Open access peer-reviewed chapter

Hypoxic Brain Injury

Written By

Zeynep Özözen Ayas, Gülgün Uncu and Demet Özbabalık Adapınar

Submitted: 10 August 2019 Reviewed: 02 September 2019 Published: 11 October 2019

DOI: 10.5772/intechopen.89487

From the Edited Volume

Disorders of Consciousness - A Review of Important Issues

Edited by Kamil Hakan Dogan

Chapter metrics overview

1,040 Chapter Downloads

View Full Metrics

Abstract

Hypoxic brain injury (HBI) is a clinical condition that results from a decrease in brain blood flow and oxygenation. The damage due to cerebral hypoperfusion is caused by many possible reasons, which leads to severe wide spectrum of clinical presentations. It can be difficult to manage disease process of HBI because the clinical outcomes are poor and treatment options are limited. Neuroprotective trials against different underlying pathophysiological pathways are promising. In spite of all the difficulties, promising signals are obtained in the recent studies. In this article, we aim to provide the details of neurotoxic mechanisms and new interventions for neuroprotection of HBI.

Keywords

  • hypoxic brain injury
  • neuronal death
  • treatment

1. Introduction

Hypoxic brain injury (HBI) is a clinical condition that results from a decrease in brain blood flow and oxygenation. Energy in the brain is mainly derived from oxygen and glucose by 95% oxidative metabolism [1, 2]. About 50% of the energy obtained is used for communication and synaptic activity between neurons, 25% for the passage of ions through the cell membrane and 25% for molecular transport and biosynthesis [3, 4]. Metabolic need increases in seizure and fever but decreases in deep coma and anaesthesia. HBI can be defined as a damage to brain cells due to hypoxia. In HBI, the clinical definition is more complex, indicating hypoxia with many etiologic causes, and broad-spectrum brain injury caused by ischemia with or without reperfusion. Clinic developmental stage of the brain, condition of development of damage, regional weakness, ethology, difficult predictability of treatment and outcomes are heterogeneous due to the differences in accepted guidelines and management standards. Although effective treatment has not yet been found, progress has been made in the prevention of HBI.

Advertisement

2. Hypoxic-ischemic brain injury

Encephalopathy—the neurologic syndrome composed of abnormalities of consciousness, tone and autonomic control is the hallmark of acute HBI [5]. The stage of encephalopathy depends on the timing and severity of the hypoxia. HBI is an important cause of mortality and morbidity in the paediatric age group. Although effective treatment has not been found yet, progress has been made in the prevention of HBI. Advances in conservation have been achieved with neonatal asphyxia and mild hypothermia in ventricular fibrillation after cardiac arrest in adulthood and early use of thrombolytics after embolic stroke in adults.

Advertisement

3. Cellular mechanisms of neuronal death following HBI

Neurons consistently require a source of metabolic substrates, especially glucose and oxygen. HI brain injury results from intracellular and intracellular processes during and after the imbalance of the presence and consumption of these substrates in the brain. In animal models, neuronal death after HBI occurs in two phases [6]: Immediately after HBI, neurons begin to die rapidly, possibly a cell death process characterised by necrosis, loss of acute plasma membrane integrity and loss of ATP [7]. In the second stage, neurons die from hours to days [8], primarily through apoptosis [9], which is cascade of active, tightly regulated intracellular pathways. Neuropathological evidence of classical neuronal apoptosis after HBI is less pronounced in humans than in animal models [10]. However, there is no doubt that urgent and delayed neuronal deaths are below neurological damage after HBI. New approaches to salvage neurons following HBI have strengthened the existing understanding of HI-induced brain injury mechanisms to specifically target central mechanisms of neuronal death. We will briefly review excitotoxicity, free radical toxicity and inflammation procedures in order to place these treatments in the context of their targeted cellular mechanisms.

