Adenovirus-Mediated Transmission of a Dominant Negative Transforming Growth Factor-β Receptor Inhibits In Vitro Mouse Cranial Suture Fusion

2002 ◽  
Vol 110 (2) ◽  
pp. 506-514 ◽  
Author(s):  
Babak J. Mehrara ◽  
Jason A. Spector ◽  
Joshua A. Greenwald ◽  
Hikari Ueno ◽  
Michael T. Longaker
Blood ◽  
2002 ◽  
Vol 99 (9) ◽  
pp. 3179-3187 ◽  
Author(s):  
Catherine M. Bollard ◽  
Claudia Rössig ◽  
M. Julia Calonge ◽  
M. Helen Huls ◽  
Hans-Joachim Wagner ◽  
...  

Abstract Transforming growth factor β (TGF-β), a pleiotropic cytokine that regulates cell growth and differentiation, is secreted by many human tumors and markedly inhibits tumor-specific cellular immunity. Tumors can avoid the differentiating and apoptotic effects of TGF-β by expressing a nonfunctional TGF-β receptor. We have determined whether this immune evasion strategy can be manipulated to shield tumor-specific cytotoxic T lymphocytes (CTLs) from the inhibitory effects of tumor-derived TGF-β. As our model we used Epstein-Barr virus (EBV)–specific CTLs that are infused as treatment for EBV-positive Hodgkin disease but that are vulnerable to the TGF-β produced by this tumor. CTLs were transduced with a retrovirus vector expressing the dominant-negative TGF-β type II receptor HATGF-βRII-Δcyt. HATGF-βRII-Δcyt– but not green fluorescence protein (eGFP)–transduced CTLs was resistant to the antiproliferative and anticytotoxic effects of exogenous TGF-β. Additionally, receptor-transduced cells continued to secrete cytokines in response to antigenic stimulation. TGF-β receptor ligation results in phosphorylation of Smad2, and this pathway was disrupted in HATGF-βRII-Δcyt–transduced CTLs, confirming blockade of the signal transduction pathway. Long-term expression of TGF-βRII-Δcyt did not affect CTL function, phenotype, or growth characteristics. Tumor-specific CTLs expressing HATGF-βRII-Δcyt should have a selective functional and survival advantage over unmodified CTLs in the presence of TGF-β–secreting tumors and may be of value in treatment of these diseases.


Hepatology ◽  
2009 ◽  
Vol 50 (5) ◽  
pp. 1494-1500 ◽  
Author(s):  
Katsunori Yoshida ◽  
Guo-Xiang Yang ◽  
Weici Zhang ◽  
Masanobu Tsuda ◽  
Koichi Tsuneyama ◽  
...  

2002 ◽  
Vol 195 (10) ◽  
pp. 1247-1255 ◽  
Author(s):  
Tania Fernandez ◽  
Stephanie Amoroso ◽  
Shellyann Sharpe ◽  
Gary M. Jones ◽  
Valery Bliskovski ◽  
...  

Transforming growth factor (TGF)-β is the prototype in a family of secreted proteins that act in autocrine and paracrine pathways to regulate cell development and function. Normal cells typically coexpress TGF-β receptors and one or more isoforms of TGF-β, thus the synthesis and secretion of TGF-β as an inactive latent complex is considered an essential step in regula-ting the activity of this pathway. To determine whether intracellular activation of TGF-β results in TGF-β ligand–receptor interactions within the cell, we studied pristane-induced plasma cell tumors (PCTs). We now demonstrate that active TGF-β1 in the PCT binds to intracellular TGF-β type II receptor (TβRII). Disruption of the expression of TGF-β1 by antisense TGF-β1 mRNA restores localization of TβRII at the PCT cell surface, indicating a ligand-induced impediment in receptor trafficking. We also show that retroviral expression of a truncated, dominant-negative TβRII (dnTβRII) effectively competes for intracellular binding of active ligand in the PCT and restores cell surface expression of the endogenous TβRII. Analysis of TGF-β receptor–activated Smad2 suggests the intracellular ligand–receptor complex is not capable of signaling. These data are the first to demonstrate the formation of an intracellular TGF-β–receptor complex, and define a novel mechanism for modulating the TGF-β signaling pathway.


2006 ◽  
Vol 66 (20) ◽  
pp. 9878-9885 ◽  
Author(s):  
Claudia D. Andl ◽  
Brenton B. Fargnoli ◽  
Takaomi Okawa ◽  
Mark Bowser ◽  
Munenori Takaoka ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document