Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Leukemia ; 38(6): 1342-1352, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38491305

ABSTRACT

Thrombopoietin (Tpo), which binds to its specific receptor, the Mpl protein, is the major cytokine regulator of megakaryopoiesis and circulating platelet number. Tpo binding to Mpl triggers activation of Janus kinase 2 (Jak2) and phosphorylation of the receptor, as well as activation of several intracellular signalling cascades that mediate cellular responses. Three tyrosine (Y) residues in the C-terminal region of the Mpl intracellular domain have been implicated as sites of phosphorylation required for regulation of major Tpo-stimulated signalling pathways: Mpl-Y565, Mpl-Y599 and Mpl-Y604. Here, we have introduced mutations in the mouse germline and report a consistent physiological requirement for Mpl-Y599, mutation of which resulted in thrombocytopenia, deficient megakaryopoiesis, low hematopoietic stem cell (HSC) number and function, and attenuated responses to myelosuppression. We further show that in models of myeloproliferative neoplasms (MPN), where Mpl is required for pathogenesis, thrombocytosis was dependent on intact Mpl-Y599. In contrast, Mpl-Y565 was required for negative regulation of Tpo responses; mutation of this residue resulted in excess megakaryopoiesis at steady-state and in response to myelosuppression, and exacerbated thrombocytosis associated with MPN.


Subject(s)
Hematopoiesis , Myeloproliferative Disorders , Receptors, Thrombopoietin , Thrombopoietin , Tyrosine , Animals , Receptors, Thrombopoietin/metabolism , Receptors, Thrombopoietin/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Mice , Thrombopoietin/metabolism , Tyrosine/metabolism , Tyrosine/genetics , Phosphorylation , Mice, Inbred C57BL , Hematopoietic Stem Cells/metabolism , Signal Transduction , Mutation , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Thrombopoiesis/genetics
2.
Biochem J ; 478(13): 2555-2569, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34109974

ABSTRACT

Structural maintenance of chromosomes flexible hinge domain-containing 1 (SMCHD1) is an epigenetic regulator that mediates gene expression silencing at targeted sites across the genome. Our current understanding of SMCHD1's molecular mechanism, and how substitutions within SMCHD1 lead to the diseases, facioscapulohumeral muscular dystrophy (FSHD) and Bosma arhinia microphthalmia syndrome (BAMS), are only emerging. Recent structural studies of its two component domains - the N-terminal ATPase and C-terminal SMC hinge - suggest that dimerization of each domain plays a central role in SMCHD1 function. Here, using biophysical techniques, we demonstrate that the SMCHD1 ATPase undergoes dimerization in a process that is dependent on both the N-terminal UBL (Ubiquitin-like) domain and ATP binding. We show that neither the dimerization event, nor the presence of a C-terminal extension past the transducer domain, affect SMCHD1's in vitro catalytic activity as the rate of ATP turnover remains comparable to the monomeric protein. We further examined the functional importance of the N-terminal UBL domain in cells, revealing that its targeted deletion disrupts the localization of full-length SMCHD1 to chromatin. These findings implicate UBL-mediated SMCHD1 dimerization as a crucial step for chromatin interaction, and thereby for promoting SMCHD1-mediated gene silencing.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Protein Multimerization , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Binding Sites/genetics , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , HEK293 Cells , Humans , Immunoblotting , Microscopy, Fluorescence , Mutation , Protein Binding , Protein Domains , Scattering, Small Angle , Substrate Specificity , Ubiquitin/chemistry , Ubiquitin/metabolism , X-Ray Diffraction
3.
Sci Signal ; 13(636)2020 06 16.
Article in English | MEDLINE | ID: mdl-32546545

ABSTRACT

Structural maintenance of chromosomes flexible hinge domain containing 1 (SMCHD1) is an epigenetic regulator in which polymorphisms cause the human developmental disorder, Bosma arhinia micropthalmia syndrome, and the degenerative disease, facioscapulohumeral muscular dystrophy. SMCHD1 is considered a noncanonical SMC family member because its hinge domain is C-terminal, because it homodimerizes rather than heterodimerizes, and because SMCHD1 contains a GHKL-type, rather than an ABC-type ATPase domain at its N terminus. The hinge domain has been previously implicated in chromatin association; however, the underlying mechanism involved and the basis for SMCHD1 homodimerization are unclear. Here, we used x-ray crystallography to solve the three-dimensional structure of the Smchd1 hinge domain. Together with structure-guided mutagenesis, we defined structural features of the hinge domain that participated in homodimerization and nucleic acid binding, and we identified a functional hotspot required for chromatin localization in cells. This structure provides a template for interpreting the mechanism by which patient polymorphisms within the SMCHD1 hinge domain could compromise function and lead to facioscapulohumeral muscular dystrophy.


