4.7 Article

ROS-induced PADI2 downregulation accelerates cellular senescence via the stimulation of SASP production and NFκB activation

Journal

CELLULAR AND MOLECULAR LIFE SCIENCES
Volume 79, Issue 3, Pages -

Publisher

SPRINGER BASEL AG
DOI: 10.1007/s00018-022-04186-5

Keywords

Senescence; Reactive oxygen species; Peptidyl arginine deiminase 2; Senescence-associated secretory phenotype; NF kappa B; Osteoblast

Funding

  1. Basic Science Research Program through the National Research Foundation of Korea (NRF) - Ministry of Education [2017R1A4A1014584, 2020R1A4A1019423, 2017R1A2B3011778, 2020R1A2B5B02002658, 2021R1A2C1007715]
  2. National Research Foundation of Korea [2017R1A4A1014584, 2021R1A2C1007715, 2020R1A2B5B02002658, 2017R1A2B3011778] Funding Source: Korea Institute of Science & Technology Information (KISTI), National Science & Technology Information Service (NTIS)

Ask authors/readers for more resources

Cellular senescence is closely related to tissue aging, including bone. Oxidative stress caused by the accumulation of reactive oxygen species (ROS) accelerates cellular senescence and dysfunction of osteoblasts. This study identified PADI2 as a regulator of ROS-accelerated senescence of osteoblasts and provided new insights into excessive ROS-promoted cellular senescence and aging-related bone diseases.
Cellular senescence is closely related to tissue aging including bone. Bone homeostasis is maintained by the tight balance between bone-forming osteoblasts and bone-resorting osteoclasts, but it undergoes deregulation with age, causing age-associated osteoporosis, a main cause of which is osteoblast dysfunction. Oxidative stress caused by the accumulation of reactive oxygen species (ROS) in bone tissues with aging can accelerate osteoblast senescence and dysfunction. However, the regulatory mechanism that controls the ROS-induced senescence of osteoblasts is poorly understood. Here, we identified Peptidyl arginine deiminase 2 (PADI2), a post-translational modifying enzyme, as a regulator of ROS-accelerated senescence of osteoblasts via RNA-sequencing and further functional validations. PADI2 downregulation by treatment with H2O2 or its siRNA promoted cellular senescence and suppressed osteoblast differentiation. CCL2, 5, and 7 known as the elements of the senescence-associated secretory phenotype (SASP) which is a secretome including proinflammatory cytokines and chemokines emitted by senescent cells and a representative feature of senescence, were upregulated by H2O2 treatment or Padi2 knockdown. Furthermore, blocking these SASP factors with neutralizing antibodies or siRNAs alleviated the senescence and dysfunction of osteoblasts induced by H2O2 treatment or Padi2 knockdown. The elevated production of these SASP factors was mediated by the activation of NF kappa B signaling pathway. The inhibition of NF kappa B using the pharmacological inhibitor or siRNA effectively relieved H2O2 treatment- or Padi2 knockdown-induced senescence and osteoblast dysfunction. Together, our study for the first time uncover the role of PADI2 in ROS-accelerated cellular senescence of osteoblasts and provide new mechanistic and therapeutic insights into excessive ROS-promoted cellular senescence and aging-related bone diseases.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available