p53-dependent release of Alarmin HMGB1 is a central mediator of senescent phenotypes

AR Davalos, M Kawahara, GK Malhotra… - Journal of Cell …, 2013 - rupress.org
AR Davalos, M Kawahara, GK Malhotra, N Schaum, J Huang, U Ved, CM Beausejour
Journal of Cell Biology, 2013rupress.org
Cellular senescence irreversibly arrests proliferation in response to potentially oncogenic
stress. Senescent cells also secrete inflammatory cytokines such as IL-6, which promote age-
associated inflammation and pathology. HMGB1 (high mobility group box 1) modulates
gene expression in the nucleus, but certain immune cells secrete HMGB1 as an extracellular
Alarmin to signal tissue damage. We show that nuclear HMGB1 relocalized to the
extracellular milieu in senescent human and mouse cells in culture and in vivo. In contrast to …
Cellular senescence irreversibly arrests proliferation in response to potentially oncogenic stress. Senescent cells also secrete inflammatory cytokines such as IL-6, which promote age-associated inflammation and pathology. HMGB1 (high mobility group box 1) modulates gene expression in the nucleus, but certain immune cells secrete HMGB1 as an extracellular Alarmin to signal tissue damage. We show that nuclear HMGB1 relocalized to the extracellular milieu in senescent human and mouse cells in culture and in vivo. In contrast to cytokine secretion, HMGB1 redistribution required the p53 tumor suppressor, but not its activator ATM. Moreover, altered HMGB1 expression induced a p53-dependent senescent growth arrest. Senescent fibroblasts secreted oxidized HMGB1, which stimulated cytokine secretion through TLR-4 signaling. HMGB1 depletion, HMGB1 blocking antibody, or TLR-4 inhibition attenuated senescence-associated IL-6 secretion, and exogenous HMGB1 stimulated NF-κB activity and restored IL-6 secretion to HMGB1-depleted cells. Our findings identify senescence as a novel biological setting in which HMGB1 functions and link HMGB1 redistribution to p53 activity and senescence-associated inflammation.
rupress.org