Reduced HSPA9B Expression, a 5q31.2 Candidate Gene, in Primary Human CD34+ Cells Recapitulates Features of Ineffective Hematopoiesis Observed in MDS.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 116-116
Author(s):  
Tim H. Chen ◽  
Mark Walshauser ◽  
Amal Kambal ◽  
Gagan Raju ◽  
Matthew J. Walter

Abstract Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematopoietic disorders characterized by ineffective hematopoiesis. Deletions spanning chromosome 5q31.2 are among the most common karyotypic abnormalities in MDS, and evidence suggests that del(5q) maybe an early genetic event. We hypothesize that haploinsufficiency of one or more of the 28 candidate genes in the 5q31.2 commonly deleted segment (CDS) are important for MDS pathogenesis. Many genes in the CDS, including HSPA9B, have dose reduced mRNA levels in CD34+ cells from MDS patients with del(5q), and zebrafish carrying a heterozygous mutation in the orthologue of the human HSPA9B gene display increased apoptosis in blood cells. Therefore, we asked whether reduced HSPA9B expression in primary human hematopoietic cells can recapitulate key features of ineffective hematopoiesis (i.e. abnormal proliferation, apoptosis, and differentiation). To address this question, we purified human CD34+ hematopoietic progenitors from cord blood samples (>90% purity) and infected them with a shRNA expressing lentivirus that also carries the puromycin resistance gene. Cell culture densities were normalized after 4 days in selection medium, and 3 days later apoptosis (AnnexinV/7-AAD), cell cycle status (BrdU/7-AAD), and erythroid or myeloid differentiation was measured using flow cytometry. We performed 4–5 independent experiments using 5 individual shRNAs targeting HSPA9B (30% to >90% knockdown) and 2 control shRNAs. Cells transduced with control shRNAs expand 18.2 fold from days 4–7 in RBC unilineage differentiation culture medium (25ng/ml SCF, 10ng/ml IL-6, 10ng/ml IL-3 and 0.5U/ml Epo), vs. only a 0.8–5.2 fold expansion when HSPA9B is knocked down (N=5 for each shRNA, p≤0.007). The reduced cell numbers observed in HSPA9B knockdown cultures is associated with an increase in apoptosis and a decrease in the number of cells entering S-phase compared to control shRNA expressing cells. 8.2% of cells in control cultures were AnnexinV+/7AAD+ vs. 20–62% of cells in HSPA9B knockdown cultures (N=5 for each shRNA, p≤0.0004). Following 1 hour of BrdU exposure, 60% of cells in control cultures were in S-phase vs. only 22–48% of HSPA9B knockdown cultures (N=5 for each shRNA, p≤0.008). In addition, after 7 days in RBC unilineage differentiation culture medium, 58% of control shRNA expressing cells were CD71+/Gycophorin A+ vs. 7.1–32% of cells in the HSPA9B knockdown cultures (N=4 for each shRNA, p≤0.004). The decreased number of GPA+ cells in HSPA9B knockdown cultures was concomitant with retention of CD34+ expression on cells. Similar results were observed using myeloid unilineage culture conditions (10ng/ml SCF, 100ng/ml G-CSF, 20% FCS), where the number of cells expressing CD15+ was reduced from 47% in control cultures to 28–37% in HSPA9B knockdown cultures. A delay in erythroid and myeloid differentiation was confirmed by cell morphology. Lentiviral knockdown of the murine orthologue of HSPA9B (Hspa9a) in 2 independent transduction/transplantation experiments resulted in a loss of transduced cells over 2 months (2 shRNAs each) compared to a control shRNA, implicating a cell intrinsic defect when Hspa9a levels are reduced in vivo. Collectively, these results implicate that reduced HSPA9B expression in human CD34+ progenitor cells results in abnormal proliferation, increased apoptosis, and altered differentiation, key features of ineffective hematopoiesis, in a dose dependent manner.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1343-1343
Author(s):  
Richard Hildreth ◽  
Robert W. Georgantas ◽  
Roshan Patel ◽  
Sebastien Morisot ◽  
Jonathan Alder ◽  
...  

