Published in

Wiley, FEBS Journal, 19(281), p. 4467-4478, 2014

DOI: 10.1111/febs.12955

Links

Tools

Export citation

Search in Google Scholar

Lysophosphatidic acid stimulates cell migration of satellite cells. A role for the sphingosine kinase/sphingosine 1-phosphate axis

This paper is available in a repository.
This paper is available in a repository.

Full text: Download

Green circle
Preprint: archiving allowed
Orange circle
Postprint: archiving restricted
Red circle
Published version: archiving forbidden
Data provided by SHERPA/RoMEO

Abstract

Regulation of motility of skeletal muscle precursor cells, such as satellite cells, is critically important for their proper recruitment at the site of tissue damage and ultimately for its correct repair. Here we show that lysophosphatidic acid (LPA), well-recognized as powerful bioactive agent, strongly stimulates cell migration of activated murine satellite cells. The biological effect exerted by LPA was found to be induced via the activation of LPA1 and LPA3, being abolished by cell treatment with the antagonist Ki16425, and severely impaired by siRNA-mediated down-regulation of the two receptor isoforms. On the contrary, silencing of LPA2 potentiated the stimulation of cell motility by LPA, suggesting that it is negatively coupled to cell migration. Notably, pharmacological inhibition of both sphingosine kinase (SK) isoforms with VPC96047 or the selective blocking of SK1 with VPC96091 abolished cell responsiveness to LPA; in agreement, gene silencing of SK1 or SK2 strikingly reduced the biological effect of LPA. Moreover, the LPA-dependent stimulation of cell chemotaxis was found to be impaired by down-regulation of S1P1 or S1P4 by specific siRNA. In summary, the obtained results support the notion that SK/sphingosine 1-phosphate (S1P) axis is critically implicated in the mechanism by which LPA elicits its pro-migratory action. This study provides new compelling information on the regulatory mechanisms of satellite cell motility and reinforces the notion that SK/S1P signaling pathway plays a crucial role in the control of skeletal muscle precursor cell biology.This article is protected by copyright. All rights reserved.