Abstract
JAK2 (Janus kinase 2) initiates the intracellular signalling cascade downstream of cell surface receptor activation by cognate haematopoietic cytokines, including erythropoietin and thrombopoietin. The pseudokinase (JH2) domain of JAK2 negatively regulates the catalytic activity of the adjacent tyrosine kinase (JH1) domain and mutations within the pseudokinase domain underlie human myeloproliferative neoplasms, including polycythaemia vera and essential thrombocytosis. To date, the mechanism of JH2-mediated inhibition of JH1 kinase activation as well as the susceptibility of pathological mutant JAK2 to inhibition by the physiological negative regulator, SOCS3, have remained unclear. Here, using recombinant, purified JAK2JH1-JH2 proteins, we demonstrate that, when activated, wild-type and myeloproliferative neoplasm-associated mutants of JAK2 exhibit comparable enzymatic activity and inhibition by SOCS3 in in vitro kinase assays. Small angle X-ray scattering (SAXS) showed that JAK2JH1-JH2 exists in an elongated configuration in solution with no evidence for interaction between JH1 and JH2 domains in cis. Collectively, these data are consistent with a model in which JAK2’s pseudokinase domain binds and inhibits the activation of the tyrosine kinase domain of a neighbouring JAK2 molecule within a cytokine receptor complex, but does not influence the activity of JAK2 once it has been activated. Our data indicate that, in the absence of the N-terminal FERM domain and thus cytokine receptor association, the wild-type and pathological mutants of JAK2 are enzymatically equivalent and equally susceptible to inhibition by SOCS3.
INTRODUCTION
Nearly all patients diagnosed with polycythaemia vera, and the majority of those with the related myeloproliferative neoplasms (MPNs), essential thrombocythaemia and primary myelofibrosis, harbour a mutation in the pseudokinase domain of the protein kinase, JAK2 (Janus Kinase 2) [1–6]. The predominant acquired somatic missense mutation translates into the amino acid substitution of valine to phenylalanine at residue 617 and results in constitutive kinase activity of the protein. In contrast to wild-type JAK2, V617F mutant JAK2 initiates signalling in the absence of cytokine and this leads to aberrant proliferation of distinct haematopoietic cell subsets and gives rise to the disease phenotype.
JAK2 is a receptor-associated multi-domain kinase that initiates the intracellular signalling cascade induced by exposure to cytokines such as erythropoietin (EPO), thrombopoietin (TPO) and interleukin-3 (IL-3)-family cytokines. JAK2, like the other JAK family members (JAK1, JAK3, TYK2) consists of four distinct domains [7]. The N-terminal FERM domain binds to the intracellular Box 1 motif on cytokine receptors, such as the EPO and TPO receptors, whilst the C-terminal kinase domain is the catalytic domain responsible for phosphorylating downstream targets, including the STAT transcription factors. Between these two terminal domains is a non-standard SH2-like domain and a pseudokinase domain, which is responsible for ensuring that the catalytic domain is switched “off” in the absence of cytokine exposure. Since the discovery of the V617F JAK2 mutation, over 30 mutations in JAK2 have been identified in patients with MPNs and other proliferative blood disorders. The overwhelming majority of these occur in the pseudokinase domain of JAK2 and the linker region connecting the pseudokinase and SH2-like domains. It is presumed that these mutations disrupt the inhibitory effects of the pseudokinase domain but our understanding of how this, and the process of JAK2 activation itself, occurs mechanistically is largely speculative and key questions remain unanswered. Biochemical and structural studies of JAK2 have been hampered by the difficulty in producing full-length recombinant protein. Many structural snap-shots of the isolated kinase domains from JAK1, JAK2, JAK3 and TYK2 have been produced since the first X-ray crystal structures were determined [8–10], but only more recently have techniques been developed to produce the pseudokinase domain [11–13] and the pseudokinase-kinase (JH1-JH2) tandem domains [11]. This advancement allows us to visualize how the V617F mutation alters the conformation of the pseudokinase domain in isolation [14, 15]. In the absence of structural information on a multi-domain construct, however, it remains unclear how the JAK2 pseudokinase domain interacts with and negatively regulates the activity of the adjacent tyrosine kinase domain and, consequently, how mutations within the pseudokinase domain lead to constitutive activation of the tyrosine kinase domain.
Differential susceptibility or interaction of JAK2 mutants with cellular regulatory proteins is another possible mechanism for the disease pathogenesis. The contribution of SOCS family proteins, physiological regulators of JAK2, has been investigated but with somewhat conflicting results. SOCS proteins regulate JAK activity in a negative feedback loop. Their expression is induced by JAK activation [16] and they then down-regulate the JAK/STAT signalling cascade, either through direct inhibition of JAKs kinase activity [17, 18] or through targeting signalling components for proteasomal degradation [19]. Disruption of this regulatory system could contribute significantly to the myeloproliferative phenotype and affect the onset and/or the severity of the disease. Epigenetic silencing of SOCS1 and SOCS3 has been detected in patients with myeloproliferative disorders [20–24]. SOCS1 and SOCS3 mRNAs are also reported to be upregulated in patients with V617F-associated myeloproliferative disorders [25, 26], and protein levels similarly increase with the induction of JAK2 V617F over-expression in cell lines [27]. This latter study found that SOCS1 and SOCS3 can inhibit V617F JAK2 and reduce the expression levels of mutant JAK2. In contrast, some reports arising from over-expression studies indicate that SOCS3 is unable to inhibit V617F JAK2 [28, 29] due to tyrosine phosphorylation of SOCS3. Whether or not mutant JAK2s can be directly inhibited by SOCS1 or SOCS3 in myeloproliferative disorders is therefore a point of contention.
In this manuscript we use newly developed methods to produce recombinant purified human JAK2 tandem kinase-pseudokinase domain constructs, including a panel of 14 constructs harbouring mutations that had been identified in patients with haematological disorders. We investigate these constructs biochemically and show that, when fully activated, none of the MPN-derived mutant constructs have an increased intrinsic kinase activity compared to wild-type. This suggests that inappropriate activation is the sole mechanism that leads to aberrant downstream signaling and that, once activated, their catalytic activity is indistinguishable to that of the wild-type enzyme. In addition, we show that all of the MPN-derived mutants are inhibited by SOCS3 in vitro with similar IC50 values compared to wild-type JAK2JH1-JH2. Finally, we use small angle X-ray scattering to gain insight into the relative orientation of JAK2’s kinase and pseudokinase domains, and their interaction with SOCS3 and show that the pseudokinase and kinase domains do not exist in any fixed orientation relative to another, in vitro, but are instead connected by a flexible linker. Collectively our data are consistent with a model in which the pseudokinase domain regulates the activity of the kinase domain in trans and that the pathological mutations that affect this regulation do so by promoting inappropriate activation of the enzyme rather than altering its intrinsic catalytic activity. Our data indicate that the mechanism that promotes this aberrant activation is not encoded by the pseudokinase and kinase domains alone, as it is only evident in the context of the full-length protein and not in the tandem domain (pseudokinase-kinase) construct studied in the present work.
EXPERIMENTAL PROCEDURES
Recombinant JAK2 cloning and expression
A fragment of human JAK2 (hJAK2) that incorporates both the kinase (JH1) and pseudokinase (JH2) domains (JH1-JH2, residues 513–1132) was cloned as a His6-tagged protein in pFastBac HTb (Life Technologies). Wild-type hJAK2JH1-JH2 and a panel of 14 different mutants were produced by oligonucleotide-directed PCR mutagenesis and all insert sequences were verified by Sanger sequencing (Biomolecular Research Facility, Australian National University or Micromon, Monash University). A composite construct encoding His6 hJAK2JH1-JH2 and N-FLAG tagged hPTP1B was assembled using the MultiBac system, as described previously [30].
Viral stocks encoding His6-tagged JAK2JH1-JH2 constructs were raised in Spodoptera frugiperda (Sf21) cells using the Bac-to-Bac baculovirus expression system (Life Technologies) following bacmid preparation, as described [31]. Expressions were performed by infecting 0.5 L Sf21 shaking cultures (density 3–4 ×106 cells/mL) with 10% (v/v) viral supernatant and culturing for 48 hours. Purification of proteins from cell pellets was accomplished using a standardised two-step process: nickel-nitrilotriacetic acid (Ni-NTA) resin then Superdex 200 size-exclusion chromatography (GE Healthcare), essentially as described previously [11, 12, 32]. Proteins were eluted from the size-exclusion column in a buffer comprising 0.5 M NaCl, 20 mM Tris pH 8, 15% (v/v) glycerol and 1 mM TCEP. Fractions containing hJAK2JH1-JH2 were concentrated using centrifugal ultrafiltration, analysed by SDS-PAGE and Western blot, aliquoted, snap frozen in liquid N2 and stored at −80°C until required.
