scholarly journals Human myeloma RPMI8226 cells and normal cells have different sensitivity to bisilicate silver nanoparticles in vitro

2021 ◽  
Vol 67 (5) ◽  
pp. 724-730
Author(s):  
Anna Sherbanyuk ◽  
Sergei Moiseev ◽  
Natalia Bychkova ◽  
Nikolai Germanov ◽  
Sergei Golyandin ◽  
...  

Introduction. Silver nanoparticles due to its pronounced cytotoxicity are regarded as promising agent for anticancer therapy. Determination of normal and transformed cells sensitivity to silver nanoparticles can be the basis for the application as an adjuvant cancer treatment. The objective of the study was to investigate influence of atomic clusters of Argentum (ACA) in the form of silver bisilicate nanoparticles colloid solution on viability and proliferation of human myeloma cell line, mesenchymal stromal cells and blood lymphocytes. Material and methods. Cell viability was evaluated by MTT and LDH assay. Cell proliferation was evaluated by flow cytometry. Results. It was found that ACA had dose-depending cytotoxicity toward all investigated cell types, but normal and transformed cells varied significantly in the sensitivity to nanoparticles. IC50 for myeloma cell line RPMI8226 was 1,75 µg/ml. For MSCs of different origin IC50 was in the range of 12 to 16 µg/ml. ACA in concentration from 2 to 3 µg/ml induced RPMI8226 cells metabolic disruption and death without influence on viability and cell cycle of mesenchymal stromal cells and blood lymphocytes. Conclusion. Results of work has shown distinct differences in sensitivity to ACA between myeloma cells, mesenchymal stromal cells and blood lymphocytes. The optimal range of ACA concentration with anticancer effect without cytotoxic influence on normal cells has been determined in vitro.

Blood ◽  
1999 ◽  
Vol 94 (10) ◽  
pp. 3551-3558 ◽  
Author(s):  
Maged S. Mahmoud ◽  
Ryuichi Fujii ◽  
Hideaki Ishikawa ◽  
Michio M. Kawano

In multiple myeloma (MM), the cell surface protein, CD19, is specifically lost while it continues to be expressed on normal plasma cells. To examine the biological significance of loss of CD19 in human myeloma, we have generated CD19 transfectants of a tumorigenic human myeloma cell line (KMS-5). The CD19 transfectants showed slower growth rate in vitro than that of control transfectants. They also showed a lower capability for colony formation as evaluated by anchorage-independent growth in soft agar assay. The CD19 transfectants also had reduced tumorigenicity in vivo when subcutaneously implanted into severe combined immunodeficiency (SCID)-human interleukin-6 (hIL-6) transgenic mice. The growth-inhibitory effect was CD19-specific and probably due to CD19 signaling because this effect was not observed in cells transfected with a truncated form of CD19 that lacks the cytoplasmic signaling domain. The in vitro growth-inhibitory effect was confirmed in a nontumorigenic human myeloma cell line (U-266). However, introduction of the CD19 gene into a human erythroleukemia cell line (K-562) also induced growth inhibition, suggesting that this effect is CD19-specific, but not restricted to myeloma cells. These data suggest that the specific and generalized loss of CD19 in human myeloma cells could be an important factor contributing to the proliferation of the malignant plasma cell clones in this disease.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 742-742 ◽  
Author(s):  
Sonia D'Souza ◽  
Davide Del Prete ◽  
Flavia Esteve ◽  
Benedicte Sammut ◽  
Shibing Yu ◽  
...  

