Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters











Publication year range
1.
Eur J Cell Biol ; 91(6-7): 496-505, 2012.
Article in English | MEDLINE | ID: mdl-22093287

ABSTRACT

The function of the liver as an important constituent of the immune system involved in innate as well as adaptive immunity is warranted by different highly specialized cell populations. As the major source of acute phase proteins, including secreted pathogen recognition receptors (PRRs), short pentraxins, components of the complement system or regulators of iron metabolism, hepatocytes are essential constituents of innate immunity and largely contribute to the control of a systemic inflammatory response. The production of acute phase proteins in hepatocytes is controlled by a variety of different cytokines released during the inflammatory process with IL-1- and IL-6-type cytokines as the leading regulators operating both as a cascade and as a network having additive, inhibitory, or synergistic regulatory effects on acute phase protein expression. Hence, IL-1ß substantially modifies IL-6-induced acute phase protein production as it almost completely abrogates production of acute phase proteins such as γ-fibrinogen, α(2)-macroglobulin or α(1)-antichymotrypsin, whereas production of for example hepcidin, C-reactive protein and serum amyloid A is strongly up-regulated. This switch-like regulation of IL-6-induced acute phase protein production by IL-1ß is due to a complex processing of the intracellular signaling events activated in response to IL-6 and/or IL-1ß, with the crosstalk between STAT3- and NF-κB-mediated signal transduction being of particular importance. Recent data suggest that in this context complex formation between STAT3 and the p65 subunit of NF-κB might be of key importance. The present review summarizes the regulation of acute phase protein production focusing on the role of the crosstalk of STAT3- and NF-κB-driven pathways for transcriptional control of acute phase gene expression.


Subject(s)
Acute-Phase Proteins/metabolism , Interleukin-1/metabolism , Interleukin-6/metabolism , Liver/metabolism , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Animals , Humans , Signal Transduction
2.
J Cell Sci ; 123(Pt 6): 947-59, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20200229

ABSTRACT

The inflammatory response involves a complex interplay of different cytokines which act in an auto- or paracrine manner to induce the so-called acute phase response. Cytokines are known to crosstalk on multiple levels, for instance by regulating the mRNA stability of targeted cytokines through activation of the p38-MAPK pathway. In our study we discovered a new mechanism that answers the long-standing question how pro-inflammatory cytokines and environmental stress restrict immediate signalling of interleukin (IL)-6-type cytokines. We show that p38, activated by IL-1beta, TNFalpha or environmental stress, impairs IL-6-induced JAK/STAT signalling through phosphorylation of the common cytokine receptor subunit gp130 and its subsequent internalisation and degradation. We identify MK2 as the kinase that phosphorylates serine 782 in the cytoplasmic part of gp130. Consequently, inhibition of p38 or MK2, deletion of MK2 or mutation of crucial amino acids within the MK2 target site or the di-leucine internalisation motif blocks receptor depletion and restores IL-6-dependent STAT activation as well as gene induction. Hence, a novel negative crosstalk mechanism for cytokine signalling is described, where cytokine receptor turnover is regulated in trans by pro-inflammatory cytokines and stress stimuli to coordinate the inflammatory response.


Subject(s)
Cytokine Receptor gp130/metabolism , Endocytosis , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Protein Processing, Post-Translational , Signal Transduction , Animals , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Endocytosis/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Humans , Interleukin-1beta/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Mice , NF-kappa B/metabolism , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Serine/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Chem Biol ; 16(7): 783-94, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19635415

ABSTRACT

Dysregulated cytokine production contributes to inflammatory and proliferative diseases. Therefore, inhibition of proinflammatory mediators such as TNF, IL-1, and IL-6 is of great clinical relevance. Actual strategies are aimed at preventing receptor activation through sequestration of the ligand. Here we describe the development of an inhibitor of murine IL-6 based on fused receptor fragments. Molecular modeling-guided analysis of the murine IL-6Ralpha revealed that mutations in the Ig-like domain D1 severely affect protein function, although D1 is not directly involved in the ligand-binding interface. The resulting single chain IL-6 inhibitor (mIL-6-RFP) consisting of domains D1-D3 of mgp130, a flexible linker, and domains D1-D3 of mIL-6Ralpha is a highly potent and specific IL-6 inhibitor. mIL-6-RFP will permit further characterization of the role of IL-6 in various disease models and could ultimately lead to anti-IL-6 therapy.


Subject(s)
Drug Design , Interleukin-6/antagonists & inhibitors , Protein Engineering/methods , Receptors, Interleukin-6/genetics , Animals , Inflammation/drug therapy , Ligands , Mice , Mutation
4.
Arthritis Rheum ; 60(7): 1932-43, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19565514

ABSTRACT

OBJECTIVE: To investigate the molecular mechanisms of CCL13/monocyte chemoattractant protein 4 (MCP-4) chemokine expression through proinflammatory cytokines in different primary human fibroblasts and the contribution of CCL13 to monocyte migration. METHODS: Using RNase protection assays and enzyme-linked immunosorbent assays, we quantified the expression of CCL13 compared with that of CCL2/MCP-1 in primary human fibroblasts. Boyden chamber assays were performed to determine the importance of CCL13 for migration of primary monocytes. Pharmacologic inhibitors as well as small interfering RNA knockdown approaches were used to investigate the signaling pathways regulating CCL13 expression. RESULTS: The interleukin-6 (IL-6)-type cytokine oncostatin M (OSM) was a powerful inducer of CCL13 expression in primary synovial fibroblasts from patients with rheumatoid arthritis (RA) as well as those from healthy control subjects but not in other types of fibroblasts. Neither IL-6 nor tumor necrosis factor alpha could stimulate the expression of CCL13 in synovial fibroblasts; IL-1beta was a very weak inducer. Synovial fibroblasts from patients with RA constitutively produced low amounts of CCL13, which was partially dependent on constitutive production of OSM. By investigating the underlying molecular mechanism, we identified STAT-5, ERK-1/2, and p38 as critical factors involved in OSM-dependent transcription and messenger RNA stabilization of CCL13. CONCLUSION: In contrast to other prominent cytokines involved in the pathogenesis of RA, OSM can strongly up-regulate the expression of CCL13, a chemokine recently identified in the synovial fluid of patients with RA. Despite potent OSM-induced signal transduction in all types of fibroblasts analyzed, only synovial fibroblasts secreted CCL13, which might be indicative of tissue-specific imprinting of different fibroblasts during development.


Subject(s)
Arthritis, Rheumatoid/metabolism , Fibroblasts/metabolism , Monocyte Chemoattractant Proteins/metabolism , Oncostatin M/metabolism , Synovial Membrane/metabolism , Arthritis, Rheumatoid/pathology , Cells, Cultured , Chemokine CCL2/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Interleukin-6/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/cytology , Monocytes/metabolism , Neutrophils/cytology , Neutrophils/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/physiology , Synovial Membrane/drug effects , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Hepatology ; 50(2): 585-91, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19582813

ABSTRACT

UNLABELLED: Interleukin-27 (IL-27) is a cytokine belonging to the IL-6/IL-12 cytokine family. It is secreted by antigen-presenting cells, strongly acts on T cells, and also stimulates innate immune cells. In most studies, the effects of IL-27 on T cells were investigated; however, not much is known about possible effects of IL-27 on other cell types. IL-27 signals via the common IL-6-type cytokine receptor chain gp130 and the IL-27-specific chain WSX-1. Given the importance of gp130 in regulating liver responses such as the acute phase response or liver regeneration, we investigated whether IL-27 could also have a function in liver cells. We find that IL-27 stimulates hepatoma cells and hepatocytes by inducing a sustained signal transducer and activator of transcription (STAT)1 and STAT3 activation. Whereas the STAT3 mediated responses to IL-27 (gamma-fibrinogen and hepcidin induction) are not detectable, we observe an interferon-gamma (IFN-gamma)-like STAT1 response leading to the induction of interferon-regulated proteins such as STAT1, STAT2, interferon response factor (IRF)-1, IRF-9, myxovirus resistance A and guanylate binding protein 2. CONCLUSION: Our study provides evidence for a function of IL-27 in hepatoma cells and hepatocytes and shows that IL-27 responses are not restricted to the classical immune cells. Our results suggest that IL-27 exerts IFN-like functions in liver cells and that it can contribute to the antiviral response in these cells.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Hepatocytes/metabolism , Interleukins/metabolism , Liver Neoplasms/metabolism , STAT1 Transcription Factor/metabolism , Animals , Antimicrobial Cationic Peptides/metabolism , Cell Line, Tumor , Fibrinogen/metabolism , Gene Expression Regulation , Hepcidins , Humans , Interferon-gamma/metabolism , Interleukins/immunology , Male , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Virus Replication
6.
J Immunol ; 182(5): 2969-77, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234192

ABSTRACT

The Janus kinases, Jaks, constitutively associate with the cytoplasmic region of cytokine receptors and play an important role in a multitude of biological processes. Jak2 dysfunction has been implicated in myeloproliferative diseases and leukemia. Although Jaks were studied extensively for many years, the molecular mechanism of Jak activation upon cytokine stimulation of cells is still incompletely understood. In this study, we investigated the importance of an unusual insertion located within the kinase domain in Jak2. We found that the deletion of this insertion, which we named the Jak-specific insertion (JSI), totally abrogates Jak2 autophosphorylation. We further point mutated four residues within the JSI that are conserved in all Jak family members. Three of these mutants showed abrogated or reduced autophosphorylation, whereas the fourth displayed increased autophosphorylation. We found that the phosphorylation state of these mutants is not influenced by other domains of the kinase. Our data further suggest that the JSI is not required for the negative regulation of kinase activity by the suppressor of cytokine signaling proteins, SOCS. Most importantly, we show that mutations in this region differentially affect IFN-gamma and erythropoietin signal transduction. Taken together, the dramatic effects on the phosphorylation status of Jak2 as well as the differential effects on the signaling via different cytokines highlight the importance of this unusual region for the catalytic activity of Jaks.


