scholarly journals Fetuin-A Alleviates Neuroinflammation Against Traumatic Brain Injury-Induced Microglial Necroptosis By Regulating Nrf-2/HO-1 Pathway

Author(s):  
Pengzhan Zhao ◽  
Zhongyuan Bao ◽  
Guangchi Sun ◽  
Honglu Chao ◽  
Yiming Tu ◽  
...  

Abstract Background: The microglia-mediated inflammatory response is a vital mechanism of secondary damage following traumatic brain injury (TBI), but its underlying mechanism of microglial activation is unclear. Methods: Controlled cortical impact (CCI) was induced in adult male C57BL/6J mice, and we also used glutamate to construct a classical in vitro injury model in BV2 cell line. The activation of microglia was determined by western blot assessments and immunostaining. The inflammatory factors were determined by ELLSA. The oxidative stress marker and mitochondrial ROS were determined by immunoblotting and MitoSox Red staining. Transmission electron microscopy (TEM) was used to observe a typical morphology of necroptotic cells. Results: Our quantitative proteomics identified 2499 proteins, 157 were significantly differentially expressed between brain tissues at 6 hours after CCI (CCI6h) and sham groups, and 109 were significantly differentially expressed between CCI24h and sham brain tissues. Moreover, compared with sham groups, the terms “acute-phase response”, “inflammation”, and “protein binding” were significantly enriched in CCI groups. Interestingly, fetuin-A, a liver-secreted acute-phase glycoprotein, was involved in these biological processes. Using experimental TBI models, we found that the fetuin-A level peaked at 6 h and then decreased gradually. Importantly, we showed that fetuin-A reduced the cortical lesion volume and edema area and inhibited the inflammatory response, which was associated with suppressing microglial necroptosis, thus decreasing microglial polarization to the M1 phenotype. Furthermore, administration of fetuin-A attenuated mitochondrial oxidative stress in glutamate-treated BV2 cells, which is a critical mechanism of necroptosis suppression. In addition, we demonstrated that fetuin-A treatment promoted translocation of nuclear factor erythroid 2-related factor 2 (Nrf-2) from the cytoplasm to the nucleus in vivo; however, the Nrf-2 inhibitor ML385 and si-heme oxygenase-1 (HO-1) disrupted the regulation of oxidative stress by fetuin-A and induced increased ROS levels and necroptosis in glutamate-treated BV2 cells. Interestingly, the mechanism of fetuin-A in BV2 cells also protects neurons from adverse factors in co-culture assays.Conclusions: Our results demonstrate that fetuin-A activates Nrf-2/HO-1, suppresses oxidative stress and necroptosis levels, and thereby attenuates the abnormal inflammatory response following TBI, providing a potential therapeutic strategy for TBI treatment.

2014 ◽  
Vol 21 (10) ◽  
pp. 1201-1211 ◽  
Author(s):  
Ana Rodriguez-Rodriguez ◽  
Juan Egea-Guerrero ◽  
Francisco Murillo-Cabezas ◽  
Antonio Carrillo-Vico

2018 ◽  
Vol 17 (9) ◽  
pp. 689-695 ◽  
Author(s):  
Nidhi Khatri ◽  
Manisha Thakur ◽  
Vikas Pareek ◽  
Sandeep Kumar ◽  
Sunil Sharma ◽  
...  

Background & Objective: Traumatic Brain Injury (TBI) is one of the major causes of mortality and morbidity worldwide. It represents mild, moderate and severe effects of physical assault to brain which may cause sequential, primary or secondary ramifications. Primary injury can be due to the first physical hit, blow or jolt to one of the brain compartments. The primary injury is then followed by secondary injury which leads to biochemical, cellular, and physiological changes like blood brain barrier disruption, inflammation, excitotoxicity, necrosis, apoptosis, mitochondrial dysfunction and generation of oxidative stress. Apart from this, there is also an immediate increase in glutamate at the synapses following severe TBI. Excessive glutamate at synapses in turn activates corresponding NMDA and AMPA receptors that facilitate excessive calcium influx into the neuronal cells. This leads to the generation of oxidative stress which further leads to mitochondrial dysfunction, lipid peroxidation and oxidation of proteins and DNA. As a consequence, neuronal cell death takes place and ultimately people start facing some serious disabilies. Conclusion: In the present review we provide extensive overview of the role of reactive oxygen species (ROS)-induced oxidative stress and its fatal effects on brain after TBI.


Critical Care ◽  
2021 ◽  
Vol 25 (1) ◽  
Author(s):  
Daniel Agustin Godoy ◽  
Rafael Badenes ◽  
Paolo Pelosi ◽  
Chiara Robba

AbstractMaintaining an adequate level of sedation and analgesia plays a key role in the management of traumatic brain injury (TBI). To date, it is unclear which drug or combination of drugs is most effective in achieving these goals. Ketamine is an agent with attractive pharmacological and pharmacokinetics characteristics. Current evidence shows that ketamine does not increase and may instead decrease intracranial pressure, and its safety profile makes it a reliable tool in the prehospital environment. In this point of view, we discuss different aspects of the use of ketamine in the acute phase of TBI, with its potential benefits and pitfalls.


Biomedicines ◽  
2020 ◽  
Vol 8 (9) ◽  
pp. 361
Author(s):  
Margaux Sambon ◽  
Anna Gorlova ◽  
Alice Demelenne ◽  
Judit Alhama-Riba ◽  
Bernard Coumans ◽  
...  

Thiamine precursors, the most studied being benfotiamine (BFT), have protective effects in mouse models of neurodegenerative diseases. BFT decreased oxidative stress and inflammation, two major characteristics of neurodegenerative diseases, in a neuroblastoma cell line (Neuro2a) and an immortalized brain microglial cell line (BV2). Here, we tested the potential antioxidant and anti-inflammatory effects of the hitherto unexplored derivative O,S-dibenzoylthiamine (DBT) in these two cell lines. We show that DBT protects Neuro2a cells against paraquat (PQ) toxicity by counteracting oxidative stress at low concentrations and increases the synthesis of reduced glutathione and NADPH in a Nrf2-independent manner. In BV2 cells activated by lipopolysaccharides (LPS), DBT significantly decreased inflammation by suppressing translocation of NF-κB to the nucleus. Our results also demonstrate the superiority of DBT over thiamine and other thiamine precursors, including BFT, in all of the in vitro models. Finally, we show that the chronic administration of DBT arrested motor dysfunction in FUS transgenic mice, a model of amyotrophic lateral sclerosis, and it reduced depressive-like behavior in a mouse model of ultrasound-induced stress in which it normalized oxidative stress marker levels in the brain. Together, our data suggest that DBT may have therapeutic potential for brain pathology associated with oxidative stress and inflammation by novel, coenzyme-independent mechanisms.


2009 ◽  
Vol 1291 ◽  
pp. 122-132 ◽  
Author(s):  
Shadi Homsi ◽  
Fabiola Federico ◽  
Nicole Croci ◽  
Bruno Palmier ◽  
Michel Plotkine ◽  
...  

1997 ◽  
Vol 47 (6) ◽  
pp. 575-581 ◽  
Author(s):  
Deepak Awasthi ◽  
Daniel F. Church ◽  
Dan Torbati ◽  
Michael E. Carey ◽  
William A. Pryor

Sign in / Sign up

Export Citation Format

Share Document