Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 8(1): 1624, 2017 11 20.
Article in English | MEDLINE | ID: mdl-29158520

ABSTRACT

Myoblast fusion is an indispensable step for skeletal muscle development, postnatal growth, and regeneration. Myeloid differentiation primary response gene 88 (MyD88) is an adaptor protein that mediates Toll-like receptors and interleukin-1 receptor signaling. Here we report a cell-autonomous role of MyD88 in the regulation of myoblast fusion. MyD88 protein levels are increased during in vitro myogenesis and in conditions that promote skeletal muscle growth in vivo. Deletion of MyD88 impairs fusion of myoblasts without affecting their survival, proliferation, or differentiation. MyD88 regulates non-canonical NF-κB and canonical Wnt signaling during myogenesis and promotes skeletal muscle growth and overload-induced myofiber hypertrophy in mice. Ablation of MyD88 reduces myofiber size during muscle regeneration, whereas its overexpression promotes fusion of exogenous myoblasts to injured myofibers. Our study shows that MyD88 modulates myoblast fusion and suggests that augmenting its levels may be a therapeutic approach to improve skeletal muscle formation in degenerative muscle disorders.


Subject(s)
Myeloid Differentiation Factor 88/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Animals , Cell Differentiation , Cell Fusion , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Development , Muscle, Skeletal/metabolism , Myeloid Differentiation Factor 88/genetics , Wnt Signaling Pathway
2.
J Mol Med (Berl) ; 94(8): 853-66, 2016 08.
Article in English | MEDLINE | ID: mdl-27377406

ABSTRACT

Skeletal muscle is composed of multinucleated myofibers that arise from the fusion of myoblasts during development. Skeletal muscle is essential for various body functions such as maintaining posture, locomotion, breathing, and metabolism. Skeletal muscle undergoes remarkable adaptations in response to environmental stimuli leading to atrophy or hypertrophy. Moreover, degeneration of skeletal muscle is a common feature in a number of muscular disorders including muscular dystrophy. Emerging evidence suggests that noncoding RNAs, such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are critical for skeletal muscle physiology. Several miRNAs and lncRNAs have now been found to control skeletal muscle development and regeneration. Noncoding RNAs also play an important role in the regulation of skeletal muscle mass in adults. Furthermore, aberrant expression of miRNAs and lncRNAs has been observed in several muscular disorders. In this article, we discuss the mechanisms of action of miRNAs and lncRNAs in skeletal muscle formation, growth, regeneration, and disease. We further highlight potential therapeutic strategies for utilizing noncoding RNAs to improve skeletal muscle function.


Subject(s)
Muscle Development , Muscle, Skeletal/metabolism , Muscular Dystrophies/metabolism , RNA, Untranslated/physiology , Animals , Gene Expression , Humans , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , RNA Interference , Regeneration
3.
Am J Physiol Cell Physiol ; 308(11): C919-31, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25810257

ABSTRACT

Myoblast fusion is critical for proper muscle growth and regeneration. During myoblast fusion, the localization of some molecules is spatially restricted; however, the exact reason for such localization is unknown. Creatine kinase B (CKB), which replenishes local ATP pools, localizes near the ends of cultured primary mouse myotubes. To gain insights into the function of CKB, we performed a yeast two-hybrid screen to identify CKB-interacting proteins. We identified molecules with a broad diversity of roles, including actin polymerization, intracellular protein trafficking, and alternative splicing, as well as sarcomeric components. In-depth studies of α-skeletal actin and α-cardiac actin, two predominant muscle actin isoforms, demonstrated their biochemical interaction and partial colocalization with CKB near the ends of myotubes in vitro. In contrast to other cell types, specific knockdown of CKB did not grossly affect actin polymerization in myotubes, suggesting other muscle-specific roles for CKB. Interestingly, knockdown of CKB resulted in significantly increased myoblast fusion and myotube size in vitro, whereas knockdown of creatine kinase M had no effect on these myogenic parameters. Our results suggest that localized CKB plays a key role in myotube formation by limiting myoblast fusion during myogenesis.


Subject(s)
Creatine Kinase, BB Form/genetics , Muscle Development/genetics , Muscle Fibers, Skeletal/enzymology , Myoblasts/enzymology , Actins/genetics , Actins/metabolism , Alternative Splicing , Animals , Cell Fusion , Creatine Kinase, BB Form/antagonists & inhibitors , Creatine Kinase, BB Form/metabolism , Creatine Kinase, MM Form/genetics , Creatine Kinase, MM Form/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscle Fibers, Skeletal/cytology , Myoblasts/cytology , Polymerization , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Two-Hybrid System Techniques
4.
Dev Biol ; 382(1): 160-71, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23872330

ABSTRACT

Actin dynamics are necessary at multiple steps in the formation of multinucleated muscle cells. BAR domain proteins can regulate actin dynamics in several cell types, but have been little studied in skeletal muscle. Here, we identify novel functions for the N-BAR domain protein, Bridging integrator 3 (Bin3), during myogenesis in mice. Bin3 plays an important role in regulating myofiber size in vitro and in vivo. During early myogenesis, Bin3 promotes migration of differentiated muscle cells, where it colocalizes with F-actin in lamellipodia. In addition, Bin3 forms a complex with Rac1 and Cdc42, Rho GTPases involved in actin polymerization, which are known to be essential for myotube formation. Importantly, a Bin3-dependent pathway is a major regulator of Rac1 and Cdc42 activity in differentiated muscle cells. Overall, these data classify N-BAR domain proteins as novel regulators of actin-dependent processes in myogenesis, and further implicate BAR domain proteins in muscle growth and repair.


Subject(s)
Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Muscle Development , Neuropeptides/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Cell Movement , Endocytosis , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/deficiency , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/physiology , Protein Structure, Tertiary , Pseudopodia/metabolism , Regeneration
SELECTION OF CITATIONS
SEARCH DETAIL
...