Identifying Direct Targets of the Hoxb4 Transcription Factor Involved in Hematopoietic Cell Self Renewal.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4190-4190
Author(s):  
Jie Jiang ◽  
Yan Shou ◽  
Brian P. Sorrentino

Abstract Hoxb4 is a member of the homeobox protein family which when overexpressed, promotes expansion of hematopoietic stem cells (HSCs) both in vivo and ex vivo. The molecular mechanisms underlying the capacity of Hoxb4 to induce self-renewal of HSCs are poorly understood. In order to identify the direct transcriptional targets of Hoxb4 in primary hematopoietic progenitor cells, we created a Hoxb4-ERT2 fusion gene to allow for temporally regulated Hoxb4 activity. This gene was introduced into mouse lineage negative bone marrow (Lin− BM) cells using retroviral-mediated gene transfer. When these cells were treated with Tamoxifen (TAM), translocation of the fusion protein into the nuclei was demonstrated using confocal microscopy. Transduced cells were treated with 300nM of TAM for varying time in suspension culture and then plated into semisolid medium for myeloid progenitor assays. After 7 days, colonies from these primary CFU-C cultures were dispersed and replated for secondary CFU-C as an assay for myeloid progenitor self renewal. Treatment with TAM for 12hr in Hoxb4-ERT2 introduced cells resulted in an increase in the number (5.5 fold) and size (10 fold) of secondary colonies comparing to mock vector transduced controls, verifying that TAM treatment resulted in self-renewal of myeloid progenitors. In order to identify the target genes for this response, RNA from 12hr and 24hr TAM treated cells was analyzed for differential gene expression using Affymetrix 430v2 chips. Analysis of three independent experiments showed good reproducibility and allowed identification of candidate genes. Local pooled error test was used to analyze the data and a false discovery rate threshold at <25% was applied to identify the probe sets significantly differing in samples from TAM-treated Hoxb4-ERT2 samples versus TAM-treated control vector samples. This analysis identified 78 probe sets from cells treated for 12 hours, 103 probe sets from 24hr treated samples, and 20 common probe sets from both time points. A computer analysis of 64 candidate promoters identified in our screen and available in the NCBI database found that 50 % of them contained more than one high or moderate affinity consensus Hoxb4 binding motif. Furthermore, 22 of our candidate genes contained more than two consensus Hoxb4 binding motifs in the promoter regions. In this selected candidate gene group, genes were identified that are involved in various cellular functions: four in signal transduction, two in cell cycle regulation/apoptosis, four in enzyme activity regulation, six in cellular metabolism, two in transcriptional regulation; and eight of them associated with signaling transduction pathways for hematopoietic cells proliferation/self renewal. Therefore, our current results have identified a relatively small number of candidate genes (20) with significant potential to be direct targets of the Hoxb4 transcription factor in primary, self-renewing hematopoietic progenitor cells. We are currently testing several of these genes for direct Hoxb4 binding activity and for functional activity in hematopoietic cells.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1425-1425
Author(s):  
Alla Dolnikov ◽  
Ailyn Choo ◽  
Patricia Palladinetti ◽  
Toby Passioura ◽  
Geoff Symonds ◽  
...  

Abstract Activating mutations of the Ras genes occur at high frequency in acute myeloid leukemia (AML). We have previously shown that expression of mutant N-ras(N-rasm) in murine hematopoietic stem cells is sufficient to induce a myeloid malignancy that resembles human AML(Mackenzie et al. Blood, 1999, 93, 2043–2056). In a ’humanised’ NOD/SCID mouse model N-rasm induced a pre-leukemic condition characterised by myeloid proliferation of human hematopoietic progenitor cells in the bone marrow of recipient mice (Shen et al. Exp. Hematol., 2004, 32: 852–860). Even though Ras usually acts as a dominant transforming oncogene, in primary cells and some cancer cell lines, Ras inhibits cell growth. We have previously shown that ectopic expression of N-rasm in leukemia U937 and K562 cells leads to growth suppression (Passioura et al. Cancer Res. 2005, 65, 797–804). The expression profile induced by N-rasm in these cells included the up-regulation of transcription factor Interferon Regulatory Factor1 (IRF1) and activation of cdk inhibitor p21WAF. IRF1 was previously defined as a tumour suppressor, and as such is a target of oncogenic mutations in AML. Antisense suppression of IRF1 prevented N-rasm induced growth suppression and up-regulation of p21WAF1. These results defined a novel tumour suppressive response to oncogenic N-rasm in leukemia cells. A retroviral cDNA library screen for genes that counteract N-rasm-induced growth suppression identified the gene for the Interferon Regulatory Factor2 (IRF2), and as confirmation of the screen, over-expression of IRF2 in leukemia U937 cells acted to inhibit N-rasm-induced growth suppression (Passioura et al. Oncogene. 2005; 24: 7327–36). IRF2 is known for its oncogenic properties and can antagonise IRF1-mediated tumour suppression. In addition, IRF2 is often up-regulated in primary leukemia samples. Here we show that IRF2 gene suppression using RNA interference acts to suppress the growth of leukemia TF-1 cells bearing N-ras mutation in codon 61 and expressing high levels of IRF1 and IRF2 and low level of p21Waf1. IRF2 down-regulation confirmed at RNA (quantitative RT-PCR) and protein (Western analysis) levels resulted in up-regulation of p21Waf1 and G2/M- rather than G1/S-growth arrest. In addition, increased polyploidisation that results from discoordinated DNA synthesis in mitotically arrested cells, was observed. In addition, IRF2-down-regulation significantly reduced clonogenic growth of the leukemic blasts. Cell growth of normal hematopoietic progenitor cells that express low levels of both IRF1 and IRF2, however, was not affected by IRF2 targeting. IRF2 targeting is currently being examined in primary AML samples in an animal model of AML. We suggest that IRF2 suppression can be used for ex vivo purging of leukemia cells in the autologous stem cell transplantation setting. To the best of our knowledge, specific IRF2 inhibition in cancer cells as a potential therapeutic approach has not been tested to date. IRF2 suppression may prove to be a novel therapeutic target for leukemia therapy.


Blood ◽  
2006 ◽  
Vol 108 (6) ◽  
pp. 2087-2094 ◽  
Author(s):  
Gabriel Ghiaur ◽  
Andrew Lee ◽  
Jeff Bailey ◽  
Jose A. Cancelas ◽  
Yi Zheng ◽  
...  

AbstractRas-related Rho GTPases regulate actin cytoskeletal organization, adhesion, gene transcription, and cell-cycle progression. The Rac subfamily of Rho GTPases and Cdc42 has been shown to play essential roles in hematopoietic stem cell (HSC) engraftment and mobilization. Here, we study the role of RhoA, a related Rho GTPase, in HSC functions. Using retrovirus-mediated gene transfer of a dominant-negative (DN) mutant of RhoA (RhoAN19), we demonstrate that down-regulation of RhoA activity resulted in increased HSC engraftment and self-renewal as measured by competitive repopulation and serial transplantation assays. However, overexpression of RhoAN19 resulted in decreased migration toward SDF-1α and α4β1- and α5β2-integrin–mediated adhesion of hematopoietic progenitor cells in vitro. Low RhoA activity was associated with higher proliferation rate of hematopoietic progenitor cells and increased cells in active phases of cell cycle, most likely via decreasing p21Cip/Waf expression and increasing cyclin D1 levels. Thus, reducing RhoA activity by optimizing the balance between adhesion/migration and proliferation/self-renewal results in a net increase in HSC engraftment. This mechanism could provide a novel therapeutic target to enhance HSC therapies.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1285-1285
Author(s):  
Jianhua Wang ◽  
Russell Taichman ◽  
Younghun Jung ◽  
Aaron Havens ◽  
Yanxi Sun ◽  
...  