Subject(s)
Chromosomal Proteins, Non-Histone/chemistry , Protein Multimerization , Animals , Chromosomal Proteins, Non-Histone/genetics , Crystallography, X-Ray , Mice , Nucleic Acids/chemistry , Nucleic Acids/metabolism , Protein Domains , Protein Structure, Quaternary , Siblings
4.
Nat Commun ; 11(1): 3150, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32561755

ABSTRACT

MLKL is the essential effector of necroptosis, a form of programmed lytic cell death. We have isolated a mouse strain with a single missense mutation, MlklD139V, that alters the two-helix 'brace' that connects the killer four-helix bundle and regulatory pseudokinase domains. This confers constitutive, RIPK3 independent killing activity to MLKL. Homozygous mutant mice develop lethal postnatal inflammation of the salivary glands and mediastinum. The normal embryonic development of MlklD139V homozygotes until birth, and the absence of any overt phenotype in heterozygotes provides important in vivo precedent for the capacity of cells to clear activated MLKL. These observations offer an important insight into the potential disease-modulating roles of three common human MLKL polymorphisms that encode amino acid substitutions within or adjacent to the brace region. Compound heterozygosity of these variants is found at up to 12-fold the expected frequency in patients that suffer from a pediatric autoinflammatory disease, chronic recurrent multifocal osteomyelitis (CRMO).


Subject(s)
Hematopoietic Stem Cells/metabolism , Hematopoietic System/pathology , Necroptosis/genetics , Protein Kinases/genetics , Animals , Animals, Newborn , Hereditary Autoinflammatory Diseases , Humans , Inflammation/genetics , Mice , Mutation, Missense , Osteomyelitis/genetics , Protein Kinases/metabolism
5.
Cell Rep ; 25(7): 1912-1923.e9, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30428357

ABSTRACT

We and others have recently reported that the SMC protein Smchd1 is a regulator of chromosome conformation. Smchd1 is critical for the structure of the inactive X chromosome and at autosomal targets such as the Hox genes. However, it is unknown how Smchd1 is recruited to these sites. Here, we report that Smchd1 localizes to the inactive X via the Xist-HnrnpK-PRC1 (polycomb repressive complex 1) pathway. Contrary to previous reports, Smchd1 does not bind Xist or other RNA molecules with any specificity. Rather, the localization of Smchd1 to the inactive X is H2AK119ub dependent. Following perturbation of this interaction, Smchd1 is destabilized, which has consequences for gene silencing genome-wide. Our work adds Smchd1 to the PRC1 silencing pathway for X chromosome inactivation.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Polycomb Repressive Complex 1/metabolism , RNA, Long Noncoding/metabolism , X Chromosome Inactivation/genetics , Animals , Base Sequence , Cell Differentiation , Female , Genome , Histones/metabolism , Lysine/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Oligonucleotides/metabolism , Protein Transport
6.
J Exp Med ; 214(3): 773-791, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28190000

ABSTRACT

Genetic alterations disrupting the transcription factor IKZF1 (encoding IKAROS) are associated with poor outcome in B lineage acute lymphoblastic leukemia (B-ALL) and occur in >70% of the high-risk BCR-ABL1+ (Ph+) and Ph-like disease subtypes. To examine IKAROS function in this context, we have developed novel mouse models allowing reversible RNAi-based control of Ikaros expression in established B-ALL in vivo. Notably, leukemias driven by combined BCR-ABL1 expression and Ikaros suppression rapidly regress when endogenous Ikaros is restored, causing sustained disease remission or ablation. Comparison of transcriptional profiles accompanying dynamic Ikaros perturbation in murine B-ALL in vivo with two independent human B-ALL cohorts identified nine evolutionarily conserved IKAROS-repressed genes. Notably, high expression of six of these genes is associated with inferior event-free survival in both patient cohorts. Among them are EMP1, which was recently implicated in B-ALL proliferation and prednisolone resistance, and the novel target CTNND1, encoding P120-catenin. We demonstrate that elevated Ctnnd1 expression contributes to maintenance of murine B-ALL cells with compromised Ikaros function. These results suggest that IKZF1 alterations in B-ALL leads to induction of multiple genes associated with proliferation and treatment resistance, identifying potential new therapeutic targets for high-risk disease.


Subject(s)
Ikaros Transcription Factor/physiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Catenins/genetics , Cell Line, Tumor , Fusion Proteins, bcr-abl/analysis , Humans , Membrane Proteins/genetics , Mice , Neoplasm Proteins/genetics , RNA-Binding Proteins/genetics , Receptors, Cell Surface/genetics , Delta Catenin
8.
Growth Factors ; 32(1): 18-26, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24438083

ABSTRACT

Activation of the cell surface receptor, c-Mpl, by the cytokine, thrombopoietin (TPO), underpins megakaryocyte and platelet production in mammals. In humans, mutations in c-Mpl have been identified as the molecular basis of Congenital Amegakaryocytic Thrombocytopenia (CAMT). Here, we show that CAMT-associated mutations in c-Mpl principally lead to defective receptor presentation on the cell surface. In contrast, one CAMT mutant c-Mpl, F104S, was expressed on the cell surface, but showed defective TPO binding and receptor activation. Using mutational analyses, we examined which residues adjacent to F104 within the membrane-distal cytokine receptor homology module (CRM) of c-Mpl comprise the TPO-binding epitope, revealing residues within the predicted Domain 1 E-F and A-B loops and Domain 2 F'-G' loop as key TPO-binding determinants. These studies underscore the importance of the c-Mpl membrane-distal CRM to TPO-binding and suggest that mutations within this CRM that perturb TPO binding could give rise to CAMT.


Subject(s)
Mutation/genetics , Receptors, Thrombopoietin/genetics , Thrombocytopenia/genetics , Thrombopoietin/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Blood Platelets/metabolism , COS Cells , Cell Line , Cell Proliferation , Chlorocebus aethiops , Congenital Bone Marrow Failure Syndromes , Megakaryocytes/metabolism , Mice , Molecular Sequence Data , Oncogene Proteins/genetics , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Receptors, Prolactin/genetics , Sequence Alignment , Signal Transduction/genetics
9.
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
10.
Nucleic Acids Res ; 39(17): 7415-27, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21652639

ABSTRACT

Transcriptional control is dependent on a vast network of epigenetic modifications. One epigenetic mark of particular interest is tri-methylation of lysine 27 on histone H3 (H3K27me3), which is catalysed and maintained by Polycomb Repressive Complex 2 (PRC2). Although this histone mark is studied widely, the precise relationship between its local pattern of enrichment and regulation of gene expression is currently unclear. We have used ChIP-seq to generate genome-wide maps of H3K27me3 enrichment, and have identified three enrichment profiles with distinct regulatory consequences. First, a broad domain of H3K27me3 enrichment across the body of genes corresponds to the canonical view of H3K27me3 as inhibitory to transcription. Second, a peak of enrichment around the transcription start site (TSS) is commonly associated with 'bivalent' genes, where H3K4me3 also marks the TSS. Finally and most surprisingly, we identified an enrichment profile with a peak in the promoter of genes that is associated with active transcription. Genes with each of these three profiles were found in different proportions in each of the cell types studied. The data analysis techniques developed here will be useful for the identification of common enrichment profiles for other histone modifications that have important consequences for transcriptional regulation.


Subject(s)
Histones/metabolism , Transcription, Genetic , Animals , Cells, Cultured , Chromatin Immunoprecipitation , Cluster Analysis , Histones/chemistry , Lysine/metabolism , Methylation , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Sequence Analysis, DNA , Transcription Initiation Site
11.
Genes Dev ; 25(3): 251-62, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21245161

ABSTRACT

Although many genes are known to be critical for early hematopoiesis in the embryo, it remains unclear whether distinct regulatory pathways exist to control hematopoietic specification versus hematopoietic stem cell (HSC) emergence and function. Due to their interaction with key regulators of hematopoietic commitment, particular interest has focused on the role of the ETS family of transcription factors; of these, ERG is predicted to play an important role in the initiation of hematopoiesis, yet we do not know if or when ERG is required. Using in vitro and in vivo models of hematopoiesis and HSC development, we provide strong evidence that ERG is at the center of a distinct regulatory program that is not required for hematopoietic specification or differentiation but is critical for HSC maintenance during embryonic development. We show that, from the fetal period, ERG acts as a direct upstream regulator of Gata2 and Runx1 gene activity. Without ERG, physiological HSC maintenance fails, leading to the rapid exhaustion of definitive hematopoiesis.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Oncogene Proteins/metabolism , Animals , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/metabolism , GATA2 Transcription Factor/metabolism , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Oncogene Proteins/genetics , Transcription Factors , Transcriptional Regulator ERG
12.
Proc Natl Acad Sci U S A ; 107(50): 21689-94, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21115812

ABSTRACT

Thrombopoietin (TPO), acting through its receptor Mpl, has two major physiological roles: ensuring production of sufficient platelets via stimulation of megakaryocyte production and maintaining hematopoietic stem cell (HSC) quiescence. Mpl also controls circulating TPO concentration via receptor-mediated internalization and degradation. Here, we demonstrate that the megakaryocytosis and increased platelet mass in mice with mutations in the Myb or p300 genes causes reduced circulating TPO concentration and TPO starvation of the stem-cell compartment, which is exacerbated because these cells additionally exhibit impaired responsiveness to TPO. HSCs from Myb(Plt4/Plt4) mice show altered expression of TPO-responsive genes and, like HSCs from Tpo and Mpl mutant mice, exhibit increased cycling and a decline in the number of HSCs with age. These studies suggest that disorders of platelet number can have profound effects on the HSC compartment via effects on the feedback regulation of circulating TPO concentration.


Subject(s)
Hematopoietic Stem Cells/physiology , Animals , Blood Platelets/metabolism , Cell Differentiation/physiology , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Hematopoietic Stem Cells/cytology , Megakaryocytes/cytology , Megakaryocytes/physiology , Mice , Mice, Knockout , Microarray Analysis , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , Receptors, Thrombopoietin/metabolism , Thrombopoietin/blood
13.
Proc Natl Acad Sci U S A ; 107(38): 16625-30, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20823251

ABSTRACT

With the notable exception of humans, uric acid is degraded to (S)-allantoin in a biochemical pathway catalyzed by urate oxidase, 5-hydroxyisourate (HIU) hydrolase, and 2-oxo-4-hydroxy-4-carboxy-5-ureidoimidazoline decarboxylase in most vertebrate species. A point mutation in the gene encoding mouse HIU hydrolase, Urah, that perturbed uric acid metabolism within the liver was discovered during a mutagenesis screen in mice. The predicted substitution of cysteine for tyrosine in a conserved helical region of the mutant-encoded HIU hydrolase resulted in undetectable protein expression. Mice homozygous for this mutation developed elevated platelet counts secondary to excess thrombopoietin production and hepatomegaly. The majority of homozygous mutant mice also developed hepatocellular carcinoma, and tumor development was accelerated by exposure to radiation. The development of hepatomegaly and liver tumors in mice lacking Urah suggests that uric acid metabolites may be toxic and that urate oxidase activity without HIU hydrolase function may affect liver growth and transformation. The absence of HIU hydrolase in humans predicts slowed metabolism of HIU after clinical administration of exogenous urate oxidase in conditions of uric acid-related pathology. The data suggest that prolonged urate oxidase therapy should be combined with careful assessment of toxicity associated with extrahepatic production of uric acid metabolites.


Subject(s)
Amidohydrolases/deficiency , Amidohydrolases/genetics , Hepatomegaly/enzymology , Hepatomegaly/genetics , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/genetics , Point Mutation , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Amino Acid Sequence , Animals , Female , Genes, Tumor Suppressor , Hepatocytes/enzymology , Hepatomegaly/etiology , Liver Neoplasms, Experimental/etiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Models, Molecular , Molecular Sequence Data , Mutagenesis , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Thrombocytosis/enzymology , Thrombocytosis/genetics , Thrombopoietin/biosynthesis , Urate Oxidase/metabolism , Uric Acid/metabolism , Uric Acid/toxicity
14.
J Cell Biol ; 190(1): 129-41, 2010 Jul 12.
Article in English | MEDLINE | ID: mdl-20603330

ABSTRACT

Inducible nitric oxide (NO) synthase (iNOS; NOS2) produces NO and related reactive nitrogen species, which are critical effectors of the innate host response and are required for the intracellular killing of pathogens such as Mycobacterium tuberculosis and Leishmania major. We have identified SPRY domain-containing SOCS (suppressor of cytokine signaling) box protein 2 (SPSB2) as a novel negative regulator that recruits an E3 ubiquitin ligase complex to polyubiquitinate iNOS, resulting in its proteasomal degradation. SPSB2 interacts with the N-terminal region of iNOS via a binding interface on SPSB2 that has been mapped by nuclear magnetic resonance spectroscopy and mutational analyses. SPSB2-deficient macrophages showed prolonged iNOS expression, resulting in a corresponding increase in NO production and enhanced killing of L. major parasites. These results lay the foundation for the development of small molecule inhibitors that could disrupt the SPSB-iNOS interaction and thus prolong the intracellular lifetime of iNOS, which may be beneficial in chronic and persistent infections.


Subject(s)
DNA-Binding Proteins/metabolism , Leishmania major , Leishmaniasis, Cutaneous/metabolism , Macrophages/metabolism , Nitric Oxide Synthase Type II/metabolism , Proteasome Endopeptidase Complex/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , Gene Expression Regulation, Enzymologic/genetics , Leishmaniasis, Cutaneous/drug therapy , Leishmaniasis, Cutaneous/genetics , Macrophages/parasitology , Mice , Mice, Knockout , Mycobacterium tuberculosis , Nitric Oxide Synthase Type II/genetics , Proteasome Endopeptidase Complex/genetics , Protein Structure, Tertiary , Suppressor of Cytokine Signaling Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/genetics
15.
J Immunol ; 183(7): 4537-44, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734231

ABSTRACT

Suppressor of cytokine signaling (SOCS)-1 is a critical inhibitor of IFN-gamma signal transduction in vivo, but the precise biochemical mechanism of action of SOCS-1 is unclear. Studies in vitro have shown that SOCS-1 binds to Jaks and inhibits their catalytic activity, but recent studies indicate SOCS-1 may act in a similar manner to SOCS-3 by firstly interacting with cytokine receptors and then inhibiting Jak activity. Here, we have generated mice, termed Ifngr1(441F), in which a putative SOCS-1 binding site, tyrosine 441 (Y441), on the IFN-gamma receptor subunit 1 (IFNGR1) is mutated. We confirm that SOCS-1 binds to IFNGR1 in wild-type but not mutant cells. Mutation of Y441 results in impaired negative regulation of IFN-gamma signaling. IFN-gamma-induced STAT1 activation is prolonged in Ifngr1(441F) cells, but not to the extent seen in cells completely lacking SOCS-1, suggesting that SOCS-1 maintains activity to modulate IFN-gamma signaling via other mechanisms. Despite this, we show that hypersensitivity to IFN-gamma results in enhanced innate tumor protection in Ifngr1(441F) mice in vivo, and unregulated expression of an IFN-gamma-dependent chemokine, monokine-induced by IFN-gamma. Collectively, these data indicate that Y441 contributes to the regulation of signaling through IFNGR1 via the recruitment of SOCS-1 to the receptor.


Subject(s)
Interferon-gamma/physiology , Protein Subunits/metabolism , Receptors, Interferon/metabolism , Signal Transduction/immunology , Suppressor of Cytokine Signaling Proteins/metabolism , Tyrosine/metabolism , Animals , Cells, Cultured , Gene Knock-In Techniques , Interferon-gamma/deficiency , Interferon-gamma/genetics , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Binding/genetics , Protein Binding/immunology , Protein Subunits/deficiency , Protein Subunits/genetics , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Signal Transduction/genetics , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/genetics , Tyrosine/genetics , Interferon gamma Receptor
16.
J Mol Biol ; 386(3): 662-74, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19154741

ABSTRACT

The four mammalian SPRY (a sequence repeat in dual-specificity kinase splA and ryanodine receptors) domain-containing suppressor of cytokine signalling (SOCS) box proteins (SSB-1 to -4) are characterised by a C-terminal SOCS box and a central SPRY domain. The latter is a protein interaction module found in over 1600 proteins, with more than 70 encoded in the human genome. Here we report the crystal structure of the SPRY domain of murine SSB-2 and compare it with the SSB-2 solution structure and crystal structures of other B30.2/SPRY domain-containing family proteins. The structure is a bent beta-sandwich, consisting of two seven-stranded beta-sheets wrapped around a long loop that extends from the centre strands of the inner or concave beta-sheet; it closely matches those of GUSTAVUS and SSB-4. The structure is also similar to those of two recently determined Neuralized homology repeat (NHR) domains (also known as NEUZ domains), with detailed comparisons, suggesting that the NEUZ/NHR domains form a subclass of SPRY domains. The binding site on SSB-2 for the prostate apoptosis response-4 (Par-4) protein has been mapped in finer detail using mutational analyses. Moreover, SSB-1 was shown to have a Par-4 binding surface similar to that identified for SSB-2. Structural perturbations of SSB-2 induced by mutations affecting its interaction with Par-4 and/or c-Met have been characterised by NMR. These comparisons, in conjunction with previously published dynamics data from NMR relaxation studies and coarse-grained dynamics simulation using normal mode analysis, further refine our understanding of the structural basis for protein recognition of SPRY domain-containing proteins.


Subject(s)
DNA-Binding Proteins/chemistry , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , DNA Mutational Analysis , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Receptors, Proteinase-Activated/metabolism , Sequence Alignment
17.
Nat Immunol ; 9(7): 810-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18500345

ABSTRACT

Ets-related gene (ERG), which encodes a member of the Ets family of transcription factors, is a potent oncogene. Chromosomal rearrangements involving ERG are found in acute myeloid leukemia, acute lymphoblastic leukemia, Ewing's sarcoma and more than half of all prostate cancers; however, the normal physiological function of Erg is unknown. We did a sensitized genetic screen of the mouse for regulators of hematopoietic stem cell function and report here a germline mutation of Erg. We show that Erg is required for definitive hematopoiesis, adult hematopoietic stem cell function and the maintenance of normal peripheral blood platelet numbers.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Trans-Activators/genetics , Trans-Activators/metabolism , Animals , Flow Cytometry , Gene Expression Regulation , Humans , Mice , Mice, Mutant Strains , Mutation , Transcription, Genetic , Transcriptional Regulator ERG
18.
Blood ; 110(5): 1466-74, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17510322

ABSTRACT

Suppressor of cytokine signaling 3 (SOCS3) is a negative regulator of granulocyte-colony stimulating factor (G-CSF) signaling in vivo. SOCS proteins regulate cytokine signaling by binding, via their SH2 domains, to activated cytokine receptors or their associated Janus kinases. In addition, they bind to the elongin B/C ubiquitin ligase complex via the SOCS box. To ascertain the contribution of the SOCS box of SOCS3 to in vivo regulation of G-CSF signaling, we generated mice expressing a truncated SOCS3 protein lacking the C-terminal SOCS box (SOCS3(Delta SB/Delta SB)). SOCS3(Delta SB/Delta SB) mice were viable, had normal steady-state hematopoiesis, and did not develop inflammatory disease. Despite the mild phenotype, STAT3 activation in response to G-CSF signaling was prolonged in SOCS3(Delta SB/Delta SB) bone marrow. SOCS3(Delta SB/Delta SB) bone marrow contained increased numbers of colony-forming cells responsive to G-CSF and IL-6. Treatment of the mice with pharmacologic doses of G-CSF, which mimics emergency granulopoiesis and therapeutic use of G-CSF, revealed that SOCS3(Delta SB/Delta SB) mice were hyperresponsive to G-CSF. Compared with wild-type mice, SOCS3(Delta SB/Delta SB) mice developed a more florid arthritis when tested using an acute disease model. Overall, the results establish a role for the SOCS box of SOCS3 in the in vivo regulation of G-CSF signaling and the response to inflammatory stimuli.


Subject(s)
Arthritis/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Leukopoiesis , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism , Acute Disease , Animals , Arthritis/genetics , Arthritis/pathology , Bone Marrow/metabolism , Bone Marrow/pathology , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/genetics , Interleukin-6/metabolism , Leukopoiesis/genetics , Mice , Mice, Mutant Strains , Receptors, Cytokine/metabolism , Signal Transduction/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , src Homology Domains/genetics
19.
Blood ; 110(7): 2371-80, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17515402

ABSTRACT

A pivotal mediator of actin dynamics is the protein cofilin, which promotes filament severing and depolymerization, facilitating the breakdown of existing filaments, and the enhancement of filament growth from newly created barbed ends. It does so in concert with actin interacting protein 1 (Aip1), which serves to accelerate cofilin's activity. While progress has been made in understanding its biochemical functions, the physiologic processes the cofilin/Aip1 complex regulates, particularly in higher organisms, are yet to be determined. We have generated an allelic series for WD40 repeat protein 1 (Wdr1), the mammalian homolog of Aip1, and report that reductions in Wdr1 function produce a dramatic phenotype gradient. While severe loss of function at the Wdr1 locus causes embryonic lethality, macrothrombocytopenia and autoinflammatory disease develop in mice carrying hypomorphic alleles. Macrothrombocytopenia is the result of megakaryocyte maturation defects, which lead to a failure of normal platelet shedding. Autoinflammatory disease, which is bone marrow-derived yet nonlymphoid in origin, is characterized by a massive infiltration of neutrophils into inflammatory lesions. Cytoskeletal responses are impaired in Wdr1 mutant neutrophils. These studies establish an essential requirement for Wdr1 in megakaryocytes and neutrophils, indicating that cofilin-mediated actin dynamics are critically important to the development and function of both cell types.


Subject(s)
Actin Depolymerizing Factors/metabolism , Microfilament Proteins/metabolism , Mutation/genetics , Thrombocytopenia/metabolism , Thrombocytopenia/pathology , Alleles , Amino Acid Sequence , Animals , Blood Platelets/cytology , Cell Differentiation , Cell Movement , Conserved Sequence , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Megakaryocytes/cytology , Mice , Mice, Knockout , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Microscopy, Electron, Transmission , Molecular Sequence Data , Neutrophils/cytology , Neutrophils/metabolism , Sequence Alignment , Thrombocytopenia/genetics
20.
J Biol Chem ; 282(7): 4728-4737, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17148442

ABSTRACT

The suppressors of cytokine signaling (SOCS) proteins inhibit cytokine action by direct interaction with Janus kinases or activated cytokine receptors. In addition to the N-terminal and Src homology 2 domains that mediate these interactions, SOCS proteins contain a C-terminal SOCS box. DNA data base searches have identified a number of other protein families that possess a SOCS box, of which the ankyrin repeat and SOCS box-containing (Asb) proteins constitute the largest. Although it is known that the SOCS proteins are involved in the negative regulation of cytokine signaling, the biological and biochemical functions of the Asbs are largely undefined. Using a proteomics approach, we demonstrate that creatine kinase B (CKB) interacts with Asb-9 in a specific, SOCS box-independent manner. This interaction increases the polyubiquitylation of CKB and decreases total CKB levels within the cell. The targeting of CKB for degradation by Asb-9 was primarily SOCS box-dependent and suggests that Asb-9 acts as a specific ubiquitin ligase regulating levels of this evolutionarily conserved enzyme.


Subject(s)
Creatine Kinase/metabolism , Protein Processing, Post-Translational/physiology , Suppressor of Cytokine Signaling Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Ankyrins/metabolism , Cytokines/metabolism , Databases, Protein , Evolution, Molecular , Humans , Mice , Proteomics , Signal Transduction/physiology , src Homology Domains/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...