RT Journal Article SR Electronic T1 Mitochondrial dysfunction and autophagy responses to skeletal muscle stress JF bioRxiv FD Cold Spring Harbor Laboratory SP 597476 DO 10.1101/597476 A1 Anna S. Nichenko A1 W. Michael Southern A1 Anita E. Qualls A1 Alexandra B. Flemington A1 Grant H. Mercer A1 Amelia Yin A1 Hang Yin A1 Jarrod A. Call YR 2019 UL http://biorxiv.org/content/early/2019/04/04/597476.abstract AB Autophagy plays an important role in mitochondrial maintenance, yet many details of skeletal muscle autophagic activity are unresolved in the context of muscle stress and/or damage. Skeletal muscles from mice were stressed either by fatiguing contractions, eccentric contraction-induced injury (ECCI), or freeze injury (FI) to establish a timeline of mitochondrial function and autophagy induction after different forms of muscle stress. Only FI was sufficient to elicit a reduction in mitochondrial function (āˆ’88%, p=0.006), yet both ECCI and FI resulted in greater autophagy-related protein content (28-fold, pā‰¤0.008) suggesting a tunable autophagic response. Muscles from another cohort of mice were used to determine specific forms of autophagy, i.e., flux and mitochondrial-specific, in response to muscle damage. Mitochondrial-specific autophagy was evident by accumulation of autophagy-related proteins in mitochondrial-enriched muscle fractions following FI (37-fold, p=0.017); however, autophagy flux, assessed by LC3II accumulation with the lysosomal inhibitor chloroquine, was insignificant suggesting a physiological bottleneck in the clearance of dysfunctional organelles following FI. Ulk1 muscle-specific knockout (Ulk1 MKO) mice were used to determine if autophagy is necessary for the recovery of mitochondrial function after muscle damage. Ulk1 MKO mice were weaker (āˆ’12%, p=0.012) and demonstrated altered satellite cell dynamics (e.g., proliferation) during muscle regeneration after FI compared to littermate control mice, but determination of autophagy necessity for the recovery of mitochondrial function was inconclusive. This study concludes that autophagy is a tunable cellular response to muscle damaging stress and may influence muscle fiber regeneration through interaction with satellite cells.Key Points SummaryMuscle contractility dysfunction is well characterized after many different types of muscle stress however, the timing and magnitude of mitochondrial dysfunction and autophagy induction after different types of muscle stress is largely unknown.In this study we found that only traumatic freeze injury causes mitochondria dysfunction compared to fatigue contractions and eccentric contraction-induced injury, and that the autophagic response to muscle stress scales to the magnitude of muscle damage, i.e., freeze vs. eccentric contraction-induced injury.We determined that total autophagy-related protein content has a greater response to muscle fiber damage compared to autophagy flux likely reflecting a bottleneck of autophagosomes awaiting degradation following muscle injury.Using a skeletal gmuscle-specific autophagy knockout mouse (Ulk1), we found that muscle contractility and satellite cell activity might be influenced by cellular events within the adult muscle fiber following muscle damage.