Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Dis ; 11(5): 397, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32457286

ABSTRACT

Loss of protein folding homeostasis features many of the most prevalent neurodegenerative disorders. As coping mechanism to folding stress within the endoplasmic reticulum (ER), the unfolded protein response (UPR) comprises a set of signaling mechanisms that initiate a gene expression program to restore proteostasis, or when stress is chronic or overwhelming promote neuronal death. This fate-defining capacity of the UPR has been proposed to play a key role in amyotrophic lateral sclerosis (ALS). However, the several genetic or pharmacological attempts to explore the therapeutic potential of UPR modulation have produced conflicting observations. In order to establish the precise relationship between UPR signaling and neuronal death in ALS, we have developed a neuronal model where the toxicity of a familial ALS-causing allele (mutant G93A SOD1) and UPR activation can be longitudinally monitored in single neurons over the process of neurodegeneration by automated microscopy. Using fluorescent UPR reporters we established the temporal and causal relationship between UPR and neuronal death by Cox regression models. Pharmacological inhibition of discrete UPR processes allowed us to establish the contribution of PERK (PKR-like ER kinase) and IRE1 (inositol-requiring enzyme-1) mechanisms to neuronal fate. Importantly, inhibition of PERK signaling with its downstream inhibitor ISRIB, but not with the direct PERK kinase inhibitor GSK2606414, significantly enhanced the survival of G93A SOD1-expressing neurons. Characterization of the inhibitory properties of both drugs under ER stress revealed that in neurons (but not in glial cells) ISRIB overruled only part of the translational program imposed by PERK, relieving the general inhibition of translation, but maintaining the privileged translation of ATF4 (activating transcription factor 4) messenger RNA. Surprisingly, the fine-tuning of the PERK output in G93A SOD1-expressing neurons led to a reduction of IRE1-dependent signaling. Together, our findings identify ISRIB-mediated translational reprogramming as a new potential ALS therapy.


Subject(s)
Acetamides/pharmacology , Amyotrophic Lateral Sclerosis/pathology , Cyclohexylamines/pharmacology , Models, Biological , Neurons/pathology , Unfolded Protein Response , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/pathology , Endoplasmic Reticulum Stress/drug effects , HEK293 Cells , HeLa Cells , Humans , Indoles/pharmacology , Mice , Mutation/genetics , Neurons/drug effects , Neurons/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Superoxide Dismutase/genetics , Survival Analysis , Unfolded Protein Response/drug effects , eIF-2 Kinase/metabolism
2.
Neurobiol Dis ; 137: 104781, 2020 04.
Article in English | MEDLINE | ID: mdl-31991248

ABSTRACT

Alpha-synuclein (aSyn) protein levels are sufficient to drive Parkinson's disease (PD) and other synucleinopathies. Despite the biomedical/therapeutic potential of aSyn protein regulation, little is known about mechanisms that limit/control aSyn levels. Here, we investigate the role of a post-translational modification, N-terminal acetylation, in aSyn neurotoxicity. N-terminal acetylation occurs in all aSyn molecules and has been proposed to determine its lipid binding and aggregation capacities; however, its effect in aSyn stability/neurotoxicity has not been evaluated. We generated N-terminal mutants that alter or block physiological aSyn N-terminal acetylation in wild-type or pathological mutant E46K aSyn versions and confirmed N-terminal acetylation status by mass spectrometry. By optical pulse-labeling in living primary neurons we documented a reduced half-life and accumulation of aSyn N-terminal mutants. To analyze the effect of N-terminal acetylation mutants in neuronal toxicity we took advantage of a neuronal model where aSyn toxicity was scored by longitudinal survival analysis. Salient features of aSyn neurotoxicity were previously investigated with this approach. aSyn-dependent neuronal death was recapitulated either by higher aSyn protein levels in the case of WT aSyn, or by the combined effect of protein levels and enhanced neurotoxicity conveyed by the E46K mutation. aSyn N-terminal mutations decreased E46K aSyn-dependent neuronal death both by reducing protein levels and, importantly, by reducing the intrinsic E46K aSyn toxicity, being the D2P mutant the least toxic. Together, our results illustrate that the N-terminus determines, most likely through its acetylation, aSyn protein levels and toxicity, identifying this modification as a potential therapeutic target.


Subject(s)
Neurons/metabolism , Parkinson Disease/genetics , Protein Aggregation, Pathological/metabolism , alpha-Synuclein/metabolism , Acetylation , Cell Death/genetics , Humans , Mutation/genetics , Parkinson Disease/metabolism , Protein Aggregation, Pathological/genetics , Protein Processing, Post-Translational/genetics , Protein Stability
3.
Proc Natl Acad Sci U S A ; 114(39): E8274-E8283, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28900007

ABSTRACT

α-Synuclein (aSyn) is the main driver of neurodegenerative diseases known as "synucleinopathies," but the mechanisms underlying this toxicity remain poorly understood. To investigate aSyn toxic mechanisms, we have developed a primary neuronal model in which a longitudinal survival analysis can be performed by following the overexpression of fluorescently tagged WT or pathologically mutant aSyn constructs. Most aSyn mutations linked to neurodegenerative disease hindered neuronal survival in this model; of these mutations, the E46K mutation proved to be the most toxic. While E46K induced robust PLK2-dependent aSyn phosphorylation at serine 129, inhibiting this phosphorylation did not alleviate aSyn toxicity, strongly suggesting that this pathological hallmark of synucleinopathies is an epiphenomenon. Optical pulse-chase experiments with Dendra2-tagged aSyn versions indicated that the E46K mutation does not alter aSyn protein turnover. Moreover, since the mutation did not promote overt aSyn aggregation, we conclude that E46K toxicity was driven by soluble species. Finally, we developed an assay to assess whether neurons expressing E46K aSyn affect the survival of neighboring control neurons. Although we identified a minor non-cell-autonomous component spatially restricted to proximal neurons, most E46K aSyn toxicity was cell autonomous. Thus, we have been able to recapitulate the toxicity of soluble aSyn species at a stage preceding aggregation, detecting non-cell-autonomous toxicity and evaluating how some of the main aSyn hallmarks are related to neuronal survival.


Subject(s)
Mutation, Missense , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Protein Aggregation, Pathological/metabolism , alpha-Synuclein/metabolism , Amino Acid Substitution , Animals , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurons/pathology , Phosphorylation , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Rats , Rats, Sprague-Dawley , alpha-Synuclein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...