Abstract In a large microRNA-array and bioinformatics study, we determined all of the microRNAs (miRs) expressed by human CD34+ hematopoietic stem-progenitor cells (HSPCs) from bone marrow and G-CSF mobilized blood. When we combined miR expression data, mRNA expression data fro a previous study (Georgantas et al, Cancer Research 64:4434), and data from various published mir-target prediction algorithms, we were able to bioinformaticly predict the actions of miRs within the hematopoietic system. MicroRNA hsa-mir-16 was highly expressed in CD34+ HSPCs, and was predicted to target several HSPC-expressed mRNAs (CXCR4, HoxB7, Runx-1, ETS-1, and Myb) that encode proteins known to be critically involved specifically in myelopoiesis within the hematopoietic system. We first confirmed that protein expression from each of these putative target mRNAs was in fact regulated by mir-16. The 3′UTR sequence from each of these mRNAs was cloned behind a luciferase reporter. Each reporter construct was transfected into K562 cells, which strongly express mir-16. In all cases, protein expression from the predicted target mRNA was greatly reduced in K562 cells, as compared to controls. As a first determination of mir-16’s function in hematopoietic cells, HL60 and K562 cells were transduced with hsa-mir-16 lentivirus, then treated with various chemical differentiation inducers. As was predicted by bioinformatics, hsa-mir-16 halted myeloid differentiation of HL60 cells, but did not affect megakaryocytic differentiation or erythroid differentiation of K562 cells. These initial findings suggest that mir-16 is a specific negative regulator of myelopoiesis. We are currently evaluating the effects of mir-16 on normal human CD34+ cells by in vitro CFC and suspension culture assays, as well as in vivo by transplantation of hsa-mir-16 lentivirus transduced cells in immunodeficient mice.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 549-549
Author(s):  
Ly P. Vu ◽  
Xinyang Zhao ◽  
Fabiana Perna ◽  
Stephen D. Nimer

Abstract Abstract 549 RUNX1 (also known as AML1) is the DNA binding component of the Core Binding Factor (CBF)-transcriptional regulatory complex, which plays an important role in hematopoiesis. Upon binding to the common binding sequence -PyGpyGGTPy (Py = pyrimidine) in the regulatory regions of promoters and enhancers of its target genes, RUNX1 acts either as an activator or a repressor, depending on promoter context and its interacting partners. Thus, modulation of the network of RUNX1 interactions can influence hematopoiesis. However, how RUNX1 selects one set of partners over another to assemble a functional complex is largely unknown. Posttranslational modifications, including ubiquitination, phosphorylation, acetylation and methylation, present a viable mean to fine-tune its functions. Here we shown that RUNX1 is arginine methylated at a specific residue, R223, by PRMT4, a type I arginine methyltransferase generally thought of as a co-activator molecule. We hypothesized that arginine methylation of RUNX1 by PRMT4 affects its protein-protein interactions, therefore, to identify proteins that specifically interact with unmethylated and/or methylated-R223 RUNX1, in an unbiased manner, we performed a peptide pull-down experiment, using a methyl-R223 RUNX1 peptide and an unmodified RUNX1 peptide as bait, following by mass spectrometry analysis. We identified several proteins that preferentially interacted with the R223 methyl peptide, but focused on a novel interacting protein, DPF2 (double PhD Finger 2), which is a widely expressed member of the d4 protein family, characterized by the presence of a tandem plant-homodomain (PHD domain). We confirmed the specific interaction between methylated-RUNX1 with DPF2 in vivo by immunoprecipitation. We generated an antibody specific for the R223 methylated-RUNX1 protein, and found that RUNX1 methylation decreases during the myeloid differentiation of human CD34+ haematopoietic stem/progenitor cells (HSPCs), without a change in the total level of RUNX1 protein, and this occurred co-incident with a downregulation of PRMT4 protein expression. Having determined that PRMT4 expression declines during myeloid differentiation, we examined the role of PRMT4 in this process, using short hairpin RNAs to knockdown PRMT4 expression in CD34+ cells. Knockdown of PRMT4 accelerates the myeloid differentiation of the cells, whereas overexpression of PRMT4 in human CD34+ cells blocked their myeloid differentiation. When analyzing the expression of several “master” regulators of myeloid differentiation, we identified microRNA-223, a myeloid specific microRNA, as a common target gene of PRMT4 and RUNX1. Furthermore, we have found that by promoting the assembly of a functional complex containing R223 methylRUNX1 and DPF2 at the transcriptional regulatory region of the microRNA-223 promoter, PRMT4 can control miR-223 expression and myeloid differentiation. We have verified the role of DPF2 in this process, as DPF2 represses miR-223 expression and loss of DPF2 promotes myeloid differentiation. Thus, DPF2 acts in a common pathway with PRMT4 to regulate myeloid differentiation. In conclusion, our study elucidates a novel mechanism, where the arginine methylation of RUNX1 regulates its recruitment of interacting partner(s). In addition to demonstrating that PRMT4 can trigger repression of gene expression, we have identified a novel role for PRMT4 (aka CARM1) in myeloid differentiation. Disclosures: No relevant conflicts of interest to declare.


