Next Article in Journal
Phloridzin Docosahexaenoate Inhibits Spheroid Formation by Breast Cancer Stem Cells and Exhibits Cytotoxic Effects against Paclitaxel-Resistant Triple Negative Breast Cancer Cells
Next Article in Special Issue
Epithelial–Mesenchymal Transition in Acute Leukemias
Previous Article in Journal
Sex Differences in Brain Disorders
Previous Article in Special Issue
MiRNAs in Hematopoiesis and Acute Lymphoblastic Leukemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Structural Analysis of Janus Tyrosine Kinase Variants in Hematological Malignancies: Implications for Drug Development and Opportunities for Novel Therapeutic Strategies

by
Omar J. Rodriguez Moncivais
1,2,
Stephanie A. Chavez
1,2,
Victor H. Estrada Jimenez
1,2,
Shengjie Sun
3,4,
Lin Li
3,4,
Robert A. Kirken
1,2 and
Georgialina Rodriguez
1,2,*
1
Department of Biological Sciences, The University of Texas at El Paso, 500 W. University Ave., El Paso, TX 79902, USA
2
Border Biomedical Research Center, The University of Texas at El Paso, 500 W. University Ave., El Paso, TX 79902, USA
3
Department of Physics, The University of Texas at El Paso, 500 W. University Ave., El Paso, TX 79902, USA
4
Computational Sciences Program, The University of Texas at El Paso, 500 W. University Ave., El Paso, TX 79902, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(19), 14573; https://doi.org/10.3390/ijms241914573
Submission received: 19 August 2023 / Revised: 20 September 2023 / Accepted: 22 September 2023 / Published: 26 September 2023
(This article belongs to the Special Issue Recent Developments and Updates in Acute Lymphoblastic Leukemia)

Abstract

:
Janus tyrosine kinase (JAK) variants are known drivers for hematological disorders. With the full-length structure of mouse JAK1 being recently resolved, new observations on the localization of variants within closed, open, and dimerized JAK structures are possible. Full-length homology models of human wild-type JAK family members were developed using the Glassman et al. reported mouse JAK1 containing the V658F structure as a template. Many mutational sites related to proliferative hematological disorders reside in the JH2 pseudokinase domains facing the region important in dimerization of JAKs in both closed and open states. More than half of all JAK gain of function (GoF) variants are changes in polarity, while only 1.2% are associated with a change in charge. Within a JAK1-JAK3 homodimer model, IFNLR1 (PDB ID7T6F) and the IL-2 common gamma chain subunit (IL2Rγc) were aligned with the respective dimer implementing SWISS-MODEL coupled with ChimeraX. JAK3 variants were observed to encircle the catalytic site of the kinase domain, while mutations in the pseudokinase domain align along the JAK-JAK dimerization axis. FERM domains of JAK1 and JAK3 are identified as a hot spot for hematologic malignancies. Herein, we propose new allosteric surfaces for targeting hyperactive JAK dimers.

1. Introduction

1.1. Molecular Structure of Janus Tyrosine Kinase (JAK)

The JAK family of JAK1, JAK2, JAK3, and TYK2 are critical drivers of hematological development, differentiation, proliferation, and survival by over 40 cytokine [1] and growth factor receptors [2,3,4]. JAKs are classified as a family based on their shared seven JAK homology (JH) domains numbered sequentially from the carboxyl to the amino terminal end (Figure 1A). Sequence similarity (>40%) across seven Janus homology (JH) domains and four functional domains, enzymatic activity, shared targets, and response to cytokines defines these tyrosine kinases as a family. At the amino-terminal (N-terminal) end of JAKs (JH6–JH7) spans an erythrocyte band 4.1, ezrin, radixin, moesin (FERM) domain (Figure 1A) [5] involved in plasma membrane localization and association with cytokine receptors [6,7]. The adjacent Src-homology 2 (SH2) domain encompasses JH3-JH4 [5] of JAKs and controls protein–protein interactions including receptor association via tyrosine phosphorylated residues [8]. Carboxyl-terminal (C-terminal) JH domains harbor an inactive pseudokinase (JH2) and catalytically active kinase domain (JH1) [5]. Segments between functional domains form connective linkers between FERM-SH2 (linker of 9 to 20 amino acids), SH2-JH2 (linker of 40 to 60 amino acids), and JH2-JH1 (linker of 20 to 40 amino acids). Supplementary Table S1 contains details of individual JAK regions.
Tandem pseudokinase and kinase domains within JAKs share conserved N-lobe, C-lobe, and secondary structural motifs (Figure 1B). The central catalytic cleft between the N-lobe and C-lobe is the area where nucleotide (ATP/ADP) binds and phosphotransferase activity occurs. Secondary structures and motifs are highly conserved between JH2 and JH1 domains.

1.2. Signal Transduction and Phosphorylation of Janus Tyrosine Kinases

JAKs associate with Type I (common gamma chain family, common beta chain family, gp130 family, GH, Prl, Epo, TPO, G-CSF, leptin) and Type II (IFN family, IL-10, IL-20) receptors which lack intrinsic tyrosine kinase activity for signal transduction from the extracellular to intracellular space [1,9]. The extracellular ligand binds to homodimeric, heterodimeric, or heterotrimeric receptors, causing aggregation of JAKs which associate with intracellular proline rich and hydrophobic box motifs of the receptor [10,11,12]. JAK dimerization allows for autoactivation through transphosphorylation of tyrosine residues within the activation loop of each JH1 kinase domain. JAK and Signal Transducer and Activator of Transcription (STAT) proteins are primarily regulated by phosphorylation events. Phosphorylation of Y1034 within JAK1 [13], JAK2 Y1007 [14,15], JAK3 Y980 [16], and Y1054 [17] of TYK2 allows for the activation loop to open and promotes phospho-transferase of the gamma-phosphate of ATP to substrate tyrosine residues. Alternatively, phosphorylation of tyrosine residues within the activation loop, i.e., JAK3 Y981, is associated with termination of JAK activity [16]. For a comprehensive list of known JAK post-translational sites, refer to Supplementary Table S2. Activated JAKs are capable of phosphorylating cytokine receptors on immunoreceptor tyrosine-based activation motifs (ITAM) creating docking-sites for the SH2 domain containing proteins such as STATs [18]. STATs themselves become JAK substrates, allowing them to form hetero- or homodimers through their own SH2 domains. Some STAT proteins require serine phosphorylation for full activation and translocation to the nucleus [19] where they regulate transcription of genes related to anti-apoptotic and proliferative functions. Termination of the JAK/STAT signaling cascade involves dephosphorylation/phosphorylation, production of inhibitory proteins, and proteasomal degradation of receptors [20].

1.3. JAK Gain of Function Mutations

Gain of function (GoF) mutations within JAK proteins are associated with overproduction of immature hematological precursor cells leading to neoplasms, acute lymphoblastic leukemia (ALL), and acute myelogenous leukemia (AML). While genetic variants within the four JAK functional domains are associated with malignancies, the most well studied focus on the pseudokinase and kinase domains as these regions are considered “hot spots” for oncogenic mutations [21] GoF mutations within these regions are associated with hyperactive JAKs and JAK/STAT signaling pathways [22]. The most notable JH2 domain genetic alterations include JAK1 V658F and the homologous JAK2 V617F mutation known to cause Polycythemia vera (PV) [23,24]. The JAK pseudokinase homologs JAK2 R683G and JAK1 R724H appear to reside within an important area as more than one alteration of these residues is related to leukemia, R683S/G, and R724H/Q/S. JAK2 R683G has been highly associated with ALL cases related to down syndrome and one analysis showed that from 20 ALL children patients, 10 had this mutation. Comparably, JAK1 R724H has been related with B-ALL conferring IL-3 independence to growth and is found in T-ALL patients [25]. It is widely believed that mutations mapped to the FERM and SH2 domains join the receptor complex leading to JAK activation in the absence of cytokine or growth factors. Variants located within functional domain linkers are also capable of deleterious consequences. For example, JAK3 SH2-JH2 linker M511I, A573V, Q507P, and R657Q are transforming in nature [26]. JAK3 Q507P is associated with constitutive STAT5 activation and leads to T-cell prolymphocytic leukemia [27,28,29].

1.4. Open and Closed JAK Kinase Formations

Incomplete JAK crystal structures [30] have limited the understanding of intramolecular and intermolecular interactions within a full structure required for signal transduction. Much of what is known is based on the partial structures of FERM-SH2, pseudokinase, and kinase domains [30,31,32,33,34,35]. Recently, the full-length structure of mouse JAK1 V658F protein bound to partial Interferon lambda receptor 1 (IFNLR1) was established by cryo-EM [36]. Based on the established Glassman et al. structure, here we modeled the full-length open and closed human structure of JAK1, JAK2, JAK3, and TYK2 using SWISS-MODEL and Chimera-X. Furthermore, partial common gamma chain-containing Box1 and Box2 sequences were aligned within the JAK 3 model to predict receptor association. This study sought to understand the molecular-structural basis of gain-of-function (GoF) variants and hematopoietic neoplasms using the established full-length mouse JAK1 structure [36]. For an in-depth analysis of inflammatory disease and immunity GoF and loss-of-function (LoF), we recommend Ott N, et al. JAKs and STATs from a Clinical Perspective: Loss-of-Function Mutations, Gain-of-Function Mutations, and Their Multidimensional Consequences, J. Clin. Immunol., 2023 [37].

2. Results

2.1. Open Structure of JAK Kinases and Mutational Landscape in Hematological Malignancies

For decades, understanding the molecular mechanism of JAK mutations has been hindered by the lack of a full-length enzymatically active structure represented by the open structure bound to the cytokine receptor. The large size of JAK proteins (120–130 kDa) made previous determination of the complete JAK/receptor complex difficult. Here the wild-type (WT) full-length human JAK1, JAK2, JAK3 and TYK2 forms have been modeled using the Glassman et al. crystal structure of mouse JAK1 bound to the Interferon lambda receptor 1 (IFNLR1). ChimeraX and SWISS-MODEL were used to align individual JAK sequences with PDB 7T6F to develop the open forms of human JAK1 (Figure 2A), JAK2 (Figure 2B), JAK3 (Figure 2C), and TYK2 (Figure 2D). In this state, JAKs are observed to have accessible JH2 and JH1 domains (Figure 2) presumably to allow ATP and substrate interaction.
A mutational map of known hematological malignancy variants (partial summarized list, Table 1; for a full list see Table S3) are depicted in Figure 2. Kinase domain sites are observed to group within the β1, β2, and β3 strands of JH1 and represent 15–30% of JAK variants (Figure 2E). Among the JAK family, JAK2 contains the most linker segment variants. The SH2 domain of all JAKs contains the least amount of reported hematopoietic malignancy mutations within functional domains (Figure 2E). Unsurprisingly, the majority of JAK transforming mutations reside within the JH2 pseudokinase domain among JAKs (Figure 2E). Notably, many JAK1 and JAK2 mutations cluster near the Glassman et al. JAK dimerization region of the JH2 domain (Figure 2A,B). A small portion (2.33%) of JAK2 and JAK3 mutations switch the amino acid charge, either from positive to negative or from negative to positive (Figure 3A). Among all combined Table 1 JAK variants, 44.6% did not change either polarity or charge, while 54.2% were associated with a change in polarity and 1.2% with a change in amino acid charge (Figure 3A). Within the JH2 domains of JAK1-3, more than half of variant changes are of polarity (Figure 3B).

2.2. JAK2 GoF Mutations Cluster to the JH1-JH2 Interface of the Closed Structure

Following cytokine-receptor complex formation, JAKs undergo structural changes from closed to open states to allow for the active site of the kinase domain to bind ATP and transfer gamma-phosphate to the substrate. We examined the process by which mutations could facilitate the enzyme open state in the absence of cytokine-receptor initiation. Among JAK family members, JAK2 is associated with the most hematopoietic malignancy variants (Table S3). For this reason, the mutational landscape of JAK2 in the closed state is presented as a representative of JAK-driven hematopoietic neoplasms (Figure 4). Several mutations are observable in the region where the FERM domain, the SH2-JH2 linker, the pseudokinase domain, and the kinase domain converge. A mutational hot spot is evident in the region where the JH2 domain interacts with the JH1 domain. The SH2-JH2 linker runs through this space. In this closed state, the JAK dimerization region within the JH2 N-lobe forms an interface with the N-lobe of the JH1 kinase domain. The region spanning the JH2 domain (L545 to E627) β1, β2, β3, αC, β4, and β5, and JH1 (L849 to I951), β1-β6, αC, and αD may serve to negatively regulate the JH1 domain by binding directly and maintaining a closed state incapable of phospho-transferase activity. Interestingly, a region where JH1-JH2-FERM domains interact is observable in the closed JAK2 state.

2.3. Landscape of GoF Variants within the JAK1-JAK3 Dimer

A model of the JAK1-JAK3 dimer associated with the Type II cytokine receptor IFNLR1 and Type I cytokine receptor common gamma chain (γc) for Interleukin –2 (IL-2), respectively, (Figure 5A) was created by using SWISS-MODEL and Chimera-X based on Glassman et al. (PDB ID 7T6F). Thirteen JAK1 mutations are found in highly dynamic loops, while twelve are in well-defined secondary structures. Comparably, nineteen JAK3 variants are found in loops and twelve are in well-defined secondary structures depicted in Figure 5A. Observably, three different regions of hematopoietic proliferative mutations are recognizable: (1) FERM, (2) pseudokinase, and (3) kinase.
Variants within the FERM domain of JAK1 and JAK3 are spread across the surface and do not immediately appear to cluster. Within the Interferon lambda receptor 1 (IFNLR1) and JAK1 (PDB ID 7T6F) structure, the JAK1 I62 and S71 sites of mutation contact the receptor (Figure 5B). Additionally, the SH2 sites T478 and S512 also contact IFNLR1 near Box2 (Figure 3B). JAK1 S512 points directly toward Box2 of the IFNLR1. Presumably, these sites could impact interaction with the receptor. Similarly, alignment of the intracellular region of the IL2Rγc with JAK3 (Figure 5C) shows that the Box 1 motif of the receptor contacts JAK3 FERM residues P151 and L156, and these are proximal to R279 and Q283. JAK3 mutational sites G62, P132, and R172 of the FERM domain and SH2 R403 are near IL2Rγ Box 2. These residues lie within the same region of E183, which also does not make contact with the intracellular receptor.
As noted previously, the majority of JAK hyperactivating mutations are located in the pseudokinase domain (Figure 3). These sites lay in the same plane (Figure 5D), including some reported in the SH2-JH2 linker around the nucleotide binding motif (HRD/HRN) and the interaction interface between JAKs. This observation suggests that this region could be important in transactivation of JAKs, possibly by acting as a scaffold which permits activation of other proteins. Moreover, this could enhance the open structure and dimerization.
Lastly, JAK3 kinase domain variants encircle the enzyme active site (Figure 5E). Based on this observation, a possible mechanism of transformation may relate to ATP binding and stabilizing the phosphotransferase process.
An overlay of full-length human JAK1 and JAK3 shows a clustering of FERM-SH2 domain hematopoietic neoplasm variants along the surface pointing away from the center of the JAK dimer (Figure 6). This hot spot was not immediately observable above in Figure 5 for either independent JAK. Hot-spot residues are not near the predicted cytokine receptor alignment which lays at the center of the receptor-JAK dimer complex (Figure 5). The method of oncogenesis here is not apparent and illustrates the involvement of a multitude of mechanisms in hyperactivity.

2.4. PDBe Proteins, Interfaces, Structures, and Assemblies Analysis of the JAK1-JAK3 Dimer

Analysis of the heterodimer interfaces using the PDBe PISA tool (Proteins, Interfaces, Structures and Assemblies) was performed using default parameters (Figure 7). Two surface areas of interaction are noticeable within the PDBe PISA structure: (1) within the JAK FERM domain between JAK1 E355 and JAK3 L114 and (2) numerous JAK JH2 points. Twenty-four JAK1 and seventeen JAK3 amino acids are indicated to be dimer interface residues (Table 2). The number of residues predicted to be present within the interface regions constitute a small fraction, 1.6% and 2.1%, respectively, of the total monomer residues (Table 2). JAK1 interface residues include E355, M570, S571, Q572, L573, S574, F575, D576, R577, L580, D608, R629, D630, S632, L633, F636, E637, R643, V656, V658, R659, D660, V661, and N663. JAK3 interface residues include L114, L508, S509, Q510, M511, T512, F513, H514, L515, L563, N564, M566, E567, L570, E571, M592, and G594. JAK residues in bold font are known hematological malignancy variants. The following JAK1 interface residues are positioned proximal (<5 amino acids) to a reported hematological variant: E355 (R360), R629/D630 (I631), L633 (A634), and R643 (S646). Similarly, JAK3 residues include L508 (Q507), E571 (A572/A573), M592 (A593), and G594 (A573). Hematological variants in the latter and former lists are indicated by bold font.
In addition to information regarding accessible/buried protein surface areas and regions of interaction, the tool predicts the presence or absence of hydrogen bonds, salt bridges, and disulfide bonds (Table 2). The complexation significance score (CCS) determined under these conditions to be zero (CCS = 0) indicates that the interface interactions do not form a stable dimer complex. This is supported by the slight change observed in the complex solvation energy (Table 2). Within the dimerization interface, salt-bridges are predicted to form between JAK1 E637 and JAK3 L515 as well as R577 of JAK1 and JAK3 E571 (Table 2). Ten potential hydrogen bonds (<4 Å) are predicted to form between JAK1-JAK3 (Table 2).

3. Discussion

An abundance of variants within the JAK family are associated with hematological malignancies including T-ALL, B-ALL, ETP-ALL, AML, MPNs, PV, NKTCL, and AMKL (Table 1) and serve to emphasize their critical roles in immune development and homeostasis. With the establishment of a full-length JAK1 V658F (130 kDa) bound to partial IFNLR1 it is possible to re-examine the location of hematological malignancy driving JAK variants. Here the open-state full-length human WT JAK models were created using the mouse Glassman et al. structure (Figure 2). Variants were observed to cluster within the JH2 N-lobe described to be the region of JAK dimerization. Notably, the majority of variants are found in this area across JAK1-3 (Figure 2). Conceivably, alterations in this region might promote dimerization and cytokine-independent signal transduction. TYK2 is associated with the fewest hematological neoplasm variants overall and is more commonly associated with inflammatory disorders.
SWISS-MODEL was also used in this study to create the JAK1-JAK3 heterodimer structure (Figure 5). An alternative approach would be to employ the Alphafold2 Multimer V3 tool [38] in combination with PDB 7T6F and template mode. Comparison of the WT JAK heterodimer using these two methods showed minor differences within loops and the position/orientation of variants between the modeled FERM and JH2 domains (Figure S1). The most drastic difference between the two models occurred with the orientation of the JAK3 kinase domain (Figure S1A,B). This is unsurprising given that the PDB 7T6F template used with Alphafold2 is a JAK1 V658F homodimer. As a result, JAK3 in the Alphafold2 model looks to be in a transactivation position, while in the SWISS-MODEL structure (Figure 5), JAK3 more closely resembles JAK1 PDB 8EWY (Figure S1C). Further experimental approaches are needed to draw solid conclusions.
Adopting the SWISS-MODEL structures, many of the JAK1 and JAK3 leukemia-driving variants are located within dynamic regions including small loops (Figure 2). More than a third of JAK mutations analyzed here are located in the pseudokinase domain (Figure 3). This area affects the interface with dimerized JAK molecules, as most of the mutations of JAK3 are in the pseudokinase domain. Hyperactivating JH2 mutations may maintain the JAK-JAK interface as opposed to blocking the active site or enhancing the interaction with the cytoplasmic domain of receptors. This idea is supported by the fact that pseudokinase domains can regulate partnering JAKs in trans as opposed to solely through cis regulation as has been reported for JAK1 GoF mutations [39,40].
The closed state of JAKs was modeled using the Glassman autoinhibited model of JAK1 [36] as a template. In this configuration, most variants lie between the JH2-JH1 interface where they likely destabilize the inhibitory interface as previously suggested [4,30]. Noticeably, a region where JH1-JH2-FERM domains interact is observable in the closed JAK state (Figure 4). In this formation, JAK2 JH2 N-lobe mutation sites I682 and R683 contact the JH1 N-lobe mutational residues D873 and T875. These interactions do not occur in the open state of JAK2. We hypothesize that mutations in this region help to keep the structure open, enhancing the process of dimerization and maintaining activated JAK-STAT signaling. Experimental studies are needed to support the observations made here. Past TYK2 pseudokinase and kinase domain modeling of the closed state [30] similarly showed the majority of reported leukemia mutations face the JH1-JH2 interface.
Like previous reports based on partial JAK structures [30,41,42,43,44], we observed many transforming mutations reside within the JH2 pseudokinase domain (Figure 3). Surprisingly, less than 3% of JAK1-3 variants are related to a change in side-chain charge and less than 50% of sites experience no change in polarity or charge, while greater than 50% of variants mentioned in Table 1 change polarity of the site (Figure 3A). An overwhelming number of JAK1-3 pseudokinase mutations are a change in polarity or involve no change in polarity/charge (Figure 3B). Of note, many of the JAK1 and JAK2 mutations cluster near the dimerization region of the Glassman et al. structure and nucleotide binding site. In fact, the binding of nucleotide to the catalytically inactive JH2 is involved in oncogenesis of JAK3 mutants [45]. These mutations are in highly dynamic structures such as loops that could be difficult to target with non-covalent or even covalent inhibitors since there is no observable characteristic pocket. This could explain why even when blocked the active site serves as a scaffold for other JAKs. Most mutations are present in the loop between the β3 and the αC of the pseudokinase domain. Since these regions are highly dynamic, minor changes could affect the interaction and transactivation process between partnering JAK homo- and heterodimers. Moreover, these areas may be more susceptible to post-translation modifications and/or mutations. Their presence within the JH2 β3 and αC loop suggests the importance of these secondary structures in controlling the JAK1 function. In fact, JAK1 JH2 mutations make contact with JAK3 once they are activated (Figure 5), suggesting that hyperactivating JH2 mutations maintain the JAK1-JAK3 dimer. Analysis of protein interactions, buried/accessible dimer surfaces, and complementary binding forces established by PDBe PISA revealed a small fraction of soluble surface residues within the dimerization interface of the JAK1-JAK3 complex (Figure 7, Table 2). A number of these residues are positioned near or are known hematological malignancy JAK variants. The PDBe PISA CCS = 0 calculation and slight change in solvation energy are not surprising given that the process of transactivation is highly dynamic and occurs rapidly. However, the blockade of this region with targeted inhibitors may be useful in disrupting ligand-independent dimerization and signal transduction.
Cumulatively, the placement of JAK mutations continues to support previous explanations of the mechanism of negative regulation by the pseudokinase domain [30,46]. Variants near the JH2 catalytic cleft potentially modulate kinase activity through the nucleotide-binding site, which has been shown to stabilize and modulate activity of JAK1, JAK2, and TYK2 [35,40,47].
In comparison, kinase domain mutations are fewer in number and are closely located to the kinase active site. JAK1 mutations R879S/C/H, T901G, and K908T reside within β1, a loop between β2 and β3, and β3, respectively. It has been established that the kinase-dead JH1 mutation K908A confers a catalytically inactive enzyme that reduces phosphorylation of downstream signaling factors including STATs [39,48]. Unexplainably, one study found that JAK1 K908A in combination with the JH2 mutation S703I rescues enzyme activity and increases STAT phosphorylation in the presence of JAK2 or TYK2 [39]. The kinase domain sites are observed to group within β1–β3 sheets which could be the area of substrate association implicating yet another mechanism that causes cytokine-independent signal transduction [49,50].
JAK FERM domains house the third highest group of mutational sites. Upon first examination, there is no observable clustering of mutations within individual JAKs (Figure 2); however, an overlay of JAK1 and JAK3 reveals that variants point outward away from intracellular portions of the cytokine receptor (Figure 6). It is widely believed that mutations mapped to the FERM and SH2 domains join the receptor complex leading to JAK activation in the absence of cytokine or growth factors; however, hematological malignancy mutations plotted here are not near the aligned cytokine receptors IFNR1 or IL-2Rγc. The FERM mutational surface (Figure 6) should be considered for allosteric drug targeting as this area can clearly regulate hyper-enzymatic activity.
Variants within linker segments indicate that these regions are capable of allosterically regulating enzymatic activity. JAK3, expressed primarily in hematopoietic cells, contains transforming mutations in the SH2-JH2 linker region (Q507P, M511I, M592V/T, A593V) and one mutation in the JH2-JH1 linker segment (V765D). M511I and A593V cause T-ALL and are sensitive to the JAK inhibitors tofacitinib and ruxolitinib [26,51].
Challenges and limitations of drug development against JAKs include the use of ATP-binding pocket competitive inhibitors, which often result in drug insensitivity and harsh side-effects during treatment. Alternative allosteric inhibitors which target regions distal from the active site represent practical options for obtaining JAK selectivity and avoiding secondary side-effects. The FDA has approved the use of allosteric JAK inhibitors for the treatment of immunological disorders such as SOTYKTU (Bristol-Meyers) for moderate to severe plaque psoriasis. Similarly, new therapeutic strategies should consider blocking the JH2 dimerization region to prevent hyperactive JAK transactivation events. For example, the JAK1 pseudokinase domain mutation V666G showed diminished in vitro activity and reduced activation of JAK3 by transactivation, suggesting that this area may be targetable for hyperactive JAK signaling. In contrast, the new JAK1 inhibitor VVD-118313 allosterically binds C817 of the pseudokinase domain. Residue C817 is distal from the JH2 JAK dimerization region. This treatment partially blocks IL-2 activated STAT5 phosphorylation in immune cells [52]. Because numerous JAK1 and JAK3 variants associated with hematological malignancies are located within dynamic structural loops (Figure 2), it is conceivable that these regions may be more susceptible to allosteric drug-targeting.
In 2009, the first JAK inhibitor (JAKi), ruxolitinib, was approved by the FDA for treatment of hematological malignancy myelofibrosis (MF). Since then, JAKis such as ruxolitinib, fedratinib, and pacritinib are FDA-approved drugs for the treatment of intermediate- to high-risk myelofibrosis (MF), a rare form of chronic leukemia [53,54,55]. Ruxolitinib is a JAK1/2 kinase ATP-competitive binding inhibitor associated with grade 3–4 anemia in 45.2% of recipients, thrombocytopenia in 12.9%, and neutropenia in 7.1% [56]. The most recent JAK inhibitor for the treatment of MF is pacritinib, a multikinase inhibitor effecting JAK2/FLT3/IRAK1/CSF1R. Disappointingly, the discussed JAKis are associated with adverse events including cyopenias, disease progression, and/or low therapeutic effects [57]. Clearly, a need exists for less toxic, more specific, small molecule inhibitors of JAK kinases. Ideally, therapeutic JAK inhibitors would target a mutated protein, leaving wild-type kinases in unrelated tissues unaffected. JAK inhibitor development should consider regions involved in cytokine receptor binding, JAK dimerization, and the primary mutation causing hyperactivity to personalize and provide meaningful treatment.
Limitations of this structural analysis include the lack of consideration for tyrosine phosphorylation and the impact of post-translation modifications on structure and function. Experimental studies are needed to support the observations made.

4. Materials and Methods

4.1. Human Full-Length JAK Modeling

UniProtKD sequences for human JAK1 (P23458), human JAK2 (O60674), human JAK3 (P52333), and human TYK2 (P29597) were used for full-length JAK modeling with SWISS-MODEL (Swiss Institute of Bioinformatics, Basel, Switzerland) [58,59,60,61] and UCSF ChimeraX (Resource for Biocomputing, Visualization, and Informatics, San Francisco, CA, USA) [62,63] as follows. Wild-type human JAK1 open and closed structures were developed by aligning the crystallographic structure of mouse JAK1 dimer with IFNLR1 (PDB ID: 7T6F, [36]) and human TYK2 domains (PDB ID: 4OLI, [30]) as templates, respectively. SWISS-MODEL was used to diminish clashes and minimize the energy for the final structure of JAK1, and this model was used as a template to generate JAK2 open and closed structures and JAK3 and TYK2 open structures. The JAK3-JAK1 dimer was generated using as a template the mouse JAK1 dimer with IFNLR1 (PDB ID: 7T6F, [36]). Since JAK3 interacts with IL2Rγ, the intracellular region containing Box1 and Box2 domains was generated by using SWISS-MODEL. The resulting structure was collocated using UCSF ChimeraX. Using MatchMaker resulted in a sequence alignment score of 35.1. Molecular analysis and localization of JAK sites were performed with UCSF ChimeraX.

4.2. PDBe PISA Tool

UniProt entries for JAK3 (P23458) and JAK1 (P52333) were used to analyze a WT-WT heterodimer with the PDBe PISA tool at https://www.ebi.ac.uk/pdbe/pisa/ (accessed on 18 August 2023) (EMBL’s European Bioinformatics Institute, Hinxton, Cambridgeshire, UK) [64]. Accessible and buried protein surface areas, the presence of hydrogen bonds, salt bridges, and disulfide bonds were obtained using default parameters: CRYST1 contains a = b = c = 1.0, alpha = beta = gamma = 90 degrees, space group = P 1.

5. Conclusions

The landscape of JAK GoF variants associated with hematological malignancies was examined within the open and closed structures of JAK family members. Many GoF mutations analyzed were found in dynamic small loops and involve a change in polarity which could make inhibitor design difficult. JH2 domain mutations cluster in the N-lobe and presumably promote dimerization and independence of cytokine-receptor complex formation to become active and maintain activated JAK-STAT signaling. It is tempting to speculate that inhibitors which selectively target mutations in this region could block variant-driven dimerization while leaving a wild-type JAK protein unaffected and responsive to physiological signals. Likewise, clustering of FERM domain GoF mutations distal from cytokine receptor interactions is observable in the overlay of JAK1–JAK3. Further research is needed to address these areas for allosteric inhibitor targeting.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms241914573/s1.

Author Contributions

Conceptualization, O.J.R.M., R.A.K. and G.R.; methodology, O.J.R.M. and S.S.; formal analysis, O.J.R.M., R.A.K. and G.R.; investigation, O.J.R.M., S.A.C., V.H.E.J., S.S. and G.R.; resources, L.L, R.A.K. and G.R.; writing—original draft preparation, O.J.R.M. and G.R.; writing—review and editing, O.J.R.M., R.A.K. and G.R.; supervision, L.L. and G.R. funding acquisition, L.L., R.A.K. and G.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by grants from the National Institute on Minority Health and Health Disparities, a component of the National Institutes of Health (5U54MD007592).

Acknowledgments

This work was supported by the University of Texas at El Paso (UTEP) Border Biomedical Research Center (BBRC) Research Infrastructure Core Bioinformatics and Biostatistics computer lab.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Garrido-Trigo, A.; Salas, A. Molecular Structure and Function of Janus Kinases: Implications for the Development of Inhibitors. J. Crohns Colitis 2020, 14, S713–S724. [Google Scholar] [CrossRef] [PubMed]
  2. Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct. Target. Ther. 2021, 6, 402. [Google Scholar] [CrossRef] [PubMed]
  3. Philips, R.L.; Wang, Y.; Cheon, H.; Kanno, Y.; Gadina, M.; Sartorelli, V.; Horvath, C.M.; Darnell, J.E., Jr.; Stark, G.R.; O’Shea, J.J. The JAK-STAT pathway at 30: Much learned, much more to do. Cell 2022, 185, 3857–3876. [Google Scholar] [CrossRef] [PubMed]
  4. Morris, R.; Kershaw, N.J.; Babon, J.J. The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci. 2018, 27, 1984–2009. [Google Scholar] [CrossRef] [PubMed]
  5. Constantinescu, S.N.; 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. [Google Scholar] [CrossRef] [PubMed]
  6. Usacheva, A.; Kotenko, S.; Witte, M.M.; Colamonici, O.R. Two distinct domains within the N-terminal region of Janus kinase 1 interact with cytokine receptors. J. Immunol. 2002, 169, 1302–1308. [Google Scholar] [CrossRef] [PubMed]
  7. Richter, M.F.; Dumenil, G.; Uze, G.; Fellous, M.; Pellegrini, S. Specific contribution of Tyk2 JH regions to the binding and the expression of the interferon alpha/beta receptor component IFNAR1. J. Biol. Chem. 1998, 273, 24723–24729. [Google Scholar] [CrossRef] [PubMed]
  8. Wallweber, H.J.; Tam, C.; Franke, Y.; Starovasnik, M.A.; Lupardus, P.J. Structural basis of recognition of interferon-alpha receptor by tyrosine kinase 2. Nat. Struct. Mol. Biol. 2014, 21, 443–448. [Google Scholar] [CrossRef]
  9. Gadina, M.; Hilton, D.; Johnston, J.A.; Morinobu, A.; Lighvani, A.; Zhou, Y.J.; Visconti, R.; O’Shea, J.J. Signaling by type I and II cytokine receptors: Ten years after. Curr. Opin. Immunol. 2001, 13, 363–373. [Google Scholar] [CrossRef]
  10. Usacheva, A.; Sandoval, R.; Domanski, P.; Kotenko, S.V.; Nelms, K.; Goldsmith, M.A.; Colamonici, O.R. Contribution of the Box 1 and Box 2 motifs of cytokine receptors to Jak1 association and activation. J. Biol. Chem. 2002, 277, 48220–48226. [Google Scholar] [CrossRef]
  11. Haan, C.; Kreis, S.; Margue, C.; Behrmann, I. Jaks and cytokine receptors--an intimate relationship. Biochem. Pharmacol. 2006, 72, 1538–1546. [Google Scholar] [CrossRef]
  12. Pelletier, S.; Gingras, S.; Funakoshi-Tago, M.; Howell, S.; Ihle, J.N. Two domains of the erythropoietin receptor are sufficient for Jak2 binding/activation and function. Mol. Cell. Biol. 2006, 26, 8527–8538. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, R.; Griffin, P.R.; Small, E.C.; Thompson, J.E. Mechanism of Janus kinase 3-catalyzed phosphorylation of a Janus kinase 1 activation loop peptide. Arch. Biochem. Biophys. 2003, 410, 7–15. [Google Scholar] [CrossRef] [PubMed]
  14. Feng, J.; Witthuhn, B.A.; Matsuda, T.; Kohlhuber, F.; Kerr, I.M.; Ihle, J.N. Activation of Jak2 catalytic activity requires phosphorylation of Y1007 in the kinase activation loop. Mol. Cell. Biol. 1997, 17, 2497–2501. [Google Scholar] [CrossRef] [PubMed]
  15. Chatti, K.; Farrar, W.L.; Duhe, R.J. Tyrosine phosphorylation of the Janus kinase 2 activation loop is essential for a high-activity catalytic state but dispensable for a basal catalytic state. Biochemistry 2004, 43, 4272–4283. [Google Scholar] [CrossRef] [PubMed]
  16. Zhou, Y.J.; Hanson, E.P.; Chen, Y.Q.; Magnuson, K.; Chen, M.; Swann, P.G.; Wange, R.L.; Changelian, P.S.; O’Shea, J.J. Distinct tyrosine phosphorylation sites in JAK3 kinase domain positively and negatively regulate its enzymatic activity. Proc. Natl. Acad. Sci. USA 1997, 94, 13850–13855. [Google Scholar] [CrossRef] [PubMed]
  17. Gauzzi, M.C.; Velazquez, L.; McKendry, R.; Mogensen, K.E.; Fellous, M.; Pellegrini, S. Interferon-alpha-dependent activation of Tyk2 requires phosphorylation of positive regulatory tyrosines by another kinase. J. Biol. Chem. 1996, 271, 20494–20500. [Google Scholar] [CrossRef] [PubMed]
  18. Darnell, J.E., Jr. STATs and gene regulation. Science 1997, 277, 1630–1635. [Google Scholar] [CrossRef]
  19. O’Shea, J.J.; Schwartz, D.M.; Villarino, A.V.; Gadina, M.; McInnes, I.B.; Laurence, A. The JAK-STAT pathway: Impact on human disease and therapeutic intervention. Annu. Rev. Med. 2015, 66, 311–328. [Google Scholar] [CrossRef]
  20. Seif, F.; Khoshmirsafa, M.; Aazami, H.; Mohsenzadegan, M.; Sedighi, G.; Bahar, M. The role of JAK-STAT signaling pathway and its regulators in the fate of T helper cells. Cell Commun. Signal 2017, 15, 23. [Google Scholar] [CrossRef]
  21. Yamaoka, K.; Saharinen, P.; Pesu, M.; Holt, V.E., 3rd; Silvennoinen, O.; O’Shea, J.J. The Janus kinases (Jaks). Genome Biol. 2004, 5, 253. [Google Scholar] [CrossRef] [PubMed]
  22. Raivola, J.; Haikarainen, T.; Abraham, B.G.; Silvennoinen, O. Janus Kinases in Leukemia. Cancers 2021, 13, 800. [Google Scholar] [CrossRef] [PubMed]
  23. Staerk, J.; Kallin, A.; Demoulin, J.B.; Vainchenker, W.; Constantinescu, S.N. JAK1 and Tyk2 activation by the homologous polycythemia vera JAK2 V617F mutation: Cross-talk with IGF1 receptor. J. Biol. Chem. 2005, 280, 41893–41899. [Google Scholar] [CrossRef] [PubMed]
  24. Means, R.T. JAK2 V617F and the evolving paradigm of polycythemia vera. Korean J. Hematol. 2010, 45, 90–94. [Google Scholar] [CrossRef] [PubMed]
  25. Flex, E.; Petrangeli, V.; Stella, L.; Chiaretti, S.; Hornakova, T.; Knoops, L.; Ariola, C.; Fodale, V.; Clappier, E.; Paoloni, F.; et al. Somatically acquired JAK1 mutations in adult acute lymphoblastic leukemia. J. Exp. Med. 2008, 205, 751–758. [Google Scholar] [CrossRef] [PubMed]
  26. Martinez, G.S.; Ross, J.A.; Kirken, R.A. Transforming Mutations of Jak3 (A573V and M511I) Show Differential Sensitivity to Selective Jak3 Inhibitors. Clin. Cancer Drugs 2016, 3, 131–137. [Google Scholar] [CrossRef] [PubMed]
  27. Bergmann, A.K.; Schneppenheim, S.; Seifert, M.; Betts, M.J.; Haake, A.; Lopez, C.; Maria Murga Penas, E.; Vater, I.; Jayne, S.; Dyer, M.J.; et al. Recurrent mutation of JAK3 in T-cell prolymphocytic leukemia. Genes Chromosomes Cancer 2014, 53, 309–316. [Google Scholar] [CrossRef]
  28. Kiel, M.J.; Velusamy, T.; Rolland, D.; Sahasrabuddhe, A.A.; Chung, F.; Bailey, N.G.; Schrader, A.; Li, B.; Li, J.Z.; Ozel, A.B.; et al. Integrated genomic sequencing reveals mutational landscape of T-cell prolymphocytic leukemia. Blood 2014, 124, 1460–1472. [Google Scholar] [CrossRef]
  29. Bellanger, D.; Jacquemin, V.; Chopin, M.; Pierron, G.; Bernard, O.A.; Ghysdael, J.; Stern, M.H. Recurrent JAK1 and JAK3 somatic mutations in T-cell prolymphocytic leukemia. Leukemia 2014, 28, 417–419. [Google Scholar] [CrossRef]
  30. Lupardus, P.J.; Ultsch, M.; Wallweber, H.; Bir Kohli, P.; Johnson, A.R.; Eigenbrot, C. Structure of the pseudokinase-kinase domains from protein kinase TYK2 reveals a mechanism for Janus kinase (JAK) autoinhibition. Proc. Natl. Acad. Sci. USA 2014, 111, 8025–8030. [Google Scholar] [CrossRef]
  31. Boggon, T.J.; Li, Y.; Manley, P.W.; Eck, M.J. Crystal structure of the Jak3 kinase domain in complex with a staurosporine analog. Blood 2005, 106, 996–1002. [Google Scholar] [CrossRef]
  32. Lucet, I.S.; Fantino, E.; Styles, M.; Bamert, R.; Patel, O.; Broughton, S.E.; Walter, M.; Burns, C.J.; Treutlein, H.; Wilks, A.F.; et al. The structural basis of Janus kinase 2 inhibition by a potent and specific pan-Janus kinase inhibitor. Blood 2006, 107, 176–183. [Google Scholar] [CrossRef] [PubMed]
  33. Alicea-Velazquez, N.L.; Jakoncic, J.; Boggon, T.J. Structure-guided studies of the SHP-1/JAK1 interaction provide new insights into phosphatase catalytic domain substrate recognition. J. Struct. Biol. 2013, 181, 243–251. [Google Scholar] [CrossRef] [PubMed]
  34. McNally, R.; Toms, A.V.; Eck, M.J. Crystal Structure of the FERM-SH2 Module of Human Jak2. PLoS ONE 2016, 11, e0156218. [Google Scholar] [CrossRef] [PubMed]
  35. Min, X.; Ungureanu, D.; Maxwell, S.; Hammaren, H.; Thibault, S.; Hillert, E.K.; Ayres, M.; Greenfield, B.; Eksterowicz, J.; Gabel, C.; et al. Structural and Functional Characterization of the JH2 Pseudokinase Domain of JAK Family Tyrosine Kinase 2 (TYK2). J. Biol. Chem. 2015, 290, 27261–27270. [Google Scholar] [CrossRef] [PubMed]
  36. Glassman, C.R.; Tsutsumi, N.; Saxton, R.A.; Lupardus, P.J.; Jude, K.M.; Garcia, K.C. Structure of a Janus kinase cytokine receptor complex reveals the basis for dimeric activation. Science 2022, 376, 163–169. [Google Scholar] [CrossRef]
  37. Ott, N.; Faletti, L.; Heeg, M.; Andreani, V.; Grimbacher, B. JAKs and STATs from a Clinical Perspective: Loss-of-Function Mutations, Gain-of-Function Mutations, and Their Multidimensional Consequences. J. Clin. Immunol. 2023, 43, 1326–1359. [Google Scholar] [CrossRef]
  38. Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Zidek, A.; Potapenko, A.; et al. Highly accurate protein structure prediction with AlphaFold. Nature 2021, 596, 583–589. [Google Scholar] [CrossRef]
  39. Gruber, C.N.; Calis, J.J.A.; Buta, S.; Evrony, G.; Martin, J.C.; Uhl, S.A.; Caron, R.; Jarchin, L.; Dunkin, D.; Phelps, R.; et al. Complex Autoinflammatory Syndrome Unveils Fundamental Principles of JAK1 Kinase Transcriptional and Biochemical Function. Immunity 2020, 53, 672–684. [Google Scholar] [CrossRef]
  40. Raivola, J.; Haikarainen, T.; Silvennoinen, O. Characterization of JAK1 Pseudokinase Domain in Cytokine Signaling. Cancers 2019, 12, 78. [Google Scholar] [CrossRef]
  41. Hamaguchi, H.; Amano, Y.; Moritomo, A.; Shirakami, S.; Nakajima, Y.; Nakai, K.; Nomura, N.; Ito, M.; Higashi, Y.; Inoue, T. Discovery and structural characterization of peficitinib (ASP015K) as a novel and potent JAK inhibitor. Bioorg. Med. Chem. 2018, 26, 4971–4983. [Google Scholar] [CrossRef] [PubMed]
  42. Chrencik, J.E.; Patny, A.; Leung, I.K.; Korniski, B.; Emmons, T.L.; Hall, T.; Weinberg, R.A.; Gormley, J.A.; Williams, J.M.; Day, J.E.; et al. Structural and thermodynamic characterization of the TYK2 and JAK3 kinase domains in complex with CP-690550 and CMP-6. J. Mol. Biol. 2010, 400, 413–433. [Google Scholar] [CrossRef] [PubMed]
  43. Virtanen, A.T.; Haikarainen, T.; Sampathkumar, P.; Palmroth, M.; Liukkonen, S.; Liu, J.; Nekhotiaeva, N.; Hubbard, S.R.; Silvennoinen, O. Identification of Novel Small Molecule Ligands for JAK2 Pseudokinase Domain. Pharmaceuticals 2023, 16, 75. [Google Scholar] [CrossRef]
  44. Zhang, D.; Wlodawer, A.; Lubkowski, J. Crystal Structure of a Complex of the Intracellular Domain of Interferon lambda Receptor 1 (IFNLR1) and the FERM/SH2 Domains of Human JAK1. J. Mol. Biol. 2016, 428, 4651–4668. [Google Scholar] [CrossRef] [PubMed]
  45. Raivola, J.; Hammaren, H.M.; Virtanen, A.T.; Bulleeraz, V.; Ward, A.C.; Silvennoinen, O. Hyperactivation of Oncogenic JAK3 Mutants Depend on ATP Binding to the Pseudokinase Domain. Front. Oncol. 2018, 8, 560. [Google Scholar] [CrossRef]
  46. Saharinen, P.; Silvennoinen, O. The pseudokinase domain is required for suppression of basal activity of Jak2 and Jak3 tyrosine kinases and for cytokine-inducible activation of signal transduction. J. Biol. Chem. 2002, 277, 47954–47963. [Google Scholar] [CrossRef] [PubMed]
  47. Hammaren, H.M.; Ungureanu, D.; Grisouard, J.; Skoda, R.C.; Hubbard, S.R.; Silvennoinen, O. ATP binding to the pseudokinase domain of JAK2 is critical for pathogenic activation. Proc. Natl. Acad. Sci. USA 2015, 112, 4642–4647. [Google Scholar] [CrossRef]
  48. Grant, A.H.; Rodriguez, A.C.; Rodriguez Moncivais, O.J.; Sun, S.; Li, L.; Mohl, J.E.; Leung, M.Y.; Kirken, R.A.; Rodriguez, G. JAK1 Pseudokinase V666G Mutant Dominantly Impairs JAK3 Phosphorylation and IL-2 Signaling. Int. J. Mol. Sci. 2023, 24, 6805. [Google Scholar] [CrossRef]
  49. Yao, H.; Ma, Y.; Hong, Z.; Zhao, L.; Monaghan, S.A.; Hu, M.C.; Huang, L.J. Activating JAK2 mutants reveal cytokine receptor coupling differences that impact outcomes in myeloproliferative neoplasm. Leukemia 2017, 31, 2122–2131. [Google Scholar] [CrossRef]
  50. Pradhan, A.; Lambert, Q.T.; Griner, L.N.; Reuther, G.W. Activation of JAK2-V617F by components of heterodimeric cytokine receptors. J. Biol. Chem. 2010, 285, 16651–16663. [Google Scholar] [CrossRef]
  51. Brothers, J.; Castillo, D.R.; Jeon, W.J.; Joung, B.; Linhares, Y. Partial response to venetoclax and ruxolitinib combination in a case of refractory T-prolymphocytic leukemia. Hematology 2023, 28, 2237342. [Google Scholar] [CrossRef] [PubMed]
  52. Kavanagh, M.E.; Horning, B.D.; Khattri, R.; Roy, N.; Lu, J.P.; Whitby, L.R.; Ye, E.; Brannon, J.C.; Parker, A.; Chick, J.M.; et al. Selective inhibitors of JAK1 targeting an isoform-restricted allosteric cysteine. Nat. Chem. Biol. 2022, 18, 1388–1398. [Google Scholar] [CrossRef]
  53. Mascarenhas, J.; Hoffman, R. Ruxolitinib: The first FDA approved therapy for the treatment of myelofibrosis. Clin. Cancer Res. 2012, 18, 3008–3014. [Google Scholar] [CrossRef] [PubMed]
  54. Saha, C.; Harrison, C. Fedratinib, the first selective JAK2 inhibitor approved for treatment of myelofibrosis—an option beyond ruxolitinib. Expert Rev. Hematol. 2022, 15, 583–595. [Google Scholar] [CrossRef] [PubMed]
  55. Mascarenhas, J. Pacritinib for the treatment of patients with myelofibrosis and thrombocytopenia. Expert Rev. Hematol. 2022, 15, 671–684. [Google Scholar] [CrossRef] [PubMed]
  56. Levavi, H.; Hoffman, R.; Marcellino, B.K. JAK inhibitors in the treatment of myelofibrosis. Clin. Adv. Hematol. Oncol. 2022, 20, 456–467. [Google Scholar] [PubMed]
  57. Verstovsek, S.; Mesa, R.A.; Gotlib, J.; Gupta, V.; DiPersio, J.F.; Catalano, J.V.; Deininger, M.W.; Miller, C.B.; Silver, R.T.; Talpaz, M.; et al. Long-term treatment with ruxolitinib for patients with myelofibrosis: 5-year update from the randomized, double-blind, placebo-controlled, phase 3 COMFORT-I trial. J. Hematol. Oncol. 2017, 10, 55. [Google Scholar] [CrossRef]
  58. Schwede, T.; Kopp, J.; Guex, N.; Peitsch, M.C. SWISS-MODEL: An automated protein homology-modeling server. Nucleic Acids Res. 2003, 31, 3381–3385. [Google Scholar] [CrossRef]
  59. Waterhouse, A.; Bertoni, M.; Bienert, S.; Studer, G.; Tauriello, G.; Gumienny, R.; Heer, F.T.; de Beer, T.A.P.; Rempfer, C.; Bordoli, L.; et al. SWISS-MODEL: Homology modelling of protein structures and complexes. Nucleic Acids Res. 2018, 46, W296–W303. [Google Scholar] [CrossRef]
  60. Bertoni, M.; Kiefer, F.; Biasini, M.; Bordoli, L.; Schwede, T. Modeling protein quaternary structure of homo- and hetero-oligomers beyond binary interactions by homology. Sci. Rep. 2017, 7, 10480. [Google Scholar] [CrossRef]
  61. Guex, N.; Peitsch, M.C.; Schwede, T. Automated comparative protein structure modeling with SWISS-MODEL and Swiss-PdbViewer: A historical perspective. Electrophoresis 2009, 30 (Suppl. S1), S162–S173. [Google Scholar] [CrossRef] [PubMed]
  62. Goddard, T.D.; Huang, C.C.; Meng, E.C.; Pettersen, E.F.; Couch, G.S.; Morris, J.H.; Ferrin, T.E. UCSF ChimeraX: Meeting modern challenges in visualization and analysis. Protein Sci. 2018, 27, 14–25. [Google Scholar] [CrossRef] [PubMed]
  63. Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Meng, E.C.; Couch, G.S.; Croll, T.I.; Morris, J.H.; Ferrin, T.E. UCSF ChimeraX: Structure visualization for researchers, educators, and developers. Protein Sci. 2021, 30, 70–82. [Google Scholar] [CrossRef] [PubMed]
  64. Krissinel, E.; Henrick, K. Inference of macromolecular assemblies from crystalline state. J. Mol. Biol. 2007, 372, 774–797. [Google Scholar] [CrossRef]
Figure 1. Linear structure of seven Janus homology (JH) domains and four functional domains of JAK family members. (A) JAK amino-terminal (NT) Janus homology 7 (JH7) domain and JH6 domain form the band 4.1, ezrin, radixin, moesin (FERM) segment; followed by a Src-homology 2 (SH2) consisting of the JH4 and JH3 domains; the JH2 pseudokinase domain and carboxyl-terminal (CT) JH1 kinase domain. JH1 activation loop regulatory tyrosine residues are depicted as Y. Created with BioRender.com. (B) Representative of JAK1 JH1 and JH2 ribbon structure depicting secondary α-helices and β-sheets within the N-lobe and C-lobes.
Figure 1. Linear structure of seven Janus homology (JH) domains and four functional domains of JAK family members. (A) JAK amino-terminal (NT) Janus homology 7 (JH7) domain and JH6 domain form the band 4.1, ezrin, radixin, moesin (FERM) segment; followed by a Src-homology 2 (SH2) consisting of the JH4 and JH3 domains; the JH2 pseudokinase domain and carboxyl-terminal (CT) JH1 kinase domain. JH1 activation loop regulatory tyrosine residues are depicted as Y. Created with BioRender.com. (B) Representative of JAK1 JH1 and JH2 ribbon structure depicting secondary α-helices and β-sheets within the N-lobe and C-lobes.
Ijms 24 14573 g001
Figure 2. Mutational map of hematopoietic malignancy driving JAK variants. FERM domain (light blue), FERM-SH2 linker (yellow), SH2 domain (pink), SH2-JH2 linker (dark blue), pseudokinase domain (grey), JH2-JH1 linker (beige), and kinase domain (light green). The side chain of the residues associated with hematological mutations are depicted as red spheres. Human JAK1 (A), JAK2 (B), JAK3 (C), and TYK2 (D). (E) Bar-graph representing percentage of hematopoietic malignancy JAK mutations (from Table 1) within individual functional domains and linker regions (FERM-SH2 linker, SH2-JH2 linker, and JH2-JH1 linker combined).
Figure 2. Mutational map of hematopoietic malignancy driving JAK variants. FERM domain (light blue), FERM-SH2 linker (yellow), SH2 domain (pink), SH2-JH2 linker (dark blue), pseudokinase domain (grey), JH2-JH1 linker (beige), and kinase domain (light green). The side chain of the residues associated with hematological mutations are depicted as red spheres. Human JAK1 (A), JAK2 (B), JAK3 (C), and TYK2 (D). (E) Bar-graph representing percentage of hematopoietic malignancy JAK mutations (from Table 1) within individual functional domains and linker regions (FERM-SH2 linker, SH2-JH2 linker, and JH2-JH1 linker combined).
Ijms 24 14573 g002
Figure 3. Nature of JAK amino acid variant change. (A) The nature of change for driver mutations was quantified across the full polypeptide sequence or (B) the pseudokinase domain alone across JAK family members. The bar labeled JAKs represents the compiled figures as a family. Change of polarity (green), charge (orange), and no change (blue) of individual residues were considered. Graphic based on macros-supported data analysis of compiled mutations from Table 1. Amino acids G, A, V, L, I, P, M, F, Y, and W were classified as non-polar; S, T, C, N, and Q as polar; K, H, and R as positively charged; and D and E as negatively charged.
Figure 3. Nature of JAK amino acid variant change. (A) The nature of change for driver mutations was quantified across the full polypeptide sequence or (B) the pseudokinase domain alone across JAK family members. The bar labeled JAKs represents the compiled figures as a family. Change of polarity (green), charge (orange), and no change (blue) of individual residues were considered. Graphic based on macros-supported data analysis of compiled mutations from Table 1. Amino acids G, A, V, L, I, P, M, F, Y, and W were classified as non-polar; S, T, C, N, and Q as polar; K, H, and R as positively charged; and D and E as negatively charged.
Ijms 24 14573 g003
Figure 4. JAK2 closed structure GoF mutations associated with hemopoietic malignancies. Inactive closed-structure of JAK2 FERM domain (light blue), FERM-SH2 linker (yellow), SH2 domain (pink), SH2-JH2 linker (dark blue), JH2 domain (gray), JH2-JH1 linker (beige), and JH1 domain (light green). (A) Side chain of residues-reported mutations as drivers of enzymatic hyperactivity are shown in red spheres. JAK2 hyperactivating FERM mutations: E61K, T108A, E177V, G276A, R340Q, L393V; SH2 mutations: T514M, N533I/Y, M535I, K539L, I540T, 538–547 indels; pseudokinase mutations: F556L, R564Q, V567A, H587N, S591L, H606Q, K607N, H608Y, L611S, V617F, C618R, D620E, L624P, I645V, I682F, R683S/G, S755R, Y813D, E846D; kinase mutations: R867Q, D873N, T875N, P933R, R938Q, R1063H, N1108S are shown. (B) Closed structure was rotated 180° along the vertical axis to show the back side of JAK2.
Figure 4. JAK2 closed structure GoF mutations associated with hemopoietic malignancies. Inactive closed-structure of JAK2 FERM domain (light blue), FERM-SH2 linker (yellow), SH2 domain (pink), SH2-JH2 linker (dark blue), JH2 domain (gray), JH2-JH1 linker (beige), and JH1 domain (light green). (A) Side chain of residues-reported mutations as drivers of enzymatic hyperactivity are shown in red spheres. JAK2 hyperactivating FERM mutations: E61K, T108A, E177V, G276A, R340Q, L393V; SH2 mutations: T514M, N533I/Y, M535I, K539L, I540T, 538–547 indels; pseudokinase mutations: F556L, R564Q, V567A, H587N, S591L, H606Q, K607N, H608Y, L611S, V617F, C618R, D620E, L624P, I645V, I682F, R683S/G, S755R, Y813D, E846D; kinase mutations: R867Q, D873N, T875N, P933R, R938Q, R1063H, N1108S are shown. (B) Closed structure was rotated 180° along the vertical axis to show the back side of JAK2.
Ijms 24 14573 g004
Figure 5. Landscape of hematopoietic malignancy driving mutations in the JAK1-JAK3 dimer. (A) Dimer of JAK1–JAK3 (left–right, respectively). FERM domain is depicted in light blue, FERM-SH2 linker in yellow, SH2 domain in light pink, SH2-JH2 linker in royal blue, pseudokinase in gray, JH2-JH1 linker in beige, and kinase domains in green. JAK1 mutations are portrayed as red spheres and JAK3 mutations as orange spheres for sites reported in Table 1. JAK1 is bound to partial intracellular IFNLR1 (purple). IFNLR1 Box1 and Box2 motifs depicted in cyan. JAK3 is bound to the IL-2Rγ intracellular region (black). IL-2Rγ Box1 and Box2 motifs are in cyan. Residues composing the JAK1 L624_R629 indel are marked. (B) JAK1 FERM region (light blue) complexed with IFNLR1 (purple) with Box1 and Box2 in cyan. (C) JAK3 FERM region (light blue) showing interaction with IL-2Rγ (black). (D) JAK1 kinase domain portraying mutations from Table 1 as red spheres the sites for mutations. Transversal cut in the region where JAKs interact showing the sites for mutation at this level. (E) JAK3 kinase domain (light green) portraying mutations from Table 1 as orange spheres around the active JH1 domain.
Figure 5. Landscape of hematopoietic malignancy driving mutations in the JAK1-JAK3 dimer. (A) Dimer of JAK1–JAK3 (left–right, respectively). FERM domain is depicted in light blue, FERM-SH2 linker in yellow, SH2 domain in light pink, SH2-JH2 linker in royal blue, pseudokinase in gray, JH2-JH1 linker in beige, and kinase domains in green. JAK1 mutations are portrayed as red spheres and JAK3 mutations as orange spheres for sites reported in Table 1. JAK1 is bound to partial intracellular IFNLR1 (purple). IFNLR1 Box1 and Box2 motifs depicted in cyan. JAK3 is bound to the IL-2Rγ intracellular region (black). IL-2Rγ Box1 and Box2 motifs are in cyan. Residues composing the JAK1 L624_R629 indel are marked. (B) JAK1 FERM region (light blue) complexed with IFNLR1 (purple) with Box1 and Box2 in cyan. (C) JAK3 FERM region (light blue) showing interaction with IL-2Rγ (black). (D) JAK1 kinase domain portraying mutations from Table 1 as red spheres the sites for mutations. Transversal cut in the region where JAKs interact showing the sites for mutation at this level. (E) JAK3 kinase domain (light green) portraying mutations from Table 1 as orange spheres around the active JH1 domain.
Ijms 24 14573 g005
Figure 6. Overlay of JAK1 and JAK3 FERM-SH2 domains reveals hot spot for hematopoietic malignancy driving variants. JAK FERM domain (light blue), SH2 domain (pink), and FERM-SH2 linker (bright yellow) depict the location of JAK1 variants (left, red spheres), JAK3 variants (right, orange spheres), and an overlay of JAK1–JAK3 variants (center). JAK1 I62V (B-ALL; T-ALL), S71C (ETP-ALL), K204M (B-ALL), T478S (AML), and S512L (T-ALL) and JAK3 G62S (AML), P132A(AMKL), R172Q (ATLL), E183G (ATLL), R403H(T-ALL) variants are depicted.
Figure 6. Overlay of JAK1 and JAK3 FERM-SH2 domains reveals hot spot for hematopoietic malignancy driving variants. JAK FERM domain (light blue), SH2 domain (pink), and FERM-SH2 linker (bright yellow) depict the location of JAK1 variants (left, red spheres), JAK3 variants (right, orange spheres), and an overlay of JAK1–JAK3 variants (center). JAK1 I62V (B-ALL; T-ALL), S71C (ETP-ALL), K204M (B-ALL), T478S (AML), and S512L (T-ALL) and JAK3 G62S (AML), P132A(AMKL), R172Q (ATLL), E183G (ATLL), R403H(T-ALL) variants are depicted.
Ijms 24 14573 g006
Figure 7. JAK1-JAK3 PDBe PISA Structure. Full-length wild-type JAK1 (cyan) with contact residues depicted in green and wild-type JAK3 (blue) with contact residues depicted in red. (A) Spacefill surface. (B) Ball-stick heterodimer structure.
Figure 7. JAK1-JAK3 PDBe PISA Structure. Full-length wild-type JAK1 (cyan) with contact residues depicted in green and wild-type JAK3 (blue) with contact residues depicted in red. (A) Spacefill surface. (B) Ball-stick heterodimer structure.
Ijms 24 14573 g007
Table 1. Partial list of Summarized JAK Hematopoietic Malignancy Variants. (For the full list, refer to Table S1). Acute lymphoblastic leukemia (ALL); Early T-cell precursor (ETP); Acute myeloid leukemia (AML); Polycythemia vera (PV); Myeloproliferative neoplasm (MPN); Essential thrombocythaemia (ET); Acute megakaryoblastic leukemia (AMKL); Adult T-cell leukemia/lymphoma (ATLL); T-cell prolymphocytic leukemia (T-PLL); Juvenile myelomonocytic leukemia (JMML); Natural killer/T-cell lymphoma (NKTCL); Idiopathic myelofibrosis (IMF); Down syndrome ALL (DS-ALL).
Table 1. Partial list of Summarized JAK Hematopoietic Malignancy Variants. (For the full list, refer to Table S1). Acute lymphoblastic leukemia (ALL); Early T-cell precursor (ETP); Acute myeloid leukemia (AML); Polycythemia vera (PV); Myeloproliferative neoplasm (MPN); Essential thrombocythaemia (ET); Acute megakaryoblastic leukemia (AMKL); Adult T-cell leukemia/lymphoma (ATLL); T-cell prolymphocytic leukemia (T-PLL); Juvenile myelomonocytic leukemia (JMML); Natural killer/T-cell lymphoma (NKTCL); Idiopathic myelofibrosis (IMF); Down syndrome ALL (DS-ALL).
JAKRegionMutationHematopoietic Malignancy Subtype
JAK1FERMI62VB-ALL; T-ALL
S71CETP-ALL
K204MB-ALL
SH2T478SAML
S512LT-ALL
JH2V658FALL
S703IALL
R724HB-ALL; T-ALL
R724QB-ALL; T-ALL
R724SB-ALL; T-ALL
JH1R879ST-ALL
R879CT-ALL
R879HT-ALL
T901GT-ALL
K908TPediatric T-ALL
K908AInactive JAK1
JAK2FERME61KPutative primary erythrocytosis
T108AFound as germline mutation in V617F-positive PV patient
E177VPutative primary erythrocytosis
G276APutative primary erythrocytosis
R340QPV
FERM-SH2 LinkerL393VFound as germline mutation in V617F-positive PV patient
SH2-JH2
Linker
T514MMPNs
N533IPV (together with K539L)
N533YPV (together with K539L)
M535IAMKL
K539LMPNs
I540TPV
538–547 indelsMPNs
JH2F556LMPNs
R564QHereditary ET
V567AMPNs
H587NMPNs
S591LMPNs
H606QMPNs
K607NAML
H608YMPNs
L611SALL
V617FMPNs
C618RMPNs
D620EMPNs
L624PMPNs
I645VMPNs
I682FALL
R683GMPNs
R683SMPNs
S755RHereditary thrombocythemia (together with R938Q in cis)
JH2-JH1 LinkerY813DIMF
E846DGermline mutation found in erythrocytosis and megakaryocytic atypia
JH1R867QALL, hereditary thrombocythemia
D873NALL
T875NAMKL
P933RALL
R938QHereditary thrombocythemia (together with S755R in cis)
R1063HGermline mutation found in erythrocytosis and megakaryocytic atypia
N1108SPV
JAK3FERMG62SAML
P132AAMKL
R172QATLL
E183GATLL
SH2R403HT-ALL
JH2Q507PT-PLL
M511IT-PLL, AML, JMML, NKTCL
A573VDS-ALL, DS AMKL, NTCL
A593VTransform BaF3 cells
R657QT-PLL, AML, JMML, NKTCL
V765DJMML
Table 2. Summary of wild-type JAK heterodimer PDBe PISA analysis. Predicted accessible surfaces, dimer interface, salt bridge, and hydrogen bond formation between JAK1-JAK3 heterodimer.
Table 2. Summary of wild-type JAK heterodimer PDBe PISA analysis. Predicted accessible surfaces, dimer interface, salt bridge, and hydrogen bond formation between JAK1-JAK3 heterodimer.
MonomerJAK1 JAK3
ClassProtein Protein
Number of residues
Interface242.10%171.60%
Surface111198.90%104598.70%
total1123100.00%1059100.00%
Solvent-accessible area, Å
Interface751.71.20%897.51.50%
Total61,180.1100.00%58,264.5100.00%
Solvation energy, kcal/mol
Isolated structure−1019.9100.00%−942100.00%
Gain on complex formation−4.20.40%−5.60.60%
Average gain−1.50.001−2.50.003
p-value0.166 0.13
Interface Salt bridges
JAK1Dist. [Å]JAK3
1GLU 6372.00LYS 515
2GLU 6373.21LYS 515
3ARG 5772.93GLU 571
Interface Hydrogen Bonds
JAK1Dist. [Å]JAK3
1PHE 5753.15MET 511
2LEU 5733.36PHE 513
3SER 5713.17LYS 515
4ARG 5773.64LEU 508
5ARG 5773.20SER 509
6ARG 5773.77SER 509
7PHE 5753.39MET 511
8LEU 5733.18PHE 513
9ARG 6593.56GLU 567
10ARG 5772.93GLU 571
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rodriguez Moncivais, O.J.; Chavez, S.A.; Estrada Jimenez, V.H.; Sun, S.; Li, L.; Kirken, R.A.; Rodriguez, G. Structural Analysis of Janus Tyrosine Kinase Variants in Hematological Malignancies: Implications for Drug Development and Opportunities for Novel Therapeutic Strategies. Int. J. Mol. Sci. 2023, 24, 14573. https://doi.org/10.3390/ijms241914573

AMA Style

Rodriguez Moncivais OJ, Chavez SA, Estrada Jimenez VH, Sun S, Li L, Kirken RA, Rodriguez G. Structural Analysis of Janus Tyrosine Kinase Variants in Hematological Malignancies: Implications for Drug Development and Opportunities for Novel Therapeutic Strategies. International Journal of Molecular Sciences. 2023; 24(19):14573. https://doi.org/10.3390/ijms241914573

Chicago/Turabian Style

Rodriguez Moncivais, Omar J., Stephanie A. Chavez, Victor H. Estrada Jimenez, Shengjie Sun, Lin Li, Robert A. Kirken, and Georgialina Rodriguez. 2023. "Structural Analysis of Janus Tyrosine Kinase Variants in Hematological Malignancies: Implications for Drug Development and Opportunities for Novel Therapeutic Strategies" International Journal of Molecular Sciences 24, no. 19: 14573. https://doi.org/10.3390/ijms241914573

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop