Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Blood Adv ; 1(16): 1224-1237, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-29296762

ABSTRACT

Genetics play a significant role in venous thromboembolism (VTE), yet current clinical laboratory-based testing identifies a known heritable thrombophilia (factor V Leiden, prothrombin gene mutation G20210A, or a deficiency of protein C, protein S, or antithrombin) in only a minority of VTE patients. We hypothesized that a substantial number of VTE patients could have lesser-known thrombophilia mutations. To test this hypothesis, we performed whole-exome sequencing (WES) in 64 patients with VTE, focusing our analysis on a novel 55-gene extended thrombophilia panel that we compiled. Our extended thrombophilia panel identified a probable disease-causing genetic variant or variant of unknown significance in 39 of 64 study patients (60.9%), compared with 6 of 237 control patients without VTE (2.5%) (P < .0001). Clinical laboratory-based thrombophilia testing identified a heritable thrombophilia in only 14 of 54 study patients (25.9%). The majority of WES variants were either associated with thrombosis based on prior reports in the literature or predicted to affect protein structure based on protein modeling performed as part of this study. Variants were found in major thrombophilia genes, various SERPIN genes, and highly conserved areas of other genes with established or potential roles in coagulation or fibrinolysis. Ten patients (15.6%) had >1 variant. Sanger sequencing performed in family members of 4 study patients with and without VTE showed generally concordant results with thrombotic history. WES and extended thrombophilia testing are promising tools for improving our understanding of VTE pathogenesis and identifying inherited thrombophilias.

2.
Nat Struct Mol Biol ; 21(7): 579-84, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24918548

ABSTRACT

Janus kinase-2 (JAK2) mediates signaling by various cytokines, including erythropoietin and growth hormone. JAK2 possesses tandem pseudokinase and tyrosine-kinase domains. Mutations in the pseudokinase domain are causally linked to myeloproliferative neoplasms (MPNs) in humans. The structure of the JAK2 tandem kinase domains is unknown, and therefore the molecular bases for pseudokinase-mediated autoinhibition and pathogenic activation remain obscure. Using molecular dynamics simulations of protein-protein docking, we produced a structural model for the autoinhibitory interaction between the JAK2 pseudokinase and kinase domains. A striking feature of our model, which is supported by mutagenesis experiments, is that nearly all of the disease mutations map to the domain interface. The simulations indicate that the kinase domain is stabilized in an inactive state by the pseudokinase domain, and they offer a molecular rationale for the hyperactivity of V617F, the predominant JAK2 MPN mutation.


Subject(s)
Janus Kinase 2/physiology , Models, Genetic , Binding Sites , Computer Simulation , Humans , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , Models, Molecular , Protein Structure, Tertiary , Signal Transduction , Structure-Activity Relationship
3.
Biochim Biophys Acta ; 1834(10): 2166-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23467009

ABSTRACT

MuSK (muscle-specific kinase) is a receptor tyrosine kinase that plays a central signaling role in the formation of neuromuscular junctions (NMJs). MuSK is activated in a complex spatio-temporal manner to cluster acetylcholine receptors on the postsynaptic (muscle) side of the synapse and to induce differentiation of the nerve terminal on the presynaptic side. The ligand for MuSK is LRP4 (low-density lipoprotein receptor-related protein-4), a transmembrane protein in muscle, whose binding affinity for MuSK is potentiated by agrin, a neuronally derived heparan-sulfate proteoglycan. In addition, Dok7, a cytoplasmic adaptor protein, is also required for MuSK activation in vivo. This review focuses on the physical interplay between these proteins and MuSK for activation and downstream signaling, which culminates in NMJ formation. This article is part of a Special Issue entitled: Emerging recognition and activation mechanisms of receptor tyrosine kinases.


Subject(s)
Muscle, Skeletal/metabolism , Nerve Endings/metabolism , Neuromuscular Junction/chemistry , Receptor Protein-Tyrosine Kinases/chemistry , Receptors, Cholinergic/chemistry , Agrin/chemistry , Agrin/genetics , Agrin/metabolism , Animals , Gene Expression Regulation , Humans , LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Models, Molecular , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Synapses/metabolism , Synaptic Transmission
4.
Mol Cell Biochem ; 367(1-2): 125-40, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22584586

ABSTRACT

Jak2 mutations in the exon 14 and exon 12 regions that cause constitutive activation have been associated with myeloproliferative neoplasms. We have previously shown that a pi stacking interaction between F617 and F595 is important for the constitutive activation of Jak2-V617F (Gnanasambandan et al., Biochemistry 49:9972-9984, 2010). Here, using a combination of molecular dynamics (MD) simulations and in vitro mutagenesis, we studied the molecular mechanism for the constitutive activation of the Jak2 exon 12 mutation, H538Q/K539L. The activation levels of Jak2-H538Q/K539L were found to be similar to that of Jak2-V617F, and Jak2-H538Q/K539L/V617F. Data from MD simulations indicated a shift in the salt bridge interactions of D620 and E621 with K539 in Jak2-WT to R541 in Jak2-H538Q/K539L. When compared to Jak2-WT, K539A mutation resulted in increased activation, while K539D or K539E mutations diminished Jak2 activation by 50 %. In the context of Jak2-H538Q/K539L, R541A mutation reduced its activation by 50 %, while R541D and R541E mutations returned its activation levels to that of Jak2-WT. Collectively, these results indicate that a shift in the salt bridge interaction of D620 and E621 with K539 in Jak2-WT to R541 in Jak2-H538Q/K539L is critical for constitutive activation of this Jak2 exon 12 mutant.


Subject(s)
Aspartic Acid/chemistry , Glutamic Acid/chemistry , Janus Kinase 2/chemistry , Amino Acid Motifs , Amino Acid Substitution , Animals , Arginine/chemistry , COS Cells , Catalytic Domain , Chlorocebus aethiops , Enzyme Activation , Gene Expression Regulation , Genes, Reporter , Humans , Janus Kinase 2/genetics , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Lysine/chemistry , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Signal Transduction , Thermodynamics
5.
Cell Signal ; 23(11): 1806-15, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21726629

ABSTRACT

Jak2 tyrosine kinase plays an important role in cytokine mediated signal transduction. There are 49 tyrosine residues in Jak2 and phosphorylation of some of these are known to play important roles in the regulation of Jak2 kinase activity. Here, using mass spectrometry, we identified tyrosine residues Y372 and Y373 as novel sites of Jak2 phosphorylation. Mutation of Y372 to F (Y372F) significantly inhibited Jak2 phosphorylation, including that of Y1007, whereas the Jak2-Y373F mutant displayed only modest reduction in phosphorylation. Relative to Jak2-WT, the ability of Jak2-Y372F to bind to and phosphorylate STAT1 was decreased, resulting in reduced Jak2-mediated downstream gene transcription. While the Y372F mutation had no effect on receptor-independent, hydrogen peroxide-mediated Jak2 activation, it impaired interferon-gamma (IFNγ) and epidermal growth factor (EGF)-dependent Jak2 activation. Interestingly however, the Y372F mutant exhibited normal receptor binding properties. Finally, co-expression of SH2-Bß only partially restored the activation of the Jak2-Y372F mutant suggesting that the mechanism whereby phosphorylation of Y372 is important for Jak2 activation is via dimerization. As such, our results indicate that Y372 plays a critical yet differential role in Jak2 activation and function via a mechanism involving Jak2 dimerization and stabilization of the active conformation.


Subject(s)
Enzyme Activation/drug effects , Gene Expression Regulation , Janus Kinase 2 , STAT1 Transcription Factor/metabolism , Signal Transduction/genetics , Tyrosine/metabolism , Animals , COS Cells , Chlorocebus aethiops , Dimerization , Enzyme Activation/genetics , Epidermal Growth Factor/pharmacology , Hydrogen Peroxide/pharmacology , Interferon-gamma/pharmacology , Janus Kinase 2/chemistry , Janus Kinase 2/deficiency , Janus Kinase 2/genetics , Mass Spectrometry , Mice , Mice, Knockout , Mutation , Phosphorylation , Plasmids , Protein Binding/drug effects , Protein Binding/genetics , Transcription, Genetic , Transfection , Tyrosine/genetics , Vaccinia virus
6.
Biochemistry ; 49(46): 9972-84, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-20958061

ABSTRACT

Somatic mutations in the Jak2 allele that lead to constitutive kinase activation of the protein have been identified in human disease conditions such as the myeloproliferative neoplasms (MPNs). The most common mutation in these patients is a V617F substitution mutation, which is believed to play a causative role in the MPN pathogenesis. As such, identifying the molecular basis for the constitutive activation of Jak2-V617F is important for understanding its clinical implications and potential treatment. Here, we hypothesized that conversion of residue 617 from Val to Phe resulted in the formation of novel π stacking interactions with neighboring Phe residues. To test this, we first examined the Jak2 structure via molecular modeling and identified a potential π stacking interaction between F594, F595, and F617. Disruption of this interaction through site-directed mutagenesis impaired Jak2 autophosphorylation, Jak2-dependent gene transcription, and in vitro kinase activity of the Jak2-V617F protein. Further, substitution of F594 and F595 with Trp did not affect Jak2 function significantly, but replacement with charged residues did, showing the importance of aromaticity and hydropathy index conservation at these positions. Using molecular dynamics (MD) simulations, we found that the π stacking interaction between residues 595 and 617 in the Jak2-V617F protein was of much greater energy and conformed to the properties of π stacking, relative to the Jak2-WT or Jak2-V617F/F594A/F595A. In summary, we have identified a π stacking interaction between F595 and F617 that is specific to and is critical for the constitutive activation of Jak2-V617F.


Subject(s)
Janus Kinase 2/chemistry , Phenylalanine/chemistry , Animals , COS Cells , Chlorocebus aethiops , Computer Simulation , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Models, Molecular , Mutation, Missense , Phenylalanine/genetics , Protein Conformation , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...