;52:753–65.CrossRefPubMedPubMedCentral
31.
go back to reference Hosono T, Kamo A, Hakotani S, Minato K, Akeno H, Taguchi Y, et al. Effect of hypothermia on motor function of adult rats after neonatal hyperthermic hypoxic-ischemic brain insult. Eur J Appl Physiol. 2010;109:35–9.CrossRefPubMed Hosono T, Kamo A, Hakotani S, Minato K, Akeno H, Taguchi Y, et al. Effect of hypothermia on motor function of adult rats after neonatal hyperthermic hypoxic-ischemic brain insult. Eur J Appl Physiol. 2010;109:35–9.CrossRefPubMed
32.
go back to reference Mortola JP, Dotta A. Effects of hypoxia and ambient temperature on gaseous metabolism of newborn rats. Am J Phys. 1992;263:R267–72.CrossRef Mortola JP, Dotta A. Effects of hypoxia and ambient temperature on gaseous metabolism of newborn rats. Am J Phys. 1992;263:R267–72.CrossRef
33.
go back to reference Rumajogee P, Bregman T, Miller SP, Yager JY, Fehlings MG. Rodent hypoxia-ischemia models for cerebral palsy research: a systematic review. Front Neurol. 2016;7:57.CrossRefPubMedPubMedCentral Rumajogee P, Bregman T, Miller SP, Yager JY, Fehlings MG. Rodent hypoxia-ischemia models for cerebral palsy research: a systematic review. Front Neurol. 2016;7:57.CrossRefPubMedPubMedCentral
34.
go back to reference Balduini W, De Angelis V, Mazzoni E, Cimino M. Long-lasting behavioral alterations following a hypoxic/ischemic brain injury in neonatal rats. Brain Res. 2000;859:318–25.CrossRefPubMed Balduini W, De Angelis V, Mazzoni E, Cimino M. Long-lasting behavioral alterations following a hypoxic/ischemic brain injury in neonatal rats. Brain Res. 2000;859:318–25.CrossRefPubMed
35.
go back to reference Rojas JJ, Deniz BF, Miguel PM, Diaz R, Hermel Édo E-S, Achaval M, et al. Effects of daily environmental enrichment on behavior and dendritic spine density in hippocampus following neonatal hypoxia–ischemia in the rat. Exp Neurol. 2013;241:25–33.CrossRefPubMed Rojas JJ, Deniz BF, Miguel PM, Diaz R, Hermel Édo E-S, Achaval M, et al. Effects of daily environmental enrichment on behavior and dendritic spine density in hippocampus following neonatal hypoxia–ischemia in the rat. Exp Neurol. 2013;241:25–33.CrossRefPubMed
36.
go back to reference Takao K, Tanda K, Nakamura K, Kasahara J, Nakao K, Katsuki M, et al. Comprehensive behavioral analysis of calcium/calmodulin-dependent protein kinase IV knockout mice. PLoS One. 2010;5:e9460.CrossRefPubMedPubMedCentral Takao K, Tanda K, Nakamura K, Kasahara J, Nakao K, Katsuki M, et al. Comprehensive behavioral analysis of calcium/calmodulin-dependent protein kinase IV knockout mice. PLoS One. 2010;5:e9460.CrossRefPubMedPubMedCentral
37.
go back to reference Hattori T, Sato Y, Kondo T, Ichinohashi Y, Sugiyama Y, Yamamoto M, et al. Administration of umbilical cord blood cells transiently decreased hypoxic-ischemic brain injury in neonatal rats. Dev Neurosci. 2015;37:95–104.CrossRefPubMed Hattori T, Sato Y, Kondo T, Ichinohashi Y, Sugiyama Y, Yamamoto M, et al. Administration of umbilical cord blood cells transiently decreased hypoxic-ischemic brain injury in neonatal rats. Dev Neurosci. 2015;37:95–104.CrossRefPubMed
38.
go back to reference Chou I-C, Trakht T, Signori C, Smith J, Felt BT, Vazquez DM, et al. Behavioral/environmental intervention improves learning after cerebral hypoxia-ischemia in rats. Stroke. 2001;32:2192–7.CrossRefPubMed Chou I-C, Trakht T, Signori C, Smith J, Felt BT, Vazquez DM, et al. Behavioral/environmental intervention improves learning after cerebral hypoxia-ischemia in rats. Stroke. 2001;32:2192–7.CrossRefPubMed
39.
go back to reference Liu L, Duff K. A technique for serial collection of cerebrospinal fluid from the cisterna magna in mouse. J Vis Exp JoVE. 2008;21:960-21. Liu L, Duff K. A technique for serial collection of cerebrospinal fluid from the cisterna magna in mouse. J Vis Exp JoVE. 2008;21:960-21.
40.
go back to reference Rodríguez-Fanjul J, Fernández-Feijóo CD, Camprubí MC. A new technique for collection of cerebrospinal fluid in rat pups. J Exp Neurosci. 2015;9:37–41.CrossRefPubMedPubMedCentral Rodríguez-Fanjul J, Fernández-Feijóo CD, Camprubí MC. A new technique for collection of cerebrospinal fluid in rat pups. J Exp Neurosci. 2015;9:37–41.CrossRefPubMedPubMedCentral
41.
go back to reference Daskalopoulos EP, Hermans KCM, van Delft L, Altara R, Blankesteijn WM. In-flammation in Heart Failure, first ed. Academic Press; 2014. Daskalopoulos EP, Hermans KCM, van Delft L, Altara R, Blankesteijn WM. In-flammation in Heart Failure, first ed. Academic Press; 2014.
43.
go back to reference Ten VS, Bradley-Moore M, Gingrich JA, Stark RI, Pinsky DJ. Brain injury and neurofunctional deficit in neonatal mice with hypoxic-ischemic encephalopathy. Behav Brain Res. 2003;145:209–19.CrossRefPubMed Ten VS, Bradley-Moore M, Gingrich JA, Stark RI, Pinsky DJ. Brain injury and neurofunctional deficit in neonatal mice with hypoxic-ischemic encephalopathy. Behav Brain Res. 2003;145:209–19.CrossRefPubMed
44.
go back to reference Wagner BP, Nedelcu J, Martin E. Delayed postischemic hypothermia improves long-term behavioral outcome after cerebral hypoxia-ischemia in neonatal rats. Pediatr Res. 2002;51:354–60.CrossRefPubMed Wagner BP, Nedelcu J, Martin E. Delayed postischemic hypothermia improves long-term behavioral outcome after cerebral hypoxia-ischemia in neonatal rats. Pediatr Res. 2002;51:354–60.CrossRefPubMed
45.
go back to reference Lubics A, Reglodi D, Tamás A, Kiss P, Szalai M, Szalontay L, et al. Neurological reflexes and early motor behavior in rats subjected to neonatal hypoxic-ischemic injury. Behav Brain Res. 2005;157:157–65.CrossRefPubMed Lubics A, Reglodi D, Tamás A, Kiss P, Szalai M, Szalontay L, et al. Neurological reflexes and early motor behavior in rats subjected to neonatal hypoxic-ischemic injury. Behav Brain Res. 2005;157:157–65.CrossRefPubMed
46.
go back to reference Patel S, Player MR. Colony-stimulating factor-1 receptor inhibitors for the treatment of cancer and inflammatory disease. Curr Top Med Chem. 2009;9:599–610.CrossRefPubMed Patel S, Player MR. Colony-stimulating factor-1 receptor inhibitors for the treatment of cancer and inflammatory disease. Curr Top Med Chem. 2009;9:599–610.CrossRefPubMed
47.
go back to reference Antier D, Zhang BL, Mailliet F, Akoka S, Pourcelot L, Sannajust F. Effects of neonatal focal cerebral hypoxia-ischemia on sleep-waking pattern, ECoG power spectra and locomotor activity in the adult rat. Brain Res. 1998;807:29–37.CrossRefPubMed Antier D, Zhang BL, Mailliet F, Akoka S, Pourcelot L, Sannajust F. Effects of neonatal focal cerebral hypoxia-ischemia on sleep-waking pattern, ECoG power spectra and locomotor activity in the adult rat. Brain Res. 1998;807:29–37.CrossRefPubMed
48.
go back to reference de Paula S, Vitola AS, Greggio S, de Paula D, Mello PB, Lubianca JM, et al. Hemispheric brain injury and behavioral deficits induced by severe neonatal hypoxia-ischemia in rats are not attenuated by intravenous administration of human umbilical cord blood cells. Pediatr Res. 2009;65:631–5.CrossRefPubMed de Paula S, Vitola AS, Greggio S, de Paula D, Mello PB, Lubianca JM, et al. Hemispheric brain injury and behavioral deficits induced by severe neonatal hypoxia-ischemia in rats are not attenuated by intravenous administration of human umbilical cord blood cells. Pediatr Res. 2009;65:631–5.CrossRefPubMed
49.
go back to reference Goren B, Cakir A, Ocalan B, Serter Kocoglu S, Alkan T, Cansev M, et al. Long-term cognitive effects of uridine treatment in a neonatal rat model of hypoxic-ischemic encephalopathy. Brain Res. 2017;1659:81–7.CrossRefPubMed Goren B, Cakir A, Ocalan B, Serter Kocoglu S, Alkan T, Cansev M, et al. Long-term cognitive effects of uridine treatment in a neonatal rat model of hypoxic-ischemic encephalopathy. Brain Res. 2017;1659:81–7.CrossRefPubMed
50.
go back to reference Johnston MV, Hagberg H. Sex and the pathogenesis of cerebral palsy. Dev Med Child Neurol. 2007;49:74–8.CrossRefPubMed Johnston MV, Hagberg H. Sex and the pathogenesis of cerebral palsy. Dev Med Child Neurol. 2007;49:74–8.CrossRefPubMed
51.
go back to reference Hagberg H, Wilson MA, Matsushita H, Zhu C, Lange M, Gustavsson M, et al. PARP-1 gene disruption in mice preferentially protects males from perinatal brain injury. J Neurochem. 2004;90:1068–75.CrossRefPubMed Hagberg H, Wilson MA, Matsushita H, Zhu C, Lange M, Gustavsson M, et al. PARP-1 gene disruption in mice preferentially protects males from perinatal brain injury. J Neurochem. 2004;90:1068–75.CrossRefPubMed
52.
go back to reference Barth TM, Stanfield BB. The recovery of forelimb-placing behavior in rats with neonatal unilateral cortical damage involves the remaining hemisphere. J Neurosci. 1990;10:3449–59.CrossRefPubMedPubMedCentral Barth TM, Stanfield BB. The recovery of forelimb-placing behavior in rats with neonatal unilateral cortical damage involves the remaining hemisphere. J Neurosci. 1990;10:3449–59.CrossRefPubMedPubMedCentral
53.
go back to reference Alsina M, Martín-Ancel A, Alarcon-Allen A, Arca G, Gayá F, García-Alix A. The severity of hypoxic-ischemic encephalopathy correlates with multiple organ dysfunction in the hypothermia era. Pediatr Crit Care Med J Soc Crit Care Med World Fed Pediatr Intensive Crit Care Soc. 2017;18:234–40. Alsina M, Martín-Ancel A, Alarcon-Allen A, Arca G, Gayá F, García-Alix A. The severity of hypoxic-ischemic encephalopathy correlates with multiple organ dysfunction in the hypothermia era. Pediatr Crit Care Med J Soc Crit Care Med World Fed Pediatr Intensive Crit Care Soc. 2017;18:234–40.
55.
go back to reference Hedtjärn M, Mallard C, Hagberg H. Inflammatory gene profiling in the developing mouse brain after hypoxia-ischemia. J Cereb Blood Flow Metab Off J Int Soc Cereb Blood Flow Metab. 2004;24:1333–51.CrossRef Hedtjärn M, Mallard C, Hagberg H. Inflammatory gene profiling in the developing mouse brain after hypoxia-ischemia. J Cereb Blood Flow Metab Off J Int Soc Cereb Blood Flow Metab. 2004;24:1333–51.CrossRef
56.
57.
go back to reference Drews K, Szczapa J, Zak J, Andrzejewska R, Zak L, Mackiewicz A. Blood serum concentration of C-reactive protein and interleukin-6 in diagnosis of neonatal infections. Ann N Y Acad Sci. 1995;762:398–9.CrossRefPubMed Drews K, Szczapa J, Zak J, Andrzejewska R, Zak L, Mackiewicz A. Blood serum concentration of C-reactive protein and interleukin-6 in diagnosis of neonatal infections. Ann N Y Acad Sci. 1995;762:398–9.CrossRefPubMed
58.
go back to reference Lusyati S, Hulzebos CV, Zandvoort J, Sukandar H, Sauer PJJ. Cytokines patterns in newborn infants with late onset sepsis. J Neonatal-Perinat Med. 2013;6:153–63. Lusyati S, Hulzebos CV, Zandvoort J, Sukandar H, Sauer PJJ. Cytokines patterns in newborn infants with late onset sepsis. J Neonatal-Perinat Med. 2013;6:153–63.
59.
go back to reference Martín-Ancel A, García-Alix A, Pascual-Salcedo D, Cabañas F, Valcarce M, Quero J. Interleukin-6 in the cerebrospinal fluid after perinatal asphyxia is related to early and late neurological manifestations. Pediatrics. 1997;100:789–94.CrossRefPubMed Martín-Ancel A, García-Alix A, Pascual-Salcedo D, Cabañas F, Valcarce M, Quero J. Interleukin-6 in the cerebrospinal fluid after perinatal asphyxia is related to early and late neurological manifestations. Pediatrics. 1997;100:789–94.CrossRefPubMed
60.
go back to reference Shahkar L, Keshtkar A, Mirfazeli A, Ahani A, Roshandel G. The role of IL-6 for predicting neonatal sepsis: a systematic review and meta-analysis. Iran J Pediatr. 2011;21:411–7.PubMedPubMedCentral Shahkar L, Keshtkar A, Mirfazeli A, Ahani A, Roshandel G. The role of IL-6 for predicting neonatal sepsis: a systematic review and meta-analysis. Iran J Pediatr. 2011;21:411–7.PubMedPubMedCentral
62.
go back to reference Dimitrijevic OB, Stamatovic SM, Keep RF, Andjelkovic AV. Absence of the chemokine receptor CCR2 protects against cerebral ischemia/reperfusion injury in mice. Stroke. 2007;38:1345–53.CrossRefPubMed Dimitrijevic OB, Stamatovic SM, Keep RF, Andjelkovic AV. Absence of the chemokine receptor CCR2 protects against cerebral ischemia/reperfusion injury in mice. Stroke. 2007;38:1345–53.CrossRefPubMed
63.
go back to reference Schilling M, Strecker J-K, Ringelstein EB, Schäbitz W-R, Kiefer R. The role of CC chemokine receptor 2 on microglia activation and blood-borne cell recruitment after transient focal cerebral ischemia in mice. Brain Res. 2009;1289:79–84.CrossRefPubMed Schilling M, Strecker J-K, Ringelstein EB, Schäbitz W-R, Kiefer R. The role of CC chemokine receptor 2 on microglia activation and blood-borne cell recruitment after transient focal cerebral ischemia in mice. Brain Res. 2009;1289:79–84.CrossRefPubMed
64.
go back to reference Li S-J, Liu W, Wang J-L, Zhang Y, Zhao D-J, Wang T-J, et al. The role of TNF-α, IL-6, IL-10, and GDNF in neuronal apoptosis in neonatal rat with hypoxic-ischemic encephalopathy. Eur Rev Med Pharmacol Sci. 2014;18:905–9.PubMed Li S-J, Liu W, Wang J-L, Zhang Y, Zhao D-J, Wang T-J, et al. The role of TNF-α, IL-6, IL-10, and GDNF in neuronal apoptosis in neonatal rat with hypoxic-ischemic encephalopathy. Eur Rev Med Pharmacol Sci. 2014;18:905–9.PubMed
65.