Subject(s)
Cytokines/physiology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mutagenesis, Insertional , Amino Acid Sequence , Animals , Catalytic Domain/genetics , Catalytic Domain/immunology , Cell Line , Cell Line, Tumor , Computer Simulation , Cytokines/biosynthesis , Enzyme Activation/genetics , Enzyme Activation/immunology , Humans , Janus Kinase 2/chemistry , Mice , Models, Molecular , Molecular Sequence Data , Point Mutation
8.
J Immunol ; 181(10): 7341-9, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18981157

ABSTRACT

The recruitment of leukocytes to injured tissue is crucial for the initiation of inflammatory responses as well as for immune surveillance to fight tumor progression. In this study, we show that oncostatin M, a member of the IL-6-type cytokine family and potent proinflammatory cytokine stimulates the expression of the chemokines CCL1, CCL7, and CCL8 in primary human dermal fibroblasts at a faster kinetic than IL-1beta or TNF-alpha. The production of CCL1 and CCL8 is important for migration of monocytes, while specific Abs against CCL1 additionally inhibit the migration of T lymphocytes. We identify the mitogen-activated protein kinases ERK1/2 and p38 as crucial factors for the enhanced expression of CCL1 and CCL8. Depletion of the ERK1/2 target genes c-Jun or c-Fos strongly decrease CCL1 and CCL8 expression, while p38 MAPK prolongs the half-life of CCL1, CCL7, and CCL8 mRNA through inhibition of tristetraprolin. None of the STAT transcription factors STAT1, STAT3, or STAT5 stimulate transcription of CCL1 or CCL8. However, we identify a negative regulatory function of activated STAT5 for the gene expression of CCL1. Importantly, not STAT5 itself, but its target gene cytokine inducible SH2-domain containing protein is required for the STAT5 inhibitory effect on CCL1 expression. Finally, we show that constitutive activation of STAT5 through a mutated form of JAK2 (JAK2 V617F) occurring in patients with myeloproliferative disorders similarly suppresses CCL1 expression. Taken together, we identify novel important inflammatory target genes of OSM which are independent of STAT signaling per se, but depend on MAPK activation and are partly repressed through STAT5-dependent expression of cytokine inducible SH2-domain containing protein.


Subject(s)
Chemokines/biosynthesis , Fibroblasts/immunology , Gene Expression Regulation/immunology , Oncostatin M/metabolism , Signal Transduction/immunology , Animals , Cells, Cultured , Chemokine CCL1/biosynthesis , Chemokine CCL7/biosynthesis , Chemokine CCL8/biosynthesis , Chemotaxis, Leukocyte/immunology , Enzyme Activation/immunology , Enzyme-Linked Immunosorbent Assay , Fibroblasts/metabolism , Gene Expression , Humans , Janus Kinase 2/immunology , Janus Kinase 2/metabolism , Mice , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/immunology , STAT5 Transcription Factor/metabolism , Skin/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Suppressor of Cytokine Signaling Proteins/metabolism , Transfection
9.
Cell Signal ; 20(7): 1385-91, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18450421

ABSTRACT

The transmembrane glycoprotein signal regulatory protein/SHP2-substrate (SIRP1alpha/SHPS-1) has been implicated in growth factor- and cell adhesion-induced signalling. Here we report on the contribution of SIRP1alpha to IL-6 type cytokine signalling. SIRP1alpha binds the protein tyrosine phosphatase SHP2 upon treatment with interleukin-6 in a stimulation-dependent manner. Mouse embryonic fibroblasts expressing a SIRP1alpha protein which lacks the intracellular part show enhanced SHP2 phosphorylation and ERK1/2 activation in response to IL-6, suggesting that SIRP1alpha affects IL-6-signalling through SHP2. Whereas SHP2 phosphorylation is enhanced in SIRP1alpha-deficient cells STAT3 activation is delayed and STAT3-dependent gene induction is reduced which correlates with reduced STAT3 serine phosphorylation. Our results indicate that SIRP1alpha contributes to IL-6 signalling by counteracting SHP2 phosphorylation which consequently affects ERK-activation and STAT3-dependent transactivation as well as target gene expression. Our observations will help to understand the tight balance of MAPK- and STAT3-activation in response to IL-6 which was found to be misbalanced in many autoimmune diseases, inflammatory proliferative diseases and cancer.


