[HTML][HTML] Neurons and glial cells acquire a senescent signature after repeated mild traumatic brain injury in a sex-dependent manner

N Schwab, D Taskina, E Leung, BT Innes… - Frontiers in …, 2022 - frontiersin.org
N Schwab, D Taskina, E Leung, BT Innes, GD Bader, LN Hazrati
Frontiers in neuroscience, 2022frontiersin.org
Mild traumatic brain injury (mTBI) is an important public health issue, as it can lead to long-
term neurological symptoms and risk of neurodegenerative disease. The pathophysiological
mechanisms driving this remain unclear, and currently there are no effective therapies for
mTBI. In this study on repeated mTBI (rmTBI), we have induced three mild closed-skull
injuries or sham procedures, separated by 24 h, in C57BL/6 mice. We show that rmTBI mice
have prolonged righting reflexes and astrogliosis, with neurological impairment in the Morris …
Mild traumatic brain injury (mTBI) is an important public health issue, as it can lead to long-term neurological symptoms and risk of neurodegenerative disease. The pathophysiological mechanisms driving this remain unclear, and currently there are no effective therapies for mTBI. In this study on repeated mTBI (rmTBI), we have induced three mild closed-skull injuries or sham procedures, separated by 24 h, in C57BL/6 mice. We show that rmTBI mice have prolonged righting reflexes and astrogliosis, with neurological impairment in the Morris water maze (MWM) and the light dark test. Cortical and hippocampal tissue analysis revealed DNA damage in the form of double-strand breaks, oxidative damage, and R-loops, markers of cellular senescence including p16 and p21, and signaling mediated by the cGAS-STING pathway. This study identified novel sex differences after rmTBI in mice. Although these markers were all increased by rmTBI in both sexes, females had higher levels of DNA damage, lower levels of the senescence protein p16, and lower levels of cGAS-STING signaling proteins compared to their male counterparts. Single-cell RNA sequencing of the male rmTBI mouse brain revealed activation of the DNA damage response, evidence of cellular senescence, and pro-inflammatory markers reminiscent of the senescence-associated secretory phenotype (SASP) in neurons and glial cells. Cell-type specific changes were also present with evidence of brain immune activation, neurotransmission alterations in both excitatory and inhibitory neurons, and vascular dysfunction. Treatment of injured mice with the senolytic drug ABT263 significantly reduced markers of senescence only in males, but was not therapeutic in females. The reduction of senescence by ABT263 in male mice was accompanied by significantly improved performance in the MWM. This study provides compelling evidence that senescence contributes to brain dysfunction after rmTBI, but may do so in a sex-dependent manner.
Frontiers