go back to reference Olmos G, Lladó J. Tumor necrosis factor alpha: a link between neuroinflammation and excitotoxicity. Mediat Inflamm. 2014;2014:861231.CrossRef Olmos G, Lladó J. Tumor necrosis factor alpha: a link between neuroinflammation and excitotoxicity. Mediat Inflamm. 2014;2014:861231.CrossRef
66.
go back to reference Szaflarski J, Burtrum D, Silverstein FS. Cerebral hypoxia-ischemia stimulates cytokine gene expression in perinatal rats. Stroke. 1995;26:1093–100.CrossRefPubMed Szaflarski J, Burtrum D, Silverstein FS. Cerebral hypoxia-ischemia stimulates cytokine gene expression in perinatal rats. Stroke. 1995;26:1093–100.CrossRefPubMed
67.
go back to reference Deng YY, Lu J, Ling E-A, Kaur C. Microglia-derived macrophage colony stimulating factor promotes generation of proinflammatory cytokines by astrocytes in the periventricular white matter in the hypoxic neonatal brain. Brain Pathol Zurich Switz. 2010;20:909–25. Deng YY, Lu J, Ling E-A, Kaur C. Microglia-derived macrophage colony stimulating factor promotes generation of proinflammatory cytokines by astrocytes in the periventricular white matter in the hypoxic neonatal brain. Brain Pathol Zurich Switz. 2010;20:909–25.
68.
go back to reference Firdaus F, Zafeer MF, Ahmad M, Afzal M. Anxiolytic and anti-inflammatory role of thymoquinone in arsenic-induced hippocampal toxicity in Wistar rats. Heliyon. 2018;4:e00650.CrossRefPubMedPubMedCentral Firdaus F, Zafeer MF, Ahmad M, Afzal M. Anxiolytic and anti-inflammatory role of thymoquinone in arsenic-induced hippocampal toxicity in Wistar rats. Heliyon. 2018;4:e00650.CrossRefPubMedPubMedCentral
69.
go back to reference Rajabi M, Mohaddes G, Farajdokht F, Nayebi Rad S, Mesgari M, Babri S. Impact of loganin on pro-inflammatory cytokines and depression- and anxiety-like behaviors in male diabetic rats. Physiol Int. 2018;105:116–26.CrossRefPubMed Rajabi M, Mohaddes G, Farajdokht F, Nayebi Rad S, Mesgari M, Babri S. Impact of loganin on pro-inflammatory cytokines and depression- and anxiety-like behaviors in male diabetic rats. Physiol Int. 2018;105:116–26.CrossRefPubMed
70.
go back to reference Borjini N, Fernández M, Giardino L, Calzà L. Cytokine and chemokine alterations in tissue, CSF, and plasma in early presymptomatic phase of experimental allergic encephalomyelitis (EAE), in a rat model of multiple sclerosis. J Neuroinflammation. 2016;13:291.CrossRefPubMedPubMedCentral Borjini N, Fernández M, Giardino L, Calzà L. Cytokine and chemokine alterations in tissue, CSF, and plasma in early presymptomatic phase of experimental allergic encephalomyelitis (EAE), in a rat model of multiple sclerosis. J Neuroinflammation. 2016;13:291.CrossRefPubMedPubMedCentral
71.
go back to reference Denes A, Vidyasagar R, Feng J, Narvainen J, McColl BW, Kauppinen RA, et al. Proliferating resident microglia after focal cerebral ischaemia in mice. J Cereb Blood Flow Metab Off J Int Soc Cereb Blood Flow Metab. 2007;27:1941–53.CrossRef Denes A, Vidyasagar R, Feng J, Narvainen J, McColl BW, Kauppinen RA, et al. Proliferating resident microglia after focal cerebral ischaemia in mice. J Cereb Blood Flow Metab Off J Int Soc Cereb Blood Flow Metab. 2007;27:1941–53.CrossRef
72.
go back to reference Escamilla J, Schokrpur S, Liu C, Priceman SJ, Moughon D, Jiang Z, et al. CSF1 receptor targeting in prostate cancer reverses macrophage-mediated resistance to androgen blockade therapy. Cancer Res. 2015;75:950–62.CrossRefPubMedPubMedCentral Escamilla J, Schokrpur S, Liu C, Priceman SJ, Moughon D, Jiang Z, et al. CSF1 receptor targeting in prostate cancer reverses macrophage-mediated resistance to androgen blockade therapy. Cancer Res. 2015;75:950–62.CrossRefPubMedPubMedCentral
73.
go back to reference Sanchez-Niño MD, Sanz AB, Ortiz A. Chronicity following ischaemia-reperfusion injury depends on tubular-macrophage crosstalk involving two tubular cell-derived CSF-1R activators: CSF-1 and IL-34. Nephrol Dial Transplant. 2016;31:1409–16.CrossRefPubMed Sanchez-Niño MD, Sanz AB, Ortiz A. Chronicity following ischaemia-reperfusion injury depends on tubular-macrophage crosstalk involving two tubular cell-derived CSF-1R activators: CSF-1 and IL-34. Nephrol Dial Transplant. 2016;31:1409–16.CrossRefPubMed
74.
go back to reference Al Mamun A, Yu H, Romana S, Liu F. Inflammatory responses are sex specific in chronic hypoxic-ischemic encephalopathy. Cell Transplant. 2018;27:1328–39.CrossRefPubMedPubMedCentral Al Mamun A, Yu H, Romana S, Liu F. Inflammatory responses are sex specific in chronic hypoxic-ischemic encephalopathy. Cell Transplant. 2018;27:1328–39.CrossRefPubMedPubMedCentral
75.
go back to reference Ardalan M, Chumak T, Vexler Z, Mallard C. Sex-dependent effects of perinatal inflammation on the brain: implication for neuro-psychiatric disorders. Int J Mol Sci. 2019;20:2270. Ardalan M, Chumak T, Vexler Z, Mallard C. Sex-dependent effects of perinatal inflammation on the brain: implication for neuro-psychiatric disorders. Int J Mol Sci. 2019;20:2270.
