27.12.2012 Views

12th Congress of the European Hematology ... - Haematologica

12th Congress of the European Hematology ... - Haematologica

12th Congress of the European Hematology ... - Haematologica

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

0414<br />

COMPARATIVE ANALYSIS OF THE CONSTITUTIVE ACTIVE JAK2V617F, JAK2T875N AND<br />

MPLW515L ALLELES IN A MURINE BONE MARROW TRANSPLANT MODEL OF<br />

MYELOPROLIFERATIVE DISEASE<br />

G. Wernig, T. Mercher, Y. Pikman, R.L. Levine, D.G. Gilliland<br />

HMS, Brigham & Women's Hospital, BOSTON, USA<br />

Present in <strong>the</strong> 95% <strong>of</strong> polycy<strong>the</strong>mia vera (PV) patients and in 50-60%<br />

<strong>of</strong> patients with essential thrombocy<strong>the</strong>mia (ET) and idiopathic myel<strong>of</strong>ibrosis<br />

(IMF), <strong>the</strong> JAK2V617F mutation, resulting in constitutive activation<br />

<strong>of</strong> <strong>the</strong> JAK/STAT pathway, is <strong>the</strong> most frequent genetic event in<br />

myeloproliferative disease (MPD). Little is know about additional mutations<br />

which contribute to JAK2V617F negative MPD. Very recently, two<br />

additional mutations were identified in JAK2V617F – patients:<br />

JAK2T857N, detected in <strong>the</strong> human AMKL cell line CHRF-288-11 and<br />

<strong>the</strong> somatic mutation in <strong>the</strong> thrombopoietin receptor, MPLW515L,<br />

detected in ~10% <strong>of</strong> patients with JAK2V617F – MF. JAK2V617F,<br />

JAK2T875N and MPLW515L were analysed in murine models <strong>of</strong> bone<br />

marrow transplantation and sufficiency for <strong>the</strong> development <strong>of</strong> MPD has<br />

been shown for each <strong>of</strong> <strong>the</strong>m. Comparison <strong>of</strong> <strong>the</strong>se three models provides<br />

insight into <strong>the</strong> JAK2-positive and JAK2 – MPDs that may inform<br />

<strong>the</strong>rapeutic strategies. Fur<strong>the</strong>rmore, developing murine models <strong>of</strong> disease<br />

for each <strong>of</strong> <strong>the</strong>se alleles may provide novel insights into phenotypic<br />

variance among <strong>the</strong> MPDs. The JAK2V617F mouse model is characterized<br />

by a striking, longterm erythrocytosis, a strain dependent degree<br />

<strong>of</strong> leukocytosis and <strong>the</strong> development <strong>of</strong> bone marrow and splenic reticulin<br />

fibrosis. Despite <strong>the</strong> presence <strong>of</strong> megakaryocytic hyperplasia, <strong>the</strong>re<br />

is impaired megakaryocytic polyploidization, and platelet counts are not<br />

augmented. JAK2T875N constitutively activates downstream effectors<br />

including STAT5 in hematopoietic cells in vitro. In a murine bone marrow<br />

transplantation model JAK2T875N, although <strong>the</strong> T875N substitution<br />

is localized in a different domain <strong>of</strong> <strong>the</strong> JAK2 kinase, results in a phenocopy<br />

<strong>of</strong> JAK2V617F disease, causing a MPD with features <strong>of</strong> AMKL<br />

including megakaryocytic hyperplasia in spleen, a polyploidization<br />

defect and increased reticulin fibrosis in bone marrow and spleen, with<br />

normal platelet and leukocyte counts. Similarly, <strong>the</strong> MPLW515L mutation<br />

also transformed hematopoietic cell lines to cytokine independent<br />

growth through activation <strong>of</strong> <strong>the</strong> JAK/STAT pathway. in vivo expression<br />

<strong>of</strong> MPLW515L, caused a rapidly fatal, fully penetrant MPD that was<br />

characterized by a marked leukocytosis, splenomegaly, bone marrow<br />

reticulin fibrosis with a strain specific disease latency. However, - in contrast<br />

to both JAK2 mutations - <strong>the</strong>re was megakaryocyte hyperplasia<br />

with normal megakaryoctye ploidy and marked thrombocytosis in <strong>the</strong><br />

3-4 million/uL range that was accompanied by thrombotic complications.<br />

Our data demonstrate important phenotypic differences between<br />

JAK2V617F, JAK2T875N and MPLW515L induced MPDs. Most striking<br />

was <strong>the</strong> effect on <strong>the</strong> megakaryocyte lineage. JAK2V617F and<br />

JAK2T875N induced megakaryocytic hyperplasia with a block in<br />

megakaryocyte maturation and lack <strong>of</strong> thrombocytosis, whereas<br />

MPLW515L enhanced megakarypoiesis resulting in severe thrombocytosis.<br />

Fur<strong>the</strong>rmore, marked leukocytosis developed in MPLW515L but<br />

not in JAK2V617F and JAK2T875N expressing C57Bl/6 mice. These findings<br />

indicate that <strong>the</strong> three mutations have different impacts on proliferation<br />

and/or survival <strong>of</strong> various hematopoietic progenitors. Since all<br />

three mutations signal through <strong>the</strong> JAK/STAT pathway, <strong>the</strong>se data suggest<br />

that <strong>the</strong> MPLW515L allele activates transcriptional programs not<br />

activated by JAK2 mutations that confer <strong>the</strong> ability for megakaryocytic<br />

development and thrombocytosis.<br />

12 th <strong>Congress</strong> <strong>of</strong> <strong>the</strong> <strong>European</strong> <strong>Hematology</strong> Association<br />

