Bone Marrow Stroma Protects Myeloma Cells from Cytotoxic Damage Via Induction of the Oncoprotein MUC1

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3378-3378
Author(s):  
Michal Bar-Natan ◽  
Katarina Luptakova ◽  
Maxwell Douglas Coll ◽  
Dina Stroopinsky ◽  
Hasan Rajabi ◽  
...  

Abstract Introduction : Stromal cells in the bone marrow microenvironment of patients with multiple myeloma (MM) are thought to play a vital role in promoting cell growth and protection from cytotoxic injury. Targeting of stromal-myeloma cell interactions to enhance anti-myeloma treatment represents a promising therapeutic strategy. The MUC1 oncoprotein is a critical oncoprotein that is expressed in the majority of primary myeloma cells and regulates downstream pathways such as NFkB and β-catenin/wnt that modulate myeloma growth and survival. Inhibition of MUC1 via a cell penetrating peptide (GO-203) that blocks down stream signaling reverses resistance to bortezomib (BZT). Herein we studied the influence of bone marrow stromal cells (BMSC) on MUC1 expression on MM cells, and its link to drug resistance. Methods and Results : Coculture of MM human cell lines (RPMI and U266) with a stromal cell line (HS-5), resulted in an upregulation of MUC1 expression as determined by an approximately 2 fold increase in the mean fluorescent intensity (MFI) of MUC1 as measured by flow cytometry. Similar findings were observed following coculture of MM cells with stromal cells isolated from primary bone marrow mononuclear cells (BMSC) of MM patients. Stromal cell mediated upregulation of MUC1 expression was subsequently confirmed by Western blot analysis. Patient derived MM cells were also noted to increase their MUC1 expression 2.9 fold when co-cultured with stroma (HS-5 cell line). MUC1 expression was also increased following coculture of MM cells with stromal cells in transwell plates, suggesting the effect was mediated by soluble factors not requiring cell-cell contact. Consistent with these findings, we demonstrated that addition of recombinant IL-6, a stromal cell derived cytokine, to MM cells resulted in a 2 fold increase in MFI of MUC1 expression. Moreover, coculture of MM cells with IL-6 neutralizing antibodies abrogated the effect of BMSC on MUC1 expression. These results suggest that stromal cell secretion of IL-6 plays a role in upregulation of the oncoprotein MUC1 on MM cells. We subsequently evaluated the effect of stromal cell induction of MUC1 expression on resistance to anti-myeloma agents. Increased MUC1 expression following coculture of MM cells with BMSC was associated with a higher level of resistance to BTZ (20nM), resulting in 48% less cell death by CellTiter-Glo and annexin/propidium iodide (PI) staining. Conversely, we demonstrated that silencing of MUC1 expression using a lentiviral siRNA resulted in enhanced sensitivity to anti-myeloma agents. Cell viability in MUC1 silenced as compared to wild type RPMI cells decreased by 18%, 43%, and 50% when treated with 10mg/ml cyclophosphamide (Cy), 5nM BZT, and 0.1mM melphalan, respectively. MUC1 silenced U266 cells demonstrated a decrease in cell viability by 24%, 34%, and 45% when treated with 10mg/ml Cy, 5nM BZT, and 1mM lenalidomide respectively. Similarly, exposure of primary MM cells to the MUC1 inhibitor GO-203 resulted in enhanced MM cell sensitivity to bortezomib and cyclophosphamide evidenced by a 60% and 39% decrease in cell viability respectively, compared to each drug alone. Conclusions : Our results delineate one of the mechanisms by which the bone marrow microenvironment confers drug resistance in MM. MM cells co-cultured with BMSC have enhanced expression of MUC1, mediated by IL-6 secretion. Overexpression in turn confers MM cell resistance to standard anti-myeloma agents. Importantly inhibition of MUC1 via silencing of expression or exposure to a small molecule inhibitor can overcome drug resistance to known anti-myeloma drugs, providing the rationale for clinical evaluation of combination therapy. Disclosures Kufe: Genus Oncology: Consultancy, Equity Ownership.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 222-222
Author(s):  
Oliver Herd ◽  
Maria Abril Arredondo Garcia ◽  
James Hewitson ◽  
Karen Hogg ◽  
Saleni Pravin Kumar ◽  
...  

