Publikationsserver der Universitätsbibliothek Marburg

Titel:Cyclophilin A and serine proteases : Targets of neuronal cell death upstream of mitochondrial demise
Autor:Reuther, Christina
Weitere Beteiligte: Culmsee, Carsten (Prof. Dr.)
Veröffentlicht:2014
URI:https://archiv.ub.uni-marburg.de/diss/z2014/0354
URN: urn:nbn:de:hebis:04-z2014-03540
DOI: https://doi.org/10.17192/z2014.0354
DDC:500 Naturwissenschaften
Titel (trans.):Cyclophilin A und Serinproteasen : Neuroprotektive Zielstrukturen oberhalb der Mitochondrienschädigung
Publikationsdatum:2014-07-02
Lizenz:https://rightsstatements.org/vocab/InC-NC/1.0/

Dokument

Schlagwörter:
Mitochondrium, mitochdria, Oxidativer Stress, oxydated stress

Summary:
Many neurological disorders and neurodegenerative diseases are associated with mitochondrial abnormalities. Mitochondria are essential organelles regulating the energy metabolism of the cell, thereby, determining essential cellular functions and viability. This accounts particularly for neurons which show a pronounced energy demand and where mitochondria play a pivotal role in balancing the Ca2+ homeostasis, controlling ROS formation and providing most of the energy for neurotransmitter metabolism and maintenance of the membrane potential. Further, mitochondria are essential organelles controlling the ‘point of no return’ in intrinsic pathways of programmed cell death where the decision between cellular life and death involves post-translational protein modifications determining mitochondrial dysfunction and release of detrimental proteins such as AIF. Therefore, major aims of the present thesis included the characterization of the key regulators of protein modifications that occur upstream of mitochondrial demise and AIF release. In this context, the role of two PPIases, CypA and Pin1, and the involvement of serine proteases in paradigms of PCD were investigated. Most experiments were performed in an immortalized neuronal cell line (HT22 cells) which depicts a well-established model to study caspase-independent cell death induced by glutamate. The most prominent feature in this model of cell death is the mitochondrial AIF release and the subsequent translocation to the nucleus where it induces chromatinolysis. To substantiate the importance of these results further experiments were performed in a model of glutamate-induced excitotoxicity in primary cortical neurons. The findings of the first part of this study revealed a prominent role for CypA in glutamate-induced mitochondrial AIF release and cell death. Glutamate toxicity resulted in the translocation of CypA to the nucleus where it built a pro-apoptotic complex with AIF, thereby inducing chromatinolysis. Silencing of CypA protected HT22 cells against glutamate toxicity. Moreover, the depletion of CypA preserved mitochondrial fission and the loss of MMP and also prevented the release of AIF from the mitochondria. Furthermore, lipid peroxidation arising from the mitochondria was attenuated which supported increased cell viability. Further experiments addressed the involvement of a second family member of PPIases, Pin1, in neuronal cell death pathways. The inhibition of Pin1 with Br57 resulted in decreased susceptibility of HT22 cells to glutamate toxicity. This increase in cell viability was attended by changes at the level of mitochondria. Pin1 inhibition led to enhanced mitochondrial fission, but further, complete mitochondrial fragmentation induced by glutamate was attenuated. This elevated mitochondrial fission rate was accompanied by a slight decrease in ATP levels in Br57-treated controls, however, the strong ATP depletion that occurred after the glutamate challenge was prevented and the impairment of MMP was abolished. In summary, these findings depict a pivotal role for CypA and Pin1 in glutamate-induced cell death in HT22 cells upstream of mitochondrial demise. Since this model exhibits common features of neurological disorders the results obtained here may give a platform to investigate new neuroprotective strategies. The second part of this thesis dealt with the impact of activated trypsin-like serine proteases on cell viability and mitochondrial function. Inhibition of serine proteases with TLCK resulted in increased cell viability in HT22 cells and in primary cortical neurons. Furthermore, this work revealed that activation of trypsin-like serine proteases occurred upstream of Bid activation and moreover, in a very initial phase of this PCD pathway in HT22 cells. Further, lipid peroxidation was blocked and mitochondrial morphology alterations were prevented. In addition, ATP depletion and the impairment of the MMP were preserved and the decrease in mitochondrial respiration after glutamate toxicity was abolished. Interestingly, the interaction of Drp1 and CypA was strengthened by TLCK. However, the release of Drp1 from the actin cytoskeleton and the following redistribution of Drp1 from the cytoplasm to the mitochondria were not prevented. In conclusion, the second part of this thesis highlighted trypsin-like serine proteases as mediators of glutamate-induced cell death in HT22 cells. The initial activation in this PCD pathway is a promising target for therapeutic intervention strategies in neurological diseases.

