Constitutive Heterozygous Expression of JAK2V617F Mutated Kinase Triggers Severe Myeloproliferative Disorder in Knock-in Mice.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2902-2902
Author(s):  
Caroline Marty ◽  
Catherine Lacout ◽  
Linda Fong ◽  
Antoine Martin ◽  
William Vainchenker ◽  
...  

Abstract Abstract 2902 Poster Board II-878 Myeloproliferative neoplasms (MPNs) are frequent malignant hematopoietic pathologies. The acquired Jak2V617F mutation is found in the majority of polycythemia vera (PV), and in approximately half of essential thrombocytosis (ET) and primary myelofibrosis (PMF). Animal models using transgenic or retroviral bone marrow transplant approaches allowing for JAK2V617F expression in hematopoietic cells have demonstrated that this sole mutation is sufficient to induce a MPN in mice. They have also shown that the level of JAK2V617F expression is crucial for MPN phenotypes. Therefore, these over-expression models appeared not suited to study human MPNs. In order to circumvent this problem, we developed a Jak2V617F constitutive and a conditional Knock-in (KI) mouse models. Jak2V617F allele was introduced into the endogenous Jak2 locus, allowing physiological expression of the mutated kinase. The Jak2V617F heterozygocity status was confirmed by allele-specific fluorescent competitive probes in quantitative real-time polymerase chain reaction. Constitutively activated JAKV617F kinase activity was assessed by analyzing STAT5a phosphorylation level in bone marrow and spleen cells by Western blotting. Expression of endogenous JAK2V617F in constitutive KI mice induced a severe PV- to PMF-like phenotype disease, at 5 months of age, characterized in blood by marked polycythemia (Hct 71% ± 3.6%), granulocytosis (WBC counts 79 ± 11 × 106 cells/mL) and thrombocytosis (4.4 ± 0.7 × 109 platelets/mL). Mice displayed enlarged spleens (1.6 ± 0.3 g compared to 0.12 ± 0.01 g for WT mice). Histological examinations of the spleens and femurs revealed advanced degree of fibrosis and trilineage hyperplasia. Flow cytometry analysis showed high levels of TER-119-positive (erythroid) cells in bone marrow and spleen and a marked decrease in lymphocyte percents. The number of CFU-E increased in spleen and included an extremely high percentage of endogenous erythroid colonies (EECs). EECs were also detected in bone marrow. The numbers of BFU-E and CFU-GM were only slightly increased in the spleen but there were no significantly changed in the bone marrow. A major drawback of this constitutive and ubiquitous KI mouse model resides in the inability of female to reproduce even after recovery of normal blood parameters induced by hydroxyurea treatment. Progeny was only obtained through KI male breeding. Embryonic expression of endogenous JAK2V617F was also obtained by crossing Jak2V617F conditional KI mice with CMV-Cre transgenic mice. The offspring presented a similar phenotype than the constitutive KI mice. We performed serial competitive bone marrow (BM) transplantations (BMTs) in irradiated WT recipient mice using from 10% to 100% CMV-Cre Jak2V617F KI mouse BM cells diluted with WT mouse BM cells. Time of disease onset and survival were shorter in mice grafted with high compared to low percentages of KI-derived cells. Reduced occurrences of disease were observed along with successive BMTs or limiting dilution of Jak2V617F-positive clone, suggesting that Jak2V617F provides little or no selective advantage to HSC. This study demonstrates that embryonic endogenous JAK2V617F heterozygous expression results in a severe lethal MPN. This result may explain why no germ line transmission of the Jak2V617F-positive human disease has been reported. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5241-5241
Author(s):  
Wesley O. Greaves ◽  
Shalini Verma ◽  
Tigist Bisrat ◽  
Hamed Rahimi ◽  
Abhaya Paladugu ◽  
...  

Abstract Abstract 5241 Introduction TP53 is the most frequently mutated tumor suppressor gene in human cancers and is usually associated with an aggressive disease course. TP53 mutation has been described in a variety of hematopoietic neoplasms, and has been suggested to play a role in leukemic transformation of myeloproliferative neoplasms (MPN). However, the incidence as well as the clinical and pathogenetic implications of TP53 mutation in each sub-category of MPN, including primary myelofibrosis, have not been described. In this study, we investigated the presence and potential clinical significance of TP53 mutations in a large series of primary myelofibrosis cases. Patients and Methods We retrieved archival bone marrow DNA from 51 consecutive patients diagnosed with primary myelofibrosis at The University of Texas MD Anderson Cancer Center between the years 2005 and 2007. Diagnosis was based on morphologic, immunophenotypic, cytogenetic and molecular evaluation of bone marrow in conjunction with clinical data. Only 2 patients had blast counts >10% (11 and 14%). Twenty nine of 50 (58%) patients showed JAK2 p.V617F mutation and all patients were negative for BCR-ABL1 translocation on routine clinical testing. DNA samples were assessed for sequence variation in exons 4 through 9 of TP53 by both high resolution melting curve (HRM) analysis using LightCycler® 480 System (Roche, Indiana IN) and bidirectional Sanger sequencing using 3730XL DNA Analyzer (Life technologies, Carlsbad CA). Results The mean overall survival was 5.7 years. Five patients developed acute leukemia, all of whom died of disease. By Sanger sequencing, only one (1.9%) case showed an amino acid-altering mutation in TP53: c.707A>G (TAC to TGC) in codon 236 (p.Y236C) of exon 7. In addition, 8 cases showed silent mutations/single nucleotide polymorphisms of unknown significance - c.36G>A (CCG to CCA) in exon 4 (n=3) and c.213A>G (CGA to CGG) in exon 6 (n=6). The p.R72P polymorphism in exon 4 which has been described in other hematopoietic neoplasms was present in 1 patient. All cases with a mutant sequence by Sanger sequencing also showed a variant melting curve pattern by HRM analysis. The patient with TP53 mutation died 2 years after presentation from progressive myelofibrosis without developing acute leukemia. Conclusion TP53 mutation is very rare in primary myelofibrosis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5207-5207 ◽  
Author(s):  
Na Xu ◽  
Yuling Li ◽  
Xuan Zhou ◽  
Lin Li ◽  
Qisi Lu ◽  
...  

Abstract Backgroud and objective: Myeloproliferative neoplasms (MPNs) are malignant disorders by proliferation of one of the myeloid lineages and characteristically show an increase in bone marrow reticulin reticulin-fibrosis.Lysyl oxidases like-2(LOXL2) is copper-dependent amine oxidases that play a critical role in the biogenesis of connective tissue by crosslinking extracellular matrix proteins, collagen and elastin,and Cancer associated-fibroblasts (CAFs) are major mediators in tumor microenvironment. Studies found that loxl2 stimulate CAFs grouth solid tumor,and the expression of LOXL2 is increased in MPN patients,espessionly in PMF patients.Here, we want to evaluate whether the expression of higher LOXL2 associated to CAFs during various MPN progression. Patients and methods: We compared normal bone marrows and those from patients with chronic myeloid leukemia(CML)(include CML-CP n=20,CML-BC n=13),polycythemia vera(PV)(n=18), essential thrombocythemia(ET) (n=23), and primary myelofibrosis (PMF) (n=8). We detected α-smooth actin and reticulin protein by immunohistochemical staining, examined LOXL2 expression by western blot in bone marrow and ELIZA kit in serum. Results: LOXL2 was not detected in normal bone marrows and serum.The level of LOXL2 gene is over expressed in PMF (p<0.01) and CML-BC (p=0.02). In other MPNs a differential pattern of expression were statistically significant (P< 0.010).The level of LOXL2 expression associated with reticulin protein expression in bone marrow, especially if reticulin protein expressed higher than 2+(p=0.01). We detected α-smooth actin positive stromal cells in CML-BC and PMF patients,and the level of LOXL2 expression is related to α-smooth actin positive stromal cells(p<0.05).we also detected α-smooth actin after co-cultured mesenchymal stem cell(MSCs) with sLOXL2 for 96 hour. Conclusion: Higher level of LOXL2 could be promote MPN progression by modulating several functions of surrounding stromal cells which acquire features of cancer-associated fibroblasts involved in the pathogenesis of MPN. These findings maybe used as the basis for future targeted therapy directed against MPN progression. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4974-4974
Author(s):  
Luigi Gugliotta ◽  
Stefano Ascani ◽  
Silvia Asioli ◽  
Emanuela Boveri ◽  
G. Fraternali Orcioni ◽  
...  

Abstract Abstract 4974 Background The bone marrow trephine biopsy (BMB) has a crucial role for the diagnosis of essential thrombocythemia (ET), both according to the PVSG and the WHO criteria. The WHO 2001 criteria enhanced the role of BMB also by distinguishing the true-ET (ET) from the prefibrotic and the early fibrotic chronic idiopathic myelofibrosis. The WHO 2008 criteria, in the JAK2 era, confirmed the diagnostic and prognostic relevance of the histopathological features in ET as well as in the other Ph-neg myeloproliferative neoplasms (MPN). Otherwise, only few validated data are presently available, and the reproducibility in the evaluation of some morphological details is still controversy. Objective To validate ET diagnosis in a large registry-based series of patients by revising the BMB specimens according to the WHO criteria and to evaluate the potential relationship between the histopathological and the clinical parameters at presentation. Methods The hematological centers of the Registro Italiano Trombocitemia (RIT) were invited to participate to this study by sending the BMB specimens (hematoxylin-eosin, Giemsa and silver stains) obtained at diagnosis. The clinic-pathological panel of the RIT (with three hematopathologists as permanent members coordinated by a chairman) performed a centralized revision of the BMB specimens, concomitantly (multiheaded microscopy) and blindly (only patient sex and age were known). The panel described for each case the morphological features and gave a diagnostic conclusion according to WHO criteria as follows: true-ET (ET); or initial primary myelofibrosis (i-PMF) distinguished in prefibrotic PMF (pf-PMF/MF-0) and early fibrotic PMF (ef-PMF/MF-1); or advanced PMF (MF-2 and MF-3); or early polycythemia vera (e-PV); or MPN unclassifiable (MPN-U); or diagnosis other than MPN (No MPN). Results Thirteen centers sent the specimen of BMB at diagnosis of 272 patients registered into the RIT and diagnosed in the years 1986-2002 (group A, cases 66, 24.3%), 2003-2005 (group B, cases 95, 34.9%) and 2006-2008 (group C, cases 111, 40.8%). The patients, 104 (38%) males and 168 (62%) females, had at diagnosis: age over 50 in 64% of cases (median 58); PLT count (109/L) >1000 in16% and <= 600 in 24% of cases (median 789); Hgb level (g/dL) >17 in 1% and <=9 in 5% of cases (median 14.3); WBC count (109/L) >9 in 44% of cases (median 8.6); splenomegaly in 18% of cases. Disease symptoms, thrombotic and hemorrhagic events were reported in 40%, 21%, and 7% of cases, respectively. During the follow-up (median 2.7 years) 65% of patients received cytoreductive drugs (Hydroxyurea 59%, Anagrelide 20%, Interferon 14%, Pipobroman 6.5%, Busulfan 0.5%). The revision of the 272 BMB specimens allowed to the following diagnosis: ET 142 cases, 52.2%; i-PMF 72 cases, 26.5% (distinguished in pf-PMF 19 cases, 7% and ef-PMF 53 cases, 19.5%); e-PV 13 cases, 4.8%; MPN-U 16 cases, 14.3%; No MPN 8 cases, 2.9%. In the patients of groups A, B, and C the rate of ET was increasing (from 44%, to 50% and to 59%, respectively) while the rate of i-PMF was decreasing (from 34%, to 31% and to 18%, respectively); moreover, the rate of e-PV was 0%, 6%, and 6%, respectively. The ET patients compared with the i-PMF patients showed at diagnosis a lower rate of splenomegaly (16/142, 11.3% vs 24/72, 33.3%, p<0.0001), of age >50 years ( 84/142, 59.2% vs 53/72, 73.6%, p<0.03), and a higher rate of PLT count (109/L) <=600 (39/142, 27.5% vs 11/72, 15,3%, p<0.04 ); no significant differences were found between the two groups for sex, Hgb level, WBC count, symptoms, thrombotic and hemorrhagic events. The treatment rate was lower in ET than in i-PMF (ratio 0.7). Conclusion This preliminary report on the revision of the BMB at diagnosis in 272 patients of the Registro Italiano Trombocitemia (RIT) shows that: the rate of true ET is continously increasing (from 44% before 2003 to 59% in the period 2006-2008), with i-PMF rate concomitantly decreasing (from 34% to 18%); the ET patients, compared with the i-PMF ones, were younger, with lower PLT count and with lower rate of splenomegaly, and received a less intensive cytoreductive treatment. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4108-4108
Author(s):  
Ann Brinch Madelung ◽  
Michael Bzorek ◽  
Henrik Bondo ◽  
Eva M.K. Zetterberg ◽  
Ole Weis Bjerrum ◽  
...  

Abstract Abstract 4108 Background: In Philadelphia-negative chronic myeloproliferative neoplasms (MPN) increased microvascular density, bizarre vessel architecture and increased number of pericytes are distinct histopathological features; apart from the characteristic proliferation of myeloid cell lines and the progressive accumulation of connective tissue in the bone marrow. Pericytes express several markers such as CD146, CD271, Smooth Muscle Actin (SMA), Desmin, Platelet-derived growth factor receptor beta and Neuron-glial 2. However, these markers are also expressed in other cell types of which some are related to vascular structures. Immunofluorescence labelling is the golden standard for detection of co-expressed cellular antigens, but due to the crowded cellular environment in bone marrow and excessive autofluorescence, identification of cell types by light microscopy is preferred. Aim: This is a methodological study aiming to identify pericyte marker profiles by light microscopy in bone marrow biopsies, contributing to our understanding of the pathogenesis of MPN. Method and results: Formalin fixed, decalcified and paraffin-embedded blocks of bone marrow trephine specimens from normal donor (n=1) and patients with primary myelofibrosis (PMF) (n=3) were included. Specimens were subjected to an immunohistochemical sequential multi-labelling and erasing technique (SE-technique), inspired by the work of Glass et al. 2009 (J Histochem Cytochem). Briefly, antigens of interest in the first and/or second layer were detected with an immunoperoxidase system and visualised with Amino-Ethyl-Carbazole (AEC). After imaging, erasing of AEC with 96% alcohol and blocking of immunoreagents, the slides were stained with a traditional double immuno-labelling procedure. We successfully applied up to four layers of antibodies using CD146, SMA, CD34, CD271, and Ki67 in different combinations; either displayed as single or single followed by traditional double sequential staining runs (figure 1). In addition to the conventional light microscopy analysis we applied a Photoshop color palette, where the different immunohistochemical reactions in the staining sequence were assigned to the different color channels creating a single composite image. The SE-technique significantly improves morphological studies especially in bone marrow trephines with the cells of interest intermingled with other cells. Additionally, the SE-technique makes it possible to detect more than two antigens regardless of immunoglobulin type or animal host. Conclusion: To our knowledge, the SE-technique described in this study, is the first to multi-label antigens, identifying vessel and pericyte architecture in bone marrow trephines at light microscopic level. This technique may unravel novel aspects of the composition of the microvessel structures in patients with PMF and related neoplasms. The SE-technique displayed as single staining images and Photoshop color palette combined image. Panel A-D shows an identical area in the different steps of the method. A) CD146. Positive pericytes (arrow). B) SMA. Positive pericytes (arrow). C) CD34. Negative pericytes (arrow). D) Combined image with CD146 (green channel), CD34 (red channel), and SMA (blue channel). Coexpression of CD146/CD34 is seen as yellow reaction deposit, and coexpression of CD146/SMA as cyan reaction deposit (arrow). Note, as a negative control, the CD146 positive fat cell in A (arrowhead) - negative in B and C, and SMA positive pericyte in B (arrow) – negative in C. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5160-5160 ◽  
Author(s):  
Rebekka K Schneider ◽  
Isabelle Leisten ◽  
Susanne Ziegler ◽  
Anne Schumacher ◽  
Tim H Brümmendorf ◽  
...  

Abstract Abstract 5160 Myeloproliferative neoplasms (MPNs) are characterized by the excessive production of terminally differentiated nonlymphoid cells or platelets in the bone marrow. They represent a heterogenous group of clonal hematologic malignancies and are classified into chronic myeloid leukemia (CML), essential thrombocythemia (ET), polycythemia vera (PV) and primary myelofibrosis (PMF) and other rarer diseases. MPNs are often associated with extramedullary hematopoiesis and progressive hepatosplenomegaly due to displacement of hematopoietic progenitor cells from the BM to spleen and liver. Progenitor mobilization follows the enhanced deposition of extracellular matrix proteins, which can be found in the bone marrow of MPN patients. Nonhematopoietic bone marrow stromal cells (BMSCs) and their precursors (mesenchymal stem cells) are believed to be conditioned by abundantly released growth factors from the pathological hematopoietic clone in MPNs and in turn contribute to a modified niche environment which participates in the maintenance of the malignant clone and in disease progression. We therefore aimed at the comparative analysis of BMSCs from MPN patients (displaying myelofibrosis 0–1) and healthy donors as well as at their matrix remodelling capacity. BMSCs from PMF patients were obtained by trephine biopsies and isolated by plastic adherence in IMDM, 20% FCS and cortisone. BMSCs from PMF patients fulfilled MSC criteria according to common consensus (Dominici et al., 2007). To compare their potential to support hemato-lymphopoiesis, we analyzed their hemato-lymphotropic cytokine transcription by RT-PCR. MSCs from MPN patients and healthy donors expressed gene transcripts for M-CSF, SDF-1, LIF, FLT3L, Oct-4, SCF, IL-6 and IL-7, suggesting no significant difference in cytokine production. However, when activated through contact with collagen I/III and embedded in three-dimensional scaffolds, MSCs from MPN-patients extensively remodelled the collagenous matrix compared to healthy donors. Under osteogenic and undifferentiated culture conditions, the extracellular matrix (ECM) production by MPN-MSC was strongly enhanced compared to MSCs from healthy controls as shown by RT-PCR and immunohistochemistry for collagen I, IV, fibronectin, laminin and osteopontin. Furthermore, MSCs from MPN patients significantly contracted and densified the collagenous matrix and the ECM deposition by MSCs from MPN patients was highly comparable to the ECM changes observed in corresponding bone punches. We therefore hypothesize that MSCs from MPN patients are primed to produce enhanced ECM proteins, whereas their capacity to support hematopoiesis seems to be unchanged. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1742-1742
Author(s):  
Srdana Grgurevic ◽  
Srdan Verstovsek ◽  
Zhiming Liu ◽  
Taghi Manshouri ◽  
David Harris ◽  
...  

Abstract Abstract 1742 Primary myelofibrosis (PMF) is a stem cell–derived hematologic malignancy, characterized by an expansion of one or more myeloid lineage resulting in bone marrow (BM) hypercellularity, magakaryocyte proliferation with atypia, granulocytic proliferation, and reticulin and/or collagen fibrosis. An acquired activating mutation in Janus kinase 2 at codon V617F (JAK2V617F) is detected in BM cells of the majority of patients with PMF. Constitutively activated JAK2 induces phosphorylation and activation of STAT3. Phosphorylated STAT3 forms heterodimers, translocates to the nucleus, binds to DNA, activates STAT3-target genes, and induces production of cytokines that interact with the BM microenvironment. Hematopoietic stroma derived soluble factors provide PMF cells with survival advantage (Manshouri et al. Cancer Res 71: 3831, 2011) and, as reported previously, most of these factors activate NF-κB in a variety of cell types. NF-κB plays an important role in the survival and proliferation of normal and neoplastic cells. In several hematologic malignancies, the NF-κB p65/p50 dimers were found to be activated to variable degrees. The activation of NF-κB is mediated by either the canonical pathway or the alternative pathway. The canonical pathway is typically activated by extracellular signals that activate the β subunit of the IκB kinase (IKK) complex (IKKβ) that induces the phosphorylation and degradation of the NF-κB inhibitor IκBα. Following IκBα degradation, NF-κB heterodimers translocate to the nucleus and bind to DNA. We have recently found that in chronic lymphocytic leukemia (CLL) constitutively activated STAT3 induces the production of unphsophorylated (U) STAT3. U-STAT3 binds to the NF-κB dimers p65/p50 in competition with IκB and the U-STAT3/NF-κB complex shuttles to the nucleus where NF-κB binds to DNA and activates NF-κB-regulated genes (Liu et al. Mol Cancer Res 9: 507, 2011). Because in PMF constitutively activated JAK2 induces phosphorylation of STAT3 and this activated form of STAT3 induces the production of U-STAT3, we wondered whether, like in CLL, U-STAT3 activates NF-κB in PMF. To determine whether NF-κB is constitutively activated in PMF we obtained BM low density cells from untreated patients with PMF. First we studied low-density BM cells of 11 patients with PMF using the electrophoretic mobility shift assay (EMSA). Cells of all samples bound to a p65/NF-κB DNA-labeled probe and the addition of an unlabelled (cold) p65/NF-κB probe attenuated or completely eliminated the binding. Typically, NF-κB-DNA binding appears and disappears due to repeated degradation and re-synthesis of IκB and the consequent activation and inactivation of NF-κB, respectively. Because we found that NF-κB is constitutively activated in all PMF BM samples we hypothesized that, like in CLL cells, activation of NF-κB in PMF cells is induced by an IκB-unrelated mechanism as reported by Yang J et al. (Cancer Res 65:939, 2005). By using immunoprecipitation of two different PMF BM samples we determined that STAT3 binds to the RelA/p65 NF-κB protein, and by using EMSA we found that anti-STAT3, similar to anti- NF-κB p65 antibodies, attenuated the binding of PMF BM cell extract to the NF-κB DNA probe. Taken together, our data suggest that U-STAT3 binds the NF-κB dimers p65/p50 and constitutively activates NF-κB in PMF. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1608-1608
Author(s):  
Lily Huang ◽  
Huiyu Yao ◽  
Yue Ma

Abstract Myeloproliferative neoplasms (MPNs) are a phenotypically diverse group of pre-leukemic diseases characterized by overproduction of one or more of the myeloid cell lineages. Gain-of -function mutations in the Janus tyrosine kinase 2 (JAK2) are major determinants in MPNs, These include the V617F mutation and mutations in exon 12. Interestingly, MPN phenotype in patients with exon 12 mutations is distinct from that of patients with the V617F mutation. Mechanisms underlying the phenotypic differences are not well understood. We performed an unbiased screen for residues essential for JAK2 auto-inhibition, and identified a panel of novel gain-of-function mutations. Interestingly, three of them with similar kinase activities in vitro elicited distinctive hematopoietic abnormalities in mice. Specifically, JAK2(K539I) results primarily in erythrocytosis, JAK2(N622I) predominantly granulocytosis, and JAK2(V617F) in both. These phenotypes are consistent with clinical data showing that patients with the V617F mutation exhibit erythrocytosis and granulocytosis, whereas those with mutations in exon 12 (where K539 resides) exhibit erythrocytosis only. To determine the mechanisms underlying the phenotypic differences by different JAK2 mutants, we characterized hematopoietic progenitors and precursor subsets in these mice for their proliferation, apoptosis and differentiation. Quantification of the hematopoietic stem and progenitor population showed an increased percentage of granulocyte-monocyte progenitors (GMP) and skewing of differentiation towards the granulocytic lineage in JAK2(V617F) and JAK2(N622I) mice compared to JAK2(K539I) or wild-type JAK2 mice. Because no difference was observed in the proliferation or apoptosis of bone marrow progenitors from JAK2 mutant mice, differentiation of the common myeloid progenitors (CMP) was likely skewed towards GMP by JAK2(V617F) and JAK2(N622I). Consistent with this hypothesis, similar results were observed in colony forming assays from sorted CMP populations. In the spleen, a decrease in GMP apoptosis and an increase in apoptosis of the megakaryocyte-erythrocyte progenitors (MEP) also contributed to the skewing towards the granulocytic lineage in JAK2(N622I) mice. Similar to MPN patients, mice expressing JAK2 mutants exhibited splenomegaly. We found that JAK2 mutants caused redistribution of hematopoietic stem and progenitors from the bone marrow to spleen. As a result, more differentiated precursors were expanded in the spleens of JAK2 mutants mice compared to mice expressing wild-type JAK2. Consistent with their phenotypes, the percentage of Annexin V+7AAD-erythroblasts in JAK2(K539I) and JAK2(V617F) mice was significantly less than in JAK2(N622I) or wild-type JAK2 mice. On the other hand, both proliferation and apoptosis contribute to the differential degrees of granulocytosis among mice expressing different JAK2 mutants. In line with the different effects elicited by different JAK2 mutants in progenitor and precursor cells, signal transduction pathways were differentially activated downstream of different JAK2 mutants. In summary, our results showed that JAK2 mutants differentially skew differentiation in early stem and progenitor compartments, and also regulate apoptosis and proliferation of distinct precursor subsets to cause erythrocytosis or granulocytosis in mice. These results provide the mechanistic basis for the phenotypic diversity observed in MPNs with different JAK2 mutants. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4095-4095
Author(s):  
Edwin Chen ◽  
Lawrence J Breyfogle ◽  
Rebekka K. Schneider ◽  
Luke Poveromo ◽  
Ross L. Levine ◽  
...  

Abstract TET2 mutations are early somatic events in the pathogenesis of acute myeloid leukemia (AML), myelodysplastic syndrome (MDS) and myeloproliferative neoplasms (MPN) and are one of the most common genetic lesions found in these diseases. In MPN, TET2 mutations are enriched within more advanced disease phenotypes such as myelofibrosis and leukemic transformation and often co-occur with the JAK2V617F mutation, which is present in the majority of MPN patients. We have developed and characterized a Jak2V617F conditional knockin mouse (Jak2VF/+), the phenotype of which closely recapitulates the features of human MPN. To determine the impact of Tet2 loss on Jak2V617F-mediated MPN, we crossed Tet2 conditional knockout mice with Jak2VF/+ knockin and Vav-Cre transgenic mice and backcrossed the compound mutant animals. We then characterized the effects of heterozygous and homozygous loss of Tet2 on the phenotype of Jak2VF/+ mice. We assessed peripheral blood counts, histopathology, hematopoietic differentiation using flow cytometry, colony formation and re-plating capacity. We also evaluated the effects of Tet2 loss on the transcriptome of the HSC compartment using gene expression microarrays and on HSC function using competitive bone marrow transplantation assays. Similar to Jak2VF/+/VavCre+ mice, Tet2+/-/Jak2VF/+/VavCre+ and Tet2-/-/Jak2VF/+/VavCre+ mice develop leukocytosis, elevated hematocrits (HCT) and thrombocytosis. Tet2-/-/Jak2VF/+/VavCre+ mice demonstrate enhanced leukocytosis and splenomegaly compared to the other groups. All groups demonstrate myeloid expansion, erythroid hyperplasia and megakaryocytic abnormalities consistent with MPN in the bone marrow and spleen, while more prominent myeloid expansion and megakaryocytic morphological abnormalities are observed in Tet2-/-/Jak2VF/+/VavCre+ mice as compared to the other groups. Notably, we do not see the development of acute myelogenous leukemia (AML) in Tet2-/-/Jak2VF/+/VavCre+ mice at 6 months. We see enhanced expansion of lineagelowSca1+cKithigh (LSK) cells (enriched for HSC) most prominently in the spleens of Tet2+/-/Jak2VF/+/VavCre+ and Tet2-/-/Jak2VF/+/VavCre+ mice as compared to Jak2VF/+/VavCre+ mice. In colony forming assays, we find that Tet2-/-/Jak2VF/+/VavCre+ LSK cells have enhanced re-plating activity compared to Jak2VF/+/VavCre+ LSK cells and that Tet2-/-/Jak2VF/+/VavCre+ LSK cells form more colonies that Tet2-/-/Jak2+/+/VavCre+ cells. Gene expression analysis demonstrates enrichment of a HSC self-renewal signature inTet2-/-/Jak2VF/+/VavCre+ LSK cells. Concordant with this, we find that Tet2-/-/Jak2VF/+/VavCre+ LSK cells have enhanced competitive repopulation at 16 weeks as compared to Jak2VF/+/VavCre+ and Tet2+/-/Jak2VF/+/VavCre+ LSK cells. In aggregate these findings demonstrate that Tet2 loss promotes disease progression in MPN but is insufficient to drive full leukemic transformation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5586-5586
Author(s):  
Maria Jose Penalva Moreno ◽  
Carolina Martinez-Laperche ◽  
Santiago Osorio Prendes ◽  
Elena Buces Gonzalez ◽  
Jose Luis Diez-Martin ◽  
...  

Abstract Introduction: Calreticulin (CALR) is a multifunctional protein regulated by calcium that is located in the endoplasmic reticulum. Recently, mutations in the calreticulin gene have been described in patients with the diagnosis of essential thrombocytemia (ET) and primary myelofibrosis (PMF), mainly in JAK2-negative cases. CALR mutations are localized to exon 9 and generate deletions or insertions that lead to a frameshift change resulting in a mutant protein. The detection of these mutations helps in the actual diagnosis of JAK2 mieloproliferative syndromes (MPN). Our aim is to assess the utility of the determination of these mutations in the management of patients with diagnosis of MPN in our center. Patients and methods: This study includes 94 patients with diagnosis of JAK2-negative MPN retrospectively selected following clinical and analytical criteria between 2008 and 2014 in our center (Table 1, 2). CALR mutations were performed with the use of fluorescent PCR following the methods described by Klampf et al. (NEJM, 2013). Results: 94 patients were analyzed, 77 of them had the diagnosis of TE, 8 of PMF and 9 of others disorders of myelodisplastic/mieloproliferative. 22% of the cases of ET had mutations in CALR (Table 1). In these mutations, a total of 53% were type I mutations (52-bp deletion) and 47% were type II mutations (5-bp insertion). Only one mutation was infrequent, a 46-pb deletion. We have found statistical correlation in the number of platelets depending on the presence of the mutation and in the largest number of platelets in type II mutations. 33% of the cases of PMF had mutations in CALR, all of them type I. Among other diseases not included in MPN, one of them had a type I mutation (data not shown). Conclusions: Our results are close to recently published results regarding the frequency of mutation and as the largest number of platelets in type II mutations with respect to mutation type I. This study confirms the importance of CALR mutations determination in the diagnosis of JAK2-negative ET and PMF with high clinical suspicion. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5215-5215
Author(s):  
Munazza Rashid ◽  
Rifat Zubair Ahmed ◽  
Shariq Ahmed ◽  
Muhammad Nadeem ◽  
Nuzhat Ahmed ◽  
...  

Abstract Myeloproliferative Neoplasms (MPNs) are a heterogeneous group of clonal disorders derived from multipotent hematopoietic myeloid progenitors. Classic "BCR-ABL1-negative" MPNs is an operational sub-category of MPNs that includes polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). These three disorders are characterized by stem cell-derived clonal myeloproliferation. The most common mutation in the MPNs PV, ET and PMF is JAK2 V617F. JAK2 V617F can be detected in about 95% of patients with PV while remaining 5% of PV patients carry a somatic mutation of JAK2 exon 12. Approximately one third of patients with ET or PMF do not carryany mutation in JAK2 or MPL. In December 2013 mutations were described in calreticulin (CALR) gene in 67-71% and 56-88% of JAK2 V617F and MPL negative patients with ET and PMF, respectively. Since this discovery, CALR mutations have not only been recommended to be included in the diagnostic algorithm for MPNs, but also CALR exon 9 mutations have been recognised to have clinical utility as mutated patients have a better outcome than JAK2 V617F positive patients.CALR mutations have also been reported to be mutually exclusive with JAK2 V617F or MPL mutations. According to our knowledge so farthere have been only six reports published,which described patients harbouring concurrent JAK2 V617F and CALR exon 9 mutations; seven ET, three PMF, one PV and one MPN-U. In the present study we are reporting ET patient with coexisting JAK2 V617F and CALR exon 9 mutations from our center. In July 2011, 55-years-old female patient was referred to our hospital with a history of gradual elevation of platelet counts accompanied with pain in right hypochondriac region and feet. Bone Marrow aspirate consisted of 'Stag-horn' appearance Megakarocytes. Multiple platelets aggregates and islands were seen throughout the aspirate smear. ARMS-PCR for JAK2 V617F mutation was positive whereas bidirectional Sanger sequencing for CALR exon 9 exhibited c.1214_1225del12 (p.E405_D408del) mutation pattern. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document