Immune thrombocytopenia (ITP) is an acquired autoimmune disease characterised by low platelet counts (<100 x 109/L) and manifests as a bleeding tendency. The demand on hematopoiesis is elevated in chronic ITP, where sustained platelet destruction mediated by an activated immune system is likely to cause considerable stress on progenitor populations. Intriguingly, this increased stress does not appear to result in functional exhaustion, as chronic ITP patients do not present with pancytopenia. By using a novel murine model of chronic ITP, generated by injecting mice with anti-CD41 antibody (ITP group) or IgG (control group) every 48hrs for 4 weeks, we aimed to define the effect of chronic ITP on hematopoietic progenitors and to elucidate the mechanisms behind the preservation of hematopoiesis. The relative numbers of hematopoietic progenitors in mice with chronic ITP vs controls were analysed by flow cytometry and their fitness was assessed by measuring their relative ability to reconstitute the hematopoietic system of lethally irradiated recipients. There was a significant increase in all hematopoietic progenitors analysed in ITP: 2.96-fold increase in multipotent progenitors, 4.66-fold increase in short-term hematopoietic stem cells (ST-HSCs) and 4.93-fold increase in long-term hematopoietic stem cells (LT-HSCs), which led to an increased ability of ITP donor bone marrow to reconstitute irradiated recipients. The results indicate that chronic ITP drives LT-HSCs out of quiescence and causes increased differentiation into committed progenitors in order to meet the increased demand in platelet production. In support of this, increased megakaryopoiesis was observed in chronic ITP, with a 60.5% increase in the number of megakaryocytes observed in bone marrow sections. Interestingly, similar to the clinical manifestation of ITP, we observed no change in levels of circulating TPO in our ITP model. Next, the effect of chronic ITP on the bone marrow microenvironment was determined due to its essential role in the support and maintenance of hematopoiesis. Histological analysis of bone marrow from mice with chronic ITP (Figure 1) revealed a 66.7% increase in the numbers of LepR+/ Cxcl12-DsRed stromal cells. LepR+/ Cxcl12-DsRed stromal cells are a well characterised stromal cell subset, known to be essential for maintenance and retention of HSCs in the bone marrow microenvironment. During chronic ITP, this stromal cell subset maintained their classically defined perivascular location and retained their ability to produce high levels of hematosupportive cytokines (Cxcl12 and Kitl). Chronic ITP was associated with a significant increase in total bone marrow expression (Cxcl12=2.39-fold increase; Kitl=1.71-fold increase), pointing to perivascular stromal cell expansion as being the source of increased local hematopoietic support. Analysis of the bone marrow vascular network revealed that the average vessel area was increased in chronic ITP (54.3% increase), whilst the number of vessels remained unchanged implying that the marrow sinusoids are vasodilated. We hypothesise that an increase in blood vessel area would aid the extravasation of circulating HSCs back into the bone marrow microenvironment where they would contribute to hematopoiesis. By developing an accurate mouse model of chronic ITP, we have identified key alterations in HSCs and the bone marrow microenvironment. Our data clearly demonstrates that in chronic ITP, HSCs are driven out of quiescence and expand in number in order to contribute to the increased demand for hematopoiesis. Furthermore, the bone marrow microenvironment adapts to this increased differentiation pressure on HSCs by creating a hematosupportive, quiescence promoting environment through the expansion of bone marrow stromal cells, and an increase in blood vessel area. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4024-4024
Author(s):  
Shannon M. Matulis ◽  
Jiangxia Liu ◽  
Vikas A. Gupta ◽  
Jason E. Conage-Pough ◽  
Ajay K. Nooka ◽  
...  

