Both Stat3-Activation and Stat3-Independent BCL2 Downregulation Are Important for Interleukin-6–Induced Apoptosis of 1A9-M Cells

Blood ◽  
1999 ◽  
Vol 93 (4) ◽  
pp. 1346-1354 ◽  
Author(s):  
Kenji Oritani ◽  
Yoshiaki Tomiyama ◽  
Paul W. Kincade ◽  
Keisuke Aoyama ◽  
Takafumi Yokota ◽  
...  

Abstract A unique subclone of a bone marrow-derived stromal cell line, BMS2.4, produces soluble factors that inhibit proliferation of several types of hematopoietic cell lines. An understanding of these molecules may be informative about negative regulatory circuits that can potentially limit blood cell formation. We used expression cloning to identify interleukin-6 (IL-6) as one factor that suppressed growth of a pre-B–cell variant line, 1A9-M. Moreover, IL-6 induced macrophage-differentiation and apoptosis of 1A9-M cells. During this process, IL-6 downregulated expression of BCL2 in 1A9-M cells and stimulated BCL-XL expression, but had no effect on p53, Bax, or Bak gene expression. Mechanisms for transduction of IL-6–induced signals were then evaluated in IL-6–stimulated 1A9-M cells. Whereas the signal transducer and activator of transcription 3 (Stat3) was phosphorylated and activated, there was no effect on either Stat1 or Stat5. The importance of BCL2 and Stat3 on IL-6–induced macrophage-differentiation and apoptosis was studied with 1A9-M cells expressing human BCL2 or a dominant-negative form of Stat3, respectively. IL-6–induced apoptosis, but not macrophage-differentiation, was blocked by continuously expressed BCL2. A dominant-negative form of Stat3 inhibited both macrophage-differentiation and apoptosis induced by IL-6. However, diminished Stat3 activity did not prevent IL-6–induced downregulation of the BCL2 gene. Therefore, activation of Stat3 is essential for IL-6–induced macrophage-differentiation and programmed cell death in this model. Whereas overexpression of BCL2 abrogates the apoptotic response, Stat3-independent signals appear to downregulate expression of the BCL2 gene.

Blood ◽  
1999 ◽  
Vol 93 (4) ◽  
pp. 1346-1354 ◽  
Author(s):  
Kenji Oritani ◽  
Yoshiaki Tomiyama ◽  
Paul W. Kincade ◽  
Keisuke Aoyama ◽  
Takafumi Yokota ◽  
...  

A unique subclone of a bone marrow-derived stromal cell line, BMS2.4, produces soluble factors that inhibit proliferation of several types of hematopoietic cell lines. An understanding of these molecules may be informative about negative regulatory circuits that can potentially limit blood cell formation. We used expression cloning to identify interleukin-6 (IL-6) as one factor that suppressed growth of a pre-B–cell variant line, 1A9-M. Moreover, IL-6 induced macrophage-differentiation and apoptosis of 1A9-M cells. During this process, IL-6 downregulated expression of BCL2 in 1A9-M cells and stimulated BCL-XL expression, but had no effect on p53, Bax, or Bak gene expression. Mechanisms for transduction of IL-6–induced signals were then evaluated in IL-6–stimulated 1A9-M cells. Whereas the signal transducer and activator of transcription 3 (Stat3) was phosphorylated and activated, there was no effect on either Stat1 or Stat5. The importance of BCL2 and Stat3 on IL-6–induced macrophage-differentiation and apoptosis was studied with 1A9-M cells expressing human BCL2 or a dominant-negative form of Stat3, respectively. IL-6–induced apoptosis, but not macrophage-differentiation, was blocked by continuously expressed BCL2. A dominant-negative form of Stat3 inhibited both macrophage-differentiation and apoptosis induced by IL-6. However, diminished Stat3 activity did not prevent IL-6–induced downregulation of the BCL2 gene. Therefore, activation of Stat3 is essential for IL-6–induced macrophage-differentiation and programmed cell death in this model. Whereas overexpression of BCL2 abrogates the apoptotic response, Stat3-independent signals appear to downregulate expression of the BCL2 gene.


1994 ◽  
Vol 180 (6) ◽  
pp. 2413-2418 ◽  
Author(s):  
R P Bissonnette ◽  
A McGahon ◽  
A Mahboubi ◽  
D R Green

T cell hybridomas respond to activation signals by undergoing apoptotic cell death, and this is likely to represent comparable events related to tolerance induction in immature and mature T cells in vivo. Previous studies using antisense oligonucleotides implicated the c-Myc protein in the phenomenon of activation-induced apoptosis. This role for c-Myc in apoptosis is now confirmed in studies using a dominant negative form of its heterodimeric binding partner, Max, which we show here inhibits activation-induced apoptosis. Further, coexpression of a reciprocally mutant Myc protein capable of forming functional heterodimers with the mutant Max can compensate for the dominant negative activity and restore activation-induced apoptosis. These results imply that Myc promotes activation-induced apoptosis by obligatory heterodimerization with Max, and therefore, by regulating gene transcription.


2006 ◽  
Vol 26 (12) ◽  
pp. 4474-4488 ◽  
Author(s):  
Takashi Yano ◽  
Kosei Ito ◽  
Hiroshi Fukamachi ◽  
Xin-Zi Chi ◽  
Hee-Jun Wee ◽  
...  

ABSTRACTGenes involved in the transforming growth factor β (TGF-β) signaling pathway are frequently altered in several types of cancers, and a gastric tumor suppressorRUNX3appears to be an integral component of this pathway. We reported previously that apoptosis is notably reduced inRunx3−/−gastric epithelial cells. In the present study, we show that a proapoptotic geneBimwas transcriptionally activated by RUNX3 in the gastric cancer cell lines SNU16 and SNU719 treated with TGF-β. The humanBimpromoter contains RUNX sites, which are required for its activation. Furthermore, a dominant negative form of RUNX3 comprised of amino acids 1 to 187 increased tumorigenicity of SNU16 by inhibiting Bim expression. InRunx3−/−mouse gastric epithelium, Bim was down-regulated, and apoptosis was reduced to the same extent as that inBim−/−gastric epithelium. We confirmed comparable expression of TGF-β1 and TGF-β receptors between wild-type andRunx3−/−gastric epithelia and reduction of Bim inTGF-β1−/−stomach. These results demonstrate that RUNX3 is responsible for transcriptional up-regulation ofBimin TGF-β-induced apoptosis.


Sign in / Sign up

Export Citation Format

Share Document