Your browser doesn't support javascript.
loading
Gut Hormone GIP Induces Inflammation and Insulin Resistance in the Hypothalamus.
Fu, Yukiko; Kaneko, Kentaro; Lin, Hsiao-Yun; Mo, Qianxing; Xu, Yong; Suganami, Takayoshi; Ravn, Peter; Fukuda, Makoto.
Afiliação
  • Fu Y; Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
  • Kaneko K; Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.
  • Lin HY; Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
  • Mo Q; Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
  • Xu Y; Dan L Duncan Cancer Center and Center for Cell Gene & Therapy, Baylor College of Medicine, Houston, Texas.
  • Suganami T; Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
  • Ravn P; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
  • Fukuda M; Department of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.
Endocrinology ; 161(9)2020 09 01.
Article em En | MEDLINE | ID: mdl-32603429
ABSTRACT
The hypothalamus plays a critical role in controlling energy balance. High-fat diet (HFD) feeding increases the gene expression of proinflammatory mediators and decreases insulin actions in the hypothalamus. Here, we show that a gut-derived hormone, glucose-dependent insulinotropic polypeptide (GIP), whose levels are elevated during diet-induced obesity, promotes and mediates hypothalamic inflammation and insulin resistance during HFD-induced obesity. Unbiased ribonucleic acid sequencing of GIP-stimulated hypothalami revealed that hypothalamic pathways most affected by intracerebroventricular (ICV) GIP stimulation were related to inflammatory-related responses. Subsequent analysis demonstrated that GIP administered either peripherally or centrally, increased proinflammatory-related factors such as Il-6 and Socs3 in the hypothalamus, but not in the cortex of C57BL/6J male mice. Consistently, hypothalamic activation of IκB kinase-ß inflammatory signaling was induced by ICV GIP. Further, hypothalamic levels of proinflammatory cytokines and Socs3 were significantly reduced by an antagonistic GIP receptor (GIPR) antibody and by GIPR deficiency. Additionally, centrally administered GIP reduced anorectic actions of insulin in the brain and diminished insulin-induced phosphorylation of Protein kinase B and Glycogen synthase kinase 3ß in the hypothalamus. Collectively, these findings reveal a previously unrecognized role for brain GIP signaling in diet-induced inflammation and insulin resistance in the hypothalamus.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Receptores dos Hormônios Gastrointestinais / Resistência à Insulina / Polipeptídeo Inibidor Gástrico / Encefalite / Hipotálamo / Inflamação Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Receptores dos Hormônios Gastrointestinais / Resistência à Insulina / Polipeptídeo Inibidor Gástrico / Encefalite / Hipotálamo / Inflamação Idioma: En Ano de publicação: 2020 Tipo de documento: Article