Neutrophil Elastase Is Important for Several Activities of PML-RARα in Early Myeloid Cells.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 486-486
Author(s):  
Andrew A. Lane ◽  
Timothy J. Ley

Abstract Acute promyelocytic leukemia (APL) is associated with the accumulation of promyelocytes that usually carry the t(15;17) translocation, generating the PML-RARα fusion gene. APL develops in mice only if PML-RARα expression is targeted to early myeloid cells; when PML-RARα is expressed in all hematopoietic cells, the only malignancy that develops is APL. These observations have suggested that there may be a component(s) of the early myeloid environment that is important for the actions of PML-RARα. We recently showed that murine bone marrow extracts and a human early myeloid cell line, U937, contain a serine protease activity that cleaves PML-RARα. The dominant cleaving activity was attributable to an early myeloid-associated protease, neutrophil elastase (NE). The penetrance of APL in mice expressing PML-RARα in early myeloid cells was significantly reduced in NE-deficient animals (Lane and Ley, Cell 115:305–318, 2003). To determine whether measurable functions of PML-RARα require NE, we performed a series of experiments. Using a GFP-PML-RARα fusion construct, we determined that expression of PML-RARα caused disruption of PML oncogenic domains (PODs) in several human hematopoietic cell lines, regardless of whether they expressed NE. However, when PML-RARα was expressed at high levels in cell lines that contained NE activity, substantial toxicity was observed. In contrast, PML-RARα did not cause toxicity in cell lines without NE activity. Similarly, when we expressed an NE-resistant PML-RARα cDNA (containing valine to arginine mutations at the P1 amino acid of the two preferred NE cleavage sites), toxicity was abrogated in clonogenic assays. The toxic effects of expressing full-length PML-RARα could not be recapitulated by expressing either dominant cleavage fragment alone, or in combination, which suggests that NE-induced cleavage may not directly create the “active” fragment of PML-RARα. Primary human and mouse primary APL cells contain abundant NE activity and PML-RARα-cleaving activity. However, we determined that two commonly used APL cell line models (NB4 and U937-PR9 cells) contain abundant full length PML-RARα protein, but neither contains NE activity nor PML-RARα-cleaving activity. As expected, high levels of expression of PML-RARα in PR9 cells did not cause toxicity. Using an in vitro G-CSF-dependent myeloid differentiation assay, we found that purified hematopoietic progenitors from mCGPML-RARα knock-in mice demonstrated markedly increased proliferation of early (blasts and promyelocytes) myeloid cells compared to wild type progenitors. This increase was not observed in progenitors from mCGPML-RARαNE−/− animals. The difference was not due to an intrinsic defect in myeloid development due to NE deficiency, since NE−/− progenitors developed normally in response to G-CSF in vitro. To extend these results, we performed competitive repopulation bone marrow transplants using NE+/+ and NE−/− mice as bone marrow donors; at 3 weeks after hematopoietic reconstitution, there was no difference in the contributions of either genotype to multi-lineage hematopoiesis. Together, these data strongly suggest that NE is important for several of the measurable activities of PML-RARα in early myeloid cells; we are therefore exploring pharmacologic NE inhibition as an approach to reduce the growth of APL cells. Our data also suggest that the physiologic activities of PML-RARα should be studied in early myeloid cells that contain NE activity.

1986 ◽  
Vol 6 (9) ◽  
pp. 3221-3231
Author(s):  
R C Schwartz ◽  
L W Stanton ◽  
S C Riley ◽  
K B Marcu ◽  
O N Witte

Murine bone marrow was either singly or doubly infected with retroviral vectors expressing v-myc (OK10) or v-Ha-ras. The infected bone marrow was cultured in a system that supports the long-term growth of B-lineage lymphoid cells. While the v-myc vector by itself had no apparent effect on lymphoid culture establishment and growth, infection with the v-Ha-ras vector or coinfection with both v-myc and v-Ha-ras vectors led to the appearance of growth-stimulated cell populations. Clonal pre-B-cell lines stably expressing v-Ha-ras alone or both v-myc and v-Ha-ras grew out of these cultures. In comparison with cell lines expressing v-Ha-ras alone, cell lines expressing both v-myc and v-Ha-ras grew to higher densities, had reduced dependence on a feeder layer for growth, and had a marked increase in ability to grow in soft-agar medium. The cell lines expressing both oncogenes were highly tumorigenic in syngeneic animals. These experiments show that the v-myc oncogene in synergy with v-Ha-ras can play a direct role in the in vitro transformation of murine B lymphoid cells.


Blood ◽  
1987 ◽  
Vol 70 (6) ◽  
pp. 1777-1783 ◽  
Author(s):  
SL Kirby ◽  
SA Bentley

There is evidence indicating that stromal proteoglycans are an important functional component of the hematopoietic microenvironment. Proteoglycan synthesis was therefore investigated in the MS3–2A and D2XRII hematopoietic stromal cell lines. These lines differ in their capacity to support hematopoiesis in vitro, D2XRII supporting in vitro hematopoiesis, whereas MS3–2A does not. Cells were labeled with 35S- sulfate as precursor, and 4 mol/L guanidine HCl extracts of cells and media were analyzed by ion-exchange chromatography, cesium chloride density gradient centrifugation, and molecular sieve chromatography. Proteoglycans were further examined by enzymatic and chemical digestions. MS3–2A cells produced at least three proteoglycan species. Two chondroitin/dermatan sulfate (CS/DS) proteoglycans, Kav = 0.40 and Kav = 0.68 on Sepharose CL-2B, were present primarily in the medium. The respective glycosaminoglycan molecular weight (mol wt) values were 38 kd and 40 kd. A heparan sulfate (HS) proteoglycan of Kav = 0.58 and glycosaminoglycan mol wt 36 kd was present primarily in the cell layer extract. D2XRII cells synthesized two HS proteoglycans. The larger (Kav = 0.45; glycosaminoglycan mol wt, 30 kd) was of low density on gradient centrifugation and more prominent in the cell layer extracts, whereas the smaller (Kav = 0.68; glycosaminoglycan mol wt, 38 kd) was dense and present mainly in the culture medium. A single CS/DS proteoglycan species of Kav 0.78 and average glycosaminoglycan of mol wt 18 kd was present in roughly equal amounts in the medium and in the cell layer. MS3–2A and D2XRII thus appear phenotypically distinct with respect to proteoglycan synthesis. These differences are discussed in relation to the microenvironmental function of bone marrow stromal elements.


2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Sho Nakai ◽  
Shutaro Yamada ◽  
Hidetatsu Outani ◽  
Takaaki Nakai ◽  
Naohiro Yasuda ◽  
...  

Abstract Approximately 60–70% of EWSR1-negative small blue round cell sarcomas harbour a rearrangement of CIC, most commonly CIC-DUX4. CIC-DUX4 sarcoma (CDS) is an aggressive and often fatal high-grade sarcoma appearing predominantly in children and young adults. Although cell lines and their xenograft models are essential tools for basic research and development of antitumour drugs, few cell lines currently exist for CDS. We successfully established a novel human CDS cell line designated Kitra-SRS and developed orthotopic tumour xenografts in nude mice. The CIC-DUX4 fusion gene in Kitra-SRS cells was generated by t(12;19) complex chromosomal rearrangements with an insertion of a chromosome segment including a DUX4 pseudogene component. Kitra-SRS xenografts were histologically similar to the original tumour and exhibited metastatic potential to the lungs. Kitra-SRS cells displayed autocrine activation of the insulin-like growth factor 1 (IGF-1)/IGF-1 receptor (IGF-1R) pathway. Accordingly, treatment with the IGF-1R inhibitor, linsitinib, attenuated Kitra-SRS cell growth and IGF-1-induced activation of IGF-1R/AKT signalling both in vitro and in vivo. Furthermore, upon screening 1134 FDA-approved drugs, the responses of Kitra-SRS cells to anticancer drugs appeared to reflect those of the primary tumour. Our model will be a useful modality for investigating the molecular pathology and therapy of CDS.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 616-616 ◽  
Author(s):  
Deepa B. Shankar ◽  
Jenny C. Chang ◽  
Bertrand Parcells ◽  
Salemiz Sandoval ◽  
Junling Li ◽  
...  

