Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 179
Filter
Add more filters

Publication year range
1.
Biotechnol Lett ; 43(1): 143-152, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33130980

ABSTRACT

OBJECTIVE: To develop recombinant factor IX (FIX) variants with augmented clotting activity. RESULTS: We generated three new variants, FIX-YKALW, FIX-ALL and FIX-LLW, expressed in SK-Hep-1 cells and characterized in vitro and in vivo. FIX-YKALW showed the highest antigen expression level among the variants (2.17 µg-mL), followed by FIX-LLW (1.5 µg-mL) and FIX-ALL (0.9 µg-mL). The expression level of FIX variants was two-five fold lower than FIX-wild-type (FIX-WT) (4.37 µg-mL). However, the biological activities of FIX variants were 15-31 times greater than FIX-WT in the chromogenic assay. Moreover, the new variants FIX-YKALW, FIX-LLW and FIX-ALL also presented higher specific activity than FIX-WT (17, 20 and 29-fold higher, respectively). FIX variants demonstrated a better clotting time than FIX-WT. In hemophilia B mice, we observed that FIX-YKALW promoted hemostatic protection. CONCLUSION: We have developed three improved FIX proteins with potential for use in protein replacement therapy for hemophilia B.


Subject(s)
Coagulants , Factor IX , Recombinant Proteins , Animals , Blood Coagulation/drug effects , Cell Line , Coagulants/chemistry , Coagulants/metabolism , Coagulants/pharmacology , Factor IX/chemistry , Factor IX/genetics , Factor IX/metabolism , Factor IX/pharmacology , Humans , Mice , Mice, Inbred C57BL , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
2.
Clin Chem Lab Med ; 59(2): 365-371, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32892172

ABSTRACT

Objectives: Emicizumab, a monoclonal antibody mimicking the function of factor (F) VIII in the activation of FX by FIXa, is widely used for prophylaxis in hemophilia patients with or without inhibitors to FVIII. Although it is administered at fixed dose, its measurement could be occasionally required. In principle, the emicizumab procoagulant effect could be assessed by the one-stage assay (OSA) currently used to measure FVIII. However, the OSA for FVIII presents with limitations. Furthermore, owing to its potent FVIII-like activity, emicizumab interferes with the measurement of the inhibitor to FVIII, which is often needed in patients on emicizumab. Methods: We prepared test samples by spiking a FVIII-deficient plasma with graded amounts of emicizumab. We modified the OSA for FVIII and tested plasma samples for emicizumab concentrations. Furthermore the chromogenic assay (CA) for FVIII with bovine reagents was used to assess for the FVIII inhibitor in patients on emicizumab. Results: Slight modification of the OSA for FVIII (i.e., higher test plasma dilution and longer coagulometer acquisition time) made the regular OSA as a reliable laboratory tool to measure emicizumab concentration as shown by the identity of the regression (observed vs. expected) lines. Furthermore, the inhibitors to FVIII in patients on emicizumab, which were negative when measured by the regular Bethesda assay, were reliably measured by the CA assay employing bovine reagents. Conclusions: The methods currently used to measure FVIII can be easily modified to make the general clinical laboratory able to assist clinicians when dealing with patients on emicizumab.


Subject(s)
Antibodies, Bispecific/blood , Antibodies, Monoclonal, Humanized/blood , Coagulants/blood , Factor VIII/metabolism , Hemophilia A/diagnosis , Aged , Animals , Antibodies, Bispecific/metabolism , Antibodies, Monoclonal, Humanized/metabolism , Binding, Competitive , Blood Coagulation , Blood Coagulation Tests , Blood Specimen Collection , Cattle , Child , Coagulants/metabolism , Dose-Response Relationship, Drug , Humans , Middle Aged , Partial Thromboplastin Time , Reference Standards , Reproducibility of Results
3.
Appl Microbiol Biotechnol ; 103(23-24): 9411-9422, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31696282

ABSTRACT

Cationic polypeptide proteins found in the seeds of the tropical plant Moringa oleifera have coagulation efficiencies similar to aluminum and ferric sulfates without their recalcitrant nature. Although these proteins possess great potential to augment or replace traditional coagulants in water treatment, harvesting active protein from seeds is laborious and not cost-effective. Here, we describe an alternative method to express and secrete active M. oleifera coagulant protein (MO) in Bacillus subtilis. A plasmid library containing the MO gene and 173 different types of secretory signal peptides was created and cloned into B. subtilis strain RIK1285. Fourteen of 440 clones screened were capable of secreting MO with yields ranging from 55 to 122 mg/L of growth medium. The coagulant activity of the highest MO secreting clone was evaluated when grown on Luria broth, and cell-free medium from the culture was shown to reduce turbidity in a buffered kaolin suspension by approximately 90% compared with controls without the MO gene. The clone was also capable of secreting active MO when grown on a defined synthetic wastewater supplemented with 0.5% tryptone. Cell-free medium from the strain harboring the MO gene demonstrated more than a 2-fold reduction in turbidity compared with controls. Additionally, no significant amount of MO was observed without the addition of the synthetic wastewater, suggesting that it served as a source of nutrients for the effective expression and translocation of MO into the medium.


Subject(s)
Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Moringa oleifera/chemistry , Plant Proteins/biosynthesis , Seeds/chemistry , Coagulants/metabolism , Flocculation , Industrial Microbiology , Plant Extracts/metabolism , Wastewater/chemistry , Water Purification/methods
4.
Int J Mol Sci ; 20(19)2019 Sep 30.
Article in English | MEDLINE | ID: mdl-31574907

ABSTRACT

Snake venom enzymes of the L-amino acid oxidase (LAAO) class are responsible for tissue hemorrhage, edema, and derangement of platelet function. However, what role, if any, these flavoenzymes play in altering plasmatic coagulation have not been well defined. Using coagulation kinetomic analyses (thrombelastograph-based), it was determined that the LAAO derived from Crotalus adamanteus venom displayed a procoagulant activity associated with weak clot strength (no factor XIII activation) similar to thrombin-like enzymes. The procoagulant activity was not modified in the presence of reduced glutathione, demonstrating that the procoagulant activity was likely due to deamination, and not hydrogen peroxide generation by the LAAO. Further, unlike the raw venom of the same species, the purified LAAO was not inhibited by carbon monoxide releasing molecule-2 (CORM-2). Lastly, exposure of the enzyme to phenylmethylsulfonyl fluoride (PMSF) resulted in the LAAO expressing anticoagulant activity, preventing contact activation generated thrombin from forming a clot. In sum, this investigation for the first time characterized the LAAO of a snake venom as both a fibrinogen polymerizing and an anticoagulant enzyme acting via oxidative deamination and not proteolysis as is the case with thrombin-like enzymes (e.g., serine proteases). Using this thrombelastographic approach, future investigation of purified enzymes can define their biochemical nature.


Subject(s)
Crotalus , L-Amino Acid Oxidase/metabolism , L-Amino Acid Oxidase/pharmacology , Snake Venoms/enzymology , Animals , Anticoagulants/chemistry , Anticoagulants/metabolism , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Calcium/metabolism , Calcium/pharmacology , Coagulants/chemistry , Coagulants/metabolism , Coagulants/pharmacology , Edetic Acid/pharmacology , Glutathione/metabolism , Glutathione/pharmacology , Heparin/pharmacology , Humans , Kinetics , L-Amino Acid Oxidase/chemistry , Organometallic Compounds/pharmacology , Thrombelastography
5.
Haematologica ; 103(6): 1073-1082, 2018 06.
Article in English | MEDLINE | ID: mdl-29545340

ABSTRACT

Tissue Factor is a cell-surface glycoprotein expressed in various cells of the vasculature and is the principal regulator of the blood coagulation cascade and hemostasis. Notably, aberrant expression of Tissue Factor is associated with cardiovascular pathologies such as atherosclerosis and thrombosis. Here, we sought to identify factors that regulate Tissue Factor gene expression and activity. Tissue Factor gene expression is regulated by various transcription factors, including activating protein-1 and nuclear factor-κ B. The peptidyl-prolyl isomerase Pin1 is known to modulate the activity of these two transcription factors, and we now show that Pin1 augments Tissue Factor gene expression in both vascular smooth muscle cells and activated endothelial cells via activating protein-1 and nuclear factor-κ B signaling. Furthermore, the cytoplasmic domain of Tissue Factor contains a well-conserved phospho-Ser258-Pro259 amino-acid motif recognized by Pin1. Using co-immunoprecipitation and solution nuclear magnetic resonance spectroscopy, we show that the WW-domain of Pin1 directly binds the cytoplasmic domain of Tissue Factor. This interaction occurs via the phospho-Ser258-Pro259 sequence in the Tissue Factor cytoplasmic domain and results in increased protein half-life and pro-coagulant activity. Taken together, our results establish Pin1 as an upstream regulator of Tissue Factor-mediated coagulation, thereby opening up new avenues for research into the use of specific Pin1 inhibitors for the treatment of diseases characterized by pathological coagulation, such as thrombosis and atherosclerosis.


Subject(s)
Coagulants/metabolism , Gene Expression , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Thromboplastin/genetics , Thromboplastin/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Multiprotein Complexes/metabolism , NF-kappa B/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Proteolysis , Thromboplastin/chemistry , Transcription Factor AP-1/metabolism
6.
Rheumatology (Oxford) ; 55(12): 2096-2108, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27018059

ABSTRACT

Atherothrombosis is a recurrent complication in APS and SLE patients. Oxidative stress has been suggested as a key player underlying this process. Autoantibodies have been pointed to as the main contributors to abnormality in the oxidative status observed in APS and SLE patients, promoting the increased production of oxidant species and the reduction of antioxidant molecules. This imbalance causes vascular damage through the activation of immune cells, including monocytes, lymphocytes and neutrophils, causing the expression of pro-inflammatory and procoagulant molecules, the formation of neutrophil extracellular traps and the adhesion of these cells to the endothelium; the induction of cellular apoptosis and impaired cell clearance, which in turn enhances autoantibody neogeneration; and cytotoxicity of endothelial cells. This review describes the mechanisms underlying the role of oxidative stress in the pathogenesis of atherothrombosis associated with APS and SLE, focused on the effect of autoantibodies, the different cell types involved and the diverse effectors, including cytokines, procoagulant proteins and their main modulators, such as oxidant/antioxidant species and intracellular pathways in each pathology. We further discuss new therapies aimed at restoring the oxidative stress balance and subsequently to tackle atherothrombosis in APS and SLE.


Subject(s)
Antiphospholipid Syndrome/complications , Atherosclerosis/etiology , Lupus Erythematosus, Systemic/complications , Oxidative Stress/physiology , Thrombosis/etiology , Acetylcysteine/therapeutic use , Anticoagulants/therapeutic use , Antioxidants/therapeutic use , Atherosclerosis/drug therapy , Autoantibodies/physiology , Coagulants/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation/physiopathology , Mitochondrial Diseases/etiology , Reactive Oxygen Species/metabolism , Recurrence , Thrombosis/drug therapy , Ubiquinone/analogs & derivatives , Ubiquinone/therapeutic use , beta 2-Glycoprotein I/metabolism , beta 2-Glycoprotein I/physiology
7.
Haemophilia ; 22(4): 615-24, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26879158

ABSTRACT

INTRODUCTION: FEIBA(®) consists of zymogens and traces of activated forms of procoagulant factors II, VII, IX, X, anticoagulants protein C and TFPI, and small amounts of cofactors FV, FVIII and protein S, in a balanced ratio. As shown previously, FII-FXa complex plays a key role in FEIBA's mode of action (MoA). METHODS: Thrombin generation (TG) was measured by spiking coagulation factors, cofactors and inhibitors to high titer FVIII inhibitor plasma, and in plasma samples from patients in a phase 3 clinical study evaluating the safety and efficacy of FEIBA prophylaxis in haemophilia A patients with inhibitors. RESULTS: Increasing the FXa/FII ratio improved TG, while adding coagulation enzyme components had a negligible effect. Adding FX, FIX, and FVII increased the peak thrombin and decreased the lag time. The presence of FV and phospholipids led to faster TG, while protein C and protein S reduced the amount of peak thrombin. TFPI appeared to have no effect. Patients on prophylaxis with FEIBA(®) showed higher peak thrombin and AUC with elevated FII, FX, FIX, FVIIa, and protein C levels, and experienced significantly less bleeding episodes than those receiving on-demand treatment. CONCLUSION: These experiments showed that although the FII-FXa complex induced immediate thrombin formation on the activated platelet surface, other procoagulant components of FEIBA were necessary to achieve an optimal thrombin burst. The presence of the pro- and anti-coagulants in FEIBA provides a haemostatic balance, and is thus expected to prevent thrombotic events. Recent clinical data verified the postulated MoA of FEIBA in prophylaxis treatment.


Subject(s)
Anticoagulants/chemistry , Blood Coagulation Factors/metabolism , Coagulants/metabolism , Thrombin/analysis , Antibodies, Neutralizing/blood , Blood Coagulation Tests , Hemophilia A/blood , Hemophilia A/pathology , Humans , Models, Molecular , Severity of Illness Index , Thrombin/metabolism
8.
Biochem J ; 468(3): 459-73, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25849538

ABSTRACT

Platelets are activated by a range of stimuli that share little or no resemblance in structure to each other or to recognized ligands, including diesel exhaust particles (DEP), small peptides [4N1-1, Champs (computed helical anti-membrane proteins), LSARLAF (Leu-Ser-Ala-Arg-Leu-Ala-Phe)], proteins (histones) and large polysaccharides (fucoidan, dextran sulfate). This miscellaneous group stimulate aggregation of human and mouse platelets through the glycoprotein VI (GPVI)-FcR γ-chain complex and/or C-type lectin-like receptor-2 (CLEC-2) as shown using platelets from mice deficient in either or both of these receptors. In addition, all of these ligands stimulate tyrosine phosphorylation in GPVI/CLEC-2-double-deficient platelets, indicating that they bind to additional surface receptors, although only in the case of dextran sulfate does this lead to activation. DEP, fucoidan and dextran sulfate, but not the other agonists, activate GPVI and CLEC-2 in transfected cell lines as shown using a sensitive reporter assay confirming a direct interaction with the two receptors. We conclude that this miscellaneous group of ligands bind to multiple proteins on the cell surface including GPVI and/or CLEC-2, inducing activation. These results have pathophysiological significance in a variety of conditions that involve exposure to activating charged/hydrophobic agents.


Subject(s)
Air Pollutants/toxicity , CD36 Antigens/chemistry , Coagulants/pharmacology , Lectins, C-Type/agonists , Lectins, C-Type/chemistry , Membrane Glycoproteins/agonists , Platelet Activation/drug effects , Vehicle Emissions/toxicity , Air Pollutants/chemistry , Air Pollutants/metabolism , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cell Line , Chickens , Coagulants/antagonists & inhibitors , Coagulants/chemistry , Coagulants/metabolism , Crosses, Genetic , Genes, Reporter/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Jurkat Cells , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Ligands , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Transgenic , Molecular Conformation , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , Phosphorylation/drug effects , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/pharmacology , Protein Engineering , Protein Processing, Post-Translational/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
9.
Water Sci Technol ; 74(2): 491-9, 2016.
Article in English | MEDLINE | ID: mdl-27438255

ABSTRACT

Coagulant dosing control in drinking and wastewater treatment plants (WWTPs) is often limited to flow proportional concepts. The advanced multi-parameter-based dosing control systems have significantly reduced coagulant consumption and improved outlet qualities. Due to the long retention time in separation stages, these models are mostly based on feed-forward (FF) models. This paper demonstrates the improvement of such models with feed-back (FB) concepts with simplifications, making it possible to use even in systems with long separation stages. Full-scale case studies from a drinking water treatment plant and a WWTP are presented. The model qualities were improved by the dosage adjustment of the FB model, ranging from 66% to 197% of the FF model. Hence, the outlet qualities became more stable and coagulant consumption was further reduced in the range of 3.7%-15.5%.


Subject(s)
Coagulants/analysis , Waste Disposal, Fluid/methods , Wastewater/analysis , Water Pollutants, Chemical/chemistry , Water Purification/methods , Coagulants/chemistry , Coagulants/metabolism , Models, Theoretical , Waste Disposal, Fluid/instrumentation , Water Purification/instrumentation
10.
J Proteome Res ; 14(11): 4896-906, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26486890

ABSTRACT

This study demonstrates a direct role of venom protein expression alteration in the evolution of snake venom toxicity. Avian skeletal muscle contractile response to exogenously administered acetylcholine is completely inhibited upon exposure to South Australian and largely preserved following exposure to Queensland eastern brown snake Pseudonaja textilis venom, indicating potent postsynaptic neurotoxicity of the former and lack thereof of the latter venom. Label-free quantitative proteomics reveals extremely large differences in the expression of postsynaptic three-finger α-neurotoxins in these venoms, explaining the difference in the muscle contractile response and suggesting that the type of toxicity induced by venom can be modified by altered expression of venom proteins. Furthermore, the onset of neuromuscular paralysis in the rat phrenic nerve-diaphragm preparation occurs sooner upon exposure to the venom (10 µg/mL) with high expression of α-neurotoxins than the venoms containing predominately presynaptic ß-neurotoxins. The study also finds that the onset of rat plasma coagulation is faster following exposure to the venoms with higher expression of venom prothrombin activator subunits. This is the first quantitative proteomic study that uses extracted ion chromatogram peak areas (MS1 XIC) of distinct homologous tryptic peptides to directly show the differences in the expression of venom proteins.


Subject(s)
Coagulants/chemistry , Elapid Venoms/chemistry , Elapidae/genetics , Neurotoxins/chemistry , Peptide Fragments/chemistry , Serine Endopeptidases/chemistry , Amino Acid Sequence , Animals , Australia , Birds , Coagulants/isolation & purification , Coagulants/metabolism , Coagulants/toxicity , Computational Biology/methods , Diaphragm/drug effects , Diaphragm/physiology , Elapid Venoms/genetics , Elapid Venoms/isolation & purification , Elapid Venoms/metabolism , Elapid Venoms/toxicity , Elapidae/classification , Evolution, Molecular , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gene Expression , Molecular Sequence Data , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Neuromuscular Junction/drug effects , Neuromuscular Junction/physiology , Neurotoxins/genetics , Neurotoxins/isolation & purification , Neurotoxins/toxicity , Peptide Fragments/isolation & purification , Phrenic Nerve/drug effects , Phrenic Nerve/physiology , Rats , Sequence Alignment , Serine Endopeptidases/isolation & purification , Serine Endopeptidases/metabolism , Serine Endopeptidases/toxicity , Species Specificity , Trypsin/chemistry
11.
J Biol Chem ; 289(3): 1732-41, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24275667

ABSTRACT

Tissue factor pathway inhibitor (TFPI) is a Kunitz-type protease inhibitor that inhibits activated factor X (FXa) via a slow-tight binding mechanism and tissue factor-activated FVII (TF-FVIIa) via formation of a quaternary FXa-TFPI-TF-FVIIa complex. Inhibition of TFPI enhances coagulation in hemophilia models. Using a library approach, we selected and subsequently optimized peptides that bind TFPI and block its anticoagulant activity. One peptide (termed compound 3), bound with high affinity to the Kunitz-1 (K1) domain of TFPI (Kd ∼1 nM). We solved the crystal structure of this peptide in complex with the K1 of TFPI at 2.55-Å resolution. The structure of compound 3 can be segmented into a N-terminal anchor; an Ω-shaped loop; an intermediate segment; a tight glycine-loop; and a C-terminal α-helix that is anchored to K1 at its reactive center loop and two-stranded ß-sheet. The contact surface has an overall hydrophobic character with some charged hot spots. In a model system, compound 3 blocked FXa inhibition by TFPI (EC50 = 11 nM) and inhibition of TF-FVIIa-catalyzed FX activation by TFPI (EC50 = 2 nM). The peptide prevented transition from the loose to the tight FXa-TFPI complex, but did not affect formation of the loose FXa-TFPI complex. The K1 domain of TFPI binds and inhibits FVIIa and the K2 domain similarly inhibits FXa. Because compound 3 binds to K1, our data show that K1 is not only important for FVIIa inhibition but also for FXa inhibition, i.e. for the transition of the loose to the tight FXa-TFPI complex. This mode of action translates into normalization of coagulation of hemophilia plasmas. Compound 3 thus bears potential to prevent bleeding in hemophilia patients.


Subject(s)
Coagulants/chemistry , Factor VIIa/chemistry , Factor Xa/chemistry , Lipoproteins/antagonists & inhibitors , Peptides/chemistry , Blood Coagulation/drug effects , Coagulants/chemical synthesis , Coagulants/metabolism , Coagulants/therapeutic use , Factor VIIa/metabolism , Factor Xa/metabolism , Hemophilia A/drug therapy , Hemophilia A/metabolism , Hemorrhage/drug therapy , Hemorrhage/metabolism , Humans , Lipoproteins/chemistry , Lipoproteins/metabolism , Peptides/chemical synthesis , Peptides/metabolism , Peptides/therapeutic use , Protein Structure, Secondary , Protein Structure, Tertiary
12.
J Biol Chem ; 288(11): 7875-7884, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23362270

ABSTRACT

Previous studies have shown that the innate immune stimulant LPS augments mechanical ventilation-induced pulmonary coagulation and inflammation. Whether these effects are mediated by alveolar epithelial cells is unclear. The alveolar epithelium is a key regulator of the innate immune reaction to pathogens and can modulate both intra-alveolar inflammation and coagulation through up-regulation of proinflammatory cytokines and tissue factor (TF), the principal initiator of the extrinsic coagulation pathway. We hypothesized that cyclic mechanical stretch (MS) potentiates LPS-mediated alveolar epithelial cell (MLE-12) expression of the chemokine keratinocyte-derived cytokine (KC) and TF. Contrary to our hypothesis, MS significantly decreased LPS-induced KC and TF mRNA and protein expression. Investigation into potential mechanisms showed that stretch significantly reduced LPS-induced surface expression of TLR4 that was not a result of increased degradation. Decreased cell surface TLR4 expression was concomitant with reduced LPS-mediated NF-κB activation. Immunofluorescence staining showed that cyclic MS markedly altered LPS-induced organization of actin filaments. In contrast to expression, MS significantly increased LPS-induced cell surface TF activity independent of calcium signaling. These findings suggest that cyclic MS of lung epithelial cells down-regulates LPS-mediated inflammatory and procoagulant expression by modulating actin organization and reducing cell surface TLR4 expression and signaling. However, because LPS-induced surface TF activity was enhanced by stretch, these data demonstrate differential pathways regulating TF expression and activity. Ultimately, loss of LPS responsiveness in the epithelium induced by MS could result in increased susceptibility of the lung to bacterial infections in the setting of mechanical ventilation.


Subject(s)
Chemokines/metabolism , Coagulants/metabolism , Epithelial Cells/cytology , Keratinocytes/cytology , Lipopolysaccharides/metabolism , Lung/metabolism , Thromboplastin/biosynthesis , Animals , Cell Line , Cell Survival , Enzyme-Linked Immunosorbent Assay/methods , Inflammation , Mice , NF-kappa B/metabolism , Signal Transduction , Thromboplastin/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptors/metabolism
13.
Blood ; 120(22): 4432-40, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-22995898

ABSTRACT

Ap3A is a platelet-dense granule component released into the extracellular space during the second wave of platelet aggregation on activation. Here, we identify an uncharacterized enzyme, nucleotide pyrophosphatase/phosphodiesterase-4 (NPP4), as a potent hydrolase of Ap3A capable of stimulating platelet aggregation and secretion. We demonstrate that NPP4 is present on the surface of vascular endothelium, where it hydrolyzes Ap3A into AMP and ADP, and Ap4A into AMP and ATP. Platelet aggregation assays with citrated platelet-rich plasma reveal that the primary and secondary waves of aggregation and dense granule release are strongly induced by nanomolar NPP4 in a concentration-dependent manner in the presence of Ap3A, while Ap3A alone initiates a primary wave of aggregation followed by rapid disaggregation. NPP2 and an active site NPP4 mutant, neither of which appreciably hydrolyzes Ap3A, have no effect on platelet aggregation and secretion. Finally, by using ADP receptor blockade we confirm that NPP4 mediates platelet aggregation via release of ADP from Ap3A and activation of ADP receptors. Collectively, these studies define the biologic and enzymatic basis for NPP4 and Ap3A activity in platelet aggregation in vitro and suggest that NPP4 promotes hemostasis in vivo by augmenting ADP-mediated platelet aggregation at the site of vascular injury.


Subject(s)
Blood Coagulation , Endothelium, Vascular/enzymology , Phosphoric Diester Hydrolases/physiology , Pyrophosphatases/physiology , Adenosine Diphosphate/metabolism , Adult , Animals , Blood Coagulation/drug effects , Blood Coagulation/physiology , Coagulants/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Dinucleoside Phosphates/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/injuries , Endothelium, Vascular/metabolism , Fluorescent Antibody Technique , Humans , Hydrolysis , In Vitro Techniques , Insecta , Phosphoric Diester Hydrolases/metabolism , Phosphoric Diester Hydrolases/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Pyrophosphatases/metabolism , Pyrophosphatases/pharmacology , Tissue Distribution
14.
Food Chem ; 450: 138984, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-38642532

ABSTRACT

This study explored the effect of diverse coagulants (glucono-δ-lactone (GDL), gypsum (GYP), microbial transglutaminase (MTGase), and white vinegar (WVG)) on microstructure, quality, and digestion properties of tofu. The four kinds of tofu were significantly different in their structure, composition, and digestibility. Tofu coagulated with MTGase had the highest springiness and cohesiveness while GDL tofu had the highest enthalpy (6.54 J/g). However, the WVG and GYP groups outperformed others in terms of thermodynamic, and digestion properties. The WVG group exhibited the highest nitrogen release (84.3%), water content, denaturation temperature, and the highest free-SH content but the lowest S-S content. Compared to WVG, the GYP group had the highest ash content, hardness, and chewiness. Results demonstrated that the tofu prepared by WVG and GYP show high digestibility. Meanwhile, the former has better thermal properties and the latter has better texture properties.


Subject(s)
Digestion , Soy Foods , Soy Foods/analysis , Glycine max/chemistry , Glycine max/metabolism , Food Handling , Models, Biological , Calcium Sulfate/chemistry , Humans , Coagulants/chemistry , Coagulants/metabolism
15.
Food Funct ; 15(2): 1031-1049, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38193367

ABSTRACT

Orange juice is an important food source of bioactive compounds, mainly the flavanones hesperidin and narirutin. This study aimed to investigate the underlying molecular mechanisms of action of orange juice's health properties by analyzing changes in the plasma proteome of healthy Brazilian volunteers after consuming juices made from 'Bahia' (BOJ-source of flavanones) and 'Cara Cara' (CCOJ-source of flavanones and carotenoids) oranges cultivated in Brazil. We used an untargeted proteomic approach, with a particular emphasis on the juices' effects on blood coagulant activity. We identified 247 differentially expressed proteins, of which 170 significantly increased or decreased after BOJ consumption and 145 after CCOJ. These proteins are involved in 105 processes that can significantly regulate cell adhesion, cell signaling, cell metabolism, inflammation, or others. Bioinformatic analysis evidenced proteins with major cellular regulatory capacity (e.g., FN1 and GAPDH) and predicted transcription factors (TFs) (e.g., SP1 and CEBPA) and miRNAs (e.g., miR-1-3p and miR-615-3p) that could be involved in the regulation of differentially expressed proteins. In-silico docking analyses between flavanone metabolites and TFs evidenced the higher binding capacity of narirutin phase II metabolites with akt1 and p38, interactions that suggest how the expression of genes of differentially expressed proteins were activated or inhibited. Moreover, the study shed light on proteins of coagulation cascade that presented expression modulated by both juices, proposing the modulation of blood coagulant activity as a potential benefit of OJ (mainly CCOJ) consumption. Taken together, this study revealed that BOJ and CCOJ consumption affected plasma proteome in healthy individuals, suggesting potential molecular targets and mechanisms of OJ bioactive compounds in humans.


Subject(s)
Citrus sinensis , Coagulants , Flavanones , MicroRNAs , Humans , Citrus sinensis/chemistry , Brazil , Proteome/analysis , Proteomics , Flavanones/metabolism , Fruit and Vegetable Juices , Fruit/chemistry , MicroRNAs/metabolism , Coagulants/analysis , Coagulants/metabolism
16.
J Proteome Res ; 12(10): 4585-98, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-23998483

ABSTRACT

Snake venom proteomes/peptidomes are highly complex and subject to ontogenetic changes. Individual variation in the venom proteome of juvenile snakes is poorly known. We report the proteomic analysis of venoms from 21 juvenile specimens of Bothrops jararaca of different geographical origins and correlate it with the evaluation of important venom features. Individual venoms showed similar caseinolytic activities; however, their amidolytic activities were significantly different. Rather intriguingly, plasma coagulant activity showed remarkable variability among the venoms but not the prothrombin-activating activity. LC-MS analysis showed significant differences between venoms; however, an interesting finding was the ubiquitous presence of the tripeptide ZKW, an endogenous inhibitor of metalloproteinases. Electrophoretic profiles of proteins submitted to reduction showed significant variability in total proteins, glycoproteins, and in the subproteomes of proteinases. Moreover, identification of differential bands revealed variation in most B. jararaca toxin classes. Profiles of venoms analyzed under nonreducing conditions showed less individual variability and identification of proteins in a conserved band revealed the presence of metalloproteinases and l-amino acid oxidase as common components of these venoms. Taken together, our findings suggest that individual venom proteome variability in B. jararaca exists from a very early animal age and is not a result of ontogenetic and diet changes.


Subject(s)
Bothrops/metabolism , Proteome/metabolism , Reptilian Proteins/metabolism , Venoms/metabolism , Amino Acid Sequence , Animals , Coagulants/chemistry , Coagulants/metabolism , Coagulants/pharmacology , Female , Glycoproteins/chemistry , Glycoproteins/metabolism , Glycoproteins/pharmacology , Humans , Male , Metalloproteases/chemistry , Metalloproteases/metabolism , Metalloproteases/pharmacology , Molecular Sequence Annotation , Oligopeptides/chemistry , Oligopeptides/metabolism , Oligopeptides/pharmacology , Proteolysis , Proteome/chemistry , Proteome/pharmacology , Prothrombin/chemistry , Reptilian Proteins/chemistry , Reptilian Proteins/pharmacology , Venoms/chemistry , Venoms/pharmacology
17.
Biochem Biophys Res Commun ; 437(4): 573-8, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23850687

ABSTRACT

Chronic low-grade inflammation is a key contributor to high-fat diet (HFD)-related diseases, such as type 2 diabetes, non-alcoholic steatohepatitis, and atherosclerosis. The inflammation is characterized by infiltration of inflammatory cells, particularly macrophages, into obese adipose tissue. However, the molecular mechanisms by which a HFD induces low-grade inflammation are poorly understood. Here, we show that histone H3, a major protein component of chromatin, is released into the extracellular space when mice are fed a HFD or macrophages are stimulated with the saturated fatty acid palmitate. In a murine macrophage cell line, RAW 264.7, palmitate activated reactive oxygen species (ROS) production and JNK signaling. Inhibitors of these pathways dampened palmitate-induced histone H3 release, suggesting that the extracellular release of histone H3 was mediated, in part, through ROS and JNK signaling. Extracellular histone activated endothelial cells to express the adhesion molecules ICAM-1 and VCAM-1 and the procoagulant molecule tissue factor, which are known to contribute to inflammatory cell recruitment and thrombosis. These results suggest the possible contribution of extracellular histone to the pathogenesis of HFD-induced inflammation and thrombosis.


Subject(s)
Diet, High-Fat , Histones/metabolism , Inflammation/metabolism , Palmitic Acid/metabolism , Thrombosis/metabolism , Adipose Tissue/metabolism , Animals , Cell Adhesion , Cell Line , Cell Survival , Coagulants/metabolism , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Adhesion Molecule-1/metabolism , MAP Kinase Kinase 4/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
18.
Eur Respir J ; 42(1): 272-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23258792

ABSTRACT

Pulmonary hypertension (PH) is a fatal disease with no treatment options, characterised by elevated pulmonary vascular resistanzce and secondary right ventricular failure. The aetiology of pulmonary arterial hypertension is multiple and its pathogenesis is complex. Although the exact role of cellular microparticles remains partially understood, there is increasing evidence to suggest an active role for microparticles in PH pathophysiology. Patients with PH exhibited higher circulating levels of microparticles compared to control subjects and in vitro or in vivo generated microparticles can induce endothelial dysfunction, interfere with coagulation pathways or modulate inflammatory phenomenon. Whether or not these new conveyors of biological information contribute to the acquisition and/or maintenance of the altered endothelial phenotype is unexplored in PH and requires further study.


Subject(s)
Cell-Derived Microparticles/chemistry , Hypertension, Pulmonary/blood , Biomarkers/metabolism , Cardiovascular Diseases/metabolism , Coagulants/metabolism , Disease Progression , Familial Primary Pulmonary Hypertension , Humans , Inflammation , Phenotype , Prognosis , Vascular Resistance
19.
PLoS Pathog ; 7(10): e1002307, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22028651

ABSTRACT

Staphylococcus aureus infection is a frequent cause of sepsis in humans, a disease associated with high mortality and without specific intervention. When suspended in human or animal plasma, staphylococci are known to agglutinate, however the bacterial factors responsible for agglutination and their possible contribution to disease pathogenesis have not yet been revealed. Using a mouse model for S. aureus sepsis, we report here that staphylococcal agglutination in blood was associated with a lethal outcome of this disease. Three secreted products of staphylococci--coagulase (Coa), von Willebrand factor binding protein (vWbp) and clumping factor (ClfA)--were required for agglutination. Coa and vWbp activate prothrombin to cleave fibrinogen, whereas ClfA allowed staphylococci to associate with the resulting fibrin cables. All three virulence genes promoted the formation of thromboembolic lesions in heart tissues. S. aureus agglutination could be disrupted and the lethal outcome of sepsis could be prevented by combining dabigatran-etexilate treatment, which blocked Coa and vWbp activity, with antibodies specific for ClfA. Together these results suggest that the combined administration of direct thrombin inhibitors and ClfA-antibodies that block S. aureus agglutination with fibrin may be useful for the prevention of staphylococcal sepsis in humans.


Subject(s)
Agglutination/physiology , Sepsis/prevention & control , Staphylococcal Infections/microbiology , Staphylococcus aureus/metabolism , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antithrombins/pharmacology , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Coagulants/metabolism , Coagulase/immunology , Coagulase/metabolism , Disease Models, Animal , Heart/microbiology , Host-Pathogen Interactions , Humans , Immunization, Passive , Longevity/drug effects , Mice , Mice, Inbred BALB C , Myocardium/pathology , Protein Binding , Sepsis/immunology , Sepsis/microbiology , Staphylococcal Infections/immunology , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/ultrastructure , von Willebrand Factor/immunology , von Willebrand Factor/metabolism
20.
Blood ; 117(11): 3172-80, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21248061

ABSTRACT

On vascular damage, coagulation is initiated by extravascular tissue factor (TF). Intravascular TF, which is present on circulating cell-derived vesicles, is noncoagulant under physiologic conditions but prothrombotic under pathologic conditions. Human saliva triggers coagulation, but the mechanism and physiologic relevance are unknown. Because saliva is known to contain TF, we hypothesized that this TF may also be associated with cell-derived vesicles to facilitate coagulation when saliva directly contacts blood. The saliva-induced shortening of the clotting time of autologous plasma and whole blood from healthy subjects (n = 10) proved TF-dependent. This TF was associated with various types of cell-derived vesicles, including microparticles and exosomes. The physiologic function was shown by adding saliva to human pericardial wound blood collected from patients undergoing cardiac surgery. Addition of saliva shortened the clotting time from 300 ± 96 to 186 ± 24 seconds (P = .03). Our results show that saliva triggers coagulation, thereby reducing blood loss and the risk of pathogens entering the blood. We postulate that our reflex to lick a wound may be a mechanism to enable TF-exposing vesicles, present in saliva, to aid in the coagulation process and thus protect the organism from entering pathogens. This unique compartmentalization may be highly conserved because also animals lick their wounds.


Subject(s)
Cell-Derived Microparticles/metabolism , Coagulants/metabolism , Saliva/metabolism , Thromboplastin/metabolism , Adult , Blood Coagulation , Cell-Derived Microparticles/ultrastructure , Exosomes/metabolism , Exosomes/ultrastructure , Factor VII/metabolism , Female , Fibrinogen/metabolism , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL