MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia

Yana Pikman, Benjamin H Lee, Thomas Mercher, Elizabeth McDowell, Benjamin L Ebert, Maricel Gozo, Adam Cuker, Gerlinde Wernig, Sandra Moore, Ilene Galinsky, Daniel J DeAngelo, Jennifer J Clark, Stephanie J Lee, Todd R Golub, Martha Wadleigh, D Gary Gilliland, Ross L Levine, Yana Pikman, Benjamin H Lee, Thomas Mercher, Elizabeth McDowell, Benjamin L Ebert, Maricel Gozo, Adam Cuker, Gerlinde Wernig, Sandra Moore, Ilene Galinsky, Daniel J DeAngelo, Jennifer J Clark, Stephanie J Lee, Todd R Golub, Martha Wadleigh, D Gary Gilliland, Ross L Levine

Abstract

Background: The JAK2V617F allele has recently been identified in patients with polycythemia vera (PV), essential thrombocytosis (ET), and myelofibrosis with myeloid metaplasia (MF). Subsequent analysis has shown that constitutive activation of the JAK-STAT signal transduction pathway is an important pathogenetic event in these patients, and that enzymatic inhibition of JAK2V617F may be of therapeutic benefit in this context. However, a significant proportion of patients with ET or MF are JAK2V617F-negative. We hypothesized that activation of the JAK-STAT pathway might also occur as a consequence of activating mutations in certain hematopoietic-specific cytokine receptors, including the erythropoietin receptor (EPOR), the thrombopoietin receptor (MPL), or the granulocyte-colony stimulating factor receptor (GCSFR).

Methods and findings: DNA sequence analysis of the exons encoding the transmembrane and juxtamembrane domains of EPOR, MPL, and GCSFR, and comparison with germline DNA derived from buccal swabs, identified a somatic activating mutation in the transmembrane domain of MPL (W515L) in 9% (4/45) of JAKV617F-negative MF. Expression of MPLW515L in 32D, UT7, or Ba/F3 cells conferred cytokine-independent growth and thrombopoietin hypersensitivity, and resulted in constitutive phosphorylation of JAK2, STAT3, STAT5, AKT, and ERK. Furthermore, a small molecule JAK kinase inhibitor inhibited MPLW515L-mediated proliferation and JAK-STAT signaling in vitro. In a murine bone marrow transplant assay, expression of MPLW515L, but not wild-type MPL, resulted in a fully penetrant myeloproliferative disorder characterized by marked thrombocytosis (Plt count 1.9-4.0 x 10(12)/L), marked splenomegaly due to extramedullary hematopoiesis, and increased reticulin fibrosis.

Conclusions: Activation of JAK-STAT signaling via MPLW515L is an important pathogenetic event in patients with JAK2V617F-negative MF. The bone marrow transplant model of MPLW515L-mediated myeloproliferative disorders (MPD) exhibits certain features of human MF, including extramedullary hematopoiesis, splenomegaly, and megakaryocytic proliferation. Further analysis of positive and negative regulators of the JAK-STAT pathway is warranted in JAK2V617F-negative MPD.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. MPLW515L Mutation Is Found in…
Figure 1. MPLW515L Mutation Is Found in JAK2V617F-Negative MF and Causes Cytokine-Independent Growth in 32D and UT7 Cells, and Constitutively Activates the JAK-STAT Signaling Pathway
(A) Forward (middle trace) and reverse (lower trace) sequence traces demonstrating a heterozygous guanine to thymine substitution (arrows) present in granulocyte DNA from a patient with MF. The mutation is not present in buccal DNA from the same patient (upper trace). (B) DNA sequence and protein translation for both the wild-type and mutant MPL alleles. The mutation results in a tryptophan-to-leucine substitution at codon 515. (C)Upper: 32D cells transduced with MPLW515L exhibit cytokine-independent growth compared with MPLWT (left). Cell lines grown in the presence of IL3 show equal rates of growth (right). Error bars denote the standard deviation for each sample measured in triplicate.Lower: UT7 cells transformed with MPLW515L exhibit cytokine-independent growth compared with MPLWT (left). Cell lines grown in the presence of TPO (5 ng/mL) show equal rates of growth (right). Error bars denote the standard deviation for each sample measured in triplicate. (D) 32D cells, 32D MPLWT cells, and 32D MPLW515L cells were deprived of cytokines and then analyzed by Western blots, demonstrating phosphorylation of JAK2, STAT5, STAT3, AKT, and ERK in MPLW515L compared with MPLWT.
Figure 2. MPLW515L-Expressing Cells Show Hyper-Responsiveness to…
Figure 2. MPLW515L-Expressing Cells Show Hyper-Responsiveness to TPO
(A) 32D MPLWT cells and 32D MPLW515L cells were cultured in triplicate at an initial concentration of 1 × 10 5 cells/mL, in RPMI/10% FBS containing TPO 2 ng/mL, 1 ng/mL, 0.1 ng/mL, 0.01 ng/mL, 0.001 ng/mL, or in the absence of TPO for four days, and then cell numbers were assessed. Error bars denote the standard deviation for each sample measured in triplicate. (B) UT7 MPLWT cells and UT7 MPLW515L cells were cultured in triplicate, at an initial concentration of 1 × 10 5 cells/mL, in IMDM/10% FBS containing TPO 5 ng/mL, 1 ng/mL, 0.1 ng/mL, 0.01 ng/mL, 0.001ng/mL, or in the absence of TPO for six days, and then cell numbers were assessed. Error bars denote the standard deviation for each sample measured in triplicate. (C) 32D MPLWT or 32D MPLW515L cells were deprived of cytokine overnight and then stimulated with TPO 5 ng/mL, 1 ng/mL, 0.1 ng/mL, 0.01 ng/mL, or without TPO for seven minutes and then analyzed by Western blot for phosphorylation of JAK2 and STAT3, demonstrating increased JAK2/STAT3 phosphorylation in response to TPO in 32D MPLW515L cells as compared with 32D-MPLWT cells. (D) Phosphorylation of JAK2, TYK2, STAT3, STAT5, AKT, and ERK in response to seven-minute stimulation with TPO (50ng/mL). 32D MPLWT or 32 MPLW515L cells were deprived of cytokine overnight and then stimulated with TPO 50 ng/mL for seven minutes and then analyzed by Western blot for phosphorylation of JAK2, TYK2, STAT3, STAT5, AKT, and ERK, demonstrating increased phosphorylation of these proteins in response to TPO in 32D MPLW515L cells as compared with 32D MPLWT cells.
Figure 3. Bone Marrow Transplant with MPLW515L-Transduced…
Figure 3. Bone Marrow Transplant with MPLW515L-Transduced Bone Marrow Causes a Rapid Myeloproliferative Disease
(A) Kaplan-Meier survival plot of Balb/C mice transduced with MPLW515L (n = 9) and MPLWT (n = 6) showing death of all MPLW515L mice between 17 and 32 days post-BMT compared with MPLWT (n = 6), which were sacrificed for endpoint analysis without evidence of disease. (B) Complete blood counts show leukocytosis and thrombocytosis in MPLW515L BMT model compared with MPLWT. There is no difference in hematocrits. Standard deviation is indicated. (C) Spleen weights of MPLW515L and MPLWT mice shows splenomegaly in MPLW515L mice but not in MPLWT mice with average spleen weight equal to 1,171 mg. (D) Liver weights of MPLW515L and MPLWT mice shows hepatomegaly in MPLW515L mice but not in MPLWT mice, with average liver weight equal to 2,390 mg (compared with 1,222 mg in MPLWT mice). (E) Bone marrow shows significantly increased bone marrow fibrosis by reticulin staining at 17 days post-BMT in MPLW515L mice, but not in MPLWT-expressing mice.
