Your browser doesn't support javascript.
loading
Iron overload reduces adiponectin receptor expression via a ROS/FOXO1-dependent mechanism leading to adiponectin resistance in skeletal muscle cells.
Dahyaleh, Karam; Sung, Hye K; Prioriello, Michelle; Rengasamy, Palanivel; Lam, Nhat H; Kim, Jae B; Gross, Sean; Sweeney, Gary.
Afiliación
  • Dahyaleh K; Department of Biology, York University, Toronto, Canada.
  • Sung HK; Department of Biology, York University, Toronto, Canada.
  • Prioriello M; Department of Biology, York University, Toronto, Canada.
  • Rengasamy P; Department of Biology, York University, Toronto, Canada.
  • Lam NH; Department of Biology, York University, Toronto, Canada.
  • Kim JB; School of Biological Sciences, Seoul National University, Seoul, South Korea.
  • Gross S; Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Knight Cancer Institute, Oregon Health and Sciences University, Portland, Oregon, USA.
  • Sweeney G; Department of Biology, York University, Toronto, Canada.
J Cell Physiol ; 236(7): 5339-5351, 2021 07.
Article en En | MEDLINE | ID: mdl-33432609
ABSTRACT
Iron overload (IO) is a common yet underappreciated finding in metabolic syndrome (MetS) patients. With the prevalence of MetS continuing to rise, it is imperative to further elucidate cellular mechanisms leading to metabolic dysfunction. Adiponectin has many beneficial effects and is a therapeutic target for the treatment of MetS and cardiovascular diseases. IO positively correlates with reduced circulating adiponectin levels yet the impact of IO on adiponectin action is unknown. Here, we established a model of IO in L6 skeletal muscle cells and found that IO-induced adiponectin resistance. This was shown via reduced p38 mitogen-activated protein kinase phosphorylation in response to the small molecule adiponectin receptor (AdipoR) agonist, AdipoRon, in presence of IO. This correlated with reduced messenger RNA and protein levels of AdipoR1 and its facilitative signaling binding partner, APPL1. IO caused phosphorylation, nuclear extrusion, and thus inhibition of FOXO1, a known transcription factor regulating AdipoR1 expression. The antioxidant N-acetyl cystine attenuated the production of reactive oxygen species (ROS) by IO, and blunted its effect on FOXO1 phosphorylation and removal from the nucleus, as well as subsequent adiponectin resistance. In conclusion, our study identifies a ROS/FOXO1/AdipoR1 axis as a cause of skeletal muscle adiponectin resistance in response to IO. This new knowledge provides insight into a cellular mechanism with potential relevance to disease pathophysiology in MetS patients with IO.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Especies Reactivas de Oxígeno / Fibras Musculares Esqueléticas / Sobrecarga de Hierro / Adiponectina / Proteína Forkhead Box O1 Tipo de estudio: Prognostic_studies / Risk_factors_studies Límite: Animals Idioma: En Revista: J Cell Physiol Año: 2021 Tipo del documento: Article País de afiliación: Canadá

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Especies Reactivas de Oxígeno / Fibras Musculares Esqueléticas / Sobrecarga de Hierro / Adiponectina / Proteína Forkhead Box O1 Tipo de estudio: Prognostic_studies / Risk_factors_studies Límite: Animals Idioma: En Revista: J Cell Physiol Año: 2021 Tipo del documento: Article País de afiliación: Canadá