Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Mol Immunol ; 17(10): 1063-1076, 2020 10.
Article in English | MEDLINE | ID: mdl-31467416

ABSTRACT

Comprehensive immune responses are essential for eliminating pathogens but must be tightly controlled to avoid sustained immune activation and potential tissue damage. The engagement of TLR4, a canonical pattern recognition receptor, has been proposed to trigger inflammatory responses with different magnitudes and durations depending on TLR4 cellular compartmentalization. In the present study, we identify an unexpected role of Lamtor5, a newly identified component of the amino acid-sensing machinery, in modulating TLR4 signaling and controlling inflammation. Specifically, Lamtor5 associated with TLR4 via their LZ/TIR domains and facilitated their colocalization at autolysosomes, preventing lysosomal tethering and the activation of mTORC1 upon LPS stimulation and thereby derepressing TFEB to promote autophagic degradation of TLR4. The loss of Lamtor5 was unable to trigger the TFEB-driven autolysosomal pathway and delay degradation of TLR4, leading to sustained inflammation and hence increased mortality among Lamtor5 haploinsufficient mice during endotoxic shock. Intriguingly, nutrient deprivation, particularly leucine deprivation, blunted inflammatory signaling and conferred protection to endotoxic mice. This effect, however, was largely abrogated upon Lamtor5 deletion. We thus propose a homeostatic function of Lamtor5 that couples pathogenic insults and nutrient availability to optimize the inflammatory response; this function may have implications for TLR4-associated inflammatory and metabolic disorders.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Inflammation/metabolism , Proteolysis , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Toll-Like Receptor 4/metabolism , Amino Acids/deficiency , Animals , Autophagosomes/metabolism , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Humans , Lipopolysaccharides , Lysosomes/metabolism , Mice , Mice, Knockout , Organelle Biogenesis , Protein Binding , RAW 264.7 Cells , Shock, Septic/immunology , Shock, Septic/pathology
2.
Sci Rep ; 7: 46904, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28928451

ABSTRACT

This corrects the article DOI: 10.1038/srep18778.

3.
Sci Rep ; 6: 18778, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26728228

ABSTRACT

The coordination of restraining and priming of antiviral signaling constitute a fundamental aspect of immunological functions. However, we currently know little about the molecular events that can translate the pathogenic cues into the appropriate code for antiviral defense. Our present study reports a specific role of B cell lymphoma (Bcl)6 as a checkpoint in the initiation of the host response to cytosolic RNA viruses. Remarkably, Bcl6 specifically binds to the interferon-regulatory factor (IRF)7 loci and restrains its transcription, thereby functioning as a negative regulator for interferon (IFN)-ß production and antiviral responses. The signal-controlled turnover of the Bcl6, most likely mediated by microRNA-127, coordinates the antiviral response and inflammatory sequelae. Accordingly, de-repression of Bcl6 resulted in a phenotypic conversion of macrophages into highly potent IFN-producing cells and rendered mice more resistant to pathogenic RNA virus infection. The failure to remove the Bcl6 regulator, however, impedes the antiviral signaling and exaggerates viral pneumonia in mice. We thus reveal a novel key molecular checkpoint to orchestrate antiviral innate immunity.


Subject(s)
Disease Resistance , Host-Pathogen Interactions , Interferon Regulatory Factor-7/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Signal Transduction , Animals , Cell Line , Co-Repressor Proteins/metabolism , Disease Resistance/genetics , Disease Resistance/immunology , Gene Expression Regulation , Histone Deacetylases/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Interferon Regulatory Factor-7/metabolism , Interferon Type I/biosynthesis , Membrane Proteins/metabolism , Mice , MicroRNAs/genetics , Models, Biological , Nerve Tissue Proteins/metabolism , Protein Binding , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptors, Cell Surface , Transcription, Genetic , Virus Diseases/metabolism , Virus Diseases/pathology , Virus Diseases/virology
4.
Biochem Biophys Res Commun ; 472(1): 11-8, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26549234

ABSTRACT

MicroRNAs play an important role in regulating the inflammatory response, and are critically involved in the development of inflammatory disorders, including those affecting the lungs. While the microRNA miR-221 is involved in embryonic lung branching morphogenesis and epithelial cell development, its importance in lung inflammation has not been previously explored. In our current study, expression of miR-221 was selectively decreased by exposure to lipopolysaccharides (LPS) both in vitro and in vivo. Enforced expression of miR-221 significantly increased the production of proinflammatory cytokines TNF-α and IL-6, and enhanced the activation of NF-κB and MAPKs upon LPS stimulation. Accordingly, intratracheal stimulation of miR-221 was shown to aggravate endotoxin-induced acute lung injuries and inflammation in mice. Mechanistic studies showed that miR-221 directly targets A20, a master regulator of NF-κB and MAPK signaling, and thus represses inflammatory signaling. Restoration of A20 in macrophages abolished the stimulatory effect of miR-221 on production of proinflammatory cytokines. Together, these results indicate the presence of a novel miRNA-mediated feed-back mechanism that controls inflammation, and suggest involvement of aberrant miR-221 expression in the development of inflammatory lung disorders.


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Animals , Cells, Cultured , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Cytokines/biosynthesis , Inflammation Mediators/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/toxicity , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Signal Transduction/drug effects , Toll-Like Receptors/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3
5.
PLoS One ; 10(5): e0127329, 2015.
Article in English | MEDLINE | ID: mdl-25993287

ABSTRACT

Hepatitis B virus (HBV) infection causes hepatocyte death and liver damage, which may eventually lead to cirrhosis and liver cancer. Hepatitis B virus X protein (HBx) is a key antigen that is critically involved in HBV-associated liver diseases. However, the molecular basis for its pathogenesis, particularly in liver damage, has not been well defined. Herein, we report that HBx was able to enhance the susceptibility of hepatocytes to TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis. Increased sensitivity to TRAIL was associated with HBx-induced upregulation of miR-125a, which, in turn, suppressed the expression of its putative target gene, A20 E3 ligase. Importantly, we demonstrate that the defective expression of A20 impaired the K63-linked polyubiquitination of caspase-8, which reciprocally enhanced the activation of caspase-8, the recruitment of Fas-associated death domain (FADD), and the formation of death-inducing signaling complex (DISC), thereby promoting HBx-mediated apoptotic signaling. Accordingly, antagonizing miR-125a or ectopically expressing A20 in hepatocytes abolished the pro-apoptotic effect of HBx. Conversely, the overexpression of miR-125a or knockdown of A20 mimicked HBx to enhance TRAIL susceptibility in hepatocytes. Thus, we establish, for the first time, a miR-125a/A20-initiated and caspase-8-targeted mechanism by which HBx modulates apoptotic signaling and increases hepatic susceptibility to the damaging agent, which might provide novel insight into HBV-related liver pathology.


Subject(s)
Apoptosis/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Hepatocytes/cytology , Hepatocytes/enzymology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Trans-Activators/metabolism , Caspase 8/metabolism , DNA Methylation/drug effects , DNA Methylation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Hepatocytes/drug effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lysine/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polyubiquitin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic/drug effects , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitination/drug effects , Up-Regulation/drug effects , Viral Regulatory and Accessory Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...