Human Bone Marrow Stromal Cells and Skin Fibroblasts Inhibit Natural Killer Cell Proliferation and Cytotoxic Activity

2011 ◽  
Vol 20 (5) ◽  
pp. 681-691 ◽  
Author(s):  
Amandine Pradier ◽  
Jakob Passweg ◽  
Jean Villard ◽  
Vincent Kindler
2012 ◽  
Vol 92 (9) ◽  
pp. 1208-1219 ◽  
Author(s):  
Cheng Zhong ◽  
Xin Zhang ◽  
Zhengjian Xu ◽  
Rongxin He

Background Electromagnetic fields (EMFs) used in stem-cell tissue engineering can help elucidate their biological principles. Objective The aim of this study was to investigate the effects of low-intensity EMFs on cell proliferation, differentiation, and cycle in mouse bone marrow stromal cells (BMSCs) and the in vivo effects of EMFs on BMSC. Methods Harvested BMSCs were cultured for 3 generations and divided into 4 groups. The methylthiotetrazole (MTT) assay was used to evaluate cell proliferation, and alkaline phosphatase activity was measured via a colorimetric assay on the 3rd, 7th, and 10th days. Changes in cell cycle also were analyzed on the 7th day, and bone nodule formation was analyzed on the 12th day. Additionally, the expression of the collagen I gene was examined by reverse transcription-polymerase chain reaction (RT-PCR) on the 10th day. The BMSCs of the irradiated group and the control group were transplanted into cortical bone of different mice femurs separately, with poly(lactic-co-glycolic acid) (PLGA) serving as a scaffold. After 4 and 8 weeks, bone the bone specimens of mice were sliced and stained by hematoxylin and eosin separately. Results The results showed that EMFs (0.5 mT, 50 Hz) accelerated cellular proliferation, enhanced cellular differentiation, and increased the percentage of cells in the G2/M+S (postsynthetic gap 2 period/mitotic phase + S phase) of the stimulation. The EMF-exposed groups had significantly higher collagen I messenger RNA levels than the control group. The EMF + osteogenic medium–treated group readily formed bone nodules. Hematoxylin and eosin staining showed a clear flaking of bone tissue in the irradiated group. Conclusion Irradiation of BMSCs with low-intensity EMFs (0.5 mT, 50 Hz) increased cell proliferation and induced cell differentiation. The results of this study did not establish a stricter animal model for studying osteogenesis, and only short-term results were investigated. Further study of the mechanism of EMF is needed.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4987-4987
Author(s):  
Hiroshi Ikeda ◽  
Yuka Aoki ◽  
Nasanori Nojima ◽  
Hiroshi Yasui ◽  
Toshiaki Hayashi ◽  
...  

Abstract Abstract 4987 The Bone marrow (BM) microenvironment plays crucial role in pathogenesis of Multiple myeloma(MM). Myeloma cells contacts with bone marrow stromal cells (BMSCs), which secrete factors/cytokines, promoting tumor cell growth and survival. Paracrine secretion of cytokines(i. e., interleukin-6 (IL-6) insulin-like growth factor-1, inflammatory protein-1a) in BM stromal cells promotes multiple myeloma cell proliferation and protects against drug-induced cytotoxicity. These cytokines provide stimulatory signals for multiple myeloma growth and survival. Bone involvement is a common feature in MM patient, solid and hematologic cancers. MM localizes to the bone in nearly all patients ranges between 40% and 75%. Disease-related skeletal complications result in significant morbidity due to pain, pathologic fractures and spinal cord compression. The bone microenvironment creates a supportive niche for tumor growth. Osteoclasts and bone marrow stromal cells, along with extracellular matrix and cytokines stimulate tumor cell proliferation and confer chemoresistance. Therefore, the reciprocal interactions between tumor cells, osteoclasts, osteoblasts, and bone marrow stromal cells present an important. In current study, monocyte can directly promote mesenchymal stem cells osteogenic differentiation through cell contact interactions, thus resulting in the production of osteogenic factors by the monocytes. This mechanism is mediated by the activation of STAT3 signaling pathway in the mesechymal stem cells that leads to the upregulation of Osteoblasts-associated genes such as Runx2 and alkaline phosphatase (ALP), and the down-regulation of inhibitors such as DKK1 to drive the differentiation of mesechymal stem cells into osteoblasts. In this study, we examined the role of monocyte, component of BM cells, as a potential niche component that supports myeloma cells. We investigated the proliferation of MM cell lines cultured alone or co-cultured with BM stromal cells, monocytes, or a combination of BM stromal cells and monocytes. Consistently, we observed increased proliferation of MM cell lines in the presence of either BM stromal cells or monocytes compared to cell line-only control. Furthermore, the co-culture of BM stromal cells plus monocytes induced the greatest degree of proliferation of myeloma cells. In addition to increased proliferation, BMSCs and monocytes decreased the rate of apoptosis of myeloma cells. Our results therefore suggest that highlights the role of monocyte as an important component of the BM microenvironment. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3253-3253
Author(s):  
Xuanru Lin ◽  
Xing Guo ◽  
Jing Chen ◽  
Qingxiao Chen ◽  
Enfan Zhang ◽  
...  

Abstract Background: Multiple myeloma (MM) is closely associated with inflammation. Patients with auto-immune disease、history of infection and other inflammatory disease have higher incidence of MM. IL-6 is the most important inflammatory factor in MM which plays a key role in the proliferation and progression. We previously demonstrated that MM cells were modified by bone marrow stromal cells (BMSCs) that formulate a inflammatory microenvironment in bone marrow (BM) and secret IL-6. How the inflammation makes BMSCs secret IL-6, however, remained undocumented. Our subsequent study compared the differential secretion of peripheral blood (PB) between MM patients and normal people by cytokine array, and showed that interleukin 32(IL-32) is highly expressed in MM patients. IL-32, also named natural killer 4(NK-4), is a newly found inflammatory factor. It was reported in solid tumors IL-32 is a pro-inflammatory factor which triggers a massive amplification of inflammatory process resulting in the change of other inflammatory factor including IL-6,IL-10,TNF-α. In this study, we examined BMSCs cytokines in MM BM and found that IL-32 was a functional factor in the process of inflammation in MM BM microenvironment. Results: First, to test our previous study, we detected IL-32 in BM supernatant and PB supernatant in both MM patients (n=45) and healthy controls (n=13) by ELISA. Result showed that in both BM and PB, MM patients have higher expression of IL-32 compared to healthy controls (P<0.05). Next, total BM cells(both CD138+ and CD138- cells) from MM patients were assayed by qRT-PCR for gene expression analysis.IL-32 were highly expressed in MM BM cells and the CD138+ cells (P<0.05). We also detected IL-32 in MM cell lines (RPMI 8226,OPM-2) and BMSCs isolated from MM patients by qRT-PCR, Western blot, and ELISA, and found that IL-32 was highly expressed in MM cell lines than BMSCs. In contrast, proteinase 3(PR3, receptor of IL-32) was highly expressed in BMSCs compared to MM cell lines. Second, we stimulated the MM BMSCs with recombinant IL-32α, and found that the secretion of IL-6,CCL3 (MIP1-α), CCL4(MIP-1β) were significantly increased and CCL-5(RANTES)and IL-10 were decreased (P<0.05). Further, Western blot was applied to detect the inflammation molecular pathway in BMSCs. JAK-STAT pathway and NF-κB pathway were activated, and the phosphorylation of STAT3 was increased. After we knock down the PR3 in BMSCs, these changes were reduced. We repeated these experiments in BMSCs isolated from 15 different MM patients, the phenomenon mentioned above showed in 11 patients. The recombinant IL-32α was also used to stimulate 8226 and OPM-2 cells, but these two kinds of MM cells didn't secret IL-6, and no significant change in cell proliferation or cell apoptosis. Finally, our group co-cultured the MM BMSCs with 8226 and OPM-2 cells. The secretion of IL-6 and the phosphorylation of JAK-STAT pathway in BMSCs were also increased. Knockdown of IL-32 in 8226 and OPM-2 cells weakened these changes.MM Cell proliferation and cell cycles after co-culture with MM BMSCs are under investigation. Conclusion: our findings suggest that IL-32 is mainly secreted by MM cells. It may not directly promote the MM cells to grow. However, IL-32 promote the MM BMSCs to secret more cytokines including IL-6,CCL3,CCL4 by activating the JAK-STAT3 pathway, which lead to a amplification of inflammation in BM environment, resulting in the cell proliferation . Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5407-5407
Author(s):  
Gaya Narendran ◽  
Matthew F. Clarkson ◽  
Randal Johnston

Abstract Introduction Acute lymphoblastic leukemia (ALL) is the most common malignant disease in children, constituting more than a quarter of all childhood cancers. The survival rate of standard/good risk ALL now exceeds 80%. In spite of the significant improvements in outcome, the emergence of resistant disease remains the most common cause of death. Escalation of the intensity of combination chemotherapy with the introduction of second-line drugs is accompanied by cumulative toxicity, with marginal incremental benefits. Hence novel therapeutic approaches are urgently needed to improve the outcome in these patients. In addition to the inherent molecular alterations, recent reports have suggested an active role for the bone marrow stromal microenvironment in promoting both leukemia cell viability and drug resistance. Previous studies have shown a pivotal role for the stromal chemokine CXCL12 (SDF-1) and its receptor CXCR4 in this process. In the multiple myeloma model, inhibition of CXCR4 by the targeted agent AMD3100 (Plerixafor) abrogates the chemotherapy protection conferred by stromal cells. Recent years have seen evidence for the effectiveness of oncolytic viruses as cytotoxic agents against refractory tumors. The strategy behind this approach is to develop viruses that can replicate and cause cell lysis specifically in cancer cells while leaving non-malignant cells unaffected. In this study we explore the utility of reovirus mediated anti-leukemic therapy in an experimental model of pediatric leukemia in the context of co-culture with bone marrow stromal cells. Methods A panel of leukemia cell lines (n=4) representing pediatric ALL and acute myeloid leukemia (AML) were evaluated when co-cultured in the presence of stromal conditioned medium, live bone marrow stromal cells, stromal cells fixed with 0.5% glutaraldehyde, or control human skin fibroblasts (1Br3 cells). After three days in culture, cell proliferation kinetics under each condition was measured by WST-1 assay. The influence of nonspecific leukemia cell attachment was evaluated by the culture of leukemia cells in poly-L-lysine coated plates. Next the leukemic cells were treated with oncolytic reovirus, at various concentrations with and without co-cultures conditions to evaluate the potential of leukemia cells to escape reovirus mediated cytolysis. Finally, the ability of stromal cells to modulate reovirus anti-leukemic activity was evaluated in the presence of various concentrations of AMD3100. Results and Discussion An average of a 2-fold increase in leukemia cell proliferation (in the absence of reovirus) was seen with live stromal co-cultures compared to conditioned media or fixed stromal cells, indicating the requirement of live cell-cell contact with stromal cells in this process. Poly-L lysine coated plates did not increase leukemia cell growth. Our data reveals that, as previously seen with chemotherapeutic drug exposure, the presence of stromal cells is able to decrease the cytolytic activity of reovirus on leukemic cells. Leukemia cells alone exhibit a survival of 10% following a three-day exposure to reovirus (50 MOI). However, in the presence of stromal the survival rate was increased to 40% (p < 0.05). Furthermore, the addition of AMD3100 provided partial restoration of the oncolytic activity (mean 15% survival, n=3 experiments). Stromal conditioned media also decreased reovirus infectivity, although to a lower extent (mean 17% cell survival). Co-culture of leukemia cells with control 1br3 human skin fibroblast cells provided no protection from reoviral cytolysis (mean 10% survival). In this proof-of-concept study, we provide evidence for the first time that the bone marrow niche may provide at least partial protection from oncolytic virotherapy. This indicates the importance of evaluating the efficacy of oncolytic viruses in the context of the tumor microenvironment in future studies. Furthermore, we provide evidence for the consideration of combination therapies with targeted agents such as AMD3100, which may disrupt the stromal protective activities, to increase the effectiveness virotherapy in the treatment of refractory leukemia. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 5012-5012
Author(s):  
Bhagavathi A Narayanan ◽  
Nicole A Doudican ◽  
Jeesun Park ◽  
Narayanan K Narayanan ◽  
Dasgupta Ramanuj ◽  
...  

Abstract Abstract 5012 Development and progression of multiple myeloma is dependent on the bone marrow (BM) microenvironment. Bone marrow stromal cells (BMSC) secrete Wnt ligands that activate the Wnt signaling pathway. The canonical Wnt pathway, which is mediated through the key transcriptional effector β-catenin (β-cat), is commonly deregulated in many cancers. Cells with β-cat-regulated transcription (CRT) are protected against apoptosis; conversely inhibition of CRT may inhibit cell proliferation. In this study we tested the efficacy of recently described inhibitors of CRT (iCRTs) for their selective antagonistic effect on Wnt-β-cat in MM cells. Although, earlier studies have documented Wnt signaling in human MM cells, in order to test the chemosensitivity of iCRTs in myeloma cells, we first confirmed the expression of β-cat in human MM cell types. An immunofluorescence detection of β-cat in U266 cells showed nuclear localization in > 70% of the cells, a similar trend of nuclear β-cat was observed in MM1 and patient derived BMMC cells. This observation is consistent with the Western blot analysis of the total protein from three cell types. The above data on the expression of nuclear β-cat in MM cell lines and cells from patient sample (n=16) provides the rationale for using these cells to test the efficacy of iCRTs (Oxazole-iCRT-3 and Thiazole-iCRT-5), that are designed to target β-cat signaling. Wnt reporter plasmid STF16-transfected MM cells treated with iCRTs at the doses of 10, 25, and 50 μM showed a dose dependent decrease (2–3 fold) in the Wnt/β-cat reporter activity, with a significant decline at a maximum dose of 50 μM (p<0. 001). Given the role of Wnt and the upstream antagonist DKK1 in normal bone formation and MM development, we examined whether inhibition of Wnt-β-cat response by iCRTs indirectly affects the protein expression of DKK1. As expected, our data from Western blot analysis of MM cells treated with iCRTs showed no significant change in the expression of DKK1, thereby confirming the effect of iCRTs on β-cat itself. While bone marrow angiogenesis is a hallmark in multiple myeloma, it is also correlated with the severity of the disease. Since we observed an elevated level of VEGF in the serum samples ranging from 300 pg/ml to 500 pq/ml in patients (n=16) with late stage MM, we tested the effect of iCRTs on VEGF level in MM cells in the cell culture medium obtained from the co-culture of bone marrow stromal cells (BMSC) with human U266 cells. Treatment of BMSC and U266 in co-culture with iCRT-3 and iCRT-5 at a dose of 50μM of each showed a decrease in the VEGF level and down regulated VEGF at the mRNA level by >3 fold determined by qRT-PCR analysis. To determine whether the effect of iCRTs on the VEGF activity is specific to its ability to antagonize b-cat activity, we used culture medium collected from cells transiently transfected with siRNA for β-cat. siRNA mediated down regulation of β-cat showed a decrease in the VEGF levels, thereby confirming that the effect of iCRTs is indeed mediated by their inhibitory effect on b-cat. Although several aspect of our key findings are yet to be confirmed in preclinical in vivo models for MM, this part of the study provide evidence that indicate a VEGF-dependent increase in cell proliferation and migration of MM cells that can be antagonized by specific inhibitors of nuclear b-cat, thereby underscoring the importance of developing iCRTs as a novel class of Wnt-directed therapeutics in human MM. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document