Abstract Osteoblasts constitute part of the stromal cell support system in marrow for hematopoiesis, however little is known as to how they interact with hematopoietic stem cells (HSCs). In vitro studies have demonstrated that the survival of HSCs in co-culture with osteoblasts requires intimate cell-to-cell contact. This suggests that the osteoblast-derived factor(s) that supports stem cell activities are either produced in very small quantities, are rapidly turned over, may be membrane-anchored and/or requires the engagement of cell-cell adhesion molecules yet to be determined. In the present report we found that survival of hematopoietic progenitor cells on osteoblasts is dependent upon the engagement of VLA-4 (α4β1) and VLA-5 (α5ß1) receptors using function blocking antibodies. Surprisingly, cell-to-cell contact is not absolutely required to support progenitor activity, but does not require receptor-ligand engagement of the VLA-4 and LFA-1 complexes, which can in part be replaced through the use of recombinant ligands (fibronectin, ICAM-1, VCAM-1). Moreover conditioned once these receptors were engaged, medium derived from HSCs grown on osteoblasts ligands supported significantly greater hematopoietic progenitors in vitro than did osteoblast-conditioned or HSC-conditioned medium alone. As an initial attempt to identify the activity we examined which genes are activated following the establishment of osteoblast-CD34+ cell co-cultures nine separate co-cultures were establsihed and the RNA was pooled and analyized on Affymetrix HG-U133A chips at 24 hours. Initially our analysis revealed that there were 259 genes that are up regulated at 24 hours, and 14 genes that are down regulated. Inspection revealed that 30 of these signals were repeated at least once suggesting that 206 genuine gene candidates were differentially expressed resulting from the co-culture. A significant proportion of the differentially expressed cDNAs represent intracellular signaling ligands 16.5% (n=34) and cell surface receptors 13.5% (n=28). Molecules associated with assembly of the extra cellular matrix or its degradation comprised 7.2% (n=15) of the differentially up regulated molecules. Molecules associated with intracellular signaling, novel sequences and intermediate metabolism comprised the majority of the remaining activities. Amoung the candidates of extra cellular signaling molecules, we noted that IL-6, LIF, MIP-1alpha and SDF-1 were identified in the microarray analysis. This observation was most gratifying as we had previously reported that IL-6, LIF and MIP-1α activities are critical components of an HSC-osteoblast microenvironment. Other notable cytokine messages for BMP-2, CCL7, FGF2b, GRO1α, GRO3, IGF1, IL1ß, IL-8, IL-11, LIF, PDGF-D and the receptors for CCL7 (CCR7). Elevations in mRNA for fibronectin, lysine hydroxylase-like proteins, laminin and Type I collagen suggest that the presence of hematopoietic cells also induces osteoblastic activities. While the identity of those molecules present in the co-cultured medium remain to be identified, the data suggests that hematopoietic cells cooperate with osteoblasts to assemble the various marrow microenvironments by directing the synthesis of osteoblast-derived cytokines to improve HSC survival.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 647-647 ◽  
Author(s):  
Junping Wei ◽  
Mark Wunderlich ◽  
James C. Mulloy

Abstract Telomerase is a reverse transcriptase that adds telomeric sequences onto chromosome ends. During hematopoiesis, telomerase activity is high in proliferating hematopoietic stem and progenitor cells, but decreases sharply as cells differentiate. Approximately 90% of all human cancers are telomerase positive, including samples from AML patients. To understand the role of telomerase in human hematopoietic stem and progenitor cell proliferation and differentiation, and in the development of leukemia, we established stable long-term cultures expressing high levels of telomerase by retrovirus-mediated transduction of the h-TERT (catalytic subunit of telomerase) gene into AML1-ETO-expressing CD34+ (AE) cells. We have previously shown that these AE cells maintain CD34 expression and proliferate for 7 months in culture but gradually lose telomere length and are not immortal, providing a good model for studying the molecular mechanisms involved in telomere shortening. Strikingly, telomerase expression enhanced the self-renewal ability of the human hematopoietic progenitor cells, as shown by continuous replating ability in methylcellulose assays. We also detected a pronounced delay in the differentiation of the progenitor cells upon telomerase expression, and an expansion of the progenitor pool. Telomerase expression promoted proliferation due to increased cell cycle progression as well as a survival advantage. At the same time, these cells demonstrated a progressive decline in telomere length, despite telomerase activity equivalent to that detected in leukemia cell lines. We conclude that expression of hTERT expands the pool of hematopoietic progenitors but does not maintain long telomeres in human CD34+ cells. hTERT plays a critical role not only in telomere homeostasis for genetic stability but also in cellular proliferation, differentiation, and self-renewal, functions that may be part of hTERT involvement in leukemogenesis. These data open an important debate regarding the specific contribution of telomerase expression to the leukemic phenotype and the potential timing of this essential hit in the progression of the disease.