0415<br />

EXPRESSION AND EFFECTS OF NUCLEAR FACTOR-ERYTHROID DERIVED 2 (NF-E2) IN<br />

MURINE AND HUMAN HEMATOPOIESIS<br />

B. Will, 1 M. Mutschler, 2 C. Scholl, 3 U. Steidl, 4 E. Levantini, 4<br />

C.S. Huettner, 5 D.G. Tenen, 4 H.L. Pahl6 1 Harvard Medical School, BOSTON, USA; 2 University <strong>of</strong> Freiburg,<br />

FREIBURG, Germany; 3 Brigham and Women's Hospital, BOSTON, USA;<br />

4 Harvard Institutes <strong>of</strong> Medicine, BOSTON, USA; 5 Medical College <strong>of</strong> Wisconsin,<br />

MILWAUKEE, USA; 6 University Hospital <strong>of</strong> Freiburg, FREIBURG, Germany<br />

The basic region-leucine zipper transcription factor NF-E2 plays an<br />

important role in terminal erythroid and megakaryocytic maturation<br />

and regulates <strong>the</strong> differentiation <strong>of</strong> erythroblasts and megakaryocytes.<br />

NF-E2 knock out mice die at birth due to lack <strong>of</strong> platelets, but only display<br />

mild defects in <strong>the</strong> erythroid lineage. NF-E2 expression has been<br />

reported in murine cKit+/Sca-1+/Lin- (KSL) cells. However, its specific<br />

role in stem cells and early progenitors remains unclear. NF-E2 is overexpressed<br />

in patients with Polycy<strong>the</strong>mia vera (PV). Several studies have<br />

demonstrated that <strong>the</strong> degree <strong>of</strong> NF-E2 overexpression in individual PV<br />

patients correlates with <strong>the</strong> proportion <strong>of</strong> Jak2V617F'positive granulocytes<br />

and <strong>the</strong> ability to form endogenous erythropoietin-independent<br />

colonies in vitro. Interestingly, <strong>the</strong> Jak2V617F activating mutation is present<br />

in uncommitted progenitor cells (Lin-/CD34 + /CD38 – /CD90 + ) and<br />

results in enhanced erythroid differentiation capacity. However, <strong>the</strong> role<br />

<strong>of</strong> NF-E2 in stem and progenitor cells and its potential cooperation with<br />

Jak2V617F in PV is unclear. In an approach to clarify <strong>the</strong> function <strong>of</strong> NF-<br />

E2 in normal hematopoiesis and PV we investigated nf-e2 mRNA expression<br />

in human and murine hematopoietic cells at different developmental<br />

stages as well as <strong>the</strong> effects <strong>of</strong> NF-E2 overexpression in murine models.<br />

Nf-e2 expression was evaluated in FACS sorted human and murine<br />

stem and progenitor populations by qRT-PCR. A lentiviral construct (NF-<br />

E2-IRES-GFP) was used to transduce KSL and cKit+/Sca-1-/Lin- cells,<br />

which were subsequently assayed for colony formation in methylcellulose.<br />

A tetracycline-inducible mouse model (TET-OFF) was generated by<br />

introducing an nf-e2 transgene under control <strong>of</strong> <strong>the</strong> tetracycline responsive<br />

element and subsequent crossing to a CD34-promoter-tTA transgenic<br />

strain. Nf-e2 displayed a similar differentiation-specific expression<br />

pattern in murine and human hematopoiesis: nf-e2 mRNA was detected<br />

in LT-HSCs and ST-HSCs. During lineage commitment nf-e2 was<br />

downregulated in both CMPs and GMPs but was again upregulated in<br />

MEPs. In murine cells, highest nf-e2 expression levels were found in<br />

proerythroblasts followed by a consecutive decrease during maturation<br />

to orthochromatic erythroblasts. Enforced expression <strong>of</strong> NF-E2 following<br />

lentiviral transduction <strong>of</strong> murine KSL and cKit+/Sca-1-/Lin- progenitor<br />

cells caused markedly decreased BFU-E and CFU-E colony formation<br />

in <strong>the</strong> presence <strong>of</strong> erythropoietin. In <strong>the</strong> tetracycline-inducible mouse<br />

model NF-E2 overexpression in KSL and cKit+/Sca-1-/Lin- cells similarly<br />

led to impaired erythroid colony formation in vitro. Lentiviral NF-E2<br />

overexpression did not appear to affect maturation <strong>of</strong> committed erythroid<br />

progenitor cells in in vitro differentiation assays <strong>of</strong> murine Ter119spleen<br />

and fetal liver (E14.5) cells. In <strong>the</strong>se assays, Jak2V617F likewise<br />

showed no effect. Our study shows that NF-E2 expression is regulated<br />

in a biphasic manner in both murine and human erythropoiesis. Forces<br />

expression <strong>of</strong> NF-E2 in various human and murine stem- and progenitor<br />

cells decreases erythroid colony cloning efficiency.<br />

haematologica/<strong>the</strong> hematology journal | 2007; 92(s1) | 153

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!