An age-related impairment of the regenerative capacity of aged muscle is a major contributor to declines in functional mobility and is associated with an increased morbidity in an elderly population. Following an acute injury, young skeletal muscle initiates a highly effective regenerative response, which largely restores the original architecture of the damaged fibers. Conversely, with increasing age, the regenerative response to injury results in a considerable scar tissue deposition at the expense of functional contractile tissue. Much of this healing defect has been attributed to an age-related decrease in muscle stem, or satellite, cell (MuSC) functionality. In response to skeletal muscle injury, MuSCs become activated from a quiescent state to repair damaged myofibers. However, it has been suggested that the increased fibrosis deposition following injury is a result of a myogenic-to-fibrogenic conversion of MuSCs. Fortunately, these age-related changes are reversible. Elegant studies employing heterochronic parabiosis, in which the circulatory systems of young and aged animals are conjoined, have revealed that rejuvenation of the systemic microenvironment significantly restores both whole tissue and MuSC regenerative capacity in aged muscle. These findings implicate that circulating factors, such as Klotho, play a critical rol in dictating skeletal muscle regenerative potential over time. Elucidation of the origin and nature of circulating factors contributing to the aged muscle phenotype is critical for the development of strategies to prevent, delay or reverse age-related declines. Consistent with the objective of the FOA, the overarching goal of this study is to identify a novel role for the anti-geronic protein, Klotho, in mediating declines in muscle healing capacity with increasing age, and to mechanistically test our hypothesis that age-related declines in neuromuscular activity contribute to attenuated Klotho expression. Specifically, in Aim 1, we will determine the direct effect of Klotho expression on MuSC fate determination and skeletal muscle regenerative potential.
In Aim 2, we will identify a role for Klotho-mediated regulation of MuSC senescence and impaired bioenergetics in dictating skeletal muscle regeneration. Finally, in Aim 3, we will investigate the molecular mechanism by which contractile activity regulates Klotho expression, both locally and systemically. These studies, when completed, will have a long and lasting impact on the field as they will establish Klotho as an important anti-geronic factor that regulate MuSC activity essential for functional muscle regeneration after injury. In addition, these experiments will lay the groundwork for future studies in which muscle stimulation in geriatric populations may be used to prevent, delay or reverse age-related declines in muscle function through improved regenerative capacity.

Public Health Relevance

Aging commonly results in a dramatically impaired healing response following an acute skeletal muscle injury. The long-term goal of this project is to better understand the role of the anti-aging protein, Klotho, on skeletal muscle regeneration. These experiments have been designed to lay the groundwork for the future development of strategies to prevent, delay or reverse declines in muscle vitality that are associated with increasing age.

Agency
National Institute of Health (NIH)
Institute
National Institute on Aging (NIA)
Type
High Priority, Short Term Project Award (R56)
Project #
1R56AG052978-01
Application #
9248756
Study Section
Special Emphasis Panel (ZAG1)
Program Officer
Kohanski, Ronald A
Project Start
2016-05-01
Project End
2017-04-30
Budget Start
2016-05-01
Budget End
2017-04-30
Support Year
1
Fiscal Year
2016
Total Cost
Indirect Cost
Name
University of Pittsburgh
Department
Physical Medicine & Rehab
Type
Schools of Medicine
DUNS #
004514360
City
Pittsburgh
State
PA
Country
United States
Zip Code
15213
Sahu, A; Mamiya, H; Shinde, S N et al. (2018) Age-related declines in ?-Klotho drive progenitor cell mitochondrial dysfunction and impaired muscle regeneration. Nat Commun 9:4859
Dalise, Stefania; Cavalli, Loredana; Ghuman, Harmanvir et al. (2017) Biological effects of dosing aerobic exercise and neuromuscular electrical stimulation in rats. Sci Rep 7:10830