Blood ◽  
2009 ◽  
Vol 114 (2) ◽  
pp. 290-298 ◽  
Author(s):  
Anja Köhler ◽  
Vince Schmithorst ◽  
Marie-Dominique Filippi ◽  
Marnie A. Ryan ◽  
Deidre Daria ◽  
...  

Abstract Aged hematopoietic stem cells (HSCs) are impaired in supporting hematopoiesis. The molecular and cellular mechanisms of stem cell aging are not well defined. HSCs interact with nonhematopoietic stroma cells in the bone marrow forming the niche. Interactions of hematopoietic cells with the stroma/microenvironment inside bone cavities are central to hematopoiesis as they regulate cell proliferation, self-renewal, and differentiation. We recently hypothesized that one underlying cause of altered hematopoiesis in aging might be due to altered interactions of aged stem cells with the microenvironment/niche. We developed time-lapse 2-photon microscopy and novel image analysis algorithms to quantify the dynamics of young and aged hematopoietic cells inside the marrow of long bones of mice in vivo. We report in this study that aged early hematopoietic progenitor cells (eHPCs) present with increased cell protrusion movement in vivo and localize more distantly to the endosteum compared with young eHPCs. This correlated with reduced adhesion to stroma cells as well as reduced cell polarity upon adhesion of aged eHPCs. These data support a role of altered eHPC dynamics and altered cell polarity, and thus altered niche biology in mechanisms of mammalian aging.


Blood ◽  
1998 ◽  
Vol 91 (11) ◽  
pp. 4099-4105 ◽  
Author(s):  
Johannes F.M. Pruijt ◽  
Yvette van Kooyk ◽  
Carl G. Figdor ◽  
Ivan J.D. Lindley ◽  
Roel Willemze ◽  
...  

Abstract Previously, we have shown that interleukin (IL)-8 induces the rapid (15 to 30 minutes) mobilization of hematopoietic progenitor cells (HPC) in mice. Because integrins are essential for adhesion and transendothelial migration of HPC, we studied the involvement of the β2-integrin leukocyte function-associated antigen-1 (LFA-1) in IL-8–induced mobilization. After a single injection of blocking anti–LFA-1 antibodies, no mobilization of colony-forming cells was observed. In addition, when mice were pretreated with anti–LFA-1 or saline and subsequently injected with 30 μg of IL-8, mobilization of HPC was completely blocked. We showed that this was not due to anti–LFA-1 antibodies affecting colony formation, as addition of anti–LFA-1 antibodies to colony cultures in semisolid medium had no inhibitory activity. Also, anti-intercellular adhesion molecule (ICAM)-1 antibodies, directed to the main ligand of LFA-1 significantly inhibited the IL-8–induced mobilization. Furthermore, IL-1–induced mobilization was significantly inhibited by anti–LFA-1 antibodies. Because LFA-1 is reported to be expressed on more differentiated HPC, it was considered that the IL-8–induced mobilization of more primitive HPC would not be blocked by anti–LFA-1 antibodies. Transplantation of blood-derived mononuclear cells (MNC) from IL-8–mobilized animals pretreated with anti–LFA-1 antibodies protected only 25% of lethally irradiated recipient mice, whereas the radioprotection rate of control mice transplanted with MNC derived from IL-8-mobilized animals was 86% (P < .01). Anti-LFA–1 antibodies did not interfere with stem cell homing, as transplantation of IL-8-mobilized blood MNC, incubated in vitro with these antibodies resulted in 100% radioprotection. We conclude that anti–LFA-1 antibodies completely prevent the rapid mobilization of colony-forming cells and of cells with radioprotective capacity induced by IL-8. These results indicate a major role for the β2-integrin LFA-1 in the IL-8–induced mobilization of hematopoietic stem cells.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 724-724
Author(s):  
Michihiro Kobayashi ◽  
Yunpeng Bai ◽  
Sisi Chen ◽  
Sarah C Nabinger ◽  
Chonghua Yao ◽  
...  

Abstract Acute myeloid leukemia (AML) is a heterogeneous disease with multiple signaling pathways contributing to its pathogenesis. Mutations in receptor tyrosine kinase KIT and FLT3 are found in approximately 40% of AML patients and targeted therapies for inhibiting KIT and FLT3 have failed, thus new targets for therapeutic intervention need to be identified. The phosphatase of regenerating liver (PRL) family of phosphatases, consisting of PRL1, PRL2, and PRL3, represents an intriguing group of proteins being validated as biomarkers and therapeutic targets in human cancer. While PRL2 is highly expressed in some subtypes of human AML, including AML1-ETO+ AML and AML with mixed lineage leukemia (MLL) translocations, its role in AML is largely unknown. To determine the role of PRL2 in the pathogenesis of AML, we utilized two murine models of human AML induced by transducing mouse HSCs with AML1-ETO or MLL-AF9. We found that PRL2 is important for the progression and maintenance of leukemia induced by AML1-ETO or MLL-AF9 through enhancing leukemia stem cell (LSC) self-renewal. To elucidate the mechanisms by which PRL2 promotes LSC maintenance, we performed genome wide RNA-seq analysis of MLL-AF9+ LSCs. Gene Set Enrichment Analysis (GESA) indicates that PRL2 deficiency alters the MLL-AF9 signature essential for LSC self-renewal. We have recently identified PRL2 to be important for the proliferation and self-renewal of hematopoietic stem cells (HSCs) through the regulation of KIT signaling. Notably, PRL2 null hematopoietic progenitor cells showed decreased KIT phosphorylation as well as ERK phosphorylation following SCF stimulation, suggesting that PRL2 is important for KIT activation. Given that KIT inactivation could be mediated by removal from the cell surface and intracellular degradation, we reasoned that PRL2 may regulate KIT receptor internalization and stability. That was indeed the case. We found that the KIT protein half-life in PRL2 null hematopoietic progenitor cells (Kit+) was significantly decreased compared to WT cells. Furthermore, PRL2 null progenitor cells showed enhanced KIT ubiquitination compared to WT cells and less KIT was found on the surface of PRL2 null progenitor cells compared to WT cells following SCF stimulation. We also found that loss of PRL2 in human AML cells resulted in enhanced internalization of KIT. These observations demonstrate that PRL2 deficiency results in less KIT on the cell surface and a lower global KIT level in the cell. Upon SCF stimulation, KIT binds to and induces the phosphorylation of CBL proteins, which in turn act as E3 ligases, mediating the ubiquitination and degradation of KIT. To understand how PRL2 modulates the turnover of KIT in hematopoietic cells, we performed GST-pulldown assays and found that the substrate-trapping mutant PRL2/CS-DA showed an increased association with KIT and CBL compared to wild-type PRL2 in Kasumi-1 cells, suggesting that KIT and CBL may be PRL2 substrates. Furthermore, we found that PRL2/CS-DA mutant showed enhanced association with FLT3 and CBL compared to wild-type PRL2 in MV4-11 cells. Our data suggest that PRL2 dephosphorylates CBL and inhibits CBL activity toward KIT and FLT3, leading to sustained activation of downstream signaling pathways. To determine the functional significance of PRL2 in human AML with KIT and FLT3 mutations, we utilized two well-established murine model of myeloproliferative neoplasms (MPN) induced by KITD814V or FLT3-ITD. We found that loss of Prl2 decreased the ability of oncogenic KITD814V and FLT3-ITD to promote mouse hematopoietic stem and progenitor cell (HSPC) proliferation in vitro andthe development of MPN in vivo. Furthermore, we found that genetic and pharmacological inhibition of PRL2 decreased the proliferation and survival of human AML cells bearing KIT or FLT3 mutations. Taken together, we demonstrate that PRL2 promotes leukemia stem cell (LSC) self-renewal and maintenance through sustaining the activity of oncogenic KIT and FLT3 signals. Our findings suggest that pharmacological inhibition of PRL2 holds potential as a novel therapy for acute myeloid leukemia, and might also be applicable to the treatment of other human cancers. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 39 (03) ◽  
Author(s):  
S Nandhini ◽  
Gowri A Mangala ◽  
G R Baranidharan ◽  
T V Meenambigai

Blood cells are responsible for constant maintenance and immune protection of every cell type of the body and this relentless and brutal work requires cells that have the greatest powers of self-renewal and are designated as Hematopoietic progenitor cells (HPCs).Peripheral blood stem cells in circulation have become the most common source of hematopoietic stem cells intended for transplantation after minimal manipulation. Homeo box (Hox), sonic hedgehog (SHH), and Wingless-type MMTV integration site family (Wnt) are known to modulate the self-renewal and expansion of hematopoietic progenitor/stem cells in humans and mice. Unlike cytokines, Hox, SHH, and Wnt are highly conserved among species from flies to humans but studies regarding the self-renewal and expansion of the HSC are extremely limited in dogs.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4042-4042
Author(s):  
Lingbo Liu ◽  
Yong Wang ◽  
Daohong Zhou

Bone marrow (BM) suppression is the most common dose-limiting side effect of conventional cancer therapy, particularly with certain alkylating agents and/or ionizing radiation (IR). It has been shown that p38 mitogen-activated protein kinase (p38 MAPK) plays a critical role in regulation of hematopoiesis and its activation mediates IR- and chemotherapy-induced cell injury and oxidative stress-induced hematopoietic stem cell (HSC) exhaustion. Therefore, we examined the role of p38 MAPK in radiation-induced BM injury. First, we monitored the activation of p38 MAPK in BM hematopoietic cells at different time after exposure to IR using our well-established long-term BM cell culture (LTBMC) model system. The activation of p38 MAPK was detected within 24 hours after BM hematopoietic cells were exposed to 4 Gy IR and this activation sustained up to 5 weeks after radiation. Inhibition of p38 MAPK activity with a p38 MAPK specific inhibitor (SB202190) attenuated IR-induced suppression of the hematopoietic function of BM hematopoietic cells in an in vitro colony forming cell (CFC) assay. Moreover, the number of hematopoietic progenitor cells produced by SB202190-treated BM cells was significantly greater than that by the cells without SB202190 treatment after exposure to IR and followed by a five-week LTBMC. Interestingly, p38 MAPK inhibition showed no effect on IR-induced apoptosis in both HSCs and hematopoietic progenitor cells (HPCs), whereas the radioprotection effect of SB202190 was associated with a significantly reduction of p16INK4a expression and senescence-associated β-galactosidase (SA-β-Gal) activity in irradiated BM cells after five weeks of LTBMC. These findings suggest that activation of p38 MAPK may mediate IR-induced hematopoietic suppression by induction of hematopoietic stem/progenitor cell senescence and pharmacological inhibition of the p38 MAPK pathway may have the potential to be developed as an innovatively therapeutic strategy to ameliorate IR- and chemotherapy-induced BM toxicity.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1296-1296
Author(s):  
Laleh S. Arabanian ◽  
Michael Haase ◽  
Ivonne Habermann ◽  
Malte von Bonin ◽  
Claudia Waskow ◽  
...  

Abstract Abstract 1296 Understanding the transcriptional mechanisms that control hematopoiesis and the interaction between hematopoietic stem cells and the bone marrow microenvironment in vivo is of considerable interest. We have previously shown that aged mice lacking the transcription factor NFATc2 develop bone marrow hypoplasia, anemia, and extramedullary hematopoiesis in spleen and liver. The proliferation and differentiation of NFATc2-deficient hematopoietic progenitor cells (HPC) ex vivo, however, was found to be intact. It remained therefore unclear whether the disturbed hematopoiesis in NFATc2-deficient mice was caused by the hematopoietic or the stroma component of the bone marrow hematopoietic niche. In the current study we dissected the relative contribution of hematopoietic and stroma cells to the phenotype of the NFATc2-deficent mice by transplanting immunomagnetically purified NFATc2-deficient (ko) HPCs to lethally irradiated wildtype (wt) mice, and vice versa. After a posttransplantation period of 6–8 months, peripheral blood, bone marrow as well as spleen and liver of the transplanted animals were analyzed and compared to wt and ko mice transplanted with control cells. Transplantation of NFATc2-deficient HPCs into wt recipients (ko → wt) induced similar hematological abnormalities as those occurring in non-transplanted ko mice or in ko mice transplanted with ko cells (ko → ko). Compared to wt mice transplanted with wt cells (wt → wt), ko → wt mice showed evidence of anemia, thrombocytopenia and a significantly reduced number of hematopoietic cells in their bone marrow. Likewise, ko → wt mice developped clear signs of extramedullary hematopoiesis in spleen and liver, which was not the case in wt → wt control animals. Our data demonstrate for the first time, that NFAT transcription factors directly regulate the intrinsic function of hematopoietic progenitor cells in vivo. The transcriptional targets for NFAT in these cells are yet unknown and are the focus of further investigations. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document