Bibliographie / References

  1. Chan DC. Fusion and fission: interlinked processes critical for mitochondrial health. Annu Rev Genet. 2012;46:265–87.
  2. Wulf G, Ryo A, Liou Y, et al. The prolyl isomerase Pin1 in breast development and cancer. Breast Cancer Research. 2003;5:76–88.
  3. Miao Q, Sun Y, Wei T, et al. Chymotrypsin B Cached in Rat Liver Lysosomes and Involved in Apoptotic Regulation through a Mitochondrial Pathway. Journal of Biological Chemistry. 2008;283:8218–28. Accessed January 27, 2014.
  4. Frydrych I, Mlejnek P. Serine protease inhibitors N-alpha-tosyl-L-lysinyl- chloromethylketone (TLCK) and N-tosyl-L-phenylalaninyl-chloromethylketone (TPCK) are potent inhibitors of activated caspase proteases. J Cell Biochem. References 138
  5. Mitsui C, Sakai K, Ninomiya T, et al. Involvement of TLCK-sensitive serine protease in colchicine-induced cell death of sympathetic neurons in culture. J Neurosci Res. 2001;66:601–11.
  6. Saldeen J, Welsh N. Interleukin-1β Induced Activation of NF-κB in Insulin- Producing RINm5F Cells Is Prevented by the Protease Inhibitor Nα-< i> p- Tosyl-L-Lysine Chloromethylketone. Biochemical and Biophysical Research Communications. 1994;203:149–55.
  7. Walker JM. The bicinchoninic acid (BCA) assay for protein quantitation. Methods Mol Biol. 1994;32:5–8.
  8. Kinzel V, Konig N. Interaction of protease inhibitors with the catalytic subunit of cAMP-dependent protein kinase. Biochem Biophys Res Commun. 1980;93:349–53.
  9. Chiu L, Ho F, Shiah S, et al. Oxidative stress initiates DNA damager MNNG- induced poly (ADP-ribose) polymerase-1-dependent parthanatos cell death. Biochemical pharmacology. 2011;81:459–70.
  10. Fukui M, Song J, Choi J, et al. Mechanism of glutamate-induced neurotoxicity in HT22 mouse hippocampal cells. European journal of pharmacology. 2009;617:1–11.
  11. Yamashima T, Oikawa S. The role of lysosomal rupture in neuronal death. Progress in neurobiology. 2009;89:343–58.
  12. Blomgren K, Zhu C, Hallin U, et al. Mitochondria and ischemic reperfusion damage in the adult and in the developing brain. Biochem Biophys Res Commun. 2003;304:551–59.
  13. Zhao M, Brunk UT, Eaton JW. Delayed oxidant-induced cell death involves activation of phospholipase A2. FEBS letters. 2001;509:399–404.
  14. Neumann H, Medana IM, Bauer J, et al. Cytotoxic T lymphocytes in autoimmune and degenerative CNS diseases. Trends in neurosciences. 2002;25:313–19.
  15. Rideout HJ, Zang E, Yeasmin M, et al. Inhibitors of trypsin-like serine proteases prevent DNA damage-induced neuronal death by acting upstream of the mitochondrial checkpoint and of p53 induction. Neuroscience. 2001;107:339– 52.
  16. Roberg K. Relocalization of cathepsin D and cytochrome c early in apoptosis revealed by immunoelectron microscopy. Lab Invest. 2001;81:149–58.
  17. Kazhdan I, Long L, Montellano R, et al. Targeted gene therapy for breast cancer with truncated Bid. Cancer Gene Ther. 2006;13:141–49.
  18. Chen J, Jin K, Chen M, et al. Early detection of DNA strand breaks in the brain after transient focal ischemia: implications for the role of DNA damage in apoptosis and neuronal cell death. Journal of Neurochemistry. 1997;69:232–45.
  19. Polster BM, Basanez G, Etxebarria A, et al. Calpain I induces cleavage and release of apoptosis-inducing factor from isolated mitochondria. J Biol Chem. 2005;280:6447–54.
  20. Zhu C, Wang X, Deinum J, et al. Cyclophilin A participates in the nuclear translocation of apoptosis-inducing factor in neurons after cerebral hypoxia- ischemia. The Journal of experimental medicine. 2007;204:1741–48.
  21. Plesnila N, Zhu C, Culmsee C, et al. Nuclear translocation of apoptosis-inducing factor after focal cerebral ischemia. J Cereb Blood Flow Metab. 2004;24:458– References References 128
  22. Rodriguez J, Lazebnik Y. Caspase-9 and APAF-1 form an active holoenzyme.
  23. Butterfield DA, Abdul HM, Opii W, et al. REVIEW: Pin1 in Alzheimer's disease. Journal of Neurochemistry. 2006;98:1697–706.
  24. Tanaka H, Shimazaki H, Kimura M, et al. Apoptosis‐Inducing Factor and Cyclophilin A Cotranslocate to the Motor Neuronal Nuclei in Amyotrophic Lateral Sclerosis Model Mice. CNS neuroscience & therapeutics. 2011;17:294– 304.
  25. Semba S, Huebner K. Protein expression profiling identifies cyclophilin A as a molecular target in Fhit-mediated tumor suppression. Mol Cancer Res. 2006;4:529–38.
  26. Stoka V, Turk V, Turk B. Lysosomal cysteine cathepsins: signaling pathways in apoptosis. Biological chemistry. 2007;388:555–60.
  27. Zhao K, Zhao X, Tu Y, et al. Lysosomal chymotrypsin B potentiates apoptosis via cleavage of Bid. Cell. Mol. Life Sci. 2010;67:2665–78. Accessed January 27, 2014.
  28. Ovaere P, Lippens S, Vandenabeele P, et al. The emerging roles of serine protease cascades in the epidermis. Trends in biochemical sciences. 2009;34:453–63.
  29. Renzing J, Hansen S, Lane DP. Oxidative stress is involved in the UV activation of p53. Journal of Cell Science. 1996;109:1105–12.
  30. Li J, Li Q, Xie C, et al. β-actin is required for mitochondria clustering and ROS generation in TNF-induced, caspase-independent cell death. Journal of Cell Science. 2004;117:4673–80.
  31. Zou H. An APAF-1 Cytochrome c Multimeric Complex Is a Functional Apoptosome That Activates Procaspase-9. Journal of Biological Chemistry. 1999;274:11549–56.
  32. Ide T, Morikawa E, Kirino T. An immunosuppressant, FK506, protects hippocampal neurons from forebrain ischemia in the mongolian gerbil. Neurosci Lett. 1996;204:157–60.
  33. Hedstrom L. An overview of serine proteases. Curr Protoc Protein Sci. 2002;Chapter 21:Unit 21.10.
  34. Cheung, Eric C C, Melanson-Drapeau L, Cregan SP, et al. Apoptosis-inducing factor is a key factor in neuronal cell death propagated by BAX-dependent and BAX-independent mechanisms. J Neurosci. 2005;25:1324–34.
  35. Haupt S, Berger M, Goldberg Z, et al. Apoptosis-the p53 network. Journal of Cell Science. 2003;116:4077–85.
  36. Williams MS, Henkart PA. Apoptotic cell death induced by intracellular proteolysis. The Journal of Immunology. 1994;153:4247–55.
  37. Daum S, Erdmann F, Fischer G, et al. Aryl Indanyl Ketones: Efficient Inhibitors of the Human Peptidyl Prolylcis/trans Isomerase Pin1. Angew. Chem. Int. Ed. 2006;45:7454–58. Accessed January 27, 2014.
  38. Vander Heiden, Matthew G, Chandel NS, Williamson EK, et al. Bcl-x< sub> L Regulates the Membrane Potential and Volume Homeostasis of Mitochondria. Cell. 1997;91:627–37.
  39. Landshamer S, Hoehn M, Barth N, et al. Bid-induced release of AIF from mitochondria causes immediate neuronal cell death. Cell Death Differ. 2008;15:1553–63.
  40. Guicciardi ME, Bronk SF, Werneburg NW, et al. Bid is upstream of lysosome- mediated caspase 2 activation in tumor necrosis factor alpha-induced hepatocyte apoptosis. Gastroenterology. 2005;129:269–84.
  41. 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. 2011;18:282–92.
  42. Martinou J, Green DR. Breaking the mitochondrial barrier. Nature reviews Molecular cell biology. 2001;2:63–67.
  43. Bidere N, Lorenzo HK, Carmona S, et al. Cathepsin D triggers Bax activation, resulting in selective apoptosis-inducing factor (AIF) relocation in T lymphocytes entering the early commitment phase to apoptosis. J Biol Chem. 2003;278:31401–11.
  44. Schreiber SL. Chemistry and biology of the immunophilins and their immunosuppressive ligands. Science. 1991;251:283–87.
  45. Kim H, Lee HS, Chang KT, et al. Chloromethyl ketones block induction of nitric oxide synthase in murine macrophages by preventing activation of nuclear factor-kappa B. J Immunol. 1995;154:4741–48.
  46. Lu P, Zhou XZ, Liou Y, et al. Critical role of WW domain phosphorylation in regulating phosphoserine binding activity and Pin1 function. Journal of Biological Chemistry. 2002;277:2381–84.
  47. Handschumacher RE, Harding MW, Rice J, et al. Cyclophilin: a specific cytosolic binding protein for cyclosporin A. Science. 1984;226:544–47.
  48. Abraham MC, Shaham S. Death without caspases, caspases without death. Trends Cell Biol. 2004;14:184–93.
  49. Karbowski M, Youle RJ. Dynamics of mitochondrial morphology in healthy cells and during apoptosis. Cell Death & Differentiation. 2003;10:870–80.
  50. Frank S. Dysregulation of mitochondrial fusion and fission: an emerging concept in neurodegeneration. Acta neuropathologica. 2006;111:93–100.
  51. Morimoto BH, Koshland, D E Jr. Excitatory amino acid uptake and N-methyl-D- aspartate-mediated secretion in a neural cell line. Proc Natl Acad Sci U S A. 1990;87:3518–21.
  52. Steiner JP, Dawson TM, Fotuhi M, et al. High brain densities of the immunophilin FKBP colocalized with calcineurin. Nature. 1992;358:584–87.
  53. Asai A, Qiu J, Narita Y, et al. High level calcineurin activity predisposes neuronal cells to apoptosis. Journal of Biological Chemistry. 1999;274:34450– 58.
  54. Diemert S, Dolga AM, Tobaben S, et al. Impedance measurement for real time detection of neuronal cell death. J Neurosci Methods. 2012;203:69–77.
  55. Gorman JJ. Inhibition of human thrombin assessed with different substrates and inhibitors. Characterization of fibrinopeptide binding interaction. Biochim Biophys Acta. 1975;412:273–82.
  56. Gong B, Chen Q, Endlich B, et al. Ionizing radiation-induced, Bax-mediated cell death is dependent on activation of cysteine and serine proteases. Cell Growth Differ. 1999;10:491–502.
  57. Liu Y, Peterson DA, Kimura H, et al. Mechanism of cellular 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction. J Neurochem. 1997;69:581–93.
  58. Yu S, Wang H, Poitras MF, et al. Mediation of poly(ADP-ribose) polymerase-1- dependent cell death by apoptosis-inducing factor. Science. 2002;297:259–63.
  59. Green DR, Reed JC. Mitochondria and apoptosis. Science-AAAS-Weekly Paper Edition. 1998;281:1309–11.
  60. Desagher S, Martinou J. Mitochondria as the central control point of apoptosis. Trends in cell biology. 2000;10:369–77.
  61. Martins LM, Morrison A, Klupsch K, et al. Neuroprotective role of the Reaper- related serine protease HtrA2/Omi revealed by targeted deletion in mice. Molecular and cellular biology. 2004;24:9848–62.
  62. 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;28:249–55.
  63. Ferri KF, Kroemer G. Organelle-specific initiation of cell death pathways. Nat Cell Biol. 2001;3:E255-63.
  64. Göthel SF, Marahiel MA. Peptidyl-prolyl cis-trans isomerases, a superfamily of ubiquitous folding catalysts. Cellular and Molecular Life Sciences CMLS. References 131
  65. Lu KP, Liou Y, Zhou XZ. Pinning down proline-directed phosphorylation signaling. Trends in cell biology. 2002;12:164–72.
  66. Hara A, Niwa M, Nakashima M, et al. Protective effect of apoptosis-inhibitory agent, N-tosyl-L-phenylalanyl chloromethyl ketone against ischemia-induced hippocampal neuronal damage. J Cereb Blood Flow Metab. 1998;18:819–23.
  67. Aoshiba K, Yasuda K, Yasui S, et al. Serine proteases increase oxidative stress in lung cells. Am J Physiol Lung Cell Mol Physiol. 2001;281:L556-64.
  68. Egger L, Schneider J, Rheme C, et al. Serine proteases mediate apoptosis-like cell death and phagocytosis under caspase-inhibiting conditions. Cell Death & Differentiation. 2003;10:1188–203.
  69. Turgeon VL, Houenou LJ. The role of thrombin-like (serine) proteases in the development, plasticity and pathology of the nervous system. Brain Res Brain Res Rev. 1997;25:85–95.
  70. Stoka V, Turk B, Schendel SL, et al. Lysosomal protease pathways to apoptosis. Cleavage of bid, not pro-caspases, is the most likely route. J Biol Chem. 2001;276:3149–57.
  71. Leist M, Jäättelä M. Triggering of apoptosis by cathepsins. Cell death and differentiation. 2001;8:324–26.
  72. Guicciardi ME, Leist M, Gores GJ. Lysosomes in cell death. Oncogene. References 137
  73. Göldner FM, Patrick JW. Neuronal localization of the cyclophilin A protein in the adult rat brain. Journal of Comparative Neurology. 1996;372:283–93.
  74. Lippens G, Landrieu I, Smet C. Molecular mechanisms of the phospho‐dependent prolyl cis/trans isomerase Pin1. Febs Journal. 2007;274:5211–22.
  75. Bleackley RC, Heibein JA. Enzymatic control of apoptosis. Natural product reports. 2001;18:431–40.
  76. Moffitt KL, Martin SL, Walker B. The emerging role of serine proteases in apoptosis. Biochemical Society Transactions. 2007;35:559–60.
  77. Klettner A, Herdegen T. FK506 and its analogs-therapeutic potential for neurological disorders. Current Drug Targets-CNS & Neurological Disorders. 2003;2:153–62.
  78. Agarwal ML, Taylor WR, Chernov MV, et al. The p53 network. Journal of Biological Chemistry. 1998;273:1–4.
  79. Arii T, Kamiya T, Arii K, et al. Neuroprotective effect of immunosuppressant FK506 in transient focal ischemia in rat: therapeutic time window for FK506 in transient focal ischemia. Neurol Res. 2001;23:755–60.
  80. Kroemer G. The proto-oncogene Bcl-2 and its role in regulating apoptosis. Nature medicine. 1997;3:614–20.
  81. Ricci JE, Waterhouse N, Green. Mitochondrial functions during cell death, a complex (IV) dilemma. Cell Death & Differentiation. 2003;10.
  82. Siekierka JJ, Hung SHY, Poe M, et al. A cytosolic binding protein for the immunosuppressant FK506 has peptidyl-prolyl isomerase activity but is distinct from cyclophilin. 1989.
  83. Lu KP, Zhou XZ. The prolyl isomerase PIN1: a pivotal new twist in phosphorylation signalling and disease. Nature reviews Molecular cell biology. References References 133
  84. Knott AB, Perkins G, Schwarzenbacher R, et al. Mitochondrial fragmentation in neurodegeneration. Nature Reviews Neuroscience. 2008;9:505–18.
  85. Doti N, Reuther C, Scognamiglio PL, et al. Inhibition of the AIF/CypA complex protects against intrinsic death pathways induced by oxidative stress. Cell Death Dis. 2014;5:e993.
  86. Zhou Q, Salvesen G. Activation of pro-caspase-7 by serine proteases includes a non-canonical specificity. Biochem. J. 1997;324:361–64.
  87. Capano M, Virji S, Crompton M. Cyclophilin-A is involved in excitotoxin-induced caspase activation in rat neuronal B50 cells. Biochem. J. 2002;363:29–36.
  88. Tan S, Sagara Y, Liu Y, et al. The regulation of reactive oxygen species production during programmed cell death. The Journal of cell biology. 1998;141:1423–32.
  89. Barone MC, Desouza LA, Freeman RS. Pin1 promotes cell death in NGF‐dependent neurons through a mechanism requiring c‐Jun activity. Journal of Neurochemistry. 2008;106:734–45.
  90. Slemmer JE, Zhu C, Landshamer S, et al. Causal role of apoptosis-inducing factor for neuronal cell death following traumatic brain injury. Am J Pathol. 2008;173:1795–805.
  91. Barbier S, Chatre L, Bras M, et al. Caspase-independent type III programmed cell death in chronic lymphocytic leukemia: the key role of the F-actin cytoskeleton. haematologica. 2009;94:507–17.
  92. Becker EBE, Bonni A. Pin1 in neuronal apoptosis. Cell cycle (Georgetown, Tex.). 2007;6:1332.
  93. Artus C, Boujrad H, Bouharrour A, et al. AIF promotes chromatinolysis and caspase‐independent programmed necrosis by interacting with histone H2AX.
  94. Ding X, Patel M, Shen D, et al. Enhanced HtrA2/Omi Expression in Oxidative Injury to Retinal Pigment Epithelial Cells and Murine Models of Neurodegeneration. Investigative Ophthalmology & Visual Science. 2009;50:4957–66.
  95. Davis TL, Walker JR, Campagna-Slater V, et al. Structural and biochemical characterization of the human cyclophilin family of peptidyl-prolyl isomerases. PLoS biology. 2010;8:e1000439.
  96. Wang Y, Kim NS, Haince J, et al. Poly (ADP-ribose)(PAR) binding to apoptosis- inducing factor is critical for PAR polymerase-1-dependent cell death (parthanatos). Science signaling. 2011;4:ra20.
  97. Grohm J, Kim S, Mamrak U, et al. Inhibition of Drp1 provides neuroprotection in vitro and in vivo. Cell Death Differ. 2012;19:1446–58.
  98. Kupfer A, Gani V, Jimenez JS, et al. Affinity labeling of the catalytic subunit of cyclic AMP-dependent protein kinase by N alpha-tosyl-L-lysine chloromethyl ketone. Proceedings of the National Academy of Sciences. 1979;76:3073–77.
  99. Heggeness MH, Simon M, Singer SJ. Association of mitochondria with microtubules in cultured cells. Proceedings of the National Academy of Sciences. 1978;75:3863–66.
  100. Dawson TM, Steiner JP, Dawson VL, et al. Immunosuppressant FK506 enhances phosphorylation of nitric oxide synthase and protects against glutamate neurotoxicity. Proc Natl Acad Sci U S A. 1993;90:9808–12.
  101. Barry M, Heibein JA, Pinkoski MJ, et al. Granzyme B short-circuits the need for caspase 8 activity during granule-mediated cytotoxic T-lymphocyte killing by directly cleaving Bid. Mol Cell Biol. 2000;20:3781–94.
  102. Solomon DH, O'Brian CA, Weinstein IB. N-alpha-Tosyl-L-lysine chloromethyl ketone and N-alpha-tosyl-L-phenylalanine chloromethyl ketone inhibit protein kinase C. FEBS Lett. 1985;190:342–44.
  103. Liu J, Farmer Jr, Jesse D, Lane WS, et al. Calcineurin is a common target of cyclophilin-cyclosporin A and FKBP-FK506 complexes. Cell. 1991;66:807–15.
  104. 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;2:1547–58.
  105. Anesti V, Scorrano L. The relationship between mitochondrial shape and function and the cytoskeleton. Biochimica et Biophysica Acta (BBA)- Bioenergetics. 2006;1757:692–99.
  106. Movsesyan VA, Yakovlev AG, Fan L, et al. Effect of Serine Protease Inhibitors on Posttraumatic Brain Injury and Neuronal Apoptosis. Experimental Neurology. 2001;167:366–75. Accessed January 27, 2014.
  107. Turk B, Turk D, Turk V. Lysosomal cysteine proteases: more than scavengers. Biochimica et Biophysica Acta (BBA)-Protein Structure and Molecular Enzymology. 2000;1477:98–111.
  108. 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, Behavior, and Immunity. 2010;24:831–38.
  109. Frank S, Gaume B, Bergmann-Leitner ES, et al. The role of dynamin-related protein 1, a mediator of mitochondrial fission, in apoptosis. Dev Cell. 2001;1:515–25.
  110. Youle RJ, Van Der Bliek, Alexander M. Mitochondrial fission, fusion, and stress. Science. 2012;337:1062–65.
  111. Rousalova I, Krepela E. Granzyme B-induced apoptosis in cancer cells and its regulation (review). Int J Oncol. 2010;37:1361–78.
  112. Takahashi K. Antioxidative action of N-?-tosyl-L-lysine chloromethyl ketone prevents death of glutathione-depleted cardiomyocytes induced by hydrogen peroxide. JBPC. 2010;01:164–71. Accessed January 27, 2014. References 136


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