scholarly journals CCL5 secreted from bone marrow stromal cells stimulates the migration and invasion of Huh7 hepatocellular carcinoma cells via the PI3K-Akt pathway

2014 ◽  
Vol 45 (1) ◽  
pp. 333-343 ◽  
Author(s):  
HUILI BAI ◽  
YAGUANG WENG ◽  
SHUNJIE BAI ◽  
YINGJIU JIANG ◽  
BAOLIN LI ◽  
...  
2019 ◽  
Vol 53 (2) ◽  
pp. 65-70
Author(s):  
Shokoufeh Taherkhani ◽  
Fatemeh Moradi ◽  
Masoumeh Hosseini ◽  
Mohsen Alipour ◽  
Hadi Feizi

AbstractObjective. Ghrelin, a 28 amino acid peptide, has diverse physiological roles. Phosphatidylino-sitol-bisphosphate 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) are involved in some of the recognized actions of ghrelin. It has been shown that ghrelin upregulates HOXB4 gene expression but the real mechanism of this effect is not clear.Methods. Rat bone marrow stromal cells (BMSCs) were cultured in DMEM. BMSCs were treated with ghrelin (100 μM) for 48 h. Real-time PCR for HOXB4 was performed from Control (untreated BMSCs), BG (BMSCs treated with 100 µM ghrelin), PD (BMSCs treated with 10 µM PD98059, a potent inhibitor of mitogen-activated protein kinase, and 100 µM ghrelin), LY (BM-SCs treated with 10 µM LY294002, a strong inhibitor of phosphoinositide 3-kinase, and 100 µM ghrelin) and SY (BMSCs treated with 10 µM LY294002 plus 10 µM PD98059, and 100 µM ghrelin) groups. Relative gene expression changes were determined using Relative expression software tool 9 (REST 9).Results. HOXB4 gene has been overexpressed in ghrelin-treated BMSCs (p<0.05). PI3K inhi-bition by LY294002 significantly downregulated the ghrelin-induced overexpression of HOXB4 (p<0.05).Conclusion. We can conclude that ghrelin, through PI3K/Akt pathway, may improve BMSC transplantation potency by reducing its apoptosis. Moreover, upregulating HOXB4 in BMSC and its possible differentiation to HSCs might in the future open the doors to new treatment for hematologic disorders. Therefore, activating the PI3K/Akt pathway, instead of using a non-specific inducer, could be the principal point to increase the efficiency of BMSC-based cell therapies in the future.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2753-2753 ◽  
Author(s):  
Hiroshi Ikeda ◽  
Teru Hideshima ◽  
Yutaka Okawa ◽  
Sonia Vallet ◽  
Samantha Pozzi ◽  
...  

Abstract The phosphatidylinositide 3-kinase (PI3K)/Akt pathway mediates cell proliferation, cell cycle regulation, apoptosis, and autophagy. Of the 8 distinct mammalian isoforms of PI3K, it is the class I PI3Ks (p110α, p110β, p110δ, and p110γ) that are responsible for Akt activation. Recently, p110δ expression has been shown in colon and bladder carcinoma, glioblastoma, and acute myeloid leukemia cells. In this study, we demonstrate expression and examined biologic sequelae of p110δ signaling in MM using a novel specific inhibitor CAL-101. Overexpression of p110δ protein was observed in 24/24 primary tumor cells from MM patients. High level of p110δ expression was detected in two (INA-6 and LB) of the 11 MM cell lines examined. Treatment of INA-6 and LB cells with CAL-101 (1.0 μM for 1.5, 3, 6 hr) significantly inhibited phosphorylation of Akt at serine 473 and threonine 308, suggesting that p110δ is the isoform responsible for PI3K signaling in these cells. Furthermore, CAL-101 induced cytotoxicity in INA-6 cells in a dose-dependent fashion, with an IC50 of 0.625 μM-1.25 μM at 48 h. In contrast, minimal cytotoxicity was observed in p110δ-negative cell lines. Importantly, CAL-101 (0.625 μM) triggered cytotoxicity against tumor cells from MM patients without affecting the survival of normal peripheral blood mononuclear cells at concentrations as high as 20 μM. The specificity of CAL-101 towards p110δ was confirmed by the observation that p110δ small interfering RNA knock-down, but not control, caused inhibition of cell growth and cytotoxicity that was consistent with CAL-101 treatment, further confirming the key role of p110δ in MM cells. Interleukin 6 (IL-6) and insulin-like growth factor I (IGF-I) and cell-cell contact with stromal cells have been shown to induce proliferative and anti-apoptotic responses in MM cells that abrogate cytotoxic effects of conventional therapies. Importantly, neither either IL-6 and IGF-1 treatment nor co-culture of MM cells with bone marrow stromal cells abrogated CAL-101-induced MM cell growth inhibition. Moreover, CAL-101 inhibited adherence of MM cells to bone marrow stromal cells. Since the PI3K/Akt pathway is involved in autophagy, we also examined whether CAL-101 induced autophagic cell death in MM cells. CAL-101 (6 hr treatment) significantly increased LC3-II expression, assessed by western blot and immunofluorescent analyses; conversely, autophagy inhibitor 3-MA (100 μM) blocked LC3-II expression induced by CAL-101. Acridine orange staining and electron microscopic analysis to assess the relative number of autolysosomes further confirmed induction of autophagy by CAL-101. Finally, combined CAL-101 with bortezomib showed synergistic cytotoxicity against MM cells (combination index = 0.64). In conclusion, our studies showed that p110δ is a novel therapeutic target in MM and provide the basis for clinical evaluation of CAL-101 in ongoing Phase 1/2 trial in hematologic malignancies including MM.


2013 ◽  
Author(s):  
Sylvia Thiele ◽  
Alexander Rauch ◽  
Jan P Tuckermann ◽  
Lorenz C Hofbauer ◽  
Martina Rauner

Sign in / Sign up

Export Citation Format

Share Document