3.1 Excitotoxicity

Glutamate is a stimulating neurotransmitter everywhere in the brain. Under pathological conditions, including HI, neuronal receptors for glutamate are overactive due to pathologically high glutamate concentration in the extraneuronal domain. This high concentration occurs as a result of synaptic release of glutamate pathologically, dysfunction of glutamate uptake mechanisms and release of glutamate from the intracellular metabolic pool. Glutamate receptor overactivation results in neuronal death, hence excitotoxicity. Overactivation of the N-methyl-D-aspartate (NMDA), subtype of the glutamate receptor, was highly effective in neuronal death after HI. NMDA receptor overactivation allows intracellular calcium to rise to toxic levels and causes cell death by activating cytotoxic phospholipases, proteases, lipases and endonucleases. Calcium is also absorbed by the mitochondria, causing loss of ATP synthesis, oxidative stress, release of proapoptotic factors and activation of the apoptotic cascade.

3.2 Free radical toxicity

Free radicals are molecules containing one or more unpaired electrons that allow increased intermolecular reactivity. Primary oxygen-free radical superoxide anion is produced in cells (O2 ). Superoxide is an important intracellular signalling molecule, as is the metabolite hydrogen peroxide (H2O2). Together with the highly reactive hydroxyl radical, O2 and H2O2 are oxygen-derived free radicals present in the cell. Oxidative stress refers to increased levels of these radicals. Oxidative stress contributes to neuron death after HI [11], by breaking down cellular proteins and DNA.

In addition to oxidative stress, increased nitric oxide (NO), nitrogen-free radical, production is a central mechanism of HI-induced neuronal death [12]. Increased NO production is mediated by neuron-specific NO synthase (nNOS) and elevated by HI (and excitotoxicity)-induced intracellular calcium concentrations. Endothelial NOS (eNOS), a second NO synthase isoform, controls vascular resistance in all organs, including the brain. Maintaining eNOS activity during and after experimental HI improves cerebral blood flow and neuronal survival [13]; therefore, treatments aimed at reducing neuronal NO production should specifically target nNOS and maintain eNOS activity. In addition to its direct effects, NO interacts with O2 to form highly reactive and toxic radical peroxynitrite [14]. Peroxynitrite-mediated peroxidation of lipid components of cellular membranes [15] and mitochondrial proteins oxidative modification [16] are important mechanisms of neuronal damage. In particular, lipid peroxidation changes the cellular membrane structure and function that triggers cellular necrosis or apoptosis.

3.3 Inflammation

Improved results in HBI animal models following inflammation inhibition [17] show that inflammation is an important mechanism of HI-induced neuronal death. After HBI, microglia is activated [18], proinflammatory cytokines, e.g. IL-1 and TNF-alpha. In addition, microglia-derived chemokines increase acutely to receive peripheral immune cells into the brain [19]. HBI activates the complementary stage in the brain [20]. Complement activation results in the formation of membrane attack complexes that form pores within the plasma membranes and lead to cell lysis [21]. Therefore, after HBI, a coordinated inflammatory response emerges, which makes a significant contribution to HBI-induced neuron death in the brain.

Advertisement

4. New treatments for HBI

The understanding of the mechanisms of HI-induced neuronal approaches to neuroprotection have shown promise in pre-clinical studies and early clinical trials. Below, we review some of the most promising approaches at different stages of development from early stage research to clinical studies and FDA approval. Since these therapies may address different mechanisms than those mediating hypothermized neuroprotection, these novel therapies also provide additional neuroprotection to those available from hypothermia therapy.

4.1 Erythropoietin

Erythropoietin (EPO) is an endogenous, hypoxia-derived glycoprotein produced in the kidney that has been shown to first regulate haematopoietic function through EPO-specific receptors. [22]. Recombinant EPO (r-EPO), currently approved to increase erythropoietin in anaemia, has also been shown in animal studies where HBI is neuroprotective [23, 24]. Activation of neuronal EPO receptors prevents HBI-induced activation of NMDA receptors and increases expression of anti-apoptotic proteins, potentially reduces excitotoxicity and reduces apoptosis [24, 25]. EPO receptor activation also inhibits HBI-induced stimulation of peroxynitrite (oxidative stress) and inflammatory cytokines, potentially reducing free radical toxicity and inflammation. [25]. EPO receptor expression, which is of particular importance for neonatal HBI, is abundant in the developing mammalian brain [26]. Systemically administered r-EPO after HBI has been shown to cross the blood-brain barrier [27]. In one study, the pharmacokinetics of EPO levels in cerebrospinal fluid in babies treated with EPO after HBI was parallel to that observed in serum [28], suggesting that r-EPO could cross the blood-brain barrier in humans.