A shorter form of hJAK2JH1-JH2 (residues 536–1132) was used in Small Angle X-ray Scattering (SAXS) experiments. In these constructs the linker region between the SH2-like domain and JH2 was excluded. These hJAK2 protein constructs were expressed and purified as described above, with the following modifications: 0.4 μM of the JAK inhibitor CMP-6 (EMD Biosciences) was added to Sf21 protein expression cultures for enhanced protein yield and stability; and following purification with Ni-NTA the His6 tag was removed by incubation with 1 mg of Tobacco Etch Virus protease overnight at 4°C. Recombinant purified SOCS3 SH2 domain was prepared as described previously, with refolding in complex with the gp130 pY757 peptide [33]. hJAK2JH1-JH2:SOCS3 SH2:gp130 phosphopeptide complex, was prepared by mixing hJAK2JH1-JH2 with an excess of SOCS3 SH2:gp130, prior to purification by Superdex-200 size-exclusion chromatography. As a final purification step, both apo JAK2JH1-JH2 and hJAK2JH1-JH2:SOCS3 complex were subjected to ion-exchange chromatography (MonoQ; GE Healthcare) in a buffer system containing 20 mM Tris pH 8, 15% (v/v) glycerol and 1 mM TCEP over a gradient of 0 to 500 mM NaCl. Protein fractions were concentrated to 5–9 mg/mL by centrifugal ultrafiltration, aliquoted, snap frozen and stored at −80°C until required.
In vitro kinase inhibition assay
hJAK2JH1-JH2 constructs were tested for their ability to be inhibited by SOCS3 by in vitro kinase assay, essentially as described [34],. For these assays a modified murine SOCS3, lacking the first 21 amino acids and with its PEST motif (residues 129–163) replaced by a 4xGly-Ser linker, and co-expressed and purified with elongins B and C for stability and solubility from E. coli (hereafter SOCS3/elonginBC; as described in [35]). Briefly, 4 nM of hJAK2JH1-JH2 was incubated in kinase buffer (20 mM Tris pH 8.0, 100 mM NaCl, 100 μM 2-Mercaptoethanol (Sigma), and 4 mM MgCl2) with 1 mM ATP and 1 μCi γ[32P]-ATP and used to phosphorylate 0.67mM of Stat5b peptide (residues 693–708;[36]). SOCS3/elonginBC inhibitor was included at 0–1.35 μM. Reactions were carried out at 22°C and terminated at two time points (7.5 and 15 min) to ensure linearity of product formation. The reactions were quenched by spotting onto P81 phosphocellulose paper (Whatman) and washed four times for 15 min in 5% (v/v) phosphoric acid. Following a final acetone wash, the paper was air-dried before phosphorimaging.
Small angle X-ray Scattering (SAXS) data collection and analyses
SAXS data were collected on the SAXS/WAXS beamline at the Australian Synchrotron [37] using the inline size-exclusion chromatography setup, as described previously [30, 38, 39]. Protein samples were injected onto a Superdex 200 5/150 (GE Healthcare) column, pre-equilibrated in 0.5 M NaCl, 20 mM Tris pH 8, 15% (v/v) glycerol and 1 mM TCEP to eliminate any aggregates prior to X-ray scattering data collection, and to ensure proper buffer matching between blanks and protein samples. Protein samples were eluted via a 1.5 mm glass capillary at 16°C through which the 1.033 Å X-ray beam was positioned. The beamline was equipped with a 1 M, 170 mm×170 mm Pilatus detector (Dektris) at a distance of 1.6 m from the scattering source, providing a q range of 0.0114 – 0.4 Å. Data were collected at 2 s intervals over the course of the elution. 2D scattering data from the chromatography elution peak were radially averaged, normalized to sample transmission and an average of scattering profiles from prior to protein elution from the column was subtracted as background using the ScatterBrain software (Stephen Mudie, Australian Synchrotron). The ATSAS suite of software was used for all subsequent SAXS data analyses [40]. For an initial assessment of data quality, Guinier analysis was performed using PRIMUS [41]. The program GNOM [42] applied an indirect Fourier transform to the scattering profile to calculate the real-space, pairwise intra-atomic distance distribution function P(r) and the maximum dimension of the scattering particle, Dmax. Rigid body modelling was performed using BUNCH [43].
RESULTS
Enzymatic analyses of wild-type and V617F JAK2
JAK2V617F is known to be aberrantly active in vivo. Whilst it is clear that this is due to inappropriate activation of the mutant enzyme when bound to homodimeric receptors, even in the absence of cytokines [44], it is also possible that, when fully activated, JAK2V617F may also have a higher catalytic activity than wild-type JAK2. The Michaelis-Menten analyses that could answer such questions have been hampered, to date, by an inability to produce recombinant fragments of JAK2 that include the pseudokinase domain. We recently developed an expression and purification protocol for JAK2 constructs that contain both the pseudokinase (JH2) and kinase (JH1) domains [11] and used this to produce wild-type and mutant (V617F) JAK2JH2-JH1 as well as the JAK2JH1 domain alone. These constructs were purified to near homogeneity and subjected to Michaelis-Menten analysis using in vitro kinase assays. All constructs were incubated with ATP prior to the final step of their purification to ensure they were fully phosphorylated at pY1007/1008 in their activation loop in JH1. Both ATP and substrate concentrations were varied in order for both KMATP and KMSubstrate to be measured. The substrate used was a tyrosine-containing peptide from STAT5b described previously [36]. We repeated the experiments 4–6 times, each time using a different preparation of enzyme, in order to ensure that any potential differences were not due to batch-specific differences in enzymatic activity. As shown in Table 1 (and Supplementary Figure 1) there was no reproducible, marked difference between the KM values of all three constructs for either ATP or peptide substrate. The other kinetic constant accessible via Michaelis-Menten analyses, kcat, could not be reliably ascertained as the specific activity of individual JAK2JH1-JH2 preparations (both wild-type and mutant) varied significantly. Nevertheless, measurements of kcat for JAK2JH1, JAK2JH1-JH2 and V617F JAK2JH1-JH2, were always within three-fold of each other (approximately 2 s−1), suggesting that there is no dramatic difference in the kinetic parameters (data not shown). We conclude that there are no significant differences in the enzymatic activities of activated wild-type and mutant (V617F) JAK2JH1-JH2 and that the aberrant activity seen in vivo is due purely to inappropriate activation of JAKs.
Table 1.
Kinetic parameters of JAK2 constructs
| KMATP (μM)* | KMSubstrate (μM)* | |
|---|---|---|
| JAK2JH1-JH2 (wild-type) | 216 ± 40 | 620 ± 230 |
| JAK2JH1-JH2 (V617F) | 144 ± 45 | 402 ± 92 |
| JAK2JH1 | 105 ± 36 | 390 ±111 |
error indicates 95% CI. n=4
Biochemical analyses of MPN-derived JAK2 mutants
Given that V617F JAK2JH1-JH2 showed no obvious difference in terms of catalytic activity compared to wild-type, we wished to investigate whether our JAK2JH1-JH2 constructs were more easily autoactivated. The primary readout for activation of JAK is phosphorylation of the JH1 activation loop tyrosines 1007/1008. Although it is known that full-length V617F JAK2 is aberrantly activated in vivo, it was unclear whether this mutation would induce increased autoactivation of our JAK2JH1-JH2 constructs, given the absence of upstream FERM and SH2-like domains in the absence of associated receptors. In addition to V617F, there are now over a dozen JAK2 mutants associated with myeloproliferative disease. We therefore extended our studies to include a number of these different mutations. Fourteen different MPN-derived JAK2 mutants were expressed and purified; these mutants incorporated many of the missense, insertion and deletion mutations reported in patients with MPNs and other blood disorders (Figure 1A). These included a selection from the three pseudokinase domain mutational “hotspots”, in exons 12, 14 and 16, as well as three kinase domain mutants (see Table 2). All purified JAK2JH1-JH2 constructs were active, indicating they were correctly folded, and displayed similar KMATP and KMSubstrate values to those described above when tested in in vitro assays (data not shown).
Figure 1. Characterization of purified recombinant JAK2JH1-JH2 preparations.
His-tagged JAK2 JH1-JH2 domains were expressed in Sf21 cells and purified by Ni-NTA affinity and size-exclusion chromatography. 0.5 μg of purified protein was analysed by SDS-PAGE and Coomassie stained (A) or transferred to PVDF membranes and treated with antibodies against phosphorylated activation loop Y1007/1008 (B: top panel) or phosphorylated Y570 (B: bottom panel). All JAK2JH1-JH2 proteins were N-terminally His6 tagged, except for the C-terminally tagged wild-type control, WT-His. Western blots are representative of two independent experiments.
Table 2.
Human JAK2 mutations associated with proliferative blood disorders cloned in this study.
| Mutation | c.DNA | Exon | Domain | Disease | Reference |
|---|---|---|---|---|---|
| FHK537-9>L | c.1611_1616del TCACAA | 12 | SH2-like:JH2 linker | IE, PV | [57] |
| K539L | c.1615_1616AA >CT | 12 | SH2-like:JH2 linker | IE, PV | [57, 58] |
| K607N | c.1821G>C | 14 | JH2 | AML | [59] |
| L611S | c.1832T>C | 14 | JH2 | ALL, PV | [60, 61] |
| V617F | c.1849G>T | 14 | JH2 | PV, ET, MF, Atypical CML | [1, 3–6, 59] |
| D620E | c.1860C>A | 14 | JH2 | PV | [62, 63] |
| I682del insMPAP | c.2046_2046C>GCCCGCCCCT | 16 | JH2 | ALL | [64] |
| I682F | c.2044A>T | 16 | JH2 | Paediatric B-ALL | [65, 66] |
| R683S | c.2049A>T | 16 | JH2 | Paediatric B-ALL, ALL | [64–67] |
| R683G | c.2047A>G | 16 | JH2 | Paediatric B-ALL, ALL | [64–67] |
| R683K | c.2048G>A | 16 | JH2 | ALL | [64] |
| R867Q | c.2600G>A | 20 | JH1 | Paediatric B-ALL | [65, 66] |
| D873N | c.2617G>A | 20 | JH1 | Paediatric B-ALL | [65, 66] |
| P933R | c.2798C>G | 21 | JH1 | Paediatric B-ALL | [65, 66] |
IE, idiopathic erythrocytosis; ALL, acute lymphoblastic leukaemia; PV, polycythaemia vera; ET, essential thrombocytosis; MF, myelofibrosis; CML, chronic myelogenous leukaemia; AML, acute myelogenous leukaemia; B-ALL, B-cell acute lymphoblastic leukaemia.
In order to determine whether these mutations were sufficient to induce aberrant auto-activation, we examined the phosphorylation of their activation loop tyrosines using a pY1007/1008 specific antibody. As shown in (Figure 1B, upper panel) the 14 JAK2JH1-JH2 mutants were autophosphorylated at pY1007/1008 at similar levels to that seen in wild-type JAK2JH1-JH2. To verify that any phosphorylation of the activation loop was due to auto-activation and not a contaminating insect cell kinase, K882R JAK2JH1-JH2, which is known to be catalytically inactive [45] was included as a control. No autophosphorylation of Y1007/1008 in K882R JAK2JH1-JH2 could be detected, confirming that the phosphorylation of the activation loop of the other JAK2 constructs occurred via auto-activation (see Figure 1B, lane 5). These results are consistent with Sanz and colleagues’ finding for recombinant purified V617F JAK2JH1-JH2 [11] and suggests that the higher level of activation loop phosphorylation in full-length mutant JAK2 is due to the requirement that it be scaffolded to a receptor for constitutive trans activation [46] or that the high concentrations of recombinant JAKs in our expression system allow phosphorylation in trans even in the absence of receptor association.
Another key event in the control of JAK2 activation is the phosphorylation of tyrosine 570, an event thought to be an inhibitory signal [47, 48]. Phosphorylation of Y570 was recently suggested to be an auto-activation event catalysed by the pseudokinase (JH2) domain, which is down-regulated in MPN-derived JAK2 mutants [12, 14]. Using an antibody specific to pY570, we did not observe any reproducible difference in the level of Y570 phosphorylation for any JAK mutants with the exception of Y570F JAK2JH1-JH2 (negative control) and K882R JAK2JH1-JH2 (Figure 1B, lower panel). The latter result was unexpected as it suggests that kinase domain activity or an active conformation of the kinase domain is absolutely required for Y570 phosphorylation.
SOCS3 is a potent inhibitor of MPN-derived JAK2JH1-JH2 mutants in vitro
One outstanding question in the field relates to the potentially ameliorating effect that the action of the physiological inhibitor of JAK2 activity, SOCS3, may have in regulating the onset or severity of the myeloproliferative phenotype induced by the V617F mutation. Some studies have suggested that that V617F JAK2 is immune to regulation by SOCS3 [28, 29] whilst others have indicated the contrary [27]. We therefore tested the ability of SOCS3 to inhibit V617F JAK2JH1-JH2 in vitro, using our previously reported assay system that measures JAK2 phosphorylation of the STAT5b peptide [31, 34]. A complex of SOCS3 with elongins B and C was used as these are the physiological ligands for SOCS3 and improve its solubility and stability in vitro [35]. As shown in Figure 2, SOCS3 inhibited all JAK2JH1-JH2 mutant proteins with a similar inhibitory concentration for 50% maximal activity (IC50) to wild-type JAK2JH1-JH2. This shows that the MPN-derived mutations do not disrupt the SOCS3 binding site, an idea supported by the fact that our wild-type JAK2JH1-JH2 and V617F JAK2JH1-JH2 constructs both bound SOCS3 with similar affinity as judged by surface plasmon resonance analysis (Supplementary Figure 1B).
Figure 2. JAK2JH1-JH2 mutants are inhibited by SOCS3.
A. SOCS3 had a comparable inhibitory effect on phosphotransfer to a STAT5b peptide substrate catalysed by purified recombinant wild-type and MPN mutant JAK2JH1-JH2 constructs in in vitro kinase assays. B. An overlay of the data from panel A illustrates that SOCS3-mediated inhibition of mutant JAK2 proteins is comparable to that toward wild-type JAK2JH1-JH2, with all IC50 values ~100 nM. Data are normalized to no inhibitor controls and averages of two independent experiments are shown. Error bars represent the range.
Structural studies of JAK2JH1-JH2 uncomplexed (apo) and in complex with SOCS3 by small angle X-ray scattering
A number of outstanding questions surround the molecular mechanism by which JAK2’s tyrosine kinase (JH1) domain is negatively regulated by the pseudokinase (JH2) domain. To date, two recurrent models for have been proposed to describe the binding and inhibition of JH1 by JH2, in which JH2 either binds in cis to the adjacent JH1 domain within the same JAK2 molecule (Figure 3A) or to the JH1 domain of another JAK2 molecule in trans (Figure 3B), although neither model has been adequately tested experimentally. Similarly, we sought to examine whether the SOCS3 SH2 domain binds JAK2JH1-JH2 in a mode comparable to that observed for the isolated JAK2JH1 domain [30, 34, 49]. Having prepared recombinant JAK2 comprising the JH2 and JH1 domains and the related SOCS3 SH2 domain complex (Figure 3), we proceeded to characterize the arrangement of domains in solution using small angle X-ray scattering (SAXS) experiments. SAXS data were collected at the Australian Synchrotron using the inline size-exclusion chromatography setup [37], with protein eluted in a buffer comprising 0.5 M NaCl, 20 mM Tris pH 8, 15% glycerol, 0.5 mM TCEP (summary statistics recorded in Table 3; Figure 4A, B). Comparable scatter patterns were obtained from experiments in which ion-exchange chromatography fractions (in 20 mM Tris pH 8, 10% glycerol, 0.5 mM TCEP containing ~0.2 M NaCl) were loaded directly into the capillary (data not shown). As shown in the insets accompanying the scatter profiles in Figures 4A and B, the Guinier plots for each of the apo JAK2JH1-JH2 and JAK2JH1-JH2:SOCS3 complex SAXS profiles are linear, consistent with protein monodispersity and an absence of aggregation that would confound data analyses. Indirect Fourier transform analyses of these data using GNOM enabled us to generate plots of real-space interatomic distance distributions (P(r) plots, Figure 4C). As shown in Figure 4C, both the apo JAK2JH1-JH2 and JAK2JH1-JH2:SOCS3 SH2 domain complex exhibit P(r) distributions representative of elongated, non-spherical particles in solution. Furthermore, these analyses indicated that the maximum dimension of the scattering particle, Dmax, is longer for the JAK2JH1-JH2:SOCS3 complex (150 Å) than the apo JAK2JH1-JH2 (135 Å). Based on our previous characterisation of JAK2JH1 inhibition by the SOCS3 SH2 domain [30, 34, 49, 50] and the JAK2JH1-JH2 inhibition studies described above, we predicted that the SOCS3 SH2 domain would engage the JH1 domain within the JAK2JH1-JH2 construct via an analogous, phospho-independent binding mode. To test this hypothesis, we used size exclusion chromatography to examine whether SOCS3 SH2 domain could form a stable complex with dephosphorylated JAK2JH1-JH2 as we observed for phosphorylated JAK2JH1-JH2. Indeed dephosphorylation of the JAK2JH1 activation loop residues, Y1007 and Y1008, by co-expression of JAK2JH1-JH2 with the cognate phosphatase, PTP1B, in insect cells did not impact the capacity of JAK2JH1-JH2 to form a stable complex with SOCS3 SH2 domain (Figure 4D and Supplementary Figure 2A). These data are consistent with our earlier interrogation of JAK2JH1 engagement by the SOCS3 SH2 domain [30] and suggest that an analogous mode of interaction occurs within the context of the JAK2JH1-JH2 tandem domains. Additionally, relative to molecular weight standards, these data illustrate that apo JAK2JH1-JH2 elutes at a volume consistent with the protein behaving as a monomer, with a small, but measurable, increase in molecular mass evident upon SOCS3 SH2 binding (Figure 4D).
