Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
FEBS Open Bio ; 13(1): 185-194, 2023 01.
Article in English | MEDLINE | ID: mdl-36416450

ABSTRACT

Macrophages distributed in tissues throughout the body contribute to homeostasis. In the inflammatory state, macrophages undergo mechanical stress that regulates the signal transduction of immune responses and various cellular functions. However, the effects of the inflammatory response on macrophages under physiological cyclic stretch are unclear. We found that physiological cyclic stretch suppresses inflammatory cytokine expression in macrophages by regulating NF-κB activity. NF-κB phosphorylation at Ser536 in macrophages was inhibited, suggesting that tank-binding kinase (TBK1) regulates NF-κB activity during physiological stress. Moreover, TBK1 expression was suppressed by physiological stretch, and TBK1 knockdown by siRNA induced the suppression of NF-κB phosphorylation at Ser536. In conclusion, physiological stretch triggers suppression of a TBK1-dependent excessive inflammatory response, which may be necessary to maintain tissue homeostasis.


Subject(s)
Lipopolysaccharides , NF-kappa B , NF-kappa B/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Signal Transduction , Immunity
2.
Sci Rep ; 12(1): 1377, 2022 01 26.
Article in English | MEDLINE | ID: mdl-35082348

ABSTRACT

Muscle wasting is a major problem leading to reduced quality of life and higher risks of mortality and various diseases. Muscle atrophy is caused by multiple conditions in which protein degradation exceeds its synthesis, including disuse, malnutrition, and microgravity. While Vitamin D receptor (VDR) is well known to regulate calcium and phosphate metabolism to maintain bone, recent studies have shown that VDR also plays roles in skeletal muscle development and homeostasis. Moreover, its expression is upregulated in muscle undergoing atrophy as well as after muscle injury. Here we show that VDR regulates simulated microgravity-induced atrophy in C2C12 myotubes in vitro. After 8 h of microgravity simulated using 3D-clinorotation, the VDR-binding motif was associated with chromatin regions closed by the simulated microgravity and enhancer regions inactivated by it, which suggests VDR mediates repression of enhancers. In addition, VDR was induced and translocated into the nuclei in response to simulated microgravity. VDR-deficient C2C12 myotubes showed resistance to simulated microgravity-induced atrophy and reduced induction of FBXO32, an atrophy-associated ubiquitin ligase. These results demonstrate that VDR contributes to the regulation of simulated microgravity-induced atrophy at least in part by controlling expression of atrophy-related genes.


Subject(s)
Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Myoblasts, Skeletal/metabolism , Receptors, Calcitriol/metabolism , Signal Transduction/genetics , Weightlessness Simulation/adverse effects , Animals , Cell Line , Gene Knockout Techniques/methods , Homeostasis/genetics , Mice , Muscle Development/genetics , Muscular Atrophy/genetics , Receptors, Calcitriol/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...