PT - JOURNAL ARTICLE AU - Roselyne Castonguay AU - Jennifer Lachey AU - Samantha Wallner AU - Jamie Strand AU - Katia Liharska AU - Abigail E. Watanabe AU - Marishka Cannell AU - Monique V. Davies AU - Dianne Sako AU - Megan E. Troy AU - Lavanya Krishnan AU - Aaron W. Mulivor AU - Huiming Li AU - Sarah Keates AU - Mark J. Alexander AU - R. Scott Pearsall AU - Ravi Kumar TI - Follistatin-288-Fc Fusion Protein Promotes Localized Growth of Skeletal Muscle AID - 10.1124/jpet.118.252304 DP - 2018 Jan 01 TA - Journal of Pharmacology and Experimental Therapeutics PG - jpet.118.252304 4099 - http://jpet.aspetjournals.org/content/early/2018/12/18/jpet.118.252304.short 4100 - http://jpet.aspetjournals.org/content/early/2018/12/18/jpet.118.252304.full AB - Follistatin is an endogenous glycoprotein that promotes growth and repair of skeletal muscle by sequestering inhibitory ligands of the transforming growth factor-β (TGF-β) superfamily and may therefore have therapeutic potential for neuromuscular diseases. Here, we sought to determine the suitability of a newly engineered follistatin fusion protein (FST288-Fc) to promote localized – rather than systemic – growth of skeletal muscle by capitalizing on the intrinsic heparin-binding ability of the follistatin-288 isoform. As determined by surface plasmon resonance and cell-based assays, FST288-Fc binds to activin A, activin B, myostatin (GDF8) and GDF11 with high affinity and neutralizes their activity in vitro. Intramuscular administration of FST288-Fc in mice induced robust, dose-dependent growth of the targeted muscle but not of surrounding or contralateral muscles, in contrast to the systemic effects of a locally administered fusion protein incorporating activin receptor type IIB (ActRIIB-Fc). Furthermore, systemic administration of FST288-Fc in mice did not alter muscle mass or body composition as determined by nuclear magnetic resonance, which again contrasts with the pronounced systemic activity of ActRIIB-Fc when administered by the same route. Subsequent analysis revealed that FST288-Fc in the circulation undergoes rapid proteolysis, thereby restricting its activity to individual muscles targeted by intramuscular administration. These results indicate that FST288-Fc can produce localized growth of skeletal muscle in a targeted manner with reduced potential for undesirable systemic effects. Thus, FST288-Fc and similar agents may be beneficial in the treatment of disorders with muscle atrophy that is focal, asymmetric or otherwise heterogeneous.