Abstract Abstract 742 Multiple Myeloma (MM) is a plasma cell malignancy characterized by formation of lytic bone lesions in approximately 90% of the patients which do not heal even after prolonged complete remission. The basis for this selective and protracted suppression of osteoblast (OBL) differentiation from immature bone marrow stromal cells (MSC) is unknown. Although factors that inhibit OBL differentiation in MM have been identified, such as DKK-1, sFRP2, IL-3, IL-7, and TNF-a, none of these factors have been shown to be responsible for the protracted suppression of OBL differentiation in MM. Further, inhibition of Runx2 activity, a critical transcription factor required for OBL differentiation has been reported in MM, but the mechanisms responsible are still unclear. To address the basis for the protracted inhibition of OBL differentiation in MM, we have developed a murine model of MM-induced OBL suppression using a genetically modified murine myeloma cell line that expresses GFP and thymidine kinase (5TGM1-GFP-TK MM cells). Injection of these 5TGM1-GFP-TK MM cells into SCID mice resulted in persistent inhibition of OBL differentiation even when the MM cells were totally depleted by ganciclovir treatment. The MSC from these mice had selective inhibition of OBL differentiation, but not adipogenesis, and minimally differentiated to OBL even when treated with BMP2. These MSC expressed elevated levels of the SNAG family Zn-finger containing transcriptional repressor, Gfi-1, which we found can cause both acute and protracted suppression of RUNX2. In support of these results, decreased RUNX2 expression and elevated GFI-1 levels were also protracted in MSC from 7 MM patients with impaired OBL differentiation compared to normals. Further, 5TGM1 inhibition of OBL differentiation in vitro was dependent on TNF-a and IL-7, and neutralizing antibodies to TNF-a and IL-7 blocked MM-induced Runx2 suppression. In addition, TNF-a and IL-7 increased Gfi-1 in a murine OBL precursor cell line (MC4). Deletion analysis of the Runx2 P1 promoter revealed that a 943-bp region containing 27 putative Gfi-1 binding sites (AA(T/G)C core) was responsible for MM repression of Runx2 expression. Importantly, siRNA knockdown of GFI-1 expression restored RUNX2, OCN, BSP and OSX expression in both MM exposed MC4 cells and in MSC from MM patients. These results support an important role for GFI-1 in repressing RUNX2 expression in MSC exposed to MM cells, thereby inhibiting osteoblastogenesis in MM. Disclosures: Roodman: Novartis: Consultancy, Research Funding, Speakers Bureau; Amgen: Consultancy; Celgene: Consultancy; Acceleron: Consultancy.


2019 ◽  
Vol 71 (2) ◽  
pp. 573-581 ◽  
Author(s):  
Akira Iwamoto ◽  
Yuichi Inoue ◽  
Hirofumi Tachibana ◽  
Hiroharu Kawahara

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
S. Marozin ◽  
B. Simon-Nobbe ◽  
S. Irausek ◽  
L. W. K. Chung ◽  
G. Lepperdinger

AbstractThe human fetal osteoblast cell line (hFOB 1.19) has been proposed as an accessible experimental model for study of osteoblast biology relating to drug development and biomaterial engineering. For their multilineage differentiation potential, hFOB has been compared to human mesenchymal progenitor cells and used to investigate bone-metabolism in vitro. Hereby, we studied whether and to what extent the conditionally immortalized cell line hFOB 1.19 can serve as a surrogate model for bone-marrow derived mesenchymal stromal cells (bmMSC). hFOB indeed exhibit specific characteristics reminiscent of bmMSC, as colony formation, migration capacity and the propensity to grow as multicellular aggregates. After prolonged culture, in contrast to the expected effect of immortalization, hFOB acquired a delayed growth rate. In close resemblance to bmMSC at increasing passages, also hFOB showed morphological abnormalities, enlargement and finally reduced proliferation rates together with enhanced expression of the cell cycle inhibitors p21 and p16. hFOB not only have the ability to undergo multilineage differentiation but portray several important aspects of human bone marrow mesenchymal stromal cells. Superior to primary MSC and osteoblasts, hFOB enabled the generation of continuous cell lines. These provide an advanced basis for investigating age-related dysfunctions of MSCs in an in vitro 3D-stem cell microenvironment.


Blood ◽  
2003 ◽  
Vol 102 (7) ◽  
pp. 2581-2592 ◽  
Author(s):  
Paula A. Croonquist ◽  
Michael A. Linden ◽  
Fangyi Zhao ◽  
Brian G. Van Ness

Abstract ANBL-6, a myeloma cell line, proliferates in response to interleukin 6 (IL-6) stimulation, coculture with bone marrow stromal cells, and when harboring a constitutively active mutant N-ras gene. Eighteen samples, including 4 IL-6-treated, 3 mutant N-ras-transfected, 3 normal stroma-stimulated, 2 multiple myeloma (MM) stroma-stimulated, and 6 untreated controls were profiled using microarrays interrogating 12 626 genes. Global hierarchical clustering analysis distinguished at least 6 unique expression signatures. Notably, the different stimuli altered distinct functional gene programs. Class comparison analysis (P = .001) revealed 138 genes (54% involved in cell cycle) that distinguished IL-6-stimulated versus nontreated samples. Eighty-seven genes distinguished stroma-stimulated versus IL-6-treated samples (22% encoded for extracellular matrix [ECM] proteins). A total of 130 genes distinguished N-ras transfectants versus IL-6-treated samples (26% involved in metabolism). A total of 157 genes, 20% of these involved in signaling, distinguished N-ras from stroma-interacting samples. All 3 stimuli shared 347 genes, mostly of metabolic function. Genes that distinguished MM1 from MM4 clinical groups were induced at least by one treatment. Notably, only 3 genes (ETV5, DUSP6, and KIAA0735) are uniquely induced in mutant ras-containing cells. We have demonstrated gene expression patterns in myeloma cells that distinguish an intrinsic genetic transformation event and patterns derived from both soluble factors and cell contacts in the bone marrow microenvironment. (Blood. 2003;102:2581-2592)


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1568-1568 ◽  
Author(s):  
Rajni Sinha ◽  
Ebenezer David ◽  
Emily Zeilter ◽  
Claire Torre ◽  
Jonathan L. Kaufman ◽  
...  

Abstract Introduction Multiple myeloma is a clonal plasma cell malignancy characterized by proliferation and accumulation of plasma cells in the bone marrow. Most patients are incurable with the current treatment modalities. Clearly novel agents are needed to improve the outcome for patients with myeloma. We have previously shown that the combination of bortezomib and tipifarnib results in synergistic myeloma cell death. This increase in apoptosis is associated with down regulation of phosphorylated AKT, a potent anti-apoptotic signaling molecule. Therefore, agents that target AKT represent ideal compounds for further study in myeloma. Perifosine is a novel, oral bioavailable alkylphospholipid. Perifosine has displayed apoptotic and antipropliferative activity in vitro and in vivo in several human cancer models including leukemia. Perifosine exerts its actions by interfering with key intracellular pathways including AKT, MAPK, JNK, p21waf1. Our hypothesis is that targeting AKT via multiple upstream pathways will result in increased myeloma cell apoptosis. Therefore, we assessed the effects of single agent perifosine with and without tipifarnib on multiple myeloma cell lines. Method The myeloma cell line RPMI8226 was used. Cell viability and proliferation were assessed using MTT assays. Cells were incubated with increasing concentrations of both agents alone and in combination. Cell proliferation was assayed at 24, 48 and 72 hours. Western blots were then carried out to evaluate the effects of the intracellular protein PDK1, one of the critical signaling molecules that phosphorylates and activates AKT. Results As we and others have previously shown, tipifarnib at concentrations that can be achieved clinically is associated with minimal cytotoxicity. At 5 μM, tipifarnib decrease proliferation by only 20%. In contrast, there is a potent dose response effect of single agent perifosine (Fig. 1). These results were apparent as early as 24 hours. When tipifarnib at 5 μM is used in combination with a subtherapeutic dose of perifosine (2 μM), there is a marked decrease in cell proliferation (Fig. 2). In addition, combination therapy resulted in a reduction in the phosphorylated form of PDK1, a critical finding that was not seen with either drug alone. Conclusion Combination therapy with tipifarnib and perifosine results in less cell proliferation compared to either agent used alone in the RPMI8226 myeloma cell line. The dosages employed in these in-vitro studies are lower than those used in previously published data and are clinically achievable. Studies targeting other cell lines including MM.1R, MM.1S, and U266 are in progress. Analysis of AKT, Caspase 3, 8 and 9 are being explored to help delineate the mechanism of this novel combination. The goal is to develop further effective treatment options for patients with myeloma. Figure 1 Figure 1. Figure 2 Figure 2.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2949-2949
Author(s):  
Akira Sakai ◽  
Miyo Oda ◽  
Noriaki Yoshida ◽  
Mitsuhiro Itagaki ◽  
Koji Arihiro ◽  
...  

