Chronic morphine application is protective against cell death in primary human neurons

Neuroreport ◽  
2008 ◽  
Vol 19 (18) ◽  
pp. 1745-1749 ◽  
Author(s):  
Jia Cui ◽  
Qiuyue Chen ◽  
Long-Chuan Yu ◽  
Yan Zhang
PLoS ONE ◽  
2011 ◽  
Vol 6 (11) ◽  
pp. e27621 ◽  
Author(s):  
Roman Fischer ◽  
Olaf Maier ◽  
Martin Siegemund ◽  
Harald Wajant ◽  
Peter Scheurich ◽  
...  

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Jeremy W. Linsley ◽  
Kevan Shah ◽  
Nicholas Castello ◽  
Michelle Chan ◽  
Dominik Haddad ◽  
...  

AbstractCell death is a critical process that occurs normally in health and disease. However, its study is limited due to available technologies that only detect very late stages in the process or specific death mechanisms. Here, we report the development of a family of fluorescent biosensors called genetically encoded death indicators (GEDIs). GEDIs specifically detect an intracellular Ca2+ level that cells achieve early in the cell death process and that marks a stage at which cells are irreversibly committed to die. The time-resolved nature of a GEDI delineates a binary demarcation of cell life and death in real time, reformulating the definition of cell death. We demonstrate that GEDIs acutely and accurately report death of rodent and human neurons in vitro, and show that GEDIs enable an automated imaging platform for single cell detection of neuronal death in vivo in zebrafish larvae. With a quantitative pseudo-ratiometric signal, GEDIs facilitate high-throughput analysis of cell death in time-lapse imaging analysis, providing the necessary resolution and scale to identify early factors leading to cell death in studies of neurodegeneration.


2020 ◽  
Vol 27 (10) ◽  
pp. 2781-2796 ◽  
Author(s):  
Plamena R. Angelova ◽  
Minee L. Choi ◽  
Alexey V. Berezhnov ◽  
Mathew H. Horrocks ◽  
Craig D. Hughes ◽  
...  

Abstract Protein aggregation and abnormal lipid homeostasis are both implicated in neurodegeneration through unknown mechanisms. Here we demonstrate that aggregate-membrane interaction is critical to induce a form of cell death called ferroptosis. Importantly, the aggregate-membrane interaction that drives ferroptosis depends both on the conformational structure of the aggregate, as well as the oxidation state of the lipid membrane. We generated human stem cell-derived models of synucleinopathy, characterized by the intracellular formation of α-synuclein aggregates that bind to membranes. In human iPSC-derived neurons with SNCA triplication, physiological concentrations of glutamate and dopamine induce abnormal calcium signaling owing to the incorporation of excess α-synuclein oligomers into membranes, leading to altered membrane conductance and abnormal calcium influx. α-synuclein oligomers further induce lipid peroxidation. Targeted inhibition of lipid peroxidation prevents the aggregate-membrane interaction, abolishes aberrant calcium fluxes, and restores physiological calcium signaling. Inhibition of lipid peroxidation, and reduction of iron-dependent accumulation of free radicals, further prevents oligomer-induced toxicity in human neurons. In summary, we report that peroxidation of polyunsaturated fatty acids underlies the incorporation of β-sheet-rich aggregates into the membranes, and that additionally induces neuronal death. This suggests a role for ferroptosis in Parkinson’s disease, and highlights a new mechanism by which lipid peroxidation causes cell death.


2019 ◽  
Vol 15 ◽  
pp. P1006-P1006
Author(s):  
Panos Theofilas ◽  
Mifrah Hayath ◽  
Chao Wang ◽  
David Butler ◽  
Salome McAllen ◽  
...  

2002 ◽  
Vol 156 (3) ◽  
pp. 519-529 ◽  
Author(s):  
Yan Zhang ◽  
Richard McLaughlin ◽  
Cynthia Goodyer ◽  
Andréa LeBlanc

Extracellular amyloid β peptides (Aβs) have long been thought to be a primary cause of Alzheimer's disease (AD). Now, detection of intracellular neuronal Aβ1–42 accumulation before extracellular Aβ deposits questions the relevance of intracellular peptides in AD. In the present study, we directly address whether intracellular Aβ is toxic to human neurons. Microinjections of Aβ1–42 peptide or a cDNA-expressing cytosolic Aβ1–42 rapidly induces cell death of primary human neurons. In contrast, Aβ1–40, Aβ40–1, or Aβ42–1 peptides, and cDNAs expressing cytosolic Aβ1–40 or secreted Aβ1–42 and Aβ1–40, are not toxic. As little as a 1-pM concentration or 1500 molecules/cell of Aβ1–42 peptides is neurotoxic. The nonfibrillized and fibrillized Aβ1–42 peptides are equally toxic. In contrast, Aβ1–42 peptides are not toxic to human primary astrocytes, neuronal, and nonneuronal cell lines. Inhibition of de novo protein synthesis protects against Aβ1–42 toxicity, indicating that programmed cell death is involved. Bcl-2, Bax-neutralizing antibodies, cDNA expression of a p53R273H dominant negative mutant, and caspase inhibitors prevent Aβ1–42-mediated human neuronal cell death. Taken together, our data directly demonstrate that intracellular Aβ1–42 is selectively cytotoxic to human neurons through the p53–Bax cell death pathway.


2019 ◽  
Author(s):  
Jeremy W. Linsley ◽  
Kevan Shah ◽  
Nicholas Castello ◽  
Michelle Chan ◽  
Dominic Haddad ◽  
...  

AbstractCell death is a critical process that occurs normally in health and disease. However, its study is limited due to available technologies that only detect very late stages in the process or specific death mechanisms. Here, we report the development of a new fluorescent biosensor called genetically encoded death indicator (GEDI). GEDI specifically detects an intracellular Ca2+ level that cells achieve early in the cell death process and marks a stage at which cells are irreversibly committed to die. The time-resolved nature of GEDI delineates a binary demarcation of cell life and death in real time, reformulating the definition of cell death. We demonstrate that GEDI acutely and accurately reports death of rodent and human neurons in vitro, and show GEDI enables a novel automated imaging platform for single cell detection of neuronal death in vivo in zebrafish larvae. With a quantitative pseudo-ratiometric signal, GEDI facilitates high-throughput analysis of cell death in time lapse imaging analysis, providing the necessary resolution and scale to identify early factors leading to cell death in studies of neurodegeneration.


Author(s):  
N. Braidy ◽  
S. Subash ◽  
M.M. Essa ◽  
R. Vaishnav ◽  
S. Al-Adawi ◽  
...  

Background: Quinolinic acid (QUIN) excitotoxicity is mediated by elevated intracellular Ca2+ levels, and nitric oxide (NO•) mediated oxidative stress leading to DNA damage, and cell death due to energy restriction. Methods: We evaluated the effect of a series of pomegranate juice extracts (PJE), Helow, Malasi, Qusum, and Hamedh, with antioxidant properties on QUIN induced excitotoxicity on primary cultures of human neurons. Results: We showed that Helow and Malasi can attenuate QUIN-induced excitotoxicity to a greater extent than Qusum and Hamedh from Oman. Similarly, both Helow and Malasi were able to attenuate QUIN-induced Ca2+ influx and nNOS activity to a greater extent compared to Qusum, and Hamedh. All extracts reduced the oxidative effects of increased NO• production, and hence preventing NAD+ depletion and cell death. Conclusion: In addition to the well-known antioxidant properties of these natural phytochemicals, the inhibitory effect of some of these compounds on specific excitotoxic processes such as calcium influx provides additional evidence for the beneficial health effects of PJE in excitable tissue, particularly within the CNS.


2019 ◽  
Vol 54 (8) ◽  
pp. 453-463 ◽  
Author(s):  
Danielle V. Bower ◽  
Johannes K. Richter ◽  
Hendrik von Tengg-Kobligk ◽  
Johannes T. Heverhagen ◽  
Val M. Runge

Sign in / Sign up

Export Citation Format

Share Document