Publikationsserver der Universitätsbibliothek Marburg

Titel:Inhibitors of p53 and HIF-prolyl-4-hydroxylases provide mitochondrial protection in a model of oxytosis
Autor:Neitemeier, Sandra
Weitere Beteiligte: Culmsee, Carsten (Prof. Dr.)
Veröffentlicht:2015
URI:https://archiv.ub.uni-marburg.de/diss/z2015/0354
DOI: https://doi.org/10.17192/z2015.0354
URN: urn:nbn:de:hebis:04-z2015-03549
DDC: Medizin
Titel (trans.):Inhibitoren für p53 und HIF-prolyl-4-hydroxylasen schützen Mitochondrien in einem Modelsystem der Oxytose
Publikationsdatum:2016-01-25
Lizenz:https://rightsstatements.org/vocab/InC-NC/1.0/

Dokument

Schlagwörter:
oxytosis, p53, Zelltod, Mitochondrium, neuronal cell death, Protein p53, Oxidativer Stress, HIF-prolyl-4-hydroxylase, mitochondria

Summary:
Mitochondrial dysfunction and demise are hallmarks of many neurological disorders and neurodegenerative diseases. Since mitochondria are the key organelles providing energy, they are regarded as the power house of the cell. Under conditions of stress, mitochondria regulate the ‘point of no return’ in intrinsic cell death pathways, which marks the decision point between life and death. Once the mitochondria are irretrievably damaged, they drive the cell into death. Mitochondrial protection displays a promising strategy to protect neurons from death. Previously, the two proteins p53 and PHD1 have been associated with neurodegeneration. Hence, the aim of this study was to investigate the role of those two proteins in neuronal cell death and to evaluate the neuroprotective potential of their particular inhibitors PFTα and DFO, DHB, CPO and oxyquinoline, respectively. Additionally, underlying mechanisms should be elucidated by which these inhibitors mediate their neuroprotective effects. In this study, immortalised mouse hippocampal HT-22 cells were used since they represent an established model of caspase-independent cell death induced by glutamate, termed oxytosis. Moreover, erastin was applied in the same cell line to induce a mode of cell death called ferroptosis. The first part of this study revealed that siRNA-mediated knockdown of p53 delayed glutamate-induced cell death in HT-22 cells for about 2 h, but failed to prevent lipid peroxidation or mitochondrial damage depicted as enhanced mitochondrial fragmentation, depolarisation of the mitochondrial membrane, increased mitochondrial ROS formation and a loss of ATP levels. Both p53 and phospho-p53 did not translocate to the mitochondria upon glutamate challenge indicating that oxytosis was not attributed to a direct action of p53 at the level of mitochondria. The inhibition of p53 transcriptional activity by knockdown of p53, which was determined by a reporter assay established in this work, could serve as a possible explanation for the observed delay of cell death. In contrast, the pharmacological p53-inhibitor PFTα prevented glutamate-induced cell death of HT-22 cells more efficiently and was still able to rescue these cells when applied up to 4 h after the onset of glutamate treatment. Furthermore, PFTα abolished lipid peroxidation and subsequently preserved mitochondrial integrity which was indicated by reduced mitochondrial fission, attenuated formation of mitochondrial ROS and restored mitochondrial membrane potential and ATP levels. Notably, PFTα rescued HT-22 cells depleted of p53 from glutamate-induced cell death. These results exposed a pronounced neuroprotective potential of PFTα which occurred at the level of mitochondria and independently of p53. The second part of this thesis demonstrated the neuroprotective potential of PHD inhibition by the use of structural diverse PHD-inhibitors and siRNAs selectively targeting PHD1. Both concepts of PHD inhibition reduced generation of lipid peroxides and preserved mitochondrial morphology and function indicated by restored mitochondrial respiration and membrane potential and abolished mitochondrial ROS formation, revealing that PHD inhibition acts upstream of mitochondrial demise. Remarkably, the effects by siRNA-mediated PHD1 silencing were less pronounced than those achieved by pharmacological inhibitors. These differences in efficacy were likely attributed to the insufficient knockdown by the siRNA approach. Nevertheless, these findings exposed the selective inhibition of PHD1 and the broad pharmacological inhibition of the PHD enzyme family as promising strategies to achieve mitochondrial rescue and subsequent neuroprotection. Previously, PHDs have been shown to interact with the transcription factor ATF4. The present study revealed that oxyquinoline was able to prevent the glutamate-induced down regulation of ATF4. However, oxyquinoline was still able to prevent oxytosis in cells depleted of ATF4. Therefore, the observed regulation of ATF4 protein levels after oxyquinoline application emerged as dispensable for oxyquinoline mediated protection in oxytosis, although previous studies in vivo suggested a modification of ATF4 transcriptional activity as mode of action for oxyquinoline. Overall, the exact mechanism by which PHD inhibition and, in particular oxyquinoline induced neuroprotection in the paradigm of oxytosis remains elusive so far. Finally, PHD inhibition was also shown to protect HT-22 cells against erastin-induced ferroptosis further supporting the pivotal role of PHDs in neuronal demise and the potential of PHD inhibition as a promising therapeutic strategy in the treatment of neurodegenerative diseases, where oxidative stress contributes to progressive mitochondrial dysfunction and neuronal death.

Bibliographie / References

  1. Martinou, J.-C. & Youle, R.J. Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics. Developmental cell 21, 92–101 (2011).
