Role for interleukin-6 and insulin-like growth factor-I via PI3-K/Akt pathway in the proliferation of CD56− and CD56+ multiple myeloma cells

2006 ◽  
Vol 34 (6) ◽  
pp. 736-744 ◽  
Author(s):  
Naohi Sahara ◽  
Akihiro Takeshita ◽  
Takaaki Ono ◽  
Yuya Sugimoto ◽  
Miki Kobayashi ◽  
...  
2002 ◽  
Vol 278 (8) ◽  
pp. 5794-5801 ◽  
Author(s):  
Klaus Podar ◽  
Yu-Tzu Tai ◽  
Craig E. Cole ◽  
Teru Hideshima ◽  
Martin Sattler ◽  
...  

2008 ◽  
Vol 141 (4) ◽  
pp. 470-482 ◽  
Author(s):  
Patricia Maiso ◽  
Enrique M. Ocio ◽  
Mercedes Garayoa ◽  
Juan C. Montero ◽  
Francesco Hofmann ◽  
...  

Blood ◽  
2004 ◽  
Vol 103 (6) ◽  
pp. 2291-2298 ◽  
Author(s):  
Saeid Abroun ◽  
Hideaki Ishikawa ◽  
Naohiro Tsuyama ◽  
Shangqin Liu ◽  
Fu-Jun Li ◽  
...  

Abstract Interleukin-6 (IL-6) is a growth and antiapoptotic factor for human myeloma cells. The autocrine loop and increased expression of the growth factor receptors have been postulated as the mechanisms of tumorigenesis. Here we show that IL-6 stimulation induced the phosphorylation of insulin-like growth factor-I (IGF-I) receptors in a human myeloma cell line, NOP2, highly expressing IL-6 receptor α (IL-6Rα) and in the IL-6Rα–transfected U266 cell line. IL-6–dependent complex formation of IL-6Rα with IGF-I receptor β was found in NOP2 where IL-6Rα colocalized with IGF-I receptors at lipid rafts. Moreover, the IL-6–induced phosphorylation of IGF-I receptor β was not blocked by a Janus kinase 2 (Jak2) inhibitor. In addition to the activation of the signal transducer and activator of transcription 3 and extracellular signal-regulated kinase 1/2, IL-6 stimulation led to the activation of Akt, presumably following the phosphorylation of IGF-I receptors. Thus, our results suggest that in NOP2, IL-6Rα and IGF-I receptors exist on the plasma membrane in close proximity, facilitating the efficient assembly of 2 receptors in response to IL-6. The synergistic effects of highly expressed IL-6Rα on IGF-I receptor–mediated signals provide a novel insight into the Jak-independent IL-6 signaling mechanism of receptor cross-talk in human myeloma cells.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3365-3365
Author(s):  
Naohi Sahara ◽  
Akihiro Takeshita ◽  
Miki Kobayashi ◽  
Kazuyuki Shigeno ◽  
Satoki Nakamura ◽  
...  

Abstract Several studies including ours have suggested that lack of CD56 expression in multiple myeloma (MM) defines a unique patient subset with poorer prognosis. However, the mechanism underlying this aggressive behavior of CD56− MM has not been well elucidated. In this study, we sorted out both CD56− and CD56+ fractions from MM cell lines or patients with MM, and investigated their different responsiveness to interleukin-6 (IL-6) or insulin like growth factor-I (IGF-I), and tried to clarify the course of action in cell cycle distribution. After stained with PE-CD56, CD56− and CD56+ fractions in KMS-21-BM and U-266 cell lines were isolated by the cell sorter, and cultured separately either in the presence or absence of IL-6 (2 ng/ml and 10 ng/ml, respectively). Although CD56− cells in both KMS-21-BM and U-266 cell lines responded significantly to IL-6 (P=0.001 and 0.009, respectively), CD56+ cells did not. Ki-67+ cells in CD56− KMS-21-BM cells, that were significantly fewer than that in CD56+ ones (P=0.0003), increased significantly upon 24-hour incubation with IL-6 (P<0.0001). Western blotting analysis showed that the level of cyclin D1 and p27 protein in CD56− KMS-21-BM cells were up- and down-regulated by IL-6 in a time dependent manner, respectively. IL-6 also brought phosphorylation of Akt (ser473) in the CD56− cells. LY-294002 completely blocked these effects of IL-6. On the other hand, Ki-67+ cells in the CD56+ cells did not respond to IL-6. Although IGF-I did not increase Ki-67+ cells either in the CD56− and CD56+ cells from KMS-21-BM, anti-IGF-I mAb significantly reduced Ki-67+ cells only in the CD56+ cells (P=0.006). IGF-I up-regulated the level of cyclin D1 and phosphorylated Akt in CD56+ KMS-21-BM cells. LY294002 completely blocked these effects of IGF-I. Same results were obtained in the analysis of U-266 cell lines. The MM cells sorted from 17 patients with MM were also examined for CD56 and Ki-67 expression. Four and 13 patients were distributed to the CD56− and CD56+ group, respectively. These MM cells from the patients were cultured with or without IL-6 (10 ng/ml) or IGF-I (500ng/ml) for 24 hours. IL-6 increased the percentage of Ki-67+ cells in the CD56− group more than those in the CD56+ group (P=0.007). Although MM cells did not respond to IL-6, IGF-I significantly increased Ki-67+ cells in the CD56+ group (P=0.005). These results suggest that CD56− and CD56+ MM cells could be stimulated by different cytokines. We here found that CD56− MM cells were proliferated more than CD56+ MM cells in the presence of IL-6, and that this effect of IL-6 was mainly mediated by the activation of PI3-K/Akt pathway. In addition, our results suggest that IGF-I play an important role in the proliferation of CD56+ MM cells via PI3-K/Akt pathway.


