scholarly journals Nicotine Increases Osteoblast Activity of Induced Bone Marrow Stromal Cells in a Dose-Dependent Manner: An in vitro Cell Culture Experiment

2012 ◽  
Vol 2 (3) ◽  
pp. 153-158 ◽  
Author(s):  
Scott D. Daffner ◽  
Stacey Waugh ◽  
Timothy L. Norman ◽  
Nilay Mukherjee ◽  
John C. France
Author(s):  
David A. Hoey ◽  
Christopher R. Jacobs

Osteoporosis is a debilitating bone disease which occurs in part when bone marrow stromal cells (BMSCs) fail to produce sufficient numbers of osteoblasts to counteract bone resorption by osteoclasts. The majority of research to date has described chemically induced differentiation of BMSCs but a key regulator of stromal cell differentiation is physical loading. BMSCs experience both hydrostatic pressure and fluid flow within the marrow cavity and such modes of loading have been shown to significantly alter gene expression in vitro [1,2]. In particular, the effect of oscillatory fluid flow (OFF) induced shear stress results in the upregulation of osteogenic genes in preosteoblastic cells; however the effect of this mode of loading is not well characterized in human MSCs (hMSCs) [3].


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3848-3848 ◽  
Author(s):  
Yu-Tzu Tai ◽  
Kihyun Kim ◽  
Xian-Feng Li ◽  
Mariateresa Fulciniti ◽  
Weihua Song ◽  
...  

Abstract Abstract 3848 Poster Board III-784 The mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling pathway plays a crucial role in the pathogenesis of human multiple myeloma (MM) by promoting interactions of MM cells with bone marrow stromal cells (BMSCs) that secrete cytokines and growth factors for MM cell growth, survival, and resistance to chemotherapeutic drugs. Accumulating studies have supported targeting this signaling pathway in MM. Here we investigate cytotoxicity of AS703026, a novel selective MEK1/2 inhibitor with highly oral bioavailability, in MM cell lines and patient MM cells and define its mechanisms of action. AS703026, more potently (∼9-10 fold) than AZD6244, inhibits growth and survival of MM cells and cytokine-induced osteoclast differentiation. It specifically blocks baseline and adhesion-induced pERK1/2, but not pSTAT3. Selective MEK1/2 inhibition by AS703026 led to a cessation of cell proliferation accompanied by G0-G1 cell cycle arrest, as shown by increased subG0 cells, and concurrently abolished S phase cells. AS703026 also reduced expression of c-maf oncogene in a time-dependent manner, suggesting a MEK1/2-dependent regulation of c-maf that may contribute MM cell growth inhibition. AS703026 further induced apoptosis in MM cells, as manifested by caspase 3 and PARP cleavages in a time-dependent manner. It blocked osteoclastogenesis in vitro, as measured by number of TRAP-positive multinuclear cells following culturing PBMCs with RANKL and M-CSF. Importantly, AS703026 sensitized drug-resistant MM cells to a broad spectrum of conventional (dexamethasone, melphalan), as well as novel or emerging (lenalidomide, perifosine, bortezomib, rapamycin) anti-MM therapies. Synergistic or additive cytotoxicity (combination index < 1) induced by these combinations was further validated by annexin-V/PI staining and flow cytometric analysis. Combining these agents led to a significantly increased apoptosis and cell death than AS703026 alone, confirming enhanced cytotoxicity against MM cells. In vivo studies demonstrate that treatment of MM cell line H929-bearing mice with AS703026 (n=4 at 30 mg/kg; n=6 at 15 mg/kg), but not vehicle alone (n=6), blocked MM tumor growth in a dose-dependent manner (p<0.008 at 30 mg/kg; p<0.02 at 15 mg/kg). Immunoblotting and immunohistochemistrical staining showed that AS703026-reduced tumor growth was associated with downregulated pERK1/2, induced PARP cleavage, and decreased microvessels in vivo. Moreover, AS703026 (<200 nM) triggered significant cytotoxicity against the majority of patients with relapsed and refractory MM (>84%, n=18), regardless mutation status of 3 RAS and BRAF genes. Bone marrow stromal cells-induced viability of MM patient cells is similarly blocked within the same dose range. Our results therefore strongly support clinical protocols evaluating AS703026, alone or with other anti-MM agents, to improve patient outcome in MM. Disclosures: Chauhan: Progenra, Inc: Consultancy. Richardson:Keryx Biopharmaceuticals: Honoraria. Clark:EMD Serono: Employment. Ogden:EMD Serono: Employment. Andreas:EMD Serono: Employment. Rastelli:EMD Serono: Employment. Anderson:Millennium Pharmaceuticals: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2496-2496 ◽  
Author(s):  
Anne-Sophie Moreau ◽  
Xiaoying Jia ◽  
Garrett O’Sullivan ◽  
Xavier Leleu ◽  
Klaus Podar ◽  
...  

Abstract Background: Waldenstrom’s Macroglobulinemia is an incurable lymphoplasmacytic lymphoma with limited options of therapy. We have previously demonstrated upregulation of PKCβ protein in WM using protein array techniques, and confirmed increased expression in WM using immunohistochemistry. PKCβ regulates cell survival and growth, as well as migration and homing in many B-cell malignancies. We therefore hypothesized that inhibition of PKCβ will induce cytotoxicity in WM. Methods: In this study, we examined the effect of serial dilutions of the PKCβ enzastaurin (2.5 uM to 20 uM) on WM cell lines (BCWM1 and WM-WSU), IgM secreting low-grade lymphoma cell lines (MEC-1, RL), as well as primary CD19+ WM cells and WM cells adherent to bone marrow stromal cells (BMSCs), which induce resistance to conventional therapy. Cytotoxicity was measured by MTT assay and inhibition of cell proliferation was determined by thymidine uptake assay. Apoptosis was measured by flow cytometry using Annexin V and DAPI staining at 48 h. Cell DNA content analysis was performed using DAPI staining on fresh cells. Cell signaling pathways targeted by enzastaurin were determined using immunoblotting at 6 h (2.5 to 10 uM) and at 7.5 uM (10 min to 12 h). The effect of enzastaurin in vivo was determined using a subcutaneous WM model in SCID mice. Enzastaurin was given by oral gavage (80mg/kg twice daily). Results: Enzastaurin demonstrated time and dose-dependent inhibition of PKCβ in WM cells. It induced a significant decrease of proliferation at 24 and 48 h in all cell lines tested with an IC50 of 2.5 to 10 uM, even in the presence of DOPPA, a specific PKCβ stimulator. Similar effects were demonstrated in primary CD19+ WM cells, with no cytotoxicity on peripheral blood mononuclear cells indicating selective toxicity on malignant cells. Enzastaurin induced dose-dependent apoptosis at 24 and 48 h with induction of caspases 3, 8, 9 and PARP cleavage as well as a decrease in Bcl-xL. Analysis of cell DNA content confirmed apoptosis at low doses of enzastaurin (5 uM). To further determine the mechanism of action of enzastaurin in WM, we examined downstream molecules. It significantly inhibited AKT phosphorylation and AKT kinase activity, as determined by inhibition of phosphorylation of GSKα/β fusion protein. In addition, enzastaurin inhibited p-MARCK, and ribosomal p-S6. Enzastaurin overcame resistance induced by co-culture of WM cells with bone marrow stromal cells. In addition, enzastaurin (2.5 to 5 uM) in combination with bortezomib (2.5 to 10 nM), another active agent in WM, demonstrated strong synergistic activity using the Calcusyn software for synergy. Given that PKCβ regulates migration and homing of B-cells, we next determined the effect of enzastaurin on in vitro migration of WM cells. In the transwell migration assay, enzastaurin inhibited migration in a dose-dependent fashion (p=0.041). Finally, in vivo animal studies demonstrated significant inhibition of WM tumor growth in the enzastaurin treated mice (n=11), compared to control mice (n=8) (p=0.028). Conclusion: Enzastaurin has significant antitumor activity in WM in vitro and in vivo, providing the framework for clinical trials evaluating enzastaurin as a new therapeutic agent in patients with WM.


2009 ◽  
Vol 9 (10) ◽  
pp. 28S
Author(s):  
Scott Daffner ◽  
John France ◽  
Chad Smalley ◽  
Stacey Waugh ◽  
Timothy Norman ◽  
...  

2004 ◽  
Vol 181 (3) ◽  
pp. 477-492 ◽  
Author(s):  
AA Fouladi Nashta ◽  
CV Andreu ◽  
N Nijjar ◽  
JK Heath ◽  
SJ Kimber

Decidualisation of uterine stromal cells is a prerequisite for implantation of the embryo in mice. Here we have used an in vitro culture system in which stromal cells decidualise as indicated by a number of markers, including an increase in alkaline phosphatase (ALP) activity. The latter was used as a quantitative marker of decidualisation in the presence of low (2%) fetal calf serum. Prostaglandin E(2) (PGE(2)), which is known to induce decidualisation, increased ALP activity, and this effect was blocked in a dose-dependent manner by indomethacin. Leukemia inhibitory factor (LIF) was then examined, but it had no effect on PGE(2) secretion. However, LIF suppressed ALP activity in a dose-dependent manner in the presence of 2% serum, while an inhibitor of LIF that competes for binding to its receptor reversed the effect of LIF and increased ALP activity above the control level. In serum-free cultures, stromal cells differentiated rapidly, and no differences were observed between LIF-treated and untreated cultures. Stromal cells produce LIF during in vitro culture, and this peaked at 48 h. Freshly collected stromal cells from both day-2 and -4 pregnant mice expressed mRNA for the LIF receptor, and the transcript level was higher in cells isolated on day 4. However, no differences were observed in the relative levels of transcripts in cells from day 2 and day 4 after culture, nor were there differences between the LIF-treated cultures and controls. Therefore, in this study, we have shown that LIF suppresses decidualisation of murine uterine stromal cells in the presence of serum, this is not due to the regulation of PGE(2) secretion by stromal cells.


2013 ◽  
Vol 18 (6) ◽  
pp. 637-646 ◽  
Author(s):  
Kristine Misund ◽  
Katarzyna A. Baranowska ◽  
Toril Holien ◽  
Christoph Rampa ◽  
Dionne C. G. Klein ◽  
...  

The tumor microenvironment can profoundly affect tumor cell survival as well as alter antitumor drug activity. However, conventional anticancer drug screening typically is performed in the absence of stromal cells. Here, we analyzed survival of myeloma cells co-cultured with bone marrow stromal cells (BMSC) using an automated fluorescence microscope platform, ScanR. By staining the cell nuclei with DRAQ5, we could distinguish between BMSC and myeloma cells, based on their staining intensity and nuclear shape. Using the apoptotic marker YO-PRO-1, the effects of drug treatment on the viability of the myeloma cells in the presence of stromal cells could be measured. The method does not require cell staining before incubation with drugs, and less than 5000 cells are required per condition. The method can be used for large-scale screening of anticancer drugs on primary myeloma cells. This study shows the importance of stromal cell support for primary myeloma cell survival in vitro, as half of the cell samples had a marked increase in their viability when cultured in the presence of BMSC. Stromal cell–induced protection against common myeloma drugs is also observed with this method.


2021 ◽  
Vol 363 ◽  
pp. 109340
Author(s):  
Abeer Sallam ◽  
Thangirala Sudha ◽  
Noureldien H.E. Darwish ◽  
Samar Eghotny ◽  
Abeer E-Dief ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document