Abstract Abstract 2949 Adhesion of multiple myeloma (MM) cells to bone marrow stromal cells triggers cytokine-mediated tumor cell growth, survival, and drug resistance. In particular, integrin a4b1, very late antigen 4 (VLA-4)-mediated fibronectin adhesion confers a survival advantage to myeloma cells. One of the problems in treating patients with MM is that it is very hard to eliminate residual myeloma cells, even following high-dose chemotherapy followed by auto-stem cell transplantation. Importantly, cell adhesion-mediated drug resistance (CAM-DR) must be overcome in order to eliminate the minimal residual disease of MM. Here we characterized a multiple myeloma cell line, MSG1, which depends on HS23 stromal cells for its survival, was established from the pleural effusion of a patient with MM who expressed the M-protein of IgA-λ in his serum. During the first two months of culture, the myeloma cells survived on adhesive cells from the pleural effusion and subsequently, they continued to proliferate on HS23 stromal cells but not on HS27A stromal cells (both stromal cells were established by Torok-Storb B, Blood 1995). The phenotype of the established MSG1 cell line was: CD138+, CD38++, CD19−, CD56−, VLA-4+, VEGFR1+, and VEGFR2+. Furthermore, immunohistochemical staining demonstrated expression of IgA and λ chain in the cytoplasm. Karyotype analysis indicated complex chromosomal abnormalities, basically hypertriploidy including the deletion of chromosome 13 and 17, and c-myc translocation. MSG1 cells continued to proliferate, not only when co-cultured with HS23 cells, but also when cultured on fibronectin-coated plates with the supernatant of HS23 cells or RPMI1640 medium supplemented with 10% FBS (control medium) containing IL-6 (10 ng/ml). Notably, MSG1 could not survive in control medium containing IL-6 or in HS23 supernatant unless bound to fibronectin, which was also expressed on HS23 and HS27A cells. IL-6 and VEGF production were detected in the supernatants of both HS23 and HS27A stromal cells (36.8±4.5 pg/ml and 131±5.8 pg/ml; 13.2±1.9 pg/ml and 16664±418 pg/ml, respectively). Next, we analyzed the effect of tocilizumab, an anti-IL-6R antibody, and bevacizumab, an anti-VEGF antibody on MSG1 survival. Tocilizumab (50 μ g/ml) inhibited MSG1 survival when cultured on fibronectin-coated plates in control medium containing IL-6 (10 ng/ml), and tocilizumab (10 μ g/ml) inhibited MSG1 survival when cultured on HS23 stromal cells. However, bevacizumab (500 μ g/ml) did not show such inhibition. Therefore, MSG1 survival depends on HS23 stromal cells: in other words, it depends on binding to fibronectin and IL-6. If these factors induced CAM-DR in myeloma cells, MSG1 may be a unique myeloma cell line that will be useful for analysis of CAM-DR, and tocilizumab might be a useful drug for treatment of MM. Furthermore, since MSG1 could survive on irradiated HS27A cells, and since HS23 and HS27A express similar adhesion molecules (Torok-Storb B et al., Blood 1995), these data suggest that HS27A might secrete factors that are detrimental to MSG1 survival. The identification of such an inhibitory factors could be of interest in terms of the regulation of myeloma proliferation. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document