Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Arterioscler Thromb Vasc Biol ; 41(6): e338-e353, 2021 06.
Article in English | MEDLINE | ID: mdl-33792343
2.
Br J Pharmacol ; 175(11): 2063-2076, 2018 06.
Article in English | MEDLINE | ID: mdl-29532457

ABSTRACT

BACKGROUND AND PURPOSE: Previously, we demonstrated that exogenous heat shock protein 27 (HSP27/gene, HSPB1) treatment of human endothelial progenitor cells (EPCs) increases the synthesis and secretion of VEGF, improves EPC-migration/re-endothelialization and decreases neo-intima formation, suggesting a role for HSPB1 in regulating EPC function. We hypothesized that HSPB1 also affects mature endothelial cells (ECs) to alter EC-mediated vasoreactivity in vivo. Our work focused on endothelial NOS (eNOS)/NO-dependent relaxation induced by ACh and the coagulation pathway-activated receptor, proteinase-activated receptor 2 (PAR2). EXPERIMENTAL APPROACH: Aorta rings from male and female wild-type, HSPB1-null and HSPB1 overexpressing (HSPB1o/e) mice were contracted with phenylephrine, and NOS-dependent relaxation responses to ACh and PAR2 agonist, 2-furoyl-LIGRLO-NH2 , were measured without and with L-NAME and ODQ, either alone or in combination to block NO synthesis/action. Tissues from female HSPB1-null mice were treated in vitro with recombinant HSP27 and then used for bioassay as above. Furthermore, oestrogen-specific effects were evaluated using a bioassay of aorta isolated from ovariectomized mice. KEY RESULTS: Relative to males, HSPB1-null female mice exhibited an increased L-NAME-resistant relaxation induced by activation of either PAR2 or muscarinic ACh receptors that was blocked in the concurrent presence of both L-NAME and ODQ. mRNAs (qPCR) for eNOS and ODQ-sensitive guanylyl-cyclase were increased in females versus males. Treatment of isolated aorta tissue with HSPB1 improved tissue responsiveness in the presence of L-NAME. Ovariectomy did not affect NO sensitivity, supporting an oestrogen-independent role for HSPB1. CONCLUSIONS AND IMPLICATIONS: HSPB1 can regulate intact vascular endothelial function to affect NO-mediated vascular relaxation, especially in females.


Subject(s)
Enzyme Inhibitors/pharmacology , HSP27 Heat-Shock Proteins/antagonists & inhibitors , Nitric Oxide Synthase Type III/antagonists & inhibitors , Receptor, PAR-2/antagonists & inhibitors , Receptors, Muscarinic/metabolism , Vasodilation/drug effects , Animals , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Inhibitors/chemistry , Female , HSP27 Heat-Shock Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NG-Nitroarginine Methyl Ester/chemistry , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/metabolism , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Quinoxalines/chemistry , Quinoxalines/pharmacology , Receptor, PAR-2/metabolism , Structure-Activity Relationship
3.
Mol Cell Biol ; 37(11)2017 06 01.
Article in English | MEDLINE | ID: mdl-28265004

ABSTRACT

The mRNA-destabilizing factor tristetraprolin (TTP) binds in a sequence-specific manner to the 3' untranslated regions of many proinflammatory mRNAs and recruits complexes of nucleases to promote rapid mRNA turnover. Mice lacking TTP develop a severe, spontaneous inflammatory syndrome characterized by the overexpression of tumor necrosis factor and other inflammatory mediators. However, TTP also employs the same mechanism to inhibit the expression of the potent anti-inflammatory cytokine interleukin 10 (IL-10). Perturbation of TTP function may therefore have mixed effects on inflammatory responses, either increasing or decreasing the expression of proinflammatory factors via direct or indirect mechanisms. We recently described a knock-in mouse strain in which the substitution of 2 amino acids of the endogenous TTP protein renders it constitutively active as an mRNA-destabilizing factor. Here we investigate the impact on the IL-10-mediated anti-inflammatory response. It is shown that the gain-of-function mutation of TTP impairs IL-10-mediated negative feedback control of macrophage function in vitro However, the in vivo effects of TTP mutation are uniformly anti-inflammatory despite the decreased expression of IL-10.


Subject(s)
Feedback, Physiological , Inflammation/pathology , Macrophages/metabolism , Macrophages/pathology , Mutation/genetics , Tristetraprolin/genetics , Animals , Bone Marrow Cells/metabolism , Cytokines/metabolism , Dual Specificity Phosphatase 1/deficiency , Dual Specificity Phosphatase 1/metabolism , Gene Expression Profiling , Inflammation/genetics , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Transcription, Genetic
4.
Biochem Soc Trans ; 44(5): 1321-1337, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27911715

ABSTRACT

Twenty years ago, the first description of a tristetraprolin (TTP) knockout mouse highlighted the fundamental role of TTP in the restraint of inflammation. Since then, work from several groups has generated a detailed picture of the expression and function of TTP. It is a sequence-specific RNA-binding protein that orchestrates the deadenylation and degradation of several mRNAs encoding inflammatory mediators. It is very extensively post-translationally modified, with more than 30 phosphorylations that are supported by at least two independent lines of evidence. The phosphorylation of two particular residues, serines 52 and 178 of mouse TTP (serines 60 and 186 of the human orthologue), has profound effects on the expression, function and localisation of TTP. Here, we discuss the control of TTP biology via its phosphorylation and dephosphorylation, with a particular focus on recent advances and on questions that remain unanswered.


Subject(s)
Inflammation/metabolism , Phosphoric Monoester Hydrolases/metabolism , Serine/metabolism , Tristetraprolin/metabolism , Amino Acid Sequence , Animals , Humans , Inflammation/genetics , Mice, Knockout , Phosphorylation , Serine/genetics , Signal Transduction/genetics , Tristetraprolin/genetics
5.
Proc Natl Acad Sci U S A ; 113(16): 4350-5, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27044108

ABSTRACT

Neutrophils are the first and most numerous cells to arrive at the site of an inflammatory insult and are among the first to die. We previously reported that alpha defensins, released from apoptotic human neutrophils, augmented the antimicrobial capacity of macrophages while also inhibiting the biosynthesis of proinflammatory cytokines. In vivo, alpha defensin administration protected mice from inflammation, induced by thioglychollate-induced peritonitis or following infection withSalmonella entericaserovar Typhimurium. We have now dissected the antiinflammatory mechanism of action of the most abundant neutrophil alpha defensin, Human Neutrophil Peptide 1 (HNP1). Herein we show that HNP1 enters macrophages and inhibits protein translation without inducing the unfolded-protein response or affecting mRNA stability. In a cell-free in vitro translation system, HNP1 powerfully inhibited both cap-dependent and cap-independent mRNA translation while maintaining mRNA polysomal association. This is, to our knowledge, the first demonstration of a peptide released from one cell type (neutrophils) directly regulating mRNA translation in another (macrophages). By preventing protein translation, HNP1 functions as a "molecular brake" on macrophage-driven inflammation, ensuring both pathogen clearance and the resolution of inflammation with minimal bystander tissue damage.


Subject(s)
Macrophages/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Salmonella Infections/metabolism , Salmonella typhimurium , alpha-Defensins/metabolism , Animals , Humans , Macrophages/pathology , Mice , Salmonella Infections/pathology , alpha-Defensins/pharmacology
6.
J Immunol ; 195(1): 277-88, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26019272

ABSTRACT

Dual-specificity phosphatase (DUSP) 1 dephosphorylates and inactivates members of the MAPK superfamily, in particular, JNKs, p38α, and p38ß MAPKs. It functions as an essential negative regulator of innate immune responses, hence disruption of the Dusp1 gene renders mice extremely sensitive to a wide variety of experimental inflammatory challenges. The principal mechanisms behind the overexpression of inflammatory mediators by Dusp1(-/-) cells are not known. In this study, we use a genetic approach to identify an important mechanism of action of DUSP1, involving the modulation of the activity of the mRNA-destabilizing protein tristetraprolin. This mechanism is key to the control of essential early mediators of inflammation, TNF, CXCL1, and CXCL2, as well as the anti-inflammatory cytokine IL-10. The same mechanism also contributes to the regulation of a large number of transcripts induced by treatment of macrophages with LPS. These findings demonstrate that modulation of the phosphorylation status of tristetraprolin is an important physiological mechanism by which innate immune responses can be controlled.


Subject(s)
Dual Specificity Phosphatase 1/immunology , Lipopolysaccharides/pharmacology , Macrophages/immunology , RNA, Messenger/immunology , Tristetraprolin/immunology , Animals , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Chemokine CXCL2/genetics , Chemokine CXCL2/immunology , Dual Specificity Phosphatase 1/genetics , Gene Expression Regulation , Immunity, Innate , Interleukin-10/genetics , Interleukin-10/immunology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 11/immunology , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/immunology , Phosphorylation , Primary Cell Culture , RNA Stability , RNA, Messenger/genetics , Signal Transduction , Tristetraprolin/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
7.
J Immunol ; 195(1): 265-76, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26002976

ABSTRACT

In myeloid cells, the mRNA-destabilizing protein tristetraprolin (TTP) is induced and extensively phosphorylated in response to LPS. To investigate the role of two specific phosphorylations, at serines 52 and 178, we created a mouse strain in which those residues were replaced by nonphosphorylatable alanine residues. The mutant form of TTP was constitutively degraded by the proteasome and therefore expressed at low levels, yet it functioned as a potent mRNA destabilizing factor and inhibitor of the expression of many inflammatory mediators. Mice expressing only the mutant form of TTP were healthy and fertile, and their systemic inflammatory responses to LPS were strongly attenuated. Adaptive immune responses and protection against infection by Salmonella typhimurium were spared. A single allele encoding the mutant form of TTP was sufficient for enhanced mRNA degradation and underexpression of inflammatory mediators. Therefore, the equilibrium between unphosphorylated and phosphorylated TTP is a critical determinant of the inflammatory response, and manipulation of this equilibrium may be a means of treating inflammatory pathologies.


Subject(s)
Macrophages/immunology , Mutation , RNA, Messenger/immunology , Salmonella Infections, Animal/immunology , Tristetraprolin/immunology , Alanine/genetics , Alanine/metabolism , Amino Acid Substitution , Animals , Cell Line , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/immunology , Female , Gene Expression , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Phosphoproteins/genetics , Phosphoproteins/immunology , Phosphorylation , Primary Cell Culture , RNA Stability , RNA, Messenger/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Salmonella Infections, Animal/genetics , Salmonella Infections, Animal/pathology , Salmonella typhimurium/immunology , Serine/genetics , Serine/metabolism , Tristetraprolin/genetics
8.
Blood ; 124(24): 3646-55, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25293769

ABSTRACT

Tissue factor (TF) (CD142) is a 47 kDa transmembrane cell surface glycoprotein that triggers the extrinsic coagulation cascade and links thrombosis with inflammation. Although macrophage TF expression is known to be regulated at the RNA level, very little is known about the mechanisms involved. Poly(adenosine 5'-diphosphate [ADP]-ribose)-polymerase (PARP)-14 belongs to a family of intracellular proteins that generate ADP-ribose posttranslational adducts. Functional screening of PARP-14-deficient macrophages mice revealed that PARP-14 deficiency leads to increased TF expression and functional activity in macrophages after challenge with bacterial lipopolysaccharide. This was related to an increase in TF messenger RNA (mRNA) stability. Ribonucleoprotein complex immunoprecipitation and biotinylated RNA pull-down assays demonstrated that PARP-14 forms a complex with the mRNA-destabilizing protein tristetraprolin (TTP) and a conserved adenylate-uridylate-rich element in the TF mRNA 3' untranslated region. TF mRNA regulation by PARP-14 was selective, as tumor necrosis factor (TNF)α mRNA, which is also regulated by TTP, was not altered in PARP-14 deficient macrophages. Consistent with the in vitro data, TF expression and TF activity, but not TNFα expression, were increased in Parp14(-/-) mice in vivo. Our study provides a novel mechanism for the posttranscriptional regulation of TF expression, indicating that this is selectively regulated by PARP-14.


Subject(s)
Gene Expression Regulation , Macrophages/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Thromboplastin/biosynthesis , Tristetraprolin/metabolism , 3' Untranslated Regions/physiology , Animals , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics , RNA Stability/drug effects , RNA Stability/physiology , Thromboplastin/genetics , Tristetraprolin/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
9.
Arthritis Rheumatol ; 66(10): 2728-38, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24965881

ABSTRACT

OBJECTIVE: Tumor necrosis factor (TNF) signals via 2 receptors, TNFR type I (TNFRI) and TNFRII, with distinct cellular distribution and signaling functions. In rheumatoid arthritis (RA), the net effect of TNFR signaling favors inflammatory responses while inhibiting the activity of regulatory T cells. TNFRII signaling has been shown to promote Treg cell function. To assess the relative contributions of TNFRI and TNFRII signaling to inflammatory and regulatory responses in vivo, we compared the effect of TNF blockade, hence TNFRI/II, versus TNFRI alone in collagen-induced arthritis (CIA) as a model of RA. METHODS: Mice with established arthritis were treated for 10 days with anti-mouse TNFRI domain antibody (dAb; DMS5540), an isotype control dAb (DMS5538), or murine TNFRII genetically fused with mouse IgG1 Fc domain (mTNFRII-Fc) beginning on the day of arthritis onset, and disease progression was monitored. Systemic cytokine concentrations and numbers of T cell subsets in lymph nodes and spleens were measured, and intrinsic Treg cell function was determined by ex vivo suppression assays. RESULTS: Progression of CIA was suppressed similarly by TNFRI (DMS5540) and TNFRI/II (mTNFRII-Fc) blockade. However, blockade of TNFRI/II led to increased effector T cell activity, which was not observed after selective TNFRI blockade, suggesting an immunoregulatory role of TNFRII. In support of this, TNFRI blockade, but not TNFRI/II blockade, expanded and activated Treg cells. Furthermore, a dramatic increase in expression of the Treg cell signature genes FoxP3 and TNFRII was observed in joints undergoing remission, which supports the notion that these molecules have a physiologic role in the resolution of inflammation. CONCLUSION: We propose that a therapeutic strategy that targets TNFRI while sparing TNFRII has the potential to both inhibit inflammation and promote Treg cell activity, which might be superior to TNF blockade.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , Single-Domain Antibodies/therapeutic use , T-Lymphocytes, Regulatory/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/immunology , Cell Proliferation/drug effects , Forkhead Transcription Factors/metabolism , Inflammation/drug therapy , Inflammation/immunology , Male , Mice , Mice, Inbred DBA , Recombinant Fusion Proteins/pharmacology , Single-Domain Antibodies/pharmacology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/immunology
10.
Cell Signal ; 26(4): 683-90, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24378531

ABSTRACT

Tumour necrosis factor (p55 or p60) receptor (TNFR) 1 is the major receptor that activates pro-inflammatory signalling and induces gene expression in response to TNF. Consensus is lacking for the function of (p75 or p80) TNFR2 but experiments in mice have suggested neuro-, cardio- and osteo-protective and anti-inflammatory roles. It has been shown in various cell types to be specifically required for the induction of TNFR-associated factor-2 (TRAF2) degradation and activation of the alternative nuclear factor (NF)-kappaB pathway, and to contribute to the activation of mitogen-activated protein kinases (MAPK) and the classical NF-kappaB pathway. We have investigated the signalling functions of TNFR2 in primary human and murine macrophages. We find that in these cells TNF induces TRAF2 degradation, and this is blocked in TNFR2(-/-) macrophages. TRAF2 has been previously reported to be required for TNF-induced activation of p38 MAPK. However, TRAF2 degradation does not inhibit TNF-induced tolerance of p38 MAPK activation. Neither TNF, nor lipopolysaccharide treatment, induced activation of the alternative NF-kappaB pathway in macrophages. Activation by TNF of the p38 MAPK and NF-kappaB pathways was blocked in TNFR1(-/-) macrophages. In contrast, although TNFR2(-/-) macrophages displayed robust p38 MAPK activation and IkappaBα degradation at high concentrations of TNF, at lower doses the concentration dependence of signalling was weakened by an order of magnitude. Our results suggest that, in addition to inducing TRAF2 protein degradation, TNFR2 also plays a crucial auxiliary role to TNFR1 in sensitising macrophages for the ligand-induced activation of the p38 MAPK and classical NF-kappaB pro-inflammatory signalling pathways.


Subject(s)
Macrophages/metabolism , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , TNF Receptor-Associated Factor 2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Enzyme Activation/drug effects , Humans , I-kappa B Proteins/metabolism , Ligands , Lipopolysaccharides/pharmacology , Macrophages/cytology , Mice , NF-KappaB Inhibitor alpha , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
11.
PLoS One ; 8(10): e77383, 2013.
Article in English | MEDLINE | ID: mdl-24143227

ABSTRACT

There is large literature describing in vitro experiments on heat shock protein (hsp)B1 but understanding of its function in vivo is limited to studies in mice overexpressing human hspB1 protein. Experiments in cells have shown that hspB1 has chaperone activity, a cytoprotective role, regulates inflammatory gene expression, and drives cell proliferation. To investigate the function of the protein in vivo we generated hspB1-deficient mice. HspB1-deficient fibroblasts display increased expression of the pro-inflammatory cytokine, interleukin-6, compared to wild-type cells, but reduced proliferation. HspB1-deficient fibroblasts exhibit reduced entry into S phase and increased expression of cyclin-dependent kinase inhibitors p27(kip1) and p21(waf1). The expression of hspB1 protein and mRNA is also controlled by the cell cycle. To investigate the physiological function of hspB1 in regulating inflammation and cell proliferation we used an excisional cutaneous wound healing model. There was a significant impairment in the rate of healing of wounds in hspB1-deficient mice, characterised by reduced re-epithelialisation and collagen deposition but also increased inflammation. HspB1 deficiency augments neutrophil infiltration in wounds, driven by increased chemokine (C-X-C motif) ligand 1 expression. This appears to be a general mechanism as similar results were obtained in the air-pouch and peritonitis models of acute inflammation.


Subject(s)
HSP27 Heat-Shock Proteins/deficiency , Wound Healing , Animals , Cell Cycle/drug effects , Cell Proliferation , Collagen/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Exons/genetics , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , HSP27 Heat-Shock Proteins/genetics , Interleukin-1/pharmacology , Interleukin-6/metabolism , Mice , Peritonitis/chemically induced , Peritonitis/pathology , Peritonitis/physiopathology , Skin/cytology , Skin/injuries , Skin/metabolism , Zymosan/adverse effects
12.
Mol Cell Biol ; 33(21): 4152-65, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23979601

ABSTRACT

The stimulation of Toll-like receptors (TLRs) on macrophages by pathogen-associated molecular patterns (PAMPs) results in the activation of intracellular signaling pathways that are required for initiating a host immune response. Both phosphatidylinositol 3-kinase (PI3K)-Akt and p38 mitogen-activated protein kinase (MAPK) signaling pathways are activated rapidly in response to TLR activation and are required to coordinate effective host responses to pathogen invasion. In this study, we analyzed the role of the p38-dependent kinases MK2/3 in the activation of Akt and show that lipopolysaccharide (LPS)-induced phosphorylation of Akt on Thr308 and Ser473 requires p38α and MK2/3. In cells treated with p38 inhibitors or an MK2/3 inhibitor, phosphorylation of Akt on Ser473 and Thr308 is reduced and Akt activity is inhibited. Furthermore, BMDMs deficient in MK2/3 display greatly reduced phosphorylation of Ser473 and Thr308 following TLR stimulation. However, MK2/3 do not directly phosphorylate Akt in macrophages but act upstream of PDK1 and mTORC2 to regulate Akt phosphorylation. Akt is recruited to phosphatidylinositol 3,4,5-trisphosphate (PIP3) in the membrane, where it is activated by PDK1 and mTORC2. Analysis of lipid levels in MK2/3-deficient bone marrow-derived macrophages (BMDMs) revealed a role for MK2/3 in regulating Akt activity by affecting availability of PIP3 at the membrane. These data describe a novel role for p38α-MK2/3 in regulating TLR-induced Akt activation in macrophages.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Toll-Like Receptors/metabolism , Animals , Cell Line , Enzyme Activation , Heat-Shock Proteins/metabolism , Imidazoles/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Molecular Chaperones , Neoplasm Proteins/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Protein Processing, Post-Translational , Pyridazines/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptor Cross-Talk , Signal Transduction , Toll-Like Receptors/agonists
13.
Circ Res ; 112(12): 1583-91, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23564640

ABSTRACT

RATIONALE: Hypoxia followed by reoxygenation promotes inflammation by activating nuclear factor κB transcription factors in endothelial cells (ECs). This process involves modification of the signaling intermediary tumor necrosis factor receptor-associated factor 6 with polyubiquitin chains. Thus, cellular mechanisms that suppress tumor necrosis factor receptor-associated factor 6 ubiquitination are potential therapeutic targets to reduce inflammation in hypoxic tissues. OBJECTIVE: In this study, we tested the hypothesis that endothelial activation in response to hypoxia-reoxygenation can be influenced by Cezanne, a deubiquitinating enzyme that cleaves ubiquitin from specific modified proteins. METHODS AND RESULTS: Studies of cultured ECs demonstrated that hypoxia (1% oxygen) induced Cezanne via p38 mitogen-activated protein kinase-dependent transcriptional and post-transcriptional mechanisms. Hypoxia-reoxygenation had minimal effects on proinflammatory signaling in unmanipulated ECs but significantly enhanced Lys63 polyubiquitination of tumor necrosis factor receptor-associated factor 6, activation of nuclear factor κB, and expression of inflammatory genes after silencing of Cezanne. Thus, although hypoxia primed cells for inflammatory activation, it simultaneously induced Cezanne, which impeded signaling to nuclear factor κB by suppressing tumor necrosis factor receptor-associated factor 6 ubiquitination. Similarly, ischemia induced Cezanne in the murine kidney in vascular ECs, glomerular ECs, podocytes, and epithelial cells, and genetic deletion of Cezanne enhanced renal inflammation and injury in murine kidneys exposed to ischemia followed by reperfusion. CONCLUSIONS: We conclude that inflammatory responses to ischemia are controlled by a balance between ubiquitination and deubiquitination, and that Cezanne is a key regulator of this process. Our observations have important implications for therapeutic targeting of inflammation and injury during ischemia-reperfusion.


Subject(s)
Endopeptidases/metabolism , Endothelial Cells/enzymology , Inflammation/prevention & control , Kidney/blood supply , Reperfusion Injury/enzymology , TNF Receptor-Associated Factor 6/metabolism , Animals , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Endopeptidases/deficiency , Endopeptidases/genetics , Endothelial Cells/immunology , Humans , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Oxygen/metabolism , RNA Interference , Rats , Rats, Inbred F344 , Reperfusion Injury/genetics , Reperfusion Injury/immunology , Signal Transduction , TNF Receptor-Associated Factor 6/genetics , Time Factors , Transcription, Genetic , Transfection , Ubiquitination , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Mol Cell Biol ; 33(7): 1456-67, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23382072

ABSTRACT

Prostaglandin production is catalyzed by cyclooxygenase 2 (cox-2). We demonstrate here that MSK1 and MSK2 (MSK1/2) can exert control on the induction of cox-2 mRNA by Toll-like receptor (TLR) agonists. In the initial phase of cox-2 induction, MSK1/2 knockout macrophages confirmed a role for MSK in the positive regulation of transcription. However, at later time points both lipopolysaccharide (LPS)-induced prostaglandin and cox-2 protein levels were increased in MSK1/2 knockout. Further analysis found that while MSKs promoted cox-2 mRNA transcription, following longer LPS stimulation MSKs also promoted degradation of cox-2 mRNA. This was found to be the result of an interleukin 10 (IL-10) feedback mechanism, with endogenously produced IL-10 promoting cox-2 degradation. The ability of IL-10 to do this was dependent on the mRNA binding protein TTP through a p38/MK2-mediated mechanism. As MSKs regulate IL-10 production in response to LPS, MSK1/2 knockout results in reduced IL-10 secretion and therefore reduced feedback from IL-10 on cox-2 mRNA stability. Following LPS stimulation, this increased mRNA stability correlated to an elevated induction of both of cox-2 protein and prostaglandin secretion in MSK1/2 knockout macrophages relative to that in wild-type cells. This was not restricted to isolated macrophages, as a similar effect of MSK1/2 knockout was seen on plasma prostaglandin E2 (PGE2) levels following intraperitoneal injection of LPS.


Subject(s)
Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Prostaglandins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Interleukin-10/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages , Mice , Mice, Inbred C57BL , Prostaglandins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteolysis , RNA Stability , RNA, Messenger/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Toll-Like Receptors/agonists , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
J Biol Chem ; 285(36): 27590-600, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20595389

ABSTRACT

Tristetraprolin (TTP) directs its target AU-rich element (ARE)-containing mRNAs for degradation by promoting removal of the poly(A) tail. The p38 MAPK pathway regulates mRNA stability via the downstream kinase MAPK-activated protein kinase 2 (MAPKAP kinase 2 or MK2), which phosphorylates and prevents the mRNA-destabilizing function of TTP. We show that deadenylation of endogenous ARE-containing tumor necrosis factor mRNA is inhibited by p38 MAPK. To investigate whether phosphorylation of TTP by MK2 regulates TTP-directed deadenylation of ARE-containing mRNAs, we used a cell-free assay that reconstitutes the mechanism in vitro. We find that phosphorylation of Ser-52 and Ser-178 of TTP by MK2 results in inhibition of TTP-directed deadenylation of ARE-containing RNA. The use of 14-3-3 protein antagonists showed that regulation of TTP-directed deadenylation by MK2 is independent of 14-3-3 binding to TTP. To investigate the mechanism whereby TTP promotes deadenylation, it was necessary to identify the deadenylases involved. The carbon catabolite repressor protein (CCR)4.CCR4-associated factor (CAF)1 complex was identified as the major source of deadenylase activity in HeLa cells responsible for TTP-directed deadenylation. CAF1a and CAF1b were found to interact with TTP in an RNA-independent fashion. We find that MK2 phosphorylation reduces the ability of TTP to promote deadenylation by inhibiting the recruitment of CAF1 deadenylase in a mechanism that does not involve sequestration of TTP by 14-3-3. Cyclooxygenase-2 mRNA stability is increased in CAF1-depleted cells in which it is no longer p38 MAPK/MK2-regulated.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribonucleases/metabolism , Transcription Factors/metabolism , Tristetraprolin/metabolism , 14-3-3 Proteins/metabolism , Animals , Base Composition , Cyclooxygenase 2/genetics , HeLa Cells , Humans , MAP Kinase Signaling System , Mice , Phosphorylation , RNA Stability , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Receptors, CCR4/metabolism , Tumor Necrosis Factor-alpha/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
17.
FEBS Lett ; 583(12): 1933-8, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19416727

ABSTRACT

p38 mitogen-activated protein kinase (MAPK) stabilises pro-inflammatory mediator mRNAs by inhibiting AU-rich element (ARE)-mediated decay. We show that in bone-marrow derived murine macrophages tristetraprolin (TTP) is necessary for the p38 MAPK-sensitive decay of several pro-inflammatory mRNAs, including cyclooxygenase-2 and the novel targets interleukin (IL)-6, and IL-1alpha. TTP(-/-) macrophages also strongly overexpress IL-10, an anti-inflammatory cytokine that constrains the production of the IL-6 despite its disregulation at the post-transcriptional level. TTP directly controls IL-10 mRNA stability, which is increased and insensitive to inhibition of p38 MAPK in TTP(-/-) macrophages. Furthermore, TTP enhances deadenylation of an IL-10 3'-untranslated region RNA in vitro.


Subject(s)
Inflammation Mediators/metabolism , Interleukin-10/genetics , Macrophages/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tristetraprolin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Base Sequence , In Vitro Techniques , Interleukin-10/antagonists & inhibitors , Interleukin-12 Subunit p40/biosynthesis , Interleukin-6/biosynthesis , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA Stability , Tristetraprolin/deficiency , Tristetraprolin/genetics
18.
J Biol Chem ; 283(11): 7036-45, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18178551

ABSTRACT

Transcription factors belonging to the NF-kappaB family regulate inflammation by inducing pro-inflammatory molecules (e.g. interleukin (IL)-8) in response to cytokines (e.g. tumor necrosis factor (TNF) alpha, IL-1) or other stimuli. Several negative regulators of NF-kappaB, including the ubiquitin-editing enzyme A20, participate in the resolution of inflammatory responses. We report that Cezanne, a member of the A20 family of the deubiquitinating cysteine proteases, can be induced by TNFalpha in cultured cells. Silencing of endogenous Cezanne using small interfering RNA led to elevated NF-kappaB luciferase reporter gene activity and enhanced expression of IL-8 transcripts in TNFalpha-treated cells. Thus we conclude that endogenous Cezanne can attenuate NF-kappaB activation and the induction of pro-inflammatory transcripts in response to TNF receptor (TNFR) signaling. Overexpression studies revealed that Cezanne suppressed NF-kappaB nuclear translocation and transcriptional activity by targeting the TNFR signaling pathway at the level of the IkappaB kinase complex or upstream from it. These effects were not observed in a form of Cezanne that was mutated at the catalytic cysteine residue (Cys209), indicating that the deubiquitinating activity of Cezanne is essential for NF-kappaB regulation. Finally, we demonstrate that Cezanne can be recruited to activated TNFRs where it suppresses the build-up of polyubiquitinated RIP1 signal adapter proteins. Thus we conclude that Cezanne forms a novel negative feedback loop in pro-inflammatory signaling and that it suppresses NF-kappaB activation by targeting RIP1 signaling intermediaries for deubiquitination.


Subject(s)
Endopeptidases/metabolism , Gene Expression Regulation, Enzymologic , Inflammation , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Ubiquitin/metabolism , DNA-Binding Proteins , Endothelial Cells/cytology , Humans , Interleukin-8/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lysine/chemistry , Models, Biological , Mutation , Nuclear Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Tumor Necrosis Factor alpha-Induced Protein 3
19.
J Biol Chem ; 282(9): 6232-41, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17202147

ABSTRACT

Heat shock protein (HSP) 27 has long been known to be a component of the p38 mitogen-activated protein kinase (MAPK) signaling pathway. p38 MAPK has important functions in the inflammatory response, but the role of HSP27 in inflammation has remained unknown. We have used small interfering RNAs to suppress HSP27 expression in HeLa cells and fibroblasts and found that it is required for pro-inflammatory cell signaling and the expression of pro-inflammatory genes. HSP27 is needed for the activation by interleukin (IL)-1 of TAK1 and downstream signaling by p38 MAPK, JNK, and their activators (MKK-3, -4, -6, -7) and IKKbeta. IL-1-induced ERK activation appears to be independent of HSP27. HSP27 is required for both IL-1 and TNF-induced signaling pathways for which the most upstream common signaling protein is TAK1. HSP27 is also required for IL-1-induced expression of the pro-inflammatory mediators, cyclooxygenase-2, IL-6, and IL-8. HSP27 functions to drive cyclooxygenase-2 and IL-6 expression by augmenting the activation of the kinase downstream of p38 MAPK, MK2, resulting in stabilization of cyclooxygenase-2 and IL-6 mRNAs. The mechanism may not occur in cells of myeloid lineage because HSP27 protein was undetectable in human monocytes and murine macrophages.


Subject(s)
Gene Expression Regulation , Heat-Shock Proteins/physiology , Inflammation/genetics , MAP Kinase Kinase Kinases/metabolism , Neoplasm Proteins/physiology , Signal Transduction , Cells, Cultured , Cyclooxygenase 2/genetics , Fibroblasts , HSP27 Heat-Shock Proteins , HeLa Cells , Humans , Interleukin-1/metabolism , Interleukin-6/genetics , Mitogen-Activated Protein Kinases/metabolism , Molecular Chaperones , RNA Stability , Tumor Necrosis Factor-alpha/metabolism
20.
Biochemistry ; 44(9): 3636-43, 2005 Mar 08.
Article in English | MEDLINE | ID: mdl-15736973

ABSTRACT

A mutant (D165N) of clostridial glutamate dehydrogenase (GDH) in which the catalytic Asp is replaced by Asn surprisingly showed a residual 2% of wild-type activity when purified after expression in Escherichia coli at 37 degrees C. This low-level activity also displayed Michaelis constants for substrates that were remarkably similar to those of the wild-type enzyme. Expression at 8 degrees C gave a mutant enzyme preparation 1000 times less active than the first preparation, but progressively, over 2 weeks' incubation at 37 degrees C in sealed vials, this enzyme regained 90% of the specific activity of wild type. This suggested that the mutant might undergo spontaneous deamidation. Mass spectrometric analysis of tryptic peptides derived from D165N samples treated in various ways showed (i) that the Asn is in place in D165N GDH freshly prepared at 8 degrees C; (ii) that there is a time-dependent reversion of this Asn to Asp over the 2-week incubation period; (iii) that detectable deamidation of other Asn residues, in Asn-Gly sequences, mainly occurred in sample workup rather than during the 2-week incubation; (iv) that there is no significant deamidation of other randomly chosen Asn residues in this mutant over the same period; and (v) that when the protein is denatured before incubation, no deamidation at Asn-165 is detectable. It appears that this deamidation depends on the residual catalytic machinery of the mutated GDH active site. A literature search indicates that this finding is not unique and that Asn may not be a suitable mutational replacement in the assessment of putative catalytic Asp residues by site-directed mutagenesis.


Subject(s)
Amides/metabolism , Asparagine/metabolism , Aspartic Acid/genetics , Catalytic Domain/genetics , Glutamate Dehydrogenase/genetics , Glutamate Dehydrogenase/metabolism , Mutagenesis, Site-Directed , Amino Acid Sequence , Asparagine/genetics , Binding Sites/genetics , Clostridium/enzymology , Clostridium/genetics , Enzyme Stability , Glutamate Dehydrogenase/isolation & purification , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Temperature , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...