Abstract Children with acute myeloid leukemia (AML) have less than 60% overall survival despite aggressive chemotherapy and bone marrow transplantation. Only one third of the adult patients diagnosed with AML will be cured. AML blast cells from up to 30% of patients express a constitutively active receptor tyrosine kinase, FLT3-ITD, which contains an internal tandem duplication in the juxtamembrane domain. Patients with FLT3-ITD have a worse prognosis. ABT-869 is a novel multi-targeted small molecule inhibitor of receptor tyrosine kinases and is a potent inhibitor of FLT3, c-Kit, and all members of the VEGF and PDGF receptor families. To determine the effects of ABT-896 on AML cells, we treated AML cell lines, primary cells, and tumors in xenograft models with varying concentrations of the drug. In vitro viability assays showed that ABT-869 inhibited the growth of two different cell lines, MV-4-11 (human AML cell line that expresses FLT3-ITD) and BAF3-ITD (murine B-cell line stably transfected with the FLT3-ITD) at an IC50 of 10nM. ABT-869 was also effective against another mutation of FLT3, D835V, but at higher concentrations (IC50 of 100nM). Phosphorylation of FLT3 and activation of downstream signaling molecules, STAT5 and ERK, were inhibited by ABT-869 in a concentration-dependent manner. Cells were also stained with Annexin V-FITC and Propidium Iodide, and analyzed using FACS. ABT-869 induced apoptosis, caspase-3 activation, and PARP cleavage after 48 hours. To examine the in vitro effects of ABT-869 on normal hematopoietic progenitor cells, we performed methylcellulose-based colony assays with human bone marrow. No significant difference was observed in the number and type of colonies formed using BM cells treated with ABT-869 or control, up to a concentration of 1 micromolar. These results suggest that ABT-869 is not toxic to normal bone marrow progenitor cells at concentrations that are effective against AML cells. To examine the effects of ABT-869 in vivo, we treated SCID mice injected with MV-4-11, Baf3-ITD, Baf3-D835V, or Baf3-WT cells, with oral preparations of ABT-869. Complete regression of MV-4-11 tumors was observed in mice treated with ABT-869 at 20 and 40 mg/kg/day. No adverse effects were detected in the peripheral blood counts, bone marrow, spleen or liver. Histology of the tumors from the control-treated group showed a high degree of proliferation by Ki-67 staining, increased mitotic figures, and a well-defined tumor mass. In contrast, the tumors from mice treated with ABT-869 showed a number of apoptotic bodies by TUNEL staining and the presence of reactive, inflammatory cells. Interestingly, we also observed that mice that received ABT-869 the day after injection of AML cells remained tumor-free for over 2 months in contrast to the mice receiving the vehicle alone. Inhibition of FLT3 phosphorylation was demonstrated in the tumors from mice treated with ABT-869. We are evaluating the activity of ABT-869 treatment of SCID mice injected with Baf3-ITD, Baf3-D835V, or Baf3-WT cells. NOD-SCID mouse models are currently being used to analyze the effects of ABT-869 on primary AML cells in vivo. Our preclinical studies demonstrate that ABT-869 is effective and nontoxic, and provide rationale for the treatment and prevention of relapse in AML patients.


Blood ◽  
1985 ◽  
Vol 66 (2) ◽  
pp. 447-455 ◽  
Author(s):  
D Zipori ◽  
J Toledo ◽  
K von der Mark

Abstract Study of a series of stromal cell lines from mouse bone marrow (MBA) verified and extended their classification as phenotypically distinct subtypes. Production of extracellular matrix proteins was examined using specific antibodies. Fibronectin and laminin were detected in all of the cell lines tested, yet 14F1.1 adipocytes exhibited particularly prominent extracellular deposition. This cell line and MBA-13.2 cells were positive to both collagen types I and IV, whereas MBA-1 and MBA- 2.1 were stained with anticollagen type I antibodies only. Coculture experiments revealed differences among the lines in their effects on normal myeloid cells and leukemic cell lines. In promoting the in vitro accumulation of myeloid progenitors (CFU-C), 14F1.1 cells surpassed the others. The MBA-2.1 cell line was particularly inhibitory to MPC-11 plasmacytoma and Friend erythroleukemia cells. However, the latter were refractory to other stromal cell lines, whereas MPC-11 cells were inhibited to various degrees by virtually all of the cell lines. Physical separation between the interacting cells reduced the inhibition in some but not all cases, and no inhibitory activity was detected in conditioned media. The MBA-13 stromal cells synergistically promoted the differentiation of dimethylsulfoxide (Me2SO)-induced Friend erythroleukemia. The latter cells themselves, at high concentrations, as well as some of the stromal cell lines and unrelated adherent cells, antagonized the Me2SO effect, revealing possible reversible stages in the Friend cell differentiation pathway.


