Molecular Mechanisms of JAK2V617F Oncogenic Activation: The JAK2- V617F Mutant Utilizes the SH2 Domain for Constitutive Kinase Activity in the Presence of Unstimulated Heterodimeric Cytokine Receptor.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 175-175
Author(s):  
Sivahari P Gorantla ◽  
Tobias Dechow ◽  
Rebekka Grundler ◽  
Christian Peschel ◽  
Justus Duyster

Abstract The JAK2-V617F mutation has been reported in the majority of MPDs including PV, ET, and IMF. This mutation leads to the constitutive activation of the JAK2 tyrosine kinase activity and overexpression of JAK2V617F renders hematopoietic cell lines growth factor-independent. However, the molecular mechanism leading to constitutive activation of JAK2V617F is largely unclear and the requirement of homodimeric or heterodimeric cytokine receptors needs to be determined. Here we show that oncogenic JAK2-V617F requires an intact SH2 domain for constitutive kinase activity. To this end we mutated the conserved arginine 426 within the SH2 domain to a lysine. Ba/F3 cells expressing JAK2V617F grew IL-3-independent and showed constitutive activation of JAK2, STAT5, and ERK1/2. In contrast, introduction of the SH2 mutation in JAK2V617F abrogated both transformation as well as constitutive activation of downstream signaling pathways. Accordingly, reconstitution of JAK2 mutants in a JAK2-negative cell line with IL-3R co-expression revealed reduced activation of JAK2 when the SH2 domain was mutated. It has been reported that JAK2 binding to homodimeric type I cytokine receptor may facilitate JAK2V617F-mediated transformation. Interestingly, co-expression of the homodomeric EpoR with SH2 mutated JAK2V617F rescues the phenotype indicating that the SH2 domain is required for JAK2 signaling in the presence of heterodimeric but not homodimeric cytokine receptors. Membrane localization studies showed equal membrane distribution of SH2-mutated and unmutated JAK2-V617F indicating that the SH2 domain mutation does not affect subcellular distribution of JAK2. However, co-IP experiments revealed a possible role for the SH2 domain in the dimerization and transphosphorylation of JAK2. Consequently, reduced transphosphorylation was seen in IL-3R- but not in EpoR-expressing cells. In a BM transplantation model we found that an intact SH2 domain in JAK2V617F was required for the induction of a MPD-like disease. Thus, our results points to an important role of the SH2 domain for the constitutive activation of JAK2V617F in cells expressing heterodimeric cytokine receptors.

Blood ◽  
2010 ◽  
Vol 116 (22) ◽  
pp. 4600-4611 ◽  
Author(s):  
Sivahari P. Gorantla ◽  
Tobias N. Dechow ◽  
Rebekka Grundler ◽  
Anna Lena Illert ◽  
Christian Meyer zum Büschenfelde ◽  
...  

Abstract The oncogenic JAK2V617F mutation is found in myeloproliferative neoplasms (MPNs) and is believed to be critical for leukemogenesis. Here we show that JAK2V617F requires an intact SH2 domain for constitutive activation of downstream signaling pathways. In addition, there is a strict requirement of cytokine receptor expression for the activation of this oncogene. Further analysis showed that the SH2 domain mutation did not interfere with JAK2 membrane distribution. However, coimmunoprecipitated experiments revealed a role for the SH2 domain in the aggregation and cross-phosphorylation of JAK2V617F at the cell membrane. Forced overexpression of cytokine receptors could rescue the JAK2V617F SH2 mutant supporting a critical role of JAK2V617F abundance for constitutive activation. However, under physiologic cytokine receptor expression the SH2 domain is absolutely necessary for oncogenic JAK2V617F activation. This is demonstrated in a bone marrow transplantation model, in which an intact SH2 domain in JAK2V617F is required for the induction of an MPN-like disease. Thus, our results points to an indispensable role of the SH2 domain in JAK2V617F-induced MPNs.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3608-3608 ◽  
Author(s):  
Sivahari P. Gorantla ◽  
Tobias Dechow ◽  
Christian Peschel ◽  
Justus Duyster

