Targeted reduction of senescent cell burden alleviates focal radiotherapy‐related bone loss

A Chandra, AB Lagnado, JN Farr… - Journal of Bone and …, 2020 - Wiley Online Library
A Chandra, AB Lagnado, JN Farr, DG Monroe, S Park, C Hachfeld, T Tchkonia, JL Kirkland…
Journal of Bone and Mineral Research, 2020Wiley Online Library
Clinical radiotherapy treats life‐threatening cancers, but the radiation often affects
neighboring normal tissues including bone. Acute effects of ionizing radiation include
oxidative stress, DNA damage, and cellular apoptosis. We show in this study that a large
proportion of bone marrow cells, osteoblasts, and matrix‐embedded osteocytes recover from
these insults only to attain a senescent profile. Bone analyses of senescence‐associated
genes, senescence‐associated beta‐galactosidase (SA‐β‐gal) activity, and presence of …
Abstract
Clinical radiotherapy treats life‐threatening cancers, but the radiation often affects neighboring normal tissues including bone. Acute effects of ionizing radiation include oxidative stress, DNA damage, and cellular apoptosis. We show in this study that a large proportion of bone marrow cells, osteoblasts, and matrix‐embedded osteocytes recover from these insults only to attain a senescent profile. Bone analyses of senescence‐associated genes, senescence‐associated beta‐galactosidase (SA‐β‐gal) activity, and presence of telomere dysfunction‐induced foci (TIF) at 1, 7, 14, 21, and 42 days post–focal radiation treatment (FRT) in C57BL/6 male mice confirmed the development of senescent cells and the senescence‐associated secretory phenotype (SASP). Accumulation of senescent cells and SASP markers were correlated with a significant reduction in bone architecture at 42 days post‐FRT. To test if senolytic drugs, which clear senescent cells, alleviate FRT‐related bone damage, we administered the senolytic agents, dasatinib (D), quercetin (Q), fisetin (F), and a cocktail of D and Q (D+Q). We found moderate alleviation of radiation‐induced bone damage with D and Q as stand‐alone compounds, but no such improvement was seen with F. However, the senolytic cocktail of D+Q reduced senescent cell burden as assessed by TIF+ osteoblasts and osteocytes, markers of senescence (p16 Ink4a and p21), and key SASP factors, resulting in significant recovery in the bone architecture of radiated femurs. In summary, this study provides proof of concept that senescent cells play a role in radiotherapy‐associated bone damage, and that reduction in senescent cell burden by senolytic agents is a potential therapeutic option for alleviating radiotherapy‐related bone deterioration. © 2020 American Society for Bone and Mineral Research.
Wiley Online Library