Abstract

Regenerative medicine is a novel approach for treating conditions in which enhanced bone regeneration is required. We identified transgelin (TAGLN), a transforming growth factor beta (TGFβ)-inducible gene, as an upregulated gene during in vitro osteoblastic and adipocytic differentiation of human bone marrow-derived stromal (skeletal) stem cells (hMSC). siRNA-mediated gene silencing of TAGLN impaired lineage differentiation into osteoblasts and adipocytes but enhanced cell proliferation. Additional functional studies revealed that TAGLN deficiency impaired hMSC cell motility and in vitro transwell cell migration. On the other hand, TAGLN overexpression reduced hMSC cell proliferation, but enhanced cell migration, osteoblastic and adipocytic differentiation, and in vivo bone formation. In addition, deficiency or overexpression of TAGLN in hMSC was associated with significant changes in cellular and nuclear morphology and cytoplasmic organelle composition as demonstrated by high content imaging and transmission electron microscopy that revealed pronounced alterations in the distribution of the actin filament and changes in cytoskeletal organization. Molecular signature of TAGLN-deficient hMSC showed that several genes and genetic pathways associated with cell differentiation, including regulation of actin cytoskeleton and focal adhesion pathways, were downregulated. Our data demonstrate that TAGLN has a role in generating committed progenitor cells from undifferentiated hMSC by regulating cytoskeleton organization. Targeting TAGLN is a plausible approach to enrich for committed hMSC cells needed for regenerative medicine application.

Highlights

  • Regenerative medicine through employing stem cell transplantation is a novel approach for treating conditions in which enhanced bone regeneration is required

  • We have identified TAGLN as one out of 11 genes that were upregulated during osteogenic differentiation and adipogenic differentiation of hMSC as well as enriched in the hMSC clone 1 high osteogenic cell (CL1) cell line, which is an hMSC cell line that exhibits enhanced osteogenic and adipogenic differentiation (Figure 1a)

  • Given the known role of TGFβ signaling in regulating TAGLN expression, we subsequently assessed the effect of TGFβ treatment on TAGLN expression and hMSC differentiation

Read more

Summary

Introduction

Regenerative medicine through employing stem cell transplantation is a novel approach for treating conditions in which enhanced bone regeneration is required. Optimal use of hMSC in therapy requires detailed understanding of molecular mechanisms of lineage commitment and differentiation as well as identifying regulatory factors that can be targeted for controlling hMSC differentiation and functions. Received 13.1.16; revised 08.4.16; accepted 30.5.16; Edited by Y Shi mouse embryogenesis.[9] It is known as one of the earliest commitment markers of differentiated smooth muscle cells,[10,11,12] and has been suggested to regulate their contractile functions. TAGLN-deficient mice were fertile and developed normally;[13] vascular smooth muscle cells exhibited a pronounced alterations in the distribution of the actin filament and changes in cytoskeletal organization.[14]. Our data suggest that TAGLN is important for differentiation progression of hMSC through regulation of distribution of actin filaments and cytoskeletal organization

Objectives
Methods
Results
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