Abstract
Myeloproliferative neoplasms such as polycythemia vera (PV), which are associated with the JAK mutation V617F, remain incurable despite progress in the use of JAK2 inhibitors for treatment of some of these diseases. In this study, we employed mice that undergo JAK2V617F-induced PV as a tool to explore new candidate targets for therapy. Our investigations focused on the lipid metabolic enzyme arachidonate 5-lipoxygenase (Alox5), which we found to be strongly upregulated by JAK2V617F in hematopoietic cells in vitro and in vivo. Notably, genetic deletion of Alox5 or its inhibition in mice with a bioactive small-molecule inhibitor was sufficient to attenuate PV development. This therapeutic effect was associated with induction of a blockade in cell-cycle progression and also with apoptosis in PV cells. Genetic loss exerted an inhibitory effect on PV-initiating cells. Similarly, Alox5 inhibition was sufficient to suppress colony formation in human JAK2V617F-expressing CD34+ cells. Mechanistic investigations showed that Alox5 inhibition reduced AKT activation and decreased β-catenin expression in JAK2V617F-expressing cells. Together, our results define Alox5 as a key genetic effector of JAK2V617F in driving PV, and they identify this enzyme as a candidate therapeutic target to treat this refractory myeloproliferative neoplasm. Cancer Res; 77(1); 164–74. ©2016 AACR.
Introduction
The myeloproliferative neoplasms (MPN) are hematologic malignancies with a chronic clinical course and a risk of developing thrombosis and acute leukemia. JAK2 is a member of the Janus family of non–receptor tyrosine kinases and is required for signaling from the erythropoietin (Epo) receptor and other type I cytokine receptors (1). The somatic activating point mutation of JAK2 (JAK2V617F) has been found in Philadelphia chromosome-negative MPNs (2, 3) in approximately 95% of patients with polycythemia vera (PV), 60% of patients with essential thrombocytosis, and 50% of patients with primary myelofibrosis (3, 4). JAK2V617F-positive cells outpace normal hematopoietic cells as a result of its constitutively active kinase activity and activation of growth factor signaling (5–7). This acquired somatic mutation stimulates independent growth of hematopoietic cells (1, 5, 8). Although JAK2 inhibitors have been shown to help to improve symptoms and quality of life in patients (9, 10), it is unlikely that these inhibitors are curative.
Mouse models of JAK2V617F-induced MPNs have been developed in recent years. In the retroviral transduction/transplantation mouse model, JAK2V617F was shown to induce MPN reminiscent of human PV, characterized by erythrocytosis and granulocytosis (11–13). JAK2V617F induces PV-like myeloproliferative disease in mice, although the severity of the disease was variable among different inbred mouse strains. JAK2V617F induces more robust leukocytosis and neutrophilia in BALB/c mice than in C57BL/6 mice (11–13). However, JAK2V617F does not induce thrombocytosis in these mice (11–13). Nevertheless, available mouse models mostly recapitulate myeloproliferative disorder induced by JAK2V617F. In JAK2V617F knock-in mice, hematopoietic stem cells (HSC) were shown to contain PV-initiating cells, but a JAK2 kinase inhibitor failed to eliminate this cell population (7). New therapeutic strategies need to be developed for treating PV more effectively. Because HSCs harbor JAK2V617F in patients with PV (5), PV should be considered as a stem cell–derived disease. Compared with PV, Philadelphia chromosome–positive chronic myeloid leukemia (CML) is also derived from HSCs and has a myeloproliferative phenotype similar to PV in mice (14). Therefore, we reasoned that a gene essential for CML development might also play a critical role in PV development. We have shown that the survival and self-renewal of CML-initiating cells require the arachidonate 5-lipoxygenase gene (Alox5) and that Alox5 is essential for CML development (15). In this study, we investigated the role of Alox5 in pathogenesis of PV in mice. We show that deletion of Alox5 or inhibition of Alox5 function attenuates PV development, suggesting a new strategy for treating PV.
Materials and Methods
Cell lines
JAK2V617F-expressing Ba/F3 cell line that expresses both JAK2V617F and GFP was kindly provided by Dr. Ross Levine (Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY) in 2014. Ba/F3 cells were transduced with pMSCV-IRES-GFP to only express GFP and used as a control, and this GFP-expressing Ba/F3 cell line was generated about 5 years ago. HEL, a human JAK2V617F-expressing cell line, was purchased from ATCC in 2016. All cell lines were grown in RPMI-1640 medium containing 10% FCS and 50 μmol/L 2-mercaptoethanol without exogenous cytokines, except for Ba/F3 cells that were grown in the presence of IL3. Ba/F3 cell line was obtained in 2001. All these cell lines were authenticated by us for confirming expression of JAK2V617F by Western blot analysis and of GFP by FACS analysis.
Mice
C57BL/6J, BALB/c, and homozygous Alox5 knockout (Alox5−/− in C57BL/6 background) mice were obtained from The Jackson Laboratory.
Retroviral bone marrow transduction and transplantation model of PV
pMSCV-JAK2V617F-GFP DNA construct was kindly provided by Dr. Richard Van Etten (Tufts-New England Medical Center). To induce PV in mice, the JAK2V617F retrovirus was generated by co-transfection of 293T cells with pMSCV-JAK2V617F-GFP and an ecotropic packaging construct and used to transduce bone marrow cells from donor mice, and after normalizing GFP+ donor cells, equal number of the transduced GFP+ cells were transplanted into each lethally irradiated recipient mouse as described previously (13).
Flow cytometric analysis of hematopoietic cells in normal and PV mice
Prior to FACS analysis, red blood cells (RBC) in bone marrow or peripheral blood cells from PV or normal mice were eliminated using NH4Cl/NH4HCO3 RBC lysis buffer (pH 7.4). For FACS analysis, hematopoietic cells were washed with PBS and stained with fluorescence-labeled antibodies (B220-PE for B cells, Gr-1-APC and Mac-1-PE for neutrophils). For stem cell analysis, bone marrow cells were suspended in staining medium [Hank Balanced Salt Solution (HBSS) with 2% heat-inactivated calf serum] and incubated with biotin-labeled lineage antibodies containing a mixture of antibodies against CD3, CD4, CD8, B220, Gr-1, Mac-1, and Ter119. After washing, the fluorochrome-labeled secondary antibody (APC-Cy7-conjugated streptavidin) for recognizing biotin and phycoerythrin (PE)-conjugated c-Kit and allophycocyanin (APC)-conjugated Sca-1 antibodies were added to the cells. All these antibodies were purchased from eBioscience Biotechnology. Human CD34+ cells from patients with PV or healthy individuals were cultured in serum-free medium (SFM) containing SCF, TPO, Flt-3 ligand, and IL3 at 50 ng/mL, and cells were treated with various doses of zileuton for 48 hours. Cells were stained with a CD34 monoclonal antibody (BD Biosciences) initially and then washed with the binding buffer provided by the manufacturer before staining with Annexin-V (BD Biosciences). Data were acquired on a FACS Calibur flow cytometric analyzer (Becton Dickinson).