Blood ◽  
2004 ◽  
Vol 103 (1) ◽  
pp. 301-308 ◽  
Author(s):  
Ya-Wei Qiang ◽  
Lei Yao ◽  
Giovanna Tosato ◽  
Stuart Rudikoff

Abstract Multiple myeloma (MM) is an incurable form of cancer characterized by accumulation of malignant plasma cells in the bone marrow. During the course of this disease, tumor cells cross endothelial barriers and home to the bone marrow. In latter stages, myeloma cells extravasate through blood vessels and may seed a variety of organs. Insulin-like growth factor I (IGF-I) is one of several growth factors shown to promote the growth of MM cells. In the current study, we have assessed the ability of IGF-I to serve additionally as a chemotactic factor affecting the mobility and invasive properties of these cells. Results indicate that IGF-I promotes transmigration through vascular endothelial cells and bone marrow stromal cell lines. Analysis of endogenous signaling pathways revealed that protein kinase D/protein kinase Cμ (PKD/PKCμ) and RhoA were both activated in a phosphatidylinositol 3-kinase (PI-3K)–dependent manner. Inhibition of PI-3K, PKCs, or Rho-associated kinase by pharmacologic inhibitors abrogated migration, whereas mitogen-activated protein kinase (MAPK), Akt, and p70S6 kinase inhibitors had no effect. These results suggest that IGF-I promotes myeloma cell migration by activation of PI-3K/PKCμ and PI-3K/RhoA pathways independent of Akt. The identification of IGF-I as both a proliferative and migratory factor provides a rational basis for the development of targeted therapeutic strategies directed at IGF-I in the treatment of MM.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1513-1513
Author(s):  
Shori Abe ◽  
Hideo Harigae ◽  
Tadao Funato ◽  
Junichi Kameoka ◽  
Shinichiro Takahashi ◽  
...  

Abstract Cytosine arabinoside (Ara-C) is one of the most common drugs used in the treatment of acute myeloid leukemia (AML). Therefore, the resistance to Ara-C is a major obstacle to improvement of the cure rate of AML. However, the molecular mechanism for Ara-C resistance has not been fully understood. In order to investigate the mechanism involved in Ara-C resistance, the gene expression profile of Ara-C-resistant human K562 leukemia cells (K562/AC cells) was compared with that of the Ara-C-sensitive K562 parental cells by using a cDNA microarray platform. Correspondence analysis demonstrated that the insulin-like growth factor I (IGF-I) gene was significantly upregulated in K562/AC cells. The increased expression of IGF-I in K562/AC cells was confirmed by quantitative RT-PCR (RQ-PCR). We then studied the biological significance of IGF-I upregulation in Ara-C resistance. First, K562/AC cells were treated by suramin, a nonspecific growth factor antagonist. The treatment with suramine reduced viability and induced apoptosis of K562/AC cells. Furthermore, the addition of IGF-I neutralizing antibody exhibited the same effect as suramine. Moreover, the IC50 of Ara-C sensitive K562 parental cells was increased by the addition of exogenous IGF-I ligand. These results strongly suggest that IGF-I-IGF-I-R pathway is directly involved in Ara-C resistance. Next, the activation status of phosphatidylinositol 3-kinase (PI3-kinase)/Akt pathway, which is the downstream of IGF-I-IGFI-R signal, in K562/AC cells was examined. Western blot analysis showed that the phosphorylation level of Akt is higher in K562/AC cells than K562 sensitive cells. In addition, LY294002, the specific inhibitor for PI3-kinase reduced the survival of K562/AC cells. Since activation of PI3-kinase/Akt pathway has been shown to exhibit an anti-apoptotic effect in tumor cells, it is possible that IGF-I induces Ara-C resistance by escaping from apoptosis through PI3-kinase/Akt pathway in an autocrine or paracrine fashion. Finally, the expression level of IGF-I was examined in 27 clinical samples by RQ-PCR. The results showed that IGF-I expression level was higher in leukemic cells in refractory or relapsed AML patients, who received Ara-C combined chemotherapy, than that at diagnosis. These results suggest that the inhibition of the IGF-I-IGF-I receptor pathway is a valuable therapeutic approach to overcome Ara-C resistance in AML.


Sign in / Sign up

Export Citation Format

Share Document