  2. Dringen, R. & Hirrlinger, J. Glutathione pathways in the brain. Biological chemistry 384, 505–516 (2003).
  3. Natarajan, R., Salloum, F.N., Fisher, B.J., Kukreja, R.C. & Fowler, A.A. Hypoxia inducible factor-1 activation by prolyl 4-hydroxylase-2 gene silencing attenuates myocardial ischemia reperfusion injury. Circulation research 98, 133– 140 (2006).
  4. Tan, S., Somia, N., Maher, P. & Schubert, D. Regulation of antioxidant metabolism by translation initiation factor 2alpha. The Journal of cell biology 152, 997–1006 (2001).
  5. Harding, H.P. et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Molecular cell 11, 619–633 (2003).
  6. Morris, E.J. et al. Cyclin-dependent kinases and P53 pathways are activated independently and mediate Bax activation in neurons after DNA damage. The Journal of neuroscience : the official journal of the Society for Neuroscience 21, 5017–5026 (2001).
  7. Xiang, H. et al. Evidence for p53-mediated modulation of neuronal viability. J. Neurosci. 16, 6753–6765 (1996).
  8. Xiang, H. et al. Bax involvement in p53-mediated neuronal cell death. The Journal of neuroscience : the official journal of the Society for Neuroscience 18, 1363–1373 (1998).
  9. Murray-Zmijewski, F., Lane, D.P. & Bourdon, J.-C. p53/p63/p73 isoforms: an orchestra of isoforms to harmonise cell differentiation and response to stress. Cell death and differentiation 13, 962–972 (2006).
  10. Rocha, S., Campbell, K.J., Roche, K.C. & Perkins, N.D. The p53-inhibitor pifithrin-alpha inhibits firefly luciferase activity in vivo and in vitro. BMC Mol. Biol. 4, 9 (2003).
  11. Vanden Berghe, T., Linkermann, A., Jouan-Lanhouet, S., Walczak, H. & Vandenabeele, P. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nature reviews. Molecular cell biology 15, 135–147 (2014).
  12. Bonini, P. et al. Oxidative stress induces p53-mediated apoptosis in glia: p53 transcription-independent way to die. Journal of neuroscience research 75, 83–95 (2004).
  13. Öxler, E.-M., Dolga, A. & Culmsee, C. AIF depletion provides neuroprotection through a preconditioning effect. Apoptosis 17, 1027–1038 (2012).
  14. Reuther, C., Ganjam, G.K., Dolga, A.M. & Culmsee, C. The serine protease inhibitor TLCK attenuates intrinsic death pathways in neurons upstream of mitochondrial demise. Apoptosis (2014).
  15. Rohrbach, S., Simm, A., Pregla, R., Franke, C. & Katschinski, D.M. Age- dependent increase of prolyl-4-hydroxylase domain (PHD) 3 expression in human and mouse heart. Biogerontology 6, 165–171 (2005).
  16. Racay, P., Tatarkova, Z., Drgova, A., Kaplan, P. & Dobrota, D. Effect of ischemic preconditioning on mitochondrial dysfunction and mitochondrial p53 translocation after transient global cerebral ischemia in rats. Neurochemical research 32, 1823–1832 (2007).
  17. Yu, J. & Zhang, L. The transcriptional targets of p53 in apoptosis control. Biochem. Biophys. Res. Commun. 331, 851–858 (2005).
  18. Cervera, A.M. et al. An alternatively spliced transcript of the PHD3 gene retains prolyl hydroxylase activity. Cancer letters 233, 131–138 (2006).
  19. Smirnova, N.A. et al. Utilization of an in vivo reporter for high throughput identification of branched small molecule regulators of hypoxic adaptation. Chemistry & biology 17, 380–391 (2010).
  20. Fukui, M., Song, J.-H., Choi, J., Choi, H.J. & Zhu, B.T. Mechanism of glutamate- induced neurotoxicity in HT22 mouse hippocampal cells. Eur. J. Pharmacol. 617, 1–11 (2009).
  21. Yang, L.-Y. et al. Post-trauma administration of the pifithrin-α oxygen analog improves histological and functional outcomes after experimental traumatic brain injury. Experimental neurology 269, 56–66 (2015).
  22. Li, Z. et al. ROS leads to MnSOD upregulation through ERK2 translocation and p53 activation in selenite-induced apoptosis of NB4 cells. FEBS letters 584, 2291–2297 (2010).
  23. Speer, R.E. et al. Hypoxia-inducible factor prolyl hydroxylases as targets for neuroprotection by "antioxidant" metal chelators: From ferroptosis to stroke. Free Radic. Biol. Med. 62, 26–36 (2013).
  24. Checler, F. & Alves da Costa, Cristine. p53 in neurodegenerative diseases and brain cancers. Pharmacol. Ther. 142, 99–113 (2014).
  25. Xie, L. et al. Depletion of PHD3 protects heart from ischemia/reperfusion injury by inhibiting cardiomyocyte apoptosis. Journal of molecular and cellular cardiology 80C, 156–165 (2015).
  26. Epstein, A.C. et al. C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 107, 43–54 (2001).
  27. Mattson, M.P. Modification of ion homeostasis by lipid peroxidation: roles in neuronal degeneration and adaptive plasticity. Trends Neurosci. 21, 53–57 (1998).
  28. Yukawa, K., Tanaka, T., Tsuji, S. & Akira, S. Regulation of transcription factor C/ATF by the cAMP signal activation in hippocampal neurons, and molecular interaction of C/ATF with signal integrator CBP/p300. Brain research. Molecular brain research 69, 124–134 (1999).
  29. Rabinowitz, M.H. Inhibition of hypoxia-inducible factor prolyl hydroxylase domain oxygen sensors: tricking the body into mounting orchestrated survival and repair responses. Journal of medicinal chemistry 56, 9369–9402 (2013).
  30. Jansson, M. et al. Arginine methylation regulates the p53 response. Nat. Cell Biol. 10, 1431–1439 (2008).
  31. Aragonés, J. et al. Deficiency or inhibition of oxygen sensor Phd1 induces hypoxia tolerance by reprogramming basal metabolism. Nature genetics 40, 170– 180 (2008).
  32. Stambolsky, P. et al. Regulation of AIF expression by p53. Cell death and differentiation 13, 2140–2149 (2006).
  33. Plesnila, N. et al. Delayed neuronal death after brain trauma involves p53- dependent inhibition of NF-kappaB transcriptional activity. Cell Death Differ. 14, 1529–1541 (2007).
  34. Yonekura, I., Takai, K., Asai, A., Kawahara, N. & Kirino, T. p53 potentiates hippocampal neuronal death caused by global ischemia. J. Cereb. Blood Flow Metab. 26, 1332–1340 (2006).
  35. Semont, A. et al. Involvement of p53 and Fas/CD95 in murine neural progenitor cell response to ionizing irradiation. Oncogene 23, 8497–8508 (2004).
  36. Endo, H., Saito, A. & Chan, P.H. Mitochondrial translocation of p53 underlies the selective death of hippocampal CA1 neurons after global cerebral ischaemia. Biochemical Society transactions 34, 1283–1286 (2006).
  37. Culmsee, C. et al. A synthetic inhibitor of p53 protects neurons against death induced by ischemic and excitotoxic insults, and amyloid beta-peptide. J. Neurochem. 77, 220–228 (2001).
  38. Wolpaw, A.J. et al. Modulatory profiling identifies mechanisms of small molecule- induced cell death. Proceedings of the National Academy of Sciences of the United States of America 108, E771-80 (2011).
  39. Keramaris, E., Hirao, A., Slack, R.S., Mak, T.W. & Park, D.S. Ataxia telangiectasia-mutated protein can regulate p53 and neuronal death independent of Chk2 in response to DNA damage. The Journal of biological chemistry 278, 37782–37789 (2003).
  40. Chen, H. & Chan, D.C. Mitochondrial dynamics--fusion, fission, movement, and mitophagy--in neurodegenerative diseases. Human molecular genetics 18, R169-76 (2009).
  41. Haupt, Y., Rowan, S., Shaulian, E., Vousden, K.H. & Oren, M. Induction of apoptosis in HeLa cells by trans-activation-deficient p53. Genes & development 9, 2170–2183 (1995).
  42. Wottawa, M., Köditz, J. & Katschinski, D.M. Normoxic destabilization of ATF-4 depends on proteasomal degradation. Acta physiologica (Oxford, England) 198, 457–463 (2010).
  43. Chen, R.-L. et al. Roles of individual prolyl-4-hydroxylase isoforms in the first 24 hours following transient focal cerebral ischaemia: insights from genetically References 160
  44. Oppermann, S. et al. Novel N-phenyl-substituted thiazolidinediones protect neural cells against glutamate-and tBid-induced toxicity. J. Pharmacol. Exp. Ther. 350, 273–289 (2014).
  45. Matoba, S. et al. p53 regulates mitochondrial respiration. Science (New York, N.Y.) 312, 1650–1653 (2006).
  46. Lieb, M.E., Menzies, K., Moschella, M.C., Ni, R. & Taubman, M.B. Mammalian EGLN genes have distinct patterns of mRNA expression and regulation.
  47. Metzen, E. et al. Intracellular localisation of human HIF-1 alpha hydroxylases: implications for oxygen sensing. Journal of cell science 116, 1319–1326 (2003).
  48. Gilman, C.P. et al. p53 is present in synapses where it mediates mitochondrial dysfunction and synaptic degeneration in response to DNA damage, and oxidative and excitotoxic insults. Neuromolecular Med. 3, 159–172 (2003).
  49. Steckley, D. et al. Puma is a dominant regulator of oxidative stress induced Bax activation and neuronal apoptosis. The Journal of neuroscience : the official journal of the Society for Neuroscience 27, 12989–12999 (2007).
  50. Tan, S., Schubert, D. & Maher, P. Oxytosis: A novel form of programmed cell death. Curr Top Med Chem 1, 497–506 (2001).
  51. Aoyama, K. & Nakaki, T. Impaired glutathione synthesis in neurodegeneration. International journal of molecular sciences 14, 21021–21044 (2013).
  52. Chou, J., Greig, N.H., Reiner, D., Hoffer, B.J. & Wang, Y. Enhanced survival of dopaminergic neuronal transplants in hemiparkinsonian rats by the p53 inactivator PFT-α. Cell Transplant 20, 1351–1359 (2011).
  53. Kim, E.Y. et al. Proteomic analysis of oxidative stress-induced neuronal cell death by using two-dimensional fluorescence difference gel electrophoresis. Int. J.
  54. Bensaad, K. et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell 126, 107–120 (2006).
  55. Nakano, K. & Vousden, K.H. PUMA, a novel proapoptotic gene, is induced by p53. Mol. Cell 7, 683–694 (2001).
  56. Vousden, K.H. & Lane, D.P. p53 in health and disease. Nature reviews. Molecular cell biology 8, 275–283 (2007).
  57. Vousden, K.H. & Prives, C. Blinded by the Light: The Growing Complexity of p53. Cell 137, 413–431 (2009).
  58. Schlisio, S. et al. The kinesin KIF1Bbeta acts downstream from EglN3 to induce apoptosis and is a potential 1p36 tumor suppressor. Genes & development 22, 884–893 (2008).
  59. Bishop, T. et al. Abnormal sympathoadrenal development and systemic hypotension in PHD3-/-mice. Molecular and cellular biology 28, 3386–3400 (2008).
  60. Seiler, A. et al. Glutathione peroxidase 4 senses and translates oxidative stress into 12/15-lipoxygenase dependent-and AIF-mediated cell death. Cell Metab. 8, 237–248 (2008).
  61. Vande Walle, L. et al. Proteome-wide Identification of HtrA2/Omi Substrates. Journal of proteome research 6, 1006–1015 (2007).
  62. Hoagland, M.S., Hoagland, E.M. & Swanson, H.I. The p53 inhibitor pifithrin- alpha is a potent agonist of the aryl hydrocarbon receptor. J. Pharmacol. Exp.
  63. Neitemeier, S., Ganjam, G.K., Diemert, S. & Culmsee, C. Pifithrin-α provides neuroprotective effects at the level of mitochondria independently of p53 inhibition. Apoptosis : an international journal on programmed cell death 19, 1665–1677 (2014).
  64. Morrison, R.S., Kinoshita, Y., Johnson, M.D., Guo, W. & Garden, G.A. p53- dependent cell death signaling in neurons. Neurochemical research 28, 15–27 (2003).
  65. 11 th International Conference on Alzheimer's and Parkinson's Diseases, Florence, Italy, 06.-10.03.2013
  66. Ameri, K. & Harris, A.L. Activating transcription factor 4. The international journal of biochemistry & cell biology 40, 14–21 (2008).
  67. Adaptaquin is an inhibitor of oxygen-sensing prolyl-hydroxylase domain enzymes that abrogates ATF4-dependent death and improves outcomes from brain hemorrhage, submitted Neitemeier S # , Oppermann S # , Laino V # , Jelinek A, Ganjam GK, Dolga AM,
  68. van Heemst, D., den Reijer, P M & Westendorp, R G J. Ageing or cancer: a review on the role of caretakers and gatekeepers. European journal of cancer (Oxford, England : 1990) 43, 2144–2152 (2007).
  69. Wallace, D.C. A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annual review of genetics 39, 359–407 (2005).
  70. Zhu, C. et al. Apoptosis-inducing factor is a major contributor to neuronal loss induced by neonatal cerebral hypoxia-ischemia. Cell death and differentiation 14, 775–784 (2007).
  71. Crumrine, R.C., Thomas, A.L. & Morgan, P.F. Attenuation of p53 expression protects against focal ischemic damage in transgenic mice. J. Cereb. Blood Flow Metab. 14, 887–891 (1994).
  72. Behl, C., Hovey, L., Krajewski, S., Schubert, D. & Reed, J.C. BCL-2 prevents killing of neuronal cells by glutamate but not by amyloid beta protein. Biochemical and biophysical research communications 197, 949–956 (1993).
  73. Yin, X.-M. Bid, a BH3-only multi-functional molecule, is at the cross road of life and death. Gene 369, 7–19 (2006).
  74. Neitemeier S, Laino V, Oppermann S, Ganjam GK, Dolga AM, Culmsee C: Bid links ferroptosis to mitochondrial cell death pathways in neurons. DPhG Jahrestagung 2014, Frankfurt, Germany, 24.-26.09.2014
  75. Culmsee C: Bid links ferroptosis to mitochondrial cell death pathways in neurons, submitted # contributed equally 9.2 Books Dolga AM, Oppermann S, Richter M, Honrath B, Neitemeier S, Jelinek A,
  76. Laino, V. Bid links ferroptosis to mitochondrial demise and intrinsic pathways of cell death. Bachelor thesis (Perugia, 2014).
  77. Ganjam GK, Dolga AM, Neitemeier S, Bolte K, Höllerhage M, Oertel WE, Hoeglinger GU, Culmsee C: Caspase inhibition mitigates alpha synuclein cytotoxicity and mitochondrial demise in human dopaminergic neurons. Society for Neuroscience Annual Meeting, Neuroscience 2014, Washington, USA, 15.-19.11.2014
  78. Ali, S.H. & DeCaprio, J.A. Cellular transformation by SV40 large T antigen: interaction with host proteins. Seminars in cancer biology 11, 15–23 (2001).
  79. Sato, H., Tamba, M., Ishii, T. & Bannai, S. Cloning and expression of a plasma membrane cystine/glutamate exchange transporter composed of two distinct proteins. The Journal of biological chemistry 274, 11455–11458 (1999).
  80. D-aspartate-mediated secretion in a neural cell line. Proc. Natl. Acad. Sci. U.S.A. 87, 3518–3521 (1990).
  81. Takeda, K., Cowan, A. & Fong, G.-H. Essential role for prolyl hydroxylase domain protein 2 in oxygen homeostasis of the adult vascular system. Circulation 116, 774–781 (2007).
  82. factor-dependent neurons. The Journal of biological chemistry 276, 5085–5092 (2001).
  83. Dixon, S.J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012).
  84. Skouta, R. et al. Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models. J. Am. Chem. Soc. 136, 4551–4556 (2014).
  85. Murphy, T.H., Miyamoto, M., Sastre, A., Schnaar, R.L. & Coyle, J.T. Glutamate toxicity in a neuronal cell line involves inhibition of cystine transport leading to oxidative stress. Neuron 2, 1547–1558 (1989).
  86. Ganjam GK, Dolga AM, Neitemeier S, de Andrade A, Bolte K, Oertel W, Höglinger GU, Culmsee C: Human dopaminergic neurons as a model system for Parkinson's disease. 8th International Symposium on Neuroprotection and Neurorepair, Magdeburg, Germany, 09.-12.04.2014
  87. Miyashita, T., Harigai, M., Hanada, M. & Reed, J.C. Identification of a p53- dependent negative response element in the bcl-2 gene. Cancer research 54, 3131–3135 (1994).
  88. Diemert, S. et al. Impedance measurement for real time detection of neuronal cell death. J. Neurosci. Methods 203, 69–77 (2012).
  89. Straub, J.A., Lipscomb, E.A., Yoshida, E.S. & Freeman, R.S. Induction of SM- 20 in PC12 cells leads to increased cytochrome c levels, accumulation of cytochrome c in the cytosol, and caspase-dependent cell death. Journal of neurochemistry 85, 318–328 (2003).
  90. Lee, D.W. et al. Inhibition of prolyl hydroxylase protects against 1-methyl-4- phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity: model for the potential involvement of the hypoxia-inducible factor pathway in Parkinson disease. The Journal of biological chemistry 284, 29065–29076 (2009).
  91. Neitemeier S, Dolga AM, Karuppagounder S, Ratan R, Culmsee C: Inhibitors of HIF- proly-4-hydroxylases (HIF-PHD) mediate mitochondrial protection and prevent oxytosis in neuronal cells. Society for Neuroscience Annual Meeting, Neuroscience 2014, Washington, USA, 15.-19.11.2014
  92. Andersen, H.H., Johnsen, K.B. & Moos, T. Iron deposits in the chronically inflamed central nervous system and contributes to neurodegeneration. Cellular and molecular life sciences : CMLS 71, 1607–1622 (2014).
  93. Mielke, K. & Herdegen, T. JNK and p38 stresskinases--degenerative effectors of signal-transduction-cascades in the nervous system. Progress in neurobiology 61, 45–60 (2000).
  94. Morrison, R.S. et al. Loss of the p53 tumor suppressor gene protects neurons from kainate-induced cell death. The Journal of neuroscience : the official journal of the Society for Neuroscience 16, 1337–1345 (1996).
  95. Kim, G.W., Kondo, T., Noshita, N. & Chan, P.H. Manganese superoxide dismutase deficiency exacerbates cerebral infarction after focal cerebral ischemia/reperfusion in mice: implications for the production and role of superoxide radicals. Stroke; a journal of cerebral circulation 33, 809–815 (2002).
  96. Flamme, I. et al. Mimicking hypoxia to treat anemia: HIF-stabilizer BAY 85- 3934 (Molidustat) stimulates erythropoietin production without hypertensive effects. PloS one 9, e111838 (2014).
  97. Pfeiffer, A. et al. Mitochondrial function and energy metabolism in neuronal HT22 cells resistant to oxidative stress. British journal of pharmacology 171, 2147–2158 (2014).
  98. Fleury, C., Mignotte, B. & Vayssière, J.-L. Mitochondrial reactive oxygen species in cell death signaling. Biochimie 84, 131–141 (2002).
  99. Endo, H., Kamada, H., Nito, C., Nishi, T. & Chan, P.H. Mitochondrial translocation of p53 mediates release of cytochrome c and hippocampal CA1 neuronal death after transient global cerebral ischemia in rats. The Journal of neuroscience : the official journal of the Society for Neuroscience 26, 7974–7983 (2006).
  100. Sancar, A., Lindsey-Boltz, L.A., Unsal-Kaçmaz, K. & Linn, S. Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Annual review of biochemistry 73, 39–85 (2004).
  101. Galluzzi, L., Kepp, O., Krautwald, S., Kroemer, G. & Linkermann, A. Molecular mechanisms of regulated necrosis. Semin. Cell Dev. Biol. (2014).
  102. Ganjam GK, Culmsee C: 'Molecular mechanisms underlying oxytosis' in 'Apoptosis and Beyond: the way cells die, Springer (submitted)
  103. Grohm, J. Molecular regulation of mitochondrial dynamics by dynamin-related protein 1 (Drp1) and Bid in model systems of neuronal cell death. Dissertation (Marburg, 2011).
  104. Lee, S. et al. Neuronal apoptosis linked to EglN3 prolyl hydroxylase and familial pheochromocytoma genes: developmental culling and cancer. Cancer cell 8, 155–167 (2005).
  105. Kunze, R. et al. Neuron-specific prolyl-4-hydroxylase domain 2 knockout reduces brain injury after transient cerebral ischemia. Stroke; a journal of cerebral circulation 43, 2748–2756 (2012).
  106. Zhu, X. et al. Novel p53 Inactivators with Neuroprotective Action: Syntheses and Pharmacological Evaluation of 2-Imino-2,3,4,5,6,7-hexahydrobenzothiazole and 2-Imino-2,3,4,5,6,7-hexahydrobenzoxazole Derivatives ⊥. J. Med. Chem. 45, 5090–5097 (2002).
  107. Copin, J.C., Gasche, Y. & Chan, P.H. Overexpression of copper/zinc superoxide dismutase does not prevent neonatal lethality in mutant mice that lack manganese superoxide dismutase. Free radical biology & medicine 28, 1571– 1576 (2000).
  108. Ratan, R.R., Murphy, T.H. & Baraban, J.M. Oxidative stress induces apoptosis in embryonic cortical neurons. Journal of neurochemistry 62, 376–379 (1994).
  109. Culmsee, C. & Mattson, M.P. p53 in neuronal apoptosis. Biochem. Biophys. Res. Commun. 331, 761–777 (2005).
  110. Bae, B.-I. et al. p53 mediates cellular dysfunction and behavioral abnormalities in Huntington's disease. Neuron 47, 29–41 (2005).
  111. Kelly, K.J. P53 Mediates the Apoptotic Response to GTP Depletion after Renal Ischemia-Reperfusion: Protective Role of a p53 Inhibitor. Journal of the American Society of Nephrology 14, 128–138 (2003).
  112. Dixon, S.J. et al. Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. eLife 3, e02523 (2014).
  113. Zaman, K. et al. Protection from oxidative stress-induced apoptosis in cortical neuronal cultures by iron chelators is associated with enhanced DNA binding of hypoxia-inducible factor-1 and ATF-1/CREB and increased expression of glycolytic enzymes, p21(waf1/cip1), and erythropoietin. The Journal of neuroscience : the official journal of the Society for Neuroscience 19, 9821–9830 (1999).
  114. Davis, J.B. & Maher, P. Protein kinase C activation inhibits glutamate-induced cytotoxicity in a neuronal cell line. Brain research 652, 169–173 (1994).
  115. Culmsee, C. et al. Reciprocal inhibition of p53 and nuclear factor-kappaB transcriptional activities determines cell survival or death in neurons. J. Neurosci. 23, 8586–8595 (2003).
  116. References 147
  117. References 148
  118. References 150
  119. References 151
  120. References 152
  121. References 153
  122. References 155
  123. References 156
  124. References 158
  125. References 159
  126. References 162
  127. References 163
  128. References 165
  129. References 166
  130. Yang, W.S. et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014).
  131. Siddiq, A. et al. Selective inhibition of hypoxia-inducible factor (HIF) prolyl- hydroxylase 1 mediates neuroprotection against normoxic oxidative death via HIF- and CREB-independent pathways. J. Neurosci. 29, 8828–8838 (2009).
  132. Lipscomb, E.A., Sarmiere, P.D. & Freeman, R.S. SM-20 is a novel mitochondrial protein that causes caspase-dependent cell death in nerve growth References 161
  133. Strom, E. et al. Small-molecule inhibitor of p53 binding to mitochondria protects mice from gamma radiation. Nat. Chem. Biol. 2, 474–479 (2006).
  134. Takeda, K. et al. Placental but not heart defects are associated with elevated hypoxia-inducible factor alpha levels in mice lacking prolyl hydroxylase domain protein 2. Molecular and cellular biology 26, 8336–8346 (2006).
  135. Friedmann Angeli, Jose Pedro et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nature cell biology 16, 1180–1191 (2014).
  136. Galluzzi, L. et al. Mitochondrial liaisons of p53. Antioxidants & redox signaling 15, 1691–1714 (2011).
  137. Duan, W. et al. p53 inhibitors preserve dopamine neurons and motor function in experimental parkinsonism. Ann. Neurol. 52, 597–606 (2002).
  138. Liu, Y., Fiskum, G. & Schubert, D. Generation of reactive oxygen species by the mitochondrial electron transport chain. Journal of neurochemistry 80, 780–787 (2002).
  139. Scortegagna, M. et al. HIF-2alpha regulates murine hematopoietic development in an erythropoietin-dependent manner. Blood 105, 3133–3140 (2005).
  140. Masuoka, H.C. & Townes, T.M. Targeted disruption of the activating transcription factor 4 gene results in severe fetal anemia in mice. Blood 99, 736– 745 (2002).
  141. Oriowo, B. et al. Targeted gene deletion of prolyl hydroxylase domain protein 3 triggers angiogenesis and preserves cardiac function by stabilizing hypoxia inducible factor 1 alpha following myocardial infarction. Current pharmaceutical design 20, 1305–1310 (2014).
  142. Komarova, E.A. et al. p53 inhibitor pifithrin alpha can suppress heat shock and glucocorticoid signaling pathways. J. Biol. Chem. 278, 15465–15468 (2003).
  143. Murphy, Patrick J M et al. Pifithrin-alpha inhibits p53 signaling after interaction of the tumor suppressor protein with hsp90 and its nuclear translocation. J. Biol.
  144. Lewerenz, J. & Maher, P. Basal Levels of eIF2 Phosphorylation Determine Cellular Antioxidant Status by Regulating ATF4 and xCT Expression. Journal of Biological Chemistry 284, 1106–1115 (2009).
  145. Maher, P. & Davis, J.B. The role of monoamine metabolism in oxidative glutamate toxicity. The Journal of neuroscience : the official journal of the Society for Neuroscience 16, 6394–6401 (1996).
  146. Dixon, S.J. & Stockwell, B.R. The role of iron and reactive oxygen species in cell death. Nat. Chem. Biol. 10, 9–17 (2014).
  147. Patel, S.A. & Simon, M.C. Biology of hypoxia-inducible factor-2alpha in development and disease. Cell death and differentiation 15, 628–634 (2008).
  148. Sohn, D. et al. Pifithrin-α protects against DNA damage-induced apoptosis downstream of mitochondria independent of p53. Cell Death Differ 16, 869–878 (2009).
  149. Green, D.R. & Kroemer, G. Cytoplasmic functions of the tumour suppressor p53. Nature 458, 1127–1130 (2009).
  150. Vousden, K.H. & Vande Woude, G F. The ins and outs of p53. Nature cell biology 2, E178-80 (2000).
  151. Sax, J.K. et al. BID regulation by p53 contributes to chemosensitivity. Nature cell biology 4, 842–849 (2002).
  152. Toledo, F. & Wahl, G.M. Regulating the p53 pathway: in vitro hypotheses, in vivo veritas. Nat. Rev. Cancer 6, 909–923 (2006).
  153. Kang, Y., Tiziani, S., Park, G., Kaul, M. & Paternostro, G. Cellular protection using Flt3 and PI3Kα inhibitors demonstrates multiple mechanisms of oxidative glutamate toxicity. Nat Commun 5, 3672 (2014).
  154. Tian, Y.-M., Mole, D.R., Ratcliffe, P.J. & Gleadle, J.M. Characterization of different isoforms of the HIF prolyl hydroxylase PHD1 generated by alternative initiation. The Biochemical journal 397, 179–186 (2006).
  155. Yu, J., Wang, Z., Kinzler, K.W., Vogelstein, B. & Zhang, L. PUMA mediates the apoptotic response to p53 in colorectal cancer cells. Proc. Natl. Acad. Sci. U.S.A. 100, 1931–1936 (2003).
  156. Tan, S., Sagara, Y., Liu, Y., Maher, P. & Schubert, D. The regulation of reactive oxygen species production during programmed cell death. J. Cell Biol. 141, 1423– 1432 (1998).
  157. Fortin, A. et al. APAF1 is a key transcriptional target for p53 in the regulation of neuronal cell death. The Journal of cell biology 155, 207–216 (2001).
  158. Takeda, K. et al. Regulation of adult erythropoiesis by prolyl hydroxylase domain proteins. Blood 111, 3229–3235 (2008).
  159. Mikhaylova, O. et al. The von Hippel-Lindau tumor suppressor protein and Egl- 9-Type proline hydroxylases regulate the large subunit of RNA polymerase II in response to oxidative stress. Molecular and cellular biology 28, 2701–2717 (2008).
  160. Lange, P.S. et al. ATF4 is an oxidative stress-inducible, prodeath transcription factor in neurons in vitro and in vivo. The Journal of experimental medicine 205, 1227–1242 (2008).
  161. Siddiq, A., Aminova, L.R. & Ratan, R.R. Hypoxia inducible factor prolyl 4- hydroxylase enzymes: center stage in the battle against hypoxia, metabolic compromise and oxidative stress. Neurochemical research 32, 931–946 (2007).
  162. Niizuma, K., Endo, H., Nito, C., Myer, D.J. & Chan, P.H. Potential role of PUMA in delayed death of hippocampal CA1 neurons after transient global cerebral ischemia. Stroke 40, 618–625 (2009).
  163. Yang, W.S. & Stockwell, B.R. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chemistry & biology 15, 234–245 (2008).
  164. Bensaad, K., Cheung, E.C. & Vousden, K.H. Modulation of intracellular ROS levels by TIGAR controls autophagy. The EMBO journal 28, 3015–3026 (2009).
  165. Pallast, S., Arai, K., Wang, X., Lo, E.H. & van Leyen, K. 12/15-Lipoxygenase targets neuronal mitochondria under oxidative stress. J. Neurochem. 111, 882– 889 (2009).
  166. Zhang, Q. et al. Control of cyclin D1 and breast tumorigenesis by the EglN2 prolyl hydroxylase. Cancer cell 16, 413–424 (2009).
  167. Landshamer, S. et al. Bid-induced release of AIF from mitochondria causes immediate neuronal cell death. Cell Death Differ. 15, 1553–1563 (2008).
  168. Beckerman, R. & Prives, C. Transcriptional regulation by p53. Cold Spring Harb Perspect Biol 2, a000935 (2010).
  169. Yagoda, N. et al. RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature 447, 864–868 (2007).
  170. Tobaben, S. 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. 18, 282–292 (2011).
  171. Engel, T. et al. Reduced hippocampal damage and epileptic seizures after status epilepticus in mice lacking proapoptotic Puma. FASEB J. 24, 853–861 (2010).
  172. Lewerenz, J. et al. Mutation of ATF4 mediates resistance of neuronal cell lines against oxidative stress by inducing xCT expression. Cell Death Differ 19, 847– 858 (2011).
  173. Vaseva, A.V. et al. p53 opens the mitochondrial permeability transition pore to trigger necrosis. Cell 149, 1536–1548 (2012).
  174. Karuppagounder, S.S. & Ratan, R.R. Hypoxia-inducible factor prolyl hydroxylase inhibition: robust new target or another big bust for stroke therapeutics? J. Cereb. Blood Flow Metab. 32, 1347–1361 (2012).
  175. Grohm, J. et al. Inhibition of Drp1 provides neuroprotection in vitro and in vivo. Cell Death Differ. 19, 1446–1458 (2012).
  176. Henke, N. et al. The plasma membrane channel ORAI1 mediates detrimental calcium influx caused by endogenous oxidative stress. Cell death & disease 4, e470 (2013).
  177. Dolga, A.M. et al. Mitochondrial small conductance SK2 channels prevent glutamate-induced oxytosis and mitochondrial dysfunction. The Journal of biological chemistry 288, 10792–10804 (2013).
  178. Li, D., Bai, T. & Brorson, J.R. Adaptation to moderate hypoxia protects cortical neurons against ischemia-reperfusion injury and excitotoxicity independently of HIF-1α. Experimental neurology 230, 302–310 (2011).
  179. Durán, R.V. et al. HIF-independent role of prolyl hydroxylases in the cellular response to amino acids. Oncogene 32, 4549–4556 (2013).
  180. Kreiner, G. et al. A neuroprotective phase precedes striatal degeneration upon nucleolar stress. Cell Death Differ. 20, 1455–1464 (2013).
  181. Guo, X. et al. Inhibition of mitochondrial fragmentation diminishes Huntington's disease-associated neurodegeneration. The Journal of clinical investigation 123, 5371–5388 (2013).
  182. McBride, H.M., Neuspiel, M. & Wasiak, S. Mitochondria: more than just a powerhouse. Current biology : CB 16, R551-60 (2006).
  183. Linkermann, A. et al. Synchronized renal tubular cell death involves ferroptosis. Proceedings of the National Academy of Sciences of the United States of America 111, 16836–16841 (2014).
  184. Poster presentations 2014
  185. Schödel, J. et al. HIF-prolyl hydroxylases in the rat kidney: physiologic expression patterns and regulation in acute kidney injury. The American journal of pathology 174, 1663–1674 (2009).
  186. Ord, D., Meerits, K. & Ord, T. TRB3 protects cells against the growth inhibitory and cytotoxic effect of ATF4. Experimental cell research 313, 3556–3567 (2007).
  187. Hiwatashi, Y. et al. PHD1 interacts with ATF4 and negatively regulates its transcriptional activity without prolyl hydroxylation. Experimental cell research 317, 2789–2799 (2011).
  188. Leker, R.R., Aharonowiz, M., Greig, N.H. & Ovadia, H. The role of p53-induced apoptosis in cerebral ischemia: effects of the p53 inhibitor pifithrin alpha. Exp. Neurol. 187, 478–486 (2004).
  189. Vousden, K.H. p53: death star. Cell 103, 691–694 (2000).
  190. Chan, D.C. Mitochondria: dynamic organelles in disease, aging, and development. Cell 125, 1241–1252 (2006).
  191. Schutte, B., Nuydens, R., Geerts, H. & Ramaekers, F. Annexin V binding assay as a tool to measure apoptosis in differentiated neuronal cells. Journal of Neuroscience Methods 86, 63–69 (1998).
  192. Tomasevic, G., Shamloo, M., Israeli, D. & Wieloch, T. Activation of p53 and its target genes p21WAF1/Cip1 and PAG608/Wig-1 in ischemic preconditioning. Molecular Brain Research 70, 304–313 (1999).
  193. 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. 24, 831–838 (2010).
  194. Wang, D.B., Kinoshita, C., Kinoshita, Y. & Morrison, R.S. p53 and mitochondrial function in neurons. Biochim. Biophys. Acta (2014).
  195. Moll, U.M., Wolff, S., Speidel, D. & Deppert, W. Transcription-independent pro- apoptotic functions of p53. Current opinion in cell biology 17, 631–636 (2005).
  196. Hermeking, H. et al. 14-3-3 sigma is a p53-regulated inhibitor of G2/M progression. Mol. Cell 1, 3–11 (1997).
  197. Kim, J.-w., Tchernyshyov, I., Semenza, G.L. & Dang, C.V. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell metabolism 3, 177–185 (2006).
  198. Mendjargal, A. et al. Pifithrin-α, a pharmacological inhibitor of p53, downregulates lipopolysaccharide-induced nitric oxide production via impairment of the MyD88-independent pathway. Int. Immunopharmacol. 15, 671–678 (2013).
  199. Komarov, P.G. et al. A chemical inhibitor of p53 that protects mice from the side effects of cancer therapy. Science 285, 1733–1737 (1999).
  200. Bruick, R.K. & McKnight, S.L. A conserved family of prolyl-4-hydroxylases that modify HIF. Science (New York, N.Y.) 294, 1337–1340 (2001).
  201. Köditz, J. et al. Oxygen-dependent ATF-4 stability is mediated by the PHD3 oxygen sensor. Blood 110, 3610–3617 (2007).
  202. Rieker, C. et al. Nucleolar disruption in dopaminergic neurons leads to oxidative damage and parkinsonism through repression of mammalian target of rapamycin signaling. J. Neurosci. 31, 453–460 (2011).


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