Your browser doesn't support javascript.
loading
The hepatocyte-specific HNF4α/miR-122 pathway contributes to iron overload-mediated hepatic inflammation.
Li, Min; Tang, Yuxiao; Wu, Lusha; Mo, Fengfeng; Wang, Xin; Li, Hongxia; Qi, Ruirui; Zhang, Hongwei; Srivastava, Arun; Ling, Chen.
Afiliação
  • Li M; Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China.
  • Tang Y; Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China.
  • Wu L; Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China.
  • Mo F; Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China.
  • Wang X; Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China.
  • Li H; Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China.
  • Qi R; Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China.
  • Zhang H; Military Hygiene Department, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China.
  • Srivastava A; Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL.
  • Ling C; State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China; and.
Blood ; 130(8): 1041-1051, 2017 08 24.
Article em En | MEDLINE | ID: mdl-28655781
ABSTRACT
Hepatic iron overload (IO) is a major complication of transfusional therapy. It was generally thought that IO triggers substantial inflammatory responses by producing reactive oxygen species in hepatic macrophages. Recently, a decrease in microRNA-122 (miR-122) expression was observed in a genetic knockout (Hfe-/-) mouse model of IO. Because hepatocyte-enriched miR-122 is a key regulator of multiple hepatic pathways, including inflammation, it is of interest whether hepatocyte directly contributes to IO-mediated hepatic inflammation. Here, we report that IO induced similar inflammatory responses in human primary hepatocytes and Thp-1-derived macrophages. In the mouse liver, IO resulted in altered expression of not only inflammatory genes but also >230 genes that are known targets of miR-122. In addition, both iron-dextran injection and a 3% carbonyl iron-containing diet led to upregulation of hepatic inflammation, which was associated with a significant reduction in HNF4α expression and its downstream target, miR-122. Interestingly, the same signaling pathway was changed in macrophage-deficient mice, suggesting that macrophages are not the only target of IO. Most importantly, hepatocyte-specific overexpression of miR-122 rescued IO-mediated hepatic inflammation. Our findings indicate the direct involvement of hepatocytes in IO-induced hepatic inflammation and are informative for developing new molecular targets and preventative therapies for patients with major hemoglobinopathy.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Hepatócitos / MicroRNAs / Fator 4 Nuclear de Hepatócito / Inflamação / Ferro / Fígado Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2017 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Hepatócitos / MicroRNAs / Fator 4 Nuclear de Hepatócito / Inflamação / Ferro / Fígado Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2017 Tipo de documento: Article