76.
go back to reference Hoogland ICM, Houbolt C, van Westerloo DJ, van Gool WA, van de Beek D. Systemic inflammation and microglial activation: systematic review of animal experiments. J Neuroinflammation. 2015;12:114.CrossRefPubMedPubMedCentral Hoogland ICM, Houbolt C, van Westerloo DJ, van Gool WA, van de Beek D. Systemic inflammation and microglial activation: systematic review of animal experiments. J Neuroinflammation. 2015;12:114.CrossRefPubMedPubMedCentral
77.
go back to reference Massella A, D’Intino G, Fernández M, Sivilia S, Lorenzini L, Giatti S, et al. Gender effect on neurodegeneration and myelin markers in an animal model for multiple sclerosis. BMC Neurosci. 2012;13:12.CrossRefPubMedPubMedCentral Massella A, D’Intino G, Fernández M, Sivilia S, Lorenzini L, Giatti S, et al. Gender effect on neurodegeneration and myelin markers in an animal model for multiple sclerosis. BMC Neurosci. 2012;13:12.CrossRefPubMedPubMedCentral
78.
go back to reference Caruso D, D’Intino G, Giatti S, Maschi O, Pesaresi M, Calabrese D, et al. Sex-dimorphic changes in neuroactive steroid levels after chronic experimental autoimmune encephalomyelitis. J Neurochem. 2010;114:921–32.CrossRefPubMed Caruso D, D’Intino G, Giatti S, Maschi O, Pesaresi M, Calabrese D, et al. Sex-dimorphic changes in neuroactive steroid levels after chronic experimental autoimmune encephalomyelitis. J Neurochem. 2010;114:921–32.CrossRefPubMed
79.
go back to reference Lee WLA, Michael-Titus AT, Shah DK. Hypoxic-ischaemic encephalopathy and the blood-brain barrier in neonates. Dev Neurosci. 2017;39:49–58.CrossRefPubMed Lee WLA, Michael-Titus AT, Shah DK. Hypoxic-ischaemic encephalopathy and the blood-brain barrier in neonates. Dev Neurosci. 2017;39:49–58.CrossRefPubMed
80.
go back to reference Rajendran P, Rengarajan T, Thangavel J, Nishigaki Y, Sakthisekaran D, Sethi G, et al. The vascular endothelium and human diseases. Int J Biol Sci. 2013;9:1057–69.CrossRefPubMedPubMedCentral Rajendran P, Rengarajan T, Thangavel J, Nishigaki Y, Sakthisekaran D, Sethi G, et al. The vascular endothelium and human diseases. Int J Biol Sci. 2013;9:1057–69.CrossRefPubMedPubMedCentral
81.
go back to reference Ho MCY, Lo ACY, Kurihara H, Yu ACH, Chung SSM, Chung SK. Endothelin-1 protects astrocytes from hypoxic/ischemic injury. FASEB J. 2001;15:618–26.CrossRefPubMed Ho MCY, Lo ACY, Kurihara H, Yu ACH, Chung SSM, Chung SK. Endothelin-1 protects astrocytes from hypoxic/ischemic injury. FASEB J. 2001;15:618–26.CrossRefPubMed
82.
go back to reference Tsang MC, Lo AC, Cheung PT, Chung SS, Chung SK. Perinatal hypoxia-/ischemia-induced endothelin-1 mRNA in astrocyte-like and endothelial cells. Neuroreport. 2001;12:2265–70.CrossRefPubMed Tsang MC, Lo AC, Cheung PT, Chung SS, Chung SK. Perinatal hypoxia-/ischemia-induced endothelin-1 mRNA in astrocyte-like and endothelial cells. Neuroreport. 2001;12:2265–70.CrossRefPubMed
83.
go back to reference Spoljaric A, Seja P, Spoljaric I, Virtanen MA, Lindfors J, Uvarov P, et al. Vasopressin excites interneurons to suppress hippocampal network activity across a broad span of brain maturity at birth. Proc Natl Acad Sci U S A. 2017;114:E10819–28.CrossRefPubMedPubMedCentral Spoljaric A, Seja P, Spoljaric I, Virtanen MA, Lindfors J, Uvarov P, et al. Vasopressin excites interneurons to suppress hippocampal network activity across a broad span of brain maturity at birth. Proc Natl Acad Sci U S A. 2017;114:E10819–28.CrossRefPubMedPubMedCentral
84.
go back to reference Hu J, Discher DJ, Bishopric NH, Webster KA. Hypoxia regulates expression of the endothelin-1 gene through a proximal hypoxia-inducible factor-1 binding site on the antisense strand. Biochem Biophys Res Commun. 1998;245:894–9.CrossRefPubMed Hu J, Discher DJ, Bishopric NH, Webster KA. Hypoxia regulates expression of the endothelin-1 gene through a proximal hypoxia-inducible factor-1 binding site on the antisense strand. Biochem Biophys Res Commun. 1998;245:894–9.CrossRefPubMed
85.
go back to reference Semenza GL. Hypoxia-inducible factor 1: oxygen homeostasis and disease pathophysiology. Trends Mol Med. 2001;7:345–50.CrossRefPubMed Semenza GL. Hypoxia-inducible factor 1: oxygen homeostasis and disease pathophysiology. Trends Mol Med. 2001;7:345–50.CrossRefPubMed
86.
go back to reference Barteczek P, Li L, Ernst A-S, Böhler L-I, Marti HH, Kunze R. Neuronal HIF-1α and HIF-2α deficiency improves neuronal survival and sensorimotor function in the early acute phase after ischemic stroke. J Cereb Blood Flow Metab. 2017;37:291–306.CrossRefPubMed Barteczek P, Li L, Ernst A-S, Böhler L-I, Marti HH, Kunze R. Neuronal HIF-1α and HIF-2α deficiency improves neuronal survival and sensorimotor function in the early acute phase after ischemic stroke. J Cereb Blood Flow Metab. 2017;37:291–306.CrossRefPubMed
87.
go back to reference Cao Y, Li Z, Li H, Ni C, Li L, Yang N, et al. Hypoxia-inducible factor-1α is involved in isoflurane-induced blood-brain barrier disruption in aged rats model of POCD. Behav Brain Res. 2018;339:39–46.CrossRefPubMed Cao Y, Li Z, Li H, Ni C, Li L, Yang N, et al. Hypoxia-inducible factor-1α is involved in isoflurane-induced blood-brain barrier disruption in aged rats model of POCD. Behav Brain Res. 2018;339:39–46.CrossRefPubMed
88.
go back to reference Geng J, Wang L, Qu M, Song Y, Lin X, Chen Y, et al. Endothelial progenitor cells transplantation attenuated blood-brain barrier damage after ischemia in diabetic mice via HIF-1α. Stem Cell Res Ther. 2017;8:163.CrossRefPubMedPubMedCentral Geng J, Wang L, Qu M, Song Y, Lin X, Chen Y, et al. Endothelial progenitor cells transplantation attenuated blood-brain barrier damage after ischemia in diabetic mice via HIF-1α. Stem Cell Res Ther. 2017;8:163.CrossRefPubMedPubMedCentral
90.
go back to reference Bauer AT, Bürgers HF, Rabie T, Marti HH. Matrix metalloproteinase-9 mediates hypoxia-induced vascular leakage in the brain via tight junction rearrangement. J Cereb Blood Flow Metab Off J Int Soc Cereb Blood Flow Metab. 2010;30:837–48.CrossRef Bauer AT, Bürgers HF, Rabie T, Marti HH. Matrix metalloproteinase-9 mediates hypoxia-induced vascular leakage in the brain via tight junction rearrangement. J Cereb Blood Flow Metab Off J Int Soc Cereb Blood Flow Metab. 2010;30:837–48.CrossRef
91.
go back to reference Higashida T, Kreipke CW, Rafols JA, Peng C, Schafer S, Schafer P, et al. The role of hypoxia-inducible factor-1α, aquaporin-4, and matrix metalloproteinase-9 in blood-brain barrier disruption and brain edema after traumatic brain injury. J Neurosurg. 2010;114:92–101.CrossRefPubMed Higashida T, Kreipke CW, Rafols JA, Peng C, Schafer S, Schafer P, et al. The role of hypoxia-inducible factor-1α, aquaporin-4, and matrix metalloproteinase-9 in blood-brain barrier disruption and brain edema after traumatic brain injury. J Neurosurg. 2010;114:92–101.CrossRefPubMed
92.
go back to reference Mun-Bryce S, Rosenberg GA. Gelatinase B modulates selective opening of the blood-brain barrier during inflammation. Am J Physiol-Regul Integr Comp Physiol. 1998;274:R1203–11.CrossRef Mun-Bryce S, Rosenberg GA. Gelatinase B modulates selective opening of the blood-brain barrier during inflammation. Am J Physiol-Regul Integr Comp Physiol. 1998;274:R1203–11.CrossRef
93.
go back to reference Rosenberg GA, Dencoff JE, McGuire PG, Liotta LA, Stetler-Stevenson WG. Injury-induced 92-kilodalton gelatinase and urokinase expression in rat brain. Lab Investig J Tech Methods Pathol. 1994;71:417–22. Rosenberg GA, Dencoff JE, McGuire PG, Liotta LA, Stetler-Stevenson WG. Injury-induced 92-kilodalton gelatinase and urokinase expression in rat brain. Lab Investig J Tech Methods Pathol. 1994;71:417–22.
94.
go back to reference Ziemka-Nalecz M, Janowska J, Strojek L, Jaworska J, Zalewska T, Frontczak-Baniewicz M, et al. Impact of neonatal hypoxia-ischaemia on oligodendrocyte survival, maturation and myelinating potential. J Cell Mol Med. 2018;22:207–22.CrossRefPubMed Ziemka-Nalecz M, Janowska J, Strojek L, Jaworska J, Zalewska T, Frontczak-Baniewicz M, et al. Impact of neonatal hypoxia-ischaemia on oligodendrocyte survival, maturation and myelinating potential. J Cell Mol Med. 2018;22:207–22.CrossRefPubMed
95.
go back to reference Hollborn M, Kohen L, Werschnik C, Tietz L, Wiedemann P, Bringmann A. Activated blood coagulation factor X (FXa) induces angiogenic growth factor expression in human retinal pigment epithelial cells. Invest Ophthalmol Vis Sci. 2012;53:5930–9.CrossRefPubMed Hollborn M, Kohen L, Werschnik C, Tietz L, Wiedemann P, Bringmann A. Activated blood coagulation factor X (FXa) induces angiogenic growth factor expression in human retinal pigment epithelial cells. Invest Ophthalmol Vis Sci. 2012;53:5930–9.CrossRefPubMed
96.
go back to reference Monteiro RQ, Lima LG, Gonçalves NP, De Souza MRA, Leal AC, Demasi MAA, et al. Hypoxia regulates the expression of tissue factor pathway signaling elements in a rat glioma model. Oncol Lett. 2016;12:315–22.CrossRefPubMedPubMedCentral Monteiro RQ, Lima LG, Gonçalves NP, De Souza MRA, Leal AC, Demasi MAA, et al. Hypoxia regulates the expression of tissue factor pathway signaling elements in a rat glioma model. Oncol Lett. 2016;12:315–22.CrossRefPubMedPubMedCentral
97.
go back to reference Jha NK, Jha SK, Sharma R, Kumar D, Ambasta RK, Kumar P. Hypoxia-induced signaling activation in neurodegenerative diseases: targets for new therapeutic strategies. J Alzheimers Dis. 2018;62:15–38.CrossRefPubMed Jha NK, Jha SK, Sharma R, Kumar D, Ambasta RK, Kumar P. Hypoxia-induced signaling activation in neurodegenerative diseases: targets for new therapeutic strategies. J Alzheimers Dis. 2018;62:15–38.CrossRefPubMed
99.
go back to reference Suzuki N, Gradin K, Poellinger L, Yamamoto M. Regulation of hypoxia-inducible gene expression after HIF activation. Exp Cell Res. 2017;356:182–6.CrossRefPubMed Suzuki N, Gradin K, Poellinger L, Yamamoto M. Regulation of hypoxia-inducible gene expression after HIF activation. Exp Cell Res. 2017;356:182–6.CrossRefPubMed
100.
go back to reference Wu YW, Gonzalez FF. Erythropoietin: a novel therapy for hypoxic-ischaemic encephalopathy? Dev Med Child Neurol. 2015;57(Suppl 3):34–9.CrossRefPubMed Wu YW, Gonzalez FF. Erythropoietin: a novel therapy for hypoxic-ischaemic encephalopathy? Dev Med Child Neurol. 2015;57(Suppl 3):34–9.CrossRefPubMed
102.
go back to reference Sun Y, Calvert JW, Zhang JH. Neonatal hypoxia/ischemia is associated with decreased inflammatory mediators after erythropoietin administration. Stroke. 2005;36:1672–8.CrossRefPubMed Sun Y, Calvert JW, Zhang JH. Neonatal hypoxia/ischemia is associated with decreased inflammatory mediators after erythropoietin administration. Stroke. 2005;36:1672–8.CrossRefPubMed
103.
go back to reference Nair J, Kumar VHS. Current and emerging therapies in the management of hypoxic ischemic encephalopathy in neonates. Child Basel Switz. 2018;5:99. Nair J, Kumar VHS. Current and emerging therapies in the management of hypoxic ischemic encephalopathy in neonates. Child Basel Switz. 2018;5:99.
104.
go back to reference Yiş U, Kurul SH, Kumral A, Tuğyan K, Cilaker S, Yilmaz O, et al. Effect of erythropoietin on oxygen-induced brain injury in the newborn rat. Neurosci Lett. 2008;448:245–9.CrossRefPubMed Yiş U, Kurul SH, Kumral A, Tuğyan K, Cilaker S, Yilmaz O, et al. Effect of erythropoietin on oxygen-induced brain injury in the newborn rat. Neurosci Lett. 2008;448:245–9.CrossRefPubMed
105.
go back to reference Juul SE, Beyer RP, Bammler TK, McPherson RJ, Wilkerson J, Farin FM. Microarray analysis of high-dose recombinant erythropoietin treatment of unilateral brain injury in neonatal mouse hippocampus. Pediatr Res. 2009;65:485–92.CrossRefPubMed Juul SE, Beyer RP, Bammler TK, McPherson RJ, Wilkerson J, Farin FM. Microarray analysis of high-dose recombinant erythropoietin treatment of unilateral brain injury in neonatal mouse hippocampus. Pediatr Res. 2009;65:485–92.CrossRefPubMed
106.
go back to reference Elmahdy H, El-Mashad A-R, El-Bahrawy H, El-Gohary T, El-Barbary A, Aly H. Human recombinant erythropoietin in asphyxia neonatorum: pilot trial. Pediatrics. 2010;125:e1135–42.CrossRefPubMed Elmahdy H, El-Mashad A-R, El-Bahrawy H, El-Gohary T, El-Barbary A, Aly H. Human recombinant erythropoietin in asphyxia neonatorum: pilot trial. Pediatrics. 2010;125:e1135–42.CrossRefPubMed
107.
go back to reference Zhu C, Kang W, Xu F, Cheng X, Zhang Z, Jia L, et al. Erythropoietin improved neurologic outcomes in newborns with hypoxic-ischemic encephalopathy. Pediatrics. 2009;124:e218–26.CrossRefPubMed Zhu C, Kang W, Xu F, Cheng X, Zhang Z, Jia L, et al. Erythropoietin improved neurologic outcomes in newborns with hypoxic-ischemic encephalopathy. Pediatrics. 2009;124:e218–26.CrossRefPubMed
109.
go back to reference Yan S-F, Fujita T, Lu J, Okada K, Shan Zou Y, Mackman N, et al. Egr-1, a master switch coordinating upregulation of divergent gene families underlying ischemic stress. Nat Med. 2000;6:1355–61.CrossRefPubMed Yan S-F, Fujita T, Lu J, Okada K, Shan Zou Y, Mackman N, et al. Egr-1, a master switch coordinating upregulation of divergent gene families underlying ischemic stress. Nat Med. 2000;6:1355–61.CrossRefPubMed
111.
go back to reference Jones MW, Errington ML, French PJ, Fine A, Bliss TV, Garel S, et al. A requirement for the immediate early gene Zif268 in the expression of late LTP and long-term memories. Nat Neurosci. 2001;4:289–96.CrossRefPubMed Jones MW, Errington ML, French PJ, Fine A, Bliss TV, Garel S, et al. A requirement for the immediate early gene Zif268 in the expression of late LTP and long-term memories. Nat Neurosci. 2001;4:289–96.CrossRefPubMed
113.
go back to reference Netto CA, Sanches E, Odorcyk FK, Duran-Carabali LE, Weis SN. Sex-dependent consequences of neonatal brain hypoxia-ischemia in the rat. J Neurosci Res. 2017;95:409–21.CrossRefPubMed Netto CA, Sanches E, Odorcyk FK, Duran-Carabali LE, Weis SN. Sex-dependent consequences of neonatal brain hypoxia-ischemia in the rat. J Neurosci Res. 2017;95:409–21.CrossRefPubMed
114.
go back to reference Charriaut-Marlangue C, Besson VC, Baud O. Sexually dimorphic outcomes after neonatal stroke and hypoxia-ischemia. Int J Mol Sci. 2017;19:1. Charriaut-Marlangue C, Besson VC, Baud O. Sexually dimorphic outcomes after neonatal stroke and hypoxia-ischemia. Int J Mol Sci. 2017;19:1.
115.
go back to reference Galanopoulou AS. Dissociated gender-specific effects of recurrent seizures on GABA signaling in CA1 pyramidal neurons: role of GABAA receptors. J Neurosci. 2008;28:1557–67.CrossRefPubMedPubMedCentral Galanopoulou AS. Dissociated gender-specific effects of recurrent seizures on GABA signaling in CA1 pyramidal neurons: role of GABAA receptors. J Neurosci. 2008;28:1557–67.CrossRefPubMedPubMedCentral
116.
go back to reference Mirza MA, Ritzel R, Xu Y, McCullough LD, Liu F. Sexually dimorphic outcomes and inflammatory responses in hypoxic-ischemic encephalopathy. J Neuroinflammation. 2015;12:32.CrossRefPubMedPubMedCentral Mirza MA, Ritzel R, Xu Y, McCullough LD, Liu F. Sexually dimorphic outcomes and inflammatory responses in hypoxic-ischemic encephalopathy. J Neuroinflammation. 2015;12:32.CrossRefPubMedPubMedCentral
117.
go back to reference Hill CA, Fitch RH. Sex differences in mechanisms and outcome of neonatal hypoxia-ischemia in rodent models: implications for sex-specific neuroprotection in clinical neonatal practice. Neurol Res Int. 2012;2012:867531.CrossRefPubMedPubMedCentral Hill CA, Fitch RH. Sex differences in mechanisms and outcome of neonatal hypoxia-ischemia in rodent models: implications for sex-specific neuroprotection in clinical neonatal practice. Neurol Res Int. 2012;2012:867531.CrossRefPubMedPubMedCentral
118.
go back to reference Barkhuizen M, van Mechelen R, Vermeer M, Chedraui P, Paes D, van den Hove DLA, et al. Systemic multipotent adult progenitor cells improve long-term neurodevelopmental outcomes after preterm hypoxic-ischemic encephalopathy. Behav Brain Res. 2019;362:77–81.CrossRefPubMed Barkhuizen M, van Mechelen R, Vermeer M, Chedraui P, Paes D, van den Hove DLA, et al. Systemic multipotent adult progenitor cells improve long-term neurodevelopmental outcomes after preterm hypoxic-ischemic encephalopathy. Behav Brain Res. 2019;362:77–81.CrossRefPubMed
Metadata
Title
Potential biomarkers for neuroinflammation and neurodegeneration at short and long term after neonatal hypoxic-ischemic insult in rat
Authors
Nozha Borjini
Sandra Sivilia
Alessandro Giuliani
Mercedes Fernandez
Luciana Giardino
Fabrizio Facchinetti
Laura Calzà
Publication date
01-12-2019
Publisher
BioMed Central
Keyword
Biomarkers
Published in
Journal of Neuroinflammation / Issue 1/2019
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-019-1595-0

Other articles of this Issue 1/2019

Journal of Neuroinflammation 1/2019 Go to the issue