scholarly journals Absence of Glia Maturation Factor Protects Dopaminergic Neurons and Improves Motor Behavior in Mouse Model of Parkinsonism

2015 ◽  
Vol 40 (5) ◽  
pp. 980-990 ◽  
Author(s):  
Mohammad Moshahid Khan ◽  
Smita Zaheer ◽  
Ramasamy Thangavel ◽  
Margi Patel ◽  
Duraisamy Kempuraj ◽  
...  
2019 ◽  
Vol 33 (S1) ◽  
Author(s):  
Sudhanshu P Raikwar ◽  
Mohammad E Ahmed ◽  
Iuliia Dubova ◽  
Shireen Mentor ◽  
Daniyal Saeed ◽  
...  

2018 ◽  
Vol 56 (6) ◽  
pp. 3865-3881 ◽  
Author(s):  
Govindhasamy Pushpavathi Selvakumar ◽  
Shankar S Iyer ◽  
Duraisamy Kempuraj ◽  
Mohammad Ejaz Ahmed ◽  
Ramasamy Thangavel ◽  
...  

2020 ◽  
Vol 57 (11) ◽  
pp. 4438-4450 ◽  
Author(s):  
Mohammad Ejaz Ahmed ◽  
Govindhasamy Pushpavathi Selvakumar ◽  
Duraisamy Kempuraj ◽  
Sudhanshu P. Raikwar ◽  
Ramasamy Thangavel ◽  
...  

2020 ◽  
Vol 83 ◽  
pp. 106441 ◽  
Author(s):  
Hayate Javed ◽  
Ramasamy Thangavel ◽  
Govindhasamy Pushpavathi Selvakumar ◽  
Iuliia Dubova ◽  
Noah Schwartz ◽  
...  

1983 ◽  
Vol 5 (3) ◽  
pp. 261-269 ◽  
Author(s):  
A. Fontana ◽  
E. Weber ◽  
P.J. Grob ◽  
R. Lim ◽  
J.F. Miller

2019 ◽  
Vol 3 (8) ◽  
pp. 1211-1225 ◽  
Author(s):  
Wulin Aerbajinai ◽  
Manik C. Ghosh ◽  
Jie Liu ◽  
Chutima Kumkhaek ◽  
Jianqing Zhu ◽  
...  

Abstract In macrophages, cellular iron metabolism status is tightly integrated with macrophage phenotype and associated with mitochondrial function. However, how molecular events regulate mitochondrial activity to integrate regulation of iron metabolism and macrophage phenotype remains unclear. Here, we explored the important role of the actin-regulatory protein glia maturation factor-γ (GMFG) in the regulation of cellular iron metabolism and macrophage phenotype. We found that GMFG was downregulated in murine macrophages by exposure to iron and hydrogen peroxide. GMFG knockdown altered the expression of iron metabolism proteins and increased iron levels in murine macrophages and concomitantly promoted their polarization toward an anti-inflammatory M2 phenotype. GMFG-knockdown macrophages exhibited moderately increased levels of mitochondrial reactive oxygen species (mtROS), which were accompanied by decreased expression of some mitochondrial respiration chain components, including the iron-sulfur cluster assembly scaffold protein ISCU as well as the antioxidant enzymes SOD1 and SOD2. Importantly, treatment of GMFG-knockdown macrophages with the antioxidant N-acetylcysteine reversed the altered expression of iron metabolism proteins and significantly inhibited the enhanced gene expression of M2 macrophage markers, suggesting that mtROS is mechanistically linked to cellular iron metabolism and macrophage phenotype. Finally, GMFG interacted with the mitochondrial membrane ATPase ATAD3A, suggesting that GMFG knockdown–induced mtROS production might be attributed to alteration of mitochondrial function in macrophages. Our findings suggest that GMFG is an important regulator in cellular iron metabolism and macrophage phenotype and could be a novel therapeutic target for modulating macrophage function in immune and metabolic disorders.


1990 ◽  
Vol 1 (10) ◽  
pp. 741-746 ◽  
Author(s):  
R Lim ◽  
W X Zhong ◽  
A Zaheer

Recombinant human glia maturation factor beta (GMF-beta) reversibly inhibits the proliferation of neoplastic cells in culture by arresting the cells in the G0/G1 phase. This phenomenon is not target-cell specific, as neural and nonneural cells are equally inhibited. When tested simultaneously, GMF-beta suppresses the mitogenic effect of acidic fibroblasts growth factor (aFGF), but the two are synergistic in promoting the morphologic differentiation of cultured astrocytes. GMF-beta also counteracts the growth-stimulating effect of pituitary extract and cholera toxin on Schwann cells. The results underscore the regulatory role of GMF-beta and its intricate interaction with the mitogenic growth factors.


Sign in / Sign up

Export Citation Format

Share Document