Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters

Country/Region as subject
Publication year range
2.
Cytokine ; 165: 156167, 2023 05.
Article in English | MEDLINE | ID: mdl-36934508

ABSTRACT

Suppressor Of Cytokine Signaling (SOCS) 1 is a critical negative regulator of cytokine signaling and required to protect against an excessive inflammatory response. Genetic deletion of Socs1 results in unrestrained cytokine signaling and neonatal lethality, characterised by an inflammatory immune infiltrate in multiple organs. Overexpression and structural studies have suggested that the SOCS1 kinase inhibitory region (KIR) and Src homology 2 (SH2) domain are important for interaction with and inhibition of the receptor-associated JAK1, JAK2 and TYK2 tyrosine kinases, which initiate downstream signaling. To investigate the role of the KIR and SH2 domain in SOCS1 function, we independently mutated key conserved residues in each domain and analysed the impact on cytokine signaling, and the in vivo impact on SOCS1 function. Mutation of the SOCS1-KIR or SH2 domain had no impact on the integrity of the SOCS box complex, however, mutation within the phosphotyrosine binding pocket of the SOCS1-SH2 domain specifically disrupted SOCS1 interaction with phosphorylated JAK1. In contrast, mutation of the KIR did not affect the interaction with JAK1, but did prevent SOCS1 inhibition of JAK1 autophosphorylation. In human and mouse cell lines, both mutants impacted the ability of SOCS1 to restrain cytokine signaling, and crucially, Socs1-R105A and Socs1-F59A mice displayed a neonatal lethality and excessive inflammatory phenotype similar to Socs1-null mice. This study defines a critical and non-redundant role for both the KIR and SH2 domain in endogenous SOCS1 function.


Subject(s)
Cytokines , Suppressor of Cytokine Signaling 1 Protein , src Homology Domains , Animals , Humans , Mice , Cytokines/metabolism , Phosphorylation , Signal Transduction/physiology , Suppressor of Cytokine Signaling 1 Protein/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , TYK2 Kinase/metabolism
3.
Immunity ; 39(3): 443-53, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-24012422

ABSTRACT

Mixed lineage kinase domain-like (MLKL) is a component of the "necrosome," the multiprotein complex that triggers tumor necrosis factor (TNF)-induced cell death by necroptosis. To define the specific role and molecular mechanism of MLKL action, we generated MLKL-deficient mice and solved the crystal structure of MLKL. Although MLKL-deficient mice were viable and displayed no hematopoietic anomalies or other obvious pathology, cells derived from these animals were resistant to TNF-induced necroptosis unless MLKL expression was restored. Structurally, MLKL comprises a four-helical bundle tethered to the pseudokinase domain, which contains an unusual pseudoactive site. Although the pseudokinase domain binds ATP, it is catalytically inactive and its essential nonenzymatic role in necroptotic signaling is induced by receptor-interacting serine-threonine kinase 3 (RIPK3)-mediated phosphorylation. Structure-guided mutation of the MLKL pseudoactive site resulted in constitutive, RIPK3-independent necroptosis, demonstrating that modification of MLKL is essential for propagation of the necroptosis pathway downstream of RIPK3.


Subject(s)
Apoptosis , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factors/metabolism , Animals , Catalytic Domain , Cell Line , Crystallography, X-Ray , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Phosphoprotein Phosphatases , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Kinases/chemistry , Protein Kinases/genetics , Signal Transduction
4.
Proteomics ; 21(11-12): e2000244, 2021 06.
Article in English | MEDLINE | ID: mdl-33945654

ABSTRACT

MARCH proteins are membrane-associated Ring-CH E3 ubiquitin ligases that dampen immune responses by downregulating cell surface expression of major histocompatibility complexes I and II as well as immune co-stimulatory receptors. We recently showed that MARCH2,3,4 and 9 also downregulate cell surface expression of the inflammatory cytokine receptor for interleukin-6 (IL6Rα). Here we use over-expression of these MARCH proteins in the M1 myeloid leukaemia cell line and cell surface proteomic analyses to globally analyse other potential targets of these proteins. A large range of cell surface proteins regulated by more than one MARCH protein in addition to several MARCH protein-specific cell surface targets were identified most of which were downregulated by MARCH expression. Prominent among these were several integrin complexes associated with immune cell homing, adhesion and migration. Integrin α4ß1 (VLA4 or VCAM-1 receptor) was downregulated only by MARCH2 and we showed that in MARCH2 knockout mice, Integrin α4 was upregulated specifically in mature B-lymphocytes and this was accompanied by decreased numbers of B-cells in the spleen.


Subject(s)
Integrins , Membrane Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Mice , Mice, Knockout , Proteomics
5.
Immunity ; 36(2): 239-50, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22342841

ABSTRACT

Janus kinases (JAKs) are key effectors in controlling immune responses and maintaining hematopoiesis. SOCS3 (suppressor of cytokine signaling-3) is a major regulator of JAK signaling and here we investigate the molecular basis of its mechanism of action. We found that SOCS3 bound and directly inhibited the catalytic domains of JAK1, JAK2, and TYK2 but not JAK3 via an evolutionarily conserved motif unique to JAKs. Mutation of this motif led to the formation of an active kinase that could not be inhibited by SOCS3. Surprisingly, we found that SOCS3 simultaneously bound JAK and the cytokine receptor to which it is attached, revealing how specificity is generated in SOCS action and explaining why SOCS3 inhibits only a subset of cytokines. Importantly, SOCS3 inhibited JAKs via a noncompetitive mechanism, making it a template for the development of specific and effective inhibitors to treat JAK-based immune and proliferative diseases.


Subject(s)
Cytokines/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites/genetics , Conserved Sequence , Humans , Interleukin-6/metabolism , Janus Kinases/antagonists & inhibitors , Janus Kinases/chemistry , Janus Kinases/genetics , Janus Kinases/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/chemistry , Suppressor of Cytokine Signaling Proteins/genetics
6.
Immunity ; 36(4): 646-57, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22483802

ABSTRACT

The immune system must distinguish viable cells from cells damaged by physical and infective processes. The damaged cell-recognition molecule Clec9A is expressed on the surface of the mouse and human dendritic cell subsets specialized for the uptake and processing of material from dead cells. Clec9A recognizes a conserved component within nucleated and nonnucleated cells, exposed when cell membranes are damaged. We have identified this Clec9A ligand as a filamentous form of actin in association with particular actin-binding domains of cytoskeletal proteins. We have determined the crystal structure of the human CLEC9A C-type lectin domain and propose a functional dimeric structure with conserved tryptophans in the ligand recognition site. Mutation of these residues ablated CLEC9A binding to damaged cells and to the isolated ligand complexes. We propose that Clec9A provides targeted recruitment of the adaptive immune system during infection and can also be utilized to enhance immune responses generated by vaccines.


Subject(s)
Actin Cytoskeleton/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Receptors, Immunologic/metabolism , Receptors, Mitogen/metabolism , Actins/metabolism , Adaptive Immunity , Animals , Binding Sites , Cell Line , Cell Membrane/metabolism , Dendritic Cells/cytology , Female , Humans , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Protein Structure, Secondary , Receptors, Immunologic/genetics , Receptors, Mitogen/chemistry , Receptors, Mitogen/genetics , Spectrin/metabolism
7.
Biochem J ; 476(19): 2869-2882, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31488575

ABSTRACT

Interleukin 6 (IL6) is a cytokine that regulates a number of important immune and inflammatory pathways. We used the ability of IL6 to inhibit the clonal proliferation of the mouse M1 myeloid leukemia cell line in agar to positively screen a cDNA expression library for proteins that inhibited IL6 activity. We found three clones completely resistant to IL6 that contained the cDNA for the Membrane-Associated RING-CH E3 ubiquitin ligase MARCH2. MARCH2 is a member of a family of membrane-bound E3 ubiquitin ligases that target cell surface receptors for degradation. MARCH2 overexpressing M1 clones retained responsiveness to the related cytokines leukemia inhibitory factor and oncostatin M and we showed that its inhibitory effect was a result of selective down-regulation of the IL6 receptor alpha chain and not the shared receptor subunit, gp130 or other signalling molecules. This activity of MARCH2 was also shared with related proteins MARCH4, MARCH9 and an isoform of MARCH3. The transmembrane domains and C-terminal domains, as well as a functional RING domain, of MARCH proteins were all required for substrate recognition and down-regulation. Genetic deletion of individual MARCH proteins in mice had no or little effect on IL6Rα levels but combined deletions of MARCH2,3 and 4 displayed elevated steady-state levels of IL6Rα in selected haemopoietic cell subsets including CD8+ and CD4+ T cells. These studies extend the potential immunosuppressive roles of MARCH proteins to include down-regulation of IL6 inflammatory responses.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/physiology , Receptors, Interleukin-6/metabolism , Ubiquitin-Protein Ligases/physiology , Animals , Cell Line, Tumor , Down-Regulation , Mice , Mice, Inbred C57BL , Protein Binding , Protein Domains , Protein Transport
8.
Immunol Cell Biol ; 97(9): 840-852, 2019 10.
Article in English | MEDLINE | ID: mdl-31335993

ABSTRACT

The innate immune system is our first line of defense against viral pathogens. Host cell pattern recognition receptors sense viral components and initiate immune signaling cascades that result in the production of an array of cytokines to combat infection. Retinoic acid-inducible gene-I (RIG-I) is a pattern recognition receptor that recognizes viral RNA and, when activated, results in the production of type I and III interferons (IFNs) and the upregulation of IFN-stimulated genes. Ubiquitination of RIG-I by the E3 ligases tripartite motif-containing 25 (TRIM25) and Riplet is thought to be requisite for RIG-I activation; however, recent studies have questioned the relative importance of these two enzymes for RIG-I signaling. In this study, we show that deletion of Trim25 does not affect the IFN response to either influenza A virus (IAV), influenza B virus, Sendai virus or several RIG-I agonists. This is in contrast to deletion of either Rig-i or Riplet, which completely abrogated RIG-I-dependent IFN responses. This was consistent in both mouse and human cell lines, as well as in normal human bronchial cells. With most of the current TRIM25 literature based on exogenous expression, these findings provide critical evidence that Riplet, and not TRIM25, is required endogenously for the ubiquitination of RIG-I. Despite this, loss of TRIM25 results in greater susceptibility to IAV infection in vivo, suggesting that it may have an alternative role in host antiviral defense. This study refines our understanding of RIG-I signaling in viral infections and will inform future studies in the field.


Subject(s)
Antiviral Agents/metabolism , DEAD Box Protein 58/metabolism , DNA-Binding Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , A549 Cells , Animals , Cell Line , Epithelial Cells/microbiology , Epithelial Cells/virology , Gene Deletion , Humans , Ligands , Mice, Inbred C57BL , RNA/metabolism , Receptors, Immunologic
10.
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
11.
Stem Cells ; 35(8): 1948-1957, 2017 08.
Article in English | MEDLINE | ID: mdl-28577303

ABSTRACT

The hematopoietically expressed homeobox transcription factor (Hhex) is important for the maturation of definitive hematopoietic progenitors and B-cells during development. We have recently shown that in adult hematopoiesis, Hhex is dispensable for maintenance of hematopoietic stem cells (HSCs) and myeloid lineages but essential for the commitment of common lymphoid progenitors (CLPs) to lymphoid lineages. Here, we show that during serial bone marrow transplantation, Hhex-deleted HSCs are progressively lost, revealing an intrinsic defect in HSC self-renewal. Moreover, Hhex-deleted mice show markedly impaired hematopoietic recovery following myeloablation, due to a failure of progenitor expansion. In vitro, Hhex-null blast colonies were incapable of replating, implying a specific requirement for Hhex in immature progenitors. Transcriptome analysis of Hhex-null Lin- Sca+ Kit+ cells showed that Hhex deletion leads to derepression of polycomb repressive complex 2 (PRC2) and PRC1 target genes, including the Cdkn2a locus encoding the tumor suppressors p16Ink 4a and p19Arf . Indeed, loss of Cdkn2a restored the capacity of Hhex-null blast colonies to generate myeloid progenitors in vitro, as well as hematopoietic reconstitution following myeloablation in vivo. Thus, HSCs require Hhex to promote PRC2-mediated Cdkn2a repression to enable continued self-renewal and response to hematopoietic stress. Stem Cells 2017;35:1948-1957.


Subject(s)
Cell Self Renewal , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Stress, Physiological , Transcription Factors/metabolism , Animals , Cell Proliferation , Gene Deletion , Gene Expression Regulation , Hematopoietic Stem Cell Transplantation , Mice, Inbred C57BL , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism
12.
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
13.
Semin Immunol ; 26(1): 13-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24418198

ABSTRACT

IL-6 a multi-functional cytokine with important effects in both inflammation and haematopoiesis. SOCS3 is the primary inhibitor of IL-6 signalling, interacting with gp130, the common shared chain of the IL-6 family of cytokines, and JAK1, JAK2 and TYK2 to control both the duration of signalling and the biological response. Recent biochemical and structural studies have shown SOCS3 binds to only these three JAKs, all of which are associated with IL-6 signalling, and not JAK3. This specificity is determined by a three residue "GQM" motif in the kinase domain of JAK1, JAK2 and TYK2. SOCS3 binds to JAK and gp130 simultaneously, and inhibits JAK activity in an ATP-independent manner by partially occluding the kinase's substrate binding groove with its kinase inhibitory region. We therefore propose a model in which each of gp130, JAK and SOCS3 are directly bound to the other two, allowing SOCS3 to inhibit IL6 signalling with high potency and specificity.


Subject(s)
Interleukin-6/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Catalysis , Cytokine Receptor gp130/metabolism , Humans , Interleukin-6/metabolism , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Models, Biological , Multigene Family , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Interleukin-6/metabolism , Signal Transduction , Suppressor of Cytokine Signaling Proteins/chemistry , Suppressor of Cytokine Signaling Proteins/genetics
14.
J Biol Chem ; 291(41): 21703-21716, 2016 Oct 07.
Article in English | MEDLINE | ID: mdl-27539849

ABSTRACT

Oncostatin M (OSM) and leukemia inhibitory factor (LIF) are IL-6 family members with a wide range of biological functions. Human OSM (hOSM) and murine LIF (mLIF) act in mouse cells via a LIF receptor (LIFR)-glycoprotein 130 (gp130) heterodimer. In contrast, murine OSM (mOSM) signals mainly via an OSM receptor (OSMR)-gp130 heterodimer and binds with only very low affinity to mLIFR. hOSM and mLIF stimulate bone remodeling by both reducing osteocytic sclerostin and up-regulating the pro-osteoclastic factor receptor activator of NF-κB ligand (RANKL) in osteoblasts. In the absence of OSMR, mOSM still strongly suppressed sclerostin and stimulated bone formation but did not induce RANKL, suggesting that intracellular signaling activated by the low affinity interaction of mOSM with mLIFR is different from the downstream effects when mLIF or hOSM interacts with the same receptor. Both STAT1 and STAT3 were activated by mOSM in wild type cells or by mLIF/hOSM in wild type and Osmr-/- cells. In contrast, in Osmr-/- primary osteocyte-like cells stimulated with mOSM (therefore acting through mLIFR), microarray expression profiling and Western blotting analysis identified preferential phosphorylation of STAT3 and induction of its target genes but not of STAT1 and its target genes; this correlated with reduced phosphorylation of both gp130 and LIFR. In a mouse model of spontaneous osteopenia caused by hyperactivation of STAT1/3 signaling downstream of gp130 (gp130Y757F/Y757F), STAT1 deletion rescued the osteopenic phenotype, indicating a beneficial effect of promoting STAT3 signaling over STAT1 downstream of gp130 in this low bone mass condition, and this may have therapeutic value.


Subject(s)
Bone Diseases, Metabolic/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Oncostatin M/metabolism , Osteocytes/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Animals , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Disease Models, Animal , Humans , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Mice , Oncostatin M/genetics , Oncostatin M Receptor beta Subunit/genetics , Oncostatin M Receptor beta Subunit/metabolism , Organ Size , Osteocytes/pathology , Phosphorylation/genetics , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics
15.
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
16.
Stem Cells ; 33(12): 3397-421, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26976235

ABSTRACT

A collection of tributes and remembrances from esteemed colleagues, mentees, and friends on the life and work of "the father of hematopoietic cytokines".


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells , Animals , History, 20th Century , History, 21st Century , Humans , Portraits as Topic
17.
Immunol Cell Biol ; 93(10): 909-13, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26077508

ABSTRACT

Suppressor of cytokine signaling (SOCS) proteins are key regulators of innate and adaptive immunity. Mice lacking functional SOCS4 are hypersusceptible to primary infection with influenza A virus (IAV), displaying dysregulated pro-inflammatory cytokine and chemokine production in the lungs, delayed viral clearance and impaired trafficking of influenza-specific CD8(+) T cells to the site of infection. Therefore, we postulated that SOCS4 is a critical regulator of anti-viral immunity. Unexpectedly, SOCS4 was not required for CD8(+) T-cell memory generation, nor was it required to efficiently recall those cells in response to secondary IAV infection. Wild-type or SOCS4-deficient mice primed and re-challenged with serologically different influenza strains, did not show differences in susceptibility to IAV and cleared the virus from the lungs at the same rate. We have not observed differences in trafficking or numbers of IAV-specific cells, numbers of resident memory T cells or in cytokine profiles in lungs of infected animals. Our data show that despite an impaired primary immune response in Socs4(R108X/R108X) mice, SOCS4 is dispensable for an efficient recall response to influenza virus infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lung/physiology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , CD8-Positive T-Lymphocytes/virology , Cell Movement/genetics , Cytokines/metabolism , Immunity/genetics , Immunologic Memory/genetics , Immunophenotyping , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Knockout , Suppressor of Cytokine Signaling Proteins/genetics
18.
Biochem J ; 462(1): 1-13, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25057888

ABSTRACT

The JAK (Janus kinase) family members serve essential roles as the intracellular signalling effectors of cytokine receptors. This family, comprising JAK1, JAK2, JAK3 and TYK2 (tyrosine kinase 2), was first described more than 20 years ago, but the complexities underlying their activation, regulation and pleiotropic signalling functions are still being explored. Here, we review the current knowledge of their physiological functions and the causative role of activating and inactivating JAK mutations in human diseases, including haemopoietic malignancies, immunodeficiency and inflammatory diseases. At the molecular level, recent studies have greatly advanced our knowledge of the structures and organization of the component FERM (4.1/ezrin/radixin/moesin)-SH2 (Src homology 2), pseudokinase and kinase domains within the JAKs, the mechanism of JAK activation and, in particular, the role of the pseudokinase domain as a suppressor of the adjacent tyrosine kinase domain's catalytic activity. We also review recent advances in our understanding of the mechanisms of negative regulation exerted by the SH2 domain-containing proteins, SOCS (suppressors of cytokine signalling) proteins and LNK. These recent studies highlight the diversity of regulatory mechanisms utilized by the JAK family to maintain signalling fidelity, and suggest alternative therapeutic strategies to complement existing ATP-competitive kinase inhibitors.


Subject(s)
Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Janus Kinase 3/metabolism , TYK2 Kinase/metabolism , Adaptor Proteins, Signal Transducing , Animals , Enzyme Activation , Humans , Immunologic Deficiency Syndromes/genetics , Intracellular Signaling Peptides and Proteins , Leukocyte Common Antigens/metabolism , Myeloproliferative Disorders/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Structure, Tertiary/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proteins/metabolism , Receptors, Cytokine/physiology , Signal Transduction , src Homology Domains
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.
Proc Natl Acad Sci U S A ; 109(40): 16270-5, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22988114

ABSTRACT

Synthetic CpG oligonucleotides (ODN) have potent immunostimulatory properties exploited in clinical vaccine trials. How CpG ODN are captured and delivered to the intracellular receptor TLR9, however, has been elusive. Here we show that DEC-205, a multilectin receptor expressed by a variety of cells, is a receptor for CpG ODN. When CpG ODN are used as an adjuvant, mice deficient in DEC-205 have impaired dendritic cell (DC) and B-cell maturation, are unable to make some cytokines such as IL-12, and display suboptimal cytotoxic T-cell responses. We reveal that DEC-205 directly binds class B CpG ODN and enhances their uptake. The CpG-ODN binding function of DEC-205 is conserved between mouse and man, although human DEC-205 preferentially binds a specific class B CpG ODN that has been selected for human clinical trials. Our findings identify an important receptor for class B CpG ODN and reveal a unique function for DEC-205.


Subject(s)
Antigens, CD/metabolism , B-Lymphocytes/metabolism , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Oligodeoxyribonucleotides/metabolism , Receptors, Cell Surface/metabolism , Animals , Antigens, CD/genetics , CHO Cells , Chromatography, Affinity , Chromatography, Gel , Cloning, Molecular , Cricetinae , Cricetulus , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Lectins, C-Type/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Minor Histocompatibility Antigens , Oligodeoxyribonucleotides/genetics , Receptors, Cell Surface/genetics , Species Specificity , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL