[HTML][HTML] Senescent cardiac fibroblast is critical for cardiac fibrosis after myocardial infarction

F Zhu, Y Li, J Zhang, C Piao, T Liu, HH Li, J Du - PloS one, 2013 - journals.plos.org
F Zhu, Y Li, J Zhang, C Piao, T Liu, HH Li, J Du
PloS one, 2013journals.plos.org
Senescence is a recognized mechanism of cardiovascular diseases; however, its
contribution to myocardial fibrosis and rupture after infarction and the underlying
mechanisms remain unclear. Here we showed that senescent cardiac fibroblasts markedly
accumulated in heart after myocardial infarction. The expression of key senescence
regulators, especially p53, was significantly up-regulated in the infarcted heart or hypoxia-
treated fibroblasts. Furthermore, knockdown of endogenous p53 by siRNA in fibroblasts …
Senescence is a recognized mechanism of cardiovascular diseases; however, its contribution to myocardial fibrosis and rupture after infarction and the underlying mechanisms remain unclear. Here we showed that senescent cardiac fibroblasts markedly accumulated in heart after myocardial infarction. The expression of key senescence regulators, especially p53, was significantly up-regulated in the infarcted heart or hypoxia-treated fibroblasts. Furthermore, knockdown of endogenous p53 by siRNA in fibroblasts markedly reduced hypoxia-induced cell senescence, cytokine expression but increased collagen expression, whereas increased expression of p53 protein by adenovirus infection had opposite effects. Consistent with in vitro results in cardiac fibroblasts, p53 deficiency in vivo significantly decreased the accumulation of senescent fibroblasts, the infiltration of macrophages and matrix metalloproteinases, but enhanced collagen deposition after myocardial infarction. In conclusion, these results suggest that the p53-mediated fibroblast senescence limits cardiac collagen production, and inhibition of p53 activity could represent a novel therapeutic target to increase reparative fibrosis and to prevent heart rupture after myocardial infarction.
PLOS