Abstract A point mutation in JAK2 (V617F) has been described recently in patients with myeloproliferative diseases like polycythemia vera (PV), essential thrombocythemia (ET) and chronic idiopathic myelofibrosis (IMF). This V617F point mutation in JAK2 has been shown to activate several downstream pathways including STAT5 and ERK. This mutation also renders haematopoietic progenitors cytokine-independent. The role of the V617F mutation in oncogenesis is not fully understood. In this study we aim to dissect the role of the SH2 domain in JAK2-V617F mediated transformation. Stable Ba/F3 cell lines expressing JAK2-wild type (wt), JAK2-V617F, JAK2-R439K (SH2 domain mutation) and JAK2-V617F/R439K mutants were generated. Cell proliferation assays showed that JAK2-V617F transforms Ba/F3 cells and renders them IL3 independent, while wild type JAK2 and JAK2-R439K could not. Surprisingly, JAK2-V617F/R439K was not able to induce a transformed phenotype in Ba/F3 cells. Imunoblotting revealed strong activation of JAK2, STAT5 and ERK in cells expressing JAK2-V617F, whereas no such activation could be found in JAK2-wt, JAK2-R439K and in JAK2-V617F/R439K expressing cells. Thus the SH2 domain in JAK2-V617F seems to play a crucial role in the transformation of Ba/F3 cells containing a heterodimeric (IL-3) cytokine receptor. It has been demonstrated that JAK2-V617F induces cellular transformation more efficiently in cells expressing a homodimeric cytokine receptor such as the erythropoetin receptor. We therefore established Ba/F3 cells overexpressing EpoR together with JAK2-wt, JAK2-V617F, JAK2-R439K and JAK2-V617F/R439K. In contrast to parental Ba/F3 cells, EpoR expressing Ba/F3 cells could be transformed by both JAK2-V617F as well as JAK2-V617F/R439K. Both the single and double mutant Ba/F3 cells showed strong activation of STAT5 and ERK. This suggests that an intact SH2 domain is not required for homodimeric cytokine receptor expressing cells. These results show that transformation by JAK2-V617F requires an intact SH2 domain only in cells expressing a heterodimeric cytokine receptor. In contrast, cells containing a homodimeric cytokine receptor are able to induce transformation in the presence of JAK2-V617F with an additional SH2 mutation. Further progress in understanding the role of the SH2 domain in JAK2-V617F mediated transformation may help in delineating downstream signalling with therapeutic implications.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4191-4191
Author(s):  
Lori N. Griner ◽  
Kathy L. McGraw ◽  
Joseph O. Johnson ◽  
Alan F. List ◽  
Gary W. Reuther

Abstract Abstract 4191 JAK2 is a cytoplasmic tyrosine kinase that plays an important role in signaling following activation of various cytokine receptors. JAK2 activation promotes growth, survival, and differentiation of various cell types. Mutation of JAK2 is seen in numerous hematopoietic diseases, most notably in myeloproliferative neoplasms (MPNs). JAK2-V617F is a frequent mutation found in the classical MPNs: polycythemia vera, essential thrombocythemia, and primary myelofibrosis. The single amino acid change of valine to phenylalanine occurs in the pseudokinase domain of JAK2, relieving auto-inhibition of the kinase domain and allowing constitutive kinase activity. Numerous mouse models have demonstrated that JAK2-V617F can induce MPN-like disorders in mice. Thus, this point mutation, as well as other less common JAK2 mutations, is believed to play an important etiologic role in the development of MPNs in humans. The development and use of JAK2 inhibitors in clinical trials has shown promising results, again demonstrating the important role JAK2 plays in MPNs. While the JAK2-V617F mutation, as well as other JAK2 mutations, decreases auto-inhibition of JAK2 kinase activity, it is clear that mutated JAK2 still requires the expression of cytokine receptors to induce activation of transforming signals in hematopoietic cells. Normally, JAK2 binds to homodimeric and heterodimeric cytokine receptors through specific receptor motifs and is activated by various structural changes induced by cytokine stimulation. Following activation it utilizes receptor tyrosines as substrates for phosphorylation, leading to recruitment of downstream signaling molecules, such as STAT5, among others. JAK2 then activates STAT5 via phosphorylation and STAT5 then translocates to the nucleus to regulate transcription of target genes. JAK2-V617F does not require ligand for activation, but still requires the scaffolding function of cytokine receptors to facilitate its full activation and activation of downstream signaling via phosphorylation. Lipid rafts are microdomains of the plasma membrane that are enriched in cholesterol and sphingolipids. They have gained appreciation in signal transduction as sites of localization of signaling mediators, including membrane-bound receptors. Congregation of signaling proteins in lipid rafts within the plasma membrane promotes complex formation and signaling cascade activation. We have recently demonstrated that JAK2 is present in lipid rafts during erythropoietin signaling and that lipid raft integrity is required for erythropoietin-mediated signal transduction (Blood 2009, 114: 292). In our current study, we demonstrate that constitutive JAK-STAT signaling driven by JAK2-V617F is sensitive to lipid raft disruption. Human erythroleukemia (HEL) cells express constitutive activation of JAK-STAT signaling due to the presence of JAK2-V617F. Treatment of these cells with methyl-beta-cyclodextrin to disrupt lipid rafts abolished JAK2, STAT5, and STAT3 activation. Similar results are obtained in other cell lines harboring JAK2-V617F and that exhibit JAK-STAT activation that is dependent on this activated form of JAK2. We also demonstrate that JAK2-V617F co-localizes with lipid rafts, as shown by immunofluorescence, and that this co-localization is abolished by lipid raft disruption. This suggests the loss of JAK2-V617F-mediated JAK-STAT activation we observe following lipid raft disruption may be due to an inhibition of properly localized protein complex formation in the plasma membrane that is necessary for JAK2-V617F signaling. Lipid rafts may provide a site for an accumulation of JAK2-V617F-containing signaling complexes and may be necessary for the cellular signals initiated by JAK2-V617F. Our data show JAK2-V617F-driven JAK-STAT pathway activation is vulnerable to lipid raft disrupting agents and suggest lipid raft integrity as a potential therapeutic target for JAK2-V617F positive neoplasms. Targeting lipid rafts in combination with JAK2 kinase inhibitors may allow for more effective kinase inhibition at lower doses, potentially decreasing undesirable side effects associated with kinase inhibitor treatment. Disclosures: No relevant conflicts of interest to declare.


