Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
FEBS J ; 290(15): 3802-3811, 2023 08.
Article in English | MEDLINE | ID: mdl-36938956

ABSTRACT

Dysregulated cytokine signalling is a hallmark of inflammatory bowel diseases. Inflammatory responses of the colon are regulated by the suppressor of cytokine signalling (SOCS) proteins. SOCS1 is a key member of this family, and its function is critical in maintaining an appropriate inflammatory response through the JAK/STAT signalling pathway. Dysregulation of SOCS1 protein has been identified as a causal element in colonic inflammatory diseases. Despite this, it remains unclear how SOCS1 protein is regulated. Here, we identify that SOCS1 protein is targeted for degradation by the ubiquitin proteasome system, mediated by the E3 ubiquitin ligase KIAA0317 during experimental colonic inflammation. We characterize the mechanism of protein-protein interaction and ubiquitin conjugation to SOCS1 and demonstrate that the modulation of SOCS1 protein level leads to stark effects on JAK/STAT inflammatory signalling. Together, these results provide insight into the regulation of colonic inflammation through a new mechanism of ubiquitin-based control of SOCS1 protein.


Subject(s)
Suppressor of Cytokine Signaling Proteins , Ubiquitin , Humans , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Inflammation/genetics , Cytokines/metabolism , Colon/metabolism
2.
JCI Insight ; 4(19)2019 10 03.
Article in English | MEDLINE | ID: mdl-31578312

ABSTRACT

Dysregulated proinflammatory cytokine release has been implicated in the pathogenesis of several life-threatening acute lung illnesses such as pneumonia, sepsis, and acute respiratory distress syndrome. Suppressors of cytokine signaling proteins, particularly SOCS2, have recently been described as antiinflammatory mediators. However, the regulation of SOCS2 protein has not been described. Here we describe a mechanism of SOCS2 regulation by the action of the ubiquitin E3 ligase KIAA0317. KIAA0317-mediated degradation of SOCS2 exacerbated inflammation in vitro, and depletion of KIAA0317 in vivo ameliorated pulmonary inflammation. KIAA0317-knockout mice exhibited resistance to LPS-induced pulmonary inflammation, while KIAA03017 reexpression mitigated this effect. We uncovered a small molecule inhibitor of KIAA0317 protein (BC-1365) that prevented SOCS2 degradation and attenuated LPS- and P. aeruginosa-induced lung inflammation in vivo. These studies show KIAA0317 to be a critical mediator of pulmonary inflammation through its degradation of SOCS2 and a potential candidate target for therapeutic inhibition.


Subject(s)
Lung/metabolism , Pneumonia/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Adult , Aged , Animals , Cytokines/metabolism , Female , Humans , Immunity, Innate , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Molecular Docking Simulation , Pneumonia/immunology , Pneumonia/pathology , Protein Binding , Transcriptome
3.
J Exp Med ; 213(6): 1029-46, 2016 05 30.
Article in English | MEDLINE | ID: mdl-27162139

ABSTRACT

The E3 small ubiquitin-like modifier (SUMO) protein ligase protein inhibitor of activated STAT 4 (PIAS4) is a pivotal protein in regulating the TGFß pathway. In this study, we discovered a new protein isoform encoded by KIAA0317, termed fibrosis-inducing E3 ligase 1 (FIEL1), which potently stimulates the TGFß signaling pathway through the site-specific ubiquitination of PIAS4. FIEL1 targets PIAS4 using a double locking mechanism that is facilitated by the kinases PKCζ and GSK3ß. Specifically, PKCζ phosphorylation of PIAS4 and GSK3ß phosphorylation of FIEL1 are both essential for the degradation of PIAS4. FIEL1 protein is highly expressed in lung tissues from patients with idiopathic pulmonary fibrosis (IPF), whereas PIAS4 protein levels are significantly reduced. FIEL1 overexpression significantly increases fibrosis in a bleomycin murine model, whereas FIEL1 knockdown attenuates fibrotic conditions. Further, we developed a first-in-class small molecule inhibitor toward FIEL1 that is highly effective in ameliorating fibrosis in mice. This study provides a basis for IPF therapeutic intervention by modulating PIAS4 protein abundance.


Subject(s)
Lung/metabolism , Protein Inhibitors of Activated STAT/metabolism , Pulmonary Fibrosis/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Bleomycin/adverse effects , Bleomycin/pharmacology , Cell Line , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Lung/pathology , Mice , Poly-ADP-Ribose Binding Proteins , Protein Inhibitors of Activated STAT/genetics , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C-theta , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Ubiquitin-Protein Ligases/genetics
4.
Sci Transl Med ; 7(295): 295ra109, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26157031

ABSTRACT

Invading pathogens may trigger overactivation of the innate immune system, which results in the release of large amounts of proinflammatory cytokines (cytokine storm) and leads to the development of pulmonary edema, multiorgan failure, and shock. PIAS1 is a multifunctional and potent anti-inflammatory protein that negatively regulates several key inflammatory pathways such as Janus kinase (JAK)-signal transducer and activator of transcription (STAT) and nuclear factor κB (NF-κB). We discovered a ubiquitin E3 ligase, HECTD2, which ubiquitinated and mediated the degradation of PIAS1, thus increasing inflammation in an experimental pneumonia model. We found that GSK3ß phosphorylation of PIAS1 provided a phosphodegron for HECTD2 targeting. We also identified a mislocalized HECTD2 polymorphism, HECTD2(A19P), that was present in 8.5% of the population and functioned to reduce inflammation. This polymorphism prevented HECTD2/PIAS1 nuclear interaction, thus preventing PIAS1 degradation. The HECTD2(A19P) polymorphism was also protective toward acute respiratory distress syndrome (ARDS). We then developed a small-molecule inhibitor, BC-1382, that targeted HECTD2 and attenuated lipopolysaccharide (LPS)- and Pseudomonas aeruginosa-induced lung inflammation. These studies describe an unreported innate immune pathway and suggest that mutation or antagonism of the E3 ligase HECTD2 results in reduced severity of lung inflammation by selectively modulating the abundance of the anti-inflammatory protein PIAS1.


Subject(s)
Immunity, Innate/physiology , Inflammation/physiopathology , Lung Injury/immunology , Lung Injury/physiopathology , Ubiquitin-Protein Ligases/physiology , Animals , Gene Knockdown Techniques , Lung Injury/microbiology , Mice , Phosphorylation , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Pseudomonas/pathogenicity
5.
J Biol Chem ; 290(29): 18124-18133, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26037928

ABSTRACT

The inflammasome is a multiprotein complex that augments the proinflammatory response by increasing the generation and cellular release of key cytokines. Specifically, the NALP3 inflammasome requires two-step signaling, priming and activation, to be functional to release the proinflammatory cytokines IL-1ß and IL-18. The priming process, through unknown mechanisms, increases the protein levels of NALP3 and pro-IL-1ß in cells. Here we show that LPS increases the NALP3 protein lifespan without significantly altering steady-state mRNA in human cells. LPS exposure reduces the ubiquitin-mediated proteasomal processing of NALP3 by inducing levels of an E3 ligase component, FBXO3, which targets FBXL2. The latter is an endogenous mediator of NALP3 degradation. FBXL2 recognizes Trp-73 within NALP3 for interaction and targets Lys-689 within NALP3 for ubiquitin ligation and degradation. A unique small molecule inhibitor of FBXO3 restores FBXL2 levels, resulting in decreased NALP3 protein levels in cells and, thereby, reducing the release of IL-1ß and IL-18 in human inflammatory cells after NALP3 activation. Our findings uncover NALP3 as a molecular target for FBXL2 and suggest that therapeutic targeting of the inflammasome may serve as a platform for preclinical intervention.


Subject(s)
Carrier Proteins/immunology , F-Box Proteins/immunology , Inflammasomes/immunology , Lipopolysaccharides/immunology , Ubiquitin-Protein Ligases/immunology , Carrier Proteins/metabolism , Cell Line , F-Box Proteins/metabolism , Humans , Immunity, Innate , Inflammasomes/metabolism , Interleukin-18/immunology , Interleukin-1beta/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Proteolysis , SKP Cullin F-Box Protein Ligases/immunology , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
6.
J Biol Chem ; 290(19): 11843-52, 2015 May 08.
Article in English | MEDLINE | ID: mdl-25778398

ABSTRACT

Fbxl7, a component of the Skp1·Cul1·F-box protein type ubiquitin E3 ligase, regulates mitotic cell cycle progression. Here we demonstrate that overexpression of Fbxl7 in lung epithelia decreases the protein abundance of survivin, a member of the inhibitor of apoptosis family. Fbxl7 mediates polyubiquitylation and proteasomal degradation of survivin by interacting with Glu-126 within its carboxyl-terminal α helix. Furthermore, both Lys-90 and Lys-91 within survivin serve as ubiquitin acceptor sites. Ectopically expressed Fbxl7 impairs mitochondrial function, whereas depletion of Fbxl7 protects mitochondria from actions of carbonyl cyanide m-chlorophenylhydrazone, an inhibitor of oxidative phosphorylation. Compared with wild-type survivin, cellular expression of a survivin mutant protein deficient in its ability to interact with Fbxl7 (E126A) and a ubiquitylation-resistant double point mutant (KK90RR/KK91RR) rescued mitochondria to a larger extent from damage induced by overexpression of Fbxl7. Therefore, these data suggest that the Skp1·Cul1·F-box protein complex subunit Fbxl7 modulates mitochondrial function by controlling the cellular abundance of survivin. The results raise opportunities for F-box protein targeting to preserve mitochondrial function.


Subject(s)
Apoptosis , F-Box Proteins/metabolism , Mitochondria/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Ubiquitination , Adenosine Triphosphate/metabolism , Animals , Energy Metabolism , F-Box Proteins/genetics , Gene Expression Regulation , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins/metabolism , Mice , Mutation , Protein Structure, Secondary , Repressor Proteins/metabolism , Survivin
7.
Cell Rep ; 7(2): 476-487, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24703837

ABSTRACT

Acute lung injury (ALI) is linked to mitochondrial injury, resulting in impaired cellular oxygen utilization; however, it is unknown how these events are linked on the molecular level. Cardiolipin, a mitochondrial-specific lipid, is generated by cardiolipin synthase (CLS1). Here, we show that S. aureus activates a ubiquitin E3 ligase component, Fbxo15, that is sufficient to mediate proteasomal degradation of CLS1 in epithelia, resulting in decreased cardiolipin availability and disrupted mitochondrial function. CLS1 is destabilized by the phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1), which binds CLS1 to phosphorylate and regulates CLS1 disposal. Like Fbxo15, PINK1 interacts with and regulates levels of CLS1 through a mechanism dependent upon Thr219. S. aureus infection upregulates this Fbxo15-PINK1 pathway to impair mitochondrial integrity, and Pink1 knockout mice are less prone to S. aureus-induced ALI. Thus, ALI-associated disruption of cellular bioenergetics involves bioeffectors that utilize a phosphodegron to elicit ubiquitin-mediated disposal of a key mitochondrial enzyme.


Subject(s)
B7-2 Antigen/metabolism , F-Box Proteins/metabolism , Mitochondria/metabolism , Pneumonia/metabolism , Protein Kinases/metabolism , Adolescent , Adult , Animals , B7-2 Antigen/genetics , Case-Control Studies , Cell Line , Cells, Cultured , Child , Enzyme Stability , F-Box Proteins/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Protein Kinases/genetics , Protein Subunits/genetics , Protein Subunits/metabolism
8.
J Phys Chem A ; 115(25): 6979-85, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21434660

ABSTRACT

We present potential energy curves calculated at the CCSD(T) level of theory for Ga(+)-RG and In(+)-RG complexes (RG = He-Rn). Spectroscopic parameters have been derived from these potentials and compared to previously calculated parameters for the Al(+)-RG and Tl(+)-RG complexes. Additionally, for some cases, we compare these parameters with those obtained from electronic spectroscopic studies on excited states of the neutral species, arising from atomic-based d ← p excitations. The Ga(+)-RG and In(+)-RG potentials have also been used to calculate the transport coefficients for M(+) traveling through a bath of RG atoms.

SELECTION OF CITATIONS
SEARCH DETAIL