Subject(s)
Interleukin-6/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Animals , Binding Sites , Cell Line , Cytokine Receptor gp130/metabolism , Enzyme Induction/drug effects , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression Regulation/drug effects , Humans , Interleukin-6/pharmacology , Mice , Models, Biological , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Proteins c-fos/genetics , Receptors, Immunologic/deficiency , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Transcriptional Activation
10.
J Biol Chem ; 283(28): 19465-77, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18430728

ABSTRACT

Human and murine oncostatin M (OSM) induce their bioactivities through a heterodimeric receptor complex consisting of gp130 and the OSM receptor (OSMR), which initiates a signaling pathway involving Janus kinases (JAKs) and transcription factors of the signal transducers and activators of transcription (STAT) family. In contrast to the signal transducing receptor subunit gp130, the OSMR allows strong activation of STAT5B. The underlying molecular mechanism, however, remained unclear. Here we demonstrate that the human and murine OSM receptors use distinct mechanisms for STAT5B activation. The human receptor contains a STAT5B recruiting tyrosine motif (Tyr837/Tyr839) C-terminal to the box 1/2 region, which is absent in the mouse receptor. In contrast, the murine receptor initiates STAT5 activation directly via the receptor bound Janus kinases. Intriguingly, the murine receptor preferentially recruits JAK2, whereas the human receptor seems to have a higher affinity for JAK1. We identify a single amino acid (Phe820) in the human receptor that is responsible for this preference. Exchange by the murine counterpart (Cys815) allows recruitment of JAK2 by the human receptor and consequently activation of STAT5B independently of receptor tyrosine motifs. STAT5B interacts directly with JAK2 only in response to activation of the murine OSMR or the mutated human OSMR. Additionally, we show that OSM-induced STAT1 phosphorylation occurs independently of receptor tyrosine motifs and is mediated directly by Janus kinases, whereas the two C-terminally located tyrosine residues Tyr917/Tyr945 of the OSMR are crucial for STAT3 activation.


Subject(s)
Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Receptors, Oncostatin M/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/physiology , Amino Acid Motifs/physiology , Animals , Cell Line, Tumor , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Humans , Janus Kinase 1/genetics , Janus Kinase 2/genetics , Mice , Receptors, Oncostatin M/genetics , STAT5 Transcription Factor/genetics , Species Specificity
11.
Biochem J ; 412(1): 65-72, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18271757

ABSTRACT

IL (interleukin)-6 exerts pro- as well as anti-inflammatory activities. Beside many other activities, IL-6 is the major inducer of acute phase proteins in the liver, acts as a differentiation factor for blood cells, as migration factor for T-cells and is a potent inducer of the chemokine MCP-1 (monocyte chemoattractant protein-1). Recent studies have focused on the negative regulation of IL-6 signal transduction through the IL-6-induced feedback inhibitors SOCS (suppressor of cytokine signalling) 1 and SOCS3 or the protein tyrosine phosphatases SHP-2 (Src homology 2 domain-containing protein tyrosine phosphatase 2) and TcPTP (T-cell protein tyrosine phosphatase). Studies on the cross-talk between pro-inflammatory mediators (IL-1, tumour necrosis factor, lipopolysaccharide) and IL-6 elucidated further regulatory mechanisms. Less is known about the regulation of IL-6 signal transduction by hormone/cytokine signalling through G-protein-coupled receptors. This is particularly surprising since many of these hormones (such as prostaglandins and chemokines) play an important role in inflammatory processes. In the present study, we have investigated the inhibitory activity of PGE(1) (prostaglandin E(1)) on IL-6-induced MCP-1 expression and have elucidated the underlying molecular mechanism. Surprisingly, PGE(1) does not affect IL-6-induced STAT (signal transducer and activator of transcription) 3 activation, but does affect ERK (extracellular-signal-regulated kinase) 1/2 activation which is crucial for IL-6-dependent expression of MCP-1. In summary, we have discovered a specific cross-talk between the adenylate cyclase cascade and the IL-6-induced MAPK (mitogen-activated protein kinase) cascade and have investigated its impact on IL-6-dependent gene expression.


Subject(s)
Alprostadil/pharmacology , Chemokine CCL2/genetics , Interleukin-6/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Animals , Cells, Cultured , Chemokine CCL2/metabolism , Colforsin/pharmacology , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/physiology , Down-Regulation/drug effects , Enzyme Activation/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pertussis Toxin/pharmacology , Signal Transduction/drug effects , Substrate Specificity/drug effects , src-Family Kinases/physiology
12.
J Biol Chem ; 283(10): 5985-95, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18174171

ABSTRACT

Fusion proteins of the extracellular parts of cytokine receptors, also known as cytokine traps, turned out to be promising cytokine inhibitors useful in anti-cytokine therapies. Here we present newly designed cytokine traps for murine and human leukemia inhibitory factor (LIF) as prototypes for inhibitors targeting cytokines that signal through a heterodimer of two signaling receptors of the glycoprotein 130 (gp130) family. LIF signals through a receptor heterodimer of LIF receptor (LIFR) and gp130 and induces the tyrosine phosphorylation of STAT3 leading to target gene expression. The analysis of various receptor fusion and deletion constructs revealed that a truncated form of the murine LIF receptor consisting of the first five extracellular domains was a potent inhibitor for human LIF. For the efficient inhibition of murine LIF, the cytokine-binding module of murine gp130 had to be fused to the first five domains of murine LIFR generating mLIF-RFP (murine LIFR fusion protein). The tyrosine phosphorylation of STAT3 and subsequent gene induction induced by human or murine LIF are completely blocked by the respective inhibitor. Furthermore, both inhibitors are specific and do not alter the bioactivities of the closely related cytokines interleukin (IL)-6 and oncostatin M. The gained knowledge on the construction of LIF inhibitors can be transferred to the design of inhibitors for related cytokines such as IL-31, IL-27, and oncostatin M for the treatment of inflammatory and malignant diseases.


Subject(s)
Cytokine Receptor gp130/metabolism , Leukemia Inhibitory Factor/antagonists & inhibitors , Receptors, OSM-LIF/metabolism , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/therapeutic use , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/metabolism , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary/genetics , Receptors, OSM-LIF/genetics , Receptors, OSM-LIF/therapeutic use , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
13.
J Immunol ; 180(2): 998-1007, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18178840

ABSTRACT

Jak1 is a tyrosine kinase that noncovalently forms tight complexes with a variety of cytokine receptors and is critically involved in signal transduction via cytokines. Jaks are predicted to have a 4.1, ezrin, radixin, moesin (FERM) domain at their N terminus. FERM domains are composed of three structurally unrelated subdomains (F1, F2, and F3) which are in close contact to one another and form the clover-shaped FERM domain. We generated a model structure of the Jak1 FERM domain, based on solved FERM structures and the alignments with other FERM domains. To destabilize different subdomains and to uncover their exact function, we mutated specific hydrophobic residues conserved in FERM domains and involved in hydrophobic core interactions. In this study, we show that the structural integrity of the F2 subdomain of the FERM domain of Jak1 is necessary to bind the IFN-gammaRalpha. By mutagenesis of hydrophobic residues in the hydrophobic core between the three FERM subdomains, we find that the structural context of the FERM domain is necessary for the inhibition of Jak1 phosphorylation. Thus, FERM domain mutations can have repercussions on Jak1 function. Interestingly, a mutation in the kinase domain (Jak1-K907E), known to abolish the catalytic activity, also leads to an impaired binding to the IFN-gammaRalpha when this mutant is expressed at endogenous levels in U4C cells. Our data show that the structural integrity of both the FERM domain and of the kinase domain is essential for both receptor binding and catalytic function/autoinhibition.


Subject(s)
Janus Kinase 1/metabolism , Receptors, Cytokine/metabolism , Receptors, Interferon/metabolism , Amino Acid Sequence , Humans , Interferon-gamma/metabolism , Janus Kinase 1/chemistry , Janus Kinase 1/genetics , Molecular Sequence Data , Mutation , Phosphorylation , Protein Structure, Tertiary/genetics , Interferon gamma Receptor
14.
J Cell Sci ; 120(Pt 18): 3249-61, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17726064

ABSTRACT

Persistent activation of the transcription factor STAT3 has been detected in many types of cancer and plays an important role in tumor progression, immune evasion and metastasis. To analyze persistent STAT3 activation we coexpressed STAT3 with v-Src. We found that tyrosine phosphorylation of STAT3 by v-Src is independent of Janus kinases (Jaks), the canonical activators of STATs. The STAT3-induced feedback inhibitor, suppressor of cytokine signaling 3 (SOCS3), did not interfere with STAT3 activation by v-Src. However, the protein inhibitor of activated STAT3 (PIAS3) suppressed gene induction by persistently activated STAT3. We measured nucleocytoplasmic shuttling of STAT3 in single cells by bleaching the YFP moiety of double-labelled STAT3-CFP-YFP in the cytoplasm. Analysis of the subcellular distribution of CFP and YFP fluorescence over time by mathematical modeling and computational parameter estimation revealed that activated STAT3 shuttles more rapidly than non-activated STAT3. Inhibition of exportin-1-mediated nuclear export slowed down nucleocytoplasmic shuttling of v-Src-activated STAT3 resulting in reduced tyrosine phosphorylation, decreased induction of STAT3 target genes and increased apoptosis. We propose passage of persistently activated STAT3 through the nuclear pore complex as a new target for intervention in cancer.


Subject(s)
Cytoplasm/metabolism , Nuclear Pore/metabolism , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Active Transport, Cell Nucleus/physiology , Animals , COS Cells , Chlorocebus aethiops , Cytoplasm/genetics , Humans , Karyopherins/genetics , Karyopherins/metabolism , Mice , Models, Biological , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , NIH 3T3 Cells , Nuclear Pore/genetics , Oncogene Protein pp60(v-src)/genetics , Oncogene Protein pp60(v-src)/metabolism , Phosphorylation , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , STAT3 Transcription Factor/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Exportin 1 Protein
15.
Cell Signal ; 19(9): 1866-78, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17543500

ABSTRACT

Despite the essential role of the fibrinogen gamma-chain as a blood clotting factor, the fibrinogen gamma-chain contains a number of interaction sites to recruit other factors such as leukocytes important for prevention of pathogen entry and propagation of the repair process. Interleukin-6 (IL-6) is known as the major inducer of gamma-fibrinogen synthesis in hepatocytes, whereas IL-1beta has been shown to act as a potent inhibitor of gamma-fibrinogen expression. Studies on the rat fibrinogen gamma-chain promoter suggest that nuclear factor (NF)-kappaB replaces the signal transducer and activator of transcription (STAT) 3 from binding to overlapping NF-kappaB/STAT3 binding sites within the 5' regulatory region of the rat gamma-chain gene promoter. However, despite its physiological relevance, the underlying mechanism responsible for the inhibitory effect of IL-1beta in humans is still not understood and apparently more complex. In contrast to the mechanism described for the rat gene our results indicate that IL-1beta suppresses the IL-6-induced activation of the human gamma-fibrinogen gene particularly by blocking the late phase STAT3-tyrosine phosphorylation NF-kappaB-dependently but independent from de novo protein synthesis. Consequently, blocking NF-kappaB activation restores specifically late phase STAT3 activation as well as the induction of the human gamma-fibrinogen gene. In contrast, specifically early STAT3 activation could be restored by a block of the p38 mitogen-activated protein kinase (p38(MAPK)) pathway. In summary, our results indicate that expression of the gamma-fibrinogen gene is mainly controlled by the strength of late phase STAT3 activation, which in turn is negatively regulated by the extent of IL-1beta-mediated NF-kappaB activity.


Subject(s)
Fibrinogen/genetics , Gene Expression Regulation/drug effects , Interleukin-1beta/pharmacology , Interleukin-6/pharmacology , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Animals , Base Sequence , Cell Line, Tumor , Enzyme Activation/drug effects , Fibrinogen/antagonists & inhibitors , Humans , I-kappa B Kinase/metabolism , Molecular Sequence Data , Rats , Regulatory Sequences, Nucleic Acid/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Mol Cancer Res ; 5(3): 271-81, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17374732

ABSTRACT

The growth of melanocytes and many early stage melanoma cells can be inhibited by cytokines, whereas late stage melanoma cells have often been reported to be "multi-cytokine-resistant." Here, we analyzed the melanoma cell line 1286, resistant towards the growth-inhibitory effects of interleukin 6 (IL-6), and oncostatin M (OSM), to better understand the mechanisms underlying cytokine resistance. Although the relevant receptors gp130 and OSMR are expressed at the cell surface of these cells, cytokine stimulation hardly led to the activation of Janus kinase 1 and signal transducer and activator of transcription (STAT)3 and STAT1. We found a high-level constitutive expression of suppressors of cytokine signaling 3 (SOCS3) that did not further increase after cytokine treatment. Importantly, upon suppression of SOCS3 by short interfering RNA, cells became susceptible towards OSM and IL-6: they showed an enhanced STAT3 phosphorylation and a dramatically increased STAT1 phosphorylation. Moreover, suppression of SOCS3 rendered 1286 cells sensitive to the antiproliferative action of IL-6 and OSM, but not of IFN-alpha. Interestingly, SOCS3-short interfering RNA treatment also increased the growth-inhibitory effect in cytokine-sensitive WM239 cells expressing SOCS3 in an inducible way. Thus, SOCS3 expression confers a growth advantage to these cell lines. Constitutive SOCS3 mRNA expression, although at lower levels than in 1286 cells, was found in nine additional human melanoma cell lines and in normal human melanocytes, although at the protein level, SOCS3 expression was marginal at best. However, in situ analysis of human melanoma specimens revealed SOCS3 immunoreactivity in 3 out of 10 samples, suggesting that in vivo SOCS3 may possibly play a role in IL-6 resistance in at least a fraction of tumors.


