Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 1 de 1
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Cell Rep ; 9(2): 633-45, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25373903

ABSTRACT

High-fat diets (HFDs) lead to obesity and inflammation in the central nervous system (CNS). Estrogens and estrogen receptor α (ERα) protect premenopausal females from the metabolic complications of inflammation and obesity-related disease. Here, we demonstrate that hypothalamic PGC-1α regulates ERα and inflammation in vivo. HFD significantly increased palmitic acid (PA) and sphingolipids in the CNS of male mice when compared to female mice. PA, in vitro, and HFD, in vivo, reduced PGC-1α and ERα in hypothalamic neurons and astrocytes of male mice and promoted inflammation. PGC-1α depletion with ERα overexpression significantly inhibited PA-induced inflammation, confirming that ERα is a critical determinant of the anti-inflammatory response. Physiologic relevance of ERα-regulated inflammation was demonstrated by reduced myocardial function in male, but not female, mice following chronic HFD exposure. Our findings show that HFD/PA reduces PGC-1α and ERα, promoting inflammation and decrements in myocardial function in a sex-specific way.


Subject(s)
Diet, High-Fat/adverse effects , Estrogen Receptor alpha/metabolism , Hypothalamus/metabolism , Transcription Factors/metabolism , Animals , Astrocytes/metabolism , Cell Line , Estrogen Receptor alpha/genetics , Female , Hypothalamus/cytology , Hypothalamus/drug effects , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Palmitic Acid/adverse effects , Palmitic Acid/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Sex Factors , Sphingolipids/metabolism , Transcription Factors/genetics , Ventricular Dysfunction/etiology , Ventricular Dysfunction/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...