Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Apoptosis ; 19(12): 1665-77, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25343947

ABSTRACT

Impaired mitochondrial integrity and function are key features of intrinsic death pathways in neuronal cells. Therefore, key regulators of intrinsic death pathways acting upstream of mitochondria are potential targets for therapeutic approaches of neuroprotection. The tumor suppressor p53 is a well-established regulator of cellular responses towards different kinds of lethal stress, including oxidative stress. Recent reports suggested that p53 may affect mitochondrial integrity and function through both, transcriptional activation of mitochondria-targeted pro-death proteins and direct effects at the mitochondrial membrane. In the present study, we compared the effects of pharmacological inhibition of p53 by pifithrin-α with those of selective p53 gene silencing by RNA interference. Using MTT assay and real-time cell impedance measurements we confirmed the protective effect of both strategies against glutamate-induced oxidative stress in immortalized mouse hippocampal HT-22 neurons. Further, we observed full restoration of mitochondrial membrane potential and inhibition of glutamate-induced mitochondrial fragmentation by pifithrin-α which was, in contrast, not achieved by p53 gene silencing. Downregulation of p53 by siRNA decreased p53 transcriptional activity and reduced expression levels of p21 mRNA, while pifithrin-α did not affect these endpoints. These results suggest a neuroprotective effect of pifithrin-α which occurred at the level of mitochondria and independently of p53 inhibition.


Subject(s)
Benzothiazoles/pharmacology , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Toluene/analogs & derivatives , Tumor Suppressor Protein p53/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Gene Knockout Techniques , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Lipid Peroxidation/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , RNA, Small Interfering/genetics , Toluene/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
Cell Death Dis ; 10(11): 865, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31727879

ABSTRACT

Evolving concepts on Parkinson's disease (PD) pathology suggest that α-synuclein (aSYN) promote dopaminergic neuron dysfunction and death through accumulating in the mitochondria. However, the consequence of mitochondrial aSYN localisation on mitochondrial structure and bioenergetic functions in neuronal cells are poorly understood. Therefore, we investigated deleterious effects of mitochondria-targeted aSYN in differentiated human dopaminergic neurons in comparison with wild-type (WT) aSYN overexpression and corresponding EGFP (enhanced green fluorescent protein)-expressing controls. Mitochondria-targeted aSYN enhanced mitochondrial reactive oxygen species (ROS) formation, reduced ATP levels and showed severely disrupted structure and function of the dendritic neural network, preceding neuronal death. Transmission electron microscopy illustrated distorted cristae and many fragmented mitochondria in response to WT-aSYN overexpression, and a complete loss of cristae structure and massively swollen mitochondria in neurons expressing mitochondria-targeted aSYN. Further, the analysis of mitochondrial bioenergetics in differentiated dopaminergic neurons, expressing WT or mitochondria-targeted aSYN, elicited a pronounced impairment of mitochondrial respiration. In a pharmacological compound screening, we found that the pan-caspase inhibitors QVD and zVAD-FMK, and a specific caspase-1 inhibitor significantly prevented aSYN-induced cell death. In addition, the caspase inhibitor QVD preserved mitochondrial function and neuronal network activity in the human dopaminergic neurons overexpressing aSYN. Overall, our findings indicated therapeutic effects by caspase-1 inhibition despite aSYN-mediated alterations in mitochondrial morphology and function.


Subject(s)
Dopaminergic Neurons/metabolism , Parkinson Disease/genetics , Serpins/pharmacology , Viral Proteins/pharmacology , alpha-Synuclein/genetics , Adenosine Triphosphate/genetics , Caspase 1/genetics , Cell Death/genetics , Dopaminergic Neurons/pathology , Gene Expression Regulation , Humans , Mitochondria/genetics , Mitochondria/metabolism , Oxygen Consumption/genetics , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Reactive Oxygen Species/metabolism
3.
Redox Biol ; 12: 558-570, 2017 08.
Article in English | MEDLINE | ID: mdl-28384611

ABSTRACT

Ferroptosis has been defined as an oxidative and iron-dependent pathway of regulated cell death that is distinct from caspase-dependent apoptosis and established pathways of death receptor-mediated regulated necrosis. While emerging evidence linked features of ferroptosis induced e.g. by erastin-mediated inhibition of the Xc- system or inhibition of glutathione peroxidase 4 (Gpx4) to an increasing number of oxidative cell death paradigms in cancer cells, neurons or kidney cells, the biochemical pathways of oxidative cell death remained largely unclear. In particular, the role of mitochondrial damage in paradigms of ferroptosis needs further investigation. In the present study, we find that erastin-induced ferroptosis in neuronal cells was accompanied by BID transactivation to mitochondria, loss of mitochondrial membrane potential, enhanced mitochondrial fragmentation and reduced ATP levels. These hallmarks of mitochondrial demise are also established features of oxytosis, a paradigm of cell death induced by Xc- inhibition by millimolar concentrations of glutamate. Bid knockout using CRISPR/Cas9 approaches preserved mitochondrial integrity and function, and mediated neuroprotective effects against both, ferroptosis and oxytosis. Furthermore, the BID-inhibitor BI-6c9 inhibited erastin-induced ferroptosis, and, in turn, the ferroptosis inhibitors ferrostatin-1 and liproxstatin-1 prevented mitochondrial dysfunction and cell death in the paradigm of oxytosis. These findings show that mitochondrial transactivation of BID links ferroptosis to mitochondrial damage as the final execution step in this paradigm of oxidative cell death.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Mitochondria/physiology , Neurons/cytology , Animals , CRISPR-Cas Systems , Cell Death , Cell Line , Gene Knockout Techniques , Lipid Peroxidation , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Neurons/physiology , Oxidative Stress , Piperazines/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction
4.
Sci Transl Med ; 8(328): 328ra29, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26936506

ABSTRACT

Disability or death due to intracerebral hemorrhage (ICH) is attributed to blood lysis, liberation of iron, and consequent oxidative stress. Iron chelators bind to free iron and prevent neuronal death induced by oxidative stress and disability due to ICH, but the mechanisms for this effect remain unclear. We show that the hypoxia-inducible factor prolyl hydroxylase domain (HIF-PHD) family of iron-dependent, oxygen-sensing enzymes are effectors of iron chelation. Molecular reduction of the three HIF-PHD enzyme isoforms in the mouse striatum improved functional recovery after ICH. A low-molecular-weight hydroxyquinoline inhibitor of the HIF-PHD enzymes, adaptaquin, reduced neuronal death and behavioral deficits after ICH in several rodent models without affecting total iron or zinc distribution in the brain. Unexpectedly, protection from oxidative death in vitro or from ICH in vivo by adaptaquin was associated with suppression of activity of the prodeath factor ATF4 rather than activation of an HIF-dependent prosurvival pathway. Together, these findings demonstrate that brain-specific inactivation of the HIF-PHD metalloenzymes with the blood-brain barrier-permeable inhibitor adaptaquin can improve functional outcomes after ICH in several rodent models.


Subject(s)
Activating Transcription Factor 4/metabolism , Brain/pathology , Intracranial Hemorrhages/pathology , Molecular Targeted Therapy , Neurons/pathology , Oxygen/metabolism , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Animals , Cell Death/drug effects , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/drug effects , Genes, Reporter , Hemin/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracranial Hemorrhages/physiopathology , Iron/pharmacology , Iron Chelating Agents/pharmacology , Mice , Neurons/drug effects , Neuroprotective Agents/pharmacology , Procollagen-Proline Dioxygenase/metabolism , Protein Domains , Protein Isoforms/metabolism , Rats , Recovery of Function/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL