Publikationsserver der Universitätsbibliothek Marburg

Titel:Autophagy and apoptosis contribute to neuronal survival in a model system of oxytosis in vitro
Autor:Neunteibl, Stefanie
Weitere Beteiligte: Culmsee, Carsten (Prof. Dr.)
Veröffentlicht:2014
URI:https://archiv.ub.uni-marburg.de/diss/z2014/0463
DOI: https://doi.org/10.17192/z2014.0463
URN: urn:nbn:de:hebis:04-z2014-04630
DDC: Naturwissenschaften
Titel (trans.):Autophagie und Apoptose tragen bei zu neuronalem Überleben in einem in vitro Modellsystem der Oxytose
Publikationsdatum:2014-08-07
Lizenz:https://rightsstatements.org/vocab/InC-NC/1.0/

Dokument

Schlagwörter:
Apoptosis, Apoptose, Autophagy, Autophagie

Summary:
Autophagy and apoptosis play major roles in determining the cellular fate. Accordingly, they participate in development, cellular homeostasis, and both in physiological as well as in pathological processes. Apoptosis is executed by activated caspases, which are specific enzymes that participate in signaling cascades that culminate in the rapid removal of organelles and other cellular structures. Autophagy is a highly conserved cytoprotective process whereby cytoplasmic contents are sequestered, transported via double-membrane autophagosomes to lysosomes, and degraded. Along with regulated necrosis and other forms of programmed cell death, pathological mechanisms of autophagy and apoptosis have been detected in neurodegenerative diseases, such as Parkinson’s disease or Alzheimer’s disease and acute brain injuries. The aim of this thesis was to investigate the role of autophagy and apoptosis for neuronal resilience versus neuronal cell death in model systems of glutamate toxicity in vitro. The role of autophagy was investigated in the model system of glutamate-induced oxidative stress, i.e. oxytosis in neural HT-22 cells. The objectives were to determine the effect of oxidative glutamate toxicity on autophagic flux and to investigate if oxytosis involves autophagy pathways of cell death. Moreover the neuroprotective effect of 3-Methyladenine (3-MA), a widely used autophagy inhibitor, was explored in the model systems of glutamate-induced oxytosis and excitotoxicity in neural HT-22 cells and primary cortical neurons, respectively. Glutamate clearly enhanced autophagy markers and induced cell death in HT-22 cells and PCN. Cell death was prevented by 3-MA, a widely used inhibitor of autophagy. Interestingly, 3-MA itself induced autophagy in HT-22 cells. A gene silencing approach targeting key regulators of autophagy reduced the autophagic flux, but failed to prevent cell death persistently. 3-MA prevented the glutamate-induced ROS formation, the loss of ATP, and preserved the mitochondrial membrane potential as well as mitochondrial morphology. The activation of the PI3K/Akt pathway and the MAPK/Erk-1/2 pathway do not play a role in 3-MA mediated neuroprotection. In conclusion, these data suggest that glutamate toxicity is associated with increased markers of autophagy. However, specific gene silencing of key regulators of autophagy did not provide protective effects, suggesting that the increased autophagic flux was dispensable for cell death induced by glutamate. The induction of autophagy can rather be interpreted as the ultimate attempt to adapt to lethal stress after glutamate challenge und is rather prosurvival and contributes to cellular homeostasis in HT-22 cells. Further, the findings clearly demonstrate that protective effects by 3-MA occur independently of autophagy inhibition, although the compound is widely used as an inhibitor of autophagy. Nowadays, 3-MA is used frequently in experimental studies in vivo and in vitro. Based on the present findings, caution is recommended in the interpretation of data obtained with 3-MA in the context of autophagy studies, in particular, if mitochondrial alterations are connected. The contribution of apoptosis to neuronal survival through intercellular signaling between dying cells and neurons was investigated using conditioned medium of neural progenitor cells (NPC) and HT-22 cells. Stem cells as well as progenitor cells have been widely used in model systems of neurodegenerative diseases and acute brain injuries where transplantation of these cells into the brain improved neuronal survival and brain functions in experimental settings. However, most transplanted cells die after transplantation in vivo, and the exact mechanism of action of stem cell or progenitor cell transplantation still remains unknown. To investigate mechanisms of intercellular signaling underlying the protective effects of transplanted stem cells, the transplantation conditions were mimicked in vitro by preparation of conditioned medium (CM) obtained from dying neuronal progenitor cells. This CM should contain similar cellular components as released during cell death of the progenitor cell transplants, and this CM should therefore also provide neuroprotective effects. Thus, the CM of dying/apoptotic cells was applied in model systems of cell death in vitro for to test its potential to mediate neuroprotection. Further, the composition of the conditioned medium obtained from the dying progenitor cells was analysed for identifying the most potent protective components that may be applied as neuroprotectants in vitro and in vivo instead of the CM or the cellular transplants. In order to mimic the conditions of transplantation, NPC were exposed to medium lacking growth factors such as FGF and EGF. Such growth factor deprivation induced caspase-dependent cell death in NPC in a time-dependent manner. The conditioned medium obtained from apoptotic NPC significantly attenuated cell death induced by growth factor withdrawal and glutamate exposure in HT-22 cells and cortical neurons in a dose-dependent manner. The protective effect of NPC CM against glutamate neurotoxicity was abolished by heat inactivation at 95°C for 30 min. Further, NPC CM enhanced phosphorylation of PKB/Akt and Erk 1/2 in neurons in a similar time frame as the neurotrophin BDNF. Inhibition of autophagy did not diminish the protective effect of NPC CM. Also the use of spermidine conditioned medium (Sp CM) from HT-22 cells, whose production aimed on the specific activation of autophagy, indicated that the induction of autophagy is not essential for the protective effect of conditioned medium. In total, these findings suggest that NPC secrete neuroprotective factors that stimulate neurotrophin-like survival signaling thereby providing protective effects against growth factor withdrawal and glutamate neurotoxicity. The results obtained from the in vitro model system in this thesis are transferable to the adult organism. Thus, the results are the basis for the development of a highly potent, standardized composition for the therapy of neurodegenerative diseases and acute brain injury. Therefore, CM and its active components could serve as an alternative therapeutic option to stem cell transplantation.

Bibliographie / References

  1. Mijaljica D, Prescott M, Devenish RJ. Microautophagy in mammalian cells: revisiting a 40-year-old conundrum. Autophagy 2011; 7 (7): 673–682.
  2. Reuck AVS de, Cameron MP, Duve C de. Lysosomes, London: J. & A. Churchill 1963. 7. Ciechanover A, Orian A, Schwartz AL. Ubiquitin-mediated proteolysis: biological regulation via destruction. Bioessays 2000; 22 (5): 442–451.
  3. Eisenberg-Lerner A, Bialik S, Simon H, Kimchi A. Life and death partners: apoptosis, autophagy and the cross-talk between them. Cell Death Differ 2009; 16 (7): 966–975.
  4. Han BH, Holtzman DM. BDNF protects the neonatal brain from hypoxic-ischemic injury in vivo via the ERK pathway. J. Neurosci. 2000; 20 (15): 5775–5781.
  5. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006; 126 (4): 663–676.
  6. Xu Y, Kim SO, Li Y, Han J. Autophagy contributes to caspase-independent macrophage cell death. J. Biol. Chem. 2006; 281 (28): 19179–19187.
  7. Bang OY, Lee JS, Lee PH, Lee G. Autologous mesenchymal stem cell transplantation in stroke patients. Ann. Neurol. 2005; 57 (6): 874–882.
  8. Chen WW, Blurton-Jones M. Concise review: Can stem cells be used to treat or model Alzheimer's disease? Stem Cells 2012; 30 (12): 2612–2618.
  9. Xue L, Fletcher GC, Tolkovsky AM. Autophagy is activated by apoptotic signalling in sympathetic neurons: an alternative mechanism of death execution. Mol. Cell. Neurosci. 1999; 14 (3): 180–198.
  10. Walker JM. The bicinchoninic acid (BCA) assay for protein quantitation. Methods Mol. Biol. 1994; 32: 5–8.
  11. Steiger-Barraissoul S, Rami A. Serum deprivation induced autophagy and predominantly an AIF-dependent apoptosis in hippocampal HT22 neurons. Apoptosis 2009; 14 (11): 1274–1288.
  12. Mukhopadhyay S, Panda PK, Sinha N et al. Autophagy and apoptosis: where do they meet? Apoptosis 2014; 19 (4): 555–566.
  13. Cowley S, Paterson H, Kemp P, Marshall CJ. Activation of MAP kinase kinase is necessary and sufficient for PC12 differentiation and for transformation of NIH 3T3 cells. Cell 1994; 77 (6): 841–852.
  14. Jung K, Chu K, Lee S et al. Augmentation of nitrite therapy in cerebral ischemia by NMDA receptor inhibition. Biochem. Biophys. Res. Commun. 2009; 378 (3): 507–512.
  15. Komatsu M, Ichimura Y. Physiological significance of selective degradation of p62 by autophagy. FEBS Letters 2010; 584 (7): 1374–1378.
  16. Ray J, Gage FH. Differential properties of adult rat and mouse brain-derived neural stem/progenitor cells. Mol. Cell. Neurosci. 2006; 31 (3): 560–573.
  17. Nakano N, Nakai Y, Seo T et al. Characterization of conditioned medium of cultured bone marrow stromal cells. Neurosci. Lett. 2010; 483 (1): 57–61.
  18. Wilkins A, Kemp K, Ginty M et al. Human bone marrow-derived mesenchymal stem cells secrete brain-derived neurotrophic factor which promotes neuronal survival in vitro. Stem Cell Research 2009; 3 (1): 63–70.
  19. Venkataramana NK, Kumar SK, Balaraju S et al. Open-labeled study of unilateral autologous bone-marrow-derived mesenchymal stem cell transplantation in Parkinson's disease. Translational Research 2010; 155 (2): 62–70.
  20. Robinson SR, Bishop GM. Abeta as a bioflocculant: implications for the amyloid hypothesis of Alzheimer's disease. Neurobiol. Aging 2002; 23 (6): 1051–1072.
  21. Kraft C, Peter M, Hofmann K. Selective autophagy: ubiquitin-mediated recognition and beyond. Nat Cell Biol 2010; 12 (9): 836–841.
  22. Deng W, Aimone JB, Gage FH. New neurons and new memories: how does adult hippocampal neurogenesis affect learning and memory? Nat. Rev. Neurosci. 2010; 11 (5): 339–350.
  23. Chen Y, Kramer DL, Li F, Porter CW. Loss of inhibitor of apoptosis proteins as a determinant of polyamine analog-induced apoptosis in human melanoma cells. Oncogene 2003; 22 (32): 4964–4972.
  24. Suzuki T, Nakagawa M, Yoshikawa A et al. The first molecular evidence that autophagy relates rimmed vacuole formation in chloroquine myopathy. J. Biochem. 2002; 131 (5): 647–651.
  25. Castino R, Bellio N, Follo C et al. Inhibition of PI3k class III-dependent autophagy prevents apoptosis and necrosis by oxidative stress in dopaminergic neuroblastoma cells. Toxicol. Sci. 2010; 117 (1): 152–162.
  26. Datta SR, Brunet A, Greenberg ME. Cellular survival: a play in three Akts. Genes Dev. 1999; 13 (22): 2905–2927.
  27. Dutta S, Singh G, Sreejith S et al. Cell Therapy: The Final Frontier for Treatment of Neurological Diseases. CNS Neurosci Ther 2013; 19 (1): 5–11.
  28. Rami A. Review: Autophagy in neurodegeneration: firefighter and/or incendiarist? Neuropathology and Applied Neurobiology 2009; 35 (5): 449–461.
  29. Kalia S, Dutz JP. New concepts in antimalarial use and mode of action in dermatology. Dermatol Ther 2007; 20 (4): 160–174.
  30. Mendivil-Perez M, Velez-Pardo C, Jimenez-Del-Rio M. TPEN induces apoptosis independently of zinc chelator activity in a model of acute lymphoblastic leukemia and ex vivo acute leukemia cells through oxidative stress and mitochondria caspase-3-and AIF-dependent pathways. Oxid Med Cell Longev 2012; 2012: 313275. 255.
  31. Silva GV, Litovsky S, Assad JAR et al. Mesenchymal stem cells differentiate into an endothelial phenotype, enhance vascular density, and improve heart function in a canine chronic ischemia model. Circulation 2005; 111 (2): 150–156.
  32. Madhavan L, Ourednik V, Ourednik J. Neural stem/progenitor cells initiate the formation of cellular networks that provide neuroprotection by growth factor-modulated antioxidant expression. Stem Cells 2008; 26 (1): 254–265.
  33. Tan S, Schubert D, Maher P. Oxytosis: A novel form of programmed cell death. Curr Top Med Chem 2001; 1 (6): 497–506.
  34. Luan Z, Liu W, Qu S et al. Effects of Neural Progenitor Cell Transplantation in Children With Severe Cerebral Palsy. cell transplant 2012; 21 (1): 91–98.
  35. Wang Y, Han R, Liang Z et al. An autophagic mechanism is involved in apoptotic death of rat striatal neurons induced by the non-N-methyl-D-aspartate receptor agonist kainic acid. Autophagy 2008; 4 (2): 214–226.
  36. Madeo F, Eisenberg T, Büttner S et al. Spermidine: a novel autophagy inducer and longevity elixir. Autophagy 2010; 6 (1): 160–162.
  37. Wen Y, Sheng R, Zhang L et al. Neuronal injury in rat model of permanent focal cerebral ischemia is associated with activation of autophagic and lysosomal pathways. Autophagy 2008; 4 (6): 762–769.
  38. Eisenberg T, Knauer H, Schauer A et al. Induction of autophagy by spermidine promotes longevity. Nat. Cell Biol. 2009; 11 (11): 1305–1314.
  39. Morselli E, Mariño G, Bennetzen MV et al. Spermidine and resveratrol induce autophagy by distinct pathways converging on the acetylproteome. J. Cell Biol. 2011; 192 (4): 615–629.
  40. Gage FH. Mammalian neural stem cells. Science 2000; 287 (5457): 1433–1438.
  41. Vandenabeele P, Galluzzi L, Vanden Berghe T, Kroemer G. Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol Cell Biol 2010; 11 (10): 700– 714.
  42. Windmolders S, Boeck A de, Koninckx R et al. Mesenchymal stem cell secreted platelet derived growth factor exerts a pro-migratory effect on resident Cardiac Atrial appendage Stem Cells. Journal of Molecular and Cellular Cardiology 2013.
  43. Zhu JM, Zhao YY, Chen SD et al. Functional recovery after transplantation of neural stem cells modified by brain-derived neurotrophic factor in rats with cerebral ischaemia. J. Int. Med. Res. 2011; 39 (2): 488–498.
  44. Blandini F, Porter RH, Greenamyre JT. Glutamate and Parkinson's disease. Mol. Neurobiol. 1996; 12 (1): 73–94.
  45. Lee JS, Hong JM, Moon GJ et al. A long-term follow-up study of intravenous autologous mesenchymal stem cell transplantation in patients with ischemic stroke. Stem Cells 2010; 28 (6): 1099–1106.
  46. Kahn BB, Alquier T, Carling D, Hardie DG. AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 2005; 1 (1): 15–25.
  47. Wang XJ, Yu J, Wong SH et al. A novel crosstalk between two major protein degradation systems: Regulation of proteasomal activity by autophagy. Autophagy 2013; 9 (10).
  48. Lockshin RA, Zakeri Z. Apoptosis, autophagy, and more. Int. J. Biochem. Cell Biol. 2004; 36 (12): 2405–2419.
  49. Culmsee C, Zhu C, Landshamer S et al. Apoptosis-inducing factor triggered by poly(ADP-ribose) polymerase and Bid mediates neuronal cell death after oxygen- glucose deprivation and focal cerebral ischemia. J. Neurosci. 2005; 25 (44): 10262– 10272.
  50. Hristov M, Erl W, Linder S, Weber PC. Apoptotic bodies from endothelial cells enhance the number and initiate the differentiation of human endothelial progenitor cells in vitro. Blood 2004; 104 (9): 2761–2766.
  51. Li Y, Maher P, Schubert D. A role for 12-lipoxygenase in nerve cell death caused by glutathione depletion. Neuron 1997; 19 (2): 453–463.
  52. Anderson CN, Tolkovsky AM. A role for MAPK/ERK in sympathetic neuron survival: protection against a p53-dependent, JNK-independent induction of apoptosis by cytosine arabinoside. J. Neurosci. 1999; 19 (2): 664–673.
  53. Kirkin V, McEwan DG, Novak I, Dikic I. A Role for Ubiquitin in Selective Autophagy. Molecular Cell 2009; 34 (3): 259–269.
  54. Landshamer S, Hoehn M, Barth N et al. Bid-induced release of AIF from mitochondria causes immediate neuronal cell death. Cell Death Differ 2008; 15 (10): 1553–1563.
  55. Tobaben S, Grohm J, Seiler A et al. Bid-mediated mitochondrial damage is a key mechanism in glutamate-induced oxidative stress and AIF-dependent cell death in immortalized HT-22 hippocampal neurons. Cell Death Differ 2010; 18 (2): 282–292.
  56. Degterev A, Huang Z, Boyce M et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat. Chem. Biol. 2005; 1 (2): 112– 119.
  57. Fernandes-Alnemri T, Litwack G, Alnemri ES. CPP32, a novel human apoptotic protein with homology to Caenorhabditis elegans cell death protein Ced-3 and mammalian interleukin-1 beta-converting enzyme. J. Biol. Chem. 1994; 269 (49): 30761–30764.
  58. Petiot A, Ogier-Denis E, Blommaart EF et al. Distinct classes of phosphatidylinositol 3'- kinases are involved in signaling pathways that control macroautophagy in HT-29 cells. J. Biol. Chem. 2000; 275 (2): 992–998.
  59. Matsui Y, Takagi H, Qu X et al. Distinct roles of autophagy in the heart during ischemia and reperfusion: roles of AMP-activated protein kinase and Beclin 1 in mediating autophagy. Circ. Res. 2007; 100 (6): 914–922.
  60. Morimoto BH, Koshland DE. Excitatory amino acid uptake and N-methyl-D-aspartate- mediated secretion in a neural cell line. Proc. Natl. Acad. Sci. U.S.A. 1990; 87 (9): 3518–3521.
  61. Zhu W, Chen J, Cong X et al. Hypoxia and serum deprivation-induced apoptosis in mesenchymal stem cells. Stem Cells 2006; 24 (2): 416–425.
  62. Diemert S, Dolga A, Tobaben S et al. Impedance measurement for real time detection of neuronal cell death. Journal of Neuroscience Methods 2012; 203 (1): 69–77.
  63. Oliver FJ, La Rubia G de, Rolli V et al. Importance of poly(ADP-ribose) polymerase and its cleavage in apoptosis. Lesson from an uncleavable mutant. J. Biol. Chem. 1998; 273 (50): 33533–33539.
  64. Petit I, Salman Kesner N, Karry R et al. Induced pluripotent stem cells from hair follicles as a cellular model for neurodevelopmental disorders. Stem Cell Research 2012; 8 (1): 134–140.
  65. Chen X, Li Y, Wang L et al. Ischemic rat brain extracts induce human marrow stromal cell growth factor production. Neuropathology 2002; 22 (4): 275–279.
  66. Shaw RJ. LKB1 and AMP-activated protein kinase control of mTOR signalling and growth. Acta Physiol (Oxf) 2009; 196 (1): 65–80.
  67. Liu Y, Peterson DA, Kimura H, Schubert D. Mechanism of cellular 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction. J. Neurochem. 1997; 69 (2): 581–593.
  68. Eva-Maria Öxler. Molecular and cellular preconditioning -powerful strategies for neuroprotection. Doktorarbeit, Marburg 2012. 200.
  69. Culmsee C, Gerling N, Lehmann M et al. Nerve growth factor survival signaling in cultured hippocampal neurons is mediated through TrkA and requires the common neurotrophin receptor P75. Neuroscience 2002; 115 (4): 1089–1108.
  70. Lim H, Lee S, Chu K et al. Neuroprotective effect of neural stem cell-conditioned media in in vitro model of Huntington's disease. Neuroscience Letters 2008; 435 (3): 175–180.
  71. Fan MMY, Raymond LA. N-methyl-D-aspartate (NMDA) receptor function and excitotoxicity in Huntington's disease. Prog. Neurobiol. 2007; 81 (5-6): 272–293.
  72. Gohil VM, Sheth SA, Nilsson R et al. Nutrient-sensitized screening for drugs that shift energy metabolism from mitochondrial respiration to glycolysis. Nat. Biotechnol. 2010; 28 (3): 249–255.
  73. Luo Y, DeFranco DB. Opposing roles for ERK1/2 in neuronal oxidative toxicity: distinct mechanisms of ERK1/2 action at early versus late phases of oxidative stress. J. Biol. Chem. 2006; 281 (24): 16436–16442.
  74. Coyle JT, Puttfarcken P. Oxidative stress, glutamate, and neurodegenerative disorders. Science 1993; 262 (5134): 689–695.
  75. Wang Y, Dong X, Cao Y et al. p53 induction contributes to excitotoxic neuronal death in rat striatum through apoptotic and autophagic mechanisms. Eur. J. Neurosci. 2009; 30 (12): 2258–2270.
  76. Stanciu M, Wang Y, Kentor R et al. Persistent activation of ERK contributes to glutamate-induced oxidative toxicity in a neuronal cell line and primary cortical neuron cultures. J. Biol. Chem. 2000; 275 (16): 12200–12206.
  77. Choi DW, Koh JY, Peters S. Pharmacology of glutamate neurotoxicity in cortical cell culture: attenuation by NMDA antagonists. J. Neurosci. 1988; 8 (1): 185–196.
  78. Crowder RJ, Freeman RS. Phosphatidylinositol 3-kinase and Akt protein kinase are necessary and sufficient for the survival of nerve growth factor-dependent sympathetic neurons. J. Neurosci. 1998; 18 (8): 2933–2943.
  79. Cao Y, Klionsky DJ. Physiological functions of Atg6/Beclin 1: a unique autophagy- related protein. Cell Res. 2007; 17 (10): 839–849.
  80. Seiler N, Raul F. Polyamines and apoptosis. J. Cell. Mol. Med. 2005; 9 (3): 623–642.
  81. Moschou PN, Roubelakis-Angelakis KA. Polyamines and programmed cell death. J. Exp. Bot. 2013.
  82. Cho D, Nakamura T, Fang J et al. S-nitrosylation of Drp1 mediates beta-amyloid- related mitochondrial fission and neuronal injury. Science 2009; 324 (5923): 102–105.
  83. Minois N, Carmona-Gutierrez D, Bauer MA et al. Spermidine promotes stress resistance in Drosophila melanogaster through autophagy-dependent and -independent pathways. Cell Death Dis 2012; 3: e401. 205.
  84. Danysz W, Parsons CG. The NMDA receptor antagonist memantine as a symptomatological and neuroprotective treatment for Alzheimer's disease: preclinical evidence. Int J Geriatr Psychiatry 2003; 18 (Suppl 1): S23-32.
  85. Brenner C, Grimm S. The permeability transition pore complex in cancer cell death. Oncogene 2006; 25 (34): 4744–4756.
  86. Chen J, Li Y, Wang L et al. Therapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in rats. Stroke 2001; 32 (4): 1005–1011.
  87. Steere AC, Angelis SM. Therapy for Lyme arthritis: strategies for the treatment of antibiotic-refractory arthritis. Arthritis Rheum. 2006; 54 (10): 3079–3086.
  88. Kuma A, Hatano M, Matsui M et al. The role of autophagy during the early neonatal starvation period. Nature 2004; 432 (7020): 1032–1036.
  89. Maher P, Davis JB. The role of monoamine metabolism in oxidative glutamate toxicity. J. Neurosci. 1996; 16 (20): 6394–6401.
  90. Brunet A, Datta SR, Greenberg ME. Transcription-dependent and -independent control of neuronal survival by the PI3K-Akt signaling pathway. Curr. Opin. Neurobiol. 2001; 11 (3): 297–305.
  91. Zhu Y, Yang G, Ahlemeyer B et al. Transforming growth factor-beta 1 increases bad phosphorylation and protects neurons against damage. J. Neurosci. 2002; 22 (10): 3898–3909.
  92. Lindvall O, Kokaia Z. Stem cells for the treatment of neurological disorders. Nature 2006; 441 (7097): 1094–1096.
  93. Lindvall O, Kokaia Z. Stem cell research in stroke: how far from the clinic? Stroke 2011; 42 (8): 2369–2375.
  94. Lindvall O, Kokaia Z, Martinez-Serrano A. Stem cell therapy for human neurodegenerative disorders-how to make it work. Nat. Med. 2004; 10: S42-50. 147.
  95. Darsalia V, Kallur T, Kokaia Z. Survival, migration and neuronal differentiation of human fetal striatal and cortical neural stem cells grafted in stroke-damaged rat striatum. Eur. J. Neurosci. 2007; 26 (3): 605–614.
  96. Weisová P, Dávila D, Tuffy LP et al. Role of 5'-adenosine monophosphate-activated protein kinase in cell survival and death responses in neurons. Antioxid. Redox Signal. 2011; 14 (10): 1863–1876.
  97. Chen S, Chang C, Tsai S et al. Functional Improvement of Focal Cerebral Ischemia Injury by Subdural Transplantation of Induced Pluripotent Stem Cells with Fibrin Glue. Stem Cells and Development 2010; 19 (11): 1757–1767.
  98. Kim SU, Vellis J de. Stem cell-based cell therapy in neurological diseases: A review. J. Neurosci. Res. 2009; 87 (10): 2183–2200.
  99. Park HJ, Lee PH, Bang OY et al. Mesenchymal stem cells therapy exerts neuroprotection in a progressive animal model of Parkinson's disease. Journal of Neurochemistry 2008; 107 (1): 141–151.
  100. Puyal J, Vaslin A, Mottier V, Clarke PGH. Postischemic treatment of neonatal cerebral ischemia should target autophagy. Ann. Neurol. 2009; 66 (3): 378–389.
  101. Menasché P. Stem cells for clinical use in cardiovascular medicine: current limitations and future perspectives. Thromb. Haemost. 2005; 94 (4): 697–701.
  102. Weisová P, Concannon CG, Devocelle M et al. Regulation of glucose transporter 3 surface expression by the AMP-activated protein kinase mediates tolerance to glutamate excitation in neurons. J. Neurosci. 2009; 29 (9): 2997–3008.
  103. Gupta VK, Scheunemann L, Eisenberg T et al. Restoring polyamines protects from age- induced memory impairment in an autophagy-dependent manner. Nat. Neurosci. 2013; 16 (10): 1453–1460.
  104. Yuan H, Gerencser AA, Liot G et al. Mitochondrial fission is an upstream and required event for bax foci formation in response to nitric oxide in cortical neurons. Cell Death Differ. 2007; 14 (3): 462–471.
  105. Gould E. How widespread is adult neurogenesis in mammals? Nat. Rev. Neurosci. 2007; 8 (6): 481–488.
  106. Knott AB, Perkins G, Schwarzenbacher R, Bossy-Wetzel E. Mitochondrial fragmentation in neurodegeneration. Nat. Rev. Neurosci. 2008; 9 (7): 505–518.
  107. Hendil KB, Lauridsen AM, Seglen PO. Both endocytic and endogenous protein degradation in fibroblasts is stimulated by serum/amino acid deprivation and inhibited by 3-methyladenine. Biochem. J. 1990; 272 (3): 577–581.
  108. Berliocchi L, Bano D, Nicotera P. Ca2+ signals and death programmes in neurons. Philos. Trans. R. Soc. Lond., B, Biol. Sci. 2005; 360 (1464): 2255–2258.
  109. Shintani T, Klionsky DJ. Autophagy in health and disease: a double-edged sword. Science 2004; 306 (5698): 990–995.
  110. Kopen GC, Prockop DJ, Phinney DG. Marrow stromal cells migrate throughout forebrain and cerebellum, and they differentiate into astrocytes after injection into neonatal mouse brains. Proc. Natl. Acad. Sci. U.S.A. 1999; 96 (19): 10711–10716.
  111. Majno G, Joris I. Apoptosis, oncosis, and necrosis. An overview of cell death. Am. J. Pathol. 1995; 146 (1): 3–15.
  112. Redmond DE, Bjugstad KB, Teng YD et al. Behavioral improvement in a primate Parkinson's model is associated with multiple homeostatic effects of human neural stem cells. Proc. Natl. Acad. Sci. U.S.A. 2007; 104 (29): 12175–12180.
  113. Ding W, Ni H, Gao W et al. Linking of Autophagy to Ubiquitin-Proteasome System Is Important for the Regulation of Endoplasmic Reticulum Stress and Cell Viability. The American Journal of Pathology 2007; 171 (2): 513–524.
  114. Poole B, Ohkuma S. Effect of weak bases on the intralysosomal pH in mouse peritoneal macrophages. J. Cell Biol. 1981; 90 (3): 665–669.
  115. Tan S, Sagara Y, Liu Y et al. The regulation of reactive oxygen species production during programmed cell death. J. Cell Biol. 1998; 141 (6): 1423–1432.
  116. Philpott KL, McCarthy MJ, Klippel A, Rubin LL. Activated phosphatidylinositol 3- kinase and Akt kinase promote survival of superior cervical neurons. J. Cell Biol. 1997; 139 (3): 809–815.
  117. Takeshige K, Baba M, Tsuboi S et al. Autophagy in yeast demonstrated with proteinase-deficient mutants and conditions for its induction. J. Cell Biol. 1992; 119 (2): 301–311.
  118. Helton TD, Otsuka T, Lee M et al. Pruning and loss of excitatory synapses by the parkin ubiquitin ligase. Proc. Natl. Acad. Sci. U.S.A. 2008; 105 (49): 19492–19497.
  119. Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy fights disease through cellular self-digestion. Nature 2008; 451 (7182): 1069–1075.
  120. Blurton-Jones M, Kitazawa M, Martinez-Coria H et al. Neural stem cells improve cognition via BDNF in a transgenic model of Alzheimer disease. Proc. Natl. Acad. Sci. U.S.A. 2009; 106 (32): 13594–13599.
  121. Klionsky DJ, Emr SD. Autophagy as a regulated pathway of cellular degradation. Science 2000; 290 (5497): 1717–1721.
  122. Kubota C, Torii S, Hou N et al. Constitutive reactive oxygen species generation from autophagosome/lysosome in neuronal oxidative toxicity. J. Biol. Chem. 2010; 285 (1): 667–674.
  123. Hsieh Y, Athar M, Chaudry IH. When apoptosis meets autophagy: deciding cell fate after trauma and sepsis. Trends Mol Med 2009; 15 (3): 129–138.
  124. He C, Klionsky DJ. Regulation Mechanisms and Signaling Pathways of Autophagy. Annu. Rev. Genet. 2009; 43 (1): 67–93.
  125. Concannon CG, Tuffy LP, Weisová P et al. AMP kinase-mediated activation of the BH3-only protein Bim couples energy depletion to stress-induced apoptosis. J. Cell Biol. 2010; 189 (1): 83–94.
  126. Wu Y, Tan H, Shui G et al. Dual Role of 3-Methyladenine in Modulation of Autophagy via Different Temporal Patterns of Inhibition on Class I and III Phosphoinositide 3- Kinase. Journal of Biological Chemistry 2010; 285 (14): 10850–10861.
  127. Jeong H, Then F, Melia TJ et al. Acetylation Targets Mutant Huntingtin to Autophagosomes for Degradation. Cell 2009; 137 (1): 60–72.
  128. Joyce N, Annett G, Wirthlin L et al. Mesenchymal stem cells for the treatment of neurodegenerative disease. Regen Med 2010; 5 (6): 933–946.
  129. Filosto M, Scarpelli M, Cotelli MS et al. The role of mitochondria in neurodegenerative diseases. J. Neurol. 2011; 258 (10): 1763–1774.
  130. Rohn TT, Wirawan E, Brown RJ et al. Depletion of Beclin-1 due to proteolytic cleavage by caspases in the Alzheimer's disease brain. Neurobiol. Dis. 2011; 43 (1): 68–78.
  131. Duan B, Wang Y, Yang T et al. Extracellular spermine exacerbates ischemic neuronal injury through sensitization of ASIC1a channels to extracellular acidosis. J. Neurosci. 2011; 31 (6): 2101–2112.
  132. Piemonte F, Rossi F, Carletti B. Neuroprotection: The Emerging Concept of Restorative Neural Stem Cell Biology for the Treatment of Neurodegenerative Diseases. CN 2011; 9 (2): 313–317.
  133. Galluzzi L, Vitale I, Abrams JM et al. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ 2011; 19 (1): 107–120.
  134. Denton D, Nicolson S, Kumar S. Cell death by autophagy: facts and apparent artefacts. Cell Death Differ. 2012; 19 (1): 87–95.
  135. Boomsma RA, Geenen DL, Hofmann TG. Mesenchymal Stem Cells Secrete Multiple Cytokines That Promote Angiogenesis and Have Contrasting Effects on Chemotaxis and Apoptosis. PLoS ONE 2012; 7 (4): e35685. 160.
  136. Kumari S, Mehta SL, Li PA. Glutamate induces mitochondrial dynamic imbalance and autophagy activation: preventive effects of selenium. PLoS ONE 2012; 7 (6): e39382. 209.
  137. Grohm J, Kim S, Mamrak U et al. Inhibition of Drp1 provides neuroprotection in vitro and in vivo. Cell Death Differ. 2012; 19 (9): 1446–1458.
  138. Seglen PO, Gordon PB. 3-Methyladenine: specific inhibitor of autophagic/lysosomal protein degradation in isolated rat hepatocytes. Proc. Natl. Acad. Sci. U.S.A. 1982; 79 (6): 1889–1892.
  139. Gao L, Jiang T, Guo J et al. Inhibition of autophagy contributes to ischemic postconditioning-induced neuroprotection against focal cerebral ischemia in rats. PLoS ONE 2012; 7 (9): e46092.
  140. Shaid S, Brandts CH, Serve H, Dikic I. Ubiquitination and selective autophagy. Cell Death Differ. 2013; 20 (1): 21–30.
  141. Yang Z, Klionsky DJ. Eaten alive: a history of macroautophagy. Nat Cell Biol 2010; 12 (9): 814–822.
  142. Sui X, Chen R, Wang Z et al. Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis 2013; 4: e838. 239.
  143. McBride HM, Neuspiel M, Wasiak S. Mitochondria: more than just a powerhouse. Curr. Biol. 2006; 16 (14): R551-60.
  144. Kelly S, Bliss TM, Shah AK et al. Transplanted human fetal neural stem cells survive, migrate, and differentiate in ischemic rat cerebral cortex. Proc. Natl. Acad. Sci. U.S.A. 2004; 101 (32): 11839–11844.
  145. Takagi Y, Takahashi J, Saiki H et al. Dopaminergic neurons generated from monkey embryonic stem cells function in a Parkinson primate model. J. Clin. Invest. 2005; 115 (1): 102–109.
  146. Gonzalez-Zulueta M, Feldman AB, Klesse LJ et al. Requirement for nitric oxide activation of p21(ras)/extracellular regulated kinase in neuronal ischemic preconditioning. Proc. Natl. Acad. Sci. U.S.A. 2000; 97 (1): 436–441.
  147. Murphy TH, Miyamoto M, Sastre A et al. Glutamate toxicity in a neuronal cell line involves inhibition of cystine transport leading to oxidative stress. Neuron 1989; 2 (6): 1547–1558.
  148. Kim H, Choi J, Ryu J et al. Activation of autophagy during glutamate-induced HT22 cell death. Biochem Biophys Res Commun 2009; 388 (2): 339–344.
  149. Orlando KA, Stone NL, Pittman RN. Rho kinase regulates fragmentation and phagocytosis of apoptotic cells. Exp. Cell Res. 2006; 312 (1): 5–15.
  150. Nunnari J, Suomalainen A. Mitochondria: in sickness and in health. Cell 2012; 148 (6): 1145–1159.
  151. Seaberg RM, van der Kooy D. Stem and progenitor cells: the premature desertion of rigorous definitions. Trends Neurosci. 2003; 26 (3): 125–131.
  152. Isele NB, Lee H, Landshamer S et al. Bone marrow stromal cells mediate protection through stimulation of PI3-K/Akt and MAPK signaling in neurons. Neurochem. Int. 2007; 50 (1): 243–250.
  153. Grohm J, Plesnila N, Culmsee C. Bid mediates fission, membrane permeabilization and peri-nuclear accumulation of mitochondria as a prerequisite for oxidative neuronal cell death. Brain Behav Immun 2010; 24 (5): 831–838.
  154. Wang F, Maeda N, Yasuhara T et al. The therapeutic potential of human umbilical cord blood transplantation for neonatal hypoxic-ischemic brain injury and ischemic stroke. Acta Med. Okayama 2012; 66 (6): 429–434.
  155. Renolleau S, Fau S, Goyenvalle C et al. Specific caspase inhibitor Q-VD-OPh prevents neonatal stroke in P7 rat: a role for gender. J. Neurochem. 2007; 100 (4): 1062–1071.


* Das Dokument ist im Internet frei zugänglich - Hinweise zu den Nutzungsrechten