Requirement of Lipid Raft Integrity for Signaling by JAK2-V617F

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.

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 ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3863-3863
Author(s):  
Gwenny M. Fuhler ◽  
A. Lyndsay Drayer ◽  
Paul J. Coffer ◽  
Edo Vellenga

Abstract Neutrophils from MDS patients are generally dysfunctional, resulting in increased susceptibility to bacterial infections. An important bactericidal activity of neutrophils is their production of reactive oxygen species (ROS) upon stimulation with the bacterial product fMLP. ROS production by fMLP can be enhanced by pre-treatment of neutrophils with pro-inflammatory cytokines such as GM-CSF. In MDS patients, this GM-CSF priming of ROS production is severely impaired. Furthermore, activation of the extracellular signal-regulated kinase (ERK1/2) and phosphatidylinositol 3-kinase/protein kinase B (PI3K/PKB) signal transduction pathways, involved in ROS production, are impaired in MDS (Fuhler et al, Blood, 101, 2003). In the current study we investigated the involvement of lipid rafts in neutrophil ROS production, and show that treatment of healthy neutrophils with the lipid-raft disrupting agent methyl-b-cyclodextrin (CD) abrogates fMLP-induced ROS production in unprimed cells (mean fluorescence intensity [MFI] of 16±2 vs 4±0.3, n=5, P=0.03) and GM-CSF-primed neutrophils (MFI 50±14 vs 9±1, P=0.04). We further demonstrate that activation of ERK and PKB by fMLP was abolished by CD treatment of unprimed or GM-CSF-primed neutrophils. In contrast, GM-CSF-stimulated phosphorylation of STAT5 was not affected by CD pre-treatment of healthy neutrophils. These results indicate the importance of lipid rafts in fMLP-stimulated ROS production and in activation of signal transduction pathways involved in this process. The expression of the ganglioside GM1 at the plasma membrane of cells has been described to be a marker for the presence of lipid rafts. We analyzed the expression of GM1 on healthy neutrophils by FACS analysis and show that stimulation of cells with fMLP increased the expression of GM1 at the plasma membrane (MFI 102±20 vs 161±38, n=6, P=0.03). Furthermore, pretreatment of neutrophils with GM-CSF further increased the fMLP-induced GM1 expression (MFI 161 ±38 vs 193±36, P=0.03). We next assessed the expression of GM1 at the plasma membrane, in conjunction with ROS production, in 8 MDS patients compared to healthy donors. As described previously, ROS production in response to fMLP was normal in unprimed MDS neutrophils. In contrast, in 6 patients, fMLP-induced ROS production in GM-CSF primed neutrophils was significantly impaired when compared to their healthy controls (mean MFI 128±54 vs 191±72, P=0.03). When investigating the GM1 expression in these patients, we found that although fMLP-triggered GM1 expression was normal in unprimed neutrophils from MDS patients, fMLP-induced GM1 expression on GM-CSF-primed cells was significantly lower on MDS neutrophils compared to their healthy counterpart (mean MFI 174±76 vs 214±78, P=0.03). GM-CSF primed neutrophils from one patient with normal ROS production also exhibited normal GM1 expression. Taken together, these data indicate an involvement of lipid rafts in ROS production, and suggest that in MDS neutrophils, an impaired lipid raft formation in GM-CSF primed cells correlates with impaired fMLP-induced ROS production.


2004 ◽  
Vol 78 (23) ◽  
pp. 12773-12780 ◽  
Author(s):  
Kati Pulkkinen ◽  
G. Herma Renkema ◽  
Frank Kirchhoff ◽  
Kalle Saksela

ABSTRACT We have previously reported that Nef specifically interacts with a small but highly active subpopulation of p21-activated kinase 2 (PAK2). Here we show that this is due to a transient association of Nef with a PAK2 activation complex within a detergent-insoluble membrane compartment containing the lipid raft marker GM1. The low abundance of this Nef-associated kinase (NAK) complex was found to be due to an autoregulatory mechanism. Although activation of PAK2 was required for assembly of the NAK complex, catalytic activity of PAK2 also promoted dissociation of this complex. Testing different constitutively active PAK2 mutants indicated that the conformation associated with p21-mediated activation rather than kinase activity per se was required for PAK2 to become NAK. Although association with PAK2 is one of the most conserved properties of Nef, we found that the ability to stimulate PAK2 activity differed markedly among divergent Nef alleles, suggesting that PAK2 association and activation are distinct functions of Nef. However, mutations introduced into the p21-binding domain of PAK2 revealed that p21-GTPases are involved in both of these Nef functions and, in addition to promoting PAK2 activation, also help to physically stabilize the NAK complex.


2015 ◽  
Vol 211 (6) ◽  
pp. 1193-1205 ◽  
Author(s):  
Heather Miller ◽  
Thiago Castro-Gomes ◽  
Matthias Corrotte ◽  
Christina Tam ◽  
Timothy K. Maugel ◽  
...  

Cells rapidly repair plasma membrane (PM) damage by a process requiring Ca2+-dependent lysosome exocytosis. Acid sphingomyelinase (ASM) released from lysosomes induces endocytosis of injured membrane through caveolae, membrane invaginations from lipid rafts. How B lymphocytes, lacking any known form of caveolin, repair membrane injury is unknown. Here we show that B lymphocytes repair PM wounds in a Ca2+-dependent manner. Wounding induces lysosome exocytosis and endocytosis of dextran and the raft-binding cholera toxin subunit B (CTB). Resealing is reduced by ASM inhibitors and ASM deficiency and enhanced or restored by extracellular exposure to sphingomyelinase. B cell activation via B cell receptors (BCRs), a process requiring lipid rafts, interferes with PM repair. Conversely, wounding inhibits BCR signaling and internalization by disrupting BCR–lipid raft coclustering and by inducing the endocytosis of raft-bound CTB separately from BCR into tubular invaginations. Thus, PM repair and B cell activation interfere with one another because of competition for lipid rafts, revealing how frequent membrane injury and repair can impair B lymphocyte–mediated immune responses.


Molecules ◽  
2020 ◽  
Vol 25 (6) ◽  
pp. 1398
Author(s):  
Cristian Vergallo ◽  
Elisa Panzarini ◽  
Bernardetta Anna Tenuzzo ◽  
Stefania Mariano ◽  
Ada Maria Tata ◽  
...  

One of the most relevant drawbacks in medicine is the ability of drugs and/or imaging agents to reach cells. Nanotechnology opened new horizons in drug delivery, and silver nanoparticles (AgNPs) represent a promising delivery vehicle for their adjustable size and shape, high-density surface ligand attachment, etc. AgNPs cellular uptake involves different endocytosis mechanisms, including lipid raft-mediated endocytosis. Since static magnetic fields (SMFs) exposure induces plasma membrane perturbation, including the rearrangement of lipid rafts, we investigated whether SMF could increase the amount of AgNPs able to pass the peripheral blood lymphocytes (PBLs) plasma membrane. To this purpose, the effect of 6-mT SMF exposure on the redistribution of two main lipid raft components (i.e., disialoganglioside GD3, cholesterol) and on AgNPs uptake efficiency was investigated. Results showed that 6 mT SMF: (i) induces a time-dependent GD3 and cholesterol redistribution in plasma membrane lipid rafts and modulates gene expression of ATP-binding cassette transporter A1 (ABCA1), (ii) increases reactive oxygen species (ROS) production and lipid peroxidation, (iii) does not induce cell death and (iv) induces lipid rafts rearrangement, that, in turn, favors the uptake of AgNPs. Thus, it derives that SMF exposure could be exploited to enhance the internalization of NPs-loaded therapeutic or diagnostic molecules.


2017 ◽  
Vol 312 (5) ◽  
pp. C627-C637 ◽  
Author(s):  
Alexey M. Petrov ◽  
Violetta V. Kravtsova ◽  
Vladimir V. Matchkov ◽  
Alexander N. Vasiliev ◽  
Andrey L. Zefirov ◽  
...  

Marked loss of skeletal muscle mass occurs under various conditions of disuse, but the molecular and cellular mechanisms leading to atrophy are not completely understood. We investigate early molecular events that might play a role in skeletal muscle remodeling during mechanical unloading (disuse). The effects of acute (6–12 h) hindlimb suspension on the soleus muscles from adult rats were examined. The integrity of plasma membrane lipid rafts was tested utilizing cholera toxin B subunit or fluorescent sterols. In addition, resting intracellular Ca2+ level was analyzed. Acute disuse disturbed the plasma membrane lipid-ordered phase throughout the sarcolemma and was more pronounced in junctional membrane regions. Ouabain (1 µM), which specifically inhibits the Na-K-ATPase α2 isozyme in rodent skeletal muscles, produced similar lipid raft changes in control muscles but was ineffective in suspended muscles, which showed an initial loss of α2 Na-K-ATPase activity. Lipid rafts were able to recover with cholesterol supplementation, suggesting that disturbance results from cholesterol loss. Repetitive nerve stimulation also restores lipid rafts, specifically in the junctional sarcolemma region. Disuse locally lowered the resting intracellular Ca2+ concentration only near the neuromuscular junction of muscle fibers. Our results provide evidence to suggest that the ordering of lipid rafts strongly depends on motor nerve input and may involve interactions with the α2 Na-K-ATPase. Lipid raft disturbance, accompanied by intracellular Ca2+ dysregulation, is among the earliest remodeling events induced by skeletal muscle disuse.


2006 ◽  
Vol 34 (3) ◽  
pp. 374-376 ◽  
Author(s):  
E.B. Babiychuk ◽  
A. Draeger

The spatial segregation of the plasma membrane plays a prominent role in distinguishing and sorting a large number of signals a cell receives simultaneously. The plasma membrane comprises regions known as lipid rafts, which serve as signal-transduction hubs and platforms for sorting membrane-associated proteins. Ca2+-binding proteins of the annexin family have been ascribed a role in the regulation of raft dynamics. Glycosylphosphatidylinositol-anchored 5′-nucleotidase is an extracellular, raft-associated enzyme responsible for conversion of extracellular ATP into adenosine. Our results point to a regulation of ecto-5′-nucleotidase activity by Ca2+-dependent, annexin-mediated stabilization of membrane rafts.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2686-2686
Author(s):  
A.E. Schade ◽  
H. Szpurka ◽  
A. Jankowska ◽  
J. Bauer ◽  
E.D. Hsi ◽  
...  

Abstract Cytokine receptors preferentially associate with particular JAK-STAT combinations to transduce specific signals. For example, erythropoietin receptor (Epo-R) preferentially interacts with JAK2 to initiate signaling pathways via STAT5. The JAK2 V617F mutation (mJAK2) found in some myeloproliferative disorders (MPD) still requires binding to type I cytokine receptors to initiate signaling. Consequently, aberrant JAK-STAT signaling in MPD may require physiologic interactions with other pathways. Src family kinases (SFK) interact with various cytokine receptors resulting in close association between SFK and JAK-STAT pathways. We hypothesized that SFK activity plays a role in the activation of STAT5 and, given the importance of STAT5 in the pathogenesis of MPD, targeted inhibition of SFK could provide a novel therapeutic approach. First, we examined the effect the SFK inhibitors PP2 and SU6656 on the proliferation of the HEL cell line harboring mJAK2 and the Epo-dependent AML line UT7/Epo; SFK inhibition significantly diminished proliferation in both cell lines. These results imply that despite of the constitutive activity of mJAK2 or in the presence of Epo stimulated JAK2-STAT5 induction, SFK activation is required for proliferation. Since mJAK2 requires a functionally intact type I cytokine receptor, we examined Epo-R signaling in greater detail via phospho-specific immunoblotting. SFK inhibition resulted in diminished levels of phospho-SFK, coinciding with a similar degree of diminished phospho-STAT5. Simultaneously decreased induction of AKT and ERK pathways after SFK inhibition suggested SFK activity is also regulating a more global signaling network through the Epo-R. Inhibition of JAK2 activity potently suppressed phospho-STAT5, as well as ERK and AKT, without affecting SFK phosphorylation. Thus, SFK lies upstream of JAK2, or SFK and JAK2 may be regulating the second messenger pathways in parallel. In electrophoretic mobility shift assays to examine the effect of SFK inhibition on STAT5 DNA binding, SFK inhibition resulted in decreased STAT5 DNA binding despite constitutive activity of mJAK2. Analogous results were obtained after Epo stimulation in UT7/Epo cells. As expected, inhibition of JAK2 resulted in almost complete loss of STAT5 DNA binding. To confirm these results in primary cells, we examined the effects of SFK inhibition on primary monocytes from a patient with mJAK2. Stimulation with GM-CSF resulted in increased STAT5A DNA binding, but not STAT5B. In the presence of the SFK inhibitor PP2, GM-CSF induction of STAT5A DNA binding activity was completely inhibited. It is interesting to note that a key difference between STAT5A and STAT5B is the potential for ERK regulation of STAT5A DNA binding activity. Thus, showing here that SFK regulates ERK activity, and knowing that ERK activity can positively regulate STAT5A DNA binding, we propose a model in which SFK activity may modulate the JAK2-STAT5 signaling axis via the ERK pathway. In summary, our results demonstrate that while JAK2 is essential for this process, SFK activity appears to be necessary for full activation by positively modulating the JAK2-STAT5 axis. SFK inhibitors recently approved and in clinical trials may demonstrate efficacy in hematologic diseases characterized by aberrant JAK-STAT signaling, such as MPD.


2007 ◽  
Vol 28 (5) ◽  
pp. 1792-1801 ◽  
Author(s):  
Megumi Funakoshi-Tago ◽  
Stéphane Pelletier ◽  
Hiroshi Moritake ◽  
Evan Parganas ◽  
James N. Ihle

ABSTRACT Janus kinases are essential for signal transduction by a variety of cytokine receptors and when inappropriately activated can cause hematopoietic disorders and oncogenesis. Consequently, it can be predicted that the interaction of the kinases with receptors and the events required for activation are highly controlled. In a screen to identify phosphorylation events regulating Jak2 activity in EpoR signaling, we identified a mutant (Jak2-Y613E) which has the property of being constitutively activated, as well as an inactivating mutation (Y766E). Although no evidence was obtained to indicate that either site is phosphorylated in signaling, the consequences of the Y613E mutation are similar to those observed with recently described activating mutations in Jak2 (Jak2-V617F and Jak2-L611S). However, unlike the V617F or L611S mutant, the Y613E mutant requires the presence of the receptor but not Epo stimulation for activation and downstream signaling. The properties of the Jak2-Y613E mutant suggest that under normal conditions, Jak2 that is not associated with a receptor is locked into an inactive state and receptor binding through the FERM domain relieves steric constraints, allowing the potential to be activated with receptor engagement.


2012 ◽  
Vol 87 (3) ◽  
pp. 1301-1311 ◽  
Author(s):  
David G. Meckes ◽  
Nathan F. Menaker ◽  
Nancy Raab-Traub

ABSTRACTThe Epstein-Barr virus (EBV) is an important human pathogen that is associated with multiple cancers. The major oncoprotein of the virus, latent membrane protein 1 (LMP1), is essential for EBV B-cell immortalization and is sufficient to transform rodent fibroblasts. This viral transmembrane protein activates multiple cellular signaling pathways by engaging critical effector molecules and thus acts as a ligand-independent growth factor receptor. LMP1 is thought to signal from internal lipid raft containing membranes; however, the mechanisms through which these events occur remain largely unknown. Lipid rafts are microdomains within membranes that are rich in cholesterol and sphingolipids. Lipid rafts act as organization centers for biological processes, including signal transduction, protein trafficking, and pathogen entry and egress. In this study, the recruitment of key signaling components to lipid raft microdomains by LMP1 was analyzed. LMP1 increased the localization of phosphatidylinositol 3-kinase (PI3K) and its activated downstream target, Akt, to lipid rafts. In addition, mass spectrometry analyses identified elevated vimentin in rafts isolated from LMP1 expressing NPC cells. Disruption of lipid rafts through cholesterol depletion inhibited PI3K localization to membranes and decreased both Akt and ERK activation. Reduction of vimentin levels or disruption of its organization also decreased LMP1-mediated Akt and ERK activation and inhibited transformation of rodent fibroblasts. These findings indicate that LMP1 reorganizes membrane and cytoskeleton microdomains to modulate signal transduction.


Sign in / Sign up

Export Citation Format

Share Document