Your browser doesn't support javascript.
loading
Actin-organizing protein palladin modulates C2C12 cell fate determination.
Nguyen, Ngoc Uyen Nhi; Hsu, Ching-Cheng; Ali, Shah R; Wang, Hao-Ven.
Affiliation
  • Nguyen NUN; Department of Life Sciences, National Cheng Kung University, 1 University Road, Tainan, 70101, Taiwan.
  • Hsu CC; Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA.
  • Ali SR; Institute of Basic Medical Science, National Cheng Kung University, 1 University Road, Tainan, 70101, Taiwan.
  • Wang HV; Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA.
Biochem Biophys Rep ; 39: 101762, 2024 Sep.
Article de En | MEDLINE | ID: mdl-39026565
ABSTRACT

Background:

Cell confluency and serum deprivation promote the transition of C2C12 myoblasts into myocytes and subsequence fusion into myotubes. However, despite all myoblasts undergoing the same serum deprivation trigger, their responses vary whether they become founder myocytes, remain proliferative, or evolve into fusion-competent myocytes remains unclear. We have previously shown that depletion of the scaffolding protein palladin in myoblasts inhibits cell migration and promotes premature muscle differentiation, pointing to its potential significance in muscle development and the necessity for a more in-depth examination of its function in cellular heterogeneity. Methods and

results:

Here, we showed that the subcellular localization of palladin might contribute to founder-fate cell decision in the early differentiation process. Depleting palladin in C2C12 myoblasts depleted integrin-ß3 plasma membrane localization of and focal adhesion formation at the early stage of myogenesis, decreased kindlin-2 and metavinculin expression during the myotube maturation process, leading to the inability of myocytes to fuse into preexisting mature myotubes. This aligns with previous findings where early differentiation into nascent myotubes occurred but compromised maturation. In contrast, wildtype C2C12 overexpressing the 140-kDa palladin isoform developed a polarized morphology with star-like structures toward other myoblasts. However, this behaviour was not observed in palladin-depleted cells, where the 140-kDa palladin overexpression could not recover cell migration capacity, suggesting other palladin isoforms are also needed to establish cell polarity.

Conclusion:

Our study identifies a counter-intuitive role for palladin in regulating myoblast-to-myocyte cell fate decisions and impacting their ability to form mature multinucleated myotubes by influencing cell signalling pathways and cytoskeletal organization, necessary for skeletal muscle regeneration and repair studies.
Mots clés

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Langue: En Journal: Biochem Biophys Rep Année: 2024 Type de document: Article Pays d'affiliation: Taïwan

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Langue: En Journal: Biochem Biophys Rep Année: 2024 Type de document: Article Pays d'affiliation: Taïwan