Your browser doesn't support javascript.
loading
TMEPAI/PMEPA1 Is a Positive Regulator of Skeletal Muscle Mass.
Hagg, Adam; Kharoud, Swati; Goodchild, Georgia; Goodman, Craig A; Chen, Justin L; Thomson, Rachel E; Qian, Hongwei; Gregorevic, Paul; Harrison, Craig A; Walton, Kelly L.
Affiliation
  • Hagg A; Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
  • Kharoud S; Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.
  • Goodchild G; Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
  • Goodman CA; Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
  • Chen JL; Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, VIC, Australia.
  • Thomson RE; Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
  • Qian H; Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.
  • Gregorevic P; Australian Institute for Musculoskeletal Science, Sunshine Hospital, The University of Melbourne, St Albans, VIC, Australia.
  • Harrison CA; Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
  • Walton KL; Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
Front Physiol ; 11: 560225, 2020.
Article in En | MEDLINE | ID: mdl-33250771
ABSTRACT
Inhibition of myostatin- and activin-mediated SMAD2/3 signaling using ligand traps, such as soluble receptors, ligand-targeting propeptides and antibodies, or follistatin can increase skeletal muscle mass in healthy mice and ameliorate wasting in models of cancer cachexia and muscular dystrophy. However, clinical translation of these extracellular approaches targeting myostatin and activin has been hindered by the challenges of achieving efficacy without potential effects in other tissues. Toward the goal of developing tissue-specific myostatin/activin interventions, we explored the ability of transmembrane prostate androgen-induced (TMEPAI), an inhibitor of transforming growth factor-ß (TGF-ß1)-mediated SMAD2/3 signaling, to promote growth, and counter atrophy, in skeletal muscle. In this study, we show that TMEPAI can block activin A, activin B, myostatin and GDF-11 activity in vitro. To determine the physiological significance of TMEPAI, we employed Adeno-associated viral vector (AAV) delivery of a TMEPAI expression cassette to the muscles of healthy mice, which increased mass by as much as 30%, due to hypertrophy of muscle fibers. To demonstrate that TMEPAI mediates its effects via inhibition of the SMAD2/3 pathway, tibialis anterior (TA) muscles of mice were co-injected with AAV vectors expressing activin A and TMEPAI. In this setting, TMEPAI blocked skeletal muscle wasting driven by activin-induced phosphorylation of SMAD3. In a model of cancer cachexia associated with elevated circulating activin A, delivery of AAVTMEPAI into TA muscles of mice bearing C26 colon tumors ameliorated the muscle atrophy normally associated with cancer progression. Collectively, the findings indicate that muscle-directed TMEPAI gene delivery can inactivate the activin/myostatin-SMAD3 pathway to positively regulate muscle mass in healthy settings and models of disease.
Key words

Full text: 1 Database: MEDLINE Type of study: Prognostic_studies Language: En Year: 2020 Type: Article

Full text: 1 Database: MEDLINE Type of study: Prognostic_studies Language: En Year: 2020 Type: Article