Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 205
Filter
Add more filters










Publication year range
1.
Placenta ; 34(11): 1014-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24074901

ABSTRACT

INTRODUCTION: Ectopic pregnancy is unique to humans and a leading cause of maternal morbidity and mortality. The etiology remains unknown however factors regulating embryo implantation likely contribute. Leukemia inhibitory factor (LIF) has roles in extravillous trophoblast adhesion and invasion and is present in ectopic implantation sites. We hypothesised that LIF facilitates blastocyst adhesion/invasion in the Fallopian tube, contributing to ectopic pregnancy. METHODS: We immunolocalised LIF receptor (R) in tubal ectopic pregnancy (N = 5). We used an oviduct cell line (OE-E6/E7) to model Fallopian tube epithelial cells and a trophoblast spheroid co-culture model (HTR-8/SVneo cell line formed spheroids) to model blastocyst attachment to the Fallopian tube. We examined LIF signaling pathways in OE-E6/E7 cells by Western blot. The effect of LIF and LIF inhibition (using a novel LIF inhibitor, PEGLA) on first-trimester placental outgrowth was determined. RESULTS: LIFR localised to villous and extravillous trophoblast and Fallopian tube epithelium in ectopic pregnancy. LIF activated STAT3 but not the ERK pathway in OE-E6/E7 cells. LIF stimulated HTR-8/SVneo spheroid adhesion to OE-E6/E7 cells which was significantly reduced after PEGLA treatment. LIF promoted placental explants outgrowth, while co-treatment with PEGLA blocked outgrowth. DISCUSSION: Our data suggests LIF facilitates the development of ectopic pregnancy by stimulating blastocyst adhesion and trophoblast outgrowth from placental explants. Ectopic pregnancy is usually diagnosed after 6 weeks of pregnancy, therefore PEGLA may be useful in targeting trophoblast growth/invasion. CONCLUSION: LIF may contribute to the development of ectopic pregnancies and that pharmacologically targeting LIF-mediated trophoblast outgrowth may be useful as a treatment for ectopic pregnancy.


Subject(s)
Blastocyst/metabolism , Fallopian Tubes/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Leukemia Inhibitory Factor/metabolism , Placenta/metabolism , Pregnancy, Tubal/metabolism , Signal Transduction , Adolescent , Adult , Blastocyst/drug effects , Blastocyst/pathology , Cell Adhesion/drug effects , Cell Line , Cell Proliferation/drug effects , Coculture Techniques , Embryo Implantation/drug effects , Epithelium/drug effects , Epithelium/metabolism , Epithelium/pathology , Fallopian Tubes/drug effects , Fallopian Tubes/pathology , Fallopian Tubes/surgery , Female , Humans , Leukemia Inhibitory Factor/antagonists & inhibitors , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/pharmacology , Leukemia Inhibitory Factor Receptor alpha Subunit/agonists , Leukemia Inhibitory Factor Receptor alpha Subunit/antagonists & inhibitors , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Middle Aged , Placenta/drug effects , Placenta/pathology , Polyethylene Glycols/pharmacology , Pregnancy , Pregnancy, Tubal/pathology , Pregnancy, Tubal/surgery , STAT3 Transcription Factor/agonists , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Spheroids, Cellular , Tissue Culture Techniques , Young Adult
2.
Mol Cell Biol ; 25(13): 5639-47, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15964819

ABSTRACT

The SSB family is comprised of four highly homologous proteins containing a C-terminal SOCS box motif and a central SPRY domain. No function has yet been ascribed to any member of this family in mammalian species despite a clear role for other SOCS proteins in negative regulation of cytokine signaling. To investigate its physiological role, the murine Ssb-2 gene was deleted by homologous recombination. SSB-2-deficient mice were shown to have a reduced rate of platelet production, resulting in very mild thrombocytopenia (25% decrease in circulating platelets). Tissue histology and other hematological parameters were normal, as was the majority of serum biochemistry, with the exception that blood urea nitrogen (BUN) levels were decreased in mice lacking SSB-2. Quantitative analysis of SSB mRNA levels indicated that SSB-1, -2, and -3 were ubiquitously expressed; however, SSB-4 was only expressed at very low levels. SSB-2 expression was observed in the kidney and in megakaryocytes, a finding consistent with the phenotype of mice lacking this gene. Deletion of SSB-2 thus perturbs the steady-state level of two tightly controlled homeostatic parameters and identifies a critical role for SSB-2 in regulating platelet production and BUN levels.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Deletion , Repressor Proteins/chemistry , Repressor Proteins/genetics , Thrombocytopenia/etiology , Thrombocytopenia/genetics , Trans-Activators/chemistry , Trans-Activators/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , Blood Platelets/metabolism , Blood Urea Nitrogen , DNA-Binding Proteins/physiology , Mice , Protein Structure, Tertiary/genetics , RNA, Messenger/metabolism , Recombination, Genetic , Repressor Proteins/physiology , Sequence Deletion , Stem Cells , Suppressor of Cytokine Signaling Proteins , Trans-Activators/physiology
3.
Proc Natl Acad Sci U S A ; 98(23): 13261-5, 2001 Nov 06.
Article in English | MEDLINE | ID: mdl-11606785

ABSTRACT

Suppressor of Cytokine Signaling-1 (SOCS-1) is an essential physiological inhibitor of IFN-gamma signaling. Mice lacking this gene die in the early postnatal period from a disease characterized by hyperresponsiveness to endogenous IFN-gamma. The SOCS box is a C-terminal domain shared with over 30 other proteins that links SOCS proteins to an E3 ubiquitin ligase activity and the proteasome, but whether it contributes to inhibition of cytokine signaling is currently disputed. We have deleted only the SOCS box of the SOCS-1 gene in mice and show that such mice have an increased responsiveness to IFN-gamma and slowly develop a fatal inflammatory disease. These results demonstrate that deletion of the SOCS box leads to a partial loss of function of SOCS-1.


Subject(s)
Carrier Proteins/physiology , Cytokines/antagonists & inhibitors , Repressor Proteins , Animals , Carrier Proteins/genetics , Cells, Cultured , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins
4.
Mol Cell Biol ; 21(18): 6189-97, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11509662

ABSTRACT

The Asbs are a family of ankyrin repeat proteins that, along with four other protein families, contain a C-terminal SOCS box motif, which was first identified in the suppressor of cytokine signaling (SOCS) proteins. While it is clear that the SOCS proteins are involved in the negative regulation of cytokine signaling, the biological roles of the other SOCS box-containing families are unknown. We have investigated Asb-1 function by generating mice that lack this protein, as well as mice that overexpress full-length or truncated Asb-1 in a wide range of tissues. Although Asb-1 is expressed in multiple organs, including the hematopoietic compartment in wild-type mice, Asb-1(-/-) mice develop normally and exhibit no anomalies of mature blood cells or their progenitors. While most organs in these mice appear normal, the testes of Asb-1(-/-) mice display a diminution of spermatogenesis with less complete filling of seminiferous tubules. In contrast, the widespread overexpression of Asb-1 in the mouse has no apparent deleterious effects.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Gene Expression Regulation , Amino Acid Sequence , Animals , Mice , Mice, Knockout , Molecular Sequence Data , Sequence Alignment , Suppressor of Cytokine Signaling Proteins
5.
Proc Natl Acad Sci U S A ; 98(16): 9324-9, 2001 Jul 31.
Article in English | MEDLINE | ID: mdl-11481489

ABSTRACT

Mice lacking suppressor of cytokine signaling 3 (SOCS3) exhibited embryonic lethality with death occurring between days 11 and 13 of gestation. At this stage, SOCS3(-/-) embryos were slightly smaller than wild type but appeared otherwise normal, and histological analysis failed to detect any anatomical abnormalities responsible for the lethal phenotype. Rather, in all SOCS3(-/-) embryos examined, defects were evident in placental development that would account for their developmental arrest and death. The placental spongiotrophoblast layer was significantly reduced and accompanied by increased numbers of giant trophoblast cells. Delayed branching of the chorioallantois was evident, and, although embryonic blood vessels were present in the labyrinthine layer of SOCS3(-/-) placentas, the network of embryonic vessels and maternal sinuses was poorly developed. Yolk sac erythropoiesis was normal, and, although the SOCS3(-/-) fetal liver was small at day 12.5 of gestation (E12.5), normal frequencies of erythroblasts and hematopoietic progenitor cells, including blast forming unit-erythroid (BFU-E) and, colony forming unit-erythroid (CFU-E) were present at both E11.5 and E12.5. Colony formation for both BFU-E and CFU-E from SOCS3(-/-) mice displayed wild-type quantitative responsiveness to erythropoietin (EPO), in the presence or absence of IL-3 or stem cell factor (SCF). These data suggest that SOCS3 is required for placental development but dispensable for normal hematopoiesis in the mouse embryo.


Subject(s)
Fetal Death/genetics , Proteins/genetics , Repressor Proteins , Transcription Factors , Animals , Base Sequence , DNA Primers , Erythropoietin/physiology , Hematopoiesis/genetics , Mice , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins
6.
Int J Hematol ; 73(3): 292-8, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11345194

ABSTRACT

The interaction of a cytokine with its specific cell surface receptor triggers the activation of intracellular signaling pathways that ultimately program the cellular response. Although the specific components and actions of the pathways driving these responses, such as the Janus kinase (JAK)/signal transducers and activators of transcription (STAT) pathway, are relatively well defined, it is becoming clear that important mechanisms exist to restrain these signaling cascades. This review discusses the key biochemical actions and biological roles of the phosphatase SHP-1, the protein inhibitors of activated STATs (PIAS) and the suppressor of cytokine signaling (SOCS) protein family in the negative regulation of cytokine signal transduction.


Subject(s)
Cytokines/physiology , DNA-Binding Proteins , Intracellular Signaling Peptides and Proteins , Repressor Proteins , Signal Transduction/physiology , Trans-Activators , Animals , Carrier Proteins/genetics , Carrier Proteins/physiology , Dimerization , Enzyme Activation , Gene Expression Regulation/physiology , Gene Targeting , Humans , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Phosphorylation , Protein Inhibitors of Activated STAT , Protein Processing, Post-Translational , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/physiology , Protein-Tyrosine Kinases/physiology , Proteins/genetics , Proteins/physiology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Transcription Factors/physiology , src Homology Domains
7.
J Biol Chem ; 276(25): 22086-9, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11306591

ABSTRACT

Suppressor of cytokine signaling-1 (SOCS-1) is a cytokine-inducible intracellular protein that functions to negatively regulate cytokine signal transduction pathways. Studies in vitro have shown that constitutive overexpression of SOCS-1 inhibits signaling in response to a range of cytokines, including interferons (IFN). Mice lacking SOCS-1 die from a complex disease characterized by liver degeneration and massive inflammation. Whereas there is clear evidence of increased IFNgamma signaling in SOCS-1(-/-) mice, it is unclear to what extent this is due to increased IFNgamma levels or to increased IFNgamma sensitivity. Here we have used SOCS-1(-/-) IFNgamma(-/-) mice, which remain healthy and produce no endogenous IFNgamma, to demonstrate that in vitro and in vivo hepatocytes lacking SOCS-1 exhibit a prolonged response to IFNgamma and that this correlates with a dramatically increased sensitivity to the toxic effects of IFNgamma in vivo. Thus, SOCS-1 is required for the timely attenuation of IFNgamma signaling in vivo.


Subject(s)
Carrier Proteins/physiology , Interferon-gamma/metabolism , Repressor Proteins , Signal Transduction/physiology , Animals , Hepatocytes/metabolism , In Vitro Techniques , Mice , Mice, Inbred Strains , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins
8.
Gene ; 258(1-2): 31-41, 2000 Nov 27.
Article in English | MEDLINE | ID: mdl-11111040

ABSTRACT

Members of the suppressor of cytokine signalling (SOCS) family of proteins have been shown to inhibit cytokine signalling via direct interactions with JAK kinases or activated cytokine receptors. In addition to their novel amino-terminal regions and SH2 domains that mediate these interactions, the SOCS proteins also contain carboxy-terminal regions of homology called the SOCS box. The SOCS box serves to couple SOCS proteins and their binding partners with the elongin B and C complex, possibly targeting them for degradation. Several other families of proteins also contain SOCS boxes but differ from the SOCS proteins in the type of domain or motif they contain upstream of the SOCS box. We report here the cloning, characterization, mapping and expression analysis of four members of the ankyrin repeat and SOCS box-containing (Asb) protein family.


Subject(s)
Ankyrin Repeat/genetics , Carrier Proteins/genetics , Genes/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Chromosome Mapping , Cloning, Molecular , DNA/chemistry , DNA/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , Exons , Gene Expression , Humans , Introns , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution
9.
Exp Hematol ; 28(10): 1105-12, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11027828

ABSTRACT

While positive effectors of cytokine signaling pathways are relatively well defined, negative regulation can be just as important but is poorly understood. The recently discovered suppressor of cytokine signaling (SOCS) family of proteins has been implicated in the negative regulation of several cytokine pathways, particularly the receptor-associated tyrosine kinase/signal transducer and activator of transcription (AK/STAT) pathways of transcriptional activation. Biochemical studies revealed that inhibition can occur via a variety of mechanisms. SOCS proteins bind to tyrosine-phosphorylated residues of target proteins via their SH2 domains, then inhibit JAK activity through their N-terminal domains, and are thought to induce degredation of bound molecules through a conserved SOCS-box motif that interacts with the proteasome. SOCS protein expression is induced by a wide variety of cytokines with each member displaying varying kinetics of induction. Gene modification studies in mice have demonstrated that SOCS-1 has a clear role in the negative regulation of interferon-gamma signaling, while other SOCS family members have also been shown to be involved in the regulation of T cell, growth hormone, and erythropoietin signaling systems.


Subject(s)
Carrier Proteins/physiology , Cytokines/antagonists & inhibitors , DNA-Binding Proteins , Intracellular Signaling Peptides and Proteins , Proteins/physiology , Repressor Proteins , Trans-Activators , Transcription Factors , Animals , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Feedback , Gene Expression , Humans , Proteins/genetics , Proteins/pharmacology , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins
10.
Blood ; 96(4): 1588-90, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10942411

ABSTRACT

Mice lacking both the gene encoding the shared receptor for granulocyte macrophage-colony-stimulating factor (GM-CSF), interleukin-3 (IL-3), and IL-5 common beta-chain (B(c)) and the gene for the IL-3 specific receptor (BIL3) were generated. This was achieved by targeting the B(c) locus in embryonic stem cells that were heterozygous for a null mutation of BIL3. Cells from mice generated with the doubly targeted embryonic stem cells were unresponsive to all 3 cytokines. Considerable previous data suggested a role for common beta-chain (beta(c)) in modulating signaling of cytokines including erythropoietin (EPO), G-CSF, and stem cell factor (SCF). However, bone marrow cells from mice lacking beta(c) and beta(IL3) showed normal responsiveness to these cytokines. Thus, there was no evidence for a biologically significant interaction between signaling via beta(c) or beta(IL3) and signaling by EPO, G-CSF, or SCF. Previously documented biochemical phenomena, including receptor transmodulation, receptor transphosphorylation, and even direct physical interaction, involving the beta(c)/beta IL-3 receptor systems do not reflect genuine interactions of physiological significance in primary hematopoietic cells. This study provided results that challenge conclusions previously established using a variety of biochemical assays. (Blood. 2000;96:1588-1590)


Subject(s)
Proto-Oncogene Proteins c-kit/genetics , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Interleukin-3/genetics , Signal Transduction/genetics , Animals , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoiesis/genetics , Interleukin-3/metabolism , Mice , Proto-Oncogene Proteins c-kit/metabolism , Receptor Cross-Talk , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Receptors, Interleukin-3/metabolism , Sequence Deletion , Stem Cell Factor/metabolism
11.
Nature ; 405(6790): 1069-73, 2000 Jun 29.
Article in English | MEDLINE | ID: mdl-10890450

ABSTRACT

Suppressor of cytokine signalling-2 (SOCS-2) is a member of the suppressor of cytokine signalling family, a group of related proteins implicated in the negative regulation of cytokine action through inhibition of the Janus kinase (JAK) signal transducers and activators of transcription (STAT) signal-transduction pathway. Here we use mice unable to express SOCS-2 to examine its function in vivo. SOCS-2(-/-) mice grew significantly larger than their wild-type littermates. Increased body weight became evident after weaning and was associated with significantly increased long bone lengths and the proportionate enlargement of most organs. Characteristics of deregulated growth hormone and insulin-like growth factor-I (IGF-I) signalling, including decreased production of major urinary protein, increased local IGF-I production, and collagen accumulation in the dermis, were observed in SOCS-2-deficient mice, indicating that SOCS-2 may have an essential negative regulatory role in the growth hormone/IGF-I pathway.


Subject(s)
DNA-Binding Proteins , Gigantism/etiology , Proteins/physiology , Repressor Proteins , Signal Transduction , Trans-Activators , Animals , Body Weight , Cytokines/metabolism , Female , Gigantism/genetics , Insulin-Like Growth Factor I/physiology , Male , Mice , Mice, Inbred C57BL , Mutagenesis , Proteins/genetics , Recombination, Genetic , Stem Cells , Suppressor of Cytokine Signaling Proteins
12.
Proc Natl Acad Sci U S A ; 97(12): 6493-8, 2000 Jun 06.
Article in English | MEDLINE | ID: mdl-10829066

ABSTRACT

Suppressor of cytokine signaling-3 (SOCS-3) is one member of a family of intracellular inhibitors of signaling pathways initiated by cytokines that use, among others, the common receptor subunit gp130. The SH2 domain of SOCS-3 has been shown to be essential for this inhibitory activity, and we have used a quantitative binding analysis of SOCS-3 to synthetic phosphopeptides to map the potential sites of interaction of SOCS-3 with different components of the gp130 signaling pathway. The only high-affinity ligand found corresponded to the region of gp130 centered around phosphotyrosine-757 (pY757), previously shown to be a docking site for the tyrosine phosphatase SHP-2. By contrast, phosphopeptides corresponding to other regions within gp130, Janus kinase, or signal transducer and activator of transcription proteins bound to SOCS-3 with weak or undetectable affinity. The significance of pY757 in gp130 as a biologically relevant SOCS-3 docking site was investigated by using transfected 293T fibroblasts. Although SOCS-3 inhibited signaling in cells transfected with a chimeric receptor containing the wild-type gp130 intracellular domain, inhibition was considerably impaired for a receptor carrying a Y-->F point mutation at residue 757. Taken together, these data suggest that the mechanism by which SOCS-3 inhibits the gp130 signaling pathway depends on recruitment to the phosphorylated gp130 receptor, and that some of the negative regulatory roles previously attributed to the phosphatase SHP-2 might in fact be caused by the action of SOCS-3.


Subject(s)
Antigens, CD/metabolism , Membrane Glycoproteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Proteins/metabolism , Repressor Proteins , Signal Transduction , Transcription Factors , Amino Acid Sequence , Binding Sites , Cytokine Receptor gp130 , Intracellular Signaling Peptides and Proteins , Janus Kinase 1 , Molecular Sequence Data , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein-Tyrosine Kinases/metabolism , SH2 Domain-Containing Protein Tyrosine Phosphatases , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins
13.
Protein Sci ; 9(4): 671-82, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10794409

ABSTRACT

The backbone dynamics of the four-helical bundle cytokine leukemia inhibitory factor (LIF) have been investigated using 15N NMR relaxation and amide proton exchange measurements on a murine-human chimera, MH35-LIF. For rapid backbone motions (on a time scale of 10 ps to 100 ns), as probed by 15N relaxation measurements, the dynamics parameters were calculated using the model-free formalism incorporating the model selection approach. The principal components of the inertia tensor of MH35-LIF, as calculated from its NMR structure, were 1:0.98:0.38. The global rotational motion of the molecule was, therefore, assumed to be axially symmetric in the analysis of its relaxation data. This yielded a diffusion anisotropy D(parallel)/D(perpendicular) of 1.31 and an effective correlation time (4D(perpendicular) + 2D(parallel))(-1) of 8.9 ns. The average values of the order parameters (S2) for the four helices, the long interhelical loops, and the N-terminus were 0.91, 0.84, and 0.65, respectively, indicating that LIF is fairly rigid in solution, except at the N-terminus. The S2 values for the long interhelical loops of MH35-LIF were higher than those of their counterparts in short-chain members of the four-helical bundle cytokine family. Residues involved in LIF receptor binding showed no consistent pattern of backbone mobilities, with S2 values ranging from 0.71 to 0.95, but residues contributing to receptor binding site III had relatively lower S2 values, implying higher amplitude motions than for the backbone of sites I and II. In the relatively slow motion regime, backbone amide exchange measurements showed that a number of amides from the helical bundle exchanged extremely slowly, persisting for several months in 2H2O at 37 degrees C. Evidence for local unfolding was considered, and correlations among various structure-related parameters and the backbone amide exchange rates were examined. Both sets of data concur in showing that LIF is one of the most rigid four-helical bundle cytokines.


Subject(s)
Growth Inhibitors/chemistry , Interleukin-6 , Lymphokines/chemistry , Amides/chemistry , Binding Sites , Leukemia Inhibitory Factor , Nitrogen Isotopes , Protein Conformation
14.
Biochem J ; 345 Pt 1: 25-32, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10600635

ABSTRACT

The receptor gp130 is used by the interleukin-6 (IL-6)-type cytokines, which include IL-6 and leukaemia-inhibitory factor (LIF). To investigate the role of the three extracellular membrane-proximal fibronectin-type-III-like (FNIII) modules of gp130 and the related receptor for granulocyte colony-stimulating factor (G-CSFR) in cytokine signal transduction we have transfected into murine myeloid M1-UR21 cells the chimaera (GR-FNIII)gp130, which contains the membrane-proximal FNIII modules of the G-CSFR on a gp130 backbone, and its complement, the chimaera (gp130-FNIII)GR. Whereas the binding affinities of (125)I-labelled IL-6 to (GR-FNIII)gp130, or of (125)I-Tyr1,3-G-CSF to (gp130-FNIII)GR, were similar to wild-type gp130 and wild-type G-CSFR, respectively, (125)I-LIF failed to bind with high affinity to (GR-FNIII)gp130. In assays measuring differentiation the (gp130-FNIII)GR cells were fully responsive to G-CSF, whereas the (GR-FNIII)gp130 cells responded fully to the agonistic anti-gp130 monoclonal antibody (mAb) B-S12, but not to IL-6 or LIF. Neutralizing mAbs that recognize the membrane-proximal FNIII modules of gp130 or the G-CSFR differentially interfered with signalling by B-S12, LIF and G-CSF. The data suggest that B-S12 and G-CSF induce the correct orientation or conformation for signalling by the wild-type and chimaeric homodimeric receptors, that the membrane-proximal region of gp130 is important for the correct formation of the signalling IL-6-IL-6 receptor-gp130 complex and that this region is also involved in LIF-dependent receptor heterodimerization and signalling.


Subject(s)
Antigens, CD/chemistry , Antigens, CD/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Animals , Antibodies, Monoclonal , Antigens, CD/genetics , Base Sequence , Cell Differentiation , Cell Line , Cell Membrane/metabolism , Cytokine Receptor gp130 , DNA Primers/genetics , Dimerization , Humans , Interleukin-6/metabolism , Ligands , Membrane Glycoproteins/genetics , Mice , Protein Structure, Quaternary , Receptors, Granulocyte Colony-Stimulating Factor/chemistry , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction
15.
J Leukoc Biol ; 66(4): 588-92, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10534114

ABSTRACT

SOCS-1 was originally identified as an inhibitor of interleukin-6 signal transduction and is a member of a family of proteins (SOCS-1 to SOCS-7 and CIS) that contain an SH2 domain and a conserved carboxyl-terminal SOCS box motif. Mutation studies have established that critical contributions from both the amino-terminal and SH2 domains are essential for SOCS-1 and SOCS-3 to inhibit cytokine signaling. Inhibition of cytokine-dependent activation of STAT3 occurred in cells expressing either SOCS-1 or SOCS-3, but unlike SOCS-1, SOCS-3 did not directly interact with or inhibit the activity of JAK kinases. Although the conserved SOCS box motif appeared to be dispensable for SOCS-1 and SOCS-3 action when overexpressed, this domain interacts with elongin proteins and may be important in regulating protein turnover. In gene knockout studies, SOCS-1(-/-) mice were born but failed to thrive and died within 3 weeks of age with fatty degeneration of the liver and hemopoietic infiltration of several organs. The thymus in SOCS-1(-/-) mice was small, the animals were lymphopenic, and deficiencies in B lymphocytes were evident within hemopoietic organs. We propose that the absence of SOCS-1 in these mice prevents lymphocytes and liver cells from appropriately controlling signals from cytokines with cytotoxic side effects.


Subject(s)
Carrier Proteins/physiology , Intracellular Signaling Peptides and Proteins , Repressor Proteins , Signal Transduction , Animals , Carrier Proteins/genetics , Humans , Mice , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , src Homology Domains
16.
Cell ; 98(5): 597-608, 1999 Sep 03.
Article in English | MEDLINE | ID: mdl-10490099

ABSTRACT

Mice lacking suppressor of cytokine signaling-1 (SOCS1) develop a complex fatal neonatal disease. In this study, SOCS1-/- mice were shown to exhibit excessive responses typical of those induced by interferon gamma (IFNgamma), were hyperresponsive to viral infection, and yielded macrophages with an enhanced IFNgamma-dependent capacity to kill L. major parasites. The complex disease in SOCS1-/- mice was prevented by administration of anti-IFNgamma antibodies and did not occur in SOCS1-/- mice also lacking the IFNgamma gene. Although IFNgamma is essential for resistance to a variety of infections, the potential toxic action of IFNgamma, particularly in neonatal mice, appears to require regulation. Our data indicate that SOCS1 is a key modulator of IFNgamma action, allowing the protective effects of this cytokine to occur without the risk of associated pathological responses.


Subject(s)
Carrier Proteins/physiology , Gene Expression Regulation, Developmental , Interferon-gamma/antagonists & inhibitors , Repressor Proteins , Signal Transduction , Alphavirus Infections/mortality , Alphavirus Infections/prevention & control , Animals , Disease Susceptibility , Interferon-gamma/pharmacology , Interferon-gamma/physiology , Leishmania major/immunology , Leishmaniasis/mortality , Leishmaniasis/prevention & control , Lymphopenia/mortality , Lymphopenia/prevention & control , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Semliki forest virus/immunology , Semliki forest virus/metabolism , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins
17.
J Neurochem ; 73(1): 96-104, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10386959

ABSTRACT

Schwann cells play a major role in promoting nerve survival and regeneration after injury. Their activities include providing neurotrophic factors and increasing the production of extracellular matrix components and cell surface adhesion molecules to promote axon regeneration. Following nerve transection, leukemia inhibitory factor (LIF) is up-regulated by Schwann cells at the injury site. LIF receptors are also up-regulated at the nerve injury site, but their cellular localization and function have not been fully characterized. We demonstrate that Schwann cells express mRNAs for LIF and the LIF receptor components LIF receptor subunit beta and glycoprotein 130 in vitro. We also show that although LIF is not required for the genesis of Schwann cells, it can potentiate the survival of differentiated Schwann cells in the context of neuregulin support. Not only does exogenous LIF promote survival under these conditions, but addition of the soluble LIF receptor (LIF binding protein) and anti-LIF antibodies significantly reduced cell survival, suggesting that LIF exerts autocrine effects. These results suggest that Schwann cell survival following nerve injury is potentially modulated by LIF.


Subject(s)
Cell Survival/physiology , Growth Inhibitors/physiology , Interleukin-6 , Lymphokines/physiology , Schwann Cells/physiology , Animals , Cell Division , Cells, Cultured , DNA/biosynthesis , Embryo, Mammalian , Gene Deletion , Gene Expression , Growth Inhibitors/genetics , Growth Inhibitors/pharmacology , Leukemia Inhibitory Factor , Leukemia Inhibitory Factor Receptor alpha Subunit , Lymphokines/genetics , Lymphokines/pharmacology , Mice , RNA, Messenger/metabolism , Receptors, Cytokine/genetics , Receptors, Cytokine/physiology , Receptors, OSM-LIF , Reverse Transcriptase Polymerase Chain Reaction , Sciatic Nerve/cytology
18.
Curr Biol ; 9(11): 605-8, 1999 Jun 03.
Article in English | MEDLINE | ID: mdl-10359701

ABSTRACT

Cytokines control a variety of cellular responses including proliferation, differentiation, survival and functional activation, via binding to specific receptors expressed on the surface of target cells [1]. The cytokine receptors of the haemopoietin family are defined by the presence of a conserved 200 amino acid extracellular domain known as the haemopoietin domain [2]. We report here the isolation of NR6, a haemopoietin receptor that, like the p40 subunit of interleukin-12 (IL-12) [3] and the EBI3 gene induced by Epstein-Barr virus infection in lymphocytes [4], contains a typical haemopoietin domain but lacks transmembrane and cytoplasmic domains. Although in situ hybridisation revealed NR6 expression at multiple sites in the developing embryo, mice lacking NR6 did not display obvious abnormalities and were born in the expected numbers. Neonatal NR6(-/-) mice failed to suckle, however, and died within 24 hours of birth, suggesting that NR6 is necessary for the recognition or processing of pheromonal signals or for the mechanics of suckling itself. In addition, NR6(-/-) mice had reduced numbers of haemopoietic progenitor cells, suggesting a potential role in the regulation of primitive haemopoiesis.


Subject(s)
Animals, Suckling/physiology , Carrier Proteins/physiology , Hematopoiesis/physiology , Receptors, Cell Surface , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Receptors, Leptin , Sequence Homology, Amino Acid , Solubility
19.
Leukemia ; 13(6): 926-34, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10360382

ABSTRACT

Mice with homozygous inactivation of the gene encoding the suppressor of cytokine signaling-1 (SOCS-1) protein die within 21 days of birth with low body weight, fatty degeneration and necrosis of the liver, infiltration of the lung, pancreas, heart and skin by macrophages and granulocytes and a profound depletion of T- and B-lymphocytes. In the present study, SOCS-1 -/- mice were found to have a moderate neutrophilia, and reduced platelet and hematocrit levels. Replacement of the SOCS-1 gene by a lac-Z reporter gene allowed documentation by FACS sorting that at least a proportion of granulocyte-macrophage progenitor cells transcribe SOCS-1. Most hematopoietic progenitor cell frequencies were normal in -/- marrow as were the size and cellular content of colonies formed by -/- progenitor cells in response to various stimulating factors. However, there was an increased frequency of macrophage progenitor cells in -/- mice and, abnormally, one quarter of all progenitor cells were located in the liver. Progenitor cells from -/- mice were hyper-responsive to stimulation by GM-CSF but not by M-CSF or Multi-CSF (IL-3). Progenitor cells from -/- mice were also hypersensitive to inhibition by interferon-gamma (IFN-gamma), the degree of inhibition varying markedly with the stimulating factor used. The suppressive effects of IFN-gamma therefore appear to involve interactions with particular growth factor-initiated signals in -/- cells--interactions that are strongly modulated by the action of the SOCS-1 protein.


Subject(s)
Carrier Proteins/genetics , Hematopoiesis/genetics , Repressor Proteins , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Female , Hematopoietic Stem Cells/pathology , Interferon-gamma/physiology , Leukocytes, Mononuclear/pathology , Male , Mice , Mice, Knockout , Spleen/pathology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins
20.
Proc Natl Acad Sci U S A ; 96(5): 2071-6, 1999 Mar 02.
Article in English | MEDLINE | ID: mdl-10051596

ABSTRACT

The suppressors of cytokine signaling (SOCS) family of proteins act as intracellular inhibitors of several cytokine signal transduction pathways. Their expression is induced by cytokine activation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway and they act as a negative feedback loop by subsequently inhibiting the JAK/STAT pathway either by direct interaction with activated JAKs or with the receptors. These interactions are mediated at least in part by the SH2 domain of SOCS proteins but these proteins also contain a highly conserved C-terminal homology domain termed the SOCS box. Here we show that the SOCS box mediates interactions with elongins B and C, which in turn may couple SOCS proteins and their substrates to the proteasomal protein degradation pathway. Analogous to the family of F-box-containing proteins, it appears that the SOCS proteins may act as adaptor molecules that target activated cell signaling proteins to the protein degradation pathway.


Subject(s)
Carrier Proteins/metabolism , Cysteine Endopeptidases/metabolism , Cytokines/physiology , Intracellular Signaling Peptides and Proteins , Multienzyme Complexes/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Repressor Proteins , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Binding Sites , Carrier Proteins/chemistry , Cell Line , Elongin , Humans , Mice , Models, Chemical , Molecular Sequence Data , Proteasome Endopeptidase Complex , Proteins/chemistry , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Transfection , Tumor Cells, Cultured , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...