2004 ◽  
Vol 76 (5) ◽  
pp. 1057-1065 ◽  
Author(s):  
Alexander Kiani ◽  
Ivonne Habermann ◽  
Michael Haase ◽  
Silvia Feldmann ◽  
Sabine Boxberger ◽  
...  

eLife ◽  
2019 ◽  
Vol 8 ◽  
Author(s):  
Murilo Delgobo ◽  
Daniel AGB Mendes ◽  
Edgar Kozlova ◽  
Edroaldo Lummertz Rocha ◽  
Gabriela F Rodrigues-Luiz ◽  
...  

Monocyte counts are increased during human tuberculosis (TB) but it has not been determined whether Mycobacterium tuberculosis (Mtb) directly regulates myeloid commitment. We demonstrated that exposure to Mtb directs primary human CD34+ cells to differentiate into monocytes/macrophages. In vitro myeloid conversion did not require type I or type II IFN signaling. In contrast, Mtb enhanced IL-6 responses by CD34+ cell cultures and IL-6R neutralization inhibited myeloid differentiation and decreased mycobacterial growth in vitro. Integrated systems biology analysis of transcriptomic, proteomic and genomic data of large data sets of healthy controls and TB patients established the existence of a myeloid IL-6/IL6R/CEBP gene module associated with disease severity. Furthermore, genetic and functional analysis revealed the IL6/IL6R/CEBP gene module has undergone recent evolutionary selection, including Neanderthal introgression and human pathogen adaptation, connected to systemic monocyte counts. These results suggest Mtb co-opts an evolutionary recent IFN-IL6-CEBP feed-forward loop, increasing myeloid differentiation linked to severe TB in humans.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3082-3082
Author(s):  
Kenichi Sawada ◽  
Makoto Hirokawa ◽  
Kayo Inaba ◽  
Hiroshi Fukaya ◽  
Yoshinari Kawabata ◽  
...  

Abstract Background. Tumor necrosis factor-α (TNF-α) and thrombopoietin (TPO) have been shown to sustain differentiation and proliferation of CD34+ cells toward dendritic cells (DCs) in the presence of multi-acting cytokines. We hypothesized that co-stimulation of TPO and TNF-α generate megakaryocytic progenitors and DCs together from human CD34+ cells and that interaction of these cells may provide a physiological and/or a pathological role of DCs in megakaryopoiesis. Materials and Methods. Highly purified human CD34+ cells were cultured with TPO, with or without TNF-α, in plasma-depleted medium and induced to undergo megakaryocytic differentiation. We enumerated megakaryocytic progenitor cells using the specific markers CD41, CD42b, and CD61, and DCs using CD4, CD11c, CD80, CD83, CD86, and CD123. The character and roles of co-developing non-megakaryocytic cells in the presence of TNF-α were analyzed by fluorescence-activated cell sorter, enzyme immunohistochemistry, confocal microscopy, and autologous mixed lymphocyte reaction. Cytokine production was assessed using a cytometric bead array system. Results. When CD34+ cells were cultured for 7 days in the presence of TPO, the generated cells predominantly expressed CD41 (95±2%), CD42b (54±12%), and CD61 (96±2%), while rarely expressing CD11c (1.6±1.3%), CD80 (0.1±0.1%), CD83 (0.8±0.6%), or CD86 (3.3±1.9%). The addition of TNF-α significantly decreased the number of cells expressing CD41 (3.0±0.6%), CD42b (3.3±1.0%), or CD61 (3.2±0.9%), but did not affect the number of total cells. In the presence of TNF-α, the generated cells expressed major histocompatibility complex (MHC) class I (100%) plus MHC class II (100%). A substantial number of cells became positive for CD11c (37±1%), and even co-stimulatory molecules such as CD80 (2.4±1.9%), CD83 (8±4%), and CD86 (18±7%). Immature CD11c+ DCs were physically associated with apoptotic and CD61+ cells and capable of endocytosing CD61+ cells. Most of the CD11c+ cells co-expressed the c-mpl TPO receptor, CD4, and CD123 and about one half of CD11c+ cells co-expressed CD86. The DCs generated by TNF-α and TPO, but not those by TNF-α alone, facilitated autologous T cell proliferation in some extent, although cytokine production from activated T cells were low. We also confirmed engulfment of CD61+ cells and their fragment by CD11c+ cells in bone marrow cells from patients with hemophagocytic syndromes. Conclusions. This is the first report showing that in the presence of TNF-α, the non-megakaryocytic cells with typical feature of DCs are co-generated from human CD34+ cells during megakaryocytic differentiation by TPO. The CD4+ CD11c+ CD123+ DCs physically associates with and phagocytose developing or dying immature megakaryocytic cells. Similar phenomenon showing engulfment of CD61+ fragment by CD11c+ cells was also observed in bone marrow cells from patients with hemophagocytic syndrome. Therefore, it may be conceivable that DCs with phagocytic activity during the development in bone marrow may play a crucial role in the maintenance of tolerance for self-substances derived from hematopoietic progenitor cells.


Blood ◽  
2009 ◽  
Vol 114 (8) ◽  
pp. 1596-1606 ◽  
Author(s):  
Ayten Kandilci ◽  
Gerard C. Grosveld

Abstract Forced expression of MN1 in primitive mouse hematopoietic cells causes acute myeloid leukemia and impairs all-trans retinoic acid-induced granulocytic differentiation. Here, we studied the effects of MN1 on myeloid differentiation and proliferation using primary human CD34+ hematopoietic cells, lineage-depleted mouse bone marrow cells, and bipotential (granulocytic/monocytic) human acute myeloid leukemia cell lines. We show that exogenous MN1 stimulated the growth of CD34+ cells, which was accompanied by enhanced survival and increased cell cycle traverse in cultures supporting progenitor cell growth. Forced MN1 expression impaired both granulocytic and monocytic differentiation in vitro in primary hematopoietic cells and acute myeloid leukemia cell lines. Endogenous MN1 expression was higher in human CD34+ cells compared with both primary and in vitro–differentiated monocytes and granulocytes. Microarray and real-time reverse-transcribed polymerase chain reaction analysis of MN1-overexpressing CD34+ cells showed down-regulation of CEBPA and its downstream target genes. Reintroduction of conditional and constitutive CEBPA overcame the effects of MN1 on myeloid differentiation and inhibited MN1-induced proliferation in vitro. These results indicate that down-regulation of CEBPA activity contributes to MN1-modulated proliferation and impaired myeloid differentiation of hematopoietic cells.


2009 ◽  
Vol 18 (6) ◽  
pp. 845-856 ◽  
Author(s):  
Claus S. Sondergaard ◽  
Jesper Bonde ◽  
Frederik Dagnæs-Hansen ◽  
Jan M. Nielsen ◽  
Vladimir Zachar ◽  
...  

Blood ◽  
2002 ◽  
Vol 99 (5) ◽  
pp. 1585-1593 ◽  
Author(s):  
Anna Jetmore ◽  
P. Artur Plett ◽  
Xia Tong ◽  
Frances M. Wolber ◽  
Robert Breese ◽  
...  

Differences in engraftment potential of hematopoietic stem cells (HSCs) in distinct phases of cell cycle may result from the inability of cycling cells to home to the bone marrow (BM) and may be influenced by the rate of entry of BM-homed HSCs into cell cycle. Alternatively, preferential apoptosis of cycling cells may contribute to their low engraftment potential. This study examined homing, cell cycle progression, and survival of human hematopoietic cells transplanted into nonobese diabetic severe combined immunodeficient (NOD/SCID) recipients. At 40 hours after transplantation (AT), only 1% of CD34+ cells, or their G0(G0CD34+) or G1(G1CD34+) subfractions, was detected in the BM of recipient mice, suggesting that homing of engrafting cells to the BM was not specific. BM of NOD/SCID mice receiving grafts containing approximately 50% CD34+ cells harbored similar numbers of CD34+ and CD34− cells, indicating that CD34+ cells did not preferentially traffic to the BM. Although more than 64% of human hematopoietic cells cycled in culture at 40 hours, more than 92% of cells recovered from NOD/SCID marrow were quiescent. Interestingly, more apoptotic human cells were detected at 40 hours AT in the BM of mice that received xenografts of expanded cells in S/G2+M than in recipients of G0/G1 cells (34.6% ± 5.9% and 17.1% ± 6.3%, respectively; P < .01). These results suggest that active proliferation inhibition in the BM of irradiated recipients maintains mitotic quiescence of transplanted HSCs early AT and may trigger apoptosis of cycling cells. These data also illustrate that trafficking of transplanted cells to the BM is not selective, but lodgment of BM-homed cells may be specific.


Sign in / Sign up

Export Citation Format

Share Document