4.2 Melatonin

Melatonin is a pineal gland hormone secreted in response to environmental light-dark cycles [29]. Melatonin has multiple cellular effects, two of which directly target known mechanisms of HBI. First, melatonin reduces free radical toxicity, scavenging hydroxyl radical and peroxynitrite by direct electron transfer [30]. Melatonin also reduces O2 production in brain slices in vitro following hypoxic ischemic stress [31]. Second, melatonin has anti-inflammatory activity. Thus, after umbilical cord occlusion in fatal sheep, melatonin reduced the production of 8-isoprostanes [32], a potent mediator of HBI-induced inflammation. In addition, melatonin given to rats immediately after focal cerebral ischemia decreased neutrophil migration and macrophage/activated microglial infiltration after 48 hours and decreased only in the ischemic hemisphere [33]. Finally, melatonin reduces the binding of NF-yaB to DNA, resulting in the production of proinflammatory cytokines including interleukin-2, interleukin-6 and tumour necrosis factor alpha [34]. These cellular effects have led to extensive investigation of melatonin as a treatment for hypoxic brain damage.

Short-term assessments of melatonin, infarct size and neurobehavioural outcomes in rats after focal cerebral ischemia are improved [33], suggesting that melatonin treatment may be applicable to global brain ischemia in the newborn. However, short-term improvements may reflect only the temporary inhibition of death-induced procedures without altering the final extent of neuronal death. Finally, melatonin may have a neuroproductive effect in addition to hypothermia. Following induction of global ischemia in newborn pigs, melatonin with hypothermia reduced MR spectroscopic indices of impaired cerebral energy metabolism compared to hypothermia alone [35].

4.3 Allopurinol

Allopurinol is a xanthine oxidase inhibitor that is a source of cytosolic O2 , which has attracted attention as a potential neuroprotective agent during HI, especially as it can cross the placenta to produce therapeutic levels in newborns [36]. Animal models including in vivo and in vitro rat models and in vivo sheep models have demonstrated that allopurinol is neuroprotective [37].

4.4 Topiramate

Topiramate is an anti-epileptic drug of interest as a potential neuroprotective agent for brain injury. Topiramate prevents seizures by inhibiting neuronal excitability, including blockade of glutamate receptors [38]. This potential anti-excitotoxicity effect suggests topiramate as a candidate treatment for HBI. Indeed, following carotid artery ligation in the rat, topiramate significantly reduces neuronal death through inhibition of glutamate receptor activity [39], reducing HBI-induced neuronal apoptosis [40]. Of particular interest is the observation that topiramate, when combined with hypothermia, adds neuroprotective effects in animal models. [41].

In the pilot study, topiramate associated with whole-body hypothermia in 27 asphyxia infants did not cause any adverse effects, short-term outcome differences or pathological cerebral magnetic resonance imaging incidence compared to 27 controls [42]. Further extensive clinical studies are needed to assess the efficacy of topiramate in preventing HI injury.

4.5 Xenon

Xenon is a chemically non-reactive gas that is extensively studied as a general anaesthesia in Europe [43, 44], due to its highly favourable safety profile. One of the activities of xenon is against NMDA receptor activation, which reduces excitotoxicity. This reduced activity results from the xenon glycine block that binds to its regulatory region on the receptor [45]. Following hypoxia or excitotoxicity in cultured murine neurons, increased xenon concentrations significantly increased neuronal survival [46]. In neonatal rats, xenon inhalation improved both histological and functional outcomes 2 months after global HI [47]. Similarly, following global forebrain ischemia in the newborn pig, xenon inhalation proved neuronal survival 72 hours after insult [48]. In particular, in these models, xenon-induced neuroprotection has been found to add to the neuroprotection provided by induced hypothermia.

4.6 nNOS inhibition

The central role of NO in HI-mediated neuronal injury and the presence of specific small molecule inhibitors of nNOS make nNOS inhibition a potentially attractive approach. With the discovery of the toxic role of NOS in HI, early studies of NOS inhibitors have yielded contradictory results since early inhibitors do not have isoform specificity [49]. However, newer, specific nNOS inhibitors may promise more [50]. Prophylactic use of highly specific nNOS inhibitor JI-10 in preterm fetal sheep increased neuronal survival following deep asphyxia [51]. Although initial data for selective nNOS inhibitors are promising, the extent of non-target effects, such as inhibition of eNOS activity and any accompanying reduction in cerebral blood flow, will need to be investigated to initiate clinical trials.

4.7 Pluronic co-polymers

After HBI, the functions of cellular membranes may change due to lipid peroxidation and lipid signalling changes. After severe HI, neuronal plasma membrane dysfunction leads to reduced membrane integrity, infiltration of intracellular components into the extracellular space and necrosis. When HI is not severe enough to induce necrosis, HI-mediated dysfunction of mitochondrial intracellular membranes can trigger apoptosis [52]. Recently, a class of synthetic molecules has been used to address HI-induced dysfunction of injured neuronal membranes in pluronic, in vitro and in vivo. Pluronics, which consist of poly [ethylene oxide] (PEO) and poly [propylene oxide] (PPO) chains, have been arranged in a three-block PEO-PPO-PEO structure. This structure allows the pluronics to interact with the cellular membranes [53, 54] and recovers the integrity of the plasma membrane after injury. Pluronic F-68, a member of Pluronics, has been shown to immediately rescue neurons from death in in vitro HI models by apoptosis blockage [55, 56]. Preliminary evidence also shows that Pluronic F-68, provided to animals for 1 week after HI, significantly improves neuronal survival in the hippocampus, a brain region highly sensitive to global HI, and saves hippocampus behaviour [57]. The novelty of this membrane-targeted approach and the lack of toxicity [58, 59] suggest that targeting membrane dysfunction may be a suitable treatment for future HBI.

4.8 Therapeutic hypothermia

The main mechanism underlying hypothermia in reducing ischemic tissue damage is its effect on metabolism [60]. Oxygen use decreases by 7% almost linearly with each °C reduction below normal [61]. On the other hand, ischemia becomes more tolerable due to the slowdown in metabolism, although a decrease in blood pressure of about 5% per degree has been observed. In animal experiments, the brain volume is approximately 4% less at 25°C compared to 37°C. Here, the main decreasing cerebral blood flow and volume, the CSF section increases by about 32%. In conclusion, intracranial and venous pressures decrease [62]. In addition, hypothermia reduces the release of excitatory neurotransmitters, such as glutamate and glycine, suppresses free radical toxicity, creates favourable effects on intracellular mediator systems, also reduces intracellular acidosis, inhibits the excretion of ubiquitin, which binds abnormal proteins and facilitates their excretion, anti-apoptotic effects and anti-inflammatory effects and other mechanisms by reducing ischemic neuron damage [63, 64].

Advertisement

5. Conclusion

Hypoxic-ischemic brain injury is a simple imbalance between demand and supply to brain energy. However, cellular mechanisms leading to neuronal death are complex and multifactorial. The overall effectiveness of induced hypothermia is relatively low and the need for mechanism-oriented therapies for HBI is high. Basic research may provide therapeutic targets for translation testing, while defining the underlying mechanisms of HBI-mediated neuronal death. The approaches discussed above target the cellular mechanisms of HBI-mediated neuronal death in many different ways. With ongoing research, one or more of these approaches or their derivatives may ultimately be effective treatments for HBI.

References

  1. 1. Astrup J, Sorenson PM, Sorenson HR. Oxygen and glucose consumption is related to Na+, K+ transport in canine brain. Stroke. 1981;12:726-730
  2. 2. Kassissia IG, Goresky CA, Rose CP, et al. Tracer oxygen distribution is barrier-limited in the cerebral microcirculation. Circulation Research. 1995;77:1202-1211
  3. 3. Astrup J. Energy-requiring cell functions in the ischemic brain: Their critical supply and possible inhibition in the protective therapy. Journal of Neurosurgery. 1982;56:482-497
  4. 4. Siesjo BK. Cerebral circulation and metabolism. Journal of Neurosurgery. 1984;60:883-908
  5. 5. Sarnat HB, Sarnat MS. Neonatal encephalopathy following fetal distress. A clinical and electroencephalographic study. Archives of Neurology. 1976;33:696-705
  6. 6. Northington FJ, Ferriero DM, Graham EM, et al. Early Neurodegeneration after hypoxia-ischemia in neonatal rat is necrosis while delayed neuronal death is apoptosis. Neurobiology of Disease. 2001;8:207-219
  7. 7. Fricker M, Tolkovsky AM. Cell culture techniques. In: Acsher M, Sunol C, Bal-Price A, editors. Mechanisms of neuronal and glial cell death. NeuroMethods. New York: Humana Press; 2011
  8. 8. McLean C, Ferriero D. Mechanisms of hypoxic—Ischemic injury in the term infant. Seminars in Perinatology. 2004;28:425-432
  9. 9. Ankarcrona M, Dypbukt JM, Bonfoco E, et al. Glutamate-induced neuronal death: A succession of necrosis or apoptosis depending on mitochondrial function. Neuron. 1995;15:961-973
  10. 10. Northington FJ, Chavez-Valdez R, Martin LJ. Neuronal cell death in neonatal hypoxia-ischemia. Annals of Neurology. 2011;69:743-758
  11. 11. Abramov AY, Scorziello A, Duchen MR. Three distinct mechanisms generate oxygen free radicals in neurons and contribute to cell death during anoxia and Reoxygenation. The Journal of Neuroscience. 2007;27:1129-1138
  12. 12. Moro MA, Cardenas A, Hurtado O, et al. Role of nitric oxide after brain ischaemia. Cell Calcium. 2004;36:265-275
  13. 13. Huang Z, Huang PL, Ma J, et al. Enlarged infarcts in endothelial nitric oxide synthase knockout mice are attenuated by nitro-L-arginine. Journal of Cerebral Blood Flow and Metabolism. 1996;16:981-987
  14. 14. Brown GC. Nitric oxide regulates mitochondrial respiration and cell functions by inhibiting cytochrome oxidase. FEBS Letters. 1995;369:136-139
  15. 15. Szabo C. Multiple pathways of peroxynitrite cytotoxicity. Toxicology Letters. 2003;140-141:105-112
  16. 16. Bolanos JP, Almeida A, Medina JM. Nitric oxide mediates brain mitochondrial damage during perinatal anoxia. Brain Research. 1998;787:117-122
  17. 17. Liu XH, Kwon D, Schielke GP, et al. Mice deficient in interleukin-1 converting enzyme are resistant to neonatal hypoxic-ischemic brain damage. Journal of Cerebral Blood Flow and Metabolism. 1999;19:1099-1108
  18. 18. McRae A, Gilland E, Bona E, et al. Microglia activation after neonatal hypoxic-ischemia. Brain Research. Developmental Brain Research. 1995;84:245-252
  19. 19. Bona E, Andersson AL, Blomgren K, et al. Chemokine and inflammatory cell response to hypoxia ischemia in immature rats. Pediatric Research. 1999;45:500-509
  20. 20. Cowell RM, Plane JM, Silverstein FS. Complement activation contributes to hypoxic-ischemic brain injury in neonatal rats. The Journal of Neuroscience. 2003;23:9459-9468
  21. 21. Harhausen D, Khojasteh U, Stahel PF, et al. Membrane attack complex inhibitor CD59a protects against focal cerebral ischemia in mice. Journal of Neuroinflammation. 2010;7:15
  22. 22. Koury MJ, Bondurant MC. The mechanism of erythropoietin action. American Journal of Kidney Diseases. 1991;18:20-23
  23. 23. Aydin A, Genc K, Akhisaroglu M, et al. Erythropoietin exerts neuroprotective effect in neonatal rat model of hypoxic-ischemic brain injury. Brain & Development. 2003;25:494-498
  24. 24. Kumral A, Genc S, Ozer E, et al. Erythropoietin downregulates bax and DP5 proapoptotic gene expression in neonatal hypoxic-ischemic brain injury. Biology of the Neonate. 2006;89:205-210
  25. 25. Digicaylioglu M, Lipton SA. Erythropoietin-mediated neuroprotection involves cross-talk between Jak2 and NF-kappaB signalling cascades. Nature. 2001;412:641-647
  26. 26. Juul SE, Anderson DK, Li Y, et al. Erythropoietin and erythropoietin receptor in the developing human central nervous system. Pediatric Research. 1998;43:40-49
  27. 27. Brines ML, Ghezzi P, Keenan S, et al. Erythropoietin crosses the blood-brain barrier to protect against experimental brain injury. Proceedings of the National Academy of Sciences of the United States of America. 2000;97:10526-10531
  28. 28. Zhu C, Kang W, Xu F, et al. Erythropoietin improved neurologic outcomes in newborns with hypoxic ischemic encephalopathy. Pediatrics. 2009;124:e218-e226
  29. 29. Lynch HJ, Wurtman RJ, Moskowitz MA, et al. Daily rhythm in human urinary melatonin. Science. 1975;187:169-171
  30. 30. Tan DX, Reiter RJ, Manchester LC, et al. Chemical and physical properties and potential mechanisms: Melatonin as a broad spectrum antioxidant and free radical scavenger. Current Topics in Medicinal Chemistry. 2002;2:181-197
  31. 31. Uchida K, Samejima M, Okabe A, et al. Neuroprotective effects of melatonin against anoxia/ aglycemia stress, as assessed by synaptic potentials and superoxide production in rat hippocampal slices. Journal of Pineal Research. 2004;37:215-222
  32. 32. Welin AK, Svedin P, Lapatto R, et al. Melatonin reduces inflammation and cell death in white matter in the mid-gestation fetal sheep following umbilical cord occlusion. Pediatric Research. 2007;61:153-158
  33. 33. Lee MY, Kuan YH, Chen HY, et al. Intravenous administration of melatonin reduces the intracerebral cellular inflammatory response following transient focal cerebral ischemia in rats. Journal of Pineal Research. 2007;42:297-309
  34. 34. Reiter RJ, Calvo JR, Karbownik M, et al. Melatonin and its relation to the immune system and inflammation. Annals of the New York Academy of Sciences. 2000;917:376-386
  35. 35. Robertson NJ, Faulkner S, Fleiss B, et al. Melatonin augments hypothermic neuroprotection in a perinatal asphyxia model. Brain. 2013;136:90-105
  36. 36. Boda M, Nemeth I, Boda D. The caffeine metabolic ratio as an index of xanthine oxidase activity in clinically active and silent celiac patients. Journal of Pediatric Gastroenterology and Nutrition. 1999;29:546-550
  37. 37. Kaandorp JJ, Derks JB, Oudijk MA, et al. Antenatal allopurinol reduces hippocampal brain damage after acute birth asphyxia in late gestation fetal sheep. Reproductive Sciences. 2014;21:251-259
  38. 38. Follett PL, Deng W, Dai W, et al. Glutamate receptor-mediated oligodendrocyte toxicity in periventricular leukomalacia: A protective role for topiramate. The Journal of Neuroscience. 2004;24:4412-4420
  39. 39. Noh MR, Kim SK, Sun W, et al. Neuroprotective effect of topiramate on hypoxic ischemic brain injury in neonatal rats. Experimental Neurology. 2006;201:470-478
  40. 40. Schubert S, Brandl U, Brodhun M, et al. Neuroprotective effects of topiramate after hypoxia ischemia in newborn piglets. Brain Research. 2005;1058:129-136
  41. 41. Liu Y, Barks JD, Xu G, et al. Topiramate extends the therapeutic window for hypothermia-mediated neuroprotection after stroke in neonatal rats. Stroke. 2004;35:1460-1465
  42. 42. Filippi L, Poggi C, la Marca G, et al. Oral topiramate in neonates with hypoxic ischemic encephalopathy treated with hypothermia: A safety study. The Journal of Pediatrics. 2010;157:361-366
  43. 43. Baumert JH, Hein M, Hecker KE, et al. Autonomic cardiac control with xenon anaesthesia in patients at cardiovascular risk. British Journal of Anaesthesia. 2007;98:722-727
  44. 44. Wappler F, Rossaint R, Baumert J, et al. Multicenter randomized comparison of xenon and isoflurane on left ventricular function in patients undergoing elective surgery. Anesthesiology. 2007;106:463-471
  45. 45. Banks P, Franks NP, Dickinson R. Competitive inhibition at the glycine site of the N-methyl-D-aspartate receptor mediates xenon neuroprotection against hypoxia-ischemia. Anesthesiology. 2010;112:614-622
  46. 46. Wilhelm S, Ma D, Maze M, et al. Effects of xenon on in vitro and in vivo models of neuronal injury. Anesthesiology. 2002;96:1485-1491
  47. 47. Hobbs C, Thoresen M, Tucker A, et al. Xenon and hypothermia combine additively, offering long-term functional and histopathologic neuroprotection after neonatal hypoxia/ischemia. Stroke. 2008;39:1307-1313
  48. 48. Chakkarapani E, Dingley J, Liu X, et al. Xenon enhances hypothermic neuroprotection in asphyxiated newborn pigs. Annals of Neurology. 2010;68:330-341
  49. 49. Dalkara T, Yoshida T, Irikura K, et al. Dual role of nitric oxide in focal cerebral ischemia. Neuropharmacology. 1994;33:1447-1452
  50. 50. Rao S, Lin Z, Drobyshevsky A, et al. Involvement of neuronal nitric oxide synthase in ongoing fetal brain injury following near-term rabbit hypoxia-ischemia. Developmental Neuroscience. 2011;33:288-298
  51. 51. Drury PP, Davidson JO, van den Heuij LG, et al. Partial neuroprotection by nNOS inhibition during profound asphyxia in preterm fetal sheep. Experimental Neurology. 2013;250:1-21
  52. 52. Grimm S. The ER–mitochondria interface: The social network of cell death. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research. 2012;1823:327-334
  53. 53. Wu G, Majewski J, Ege C, et al. Interaction between lipid monolayers and Poloxamer 188: An X-ray reflectivity and diffraction study. Biophysical Journal. 2005;89:3159-3173
  54. 54. Firestone MA, Seifert S. Interaction of nonionic PEO-PPO Diblock copolymers with lipid bilayers. Biomacromolecules. 2005;6:2678-2687
  55. 55. Shelat PB, Plant LD, Wang JC, et al. The membrane-active tri-block copolymer Pluronic F-68 profoundly rescues rat hippocampal neurons from oxygen–glucose deprivation-induced death through early inhibition of apoptosis. The Journal of Neuroscience. 2013;33:12287-12299
  56. 56. Marks JD, Pan CY, Bushell T, et al. Amphiphilic, tri-block copolymers provide potent, membrane-targeted neuroprotection. FASEB Journal Express Article. 2001;15(6):1107-1109. DOI: 10.1096/fj.00-0547fje
  57. 57. Marks JD, Doi A, Garcia AG, et al. Poloxamer 188, an amphiphilic tri-block co-polymer, provides profound hippocampal neuroprotection following transient global forebrain ischemia in the Mongolian gerbil. 2007 Neuroscience Meeting Planner. 2007
  58. 58. Ballas SK, Files B, Luchtman-Jones L, et al. Safety of purified poloxamer 188 in sickle cell disease: Phase I study of a non-ionic surfactant in the management of acute chest syndrome. Hemoglobin. 2004;28:85-102
  59. 59. Duvinage C, Millecamps S, Sagnier A, et al. One month intravenous toxicity studies of poloxamer 188 in male Sprague-Dawley rats and in beagle dogs. Toxicology Letters. 1996;88:101
  60. 60. Erecinska M, Thoresen M, Silver IA. Effects of hypothermia on energy metabolism in mammalian central nervous system. Journal of Cerebral Blood Flow and Metabolism. 2003;23:513-530
  61. 61. Girnsberg MD. Temperature influences on ischemic brain injury. In: Hsu CY, editor. Ischemic Stroke: From Basic Mechanism to New Drug Development. Vol. 16. Basel: Monogr Clin Neurosci. Karger; 1998. pp. 65-88
  62. 62. Rincon F, Mayer SA. Therapeutic hypothermia for brain injury after cardiac arrest. Seminars in Neurology. 2006;26:387-395
  63. 63. Berger C, Schabitz WR, Georgiadis D, et al. Effects of hypothermia on excitatory amino acids and metabolism in stroke patients: A micro-dialysis study. Stroke. 2002;33:519-524
  64. 64. Hemmen TM, Lyden PD. Induced hypothermia for acute stroke. Stroke. 2007;28:794-799

Written By

Zeynep Özözen Ayas, Gülgün Uncu and Demet Özbabalık Adapınar

Submitted: 10 August 2019 Reviewed: 02 September 2019 Published: 11 October 2019