Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
iScience ; 23(8): 101433, 2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32823063

ABSTRACT

The anti-inflammatory actions of interleukin-10 (IL10) are thought to be mediated primarily by the STAT3 transcription factor, but pro-inflammatory cytokines such as interleukin-6 (IL6) also act through STAT3. We now report that IL10, but not IL6 signaling, induces formation of a complex between STAT3 and the inositol polyphosphate-5-phosphatase SHIP1 in macrophages. Both SHIP1 and STAT3 translocate to the nucleus in macrophages. Remarkably, sesquiterpenes of the Pelorol family, which we previously described as allosteric activators of SHIP1 phosphatase activity, could induce SHIP1/STAT3 complex formation in cells and mimic the anti-inflammatory action of IL10 in a mouse model of colitis. Using crystallography and docking studies we identified a drug-binding pocket in SHIP1. Our studies reveal new mechanisms of action for both STAT3 and SHIP1 and provide a rationale for use of allosteric SHIP1-activating compounds, which mimic the beneficial anti-inflammatory actions of IL10. VIDEO ABSTRACT.

2.
PLoS One ; 8(8): e71336, 2013.
Article in English | MEDLINE | ID: mdl-23951138

ABSTRACT

The anti-inflammatory cytokine interleukin-10 (IL-10) is essential for attenuating the inflammatory response, which includes reducing the expression of pro-inflammatory microRNA-155 (miR-155) in lipopolysaccharide (LPS) activated macrophages. miR-155 enhances the expression of pro-inflammatory cytokines such as TNFα and suppresses expression of anti-inflammatory molecules such as SOCS1. Therefore, we examined the mechanism by which IL-10 inhibits miR-155. We found that IL-10 treatment did not affect the transcription of the miR-155 host gene nor the nuclear export of pre-miR-155, but rather destabilized both pri-miR-155 and pre-miR-155 transcripts, as well as interfered with the final maturation of miR-155. This inhibitory effect of IL-10 on miR-155 expression involved the contribution of both the STAT3 transcription factor and the phosphoinositol phosphatase SHIP1. This is the first report showing evidence that IL-10 regulates miRNA expression post-transcriptionally.


Subject(s)
Interleukin-10/pharmacology , Macrophages/drug effects , Macrophages/metabolism , MicroRNAs/genetics , RNA Precursors/genetics , RNA Processing, Post-Transcriptional/drug effects , RNA Stability/drug effects , Animals , Biological Transport , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Female , Gene Expression Regulation/drug effects , Inositol Polyphosphate 5-Phosphatases , Lipopolysaccharides/immunology , Macrophages/immunology , Male , Mice , MicroRNAs/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/metabolism , RNA Precursors/metabolism , STAT3 Transcription Factor/metabolism
3.
J Biol Chem ; 287(45): 38020-7, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-22955274

ABSTRACT

Production of the proinflammatory cytokine TNFα by activated macrophages is an important component of host defense. However, TNFα production must be tightly controlled to avoid pathological consequences. The anti-inflammatory cytokine IL-10 inhibits TNFα mRNA expression through activation of the STAT3 transcription factor pathway and subsequent expression of STAT3-dependent gene products. We hypothesized that IL-10 must also have more rapid mechanisms of action and show that IL-10 rapidly shifts existing TNFα mRNA from polyribosome-associated polysomes to monosomes. This translation suppression requires the presence of SHIP1 (SH2 domain-containing inositol 5'-phosphatase 1) and involves inhibition of Mnk1 (MAPK signal-integrating kinase 1). Furthermore, activating SHIP1 using a small-molecule agonist mimics the inhibitory effect of IL-10 on Mnk1 phosphorylation and TNFα translation. Our data support the existence of an alternative STAT3-independent pathway through SHIP1 for IL-10 to regulate TNFα translation during the anti-inflammatory response.


Subject(s)
Interleukin-10/pharmacology , Lipopolysaccharides/pharmacology , Phosphoric Monoester Hydrolases/metabolism , Protein Biosynthesis/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line , Cells, Cultured , Female , Immunoblotting , Inositol Polyphosphate 5-Phosphatases , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Phosphorylation/drug effects , Polyribosomes/genetics , Polyribosomes/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics
4.
FASEB J ; 26(8): 3163-77, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22651931

ABSTRACT

SH2 domain-containing inositol-5'-phosphatase-1 (SHIP1) inhibits inflammation by hydrolyzing phosphoinositide-3'-kinase generated membrane phosphatidylinositol-3,4,5-trisphosphate (PIP(3)). Bioinformatic analysis of SHIP1 from multiple species revealed a pleckstrin homololgy-related (PH-R) domain, which we hypothesize mediates SHIP1's association with the membrane, a requirement for its biological function. Recombinant murine SHIP1 PH-R domain was subjected to biophysical and biochemical analysis. Residues K370 and K397 were found to be important for PH-R domain association with membrane PIP(3). Wild-type PH-R domain bound PIP(3) with 1.9 ± 0.2 nM affinity, while the affinity of a K370A/K397A substituted mutant was too low to measure. Wild-type (but not the K370A/K397A substituted) full-length SHIP1 protein, reconstitutes normal inhibition of Fcγ receptor-mediated phagocytosis when introduced into SHIP1(-/-) murine macrophages, reducing the number of phagocytic events by 2-fold as compared to SHIP1(-/-) cells. In fact, the PH-R-mediated membrane interaction appears to be a major mechanism by which SHIP1 is recruited to the membrane, since the K370A/K397A substitution reduced the recruitment of both full-length SHIP1 and the PH-R domain by ≥2-fold. We have previously shown that SHIP1 enzyme activity can be targeted for therapeutic purposes. The current studies suggest that molecules targeting the PH-R domain can also modulate SHIP1 function.


Subject(s)
Phagocytosis/physiology , Phosphoric Monoester Hydrolases/metabolism , Receptors, IgG/physiology , Allosteric Regulation , Amino Acid Sequence , Inositol Polyphosphate 5-Phosphatases , Nuclear Magnetic Resonance, Biomolecular , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/genetics , Protein Structure, Tertiary
5.
Blood ; 110(6): 1942-9, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17502453

ABSTRACT

Because phosphoinositide 3-kinase (PI3K) plays a central role in cellular activation, proliferation, and survival, pharmacologic inhibitors targeting components of the PI3K pathway are actively being developed as therapeutics for the treatment of inflammatory disorders and cancer. These targeted drugs inhibit the activity of either PI3K itself or downstream protein kinases. However, a previously unexplored, alternate strategy is to activate the negative regulatory phosphatases in this pathway. The SH2-containing inositol-5'-phosphatase SHIP1 is a normal physiologic counter-regulator of PI3K in immune/hematopoietic cells that hydrolyzes the PI3K product phosphatidylinositiol-3,4,5-trisphosphate (PIP(3)). We now describe the identification and characterization of potent and specific small-molecule activators of SHIP1. These compounds represent the first small-molecule activators of a phosphatase, and are able to activate recombinant SHIP1 enzyme in vitro and stimulate SHIP1 activity in intact macrophage and mast cells. Mechanism of activation studies with these compounds suggest that they bind a previously undescribed, allosteric activation domain within SHIP1. Furthermore, in vivo administration of these compounds was protective in mouse models of endotoxemia and acute cutaneous anaphylaxis, suggesting that SHIP1 agonists could be used therapeutically to inhibit the PI3K pathway.


Subject(s)
Anaphylaxis/drug therapy , Endotoxemia/drug therapy , Enzyme Inhibitors/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Polycyclic Compounds/pharmacology , Sesquiterpenes/pharmacology , Allosteric Regulation , Anaphylaxis/metabolism , Anaphylaxis/pathology , Animals , Calcium/metabolism , Cells, Cultured , Endotoxemia/metabolism , Endotoxemia/pathology , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic , Humans , Immunoprecipitation , Inositol Polyphosphate 5-Phosphatases , Kidney/cytology , Kidney/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphorylation/drug effects , Polycyclic Compounds/chemistry , Porifera/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sesquiterpenes/chemistry , Signal Transduction/drug effects , Skin Tests
6.
J Interferon Cytokine Res ; 26(5): 281-90, 2006 May.
Article in English | MEDLINE | ID: mdl-16689656

ABSTRACT

The cytokine interleukin-10 (IL-10) is an important regulator of immune cell function, proliferation, and survival. The IL-10 receptor (IL-10R) consists of two subunits, IL-10R1 and IL-10R2, both belonging to the class II cytokine receptor superfamily. Like other members of the cytokine receptor superfamily, IL-10R stimulation leads to activation of Jak family kinases and Stat transcription factors. To identify additional signal transduction pathways used by the IL-10R, we purified 92-kDa and 100-kDa proteins that coprecipitated with IL-10R1 from IL-10-stimulated cells. Both proteins were found to be related to the 97-kDa subunit of the regulatory component of the 26S proteasome. Subsequent studies confirmed that the IL-10R1 undergoes ligand- dependent internalization and proteasome-mediated degradation. An IL-10R1 cytoplasmic domain mutant deficient for internalization exhibited prolonged signaling through Jak1 and Stat3, reinforcing the importance of receptor internalization for signal termination.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Receptors, Interleukin-10/metabolism , Signal Transduction , Animals , Cell Line , Down-Regulation/drug effects , Humans , Interleukin-10/pharmacology , Kinetics , Ligands , Mice , Molecular Weight , Mutation/genetics , Protein Binding , Receptors, Interleukin-10/genetics , Signal Transduction/drug effects
7.
J Biol Chem ; 281(10): 6316-24, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16352613

ABSTRACT

The cytokine interleukin-10 (IL-10) potently inhibits macrophage function through activation of the transcription factor STAT3. The expression of SOCS3 (suppressor of cytokine signaling-3) has been shown to be induced by IL-10 in a STAT3-dependent manner. However, the relevance of SOCS3 expression to the anti-inflammatory effect of IL-10 on macrophages has been controversial. Through kinetic analysis of the requirement for SOCS3 in IL-10 inhibition of lipopolysaccharide (LPS)-stimulated tumor necrosis factor-alpha (TNFalpha) transcription and translation, SOCS3 was found to be necessary for TNFalpha expression during the early phase, but not the late phase of IL-10 action. SOCS3 was essential for IL-10 inhibition of LPS-stimulated production of iNOS (inducible nitric-oxide synthase) protein and nitric oxide (NO). To determine the domains of SOCS3 protein important in mediating these effects, SOCS3-/- macrophages were reconstituted with SOCS3 mutated for the SH2, KIR, SOCS box domains, and tyrosines 204 (Tyr204) and 221 (Tyr221). The SH2 domain, SOCS box, and both Tyr204 and Tyr221 were required for IL-10 inhibition of TNFalpha mRNA and protein expression, but interestingly the KIR domain was necessary only for IL-10 inhibition of TNFalpha protein expression. In contrast, Tyr204 and Tyr221 were the only structural features of SOCS3 that were necessary in mediating IL-10 inhibition of iNOS protein expression and NO production. These data define SOCS3 as an important mediator of IL-10 inhibition of macrophage activation and that SOCS3 interferes with distinct LPS-stimulated signal transduction events through differing mechanisms.


Subject(s)
Interleukin-10/physiology , Macrophages/physiology , Nitric Oxide/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Cell Line , Lipopolysaccharides/pharmacology , Macrophage Activation/genetics , Macrophages/immunology , Mice , Mutation , Nitric Oxide/biosynthesis , Phosphorylation , RNA, Messenger/metabolism , STAT3 Transcription Factor/physiology , Signal Transduction/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/genetics , Tumor Necrosis Factor-alpha/physiology , Tyrosine/chemistry , Tyrosine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL