MAGE-C1/CT7 Inhibition Increases Myeloma Cell Line Sensitivity to Bortezomib-Induced Apoptosis: New Target for Myeloma Therapy?

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1908-1908
Author(s):  
Fabricio de Carvalho ◽  
Erico T. Costa ◽  
Anamaria A. Camargo ◽  
Juliana C. Gregorio ◽  
Cibele Masotti ◽  
...  

Abstract Abstract 1908 Introduction: MAGE-C1/CT7 encodes for a cancer/testis antigen (CTA) frequently expressed in multiple myeloma (MM) that may be a potential target for immunotherapy in this still incurable disease. The expression of this CTA is restricted to malignant plasma cells and a positive correlation between MAGEC1/CT7 expression and advanced stage has been demonstrated for MM. It has been suggested that MAGE-C1/CT7 might play a pathogenic role in MM; however, the exact function of this protein in the pathophysiology of MM is not yet understood. Objectives: (1) To clarify the role of MAGE-C1/CT7 in the control of cellular proliferation and cell cycle regulation in myeloma cell line SKO-007 and (2) to evaluate the impact of silencing MAGE-C1/CT7 on cells treated with bortezomib. Material and Methods: Short hairpin RNA (shRNA) specific for MAGE-C1/CT7 was inserted in the pRETROSUPER(pRS) retroviral vector. The pRS-shRNA-MAGE-C1/CT7 was co-transfected with pCL-amphotropic packing vector in 293T cells to produce virus particles. Sko-007 myeloma cell line was transduced for stable expression of shRNA-MAGE-C1/CT7. Downregulation of MAGE-C1/CT7 was confirmed by real time PCR (RQ-PCR) and western blot. Functional studies included cell proliferation, cell cycle analysis using propidium iodide, and analysis of apoptosis using annexin V staining. Results: SKO-007 MM cell line was transduced for stable expression of shRNA-MAGE-C1/CT7. SKO-007 cells were divided into three derivatives: empty vector (pRS) and ineffective shRNA (antisense strand deleted – GC bases) [both used as controls for all the experiments] and inhibited (shMAGE-C1/CT7). MAGE-C1/CT7 mRNA expression was ∼5 times lower in inhibited cell line than control cells by RQ-PCR. Western blot showed 70–80% decrease in MAGE-C1/CT7 protein expression in inhibited cells when compared with controls. Functional assays did not indicate a difference in cell proliferation and DNA synthesis when inhibited cells were compared with controls. We used empty vector, ineffective shRNA and inhibited cells to determine whether inhibition of MAGE-C1/CT7 was associated with cell cycle dysregulation. We detected differences between inhibited cells and both controls regarding the proportion of myeloma cells in the G2/M phase (p<0.05). When inhibited cells and controls were treated with 10 nM bortezomib for 48h, inhibited cells showed a 48% reduction of cells in the G2/M phase but control cells have 11% (empty vector) and 10% (ineffective shRNA) of reduction (p<0.05). Inhibited cells treated with 15 nM bortezomib showed an increased percentage of apoptotic cells in comparison with bortezomib treated controls (p<0.01) [Figure]. Conclusions: MAGE-C1/CT7 antigen inhibition did not change cell proliferation and DNA synthesis in SKO-007 cells. However, we found that MAGE-C1/CT7 plays in cell cycle regulation, protecting SKO-007 cells against bortezomib-induced apoptosis. Therefore, MAGE-C1/CT7 silencing by shRNA could be a strategy for future therapies in MM, i.e. in combination with proteasome inhibitors. [Supported by CNPq and LICR] Disclosures: No relevant conflicts of interest to declare.

