Your browser doesn't support javascript.
loading
Insulin regulates POMC neuronal plasticity to control glucose metabolism.
Dodd, Garron T; Michael, Natalie J; Lee-Young, Robert S; Mangiafico, Salvatore P; Pryor, Jack T; Munder, Astrid C; Simonds, Stephanie E; Brüning, Jens Claus; Zhang, Zhong-Yin; Cowley, Michael A; Andrikopoulos, Sofianos; Horvath, Tamas L; Spanswick, David; Tiganis, Tony.
Afiliação
  • Dodd GT; Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.
  • Michael NJ; Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia.
  • Lee-Young RS; Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.
  • Mangiafico SP; Department of Physiology, Monash University, Victoria, Australia.
  • Pryor JT; Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.
  • Munder AC; Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia.
  • Simonds SE; Monash Metabolic Phenotyping Facility, Monash University, Victoria, Australia.
  • Brüning JC; Department of Medicine (Austin Hospital), The University of Melbourne, Melbourne, Australia.
  • Zhang ZY; Department of Physiology, Monash University, Victoria, Australia.
  • Cowley MA; Warwick Medical School, University of Warwick, Coventry, United Kingdom.
  • Andrikopoulos S; Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.
  • Horvath TL; Department of Physiology, Monash University, Victoria, Australia.
  • Spanswick D; Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia.
  • Tiganis T; Department of Physiology, Monash University, Victoria, Australia.
Elife ; 72018 09 19.
Article em En | MEDLINE | ID: mdl-30230471
ABSTRACT
Hypothalamic neurons respond to nutritional cues by altering gene expression and neuronal excitability. The mechanisms that control such adaptive processes remain unclear. Here we define populations of POMC neurons in mice that are activated or inhibited by insulin and thereby repress or inhibit hepatic glucose production (HGP). The proportion of POMC neurons activated by insulin was dependent on the regulation of insulin receptor signaling by the phosphatase TCPTP, which is increased by fasting, degraded after feeding and elevated in diet-induced obesity. TCPTP-deficiency enhanced insulin signaling and the proportion of POMC neurons activated by insulin to repress HGP. Elevated TCPTP in POMC neurons in obesity and/or after fasting repressed insulin signaling, the activation of POMC neurons by insulin and the insulin-induced and POMC-mediated repression of HGP. Our findings define a molecular mechanism for integrating POMC neural responses with feeding to control glucose metabolism.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Pró-Opiomelanocortina / Glucose / Insulina / Plasticidade Neuronal / Neurônios Limite: Animals / Humans / Male Idioma: En Revista: Elife Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Pró-Opiomelanocortina / Glucose / Insulina / Plasticidade Neuronal / Neurônios Limite: Animals / Humans / Male Idioma: En Revista: Elife Ano de publicação: 2018 Tipo de documento: Article