Peroxisome proliferator-activated receptor γ coactivator-1α is a central negative regulator of vascular senescence

S Xiong, G Salazar, N Patrushev, M Ma… - … , and vascular biology, 2013 - Am Heart Assoc
S Xiong, G Salazar, N Patrushev, M Ma, F Forouzandeh, L Hilenski, RW Alexander
Arteriosclerosis, Thrombosis, and vascular biology, 2013Am Heart Assoc
Objective—Cellular senescence influences organismal aging and increases predisposition
to age-related diseases, in particular cardiovascular disease, a leading cause of death and
disability worldwide. Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) is
a master regulator of mitochondrial biogenesis and function, oxidative stress, and insulin
resistance. Senescence is associated with telomere and mitochondrial dysfunction and
oxidative stress, implying a potential causal role of PGC-1α in senescence pathogenesis …
Objective
Cellular senescence influences organismal aging and increases predisposition to age-related diseases, in particular cardiovascular disease, a leading cause of death and disability worldwide. Peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) is a master regulator of mitochondrial biogenesis and function, oxidative stress, and insulin resistance. Senescence is associated with telomere and mitochondrial dysfunction and oxidative stress, implying a potential causal role of PGC-1α in senescence pathogenesis.
Approach and Results
We generated a PGC-1α+/–/apolipoprotein E–/– mouse model and showed that PGC-1α deficiency promotes a vascular senescence phenotype that is associated with increased oxidative stress, mitochondrial abnormalities, and reduced telomerase activity. PGC-1α disruption results in reduced expression of the longevity-related deacetylase sirtuin 1 (SIRT1) and the antioxidant catalase, and increased expression of the senescence marker p53 in aortas. Further, angiotensin II, a major hormonal inducer of vascular senescence, induces prolonged lysine acetylation of PGC-1α and releases the PGC-1α–FoxO1 complex from the SIRT1 promoter, thus reducing SIRT1 expression. The phosphorylation-defective mutant PGC-1α S570A is not acetylated, is constitutively active for forkhead box O1-dependent SIRT1 transcription, and prevents angiotensin II–induced senescence. Acetylation of PGC-1α by angiotensin II interrupts the PGC-1α–forkhead box O1–SIRT1 feed-forward signaling circuit leading to SIRT1 and catalase downregulation and vascular senescence.
Conclusions
PGC-1α is a primary negative regulator of vascular senescence. Moreover, the central role of posttranslational modification of PGC-1α in regulating angiotensin II–induced vascular senescence may inform development of novel therapeutic strategies for mitigating age-associated diseases, such as atherosclerosis.
Am Heart Assoc