Abstract

Appropriate cellular signaling is essential to control cell proliferation, differentiation, and cell death. Aberrant signaling can have devastating consequences and lead to disease states, including cancer. The transforming growth factor-β (TGF-β) signaling pathway is a prominent signaling pathway that has been tightly regulated in normal cells, whereas its deregulation strongly correlates with the progression of human cancers. The regulation of the TGF-β signaling pathway involves a variety of physiological regulators. Many of these molecules act to alter the activity of Smad proteins. In contrast, the number of molecules known to affect the TGF-β signaling pathway at the receptor level is relatively low, and there are no known direct modulators for the TGF-β type II receptor (TβRII). Here we identify SPSB1 (a Spry domain-containing Socs box protein) as a novel regulator of the TGF-β signaling pathway. SPSB1 negatively regulates the TGF-β signaling pathway through its interaction with both endogenous and overexpressed TβRII (and not TβRI) via its Spry domain. As such, TβRII and SPSB1 co-localize on the cell membrane. SPSB1 maintains TβRII at a low level by enhancing the ubiquitination levels and degradation rates of TβRII through its Socs box. More importantly, silencing SPSB1 by siRNA results in enhanced TGF-β signaling and migration and invasion of tumor cells.

Highlights

  • TGF-␤ signaling is tightly controlled by different regulators along its signaling cascade

  • The regulation of the TGF-␤ signaling pathway involves a variety of physiological regulators

  • SPSB1 and SPSB2 have been shown to interact with inducible nitric-oxide synthase, targeting it for ubiquitination and proteasomal degradation and implicating the SPSB proteins in regulating the host response to infection [37,38,39]

Read more

Summary

Background

TGF-␤ signaling is tightly controlled by different regulators along its signaling cascade. SPSB1 negatively regulates the TGF-␤ signaling pathway through its interaction with both endogenous and overexpressed T␤RII (and not T␤RI) via its Spry domain. TGF-␤ type II receptor (T␤RII) or the negative regulator Smad as a transgene in mice resulted in tumor formation [8, 9], suggesting that complete blocking of the TGF-␤ signaling is not necessary for tumor formation. The TGF-␤ type III (T␤RIII) is an accessory, non-signaling receptor In vitro, it had no effect on the T␤RI- and T␤RII-mediated signaling activation per se. The activated ligand-receptor complex binds and phosphorylates the intracellular signaling molecules Smad and Smad through T␤RI [1, 12] Once phosphorylated, these regulatory Smads (R-Smad) form complexes with Smad ( called Dpc for deleted in pancreatic carcinoma locus 4 or common Smad) and translocate into the nucleus.

The abbreviations used are
Experimental Procedures
Results
Discussion
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