Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 289(46): 31736-31750, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25288807

ABSTRACT

Stefin B (cystatin B) is an endogenous cysteine cathepsin inhibitor, and the loss-of-function mutations in the stefin B gene were reported in patients with Unverricht-Lundborg disease (EPM1). In this study we demonstrated that stefin B-deficient (StB KO) mice were significantly more sensitive to the lethal LPS-induced sepsis and secreted higher amounts of pro-inflammatory cytokines IL-1ß and IL-18 in the serum. We further showed that increased caspase-11 gene expression and better pro-inflammatory caspase-1 and -11 activation determined in StB KO bone marrow-derived macrophages resulted in enhanced IL-1ß processing. Pretreatment of macrophages with the cathepsin inhibitor E-64d did not affect secretion of IL-1ß, suggesting that the increased cathepsin activity determined in StB KO bone marrow-derived macrophages is not essential for inflammasome activation. Upon LPS stimulation, stefin B was targeted into the mitochondria, and the lack of stefin B resulted in the increased destabilization of mitochondrial membrane potential and mitochondrial superoxide generation. Collectively, our study demonstrates that the LPS-induced sepsis in StB KO mice is dependent on caspase-11 and mitochondrial reactive oxygen species but is not associated with the lysosomal destabilization and increased cathepsin activity in the cytosol.


Subject(s)
Cystatin B/physiology , Endotoxemia/metabolism , Gene Expression Regulation , Inflammation/metabolism , Animals , Caspases/metabolism , Caspases, Initiator , Escherichia coli/metabolism , Inflammasomes/metabolism , Lipopolysaccharides , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism
2.
Protein Pept Lett ; 21(9): 957-65, 2014.
Article in English | MEDLINE | ID: mdl-24702263

ABSTRACT

Cystatin F is a unique member of the cystatin family of cysteine protease inhibitors, which is synthesized as an inactive dimer and it is activated by N-terminal cleavage in the endolysosomes. It is expressed in the cells of the immune system: myeloid cells and the cells involved in target cell killing: natural killer (NK) cells and cytotoxic T cells (CTLs). Upon activation of the NK cells with interleukin 2 (IL-2), cystatin F was found upregulated and co-localized in cytotoxic granules with cathepsin C (CatC) and CatV. However, cystatin F inhibits the CatC in cells only when its N-terminal part is processed. Although cystatin F could inhibit both CatV and CatC, the IL-2 stimulation of the YT cells resulted in an increased CatV activity, while the CatC activity was unchanged. The incubation of IL-2 activated NK cells with a cysteine proteinase inhibitor E-64d increased the cystatin F dimer formation. Our results suggest that cystatin F not only inhibits CatV, but it is processed by the CatV in order to inhibit the CatC activity in cytotoxic granules. The regulation of the CatC activity in the cytotoxic granules of the NK cells by the cystatin F could be important for the processing and activation of granule-associated serine proteases - granzymes.


Subject(s)
Biomarkers, Tumor/metabolism , Cystatins/metabolism , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/enzymology , Peptide Hydrolases/metabolism , Blotting, Western , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Microscopy, Confocal , Up-Regulation/drug effects
3.
FEBS Lett ; 588(5): 720-6, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24462687

ABSTRACT

Innate immune responses are tightly regulated to avoid excessive activation and subsequent inflammatory damage to the host, and interleukin-10 (IL-10) plays a crucial role in preventing inflammation. Stefin B (cystatin B) is an endogenous inhibitor of cysteine proteinases. In stefin B-deficient bone marrow-derived macrophages (BMDMs), we detected an increase in the induction of the LPS-induced pro-inflammatory signal nitric oxide (NO) but decreased IL-10 expression. The phosphorylation of ERK and p38 MAP-kinases was significantly decreased in stefin B-deficient macrophages, as was STAT-3 phosphorylation. These findings show that stefin B influences the expression of anti-inflammatory IL-10 in response to the TLR4 agonist LPS.


Subject(s)
Cystatin B/deficiency , Interleukin-10/metabolism , Macrophages/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cells, Cultured , Cystatin B/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Interferon-gamma/genetics , Interleukin-10/genetics , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System , Macrophage Activation , Macrophages/immunology , Mice, Knockout , Nitric Oxide/metabolism , Transcriptional Activation/immunology
4.
J Biol Chem ; 285(13): 10078-10086, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20075068

ABSTRACT

Stefin B (cystatin B) is an endogenous inhibitor of cysteine proteinases localized in the nucleus and the cytosol. Loss-of-function mutations in the stefin B gene (CSTB) gene were reported in patients with Unverricht-Lundborg disease (EPM1). We have identified an interaction between stefin B and nucleosomes, specifically with histones H2A.Z, H2B, and H3. In synchronized T98G cells, stefin B co-immunoprecipitated with histone H3, predominantly in the G(1) phase of the cell cycle. Stefin B-deficient mouse embryonic fibroblasts entered S phase earlier than wild type mouse embryonic fibroblasts. In contrast, increased expression of stefin B in the nucleus delayed cell cycle progression in T98G cells. The delay in cell cycle progression was associated with the inhibition of cathepsin L in the nucleus, as judged from the decreased cleavage of the CUX1 transcription factor. In vitro, inhibition of cathepsin L by stefin B was potentiated in the presence of histones, whereas histones alone did not affect the cathepsin L activity. Interaction of stefin B with the Met-75 truncated form of cathepsin L in the nucleus was confirmed by fluorescence resonance energy transfer experiments in the living cells. Stefin B could thus play an important role in regulating the proteolytic activity of cathepsin L in the nucleus, protecting substrates such as transcription factors from its proteolytic processing.


Subject(s)
Cathepsin L/metabolism , Cell Nucleus/metabolism , Cystatin B/metabolism , Gene Expression Regulation , Histones/chemistry , Animals , Cell Cycle , Cell Line, Tumor , Cytosol/metabolism , Fibroblasts/metabolism , Fluorescence Resonance Energy Transfer/methods , Histones/metabolism , Humans , Mice , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...