1999 ◽  
Vol 78 (1) ◽  
pp. 13-18 ◽  
Author(s):  
M. T. Petrucci ◽  
M. R. Ricciardi ◽  
C. Ariola ◽  
C. Gregorj ◽  
M. Ribersani ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 5017-5017
Author(s):  
Miki Nakamura ◽  
Hiroyasu Esumi ◽  
Jie Lu ◽  
Hiroaki Mitsuya ◽  
Hiroyuki Hata

Abstract Introduction Kigamicin (KM) is a compound isolated from Actinomycetes, which reportedly induces necrosis in pancreatic cancer cells under nutrient-starving condition but not under nutrient rich condition via PI3-kinase inhibition (Lu et al., Cancer Science 95, 547–52, 2004). Although the PI3-kinase activity is thought to be critical in the growth of myeloma cells, its actual role remains to be determined. In the present study, we evaluated KM’s anti-myeloma activity in both laboratory and primary myeloma cells and found that, contrary to the original finding in pancreatic cancer cells, KM induced necrosis in human myeloma cells both under nutrient-starving and nutrient-rich conditions. Results and discussion Myeloma cell lines (12PE and KHM-11) and primary myeloma cells purified with CD138-coated immune-magnetic beads were incubated with KM under nutrient-rich conditions. The CC50 value of KM for myeloma cells was approximately 100 nM after 24-hour exposure while pancreatic cancer cell line, PANC-1, did not show inhibition of viability even at 10 mM under nutrient-rich conditions, suggesting high sensitivity of myeloma cells to KM. When whole mononuclear cells obtained from a myeloma patient’s bone marrow were cultured with KM at a concentration of 500 nM in vitro, normal lymphocytes were spared while all myeloma cells underwent necrosis, suggesting that preferential cytotoxicity of KM in myeloma cells. Western blot analysis revealed that AKT phosphorylation decreased with KM treatment, suggesting that KM inhibits PI-3 kinase activity as previously reported. However, another pan-PI3 kinase inhibitor, LY294002, did not induce necrosis in myeloma cells, suggesting that PI3-kinase inhibition is not critically related to the cytotoxicity of KM in myeloma cells. A pan-caspase inhibitor, ZVAD-FMK, only partially inhibited cell death, suggesting that caspase is not involved in the cytotoxic function of KM, either. To further determine the mechanism of cytotoxicity in myeloma cells, a possible involvement of cyclin D1 and p21 was also examined. Western blot analysis revealed that KM completely reduced cyclin-D1 in myeloma cells. Moreover, KM induced translocation of p21 from cytoplasm to nucleus within 5 hours treatment, suggesting that KM disrupted cell cycle regulation. Finally, melphalan-resistant myeloma cell line, 11-EMS, showed significant cell death when exposed to KM even more efficiently than did melphalan-sensitive parental cell line, KHM-11. Since a number of anti-cancer reagents induce apoptosis in myeloma cells, KM induction of necrosis may represent a unique mechanism(s) and may overcome drug resistance of myeloma cells. Taking into a consideration a recent report by Lu et al. (Cancer Science 95, 547–52, 2004) showing that KM’s safe usage in a murine pancreatic cancer xenograft model, the present data suggest that KM could be a potential therapeutic agent for treatment of myeloma and warrant that further preclinial development of KM be continued.


FEBS Letters ◽  
2006 ◽  
Vol 580 (5) ◽  
pp. 1205-1214 ◽  
Author(s):  
Benyam Asefa ◽  
Jonathan M. Dermott ◽  
Philipp Kaldis ◽  
Karen Stefanisko ◽  
David J. Garfinkel ◽  
...  

BMC Cancer ◽  
2006 ◽  
Vol 6 (1) ◽  
Author(s):  
Osamu Hashimoto ◽  
Masako Shinkawa ◽  
Takuji Torimura ◽  
Toru Nakamura ◽  
Karuppaiyah Selvendiran ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3444-3444
Author(s):  
Magne Rekvig ◽  
Anne-Tove Brenne ◽  
Torstein Baade Ro ◽  
Anders Waage ◽  
Magne Borset ◽  
...  

Abstract Multiple myeloma has two distinct features: Expansion of malignant plasma cells within the bone marrow accompanied by skeletal destruction. Bone morphogenetic proteins (BMPs) have been shown to induce apoptosis and inhibit growth in myeloma cells. BMPs are members of the TGF-β superfamily of proteins capable of inducing bone formation, and regulate proliferation, differentiation and apoptosis. We have investigated myeloma cell apoptosis and proliferation with BMP-4 and −6 in concert with the myeloma cell growth factors interleukin (IL)-2, IL-6, IL-10, IL-15, IL-21, tumor necrosis factor (TNF)-α and insulin-like growth factor (IGF)-1. Eight samples of highly purified myeloma cells from patients and a human myeloma cell line, IH-1 (Brenne AT et al. Blood. 2002 May 15;99(10):3756–62.), were used in this study. Cytokine concentrations used in the referred experiments were for BMP-4 20ng/ml, BMP-6 250ng/ml, IL-15 20ng/ml and IL-6 0,1ng/ml, respectively. Growth inhibition was measured in a proliferation assay by methyl-[3H]-thymidine incorporation and apoptosis by annexin V- FITC-binding/PI-uptake on flow cytometry. IL-15 antagonized growth inhibition (Figure A) and prevented apoptosis induced by BMP-4 (Figure B) and BMP-6 in the myeloma cell line IH-1. IL-15 also antagonized the growth inhibition induced by BMP-4 and/or BMP-6 in three out of eight patient samples. Neither IL-6, nor any of the other investigated cytokines were able to rescue the myeloma cells from growth inhibition and apoptosis induced by BMP-4 and -6. Among the investigated cytokines, we found that IL-15 has a unique capability to antagonize BMP- induced apoptosis and growth inhibition in myeloma cells. We examined cleavage of the proapoptotic protein caspase-3 and found that BMP-4 activated caspase-3 in the IH-1 cell line. This activation of caspase-3 was blocked by IL-15 but not by IL-6. We have demonstrated a possible mechanism for myeloma cells to escape apoptosis and growth-inhibition within the bone marrow. Intramedullar levels of IL-15 and BMPs may play a role in the pathogenesis of multiple myeloma. Figure A. Proliferation in response to BMP-4 stimulus Figure A. Proliferation in response to BMP-4 stimulus Figure B. Apoptosis in response to BMP-4 stimulus Figure B. Apoptosis in response to BMP-4 stimulus


2016 ◽  
Vol 60 (7-8-9) ◽  
pp. 305-314
Author(s):  
Mateusz Debowski ◽  
Mohammed El Dika ◽  
Jacek Malejczyk ◽  
Robert Zdanowski ◽  
Claude Prigent ◽  
...  

2022 ◽  
Vol 55 (1) ◽  
Author(s):  
Fatemeh Safari ◽  
Bahman Akbari

Abstract Background Chinese hamster ovary cell line has been used routinely as a bioproduction factory of numerous biopharmaceuticals. So far, various engineering strategies have been recruited to improve the production efficiency of this cell line such as apoptosis engineering. Previously, it is reported that the caspase-7 deficiency in CHO cells reduces the cell proliferation rate. But the effect of this reduction on the CHO cell productivity remained unclear. Hence, in the study at hand the effect of caspase-7 deficiency was assessed on the cell growth, viability and protein expression. In addition, the enzymatic activity of caspase-3 was investigated in the absence of caspase-7. Results Findings showed that in the absence of caspase-7, both cell growth and cell viability were decreased. Cell cycle analysis illustrated that the CHO knockout (CHO-KO) cells experienced a cell cycle arrest in G2/M phase. This cell cycle arrest resulted in a 1.7-fold increase in the expression of luciferase in CHO-KO cells compared to parenteral cells. Furthermore, in the apoptotic situation the enzymatic activity of caspase-3 in CHO-KO cells was approximately 3 times more than CHO-K1 cells. Conclusions These findings represented that; however, caspase-7 deficiency reduces the cell proliferation rate but the resulted cell cycle arrest leads to the enhancement of recombinant protein expression. Moreover, increasing in the caspase-3 enzymatic activity compensates the absence of caspase-7 in the caspase cascade of apoptosis.


Sign in / Sign up

Export Citation Format

Share Document