Myeloma Cell Interaction with Osteoclasts and Mesenchymal Stem Cells Reveals Genes Associated with Post Relapse Survival

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2957-2957
Author(s):  
Igor Entin ◽  
Shmuel Yaccoby ◽  
Wen Zhining ◽  
John Shaughnessy ◽  
Bart Barlogie ◽  
...  

Abstract Abstract 2957 Myeloma is intimately associated with osteolytic bone disease, resulting from myeloma cells' interactions with osteoclasts and osteoblasts and their progenitors, and is dependent on the changes it induces in bone metabolism for progression. Myeloma cell dependence on the bone marrow microenvironment is also evident experimentally, where interaction of primary myeloma plasma cells (MMPC) with osteoclasts (OC) and with mesenchymal stem cells (MSC) support the survival of primary myeloma cells. To understand the molecular mechanisms associated with the survival of MMPC, we used Acuity 4 software to analyze Affymetrix U133 Plus2 chip data and identify changes in gene expression in induced MMPC freshly isolated from 8 patients by interaction with OC and from 8 additional patients with MSC. Expression by MMPC of 675 genes was changed following interaction with OC; 552 genes were upregulated and 123 down regulated. Expression of 296 genes was changed in MSC co cultures (161 upregulated, 135 down regulated). Comparison of the genes whose expression was similarly changed in both co culture systems identified 72 probesets, representing 58 genes, that were commonly changed; 33 were upregulated and 25 down regulated. Ingenuity Pathway Analysis assigned 54 of the 58 genes to 4 distinguished networks of interrelated genes with high probability scores. We next tested the hypothesis that the expression of genes whose expression was commonly changed in the co culture systems has clinical significance. To accomplish this, we used gene expression data available on 127 relapsed patients who had been uniformly treated on our Total Therapy 2 protocol, and for whom gene expression (GEP) data at first relapse (RL) were available. 71 of these patients also had pre treatment (BL) GEP data; for these 71 patients we calculated change in expression of the 72 probesets as the ratio of RL/BL expression signal. We identified 7 genes whose expression changes were significantly (p≤0.05) associated with survival after relapse: These genes were, in order of significance: CCNE2, PECAM1, KLHL21, ICAM1, PLAU, ANPEP, and DUSP1, with p-values ranging from 0.017 to 0.05. Up regulation of PECAM1, ANPEP, DUSP1, and down regulation of CCNE2, KLHL21, ICAM1, and PLAU were associated with longer survival. We further determined whether expression level of these 72 probesets at relapse, defined by signal intensity, correlated with post relapse survival of the 127 patients; 18 genes were significantly (p<0.05)associated with survival: of these, the top 6 genes, sorted in order of p-values of the univariate test were CCNE2 (p<1e-7, HR, defined as ratio of hazard for a twofold increased in signal) =1.83), PECAM1 (p=2e-7, HR=0.64), FOSB (p=2.3e-6, HR=0.76), HMOX1 (p=8.2e-5; HR=0.72), CISH (p<0.0002, HR=0.76), and JUN (p=0.0008, HR=0.78). Eleven other genes associated with survival had p values ranging from <0.002 to 0.047. Although not the purpose of this work, we also tested the ability of the 72 probesets to predict survival with each probeset dichotomized at the median. Using the BRB Array tool, we found that 17 genes predict post relapse survival with at p=0.01 based on log-rank tests in 100 permutations. The percent variability explained by the first 2 principal components = 42.5. Using co culture of myeloma cells with osteoclasts and MSC we identified MMPC genes whose expression is associated with the survival of patients after relapse. These genes define potential targets for improving the survival of relapsed myeloma patients. Disclosures: No relevant conflicts of interest to declare.

2019 ◽  
Vol 208 (1-2) ◽  
pp. 48-58
Author(s):  
Marzieh Hosseini ◽  
Saghar Salehpour ◽  
Marefat Ghaffari Novin ◽  
Zahra Shams Mofarahe ◽  
Mohammad-Amin Abdollahifar ◽  
...  

