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










Publication year range
3.
bioRxiv ; 2023 Aug 03.
Article in English | MEDLINE | ID: mdl-37577651

ABSTRACT

MET is a receptor tyrosine kinase (RTK) responsible for initiating signaling pathways involved in development and wound repair. MET activation relies on ligand binding to the extracellular receptor, which prompts dimerization, intracellular phosphorylation, and recruitment of associated signaling proteins. Mutations, which are predominantly observed clinically in the intracellular juxtamembrane and kinase domains, can disrupt typical MET regulatory mechanisms. Understanding how juxtamembrane variants, such as exon 14 skipping (METΔEx14), and rare kinase domain mutations can increase signaling, often leading to cancer, remains a challenge. Here, we perform a parallel deep mutational scan (DMS) of MET intracellular kinase domain in two fusion protein backgrounds: wild type and METΔEx14. Our comparative approach has revealed a critical hydrophobic interaction between a juxtamembrane segment and the kinase αC helix, pointing to differences in regulatory mechanisms between MET and other RTKs. Additionally, we have uncovered a ß5 motif that acts as a structural pivot for kinase domain activation in MET and other TAM family of kinases. We also describe a number of previously unknown activating mutations, aiding the effort to annotate driver, passenger, and drug resistance mutations in the MET kinase domain.

4.
Structure ; 30(11): 1465-1467, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36332608

ABSTRACT

TRIB2, a member of the human Tribbles pseudokinase family, functions as a molecular scaffold in diverse signaling pathways. In this issue of Structure, Jamieson et al. report the first high-resolution structure of TRIB2 bound to a nanobody that offers insights into its "active-like" state.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases , Intracellular Signaling Peptides and Proteins , Humans , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction
5.
Nat Commun ; 12(1): 7032, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857742

ABSTRACT

Suppressor of cytokine signaling (SOCS)2 protein is a key negative regulator of the growth hormone (GH) and Janus kinase (JAK)-Signal Transducers and Activators of Transcription (STAT) signaling cascade. The central SOCS2-Src homology 2 (SH2) domain is characteristic of the SOCS family proteins and is an important module that facilitates recognition of targets bearing phosphorylated tyrosine (pTyr) residues. Here we identify an exosite on the SOCS2-SH2 domain which, when bound to a non-phosphorylated peptide (F3), enhances SH2 affinity for canonical phosphorylated ligands. Solution of the SOCS2/F3 crystal structure reveals F3 as an α-helix which binds on the opposite side of the SH2 domain to the phosphopeptide binding site. F3:exosite binding appears to stabilise the SOCS2-SH2 domain, resulting in slower dissociation of phosphorylated ligands and consequently, enhances binding affinity. This biophysical enhancement of SH2:pTyr binding affinity translates to increase SOCS2 inhibition of GH signaling.


Subject(s)
Suppressor of Cytokine Signaling Proteins/chemistry , Tyrosine/chemistry , A549 Cells , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Models, Molecular , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Tyrosine/metabolism
6.
Res Sq ; 2021 May 19.
Article in English | MEDLINE | ID: mdl-34031651

ABSTRACT

The SARS-CoV-2 protein Nsp2 has been implicated in a wide range of viral processes, but its exact functions, and the structural basis of those functions, remain unknown. Here, we report an atomic model for full-length Nsp2 obtained by combining cryo-electron microscopy with deep learning-based structure prediction from AlphaFold2. The resulting structure reveals a highly-conserved zinc ion-binding site, suggesting a role for Nsp2 in RNA binding. Mapping emerging mutations from variants of SARS-CoV-2 on the resulting structure shows potential host-Nsp2 interaction regions. Using structural analysis together with affinity tagged purification mass spectrometry experiments, we identify Nsp2 mutants that are unable to interact with the actin-nucleation-promoting WASH protein complex or with GIGYF2, an inhibitor of translation initiation and modulator of ribosome-associated quality control. Our work suggests a potential role of Nsp2 in linking viral transcription within the viral replication-transcription complexes (RTC) to the translation initiation of the viral message. Collectively, the structure reported here, combined with mutant interaction mapping, provides a foundation for functional studies of this evolutionary conserved coronavirus protein and may assist future drug design.

7.
bioRxiv ; 2021 May 11.
Article in English | MEDLINE | ID: mdl-34013269

ABSTRACT

The SARS-CoV-2 protein Nsp2 has been implicated in a wide range of viral processes, but its exact functions, and the structural basis of those functions, remain unknown. Here, we report an atomic model for full-length Nsp2 obtained by combining cryo-electron microscopy with deep learning-based structure prediction from AlphaFold2. The resulting structure reveals a highly-conserved zinc ion-binding site, suggesting a role for Nsp2 in RNA binding. Mapping emerging mutations from variants of SARS-CoV-2 on the resulting structure shows potential host-Nsp2 interaction regions. Using structural analysis together with affinity tagged purification mass spectrometry experiments, we identify Nsp2 mutants that are unable to interact with the actin-nucleation-promoting WASH protein complex or with GIGYF2, an inhibitor of translation initiation and modulator of ribosome-associated quality control. Our work suggests a potential role of Nsp2 in linking viral transcription within the viral replication-transcription complexes (RTC) to the translation initiation of the viral message. Collectively, the structure reported here, combined with mutant interaction mapping, provides a foundation for functional studies of this evolutionary conserved coronavirus protein and may assist future drug design.

8.
Biochem Soc Trans ; 49(2): 645-661, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33860789

ABSTRACT

The MET receptor tyrosine kinase (RTK) and its cognate ligand hepatocyte growth factor (HGF) comprise a signaling axis essential for development, wound healing and tissue homeostasis. Aberrant HGF/MET signaling is a driver of many cancers and contributes to drug resistance to several approved therapeutics targeting other RTKs, making MET itself an important drug target. In RTKs, homeostatic receptor signaling is dependent on autoinhibition in the absence of ligand binding and orchestrated set of conformational changes induced by ligand-mediated receptor dimerization that result in activation of the intracellular kinase domains. A fundamental understanding of these mechanisms in the MET receptor remains incomplete, despite decades of research. This is due in part to the complex structure of the HGF ligand, which remains unknown in its full-length form, and a lack of high-resolution structures of the complete MET extracellular portion in an apo or ligand-bound state. A current view of HGF-dependent MET activation has evolved from biochemical and structural studies of HGF and MET fragments and here we review what these findings have thus far revealed.


Subject(s)
Hepatocyte Growth Factor/chemistry , Hepatocyte Growth Factor/metabolism , Protein Domains , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/metabolism , Animals , Hepatocyte Growth Factor/genetics , Humans , Ligands , Models, Molecular , Mutation , Protein Binding , Proto-Oncogene Proteins c-met/genetics , Signal Transduction/genetics
9.
Sci Rep ; 11(1): 9161, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33911106

ABSTRACT

Over one billion people are currently infected with a parasitic nematode. Symptoms can include anemia, malnutrition, developmental delay, and in severe cases, death. Resistance is emerging to the anthelmintics currently used to treat nematode infection, prompting the need to develop new anthelmintics. Towards this end, we identified a set of kinases that may be targeted in a nematode-selective manner. We first screened 2040 inhibitors of vertebrate kinases for those that impair the model nematode Caenorhabditis elegans. By determining whether the terminal phenotype induced by each kinase inhibitor matched that of the predicted target mutant in C. elegans, we identified 17 druggable nematode kinase targets. Of these, we found that nematode EGFR, MEK1, and PLK1 kinases have diverged from vertebrates within their drug-binding pocket. For each of these targets, we identified small molecule scaffolds that may be further modified to develop nematode-selective inhibitors. Nematode EGFR, MEK1, and PLK1 therefore represent key targets for the development of new anthelmintic medicines.


Subject(s)
Anthelmintics/pharmacology , Caenorhabditis elegans/enzymology , Drug Evaluation, Preclinical/methods , Protein Kinase Inhibitors/pharmacology , Animals , Anthelmintics/chemistry , Caenorhabditis elegans/drug effects , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinases/chemistry , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Vertebrates
10.
Growth Factors ; 36(3-4): 104-117, 2018 08.
Article in English | MEDLINE | ID: mdl-30318950

ABSTRACT

The development and activity of our immune system are largely controlled by the action of pleiotropic cytokines and growth factors, small secreted proteins, which bind to receptors on the surface of immune cells to initiate an appropriate physiological response. Cytokine signalling is predominantly executed by intracellular proteins known as the Janus kinases (JAKs) and the signal transducers and activators of transcriptions (STATs). Although the 'nuts and bolts' of cytokine-activated pathways have been well established, the nuanced way in which distinct cellular outcomes are achieved and the precise molecular details of the proteins that regulate these pathways are still being elucidated. This is highlighted by the intricate role of the suppressor of cytokine signalling (SOCS) proteins. The SOCS proteins act as negative feedback inhibitors, dampening specific cytokine signals to prevent excessive cellular responses and returning the cell to a homeostatic state. A great deal of study has demonstrated their ability to inhibit these pathways at the receptor complex, either through direct inhibition of JAK activity or by targeting the receptor complex for proteasomal degradation. Detailed analysis of individual SOCS proteins is slowly revealing the complex and highly controlled manner by which they can achieve specificity for distinct substrates. However, for many of the SOCS, a level of detail is still lacking, including confident identification of the full suite of tyrosine phosphorylated targets of their SH2 domain. This review will highlight the general mechanisms which govern SOCS specificity of action and discuss the similarities and differences between selected SOCS proteins, focusing on CIS, SOCS1 and SOCS3. Because of the functional and sequence similarities within the SOCS family, we will also discuss the evidence for functional redundancy.


Subject(s)
Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Humans , Interleukins/metabolism , Protein Binding , Signal Transduction , Suppressor of Cytokine Signaling Proteins/chemistry , src Homology Domains
11.
Biochem J ; 475(2): 429-440, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29259080

ABSTRACT

The retinoic acid-inducible gene-I (RIG-I) receptor recognizes short 5'-di- and triphosphate base-paired viral RNA and is a critical mediator of the innate immune response against viruses such as influenza A, Ebola, HIV and hepatitis C. This response is reported to require an orchestrated interaction with the tripartite motif 25 (TRIM25) B30.2 protein-interaction domain. Here, we present a novel second RIG-I-binding interface on the TRIM25 B30.2 domain that interacts with CARD1 and CARD2 (caspase activation and recruitment domains) of RIG-I and is revealed by the removal of an N-terminal α-helix that mimics dimerization of the full-length protein. Further characterization of the TRIM25 coiled-coil and B30.2 regions indicated that the B30.2 domains move freely on a flexible tether, facilitating RIG-I CARD recruitment. The identification of a dual binding mode for the TRIM25 B30.2 domain is a first for the SPRY/B30.2 domain family and may be a feature of other SPRY/B30.2 family members.


Subject(s)
B30.2-SPRY Domain/genetics , Caspase Activation and Recruitment Domain/genetics , DEAD Box Protein 58/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Recombinant Fusion Proteins/chemistry , Sequence Deletion , Amino Acid Sequence , Animals , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , HEK293 Cells , Histidine/genetics , Histidine/metabolism , Humans , Mice , Models, Molecular , Oligopeptides/genetics , Oligopeptides/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Immunologic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
Elife ; 62017 02 14.
Article in English | MEDLINE | ID: mdl-28195529

ABSTRACT

Influenza virus infections have a significant impact on global human health. Individuals with suppressed immunity, or suffering from chronic inflammatory conditions such as COPD, are particularly susceptible to influenza. Here we show that suppressor of cytokine signaling (SOCS) five has a pivotal role in restricting influenza A virus in the airway epithelium, through the regulation of epidermal growth factor receptor (EGFR). Socs5-deficient mice exhibit heightened disease severity, with increased viral titres and weight loss. Socs5 levels were differentially regulated in response to distinct influenza viruses (H1N1, H3N2, H5N1 and H11N9) and were reduced in primary epithelial cells from COPD patients, again correlating with increased susceptibility to influenza. Importantly, restoration of SOCS5 levels restricted influenza virus infection, suggesting that manipulating SOCS5 expression and/or SOCS5 targets might be a novel therapeutic approach to influenza.


Subject(s)
Cytokines/metabolism , ErbB Receptors/antagonists & inhibitors , Influenza A virus/immunology , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Body Weight , Disease Models, Animal , Humans , Mice , Mice, Knockout , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Suppressor of Cytokine Signaling Proteins/deficiency , Viral Load
13.
Cereb Cortex ; 27(1): 576-588, 2017 01 01.
Article in English | MEDLINE | ID: mdl-26503265

ABSTRACT

Mutations of the reelin gene cause severe defects in cerebral cortex development and profound intellectual impairment. While many aspects of the reelin signaling pathway have been identified, the molecular and ultimate cellular consequences of reelin signaling remain unknown. Specifically, it is unclear if termination of reelin signaling is as important for normal cortical neuron migration as activation of reelin signaling. Using mice that are single or double deficient, we discovered that combined loss of the suppressors of cytokine signaling, SOCS6 and SOCS7, recapitulated the cortical layer inversion seen in mice lacking reelin and led to a dramatic increase in the reelin signaling molecule disabled (DAB1) in the cortex. The SRC homology domains of SOCS6 and SOCS7 bound DAB1 ex vivo. Mutation of DAB1 greatly diminished binding and protected from degradation by SOCS6. Phosphorylated DAB1 was elevated in cortical neurons in the absence of SOCS6 and SOCS7. Thus, constitutive activation of reelin signaling was observed to be equally detrimental as lack of activation. We hypothesize that, by terminating reelin signaling, SOCS6 and SOCS7 may allow new cycles of reelin signaling to occur and that these may be essential for cortical neuron migration.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Suppressor of Cytokine Signaling Proteins/deficiency , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Movement/physiology , Cerebral Cortex/pathology , Extracellular Matrix Proteins/genetics , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Neurons/metabolism , Phosphorylation , Reelin Protein , Serine Endopeptidases/genetics , Suppressor of Cytokine Signaling Proteins/genetics
14.
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
15.
Biochemistry ; 54(30): 4672-82, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26173083

ABSTRACT

SOCS5 can negatively regulate both JAK/STAT and EGF-receptor pathways and has therefore been implicated in regulating both the immune response and tumorigenesis. Understanding the molecular basis for SOCS5 activity may reveal novel ways to target key components of these signaling pathways. The N-terminal region of SOCS5 coordinates critical protein interactions involved in inhibition of JAK/STAT signaling, and a conserved region within the N-terminus of SOCS5 mediates direct binding to the JAK kinase domain. Here we have characterized the solution conformation of this conserved JAK interaction region (JIR) within the largely disordered N-terminus of SOCS5. Using nuclear magnetic resonance (NMR) chemical shift analysis, relaxation measurements, and NOE analysis, we demonstrate the presence of preformed structural elements in the JIR of mouse SOCS5 (mSOCS5175-244), consisting of an α-helix encompassing residues 224-233, preceded by a turn and an extended structure. We have identified a phosphorylation site (Ser211) within the JIR of mSOCS5 and have investigated the role of phosphorylation in modulating JAK binding using site-directed mutagenesis.


Subject(s)
Suppressor of Cytokine Signaling Proteins/chemistry , Amino Acid Substitution , Animals , Mice , Mutagenesis, Site-Directed , Mutation, Missense , Phosphorylation , Protein Structure, Secondary , Protein Structure, Tertiary , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
16.
Immunol Rev ; 266(1): 123-33, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26085211

ABSTRACT

The suppressor of cytokine signaling (SOCS) family of proteins are key negative regulators of cytokine and growth factor signaling. They act at the receptor complex to modulate the intracellular signaling cascade, preventing excessive signaling and restoring homeostasis. This regulation is critical to the normal cessation of signaling, highlighted by the complex inflammatory phenotypes exhibited by mice deficient in SOCS1 or SOCS3. These two SOCS proteins remain the best characterized of the eight family members (CIS, SOCS1-7), and in particular, we now possess a sound understanding of the mechanism of action for SOCS3. Here, we review the mechanistic role of the SOCS proteins and identify examples where clear, definitive data have been generated and discuss areas where the information is less clear. From this functional viewpoint, we discuss how the SOCS proteins achieve exquisite and specific regulation of cytokine signaling and highlight outstanding questions regarding the function of the less well-studied SOCS family members.


Subject(s)
Cytokines/metabolism , Proteasome Endopeptidase Complex/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Binding, Competitive , Cytokines/genetics , Humans , Mice , Mice, Knockout , Proteolysis , Signal Transduction/genetics , Suppressor of Cytokine Signaling Proteins/genetics
17.
PLoS Pathog ; 10(5): e1004134, 2014 May.
Article in English | MEDLINE | ID: mdl-24809749

ABSTRACT

Suppressor of cytokine signaling (SOCS) proteins are key regulators of innate and adaptive immunity. There is no described biological role for SOCS4, despite broad expression in the hematopoietic system. We demonstrate that mice lacking functional SOCS4 protein rapidly succumb to infection with a pathogenic H1N1 influenza virus (PR8) and are hypersusceptible to infection with the less virulent H3N2 (X31) strain. In SOCS4-deficient animals, this led to substantially greater weight loss, dysregulated pro-inflammatory cytokine and chemokine production in the lungs and delayed viral clearance. This was associated with impaired trafficking of influenza-specific CD8 T cells to the site of infection and linked to defects in T cell receptor activation. These results demonstrate that SOCS4 is a critical regulator of anti-viral immunity.


Subject(s)
Adaptive Immunity/genetics , Cytokines/adverse effects , Cytokines/metabolism , Inflammation/prevention & control , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/immunology , Suppressor of Cytokine Signaling Proteins/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Cytoprotection/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/adverse effects , Inflammation Mediators/metabolism , Influenza A Virus, H1N1 Subtype/growth & development , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/virology , Viral Load/genetics
18.
Arthritis Rheumatol ; 66(9): 2391-402, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24839265

ABSTRACT

OBJECTIVE: To examine the impact of the gp130 cytokine family on murine articular cartilage and to explore a potential regulatory role of suppressor of cytokine signaling 3 (SOCS-3) in murine chondrocytes. METHODS: In wild-type (WT) mouse chondrocytes, baseline receptor expression levels and gp130 cytokine-induced JAK/STAT signaling were determined by flow cytometry, and expression of SOCS-3 was assessed by quantitative polymerase chain reaction. The role of endogenous SOCS-3 was examined in cartilage explants and chondrocytes from mice with conditional deletion of Socs3 driven by the Col2a1 promoter in vitro (Socs3(Δ/Δcol2) ) and from mice during CD4+ T cell-dependent inflammatory monarthritis. Bone erosions in the murine joints were analyzed by micro-computed tomography. RESULTS: On chondrocytes from WT mice, gp130 and the oncostatin M (OSM) receptor were strongly expressed, whereas the transmembrane interleukin-6 (IL-6) receptor was expressed at much lower levels. Compared to other gp130 cytokines, OSM was the most potent activator of the JAK/STAT pathway and of SOCS-3 induction. Treatment of Socs3(Δ/Δcol2) mouse cartilage explants and chondrocytes with gp130 cytokines prolonged JAK/STAT signaling, enhanced cartilage degradation, increased the expression of Adamts4, Adamts5, and RANKL, and elevated the production of IL-6, granulocyte colony-stimulating factor, CXCL1, and CCL2. Socs3(Δ/Δcol2) mice developed exacerbated inflammation and joint damage in response to gp130 cytokine injections, and these histopathologic features were also observed in mice with inflammatory monarthritis. CONCLUSION: The results of this study highlight a key role for SOCS-3 in regulating chondrocyte responses during inflammatory arthritis. Within the gp130 cytokine family, OSM is a potent stimulus of chondrocyte responses, while IL-6 probably signals via trans-signaling. The gp130 cytokine-driven production of RANKL in chondrocytes may link chondrocyte activation and bone remodeling during inflammatory arthritis. Thus, these findings suggest that the inhibition of OSM might reduce the development and severity of structural joint damage during inflammatory arthritis.


Subject(s)
Arthritis, Experimental/metabolism , Chondrocytes/metabolism , Cytokine Receptor gp130/metabolism , Signal Transduction/physiology , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cartilage, Articular/metabolism , Cytokine Receptor gp130/genetics , Knee Joint/metabolism , Mice , Mice, Knockout , Oncostatin M/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics
19.
PLoS One ; 8(8): e70536, 2013.
Article in English | MEDLINE | ID: mdl-23990909

ABSTRACT

Suppressor of Cytokine Signaling (SOCS)5 is thought to act as a tumour suppressor through negative regulation of JAK/STAT and epidermal growth factor (EGF) signaling. However, the mechanism/s by which SOCS5 acts on these two distinct pathways is unclear. We show for the first time that SOCS5 can interact directly with JAK via a unique, conserved region in its N-terminus, which we have termed the JAK interaction region (JIR). Co-expression of SOCS5 was able to specifically reduce JAK1 and JAK2 (but not JAK3 or TYK2) autophosphorylation and this function required both the conserved JIR and additional sequences within the long SOCS5 N-terminal region. We further demonstrate that SOCS5 can directly inhibit JAK1 kinase activity, although its mechanism of action appears distinct from that of SOCS1 and SOCS3. In addition, we identify phosphoTyr317 in Shc-1 as a high-affinity substrate for the SOCS5-SH2 domain and suggest that SOCS5 may negatively regulate EGF and growth factor-driven Shc-1 signaling by binding to this site. These findings suggest that different domains in SOCS5 contribute to two distinct mechanisms for regulation of cytokine and growth factor signaling.


Subject(s)
Janus Kinase 1/metabolism , Shc Signaling Adaptor Proteins/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Binding Sites , Cytokines/metabolism , Genetic Vectors , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Janus Kinase 2/metabolism , Janus Kinase 3/metabolism , Mice , Phosphopeptides/chemistry , Phosphorylation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1 , Substrate Specificity , Surface Plasmon Resonance , TYK2 Kinase/metabolism , src Homology Domains
20.
Cytokine Growth Factor Rev ; 24(3): 241-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23545160

ABSTRACT

The discovery of the Suppressor of Cytokine Signaling (SOCS) family of proteins has resulted in a significant body of research dedicated to dissecting their biological functions and the molecular mechanisms by which they achieve potent and specific inhibition of cytokine and growth factor signaling. The Australian contribution to this field has been substantial, with the initial discovery of SOCS1 by Hilton, Starr and colleagues (discovered concurrently by two other groups) and the following work, providing a new perspective on the regulation of JAK/STAT signaling. In this review, we reflect on the critical discoveries that have lead to our current understanding of how SOCS proteins function and discuss what we see as important questions for future research.


Subject(s)
Cytokines/physiology , Signal Transduction/physiology , Suppressor of Cytokine Signaling Proteins/physiology , Animals , Humans , Janus Kinases , STAT Transcription Factors/drug effects , Signal Transduction/drug effects , Suppressor of Cytokine Signaling Proteins/chemistry , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...