Blood ◽  
2004 ◽  
Vol 104 (4) ◽  
pp. 1127-1136 ◽  
Author(s):  
Rhonna M. Gurevich ◽  
Peter D. Aplan ◽  
R. Keith Humphries

AbstractChromosomal rearrangements of the 11p15 locus have been identified in hematopoietic malignancies, resulting in translocations involving the N-terminal portion of the nucleoporin gene NUP98. Fifteen different fusion partner genes have been identified for NUP98, and more than one half of these are homeobox transcription factors. By contrast, the NUP98 fusion partner in t(11;20) is Topoisomerase I (TOP1), a catalytic enzyme recognized for its key role in relaxing supercoiled DNA. We now show that retrovirally engineered expression of NUP98-TOP1 in murine bone marrow confers a potent in vitro growth advantage and a block in differentiation in hematopoietic precursors, evidenced by a competitive growth advantage in liquid culture, increased replating efficient of colony-forming cells (CFCs), and a marked increase in spleen colony-forming cell output. Moreover, in a murine bone marrow transplantation model, NUP98-TOP1 expression led to a lethal, transplantable leukemia characterized by extremely high white cell counts, splenomegaly, and mild anemia. Strikingly, a mutation to a TOP1 site to inactivate the isomerase activity essentially left unaltered the growth-promoting and leukemogenic effects of NUP98-TOP1. These findings, together with similar biologic effects reported for NUP98-HOX fusions, suggest unexpected, overlapping functions of NUP98 fusion genes, perhaps related to common DNA binding properties. (Blood. 2004;104:1127-1136)


Blood ◽  
1991 ◽  
Vol 78 (2) ◽  
pp. 304-309 ◽  
Author(s):  
A Peled ◽  
D Zipori ◽  
O Abramsky ◽  
H Ovadia ◽  
E Shezen

Human fibrotic bone marrow (BM) stroma has been shown to contain alpha- smooth muscle actin (alpha-SMA)-positive cells. These closely resemble myofibroblasts that were described in other fibrotic tissues. We studied the expression of alpha-SMA in a series of murine BM-derived stromal cell lines to investigate the cellular origin and functional significance of myofibroblast-like cells in hematopoietic tissues. Although these cell lines differed in their biologic properties, most of them expressed alpha-SMA under certain conditions. Cells expressing alpha-SMA constituted a minor population in post-confluent, growth- arrested cultures. However, the incidence of cells expressing alpha-SMA increased significantly when cultures were transferred to nonconfluent conditions. A similar increase in alpha-SMA-positive cells occurred after a strip of cells was scraped away from the confluent cell layer; the cells of the affected area acquired alpha-SMA-positive contractile phenotype. The relationship between alpha-SMA expression and hematopoietic activity was studied using a cloned cell line of BM origin (14F1.1). The ability of these endothelial-adipocyte cells to support hematopoiesis in vitro was maximal under confluent conditions, whereas their expression of alpha-SMA under such conditions was residual. Moreover, in long-term BM cultures supported by confluent 14F1.1 cells, stromal areas associated with proliferating hematopoietic precursors, known as “cobblestone areas,” were devoid of alpha-SMA- positive cells. These observations suggest that the expression of alpha- SMA is reversible and inversely related to hematopoietic activity.


1981 ◽  
Vol 50 (1) ◽  
pp. 281-297
Author(s):  
M. Lanotte ◽  
T.D. Allen ◽  
T.M. Dexter

Morphological, enzymic and antigenic data are presented regarding a human bone-marrow stromal cell line maintained for 10 months and subcultured weekly. The main characteristics are a fibroblastoid morphology, diffuse growth in collagen gels, no colony formation in soft gel media, contact inhibition of growth and conversion to adipocytes when treated with hydrocortisone. The cells are non-phagocytic and membrane Fc receptors (i.e. aggregated human immunoglobulin G receptors) are absent, but they show diffuse cytoplasmic non-specific esterase activity, a strong acid phosphatase reaction, and a negative immunofluorescence (direct and indirect) against factor VIII antigen. Other cell lines also have been isolated and maintained in culture and present similar characteristics. These cell lines are thought to be derived from the acid-phosphatase-positive marrow stroma directly associated with bone trabecular matrix and probably represent a component of the haemopoietic inductive microenvironment. As such, they may provide a useful tool for studies in vitro of cell interactions and regulatory processes in the control of human bone-marrow haemopoiesis.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2561-2561
Author(s):  
Marcin Wysoczynski ◽  
Katarzyna Miekus ◽  
Anna Marcinkowska ◽  
Anna Janowska-Wieczorek ◽  
Mariusz Z. Ratajczak

Abstract Rhabdomyosarcoma (RMS) and skeletal muscle-derived tumors frequently infiltrate bone marrow (BM). We have demonstrated that the stromal-derived factor (SDF)-1-CXCR4 receptor (Blood2002;100:2597) and hepatocyte growth factor (HGF)-c-Met receptor (Cancer Res. 2003;63:7926) play an important role in RMS metastasis to BM. Leukemia inhibitory factor (LIF) is a well known factor that plays an important role in skeletal muscle development/regeneration and similarly as SDF-1 and HGF is secreted by BM stroma. This prompted us to examine whether the LIF-LIF receptor (LIF-R) axis affects the biology/metastasis of RMS cells. We employed in our studies, human established RMS cell lines, as well as RMS samples isolated from patients and noticed that LIF-R was expressed not only on established human RMS cell lines (7/7) but more importantly, it was also detectable in patient samples (23/23). We also found that in RMS cells LIF stimulatesphosphorylation of MAPKp42/44, AKT and STAT3,chemotaxis and adhesion andincreases resistance to cytostatics (e.g., etoposide). These LIF-mediated effects were inhibited after downregulating the LIF-R by siRNA. To learn more on the biological significance of the LIF-LIF-R axis in vivo we employed two models. First, human RMS cells (RH-30) were exposed or not exposed to LIF-R siRNA and subsequently injected into SCID™-Beige immunodeficient mice. To estimate the number of RMS cells that seed to BM and liver in these animals, we isolated DNA and using real- time RT-PCR, amplified human a-satellite sequences and murine b-actin. The number of human cells present in murine organs was subsequently calculated from a standard curve derived from mixing varying numbers of human cells with a constant number of murine cells. We noticed that downregulation of LIF-R by siRNA significantly decreased the number of human RMS cells in murine BM and liver (x4 and x2 respectively). In a second model, the RH30 cell line was selected by repetitive chemotaxis for cells that are highly responsive to LIF (RH-30 L) and subsequently the cells from parental RH-30 cell line and RH-30 L cells were injected intramuscularly. Six weeks after tumour inoculation, we detected more metastasis in bone marrow and lungs in mice injected with RH-30L cells as compared to parental RH-30 clone (x6 and x3 respectively). In conclusion, we present evidence for the first time that the inhibition of LIF-LIF-R axis may decrease the invasive potential of human RMS both in vitro and in vivo. Hence, molecular targeting of LIF-LIF-R axis could possibly become a more effective new strategy to control the progression and metastasis of RMS.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 378-378 ◽  
Author(s):  
Alba Matas-Céspedes ◽  
Anna Vidal Crespo ◽  
Vanina Rodriguez ◽  
Gael Roue ◽  
Armando Lopez-Guillermo ◽  
...  

Abstract Daratumumab (DARA) is a human CD38 antibody with broad-spectrum killing activity. DARA induces killing of tumor cells, mainly via complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC) (de Weers M, J Immunol 2011). DARA is currently being evaluated in phase I/II clinical trials in patients with multiple myeloma. In these clinical studies the adverse events have been manageable and marked reductions in paraprotein and bone marrow plasma cells have been observed. We have previously reported (Blood (ASH annual meeting abstracts). Nov 2012, 120 (21): 3935) that DARA induces cytotoxic activity in vitro via ADCC in primary cells and cell lines from Mantle Cell Lymphoma (MCL), Follicular Lymphoma (FL) and Chronic Lymphoctic Leukemia (CLL). CDC induction was low, which is associated to high expression of the complement inhibitors and reduced number of CD38 molecules per cell in these indications. This suggests a threshold for CD38-targeted CDC lysis. In addition, based on known interactions between CD38-CXCR4, we also demonstrated that in the CLL subtype, with high CD38 and more migratory capacity, DARA (10-30 µg/mL) inhibited in vitro CXCL12/SDF1α-mediated migration up to 70%. Here, we present the first preclinical in vivo results of DARA in mouse models of MCL, transformed FL(tFL) and CLL. We generated heterotopic and systemic mouse models of these entities by subcutaneous or intravenous inoculation of tumor cell lines in SCID mice, that retain both NK cells and macrophages as potential effector cells. In MCL (REC cell line) and tFL (RL cell line) subcutaneous mouse models, we tested DARA activity in both prophylactic (treatment initiation simultaneous to lymphoma cell inoculation) and therapeutic settings(treatment initiation one week after lymphoma cell inoculation, when tumors were about 100 mm3 in size). In the prophylactic setting, mice received 3 doses of DARA or control IgG bi-weekly (20/10/10 mg/kg). In the therapeutic setting, treatment was intensified to 4 doses of DARA or control IgG weekly (20/10/10/10 mg/kg). In both schedules, mice were sacrificed one week after the last dose. DARA completely abrogated tumor growth of REC or RL cells in the prophylactic setting. Moreover, in the therapeutic setting, DARA induced total tumor regression of REC tumors in 4 out of 6 mice, and prevented the splenomegaly observed in control IgG treated mice. In the case of tFL and therapeutic setting, DARA reduced 60% of tumor growth compared to control IgG treated mice at day 32, when experiment was finished. In CLL, we analyzed the effect of DARA on cell homing to lymphoid organs together with its therapeutic properties in a systemic CLL mouse model. Using NOD/SCID/gamma null mice (lacking NK cells and effective macrophages), we analyzed the effect of DARA on primary CLL cell migration from Peripheral Blood (PB) to bone marrow (BM) and Spleen. In this system, NSG mice were pretreated (day 0) with DARA, control IgG or anti-CXCR4 as positive control for inhibition of cell homing, followed by fresh CLL cell inoculation (50×106 cells/per mice) on day 1. PB, BM and spleen cells were isolated on day 2 and CLL cells were identified by staining for CD45/CD19/CD5 and counted using a flow cytometer. Cell counting showed that CLL cells mainly migrate to the spleen, and that DARA significantly reduced this migration (55% inhibition on average, p<0.05). Migration of CLL cells to BM was limited and was not affected by pretreatment of mice with DARA. Finally, we tested DARA therapeutic activity in a systemic CLL mouse model (MEC2 cell line), following the schedule described before (4 doses of control IgG/ DARA weekly (20/10/10/10 mg/kg)), and assessed efficacy on mice overall survival. Mice treated with control IgG started to lose weight and showed signs of disease between days 30-40 and were sacrificed for ethical reasons. In the DARA treated group, in 7 out of 8 mice survival was extended up to day 90, when the experiment was stopped. In conclusion, DARA demonstrated strong in vivo activity in immunocompromised mouse models of MCL, tFL and CLL cell lines and interfered with homing of primary CLL cells to the spleen. These results warrant further investigation of DARA in clinical trials for these indications. Disclosures: Lopez-Guillermo: Roche: Membership on an entity’s Board of Directors or advisory committees. Lammerts van Bueren:Genmab: Employment, Stocks Other. Bakker:Genmab: Employment, Stocks Other. Parren:Genmab: Employment, Stocks Other. Perez-Galan:Genmab: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document