Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-skm99 Total loading time: 0 Render date: 2024-04-26T21:06:28.155Z Has data issue: false hasContentIssue false

2 - Mechanisms of neurodegeneration and therapeutics in animal models of neonatal hypoxic–ischemic encephalopathy

from Section 1 - Epidemiology, pathophysiology, and pathogenesis of fetal and neonatal brain injury

Published online by Cambridge University Press:  12 January 2010

David K. Stevenson
Affiliation:
Stanford University School of Medicine, California
William E. Benitz
Affiliation:
Stanford University School of Medicine, California
Philip Sunshine
Affiliation:
Stanford University School of Medicine, California
Susan R. Hintz
Affiliation:
Stanford University School of Medicine, California
Maurice L. Druzin
Affiliation:
Stanford University School of Medicine, California
Get access

Summary

Introduction

Perinatal hypoxia–ischemia (HI) and asphyxia due to umbilical cord prolapse, delivery complications, airway obstruction, asthma, drowning, and cardiac arrest are significant causes of brain damage, mortality, and morbidity in infants and young children. The incidence of HI encephalopathy (HIE), for example, is ∼ 2 to 4/1000 live term births. Term infants that experience episodes of asphyxia can have damage in the brainstem and forebrain, with the basal ganglia, particularly the striatum, and somatosensory systems showing selective vulnerability. Infants surviving with HIE can have long-term neurological disability, including disorders in movement, visual deficits, learning and cognition impairments, and epilepsy. Many of these neurological disabilities are contributors to the complex clinical syndrome of cerebral palsy. Neuroimaging studies of full-term neonates and experimental studies on animal models suggest that this pattern of selective vulnerability is related to local metabolism and brain regional interconnections that instigate and propagate the damage within specific neural systems. This idea has been called the “metabolism-connectivity concept”. The neurodegeneration is partly triggered by excitotoxic mechanisms resulting from excessive activation of excitatory glutamate receptors and oxidative stress. The ion channel N-methyl-d-aspartate (NMDA) receptor and intracellular signaling networks involving calcium, nitric oxide synthase (NOS), mitochondria, and reactive oxygen species (ROS), such as superoxide, nitric oxide (NO), peroxynitrite, and hydrogen peroxide (H2O2), appear to have instrumental roles in the neuronal cell death leading to perinatal HIE.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2009

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

Glass, HC, Ferriero, DM. Treatment of hypoxic–ischemic encephalopathy in newborns. Curr Treat Options Neurol 2007; 9: 414–23.CrossRefGoogle ScholarPubMed
Maller, AI, Hankins, LL, Yeakley, JW, et al. Rolandic type cerebral palsy in children as a pattern of hypoxic–ischemic injury in the full-term neonate. J Child Neurol 1998; 13: 313–21.CrossRefGoogle ScholarPubMed
Martin, LJ, Brambrink, A, Koehler, RC, et al. Primary sensory and forebrain motor systems in the newborn brain are preferentially damaged by hypoxia–ischemia. J Comp Neurol 1997; 377: 262–85.3.0.CO;2-1>CrossRefGoogle ScholarPubMed
Martin, LJ, Brambrink, A, Koehler, RC, et al. Neonatal asphyxic brain injury is neural system preferential and targets sensory-motor networks. In Stevenson, DK, Sunshine, P, eds., Fetal and Neonatal Brain Injury: Mechanisms, Management, and the Risks of Practice, 2nd edn. New York, NY: Oxford University Press, 1997: 374–99.Google Scholar
Volpe, JJ. Neurology of the Newborn. Philadelphia, PA: Saunders, 2001.Google Scholar
Johnston, MV, Nakajima, W, Hagberg, H. Mechanisms of hypoxic neurodegeneration in the developing brain. Neuroscientist 2002; 8: 212–20.CrossRefGoogle ScholarPubMed
Martin, LJ. Mechanisms of brain damage in animal models of hypoxia–ischemia in newborns. In Stevenson, DK, Benitz, WE, Sunshine, P, eds., Fetal and Neonatal Brain Injury: Mechanisms, Management, and the Risks of Practice, 3rd edn. Cambridge: Cambridge University Press, 2003: 30–57.CrossRefGoogle Scholar
McQuillen, PS, Ferriero, DM. Selective vulnerability in the developing central nervous system. Pediatr Neurol 2004; 30: 227–35.CrossRefGoogle ScholarPubMed
Blomgren, K, Hagberg, H. Free radicals, mitochondria, and hypoxia–ischemia in the developing brain. Free Rad Biol Med 2006; 40: 388–97.CrossRefGoogle ScholarPubMed
Hamrick, SE, Ferriero, DM. The injury response in the term newborn brain: can we neuroprotect? Curr Opin Neurol 2003; 16: 147–54.CrossRefGoogle ScholarPubMed
,Hypothermia after Cardiac Arrest Study Group. Mild therapeutic hypothermia to improve the neurologic outcome after cardiac arrest. N Engl J Med 2002; 346: 549–56.CrossRefGoogle Scholar
Shankaran, S, Laptook, AR, Ehrenkranz, RA, et al. Whole-body hypothermia for neonates with hypoxic–ischemic encephalopathy. N Engl J Med 2005; 353: 1574–84.CrossRefGoogle ScholarPubMed
Gluckman, PD, Wyatt, JS, Azzopardi, D, et al. Selective head cooling with mild systemic hypothermia after neonatal encephalopathy: multicentre randomized trial. Lancet 2005; 365: 663–70.CrossRefGoogle Scholar
Kanagawa, T, Fukuda, H, Tsubouchi, H, et al. A decrease of cell proliferation by hypothermia in the hippocampus of the neonatal rat. Brain Res 2006; 1111: 36–40.CrossRefGoogle ScholarPubMed
Lockshin, RA, Zakeri, Z. Caspase-independent cell deaths. Curr Opin Cell Biol 2002; 14: 727–33.CrossRefGoogle ScholarPubMed
Gilbert, SF. Developmental Biology. Sunderland, MA: Sinauer Associates, 2006.Google Scholar
Klionsky, DJ, Emr, SD. Autophagy as a regulated pathway of cellular degradation. Science 2000; 290: 1717–21.CrossRefGoogle ScholarPubMed
Portera-Cailliau, C, Price, DL, Martin, LJ. Excitotoxic neuronal death in the immature brain is an apoptosis–necrosis morphological continuum. J Comp Neurol 1997; 378: 70–87.Google ScholarPubMed
Portera-Cailliau, C, Price, DL, Martin, LJ. Non-NMDA and NMDA receptor-mediated excitotoxic neuronal deaths in adult brain are morphologically distinct: further evidence for an apoptosis–necrosis continuum. J Comp Neurol 1997; 378: 88–104.3.0.CO;2-G>CrossRefGoogle ScholarPubMed
Martin, LJ, Al-Abdulla, NA, Brambrink, AM, et al. Neurodegeneration in excitotoxicity, global cerebral ischemia, and target deprivation: a perspective on the contributions of apoptosis and necrosis. Brain Res Bull 1998; 46: 281–309.CrossRefGoogle ScholarPubMed
Martin, LJ. Neuronal death in amyotrophic lateral sclerosis is apoptosis: possible contribution of a programmed cell death mechanism. J Neuropathol Exp Neurol 1999; 58: 459–71.CrossRefGoogle ScholarPubMed
Nakajima, W, Ishida, A, Lange, MS, et al. Apoptosis has a prolonged role in the neurodegeneration after hypoxic ischemia in the newborn rat. J Neurosci 2000; 20: 7994–8004.CrossRefGoogle Scholar
Northington, FJ, Ferriero, DM, Graham, EM, et al. Early neurodegeneration after hypoxia–ischemia in neonatal rat is necrosis while delayed neuronal death is apoptosis. Neurobiol Dis 2001; 8: 207–19.CrossRefGoogle ScholarPubMed
Northington, FJ, Zelaya, ME, O'Riordan, DP, et al. Failure to complete apoptosis following neonatal hypoxia–ischemia manifests as “continuum” phenotype of cell death and occurs with multiple manifestations of mitochondrial dysfunction in rodent forebrain. Neuroscience 2007; 149: 822–33.CrossRefGoogle ScholarPubMed
Lennon, SV, Martin, SJ, Cotter, TG. Dose-dependent induction of apoptosis in human tumour cell lines by widely diverging stimuli. Cell Prolif 1991; 24: 203–14.CrossRefGoogle ScholarPubMed
Fernandes, RS, Cotter, TG. Apoptosis or necrosis: intracellular levels of glutathione influence mode of cell death. Biochem Pharmacol 1994; 48: 675–81.CrossRefGoogle ScholarPubMed
Bonfoco, E, Krainc, D, Ankarcrona, M, et al. Apoptosis and necrosis: two distinct events induced, respectively, by mild and intense insults with N-methyl-D-aspartate or nitric oxide/superoxide in cortical cell culture. Proc Natl Acad Sci USA 1995; 92: 7162–6.CrossRefGoogle ScholarPubMed
Raffray, M, Cohen, GM. Apoptosis and necrosis in toxicology: a continuum or distinct modes of cell death? Pharmacol Ther 1997; 75: 153–77.CrossRefGoogle ScholarPubMed
Yuan, J, Lipinski, M, Degterev, A. Diversity in the mechanisms of neuronal cell death. Neuron 2003; 40: 401–13.CrossRefGoogle ScholarPubMed
Orrenius, S, Zhivotovsky, B, Nicotera, P. Regulation of cell death: the calcium–apoptosis link. Nat Rev Mol Cell Biol 2003; 4: 552–65.CrossRefGoogle ScholarPubMed
Trump, BF, Berezesky, IK. The role of altered [Ca2+]i regulation in apoptosis, oncosis, and necrosis. Biochim Biophys Acta 1996; 1313: 173–8.CrossRefGoogle ScholarPubMed
Majno, G, Joris, I. Apoptosis, oncosis, and necrosis: an overview of cell death. Am J Pathol 1995; 146: 3–15.Google ScholarPubMed
Trump, BF, Goldblatt, PJ, Stowell, RE. Studies on necrosis of mouse liver in vitro: ultrastructural alterations in the mitochondria of hepatic parenchymal cells. Lab Invest 1964; 14: 343–71.Google Scholar
Leist, M, Single, B, Castoldi, AF, et al. Intracellular adenosine triphosphate (ATP) concentration: a switch in the decision between apoptosis and necrosis. J Exp Med 1997; 185: 1481–6.CrossRefGoogle ScholarPubMed
Martin, LJ, Brambrink, AM, Price, AC, et al. Neuronal death in newborn striatum after hypoxia–ischemia is necrosis and evolves with oxidative stress. Neurobiol Dis 2000; 7: 169–91.CrossRefGoogle ScholarPubMed
Golden, WC, Brambrink, AM, Traystman, RJ, et al. Failure to sustain recovery of Na,K-ATPase function is a possible mechanism for striatal neurodegeneration in hypoxic–ischemic newborn piglets. Brain Res Mol Brain Res 2001; 88: 94–102.CrossRefGoogle ScholarPubMed
Castro, J, Ruminot, I, Porras, OH, et al. ATP steal between cation pumps: a mechanism linking Na+ influx to the onset or necrotic Ca2+ overload. Cell Death Diff 2006; 13: 1675–85.CrossRefGoogle ScholarPubMed
Proskuryakov, SY, Konoplyannikov, AG, Gabai, VL. Necrosis: a specific form of programmed cell death. Exp Cell Res 2003; 283: 1–16.CrossRefGoogle ScholarPubMed
Ha, HC, Snyder, SH. Poly(ADP-ribose) polymerase-1 in the nervous system. Neurobiol Dis 2000; 7: 225–39.CrossRefGoogle Scholar
Crompton, M. The mitochondrial permeability transition pore and its role in cell death. Biochem J 1999; 341: 233–49.CrossRefGoogle ScholarPubMed
Gurp, M, Festjens, N, Loo, G, et al. Mitochondrial intermembrane proteins in cell death. Biochem Biophys Res Comm 2003; 304: 487–97.CrossRefGoogle ScholarPubMed
Crompton, M, Virji, S, Ward, JM. Cyclophilin-D binds strongly to complexes of the voltage-dependent anion channel and the adenine nucleotide translocase to form the permeability transition pore. Eur J Biochem 1998; 258: 729–35.CrossRefGoogle ScholarPubMed
Baines, CP, Kaiser, RA, Purcell, NH, et al. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature 2005; 434: 658–62.CrossRefGoogle ScholarPubMed
Nakagawa, T, Shimizu, S, Watanabe, T, et al. Cyclophilin D-dependent mitochondrial transition regulates some necrotic but not apoptotic cell death. Nature 2005; 434: 652–8.CrossRefGoogle Scholar
Vande Velde, C, Cizeau, J, Dubik, D, et al. BNIP3 and genetic control of necrosis-like cell death through the mitochondrial permeability transition pore. Mol Cell Biol 2000; 20: 5454–68.CrossRefGoogle ScholarPubMed
Tata, JR. Requirement for RNA and protein synthesis for induced regression of tadpole tail in organ culture. Dev Biol 1966; 13: 77–94.CrossRefGoogle ScholarPubMed
Schwartz, LM, Smith, SW, Jones, MEE, et al. Do all programmed cell deaths occur via apoptosis?Proc Natl Acad Sci USA 1993; 90: 980–4.CrossRefGoogle ScholarPubMed
Amin, F, Bowen, ID, Szegedi, Z, et al. Apoptotic and non-apoptotic modes of programmed cell death in MCF-7 human breast carcinoma cells. Cell Biol Intl 2000; 24: 253–60.CrossRefGoogle ScholarPubMed
Jacobson, M. Developmental Neurobiology. New York, NY: Plenum Press, 1991.CrossRefGoogle Scholar
Glücksmann, A. Cell deaths in normal vertebrate ontogeny. Biol Rev 1951; 26: 59–86.CrossRefGoogle ScholarPubMed
Lockshin, RA, Williams, CM. Programmed cell death: II. Endocrine potentiation of the breakdown of the intersegmental muscles of silkmoths. J Insect Physiol 1964; 10: 643–9.CrossRefGoogle Scholar
Saunders, JW. Death in embryonic systems. Science 1966; 54: 604–12.CrossRefGoogle Scholar
Clarke, PGH. Developmental cell death: morphological diversity and multiple mechanisms. Anat Embryol 1990; 181: 195–213.CrossRefGoogle ScholarPubMed
Bursch, W, Paffe, S, Putz, B, et al. Determination of the length of the histological stages of apoptosis in normal liver and in altered hepatic foci of rats. Carcinogenesis 1990; 11: 847–53.CrossRefGoogle ScholarPubMed
Wyllie, AH, Kerr, JFR, Currie, AR. Cell death: the significance of apoptosis. Int Rev Cytol 1980; 68: 251–306.CrossRefGoogle Scholar
Nagata, S. Fas ligand-induced apoptosis. Annu Rev Genet 1999; 33: 29–55.CrossRefGoogle ScholarPubMed
Kerr, JFR, Wyllie, AH, Currie, AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 1972; 26: 239–57.CrossRefGoogle ScholarPubMed
Lockshin, RA, Zakeri, Z. Programmed cell death and apoptosis: origins of the theory. Nat Rev Mol Cell Biol 2001; 2: 545–50.CrossRefGoogle Scholar
Wyllie, AH. Glucocorticoid-induced thymocyte apoptosis is associated with enodgenous endonuclease activation. Nature 1980; 284: 555–6.CrossRefGoogle Scholar
Liu, X, Zou, H, Slaughter, C, et al. DFF, a heterodimeric protein that functions downstream of caspase-3 to trigger DNA fragmentation during apoptosis. Cell 1997; 89: 175–84.CrossRefGoogle ScholarPubMed
Sakahira, H, Enari, M, Ohsawa, Y, et al. Apoptotic nuclear morphological change without DNA fragmentation. Curr Biol 1999; 9: 543–6.CrossRefGoogle ScholarPubMed
Pilar, G, Landmesser, L. Ultrastructural differences during embryonic cell death in normal and peripherally deprived ciliary ganglia. J Cell Biol 1976; 68: 339–56.CrossRefGoogle ScholarPubMed
Lockshin, RA, Zakeri, A. Programmed cell death: early changes in metamorphosing cells. Biochem Cell Biol 1994; 72: 589–96.CrossRefGoogle ScholarPubMed
Schweichel, JU, Merker, HJ. The morphology of various types of cell death in prenatal tissues. Teratology 1973; 7: 253–66.CrossRefGoogle ScholarPubMed
Xue, LZ, Fletcher, GC, Tolkovsky, AM. Autophagy is activated by apoptotic signalling in sympathetic neurons: an alternative mechanism of death execution. Mol Cell Neurosci 1999; 14: 180–98.CrossRefGoogle ScholarPubMed
Yue, Z, Horton, A, Bravin, M, et al. A novel protein complex linking the δ 2 glutamate receptor and autophagy: implications for neurodegeneration in Lurcher mice. Neuron 2002; 35: 921–33.CrossRefGoogle ScholarPubMed
Koike, M, Shibata, M, Tadakoshi, M, et al. Inhibition of autophagy prevents hippocampal pyramidal neuron death after hypoxic–ischemic injury. Am J Pathol 2008; 172: 454–69.CrossRefGoogle ScholarPubMed
Mizushima, N, Ohsumi, Y, Yoshimori, T. Autophagosome formation in mammalian cells. Cell Struct Funct 2002; 27: 421–9.CrossRefGoogle ScholarPubMed
Bursch, W. The autophagosomal–lysosomal compartment in programmed cell death. Cell Death Diff 2001; 8: 569–81.CrossRefGoogle ScholarPubMed
Inbal, B, Bialik, S, Sabanay, I, et al. DAP kinase and DRP-1 mediate membrane blebbing and the formation of autophagic vesicles during programmed cell death. J Cell Biol 2002; 157: 455–68.CrossRefGoogle ScholarPubMed
Liang, XH, Kleeman, LK, Jiang, HH, et al. Protection against fatal sindbis virus encephalitis by beclin, a novel Bcl-2-interacting protein. J Virol 1998; 72: 8586–96.Google ScholarPubMed
Ogier-Denis, E, Codogno, P. Autophagy: a barrier or an adaptive response to cancer. Biochim Biophys Acta 2003; 1603: 113–28.Google ScholarPubMed
Ameisen, JC. On the origin, evolution, and nature of programmed cell death: a timeline of four billion years. Cell Death Diff 2002; 9: 367–93.CrossRefGoogle ScholarPubMed
Metzstein, MM, Stanfield, , Horvitz, NR. Genetics of programmed cell death in C. elegans: past, present and future. Trends Genet 1998; 14: 410–16.CrossRefGoogle Scholar
Cory, S, Adams, JM. The Bcl2 family: regulators of the cellular life-or-death switch. Nat Rev Cancer 2002; 2: 647–56.CrossRefGoogle ScholarPubMed
Wolf, BB, Green, DR. Suicidal tendencies: apoptotic cell death by caspase family proteinases. J Biol Chem 1999; 274: 20049–52.CrossRefGoogle ScholarPubMed
Levrero, M, Laurenzi, V, Costanzo, A, et al. The p53/p63/p73 family of transcription factors: overlapping and distinct functions. J Cell Sci 2000; 113: 1661–70.Google ScholarPubMed
Hegde, R, Srinivasula, SM, Zhang, Z, et al. Identification of Omi/HtrA2 as a mitochondrial apoptotic serine protease that disrupts inhibitor of apoptosis protein-caspase interaction. J Biol Chem 2002; 277: 432–8.CrossRefGoogle ScholarPubMed
Li, P, Nijhawan, D, Budihardjo, I, et al. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 1997; 91: 479–89.CrossRefGoogle ScholarPubMed
Liu, X, Kim, CN, Yang, J, et al. Induction of apoptotic program in cell-free extracts: Requirement for dATP and cytochrome c. Cell 1996; 86: 147–57.CrossRefGoogle ScholarPubMed
Klein, JA, Longo-Guess, CM, Rossmann, MP, et al. The harlequin mouse mutation downregulates apoptosis-inducing factor. Nature 2002; 419: 367–74.CrossRefGoogle ScholarPubMed
Bekkum, DW. The effect of x-rays on phosphorylations in vivo. Biochim Biophys Acta 1957; 25: 487–92.CrossRefGoogle Scholar
Scorrano, L, Oakes, SA, Opferman, TJ, et al. Bax and Bak regulation of endoplasmic reticulum Ca2+: a control point for apoptosis. Science 2003; 300: 135–9.CrossRefGoogle ScholarPubMed
Muchmore, SW, Sattler, M, Liang, H, et al. X-ray and NMR structure of human Bcl-xL, an inhibitor of programmed cell death. Nature 1999; 381: 335–41.CrossRefGoogle Scholar
Martin, LJ, Price, AC, McClendon, KB. Early events of target deprivation/axotomy-induced neuronal apoptosis in vivo: oxidative stress, DNA damage, p53 phosphorylation and subcellular redistribution of death proteins. J Neurochem 2003; 85: 234–47.CrossRefGoogle ScholarPubMed
Wolter, KG, Hsu, YT, Smith, CL, et al. Movement of Bax from the cytosol to mitochondria during apoptosis. J Cell Biol 1997; 139: 1281–92.CrossRefGoogle ScholarPubMed
Nechushtan, A, Smith, CL, Lamensdorf, I, et al. Bax and Bak coalesce into novel mitochondria-associated clusters during apoptosis. J Cell Biol 2001; 153: 1265–76.CrossRefGoogle ScholarPubMed
Antonsson, B, Conti, F, Ciavatta, A, et al. Inhibition of Bax channel-forming activity by bcl-2. Science 1997; 277: 370–2.CrossRefGoogle ScholarPubMed
Shimizu, S, Ide, T, Yanagida, T, et al. Electrophysiological study of a novel large pore formed by Bax and the voltage-dependent anion channel that is permeable to cytochrome c. J Biol Chem 2000; 275: 12321–5.CrossRefGoogle ScholarPubMed
Kluck, RM, Bossy-Wetzel, E, Green, DR, et al. The release of cytochrome c from mitochondria: a primary site for bcl-2 regulation of apoptosis. Science 1997; 275: 1132–6.CrossRefGoogle ScholarPubMed
Yang, J, Liu, X, Bhalla, K, et al. Prevention of apoptosis by bcl-2: release of cytochrome c from mitochondria blocked. Science 1997; 275: 1129–32.CrossRefGoogle ScholarPubMed
Vander Heiden, MG, Chandel, NS, Williamson, EK, et al. Bcl-xL regulates the membrane potential and volume homeostasis of mitochondria. Cell 1997; 91: 627–37.CrossRefGoogle ScholarPubMed
Hu, Y, Benedict, MA, Wu, D, et al. Bcl-xL interacts with Apaf-1 and inhibits Apaf-1-dependent caspase-9 activation. Proc Natl Acad Sci USA 1998; 95: 4386–91.CrossRefGoogle ScholarPubMed
Song, Q, Kuang, Y, Dixit, VM, et al. Boo, a negative regulator of cell death, interacts with Apaf-1. EMBO J 1999; 18: 167–78.CrossRefGoogle ScholarPubMed
Wei, MC, Zong, WX, Cheng, EHY. Proapoptotic Bax and Bak: a requisite gateway to mitochondrial dysfunction and death. Science 2001; 292: 727–30.CrossRefGoogle ScholarPubMed
Letai, A, Bassik, MC, Walensky, LD. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell 2001; 2: 183–92.CrossRefGoogle Scholar
Puka-Sundvall, M, Gajkowska, B, Cholewinski, M, et al. Subcellular distribution of calcium and ultrastructural changes after cerebral hypoxia–ischemia in immature rats. Dev Brain Res 2000; 125: 31–41.CrossRefGoogle ScholarPubMed
Lithgow, T, Driel, R, Bertram, JF, et al. The protein product of the oncogene bcl-2 is a component of the nuclear envelope, the endoplasmic reticulum, and the outer mitochondrial membrane. Cell Growth Differ 1994; 5: 411–17.Google ScholarPubMed
Murakami, Y, Aizu-Yokota, E, Sonoda, Y, et al. Suppression of endoplasmic stress- induced caspase activation and cell death by overexpression of Bcl-xL or Bcl-2. J Biochem 2007; 141: 401–10.CrossRefGoogle ScholarPubMed
Zong, WX, Li, C, Hatzivassiliou, G, et al. Bax and Bak can localize to the endoplasmic reticulum to initiate apoptosis. J Cell Biol 2003; 162: 59–69.CrossRefGoogle ScholarPubMed
Haldar, S, Jena, N, Croce, CM. Inactivation of Bcl-2 by phosphorylation. Proc Natl Acad Sci USA 1995; 92: 4507–11.CrossRefGoogle ScholarPubMed
Hallin, U, Kondo, E, Ozaki, Y, et al. Bcl-2 phosphorylation in the BH4 domain precedes caspase-3 activation and cell death after neonatal cerebral hypoxic–ischemic injury. Neurobiol Dis 2006; 21: 478–86.CrossRefGoogle ScholarPubMed
Wang, HG, Rapp, UR, Reed, JC. Bcl-2 targets the protein kinase raf-1 to mitochondria. Cell 1996; 87: 629–38.CrossRefGoogle Scholar
Datta, SR, Dudek, H, Tao, X. Akt phosphorylation of Bad couples survival signals to the cell-intrinsic death machinery. Cell 1997; 91: 231–41.CrossRefGoogle ScholarPubMed
del Peso, L, Gonzlez-Garcia, M, Page, C, et al. Interleukin-3-induced phosphorylation of Bad through the protein kinase Akt. Science 1997; 278: 687–9.CrossRefGoogle ScholarPubMed
Cardone, MH, Roy, N, Stennicke, HR. Regulation of cell death protease caspase-9 by phosphorylation. Science 1998; 282: 1318–21.CrossRefGoogle ScholarPubMed
Zha, J, Harada, H, Yang, E, et al. Serine phosphorylation of death agonist Bad in response to survival factor results in binding to 14–3–3 not Bcl-xL. Cell 1996; 87: 619–28.CrossRefGoogle ScholarPubMed
Wang, H-G, Pathan, N, Ethell, IM, et al. Ca2+-induced apoptosis through calcineurin dephosphorylation of Bad. Science 1999; 284: 339–43.CrossRefGoogle ScholarPubMed
Yang, E, Zha, J, Jockel, J. Bad, a heterodimeric partner for Bcl-xL and Bcl-2, displaces Bax and promotes cell death. Cell 1995; 80: 285–91.CrossRefGoogle ScholarPubMed
Danial, NN, Gramm, CF, Scorrano, L. Bad and glucokinase reside in a mitochondrial complex that integrates glycolysis and apoptosis. Nature 2003; 424: 952–6.CrossRefGoogle Scholar
Stennicke, HR, Deveraux, QL, Humke, EW. Caspase-9 can be activated without proteolytic processing. J Biol Chem 1999; 274: 8359–62.CrossRefGoogle ScholarPubMed
Schwartz, LM, Milligan, CE. Cold thoughts of death: the role of ICE proteases in neuronal cell death. Trends Neurosci 1996; 19: 555–62.CrossRefGoogle ScholarPubMed
Zou, H, Li, Y, Liu, X, et al. An Apaf-1-cytochrome c multimeric complex is a functional apoptosome that activates procaspase-9. J Biol Chem 1999; 274: 11549–56.CrossRefGoogle ScholarPubMed
Li, H, Zhu, H, Xu, CJ, et al. Cleavage of Bid by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell 1989; 94: 491–501.CrossRefGoogle Scholar
Robertson, JD, Enoksson, M, Suomela, M, et al. Caspase-2 acts upstream of mitochondria to promote cytochrome c release during etoposide-induced apoptosis. J Biol Chem 2002; 277: 29803–9.CrossRefGoogle ScholarPubMed
Beresford, PJ, Zhang, D, Oh, DY, et al. Granzyme A activates an endoplasmic reticulum-associated caspase-independent nuclease to induce single-stranded DNA nicks. J Biol Chem 2001; 76: 43285–93.CrossRefGoogle Scholar
Fan, Z, Beresford, PJ, Oh, DY, et al. Tumor suppressor NM23-H1 is a granzyme A-activated DNase during TL-mediated apoptosis, and the nucleosome assembly protein SET is its inhibitor. Cell 2003; 112: 659–72.CrossRefGoogle Scholar
LaCasse, EC, Baird, S, Korneluk, RG, et al. The inhibitors of apoptosis (IAPs) and their emerging role in cancer. Oncogene 1998; 17: 3247–59.CrossRefGoogle Scholar
Holcik, M. The IAP proteins. Trends Genet 2002; 18: 537–8.CrossRefGoogle ScholarPubMed
Deveraux, QL, Roy, N, Stennicke, HR, et al. IAPs block apoptotic events induced by caspase-8 and cytochrome c by direct inhibition of distinct caspases. EMBO J 1998; 17: 2215–23.CrossRefGoogle ScholarPubMed
Hao, Y, Sekine, K, Kawabata, A, et al. Apollon ubiquitinates SMAC and caspase-9 and has essential cytoprotective function. Nat Cell Biol 2004; 6: 849–60.CrossRefGoogle Scholar
Jiang, Y, Bruin, A, Caldas, H, et al. Essential role for survivin in early brain development. J Neurosci 2005; 25: 6962–70.CrossRefGoogle ScholarPubMed
Xu, DG, Korneluk, RG, Tamai, K, et al. Distribution of neuronal apoptosis inhibitory protein-like immunoreactivity in the rat central nervous system. J Comp Neurol 1997; 382: 247–59.3.0.CO;2-3>CrossRefGoogle ScholarPubMed
Martin, LJ, Liu, Z, Chen, K, et al. Motor neuron degeneration in amyotrophic lateral sclerosis mutant superoxide dismutase-1 transgenic mice: mechanisms of mitochondriopathy and cell death. J Comp Neurol 2007; 500: 20–46.CrossRefGoogle ScholarPubMed
Roy, N, Mahadevan, MS, McLean, M, et al. The gene for neuronal apoptosis inhibitory protein is partially deleted in individuals with spinal muscular atrophy. Cell 1995; 80: 167–78.CrossRefGoogle ScholarPubMed
Du, C, Fang, M, Li, Y, et al. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000; 102: 33–42.CrossRefGoogle ScholarPubMed
Verhagen, AM, Ekert, PG, Pakusch, M, et al. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 2000; 102: 43–53.CrossRefGoogle ScholarPubMed
Suzuki, Y, Takahashi-Niki, K, Akagi, T, et al. Mitochondrial protease Omi/HtrA2 enhances caspase activation through multiple pathways. Cell Death Diff 2004; 11: 208–16.CrossRefGoogle ScholarPubMed
Susin, SA, Lorenzo, HK, Zamzami, N, et al. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 1999; 397: 441–6.CrossRefGoogle ScholarPubMed
Mate, MJ, Ortiz-Lombardia, M, Boitel, B, et al. The crystal structure of the mouse apoptosis-inducing factor AIF. Nat Struct Biol 2002; 9: 442–6.CrossRefGoogle ScholarPubMed
Desagher, S, Osen-Sand, A, Nichols, A, et al. Bid-induced conformational change of bax is responsible for mitochondrial cytochrome c release during apoptosis. J Cell Biol 1999; 144: 891–901.CrossRefGoogle ScholarPubMed
Troy, CM, Friedman, JE, Friedman, WJ. Mechanisms of p75-mediated death of hippocampal neurons: role of caspases. J Biol Chem 2002; 277: 34295–302.CrossRefGoogle ScholarPubMed
Giaccia, AJ, Kastan, MB. The complexity of p53 modulation: emerging patterns from divergent signals. Genes Develop 1998; 12: 2973–83.CrossRefGoogle ScholarPubMed
Chang, YC, Lee, YS, Tejima, T, et al. Mdm-2 and bax, downstream mediators of the p53 response, are degraded by the ubiquitin-proteasome pathway. Cell Growth Diff 1998; 9: 79–84.Google Scholar
Maki, CG, Huibregtse, JM, Howley, PM. In vivo ubiquitination and proteasome-mediated degradation of p53. Cancer Res 1996; 56: 2649–54.Google ScholarPubMed
Shieh, SY, Ikeda, M, Taya, Y, et al. DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell 1997; 91: 325–34.CrossRefGoogle ScholarPubMed
Villunger, A, Michalak, EM, Coultas, L, et al. p53- and drug-induced apoptotic responses mediated by BH3-only proteins Puma and Noxa. Science 2003; 302: 1036–8.CrossRefGoogle ScholarPubMed
Aloyz, RS, Bamji, SX, Pozniak, CD, et al. p53 is essential for developmental neuron death regulated by the TrkA and p75 neurotrophin receptors. J Cell Biol 1998; 143: 1691–703.CrossRefGoogle ScholarPubMed
Martin, LJ, Kaiser, A, Yu, JW, et al. Injury-induced apoptosis of neurons in adult brain is mediated by p53-dependent and p53-independent pathways and requires Bax. J Comp Neurol 2001; 433: 299–311.CrossRefGoogle ScholarPubMed
Martin, LJ, Liu, Z. Injury-induced spinal motor neuron apoptosis is preceded by DNA single-strand breaks and is p53- and Bax-dependent. J Neurobiol 2002; 50: 181–97.CrossRefGoogle ScholarPubMed
Pozniak, CD, Radinovic, S, Yang, A, et al. An anti-apoptotic role for the p53 family member, p73, during developmental neuron death. Science 2000; 289: 304–6.CrossRefGoogle ScholarPubMed
Lucas, DR, Newhouse, JP. The toxic effect of sodium L-glutamate on the inner layers of the retina. Arch Ophthal 1957; 58: 193–201.CrossRefGoogle ScholarPubMed
Olney, JW. Glutamate-induced neuronal necrosis in the infant mouse hypothalamus: an electron microscopic study. J Neuropathol Exp Neurol 1971; 30: 75–90.CrossRefGoogle Scholar
Choi, DW. Excitotoxic cell death. J Neurobiol 1992; 23: 1261–76.CrossRefGoogle ScholarPubMed
Martin, LJ, Sieber, FE, Traystman, RJ. Apoptosis and necrosis occur in separate neuronal populations in hippocampus and cerebellum after ischemia and are associated with alterations in metabotropic glutamate receptor signaling pathways. J Cereb Blood Flow Metab 2000; 20: 153–67.CrossRefGoogle ScholarPubMed
Lipton, SA, Rosenberg, PA. Excitatory amino acids as a final common pathway for neurologic disorders. N Engl J Med 1994; 330: 613–22.Google ScholarPubMed
Gwag, BJ, Koh, JY, DeMaro, JA, et al. Slowly triggered excitotoxicity occurs by necrosis in cortical cultures. Neuroscience 1997; 77: 393–401.CrossRefGoogle ScholarPubMed
Kure, S, Tominaga, T, Yoshimoto, T, et al. Glutamate triggers internucleosomal DNA cleavage in neuronal cells. Biochem Biophys Res Commun 1991; 179: 39–45.CrossRefGoogle ScholarPubMed
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: 961–73.CrossRefGoogle ScholarPubMed
Simonian, NA, Getz, RL, Leveque, JC, et al. Kainate induces apoptosis in neurons. Neuroscience 1996; 74: 675–83.CrossRefGoogle ScholarPubMed
Dessi, F, Charriaut-Marlangue, C, Khrestchatisky, M, et al. Glutamate-induced neuronal death is not a programmed cell death in cerebellar culture. J Neurochem 1993; 60: 1953–5.CrossRefGoogle Scholar
Xiang, H, Kinoshita, Y, Knudson, CM, et al. Bax involvement in p53-mediated neuronal cell death. J Neurosci 1998; 18: 1363–73.CrossRefGoogle ScholarPubMed
Miller, TM, Moulder, KL, Knudson, CM, et al. Bax deletion further orders the cell death pathway in cerebellar granule cells and suggests a caspase-independent pathway to cell death. J Cell Biol 1997; 139: 205–17.CrossRefGoogle ScholarPubMed
Dargusch, R, Piasecki, D, Tan, S, et al. The role of Bax in glutamate-induced nerve cell death. J Neurochem 2001; 76: 295–301.CrossRefGoogle ScholarPubMed
Johnson, MD, Kinoshita, Y, Xiang, H, et al. Contribution of p53-dependent caspase activation to neuronal cell death declines with neuronal maturation. J Neurosci 1999; 19: 2996–3006.CrossRefGoogle ScholarPubMed
Tenneti, L, Lipton, SA. Involvement of activated caspase-3-like proteases in N-methyl-D-aspartate-induced apoptosis in cerebrocortical neurons. J Neurochem 2001; 74: 134–42.CrossRefGoogle Scholar
Simons, M, Beinroth, S, Gleichmann, M, et al. Adenovirus-mediated gene transfer of inhibitors of apoptosis proteins delays apoptosis in cerebellar granule neurons. J Neurochem 1999; 72: 292–301.CrossRefGoogle ScholarPubMed
Lookeren Campagne, M, Lucassen, PJ, Vermeulen, JP, et al. NMDA and kainate induced internucleosomal DNA cleavage associated with both apoptotic and necrotic cell death in the neonatal rat brain. Eur J Neurosci 1995; 7: 1627–40.CrossRefGoogle ScholarPubMed
Holcik, M, Thompson, CS, Yaraghi, Z, et al. The hippocampal neurons of neuronal apoptosis inhibitory protein 1 (NAIP1)-deleted mice display increased vulnerability to kainic acid-induced injury. Proc Natl Acad Sci USA 1999; 97: 2286–90.CrossRefGoogle Scholar
Schreiber, SS, Tocco, G, Najm, I, et al. Cycloheximide prevents kainate-induced neuronal death and c-fos expression in adult rat brain. J Mol Neurosci 1993; 4: 149–59.CrossRefGoogle ScholarPubMed
Leppin, C, Finiels-Marlier, F, Crawley, JN, et al. Failure of a protein synthesis inhibitor to modify glutamate receptor-mediated neurotoxicity in vivo. Brain Res 1992; 581: 168–70.CrossRefGoogle ScholarPubMed
Lok, J, Martin, LJ. Rapid subcellular redistribution of Bax precedes caspase-3 and endonuclease activation during excitotoxic neuronal apoptosis in rat brain. J Neurotrauma 2002; 19: 815–28.CrossRefGoogle ScholarPubMed
Martin, LJ. Neuronal cell death in nervous system development, disease, and injury. Int J Mol Med 2001; 7: 455–78.Google ScholarPubMed
Natale, JE, Cheng, Y, Martin, LJ. Thalamic neuron apoptosis emerges rapidly after cortical damage in immature mice. Neuroscience 2002; 112: 665–76.CrossRefGoogle ScholarPubMed
Fujikawa, DG. Confusion between neuronal apoptosis and activation of programmed cell death mechanisms in acute necrotic insults. Trends Neurosci 2000; 23: 410–11.CrossRefGoogle ScholarPubMed
Ishimaru, MJ, Ikonomidou, C, Tenkova, TI, et al. Distinguishing excitotoxic from apoptotic neurodegeneration in the developing rat brain. J Comp Neurol 1999; 408: 461–76.3.0.CO;2-9>CrossRefGoogle ScholarPubMed
Sloviter, RS. Apoptosis: a guide for the perplexed. Trends Pharmacol Sci 2002; 23: 19–24.CrossRefGoogle ScholarPubMed
Northington, FJ, Graham, EM, Martin, LJ. Apoptosis in perinatal hypoxic–ischemic brain injury: how important is it and should it be inhibited?Brain Res Rev 2005; 50: 244–57.CrossRefGoogle ScholarPubMed
Baille, V, Clarke, PGH, Brocher, G, et al. Soman-induced convulsions: the neuropathology revisited. Toxicology 2005; 215: 1–24.CrossRefGoogle ScholarPubMed
Zhu, C, Wang, X, Xu, F, et al. The influence of age on apoptotic and other mechanisms of cell death after cerebral hypoxia–ischemia. Cell Death Diff 2005; 12: 162–76.CrossRefGoogle ScholarPubMed
Sheldon, RA, Hall, JJ, Noble, LJ, et al. Delayed cell death in neonatal mouse hippocampus from hypoxia–ischemia is neither apoptotic nor necrotic. Neurosci Lett 2001; 304: 165–8.CrossRefGoogle ScholarPubMed
Wei, L, Ying, D-J, Cui, L, et al. Necrosis, apoptosis, and hybrid death in the cortex and thalamus after barrel cortex ischemia in rats. Brain Res 2004; 1022: 54–61.CrossRefGoogle ScholarPubMed
Bruijn, LI. Amyotrophic lateral sclerosis: from disease mechanisms to therapies. BioTechniques 2002; 32: 1112–21.Google ScholarPubMed
Formigli, L, Papucci, L, Tani, A, et al. Aponecrosis: morphological and biochemical exploration of a syncretic process of cell death sharing apoptosis and necrosis. J Cell Physiol 2000; 182: 41–9.3.0.CO;2-7>CrossRefGoogle ScholarPubMed
Ohmura, A, Nakajima, W, Ishida, A, et al. Prolonged hypothermia protects neonatal rat brain against hypoxic-ischemia by reducing both apoptosis and necrosis. Brain Dev 2005; 27: 517–26.CrossRefGoogle ScholarPubMed
Takizawa, Y, Takashima, S, Itoh, M. A histopathological study of premature and mature infants with pontosubicular neuron necrosis: neuronal cell death in perinatal brain damage. Brain Res 2006; 1095: 2000–6.CrossRefGoogle ScholarPubMed
Taniguchi, H, Mohri, I, Okabe-Arahori, H, et al. Prostaglandin D2 protects neonatal mouse brain from hypoxic ischemic injury. J Neurosci 2007; 27: 4303–12.CrossRefGoogle ScholarPubMed
Squier, W, Cowan, FM. The value of autopsy in determining the cause of failure to respond to resuscitation at birth. Semin Neonatol 2004; 9: 331–45.CrossRefGoogle ScholarPubMed
Barkovich, AJ, Westmark, KD, Bedi, HS, et al. Proton spectroscopy and diffusion imaging on the first day of life after perinatal asphyxia: preliminary report. Am J Neuroradiol 2001; 22: 1786–94.Google ScholarPubMed
Meng, SZ, Ohyu, J, Takashima, S. Changes in AMPA glutamate and dopamine D2 receptors in hypoxic–ischemic basal ganglia necrosis. Pediatr Neurol 1997; 17: 139–43.CrossRefGoogle ScholarPubMed
Cheng, Y, Deshmukh, M, D'Costa, A, et al. Caspase inhibitor affords neuroprotection with delayed adminstration in a rat model of neonatal hypoxic–ischemic brain injury. J Clin Invest 1998; 101: 1992–9.CrossRefGoogle Scholar
Rice, JE, Vannucci, RC, Brierley, JB. The influence of immaturity on hypoxic–ischemic brain damage in the rat. Ann Neurol 1981; 9: 131–41.CrossRefGoogle ScholarPubMed
Clancy, B, Darlington, RB, Finlay, BL. Translating developmental time across mammalian species. Neuroscience 2001; 105: 7–17.CrossRefGoogle ScholarPubMed
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: 61–7.CrossRefGoogle ScholarPubMed
Ginsberg, MD, Busto, R. Rodent models of cerebral ischemia. Stroke 1989; 20: 1627–42.CrossRefGoogle ScholarPubMed
Yager, JY. Animal models of hypoxic-ischemic brain damage in the newborn. Semin Pediatr Neurol 2001; 11: 31–46.CrossRefGoogle Scholar
Dobbing, J, Sands, J. Comparative aspects of the brain growth spurt. Early Hum Dev 1979; 3: 79–83.CrossRefGoogle ScholarPubMed
Roland, EH, Poskitt, K, Rodriguez, E, et al. Perinatal hypoxic–ischemic thalamic injury: clinical features and neuroimaging. Ann Neurol 1998; 44: 161–6.CrossRefGoogle ScholarPubMed
Batista, CE, Chugani, HT, Juhasz, C, et al. Transient hypermetabolism of the basal ganglia following perinatal hypoxia. Pediatr Neurol 2007; 36: 330–3.CrossRefGoogle ScholarPubMed
Agnew, DM, Koehler, RC, Guerguerian, AM, et al. Hypothermia for 24 hours after asphyxic cardiac arrest in piglets provides striatal neuroprotection that is sustained 10 days after rewarming. Pediatr Res 2003; 54: 1–10.CrossRefGoogle ScholarPubMed
Thoresen, M, Satas, S, Loberg, EM, et al. Twenty-four hours of mild hypothermia in unsedated newborn pigs starting after a severe global hypoxic–ischemic insult is not neuroprotective. Pediatr Res 2001; 50: 405–11.CrossRefGoogle Scholar
Eicher, DJ, Wagner, CL, Katikaneni, LP, et al. Moderate hypothermia in neonatal encephalopathy: efficacy outcomes. Pediatr Neurol 2005; 32: 11–17.CrossRefGoogle ScholarPubMed
Blomgren, K, Zhu, C, Wang, X, et al. Synergistic activation of caspase-3 by m-calpain after neonatal hypoxia–ischemia: a mechanism of “pathological apoptosis”?J Biol Chem 2001; 276: 10191–8.CrossRefGoogle ScholarPubMed
Hu, BR, Liu, CL, Ouyang, Y, et al. Involvement of caspase-3 in cell death after hypoxia–ischemia declines during brain maturation. J Cereb Blood Flow Metab 2000; 20: 1294–300.CrossRefGoogle ScholarPubMed
Lesuisse, C, Martin, LJ. Immature and mature cortical neurons engage different apoptotic mechanisms involving caspase-3 and the mitogen-activated protein kinase pathway. J Cereb Blood Flow Metab 2002; 22: 935–50.CrossRefGoogle ScholarPubMed
Feng, Y, LeBlanc, MH. Treatment of hypoxic–ischemic brain injury in newborn rats with TPCK 3 h after hypoxia decreases caspase-9 activation and improves neuropathologic outcome. Dev Neurosci 2003; 25: 34–40.CrossRefGoogle ScholarPubMed
Feng, Y, Fratkin, JD, LeBlanc, MH. Inhibiting caspase-8 after injury reduces hypoxic–ischemic brain injury in the newborn rat. Eur J Pharmacol 2003; 481: 169–73.CrossRefGoogle ScholarPubMed
Feng, Y, Fratkin, JD, LeBlanc, MH. Inhibiting caspase-9 after injury reduces hypoxic ischemic neuronal injury in the cortex in the newborn rat. Neurosci Lett 2003; 344: 201–4.CrossRefGoogle ScholarPubMed
Han, BH, Xu, D, Choi, J, et al. Selective, reversible caspase-3 inhibitor is neuroprotective and reveals distinct pathways of cell death after neonatal hypoxic–ischemic brain injury. J Biol Chem 2002; 277: 30128–36.CrossRefGoogle ScholarPubMed
Knoblach, SM, Alroy, DA, Nikolaeva, M, et al. Caspase inhibitor z-DEVD-fmk attenuates calpain and necrotic cell death in vitro and after traumatic brain injury. J Cereb Blood Flow Metab 2004; 24: 1119–32.CrossRefGoogle ScholarPubMed
Rozman-Pungercar, J, Kopitar-Jerala, N, Bogyo, M, et al. Inhibition of papain-like cysteine proteases and legumain by caspase-specific inhibitors: when reaction mechanism is more important than specificity. Cell Death Differ 2003; 10: 881–8.CrossRefGoogle ScholarPubMed
Ostwald, K, Hagberg, H, Andine, P, et al. Upregulation of calpain activity in neonatal rat brain after hypoxic-ischemia. Brain Res 1993; 630: 289–94.CrossRefGoogle ScholarPubMed
Kawamura, M, Nakajima, W, Ishida, A, et al. Calpain inhibitor MDL 28170 protects hypoxic–ischemic brain injury in neonatal rats by inhibition of both apoptosis and necrosis. Brain Res 2005; 1037: 59–69.CrossRefGoogle ScholarPubMed
Han, Y, Giroux, A, Colucci, J, et al. Novel pyrazinone mono-amides as potent and reversible caspase-3 inhibitors. Bioorg Med Chem Lett 2005; 15: 1173–80.CrossRefGoogle ScholarPubMed
Gibson, ME, Han, BH, Choi, J, et al. Bax contributes to apoptotic-like death following neonatal hypoxia–ischemia: evidence for distinct apoptosis pathways. Mol Med 2001; 7: 644–55.Google ScholarPubMed
Sun, W, Oppenheim, RW. Response of motoneurons to neonatal sciatic nerve axotomy in Bax-knockout mice. Mol Cell Neurosci 2003; 24: 875–86.CrossRefGoogle ScholarPubMed
Ness, JM, Harvey, CA, Strasser, A, et al. Selective involvement of BH3-only Bcl-2 family members Bim and Bad in neonatal hypoxia–ischemia. Brain Res 2006; 1099: 150–9.CrossRefGoogle ScholarPubMed
Zhu, C, Xu, F, Fukuda, A, et al. X chromosome-linked inhibitor of apoptosis protein reduces oxidative stress after cerebral irradiation or hypoxia–ischemia through up-regulation of mitochondrial antioxidants. Eur J Neurosci 2007; 26: 3402–10.CrossRefGoogle ScholarPubMed
Zhu, C, Wang, X, Huang, Z, et al. Apoptosis-inducing factor is a major contributor to neuronal loss induced by neonatal cerebral hypoxia–ischemia. Cell Death Diff 2007; 14: 775–84.CrossRefGoogle ScholarPubMed
Northington, FJ, Ferriero, DM, Flock, DL, et al. Delayed neurodegeneration in neonatal rat thalamus after hypoxia–ischemia is apoptosis. J Neurosci 2001; 21: 1931–8.CrossRefGoogle ScholarPubMed
Northington, FJ, Ferriero, DM, Martin, LJ. Neurodegeneration in the thalamus following neonatal hypoxia–ischemia is programmed cell death. Dev Neurosci 2001; 23: 186–91.CrossRefGoogle ScholarPubMed
Al-Abdulla, NA, Martin, LJ. Apoptosis of retrogradely degenerating neurons occurs in association with the accumulation of perikaryal mitochondria and oxidative damage to the nucleus. Am J Pathol 1998; 153: 447–56.CrossRefGoogle ScholarPubMed
Al-Abdulla, NA, Portera-Cailliau, C, Martin, LJ. Occipital cortex ablation in adult rat causes retrograde neuronal death in the lateral geniculate nucleus that resembles apoptosis. Neuroscience 1998; 86: 191–209.CrossRefGoogle ScholarPubMed
Johnston, MV. Selective vulnerability in the neonatal brain. Ann Neurol 1998; 44: 155–6.CrossRefGoogle ScholarPubMed
Brambrink, AM, Martin, LJ, Hanley, DF, et al. Effects of the AMPA receptor antagonist NBQX on the outcome of newborn pigs after asphyxic cardiac arrest. J Cereb Blood Flow Metab 1999; 19: 927–38.CrossRefGoogle ScholarPubMed
Yang, ZJ, Torbey, M, Li, Z, et al. Dopamine receptor modulation of hypoxic–ischemic neuronal injury in striatum of newborn piglets. J Cereb Blood Flow Metab 2007; 27: 1339–51.CrossRefGoogle ScholarPubMed
Golden, WC, Branbrink, AM, Traystman, RJ, et al. Nitration of the striatal Na,K-ATPaseα3 isoform occurs in normal brain development but is not increased during hypoxia–ischemia in newborn piglets. Neurochem Res 2003; 28: 1883–9.CrossRefGoogle Scholar
Mueller-Burke, D, Koehler, RC, Martin, LJ. Rapid NMDA receptor phosphorylation and oxidative stress precede striatal neurodegeneration after hypoxic ischemia in newborn piglets and are attenuated with hypothermia. Int J Dev Neurosci 2008; 26: 67–76.CrossRefGoogle ScholarPubMed
Prybylowski, K, Wenthold, RJ. N-methyl- D-aspartate receptors: subunit assembly and trafficking to the synapse. J Biol Chem 2004; 279: 9673–6.CrossRefGoogle ScholarPubMed
Tingley, WG, Ehlers, MD, Kameyama, K, et al. Characterization of protein kinase A and protein kinase C phosphorylation of the N-methyl-D-aspartate receptor NR1 subunit using phosphorylation site-specific antibodies. J Biol Chem 1997; 272: 5157–66.CrossRefGoogle ScholarPubMed
Guerguerian, AM, Brambrink, AM, Traystman, RJ, et al. Altered expression and phosphorylation of N-methyl-D-aspartate receptors in piglet striatum after hypoxia-ischemia. Brain Res Mol Brain Res 2002; 104: 66–80.CrossRefGoogle ScholarPubMed
Trescher, WH, Ishiwa, S, Johnston, MV. Brief post-hypoxic–ischemic hypothermia markedly delays neonatal brain injury. Brain Dev 1997; 19: 326–38.CrossRefGoogle ScholarPubMed
Bona, E, Hagberg, H, Loberg, EM, et al. Protective effects of moderate hypothermia after neonatal hypoxia–ischemia: short- and long-term outcome. Pediatr Res 1998; 43: 738–45.CrossRefGoogle ScholarPubMed
Ginsberg, SD, Martin, LJ. Ultrastructural analysis of the progression of neurodegeneration in the septum following fimbria–fornix transection. Neuroscience 1998; 86; 1259–72.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
Mueller, D, Shamblott, MJ, Fox, HE, et al. Transplanted human embryonic germ cell-derived neural stem cells replace neurons and oligodendrocytes in the forebrain of neonatal mice with excitotoxic brain damage. J Neurosci Res 2005; 82: 592–608.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
Ikeda, T, Iwai, M, Hayashi, T, et al. Limited differentiation to neurons and astroglia from neural stem cells in the cortex and striatum after ischemia/hypoxia in the neonatal rat brain. Am J Obstet Gynecol 2005; 193: 849–56.CrossRefGoogle ScholarPubMed
Liu, Z, Martin, LJ. The olfactory bulb core is a rich source of neural progenitor and stem cells in adult rodent and human. J Comp Neurol 2003; 459: 368–91.CrossRefGoogle ScholarPubMed
Gritti, A, Bonfanti, L, Doetsch, F, et al. Multipotent neural stem cells reside in the rostral extension and olfactory bulb of adult rodents. J Neurosci 2002; 22: 437–45.CrossRefGoogle ScholarPubMed
Alvarez-Buylla, A, Garcia-Verdugo, JM. Neurogenesis in adult subventricular zone. J Neurosci 2002; 22: 629–34.CrossRefGoogle ScholarPubMed
Martin, LJ, Liu, Z. Adult olfactory bulb neural precursor cell grafts provide temporary protection from motor neuron degeneration, improve motor function, and extend survival in amyotrophic lateral sclerosis mice. J Neuropathol Exp Neurol 2007; 66: 1002–18.CrossRefGoogle ScholarPubMed
McEvoy, AW, Bartolucci, M, Revesz, T, et al. Intractable epilepsy and olfactory bulb hamartoma. Stereotact Funct Neurosurg 2002; 79: 88–93.CrossRefGoogle ScholarPubMed
Obeid, F, Al-Mefty, O. Recurrence of olfactory groove meningiomas. Neurosurgery 2003; 53: 534–42.CrossRefGoogle ScholarPubMed
Spektor, S, Valarezo, J, Fliss, DM, et al. Olfactory groove meningiomas from neurosurgical and ear, nose, and throat perspectives: approaches, techniques, and outcomes. Neurosurgery 2005; 57: 268–79.Google 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
×