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










Database
Type of study
Language
Publication year range
1.
Front Immunol ; 9: 818, 2018.
Article in English | MEDLINE | ID: mdl-29755460

ABSTRACT

Proteinase 3 (PR3) is the autoantigen in granulomatosis with polyangiitis, an autoimmune necrotizing vasculitis associated with anti-neutrophil cytoplasmic antibodies (ANCAs). Moreover, PR3 is a serine protease whose membrane expression can potentiate inflammatory diseases such as ANCA-associated vasculitis and rheumatoid arthritis. During apoptosis, PR3 is co-externalized with phosphatidylserine (PS) and is known to modulate the clearance of apoptotic cells through a calreticulin (CRT)-dependent mechanism. The complement protein C1q is one mediator of efferocytosis, the clearance of altered self-cells, particularly apoptotic cells. Since PR3 and C1q are both involved in the clearance of apoptotic cells and immune response modulation and share certain common ligands (i.e., CRT and PS), we examined their possible interaction. We demonstrated that C1q binding was increased on apoptotic rat basophilic leukemia (RBL) cells that expressed PR3, and we demonstrated the direct interaction between purified C1q and PR3 molecules as shown by surface plasmon resonance. To better understand the functional consequence of this partnership, we tested C1q-dependent phagocytosis of the RBL cell line expressing PR3 and showed that PR3 impaired C1q enhancement of apoptotic cell uptake. These findings shed new light on the respective roles of C1q and PR3 in the elimination of apoptotic cells and suggest a novel potential axis to explore in autoimmune diseases characterized by a defect in apoptotic cell clearance and in the resolution of inflammation.


Subject(s)
Apoptosis , Complement C1q/immunology , Myeloblastin/immunology , Animals , Cell Line, Tumor , Cell Membrane/immunology , Humans , Inflammation , Leukemia, Basophilic, Acute/immunology , Myeloblastin/genetics , Neutrophils/immunology , Phagocytosis , Protein Binding , Rats
2.
J Immunol ; 189(5): 2574-83, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22844112

ABSTRACT

Proteinase 3 (PR3) is the target of anti-neutrophil cytoplasm Abs in granulomatosis with polyangiitis, a form of systemic vasculitis. Upon neutrophil apoptosis, PR3 is coexternalized with phosphatidylserine and impaired macrophage phagocytosis. Calreticulin (CRT), a protein involved in apoptotic cell recognition, was found to be a new PR3 partner coexpressed with PR3 on the neutrophil plasma membrane during apoptosis, but not after degranulation. The association between PR3 and CRT was demonstrated in neutrophils by confocal microscopy and coimmunoprecipitation. Evidence for a direct interaction between PR3 and the globular domain of CRT, but not with its P domain, was provided by surface plasmon resonance spectroscopy. Phagocytosis of apoptotic neutrophils from healthy donors was decreased after blocking lipoprotein receptor-related protein (LRP), a CRT receptor on macrophages. In contrast, neutrophils from patients with granulomatosis with polyangiitis expressing high membrane PR3 levels showed a lower rate of phagocytosis than those from healthy controls not affected by anti-LRP, suggesting that the LRP-CRT pathway was disturbed by PR3-CRT association. Moreover, phagocytosis of apoptotic PR3-expressing cells potentiated proinflammatory cytokine in vitro by human monocyte-derived macrophages and in vivo by resident murine peritoneal macrophages, and diverted the anti-inflammatory response triggered by the phagocytosis of apoptotic cells after LPS challenge in thioglycolate-elicited murine macrophages. Therefore, membrane PR3 expressed on apoptotic neutrophils might amplify inflammation and promote autoimmunity by affecting the anti-inflammatory "reprogramming" of macrophages.


Subject(s)
Apoptosis/immunology , Autoantigens/metabolism , Calreticulin/metabolism , Granulomatosis with Polyangiitis/immunology , Macrophages/immunology , Microscopic Polyangiitis/immunology , Myeloblastin/metabolism , Neutrophils/immunology , Adjuvants, Immunologic/physiology , Animals , Granulomatosis with Polyangiitis/enzymology , Granulomatosis with Polyangiitis/pathology , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/pathology , Macrophages/enzymology , Macrophages/pathology , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopic Polyangiitis/enzymology , Microscopic Polyangiitis/pathology , Neutrophils/enzymology , Neutrophils/pathology , Rats
3.
J Biol Chem ; 287(7): 5070-8, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22184139

ABSTRACT

The ATP-binding cassette transporter ABCB4 is a phosphatidylcholine translocator specifically expressed at the bile canalicular membrane in hepatocytes, highly homologous to the multidrug transporter ABCB1. Variations in the ABCB4 gene sequence cause progressive familial intrahepatic cholestasis type 3. We have shown previously that the I541F mutation, when reproduced either in ABCB1 or in ABCB4, led to retention in the endoplasmic reticulum (ER)/Golgi. Here, Madin-Darby canine kidney cells expressing ABCB1-GFP were used as a model to investigate this mutant. We show that ABCB1-I541F is not properly folded and is more susceptible to in situ protease degradation. It colocalizes and coprecipitates with the ER chaperone calnexin and coprecipitates with the cytosolic chaperone Hsc/Hsp70. Silencing of calnexin or overexpression of Hsp70 have no effect on maturation of the mutant. We also tested potential rescue by chemical and pharmacological chaperones. Thapsigargin and sodium 4-phenyl butyrate were inefficient. Glycerol improved maturation and exit of the mutant from the ER. Cyclosporin A, a competitive substrate for ABCB1, restored maturation, plasma membrane expression, and activity of ABCB1-I541F. Cyclosporin A also improved maturation of ABCB4-I541F in Madin-Darby canine kidney cells. In HepG(2) cells transfected with ABCB4-I541F cDNA, cyclosporin A allowed a significant amount of the mutant protein to reach the membrane of bile canaliculi. These results show that the best strategy to rescue conformation-defective ABCB4 mutants is provided by pharmacological chaperones that specifically target the protein. They identify cyclosporin A as a potential novel therapeutic tool for progressive familial intrahepatic cholestasis type 3 patients.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Calnexin/metabolism , Cryoprotective Agents/pharmacology , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Glycerol/pharmacology , HSC70 Heat-Shock Proteins/metabolism , Mutation, Missense , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Amino Acid Substitution , Animals , Antineoplastic Agents/pharmacology , Calnexin/genetics , Cholestasis/drug therapy , Cholestasis/genetics , Cholestasis/metabolism , Dogs , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Gene Silencing , HSC70 Heat-Shock Proteins/genetics , Hep G2 Cells , Humans , Phenylbutyrates/pharmacology , Protein Transport/drug effects , Protein Transport/genetics , Thapsigargin/pharmacology
4.
J Leukoc Biol ; 90(5): 941-50, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21821719

ABSTRACT

PR3, also called myeloblastin, is a neutrophil serine protease that promotes myeloid cell proliferation by cleaving the cyclin-dependent kinase inhibitor p21(cip1/waf1). In addition, it is the target of ANCA in GPA, a necrotizing vasculitis. Anti-PR3 ANCA binding to membrane-expressed PR3 triggers neutrophil activation, potentiating vascular inflammation. This study performed in RBL cells identifies the structural motifs of PR3 membrane anchorage and examines its impact on PR3 proinflammatory and proliferative functions. With the use of MD simulations and mutagenesis, we demonstrate that the mutations of four hydrophobic (F180, F181, L228, F229) or four basic (R193, R194, K195, R227) amino acids abrogated PR3 membrane anchorage. The hydrophobic patch-deficient PR3 mutant (PR34H4A) was still able to cleave the synthetic substrate Boc-Ala-Pro-Val in cell lysates. However, in contrast to WT PR3, PR34H4A was not expressed at the plasma membrane after degranulation and failed to cleave extracellular fibronectin, was not externalized after apoptosis and did not impair macrophage phagocytosis of apoptotic cells, did not promote myeloid cell proliferation and failed to cleave p21/waf1. PR3 membrane insertion appears to be pivotal for its proinflammatory activities, such as extracellular proteolysis and impairment of apoptotic cell clearance, but also for myeloid cell proliferation. Targeting membrane-associated PR3 might constitute a novel, anti-inflammatory therapeutic strategy in inflammatory disease especially in vasculitis, but this approach has to be validated in mature neutrophils.


Subject(s)
Basophils/immunology , Myeloblastin , Neutrophil Activation , Animals , Apoptosis , Basophils/enzymology , Basophils/ultrastructure , Cell Line , Cell Membrane/enzymology , Cell Membrane/immunology , Cell Proliferation , Inflammation , Mutation , Myeloblastin/chemistry , Myeloblastin/genetics , Myeloblastin/immunology , Protein Structure, Tertiary , Proteolysis , Rats
5.
J Immunol ; 182(11): 7254-63, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19454722

ABSTRACT

Because neutrophil apoptosis plays a key role in resolving inflammation, identification of proteins regulating neutrophil survival should provide new strategies to modulate inflammation. Using a proteomic approach, coronin-1 was identified as a cytosolic protein cleaved during neutrophil apoptosis. Coronin-1 is an actin-binding protein that can associate with phagosomes and NADPH oxidase, but its involvement in apoptosis was currently unknown. In coronin-1-transfected PLB985 cells, coronin-1 overexpression did not modify the kinetics of granulocyte differentiation as assessed by CD11b labeling. Concerning apoptosis, increased coronin-1 expression in dimethylformamide-differentiated PLB985 significantly decreased gliotoxin-induced mitochondrial depolarization as compared with controls. Likewise, coronin-1 significantly decreased TRAIL-induced apoptosis with less mitochondrial depolarization, caspase-3 and caspase-9 activities, but not caspase-8 or Bid truncation suggesting that coronin-1 interfered with mitochondria-related events. To validate the prosurvival role of coronin-1 in a pathophysiological condition involving neutrophil-dominated inflammation, neutrophils from cystic fibrosis (CF) patients were studied. Circulating neutrophils from CF patients had more coronin-1 expression assessed by immunoblotting or proteomic analysis of cytosolic proteins. This was associated with a lower apoptosis rate than those from controls evidenced by delayed phosphatidylserine externalization and mitochondria depolarization. In addition, inflammatory neutrophils from CF patients lungs showed an intense coronin-1 immunolabeling. We concluded that coronin-1 could constitute a potential target in resolving inflammation.


Subject(s)
Apoptosis , Microfilament Proteins/analysis , Neutrophils/cytology , Cell Survival , Cystic Fibrosis/pathology , Cytosol/chemistry , Humans , Hydrolysis , Inflammation , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Mitochondria/physiology , Neutrophils/pathology , Proteomics
SELECTION OF CITATIONS
SEARCH DETAIL
...