Your browser doesn't support javascript.
loading
Glia maturation factor-γ regulates murine macrophage iron metabolism and M2 polarization through mitochondrial ROS.
Aerbajinai, Wulin; Ghosh, Manik C; Liu, Jie; Kumkhaek, Chutima; Zhu, Jianqing; Chin, Kyung; Rouault, Tracey A; Rodgers, Griffin P.
Afiliación
  • Aerbajinai W; Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute.
  • Ghosh MC; Metals Biology and Molecular Medicine Group, Eunice Kennedy Shriver National Institute of Child Health and Human Development, and.
  • Liu J; Laboratory of Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD.
  • Kumkhaek C; Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute.
  • Zhu J; Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute.
  • Chin K; Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute.
  • Rouault TA; Metals Biology and Molecular Medicine Group, Eunice Kennedy Shriver National Institute of Child Health and Human Development, and.
  • Rodgers GP; Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute.
Blood Adv ; 3(8): 1211-1225, 2019 04 23.
Article en En | MEDLINE | ID: mdl-30971398
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.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Especies Reactivas de Oxígeno / Factor de Maduración de la Glia / Hierro / Macrófagos / Mitocondrias Límite: Animals Idioma: En Revista: Blood Adv Año: 2019 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Especies Reactivas de Oxígeno / Factor de Maduración de la Glia / Hierro / Macrófagos / Mitocondrias Límite: Animals Idioma: En Revista: Blood Adv Año: 2019 Tipo del documento: Article