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-β (TGF-β1)-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 AAV:TMEPAI 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.

Highlights

  • Myostatin, activin A, activin B, and GDF11 comprise a subgroup within the transforming growth factor-β (TGF-β) family of proteins that is of great interest in the field of skeletal muscle biology (Chen et al, 2016a)

  • In previous studies conducted by our laboratories, transcriptional profiling indicated that skeletal muscle expression of transmembrane prostate androgen-induced (TMEPAI) mRNA (Pmepa1) increased following exposure to activin A (Chen et al, 2014) and decreased when activin and/or myostatin signaling pathways were inhibited (Chen et al, 2017)

  • QPCR analysis of mouse tibialis anterior (TA) muscles injected with associated viral vector (AAV):ActA confirmed that TMEPAI transcription increased markedly (6-fold) in response to activin A expression (Supplementary Figure 1A)

Read more

Summary

Introduction

Activin A, activin B, and GDF11 comprise a subgroup within the transforming growth factor-β (TGF-β) family of proteins that is of great interest in the field of skeletal muscle biology (Chen et al, 2016a). SMAD2/3 activation regulates the expression of the E3 ubiquitin ligases MuRF-1 and Atrogin-1 (Sartori et al, 2009; Lokireddy et al, 2011; Goodman et al, 2013), which mediate ubiquitination and proteasomal degradation of myofibrillar proteins, such as myosin (Clarke et al, 2007; Fielitz et al, 2007) These ActRIIA/B ligands regulate genes associated with muscle protein turnover, metabolism, and sarcomere function (Chen et al, 2015, 2017) that are transcriptionally indicative of muscle wasting associated with cancer cachexia (Baker et al, 2010; Johnston et al, 2015; Rovira Gonzalez et al, 2019)

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call