Cells ◽  
2019 ◽  
Vol 8 (8) ◽  
pp. 854 ◽  
Author(s):  
Perner ◽  
Perner ◽  
Ernst ◽  
Heidel

Clonal alterations in hematopoietic cells occur during aging and are often associated with the establishment of a subclinical inflammatory environment. Several age-related conditions and diseases may be initiated or promoted by these alterations. JAK2 mutations are among the most frequently mutated genes in blood cells during aging. The most common mutation within the JAK2 gene is JAK2-V617F that leads to constitutive activation of the kinase and thereby aberrant engagement of downstream signaling pathways. JAK2 mutations can act as central drivers of myeloproliferative neoplasia, a pre-leukemic and age-related malignancy. Likewise, hyperactive JAK-signaling is a hallmark of immune diseases and critically influences inflammation, coagulation and thrombosis. In this review we aim to summarize the current knowledge on JAK2 in clonal hematopoiesis during aging, the role of JAK-signaling in inflammation and lymphocyte biology and JAK2 function in age-related diseases and malignant transformation.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3732-3732
Author(s):  
Anuradha R. Pradhan ◽  
Que Lambert ◽  
Gary W. Reuther

Abstract We have recently identified IL27Ra as a gene from an AML patient that transforms hematopoietic cells to a state of cytokine-independent growth. Importantly, we have found that the gene product of IL27Ra, intereukin-27 receptor (IL27R), is expressed on the cell surface of the leukemic cells of AML patients. IL27R, also know as TCCR and WSX-1, is a component if a heterodimeric type I cytokine receptor that functions as the ligand-binding subunit of the receptor for IL-27. Normally, IL27R functions with the gp130 co-receptor to induce signal transduction in response to IL-27. However, we have demonstrated that IL27R can induce the transformation of hematopoietic cells in a ligand- and gp130-independent manner. IL27R, which contains a JAK-binding Box 1 motif, requires JAK family kinase activity to transform cells. In a potential mechanism of IL27R-mediated transformation, IL27R may function as a homodimer to activate JAK family members. In an effort to determine if IL27R can function as a homodimer, we tested to see if IL27R could activate the V617F mutation of JAK2 commonly found in various myeloproliferative neoplasms (MPNs). It is believed that JAK2-V617F utilizes a homodimeric cytokine receptor complex as a scaffold in order to multimerize and become fully activated via transphosphorylation. Expression of IL27R activated the kinase activity of JAK2-V617F, suggesting that IL27R may form homodimers in cells. In addition, co-expression of IL27R and JAK2-V617F led to transformation of BaF3 cells to cytokine-independence, whereas IL27R expressed with wildtype JAK2 or JAK2-V617F expressed alone did not induce rapid cytokine-independent growth of these cells. This is similar data to that obtained with co-expression of JAK2-V617F with homodimeric receptors (e.g. EpoR, GCSFR), demonstrating that a component of a heterodimeric cytokine receptor can functionally replace a homodimeric receptor in mediating JAK2-V617F activation. Chemical crosslinking showed that IL27R could dimerize suggesting that IL27R may form a scaffold for JAK2-V617F-mediated transformation. In addition to providing a scaffold for binding and activation of JAK2-V617F, it has been proposed that homodimeric receptors also provide an important substrate of the JAK2 tyrosine kinase activity to induce cell signaling and transformation. Tyrosine phosphorylation of the scaffolding receptor presumably recruits signaling molecules (e.g. STATs) to the receptor where they can be activated by JAK2 phosphorylation. We have mutated the two tyrosine residues in the cytoplasmic domain of IL27R and used these receptor mutants to determine if tyrosine phosphorylation of IL27R is required to induce the transforming signal of JAK2-V617F. Upon initial cytokine removal, there is a slight lag in the growth of JAK2-V617F cells expressing an IL27R receptor that lacks both intracellular tyrosines, suggesting these tyrosines may play a role in signaling. However, this lag is transient and cytokine-independent growth becomes similar to cells expressing wildtype IL27R and JAK2-V617F. This suggests tyrosine phosphorylation of the receptor is not required to induce the transforming signal of JAK2-V617F. Our studies demonstrate for the first time that single chain components of heterodimeric receptors can functionally replace homodimeric receptors in mediating transformation of hematopoietic cells by JAK2-V617F. Our work also suggests that JAK2-V617F does not need to phosphorylate its receptor scaffold to mediate its transforming signal. The requirement for other endogenous cytokine receptors as substrates for activated JAK2-V617F receptors remains a possibility and is currently under investigation. Together, our studies demonstrate that IL27R, and perhaps other single chain components of heterodimeric receptors, may play unappreciated roles in myeloid diseases such as AML and MPNs.


2015 ◽  
Vol 468 (2) ◽  
pp. 283-291 ◽  
Author(s):  
Sonja Lorenz ◽  
Patricia Deng ◽  
Oliver Hantschel ◽  
Giulio Superti-Furga ◽  
John Kuriyan

Constitutive activation of the tyrosine kinase c-Abl in the cell involves interactions between the SH2 (Src-homology 2) and kinase domains (KDs). We present a crystal structure of a c-Abl construct comprising these domains and analyse the functional role of their interface in vitro.


1993 ◽  
Vol 79 (2) ◽  
pp. 92-99 ◽  
Author(s):  
Vincenzo Chiarugi ◽  
Persio Dello Sbarba ◽  
Lucia Magnelli ◽  
Paola Chiarugi ◽  
Stefania Pacini ◽  
...  

Novel aspects of cytokine receptor signal transduction are discussed and cytokine receptors classified based on ligand-dependent signalling. An introductory section presents an overview of the role of cytokines in hematopoiesis. A brief explanation of basic concepts, such as redundancy, pleiotropy, synergism, etc., important for the understanding of cell response to cytokines, is also included. Three of five classes of receptors show the involvement of tyrosine kinase activity as the key step in signal transduction. The importance of tyrosine phosphorylation in cellular response to cytokines is pointed out.


2021 ◽  
Vol 7 (1) ◽  
Author(s):  
Yu Zhang ◽  
Christopher D. Kontos ◽  
Brian H. Annex ◽  
Aleksander S. Popel

AbstractThe Ang–Tie signaling pathway is an important vascular signaling pathway regulating vascular growth and stability. Dysregulation in the pathway is associated with vascular dysfunction and numerous diseases that involve abnormal vascular permeability and endothelial cell inflammation. The understanding of the molecular mechanisms of the Ang–Tie pathway has been limited due to the complex reaction network formed by the ligands, receptors, and molecular regulatory mechanisms. In this study, we developed a mechanistic computational model of the Ang–Tie signaling pathway validated against experimental data. The model captures and reproduces the experimentally observed junctional localization and downstream signaling of the Ang–Tie signaling axis, as well as the time-dependent role of receptor Tie1. The model predicts that Tie1 modulates Tie2’s response to the context-dependent agonist Ang2 by junctional interactions. Furthermore, modulation of Tie1’s junctional localization, inhibition of Tie2 extracellular domain cleavage, and inhibition of VE-PTP are identified as potential molecular strategies for potentiating Ang2’s agonistic activity and rescuing Tie2 signaling in inflammatory endothelial cells.


2020 ◽  
Vol 4 (Supplement_1) ◽  
Author(s):  
Colleen Hadley ◽  
Isin Cakir ◽  
Roger D Cone

Abstract Overweight and obesity are global concerns affecting nearly one third of the world population. These conditions are characterized by increased adiposity and are accompanied by a proportional increase in circulating leptin, an anorexigenic adipokine. Leptin is responsible for signaling peripheral energy status to the central nervous system to modulate food intake and energy expenditure. As such, neurons within the hypothalamus expressing the long isoform of leptin receptor (LepRb), a type I cytokine receptor, are primarily responsible for mediating the effects of leptin, which signal predominantly through the JAK2-STAT3 transduction mechanism. STAT3 is a latent transcription factor activated upon phosphorylation, which triggers its homodimerization and nuclear translocation. Evidence, however, for JAK2-independent, STAT3-dependent leptin receptor signaling mechanisms exist. FAK (focal adhesion kinase, Ptk2) and Pyk2 (protein tyrosine kinase 2b, Ptk2b) are a subset of nonreceptor protein tyrosine kinases and comprise the focal adhesion kinase family. FAK and Pyk2 are implicated in the regulation of cytokine receptor signaling. Furthermore, Pyk2 knockout mice have an obesity prone phenotype. Here, we studied the role of the focal adhesion kinases in leptin receptor signaling using genetic and pharmacological approaches. We found that overexpression of Pyk2 or FAK increased STAT3 phosphorylation (activation). Overexpression of a FAK or Pyk2 construct with impaired kinase activity, however, attenuated STAT3 phosphorylation, suggesting the increase in STAT3 phosphorylation is largely dependent upon kinase activity of FAK/Pyk2. Treatment of cells with a small molecule dual inhibitor of FAK and Pyk2 (PF431396) attenuated leptin-induced STAT3 phosphorylation in a mouse hypothalamic cell line. Importantly, this effect is independent of JAK2, as PF treatment of two independent JAK2-deficient cell lines exhibited similar attenuation of leptin-induced STAT3 phosphorylation. To assess the physiological relevance of FAK/Pyk2 in leptin receptor signaling in vivo, we administered PF compound to the lateral ventricle of 24-hour fasted lean wild-type mice followed by peripheral leptin administration. Intracerebroventricular (ICV) administration of PF suppressed the anorectic effect of leptin as evidenced by impaired inhibition of food intake upon refeeding. Accordingly, analysis of total hypothalamic lysates from these mice showed ICV PF impaired leptin-induced STAT3 phosphorylation. Taken together, these data suggest that Pyk2 and/or FAK play a role in leptin signal transduction.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Jin Wang ◽  
Jie Huang ◽  
Shan Yao ◽  
Jia-Hui Wu ◽  
Hui-Bin Li ◽  
...  

Abstract Background The ketogenic diet (KD)has been considered an effective treatment for epilepsy, whereas its underlying mechanisms remain obscure. We have previously reported that the KD feeding increased Neuregulin 1 (NRG1) expression in the hippocampus; disruption of NRG1 signaling by genetically deleting its receptor-ErbB4 abolished KD’s effects on inhibitory synaptic activity and seizures. However, it is still unclear about the mechanisms underlying the effect of KD on NRG1 expression and whether the effects of KD require ErbB4 kinase activity. Methods The effects of the KD on NRG1 expression were assessed via western blotting and real-time PCR. Acetylation level at the Nrg1 promoter locus was examined using the chromatin immunoprecipitation technique. Kainic acid (KA)-induced acute seizure model was utilized to examine the effects of KD and histone deacetylase inhibitor-TSA on seizures. Synaptic activities in the hippocampus were recorded with the technique of electrophysiology. The obligatory role of ErbB4 kinase activity in KD’s effects on seizures and inhibitory synaptic activity was evaluated by using ErbB kinase antagonist and transgenic mouse-T796G. Results We report that KD specifically increases Type I NRG1 expression in the hippocampus. Using the chromatin immunoprecipitation technique, we observe increased acetylated-histone occupancy at the Nrg1 promoter locus of KD-fed mice. Treatment of TSA dramatically elevates NRG1 expression and diminishes the difference between the effects of the control diet (CD) and KD. These data indicate that KD increases NRG1 expression via up-regulating histone acetylation. Moreover, both pharmacological and genetic inhibitions of ErbB4 kinase activity significantly block the KD’s effects on inhibitory synaptic activity and seizure, suggesting an essential role of ErbB4 kinase activity. Conclusion These results strengthen our understanding of the role of NRG1/ErbB4 signaling in KD and shed light on novel therapeutic interventions for epilepsy.


Sign in / Sign up

Export Citation Format

Share Document