Figure 4. MPLW515L and MPLWT Bone Marrow…
Figure 4. MPLW515L and MPLWT Bone Marrow Transplant Model Histopathology
Histology of MPLW515L-transduced and MPLWT-transduced Balb/C mice showing images of peripheral blood (A and B) and histopathology in representative sections of bone marrow (C and D), spleen (E–H), and liver (I–L). Peripheral blood smear (B) (600×, Wright-Giemsa) of a representative MPLWT animal displays an unremarkable white blood cell and platelet count. In contrast, peripheral blood smear (A) (600×, Wright-Giemsa) of a representative MPLW515L mutant animal reveals marked thrombocytosis and leukocytosis comprising a predominant population of maturing myeloid cells as well as frequent nucleated erythroid forms. Bone marrow images from MPLWT animals display preserved marrow architecture with maturing trilineage hematopoiesis (D) (600×, hematoxylin and eosin [H&E]). Comparatively, bone marrow sections from MPLW515L mutant animals demonstrate marrow elements comprising a prominent population of maturing myeloid cells with increased numbers of megakaryocytes including atypical and dysplastic forms occurring in frequent clusters (C) (600×, H&E) and showing emperipolesis of neutrophils in megakaryocyte cytoplasm. Spleen sections from MPLW515L mice display complete effacement of normal splenic architecture (E) (40×, H&E) with a marked expansion of red pulp that is composed of an admixture of maturing myeloid and erythroid elements and numerous numbers of atypical megakaryocytes (G) (600×, H&E) compared with MPLWT spleens (F and H) (40× and 600×, H&E), which display a relative preservation of normal spleen architecture and the presence of only maturing erythroid forms in the red pulp. Liver images from MPLW515L mice illustrate evidence of extensive extramedullary hematopoiesis in a perivascular and sinusoidal distribution (I) (100×, H&E) composed predominantly of a population of maturing erythroid elements with frequent large atypical megakaryocytes and occasional admixed myeloid forms (K) (600×, H&E). In comparison, only small, focal areas of nucleated erythroid cells were observed in livers from MPLWT animals (J and L) (100× and 600×, H&E).
Figure 5. Flow Cytometry Analysis of BM…
Figure 5. Flow Cytometry Analysis of BM and Spleen in Mice Transduced with MPLW515L and MPLWT
(A) Flow-cytometry analysis of bone marrow cells shows a 4-fold increase in Mac1+/Gr1+ cells and a shift to a more immature erythroid population. There is a 15-fold increase in CD41+ cells in bone marrow expressing MPLW515L compared with MPLWT. (B) Flow-cytometry analysis of spleen cells shows a 10-fold increase in Mac1+/Gr1+ cells in MPLW515L. There is also a shift to a more immature erythroid population in MPLW515L with a greater percentage of CD71+/Ter119- cells. CD41+ cells are increased 30-fold in MPLW515L spleen cells compared with MPLWT spleen cells.
Figure 6. MPLW515L Increases the Number of…
Figure 6. MPLW515L Increases the Number of Megakaryocyte and Myeloid Colonies in Spleen, without Affecting Megakaryocyte Ploidy, and Causes Cytokine-Independent Myeloid Colony Growth
(A) Megakaryocyte colony–forming assay in the presence of TPO, IL3, IL11, and IL6 demonstrates similar numbers of megakaryocyte colonies obtained from MPLW515L-expressing bone marrow and an increase in the number of megakaryocyte colonies from spleen cells compared with MPLWT. (B) Acetylcholinesterase staining of megakaryocyte colonies derived from bone marrow demonstrates much larger colony size in MPLW515L as compared with MPLWT megakaryocyte colonies. (C) Megakaryocyte ploidy analysis shows the same distribution for MPLW515L-expressing cells and MPLWT-expressing cells. (D and E) Total myeloid colony formation from bone marrow cells (D) and spleen cells (E) demonstrates cytokine-independent colony formation in MPLW515L bone marrow and spleen. There is no difference in colony distribution between MPLWT- and MPLW515L-expressing cells. Colony counts reflect only positively identifiable colonies, with thorough megakaryocyte colony analysis done in MegaCult assay ( Figure 6A), and thus are excluded from methylcellulose colony analysis. Colony numbers represent a total of three representative mice per group, in duplicate.
Figure 7. 32D MPLW515L Cells Are Sensitive…
Figure 7. 32D MPLW515L Cells Are Sensitive to JAK Inhibitor I
(A) Dose-dependent inhibition of growth of 32D MPLW515L and 32D MPLWT cells but not 32D FIP1L1-PDGFRA cells with increasing doses of JAK Inhibitor I. (B) Reduction in STAT3 phosphorylation in 32D MPLW515L cells but not 32D FIP1L1-PDGFRA cells with increasing doses of JAK Inhibitor I. Cells were incubated with varying drug concentrations for four hours and then collected for Western blot analysis.

