Neuropediatrics 2022; 53(06): 402-417
DOI: 10.1055/s-0042-1755235
Review Article

Neonatal Hypoxic–Ischemic Encephalopathy: Perspectives of Neuroprotective and Neuroregenerative Treatments

1   Departamento de Fisiología y Farmacología, Laboratorio de Neurofisiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
,
Denisse Calderón-Vallejo
1   Departamento de Fisiología y Farmacología, Laboratorio de Neurofisiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
2   Departamento de Morfología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
,
J. Luis Quintanar
1   Departamento de Fisiología y Farmacología, Laboratorio de Neurofisiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
› Author Affiliations
Funding This research did not receive any grant from funding agencies in the public, commercial, or not-for-profit sectors.

Abstract

Hypoxic–ischemic encephalopathy (HIE) is a serious condition that could have deleterious neurological outcomes, such as cerebral palsy, neuromotor disability, developmental disability, epilepsy, and sensitive or cognitive problems, and increase the risk of death in severe cases. Once HIE occurs, molecular cascades are triggered favoring the oxidative stress, excitotoxicity, and inflammation damage that promote cell death via apoptosis or necrosis. Currently, the therapeutic hypothermia is the standard of care in HIE; however, it has a small window of action and only can be used in children of more than 36 gestational weeks; for this reason, it is very important to develop new therapies to prevent the progression of the hypoxic–ischemic injury or to develop neuroregenerative therapies in severe HIE cases. The objective of this revision is to describe the emerging treatments for HIE, either preventing cell death for oxidative stress, excitotoxicity, or exacerbated inflammation, as well as describing a new therapeutic approach for neuroregeneration, such as mesenchymal stem cells, brain-derived neurotrophic factor, and gonadotropin realizing hormone agonists.

Author Contributions

All authors have participated in drafting this manuscript and have approved the final version submitted for publication.


Note

All figures were created with BioRender.com.




Publication History

Received: 24 January 2022

Accepted: 21 June 2022

Article published online:
28 August 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Kurinczuk JJ, White-Koning M, Badawi N. Epidemiology of neonatal encephalopathy and hypoxic-ischaemic encephalopathy. Early Hum Dev 2010; 86 (06) 329-338
  • 2 Millar LJ, Shi L, Hoerder-Suabedissen A, Molnár Z. Neonatal hypoxia ischaemia: mechanisms, models, and therapeutic challenges. Front Cell Neurosci 2017; 11: 78
  • 3 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
  • 4 Montaldo P, Pauliah SS, Lally PJ, Olson L, Thayyil S. Cooling in a low-resource environment: lost in translation. Semin Fetal Neonatal Med 2015; 20 (02) 72-79
  • 5 Choi DW. Calcium-mediated neurotoxicity: relationship to specific channel types and role in ischemic damage. Trends Neurosci 1988; 11 (10) 465-469
  • 6 Drury PP, Gunn ER, Bennet L, Gunn AJ. Mechanisms of hypothermic neuroprotection. Clin Perinatol 2014; 41 (01) 161-175
  • 7 Kaur C, Rathnasamy G, Ling EA. Roles of activated microglia in hypoxia induced neuroinflammation in the developing brain and the retina. J Neuroimmune Pharmacol 2013; 8 (01) 66-78
  • 8 Weinstein JR, Koerner IP, Möller T. Microglia in ischemic brain injury. Future Neurol 2010; 5 (02) 227-246
  • 9 Zhao M, Zhu P, Fujino M. et al. Oxidative stress in hypoxic-ischemic encephalopathy: Molecular mechanisms and therapeutic strategies. Int J Mol Sci 2016; 17 (12) 2078
  • 10 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 (05) 1067-1076
  • 11 Palmer C, Vannucci RC, Towfighi J. Reduction of perinatal hypoxic-ischemic brain damage with allopurinol. Pediatr Res 1990; 27 (4, pt. 1): 332-336
  • 12 Perlman JM. Intervention strategies for neonatal hypoxic-ischemic cerebral injury. Clin Ther 2006; 28 (09) 1353-1365
  • 13 Arteaga O, Álvarez A, Revuelta M, Santaolalla F, Urtasun A, Hilario E. Role of antioxidants in neonatal hypoxic–ischemic brain injury: new therapeutic approaches. Int J Mol Sci 2017; 18 (02) 265
  • 14 Liu N, Xu H, Sun Q. et al. The role of oxidative stress in hyperuricemia and xanthine oxidoreductase (XOR) inhibitors. Oxid Med Cell Longev 2021; 2021: 1470380
  • 15 Shadid M, Buonocore G, Groenendaal F. et al. Effect of deferoxamine and allopurinol on non-protein-bound iron concentrations in plasma and cortical brain tissue of newborn lambs following hypoxia-ischemia. Neurosci Lett 1998; 248 (01) 5-8
  • 16 Vázquez-Mellado J, Morales EM, Pacheco-Tena C, Burgos-Vargas R. Relation between adverse events associated with allopurinol and renal function in patients with gout. Ann Rheum Dis 2001; 60 (10) 981-983
  • 17 Palmer C, Towfighi J, Roberts RL, Heitjan DF. Allopurinol administered after inducing hypoxia-ischemia reduces brain injury in 7-day-old rats. Pediatr Res 1993; 33 (4, pt. 1): 405-411
  • 18 Peeters-Scholte C, Braun K, Koster J. et al. Effects of allopurinol and deferoxamine on reperfusion injury of the brain in newborn piglets after neonatal hypoxia-ischemia. Pediatr Res 2003; 54 (04) 516-522
  • 19 Marro PJ, McGowan JE, Razdan B, Mishra OP, Delivoria-Papadopoulos M. Effect of allopurinol on uric acid levels and brain cell membrane Na+, K(+)-ATPase activity during hypoxia in newborn piglets. Brain Res 1994; 650 (01) 9-15
  • 20 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
  • 21 Gunes T, Ozturk MA, Koklu E, Kose K, Gunes I. Effect of allopurinol supplementation on nitric oxide levels in asphyxiated newborns. Pediatr Neurol 2007; 36 (01) 17-24
  • 22 Kaandorp JJ, van Bel F, Veen S. et al. Long-term neuroprotective effects of allopurinol after moderate perinatal asphyxia: follow-up of two randomised controlled trials. Arch Dis Child Fetal Neonatal Ed 2012; 97 (03) F162-F166
  • 23 Chaudhari T, McGuire W. Allopurinol for preventing mortality and morbidity in newborn infants with hypoxic-ischaemic encephalopathy. Cochrane Database Syst Rev 2012; (07) CD006817
  • 24 Maiwald CA, Annink KV, Rüdiger M. et al; ALBINO Study Group. Effect of allopurinol in addition to hypothermia treatment in neonates for hypoxic-ischemic brain injury on neurocognitive outcome (ALBINO): study protocol of a blinded randomized placebo-controlled parallel group multicenter trial for superiority (phase III). BMC Pediatr 2019; 19 (01) 210
  • 25 Chu WY, Annink KV, Nijstad AL. et al; ALBINO Study Group. Pharmacokinetic/pharmacodynamic modelling of allopurinol, its active metabolite oxypurinol, and biomarkers hypoxanthine, xanthine and uric acid in hypoxic-ischemic encephalopathy neonates. Clin Pharmacokinet 2022; 61 (02) 321-333
  • 26 Van Bel F, Shadid M, Moison RMW. et al. Effect of allopurinol on postasphyxial free radical formation, cerebral hemodynamics, and electrical brain activity. Pediatrics 1998; 101 (02) 185-193
  • 27 Benders MJ, Bos AF, Rademaker CM. et al. Early postnatal allopurinol does not improve short term outcome after severe birth asphyxia. Arch Dis Child Fetal Neonatal Ed 2006; 91 (03) F163-F165
  • 28 Boda D. Results of and further prevention of hypoxic fetal brain damage by inhibition of xanthine oxidase enzyme with allopurinol. J Perinat Med 2011; 39 (04) 441-444
  • 29 Abe H. Role of histidine-related compounds as intracellular proton buffering constituents in vertebrate muscle. Biochemistry (Mosc) 2000; 65 (07) 757-765
  • 30 Bellia F, Vecchio G, Cuzzocrea S, Calabrese V, Rizzarelli E. Neuroprotective features of carnosine in oxidative driven diseases. Mol Aspects Med 2011; 32 (4-6): 258-266
  • 31 Hipkiss AR. Carnosine and its possible roles in nutrition and health. Adv Food Nutr Res 2009; 57: 87-154
  • 32 Zhang X, Song L, Cheng X. et al. Carnosine pretreatment protects against hypoxia-ischemia brain damage in the neonatal rat model. Eur J Pharmacol 2011; 667 (1–3): 202-207
  • 33 Zhang H, Guo S, Zhang L. et al. Treatment with carnosine reduces hypoxia-ischemia brain damage in a neonatal rat model. Eur J Pharmacol 2014; 727 (01) 174-180
  • 34 Ouyang L, Tian Y, Bao Y. et al. Carnosine decreased neuronal cell death through targeting glutamate system and astrocyte mitochondrial bioenergetics in cultured neuron/astrocyte exposed to OGD/recovery. Brain Res Bull 2016; 124: 76-84
  • 35 Rajanikant GK, Zemke D, Senut MC. et al. Carnosine is neuroprotective against permanent focal cerebral ischemia in mice. Stroke 2007; 38 (11) 3023-3031
  • 36 Byun JC, Lee SR, Kim CS. Effects of carnosine and hypothermia combination therapy on hypoxic-ischemic brain injury in neonatal rats. Clin Exp Pediatr 2021; 64 (08) 422-428
  • 37 Ma J, Bo SH, Lu XT, Xu AJ, Zhang J. Protective effects of carnosine on white matter damage induced by chronic cerebral hypoperfusion. Neural Regen Res 2016; 11 (09) 1438-1444
  • 38 Keating GM. Dexmedetomidine: a review of its use for sedation in the intensive care setting. Drugs 2015; 75 (10) 1119-1130
  • 39 Gao Y, Zhang Y, Dong Y, Wu X, Liu H. Dexmedetomidine mediates neuroglobin up-regulation and alleviates the hypoxia/reoxygenation injury by inhibiting neuronal apoptosis in developing rats. Front Pharmacol 2020; 11: 555532
  • 40 Ren X, Ma H, Zuo Z. Dexmedetomidine postconditioning reduces brain injury after brain hypoxia-ischemia in neonatal rats. J Neuroimmune Pharmacol 2016; 11 (02) 238-247
  • 41 Paris A, Mantz J, Tonner PH, Hein L, Brede M, Gressens P. The effects of dexmedetomidine on perinatal excitotoxic brain injury are mediated by the α2A-adrenoceptor subtype. Anesth Analg 2006; 102 (02) 456-461
  • 42 McAdams RM, Pak D, Lalovic B, Phillips B, Shen DD. Dexmedetomidine pharmacokinetics in neonates with hypoxic-ischemic encephalopathy receiving hypothermia. Anesthesiol Res Pract 2020; 2020: 2582965
  • 43 Rossert J, Eckardt KU. Erythropoietin receptors: their role beyond erythropoiesis. Nephrol Dial Transplant 2005; 20 (06) 1025-1028
  • 44 Brines ML, Ghezzi P, Keenan S. et al. Erythropoietin crosses the blood-brain barrier to protect against experimental brain injury. Proc Natl Acad Sci U S A 2000; 97 (19) 10526-10531
  • 45 Kumral A, Genc S, Ozer E. et al. Erythropoietin downregulates bax and DP5 proapoptotic gene expression in neonatal hypoxic-ischemic brain injury. Biol Neonate 2006; 89 (03) 205-210
  • 46 Elmahdy H, El-Mashad AR, El-Bahrawy H, El-Gohary T, El-Barbary A, Aly H. Human recombinant erythropoietin in asphyxia neonatorum: pilot trial. Pediatrics 2010; 125 (05) e1135-e1142
  • 47 Digicaylioglu M, Lipton SA. Erythropoietin-mediated neuroprotection involves cross-talk between Jak2 and NF-kappaB signalling cascades. Nature 2001; 412 (6847): 641-647
  • 48 Kumral A, Gonenc S, Acikgoz O. et al. Erythropoietin increases glutathione peroxidase enzyme activity and decreases lipid peroxidation levels in hypoxic-ischemic brain injury in neonatal rats. Biol Neonate 2005; 87 (01) 15-18
  • 49 Montero M, Poulsen FR, Noraberg J. et al. Comparison of neuroprotective effects of erythropoietin (EPO) and carbamylerythropoietin (CEPO) against ischemia-like oxygen-glucose deprivation (OGD) and NMDA excitotoxicity in mouse hippocampal slice cultures. Exp Neurol 2007; 204 (01) 106-117
  • 50 Sun Y, Calvert JW, Zhang JH. Neonatal hypoxia/ischemia is associated with decreased inflammatory mediators after erythropoietin administration. Stroke 2005; 36 (08) 1672-1678
  • 51 Xiong T, Yang X, Qu Y. et al. Erythropoietin induces synaptogenesis and neurite repair after hypoxia ischemia-mediated brain injury in neonatal rats. Neuroreport 2019; 30 (11) 783-789
  • 52 Malla RR, Asimi R, Teli MA, Shaheen F, Bhat MA. Erythropoietin monotherapy in perinatal asphyxia with moderate to severe encephalopathy: a randomized placebo-controlled trial. J Perinatol 2017; 37 (05) 596-601
  • 53 Rogers EE, Bonifacio SL, Glass HC. et al. Erythropoietin and hypothermia for hypoxic-ischemic encephalopathy. Pediatr Neurol 2014; 51 (05) 657-662
  • 54 Wu YW, Mathur AM, Chang T. et al. High-dose erythropoietin and hypothermia for hypoxic-Ischemic encephalopathy: a phase II trial. Pediatrics 2016; 137 (06) e20160191
  • 55 El Shimi MS, Awad HA, Hassanein SMA. et al. Single dose recombinant erythropoietin versus moderate hypothermia for neonatal hypoxic ischemic encephalopathy in low resource settings. J Matern Fetal Neonatal Med 2014; 27 (13) 1295-1300
  • 56 Wu YW, Bauer LA, Ballard RA. et al. Erythropoietin for neuroprotection in neonatal encephalopathy: safety and pharmacokinetics. Pediatrics 2012; 130 (04) 683-691
  • 57 Zhu C, Kang W, Xu F. et al. Erythropoietin improved neurologic outcomes in newborns with hypoxic-ischemic encephalopathy. Pediatrics 2009; 124 (02) e218-e226
  • 58 Schoener B, Borger J. Erythropoietin stimulating agents. In: StatPearls. Treasure Island, FL: StatPearls Publishing; 2022
  • 59 Benders MJ, van der Aa NE, Roks M. et al. Feasibility and safety of erythropoietin for neuroprotection after perinatal arterial ischemic stroke. J Pediatr 2014; 164 (03) 481-6.e1 , 2
  • 60 Chen YC, Tain YL, Sheen JM, Huang LT. Melatonin utility in neonates and children. J Formos Med Assoc 2012; 111 (02) 57-66
  • 61 Reiter R, Tan D. et al. Melatonin salvages neural tissue from ischemia/reperfusion injury. Open Neuroendocrinol J 2010; 3: 112-120
  • 62 Liu L, Chen H, Jin J. et al. Melatonin ameliorates cerebral ischemia/reperfusion injury through SIRT3 activation. Life Sci 2019; 239: 117036
  • 63 Manev H, Uz T, Kharlamov A, Joo JY. Increased brain damage after stroke or excitotoxic seizures in melatonin-deficient rats. FASEB J 1996; 10 (13) 1546-1551
  • 64 Cardinali DP. An assessment of melatonin's therapeutic value in the hypoxic-ischemic encephalopathy of the newborn. Front Synaptic Neurosci 2019; 11: 34
  • 65 Venegas C, García JA, Escames G. et al. Extrapineal melatonin: analysis of its subcellular distribution and daily fluctuations. J Pineal Res 2012; 52 (02) 217-227
  • 66 Cecon E, Oishi A, Jockers R. Melatonin receptors: molecular pharmacology and signalling in the context of system bias. Br J Pharmacol 2018; 175 (16) 3263-3280
  • 67 Okatani Y, Wakatsuki A, Shinohara K, Taniguchi K, Fukaya T. Melatonin protects against oxidative mitochondrial damage induced in rat placenta by ischemia and reperfusion. J Pineal Res 2001; 31 (02) 173-178
  • 68 Alonso-Alconada D, Álvarez A, Lacalle J, Hilario E. Histological study of the protective effect of melatonin on neural cells after neonatal hypoxia-ischemia. Histol Histopathol 2012; 27 (06) 771-783
  • 69 Reiter RJ, Tan DX, Burkhardt S. Reactive oxygen and nitrogen species and cellular and organismal decline: amelioration with melatonin. Mech Ageing Dev 2002; 123 (08) 1007-1019
  • 70 Fischer TW, Kleszczyński K, Hardkop LH, Kruse N, Zillikens D. Melatonin enhances antioxidative enzyme gene expression (CAT, GPx, SOD), prevents their UVR-induced depletion, and protects against the formation of DNA damage (8-hydroxy-2′-deoxyguanosine) in ex vivo human skin. J Pineal Res 2013; 54 (03) 303-312
  • 71 Balduini W, Carloni S, Perrone S. et al. The use of melatonin in hypoxic-ischemic brain damage: an experimental study. J Matern Fetal Neonatal Med 2012; 25 (Suppl. 01) 119-124
  • 72 Blanco S, Hernández R, Franchelli G, Ramos-Álvarez MM, Peinado MÁ. Melatonin influences NO/NOS pathway and reduces oxidative and nitrosative stress in a model of hypoxic-ischemic brain damage. Nitric Oxide 2017; 62: 32-43
  • 73 Wang X. The antiapoptotic activity of melatonin in neurodegenerative diseases. CNS Neurosci Ther 2009; 15 (04) 345-357
  • 74 Hu Y, Wang Z, Liu Y. et al. Melatonin reduces hypoxic-ischaemic (HI) induced autophagy and apoptosis: An in vivo and in vitro investigation in experimental models of neonatal HI brain injury. Neurosci Lett 2017; 653: 105-112
  • 75 Merchant NM, Azzopardi DV, Hawwa AF. et al. Pharmacokinetics of melatonin in preterm infants. Br J Clin Pharmacol 2013; 76 (05) 725-733
  • 76 Carloni S, Proietti F, Rocchi M. et al. Melatonin pharmacokinetics following oral administration in preterm neonates. Molecules 2017; 22 (12) 2115
  • 77 Aly H, Elmahdy H, El-Dib M. et al. Melatonin use for neuroprotection in perinatal asphyxia: a randomized controlled pilot study. J Perinatol 2015; 35 (03) 186-191
  • 78 Ahmad QM, Chishti AL, Waseem N. Role of melatonin in management of hypoxic ischaemic encephalopathy in newborns: a randomized control trial. J Pak Med Assoc 2018; 68 (08) 1233-1237
  • 79 Balduini W, Weiss MD, Carloni S. et al. Melatonin pharmacokinetics and dose extrapolation after enteral infusion in neonates subjected to hypothermia. J Pineal Res 2019; 66 (04) e12565
  • 80 Chen HSV, Wang YF, Rayudu PV. et al. Neuroprotective concentrations of the N-methyl-D-aspartate open-channel blocker memantine are effective without cytoplasmic vacuolation following post-ischemic administration and do not block maze learning or long-term potentiation. Neuroscience 1998; 86 (04) 1121-1132
  • 81 Chen HSV, Lipton SA. The chemical biology of clinically tolerated NMDA receptor antagonists. J Neurochem 2006; 97 (06) 1611-1626
  • 82 Volbracht C, van Beek J, Zhu C, Blomgren K, Leist M. Neuroprotective properties of memantine in different in vitro and in vivo models of excitotoxicity. Eur J Neurosci 2006; 23 (10) 2611-2622
  • 83 Chen HS, Pellegrini JW, Aggarwal SK. et al. Open-channel block of N-methyl-D-aspartate (NMDA) responses by memantine: therapeutic advantage against NMDA receptor-mediated neurotoxicity. J Neurosci 1992; 12 (11) 4427-4436
  • 84 Manning SM, Talos DM, Zhou C. et al. NMDA receptor blockade with memantine attenuates white matter injury in a rat model of periventricular leukomalacia. J Neurosci 2008; 28 (26) 6670-6678
  • 85 Liu C, Lin N, Wu B, Qiu Y. Neuroprotective effect of memantine combined with topiramate in hypoxic-ischemic brain injury. Brain Res 2009; 1282: 173-182
  • 86 Landucci E, Filippi L, Gerace E, Catarzi S, Guerrini R, Pellegrini-Giampietro DE. Neuroprotective effects of topiramate and memantine in combination with hypothermia in hypoxic-ischemic brain injury in vitro and in vivo. Neurosci Lett 2018; 668: 103-107
  • 87 National Library of Medicine. PubChem. Nitric oxide. Accessed April 12, 2022 at: https://pubchem.ncbi.nlm.nih.gov/compound/Nitric-oxide
  • 88 Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases: structure, function and inhibition. Biochem J 2001; 357 (Pt 3): 593-615
  • 89 Ichinose F, Roberts Jr. JD, Zapol WM. Inhaled nitric oxide: a selective pulmonary vasodilator: current uses and therapeutic potential. Circulation 2004; 109 (25) 3106-3111
  • 90 Angelis D, Savani R, Chalak L. Nitric oxide and the brain. Part 1: Mechanisms of regulation, transport and effects on the developing brain. Pediatr Res 2021; 89 (04) 738-745
  • 91 Charriaut-Marlangue C, Bonnin P, Gharib A. et al. Inhaled nitric oxide reduces brain damage by collateral recruitment in a neonatal stroke model. Stroke 2012; 43 (11) 3078-3084
  • 92 Joriot-Chekaf S, Sfeir R, Riou Y. et al. Evaluation of inhaled. NO in a model of rat neonate brain injury caused by hypoxia-ischaemia. Injury 2010; 41 (05) 517-521
  • 93 Zhu C, Sun Y, Gao J, Wang X, Plesnila N, Blomgren K. Inhaled nitric oxide protects males but not females from neonatal mouse hypoxia-ischemia brain injury. Transl Stroke Res 2013; 4 (02) 201-207
  • 94 Fukuda S, Nabetani M, Goldman RD. et al. Experience of cases with inhaled nitric oxide and therapeutic hypothermia. Pediatr Int (Roma) 2022; 64 (01) e14901
  • 95 Fernández AP, Alonso D, Lisazoaín I. et al. Postnatal changes in the nitric oxide system of the rat cerebral cortex after hypoxia during delivery. Brain Res Dev Brain Res 2003; 142 (02) 177-192
  • 96 Favié LMA, Peeters-Scholte CMPCD, Bakker A. et al. Pharmacokinetics and short-term safety of the selective NOS inhibitor 2-iminobiotin in asphyxiated neonates treated with therapeutic hypothermia. Pediatr Res 2020; 87 (04) 689-696
  • 97 Bjorkman ST, Ireland Z, Fan X. et al. Short-term dose-response characteristics of 2-iminobiotin immediately postinsult in the neonatal piglet after hypoxia-ischemia. Stroke 2013; 44 (03) 809-811
  • 98 Donovan MD, Griffin BT, Kharoshankaya L, Cryan JF, Boylan GB. Pharmacotherapy for neonatal seizures: current knowledge and future perspectives. Drugs 2016; 76 (06) 647-661
  • 99 Cilio MR, Ferriero DM. Synergistic neuroprotective therapies with hypothermia. Semin Fetal Neonatal Med 2010; 15 (05) 293-298
  • 100 Walia KS, Khan EA, Ko DH, Raza SS, Khan YN. Side effects of antiepileptics–a review. Pain Pract 2004; 4 (03) 194-203
  • 101 Noh MR, Kim SK, Sun W. et al. Neuroprotective effect of topiramate on hypoxic ischemic brain injury in neonatal rats. Exp Neurol 2006; 201 (02) 470-478
  • 102 Follett PL, Deng W, Dai W. et al. Glutamate receptor-mediated oligodendrocyte toxicity in periventricular leukomalacia: a protective role for topiramate. J Neurosci 2004; 24 (18) 4412-4420
  • 103 Jiang H, Lei JJ, Zhang YH. Protective effect of topiramate on hypoxic-ischemic brain injury in neonatal rat. Asian Pac J Trop Med 2014; 7 (06) 496-500
  • 104 Liu Y, Barks JD, Xu G, Silverstein FS. Topiramate extends the therapeutic window for hypothermia-mediated neuroprotection after stroke in neonatal rats. Stroke 2004; 35 (06) 1460-1465
  • 105 Filippi L, la Marca G, Fiorini P. et al. Topiramate concentrations in neonates treated with prolonged whole body hypothermia for hypoxic ischemic encephalopathy. Epilepsia 2009; 50 (11) 2355-2361
  • 106 Filippi L, Poggi C, la Marca G. et al. Oral topiramate in neonates with hypoxic ischemic encephalopathy treated with hypothermia: a safety study. J Pediatr 2010; 157 (03) 361-366
  • 107 Filippi L, Fiorini P, Catarzi S. et al. Safety and efficacy of topiramate in neonates with hypoxic ischemic encephalopathy treated with hypothermia (NeoNATI): a feasibility study. J Matern Fetal Neonatal Med 2018; 31 (08) 973-980
  • 108 Nuñez-Ramiro A, Benavente-Fernández I, Valverde E. et al; on behalf of the Hypotop Study Group. Topiramate plus cooling for hypoxic-ischemic encephalopathy: a randomized, controlled, multicenter, double-blinded trial. Neonatology 2019; 116 (01) 76-84
  • 109 Patsalos PN, Berry DJ, Bourgeois BFD. et al. Antiepileptic drugs–best practice guidelines for therapeutic drug monitoring: a position paper by the subcommission on therapeutic drug monitoring, ILAE Commission on Therapeutic Strategies. Epilepsia 2008; 49 (07) 1239-1276
  • 110 Marques MR, Garcia-Robles A, Usach I. et al. Topiramate pharmacokinetics in neonates undergoing therapeutic hypothermia and proposal of an optimised dosing schedule. Acta Paediatr 2020; 109 (02) 300-308
  • 111 Harris PD, Barnes R. The uses of helium and xenon in current clinical practice. Anaesthesia 2008; 63 (03) 284-293
  • 112 Dinse A, Föhr KJ, Georgieff M, Beyer C, Bulling A, Weigt HU. Xenon reduces glutamate-, AMPA-, and kainate-induced membrane currents in cortical neurones. Br J Anaesth 2005; 94 (04) 479-485
  • 113 Heurteaux C, Guy N, Laigle C. et al. TREK-1, a K+ channel involved in neuroprotection and general anesthesia. EMBO J 2004; 23 (13) 2684-2695
  • 114 Sun XL, Hu G. ATP-sensitive potassium channels: a promising target for protecting neurovascular unit function in stroke. Clin Exp Pharmacol Physiol 2010; 37 (02) 243-252
  • 115 Amer AR, Oorschot DE. Xenon combined with hypothermia in perinatal hypoxic-ischemic encephalopathy: a noble gas, a noble mission. Pediatr Neurol 2018; 84: 5-10
  • 116 de Rossi LW, Brueckmann M, Rex S, Barderschneider M, Buhre W, Rossaint R. Xenon and isoflurane differentially modulate lipopolysaccharide-induced activation of the nuclear transcription factor KB and production of tumor necrosis factor-α and interleukin-6 in monocytes. Anesth Analg 2004; 98 (04) 1007-1012
  • 117 Maze M, Laitio T. Neuroprotective properties of xenon. Mol Neurobiol 2020; 57 (01) 118-124
  • 118 Dingley J, Tooley J, Porter H, Thoresen M. Xenon provides short-term neuroprotection in neonatal rats when administered after hypoxia-ischemia. Stroke 2006; 37 (02) 501-506
  • 119 Hobbs C, Thoresen M, Tucker A, Aquilina K, Chakkarapani E, Dingley J. Xenon and hypothermia combine additively, offering long-term functional and histopathologic neuroprotection after neonatal hypoxia/ischemia. Stroke 2008; 39 (04) 1307-1313
  • 120 Yang T, Zhuang L, Rei Fidalgo AM. et al. Xenon and sevoflurane provide analgesia during labor and fetal brain protection in a perinatal rat model of hypoxia-ischemia. PLoS One 2012; 7 (05) e37020
  • 121 Yin X, Zhao J, Jiang H. et al. Impact of xenon on CLIC4 and Bcl-2 expression in lipopolysaccharide and hypoxia-ischemia-induced periventricular white matter damage. Neonatology 2018; 113 (04) 339-346
  • 122 Sabir H, Osredkar D, Maes E, Wood T, Thoresen M. Xenon combined with therapeutic hypothermia is not neuroprotective after severe hypoxia-ischemia in neonatal rats. PLoS One 2016; 11 (06) e0156759
  • 123 Dingley J, Tooley J, Liu X. et al. Xenon ventilation during therapeutic hypothermia in neonatal encephalopathy: a feasibility study. Pediatrics 2014; 133 (05) 809-818
  • 124 Chakkarapani E, Thoresen M, Hobbs CE, Aquilina K, Liu X, Dingley J. A closed-circuit neonatal xenon delivery system: a technical and practical neuroprotection feasibility study in newborn pigs. Anesth Analg 2009; 109 (02) 451-460
  • 125 Dominici M, Le Blanc K, Mueller I. et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006; 8 (04) 315-317
  • 126 Serrenho I, Rosado M, Dinis A. et al. Stem cell therapy for neonatal hypoxic-ischemic encephalopathy: a systematic review of preclinical studies. Int J Mol Sci 2021; 22 (06) 1-29
  • 127 Tapia-Bustos A, Lespay-Rebolledo C, Vío V. et al. Neonatal mesenchymal stem cell treatment improves myelination impaired by global perinatal asphyxia in rats. Int J Mol Sci 2021; 22 (06) 3275
  • 128 McDonald CA, Penny TR, Paton MCB. et al. Effects of umbilical cord blood cells, and subtypes, to reduce neuroinflammation following perinatal hypoxic-ischemic brain injury. J Neuroinflammation 2018; 15 (01) 47
  • 129 Wang XL, Zhao YS, Hu MY, Sun YQ, Chen YX, Bi XH. Umbilical cord blood cells regulate endogenous neural stem cell proliferation via hedgehog signaling in hypoxic ischemic neonatal rats. Brain Res 2013; 1518: 26-35
  • 130 Hu Y, Chen W, Wu L, Jiang L, Qin H, Tang N. Hypoxic preconditioning improves the survival and neural effects of transplanted mesenchymal stem cells via CXCL12/CXCR4 signalling in a rat model of cerebral infarction. Cell Biochem Funct 2019; 37 (07) 504-515
  • 131 Cotten CM, Murtha AP, Goldberg RN. et al. Feasibility of autologous cord blood cells for infants with hypoxic-ischemic encephalopathy. J Pediatr 2014; 164 (05) 973-979.e1
  • 132 Cotten CM, Fisher K, Kurtzberg J, Simmons R, Phase I. Trial of Allogeneic Umbilical Cord Tissue-Derived Mesenchymal Stromal Cells in Neonates with Hypoxic-Ischemic Encephalopathy. Cytotherapy 2020; 22 (5, suppl): S192
  • 133 Kabatas S, Civelek E, Savrunlu EC. et al. Feasibility of allogeneic mesenchymal stem cells in pediatric hypoxic-ischemic encephalopathy: phase I study. World J Stem Cells 2021; 13 (05) 470-484
  • 134 Lu B, Nagappan G, Lu Y. BDNF and synaptic plasticity, cognitive function, and dysfunction. Handb Exp Pharmacol 2014; 220: 223-250
  • 135 Zhao H, Alam A, San CY. et al. Molecular mechanisms of brain-derived neurotrophic factor in neuro-protection: Recent developments. Brain Res 2017; 1665: 1-21
  • 136 Xiong LL, Chen J, Du RL. et al. Brain-derived neurotrophic factor and its related enzymes and receptors play important roles after hypoxic-ischemic brain damage. Neural Regen Res 2021; 16 (08) 1453-1459
  • 137 Bae SH, Yoo MR, Kim YY. et al. Brain-derived neurotrophic factor mediates macrophage migration inhibitory factor to protect neurons against oxygen-glucose deprivation. Neural Regen Res 2020; 15 (08) 1483-1489
  • 138 Xue LL, Du RL, Hu Y. et al. BDNF promotes neuronal survival after neonatal hypoxic-ischemic encephalopathy by up-regulating Stx1b and suppressing VDAC1. Brain Res Bull 2021; 174: 131-140
  • 139 Lindholm D, Dechant G, Heisenberg C-P, Thoenen H. Brain-derived neurotrophic factor is a survival factor for cultured rat cerebellar granule neurons and protects them against glutamate-induced neurotoxicity. Eur J Neurosci 1993; 5 (11) 1455-1464
  • 140 Miguel PM, Deniz BF, Confortim HD. et al. Methylphenidate treatment increases hippocampal BDNF levels but does not improve memory deficits in hypoxic-ischemic rats. J Psychopharmacol 2020; 34 (07) 750-758
  • 141 Difato F, Tsushima H, Pesce M, Benfenati F, Blau A, Chieregatti E. The formation of actin waves during regeneration after axonal lesion is enhanced by BDNF. Sci Rep 2011; 1 (01) 183
  • 142 Im SH, Yu JH, Park ES. et al. Induction of striatal neurogenesis enhances functional recovery in an adult animal model of neonatal hypoxic-ischemic brain injury. Neuroscience 2010; 169 (01) 259-268
  • 143 Liu W, Wang X, O'Connor M, Wang G, Han F. Brain-derived neurotrophic factor and its potential therapeutic role in stroke comorbidities. Neural Plast 2020; 2020: 1969482
  • 144 Millar RP, Lu ZL, Pawson AJ, Flanagan CA, Morgan K, Maudsley SR. Gonadotropin-releasing hormone receptors. Endocr Rev 2004; 25 (02) 235-275
  • 145 Gründker C, Günthert AR, Westphalen S, Emons G. Biology of the gonadotropin-releasing hormone system in gynecological cancers. Eur J Endocrinol 2002; 146 (01) 1-14
  • 146 Badr M, Pelletier G. Characterization and autoradiographic localization of LHRH receptors in the rat brain. Synapse 1987; 1 (06) 567-571
  • 147 Dolan S, Evans NP, Richter TA, Nolan AM. Expression of gonadotropin-releasing hormone and gonadotropin-releasing hormone receptor in sheep spinal cord. Neurosci Lett 2003; 346 (1,2): 120-122
  • 148 Jennes L, Eyigor O, Janovick JA, Conn PM. Brain gonadotropin releasing hormone receptors: localization and regulation. Recent Prog Horm Res 1997; 52: 475-490 , discussion 490–491
  • 149 Deivaraju C, Temple HT, Block N, Robinson P, Schally AV. LHRH receptor expression in sarcomas of bone and soft tissue. Horm Mol Biol Clin Investig 2016; 28 (02) 105-111
  • 150 Quintanar JL, Salinas E. Neurotrophic effects of GnRH on neurite outgrowth and neurofilament protein expression in cultured cerebral cortical neurons of rat embryos. Neurochem Res 2008; 33 (06) 1051-1056
  • 151 Quintanar JL, Calderón-Vallejo D, Hernández-Jasso I. Effects of GnRH on neurite outgrowth, neurofilament and spinophilin proteins expression in cultured spinal cord neurons of rat embryos. Neurochem Res 2016; 41 (10) 2693-2698
  • 152 Periti P, Mazzei T, Mini E. Clinical pharmacokinetics of depot leuprorelin. Clin Pharmacokinet 2002; 41 (07) 485-504
  • 153 Conn PM, Crowley Jr WF. Gonadotropin-releasing hormone and its analogs. Annu Rev Med 1994; 45: 391-405
  • 154 Díaz Galindo C, Gómez-González B, Salinas E. et al. Leuprolide acetate induces structural and functional recovery of injured spinal cord in rats. Neural Regen Res 2015; 10 (11) 1819-1824
  • 155 Quintanar JL, Diaz-Galindo Mdel C, Calderon-Vallejo D, Hernandez-Jasso I. Clinical effect of leuprolide acetate, an agonist of GnRH, on sensitive and motor function in a patient with chronic spinal cord injury. J Neurol Res 2016; 6 (5,6): 111-113
  • 156 Quintanar JL, Díaz-Galindo C, Calderón-Vallejo D. et al. Neurological improvement in patients with chronic spinal cord injury treated with leuprolide acetate, an agonist of GnRH. Acta Neurobiol Exp (Warsz) 2018; 78 (04) 352-357
  • 157 Chu C, Gao G, Huang W. Ischemia-reperfusion injury effects a change in expression of GnRH and its receptor in CA1 neurons in rat hippocampus. Int J Neurosci 2008; 118 (03) 375-390
  • 158 Chu C, Xu B, Huang W. GnRH analogue attenuated apoptosis of rat hippocampal neuron after ischemia-reperfusion injury. J Mol Histol 2010; 41 (06) 387-393
  • 159 Guzmán-Soto I, Salinas E, Hernández-Jasso I, Quintanar JL. Leuprolide acetate, a GnRH agonist, improves experimental autoimmune encephalomyelitis: a possible therapy for multiple sclerosis. Neurochem Res 2012; 37 (10) 2190-2197
  • 160 Guzmán-Soto I, Salinas E, Quintanar JL. Leuprolide acetate inhibits spinal cord inflammatory response in experimental autoimmune encephalomyelitis by suppressing NF-κB activation. Neuroimmunomodulation 2016; 23 (01) 33-40