Figure 3. Models depicting candidate mechanisms of JAK2 pseudokinase (JH2) inhibition of the tyrosine kinase (JH1) domain.
Schematic representations of two candidate mechanisms by which the JAK2 JH2 domain inhibits the catalytic activity of JH1 in cis (A) within the same JAK2 molecule or in trans (B) by binding the JH1 domain of another JAK2 protomer within an unactivated, dimeric class I cytokine receptor complex (such as that of gp130). The four domains of JAK2 are shown schematically: the N-terminal receptor-bound FERM domain, the SH2-like domain of unknown function, the regulatory pseudokinase domain (JH2) and the C-terminal tyrosine kinase domain (JH1).
C. SOCS3 (yellow) binds to JAK2JH1 via its SH2 domain to inhibit its kinase activity, and is simultaneously bound to the cytokine receptor, gp130.
Schematic representations of constructs used to test the models shown in panels A–C in SAXS and size exclusion chromatography experiments (Figure 4) are shown on the right of the respective model.
Table 3.
Data collection and scattering parameters for small angle X-ray scattering
| Data collection parameters | |
|---|---|
| Instrument | Australian Synchrotron SAXS/WAXS beamline |
| Beam geometry | 120 μm point source |
| Beam wavelength (Å) | 1.033 |
| q range (Å−1) | 0.0114 – 0.4 |
| Exposure time (s) | 2 |
| Protein concentration (mg/mL)§ | 5–9 |
| Temperature (°C) | 16 |
| Structural parameters | ||
|---|---|---|
| apo JAK2JH1-JH2 | JAK2JH1-JH2: SOCS3 SH2: gp130 phosphopeptide | |
| I(0) (cm−1) [from P(r)] | 0.02948±0.00017 | 0.02027±0.00036 |
| Rg (Å) [from P(r)] | 38.11±0.35 | 41.26±0.96 |
| I(0) (cm−1) [from Guinier] | 0.0291±0.00002 | 0.02010±0.00034 |
| Rg (Å) [from Guinier] | 36.4±0.39 | 39.7±1.03 |
| Dmax (Å) | 135 | 150 |
| Software | |
|---|---|
| Primary data reduction | ScatterBrain (Australian Synchrotron) |
| Data Processing | PRIMUS, GNOM |
| Rigid body modelling | BUNCH |
| Computation of model intensities | CRYSOL |
| 3-D graphics representations | MacPyMOL |
via in-line size exclusion chromatography
Figure 4. Small angle X-ray scattering analyses of apo JAK2JH1-JH2 and the JAK2JH1-JH2:SOCS3 complex.
Scattering intensity profiles for (A) apo JAK2JH1-JH2 and (B) JAK2JH1-JH2:SOCS3 complex with inset Guinier plots for data in the range, q.Rg ≤ 1.3. Linearity of the Guinier plots indicates neither high molecular weight aggregates nor inter-particle interference contribute measurably to scattering. C. Fourier transformation of the scattering intensity yields the pair-wise inter-atomic distance distribution function, P(r), calculated using GNOM. The maximum particle dimension, Dmax, for apo JAK2JH1-JH2 is 135 Å (grey curve) and 150 Å for JAK2JH1-JH2:SOCS3 SH2 domain complex.
D. Superdex-200 size exclusion chromatography analyses of apo JAK2JH1-JH2, JAK2JH1-JH2:SOCS3 complex, dephospho-JAK2JH1-JH2:SOCS3 complex and SOCS3 SH2 domain. The elution volumes of molecular weight standards are marked above the chromatograms. Apo JAK2JH1-JH2 and apo dephosphorylated JAK2JH1-JH2 elution profiles superimpose (not shown) and as such we have omitted the latter profile for clarity. Peak fractions of JAK2JH1-JH2:SOCS3 and dephospho-JAK2JH1-JH2:SOCS3 were resolved by reducing SDS-PAGE before Coomassie Blue staining or anti-pY1007/1008 Western blot analysis (Supplementary Figure 2A).
E. Rigid-body modelling of apo JAK2 JH1-JH2 domains using BUNCH modelling to fit the experimental scattering data (A). The lower structural cartoon depicts a 90° rotation of the upper cartoon about the x-axis. The model comprises the crystal structures of JH2 (PDB: 4FVP; dark blue) and JH1 (PDB:2B7A; light blue) connected by a flexible linker of dummy atoms (grey), with relative positions of JH2 and JH1 optimised to fit experimental scattering data. The fit of BUNCH model to experimental data is shown in Supplementary Figure 2B. F. Rigid body modelling of the JAK2 JH1-JH2:SOCS3 SH2 domain complex, as described for apo JAK2 JH1-JH2 (E), but using JH2 (PDB: 4FVP; dark blue) and JH1:SOCS3 complex (PDB:4GL9; JAK2 JH1, light blue; SOCS3 SH2, yellow). The fit of BUNCH model to experimental data is shown in Supplementary Figure 2C.
We further analysed our SAXS data to define the orientations of the JH1 and JH2 domains relative to one another in solution. Using the crystal structures of JAK2JH1 and JAK2JH2 as rigid bodies, we used BUNCH [43] to fit the JH1 and JH2 models connected by a flexible bead linker to our experimental SAXS profile (Figure 4E; χ for fit to data=0.38; Supplementary Figure 2B). As anticipated from the P(r) distribution (Figure 4C), the JAK2JH1-JH2 model derived from BUNCH is elongated, rather than globular. Similarly, we used BUNCH to perform rigid body modeling with the JAK2JH2 and JAK2JH1:SOCS3 crystal structures as starting models to generate the models shown in Figure 4F (χ for fit to data=0.36; Supplementary Figure 2C). We elected to perform fitting using the JAK2JH1:SOCS3 SH2 domain complex as a single rigid body rather than the component domains, since our biochemical (Figure 2) and size exclusion chromatography (Figure 4D) studies are consistent with SOCS3 inhibition of JH1 in the context of the JAK2JH1-JH2 tandem domains occurring via the same mode as observed for JAK2JH1 alone in earlier work [30, 34]. The resulting model reveals an elongated arrangement of domains (Figure 4F), similar to that observed for apo JAK2JH1-JH2, although the longer maximum dimension, Dmax, observed for the JAK2JH1-JH2:SOCS3 complex relative to apo JAK2JH1-JH2 can be accounted for by the addition of SOCS3 SH2 domain to the complex. Overall, these data are most consistent with JAK2JH1-JH2 existing in an elongated conformation in solution, in which the JAK2JH2 and JAK2JH1 domains do not directly engage one another.
DISCUSSION
The JAK2 pseudokinase (JH2) domain was initially revealed as an essential negative regulator of the tyrosine kinase activity of the adjacent JH1 domain in 2000 [51], and coupled with subsequent identification of mutations within the JH2 domain as the molecular basis for haematological malignancies, this has ignited interest in the physiological roles of this domain. Whilst the mechanism by which it exerts its activity upon the tyrosine kinase domain has been the subject of intense speculation over the past decade, biochemical examination of the interaction between the JAK2JH1 and JAK2JH2 domains has been thwarted by a dearth of methods to produce purified, recombinant constructs that incorporate both of these domains. Methods to produce such material have only recently been developed [11, 12].
We purified a recombinant JAK2JH1-JH2 construct and examined its structure using small angle X-ray scattering (SAXS). Strikingly, these experiments showed that there was no detectable interaction between the two adjacent domains when they were part of the same polypeptide chain. This is contrary to the prevailing model in which JH2 binds and inhibits its adjacent JH1 domain in cis (Figure 3A). Our SAXS data show that the two domains adopt a “beads-on-a-string”, elongated conformation with no stable contacts with each other. This raises several possibilities, either: (A) the N-terminal FERM and SH2 domains are required to scaffold a direct JAK2JH1:JAK2JH2 interaction in cis; (B) the interaction between the two domains occurs in trans, whereby the JH2 domain of one JAK molecule inhibits the activation of the JH1 domain of a second JAK molecule; or (C) any JAK2JH1:JAK2JH2 interaction is lost upon activation of the kinase (JAK2JH1) domain. The latter hypothesis is possible given that the JAK2JH1-JH2 constructs we used in our SAXS experiments were fully phosphorylated on Y1007, while analogous analyses of the dephosphorylated protein were precluded by lower solubility. It is even possible that a combination of all three of these possibilities represents the physiological situation. We favour a trans-inhibition model in light of our data and that contained within a recent study which indicated that, within the physiological context of signalling by full length JAK proteins associated with the dimeric thrombopoietin or erythropoietin receptors, mutations within the JAK2JH2 domain preclude binding and inhibition of the JH1 domains of not only JAK2, but also the other JAK family members, JAK1 and TYK2, bound to a neighbouring receptor chain within a preformed dimeric receptor complex [52].
The fact that we see no evidence for JAK2JH1-JH2 tandem domains forming an intermolecular dimer in vitro (by size exclusion chromatography or small angle X-ray scattering) does not detract from the above hypothesis as, in vivo, individual JAK monomers are brought into close proximity by associating with dimeric (or oligomeric) receptors. This results in a much higher local concentration of JAK pairs than can be obtained by simply concentrating recombinant JAKJH1-JH2 constructs in vitro. It is notable that if the JAK2JH1:JAK2JH2 interaction occurred in cis, then it would not be concentration- or receptor-dependent, as the two domains are part of the same polypeptide chain. Indeed, the observation that recombinant JAK2JH1-JH2 proteins were purified from insect cells in an activated form, as evidenced by Y1007/1008 phosphorylation (Figure 1B), is consistent with the JAK2JH2 domain not binding and inhibiting the adjoining JAK2JH1 domain in cis, in keeping with the elongated JAK2JH1-JH2 structure observed in SAXS experiments.
The V617F mutation in the JAK2 pseudokinase domain is associated with a number of human myeloproliferative disorders, especially polycythemia vera. This mutation causes JAK2 to become constitutively active, whereas the wild-type enzyme is only activated after cytokine binds to the extracellular domain of the receptor to which JAK is bound. We conceived of two possibilities to best describe the molecular events that lead to this constitutive activation. Either (A) mutant JAK2 is hyper-activated and therefore phosphorylates itself and downstream substrates at an unusually high rate or (B) mutant JAK2 trans-phosphorylation is constitutive, while its activity in phosphorylating downstream targets is the same as that of activated wild-type JAK2. These two possibilities can also be described as (A) mutant JAK is aberrantly active or (B) mutant JAK is aberrantly activated. We investigated the first possibility by investigating the enzymatic activity of wild-type and V617F JAK2JH1-JH2. We ensured that both enzymes were fully activated before commencing these experiments by pre-incubating them with ATP and monitoring the phosphorylation of their activation loop tyrosines (Y1007/1008) by Western blot. We found the KM and Vmax of the fully activated mutant (V617F) enzyme to be the same as fully-activated wild-type JAK2JH1-JH2. This analysis was extended to a panel of fourteen different MPN-derived JAK2JH1-JH2 mutants and none of them displayed any significant difference in activity compared to wild-type JAK2JH1-JH2. Moreover, wild-type and mutant JAK2JH1-JH2 constructs exhibited comparable auto-phosphorylation of the JH1 activation loop residues, Y1007/1008, and the negative regulatory phosphorylation site in JH2, Y570, in Western blot analyses, indicating no intrinsic defects or elevations in the catalytic activities of these JAK2JH1-JH2 constructs. These observations are consistent with the fact that the N-terminal FERM and SH2-like domains and receptor association are known to be a necessary prerequisite for the enhanced auto-activation seen for mutant JAK2 [44, 53–55]. The absence of Y570 phosphorylation in the K882R protein was unexpected, since in cells Y570 phosphorylation has been shown to occur in catalytically defective JAK2 mutants expressed in JAK2-deficient cell lines [12, 48] and in recombinant purified JAK2JH2 domain [12]. It is thus possible that the ATP bound conformations of both JAK2JH1 and JAK2JH2 are required for optimal phosphorylation of Y570. We conclude that, although MPN-derived JAK2JH1-JH2 mutants are “switched on” (activated) inappropriately, their catalytic activity once they are switched on is normal.
Apart from the pseudokinase domain, the other major regulator of JAK2 activity, in vivo, is SOCS3. SOCS3 inhibits the catalytic domain of JAK2 (in addition to JAK1 and TYK2) by binding near the active site and preventing substrate access. This is believed to be independent of the pseudokinase domain although that had never been formally tested. If that is the case then SOCS3 should inhibit mutant JAK2 and act, in vivo, to ameliorate the myeloproliferative phenotype. However, there have been conflicting reports as to whether MPN-derived JAK2 mutants are susceptible to inhibition by SOCS3. We sought to investigate this in vitro by performing kinase inhibition assays using SOCS3 and our panel of MPN-derived JAK2JH1-JH2 mutants. All JAK2JH1-JH2 mutants were inhibited by SOCS3 with similar affinity. This indicates that the JAK2JH2 domain (and mutations within) do not interfere with SOCS3 binding. This observation is consistent with our SAXS analysis of a SOCS3:JAK2JH1-JH2 complex. As observed for the JAK2JH1 domain in previous studies [30, 34], SOCS3 formed a stable complex with the JAK2JH1-JH2 tandem domains (Figure 4D). This complex is elongated, like the apo JAK2JH1-JH2, but exhibits a longer maximum particle dimension (Figure 4C), owing to further extension arising from the addition of the SOCS3 SH2 domain (Figure 4F). Together these data support a model in which JAK2 binds SOCS3 via the JAK2JH1 domain exclusively, explaining why the introduction of MPN mutations have no bearing on the capacity of SOCS3 to inhibit JAK2JH1 activity within the context of the JAK2JH1-JH2 tandem domains (Figure 3C). Our data show that MPN-derived JAK2JH1-JH2 mutants have the same intrinsic susceptibility to SOCS3 inhibition as wild-type JAK2JH1-JH2 however, in vivo there may be factors that confound this inhibition. This may include hyperphosphorylation of SOCS3 by mutant JAK2 as suggested in earlier studies [28, 29].
Our recent work suggests that few pseudokinases exhibit kinase activity [32, 56], however JAK2JH2 is one of those few that does [12]. It still remains unclear whether the weak enzymatic activity of JAK2’s pseudokinase domain is intrinsic to the negative regulatory role it serves. By comparison, the pseudokinase domain of JAK1 does not undergo autophosphorylation ([13]; and DU and OS, unpublished work), suggesting that the negative regulation conferred by the JAK1 pseudokinase domain may arise principally as a consequence of its interaction with the tyrosine kinase (JH1) domain. These questions await the structural characterization of full-length JAK2, and our efforts lie in this direction.
Supplementary Material
SUMMARY STATEMENT.
Unexpectedly, wild-type and myeloproliferative neoplasm-associated mutants of JAK2 pseudokinase (JH2)-kinase (JH1) exhibited comparable catalytic activities and inhibition by SOCS3 in vitro. These data were consistent with small angle scattering, in which the JH2 and JH1 domains behaved independently in the absence of receptor association.
Acknowledgments
We thank the Australian Synchrotron SAXS/WAXS beamline staff for their insights and expert assistance with data collection.
FUNDING
This work was supported by NHMRC grants [1011804, 1016647]; a grant from the National Institutes of Health, USA (CA-22556); Leukaemia Foundation and Australian Stem Cell Centre scholarships to LNV; an NHMRC fellowship to NAN; ARC fellowships to JJB and JMM [FT110100169, FT100100100]; Medical Research Council of Academy of Finland, the Sigrid Juselius Foundation, Medical Research Fund of Tampere University Hospital, the Finnish Cancer Foundation, and Tampere Tuberculosis Foundation grants to OS; with additional support from the Victorian State Government Operational Infrastructure Support and NHMRC IRIISS grant [361646].
Footnotes
AUTHOR CONTRIBUTIONS
Leila Varghese, Nicos Nicola, Olli Silvennoinen, Jeffrey Babon and James Murphy designed the project; Leila Varghese, Nicholas Liau, Samuel Young, Artem Laktyushin, Jeffrey Babon and James Murphy performed experiments; Leila Varghese, Nicholas Liau, Isabelle Lucet, Nicos Nicola, Jeffrey Babon and James Murphy analysed data; Daniela Ungureanu, Henrik Hammaren and Olli Silvennoinen contributed vital reagents and protocols; Leila Varghese, Jeffrey Babon and James Murphy wrote the manuscript with input from all other authors.
References
- 1.Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, Vassiliou GS, Bench AJ, Boyd EM, Curtin N, Scott MA, Erber WN, Green AR. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365:1054–1061. doi: 10.1016/S0140-6736(05)71142-9. [DOI] [PubMed] [Google Scholar]
- 2.Constantinescu SN, Leroy E, Gryshkova V, Pecquet C, Dusa A. Activating Janus kinase pseudokinase domain mutations in myeloproliferative and other blood cancers. Biochem Soc Trans. 2013;41:1048–1054. doi: 10.1042/BST20130084. [DOI] [PubMed] [Google Scholar]
- 3.James C, Ugo V, Le Couédic JP, Staerk J, Delhommeau F, Lacout C, Garçon L, Raslova H, Berger R, Bennaceur-Griscelli A, Villeval JL, Constantinescu SN, Casadevall N, Vainchenker W. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434:1144–1148. doi: 10.1038/nature03546. [DOI] [PubMed] [Google Scholar]
- 4.Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg JR, Tichelli A, Cazzola M, Skoda RC. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352:1779–1790. doi: 10.1056/NEJMoa051113. [DOI] [PubMed] [Google Scholar]
- 5.Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJ, Boggon TJ, Wlodarska I, Clark JJ, Moore S, Adelsperger J, Koo S, Lee JC, Gabriel S, Mercher T, D’Andrea A, Fröhling S, Döhner K, Marynen P, Vandenberghe P, Mesa RA, Tefferi A, Griffin JD, Eck MJ, Sellers WR, Meyerson M, Golub TR, Lee SJ, Gilliland DG. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7:387–397. doi: 10.1016/j.ccr.2005.03.023. [DOI] [PubMed] [Google Scholar]
- 6.Zhao R, Xing S, Li Z, Fu X, Li Q, Krantz SB, Zhao ZJ. Identification of an acquired JAK2 mutation in polycythemia vera. The Journal of biological chemistry. 2005;280:22788–22792. doi: 10.1074/jbc.C500138200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Harpur AG, Andres AC, Ziemiecki A, Aston RR, Wilks AF. JAK2, a third member of the JAK family of protein tyrosine kinases. Oncogene. 1992;7:1347–1353. [PubMed] [Google Scholar]
- 8.Lucet IS, Fantino E, Styles M, Bamert R, Patel O, Broughton SE, Walter M, Burns CJ, Treutlein H, Wilks AF, Rossjohn J. The structural basis of Janus kinase 2 inhibition by a potent and specific pan-Janus kinase inhibitor. Blood. 2006;107:176–183. doi: 10.1182/blood-2005-06-2413. [DOI] [PubMed] [Google Scholar]
- 9.Boggon TJ, Li Y, Manley PW, Eck MJ. Crystal structure of the Jak3 kinase domain in complex with a staurosporine analog. Blood. 2005;106:996–1002. doi: 10.1182/blood-2005-02-0707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Williams NK, Bamert RS, Patel O, Wang C, Walden PM, Wilks AF, Fantino E, Rossjohn J, Lucet IS. Dissecting specificity in the Janus kinases: the structures of JAK-specific inhibitors complexed to the JAK1 and JAK2 protein tyrosine kinase domains. J Mol Biol. 2009;387:219–232. doi: 10.1016/j.jmb.2009.01.041. [DOI] [PubMed] [Google Scholar]
- 11.Sanz A, Ungureanu D, Pekkala T, Ruijtenbeek R, Touw IP, Hilhorst R, Silvennoinen O. Analysis of Jak2 Catalytic Function by Peptide Microarrays: The Role of the JH2 Domain and V617F Mutation. PLoS ONE. 2011;6:e18522. doi: 10.1371/journal.pone.0018522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ungureanu D, Wu J, Pekkala T, Niranjan Y, Young C, Jensen ON, Xu CF, Neubert TA, Skoda RC, Hubbard SR, Silvennoinen O. The pseudokinase domain of JAK2 is a dual-specificity protein kinase that negatively regulates cytokine signaling. Nat Struct Mol Biol. 2011;18:971–976. doi: 10.1038/nsmb.2099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Toms AV, Deshpande A, McNally R, Jeong Y, Rogers JM, Kim CU, Gruner SM, Ficarro SB, Marto JA, Sattler M, Griffin JD, Eck MJ. Structure of a pseudokinase-domain switch that controls oncogenic activation of Jak kinases. Nat Struct Mol Biol. 2013;20:1221–1223. doi: 10.1038/nsmb.2673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Bandaranayake RM, Ungureanu D, Shan Y, Shaw DE, Silvennoinen O, Hubbard SR. Crystal structures of the JAK2 pseudokinase domain and the pathogenic mutant V617F. Nat Struct Mol Biol. 2012;19:754–759. doi: 10.1038/nsmb.2348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Silvennoinen O, Ungureanu D, Niranjan Y, Hammaren H, Bandaranayake R, Hubbard SR. New insights into the structure and function of the pseudokinase domain in JAK2. Biochem Soc Trans. 2013;41:1002–1007. doi: 10.1042/BST20130005. [DOI] [PubMed] [Google Scholar]
- 16.Starr R, Willson TA, Viney EM, Murray LJ, Rayner JR, Jenkins BJ, Gonda TJ, Alexander WS, Metcalf D, Nicola NA, Hilton DJ. A family of cytokine-inducible inhibitors of signalling. Nature. 1997;387:917–921. doi: 10.1038/43206. [DOI] [PubMed] [Google Scholar]
- 17.Sasaki A, Yasukawa H, Suzuki A, Kamizono S, Syoda T, Kinjyo I, Sasaki M, Johnston JA, Yoshimura A. Cytokine-inducible SH2 protein-3 (CIS3/SOCS3) inhibits Janus tyrosine kinase by binding through the N-terminal kinase inhibitory region as well as SH2 domain. Genes to cells : devoted to molecular & cellular mechanisms. 1999;4:339–351. doi: 10.1046/j.1365-2443.1999.00263.x. [DOI] [PubMed] [Google Scholar]
- 18.Yasukawa H, Misawa H, Sakamoto H, Masuhara M, Sasaki A, Wakioka T, Ohtsuka S, Imaizumi T, Matsuda T, Ihle JN, Yoshimura A. The JAK-binding protein JAB inhibits Janus tyrosine kinase activity through binding in the activation loop. The EMBO journal. 1999;18:1309–1320. doi: 10.1093/emboj/18.5.1309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zhang JG, Farley A, Nicholson SE, Willson TA, Zugaro LM, Simpson RJ, Moritz RL, Cary D, Richardson R, Hausmann G, Kile BJ, Kent SB, Alexander WS, Metcalf D, Hilton DJ, Nicola NA, Baca M. The conserved SOCS box motif in suppressors of cytokine signaling binds to elongins B and C and may couple bound proteins to proteasomal degradation. Proceedings of the National Academy of Sciences of the United States of America. 1999;96:2071–2076. doi: 10.1073/pnas.96.5.2071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Capello D, Deambrogi C, Rossi D, Lischetti T, Piranda D, Cerri M, Spina V, Rasi S, Gaidano G, Lunghi M. Epigenetic inactivation of suppressors of cytokine signalling in Philadelphia-negative chronic myeloproliferative disorders. Br J Haematol. 2008;141:504–511. doi: 10.1111/j.1365-2141.2008.07072.x. [DOI] [PubMed] [Google Scholar]
- 21.Chaligné R, Tonetti C, Besancenot R, Marty C, Kiladjian JJ, Socié G, Bordessoule D, Vainchenker W, Giraudier S MPD FIf. SOCS3 inhibits TPO-stimulated, but not spontaneous, megakaryocytic growth in primary myelofibrosis. Leukemia. 2009;23:1186–1190. doi: 10.1038/leu.2009.22. [DOI] [PubMed] [Google Scholar]
- 22.Fourouclas N, Li J, Gilby D, Campbell P, Beer P, Boyd E, Goodeve A, Bareford D, Harrison C, Reilly J, Green A, Bench A. Methylation of the suppressor of cytokine signaling 3 gene (SOCS3) in myeloproliferative disorders. Haematologica. 2008;93:1635–1644. doi: 10.3324/haematol.13043. [DOI] [PubMed] [Google Scholar]
- 23.Jost E, do ON, Dahl E, Maintz CE, Jousten P, Habets L, Wilop S, Herman JG, Osieka R, Galm O. Epigenetic alterations complement mutation of JAK2 tyrosine kinase in patients with BCR/ABL-negative myeloproliferative disorders. Leukemia. 2007;21:505–510. doi: 10.1038/sj.leu.2404513. [DOI] [PubMed] [Google Scholar]
- 24.Teofili L, Martini M, Cenci T, Guidi F, Torti L, Giona F, Foa R, Leone G, Larocca LM. Epigenetic alteration of SOCS family members is a possible pathogenetic mechanism in JAK2 wild type myeloproliferative diseases. Int J Cancer. 2008;123:1586–1592. doi: 10.1002/ijc.23694. [DOI] [PubMed] [Google Scholar]
- 25.Bock O, Hussein K, Brakensiek K, Buhr T, Schlue J, Wiese B, Kreipe H. The suppressor of cytokine signalling-1 (SOCS-1) gene is overexpressed in Philadelphia chromosome negative chronic myeloproliferative disorders. Leukemia research. 2007;31:799–803. doi: 10.1016/j.leukres.2006.08.022. [DOI] [PubMed] [Google Scholar]
- 26.Kralovics R, Teo SS, Buser AS, Brutsche M, Tiedt R, Tichelli A, Passamonti F, Pietra D, Cazzola M, Skoda RC. Altered gene expression in myeloproliferative disorders correlates with activation of signaling by the V617F mutation of Jak2. Blood. 2005;106:3374–3376. doi: 10.1182/blood-2005-05-1889. [DOI] [PubMed] [Google Scholar]
- 27.Haan S, Wüller S, Kaczor J, Rolvering C, Nöcker T, Behrmann I, Haan C. SOCS-mediated downregulation of mutant Jak2 (V617F, T875N and K539L) counteracts cytokine-independent signaling. Oncogene. 2009;28:3069–3080. doi: 10.1038/onc.2009.155. [DOI] [PubMed] [Google Scholar]
- 28.Elliott J, Suessmuth Y, Scott LM, Nahlik K, McMullin MF, Constantinescu SN, Green AR, Johnston JA. SOCS3 tyrosine phosphorylation as a potential bio-marker for myeloproliferative neoplasms associated with mutant JAK2 kinases. Haematologica. 2009;94:576–580. doi: 10.3324/haematol.2008.002352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Hookham MB, Elliott J, Suessmuth Y, Staerk J, Ward AC, Vainchenker W, Percy MJ, McMullin MF, Constantinescu SN, Johnston JA. The myeloproliferative disorder-associated JAK2 V617F mutant escapes negative regulation by suppressor of cytokine signaling 3. Blood. 2007;109:4924–4929. doi: 10.1182/blood-2006-08-039735. [DOI] [PubMed] [Google Scholar]
- 30.Kershaw NJ, Murphy JM, Liau NP, Varghese LN, Laktyushin A, Whitlock EL, Lucet IS, Nicola NA, Babon JJ. SOCS3 binds specific receptor-JAK complexes to control cytokine signaling by direct kinase inhibition. Nature structural & molecular biology. 2013;20:469–476. doi: 10.1038/nsmb.2519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Babon JJ, Murphy JM. In vitro JAK kinase activity and inhibition assays. Methods Mol Biol. 2013;967:39–55. doi: 10.1007/978-1-62703-242-1_3. [DOI] [PubMed] [Google Scholar]
- 32.Murphy JM, Zhang Q, Young SN, Reese ML, Bailey FP, Eyers PA, Ungureanu D, Hammaren H, Silvennoinen O, Varghese LN, Chen K, Tripaydonis A, Jura N, Fukuda K, Qin J, Nimchuk Z, Mudgett MB, Elowe S, Gee CL, Liu L, Daly RJ, Manning G, Babon JJ, Lucet IS. A robust methodology to subclassify pseudokinases based on their nucleotide binding properties. Biochemical Journal. 2013;457:323–334. doi: 10.1042/BJ20131174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Babon JJ, Yao S, DeSouza DP, Harrison CF, Fabri LJ, Liepinsh E, Scrofani SD, Baca M, Norton RS. Secondary structure assignment of mouse SOCS3 by NMR defines the domain boundaries and identifies an unstructured insertion in the SH2 domain. FEBS J. 2005;272:6120–6130. doi: 10.1111/j.1742-4658.2005.05010.x. [DOI] [PubMed] [Google Scholar]
- 34.Babon JJ, Kershaw NJ, Murphy JM, Varghese LN, Laktyushin A, Young SN, Lucet IS, Norton RS, Nicola NA. Suppression of cytokine signaling by SOCS3: characterization of the mode of inhibition and the basis of its specificity. Immunity. 2012;36:239–250. doi: 10.1016/j.immuni.2011.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Babon JJ, Sabo JK, Soetopo A, Yao S, Bailey MF, Zhang JG, Nicola NA, Norton RS. The SOCS box domain of SOCS3: structure and interaction with the elonginBC-cullin5 ubiquitin ligase. J Mol Biol. 2008;381:928–940. doi: 10.1016/j.jmb.2008.06.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Saharinen P, Vihinen M, Silvennoinen O. Autoinhibition of Jak2 tyrosine kinase is dependent on specific regions in its pseudokinase domain. Mol Biol Cel. 2003;14:1448–1459. doi: 10.1091/mbc.E02-06-0342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kirby NM, Mudie ST, Hawley AM, Cookson DJ, Mertens HDT, Cowieson N, Samardzic-Boban V. A low-background-intensity focusing small-angle X-ray scattering undulator beamline. J Appl Cryst. 46:1670–1680. [Google Scholar]
- 38.Murphy JM, Czabotar PE, Hildebrand JM, Lucet IS, Zhang JG, Alvarez-Diaz S, Lewis R, Lalaoui N, Metcalf D, Webb AI, Young SN, Varghese LN, Tannahill GM, Hatchell EC, Majewski IJ, Okamoto T, Dobson RCJ, Hilton DJ, Babon JJ, Nicola NA, Strasser A, Silke J, Alexander WS. The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity. 2013;39:443–453. doi: 10.1016/j.immuni.2013.06.018. [DOI] [PubMed] [Google Scholar]
- 39.Murphy JM, Lucet IS, Hildebrand JM, Tanzer MC, Young SN, Sharma P, Lessene G, Alexander WS, Babon JJ, Silke J, Czabotar PE. Insights into the evolution of divergent nucleotide-binding mechanisms among pseudokinases revealed by crystal structures of human and mouse MLKL. Biochemical Journal. 2014;457:369–377. doi: 10.1042/BJ20131270. [DOI] [PubMed] [Google Scholar]
- 40.Konarev PV, Petoukhov MV, Volkov VV, Svergun DI. ATSAS 2.1, a program package for small-angle scattering data analysis. Journal of Applied Crystallography. 2006;39:277–286. doi: 10.1107/S0021889812007662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Konarev PV, Volkov VV, Sokolova AV, Koch MHJ, Svergun DI. PRIMUS: a Windows PC-based system for small-angle scattering data analysis. Journal of Applied Crystallography. 2003;36:1277–1282. [Google Scholar]
- 42.Svergun DI. Determination of the regularization parameter in indirect-transform methods using perceptual criteria. J Appl Cryst. 1992;25:495–503. [Google Scholar]
- 43.Petoukhov MV, Svergun DI. Global rigid body modeling of macromolecular complexes against small-angle scattering data. Biophys J. 2005;89:1237–1250. doi: 10.1529/biophysj.105.064154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lu X, Huang LJ, Lodish HF. Dimerization by a cytokine receptor is necessary for constitutive activation of JAK2V617F. The Journal of biological chemistry. 2008;283:5258–5266. doi: 10.1074/jbc.M707125200. [DOI] [PubMed] [Google Scholar]
- 45.Pelletier S, Gingras S, Funakoshi-Tago M, Howell S, Ihle JN. Two domains of the erythropoietin receptor are sufficient for Jak2 binding/activation and function. Molecular and cellular biology. 2006;26:8527–8538. doi: 10.1128/MCB.01035-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lu X, Levine R, Tong W, Wernig G, Pikman Y, Zarnegar S, Gilliland DG, Lodish H. Expression of a homodimeric type I cytokine receptor is required for JAK2V617F-mediated transformation. Proc Natl Acad Sci USA. 2005;102:18962–18967. doi: 10.1073/pnas.0509714102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Argetsinger LS, Kouadio JL, Steen H, Stensballe A, Jensen ON, Carter-Su C. Autophosphorylation of JAK2 on tyrosines 221 and 570 regulates its activity. Molecular and cellular biology. 2004;24:4955–4967. doi: 10.1128/MCB.24.11.4955-4967.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Feener EP, Rosario F, Dunn SL, Stancheva Z, Myers MG. Tyrosine phosphorylation of Jak2 in the JH2 domain inhibits cytokine signaling. Molecular and cellular biology. 2004;24:4968–4978. doi: 10.1128/MCB.24.11.4968-4978.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kershaw NJ, Murphy JM, Lucet IS, Nicola NA, Babon JJ. Regulation of Janus kinases by SOCS proteins. Biochem Soc Trans. 2013;41:1042–1047. doi: 10.1042/BST20130077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Babon JJ, Nicola NA. The biology and mechanism of action of suppressor of cytokine signaling 3. Growth Factors. 2012;30:207–219. doi: 10.3109/08977194.2012.687375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Saharinen P, Takaluoma K, Silvennoinen O. Regulation of the Jak2 tyrosine kinase by its pseudokinase domain. Molecular and cellular biology. 2000;20:3387–3395. doi: 10.1128/mcb.20.10.3387-3395.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Koppikar P, Bhagwat N, Kilpivaara O, Manshouri T, Adli M, Hricik T, Liu F, Saunders LM, Mullally A, Abdel-Wahab O, Leung L, Weinstein A, Marubayashi S, Goel A, Gönen M, Estrov Z, Ebert BL, Chiosis G, Nimer SD, Bernstein BE, Verstovsek S, Levine RL. Heterodimeric JAK-STAT activation as a mechanism of persistence to JAK2 inhibitor therapy. Nature. 2012;489:155–159. doi: 10.1038/nature11303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wernig G, Gonneville JR, Crowley BJ, Rodrigues MS, Reddy MM, Hudon HE, Walz C, Reiter A, Podar K, Royer Y, Constantinescu SN, Tomasson MH, Griffin JD, Gilliland DG, Sattler M. The Jak2V617F oncogene associated with myeloproliferative diseases requires a functional FERM domain for transformation and for expression of the Myc and Pim proto-oncogenes. Blood. 2008;111:3751–3759. doi: 10.1182/blood-2007-07-102186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zhao L, Ma Y, Seemann J, Huang LJ. A regulating role of the JAK2 FERM domain in hyperactivation of JAK2(V617F) The Biochemical journal. 2010;426:91–98. doi: 10.1042/BJ20090615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Gorantla SP, Dechow TN, Grundler R, Illert AL, Zum Buschenfelde CM, Kremer M, Peschel C, Duyster J. Oncogenic JAK2V617F requires an intact SH2-like domain for constitutive activation and induction of a myeloproliferative disease in mice. Blood. 2010;116:4600–4611. doi: 10.1182/blood-2009-07-236133. [DOI] [PubMed] [Google Scholar]
- 56.Lucet IS, Babon JJ, Murphy JM. Techniques to examine nucleotide binding by pseudokinases. Biochem Soc Trans. 2013;41:975–980. doi: 10.1042/BST20130075. [DOI] [PubMed] [Google Scholar]
- 57.Scott LM, Tong W, Levine RL, Scott MA, Beer PA, Stratton MR, Futreal PA, Erber WN, McMullin MF, Harrison CN, Warren AJ, Gilliland DG, Lodish HF, Green AR. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med. 2007;356:459–468. doi: 10.1056/NEJMoa065202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Martinez-Aviles L, Besses C, Alvarez-Larran A, Cervantes F, Hernandez-Boluda JC, Bellosillo B. JAK2 exon 12 mutations in polycythemia vera or idiopathic erythrocytosis. Haematologica. 2007;92:1717–1718. doi: 10.3324/haematol.12011. [DOI] [PubMed] [Google Scholar]
- 59.Lee JW, Kim YG, Soung YH, Han KJ, Kim SY, Rhim HS, Min WS, Nam SW, Park WS, Lee JY, Yoo NJ, Lee SH. The JAK2 V617F mutation in de novo acute myelogenous leukemias. Oncogene. 2006;25:1434–1436. doi: 10.1038/sj.onc.1209163. [DOI] [PubMed] [Google Scholar]
- 60.Kratz CP, Böll S, Kontny U, Schrappe M, Niemeyer CM, Stanulla M. Mutational screen reveals a novel JAK2 mutation, L611S, in a child with acute lymphoblastic leukemia. Leukemia. 2006;20:381–383. doi: 10.1038/sj.leu.2404060. [DOI] [PubMed] [Google Scholar]
- 61.Nussenzveig RH, Burjanivova T, Salama ME, Ogilvie NW, Marcinek J, Plank L, Agarwal AM, Perkins SL, Prchal JT. Detection of JAK2 mutations in paraffin marrow biopsies by high resolution melting analysis: identification of L611S alone and in cis with V617F in polycythemia vera. Leukemia Lymphoma. 2012;53:2479–2486. doi: 10.3109/10428194.2012.697562. [DOI] [PubMed] [Google Scholar]
- 62.Schnittger S, Bacher U, Kern W, Schröder M, Haferlach T, Schoch C. Report on two novel nucleotide exchanges in the JAK2 pseudokinase domain: D620E and E627E. Leukemia. 2006;20:2195–2197. doi: 10.1038/sj.leu.2404325. [DOI] [PubMed] [Google Scholar]
- 63.Grunebach F, Bross-Bach U, Kanz L, Brossart P. Detection of a new JAK2 D620E mutation in addition to V617F in a patient with polycythemia vera. Leukemia. 2006;20:2210–2211. doi: 10.1038/sj.leu.2404419. [DOI] [PubMed] [Google Scholar]
- 64.Bercovich D, Ganmore I, Scott LM, Wainreb G, Birger Y, Elimelech A, Shochat C, Cazzaniga G, Biondi A, Basso G, Cario G, Schrappe M, Stanulla M, Strehl S, Haas OA, Mann G, Binder V, Borkhardt A, Kempski H, Trka J, Bielorei B, Avigad S, Stark B, Smith O, Dastugue N, Bourquin JP, Tal NB, Green AR, Izraeli S. Mutations of JAK2 in acute lymphoblastic leukaemias associated with Down’s syndrome. Lancet. 2008;372:1484–1492. doi: 10.1016/S0140-6736(08)61341-0. [DOI] [PubMed] [Google Scholar]
- 65.Mullighan CG, Zhang J, Harvey RC, Collins-Underwood JR, Schulman BA, Phillips LA, Tasian SK, Loh ML, Su X, Liu W, Devidas M, Atlas SR, Chen IM, Clifford RJ, Gerhard DS, Carroll WL, Reaman GH, Smith M, Downing JR, Hunger SP, Willman CL. JAK mutations in high-risk childhood acute lymphoblastic leukemia. Proc Natl Acad Sci USA. 2009;106:9414–9418. doi: 10.1073/pnas.0811761106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zhang JH, Mullighan CG, Harvey RC, Wu G, Chen X, Edmonson M, Buetow KH, Carroll WL, Chen IM, Devidas M, Gerhard DS, Loh ML, Reaman GH, Relling MV, Camitta BM, Bowman WP, Smith MA, Willman CL, Downing JR, Hunger SP. Key pathways are frequently mutated in high-risk childhood acute lymphoblastic leukemia: a report from the Children’s Oncology Group. Blood. 2011;118:3080–3087. doi: 10.1182/blood-2011-03-341412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kearney L, De Castro DG, Yeung J, Procter J, Horsley SW, Eguchi-Ishimae M, Bateman CM, Anderson K, Chaplin T, Young BD, Harrison CJ, Kempski H, So CWE, Ford AM, Greaves M. Specific JAK2 mutation (JAK2R683) and multiple gene deletions in Down syndrome acute lymphoblastic leukemia. Blood. 2009;113:646–648. doi: 10.1182/blood-2008-08-170928. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.




