In this issue of Blood, Gautier and colleagues describe a novel signaling pathway in which deregulated JAK2 activity augments expression of a key regulator of the cell cycle, the CDC25A phosphatase, via a translational mechanism.1 

Six years ago, the identification of an activating point mutation in the JAK2 kinase, JAK2V617F, in a large majority of patients with myeloproliferative neoplasms (MPNs) provided a significant advance in our understanding of the molecular pathogenesis of these disorders. As is often the case, however, this seminal discovery led to more questions, many of which remain unanswered. One key controversy is whether, and if so, how, the JAK2V617F allele provides a proliferative advantage to the cell that incurs the mutation. While it was reported earlier that the JAK2V617F mutation does not provide a proliferative advantage during in vitro expansion,2  a recent study demonstrates a proliferative advantage at the single cell level, especially for those cells that have undergone conversion to the homozygous mutant state.3  In addition, mature compartments display higher JAK2V617F allele burdens than CD34-positive stem cells, supporting the hypothesis that the mutant kinase provides a proliferative advantage during maturation.3-5 

Here, Gautier and colleagues provide a significant contribution to our understanding of altered JAK2V617F function and its effect on the cell cycle in MPN cells. They report that the CDC25A phosphatase, whose activity stimulates completion of the G1 phase and activation of DNA synthesis, is overexpressed in MPN patients' cells and that this increase in CDC25A levels is dependent on augmented JAK2 signaling. Interestingly, it is not the “classic” JAK2 signaling via activation of STAT transcription factors and increased transcription of target genes that raises CDC25A levels in MPN cells. Rather, the authors show that JAK2V617F increases CDC25A translation. This is effected by a novel signaling pathway in which JAK2V617F decreases phosphorylation of elongation factor eIF2α, thereby retaining or prolonging its activity, resulting in increased CDC25A translation. Importantly, pharmacologic inhibition of JAK2V617F-mediated eIF2α dephosphorylation decreased proliferation of primary MPN cells. Furthermore, growth of EPO-independent erythroid colonies (so-called endogenous erythroid colonies [EECs]), a pathognomonic hallmark of polycythemia vera,6  was selectively and significantly inhibited by addition of a pan-CDC25 inhibitor. EPO-dependent growth of JAK2 wt and healthy control cells was not affected, although these data await verification on a larger number of samples. These data demonstrate that mutant JAK2V617F actively influences cell-cycle regulation, thereby promoting cell proliferation. Notably, the study establishes CDC25A as a novel JAK2V617F target and suggests that its inhibition may impede growth of the MPN clone.

After the initial discovery of the JAK2V617F mutation, hopes were high that a targeted therapy for MPN patients, similar in efficacy to the tyrosine kinase inhibitors that have changed the paradigm of CML treatment, was within reach. Six years later, the first JAK2 inhibitor, Ruxolitinib, has gained FDA approval but, while very effective at ameliorating disease symptoms, it does not appear to alter the natural history of these diseases. In particular, its effect on JAK2V617F allele burden, widely assumed to reflect the size of the neoplastic clone, is limited.7  Therefore, novel strategies, most likely involving novel targets, are required to foster the development of innovative therapies for MPN patients. Because, as demonstrated by the use of a pan-CDC25 inhibitor in this study, the phosphatase CDC25A is amenable to pharmacologic inhibition, and healthy hematopoiesis appears largely unaffected by this intervention, the paper by Gautier et al provides a promising lead toward the exploration of alternative targets for rational drug design in MPN treatment.

Conflict-of-interest disclosure: The author declares no competing financial interests. ■

1
Gautier
 
E-F
Picard
 
M
Laurent
 
C
, et al. 
The cell cycle regulator CDC25A is a target for the JAK2V617F oncogene.
Blood
2012
, vol. 
119
 
5
(pg. 
1190
-
1199
)
2
Gaikwad
 
A
Nussenzveig
 
R
Liu
 
E
Gottshalk
 
S
Chang
 
K
Prchal
 
JT
In vitro expansion of erythroid progenitors from polycythemia vera patients leads to decrease in JAK2 V617F allele.
Exp Hematol
2007
, vol. 
35
 
4
(pg. 
587
-
595
)
3
Anand
 
S
Stedham
 
F
Beer
 
P
, et al. 
Effects of the JAK2 mutation on the hematopoietic stem and progenitor compartment in human myeloproliferative neoplasms.
Blood
2011
, vol. 
118
 
1
(pg. 
177
-
181
)
4
Dupont
 
S
Masse
 
A
James
 
C
, et al. 
The JAK2 V617F mutation triggers erythropoietin hypersensitivity and terminal erythroid amplification in primary cells from patients with polycythemia vera.
Blood
2007
, vol. 
110
 
3
(pg. 
1013
-
1021
)
5
Moliterno
 
AR
Williams
 
DM
Rogers
 
O
Isaacs
 
MA
Spivak
 
JL
Phenotypic variability within the JAK2 V617F-positive MPD: roles of progenitor cell and neutrophil allele burdens.
Exp Hematol
2008
, vol. 
36
 
11
(pg. 
1480
-
1486
)
6
Prchal
 
JF
Adamson
 
JW
Murphy
 
S
Steinmann
 
L
Fialkow
 
PJ
Polycythemia vera. The in vitro response of normal and abnormal stem cell lines to erythropoietin.
J Clin Invest
1978
, vol. 
61
 
4
(pg. 
1044
-
1047
)
7
Verstovsek
 
S
Kantarjian
 
H
Mesa
 
RA
, et al. 
Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis.
N Engl J Med
2010
, vol. 
363
 
12
(pg. 
1117
-
1127
)
Sign in via your Institution