scholarly journals CRISPR/Cas9 Editing of Glia Maturation Factor Regulates Mitochondrial Dynamics by Attenuation of the NRF2/HO-1 Dependent Ferritin Activation in Glial Cells

2019 ◽  
Vol 14 (4) ◽  
pp. 537-550 ◽  
Author(s):  
Govindhasamy Pushpavathi Selvakumar ◽  
Mohammad Ejaz Ahmed ◽  
Sudhanshu P. Raikwar ◽  
Ramasamy Thangavel ◽  
Duraisamy Kempuraj ◽  
...  
2019 ◽  
Vol 56 (10) ◽  
pp. 6964-6975 ◽  
Author(s):  
Mohammad Ejaz Ahmed ◽  
Govindhasamy Pushpavathi Selvakumar ◽  
Duraisamy Kempuraj ◽  
Ramasamy Thangavel ◽  
Shireen Mentor ◽  
...  

1981 ◽  
Vol 6 (4) ◽  
pp. 401-412 ◽  
Author(s):  
Taiji Kato ◽  
Yosuke Yamakawa ◽  
Ramon Lim ◽  
David E. Turriff ◽  
Ryo Tanaka

2017 ◽  
Vol 45 (6) ◽  
pp. 1670-1679 ◽  
Author(s):  
Gungor Cagdas DINCEL

Objective We previously showed that Toxoplasma gondii infection induces severe neuropathology in the form of oxidative stress, high nitric oxide production, glial activation, and apoptosis. This study examined the association between glia maturation factor-beta (GMF-β) expression, activated astrocytes/microglia, and neuropathology in toxoplasmic encephalitis (TE). Methods Mouse brain GMF expression was examined by immunohistochemistry on days 10 and 30 post- T. gondii infection. Results Neuropathology of infected mice was associated with increased GMF expression in reactive glial cells and neurons compared with healthy controls. Specific up-regulation of GMF-β expression in glial cells was associated with increased gliosis in TE. Conclusions GMF up-regulation in glial cells causes neuronal destruction, suggesting a TE pathological pathway involving GMF-mediated brain cell cytotoxicity. GMF-β may therefore be a good biomarker for disease risk assessment and to estimate host neuropathy after exposure to T. gondii, as well as providing a new therapeutic target. This is the first study to demonstrate the expression of GMF-β in reactive glial cells and its association with neuropathology in TE.


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.


Biochemistry ◽  
1985 ◽  
Vol 24 (27) ◽  
pp. 8070-8074 ◽  
Author(s):  
Ramon Lim ◽  
Joyce F. Miller ◽  
Danny J. Hicklin ◽  
Andrew A. Andresen

1990 ◽  
Vol 17 (4) ◽  
pp. 559-571 ◽  
Author(s):  
Kuniko Okumura-Noji ◽  
Taiji Kato ◽  
Jin-ichi Ito ◽  
Tatsuo Suzuki ◽  
Ryo Tanaka

Sign in / Sign up

Export Citation Format

Share Document