Follicular loss and tissue degeneration are great challenges in ovarian tissue culture systems. Mesenchymal stem cells (MSC) secrete a cocktail of growth factors and cytokines which supports adjacent cells and tissues. The aim of the current study was to investigate the impact of human bone marrow (hBM)-MSC, as co-culture cells, on human follicular development in ovarian cortical tissue (OCT) culture. For this purpose, warmed OCT fragments were co-cultured with hBM-MSC for 8 days and compared to monocultured OCT. During the culture period, ovarian follicle survival and development in the OCT were evaluated using histological observation, follicular developmental-related genes expression, and estradiol production. Furthermore, cell proliferation and apoptosis were assessed. The results showed that there were no significant differences in conserved ovarian follicles with a normal morphology between the two groups. However, the percentage of developing follicles, as well as follicular developmental gene expression, significantly increased in the co-culture group compared to the monoculture group. On the other hand, compared with the monoculture group, the co-culture group demonstrated a significant increase in cell proliferation, indicated by Ki67 gene expression, as well as a dramatic decrease in apoptotic cell percentage, revealed by TUNEL assay. These findings indicated that co-culturing of hBM-MSC with OCT could improve follicular activation and early follicular development in human ovarian tissue culture systems.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2432-2432 ◽  
Author(s):  
James Q Yin ◽  
Chunji Gao ◽  
Bing Han ◽  
Jianliang Sheng

Abstract Introduction Naturally-occurring regeneration of cells and tissues is generally involved in four working mechanisms such as directed differentiation, dedifferentiation, trans-differentiation and transdetermination. The better exploring of these mechanisms could be beneficial to develop clinical strategies for regenerative medicine and to reduce the likelihood of immune rejection and relevant complications Recently, “trans-determination” has attracted great controversy, mostly in regards to whether adult stem cells can colonize other tissues after transplantation. More importantly, how to generate large amounts of a particular stem cell type through a transdetermination process remained to be unsolved. Similarly, it is unclear whether mesenchymal stem cells (MSCs) can transdeterminate into hematopoietic stem cells (HSCs). Methods Many technologies were used to validate the transdetermination of adipose-derived mesenchymal stem cells (AD-MSCs) into hematopoietic stem cells (HSCs) from different aspects. They include FACS analysis, PCR tests, immunostaining, expansion and repopulating assays, transplantation analysis and others, showing their in vivo and in vitro potentials for long-term self-renewal and differentiation into multi-lineages of blood cells. Moreover, these AD-HSCs can reconstitute hematopoietic function in six patients. Results We report firstly here that a huge number of human AD-MSCs that are CD44+,CD29+, CD105+, CD166+,CD133-,CD34- could rapidly transdifferentiate into hematopoietic stem cells (CD49f+/CD133+/CD34+) and their descending blood cells in vitro, after transfected with two small RNAs. The sRNAs were high-effectively delivered into MSCs by a novel peptide means. These adipose-derived HSCs (AD-HSCs) could form different types of hematopoietic colonies as nature-occurring HSCs did. Upon the primary and secondary transplantation into sublethally or lethally irradiated mice, these MSC-HSCs engrafted and differentiated into all hematopoietic lineages such as erythrocytes, lymphocytes, myelocytes and thrombocytes. Furthermore, we demonstrated the first evidence that the transdetermination of MSCs was induced by acetylation of histone proteins and activation of many transcriptional factors. More excitingly, these MSC-derived HSCs can reconstitute hematopoietic function in six patients with severe aplastic anemia. Conclusion our findings identify the molecular mechanisms that regulate the directed transdifferentiation of MSCs toward HSCs, create a new source for individual HSC transplantation used for the treatment of blood diseases and cancers, and break the stalemate caused by bone marrow match and graft-versus-host disease. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2458-2458 ◽  
Author(s):  
Bruno B. Sangiorgi ◽  
Vitor Leão ◽  
Josiane Lilian dos Santos Schiavinato ◽  
Maristela Delgado Orellana ◽  
Marco A. Zago ◽  
...  

Abstract Mesenchymal stem cells (MSC) represent a promising cell-based therapy for Graft Versus Host Disease (GVHD), for they can suppress the proliferation of activated lymphocytes in vitro, through either direct contact or secretion of anti-inflammatory cytokines. However, clinical trials performed in the past decade evaluating the effectiveness of infused MSC to improve the outcome of GVHD patients had variable results so far. Since the presence of associated infections are frequent in patients with GVHD, and given that priming to different Toll-like receptors (TLR) may elicit opposite immunosuppressory proprieties in MSC, in this study we investigated the modulation of the immune suppressive potential of bone marrow MSC (BM-MSC) by different TLR ligands. Thereby, BM-MSC (n=3) were primed for 24h with LPS (1µg/ml) and/or the CpG oligonucleotide CpG ODN DSP30 (1µM), to stimulate the TLR4 and TLR9, respectively. After that, peripheral blood CD3+ lymphocytes were labeled with the fluorescent dye CFSE, activated with antibody-loaded beads anti CD2, CD3 and CD28, and cocultured for 5 days with treated or untreated BM-MSC. Suppression of lymphocyte proliferation, by BM-MSC in different conditions, was evaluated by flow cytometry. As compared to untreated control MSC, the immunosuppressive capacity of TLR4 primed BM-MSC was inhibited by 30% (p<0.05), while that of TLR9 primed BM-MSC was enhanced by 32% (p<0.05). As a result, simultaneous priming of MSC with both ligands kept immunosuppression at levels comparable to that of untreated BM-MSC. In order to identify the possible mechanisms involved in this modulatory activity, BM-MSC (n=3) in same conditions were evaluated for changes in proliferation, using an assay of EdU incorporation into DNA and by direct cell count of fluorescently labeled cells, using an automated High Content Screening (HCS) imaging device. As results, we observed that TLR9 priming significantly promoted the proliferation of BM-MSC, as demonstrated by higher (p<0.05) EdU incorporation and cell count; while TLR4 priming had no effect in these parameters. Since priming by TLR4 and TLR9 may lead to the respective activation of canonical and non-canonical NF-kB pathways (which are mediated by the transcription factors RelA and RelB, respectively); we used chromatin immunoprecipitation (ChIP) to evaluate the binding of RelA and RelB to the gene promoter of the vascular cell adhesion molecule-1 (VCAM-1) in BM-MSC (n=3) primed of TLR4 and/or TLR9. In addition, expression of RelA or RelB in BM-MSC (n=3) in the same conditions, was quantified using a HCS device. Priming of TLR4 led, exclusively, to the binding of RelA, (three fold enrichment, compared to unprimed BM-MSC). Moreover, simultaneous priming of TLR4 and TLR9 resulted in an increased binding of both RelA and RelB. Finally, increased staining of RelB was also verified by HCS, in simultaneously primed with ligands for TLR4 and TLR9. Altogether, our results indicates that BM-MSC had its immunosuppressory capacity hampered by priming of TLR4 by LPS (possibly due to the activation of canonical NF-Kb pathway), while TLR9 priming by CpG oligonucleotide enhances the immune suppression, which may be related to an increase in proliferation and, possibly, due to the activation of non-canonical NF-Kb pathway in MSC simultaneously primed for TLR4 and TLR9. These observations may help to elucidate the cellular and molecular mechanisms underlying the heterogeneous results in clinical studies of GVHD patients receiving BM-MSC infusions. As patients subjected to bone marrow transplantation are more prompt to infections by different pathogens (potentially exposing BM-MSC to different TLR ligands), the identification of the CpG oligonucleotide as a TLR9 ligand capable of enhancing and protecting the immunosuppressive activity of BM-MSC, may lead to the establishment of a laboratory protocol aiming the priming to TLR9, or the specific activation of non-canonical NF-Kb pathways, in order to boost BM-MSC efficiency in clinical settings aiming at the treatment of GVHD patients. Disclosures: No relevant conflicts of interest to declare.


Molecules ◽  
2021 ◽  
Vol 26 (3) ◽  
pp. 606
Author(s):  
Nihal AlMuraikhi ◽  
Hanouf Alaskar ◽  
Sarah Binhamdan ◽  
Amal Alotaibi ◽  
Moustapha Kassem ◽  
...  

Several signalling pathways, including the JAK/STAT signalling pathway, have been identified to regulate the differentiation of human bone marrow skeletal (mesenchymal) stem cells (hBMSCs) into bone-forming osteoblasts. Members of the JAK family mediate the intracellular signalling of various of cytokines and growth factors, leading to the regulation of cell proliferation and differentiation into bone-forming osteoblastic cells. Inhibition of JAK2 leads to decoupling of its downstream mediator, STAT3, and the subsequent inhibition of JAK/STAT signalling. However, the crucial role of JAK2 in hBMSCs biology has not been studied in detail. A JAK2 inhibitor, Fedratinib, was identified during a chemical biology screen of a small molecule library for effects on the osteoblastic differentiation of hMSC-TERT cells. Alkaline phosphatase activity and staining assays were conducted as indicators of osteoblastic differentiation, while Alizarin red staining was used as an indicator of in vitro mineralised matrix formation. Changes in gene expression were assessed using quantitative real-time polymerase chain reaction. Fedratinib exerted significant inhibitory effects on the osteoblastic differentiation of hMSC-TERT cells, as demonstrated by reduced ALP activity, in vitro mineralised matrix formation and downregulation of osteoblast-related gene expression, including ALP, ON, OC, RUNX2, OPN, and COL1A1. To identify the underlying molecular mechanisms, we examined the effects of Fedratinib on a molecular signature of several target genes known to affect hMSC-TERT differentiation into osteoblasts. Fedratinib inhibited the expression of LIF, SOCS3, RRAD, NOTCH3, TNF, COMP, THBS2, and IL6, which are associated with various signalling pathways, including TGFβ signalling, insulin signalling, focal adhesion, Notch Signalling, IL-6 signalling, endochondral ossification, TNF-α, and cytokines and inflammatory response. We identified a JAK2 inhibitor (Fedratinib) as a powerful inhibitor of the osteoblastic differentiation of hMSC-TERT cells, which may be useful as a therapeutic option for treating conditions associated with ectopic bone formation or osteosclerotic metastases.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3932-3932
Author(s):  
Erming Tian ◽  
Saad Usmani ◽  
Yiming Zhou ◽  
Bart Barlogie ◽  
John D Shaughnessy

Abstract Abstract 3932 Studies on the longevity of plasma cells (PCs) have provided evidence that PCs can survive for several months in the absence of repopulation by memory B cells, and PCs in the bone marrow appear to have a much longer lifespan than those found in the extramedullary environments. The concept of myeloma stem cells is based on the theory that a subset of B cells (plasmablasts) with the CD138−/CD20+/CD27+ phenotype coexisting with the majority of terminally differentiated myeloma cells give rise to tumor cells via constant clonogenic reproduction and differentiation. In previous studies, by depleting a large portion of CD138+/CD19− myeloma PCs, a small number of CD138−/CD19+ cells remained. These cells proliferated with high efficiency, giving rise to progeny with strong CD138 expression (CD138++) both in vitro and in vivo. By contrast, the counterpart of terminally differentiated CD138++ myeloma cells had limited or no clonal potential to recapitulate growth. The researchers also found that identical cytogenetic aberrations were maintained indefinitely throughout the replications. Nevertheless, the developmental path of the myeloma stem cells to its fully differentiated progeny is still unknown; more intriguingly, the mechanism that modulates myeloma cell maturation in vitro has yet to be elucidated. Using a combination of array-comparative genomic hybridization (aCGH) and gene expression profiles (GEP) we discovered that PAR1 (coagulation factor II [thrombin] receptor, or F2R; 5q13.3) expression is strongly correlated with gains of chromosome 5, a consistent feature of the hyperdiploid (HY) molecular subtype of multiple myeloma. At the diagnosis, PAR1 gene expression is elevated in 50% of primary myeloma cases. In myeloma bone marrow and human myeloma cell lines (HMCLs), we have been able to identify a subset of cells that are strongly positive for PAR1 and have weak CD138 and CD38 surface markers. The PAR1-positive phenotype defines a distinct subpopulation in heterogeneous bone marrow cells, and, to a greater degree, in a homogenous HMCL. Significantly, we have been able to demonstrate proliferation inhibition when HMCLs that express high levels of PAR1 are exposed to thrombin. Studies of PAR1 functions indicate it is not only a thrombin receptor, but also modulates the phosphorylation of ß-catenin, either parallel to or competing with the well-known Wnt pathway. The destination of stabilized ß-catenin is crucial to the formation of adherens junctions (AJs) at the cell membrane and for gene transcription regulation in the nucleus. In this study, we also discovered that most HMCLs and primary myeloma PCs retain high levels of ß-catenin, which is primarily accumulated in the nucleus, and thrombin-catalyzed stimulation can redistribute ß-catenin into cytoplasm to strongly enhance the AJs between myeloma cells and stromal cells. Based on this evidence, we believe that myeloma cells are in fact capable of transforming into a quiescent stem-like phenotype; and thrombin-induced PAR1 signaling modulates the intracellular distribution of ß-catenin that plays a major role in the reversible transformation of primary myeloma cells to indolent and drug resistant myelomablasts. Disclosures: Barlogie: Celgene: Consultancy, Honoraria, Research Funding; IMF: Consultancy, Honoraria; MMRF: Consultancy; Millennium: Consultancy, Honoraria, Research Funding; Genzyme: Consultancy; Novartis: Research Funding; NCI: Research Funding; Johnson & Johnson: Research Funding; Centocor: Research Funding; Onyx: Research Funding; Icon: Research Funding. Shaughnessy:Myeloma Health, Celgene, Genzyme, Novartis: Consultancy, Employment, Equity Ownership, Honoraria, Patents & Royalties.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3304-3304 ◽  
Author(s):  
Syed Mehdi ◽  
Sharmin Khan ◽  
Wen Ling ◽  
Randal S Shelton ◽  
Joshua Epstein ◽  
...  

Abstract Introduction: Each disease stage in myeloma (MM) is associated with parallel changes in both the MM clone and the bone marrow (BM) microenvironment. Mesenchymal cell lineages derived from mesenchymal stem cells (MSCs), including osteoblasts, adipocytes and pericytes play an important role in MM cell growth mediated by the modification of the MM niche in the BM. The overall goal of the study was to test and identify changes induced in MSCs by high-risk (HR) MM cells that impact MSC function and promote oncogenic pathways capable of supporting low-risk (LR) MM cells. Methods: Normal MSCs were either cultured alone ("unconditioned") or co-cultured with MM cells for 5 days. The cultured and co-cultured cells were trypsinized, replated for 40 min, followed by serial washing to remove MM cells from the adherent MSCs. More than 95% of the remaining adherent cells after co-culture were MSCs ("preconditioned"). The unconditioned and preconditioned MSCs or their 24 hrs conditioned media (CM; 50%) were tested for their ability to support the 5-days growth of CD138+ MM cells from LR (n=4) and HR (n=3) patients. To identify factors altered in MSCs by HR MM cells, the unconditioned and preconditioned MSCs and their serum-free conditioned media (n=4) underwent gene expression profiling and proteomic analysis. Whole bone biopsy gene expression profiles from newly diagnosed patients with MM enrolled in Total Therapy clinical trials were used to correlate the altered expression of factors in preconditioned MSCs with their expression in clinical samples. Results: Growth of all MM cells tested was increased by inclusion of MSCs preconditioned with HR MM cells by 2.2± 0.2 (p<0.0004) and by CM from these MSCs by 9.6±2.0 (p<0.006), compared to culture of MM cells in fresh media. In contrast, CM from unconditioned MSCs increased growth of HR MM cells by 2.6±0.6 (p<0.01) fold but had minor effect on growth of LR MM cells. CM from MSCs preconditioned with HR MM cells increased growth of LR and HR MM cells by 5.7±0.1 (p<0.0002) and 2.6±1.2 (p<0.04), compared to culture of MM cells in CM from unconditioned MSCs, respectively. Growth of LR MM cells was higher by 2.9±0.3 fold using CM from MSCs preconditioned with HR MM cells than by using CM from MSCs preconditioned with LR MM cells (p<0.005). To determine the role of cell-cell contact, we compared the effect of the preconditioned MSCs and their CM on growth of LR and HR MM cells. Growth of LR MM cells (p< 0.003) and HR MM cells (p< 0.005) was higher when cultured in CM than in co-culture with MSCs. These data imply that soluble factors from preconditioned MSCs support MM cell proliferation and that adhesion of MM cells to MSCs may restrain proliferation. Genes overexpressed in preconditioned MSCs included growth factors (e.g. IL6) and receptors (e.g. EDNRA); genes underexpressed include factors associated with activity of osteoblasts (e.g. ITGBL1) and adipocytes (IGFBP2). A proteomic analysis showed a reduced level of the secreted factors IGFBP2 and ITGBL1 and increase level of IL6 in CM from MSCs preconditioned with HR MM cells compared to CM from unconditioned MSCs. IGFBP2 mediates local bioavailability of IGF1 and IGF2 and is also involved in bone formation and angiogenesis independently of the IGF axis. ITGBL1 is involved in osteoblastogenesis whereas EDNRA is known to be expressed by pericytes. Global gene expression profiles from patient material showed that EDNRA and IGFBP2 are not expressed in MM cells but are highly expressed in cultured MSCs compared to hematopoietic cells in buffy coat BM samples. EDNRA is overexpressed (p<0.005) whereas IGFBP2 is underexpressed (p<0.005) in whole BM biopsy samples from MM patients with HR disease compared to patients with LR disease (p<0.005) and in Focal Lesion compared to random BM biopsies taken from the same patients (p<0.0000006 for EDNRA and p<0.02 for IGFBP2). IHC staining of patients' bone biopsies showed higher numbers of EDNRA+ mesenchymal-like cells in MM (n=10) than MGUS/AMM (n=10, p<0.0003) and in HR MM BM than LR MM BM (p<0.03). IHC analysis also revealed that IGFBP2 is highly expressed by immature adipocytes and that its expression in these cells is reduced in HR MM BM. Conclusion: Preconditioning of MSCs is essential for promoting growth of MM cells from LR patients. Factors altered in MSCs by HR MM cells are linked to signaling pathways known to directly stimulate MM cell growth and markers associated with distinct MSC lineages changed in HR MM niche. Disclosures Davies: Janssen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Celgene: Consultancy, Honoraria. Barlogie:Signal Genetics: Patents & Royalties. Morgan:Janssen: Research Funding; Univ of AR for Medical Sciences: Employment; Celgene: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria; Bristol Meyers: Consultancy, Honoraria.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2191-2191
Author(s):  
Takuo Katsumoto ◽  
Kazutsune Yamagata ◽  
Yoko Ogawara ◽  
Takuro Nakamura ◽  
Issay Kitabayashi

Abstract Monocytic leukemia Zinc finger protein (MOZ), a histone acetyltransferase, is involved in chromosome translocations associated with FAB M4/M5 types of acute myeloid leukemia (AML). In normal hematopoiesis, MOZ is essential for self-renewal of hematopoietic stem cells (HSCs) and for expression of HoxA9/Meis1 in hematopoietic stem/progenitor cells (HSPCs). Previously we found that endogenous MOZ is critical for MOZ-TIF2-induced AML. Although MOZ-/- cells expressing the MOZ-fusion serially generated colonies in vitro, they did not induce AML after transplantation into recipient mice. In these cells, up-regulation of Meis1 was impaired, while HoxA9 expression was induced. However, roles of endogenous MOZ in MOZ fusion induced leukemia remained unclear. To elucidate molecular mechanisms, we performed experiments described below. First, to reveal mechanisms in defect of Meis1 expression in MOZ-/- MOZ-fusion leukemia cells, we performed chromatin immune-precipitation assays on Meis1 locus. Coincident with gene expression, active histone marks (H3K9ac, H3K27ac etc.) were disrupted. In contrast, repressive histone modifications (H3K9me2, H3K27me3) were elevated. Next we analyzed requirement of HoxA9 and Meis1 in MOZ fusion induced AML development. When mice were transplanted with MOZ-/- HSPCs simultaneously introduced with MOZ-fusion and Meis1 genes, AML development were induced. On the other hand, when Meis1 was conditionally deleted in MOZ-fusion leukemia cells, AML development was significantly delayed. Mice transplanted with MOZ-/- HSPCs, which were introduced with both HoxA9 and Meis1 genes elicited AML development. Furthermore, we analyzed gene expression profiles of MOZ-/- MOZ fusion leukemia cells. In these cells, expressions of monocyte/macrophage lineage characteristic genes (C/EBPa, Irf8, CD68 etc.) and MLL fusion target genes (Meis1, Mef2c) were decreased. In contract, other hematopoietic lineage characteristic genes (GATA1-3, FOG-1, CD41, Aiolos, Helios, Eag, Epx etc.) were increased. In addition, expression of CDK inhibitor INK4A was also up-regulated. Finally, we tested requirement of endogenous MOZ in various cellular conditions. Previous report showed that AML development was induced by introduction of MOZ-TIF2 not only in hematopoietic stem cells but also in more differentiated Common myeloid progenitors (CMPs) and Granulocyte/Monocyte progenitors (GMPs) (Huntly et al, Cancer Cell 2004). So we introduced MOZ fusion genes in HSCs and CMPs collected from E14.5 MOZ-/- fetal liver. MOZ-/- HSCs, not CMPs, expressing MOZ-TIF2 continuously formed colonies in vitro. In the CMPs expressing MOZ-TIF2, expression of both Meis1 and HoxA9, were abolished. These results suggest that high levels of HoxA9 and Meis1 expressions were respectively required for MOZ-TIF2-induced AML development, and that endogenous MOZ is critical for MOZ-TIF2-induced AML development. Disclosures No relevant conflicts of interest to declare.


2011 ◽  
Vol 26 (10) ◽  
pp. 2552-2563 ◽  
Author(s):  
Pei-Chi Tseng ◽  
Sheng-Mou Hou ◽  
Ruey-Jien Chen ◽  
Hsiao-Wen Peng ◽  
Chi-Fen Hsieh ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document