Critical Role of the Gab2/PI3K/mTOR Pathway in the Pathogenesis of Ptpn11 (Shp2) Mutation-Induced Myeloproliferative Disease.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2856-2856
Author(s):  
Wei Liu ◽  
Xia Liu ◽  
Cheng-Kui Qu

Abstract Abstract 2856 Germline and somatic mutations (heterozygous) in Ptpn11 (Shp2), a protein tyrosine phosphatase implicated in multiple cell signaling processes, have been identified in juvenile myelomonocytic leukemia (JMML), a childhood myeloproliferative disease (MPD), and pediatric acute leukemias. These mutations cause hyperactivation of Shp2 catalytic activity and enhance the binding of mutant Shp2 to signaling partners. Ptpn11 mutations are sufficient to drive the development of JMML-like MPD and acute leukemias in mice, suggesting that they play a causal role in the pathogenesis of hematological malignancies. However, the mechanisms by which Ptpn11 mutations induce these malignancies are not completely understood and the signaling partners that mediate the pathogenic effects of Ptpn11 mutations have not been explored. We previously generated a line of conditional knock-in mice with Ptpn11E76K mutation, the most common and most active Ptpn11 mutation found in JMML and acute leukemias. Induced knock-in of this mutation in hematopoietic cells resulted in MPD with full penetrance as a result of aberrant activation of hematopoietic stem cells (HSCs) and myeloid progenitors (J. Exp. Med., 2011). Recently, we discovered that the interaction between Shp2 E76K and Gab2, a prominent interacting protein of Shp2 and a scaffolding protein important for cytokine-induced PI3K/Akt/mTOR signaling, was greatly enhanced, and that mTOR was highly activated in Ptpn11E76K/+ MPD cells. To address the role of Gab2 and mTOR in the pathogenesis of Ptpn11E76K/+ mutation-induced MPD, Ptpn11E76K/+/Gab2-/- double mutant mice were generated and their phenotypes were compared with those of Ptpn11E76K/+ single mutant mice. MPD phenotypes were markedly attenuated in Ptpn11E76K/+/Gab2-/- double mutant mice. Overproduction of myeloid cells in the bone marrow was alleviated, and splenomegaly was diminished in the double mutants. Myeloid cell infiltration in the liver also decreased. Cytokine (IL-3 and GM-CSF) sensitivity of myeloid progenitors was significantly decreased in Ptpn11E76K/+/Gab2−/− mice as compared to that in Ptpn11E76K/+ mice. Hyperactivation of HSCs and excessive myeloid differentiation caused by Ptpn11E76K mutation were largely corrected by deletion of Gab2. Furthermore, we treated Ptpn11E76K/+ mice with Rapmycin, a specific and potent mTOR inhibitor, which substantially diminished MPD phenotypes. Collectively, this study reveals the essential role of the Gab2/PI3K/mTOR pathway in mediating the pathogenic effects of Ptpn11E76K/+ mutation and suggests that Gab2 and mTOR are potential therapeutic targets for the treatment of Ptpn11-associated hematological malignancies. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2000 ◽  
Vol 96 (2) ◽  
pp. 705-710 ◽  
Author(s):  
Patricia Isnard ◽  
Nathalie Coré ◽  
Philippe Naquet ◽  
Malek Djabali

Some chromosomal translocations in acute leukemias involve the fusion of the trithorax-related protein Mll (also called HRX, All1, Htrx,) with a variety of heterologous proteins. In acute lymphoblastic leukemia associated with the t(4;11)(q21;q23) translocation, the4q21 gene that fuses with Mll is AF4. To gain insight into the potential role of AF4 in leukemogenesis and development, this gene was inactivated by homologous recombination in mice. As expected from the tissue distribution of the AF4 transcript, development of both B and T cells is affected in AF4 mutant mice. A severe reduction of the thymic double positive CD4/CD8 (CD4+/CD8+) population was observed; in addition most double- and single-positive cells expressed lower levels of CD4 and CD8 coreceptors. Most importantly, the reconstitution of the double-positive compartment by expansion of the double-negative cell compartment was severely impaired in these mutant mice. In the bone marrow pre-B and mature B-cell numbers are reduced. These results demonstrate that the function of the mAF4 gene is critical for normal lymphocyte development. This raises the possibility that the disruption of the normal AF4 gene or its association with Mll function by translocation may orient the oncogenic process toward the lymphoid lineage. This represents the first functional study using a knock-out strategy on one of the Mll partner genes in translocation-associated leukemias.


2009 ◽  
Vol 16 (2) ◽  
pp. 483-490 ◽  
Author(s):  
Monica Fedele ◽  
Dario Palmieri ◽  
Gennaro Chiappetta ◽  
Rosa Pasquinelli ◽  
Ivana De Martino ◽  
...  

Impairment of the p27kip1 function, caused by a drastic reduction of its expression or cytoplasmic mislocalization, has been frequently observed in thyroid carcinomas. To understand the role of p27kip1 impairment in thyroid carcinogenesis, we investigated the consequences of the loss of p27kip1 expression in the context of a mouse modeling of papillary thyroid cancer, expressing the TRK-T1 oncogene under the transcriptional control of thyroglobulin promoter. We found that double mutant mice homozygous for a p27kip1 null allele (TRK-T1/p27−/−) display a higher incidence of papillary thyroid carcinomas, with a shorter latency period and increased proliferation index, compared with p27kip1 wild-type compounds (TRK-T1/p27+/+). Consistently, double mutant mice heterozygous for a p27kip1 null allele (TRK-T1/p27+/−) show an incidence of thyroid carcinomas that is intermediate between TRK-T1/p27−/− and TRK-T1/p27+/+ mice. Therefore, our findings suggest a dose-dependent role of p27kip1 function in papillary thyroid cancer development.


2001 ◽  
Vol 21 (13) ◽  
pp. 4391-4398 ◽  
Author(s):  
Makoto Tanaka ◽  
Martina Schinke ◽  
Hai-Sun Liao ◽  
Naohito Yamasaki ◽  
Seigo Izumo

ABSTRACT Nkx2.5 and Nkx2.6 are murine homologs of Drosophilatinman. Their genes are expressed in the ventral region of the pharynx at early stages of embryogenesis. However, no abnormalities in the pharynges of embryos with mutations in either Nkx2.5 or Nkx2.6 have been reported. To examine the function of Nkx2.5 and Nkx2.6 in the formation of the pharynx, we generated and analyzed Nkx2.5 and Nkx2.6 double-mutant mice. Interestingly, in the double-mutant embryos, the pharynx did not form properly. Pharyngeal endodermal cells were largely missing, and the mutant pharynx was markedly dilated. Moreover, we observed enhanced apoptosis and reduced proliferation in pharyngeal endodermal cells of the double-mutant embryos. These results demonstrated a critical role of the NK-2 homeobox genes in the differentiation, proliferation, and survival of pharyngeal endodermal cells. Furthermore, the development of the atrium was less advanced in the double-mutant embryos, indicating that these two genes are essential for both pharyngeal and cardiac development.


Blood ◽  
2010 ◽  
Vol 116 (18) ◽  
pp. 3611-3621 ◽  
Author(s):  
Dan Xu ◽  
Siying Wang ◽  
Wen-Mei Yu ◽  
Gordon Chan ◽  
Toshiyuki Araki ◽  
...  

Abstract Germline and somatic gain-of-function mutations in tyrosine phosphatase PTPN11 (SHP-2) are associated with juvenile myelomonocytic leukemia (JMML), a myeloproliferative disease (MPD) of early childhood. The mechanism by which PTPN11 mutations induce this disease is not fully understood. Signaling partners that mediate the pathogenic effects of PTPN11 mutations have not been explored. Here we report that germ line mutation Ptpn11D61G in mice aberrantly accelerates hematopoietic stem cell (HSC) cycling, increases the stem cell pool, and elevates short-term and long-term repopulating capabilities, leading to the development of MPD. MPD is reproduced in primary and secondary recipient mice transplanted with Ptpn11D61G/+ whole bone marrow cells or purified Lineage−Sca-1+c-Kit+ cells, but not lineage committed progenitors. The deleterious effects of Ptpn11D61G mutation on HSCs are attributable to enhancing cytokine/growth factor signaling. The aberrant HSC activities caused by Ptpn11D61G mutation are largely corrected by deletion of Gab2, a prominent interacting protein and target of Shp-2 in cell signaling. As a result, MPD phenotypes are markedly ameliorated in Ptpn11D61G/+/Gab2−/− double mutant mice. Collectively, our data suggest that oncogenic Ptpn11 induces MPD by aberrant activation of HSCs. This study also identifies Gab2 as an important mediator for the pathogenic effects of Ptpn11 mutations.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3612-3612
Author(s):  
Fengchun Yang ◽  
Jayme Allen ◽  
Shi Chen ◽  
Yan Li ◽  
Jin Yuan ◽  
...  

Abstract Neurofibromin, the protein encoded by the NF1 tumor-suppressor gene, negatively regulates the output of p21ras proteins by accelerating the hydrolysis of active Ras-guanosine triphosphate to inactive Ras-guanosine diphosphate. Children with neurofibromatosis type 1 (NF1) are predisposed to juvenile myelomonocytic leukemia (JMML) associated with loss of the normal NF1 allele. Genetically engineered mice containing nullizygous loss of Nf1 in the hematopoietic system develop a leukemia with complete penetrance that is similar to JMML, including hepatosplenomegaly, elevated peripheral blood cell counts, and elevated numbers of myeloid progenitors that are hypersensitive to multiple cytokines, particularly GM-CSF. Though we and others have found that p21ras is hyperactivated in Nf1−/− myeloid progenitors, inhibiting Ras specifically as a therapeutic target has been challenging. Therefore, identification of alterations in distinct p21ras effector pathways that control leukemia progression in Nf1-deficient cells is critical for understanding disease pathogenesis and identifying therapeutic targets. Here we intercrossed MxCre; Nf1flox/flox mice with mice that are deficient in the small Rho GTPases Rac1 or Rac2 to generate syngeneic progeny that were MxCre; Nf1flox/flox, MxCre; Nf1flox/flox;Rac1flox flox or MxCre; Nf1flox/flox; Rac2 −/−. Consistent with previous studies (Le, Blood 2004) MxCre; Nf1flox/flox mice develop a progressive myeloproliferative disease with 100% penetrance 6 months following interferon inducible induction of the MxCre transgene to disrupt the Nf1flox alleles. Eighty percent of MxCre; Nf1flox/flox die by 9 months after inactivation. Genetic disruption of Rac2 was not sufficient to diminish the onset or severity of the characteristic myeloproliferative disease of MxCre; Nf1flox/flox mice. In contrast, MxCre; Nf1flox/flox; Rac1flox/flox mice followed for 11 months all survived and had normal bone marrow cellularity, spleen weight and splenic architecture. MxCre; Nf1flox/flox mice have elevated numbers of both HPP-CFC and LPP-CFC in the bone marrow and spleen. In contrast, MxCre; Nf1flox/flox; Rac1flox/flox mice had myeloid progenitor numbers that were comparable to wildtype, age-matched controls. Further, though myeloid progenitors from MxCre; Nf1flox/flox mice are hypersensitive to GM-CSF, myeloid progenitors from MxCre; Nf1flox/flox; Rac1flox/flox have a sensitivity to GM-CSF comparable to wildtype controls. The correction in the myeloproliferative phenotype isolated from MxCre; Nf1flox/flox; Rac1flox/flox mice was associated with the reduction in Rac-GTP and prolonged Erk phosphorylation, a MAPK effector that is characteristically elevated in Nf1−/− myeloid progenitors. Collectively, these genetic data identify Ras-Rac1 signaling pathway as a key axis in the genesis of juvenile myelomonocytic leukemia and provide evidence that Rac1 is a therapeutic molecular target for this myeloproliferative disease that currently has no effective therapies.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 392-392
Author(s):  
Dan Xu ◽  
Xia Liu ◽  
Wen-Mei Yu ◽  
Howard J. Meyerson ◽  
Stanton L. Gerson ◽  
...  

Abstract Abstract 392 SHP2, a protein tyrosine phosphatase implicated in multiple cell signaling processes, plays an essential role in hematopoietic cell development. Our previous studies have demonstrated that this phosphatase is required for erythroid, myeloid, and lymphoid development and that it functions in cytokine signaling in both catalytically-dependent and –independent manners. Notably, germline and somatic mutations (heterozygous) in PTPN11 (encoding SHP2) have been identified in 35% of the patients with juvenile myelomonocytic leukemia (JMML), a childhood myeloproliferative disorder (MPD). Furthermore, PTPN11 mutations are also found in pediatric myelodysplastic syndromes (10%), B cell lymphoblastic leukemia (B-ALL) (7%), acute myeloid leukemia (AML) (4%), and sporadic solid tumors. These mutations result in hyperactivation of SHP2 catalytic activity. In addition, PTPN11 disease mutations, especially leukemia mutations, enhance the binding of mutant SHP2 to signaling partners. Although previous studies have shown that Ptpn11 mutations induce cytokine hypersensitivity in myeloid progenitors and MPD in mice, it is unclear whether Ptpn11 mutations also play a causal role in the pathogenesis of acute leukemias. If so, the underlying mechanisms and the cell origin of leukemia initiating/stem cells (LSCs) remain to be determined. PTPN11E76K mutation is the most common and most active PTPN11 mutation found in JMML and acute leukemias. However, the pathogenic effects of this mutation have not been well characterized. We created Ptpn11E76K conditional knock-in mice. Global Ptpn11E76K/+ mutation resulted in early embryonic lethality associated with enhanced ERK signaling. Induced knock-in of this mutation in pan hematopoietic cells led to MPD as a result of aberrant activation of hematopoietic stem cells (HSCs) and myeloid progenitors. These animals subsequently progressed to acute leukemias. Intriguingly, in addition to AML, T-ALL and B-ALL were evolved. PTPN11E76K/+ mutation induced LSC development not only in stem cells but also in lineage committed progenitors as tissue-specific knock-in of Ptpn11E76K/+ mutation in myeloid, T lymphoid, and B lymphoid progenitors also resulted in AML, T-ALL, and B-ALL, respectively. Further analyses revealed that Shp2 was distributed to centrosomes and that Ptpn11E76K/+ mutation promoted LSC development partly by causing centrosome amplification and genomic instability. Thus, Ptpn11E76K mutation has non-lineage specific effects on malignant transformation of hematopoietic cells and initiates acute leukemias at various stages of hematopoiesis. This mutation may play an initiating role in the pathogenesis of pediatric acute leukemias. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2000 ◽  
Vol 96 (2) ◽  
pp. 705-710 ◽  
Author(s):  
Patricia Isnard ◽  
Nathalie Coré ◽  
Philippe Naquet ◽  
Malek Djabali

Abstract Some chromosomal translocations in acute leukemias involve the fusion of the trithorax-related protein Mll (also called HRX, All1, Htrx,) with a variety of heterologous proteins. In acute lymphoblastic leukemia associated with the t(4;11)(q21;q23) translocation, the4q21 gene that fuses with Mll is AF4. To gain insight into the potential role of AF4 in leukemogenesis and development, this gene was inactivated by homologous recombination in mice. As expected from the tissue distribution of the AF4 transcript, development of both B and T cells is affected in AF4 mutant mice. A severe reduction of the thymic double positive CD4/CD8 (CD4+/CD8+) population was observed; in addition most double- and single-positive cells expressed lower levels of CD4 and CD8 coreceptors. Most importantly, the reconstitution of the double-positive compartment by expansion of the double-negative cell compartment was severely impaired in these mutant mice. In the bone marrow pre-B and mature B-cell numbers are reduced. These results demonstrate that the function of the mAF4 gene is critical for normal lymphocyte development. This raises the possibility that the disruption of the normal AF4 gene or its association with Mll function by translocation may orient the oncogenic process toward the lymphoid lineage. This represents the first functional study using a knock-out strategy on one of the Mll partner genes in translocation-associated leukemias.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3585-3585
Author(s):  
Y. Lucy Liu ◽  
Yan Yan ◽  
Cody Webster ◽  
Lijian Shao ◽  
Shelly Y. Lensing ◽  
...  

Abstract Cancer is caused by accumulated genomic and epigenetic abnormalities during the development of an individual, particularly during the neonatal period, when developmental plasticity is actively occurring. Myeloid-specific deletion of pten in embryos or after 3 weeks of age causes acute monocytic or myeloid leukemia (AML) or acute lymphoblastic leukemia (ALL) following a transient myeloproliferative neoplasm (MPN) in adult mice, which can mimic the human diseases to varying degrees. However, it is not clear how the timing of genomic and epigenetic abnormalities contributes to the disease phenotype in a mouse that is of an age comparable to human children. We hypothesized that during the development/aging process, the timing of when the genomic abnormality or “hit” occurs, such as loss of Pten or Nf1, is a critical determinant of the disease phenotype. We tested this by investigating the effect of somatic deletion of Pten at an age of 8 days, one of the most vulnerable stages for malignancy development in mice with or without a germline mutant Nf1. Through crossbreeding, we generated mice with Ptenfl/flNf1Fcr/+Mx1-Cre+ on a C57BL6/129 genetic background, and conditionally deleted Pten in a myeloid-specific manner by intraperitoneal injection of Poly(I:C). Mice with a pten deletion and mutant Nf1 (ptenkoNf1mut, hereafter referred as double mutant) showed signs of sickness at the end of the 2nd week of life, and all died by age 3-5 weeks (equivalent to 1-3 years old in humans). The natural survival in double mutant mice (n=10) was significantly shorter than those with wild type pten and Nf1 (ptenwt; Nf1wt, hereafter referred as WT, n=6, median 0.9 vs >14 months, p<0.001). At autopsy, the double mutant mice had obvious hepatosplenomegaly compared to the WT littermates, but no lymphadenopathy nor thymus enlargement was detected. We then analyzed 15 littermate pairs with/without mutations at age 3-4 weeks. The spleen and liver sizes in the double mutants were significantly increased compared to that in WT (median weights: spleen-198 vs 69mg, p< 0.001; liver-836 vs 532mg, p=0.013). Peripheral blood (PB) analysis showed no difference in WBC in both groups, but significant decreases in double mutant mice in RBC and Hgb (median RBC-5.6 vs 7.6x106/ul, p=0.010; Hgb-10.1 vs 12.6g/ul, p=0.022), but with a normal MCV. HE and IHC stained tissue sections of formalin-fixed organs with antibodies to B220, CD3e, or F4/80 found increased cellularity in bone marrow (BM), and severe infiltration of mature myeloid cells in double mutant spleens, livers, and lungs. Flow cytometric analysis showed that double mutant mice had significantly increased cells marking for monocyte/macrophages(Mac-1+Gr-1int, or Mac-1+CD115+) and granulocytes (Mac-1+ Gr-1+)[p≤0.001] in BM and PB; both T-cells (CD3e+) and B-cells (CD19) were significantly reduced in PB and spleen (p<0.01); platelets (CD41+Ter119-) were reduced in PB. An erythroid differentiation block [indicative of myelodysplasia (MDS) or leukemia] was found in double mutant mice (significant increases in Ter119+CD71high cells). Pten deleted mice without mutant Nf1had a similar phenotype, but survived longer than double mutant mice (median 1.2 vs 0.9 months, p<0.001). Colony formation assays showed no cytokine hypersensitivity in BM or spleen cells in double mutants in response to GM-CSF or IL-3. Based on the Bethesda proposals for classification of non-lymphoid hematopoietic neoplasms in mice, we believe that we have developed a mouse model of myeloproliferative disease with features of a pediatric unclassifiable mixed MDS/MPN. It also mimics to some degree human Juvenile Myelomonocytic Leukemia (JMML), in terms of age of onset and organ infiltration with myelomonocytic cells, but fails to mimic JMML with respect to GM-CSF hypersensitivity. We demonstrate that deletion of pten at an early age in the mouse causes a fatal mixed MDS/MPN that is different from the previously reported mouse models, in which myeloid-specific deletions of Pten were induced in embryos or after 3 weeks of age, and these mice developed only a transient MPN, all eventually dying of AML or ALL at adult ages. To the best of our knowledge, this is the first model in young mice bearing a somatic mutation that resembles a human pediatric unclassifiable MDS/MPN and more closely represents the true genetic and epigenetic background, thus providing a novel tool to study these mechanisms underlying pediatric MDS/MPN. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 818-818 ◽  
Author(s):  
Wei Liu ◽  
Xia Liu ◽  
Wen-Mei Yu ◽  
Kevin D. Bunting ◽  
Cheng-Kui Qu

Abstract Effective therapeutic interventions for juvenile myelomonocytic leukemia (JMML), a fatal childhood malignancy, are lacking. Relapse is the most frequent cause of treatment failure, most likely due to the persistence of leukemic stem cells (LSCs), a small population of self-renewing precursor cells that give rise to the bulk of tumor cells. This reservoir of tumor cells are responsible for long-term maintenance of leukemia growth, and are also a major source of drug resistance. Clearly, a novel approach focused on the unique properties of LSCs is needed. However, it remains a critical challenge how such cells may be eradicated. JMML is known to be caused by genetic mutations in cell signaling proteins involved in the Ras pathway, among which the phosphatase Ptpn11 (Shp2) is most frequently mutated. Ptpn11 mutations (heterozygous) cause greatly increased catalytic activity. We have recently created conditional knock-in mice with the Ptpn11E76K mutation, the most common Ptpn11 mutation found in JMML. Induction of the Ptpn11E76K/+ mutation in these mice (Ptpn11E76K/+/Mx1-Cre+) leads to JMML-like myeloid malignancy with full penetrance by aberrant activation of stem cells and myeloid progenitors. In an effort to understand the biological properties of LSCs in JMML, we studied leukemic hematopoietic stem cells (Lin-Sca-1+c-Kit+Flk2-CD150+CD48- cells)(referred to as LSCs since they reproduce the same disease in sub-lethally-irradiated transplants) in this mouse model. We found that Stat5 was hyper-activated in LSCs in Ptpn11E76K/+/Mx1-Cre+ mice in the chronic phase. Stat5 hyperactivation is likely to be mediated through Jak2 kinase as Jak2 is highly activated in Shp2 E76K-expressing cells. More importantly, these LSCs appear to rely on hyperactivation of Stat5 for maintenance and self-renewal because deletion of Stat5ab in Ptpn11E76K/+/Mx1-Cre+/Stat5abfl/fl double mutant mice resulted in massive cell death in LSCs while neither Stat5ab knockout nor Ptpn11E76K/+ knock-in alone mice had these stem cell phenotypes. Apoptotic cells in the stem cell population were 6.38±1.42, 4.06±0.51, 10.52±6.88, and 24.50±10.27% in Ptpn11+/+/Mx1-Cre+, Ptpn11E76K/+/Mx1-Cre+, Stat5abfl/fl/Mx1-Cre+, and Ptpn11E76K/+/Mx1-Cre+/Stat5abfl/fl mice, respectively. Consequently, the stem cell pool in Ptpn11E76K/+/Mx1-Cre+/Stat5abfl/fl double mutants was drastically decreased. Numbers of stem cells per femur were 3.36±1.51, 0.94±0.63, 1.15±0.39, and 0.17±0.20 (x103) in Ptpn11+/+/Mx1-Cre+, Ptpn11E76K/+/Mx1-Cre+, Stat5abfl/fl/Mx1-Cre+, and Ptpn11E76K/+/Mx1-Cre+/Stat5abfl/fl mice, respectively. Ptpn11E76K/+/Mx1-Cre+/Stat5abfl/fl mice died of pan cytopenia within 4-8 weeks of Stat5 deletion while none of other groups of mice did. Repopulation capabilities of the double mutant stem cells were essentially lost in sub-lethally-irradiated recipient animals. Further mechanistic investigations revealed that tyrosine phosphorylation levels (indicative of activity) of Stat3, one of the substrates of the Shp2 phosphatase, were decreased by ~7.50 fold in Ptpn11E76K/+ LSCs as compared to Ptpn11+/+ control cells due to the enhanced dephosphorylation by the hyperactive Shp2 E76K mutant. Thus, diminished Stat3 activity sensitized Ptpn11E76K/+ LSCs to Stat5 depletion-induced cell death. Taken together, this study suggests a crucial role of Stat5 in the maintenance of LSCs in Ptpn11-associated JMML. The synthetic lethality induced by loss of Stat5 in Ptpn11-mutated JMML raises the possibility that clinically-used inhibitors of upstream Jak2 kinase may be effective in eradicating LSCs in this particular subtype of JMML. Disclosures No relevant conflicts of interest to declare.


2016 ◽  
Vol 7 (1-2) ◽  
pp. 141-151
Author(s):  
Mario Morales-Martinez ◽  
Luz A. Franco-Cea ◽  
Liliana Moreno Vargas ◽  
Otoniel Martinez-Maza ◽  
Sara Huerta-Yepez ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document