Abstract Abstract 4024 The survival of multiple myeloma cells in the bone marrow is largely dependent on its interactions with cellular components of the microenvironment. These interactions, in the form of cell-cell interactions and soluble factors support myeloma cell survival and allow for the mutations associated with transformation to drive plasma cell growth and proliferation. Moreover survival signals from the bone marrow microenvironment influence the response to therapeutic agents, therefore understanding the molecular basis of these interactions is necessary to improve the depth of responses to therapy in myeloma patients. Cell survival is regulated by the Bcl-2 family of proteins and we previously reported that in myeloma cells one can map which Bcl-2 protein is necessary for cell survival by determining which protein is sequestering the pro-apoptotic molecule, Bim. This could also be used to predict myeloma cell sensitivity to th Bcl-2/Bcl-xL antagonist ABT-737. While studying the response of freshly isolated patient samples to ABT-737, we found that in 40% of the samples tested (N=10) as well as in 5 myeloma cell lines, inclusion of stromal components significantly inhibited ABT-737-induced apoptosis. Thus the microenvironment can influence survival by altering the pattern of Bcl-2 dependence in cells. We have determined that a soluble factor secreted by stromal cells is necessary for this activity and now report that this factor is interleukin-6. We found that addition of conditioned medium from the HS-5 stromal line or freshly isolated bone marrow stromal cells (N=4) from myeloma patients or normal donors (N=1) to human myeloma cell lines all resulted significant resistance to ABT-737. Since all of these cells are a rich source of IL-6, we tested if the addition of recombinant IL-6 mimicked the addition of stromal cell supernatant. Indeed rIL-6 (10 ng/ml) was as potent as co-culture with HS-5 cells or addition of HS-5 or stromal cell conditioned medium at blocking ABT-737-induced apoptosis in MM.1s cells. Thus IL-6 is sufficient to block ABT-737-induced apoptosis. To demonstrate that IL-6 was necessary we determined the effect of the addition of an IL-6 neutralizing antibody on stromal cell-induced resistance to ABT-737. We found that addition of the antibody to HS-5 conditioned medium blocked resistance to ABT-737 in a dose-dependent fashion. Moreover we found that addition of anti-IL6 to a patient sample had a similar effect as removing the stromal cells on the response to ABT-737-induced death. Taken together these data suggest that IL-6 is both necessary and sufficient for regulating Bcl-2 dependence in multiple myeloma. To define the molecular basis for these findings we initially investigated the expression of the key Bcl-2 proteins involved in Bcl-2 dependence and myeloma cell survival. Despite their profound effects on ABT-737-induced apoptosis we found that neither co-culture of MM.1s cells with HS-5 cells, HS-5 conditioned medium or rIL-6 had any effect on the expression of Mcl-1, Bcl-xL or Bim. We then investigated the patterns of interaction between these proteins and found that the addition of stromal components or IL-6 resulted in a shift of Bim binding from Bcl-xL to Mcl-1. This is consistent with our previous findings that when Bim is bound to Mcl-1, myeloma cells are less sensitive to ABT-737. Finally we set out to determine how IL-6 signaling results in a change in ABT-737 sensitivity. We treated MM.1s cells with ABT-737 and IL-6 in the presence of inhibitors of the MEK/ERK, p38, PI3K/AKT, and JAK/STAT pathways. We found that only the MEK inhibitor, U0126 could re-sensitize IL-6-treated cells to ABT-737-induced death at concentrations that did not kill cells or have additive effects with ABT-737 in the absence of IL-6 (P<0.05 2-way ANOVA). Taken together, these data indicate that stromal cells regulate myeloma cell survival through IL-6 by altering Bim binding in a MEK-dependent fashion. This results in a cell that is highly dependent on Mcl-1. These data offer an explanation for why proteasome inhibitors are so effective in this disease. While the bone marrow microenvironment is enforcing Mcl-1 dependence, proteasome inhibitors induce the Mcl-1 inhibitor Noxa, which overcomes this activity of IL-6. Moreover these data point to potential ways of combining the targeting of Bcl-2 proteins and IL-6 signaling as a means to effectively kill myeloma cells in the bone marrow microenvironment. Disclosures: Kaufman: Millenium: Consultancy; Celgene: Consultancy; Novartis: Consultancy; Onyx Pharmaceuticals: Consultancy.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1354-1354
Author(s):  
Moaaz Soliman ◽  
Ella J. Born ◽  
Sara V. Hartman ◽  
Sarah A. Holstein

