Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
3.
Nat Commun ; 14(1): 5053, 2023 08 19.
Article in English | MEDLINE | ID: mdl-37598178

ABSTRACT

Brain exposure of systemically administered biotherapeutics is highly restricted by the blood-brain barrier (BBB). Here, we report the engineering and characterization of a BBB transport vehicle targeting the CD98 heavy chain (CD98hc or SLC3A2) of heterodimeric amino acid transporters (TVCD98hc). The pharmacokinetic and biodistribution properties of a CD98hc antibody transport vehicle (ATVCD98hc) are assessed in humanized CD98hc knock-in mice and cynomolgus monkeys. Compared to most existing BBB platforms targeting the transferrin receptor, peripherally administered ATVCD98hc demonstrates differentiated brain delivery with markedly slower and more prolonged kinetic properties. Specific biodistribution profiles within the brain parenchyma can be modulated by introducing Fc mutations on ATVCD98hc that impact FcγR engagement, changing the valency of CD98hc binding, and by altering the extent of target engagement with Fabs. Our study establishes TVCD98hc as a modular brain delivery platform with favorable kinetic, biodistribution, and safety properties distinct from previously reported BBB platforms.


Subject(s)
Blood-Brain Barrier , Brain , Animals , Mice , Tissue Distribution , Antibodies , Engineering , Macaca fascicularis
4.
Commun Biol ; 6(1): 641, 2023 06 14.
Article in English | MEDLINE | ID: mdl-37316570

ABSTRACT

Protein Tyrosine Phosphatase 1B (PTP1B) is the prototypical protein tyrosine phosphatase and plays an essential role in the regulation of several kinase-driven signalling pathways. PTP1B displays a preference for bisphosphorylated substrates. Here we identify PTP1B as an inhibitor of IL-6 and show that, in vitro, it can dephosphorylate all four members of the JAK family. In order to gain a detailed understanding of the molecular mechanism of JAK dephosphorylation, we undertook a structural and biochemical analysis of the dephosphorylation reaction. We identified a product-trapping PTP1B mutant that allowed visualisation of the tyrosine and phosphate products of the reaction and a substrate-trapping mutant with a vastly decreased off-rate compared to those previously described. The latter mutant was used to determine the structure of bisphosphorylated JAK peptides bound to the enzyme active site. These structures revealed that the downstream phosphotyrosine preferentially engaged the active site, in contrast to the analogous region of IRK. Biochemical analysis confirmed this preference. In this binding mode, the previously identified second aryl binding site remains unoccupied and the non-substrate phosphotyrosine engages Arg47. Mutation of this arginine disrupts the preference for the downstream phosphotyrosine. This study reveals a previously unappreciated plasticity in how PTP1B interacts with different substrates.


Subject(s)
Janus Kinases , Phosphoric Monoester Hydrolases , Phosphotyrosine , Arginine , Binding Sites
5.
Bioorg Med Chem Lett ; 89: 129277, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37105490

ABSTRACT

Inhibition of NF-κB inducing kinase (NIK) has been pursued as a promising therapeutic target for autoimmune disorders due to its highly regulated role in key steps of the NF-κB signaling pathway. Previously reported NIK inhibitors from our group were shown to be potent, selective, and efficacious, but had higher human dose projections than desirable for immunology indications. Herein we report the clearance-driven optimization of a NIK inhibitor guided by metabolite identification studies and structure-based drug design. This led to the identification of an azabicyclo[3.1.0]hexanone motif that attenuated in vitro and in vivo clearance while maintaining NIK potency and increasing selectivity over other kinases, resulting in a greater than ten-fold reduction in predicted human dose.


Subject(s)
NF-kappa B , Signal Transduction , Humans , NF-kappa B/metabolism , Half-Life , Drug Design
6.
ACS Chem Biol ; 18(4): 772-784, 2023 04 21.
Article in English | MEDLINE | ID: mdl-36893429

ABSTRACT

Wnt ligands are critical for tissue homeostasis and form a complex with LRP6 and frizzled coreceptors to initiate Wnt/ß-catenin signaling. Yet, how different Wnts achieve various levels of signaling activation through distinct domains on LRP6 remains elusive. Developing tool ligands that target individual LRP6 domains could help elucidate the mechanism of Wnt signaling regulation and uncover pharmacological approaches for pathway modulation. We employed directed evolution of a disulfide constrained peptide (DCP) to identify molecules that bind to the third ß-propeller domain of LRP6. The DCPs antagonize Wnt3a while sparing Wnt1 signaling. Using PEG linkers with different geometries, we converted the Wnt3a antagonist DCPs to multivalent molecules that potentiated Wnt1 signaling by clustering the LRP6 coreceptor. The mechanism of potentiation is unique as it occurred only in the presence of extracellular secreted Wnt1 ligand. While all DCPs recognized a similar binding interface on LRP6, they displayed different spatial orientations that influenced their cellular activities. Moreover, structural analyses revealed that the DCPs exhibited new folds that were distinct from the parent DCP framework they were evolved from. The multivalent ligand design principles highlighted in this study provide a path for developing peptide agonists that modulate different branches of cellular Wnt signaling.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-6 , Wnt Proteins , Ligands , Wnt Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , beta Catenin/metabolism , Protein Binding , Wnt Signaling Pathway , Peptides/pharmacology , Peptides/metabolism
7.
Nature ; 609(7926): 400-407, 2022 09.
Article in English | MEDLINE | ID: mdl-35768504

ABSTRACT

The RAS-RAF pathway is one of the most commonly dysregulated in human cancers1-3. Despite decades of study, understanding of the molecular mechanisms underlying dimerization and activation4 of the kinase RAF remains limited. Recent structures of inactive RAF monomer5 and active RAF dimer5-8 bound to 14-3-39,10 have revealed the mechanisms by which 14-3-3 stabilizes both RAF conformations via specific phosphoserine residues. Prior to RAF dimerization, the protein phosphatase 1 catalytic subunit (PP1C) must dephosphorylate the N-terminal phosphoserine (NTpS) of RAF11 to relieve inhibition by 14-3-3, although PP1C in isolation lacks intrinsic substrate selectivity. SHOC2 is as an essential scaffolding protein that engages both PP1C and RAS to dephosphorylate RAF NTpS11-13, but the structure of SHOC2 and the architecture of the presumptive SHOC2-PP1C-RAS complex remain unknown. Here we present a cryo-electron microscopy structure of the SHOC2-PP1C-MRAS complex to an overall resolution of 3 Å, revealing a tripartite molecular architecture in which a crescent-shaped SHOC2 acts as a cradle and brings together PP1C and MRAS. Our work demonstrates the GTP dependence of multiple RAS isoforms for complex formation, delineates the RAS-isoform preference for complex assembly, and uncovers how the SHOC2 scaffold and RAS collectively drive specificity of PP1C for RAF NTpS. Our data indicate that disease-relevant mutations affect complex assembly, reveal the simultaneous requirement of two RAS molecules for RAF activation, and establish rational avenues for discovery of new classes of inhibitors to target this pathway.


Subject(s)
Intracellular Signaling Peptides and Proteins , Protein Phosphatase 1 , Signal Transduction , ras Proteins , Cryoelectron Microscopy , Guanosine Triphosphate/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Mutation , Phosphoserine , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/ultrastructure , Protein Phosphatase 1/chemistry , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/ultrastructure , Substrate Specificity , raf Kinases/metabolism , ras Proteins/chemistry , ras Proteins/genetics , ras Proteins/metabolism , ras Proteins/ultrastructure
8.
Cell Rep ; 38(6): 110351, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35139374

ABSTRACT

KRAS, which is mutated in ∼30% of all cancers, activates the RAF-MEK-ERK signaling cascade. CRAF is required for growth of KRAS mutant lung tumors, but the requirement for CRAF kinase activity is unknown. Here, we show that subsets of KRAS mutant tumors are dependent on CRAF for growth. Kinase-dead but not dimer-defective CRAF rescues growth inhibition, suggesting that dimerization but not kinase activity is required. Quantitative proteomics demonstrates increased levels of CRAF:ARAF dimers in KRAS mutant cells, and depletion of both CRAF and ARAF rescues the CRAF-loss phenotype. Mechanistically, CRAF depletion causes sustained ERK activation and induction of cell-cycle arrest, while treatment with low-dose MEK or ERK inhibitor rescues the CRAF-loss phenotype. Our studies highlight the role of CRAF in regulating MAPK signal intensity to promote tumorigenesis downstream of mutant KRAS and suggest that disrupting CRAF dimerization or degrading CRAF may have therapeutic benefit.


Subject(s)
Carcinogenesis/metabolism , Dimerization , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Carcinogenesis/drug effects , Cell Line, Tumor , Humans , MAP Kinase Signaling System/drug effects , Mice , Phosphorylation/physiology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , ras Proteins/genetics
9.
Nature ; 594(7863): 418-423, 2021 06.
Article in English | MEDLINE | ID: mdl-33953400

ABSTRACT

Although RAF monomer inhibitors (type I.5, BRAF(V600)) are clinically approved for the treatment of BRAFV600-mutant melanoma, they are ineffective in non-BRAFV600 mutant cells1-3. Belvarafenib is a potent and selective RAF dimer (type II) inhibitor that exhibits clinical activity in patients with BRAFV600E- and NRAS-mutant melanomas. Here we report the first-in-human phase I study investigating the maximum tolerated dose, and assessing the safety and preliminary efficacy of belvarafenib in BRAFV600E- and RAS-mutated advanced solid tumours (NCT02405065, NCT03118817). By generating belvarafenib-resistant NRAS-mutant melanoma cells and analysing circulating tumour DNA from patients treated with belvarafenib, we identified new recurrent mutations in ARAF within the kinase domain. ARAF mutants conferred resistance to belvarafenib in both a dimer- and a kinase activity-dependent manner. Belvarafenib induced ARAF mutant dimers, and dimers containing mutant ARAF were active in the presence of inhibitor. ARAF mutations may serve as a general resistance mechanism for RAF dimer inhibitors as the mutants exhibit reduced sensitivity to a panel of type II RAF inhibitors. The combination of RAF plus MEK inhibition may be used to delay ARAF-driven resistance and suggests a rational combination for clinical use. Together, our findings reveal specific and compensatory functions for the ARAF isoform and implicate ARAF mutations as a driver of resistance to RAF dimer inhibitors.


Subject(s)
Drug Resistance, Neoplasm/genetics , Melanoma/drug therapy , Melanoma/genetics , Mutation , Proto-Oncogene Proteins A-raf/antagonists & inhibitors , Proto-Oncogene Proteins A-raf/genetics , raf Kinases/antagonists & inhibitors , Animals , Cell Line , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Humans , Melanoma/pathology , Mice , Protein Multimerization/drug effects , Proto-Oncogene Proteins A-raf/chemistry , raf Kinases/chemistry
10.
Biochemistry ; 59(41): 3982-3992, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32970425

ABSTRACT

The Ras-RAF-MEK-ERK signaling axis, commonly mutated in human cancers, is highly regulated to prevent aberrant signaling in healthy cells. One of the pathway modulators, 14-3-3, a constitutive dimer, induces RAF dimerization and activation by binding to a phosphorylated motif C-terminal to the RAF kinase domain. Recent work has suggested that a C-terminal "DTS" region in BRAF is necessary for this 14-3-3-mediated activation. We show that the catalytic activity and ATP binding affinity of the BRAF:14-3-3 complex is insensitive to the presence or absence of the DTS, while the ATP sites of both BRAF molecules are identical and available for binding. We also present a crystal structure of the apo BRAF:14-3-3 complex showing that the DTS is not required to attain the catalytically active conformation of BRAF. Rather, BRAF dimerization induced by 14-3-3 is the key step in activation, allowing the active BRAF:14-3-3 tetramer to achieve catalytic activity comparable to the constitutively active oncogenic BRAF V600E mutant.


Subject(s)
14-3-3 Proteins/chemistry , 14-3-3 Proteins/metabolism , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Adenosine Triphosphate/metabolism , Catalysis , Humans , Protein Binding , Protein Multimerization , Signal Transduction
11.
Nat Struct Mol Biol ; 27(2): 134-141, 2020 02.
Article in English | MEDLINE | ID: mdl-31988522

ABSTRACT

The RAS-RAF-MEK-ERK signaling axis is frequently activated in human cancers. Physiological concentrations of ATP prevent formation of RAF kinase-domain (RAFKD) dimers that are critical for activity. Here we present a 2.9-Å-resolution crystal structure of human BRAFKD in complex with MEK and the ATP analog AMP-PCP, revealing interactions between BRAF and ATP that induce an inactive, monomeric conformation of BRAFKD. We also determine how 14-3-3 relieves the negative regulatory effect of ATP through a 2.5-Å-resolution crystal structure of the BRAFKD-14-3-3 complex, in which dimeric 14-3-3 enforces a dimeric BRAFKD assembly to increase BRAF activity. Our data suggest that most oncogenic BRAF mutations alter interactions with ATP and counteract the negative effects of ATP binding by lowering the threshold for RAF dimerization and pathway activation. Our study establishes a framework for rationalizing oncogenic BRAF mutations and provides new avenues for improved RAF-inhibitor discovery.


Subject(s)
14-3-3 Proteins/metabolism , Adenosine Triphosphate/metabolism , Proto-Oncogene Proteins B-raf/metabolism , 14-3-3 Proteins/chemistry , Adenosine Triphosphate/analogs & derivatives , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/metabolism , Crystallography, X-Ray , Humans , Molecular Dynamics Simulation , Protein Conformation , Protein Multimerization , Proto-Oncogene Proteins B-raf/chemistry
12.
Cancers (Basel) ; 11(11)2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31683831

ABSTRACT

Janus kinases (JAKs) are found constitutively associated with cytokine receptors and are present in an inactive state prior to cytokine exposure. Activating mutations of JAKs are causative for a number of leukemias, lymphomas, and myeloproliferative diseases. In particular, the JAK2V617F mutant is found in most human cases of polycythemia vera, a disease characterized by over-production of erythrocytes. The V617F mutation is found in the pseudokinase domain of JAK2 and it leads to cytokine-independent activation of the kinase, as does the orthologous mutation in other JAK-family members. The mechanism whereby this mutation hyperactivates these kinases is not well understood, primarily due to the fact that the full-length JAK proteins are difficult to produce for structural and kinetic studies. Here we have overcome this limitation to perform a series of enzymatic analyses on full-length JAK1 and its constitutively active mutant form (JAK1V658F). Consistent with previous studies, we show that the presence of the pseudokinase domain leads to a dramatic decrease in enzymatic activity with no further decrease from the presence of the FERM or SH2 domains. However, we find that the mutant kinase, in vitro, is indistinguishable from the wild-type enzyme in every measurable parameter tested: KM (ATP), KM (substrate), kcat, receptor binding, thermal stability, activation rate, dephosphorylation rate, and inhibitor affinity. These results show that the V658F mutation does not enhance the intrinsic enzymatic activity of JAK. Rather this data is more consistent with a model in which there are cellular processes and interactions that prevent JAK from being activated in the absence of cytokine and it is these constraints that are affected by disease-causing mutations.

13.
Sci Adv ; 4(11): eaat3834, 2018 11.
Article in English | MEDLINE | ID: mdl-30498775

ABSTRACT

Treatment of patients with myelofibrosis with the type I JAK (Janus kinase) inhibitor ruxolitinib paradoxically induces JAK2 activation loop phosphorylation and is associated with a life-threatening cytokine-rebound syndrome if rapidly withdrawn. We developed a time-dependent assay to mimic ruxolitinib withdrawal in primary JAK2V617F and CALR mutant myelofibrosis patient samples and observed notable activation of spontaneous STAT signaling in JAK2V617F samples after drug washout. Accumulation of ruxolitinib-induced JAK2 phosphorylation was dose dependent and correlated with rebound signaling and the presence of a JAK2V617F mutation. Ruxolitinib prevented dephosphorylation of a cryptic site involving Tyr1007/1008 in JAK2 blocking ubiquitination and degradation. In contrast, a type II JAK inhibitor, CHZ868, did not induce JAK2 phosphorylation, was not associated with withdrawal signaling, and was superior in the eradication of flow-purified JAK2V617F mutant CD34+ progenitors after drug washout. Type I inhibitor-induced loop phosphorylation may act as a pathogenic signaling node released upon drug withdrawal, especially in JAK2V617F patients.


Subject(s)
Janus Kinase 2/metabolism , Janus Kinase Inhibitors/pharmacology , Primary Myelofibrosis/metabolism , Pyrazoles/pharmacology , Substance Withdrawal Syndrome/pathology , Apoptosis , Cell Proliferation , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Mutation , Nitriles , Phosphorylation , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/pathology , Pyrimidines , Signal Transduction , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/metabolism , Tumor Cells, Cultured
14.
Nat Commun ; 9(1): 1558, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29674694

ABSTRACT

The SOCS family of proteins are negative-feedback inhibitors of signalling induced by cytokines that act via the JAK/STAT pathway. SOCS proteins can act as ubiquitin ligases by recruiting Cullin5 to ubiquitinate signalling components; however, SOCS1, the most potent member of the family, can also inhibit JAK directly. Here we determine the structural basis of both these modes of inhibition. Due to alterations within the SOCS box domain, SOCS1 has a compromised ability to recruit Cullin5; however, it is a direct, potent and selective inhibitor of JAK catalytic activity. The kinase inhibitory region of SOCS1 targets the substrate binding groove of JAK with high specificity and thereby blocks any subsequent phosphorylation. SOCS1 is a potent inhibitor of the interferon gamma (IFNγ) pathway, however, it does not bind the IFNγ receptor, making its mode-of-action distinct from SOCS3. These findings reveal the mechanism used by SOCS1 to inhibit signalling by inflammatory cytokines.


Subject(s)
Janus Kinase 1/chemistry , Janus Kinase 2/chemistry , Janus Kinase Inhibitors/chemistry , Suppressor of Cytokine Signaling 1 Protein/chemistry , Binding Sites , Crystallography, X-Ray , Cullin Proteins/genetics , Cullin Proteins/metabolism , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Janus Kinase Inhibitors/metabolism , Models, Molecular , Phosphorylation , Protein Domains , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/chemistry , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism
15.
Methods Mol Biol ; 1725: 267-280, 2018.
Article in English | MEDLINE | ID: mdl-29322424

ABSTRACT

Interferon gamma (IFNγ) is a potent inflammatory and immune cytokine. IFNγ signals via the interferon gamma receptor (IFNGR), which is constitutively bound to Janus Kinase (JAK) 1 and JAK2 via its intracellular domain. These two JAK proteins then initiate the inflammatory signaling cascade. The most potent inhibitor of IFNγ signaling is Suppressor of Cytokine Signaling 1 (SOCS1). SOCS1 negatively regulates IFNγ signaling pathway (and other pathways) by directly inhibiting JAKs. Here, we describe a protocol for the recombinant production and purification of the JAK1 kinase domain and its inhibitor SOCS1, for structural and biochemical studies.


Subject(s)
Janus Kinase 1/isolation & purification , Janus Kinase 1/metabolism , Suppressor of Cytokine Signaling 1 Protein/isolation & purification , Suppressor of Cytokine Signaling 1 Protein/metabolism , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Cells, Cultured , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Janus Kinase 1/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sf9 Cells , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/genetics
16.
Methods Mol Biol ; 1555: 173-182, 2017.
Article in English | MEDLINE | ID: mdl-28092033

ABSTRACT

Src Homology 2 (SH2) domains are protein domains which have a high binding affinity for specific amino acid sequences containing a phosphorylated tyrosine residue. The Suppressors of Cytokine Signaling (SOCS) proteins use an SH2 domain to bind to components of certain cytokine signaling pathways to downregulate the signaling cascade. The recombinantly produced SH2 domains of various SOCS proteins have been used to undertake structural and functional studies elucidating the method of how such targeting occurs. Here, we describe the protocol for the recombinant production and purification of SOCS SH2 domains, with an emphasis on SOCS3.


Subject(s)
Recombinant Proteins , Suppressor of Cytokine Signaling Proteins/isolation & purification , Cloning, Molecular , Cytokines/chemistry , Cytokines/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Janus Kinases/chemistry , Janus Kinases/genetics , Janus Kinases/isolation & purification , Janus Kinases/metabolism , Protein Binding , Protein Refolding , Recombinant Fusion Proteins , Signal Transduction , Solubility , Structure-Activity Relationship , Suppressor of Cytokine Signaling Proteins/chemistry , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , src Homology Domains
17.
Structure ; 24(6): 840-2, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27276423

ABSTRACT

Janus kinases (JAKs) initiate the intracellular signaling cascade triggered by exposure of cells to cytokines and interferons. In order to achieve this, JAKs are bound to the intracellular domain of specific cytokine receptors immediately adjacent to the cell membrane. In this issue of Structure, Ferrao et al. (2016) provide structural details of such an interaction and in doing so, identify for the first time the motif used by type II cytokine receptors to recruit JAK1.


Subject(s)
Janus Kinase 1/chemistry , Receptors, Cytokine/chemistry , Cell Membrane , Protein-Tyrosine Kinases/chemistry , Signal Transduction
18.
Nat Immunol ; 17(7): 816-24, 2016 07.
Article in English | MEDLINE | ID: mdl-27213690

ABSTRACT

The detection of aberrant cells by natural killer (NK) cells is controlled by the integration of signals from activating and inhibitory ligands and from cytokines such as IL-15. We identified cytokine-inducible SH2-containing protein (CIS, encoded by Cish) as a critical negative regulator of IL-15 signaling in NK cells. Cish was rapidly induced in response to IL-15, and deletion of Cish rendered NK cells hypersensitive to IL-15, as evidenced by enhanced proliferation, survival, IFN-γ production and cytotoxicity toward tumors. This was associated with increased JAK-STAT signaling in NK cells in which Cish was deleted. Correspondingly, CIS interacted with the tyrosine kinase JAK1, inhibiting its enzymatic activity and targeting JAK for proteasomal degradation. Cish(-/-) mice were resistant to melanoma, prostate and breast cancer metastasis in vivo, and this was intrinsic to NK cell activity. Our data uncover a potent intracellular checkpoint in NK cell-mediated tumor immunity and suggest possibilities for new cancer immunotherapies directed at blocking CIS function.


Subject(s)
Immunotherapy/methods , Killer Cells, Natural/immunology , Neoplasms/therapy , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cell Proliferation/genetics , Cytotoxicity, Immunologic/genetics , Immunologic Surveillance , Interferon-gamma/metabolism , Interleukin-15/metabolism , Janus Kinase 1/metabolism , Lymphocyte Activation/genetics , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Neoplasms/immunology , Signal Transduction/genetics , Suppressor of Cytokine Signaling Proteins/genetics
19.
Biochem J ; 458(2): 395-405, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24354892

ABSTRACT

JAK2 (Janus kinase 2) initiates the intracellular signalling cascade downstream of cell surface receptor activation by cognate haemopoietic cytokines, including erythropoietin and thrombopoietin. The pseudokinase domain (JH2) of JAK2 negatively regulates the catalytic activity of the adjacent tyrosine kinase domain (JH1) and mutations within the pseudokinase domain underlie human myeloproliferative neoplasms, including polycythaemia vera and essential thrombocytosis. To date, the mechanism of JH2-mediated inhibition of JH1 kinase activation as well as the susceptibility of pathological mutant JAK2 to inhibition by the physiological negative regulator SOCS3 (suppressor of cytokine signalling 3) have remained unclear. In the present study, using recombinant purified JAK2JH1-JH2 proteins, we demonstrate that, when activated, wild-type and myeloproliferative neoplasm-associated mutants of JAK2 exhibit comparable enzymatic activity and inhibition by SOCS3 in in vitro kinase assays. SAXS (small-angle X-ray scattering) showed that JAK2JH1-JH2 exists in an elongated configuration in solution with no evidence for interaction between JH1 and JH2 domains in cis. Collectively, these data are consistent with a model in which JAK2's pseudokinase domain does not influence the activity of JAK2 once it has been activated. Our data indicate that, in the absence of the N-terminal FERM domain and thus cytokine receptor association, the wild-type and pathological mutants of JAK2 are enzymatically equivalent and equally susceptible to inhibition by SOCS3.


Subject(s)
Hematologic Neoplasms/prevention & control , Janus Kinase 2/antagonists & inhibitors , Mutation, Missense/genetics , Myeloproliferative Disorders/prevention & control , Suppressor of Cytokine Signaling Proteins/physiology , Catalytic Domain/genetics , Genetic Predisposition to Disease , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Protein Structure, Secondary/genetics , Recombinant Proteins/genetics , Scattering, Small Angle , Signal Transduction/genetics , Signal Transduction/physiology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , X-Ray Diffraction
20.
Nat Struct Mol Biol ; 20(4): 469-76, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23454976

ABSTRACT

The inhibitory protein SOCS3 plays a key part in the immune and hematopoietic systems by regulating signaling induced by specific cytokines. SOCS3 functions by inhibiting the catalytic activity of Janus kinases (JAKs) that initiate signaling within the cell. We determined the crystal structure of a ternary complex between mouse SOCS3, JAK2 (kinase domain) and a fragment of the interleukin-6 receptor ß-chain. The structure shows that SOCS3 binds JAK2 and receptor simultaneously, using two opposing surfaces. While the phosphotyrosine-binding groove on the SOCS3 SH2 domain is occupied by receptor, JAK2 binds in a phosphoindependent manner to a noncanonical surface. The kinase-inhibitory region of SOCS3 occludes the substrate-binding groove on JAK2, and biochemical studies show that it blocks substrate association. These studies reveal that SOCS3 targets specific JAK-cytokine receptor pairs and explains the mechanism and specificity of SOCS action.


Subject(s)
Cytokines/metabolism , Janus Kinase 2/metabolism , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism , Amino Acid Sequence , Binding Sites , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/chemistry , Models, Molecular , Molecular Sequence Data , Protein Binding , Sequence Homology, Amino Acid , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...