Am J Perinatol
DOI: 10.1055/a-2159-0488
Original Article

Neuroserpin As an Adjuvant Therapy for Hypothermia on Brain Injury in Neonatal Hypoxic–Ischemic Rats

Hasan Kilicdag
1   Division of Neonatology, Department of Pediatrics, Baskent University Faculty of Medicine, Ankara, Turkey
,
Kubra Akillioglu
2   Division of Neurophysiology, Department of Physiology, Medical Faculty, University of Cukurova, Turkey
,
Emine Kilic Bagır
3   Department of Pathology, Cukurova University, Medical Faculty, Adana, Turkey
,
Seda Kose
2   Division of Neurophysiology, Department of Physiology, Medical Faculty, University of Cukurova, Turkey
,
Seyda Erdogan
3   Department of Pathology, Cukurova University, Medical Faculty, Adana, Turkey
› Author Affiliations

Abstract

Objective We aimed to assess the effects of neuroserpin and its combination with hypothermia on hypoxic-ischemic (HI) brain injury in neonatal rats. Neuroserpin is an axon-secreted serine protease inhibitor and is important for brain development, neuronal survival, and synaptic plasticity.

Study Design Male Wistar–Albino rats on postnatal day 7 (P7) were randomly divided into five groups: sham group (n = 10), (HI; n = 10), hypoxic-ischemic hypothermia (HIH; n = 10), hypoxic-ischemic neuroserpin (HIN; n = 10), and hypoxic-ischemic neuroserpin-hypothermia (HINH; n = 10). The P7 rat brain's maturation is similar to a late preterm human brain at 34 to 36 weeks of gestation. HI was induced in rats on P7 as previously described. A single dose of 0.2 µM neuroserpin (HINH and HIN) or an equivalent volume of phosphate-buffered saline (sham, HIH, and HI) was administered intraventricularly by a Hamilton syringe immediately after hypoxia. In the follow-up, pups were subjected to systemic hypothermia or normothermia for 2 hours. Euthanasia was performed for histopathological evaluation on P10. Apoptosis was detected by caspase-3 activity and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) staining and was counted in the hippocampus.

Results In comparison to the HI group, the TUNEL-positive and caspase-3-positive neurons in the sham, HIN, HIH, and HINH groups were considerably lower (13.4 ± 1.0 vs. 1.9 ± 0.9, 6.0 ± 0.9, 5.3 ± 1.6, and 4.0 ± 1.1; p < 0.001) and (13.5 ± 1.7 vs. 1.2 ± 0.7, 9.1 ± 2.7, 4.8 ± 1.0, and 3.9 ± 1.6; p < 0.001). HIN, HIH, and HINH, compared to the sham group, showed more TUNEL-positive and caspase-3-positive neurons (6.0 ± 0.9, 5.3 ± 1.6, 4.0 ± 1.1 vs. 1.9 ± 0.9 and 9.1 ± 2.7, 4.8 ± 1.0, 3.9 ± 1.6 vs. 1.2 ± 0.7; p < 0.001). The HINH group (synergistic effect) had significantly fewer TUNEL-positive neurons and caspase-3-positive neurons than the HIN group (4.0 ± 1.1vs. 6.0 ± 0.9 and 3.9 ± 1.6 vs. 9.1 ± 2.7; p < 0.001).

Conclusion Our study showed that both neuroserpin alone and as an adjuvant treatment for hypothermia may have a neuroprotective effect on brain injury.

Key Points

  • Neuroserpin decreased brain injury.

  • Neuroserpin showed a synergistic effect when used as an adjuvant treatment for hypothermia.

  • The neuroprotective effect of neuroserpine was related to its antiapoptotic properties.



Publication History

Received: 12 May 2023

Accepted: 22 August 2023

Accepted Manuscript online:
23 August 2023

Article published online:
25 September 2023

© 2023. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Inder TE, Volpe JJ. Chapter 20—Hypoxic-ischemic injury in the term infant: clinical-neurological features, diagnosis, imaging, prognosis, therapy. In: Volpe JJ, Inder TE, Darras BT, de Vries LS, du Plessis AJ, Neil JJ. et al., eds. Volpe's Neurology of the Newborn (Sixth Edition). Elsevier; 2018: 510-563.e15
  • 2 Kurinczuk JJ, White-Koning M, Badawi N. Epidemiology of neonatal encephalopathy and hypoxic-ischaemic encephalopathy. Early Hum Dev 2010; 86 (06) 329-338
  • 3 Lawn JE, Osrin D, Adler A, Cousens S. Four million neonatal deaths: counting and attribution of cause of death. Paediatr Perinat Epidemiol 2008; 22 (05) 410-416
  • 4 Polglase GR, Ong T, Hillman NH. Cardiovascular alterations and multiorgan dysfunction after birth asphyxia. Clin Perinatol 2016; 43 (03) 469-483
  • 5 Jacobs SE, Berg M, Hunt R, Tarnow-Mordi WO, Inder TE, Davis PG. Cooling for newborns with hypoxic ischaemic encephalopathy. Cochrane Database Syst Rev 2013; 2013 (01) CD003311
  • 6 Thoresen M. Who should we cool after perinatal asphyxia?. Semin Fetal Neonatal Med 2015; 20 (02) 66-71
  • 7 Shankaran S, Pappas A, McDonald SA. et al; Eunice Kennedy Shriver NICHD Neonatal Research Network. Childhood outcomes after hypothermia for neonatal encephalopathy. N Engl J Med 2012; 366 (22) 2085-2092 Erratum in: N Engl J Med. 2012;367(11):1073
  • 8 Shankaran S, Laptook AR, Pappas A. et al; Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network. Effect of depth and duration of cooling on death or disability at age 18 months among neonates with hypoxic-ischemic encephalopathy: a randomized clinical trial. JAMA 2017; 318 (01) 57-67
  • 9 Osterwalder T, Contartese J, Stoeckli ET, Kuhn TB, Sonderegger P. Neuroserpin, an axonally secreted serine protease inhibitor. EMBO J 1996; 15 (12) 2944-2953
  • 10 Gelderblom M, Neumann M, Ludewig P. et al. Deficiency in serine protease inhibitor neuroserpin exacerbates ischemic brain injury by increased postischemic inflammation. PLoS One 2013; 8 (05) e63118
  • 11 Lee TW, Tsang VWK, Loef EJ, Birch NP. Physiological and pathological functions of neuroserpin: regulation of cellular responses through multiple mechanisms. Semin Cell Dev Biol 2017; 62: 152-159
  • 12 Wu J, Echeverry R, Guzman J, Yepes M. Neuroserpin protects neurons from ischemia-induced plasmin-mediated cell death independently of tissue-type plasminogen activator inhibition. Am J Pathol 2010; 177 (05) 2576-2584
  • 13 Krueger SR, Ghisu GP, Cinelli P. et al. Expression of neuroserpin, an inhibitor of tissue plasminogen activator, in the developing and adult nervous system of the mouse. J Neurosci 1997; 17 (23) 8984-8996
  • 14 Cinelli P, Madani R, Tsuzuki N. et al. Neuroserpin, a neuroprotective factor in focal ischemic stroke. Mol Cell Neurosci 2001; 18 (05) 443-457
  • 15 Yepes M, Sandkvist M, Wong MK. et al. Neuroserpin reduces cerebral infarct volume and protects neurons from ischemia-induced apoptosis. Blood 2000; 96 (02) 569-576
  • 16 Zhang Z, Zhang L, Yepes M. et al. Adjuvant treatment with neuroserpin increases the therapeutic window for tissue-type plasminogen activator administration in a rat model of embolic stroke. Circulation 2002; 106 (06) 740-745
  • 17 Millar LJ, Shi L, Hoerder-Suabedissen A, Molnár Z. Neonatal hypoxia ischaemia: mechanisms, models, and therapeutic challenges. Front Cell Neurosci 2017; 11: 78
  • 18 Burd I, Welling J, Kannan G, Johnston MV. Excitotoxicity as a common mechanism for fetal neuronal injury with hypoxia and intrauterine inflammation. Adv Pharmacol 2016; 76: 85-101
  • 19 Chamorro Á, Dirnagl U, Urra X, Planas AM. Neuroprotection in acute stroke: targeting excitotoxicity, oxidative and nitrosative stress, and inflammation. Lancet Neurol 2016; 15 (08) 869-881
  • 20 Lebeurrier N, Liot G, Lopez-Atalaya JP. et al. The brain-specific tissue-type plasminogen activator inhibitor, neuroserpin, protects neurons against excitotoxicity both in vitro and in vivo. Mol Cell Neurosci 2005; 30 (04) 552-558
  • 21 Cheng Y, Loh YP, Birch NP. Neuroserpin attenuates H2O2-induced oxidative stress in hippocampal neurons via AKT and BCL-2 signaling pathways. J Mol Neurosci 2017; 61 (01) 123-131
  • 22 Mohsenifar A, Lotfi AS, Ranjbar B. et al. A study of the oxidation-induced conformational and functional changes in neuroserpin. Iran Biomed J 2007; 11 (01) 41-46
  • 23 Munuswamy-Ramanujam G, Dai E, Liu L. et al. Neuroserpin, a thrombolytic serine protease inhibitor (serpin), blocks transplant vasculopathy with associated modification of T-helper cell subsets. Thromb Haemost 2010; 103 (03) 545-555
  • 24 Fan X, Kavelaars A, Heijnen CJ, Groenendaal F, van Bel F. Pharmacological neuroprotection after perinatal hypoxic-ischemic brain injury. Curr Neuropharmacol 2010; 8 (04) 324-334
  • 25 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
  • 26 Rice III JE, Vannucci RC, Brierley JB. The influence of immaturity on hypoxic-ischemic brain damage in the rat. Ann Neurol 1981; 9 (02) 131-141
  • 27 Gunn AJ, Thoresen M. Animal studies of neonatal hypothermic neuroprotection have translated well in to practice. Resuscitation 2015; 97: 88-90
  • 28 Jacobs SE, Morley CJ, Inder TE. et al; Infant Cooling Evaluation Collaboration. Whole-body hypothermia for term and near-term newborns with hypoxic-ischemic encephalopathy: a randomized controlled trial. Arch Pediatr Adolesc Med 2011; 165 (08) 692-700
  • 29 Tucker AM, Aquilina K, Chakkarapani E, Hobbs CE, Thoresen M. Development of amplitude-integrated electroencephalography and interburst interval in the rat. Pediatr Res 2009; 65 (01) 62-66
  • 30 Hagberg H, Ichord R, Palmer C, Yager JY, Vannucci SJ. Animal models of developmental brain injury: relevance to human disease. A summary of the panel discussion from the Third Hershey Conference on Developmental Cerebral Blood Flow and Metabolism. Dev Neurosci 2002; 24 (05) 364-366
  • 31 Nie X, Lowe DW, Rollins LG. et al. Sex-specific effects of N-acetylcysteine in neonatal rats treated with hypothermia after severe hypoxia-ischemia. Neurosci Res 2016; 108: 24-33
  • 32 Gunn AJ, Bennet L. Fetal hypoxia insults and patterns of brain injury: insights from animal models. Clin Perinatol 2009; 36 (03) 579-593
  • 33 Davidson JO, Wassink G, van den Heuij LG, Bennet L, Gunn AJ. Therapeutic Hypothermia for Neonatal Hypoxic-Ischemic Encephalopathy - Where to from Here?. Front Neurol 2015; 6: 198
  • 34 Jänicke RU, Sprengart ML, Wati MR, Porter AG. Caspase-3 is required for DNA fragmentation and morphological changes associated with apoptosis. J Biol Chem 1998; 273 (16) 9357-9360
  • 35 Rodríguez-Fanjul J, Durán Fernández-Feijóo C, Lopez-Abad M. et al. Neuroprotection with hypothermia and allopurinol in an animal model of hypoxic-ischemic injury: is it a gender question?. PLoS One 2017; 12 (09) e0184643
  • 36 Charriaut-Marlangue C, Besson VC, Baud O. Sexually dimorphic outcomes after neonatal stroke and hypoxia-ischemia. Int J Mol Sci 2017; 19 (01) 61
  • 37 Bona E, Hagberg H, Løberg EM, Bågenholm R, Thoresen M. Protective effects of moderate hypothermia after neonatal hypoxia-ischemia: short- and long-term outcome. Pediatr Res 1998; 43 (06) 738-745
  • 38 Fan X, van Bel F, van der Kooij MA, Heijnen CJ, Groenendaal F. Hypothermia and erythropoietin for neuroprotection after neonatal brain damage. Pediatr Res 2013; 73 (01) 18-23
  • 39 Wang L, Zhang Y, Asakawa T. et al. Neuroprotective effect of neuroserpin in oxygen-glucose deprivation- and reoxygenation-treated rat astrocytes in vitro. PLoS One 2015; 10 (04) e0123932
  • 40 Patel SD, Pierce L, Ciardiello AJ, Vannucci SJ. Neonatal encephalopathy: pre-clinical studies in neuroprotection. Biochem Soc Trans 2014; 42 (02) 564-568
  • 41 Romijn HJ, Hofman MA, Gramsbergen A. At what age is the developing cerebral cortex of the rat comparable to that of the full-term newborn human baby?. Early Hum Dev 1991; 26 (01) 61-67
  • 42 Patel SD, Pierce L, Ciardiello A. et al. Therapeutic hypothermia and hypoxia-ischemia in the term-equivalent neonatal rat: characterization of a translational preclinical model. Pediatr Res 2015; 78 (03) 264-271
  • 43 Gonzalez FF, Larpthaveesarp A, McQuillen P. et al. Erythropoietin increases neurogenesis and oligodendrogliosis of subventricular zone precursor cells after neonatal stroke. Stroke 2013; 44 (03) 753-758
  • 44 Osredkar D, Sall JW, Bickler PE, Ferriero DM. Erythropoietin promotes hippocampal neurogenesis in in vitro models of neonatal stroke. Neurobiol Dis 2010; 38 (02) 259-265
  • 45 Wu YW, Comstock BA, Gonzalez FF. et al; HEAL Consortium. Trial of erythropoietin for hypoxic-ischemic encephalopathy in newborns. N Engl J Med 2022; 387 (02) 148-159