Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 35(5): e21569, 2021 05.
Article in English | MEDLINE | ID: mdl-33864420

ABSTRACT

Aberrant accumulation of amyloid-ß (Aß) in brain is the major trigger for pathogenesis in Alzheimer's disease (AD). It is imperative to understand how Aß attains such toxic levels in the brain parenchyma. We detected that a subtle and tolerable amount of DNA damage, related to aging, increased intraneuronal Aß1-42 production both in cultured neuron and in cortex of rodent brain. Strikingly, we also observed elevated levels of mitochondrial fusion and of its major driver protein, MFN2. Hyperfusion of mitochondria may be seen as an adaptive stress response resulting from the induction of ER stress since we detected the activation of both PERK and IRE1α arms of unfolded protein response of ER stress. We found increased phosphorylation of PERK substrate eukaryotic initiation factor 2 α (eIF2α), and upregulation of the downstream effector proteins, ATF4 and CHOP. Concomitantly, increased XBP1 level, the direct effecter protein of IRE-1α, was observed. Reports suggest that eIF2α phosphorylation can increase BACE1 activity, the rate limiting enzyme in Aß production. Here, we show that inhibiting PERK, decreased Aß1-42 level while direct BACE1 inhibition, reduced the mitochondrial fusion. We found increased MFN2 expression in young 5xFAD mice when Aß plaques and neurodegeneration were absent. Thus, our study indicates that mild DNA damage leads to increased Aß1-42 production almost as a consequence of an initial ER stress-directed protective mitochondrial fusion in brain. We propose that an age-related subtle genomic DNA damage may trigger enhanced intraneuronal Aß1-42 production in an apparently healthy neuron way before the appearance of clinical symptoms in AD.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , DNA Damage , Neurons/metabolism , Peptide Fragments/metabolism , Amyloid Precursor Protein Secretases/genetics , Animals , Brain/pathology , Disease Models, Animal , Genomics , Humans , Male , Mice , Mice, Transgenic , Neurons/pathology , Phosphorylation , Rats , Rats, Sprague-Dawley
2.
Free Radic Res ; 54(7): 477-496, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32842814

ABSTRACT

Smokeless tobacco (SLT) or chewing tobacco has been a highly addictive practice in India across ages, posing major threat to the systemic health and possibly neurodegeneration. Earlier studies showed components of SLT could be harmful to neuronal health. However, mechanism of SLT in neurodegeneration remained unexplored. This study investigated the detrimental role of SLT on differentiated neuronal cell lines, PC12 and SH-SY5Y by using graded doses of water soluble lyophilised SLT. Reduced cell viability, compromised mitochondrial structure and functions were observed when neuronal cell lines were treated with SLT (6 mg/mL) for 24 h. There was reduction of oxidative phosphorylation and aerobic glycolysis as determined by diminution of ATP production (2.5X) and basal respiration (1.9X). Mitochondrial membrane potential was dropped by 3.5 times. Bid, a pro-apoptotic Bcl-2 family protein, has imperative role in regulating mitochondrial outer membrane permeabilization and subsequent cytochrome c release leading to apoptosis. This article for the first time indicated the involvement of Bid in SLT mediated neurotoxicity and possibly neurodegeneration. SLT treatment enhanced expression of cleaved-Bid in time dependent manner. The involvement of Bid was further confirmed by using Bid specific shRNA which reversed the effects of SLT and conferred significant protection from apoptosis up to 72 h. Thus, our results clearly indicated that SLT induced neuronal cell death occurred via production of ROS, alteration of mitochondrial morphology, membrane potential and oxidative phosphorylation, inactivation of survival pathway and activation of apoptotic markers mediated by Bid. Therefore, Bid could be a potential future therapeutic target for SLT induced neurodegeneration.


Subject(s)
Neurons/pathology , Tobacco, Smokeless/toxicity , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cytochromes c/metabolism , DNA Damage , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/pathology , Neurons/drug effects , Neurons/metabolism , Oxidative Phosphorylation , PC12 Cells , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Reactive Oxygen Species/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Tumor Suppressor Protein p53/metabolism
3.
FEBS J ; 287(11): 2386-2413, 2020 06.
Article in English | MEDLINE | ID: mdl-31747135

ABSTRACT

ß-amyloid (Aß) aggregates involved in Alzheimer's disease (AD) are resistant to proteases but could be destabilized by small peptides designed to target specific hydrophobic regions of Aß that take part in aggregate assembly. Since thrombin and AD are intricately connected, and elastase modulates thrombin activity, elastase-digested thrombin peptides were verified for intervention in the Aß-aggregation pathway. Intact or elastase-digested thrombin destabilized Aß fibril, as demonstrated by thioflavin T assay. Peptides were synthesized employing thrombin as a template, of which, a hexapeptide (T3) showed maximum destabilization at 1 µm. ExPASy peptide cutter software coupled with mass spectrometric analysis confirmed the generation of T3 peptide from elastase-digested thrombin. TEM micrographs revealed that 30-day incubation of preformed Aß fibrils or monomers with T3 resulted in destabilization or inhibition, respectively, leading mostly to particles of 1.74 ± 0.17 nm, which roughly corresponded to Aß monomer. Surface plasmon resonance employing CM5 chip coupled with Aß40 mouse monoclonal antibody showed a drop in response when T3 was incubated with Aß fibrils between 2 and 8 h. 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide and confocal microscopy demonstrated the ability of T3 to rescue neuroblastoma cells from Aß oligomer-induced cytotoxic damage. Although no [Aß-T3] adduct could be detected by mass spectrometry, an initial interaction appeared to facilitate the process of destabilization/inhibition of aggregation. T3 was comparable to standard ß-sheet breaker peptides, LPFFD and KLVFF in terms of Aß aggregate destabilization. High hydrophobicity values coupled with recognition and breaking elements make T3 a potential candidate for future therapeutic applications.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/genetics , Protein Aggregation, Pathological/drug therapy , Thrombin/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid/antagonists & inhibitors , Amyloid/genetics , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Benzothiazoles/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions/drug effects , Mice , Neuroprotection/drug effects , Peptide Hydrolases/genetics , Peptides/pharmacology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Thrombin/genetics
4.
J Neurochem ; 134(6): 1091-103, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25891762

ABSTRACT

Neuronal loss in selective areas of brain underlies the pathology of Alzheimer's disease (AD). Recent evidences place oligomeric ß-amyloid (Aß) central to the disease. However, mechanism of neuron death in response to Aß remains elusive. Activation of the c-Jun N-terminal kinase (JNK) pathway and induction of the AP-1 transcription factor c-Jun are reported in AD. However, targets of JNK/c-Jun in Aß-induced neuron death are mostly unknown. Our study shows that pro-apoptotic proteins, Bim (Bcl-2 interacting mediator of cell death) and Puma (p53 up-regulated modulator of apoptosis) are targets of c-Jun in Aß-treated neurons. We demonstrate that the JNK/c-Jun pathway is activated, in cultures of cortical neurons following treatment with oligomeric Aß and in AD transgenic mice, and that inhibition of this pathway by selective inhibitor blocks induction of Puma by Aß. We also find that both JNK and p53 pathways co-operatively regulate Puma expression in Aß-treated neurons. Moreover, we identified a novel AP1-binding site on rat puma gene which is necessary for direct binding of c-Jun with Puma promoter. Finally, we find that knocking down of c-Jun by siRNA provides significant protection from Aß toxicity and that induction of Bim and Puma by Aß in neurons requires c-Jun. Taken together, our results suggest that both Bim and Puma are target of c-Jun and elucidate the intricate regulation of Puma expression by JNK/c-Jun and p53 pathways in neurons upon Aß toxicity. JNK/c-Jun pathway is shown to be activated in neurons of the Alzheimer's disease (AD) brain and plays a vital role in neuron death in AD models. However, downstream targets of c-Jun in this disease have not been thoroughly elucidated. Our study shows that two important pro-apoptotic proteins, Bim (Bcl-2 interacting mediator of cell death) and Puma (p53 up-regulated modulator of apoptosis) are targets of c-Jun in Aß-treated neurons. We demonstrate that the JNK/c-jun pathway is activated, in cultures of cortical neurons following treatment with oligomeric Aß and in AD transgenic mice, and that inhibition of this pathway by selective inhibitor blocks induction of Puma by Aß. We have also observed functional co-operation of both JNK and p53 pathway in regulation of Puma under Aß toxicity. Most importantly, we identified a novel AP1-binding site on rat puma gene which is necessary for direct binding of c-Jun with Puma promoter. Thus, our results suggest that both Bim and Puma are target of c-Jun and elucidate the intricate regulation of Puma expression by JNK/c-Jun and p53 pathways in neurons upon Aß toxicity.


Subject(s)
Alzheimer Disease/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Gene Expression Regulation/physiology , MAP Kinase Signaling System/physiology , Neurons/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Blotting, Western , Chromatin Immunoprecipitation , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Neurons/pathology , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Transfection , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...