Abstract Abstract 1354 Multiple myeloma is highly dependent on the interaction of the malignant plasma cells with the bone marrow environment. In particular, adhesion to bone marrow stromal cells stimulates myeloma cell proliferation and contributes to drug resistance. New therapeutic strategies which target the microenvironment that harbors myeloma are needed. We have previously demonstrated that isoprenoid biosynthetic pathway (IBP) and prenyltransferase inhibitors cause inhibitory effects in myeloma cells, particularly those agents which disrupt Rab geranylgeranylation. Our prior studies focused on the effects of these agents in disrupting monoclonal protein secretion and inducing the unfolded protein response. Whether disruption of Rab geranylgeranylation affects other key cellular processes in myeloma cells or the bone marrow microenvironment remains to be determined. In this study, we examined the effects of IBP and prenyltransferase inhibitors on several key aspects of myeloma-stromal cell interactions, including adhesion, proliferation, and cytokine secretion. The specific inhibitors utilized included lovastatin (HMG-CoA reductase inhibitor), zoledronic acid (farnesyl pyrophosphate (FPP) synthase inhibitor), digeranyl bisphosphonate (DGBP) (geranylgeranyl pyrophosphate (GGPP) synthase inhibitor), FTI-277 (farnesyl transferase (FTase) inhibitor), GGTI-2133 (geranylgeranyl transferase (GGTase) I inhibitor) and 3-PEHPC (GGTase II inhibitor). Studies were performed with the HS-5 human bone marrow stromal cell line as well as the RPMI-8226 and U266 human myeloma cell lines. MTT cytotoxicity assays demonstrated that the stromal cells are more sensitive to agents which deplete cells of isoprenoids than to direct prenyltransferase inhibitors. Co-incubation studies utilizing the isoprenoid intermediates mevalonate, FPP, and GGGP, revealed that depletion of GGPP, but not FPP, is responsible for IBP inhibitor-induced cytotoxicity in HS-5 stromal cells. Disruption of protein prenylation at the relevant drug concentrations was confirmed via immunoblot analysis of cytosolic and membranous fractions obtained from Triton X-114 lysis of HS-5 cells. Pre-treatment of U266 cells with lovastatin or 3-PEHPC for 24 hours significantly decreased the adhesion of those cells to either the HS-5 stromal cell layer or to fibronectin. To determine whether IBP or prenyltransferase inhibitors disrupt secretion of cytokines from the HS-5 cells, IL-6 ELISA studies were performed. Treatment of HS-5 cells for 24 hours with concentrations of IBP inhibitors that induce less than a 20% decrease in cell viability resulted in a decrease in IL-6 secretion by 20–40%. Of note, 3-PEHPC, but not the FTase or GGTase I inhibitors, significantly decreased IL-6 secretion. The sensitivity of RPMI-8226 and U266 cells to lovastatin or 3-PEHPC was not significantly altered when co-cultured with HS-5 cells as determined by a BrdU incorporation assay. In conclusion, these studies are the first to provide evidence for the role of Rab proteins in regulating myeloma cell adhesion and to demonstrate that disruption of Rab geranylgeranylation results in diminished stromal cell cytokine secretion. Further studies are required to determine the identity of the specific Rabs mediating these effects and the underlying mechanisms. This work supports the further investigation into agents which inhibit Rab geranylgeranylation as a novel strategy with which to target myeloma cells and the bone marrow microenvironment. Disclosures: No relevant conflicts of interest to declare.


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.


Blood ◽  
2000 ◽  
Vol 96 (5) ◽  
pp. 1953-1960 ◽  
Author(s):  
Toshimi Michigami ◽  
Nobuaki Shimizu ◽  
Paul J. Williams ◽  
Maria Niewolna ◽  
Sarah L. Dallas ◽  
...  

Abstract Myeloma is a unique hematologic malignancy that exclusively homes in the bone marrow and induces massive osteoclastic bone destruction presumably by producing cytokines that promote the differentiation of the hematopoietic progenitors to osteoclasts (osteoclastogenesis). It is recognized that neighboring bone marrow stromal cells influence the expression of the malignant phenotype in myeloma cells. This study examined the role of the interactions between myeloma cells and neighboring stromal cells in the production of osteoclastogenic factors to elucidate the mechanism underlying extensive osteoclastic bone destruction. A murine myeloma cell line 5TGM1, which causes severe osteolysis, expresses α4β1-integrin and tightly adheres to the mouse marrow stromal cell line ST2, which expresses the vascular cell adhesion molecule-1 (VCAM-1), a ligand for α4β1-integrin. Co-cultures of 5TGM1 with primary bone marrow cells generated tartrate-resistant acid phosphatase-positive multinucleated bone-resorbing osteoclasts. Co-cultures of 5TGM1 with ST2 showed increased production of bone-resorbing activity and neutralizing antibodies against VCAM-1 or α4β1-integrin inhibited this. The 5TGM1 cells contacting recombinant VCAM-1 produced increased osteoclastogenic and bone-resorbing activity. The activity was not blocked by the neutralizing antibody to known osteoclastogenic cytokines including interleukin (IL)-1, IL-6, tumor necrosis factor, or parathyroid hormone-related peptide. These data suggest that myeloma cells are responsible for producing osteoclastogenic activity and that establishment of direct contact with marrow stromal cells via α4β1-integrin/VCAM-1 increases the production of this activity by myeloma cells. They also suggest that the presence of stromal cells may provide a microenvironment that allows exclusive colonization of myeloma cells in the bone marrow.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2328-2328
Author(s):  
Katja C. Weisel ◽  
Ying Gao ◽  
Jae-Hung Shieh ◽  
Lothar Kanz ◽  
Malcolm A.S. Moore

Abstract The aorta-gonads-mesonephros (AGM) region autonomously generates adult repopulating hematopoietic stem cells (HSC) in the mouse embryo and provides its own HSC-supportive microenvironment. Stromal cells from adult bone marrow, yolk sac, fetal liver and AGM have been used in coculture systems for analysing growth, maintenance and differentiation of hematopoietic stem cells. We generated &gt;100 cloned stromal cell lines from the AGM of 10.5 dpc mouse embryos. In previous studies, we tested these for support of murine adult and human cord blood (CB) CD34+ cells. We could demonstrate that 25 clones were superior to the MS5 bone marrow stromal cell line in supporting progenitor cell expansion of adult mouse bone marrow both, in 2ndry CFC and CAFC production. In addition we demonstrated that 5 AGM lines promoted in absence of exogenous growth factors the expansion of human CB cells with progenitor (CFC production for at least 5 weeks) and stem cell (repopulation of cocultured cells in NOD/SCID assay) function. Now, we could show that one of the isolated stromal cell lines (AGM-S62) is capable in differentiating undifferentiated murine embryonic stem (mES) cells into cells of the hematopoietic lineage. A sequential coculture of mES-cells with AGM-S62 showed production of CD41+ hematopoietic progenitor cells at day 10 as well as 2ndry CFC and CAFC production of day 10 suspension cells. Hematopoietic cell differentiation was comparable to standard OP9 differentiation assay. With these data, we can describe for the first time, that a stromal cell line other than OP9 can induce hematopoietic differentiation of undifferentiated mES cells. Hematopoietic support occurs independently of M-CSF deficiency, which is the characteristic of OP9 cells, because it is strongly expressed by AGM-S62. To evaluate genes responsible for hematopoietic cell support, we compared a supporting and a non-supporting AGM stromal cell line by microarray analysis. The cell line with hematopoietic support clearly showed a high expression of mesenchymal markers (laminins, thrombospondin-1) as well as characteristic genes for the early vascular smooth muscle phenotype (Eda). Both phenotypes are described for stromal cells with hematopoietic support generated from bone marrow and fetal liver. In addition, the analysed supporting AGM stromal cell line interestingly expressed genes important in early B-cell differentiation (osteoprotegerin, early B-cell factor 1, B-cell stimulating factor 3), which goes in line with data demonstrating early B-cell development in the AGM-region before etablishing of fetal liver hematopoiesis. Further studies will show the significance of single factors found to be expressed in microarray analyses. This unique source of &gt; 100 various cell lines will be of value in elucidating the molecular mechanisms regulating embryonic and adult hematopoiesis in mouse and man.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3473-3473
Author(s):  
Peter M. Voorhees ◽  
George W. Small ◽  
Deborah J. Kuhn ◽  
Qing Chen ◽  
Sally A. Hunsucker ◽  
...  

Abstract Given the critical role that IL-6 plays in MM cell proliferation, survival, and resistance to GCs, we evaluated the ability of CNTO328, a chimeric monoclonal IL-6 neutralizing antibody, to overcome GC resistance in cell line models of human MM. In the presence of IL-6, the MM cell lines ANBL-6 and KAS-6 were resistant to the cytotoxic activity of dexamethasone (Dex) as assessed by cell viability assays both in suspension culture and in the context of patient-derived stromal cells. Resistance to dexamethasone was readily reversed by CNTO328, but not an isotype control antibody, in suspension culture. For example, in the case of the ANBL-6 model, viability was reduced by 12% with CNTO328 alone, 8% with Dex, but 74% with the combination, consistent with a synergistic interaction Given the ability of other growth factors in the bone marrow microenvironment to confer GC resistance in preclinical models of MM, we evaluated the activity of the CNTO328 and Dex combination in ANBL-6 and KAS-6 cells using a physiologically-relevant MM cell/patient-derived bone marrow stromal cell co-culture system. Importantly, bone marrow stromal cells rendered ANBL-6 and KAS-6 cells resistant to Dex in cell viability assays, and CNTO328 was able to reestablish Dex sensitivity, thus confirming a central role of IL-6 in bone marrow stroma-mediated GC resistance. Furthermore, treatment of ANBL-6 and KAS-6 cells with Dex alone did not induce apoptosis in this co-culture system, whereas the combination of CNTO328 and Dex led to a synergistic induction of apoptosis. In KAS-6 cells, IL-6-mediated Dex resistance was not overcome using pharmacologic inhibitors to p38, PI-3 kinase, mTor or MEK, suggesting that other IL-6 signaling pathways are likely involved. In contrast, the mTor inhibitor rapamycin was capable of sensitizing ANBL-6 cells to Dex in the presence of IL-6, suggesting that this pathway may be relevant to IL-6-mediated GC resistance in these cells. Induction of the pro-apoptotic Bcl-2 family member, Bim, has been shown to play an important role in GC-mediated cell death in lymphocytes as well as preclinical lymphoma and acute lymphoblastic leukemia models. Interestingly, although treatment of ANBL-6 cells in the presence of IL-6 with either CNTO328 or dexamethasone did not lead to induction of Bim, the combination led to a 3.3-fold increase in its expression. Taken together, the above data demonstrate that inhibition of IL-6 signaling with CNTO328 can effectively overcome IL-6-mediated GC resistance even in the presence of bone marrow stroma, and provide a compelling rationale for translation of this combination into clinical trials for patients suffering from MM. Furthermore, we show that the ability of CNTO328 to overcome GC resistance may be mediated in part by its ability to reverse IL-6-mediated repression of GC-induced Bim expression. Studies evaluating the relevance of Bim modulation in IL-6-mediated GC resistance and the molecular pathways that mediate this effect are on-going.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4040-4040
Author(s):  
Szabolcs Fatrai ◽  
Simon M.G.J. Daenen ◽  
Edo Vellenga ◽  
Jan J. Schuringa

Abstract Mucin1 (Muc1) is a membrane glycoprotein which is expressed on most of the normal secretory epithelial cells as well as on hematopoietic cells. It is involved in migration, adhesion and intracellular signalling. Muc1 can be cleaved close to the membrane-proximal region, resulting in an intracellular Muc1 that can associate with or activate various signalling pathway components such as b-catenin, p53 and HIF1a. Based on these properties, Muc1 expression was analysed in human hematopoietic stem/progenitor cells. Muc1 mRNA expression was highest in the immature CD34+/CD38− cells and was reduced upon maturation towards the progenitor stage. Cord blood (CB) CD34+ cells were sorted into Muc1+ and Muc1− populations followed by CFC and LTC-IC assays and these experiments revealed that the stem and progenitor cells reside predominantly in the CD34+/Muc1+ fraction. Importantly, we observed strongly increased Muc1 expression in the CD34+ subfraction of AML mononuclear cells. These results tempted us to further study the role of Muc1 overexpression in human CD34+ stem/progenitor cells. Full-length Muc1 (Muc1F) and a Muc1 isoform with a deleted extracellular domain (DTR) were stably expressed in CB CD34+ cells using a retroviral approach. Upon coculture with MS5 bone marrow stromal cells, a two-fold increase in expansion of suspension cells was observed in both Muc1F and DTR cultures. In line with these results, we observed an increase in progenitor counts in the Muc1F and DTR group as determined by CFC assays in methylcellulose. Upon replating of CFC cultures, Muc1F and DTR were giving rise to secondary colonies in contrast to empty vector control groups, indicating that self-renewal was imposed on progenitors by expression of Muc1. A 3-fold and 2-fold increase in stem cell frequencies was observed in the DTR and Muc1F groups, respectively, as determined by LTC-IC assays. To determine whether the above mentioned phenotypes in MS5 co-cultures were stroma-dependent, we expanded Muc1F and DTR-transduced cells in cytokine-driven liquid cultures. However, no proliferative advantage or increase in CFC frequencies was observed suggesting that Muc1 requires bone marrow stromal cells. In conclusion, our data indicate that HSCs as well as AML cells are enriched for Muc1 expression, and that overexpression of Muc1 in CB cells is sufficient to increase both progenitor and stem cell frequencies.


2001 ◽  
Vol 93 (5) ◽  
pp. 674-680 ◽  
Author(s):  
Dirk H�nemann ◽  
Manik Chatterjee ◽  
Rocco Savino ◽  
Kurt Bommert ◽  
Renate Burger ◽  
...  

Blood ◽  
1992 ◽  
Vol 80 (12) ◽  
pp. 3079-3089
Author(s):  
J Mladenovic ◽  
SM Anderson

The S17 murine stromal cell line was infected with retroviral vectors encoding the v-src and c-src oncogenes and cells expressing high levels of either pp60v-src or pp60c-src were isolated. Long-term bone marrow cultures (LTBMCs) established with these different stromal cell lines showed that progenitor cells proliferated to a greater extent in cultures with stromal cells that over-expressed either c-src or v-src. An increase in the number of granulocytes, monocytes, and colony- forming units granulocyte-macrophage (CFU-GM) in the nonadherent cell population of LTBMCs prepared with S17/v-src or S17/c-src stromal cells was observed. Conditioned media from the S17/v-src and S17/src stromal cell lines stimulated the formation of CFU-GM in the absence of additional hematopoietic cell growth factors. Conditioned media from S17/v-src and S17/c-src stimulated proliferation of the granulocyte- macrophage colony-stimulating factor (GM-CSF)-responsive cell line FDCP-1 and this stimulation was inhibited by neutralizing antisera to murine GM-CSF. An increase in the concentration of GM-CSF was confirmed by enzyme-linked immunosorbent assay. No secretion of interleukin-1 alpha (IL-1 alpha) or tumor necrosis factor-alpha was detected by any of the stromal cell lines. There was no increase in the secretion of either CSF-1 or IL-6 by either S17/v-src or S17/c-src. The addition of 1 micrograms/mL monoclonal anti-GM-CSF antibody to LTBMCs caused a decrease in the number of nonadherent cells in cultures established with each of the different stromal cell lines. Northern blot analysis showed no difference in the level of GM-CSF RNA among the different stromal cell lines. These studies suggest that the increased proliferation of hematopoietic progenitor cells in LTBMCs with S17/v-src or S17/c-src cells may result from a posttranscriptional event that elevates production of GM-CSF by the S17/c-src and S17/v-src stromal cells.


Sign in / Sign up

Export Citation Format

Share Document