Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-c9gpj Total loading time: 0 Render date: 2024-07-10T00:08:49.527Z Has data issue: false hasContentIssue false

18 - Novel neuroprotective therapies

from Section 3 - The future

Published online by Cambridge University Press:  05 March 2013

A. David Edwards
Affiliation:
Institute of Reproductive and Developmental Biology, Imperial College, London
Denis V. Azzopardi
Affiliation:
Institute of Reproductive and Developmental Biology, Imperial College, London
Alistair J. Gunn
Affiliation:
School of Medical Sciences, University of Auckland
Get access

Summary

Introduction

This chapter will review the evidence for selected novel neuroprotective therapies, providing background information, in vitro and in vivo data for safety, efficacy, clinical feasibility and where available, a review of clinical studies. Examples of growth factors (erythropoietin), cell-based therapies and inhaled agents (xenon) will be discussed. We aim to give an overview of therapies that may be added to our armamentarium in the next decade. Ideally, in the future, the approach to neuroprotection will be integrated, with combined therapies timed to target specific mechanisms of injury and repair.

Introduction

Erythropoietin (Epo) is a 34-kDa glycoprotein originally identified for its role in erythropoiesis, but which has since been found to have other functions. During embryologic and fetal development, Epo receptors (EpoR) are widespread [1] and Epo appears to have trophic effects on the vascular and nervous systems among others [2,3]. As the fetus develops, EpoR become increasingly regionally and cell-specific [4]. The nonhaematopoietic function of Epo as it interacts with these receptors has been the subject of extensive investigation over the past 15 years. In particular, the neuroprotective effects of Epo and the mechanisms by which these effects occur have been researched in experimental paradigms ranging from cell culture to knockout mice, to small and large animal models of brain injury.

Type
Chapter
Information
Neonatal Neural Rescue
A Clinical Guide
, pp. 195 - 207
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Juul, SE, Yachnis, AT, Christensen, RD. Tissue distribution of erythropoietin and erythropoietin receptor in the developing human fetus. Early Hum Dev 1998;52:235–49.CrossRefGoogle ScholarPubMed
Yu, X, Shacka, JJ, Eells, JB, et al. Erythropoietin receptor signalling is required for normal brain development. Development 2002;129:505–16.Google ScholarPubMed
Chen, ZY, Asavaritikrai, P, Prchal, JT, Noguchi, CT. Endogenous erythropoietin signaling is required for normal neural progenitor cell proliferation. J Biol Chem 2007;282:25875–83.CrossRefGoogle ScholarPubMed
Juul, SE, Yachnis, AT, Rojiani, AM, Christensen, RD. Immunohistochemical localization of erythropoietin and its receptor in the developing human brain. Pediatr Dev Pathol 1999;2:148–58.CrossRefGoogle ScholarPubMed
Wang, L, Zhang, Z, Wang, Y, Zhang, R, Chopp, M. Treatment of stroke with erythropoietin enhances neurogenesis and angiogenesis and improves neurological function in rats. Stroke 2004;35:1732–7.CrossRefGoogle ScholarPubMed
Wallach, I, Zhang, J, Hartmann, A, et al. Erythropoietin-receptor gene regulation in neuronal cells. Pediatr Res 2009;65:619–24.CrossRefGoogle ScholarPubMed
Sugawa, M, Sakurai, Y, Ishikawa-Ieda, Y, Suzuki, H, Asou, H. Effects of erythropoietin on glial cell development; oligodendrocyte maturation and astrocyte proliferation. Neurosci Res 2002;44:391–403.CrossRefGoogle ScholarPubMed
Nagai, A, Nakagawa, E, Choi, HB, et al. Erythropoietin and erythropoietin receptors in human CNS neurons, astrocytes, microglia and oligodendrocytes grown in culture. J Neuropathol Exp Neurol 2001;60:386–92.CrossRefGoogle ScholarPubMed
Chong, ZZ, Kang, JQ, Maiese, K. Erythropoietin fosters both intrinsic and extrinsic neuronal protection through modulation of microglia, Akt1, Bad and caspase-mediated pathways. Br J Pharmacol 2003;138:1107–18.CrossRefGoogle ScholarPubMed
Digicaylioglu, M, Lipton, SA. Erythropoietin-mediated neuroprotection involves cross-talk between Jak2 and NF-kappaB signalling cascades. Nature 2001;412:641–7.CrossRefGoogle ScholarPubMed
Genc, K, Genc, S, Baskin, H, Semin, I. Erythropoietin decreases cytotoxicity and nitric oxide formation induced by inflammatory stimuli in rat oligodendrocytes. Physiol Res 2006;55:33–8.Google ScholarPubMed
Vitellaro-Zuccarello, L, Mazzetti, S, Madaschi, L, et al. Chronic erythropoietin-mediated effects on the expression of astrocyte markers in a rat model of contusive spinal cord injury. Neuroscience 2008;151:452–66.CrossRefGoogle Scholar
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;97:10526–31.CrossRefGoogle ScholarPubMed
Juul, SE, McPherson, RJ, Farrell, FX, et al. Erythropoietin concentrations in cerebrospinal fluid of nonhuman primates and fetal sheep following high-dose recombinant erythropoietin. Biol Neonate 2004;85:138–44.CrossRefGoogle ScholarPubMed
Statler, PA, McPherson, RJ, Bauer, LA, Kellert, BA, Juul, SE. Pharmacokinetics of high-dose recombinant erythropoietin in plasma and brain of neonatal rats. Pediatr Res 2007;61:671–5.CrossRefGoogle ScholarPubMed
Kellert, BA, McPherson, RJ, Juul, SE. A comparison of high-dose recombinant erythropoietin treatment regimens in brain-injured neonatal rats. Pediatr Res 2007;61:451–5.CrossRefGoogle ScholarPubMed
Weber, A, Dzietko, M, Berns, M, et al. Neuronal damage after moderate hypoxia and erythropoietin. Neurobiol Dis 2005;20:594–600.CrossRefGoogle ScholarPubMed
Keller, M, Yang, J, Griesmaier, E, et al. Erythropoietin is neuroprotective against NMDA-receptor-mediated excitotoxic brain injury in newborn mice. Neurobiol Dis 2006;24:357–66.CrossRefGoogle ScholarPubMed
Kawakami, M, Iwasaki, S, Sato, K, Takahashi, M. Erythropoietin inhibits calcium-induced neurotransmitter release from clonal neuronal cells. Biochem Biophys Res Commun 2000;279:293–7.CrossRefGoogle ScholarPubMed
Sun, Y, Calvert, JW, Zhang, JH. Neonatal hypoxia/ischemia is associated with decreased inflammatory mediators after erythropoietin administration. Stroke 2005;36:1672–8.CrossRefGoogle ScholarPubMed
Dzietko, M, Felderhoff-Mueser, U, Sifringer, M, et al. Erythropoietin protects the developing brain against N-methyl-D-aspartate receptor antagonist neurotoxicity. Neurobiol Dis 2004;15:177–87.CrossRefGoogle ScholarPubMed
Wang, L, Chopp, M, Gregg, SR, et al. Neural progenitor cells treated with EPO induce angiogenesis through the production of VEGF. J Cereb Blood Flow Metab 2008;28:1361–8.CrossRefGoogle ScholarPubMed
Bocker-Meffert, S, Rosenstiel, P, Rohl, C, et al. Erythropoietin and VEGF promote neural outgrowth from retinal explants in postnatal rats. Invest Ophthalmol Vis Sci 2002;43:2021–6.Google ScholarPubMed
Wang, L, Zhang, ZG, Zhang, RL, et al. Matrix metalloproteinase 2 (MMP2) and MMP9 secreted by erythropoietin-activated endothelial cells promote neural progenitor cell migration. J Neurosci 2006;26:5996–6003.CrossRefGoogle ScholarPubMed
Xiong, Y, Mahmood, A, Qu, C, et al. Erythropoietin improves histological and functional outcomes after traumatic brain injury in mice in the absence of the neural erythropoietin receptor. J Neurotrauma 2010;27:205–15.CrossRefGoogle ScholarPubMed
Gonzalez, FF, Abel, R, Almli, CR, et al. Erythropoietin sustains cognitive function and brain volume after neonatal stroke. Dev Neurosci 2009;31:403–11.CrossRefGoogle ScholarPubMed
Tsai, PT, Ohab, JJ, Kertesz, N, et al. A critical role of erythropoietin receptor in neurogenesis and post-stroke recovery. J Neurosci 2006;26:1269–74.CrossRefGoogle ScholarPubMed
Rees, S, Hale, N, De Matteo, R, et al. Erythropoietin is neuroprotective in a preterm ovine model of endotoxin-induced brain injury. J Neuropathol Exp Neurol 2010;69:306–19.CrossRefGoogle Scholar
Juul, SE, Aylward, E, Richards, T, et al. Prenatal cord clamping in newborn Macaca nemestrina: a model of perinatal asphyxia. Dev Neurosci 2007;29:311–20.CrossRefGoogle ScholarPubMed
Juul, SE, McPherson, RJ, Bauer, LA, et al. A phase I/II trial of high-dose erythropoietin in extremely low birth weight infants: pharmacokinetics and safety. Pediatrics 2008;122:383–91.CrossRefGoogle ScholarPubMed
Fauchere, JC, Dame, C, Vonthein, R, et al. An approach to using recombinant erythropoietin for neuroprotection in very preterm infants. Pediatrics 2008;122:375–82.CrossRefGoogle ScholarPubMed
Zhu, C, Kang, W, Xu, F, et al. Erythropoietin improved neurologic outcomes in newborns with hypoxic-ischemic encephalopathy. Pediatrics 2009;124:e218–26.CrossRefGoogle ScholarPubMed
Elmahdy, H, El-Mashad, AR, El-Bahrawy, H, et al. Human recombinant erythropoietin in asphyxia neonatorum: pilot trial. Pediatrics 2010;125:e1135–42.CrossRefGoogle ScholarPubMed
Ohls, RK, Ehrenkranz, RA, Das, A, et al. Neurodevelopmental outcome and growth at 18 to 22 months’ corrected age in extremely low birth weight infants treated with early erythropoietin and iron. Pediatrics 2004;114:1287–91.CrossRefGoogle ScholarPubMed
Bierer, R, Peceny, MC, Hartenberger, CH, Ohls, RK. Erythropoietin concentrations and neurodevelopmental outcome in preterm infants. Pediatrics 2006;118:e635–40.CrossRefGoogle ScholarPubMed
Brown, MS, Eichorst, D, Lala-Black, B, Gonzalez, R. Higher cumulative doses of erythropoietin and developmental outcomes in preterm infants. Pediatrics 2009;124:e681–7.CrossRefGoogle ScholarPubMed
Mennini, T, De Paola, M, Bigini, P, et al. Nonhematopoietic erythropoietin derivatives prevent motoneuron degeneration in vitro and in vivo. Mol Med 2006;12:153–60.CrossRefGoogle ScholarPubMed
Sturm, B, Helminger, M, Steinkellner, H, et al. Carbamylated erythropoietin increases frataxin independent from the erythropoietin receptor. Eur J Clin Invest 2010;40:561–5.CrossRefGoogle ScholarPubMed
Wang, L, Zhang, ZG, Gregg, SR, et al. The Sonic hedgehog pathway mediates carbamylated erythropoietin-enhanced proliferation and differentiation of adult neural progenitor cells. J Biol Chem 2007;282:32462–70.CrossRefGoogle ScholarPubMed
Pankratova, S, Kiryushko, D, Sonn, K, et al. Neuroprotective properties of a novel, non-haematopoietic agonist of the erythropoietin receptor. Brain 2010;133:2281–94.CrossRefGoogle ScholarPubMed
Ohls, RK. The use of erythropoietin in neonates. Clin Perinatol 2000;27:681–96.CrossRefGoogle ScholarPubMed
McPherson, RJ, Demers, EJ, Juul, SE. Safety of high-dose recombinant erythropoietin in a neonatal rat model. Neonatology 2007;91:36–43.CrossRefGoogle Scholar
Ribatti, D, Presta, M, Vacca, A, et al. Human erythropoietin induces a pro-angiogenic phenotype in cultured endothelial cells and stimulates neovascularization in vivo. Blood 1999;93:2627–36.Google ScholarPubMed
Ohlsson, A, Aher, SM. Early erythropoietin for preventing red blood cell transfusion in preterm and/or low birth weight infants. Cochrane Database Syst Rev 2006;3:CD004863.Google Scholar
Chen, J, Smith, LE. A double-edged sword: erythropoietin eyed in retinopathy of prematurity. J AAPOS 2008;12:221–2.CrossRefGoogle ScholarPubMed
Slusarski, JD, McPherson, RJ, Wallace, GN, Juul, SE. High-dose erythropoietin does not exacerbate retinopathy of prematurity in rats. Pediatr Res 2009;66:625–30.CrossRefGoogle Scholar
Cariou, A, Claessens, YE, Pene, F, et al. Early high-dose erythropoietin therapy and hypothermia after out-of-hospital cardiac arrest: a matched control study. Resuscitation 2008;76:397–404.CrossRefGoogle ScholarPubMed
Tseng, MY, Hutchinson, PJ, Richards, HK, et al. Acute systemic erythropoietin therapy to reduce delayed ischemic deficits following aneurysmal subarachnoid hemorrhage: a Phase II randomized, double-blind, placebo-controlled trial. Clinical article. J Neurosurg 2009;111:171–80.CrossRefGoogle ScholarPubMed
Pimentel-Coelho, PM, Mendez-Otero, R. Cell therapy for neonatal hypoxic-ischemic encephalopathy. Stem Cells Dev 2010;19:299–310.CrossRefGoogle ScholarPubMed
Sharp, FR, Gonzalez, MF, Ferriero, DM, Sagar, SM. Injured adult neocortical neurons sprout fibers into surviving fetal frontal cortex transplants: evidence using NADPH-diaphorase staining. Neurosci Lett 1986;65:204–8.CrossRefGoogle ScholarPubMed
Elsayed, MH, Hogan, TP, Shaw, PL, Castro, AJ. Use of fetal cortical grafts in hypoxic-ischemic brain injury in neonatal rats. Exp Neurol 1996;137:127–41.CrossRefGoogle ScholarPubMed
Jansen, EM, Solberg, L, Underhill, S, et al. Transplantation of fetal neocortex ameliorates sensorimotor and locomotor deficits following neonatal ischemic-hypoxic brain injury in rats. Exp Neurol 1997;147:487–97.CrossRefGoogle ScholarPubMed
Park, KI, Teng, YD, Snyder, EY. The injured brain interacts reciprocally with neural stem cells supported by scaffolds to reconstitute lost tissue. Nat Biotechnol 2002;20:1111–7.CrossRefGoogle ScholarPubMed
Hoehn, M, Kustermann, E, Blunk, J, et al. Monitoring of implanted stem cell migration in vivo: a highly resolved in vivo magnetic resonance imaging investigation of experimental stroke in rat. Proc Natl Acad Sci U S A 2002;99:16267–72.CrossRefGoogle ScholarPubMed
Park, KI, Himes, BT, Stieg, PE, et al. Neural stem cells may be uniquely suited for combined gene therapy and cell replacement: evidence from engraftment of Neurotrophin-3-expressing stem cells in hypoxic-ischemic brain injury. Exp Neurol 2006;199:179–90.CrossRefGoogle ScholarPubMed
Park, KI, Hack, MA, Ourednik, J, et al. Acute injury directs the migration, proliferation and differentiation of solid organ stem cells: evidence from the effect of hypoxia-ischemia in the CNS on clonal “reporter” neural stem cells. Exp Neurol 2006;199:156–78.CrossRefGoogle ScholarPubMed
Capone, C, Frigerio, S, Fumagalli, S, et al. Neurosphere-derived cells exert a neuroprotective action by changing the ischemic microenvironment. PLoS One 2007;2:e373.CrossRefGoogle ScholarPubMed
Hicks, AU, Hewlett, K, Windle, V, et al. Enriched environment enhances transplanted subventricular zone stem cell migration and functional recovery after stroke. Neuroscience 2007;146:31–40.CrossRefGoogle ScholarPubMed
Imitola, J, Raddassi, K, Park, KI, et al. Directed migration of neural stem cells to sites of CNS injury by the stromal cell-derived factor 1alpha/CXC chemokine receptor 4 pathway. Proc Natl Acad Sci U S A 2004;101:18117–22.CrossRefGoogle ScholarPubMed
Mueller, FJ, Serobyan, N, Schraufstatter, IU, et al. Adhesive interactions between human neural stem cells and inflamed human vascular endothelium are mediated by integrins. Stem Cells 2006;24:2367–72.CrossRefGoogle ScholarPubMed
Zheng, T, Rossignol, C, Leibovici, A, et al. Transplantation of multipotent astrocytic stem cells into a rat model of neonatal hypoxic-ischemic encephalopathy. Brain Res 2006;1112:99–105.CrossRefGoogle ScholarPubMed
Sato, Y, Nakanishi, K, Hayakawa, M, et al. Reduction of brain injury in neonatal hypoxic-ischemic rats by intracerebroventricular injection of neural stem/progenitor cells together with chondroitinase ABC. Reprod Sci 2008;15:613–20.CrossRefGoogle ScholarPubMed
Modo, M, Mellodew, K, Cash, D, et al. Mapping transplanted stem cell migration after a stroke: a serial, in vivo magnetic resonance imaging study. Neuroimage 2004;21:311–7.CrossRefGoogle ScholarPubMed
Guzman, R, Uchida, N, Bliss, TM, et al. Long-term monitoring of transplanted human neural stem cells in developmental and pathological contexts with MRI. Proc Natl Acad Sci U S A 2007;104:10211–6.CrossRefGoogle ScholarPubMed
Rice, HE, Hsu, EW, Sheng, H, et al. Superparamagnetic iron oxide labeling and transplantation of adipose-derived stem cells in middle cerebral artery occlusion-injured mice. AJR Am J Roentgenol 2007;188:1101–8.CrossRefGoogle ScholarPubMed
Obenaus, A, Robbins, M, Blanco, G, et al. Multi-modal magnetic resonance imaging alterations in two rat models of mild neurotrauma. J Neurotrauma 2007;24:1147–60.CrossRefGoogle ScholarPubMed
Phinney, DG, Baddoo, M, Dutreil, M, et al. Murine mesenchymal stem cells transplanted to the central nervous system of neonatal versus adult mice exhibit distinct engraftment kinetics and express receptors that guide neuronal cell migration. Stem Cells Dev 2006;15:437–7.CrossRefGoogle ScholarPubMed
Liesz, A, Suri-Payer, E, Veltkamp, C, et al. Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke. Nat Med 2009;15:192–9.CrossRefGoogle ScholarPubMed
de Paula, S, Vitola, AS, Greggio, S, 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.CrossRefGoogle Scholar
Yasuhara, T, Hara, K, Maki, M, et al. Mannitol facilitates neurotrophic factor up-regulation and behavioural recovery in neonatal hypoxic-ischaemic rats with human umbilical cord blood grafts. J Cell Mol Med 2010;14:914–21.CrossRefGoogle ScholarPubMed
Lee, JA, Kim, BI, Jo, CH, et al. Mesenchymal stem-cell transplantation for hypoxic-ischemic brain injury in neonatal rat model. Pediatr Res 2010;67:42–6.CrossRefGoogle ScholarPubMed
van Velthoven, CT, Kavelaars, A, van Bel, F, Heijnen, CJ. Repeated mesenchymal stem cell treatment after neonatal hypoxia-ischemia has distinct effects on formation and maturation of new neurons and oligodendrocytes leading to restoration of damage, corticospinal motor tract activity and sensorimotor function. J Neurosci 2010;30:9603–11.CrossRefGoogle ScholarPubMed
Yasuhara, T, Hara, K, Maki, M, et al. Intravenous grafts recapitulate the neurorestoration afforded by intracerebrally delivered multipotent adult progenitor cells in neonatal hypoxic-ischemic rats. J Cereb Blood Flow Metab 2008;28:1804–10.CrossRefGoogle ScholarPubMed
Ma, J, Wang, Y, Yang, J, et al. Treatment of hypoxic-ischemic encephalopathy in mouse by transplantation of embryonic stem cell-derived cells. Neurochem Int 2007;51:57–65.CrossRefGoogle ScholarPubMed
Gonzalez, FF, McQuillen, P, Mu, D, et al. Erythropoietin enhances long-term neuroprotection and neurogenesis in neonatal stroke. Dev Neurosci 2007;29:321–30.CrossRefGoogle ScholarPubMed
Jin-qiao, S, Bin, S, Wen-hao, Z, Yi, Y. Basic fibroblast growth factor stimulates the proliferation and differentiation of neural stem cells in neonatal rats after ischemic brain injury. Brain Dev 2009;31:331–40.CrossRefGoogle ScholarPubMed
Yata, K, Matchett, GA, Tsubokawa, T, et al. Granulocyte-colony stimulating factor inhibits apoptotic neuron loss after neonatal hypoxia-ischemia in rats. Brain Res 2007;1145:227–38.CrossRefGoogle ScholarPubMed
Katsuragi, S, Ikeda, T, Date, I, et al. Grafting of glial cell line-derived neurotrophic factor secreting cells for hypoxic-ischemic encephalopathy in neonatal rats. Am J Obstet Gynecol 2005;192:1137–45.CrossRefGoogle ScholarPubMed
Katsuragi, S, Ikeda, T, Date, I, et al. Implantation of encapsulated glial cell line-derived neurotrophic factor-secreting cells prevents long-lasting learning impairment following neonatal hypoxic-ischemic brain insult in rats. Am J Obstet Gynecol 2005;192:1028–37.CrossRefGoogle ScholarPubMed
Lorek, A, Takei, Y, Cady, EB, et al. Delayed (“secondary”) cerebral energy failure after acute hypoxia-ischemia in the newborn piglet: continuous 48-hour studies by phosphorus magnetic resonance spectroscopy. Pediatr Res 1994;36:699–706.CrossRefGoogle ScholarPubMed
Todd, MM, Chadwick, HS, Shapiro, HM, et al. The neurologic effects of thiopental therapy following experimental cardiac arrest in cats. Anesthesiology 1982;57:76.CrossRefGoogle ScholarPubMed
Franks, NP. General anaesthesia: from molecular targets to neuronal pathways of sleep and arousal. Nat Rev Neurosci 2008;9:370–86.CrossRefGoogle ScholarPubMed
Franks, NP, Dickinson, R, de Sousa, SL, Hall, AC, Lieb, WR. How does xenon produce anaesthesia?Nature 1998;396:324.CrossRefGoogle ScholarPubMed
Wilhelm, S, Ma, D, Maze, M, Franks, NP. Effects of xenon on in vitro and in vivo models of neuronal injury. Anesthesiology 2002;96:1485–91.CrossRefGoogle ScholarPubMed
Ma, D, Wilhelm, S, Maze, M, Franks, NP. Neuroprotective and neurotoxic properties of the ‘inert’ gas, xenon. Br J Anaesth 2002;89:739–46.CrossRefGoogle ScholarPubMed
Ma, D, Yang, H, Lynch, J, et al. Xenon attenuates cardiopulmonary bypass-induced neurologic and neurocognitive dysfunction in the rat. Anesthesiology 2003;98:690–8.CrossRefGoogle ScholarPubMed
Homi, HM, Yokoo, N, Ma, D, et al. The neuroprotective effect of xenon administration during transient middle cerebral artery occlusion in mice. Anesthesiology 2003;99:876–81.CrossRefGoogle ScholarPubMed
David, HN, Leveille, F, Chazalviel, L, et al. Reduction of ischemic brain damage by nitrous oxide and xenon. J Cereb Blood Flow Metab 2003;23:1168–73.CrossRefGoogle ScholarPubMed
Schmidt, M, Marx, T, Gloggl, E, Reinelt, H, Schirmer, U. Xenon attenuates cerebral damage after ischemia in pigs. Anesthesiology 2005;102:929–36.CrossRefGoogle ScholarPubMed
Sakamoto, S, Nakao, S, Masuzawa, M, et al. The differential effects of nitrous oxide and xenon on extracellular dopamine levels in the rat nucleus accumbens: a microdialysis study. Anesth Analg 2006;103:1459–63.CrossRefGoogle ScholarPubMed
Bantel, C, Maze, M, Trapp, S. Noble gas xenon is a novel adenosine triphosphate-sensitive potassium channel opener. Anesthesiology 2010;112:623–30.CrossRefGoogle ScholarPubMed
Gruss, M, Bushell, TJ, Bright, DP, et al. Two-pore-domain K+ channels are a novel target for the anesthetic gases xenon, nitrous oxide and cyclopropane. Mol Pharmacol 2004;65:443–52.CrossRefGoogle ScholarPubMed
Ma, D, Lim, T, Xu, J, et al. Xenon preconditioning protects against renal ischemic-reperfusion injury via HIF-1alpha activation. J Am Soc Nephrol 2009;20:713–20.CrossRefGoogle ScholarPubMed
Ma, D, Hossain, M, Chow, A, et al. Xenon and hypothermia combine to provide neuroprotection from neonatal asphyxia. Ann Neurol 2005;58:182–93.CrossRefGoogle ScholarPubMed
Dingley, J, Tooley, J, Porter, H, Thoresen, M. Xenon provides short-term neuroprotection in neonatal rats when administered after hypoxia-ischemia. Stroke 2006;37:501–6.CrossRefGoogle ScholarPubMed
Ma, D, Hossain, M, Pettet, GK, et al. Xenon preconditioning reduces brain damage from neonatal asphyxia in rats. J Cereb Blood Flow Metab 2006;26:199–208.CrossRefGoogle ScholarPubMed
Jawad, N, Rizvi, M, Gu, J, et al. Neuroprotection (and lack of neuroprotection) afforded by a series of noble gases in an in vitro model of neuronal injury. Neurosci Lett 2009;460:232–36.CrossRefGoogle Scholar
Luo, Y, Ma, D, Ieong, E, et al. Xenon and sevoflurane protect against brain injury in a neonatal asphyxia model. Anesthesiology 2008;109:782–9.CrossRefGoogle Scholar
Jevtovic-Todorovic, V, Hartman, RE, Izumi, Y, et al. Early exposure to common anesthetic agents causes widespread neurodegeneration in the developing rat brain and persistent learning deficits. J Neurosci 2003;23:876–82.CrossRefGoogle ScholarPubMed
Liang, G, Ward, C, Peng, J, et al. Isoflurane causes greater neurodegeneration than an equivalent exposure of sevoflurane in the developing brain of neonatal mice. Anesthesiology 2010;112:1325–34.CrossRefGoogle ScholarPubMed
Cattano, D, Young, C, Straiko, MM, Olney, JW. Subanesthetic doses of propofol induce neuroapoptosis in the infant mouse brain. Anesth Analg 2008;106:1712–4.CrossRefGoogle ScholarPubMed
Young, C, Jevtovic-Todorovic, V, Qin, YQ, et al. Potential of ketamine and midazolam, individually or in combination, to induce apoptotic neurodegeneration in the infant mouse brain. Br J Pharmacol 2005;146:189–97.CrossRefGoogle ScholarPubMed
Vutskits, L, Gascon, E, Tassonyi, E, Kiss, JZ. Effect of ketamine on dendritic arbor development and survival of immature GABAergic neurons in vitro. Toxicol Sci 2006;91:540–9.CrossRefGoogle ScholarPubMed
Ma, D, Williamson, P, Januszewski, A, et al. Xenon mitigates isoflurane-induced neuronal apoptosis in the developing rodent brain. Anesthesiology 2007;106:746–53.CrossRefGoogle ScholarPubMed
Sanders, RD, Xu, J, Shu, Y, et al. Dexmedetomidine attenuates isoflurane-induced neurocognitive impairment in neonatal rats. Anesthesiology 2009;110:1077–85.CrossRefGoogle ScholarPubMed
Lu, LX, Yon, JH, Carter, LB, Jevtovic-Todorovic, V. General anesthesia activates BDNF-dependent neuroapoptosis in the developing rat brain. Apoptosis 2006;11:1603–15.CrossRefGoogle ScholarPubMed
Sall, JW, Stratmann, G, Leong, J, et al. Isoflurane inhibits growth but does not cause cell death in hippocampal neural precursor cells grown in culture. Anesthesiology 2009;110:826–33.CrossRefGoogle Scholar
Loepke, AW, Soriano, SG. An assessment of the effects of general anesthetics on developing brain structure and neurocognitive function. Anesth Analg 2008;106:1681–707.CrossRefGoogle ScholarPubMed
Loepke, AW, McGowan, FX, Soriano, SG. CON: the toxic effects of anesthetics in the developing brain: the clinical perspective. Anesth Analg 2008;106:1664–9.CrossRefGoogle ScholarPubMed
Olney, JW, Young, C, Wozniak, DF, Ikonomidou, C, Jevtovic-Todorovic, V. Anesthesia-induced developmental neuroapoptosis. Does it happen in humans?Anesthesiology 2004;101:273–5.CrossRefGoogle ScholarPubMed
Todd, MM. Anesthetic neurotoxicity: the collision between laboratory neuroscience and clinical medicine. Anesthesiology 2004;101:272–3.CrossRefGoogle ScholarPubMed
Wilder, RT, Flick, RP, Sprung, J, et al. Early exposure to anesthesia and learning disabilities in a population-based birth cohort. Anesthesiology 2009;110:796–804.CrossRefGoogle Scholar
Bartels, M, Althoff, RR, Boomsma, DI. Anesthesia and cognitive performance in children: no evidence for a causal relationship. Twin Res Hum Genet 2009;12:246–53.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×