(provided by PI): Myoblast fusion is a critical event that is required not only for the development of skeletal muscle during embryogenesis but also for the regeneration of adult myofibers upon injury and for load-induced skeletal muscle hypertrophy. Augmenting the natural process of muscle cell fusion in existing or introduced myogenic cells has enormous therapeutic potential to treat degenerative muscle diseases. However, the mechanisms of myoblast fusion in vertebrates remain less understood. Accumulating evidence suggests that myoblast fusion in mammalian system involves the activation of specific cell signaling pathways such as MAPK, non-canonical NF-?B, canonical Wnt signaling. However, the upstream signaling mechanisms leading to their activation and how the activation of these pathways promotes myoblast fusion during myogenesis remain poorly understood. Our preliminary studies have identified that myeloid differentiation primary response gene 88 (Myd88) is a critical regulator of myoblast fusion both in vitro and in vivo. Our results show that the levels of Myd88 are increased during myogenesis. Genetic ablation of Myd88 impairs myoblast fusion whereas overexpression of Myd88 enhances the formation of myotubes in cultured myoblasts. Furthermore, we have obtained initial evidence that Myd88 regulates the activation of specific profusion signaling pathways during myogenesis. Based on our preliminary studies, we have proposed to establish the role and delineate the signaling and molecular mechanisms by which Myd88 mediates skeletal muscle formation in vitro and in vivo. Our working hypotheses are: (I) Myd88 promotes myoblast fusion in multiple conditions through augmenting the gene expression of specific profusion molecules; (II) Myd88 coordinates the activation of canonical Wnt and non-canonical NF-?B signaling to promote myoblast fusion during myogenesis; (III) Both transcriptional and post-transcriptional mechanisms regulate the levels of Myd88 in differentiating myoblasts. We will test these hypotheses by addressing the following three specific aims.
Aim I : Establish the role and investigate molecular mechanisms by which Myd88 mediates myoblast fusion;
Aim II : Investigate the signaling networks through which Myd88 promotes myoblast fusion;
and Aim III : Investigate the mechanisms by which levels of Myd88 are increased during myogenesis. Successful completion of this project will provide critical insights into the mechanisms of myoblast fusion and will lead to the identification of novel drug targets for treatment of muscle disorders.

Public Health Relevance

Myoblast fusion is a fundamental process that is crucial for the repair and growth of skeletal muscle upon injury. Understanding of the mechanisms of myoblast fusion has enormous potential for the development of novel therapeutics aimed at treating various genetic and acquired degenerative muscle disorders. The primary goal of the proposed research is to understand the signaling mechanisms of myoblast fusion and thereby providing fundamental knowledge required to enhance the therapeutic potential of myogenic cells to treat muscle wasting in various muscle disorders.

Agency
National Institute of Health (NIH)
Institute
National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
Type
Research Project (R01)
Project #
1R01AR068313-01
Application #
8941165
Study Section
Special Emphasis Panel (ZRG1-MOSS-V (03))
Program Officer
Boyce, Amanda T
Project Start
2015-09-14
Project End
2020-08-31
Budget Start
2015-09-14
Budget End
2016-08-31
Support Year
1
Fiscal Year
2015
Total Cost
$335,541
Indirect Cost
$115,541
Name
University of Louisville
Department
Anatomy/Cell Biology
Type
Schools of Medicine
DUNS #
057588857
City
Louisville
State
KY
Country
United States
Zip Code
40202
Bohnert, Kyle R; McMillan, Joseph D; Kumar, Ashok (2018) Emerging roles of ER stress and unfolded protein response pathways in skeletal muscle health and disease. J Cell Physiol 233:67-78
Hindi, Sajedah M; Sato, Shuichi; Xiong, Guangyan et al. (2018) TAK1 regulates skeletal muscle mass and mitochondrial function. JCI Insight 3:
Gallot, Yann S; Straughn, Alex R; Bohnert, Kyle R et al. (2018) MyD88 is required for satellite cell-mediated myofiber regeneration in dystrophin-deficient mdx mice. Hum Mol Genet 27:3449-3463
Xiong, Guangyan; Hindi, Sajedah M; Mann, Aman K et al. (2017) The PERK arm of the unfolded protein response regulates satellite cell-mediated skeletal muscle regeneration. Elife 6:
Hindi, Sajedah M; Shin, Jonghyun; Gallot, Yann S et al. (2017) MyD88 promotes myoblast fusion in a cell-autonomous manner. Nat Commun 8:1624
Hindi, Lubna; McMillan, Joseph D; Afroze, Dil et al. (2017) Isolation, Culturing, and Differentiation of Primary Myoblasts from Skeletal Muscle of Adult Mice. Bio Protoc 7:
Gallot, Yann S; McMillan, Joseph D; Xiong, Guangyan et al. (2017) Distinct roles of TRAF6 and TAK1 in the regulation of adipocyte survival, thermogenesis program, and high-fat diet-induced obesity. Oncotarget 8:112565-112583
Simionescu-Bankston, Adriana; Kumar, Ashok (2016) Noncoding RNAs in the regulation of skeletal muscle biology in health and disease. J Mol Med (Berl) 94:853-66
Hindi, Sajedah M; Kumar, Ashok (2016) TRAF6 regulates satellite stem cell self-renewal and function during regenerative myogenesis. J Clin Invest 126:151-68
Gallot, Yann Simon; Hindi, Sajedah M; Mann, Aman K et al. (2016) Isolation, Culture, and Staining of Single Myofibers. Bio Protoc 6:

Showing the most recent 10 out of 13 publications