Subject(s)
Drug Resistance, Neoplasm/immunology , Interleukin-6/pharmacology , Melanoma/pathology , Skin Neoplasms/pathology , Suppressor of Cytokine Signaling Proteins/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cytokines/pharmacology , Drug Resistance, Neoplasm/genetics , Humans , Melanoma/chemistry , Melanoma/metabolism , Oncostatin M/pharmacology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Skin Neoplasms/chemistry , Skin Neoplasms/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/analysis , Suppressor of Cytokine Signaling Proteins/genetics
17.
J Immunol ; 178(5): 2813-26, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17312125

ABSTRACT

The potential of some proinflammatory mediators to inhibit gp130-dependent STAT3 activation by enhancing suppressor of cytokine signaling (SOCS) 3 expression represents an important molecular mechanism admitting the modulation of the cellular response toward gp130-mediated signals. Thus, it is necessary to understand the mechanisms involved in the regulation of SOCS3 expression by proinflammatory mediators. In this study, we investigate SOCS3 expression initiated by the proinflammatory cytokine TNF-alpha. In contrast to IL-6, TNF-alpha increases SOCS3 expression by stabilizing SOCS3 mRNA. Activation of the MAPK kinase 6 (MKK6)/p38(MAPK)-cascade is required for TNF-alpha-mediated stabilization of SOCS3 mRNA and results in enhanced SOCS3 protein expression. In fibroblasts or macrophages deficient for MAPK-activated protein kinase 2 (MK2), a downstream target of the MKK6/p38(MAPK) cascade, basal SOCS3-expression is strongly reduced and TNF-alpha-induced SOCS3-mRNA stabilization is impaired, indicating that MK2 is crucial for the control of SOCS3 expression by p38(MAPK)-dependent signals. As a target for SOCS3 mRNA stability-regulating signals, a region containing three copies of a pentameric AUUUA motif in close proximity to a U-rich region located between positions 2422 and 2541 of the 3' untranslated region of SOCS3 is identified. One factor that could target this region is the zinc finger protein tristetraprolin (TTP), which is shown to be capable of destabilizing SOCS3 mRNA via this region. However, data from TTP-deficient cells suggest that TTP does not play an irreplaceable role in the regulation of SOCS3 mRNA stability by TNF-alpha. In summary, these data indicate that TNF-alpha regulates SOCS3 expression on the level of mRNA stability via activation of the MKK6/p38(MAPK) cascade and that the activation of MK2, a downstream target of p38(MAPK), is important for the regulation of SOCS3 expression.


Subject(s)
Gene Expression Regulation/physiology , MAP Kinase Signaling System/physiology , RNA Stability/physiology , RNA, Messenger/biosynthesis , Suppressor of Cytokine Signaling Proteins/biosynthesis , Tumor Necrosis Factor-alpha/immunology , 3T3 Cells , Animals , Gene Expression Regulation/drug effects , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase 6/immunology , MAP Kinase Kinase 6/metabolism , MAP Kinase Signaling System/drug effects , Macrophages/enzymology , Macrophages/immunology , Mice , Protein Kinases/immunology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , RNA Stability/drug effects , RNA, Messenger/immunology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/immunology , Tristetraprolin/immunology , Tristetraprolin/metabolism , Tumor Necrosis Factor-alpha/pharmacology
18.
Mol Cancer Res ; 5(12): 1331-41, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18171991

ABSTRACT

Signal transducers and activators of transcriptions (STAT) are key mediators of cytokine signaling. Moreover, these transcription factors play a crucial role in oncogenic signaling where inappropriate and sustained activation of STATs, especially STAT3, is a trait of many different cancers and their derived cell lines. Constitutively active STAT3 has been reported to prevent programmed cell death and enhance cell proliferation, whereas the disruption of STAT3 signaling can inhibit tumor growth. The physiologic activation of STAT3 by cytokines has been well established; however, little is known about altered, stimulation-independent STAT3 activation. Here, we show that, in most but not all melanoma cell lines, STAT3 phosphorylation increased substantially with cell density and that this STAT3 was able to bind to DNA and to activate transcription. Inhibitor studies showed that the cell density-dependent STAT3 activation relies on Janus kinases (JAK) rather than Src kinases. Using a specific JAK inhibitor, sustained STAT3 activation was completely abrogated in all tested melanoma lines, whereas inhibition of Src or mitogen-activated protein kinase/extracellular signal-regulated kinase kinase 1/2 had no effect on constitutively tyrosine-phosphorylated STAT3 levels. Although STAT3 activation was completely blocked with JAK inhibitor I and to a lesser extent with the common JAK inhibitor AG490, only the latter compound markedly decreased proliferation and induced apoptosis. Taken together, variations in cell density can profoundly modify the extent of JAK-mediated persistent STAT3 phosphorylation; however, STAT3 activation was not sufficient to provide critical growth and survival signals in melanoma cell lines.


Subject(s)
Janus Kinases/metabolism , Melanoma/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Skin Neoplasms/metabolism , Apoptosis/physiology , Carcinoma, Hepatocellular , Cell Count , Cell Division/physiology , Cell Survival/physiology , HeLa Cells , Humans , Liver Neoplasms , Melanoma/pathology , Phosphorylation , Skin Neoplasms/pathology , src-Family Kinases/metabolism
19.
J Biol Chem ; 282(2): 1238-48, 2007 Jan 12.
Article in English | MEDLINE | ID: mdl-17085445

ABSTRACT

Although fusion proteins of the extracellular parts of receptor subunits termed cytokine traps turned out to be promising cytokine inhibitors for anti-cytokine therapies, their mode of action has not been analyzed. We developed a fusion protein consisting of the ligand binding domains of the IL-6 receptor subunits IL-6Ralpha and gp130 that acts as a highly potent IL-6 inhibitor. Gp130 is a shared cytokine receptor also used by the IL-6-related cytokines oncostatin M and leukemia inhibitory factor. In this study, we have shown that the IL-6 receptor fusion protein (IL-6-RFP) is a specific IL-6 inhibitor that does not block oncostatin M or leukemia inhibitory factor. We characterized the complex of IL-6-RFP and fluorescently labeled IL-6 (YFPIL-6) by blue native PAGE and gel filtration. A 2-fold molar excess of IL-6-RFP over IL-6 was sufficient to entirely bind IL-6 in a complex with IL-6-RFP. As shown by treatment with urea and binding competition experiments, the complex of IL-6 and IL-6-RFP is more stable than the complex of IL-6, soluble IL-6Ralpha, and soluble gp130. By live cell imaging, we have demonstrated that YFP-IL-6 bound to the surface of cells expressing gp130-CFP is removed from the plasma membrane upon the addition of IL-6-RFP. The apparent molecular mass of the IL-6.IL-6-RFP complex determined by blue native PAGE and gel filtration suggests that IL-6 is trapped in a structure analogous to the native hexameric IL-6 receptor complex. Thus, fusion of the ligand binding domains of heteromeric receptors leads to highly specific cytokine inhibitors with superior activity compared with the separate soluble receptors.


Subject(s)
Cytokine Receptor gp130/genetics , Drug Design , Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/genetics , Recombinant Fusion Proteins/genetics , Acute-Phase Reaction/genetics , Animals , Antineoplastic Agents/pharmacology , Bacterial Proteins/genetics , Baculoviridae/genetics , COS Cells , Carcinoma, Hepatocellular , Cell Line, Tumor , Chlorocebus aethiops , Cytokine Receptor gp130/chemistry , Cytokine Receptor gp130/metabolism , Gene Expression/immunology , Humans , Interleukin-6/metabolism , Interleukin-6/pharmacology , Leukemia Inhibitory Factor/pharmacology , Liver Neoplasms , Luminescent Proteins/genetics , Oncostatin M/pharmacology , Protein Structure, Tertiary , Receptors, Interleukin-6/chemistry , Receptors, Interleukin-6/metabolism , Recombinant Fusion Proteins/metabolism , STAT3 Transcription Factor/metabolism
20.
J Allergy Clin Immunol ; 118(4): 930-7, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17030248

ABSTRACT

BACKGROUND: IL-31 is produced by activated T lymphocytes, preferentially by TH2 cells. Transgenic mice overexpressing IL-31 have a phenotype resembling allergic dermatitis in human subjects. OBJECTIVE: We sought to evaluate the potential importance of IL-31 in the pathogenesis of human T cell-mediated skin diseases. METHODS: We analyzed total RNA taken from 149 skin biopsy specimens from patients with atopic dermatitis (AD), allergic contact dermatitis (ACD), or psoriasis in comparison with specimens taken from patients with healthy skin (n = 13) by using quantitative real-time PCR for the expression of TH1/TH2 cytokines. RESULTS: We found statistically increased mRNA levels of IL-31 in biopsy specimens taken from patients with AD, irrespective of the severity of the disease and serum IgE levels. Moreover, IL-31 mRNA levels were strongly increased in many biopsy specimens taken from patients with ACD. However, no increased transcription of IL-31 could be detected in biopsy specimens taken from psoriatic plaques. A comparison of mRNA levels of IL-31 with TH1 or TH2 cytokines demonstrates a correlation of the expression of IL-31 with IL-4 and IL-13 but not with IFN-gamma. No significant increase of IL-31 receptor mRNA could be detected in any disease, whereas the second receptor subunit of IL-31, the oncostatin M receptor, seems to be enhanced transcribed in patients with psoriasis. CONCLUSION: IL-31 expression is not only increased in patients with AD but also in those with ACD, 2 pruritic skin disorders. In both types of eczema, expression of IL-31 is associated with the expression of the TH2 cytokines IL-4 and IL-13. CLINICAL IMPLICATIONS: IL-31 might contribute not only to the development of AD but also to ACD-provoked skin inflammation.


Subject(s)
Dermatitis, Allergic Contact/immunology , Dermatitis, Atopic/immunology , Interleukin-13/biosynthesis , Interleukin-4/biosynthesis , Interleukins/biosynthesis , Female , Gene Expression , Gene Expression Profiling , Humans , Immunoglobulin E/blood , Male , Middle Aged , Psoriasis/immunology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Skin/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL