Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Thromb Haemost ; 18(1): 104-113, 2020 01.
Article in English | MEDLINE | ID: mdl-31549480

ABSTRACT

BACKGROUND: Recombinant factor VIIa (rFVIIa) enhances thrombin generation in a platelet-dependent manner; however, rFVIIa binds activated platelets with relatively low affinity. Triggering receptor expressed on myeloid cells (TREM)-like transcript (TLT)-1 is expressed exclusively on activated platelets. OBJECTIVE: To enhance the potency of rFVIIa via binding TLT-1. METHODS: Recombinant FVIIa was conjugated to a TLT-1 binding Fab. In vitro potency of this platelet-targeted rFVIIa (PT-rFVIIa) was evaluated using factor X activation assays and by measuring viscoelastic changes in whole blood. In vivo potency was evaluated using a tail vein transection model in F8-/- mice expressing human TLT-1. RESULTS: PT-rFVIIa and rFVIIa had similar dissociation constant values for tissue factor binding and similar tissue factor-dependent factor X activation. However, PT-rFVIIa had increased catalytic efficiency on TLT-1-loaded vesicles and activated platelets. The in vitro potency in normal human blood with antibody-induced hemophilia A was dependent on assay conditions used; with maximally activated platelets, the half maximal effective concentration for clot time for PT-rFVIIa was 49-fold lower compared with rFVIIa. In the murine bleeding model, a 53-fold lower half maximal effective concentration was observed for blood loss for PT-rFVIIa, supporting the relevance of the assay conditions with maximally activated platelets. In vitro analysis of blood from subjects with hemophilia A confirmed the data obtained with normal blood. CONCLUSIONS: Increasing the affinity of rFVIIa to activated platelets resulted in approximately 50-fold increased potency both in vitro and in the mouse model. The correlation of in vivo with in vitro data using maximally activated platelets supports that these assay conditions are relevant when evaluating platelet-targeted hemostatic concepts.


Subject(s)
Blood Platelets , Hemophilia A , Animals , Factor VIIa , Hemophilia A/drug therapy , Mice , Recombinant Proteins , Thrombin
2.
MAbs ; 10(1): 118-128, 2018 01.
Article in English | MEDLINE | ID: mdl-29185848

ABSTRACT

Decysin-1 (ADAMDEC1) is an orphan ADAM-like metalloprotease with unknown biological function and a short domain structure. ADAMDEC1 mRNA has previously been demonstrated primarily in macrophages and mature dendritic cells. Here, we generated monoclonal antibodies (mAbs) against the mature ADAMDEC1 protein, as well as mAbs specific for the ADAMDEC1 pro-form, enabling further investigations of the metalloprotease. The generated mAbs bind ADAMDEC1 with varying affinity and represent at least six different epitope bins. Binding of mAbs to one epitope bin in the C-terminal disintegrin-like domain efficiently reduces the proteolytic activity of ADAMDEC1. A unique mAb, also recognizing the disintegrin-like domain, stimulates the caseinolytic activity of ADAMDEC1 while having no significant effect on the proteolysis of carboxymethylated transferrin. Using two different mAbs binding the disintegrin-like domain, we developed a robust, quantitative sandwich ELISA and demonstrate secretion of mature ADAMDEC1 protein by primary human macrophages. Surprisingly, we also found ADAMDEC1 present in human plasma with an approximate concentration of 0.5 nM. The presence of ADAMDEC1 both in human plasma and in macrophage cell culture supernatant were biochemically validated using immunoprecipitation and Western blot analysis demonstrating that ADAMDEC1 is secreted in a mature form.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Epitopes/immunology , Protease Inhibitors/pharmacology , Proteolysis/drug effects , ADAM Proteins/blood , ADAM Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Binding Sites, Antibody , Carboxylic Acids/metabolism , Cells, Cultured , Humans , Kinetics , Macrophages/drug effects , Macrophages/enzymology , Methylation , Mice , Protease Inhibitors/immunology , Protein Binding , Protein Interaction Domains and Motifs , Substrate Specificity , Transferrin/analogs & derivatives , Transferrin/metabolism
5.
Biochem J ; 407(1): 41-8, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17650073

ABSTRACT

Most known organisms encode proteases that are crucial for constitutive proteolytic events. In the present paper, we describe a method to define these events in proteomes from Escherichia coli to humans. The method takes advantage of specific N-terminal biotinylation of protein samples, followed by affinity enrichment and conventional LC (liquid chromatography)-MS/MS (tandem mass spectrometry) analysis. The method is simple, uses conventional and easily obtainable reagents, and is applicable to most proteomics facilities. As proof of principle, we demonstrate profiles of proteolytic events that reveal exquisite in vivo specificity of methionine aminopeptidase in E. coli and unexpected processing of mitochondrial transit peptides in yeast, mouse and human samples. Taken together, our results demonstrate how to rapidly distinguish real proteolysis that occurs in vivo from the predictions based on in vitro experiments.


Subject(s)
Chromatography, Liquid/methods , Peptide Hydrolases/metabolism , Proteomics/methods , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Aminopeptidases/chemistry , Aminopeptidases/metabolism , Animals , Aprotinin/chemistry , Aprotinin/metabolism , Blood Proteins/chemistry , Blood Proteins/metabolism , Caspases/chemistry , Caspases/metabolism , Cells, Cultured , Genome, Fungal , Humans , Methionyl Aminopeptidases , Methylurea Compounds/pharmacology , Mice , Mitochondria/metabolism , Models, Biological , Molecular Sequence Data , Peptides/analysis , Peptides/chemistry , Peptides/metabolism , Proteome/analysis , Proteome/metabolism
6.
J Proteome Res ; 6(7): 2850-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17547438

ABSTRACT

The identification of natural substrates and their cleavage sites is pivotal to defining proteolytic pathways. Here we report a novel strategy for the identification of the signature of proteolytic cleavage events based on quantitative proteomics. Lysine residues in proteins are blocked by guanidination so that free N-terminals can be labeled with amine-specific iTRAQ reagents. The quantitative nature of iTRAQ reagents allows us to distinguish N-terminals newly formed by proteolytic treatment (neoepitopes) from original N-terminals in proteins. Proteins are digested with trypsin and analyzed using MALDI-TOF/TOF mass spectrometry. Peptides labeled with iTRAQ reagents are distinguished from other peptides by exhibiting intense signature ions in tandem mass spectrometry analysis. A corresponding data acquisition strategy was developed to specifically analyze iTRAQ tagged N-terminal peptides. To validate the procedure, we examined a set of recombinant Escherichia coli proteins that have predicted caspase-3 cleavage motifs. The protein mixture was treated with active or inactive caspase-3 and subsequently labeled with two different iTRAQ reagents. Mass spectrometric analysis located 10 cleavage sites, all corresponding to caspase-3 consensus. Spiking caspase-cleaved substrate into a human cell lysate demonstrated the high sensitivity of the procedure. Moreover, we were able to identify proteolytic cleavage products associated with the induction of cell-free apoptosis. Together, these data reveal a novel application for iTRAQ technology for the detection of proteolytic substrates.


Subject(s)
Peptide Hydrolases/metabolism , Proteomics/methods , Amino Acid Sequence , Caspase 3/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Hydrolysis , Lysine/chemistry , Molecular Sequence Data , Peptides/chemistry , Proteins/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity
7.
Biochem Biophys Res Commun ; 327(3): 774-9, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15649413

ABSTRACT

Human mitochondrial glutaredoxin 2 (Grx2) catalyzes glutathione-dependent dithiol reaction mechanisms, reducing protein disulfides, and monothiol reactions, reducing mixed disulfides between proteins and GSH (de-/glutathionylation). Here, we have overexpressed Grx2 in HeLa cells in its mitochondrial form (mGrx2-HeLa) as well as a truncated cytosolic form, lacking the mitochondrial translocation signal (tGrx2-HeLa). The resulting clones were less susceptible to apoptosis induced by 2-deoxy-d-glucose (2-DG) or doxorubicin (Dox). Overexpression of Grx2 inhibited cytochrome c release and caspase activation induced by both agents. In addition, Grx2 prevented 2-DG- and Dox-induced loss of cardiolipin, the phospholipid anchoring cytochrome c to the inner mitochondrial membrane. Overexpression of mGrx2 provided better protection than tGrx2 overexpression, especially after treatment with 2-DG. We propose that Grx2 facilitates the maintenance of cellular redox homeostasis upon treatment with apoptotic agents, thereby preventing cardiolipin oxidation and cytochrome c release.


Subject(s)
Apoptosis/drug effects , Cytochromes c/metabolism , Oxidoreductases/pharmacology , Apoptosis/physiology , Cardiolipins/metabolism , Caspases/metabolism , Cytochromes c/antagonists & inhibitors , Cytosol/metabolism , Deoxyglucose/pharmacology , Doxorubicin/pharmacology , Gene Expression Regulation , Glutaredoxins , Humans , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Oxidoreductases/genetics , Oxidoreductases/metabolism , Tumor Cells, Cultured
8.
J Biol Chem ; 279(48): 49575-8, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15475367

ABSTRACT

Caspases are cysteine proteases that play a central role in the execution of apoptosis. Recent evidence indicates that caspase-2 is activated early in response to genotoxic stress and can function as an upstream modulator of the mitochondrial apoptotic pathway. In particular, we have shown previously that fully processed caspase-2 can permeabilize the outer mitochondrial membrane and cause cytochrome c and Smac/DIABLO release from these organelles. Using permeabilized cells, isolated mitochondria, and protein-free liposomes, we now report that this effect is direct and depends neither on the presence or cleavage of other proteins nor on a specific phospholipid composition of the liposomal membrane. Interestingly, caspase-2 was also shown to disrupt the interaction of cytochrome c with anionic phospholipids, notably cardiolipin, and thereby enhance the release of the hemoprotein caused by treatment of mitochondria with digitonin or the proapoptotic protein Bax. Combined, our data suggest that caspase-2 possesses an unparalleled ability to engage the mitochondrial apoptotic pathway by permeabilizing the outer mitochondrial membrane and/or by breaching the association of cytochrome c with the inner mitochondrial membrane.


Subject(s)
Caspases/metabolism , Cytochromes c/metabolism , Phospholipids/metabolism , Caspase 2 , Dextrans/metabolism , Digitonin , Humans , Indicators and Reagents , Jurkat Cells , Liposomes/metabolism , Liver/enzymology , Liver/metabolism , Mitochondria/metabolism
10.
Proc Natl Acad Sci U S A ; 101(36): 13227-32, 2004 Sep 07.
Article in English | MEDLINE | ID: mdl-15328416

ABSTRACT

Glutaredoxin (Grx) belongs to the thioredoxin fold superfamily and catalyzes glutathione-dependent oxidoreductions. The recently discovered mitochondrial and nuclear Grx (Grx2) differs from the more abundant cytosolic Grx (Grx1) by its higher affinity toward S-glutathionylated proteins and by being a substrate for thioredoxin reductase. Here, we have successfully established a method to silence the expression of Grx2 in HeLa cells by using short interfering RNA to study its role in the cell. Cells with levels of Grx2 <3% of the control were dramatically sensitized to cell death induced by doxorubicin/adriamycin and phenylarsine oxide but did not show signs of a general increase in oxidative damage with respect to carbonylation and glutathionylation. The ED(50) for doxorubicin dropped from 40 to 0.7 microM and for phenylarsine oxide from 200 to 5 nM. However, no differences were detected after treatment with cadmium, a known inhibitor of Grx1. These results indicate a crucial role of Grx2 in the regulation of the mitochondrial redox status and regulation of cell death at the mitochondrial checkpoint.


Subject(s)
Arsenicals/pharmacology , Doxorubicin/pharmacology , Gene Silencing , Oxidoreductases/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Cadmium/pharmacology , Glutaredoxins , HeLa Cells , Humans , Oxidative Stress , Oxidoreductases/genetics
11.
Eur J Neurosci ; 19(10): 2613-21, 2004 May.
Article in English | MEDLINE | ID: mdl-15147295

ABSTRACT

Despite an increasing interest in neural stem cell (NSC) research, relatively little is known about the biochemical regulation of cell death pathways in these cells. We demonstrate here, using murine-derived multipotent C17.2 NSCs, that cells undergo mitochondria-mediated cell death in response to apoptotic stimuli such as oxidative stress induced by 2,3-dimethoxy-1,4-naphthoquinone (DMNQ). In particular, treated cells exhibited apoptotic features, including Bax translocation, cytochrome c release, activation of caspase-9 and -3, chromatin condensation and DNA fragmentation. Although C17.2 cells possess the Fas receptor and express procaspase-8, agonistic Fas mAb treatment failed to induce apoptosis. Fas treatment activated the extracellular signal-regulated protein kinase (ERK) pathway, which may have an antiapoptotic as well as a growth stimulating role. Combined, our findings indicate that while NSCs are sensitive to cytotoxic stimuli that involve an engagement of mitochondria, Fas treatment does not induce death and may have an alternative role.


Subject(s)
Cell Death/physiology , Cell Differentiation/physiology , Mitochondria/metabolism , Neurons/cytology , Stem Cells/cytology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Annexin A5/metabolism , Antibodies, Monoclonal/pharmacology , Benzimidazoles/metabolism , Blotting, Western/methods , Caspase Inhibitors , Caspases/metabolism , Cell Line , Chromatin/drug effects , Coumarins/pharmacology , Cytochromes c/metabolism , Drug Interactions , Electrophoresis, Gel, Pulsed-Field/methods , Enzyme Inhibitors/toxicity , Flavonoids/pharmacology , Fluorescent Dyes/pharmacology , Free Radical Scavengers/pharmacology , Immunohistochemistry/methods , Metalloporphyrins/pharmacology , Mice , Mitogen-Activated Protein Kinases/metabolism , Naphthoquinones/pharmacology , Neurons/metabolism , Oligopeptides/pharmacology , Propidium/metabolism , Staurosporine/toxicity , Stem Cells/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Time Factors , fas Receptor/metabolism
12.
Biochem J ; 378(Pt 1): 213-7, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14629197

ABSTRACT

The mechanisms regulating mitochondrial outer-membrane permeabilization and the release of cytochrome c during apoptosis remain controversial. In the present study, we show in an in vitro model system that the release of cytochrome c may occur via moderate modulation of mitochondrial volume, irrespective of the mechanism leading to the mitochondrial swelling. In contrast with mitochondrial permeability transition-dependent release of cytochrome c, in the present study mitochondria remain intact and functionally active.


Subject(s)
Cytochromes c/metabolism , Ion Channels/physiology , Mitochondria/metabolism , Animals , Calcium/pharmacology , Ionophores/pharmacology , Male , Mitochondria/physiology , Mitochondria/ultrastructure , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Osmotic Pressure , Rats , Rats, Sprague-Dawley , Valinomycin/pharmacology
13.
J Biol Chem ; 279(2): 1100-7, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14551208

ABSTRACT

Cardiolipin (CL) is an inner mitochondrial membrane phospholipid that contributes to optimal mitochondrial function and is gaining widespread attention in studies of mitochondria-mediated apoptosis. Divergent hypotheses describing the role of CL in cytochrome c release and apoptosis have evolved. We addressed this controversy directly by comparing the spontaneous- and Bax-mediated cytochrome c release from mitochondria isolated from two strains of Saccharomyces cerevisiae: one lacking CL-synthase and therefore CL (DeltaCRD1) and the other, its corresponding wild type (WT). We demonstrated by liquid chromatography-mass spectrometry that the main yeast CL species [(16:1)2(18:1)2] differs in fatty acid composition from mammalian CL [(18:2)4], and we verified the absence of the yeast CL species in the DeltaCRD1 strain. We also demonstrated that the mitochondrial association of Bax and the resulting cytochrome c release is not dependent on the CL content of the yeast mitochondrial membranes. Bax inserted equally into both WT and DeltaCRD1 mitochondrial membranes under conditions that lead to the release of cytochrome c from both strains of yeast mitochondria. Furthermore, using models of synthetic liposomes and isolated yeast mitochondria, we found that cytochrome c was bound more "loosely" to the CL-deficient systems compared with when CL is present. These data challenge recent studies implicating that CL is required for Bax-mediated pore formation leading to the release of proteins from the mitochondrial intermembrane space. In contrast, they support our recently proposed two-step mechanism of cytochrome c release, which suggests that CL is required for binding cytochrome c to the inner mitochondrial membrane.


Subject(s)
Cardiolipins/physiology , Cytochromes c/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Animals , Apoptosis , Cattle , Chromatography, Liquid , Chromatography, Thin Layer , Intracellular Membranes/metabolism , Ions , Mass Spectrometry , Mitochondria/metabolism , Models, Chemical , Myocardium/metabolism , Oxygen Consumption , Phospholipids/metabolism , Potassium Chloride/chemistry , Protein Binding , Sucrose/metabolism , Time Factors , bcl-2-Associated X Protein
14.
FASEB J ; 17(1): 124-6, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12475911

ABSTRACT

Fluorescence microscopy of A549 cells stained with a glutathione (L-gamma-glutamyl-L-cysteinylglycine, GSH)-specific polyclonal antibody displayed uniform staining of the peri-nuclear cytosol, with the nuclear region apparently lacking GSH staining. This discontinuous staining was confirmed in other cell types and also corroborated in A549 cells stained with the thiol-reactive dye mercury orange. The selectivity of antibody binding was confirmed by buthionine sulfoximine (BSO)-dependent inhibition of GSH synthesis. However, confocal visualization of antibody-stained A549 cells in the z-plane revealed the majority of the peri-nuclear staining intensity in the upper half of the cell to be associated with mitochondria, as confirmed by double staining for cytochrome oxidase. Integration of the confocal signals from the nuclear and cytosolic regions halfway down the z-plane showed that the GSH concentrations of these compartments are close to equilibrium. Confirmation of the relatively high levels of mitochondrial glutathione was provided in cells treated with BSO and visualized in z-section, revealing the mitochondrial GSH content of these cells to be well preserved in apposition to near-complete depletion of cytosolic/nuclear GSH. Localized gradients within the cytosolic compartment were also visible, particularly in the z-plane. The antibody also provided initial visualization of the compartmentalization of protein-GSH mixed disulfides formed in A549 cells exposed to diamide. Discontinuous staining was again evident, with heavy staining in membrane blebs and in the nuclear region. Using FACS analysis of anti-GSH antibody-stained Jurkat T lymphocytes, we also demonstrated population variations in the cellular compliment of GSH and protein-GSH mixed disulfides, formed in response to diamide. In addition, we showed cell-cycle variation in GSH content of the cells, with the highest levels of GSH associated with the G2/M mitotic phase of the cell cycle, using double staining with propidium iodide. Similar FACS analyses performed in isolated mitochondria presented a considerable variation in GSH content within mitochondria of uniform granularity from the same preparation.


Subject(s)
Glutathione Disulfide/analysis , Glutathione/analysis , Animals , Cell Compartmentation , Cell Cycle , Cell Line , Cells, Cultured , Diamide/pharmacology , Flow Cytometry , Glutathione/chemistry , Microscopy, Confocal , Mitochondria/chemistry , Models, Biological , Oxidative Stress , Proteins/analysis
15.
J Biol Chem ; 277(33): 29803-9, 2002 Aug 16.
Article in English | MEDLINE | ID: mdl-12065594

ABSTRACT

DNA damage induced by the cancer chemotherapeutic drug etoposide triggers the onset of a series of intracellular events characteristic of apoptosis. Among the early changes observed is the release of cytochrome c from mitochondria, although the mechanism responsible for this effect is unclear. We demonstrate here a role for caspase-2 in etoposide-induced cytochrome c release. In particular, Jurkat T-lymphocytes treated with an irreversible caspase-2 inhibitor, benzyloxycarbonyl-Val-Asp-Val-Ala-Asp-fluoromethyl ketone (z-VDVAD-fmk), or stably transfected with pro-caspase-2 antisense (Casp-2/AS) are refractory to cytochrome c release stimulated by etoposide. Experiments performed using a reconstituted cell-free system indicate that etoposide-induced cytochrome c release by way of caspase-2 occurs independently of cytosolic factors, suggesting that the nuclear pool of pro-caspase-2 is critical to this process. Apart from inhibiting cytochrome c release, undermining caspase-2 activity results in an attenuation of downstream events, such as pro-caspase-9 and -3 activation, phosphatidylserine exposure on the plasma membrane, and DNA fragmentation. Taken together, our data indicate that caspase-2 provides an important link between etoposide-induced DNA damage and the engagement of the mitochondrial apoptotic pathway.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Cytochrome c Group/metabolism , Etoposide/pharmacology , Mitochondria/metabolism , Caspase 2 , Humans , Jurkat Cells , Mitochondria/enzymology , Oligopeptides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...