Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Neuroscience ; 545: 158-170, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38513765

ABSTRACT

Thioredoxin-reductase 2 (Txnrd2) belongs to the thioredoxin-reductase family of selenoproteins and is a key antioxidant enzyme in mammalian cells to regulate redox homeostasis. Here, we reported that Txnrd2 exerted a major influence in brain damage caused by Intracerebral hemorrhage (ICH) by suppressing endoplasmic reticulum (ER) stress oxidative stress and via Trx2/Prx3 pathway. Furthermore, we demonstrated that pharmacological selenium (Se) rescued the brain damage after ICH by enhancing Txnrd2 expression. Primarily, expression and localization of Txnrd2, Trx2 and Prx3 were determined in collagenase IV-induced ICH model. Txnrd2 was then knocked down using siRNA interference in rats which were found to develop more severe encephaledema and neurological deficits. Mechanistically, we observed that loss of Txnrd2 leads to increased lipid peroxidation levels and ER stress protein expression in neurons and astrocytes. Additionally, it was revealed that Se effectively restored the expression of Txnrd2 in brain and inhibited both the activity of ER stress protein activity and the generation of reactive oxygen species (ROS) by promoting Trx2/Prx3 kilter when administrating sodium selenite in lateral ventricle. This study shed light on the effect of Txnrd2 in regulating oxidative stress and ER stress via Trx2/Prx3 pathway upon ICH and its promising potential as an ICH therapeutic target.


Subject(s)
Cerebral Hemorrhage , Endoplasmic Reticulum Stress , Oxidative Stress , Rats, Sprague-Dawley , Thioredoxin Reductase 2 , Thioredoxins , Animals , Male , Rats , Astrocytes/metabolism , Astrocytes/drug effects , Brain/metabolism , Brain/drug effects , Brain/pathology , Brain Injuries/metabolism , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Disease Models, Animal , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum Stress/drug effects , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Peroxiredoxin III/metabolism , Reactive Oxygen Species/metabolism , Selenium/pharmacology , Signal Transduction/physiology , Signal Transduction/drug effects , Thioredoxin Reductase 2/metabolism , Thioredoxins/metabolism
2.
Free Radic Biol Med ; 199: 67-80, 2023 04.
Article in English | MEDLINE | ID: mdl-36805044

ABSTRACT

Intracerebral hemorrhage (ICH) induces high mortality and disability. Neuronal death is the principal factor to unfavourable prognosis in ICH. However, the mechanisms underlying this association remain unclear. In this study, we investigated the molecular mechanisms by which neuronal ferroptosis occurs after ICH and whether the use of corresponding modulators can inhibit neuronal death and improve early outcomes in a rat ICH model. Our findings indicated that Nox4 and TF/TfR were upregulated in the perihematomal tissues of ICH rats. Oxidative stress and iron overload induced by Nox4 and TF/TfR promoted neuronal ferroptosis post-ICH. In contrast, application of Nox4-siRNA and the deferoxamine (DFO) attenuated peroxidation and iron deposition in the hemorrhagic brain, alleviated neuronal ferroptosis, and improved sensorimotor function in ICH rats. Additionally, our findings indicated that the post-ICH neuronal reduced glutathione (GSH) depletion were not related to dysfunctional glutamine delivery in astrocytes but rather to downregulation of EAAT3 due to lipid peroxidation-induced dysfunction in the neuronal membrane. These findings indicate that ferroptosis is involved in neuronal death in model rats with collagenase-induced ICH. Oxidative stress and iron overload induced by Nox4 and TF/TfR exacerbate ferroptosis after ICH, while Nox4 downregulation and iron chelation exert neuroprotective effects. The present results highlight the cysteine importer EAAT3 as a potential biomarker of ferroptosis and provide insight into the neuronal death process that occurs following ICH, which may aid in the development of translational treatment strategies for ICH.


Subject(s)
Ferroptosis , Iron Overload , Animals , Rats , Cell Death , Cerebral Hemorrhage/genetics , Ferroptosis/genetics , Iron Overload/genetics , NADPH Oxidase 4/genetics , Oxidative Stress/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...