References

    1. Dameshek W. Some speculations on the myeloproliferative syndromes. Blood. 1951;6:372–375.
    1. Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7:387–397.
    1. Jones AV, Kreil S, Zoi K, Waghorn K, Curtis C, et al. Widespread occurrence of the JAK2 V617F mutation in chronic myeloproliferative disorders. Blood. 2005;106:2162–2168.
    1. James C, Ugo V, Le Couedic JP, StaERK J, Delhommeau F, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434:1144–1148.
    1. Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365:1054–1061.
    1. Kralovics R, Passamonti F, Buser AS, Teo S, Tiedt R, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352:1779–1790.
    1. Levine RL, Belisle C, Wadleigh M, Zahrieh D, Lee S, et al. X-inactivation based clonality analysis and quantitative JAK2V617F assessment reveals a strong association between clonality and JAK2V617F in PV but not ET/MMM, and identifies a subset of JAK2V617F negative ET and MMM patients with clonal hematopoiesis. Blood. 2006;107:4139–41.
    1. Campbell PJ, Griesshammer M, Dohner K, Dohner H, Kusec R, et al. V617F mutation in JAK2 is associated with poorer survival in idiopathic myelofibrosis. Blood. 2005;107:2098–2100.
    1. Zhao R, Xing S, Li Z, Fu X, Li Q, et al. Identification of an Acquired JAK2 Mutation in Polycythemia Vera. J Biol Chem. 2005;280:22788–22792.
    1. Lu X, Levine R, Tong W, Wernig G, Pikman Y, et al. Expression of a homodimeric type I cytokine receptor is required for JAK2V617F-mediated transformation. Proc Natl Acad Sci U S A. 2005;102:18962–18967.
    1. Wernig G, Mercher T, Okabe R, Levine RL, Lee BH, et al. Expression of JAK2V617F causes a polycythemia vera-like disease with associated myelofibrosis in a murine bone marrow transplant model. Blood. 2006;107:4274–4281.
    1. Scott LM, Campbell PJ, Baxter EJ, Todd T, Stephens P, et al. The V617F JAK2 mutation is uncommon in cancers and in myeloid malignancies other than the classic myeloproliferative disorders. Blood. 2005;106:2920–2921.
    1. Taksin AL, Couedic JP, Dusanter-Fourt I, Masse A, Giraudier S, et al. Autonomous megakaryocyte growth in essential thrombocythemia and idiopathic myelofibrosis is not related to a c-mpl mutation or to an autocrine stimulation by Mpl-L. Blood. 1999;93:125–139.
    1. Le Couedic JP, Mitjavila MT, Villeval JL, Feger F, Gobert S, et al. Missense mutation of the erythropoietin receptor is a rare event in human erythroid malignancies. Blood. 1996;87:1502–1511.
    1. Hess G, Rose P, Gamm H, Papadileris S, Huber C, et al. Molecular analysis of erythropoietin receptor system in patients with polycythaemia vera. Br J Haematol. 1994;88:794–802.
    1. Cools J, DeAngelo DJ, Gotlib J, Stover EH, Legare RD, et al. A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome. N Engl J Med. 2003;348:1201–1214.
    1. Kelly LM, Liu Q, Kutok JL, Williams IR, Boulton CL, et al. FLT3 internal tandem duplication mutations associated with human acute myeloid leukemias induce myeloproliferative disease in a murine bone marrow transplant model. Blood. 2002;99:310–318.
    1. Stover EH, Chen J, Lee BH, Cools J, McDowell E, et al. The small molecule tyrosine kinase inhibitor AMN107 inhibits TEL-PDGFRbeta and FIP1L1-PDGFRalpha in vitro and in vivo. Blood. 2005;106:3206–3213.
    1. Growney JD, Shigematsu H, Li Z, Lee BH, Adelsperger J, et al. Loss of Runx1 perturbs adult hematopoiesis and is associated with a myeloproliferative phenotype. Blood. 2005;106:494–504.
    1. The International HapMap Project. Nature. 2003;426:789–796.
    1. Thompson JE, Cubbon RM, Cummings RT, Wicker LS, Frankshun R, et al. Photochemical preparation of a pyridone containing tetracycle: A Jak protein kinase inhibitor. Bioorg Med Chem Lett. 2002;12:1219–1223.
    1. Ding J, Komatsu H, Wakita A, Kato-Uranishi M, Ito M, et al. Familial essential thrombocythemia associated with a dominant-positive activating mutation of the c-MPL gene, which encodes for the receptor for thrombopoietin. Blood. 2004;103:4198–4200.
    1. Wiestner A, Schlemper RJ, van der Maas AP, Skoda RC. An activating splice donor mutation in the thrombopoietin gene causes hereditary thrombocythaemia. Nat Genet. 1998;18:49–52.
    1. Abe M, Suzuki K, Inagaki O, Sassa S, Shikama H. A novel MPL point mutation resulting in thrombopoietin-independent activation. Leukemia. 2002;16:1500–1506.
    1. Staerk J, Lacout C, Sato T, Smith SO, Vainchenker W, et al. An amphipathic motif at the transmembrane-cytoplasmic junction prevents autonomous activation of the thrombopoietin receptor. Blood. 2005;107:1864–1871.
    1. Tefferi A. Pathogenesis of myelofibrosis with myeloid metaplasia. J Clin Oncol. 2005;23:8520–8530.
    1. Tefferi A. Myelofibrosis with myeloid metaplasia. N Engl J Med. 2000;342:1255–1265.
    1. Schwaller J, Parganas E, Wang D, Cain D, Aster JC, et al. STAT5 is essential for the myelo- and lymphoproliferative disease induced by TEL/JAK2. Mol Cell. 2000;6:693–704.
    1. Cools J, Stover EH, Boulton CL, Gotlib J, Legare RD, et al. PKC412 overcomes resistance to imatinib in a murine model of FIP1L1-PDGFRalpha-induced myeloproliferative disease. Cancer Cell. 2003;3:459–469.
    1. Daley GQ, Van Etten RA, Baltimore D. Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome. Science. 1990;247:824–830.
    1. Kakumitsu H, Kamezaki K, Shimoda K, Karube K, Haro T, et al. Transgenic mice overexpressing murine thrombopoietin develop myelofibrosis and osteosclerosis. Leuk Res. 2005;29:761–769.
    1. Villeval JL, Cohen-Solal K, Tulliez M, Giraudier S, Guichard J, et al. High thrombopoietin production by hematopoietic cells induces a fatal myeloproliferative syndrome in mice. Blood. 1997;90:4369–4383.
    1. Yan XQ, Lacey D, Fletcher F, Hartley C, McElroy P, et al. Chronic exposure to retroviral vector encoded MGDF (mpl-ligand) induces lineage-specific growth and differentiation of megakaryocytes in mice. Blood. 1995;86:4025–4033.
    1. Vannucchi AM, Bianchi L, Cellai C, Paoletti F, Rana RA, et al. Development of myelofibrosis in mice genetically impaired for GATA-1 expression ( GATA-1(low) mice) . Blood. 2002;100:1123–1132.
    1. Marchetti M, Barosi G, Balestri F, Viarengo G, Gentili S, et al. Low-dose thalidomide ameliorates cytopenias and splenomegaly in myelofibrosis with myeloid metaplasia: A phase II trial. J Clin Oncol. 2004;22:424–431.
    1. Deeg HJ, Gooley TA, Flowers ME, Sale GE, Slattery JT, et al. Allogeneic hematopoietic stem cell transplantation for myelofibrosis. Blood. 2003;102:3912–3918.

Source: PubMed

3
Abonnere