SCF and IL-5 Synergize with FIP1L1/PDGFRα To Induce Mastocytosis in a Chronic Eosinophilic Leukemia Murine Model.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3631-3631
Author(s):  
Yoshiyuki Yamada ◽  
Marc E. Rothenberg ◽  
Andrew W. Lee ◽  
Nabeel J.H. Al-Moamen ◽  
David A. Williams ◽  
...  

Abstract FIP1L1/PDGFRα (F/P) fusion protein has been identified as a cause of chronic eosinophilic leukemia (CEL)/hypereosinophilic syndrome (HES). Our group has previously described a murine CEL/HES model based on the expression of both F/P and T-cell-dependent overexpression of IL-5 (Yamada Y et al., Blood 2006), which develops a full hypereosinophilic syndrome with eosinophil infiltration into multiple organs similar to that observed in patients with CEL/HES (CEL-like mouse). Patients with F/P+ HES/CEL also display mastocytosis. Since mast cell (MC) development largely depends on c-kit signaling induced by its ligand SCF, we aimed to determine if SCF collaborates with the F/P fusion protein and IL-5 to enhance MC development. The CEL-like mice showed higher levels of MC infiltration in small intestine compared to transplanted mice with IL-5 transgenic HSC/P (control, Table 1). Interestingly, the intestinal MC infiltration of CEL-like mice was primarily associated with MC residing in the lamina propria and intraepithelial locations associated with villi; whereas MC in control mice were primarily in the crypt areas (47-fold higher mast cell levels in villi compared to those of control mice). In addition to the small intestine, skin MC infiltration was also significantly increased in CEL-like mice (Table 1). Notably, F/P+ BM hematopoietic stem cell/progenitor (HSC/P) showed proliferation and MC differentiation in vitro in the absence of cytokines (2.3-fold cell expansion, 54% c-kit+/FcεRIα+ cells) while empty vector (EGFP alone)-transduced (control) HSC/P did not survive in this culture condition. Such an expansion became even higher (36-fold expansion, 90% c-kit+/FcεRIα+ cells and 2,000-fold higher than control HSC/P) in the presence of low-concentration of SCF (10 ng/ml) for 3 weeks in culture. In contrast, culture with no cytokines or low-dose of SCF did not induce any MC development from control HSC/P. Unlike low-concentration of SCF, IL-3 (100 ng/ml), which induces a rapid and significant MC expansion in control cells, impaired MC development of F/P+ HSC/P (129-fold lower expansion at 3 weeks of culture), suggesting that MC proliferation induced by F/P expression may trigger different signaling pathways than during normal MC differentiation induced by IL-3. In summary, the F/P fusion protein induces murine mastocytosis via SCF/c-kit signaling, which is synergistically enhanced by IL-5 overexpression. Table 1. Tissue Mast Cell Infiltration (Mast Cells/mm2) Control (IL-5 overexpressing) mice CEL-like mice *p<0.05 compared to control; n=4, per group Small Intestine 144 ± 70.2 964 ± 316.5* Skin 129 ± 33.2 208 ± 42.8*

Blood ◽  
2008 ◽  
Vol 112 (6) ◽  
pp. 2500-2507 ◽  
Author(s):  
Yoshiyuki Yamada ◽  
Abel Sanchez-Aguilera ◽  
Eric B. Brandt ◽  
Melissa McBride ◽  
Nabeel J. H. Al-Moamen ◽  
...  

Abstract Expression of the fusion gene FIP1-like 1/platelet-derived growth factor receptor alpha (FIP1L1/PDGFRα, F/P) and dysregulated c-kit tyrosine kinase activity are associated with systemic mastocytosis (SM) and chronic eosinophilic leukemia (CEL)/hypereosinophilic syndrome (HES). We analyzed SM development and pathogenesis in a murine CEL model induced by F/P in hematopoietic stem cells and progenitors (HSCs/Ps) and T-cell overexpression of IL-5 (F/P-positive CEL mice). These mice had more mast cell (MC) infiltration in the bone marrow (BM), spleen, skin, and small intestine than control mice that received a transplant of IL-5 transgenic HSCs/Ps. Moreover, intestinal MC infiltration induced by F/P expression was severely diminished, but not abolished, in mice injected with neutralizing anti–c-kit antibody, suggesting that endogenous stem cell factor (SCF)/c-kit interaction synergizes with F/P expression to induce SM. F/P-expressing BM HSCs/Ps showed proliferation and MC differentiation in vitro in the absence of cytokines. SCF stimulated greater migration of F/P-expressing MCs than mock vector–transduced MCs. F/P-expressing bone marrow–derived mast cells (BMMCs) survived longer than mock vector control BMMCs in cytokine-deprived conditions. The increased proliferation and survival correlated with increased SCF-induced Akt activation. In summary, F/P synergistically promotes MC development, activation, and survival in vivo and in vitro in response to SCF.


Blood ◽  
2002 ◽  
Vol 99 (1) ◽  
pp. 15-23 ◽  
Author(s):  
James C. Mulloy ◽  
Jörg Cammenga ◽  
Karen L. MacKenzie ◽  
Francisco J. Berguido ◽  
Malcolm A. S. Moore ◽  
...  

The acute myelogenous leukemia–1 (AML1)–ETO fusion protein is generated by the t(8;21), which is found in 40% of AMLs of the French-American-British M2 subtype. AML1-ETO interferes with the function of the AML1 (RUNX1, CBFA2) transcription factor in a dominant-negative fashion and represses transcription by binding its consensus DNA–binding site and via protein-protein interactions with other transcription factors. AML1 activity is critical for the development of definitive hematopoiesis, and haploinsufficiency of AML1 has been linked to a propensity to develop AML. Murine experiments suggest that AML1-ETO expression may not be sufficient for leukemogenesis; however, like the BCR-ABL isoforms, the cellular background in which these fusion proteins are expressed may be critical to the phenotype observed. Retroviral gene transfer was used to examine the effect of AML1-ETO on the in vitro behavior of human hematopoietic stem and progenitor cells. Following transduction of CD34+ cells, stem and progenitor cells were quantified in clonogenic assays, cytokine-driven expansion cultures, and long-term stromal cocultures. Expression of AML1-ETO inhibited colony formation by committed progenitors, but enhanced the growth of stem cells (cobblestone area-forming cells), resulting in a profound survival advantage of transduced over nontransduced cells. AML1-ETO–expressing cells retained progenitor activity and continued to express CD34 throughout the 5-week long-term culture. Thus, AML1-ETO enhances the self-renewal of pluripotent stem cells, the physiological target of many acute myeloid leukemias.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1540-1540
Author(s):  
Yoshiyuki Yamada ◽  
Jose A. Cancelas ◽  
Eric B. Brandt ◽  
Abel Sanchez-Aguilera ◽  
Melissa McBride ◽  
...  

Abstract Systemic mastocytosis (SM) associated with chronic eosinophilic leukemia (CEL)/hypereosinophilic syndrome (HES) is a result of expression of the Fip1-like1 (FIP1L1)/platelet-derived growth factor receptor alpha (PDGFRa) (F/P) fusion gene. We have previously described a murine CEL/HES model (CEL-like mice) induced by F/P fusion gene transduction and T-cell overexpression of IL-5 (Yamada Y et al., Blood 2006). We have now validated a preclinical murine model of F/P-induced SM/CEL and analyzed the pathogenesis of SM in this model. F/P+ mast cells (MC, defined as EGFP+/c-kit+/FceRI+) were significantly increased in the small intestine, bone marrow (BM) and spleen of CEL-like mice compared to wild-type mice (Table). CEL-like mice also developed cutaneous MC infiltration. In addition, mMCP-1 serum levels, which correlate well with MC expansion and activation in vivo, were significantly higher in CEL-like mice than in wild-type mice (64,000 ± 23,800 and 38 ± 41.4 pg/ml, respectively). F/P induces increased expansion of BM-derived MC in vitro (∼2,000-fold) and F/P+ BM-derived MC survive longer than wild-type MC in cytokine-deprived medium (28.0 ± 2.3% vs. 8.7 ± 3.1% 7AAD−/Annexin V− cells after 48 hours). This correlated with increased Akt phosphorylation in the F/P+ MC. Since c-kit mutations are the most frequent cause of SM, we analyzed the possible synergistic role of SCF and F/P signaling. F/P and SCF/c-kit signaling indeed synergize in the development of BM-derived MC (16-fold greater expansion than in the absence of SCF) and F/P+ BM-derived MC showed a 3.7-fold greater migratory response to SCF than wild-type BM-derived MC. In order to determine the role of SCF/c-kit signaling in F/P+ MC development, activation and tissue infiltration in vivo,these responses were evaluated in mice that were treated with a blocking anti-c-kit blocking antibody, ACK-2, or an isotype-matched control antibody. ACK-2 treatment suppressed intestinal MC infiltration and elevated plasma levels of mMCP-1 induced by F/P expression by 95 ± 6.0% and 98 ± 0.76%, respectively, whereas MC and plasma mMCP-1 were completely undetectable in wild-type mice treated with ACK2. This suggests that SCF/c-kit interactions may synergize with F/P to induce SM. In summary, mice with CEL-like disease also develop SM. F/P-induced SM is a result of increased in vivo MC proliferation, survival, activation and tissue infiltration. SCF/c-kit signaling synergizes with F/P in vivo and in vitro to promote mast cell development, activation and survival. EGFP+/c-kit+/FcεRI+ cell frequency in tissues of control and CEL-like mice (%) Control mice CEL-like mice Small intestine 1.0±0.95 47±21.4* Bone marrow 0.2±0.14 3±1.9* Spleen 0.05±0.01 3±0.8*


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4039-4039
Author(s):  
Ri Zhang ◽  
Wenjin Gao ◽  
Yuanyuan Sun ◽  
Jingcheng Miao ◽  
Xueguang Zhang

Abstract Transforming growth factor-beta 1 (TGF-β1) is known to maintain primitive human hematopoietic stem/progenitor cells with polyfunctional role in a quiescent state and CD133 is a new stem cell antigen that may provide an alternative to CD34 for the selection and expansion of hematopoietic cells for transplantation. To investigate the specific effect of TGF-β1 on proliferation and differentiation of CD133 positive cells derived from umbilical cord blood (UCB) during short-term culture in vitro, CD133 positive cells from 20 fresh UCB samples were selected using Miltenyi Biotec’s CliniMACS separation device and were cultured in IMDM medium with 20% FCS in the presence of a cytokine combination of SCF, IL-6, thrombopoietin, IL-3 and Flt3-ligand for up to 2 weeks and TGF-β1 with low concentration was also added to the mediumon day 4. The proliferative response was assessed at day 7, day 10 and day 14 by evaluating the following parameters: nucleated cells (NC), clonogenic progenitors (CFU-GEMM,CFU-GM and BFU-E), and immunophenotypes (CD133 and CD34). The results showed that efficacious expansion of various hematopoietic stem/progenitor cells was constantly observed during the culture. The fold expansion of NC on day7, day10 and day14 expansion were 33.59,224.26 and 613.48, respectively. The fold expansion of CFU-GEMM, CFU-GM and BFU-E on day 10 were 24.89, 41.62 and 49.28, respectively, obviously higher than that without ex vivo expansion (P<0.05). The expansions of CD133+, CD133+CD34+ and CD34+ subpopulation on day 14 were up to 25.83-fold, 16.16-fold and 60.54-fold, respectively. Furthermore the expansion systems with TGF-β1 showed more CD133+ cells than control at every time points. Our datas suggested that the CD133+ cells from human UCB have great expansion potential for ex-vivo expansion. The low concentration of TGF-β1 may delay over-differentiation of hematopoietic stem/progenitor cells.


1991 ◽  
Vol 173 (5) ◽  
pp. 1205-1211 ◽  
Author(s):  
P de Vries ◽  
K A Brasel ◽  
J R Eisenman ◽  
A R Alpert ◽  
D E Williams

Pluripotent hematopoietic stem cells (PHSC) are very rare cells whose functional capabilities can only be analyzed indirectly. For a better understanding and possible manipulation of mechanisms that regulate self-renewal and commitment to differentiation of PHSC, it is necessary to purify these cells and to develop assays for their growth in vitro. In the present study, a rapid and simple, widely applicable procedure to highly purify day 14 spleen colony-forming cells (day 14 CFU-S) is described. Low density bone marrow cells (rho less than or equal to 1.078 g/cm3) were enriched by two successive light-activated cell sorting procedures. In the first sort, cells within a predetermined light scatter (blast cell) window that are wheat germ agglutinin/Texas Red (WGA/TxR) positive and mAb 15-1.4.1/fluorescein isothiocyanate negative (granulocyte-monocyte marker) were selected. In the second sort, cells were selected on the basis of retention of the supravital dye rhodamine 123 (Rh123). Cells that take up little Rh123 (Rh123 dull cells) and those that take up more Rh123 (Rh123 bright cells) were 237-fold and 132-fold enriched, respectively, for day 14 CFU-S. Both Rh123 fractions were cultured for various time periods in vitro in the presence of mast cell growth factor (MGF), with or without interleukin 3 (IL-3) or IL-1 alpha. Both Rh123 fractions proliferated in response to MGF alone as determined by a [3H]TdR assay or by counting nucleated cells present in the cultures over time. MGF also acted synergistically with both IL-3 and IL-1 alpha to promote stem cell proliferation. Stimulation of both Rh123 fractions with MGF alone did not result in a net increase of day 14 CFU-S. Stimulation with MGF + IL-3 or MGF + IL-alpha resulted in a 4.4- or 2.6-fold increase of day 14 CFU-S in the Rh123 dull fraction, and an 11.6-fold or 2.6-fold increase of day 14 CFU-S in the Rh123 bright fraction, respectively. The data presented in this paper indicate that in vitro MGF acts on primitive hematopoietic stem cells by itself and also is a potent synergistic factor in combination with IL-3 or IL-1 alpha.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2422-2422
Author(s):  
Elizabeth H. Stover ◽  
Jan Cools ◽  
D. Gary Gilliland

Abstract We recently identified the FIP1L1-PDGFRα fusion protein as a frequent cause of hypereosinophilic syndrome (HES), a disorder characterized by persistent eosinophilia and organ dysfunction. ~60% of HES patients who are sensitive to imatinib, a selective tyrosine kinase inhibitor, harbor the FIP1L1-PDGFRα fusion. The fusion is expressed as a consequence of an interstitial chromosomal deletion of human chromosome 4 that fuses a novel protein FIP1L1 to the cytoplasmic domain of the tyrosine kinase PDGFRα, a type III receptor tyrosine kinase (RTK). The fusion protein is a constitutively active kinase and has transforming properties in vitro and in vivo that are inhibited by imatinib. In order to understand the mechanism of activation of the FIP1L1-PDGFRα fusion kinase, a series of deletions of FIP1L1 were fused to PDGFRα and the fusions were tested for transforming activity. The data suggested that the FIP1L1 moiety was in fact dispensable for PDGFRα activation. For example, substitution of FIP1L1 with the Myc-epitope fused to PDGFRA still resulted in PDGFRα autophosphorylation; transformation of Ba/F3 cells to IL-3 independent growth; and induction of a myeloproliferative disease in a murine bone marrow transplant assay. Structural analyses of other type III RTKs, such as FLT3, have identified an autoinhibitory function of the juxtamembrane (JM) domain. Thus, the lack of requirement for FIP1L1 suggests that activation of the FIP1L1-PDGFRa fusion kinase might be caused by disruption of an autoinhibitory JM domain in PDGFRα. Indeed, in patients with the FIP1L1-PDGFRα fusion, the breakpoints in PDGFRA are tightly clustered within exon 12, which encompasses the JM domain. Whereas activation of most known fusion kinases relies on enforced dimerization by a fusion partner, disruption of an autoinhibitory JM domain may be an alternative mode of fusion kinase activation.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1283-1283
Author(s):  
Catherine L Carmichael ◽  
Donald Metcalf ◽  
Katya J. Henley ◽  
Elizabeth A Kruse ◽  
Ladina Di Rago ◽  
...  

Abstract Abstract 1283 ETS-related gene, ERG, is a key regulator of hematopoietic stem cell (HSC) function, and a potent oncogene. It is involved in chromosomal translocations with the EWS gene in Ewing's sarcoma, the TLS gene in Acute Myeloid Leukemia (AML) and the TMPRSS2 gene in more than half of all prostate cancers. In addition, increased ERG levels are associated with poor prognosis in cytogenetically normal AML and T-cell Acute Lymphoblastic Leukemia; and our recent data suggests that trisomy of ERG is important for development of myeloproliferative disease and Acute Megakaryocytic Leukemia (AMKL) in Down syndrome individuals. The role ERG, and the oncogenic fusion protein TLS-ERG, play during hematopoietic transformation remains unclear. Hematopoietic overexpression of ERG has been shown to induce T-cell leukemia in mice. Development of a non-lymphoid disease has also been described, however this disease was reported to be an AMKL by one group, and a non-malignant erythroid hyperplasia by another. Hematopoietic overexpression of TLS-ERG in mice has not been described. This fusion has been shown to perturb differentiation and increase self-renewal of human myeloid progenitor cells in vitro, and enable the IL-3 dependent L-G murine myeloid progenitor cell line to induce a leukemia-like disease in vivo. In order to clarify and compare the role of wild-type and rearranged forms of ERG in leukemia development, we injected lethally irradiated mice with fetal liver cells (FLCs) transduced with retrovirus carrying either Erg or TLS-ERG. These mice succumbed to disease with a median latency of 80 days after receiving Erg-transduced FLCs, or 44 days after receiving TLS-ERG-transduced FLCs. Consistent with published data, 30% of Erg mice developed T-cell leukemias. Interestingly, no TLS-ERG mice developed this disease. Strikingly, 100% of Erg and TLS-ERG mice developed an identical non-lymphoid disease characterised by hepatosplenomegaly, anemia and leukocytosis. Histopathological and flow cytometric analysis revealed infiltration of the bone marrow, spleen, lung and liver by nucleated erythroblasts, expressing a high level of CD71 and varying levels of Ter119. Interestingly, in some mice a subset of these cells also expressed the megakaryocytic marker CD41. Primary spleen cells were capable of transplanting disease in non-irradiated mice, demonstrating that this disease was malignant. Spleen cells from Erg and TLS-ERG leukemic mice, but not controls, were capable of generating large numbers of small colonies when cultured in methylcellulose stimulated with IL-3/SCF/EPO. These colonies primarily contained erythroblasts (CD71+Ter119+/−), however some cells also expressed CD41 and were acetylcholinesterase positive. Most acetylcholinesterase positive cells were of small size, indicating either incomplete or early megakaryocyte maturation. Combined, these data suggest that hematopoietic overexpression of Erg or TLS-ERG in mice leads to transformation of a bi-potential erythroid-megakaryocyte progenitor. Following transformation, this cell appears to retain some bi-potentiality in vitro, however in vivo it primarily develops along the erythroid lineage. Thus, Erg- and TLS-ERG- induced non-lymphoid disease may be best described as an erythro-megakaryocytic leukemia. Finally, the data also indicate that truncation and fusion to TLS abrogates ERG's ability to transform lymphoid progenitors, however TLS-ERG retains the ability to transform myeloid progenitors in a manner that strongly resembles that of wild-type Erg. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5222-5222
Author(s):  
Jiannong Cen ◽  
Zixing Chen ◽  
Xiaofei Qi ◽  
Li Yao ◽  
Jun He ◽  
...  

Abstract Idiopathic hypereosinophilic syndromes (HES) or chronic eosinophilic leukemia (CEL) comprise a spectrum of indolent to aggressive diseases characterized by persistent hypereosinophilia. Hypereosinophilia can result from the presence of a defect in the hematopoietic stem cell giving rise to eosinophilia, it can present in many myeloproliferative disorders or alternatively it may be a reactive form, secondary to many clinical conditions. The fusion gene FIP1L1-PDGFR alpha was identified in a subset of patients presenting with HES/CEL. In spite of this, the majority of HES/CEL patients do not present detectable molecular lesions and for many of them the diagnosis is based on exclusion criteria and sometimes it remains doubt. CD34-positive progenitor cells from bone marrow (BM) express BAALC and WT1. Overexpression of BAALC and WT1 were seen in patients with AML and ALL. In a subset of AML it marked poor prognosis, suggesting a role for BAALC or WT1 overexpression in acute leukemia. To explored the possibility to distinguish between HES/CEL and reactive hypereosinophilia based on the measurement of BAALC and WT1 transcript amount. Twenty-two patients with hypereosinophilia were characterized at the molecular level and analyzed for BAALC and WT1 expression. The transcription of FIP1L1-PDGFRalpha fusion gene was detected by nested RT-PCR. The relative transcript amount of BAALC and WT1 were determined by real time PCR analyses. The FIP1L1-PDGFRalpha fusion gene expressed has been identified in bone marrow mononuclear cells of 4 cases. The relative expression level of BAALC and WT1 in these 4 cases with positive FIP1L1-PDGFRalpha fusion gene expression were 2.27(0.27–6.8) and 0.39(0.002–0.90), respectively. Whereas the relative amount of transcripts of BAALC and WT1 in 18 patients with negative FIP1L1-PDGFRalpha fusion gene were 0.069(0.015–0.11) and 0.054(0–0.34) respectively. The relative amount of transcripts of BAALC and WT1 in patients with HES/CEL were 32 times and 7 times than that in those with negative FIP1L1-PDGFRalpha fusion gene, respectively. These results clearly demonstrates that BAALC and WT1 quantitative assessment allows to discriminate between HES/CEL and reactive eosinophilia and represents a useful tool for disease monitoring especially in the patients lacking a marker of clonality.


Sign in / Sign up

Export Citation Format

Share Document