Western blot analysis
Antibodies against p-JAK2, JAK2, p-STAT5, STAT5, p-AKT, AKT, p-PI3K, PI3K, p-MAPK, and MAPK were purchased from Cell Signaling Technology. Antibodies against ALOX5 (5-LO) and ACTIN were purchased from Santa Cruz Biotechnology. Protein lysates were prepared by lysing cells in RIPA buffer, and Western blot analysis was carried out as described previously (16).
Drug treatment
For treating PV mice, zileuton (Critical Therapeutics) was dissolved in water and given orally in a volume of less than 0.3 mL by gavage (300 mg/kg, once a day) beginning at 2 months after induction of PV. Water was used as a placebo. For in vitro assay, zileuton (Cayman Chemicals) was dissolved in DMSO to make 100 mmol/L stock solution before it was diluted in culture medium for use. DMSO was used as a control. Ruxolitinib (Cayman Chemicals) was dissolved in ethanol to make 10 μmol/L stock solution before it was diluted in culture medium for use.
EPO measurement
Plasma was collected by centrifugation of peripheral blood from mice at 1,000 × g for 5 minutes. EPO levels in plasma were determined by ELISA kit (R&D Systems).
Leukotriene B4 measurement
Plasma samples were collected from mice and stored at −80°C before use. The levels of leukotriene B4 (LTB4) in plasma were determined using an ELISA kit (Cayman Chemical) following the manufacturer's instruction. Briefly, 50 μL of each standard or plasma sample was added to each well of a 96-well plate and then 50 μL of LTB4 AchE Tracer was added to each well. Next, 50 μL of LTB4 EIA antiserum was added to each well, and the 96-well plates were incubated at 4°C overnight. The plates were read using an ELISA microplate reader.
Specimen collection and cell preparation
Peripheral blood was collected from patients with PV at the Icahn School of Medicine at Mount Sinai (ISMMS; Mount Sinai, NY). Written informed consent was obtained from all patients according to guidelines established by the Institutional Review Board of the ISMMS. All patients met the World Health Organization diagnostic criteria for PV. The blood samples were layered onto Ficoll-Hypaque (1.077 g/mL; GE Healthcare) and low-density mononuclear cells were separated after centrifugation. The CD34+ cell population was isolated using a human CD34+ cell selection kit (StemCell Technologies) according to the manufacturer's instructions. Normal human CD34+ cells were purchased from AllCells.
Cell proliferation assay for human PV cells
Mononuclear cells isolated from patients with PV or healthy donors were cultured in 12-well plates with IMDM and supplemented with 20% FBS, 100 U/mL penicillin, and 10 μg/mL streptomycin (Invitrogen). Equal cell numbers were plated in both untreated and treated well at day zero. Cells were treated with various doses of zileuton (50–500 μmol/L) for 4 days. Cell proliferation was determined using the PrestoBlue Cell Viability Reagent (Life Technologies) according to the instructions provided by the manufacturer. Briefly, the reagent was added directly to the cell culture wells at 1:10 dilution, and the cells were incubated for 2 hours. The viable cell number was determined by measuring fluorescence intensity. The fluorescence intensity was measured at 560 nm (excitation) and 590 nm (emission) using a Spectra Max M5e Microplate Reader and was corrected for background fluorescence by including control wells containing culture media alone (no cells) for each plate. The data were analyzed using SoftMax Pro software (Molecular Devices). Absolute cell number also was determined using trypan blue solution (Sigma). The proportion of CD34+ cells was measured flow cytometrically after staining with an anti-CD34 antibody (BD Biosciences). Data were acquired on a FACS Calibur flow cytometric analyzer (Becton Dickinson).
Colony formation assay
Bone marrow cells were collected from PV mice and then normalized to seed 1 × 104 GFP+ cells and then plated into each well of a 6-well plate and cultured in M3434 medium in the presence and absence of zileuton (100 μmol/L; n = 3 for each treatment group). The colonies were counted at day 7 after drug treatment. For CFU-GM and CFU-E/BFU-E assays, bone marrow cells collected from PV mice were analyzed by FACS for the percentage of GFP+ cells and then normalized to seed 1 × 104 GFP+ cells in each dish in Stem Cell Methocult with 3 dishes for each sample in the presence and absence of zileuton. Colonies derived from CFU-E were counted after 48 hours of culture, and colonies from BFU-E and CFU-GM were counted after 10 and 7 days, respectively.
In some cases, CD34+ cells were assayed for colony formation assay in semisolid media as previously described (17). Briefly, 500 CD34+ cells were plated in duplicate in tissue culture dishes (30 mm diameter) containing 1-mL IMDM with 1.1% methylcellulose and 20% FBS, to which stem cell factor (SCF), thrombopoietin (TPO), fms-like tyrosine kinase 3 ligand (Flt-3 ligand), IL3, granulocyte macrophage-colony stimulating factor (GM-CSF) at 50 ng/mL, and 2 U/mL EPO were added. Various doses of zileuton (50–500 μmol/L; Cayman) were added to the medium. Colonies were enumerated after 14 days of incubation, and individual colonies were plucked and genotyped for JAK2V617F as previously described (17).
Nested allele-specific PCR for JAK2V617F
Genomic DNA was isolated from randomly plucked colonies using the Extract-N-Amp Blood PCR Kits (Sigma). JAK2V617F was detected by using a nested allele-specific PCR. The final PCR products were analyzed on 2.0% agarose gels. The nested PCR product had a size of 453 bp. A 279-bp product indicated allele-specific JAK2V617F-positive, whereas a 229-bp product denoted allele-specific wild-type product. Colonies were classified as homozygous for JAK2V617F, if they contained only the 279-bp band, whereas heterozygous colonies were identified on the basis of the presence of both the 279-bp and 229-bp bands (17).
Flow cytometric assay of human normal and PV cells
CD34+ cells from patients with PV and normal CD34+ human cells were cultured in SFM containing SCF, TPO, Flt-3 ligand, and IL3 at 50 ng/mL, and cells were treated with various doses of zileuton (50–500 μmol/L). Cells were stained with a CD34 monoclonal antibody (BD Biosciences) initially and then washed again with binding buffer before staining with Annexin-V (BD Biosciences). Data were acquired on a FACS Calibur flow cytometric analyzer (Becton Dickinson).
Statistical analysis
Statistical analysis was performed by using the Student t test (GraphPad Prism v5.01 software for Windows, GraphPad Software).
Results
Alox5 expression is upregulated by JAK2V617F and Alox5 inhibition abrogates JAK2V617F-stimulated cell growth
To study the role of Alox5 in JAK2V617F-induced PV, we decided to use Ba/F3 and JAK2V617F-expressing Ba/F3 cells, as well as JAK2V617F-expressing human HEL cells. We characterized these cells for the activation of JAK/STAT signaling by JAK2V617F using a JAK2 inhibitor ruxolitinib. We found that compared with Ba/F3 cells, phosphorylation of JAK2 and STAT5 was greatly increased in JAK2V617F-expressing Ba/F3 and HEL cells and that inhibition of JAK2 kinase activity by ruxolitinib largely reduced the phosphorylation (Fig. 1A), indicating that JAK/STAT signaling is highly activated by JAK2V617F in these cells. In addition, expression of JAK2V617F also led to activation of β-catenin and AKT (Fig. 1B). Using these cells, we then tested whether Alox5 expression is regulated by JAK2V617F. We found that in JAK2V617F-expressing Ba/F3 cells, expression of 5-LO encoded by Alox5 was significantly increased as compared with Ba/F3 cells (Fig. 1C), suggesting that Alox5 is involved in JAK2V617F signaling. Next, we wondered whether upregulation of 5-LO expression by JAK2V617F is abolished upon inhibition of JAK2V617F signaling. We treated JAK2V617F-expressing Ba/F3 and HEL cells with ruxolitinib using Ba/F3 cells as control and found that inhibition of JAK2 phosphorylation by Ruxolitinib did not downregulate 5-LO expression (Fig. 1C). This result suggests that JAK2V617F regulates 5-LO expression independently of its kinase activity, providing a unique mechanism for the involvement of Alox5 in JAK2V617F signaling. To study the functional relevance of JAK2V617F/Alox5 signaling, we further tested whether inhibition of Alox5 function abrogates JAK2V617F-stimulated cell proliferation by treating JAK2V617F-expressing Ba/F3 and HEL cells with the Alox5 inhibitor zileuton and observed a dose-dependent growth inhibition of the cells (Fig. 1D). In contrast, zileuton had no significant inhibitory effect on parental Ba/F3 cells that do not express JAK2V617F (Fig. 1D), indicating that zileuton inhibits cell growth stimulated by JAK2V617F. Furthermore, we examined the effect of Alox5 inhibition on apoptosis and cell-cycle progression of Ba/F3, JAK2V617F-expressing Ba/F3 and HEL cells. We found that zileuton treatment induced apoptosis of JAK2V617F-expressing Ba/F3 and HEL cells with a minimal effect on Ba/F3 cells (Fig. 1E). In addition, zileuton treatment caused a blockade of cell-cycle progression of JAK2V617F-expressing Ba/F3 cells from G0 to G1 phase but did not affect the S + G2–M phase (Fig. 1F). No effect of zileuton on the S + G2–M phase was confirmed by staining the cells with propidium iodide (PI) and subsequent FACS analysis (Fig. 1G).
Alox5 expression is upregulated by JAK2V617F and Alox5 inhibition abrogates JAK2V617F-stimulated cell growth. A, Western blot analysis showed inhibition of JAK/STAT signaling by ruxolitinib in JAK2V617F-expressing Ba/F3 and HEL cells. Two consistent independent experiments were done, and the results from a representative experiment are shown. B, Western blot analysis showed that expression of JAK2V617F increased expression of β-catenin and activated AKT. P-AKT, phosphorylated AKT. Two consistent independent experiments were done, and the results from a representative experiment are shown. C, Western blot analysis showed that ruxolitinib inhibited JAK2/STAT5 signaling after 24-hour treatment but did not reduce 5-LO expression in JAK2V617F-expressing Ba/F3 and HEL cells. Two consistent independent experiments were done, and the results from a representative experiment are shown. D, Alox5 inhibition abrogates proliferation of JAK2V617F-expressing mouse Ba/F3 and human HEL cells. The cells were treated with DMSO or zileuton (100, 250, and 500 μmol/L) for 48 hours, and live cells were counted by trypan blue staining. **, P < 0.01. Three consistent independent experiments were done, and the results from a representative experiment are shown. Note that zileuton had no significant effect on Ba/F3 cells. P values are 0.7610 for 100 μmol/L zileuton, 0.2253 for 250 μmol/L, and 0.0518 for 500 μmol/L, respectively. E, Ba/F3, JAK2V617F-expressing Ba/F3 cells, and HEL cells were treated with DMSO or zileuton (100, 250, or 500 μmol/L) for 48 hours. The cells were stained with Annexin-V+ and 7-aminoactinomycin D (7-AAD)+ and apoptotic cells were analyzed by FACS. Three consistent independent experiments were done, and the results from a representative experiment are shown. F, JAK2V617F-expressing Ba/F3 cells were treated with DMSO or zileuton (250 μmol/L) for 48 hours, and the cells were stained with Pyronin/Hoechst. Cell-cycle progression was analyzed by FACS. Two consistent independent experiments were done, and the results from a representative experiment are shown. G, JAK2V617F-expressing Ba/F3 cells were treated with DMSO or zileuton (100 or 250 μmol/L) for 48 hours, and the cells were stained with PI. Cell-cycle progression was analyzed by FACS. Two consistent independent experiments were done, and the results from a representative experiment are shown.
Alox5 expression is upregulated by JAK2V617F and Alox5 inhibition abrogates JAK2V617F-stimulated cell growth. A, Western blot analysis showed inhibition of JAK/STAT signaling by ruxolitinib in JAK2V617F-expressing Ba/F3 and HEL cells. Two consistent independent experiments were done, and the results from a representative experiment are shown. B, Western blot analysis showed that expression of JAK2V617F increased expression of β-catenin and activated AKT. P-AKT, phosphorylated AKT. Two consistent independent experiments were done, and the results from a representative experiment are shown. C, Western blot analysis showed that ruxolitinib inhibited JAK2/STAT5 signaling after 24-hour treatment but did not reduce 5-LO expression in JAK2V617F-expressing Ba/F3 and HEL cells. Two consistent independent experiments were done, and the results from a representative experiment are shown. D, Alox5 inhibition abrogates proliferation of JAK2V617F-expressing mouse Ba/F3 and human HEL cells. The cells were treated with DMSO or zileuton (100, 250, and 500 μmol/L) for 48 hours, and live cells were counted by trypan blue staining. **, P < 0.01. Three consistent independent experiments were done, and the results from a representative experiment are shown. Note that zileuton had no significant effect on Ba/F3 cells. P values are 0.7610 for 100 μmol/L zileuton, 0.2253 for 250 μmol/L, and 0.0518 for 500 μmol/L, respectively. E, Ba/F3, JAK2V617F-expressing Ba/F3 cells, and HEL cells were treated with DMSO or zileuton (100, 250, or 500 μmol/L) for 48 hours. The cells were stained with Annexin-V+ and 7-aminoactinomycin D (7-AAD)+ and apoptotic cells were analyzed by FACS. Three consistent independent experiments were done, and the results from a representative experiment are shown. F, JAK2V617F-expressing Ba/F3 cells were treated with DMSO or zileuton (250 μmol/L) for 48 hours, and the cells were stained with Pyronin/Hoechst. Cell-cycle progression was analyzed by FACS. Two consistent independent experiments were done, and the results from a representative experiment are shown. G, JAK2V617F-expressing Ba/F3 cells were treated with DMSO or zileuton (100 or 250 μmol/L) for 48 hours, and the cells were stained with PI. Cell-cycle progression was analyzed by FACS. Two consistent independent experiments were done, and the results from a representative experiment are shown.
Alox5 is required for the development of PV induced by JAK2V617F in mice
Next, we examined whether Alox5 is required for induction of PV by JAK2V617F in the retrovirus transduction/transplantation mouse model (13). We transduced bone marrow cells from wild-type (WT) or Alox5 homozygous knockout (Alox5−/−) mice in C57BL/6 background with retrovirus expressing JAK2V617F, followed by transplantation into lethally irradiated recipient mice. We found that the white blood cell counts (WBC) in recipients of JAK2V617F-transduced Alox5−/− donor bone marrow cells was significantly lower than that in recipients of JAK2V617F-transduced WT donor bone marrow cells (Fig. 2A), indicating that Alox5 is required for JAK2V617F-induced myeloproliferative disorder. In addition, we found that the average percentage of JAK2V617F-expressing Gr-1+ WBC count (GFP+Gr-1+ WBCs) in recipients of JAK2V617F-transduced Alox5−/− donor bone marrow cells was significantly lower than that in recipients of JAK2V617F-transduced WT donor bone marrow cells (Fig. 2B; P < 0.01), indicating further that Alox5 is required for JAK2V617F-induced myeloproliferative disorder. We also analyzed WBCs that did not express JAK2V617F (GFP−) in mice and found that the percentage of GFP− Gr-1+ WBCs was not reduced in the recipients of JAK2V617F-transduced Alox5−/− donor bone marrow cells (Fig. 2B), suggesting that Alox5 deficiency does not significantly affect normal Gr-1+ WBCs. The effect of Alox5 deficiency on PV development was further supported by the lower number of JAK2V617F-expressing (GFP+) Gr-1+ WBCs compared with the WT group (Fig. 2C). When we compared total numbers of cells in these PV mice, we found that RBCs (Fig. 2D), hemoglobin (Fig. 2E), and hematocrit (Fig. 2F) were significantly lower in recipients of JAK2V617F-transduced Alox5−/− bone marrow cells than in recipients of the transduced WT bone marrow cells, further indicating that Alox5 is required for PV development. In bone marrow, the average percentage of JAK2V617F-expressing Gr-1+ WBCs (GFP+ Gr-1+ WBCs) in recipients of JAK2V617F-transduced Alox5−/− donor bone marrow cells was also significantly lower than that in recipients of JAK2V617F-transduced WT donor bone marrow cells (Fig. 2G; P < 0.01). In addition, the number of bone marrow erythroid cells (Ter119+GFP+) in mice receiving JAK2V617F-transduced Alox5−/− bone marrow cells was significantly lower than that in mice receiving JAK2V617F-transduced WT bone marrow cells (P < 0.05; Fig. 2H) and the same was true in the spleen (Fig. 2I), which was consistent with the smaller spleen size in the absence of Alox5 (Fig. 2J). Because JAK2V617F-transformed HSCs is shown to be a PV-initiating cell population (7), we further tested whether loss of Alox5 affects this cell population in PV mice. We found that the number of HSCs (Lin−Sca-1+c-Kit+) in mice receiving JAK2V617F-transduced Alox5−/− bone marrow cells was significantly lower than that in mice receiving JAK2V617F-transduced WT bone marrow cells (Fig. 2K).
Alox5 is required for the development of PV induced by JAK2V617F in mice. A, FACS analysis showed WBCs in peripheral blood of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice. Mean percentage for each group (*, P < 0.05; **, P < 0.01; n = 5 for each group) is shown. B, FACS analysis showed GFP+Gr-1+ cells in peripheral blood of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice. Mean percentage for each group (*, P < 0.05; n = 5 for each group) is shown. C, Total numbers of WBCs in peripheral blood of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice were counted at different time points after induction of PV. Mean value for each group (*, P < 0.05; **, P < 0.01; n = 5 for each group) is shown. RBC counts (D), hemoglobin (E), and hematocrit (F) in peripheral blood of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice were measured at different time points after induction of PV. Mean value for each group (*, P < 0.05; **, P < 0.01; n = 5 for each group) is shown. G and H, Two months after induction of PV, FACS analysis showed the percentage of WBCs (G) and number of Ter119+GFP+ cells (H) in bone marrow of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice. Mean value is shown for each group (*, P < 0.05, n = 5 for G; **, P < 0.01, n = 5 for H). I, Two months after induction of PV, FACS analysis showed the number of Ter119+GFP+ cells in the spleen of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice. Mean value is shown for each group (*, P < 0.05; n = 5). J, Two months after induction of PV, spleen weight was compared between recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice. Mean value is shown for each group (**, P < 0.01; n = 5). K, Three months after induction of PV, FACS analysis showed the numbers of GFP+Lin−Sca1+c-Kit+ cells in bone marrow of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice. Mean value is shown for each group (P < 0.05; n = 3).
Alox5 is required for the development of PV induced by JAK2V617F in mice. A, FACS analysis showed WBCs in peripheral blood of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice. Mean percentage for each group (*, P < 0.05; **, P < 0.01; n = 5 for each group) is shown. B, FACS analysis showed GFP+Gr-1+ cells in peripheral blood of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice. Mean percentage for each group (*, P < 0.05; n = 5 for each group) is shown. C, Total numbers of WBCs in peripheral blood of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice were counted at different time points after induction of PV. Mean value for each group (*, P < 0.05; **, P < 0.01; n = 5 for each group) is shown. RBC counts (D), hemoglobin (E), and hematocrit (F) in peripheral blood of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice were measured at different time points after induction of PV. Mean value for each group (*, P < 0.05; **, P < 0.01; n = 5 for each group) is shown. G and H, Two months after induction of PV, FACS analysis showed the percentage of WBCs (G) and number of Ter119+GFP+ cells (H) in bone marrow of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice. Mean value is shown for each group (*, P < 0.05, n = 5 for G; **, P < 0.01, n = 5 for H). I, Two months after induction of PV, FACS analysis showed the number of Ter119+GFP+ cells in the spleen of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice. Mean value is shown for each group (*, P < 0.05; n = 5). J, Two months after induction of PV, spleen weight was compared between recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice. Mean value is shown for each group (**, P < 0.01; n = 5). K, Three months after induction of PV, FACS analysis showed the numbers of GFP+Lin−Sca1+c-Kit+ cells in bone marrow of recipients of JAK2V617F-transduced bone marrow cells from WT or Alox5−/− donor mice. Mean value is shown for each group (P < 0.05; n = 3).
Inhibition of Alox5 function suppresses the development of PV induced by JAK2V617F in mice
Because deletion of Alox5 caused inhibition of the development of JAK2V617F-induced PV in mice (Fig. 2), we tested whether Alox5 is a potential target gene for PV by treating PV mice with zileuton (300 mg/kg, once a day) or a placebo. BALB/c mice were used because JAK2V617F induces PV more robustly in BALB/c mice than in C57BL/6 mice (13). Western blot analysis of protein lysates from the spleens of PV mice showed that 5-lipoxyginase (5-LO, a protein product of the Alox5 gene) is upregulated by JAK2V617F, and inhibition of Alox5 function by zileuton restored the 5-LO level back to the control level (Fig. 3A). We believe that the reduced 5-LO levels in zileuton-treated PV mice simply reflect the reduction or disappearance of JAK2V617F-expressing cells in the spleen of the treated mice, as zileuton is thought to inhibit the enzymatic activity of 5-LO with no known effect on 5-LO expression. In these mice, peripheral blood smears and tissue sections of bone marrow, spleen, and liver showed a reduced number of peripheral blood WBCs and less tissue infiltration in zileuton-treated PV mice as compared with placebo-treated PV mice (Fig. 3B). We also compared WBC counts and found that the number of WBCs in placebo-treated mice rose significantly with time, whereas the number of WBCs and percentage of JAK2V617F-expressing (GFP+) Gr-1+ WBCs in zileuton-treated mice did not (Fig. 3C and D). The inhibition of PV development by zileuton correlated with much smaller infiltrated spleen in zileuton-treated PV mice than in placebo-treated PV mice (Fig. 3E). To further demonstrate the effect of Alox5 inhibition by zileuton on PV development, we compared the levels of GFP+ cells in PV mice. FACS analysis showed that the percentage of Gr-1+ or Mac+ PV cells in peripheral blood was significantly lower in zileuton-treated PV mice than in placebo-treated PV mice (Fig. 3F), although there was no significant difference in the percentage of B220+ cells between the 2 treatment groups (Fig. 3F). Similar result was observed in bone marrow of these mice (Fig. 3G). Consistent with the inhibition of PV cells by zileuton in mice, the average LTB4 level in zileuton-treated PV mice was much lower than that in placebo-treated PV mice (Fig. 3H), and survival of zileuton-treated PV mice was significantly improved compared with placebo-treated PV mice (Fig. 3I). Beginning 6 months after induction of PV, about 40% placebo-treated PV mice gradually died, but all zileuton-treated mice survived (Fig. 3I). In addition, quantitative colony-forming assay showed that zileuton inhibits PV cell growth in vitro (Fig. 3J). Furthermore, the colony formation of granuloctye macrophage progenitor (CFU-GM) and erythroid cells (BFU-E/CFU-E) was also inhibited by zileuton (Fig. 3K). Together, these results indicate that inhibition of Alox5 function suppresses the growth of JAK2V617F-expressing cells, providing a novel therapeutic strategy for JAK2V617F-induced PV.
Inhibition of Alox5 function suppresses the development of PV induced by JAK2V617F in mice. A, Alox5 expression was upregulated by JAK2V617F in PV mice. Mice with JAK2V617F -induced PV were treated with placebo or zileuton (300 mg/kg/d) for 2 months beginning at 2 months after induction of PV. Protein lysates were isolated from the spleens of normal (control; n = 4) and PV mice (n = 4 for placebo-treated group; n = 5 for zileuton-treated group), and Western blot analysis showed that 5-LO was upregulated by JAK2V617F and this upregulation was reverted by zileuton treatment. B, Hematoxylin and eosin staining of peripheral blood smears, bone marrow, spleen, and liver sections from placebo- or zileuton-treated PV mice after 4-month treatment. C, WBC counts of placebo- or zileuton-treated PV mice were compared at different time points after induction of PV. *, P < 0.05. D, Percentages (left) and numbers (right) of GFP+Gr-1+ WBCs in peripheral blood of placebo- or zileuton-treated PV mice were calculated on the basis of WBC counts and FACS analysis of GFP+ in peripheral blood of the treated PV mice (WBC count × % of GFP+ cells) at 30 and 90 days (P < 0.01; n = 5 for each treatment group). Note that prior to treatments, GFP+ cells in peripheral blood were assessed by distributing the mice into two treatment groups. On average, the percentages of GFP+ cells were 17.5% for the placebo group and 19.2% for the zileuton group. E, Gross appearance and weight of the spleens of placebo- or zileuton-treated PV mice. FACS analysis of GFP+ cells in peripheral blood (F) and bone marrow (G) of placebo- or zileuton-treated PV mice. *, P < 0.05; **, P < 0.01. H, PV mice were treated with placebo or zileuton, beginning 1 month after PV induction, and the levels of LTB4 in plasma of the mice were measured at the days 0 and 60 posttreatments. I, Kaplan–Meier survival curves for placebo- (n = 8) and zileuton-treated (n = 9) PV mice. **, P < 0.01. J, Quantitative colony-forming assay showed that zileuton inhibits PV cell growth in vitro. Bone marrow cells were collected from PV mice, and 1 × 104 GFP+ cells were cultured in M3434 medium in the presence and absence of zileuton (100 μmol/L; n = 3 for each group). *, P < 0.05. The colonies were counted at day 7 after drug treatment. K, Quantitative colony-forming assay for CFU-GM or BFU-E/CFU-E. bone marrow cells were collected from PV mice, and 1 × 104 cells were cultured in M3434 medium in the presence and absence of zileuton (100 μmol/L; n = 3 for each group). *, P < 0.05. The CFU-GM colonies were counted at day 7 after drug treatment, BFU-E colonies counted at day 10, and CFU-E colonies counted at 48 hours.
Inhibition of Alox5 function suppresses the development of PV induced by JAK2V617F in mice. A, Alox5 expression was upregulated by JAK2V617F in PV mice. Mice with JAK2V617F -induced PV were treated with placebo or zileuton (300 mg/kg/d) for 2 months beginning at 2 months after induction of PV. Protein lysates were isolated from the spleens of normal (control; n = 4) and PV mice (n = 4 for placebo-treated group; n = 5 for zileuton-treated group), and Western blot analysis showed that 5-LO was upregulated by JAK2V617F and this upregulation was reverted by zileuton treatment. B, Hematoxylin and eosin staining of peripheral blood smears, bone marrow, spleen, and liver sections from placebo- or zileuton-treated PV mice after 4-month treatment. C, WBC counts of placebo- or zileuton-treated PV mice were compared at different time points after induction of PV. *, P < 0.05. D, Percentages (left) and numbers (right) of GFP+Gr-1+ WBCs in peripheral blood of placebo- or zileuton-treated PV mice were calculated on the basis of WBC counts and FACS analysis of GFP+ in peripheral blood of the treated PV mice (WBC count × % of GFP+ cells) at 30 and 90 days (P < 0.01; n = 5 for each treatment group). Note that prior to treatments, GFP+ cells in peripheral blood were assessed by distributing the mice into two treatment groups. On average, the percentages of GFP+ cells were 17.5% for the placebo group and 19.2% for the zileuton group. E, Gross appearance and weight of the spleens of placebo- or zileuton-treated PV mice. FACS analysis of GFP+ cells in peripheral blood (F) and bone marrow (G) of placebo- or zileuton-treated PV mice. *, P < 0.05; **, P < 0.01. H, PV mice were treated with placebo or zileuton, beginning 1 month after PV induction, and the levels of LTB4 in plasma of the mice were measured at the days 0 and 60 posttreatments. I, Kaplan–Meier survival curves for placebo- (n = 8) and zileuton-treated (n = 9) PV mice. **, P < 0.01. J, Quantitative colony-forming assay showed that zileuton inhibits PV cell growth in vitro. Bone marrow cells were collected from PV mice, and 1 × 104 GFP+ cells were cultured in M3434 medium in the presence and absence of zileuton (100 μmol/L; n = 3 for each group). *, P < 0.05. The colonies were counted at day 7 after drug treatment. K, Quantitative colony-forming assay for CFU-GM or BFU-E/CFU-E. bone marrow cells were collected from PV mice, and 1 × 104 cells were cultured in M3434 medium in the presence and absence of zileuton (100 μmol/L; n = 3 for each group). *, P < 0.05. The CFU-GM colonies were counted at day 7 after drug treatment, BFU-E colonies counted at day 10, and CFU-E colonies counted at 48 hours.
We further evaluated the effect of Alox5 inhibition by zileuton on PV development by examining RBCs and platelets. We found that zileuton treatment did not significantly affect the level of RBCs (Fig. 4A) but had a significant inhibitory effect on mean corpuscular volume (MCV; Fig. 4B), hematocrit (Fig. 4C), mean corpuscular hemoglobin (MCH; Fig. 4D), and hemoglobin (Fig. 4E) as compared with placebo-treated PV mice. Zileuton treatment did not have an inhibitory effect on the level of PLT in PV mice (Fig. 4F). Furthermore, we found that in both bone marrow and spleen, the number of JAK2V617F-expressed erythroid cells (Ter-119+GFP+) in zileuton-treated PV mice was significantly lower than that in placebo-treated PV mice (P < 0.01 and P < 0.001; Fig. 4G and H). The above-described inhibitory effect of zileuton was consistent with the smaller size of the spleen in zileuton-treated PV mice than in placebo-treated PV mice (Fig. 4I).
Characterization of zileuton-treated PV mice. Levels of RBC (A), MCV (B), hematocrit (C), MCH (D), hemoglobin (E), and platelets (F) in placebo- or zileuton-treated PV mice were measured at different time points after induction of PV. Mean value for each time point (*, P < 0.05; **, P < 0.01; n = 5 for each group) is shown. Note that prior to treatments, the levels of RBC, MCV, hematocrit, MCH, hemoglobin, and platelets were assessed by equally distributing the mice into two treatment groups. On average, RBC was 9.3 M/uL, MCV 71.9 fL, hematocrit 83.5%, MCH 18.7 pg, hemoglobin 20.0 g/dL, and platelets 611.9 K/uL. Number of JAK2V617F-expressed erythroid cells (Ter119+GFP+) in bone marrow (G) and spleen (H) of placebo- or zileuton-treated PV mice. **, P < 0.01; ***, P < 0.001; n = 4 for each group. I, Spleen weight comparison between placebo- or zileuton-treated PV mice. PV mice were treated with a placebo or zileuton for 30 days. **, P < 0.01; n = 5 for each group.
Characterization of zileuton-treated PV mice. Levels of RBC (A), MCV (B), hematocrit (C), MCH (D), hemoglobin (E), and platelets (F) in placebo- or zileuton-treated PV mice were measured at different time points after induction of PV. Mean value for each time point (*, P < 0.05; **, P < 0.01; n = 5 for each group) is shown. Note that prior to treatments, the levels of RBC, MCV, hematocrit, MCH, hemoglobin, and platelets were assessed by equally distributing the mice into two treatment groups. On average, RBC was 9.3 M/uL, MCV 71.9 fL, hematocrit 83.5%, MCH 18.7 pg, hemoglobin 20.0 g/dL, and platelets 611.9 K/uL. Number of JAK2V617F-expressed erythroid cells (Ter119+GFP+) in bone marrow (G) and spleen (H) of placebo- or zileuton-treated PV mice. **, P < 0.01; ***, P < 0.001; n = 4 for each group. I, Spleen weight comparison between placebo- or zileuton-treated PV mice. PV mice were treated with a placebo or zileuton for 30 days. **, P < 0.01; n = 5 for each group.
Because JAK2V617F affects plasma levels of Epo in patients with PV (12), we also investigated whether zileuton treatment alters the level of Epo regulated by JAK2V617F by using mice receiving empty vector–transduced bone marrow cells as a control. We found that the EPO level in placebo-treated PV mice was significantly lower than that in the control mice and that zileuton treatment largely restored the plasma EPO level after 30-day treatment (Fig. 5A). To investigate whether JAK2V617F regulates the EPO signaling pathway via Alox5, we treated JAK2V617F-expressing BaF/3 and HEL cells with zileuton using Ba/F3 cells as control because the EPO signaling is linked to the JAK2/STAT5 pathway and subsequently the PI3K/AKT pathway in JAK2V617F-expressing cells (2, 3). We found that at higher dose of zileuton with 6-hour treatment, JAK2/STAT5 signaling was inhibited (Fig. 5B). In JAK2V617F-expressing Ba/F3 cells, zileuton also inhibited activation of AKT and β-catenin by JAK2V617F (Fig. 5C) and did not affect activation of PI3K and MAPK (Fig. 5D).
Mechanistic study of pathways affected by zileuton in JAK2V617F-expressing cells. A, Levels of EPO in peripheral blood of placebo- or zileuton-treated PV mice at day 30 after the treatment. *, P < 0.05; n = 3 for each group. Mice receiving empty vector–transduced bone marrow cells were used as a control. Two consistent independent experiments were done, and the results from a representative experiment are shown. B, Ba/F3, JAK2V617F-expressing Ba/F3, and HEL cells were treated with zileuton for 3 or 6 hours. Protein lysates were analyzed by Western blotting for expression of the proteins indicated. Two consistent independent experiments were done, and the results from a representative experiment are shown. C and D, JAK2V617F-expressing Ba/F3 cells were treated with DMSO or zileuton (250 μmol/L) for 48 hours. Protein lysates were analyzed by Western blotting for expression of the proteins indicated. Two consistent independent experiments were done, and the results from a representative experiment are shown.
Mechanistic study of pathways affected by zileuton in JAK2V617F-expressing cells. A, Levels of EPO in peripheral blood of placebo- or zileuton-treated PV mice at day 30 after the treatment. *, P < 0.05; n = 3 for each group. Mice receiving empty vector–transduced bone marrow cells were used as a control. Two consistent independent experiments were done, and the results from a representative experiment are shown. B, Ba/F3, JAK2V617F-expressing Ba/F3, and HEL cells were treated with zileuton for 3 or 6 hours. Protein lysates were analyzed by Western blotting for expression of the proteins indicated. Two consistent independent experiments were done, and the results from a representative experiment are shown. C and D, JAK2V617F-expressing Ba/F3 cells were treated with DMSO or zileuton (250 μmol/L) for 48 hours. Protein lysates were analyzed by Western blotting for expression of the proteins indicated. Two consistent independent experiments were done, and the results from a representative experiment are shown.
Inhibition of Alox5 function reduces proliferation of human CD34+ cells from patients with PV
We next evaluated whether inhibition of Alox5 function by zileuton reduces proliferation of human CD34+ cells from patients with PV. We cultured peripheral blood cells from patients with PV under the stem cell culture conditions in the absence or presence of zileuton (100 and 250 μmol/L) for 48 hours. The percentages of CD34+ cells were analyzed by FACS, and total cell numbers were compared at the end of the culture. We found that zileuton treatment significantly reduced proliferation of CD34+ cells (Fig. 6A). Human CD34+ cells from patients with PV were also analyzed in a colony-forming assay. The colonies were enumerated after 14 days of incubation (Table 1), and individual colonies were plucked and genotyped for detecting the presence of JAK2V617F. We found that zileuton treatment significantly reduced colony number of CD34+ progenitor cells carrying 2 JAK2V617F mutant alleles (Fig. 6B) but had no effect on colony formation of normal CD34+ progenitor cells. Zileuton treatment also reduced colony formation of human CFU-GM and BFU-E (Fig. 6C).
Inhibition of Alox5 function reduces proliferation of human CD34+ cells from patients with PV. A, Peripheral blood mononuclear cells from patients with PV were cultured under the stem cell conditions in the absence or presence of zileuton (100 and 250 μmol/L) for 48 hours. A total of 1 × 106 human peripheral blood mononuclear cells were cultured in a 24-well plate with SFM containing SCF, TPO, Flt-3 ligand, and IL3 at 50 ng/mL, and cells were treated with vehicle (control) or various doses of zileuton. CD34+ cells were analyzed by FACS, and total number of CD34+ cells at the end of the culture was compared between treatment groups. B, CD34+ cells were assayed for colony formation in the presence of various concentrations of zileuton (50–250 μmol/L). Colonies were enumerated after 14 days of incubation, and individual colonies were plucked and genotyped for JAK2V617F. *, P < 0.05. C, Quantitative colony-forming assay for CFU-GM and CFU-E in the absence and presence of various concentrations of zileuton (50–500 μmol/L). Colonies were counted at day 14 after drug treatment. (**, P < 0.01).
Inhibition of Alox5 function reduces proliferation of human CD34+ cells from patients with PV. A, Peripheral blood mononuclear cells from patients with PV were cultured under the stem cell conditions in the absence or presence of zileuton (100 and 250 μmol/L) for 48 hours. A total of 1 × 106 human peripheral blood mononuclear cells were cultured in a 24-well plate with SFM containing SCF, TPO, Flt-3 ligand, and IL3 at 50 ng/mL, and cells were treated with vehicle (control) or various doses of zileuton. CD34+ cells were analyzed by FACS, and total number of CD34+ cells at the end of the culture was compared between treatment groups. B, CD34+ cells were assayed for colony formation in the presence of various concentrations of zileuton (50–250 μmol/L). Colonies were enumerated after 14 days of incubation, and individual colonies were plucked and genotyped for JAK2V617F. *, P < 0.05. C, Quantitative colony-forming assay for CFU-GM and CFU-E in the absence and presence of various concentrations of zileuton (50–500 μmol/L). Colonies were counted at day 14 after drug treatment. (**, P < 0.01).
Effect of zileuton on colony formation of CD34+ cells from patients with PV
. | . | . | JAK2V617F genotype . | . | . |
---|---|---|---|---|---|
. | . | Colony number . | |||
Cases . | Zileuton, μmol/L . | Homo . | Hete . | Wild . | Total . |
T131 | 0 | 4 | 1 | 19 | 24 |
50 | 4 | 0 | 20 | 24 | |
250 | 2 | 2 | 20 | 24 | |
T136 | 0 | 6 | 6 | 10 | 22 |
50 | 7 | 7 | 9 | 23 | |
250 | 2 | 6 | 10 | 18 | |
T139 | 0 | 19 | 3 | 1 | 23 |
50 | 20 | 1 | 2 | 23 | |
250 | 21 | 1 | 2 | 24 | |
T148 | 0 | 9 | 5 | 7 | 21 |
50 | 10 | 5 | 9 | 24 | |
250 | 5 | 7 | 7 | 19 |
. | . | . | JAK2V617F genotype . | . | . |
---|---|---|---|---|---|
. | . | Colony number . | |||
Cases . | Zileuton, μmol/L . | Homo . | Hete . | Wild . | Total . |
T131 | 0 | 4 | 1 | 19 | 24 |
50 | 4 | 0 | 20 | 24 | |
250 | 2 | 2 | 20 | 24 | |
T136 | 0 | 6 | 6 | 10 | 22 |
50 | 7 | 7 | 9 | 23 | |
250 | 2 | 6 | 10 | 18 | |
T139 | 0 | 19 | 3 | 1 | 23 |
50 | 20 | 1 | 2 | 23 | |
250 | 21 | 1 | 2 | 24 | |
T148 | 0 | 9 | 5 | 7 | 21 |
50 | 10 | 5 | 9 | 24 | |
250 | 5 | 7 | 7 | 19 |
Discussion
In this study, we find that loss of the Alox5 gene or pharmacologic inhibition of Alox5 function attenuates the development of PV induced by JAK2V617F, providing a strong rationale for targeting Alox5 in PV treatment. This idea is supported by our finding that inhibition of Alox5 function reduces proliferation of human CD34+ cells from patients with PV. Recently, Alox5 has been shown to play a role in functional regulation of disease-initiating cells for BCR-ABL- or TEL-PDGFR–induced myeloid leukemia and human glioma cells (15, 18, 19). Similar to CML induced by BCR-ABL, JAK2V617F was detected in HSCs of patients with PV (5), and JAK2V617F-expressing long-term HSCs were shown to be responsible for the initiation and maintenance of PV in mice (7). The effect of Alox5 loss on PV-initiating cells could complement the benefit from the use of a JAK2 inhibitor that does not have an inhibitory effect on PV-initiating cells (7).
It is interesting for us to observe that inhibition of JAK2 activity by ruxolitinib inhibits JAK2 phosphorylation but does not cause a reduced 5-LO expression, suggesting that JAK2V617F stimulates 5-LO expression independently of its kinase activity. This result supports a rationale for simultaneously inhibiting both JAK2V617F and 5-LO in treating PV. A number of JAK2 inhibitors (such as SAR302503 and Jakafi) have been developed and tested in clinic (20), and a long-term treatment benefit of these inhibitors still remains to be seen. Thus, an anti-Alox5 agent alone or in combination with a JAK2 inhibitor will provide a new therapeutic strategy for PV.
β-Catenin pathway plays a critical role in self-renewal and differentiation of both HSCs (21) and leukemic stem cells (LSC) in CML (22–24). We show that the molecular mechanism for the role of Alox5 in PV is linked to the β-catenin pathway. Our previous study shows that Alox5 regulates the level of β-catenin expression in LSCs of CML (15), and similarly, we find that JAK2V617F also induces Alox5 expression. These findings suggest that the Alox5 may represent a shared signaling pathway by Philadelphia chromosome–positive and -negative MPNs. Besides β-catenin, we also show that JAK2V617F activates the AKT pathway but does not activate PI3K and MAPK, which is different from BCR-ABL that activates these two pathways in CML cells (25). This signaling difference may reflect pathologic differences between PV and CML and provides clues for better understanding of these 2 types of MPNs. It is likely that Alox5 functions upstream of β-catenin and AKT because inhibition of Alox5 function by zileuton causes downregulation of β-catenin expression and reduced AKT phosphorylation in JAK2V617F-expressing cells. Our future study will focus on explaining how Alox5 signals to β-catenin and AKT in PV cells.
Alox5 is involved in numerous physiologic and pathologic processes, including oxidative stress response, inflammation, and cancer (26–30). Increasing evidence shows that the metabolites of the arachidonic acid cascade play a role in epithelial cell proliferation and tumorigenesis (31, 32). A known function of Alox5 is to produce inflammatory metabolites called leukotrienes. There are several types of leukotrienes (33), among which LTB4 is a major product of the Alox5 pathway. We show that the level of LTB4 is decreased in PV mice treated with zileuton. Although there is no available evidence indicating that LTB4 is a mediator of Alox5 signaling in activation of β-catenin and AKT, this possibility needs to be examined in the future. At least, it is reasonable to think that LTB4 could serve as a biologic indicator for PV pathogenesis.
In summary, we show that Alox5 plays critical role in the development of PV induced by JAK2V617F, which identified Alox5 as a potential target gene for PV. Future studies will test more robustly whether inhibition of Alox5 function is effective in suppressing human PV cells.
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Authors' Contributions
Conception and design: Y. Chen, A. Liang, S. Li
Development of methodology: Y. Chen, Y. Shan, N. DeSouza, R. Hoffman
Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Y. Chen, Y. Shan, N. DeSouza, R. Hoffman, S. Li
Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Y. Chen, Y. Shan, N. DeSouza, R. Hoffman, S. Li
Writing, review, and/or revision of the manuscript: Y. Chen, M. Lu, N. DeSouza, R. Hoffman, A. Liang, S. Li
Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Y. Chen, M. Lu, S. Li
Study supervision: Y. Chen, S. Li
Other (technical support for animal experiment): Z. Guo
Acknowledgments
We thank Dr. Ross Levine (Memorial Sloan-Kettering Cancer Center) for providing JAK2V617F-expressing Ba/F3 cell line.
Grant Support
This work was supported mostly by a grant from the MPN Research Foundation (S. Li). The work was also partially supported by grants from the Leukemia & Lymphoma Society and NIH grants R21-HL113603, R01-CA122142, and R01-CA176179 (S. Li).
The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.