Abstract

BackgroundContinuous low-intensity ultrasound (cLIUS) facilitates the chondrogenic differentiation of human mesenchymal stromal cells (MSCs) in the absence of exogenously added transforming growth factor-beta (TGFβ) by upregulating the expression of transcription factor SOX9, a master regulator of chondrogenesis. The present study evaluated the molecular events associated with the signaling pathways impacting SOX9 gene and protein expression under cLIUS.MethodsHuman bone marrow-derived MSCs were exposed to cLIUS stimulation at 14 kPa (5 MHz, 2.5 Vpp) for 5 min. The gene and protein expression of SOX9 was evaluated. The specificity of SOX9 upregulation under cLIUS was determined by treating the MSCs with small molecule inhibitors of select signaling molecules, followed by cLIUS treatment. Signaling events regulating SOX9 expression under cLIUS were analyzed by gene expression, immunofluorescence staining, and western blotting.ResultscLIUS upregulated the gene expression of SOX9 and enhanced the nuclear localization of SOX9 protein when compared to non-cLIUS-stimulated control. cLIUS was noted to enhance the phosphorylation of the signaling molecule ERK1/2. Inhibition of MEK/ERK1/2 by PD98059 resulted in the effective abrogation of cLIUS-induced SOX9 expression, indicating that cLIUS-induced SOX9 upregulation was dependent on the phosphorylation of ERK1/2. Inhibition of integrin and TRPV4, the upstream cell-surface effectors of ERK1/2, did not inhibit the phosphorylation of ERK1/2 and therefore did not abrogate cLIUS-induced SOX9 expression, thereby suggesting the involvement of other mechanoreceptors. Consequently, the effect of cLIUS on the actin cytoskeleton, a mechanosensitive receptor regulating SOX9, was evaluated. Diffused and disrupted actin fibers observed in MSCs under cLIUS closely resembled actin disruption by treatment with cytoskeletal drug Y27632, which is known to increase the gene expression of SOX9. The upregulation of SOX9 under cLIUS was, therefore, related to cLIUS-induced actin reorganization. SOX9 upregulation induced by actin reorganization was also found to be dependent on the phosphorylation of ERK1/2.ConclusionsCollectively, preconditioning of MSCs by cLIUS resulted in the nuclear localization of SOX9, phosphorylation of ERK1/2 and disruption of actin filaments, and the expression of SOX9 was dependent on the phosphorylation of ERK1/2 under cLIUS.

Highlights

  • Continuous low-intensity ultrasound facilitates the chondrogenic differentiation of human mesenchymal stromal cells (MSCs) in the absence of exogenously added transforming growth factor-beta (TGFβ) by upregulating the expression of transcription factor Sex determining region Y-box 9 (SOX9), a master regulator of chondrogenesis

  • No significant difference in the expression of osteogenic markers (RUNX2, COL1A1) and adipogenic markers (CEBPA, PPARγ) in MSCs was observed following 10 days of Continuous low-intensity ultrasound (cLIUS) (5 MHz) stimulation whereas chondrogenic markers (SOX9 and COL2A1) remained significantly elevated under cLIUS stimulation when compared to non-cLIUS-stimulated controls (Additional file 1: Figure S1)

  • In summary, our study identified the phosphorylation of Extracellular signal-regulated kinase 1 and 2 (ERK1/2), increased nuclear localization of the SOX9 protein, and disrupted actin as the events mediating increased SOX9 gene expression under cLIUS

Read more

Summary

Introduction

Continuous low-intensity ultrasound (cLIUS) facilitates the chondrogenic differentiation of human mesenchymal stromal cells (MSCs) in the absence of exogenously added transforming growth factor-beta (TGFβ) by upregulating the expression of transcription factor SOX9, a master regulator of chondrogenesis. The clinical outcomes of current strategies of cartilage repair autologous chondrocyte implantation (ACI) or matrix-assisted autologous chondrocyte implantation (MACI) are compromised by the phenotypic instability of expanded autologous chondrocytes ex vivo [3, 4] that leads to graft hypertrophy [5] and the formation of a mechanically inferior tissue in vivo. Taking cues from the in vivo regulation of MSC chondrogenesis, current in vitro protocols include select growth factors (i.e., TGFβ) for differentiation of MSCs [6]. Chondroinductive protocols that do not rely on growth factors are of interest for the eventual development of ex vivo differentiation protocols for ACI and in situ repair strategies like microfracture

Objectives
Methods
Results
Discussion
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