Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters











Publication year range
1.
Blood ; 136(15): 1773-1782, 2020 10 08.
Article in English | MEDLINE | ID: mdl-32542378

ABSTRACT

G protein-coupled receptors are critical mediators of platelet activation whose signaling can be modulated by members of the regulator of G protein signaling (RGS) family. The 2 most abundant RGS proteins in human and mouse platelets are RGS10 and RGS18. While each has been studied individually, critical questions remain about the overall impact of this mode of regulation in platelets. Here, we report that mice missing both proteins show reduced platelet survival and a 40% decrease in platelet count that can be partially reversed with aspirin and a P2Y12 antagonist. Their platelets have increased basal (TREM)-like transcript-1 expression, a leftward shift in the dose/response for a thrombin receptor-activating peptide, an increased maximum response to adenosine 5'-diphosphate and TxA2, and a greatly exaggerated response to penetrating injuries in vivo. Neither of the individual knockouts displays this constellation of findings. RGS10-/- platelets have an enhanced response to agonists in vitro, but platelet count and survival are normal. RGS18-/- mice have a 15% reduction in platelet count that is not affected by antiplatelet agents, nearly normal responses to platelet agonists, and normal platelet survival. Megakaryocyte number and ploidy are normal in all 3 mouse lines, but platelet recovery from severe acute thrombocytopenia is slower in RGS18-/- and RGS10-/-18-/- mice. Collectively, these results show that RGS10 and RGS18 have complementary roles in platelets. Removing both at the same time discloses the extent to which this regulatory mechanism normally controls platelet reactivity in vivo, modulates the hemostatic response to injury, promotes platelet production, and prolongs platelet survival.


Subject(s)
Blood Platelets/metabolism , Platelet Activation/genetics , RGS Proteins/genetics , Thrombopoiesis/genetics , Animals , Blood Platelets/drug effects , Cell Survival/genetics , Mice , Mice, Knockout , Phosphorylation , Platelet Activating Factor/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Count , RGS Proteins/metabolism , Thrombopoiesis/drug effects
2.
Vet Immunol Immunopathol ; 221: 110011, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32007672

ABSTRACT

Myelosupression resulting from chemotherapy has been widely described in veterinary medicine; however, there is limited information relating to alterations in neutrophil function after chemotherapy in dogs with cancer. The aim of this study was to determine the non-proliferative effects of vincristine, carboplatin, and cisplatin on canine neutrophils by evaluating activation of oxidative and non-oxidative responses. Neutrophils were isolated from venous blood. Levels of reactive oxygen species (ROS) and metalloproteinase 9 (MMP-9) were measured in vitro during neutrophil exposure to these chemotherapeutic agents for 15 min followed by stimulation with platelet activating factor (PAF). ROS production was detected via luminescence, and MMP- 9 liberation was determined by zymography. The chemotherapeutic agents caused an increase in PAF-induced ROS production, but no change in the non-oxidative response was observed. These results suggest that these chemotherapeutic agents may act as priming agents by increasing the oxidative response. These effects could be beneficial for dogs with cancer by supporting their immune systems; however, excessive ROS liberation has been associated with inflammation, neutrophil-mediated cell injury, carcinogenesis, and metastasis. Clinical studies are necessary to evaluate the significance of these findings.


Subject(s)
Carboplatin/pharmacology , Cisplatin/pharmacology , Neutrophils/drug effects , Neutrophils/immunology , Respiratory Burst/drug effects , Vincristine/pharmacology , Animals , Antineoplastic Agents/pharmacology , Dogs , Matrix Metalloproteinase 9/analysis , Platelet Activating Factor/pharmacology , Reactive Oxygen Species/analysis
3.
Biomed Res Int ; 2017: 4315832, 2017.
Article in English | MEDLINE | ID: mdl-29226136

ABSTRACT

This work reports the purification and functional characterization of BmooPAi, a platelet-aggregation-inhibiting factor from Bothrops moojeni snake venom. The toxin was purified by a combination of three chromatographic steps (ion-exchange on DEAE-Sephacel, molecular exclusion on Sephadex G-75, and affinity chromatography on HiTrap™ Heparin HP). BmooPAi was found to be a single-chain protein with an apparent molecular mass of 32 kDa on 14% SDS-PAGE, under reducing conditions. Sequencing of BmooPAi by Edman degradation revealed the amino acid sequence LGPDIVPPNELLEVM. The toxin was devoid of proteolytic, haemorrhagic, defibrinating, or coagulant activities and induced no significant oedema or hyperalgesia. BmooPAi showed a rather specific inhibitory effect on ristocetin-induced platelet aggregation in human platelet-rich plasma, whereas it had little or no effect on platelet aggregation induced by collagen and adenosine diphosphate. The results presented in this work suggest that BmooPAi is a toxin comprised of disintegrin-like and cysteine-rich domains, originating from autolysis/proteolysis of PIII SVMPs from B. moojeni snake venom. This toxin may be of medical interest because it is a platelet aggregation inhibitor, which could potentially be developed as a novel therapeutic agent to prevent and/or treat patients with thrombotic disorders.


Subject(s)
Bothrops/metabolism , Platelet Activating Factor/isolation & purification , Platelet Activating Factor/pharmacology , Platelet Aggregation Inhibitors/isolation & purification , Platelet Aggregation Inhibitors/pharmacology , Snake Venoms/metabolism , Adenosine Diphosphate/metabolism , Amino Acid Sequence , Animals , Blood Platelets/drug effects , Hemorrhage/drug therapy , Humans , Male , Mice , Molecular Weight , Platelet Aggregation/drug effects , Proteolysis/drug effects , Rats , Rats, Wistar
4.
Vet Immunol Immunopathol ; 191: 68-73, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28895869

ABSTRACT

Cytoplasmic pH homeostasis is required for an appropriate response in polymorphonuclear neutrophils (PMNs). In these cells, chemotaxis and reactive oxygen species (ROS) production are reduced by the use of Na+/H+ exchanger (NHE-1) inhibitors, but these results are mainly obtained using amiloride, a non-selective NHE-1 inhibitor. In bovine PMNs, the role of NHE-1 in functional responses has not been confirmed yet. The aim of this study was to determine the role of NHE-1 using amiloride and zoniporide in pH regulation, ROS production, matrix metalloproteinase 9 (MMP-9) release and calcium flux in bovine PMNs induced by the platelet activation factor (PAF), additionally we evaluated the presence of NHE-1 and NHE-2 mRNA Our data show the presence only of NHE-1 but not NHE-2 in bovine PMNs. Amiloride or zoniporide inhibited the intracellular alkalization induced by PAF without affecting calcium flux. Amiloride diminished ROS production and MMP-9 release, while zoniporide enhanced ROS production without change the MMP-9 release induced by PAF. Our work led us to conclude that changes in intracellular pH induced by PAF are regulated by NHE-1 in bovine neutrophils, but the effects of amiloride on ROS production and MMP-9 release induced by PAF are not NHE-1 dependent.


Subject(s)
Amiloride/pharmacology , Matrix Metalloproteinase 9/metabolism , Neutrophils/drug effects , Platelet Activating Factor/antagonists & inhibitors , Respiratory Burst/drug effects , Sodium-Hydrogen Exchanger 1/antagonists & inhibitors , Animals , Calcium/metabolism , Cattle , Female , Guanidines/pharmacology , Neutrophils/enzymology , Neutrophils/metabolism , Platelet Activating Factor/pharmacology , Polymerase Chain Reaction/veterinary , Pyrazoles/pharmacology , Reactive Oxygen Species/metabolism , Sodium-Hydrogen Exchanger 1/metabolism
5.
Pathog Dis ; 75(7)2017 09 29.
Article in English | MEDLINE | ID: mdl-28830073

ABSTRACT

Platelet-activating factor (PAF) is produced by macrophages during inflammation and infections. We evaluated whether PAF is able to modulate the infection of human macrophages by Leishmania braziliensis, the main Leishmania sp. in Brazil. Monocyte-derived macrophages were incubated with promastigote forms in absence or presence of exogenous PAF. We observed that the treatment of macrophages with low concentrations of PAF prior to infection increased the phagocytosis of L. braziliensis. More importantly, exogenous PAF reduced the parasitism when it was added before, during or after infection. In addition, treatment with a PAF antagonist (PCA 4248) resulted in a significant increase of macrophage infection in a concentration-dependent manner, suggesting that endogenous PAF is important to control L. braziliensis infection. Mechanistically, while exogenous PAF increased production of reactive oxygen species (ROS) treatment with PCA 4248 reduced oxidative burst during L. braziliensis infection. The microbicidal effects of exogenous PAF were abolished when macrophages were treated with apocynin, an NADPH oxidase inhibitor. The data show that PAF promotes the production of ROS induced by L. braziliensis, suggesting that this lipid mediator may be relevant to control L. braziliensis infection in human macrophages.


Subject(s)
Leishmania braziliensis/drug effects , Macrophages/drug effects , Phagocytosis/drug effects , Platelet Activating Factor/pharmacology , Reactive Oxygen Species/agonists , Acetophenones/pharmacology , Dihydropyridines/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression , Humans , Leishmania braziliensis/growth & development , Macrophages/immunology , Macrophages/parasitology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Platelet Activating Factor/antagonists & inhibitors , Primary Cell Culture , Reactive Oxygen Species/metabolism , Respiratory Burst/drug effects
6.
Sci Rep ; 6: 32092, 2016 08 24.
Article in English | MEDLINE | ID: mdl-27554194

ABSTRACT

Platelet-activating factor receptor (PAFR) is a G protein-coupled receptor (GPCR) implicated in many diseases. Toll-like receptors (TLRs) play a critical role in shaping innate and adaptive immune responses. In this study, we investigated whether PAFR signaling changes the macrophages responsiveness to agonists of TLR2 (Pam3Cys), TLR4 (LPS), and TLR3 agonist Poly(I:C). Exogenous PAF inhibited the production of pro-inflammatory cytokines (IL-12p40, IL-6, and TNF-α) and increased anti-inflammatory IL-10 in macrophages challenged with Pam3Cys and LPS, but not with Poly (I:C). PAF did not affect mRNA expression of MyD88, suggesting that PAF acts downstream the adaptor. PAF inhibited LPS-induced phosphorylation of NF-κB p65 and increased NF-κB p105 phosphorylation, which is processed in the proteasome to generate p50 subunit. The PAF potentiation of IL-10 production was dependent on proteasome processing but independent of NF-κB transactivation domain. Inhibition of p50 abolished the PAF-induced IL-10 production. These findings indicate that the impaired transcriptional activity of the p65 subunit and the enhanced p105 phosphorylation induced by PAF are responsible for down regulation of pro-inflammatory cytokines and up regulation of IL-10, respectively, in LPS-challenged macrophages. Together, our data unveil a heretofore unrecognized role for PAFR in modulating activation of NF-κB in macrophages.


Subject(s)
Inflammation/metabolism , NF-kappa B p50 Subunit/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Toll-Like Receptors/metabolism , Transcription Factor RelA/metabolism , Animals , Cytokines/metabolism , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Lipoproteins/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Platelet Activating Factor/metabolism , Platelet Activating Factor/pharmacology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
7.
Vet Immunol Immunopathol ; 176: 18-27, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27288853

ABSTRACT

Increased short-chain fatty acid (SCFA) production is associated with subacute ruminal acidosis (SARA) and activation of inflammatory processes. In humans and rodents, SCFAs modulate inflammatory responses in the gut via free fatty acid receptor 2 (FFA2). In bovines, butyric acid is one of the most potent FFA2 agonists. Its expression in bovine neutrophils has recently been demonstrated, suggesting a role in innate immune response in cattle. This study aimed to evaluate if butyric acid modulates oxidative and non-oxidative functions or if it can potentiate other inflammatory mediators in bovine neutrophils. Our results showed that butyric acid can activate bovine neutrophils, inducing calcium (Ca(2+)) influx and mitogen-activated protein kinase (MAPK) phosphorylation, two second messengers involved in FFA2 activation. Ca(2+) influx induced by butyric acid was dependent on the extracellular and intracellular Ca(2+) source and phospholipase C (PLC) activation. Butyric acid alone had no significant effect on reactive oxygen species (ROS) production and chemotaxis; however, a priming effect on platelet-activating factor (PAF), a potent inflammatory mediator, was observed. Butyric acid increased CD63 expression and induced the release of neutrophil granule markers matrix metalloproteinase-9 (MMP-9) and lactoferrin. Finally, we observed that butyric acid induced neutrophil extracellular trap (NET) formation without affecting cellular viability. These findings suggest that butyric acid, a component of the ruminal fermentative process, can modulate the innate immune response of ruminants.


Subject(s)
Butyric Acid/pharmacology , Cattle/immunology , Neutrophils/drug effects , Platelet Activating Factor/pharmacology , Animals , Calcium/metabolism , Chemotaxis, Leukocyte , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Traps/drug effects , Fatty Acids, Nonesterified/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Vet Sci ; 15(2): 217-24, 2014.
Article in English | MEDLINE | ID: mdl-25013355

ABSTRACT

Reactive oxygen species (ROS) production is one of the main mechanisms used to kill microbes during innate immune response. D-lactic acid, which is augmented during acute ruminal acidosis, reduces platelet activating factor (PAF)-induced ROS production and L-selectin shedding in bovine neutrophils in vitro. This study was conducted to investigate whether acute ruminal acidosis induced by acute oligofructose overload in heifers interferes with ROS production and L-selectin shedding in blood neutrophils. Blood neutrophils and plasma were obtained by jugular venipuncture, while ruminal samples were collected using rumenocentesis. Lactic acid from plasma and ruminal samples was measured by HPLC. PAF-induced ROS production and L-selectin shedding were measured in vitro in bovine neutrophils by a luminol chemiluminescence assay and flow cytometry, respectively. A significant increase in ruminal and plasma lactic acid was recorded in these animals. Specifically, a decrease in PAF-induced ROS production was observed 8 h after oligofructose overload, and this was sustained until 48 h post oligofructose overload. A reduction in PAF-induced L-selectin shedding was observed at 16 h and 32 h post oligofructose overload. Overall, the results indicated that neutrophil PAF responses were altered in heifers with ruminal acidosis, suggesting a potential dysfunction of the innate immune response.


Subject(s)
Acidosis/veterinary , Cattle Diseases/immunology , Immunity, Innate , Neutrophils/drug effects , Oligosaccharides/pharmacology , Platelet Activating Factor/pharmacology , Acidosis/chemically induced , Acidosis/immunology , Animals , Blood , Cattle , Cattle Diseases/chemically induced , Female , Flow Cytometry/veterinary , L-Selectin/metabolism , Oligosaccharides/toxicity , Reactive Oxygen Species/metabolism , Rumen
9.
Cell Microbiol ; 16(8): 1244-54, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24612488

ABSTRACT

ExoU is an important virulence factor in acute Pseudomonas aeruginosa infections. Here, we unveiled the mechanisms of ExoU-driven NF-κB activation by using human airway cells and mice infected with P. aeruginosa strains. Several approaches showed that PAFR was crucially implicated in the activation of the canonical NF-κB pathway. Confocal microscopy of lungs from infected mice revealed that PAFR-dependent NF-κB activation occurred mainly in respiratory epithelial cells, and reduced p65 nuclear translocation was detected in mice PAFR-/- or treated with the PAFR antagonist WEB 2086. Several evidences showed that ExoU-induced NF-κB activation regulated PAFR expression. First, ExoU increased p65 occupation of PAFR promoter, as assessed by ChIP. Second, luciferase assays in cultures transfected with different plasmid constructs revealed that ExoU promoted p65 binding to the three κB sites in PAFR promoter. Third, treatment of cell cultures with the NF-κB inhibitor Bay 11-7082, or transfection with IκBα negative-dominant, significantly decreased PAFR mRNA. Finally, reduction in PAFR expression was observed in mice treated with Bay 11-7082 or WEB 2086 prior to infection. Together, our data demonstrate that ExoU activates NF-κB by PAFR signalling, which in turns enhances PAFR expression, highlighting an important mechanism of amplification of response to this P. aeruginosa toxin.


Subject(s)
Bacterial Proteins/metabolism , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/genetics , Pseudomonas aeruginosa/pathogenicity , Receptors, G-Protein-Coupled/genetics , Transcription Factor RelA/metabolism , Animals , Azepines/pharmacology , Bacterial Toxins/metabolism , Cell Line , Enzyme Activation , Female , Gene Expression Regulation , Humans , Lung/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Platelet Activating Factor/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/biosynthesis , Promoter Regions, Genetic , Protein Binding , Pseudomonas Infections/pathology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/biosynthesis , Signal Transduction/genetics , Triazoles/pharmacology
10.
Life Sci ; 93(9-11): 416-22, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23871985

ABSTRACT

AIMS: The relationship between the activity of eosinophils and platelets has been observed in recent decades by many scientists. These observations include increased numbers of eosinophils associated with platelet disorders, including changes in the coagulation cascade and platelet aggregation. Based on these observations, the interaction between eosinophils and platelets in platelet aggregation was analyze. MAIN METHODS: Human platelets were incubated with eosinophil cytosolic fraction, promyelocytic human HL-60 clone 15 cell lineage, and eosinophil cationic protein (ECP). Platelet rich plasma (PRP) aggregation was induced by adenosine diphosphate, platelet activating factor, arachidonic acid, and collagen, and washed platelets (WP) were activated by thrombin. KEY FINDINGS: Aggregation induced by all agonists was dose dependently inhibited by eosinophil cytosolic fraction. This inhibition was only partially reversed by previous incubation of the eosinophils with l-Nitro-Arginine-Methyl-Ester (l-NAME). Previous incubation with indomethacin did not prevent the cytosolic fraction induced inhibition. The separation of eosinophil cytosolic fraction by gel filtration on Sephadex G-75 showed that the inhibitory activity was concentrated in the lower molecular weight fraction. HL-60 clone 15 cells differentiated into eosinophils for 5 and 7 day were able to inhibit platelet aggregation. The ECP protein inhibited the platelet aggregation on PRP and WP. This inhibition was more evident in WP, and the citotoxicity MTT assay proved the viability of tested platelets, showing that the observed inhibition by the ECP protein does not occur simply by cell death. SIGNIFICANCE: Our results indicate that eosinophils play a fundamental role in platelet aggregation inhibition.


Subject(s)
Blood Platelets/metabolism , Cytosol/metabolism , Eosinophils/metabolism , Platelet Aggregation/physiology , Adenosine Diphosphate/pharmacology , Animals , Arachidonic Acid/pharmacology , Blood Platelets/drug effects , Chromatography, Gel/methods , Collagen/pharmacology , Dextrans , HL-60 Cells , Humans , Indomethacin/pharmacology , Male , Molecular Weight , NG-Nitroarginine Methyl Ester/pharmacology , Platelet Activating Factor/pharmacology , Platelet Aggregation/drug effects , Rats , Rats, Wistar , Thrombin/pharmacology , Time Factors
11.
Mini Rev Med Chem ; 13(7): 997-1004, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23544467

ABSTRACT

Chagas Disease (CD), a tropical parasitic disease caused by the flagellate protozoan Trypanosoma cruzi, accounts for the highest burden of parasitic diseases in the Western hemisphere. Current drug treatments for CD are highly toxic and often ineffective, particularly for the chronic stage of the disease, a fact that clearly emphasizes the importance of identification/validation of molecular targets for the development of new drugs to treat the disease. Here, we review in details the evidences that suggest the existence of specific receptors for platelet-activating factor (PAF) in T. cruzi, the role of PAF on the control of parasite differentiation and the potential of exploring these putative receptors as new targets for the chemotherapy of CD.


Subject(s)
Acrylates/pharmacology , Chagas Disease/drug therapy , Chagas Disease/parasitology , Platelet Activating Factor/analogs & derivatives , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Trypanosoma cruzi/drug effects , Acrylates/chemistry , Animals , Humans , Models, Biological , Molecular Structure , Platelet Activating Factor/chemistry , Platelet Activating Factor/pharmacology , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Trypanosoma cruzi/metabolism
12.
Vet Immunol Immunopathol ; 145(1-2): 540-5, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22226550

ABSTRACT

2-Aminoethoxydiphenyl borate (2-APB) interferes with the Ca(2+) influx and reduces the ROS production, gelatinase secretion and CD11b expression in bovine neutrophils. Moreover, it has been suggested that inhibition of the Ca(2+) channel involved in the store operated Ca(2+) entry (SOCE) is a potential target for the development of new anti-inflammatory drugs in cattle, however it is unknown whether 2-APB affects neutrophil functions associated with the innate immune response. This study describes the effect of 2-APB, a putative SOCE inhibitor, on alkaline phosphatase activity a marker of secretory vesicles, CD63 a marker for azurophil granules, F-actin polymerization and in vitro chemotaxis in bovine neutrophils stimulated with platelet-activating factor (PAF). Also, we evaluated the effect of 2-APB in the phagocytic activity against Escherichia coli and Staphylococcus aureus bioparticles. We observed that doses of 2-APB ≥10 µM significantly reduced alkaline phosphatase activity and in vitro chemotaxis, whereas concentrations of 2-APB ≥50 µM reduced CD63 expression and F-actin polymerization. Finally, we observed that 2-APB did not affect the phagocytic activity in neutrophils incubated with E. coli and S. aureus bioparticles. We concluded that inhibition of Ca(2+) influx could be a useful strategy to reduce inflammatory process in cattle.


Subject(s)
Actins/drug effects , Alkaline Phosphatase/antagonists & inhibitors , Boron Compounds/pharmacology , Calcium Channel Blockers/pharmacology , Chemotaxis/drug effects , Neutrophils/drug effects , Phagocytosis/drug effects , Tetraspanin 30/biosynthesis , Actins/metabolism , Alkaline Phosphatase/metabolism , Animals , Calcium/metabolism , Cattle , Chemotaxis/physiology , Dose-Response Relationship, Drug , Female , Flow Cytometry/veterinary , Neutrophils/enzymology , Neutrophils/metabolism , Neutrophils/physiology , Phagocytosis/physiology , Platelet Activating Factor/pharmacology , Polymerization/drug effects
13.
Res Vet Sci ; 93(1): 393-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21820686

ABSTRACT

Phagocytosis exerted by alveolar macrophages and neutrophils is crucial in the clearance of exogenous particles deposited in the airways. Therefore, substances that activate these phagocytes in the airways can exert important effects on the particle clearance rate. PAF, particularly, was proved to be a potent activator of several immune cells and was shown to be present in the equine lower airways in specific conditions, such as after exercise. The present study aimed to investigate if PAF is able to increase the phagocytic capacity and the production of superoxide anion in equine alveolar macrophage and blood neutrophils. The results show that PAF increased these parameters in both phagocytes even in concentrations as low as 0.1 and 1.0 nM. On that ground, the present work suggests that PAF is involved in the process of particle clearance in equine lower airways.


Subject(s)
Macrophages, Alveolar/drug effects , Neutrophils/drug effects , Phagocytosis/drug effects , Platelet Activating Factor/pharmacology , Superoxides/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Horses , Macrophages, Alveolar/chemistry , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/physiology , Neutrophils/chemistry , Neutrophils/metabolism , Neutrophils/physiology , Superoxides/analysis
14.
Vet Immunol Immunopathol ; 144(1-2): 68-78, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21816488

ABSTRACT

D-lactic acidosis occurs in ruminants, such as cattle, with acute ruminal acidosis caused by ingestion of excessive amounts of highly fermentable carbohydrates. Affected animals show clinical signs similar to those of septic shock, as well as acute laminitis and liver abscesses. It has been proposed that the inflammatory response and susceptibility to infection could both be caused by the inhibition of phagocytic mechanisms. To determine the effects of d-lactic acid on bovine neutrophil functions, we pretreated cells with different concentrations of D-lactic acid and measured intracellular pH using 2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein acetoxymethyl ester (BCECF-AM) and calcium flux using FLUO-3 AM-loaded neutrophils. Reactive oxygen species (ROS) production was measured using a luminol chemiluminescence assay, and MMP-9/gelatinase-B granule release was measured by zymography. CD11b and CD62L/l-selectin expression, changes in cell shape, superoxide anion production, phagocytosis of Escherichia coli-Texas red bioparticles, and apoptosis were all measured using flow cytometry. Our results demonstrated that D-lactic acid reduced ROS production, CD11b upregulation and MMP-9 release in bovine neutrophils treated with 100 nM platelet-activating factor (PAF). D-lactic acid induced MMP-9 release and, at higher concentrations, upregulated CD11b expression, decrease L-selectin expression, and induces late apoptosis. We concluded that D-lactic acid can interfere with neutrophil functions induced by PAF, leading to reduced innate immune responses during bacterial infections. Moreover, the increase of MMP-9 release and CD11b expression induced by 10mM D-lactic acid could promote an nonspecific neutrophil-dependent inflammatory reaction in cattle with acute ruminal acidosis.


Subject(s)
Lactic Acid/pharmacology , Neutrophils/drug effects , Platelet Activating Factor/antagonists & inhibitors , Acidosis, Lactic/metabolism , Acidosis, Lactic/veterinary , Animals , CD11b Antigen/analysis , Calcium/analysis , Cattle , Cattle Diseases/metabolism , Female , Flow Cytometry/veterinary , Hydrogen-Ion Concentration , L-Selectin/analysis , Luminescent Measurements/veterinary , Matrix Metalloproteinase 9/metabolism , Neutrophils/chemistry , Phagocytosis/drug effects , Platelet Activating Factor/pharmacology , Reactive Oxygen Species/analysis
15.
Nat Prod Commun ; 5(3): 383-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20420312

ABSTRACT

The in vitro anti-inflammatory effect of eight aporphine alkaloids isolated from the leaves of two Lauraceae plants (Pleurothyrium cinereum and Ocotea macrophylla) was evaluated through inhibition of two isozymes of cyclooxygenase (COX-1 and COX-2), 5-lipoxygenase (5-LOX), and platelet aggregation induced by PAF, AA and ADP. All alkaloids exhibited inhibitory activities against COX-2 (IC50 25.9-116 microM range) and PAF- and AA-induced platelet aggregation, while only four and three of them were good COX-1 and 5-LOX inhibitors, respectively. (+)-N-acetyl-nornantenine 6 was the most potent COX-2, 5-LOX, AA and PAF inhibitor.


Subject(s)
Alkaloids/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Aporphines/chemistry , Lauraceae/chemistry , Adenosine Diphosphate/pharmacology , Alkaloids/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aporphines/pharmacology , Arachidonic Acid/pharmacology , Circular Dichroism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/pharmacology , Female , In Vitro Techniques , Lipoxygenase Inhibitors , Magnetic Resonance Spectroscopy , Plant Extracts/chemistry , Plant Leaves/chemistry , Platelet Activating Factor/pharmacology , Platelet Aggregation/drug effects , Rabbits
16.
Biochem Pharmacol ; 77(7): 1223-35, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19283893

ABSTRACT

Platelet-activating factor (PAF) is an inflammatory mediator widely known to exert relevant pathophysiological functions. However, the relevance of PAF in nociception has received much less attention. Herein, we have investigated the mechanisms underlying PAF-induced spontaneous nociception and mechanical hypersensitivity in the rat paw. PAF injection (1- 30 nmol/paw) resulted in a dose-related overt nociception, whilst only the dose of 10 nmol/ paw produced a significant and time-related mechanical hypersensitivity. Local coinjection of PAF antagonist WEB2086 significantly inhibited both spontaneous nociception and mechanical hypersensitivity. Moreover, the coinjection of the natural IL-1beta receptor antagonist (IRA) notably prevented both PAF-induced nociceptive responses, whilst these responses were not altered by anti-TNFalpha coinjection. Interestingly, pretreatment with the ultrapotent vaniloid agonist resiniferotoxin, coinjection of the TRPV1 receptor antagonist SB366791, or mast cell depletion with compound 48/80 markedly prevented PAF-induced spontaneous nociception. Conversely, PAF-elicited mechanical hypersensitivity was strikingly susceptible to distinct antineutrophil-related strategies, namely the antineutrophil antibody, the selectin blocker fucoidin, the chemokine CXCR2 receptor antagonist SB225002, and the C5a receptor antibody anti-CD88. Notably, the same antineutrophil migration strategies significantly prevented the increase of myeloperoxidase activity induced by PAF. The mechanical hypersensitivity caused by PAF was also prevented by the cyclooxygenase inhibitors indomethacin or celecoxib, and by the selective beta(1) adrenergic receptor antagonist atenolol. Collectively, the present results provide consistent evidence indicating that distinct mechanisms are involved in the spontaneous nociception and mechanical hypersensitivity caused by PAF. They also support the concept that selective PAF receptor antagonists might constitute interesting targets for the development of new analgesic drugs.


Subject(s)
Forelimb/metabolism , Pain Measurement/methods , Pain/chemically induced , Pain/metabolism , Platelet Activating Factor/toxicity , Animals , Azepines/pharmacology , Azepines/therapeutic use , Forelimb/drug effects , Forelimb/pathology , Male , Pain/prevention & control , Pain Measurement/drug effects , Platelet Activating Factor/pharmacology , Platelet Membrane Glycoproteins/agonists , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/metabolism , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Triazoles/pharmacology , Triazoles/therapeutic use
17.
J Insect Physiol ; 55(6): 532-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19232405

ABSTRACT

The effects of physalin B (a natural secosteroidal chemical from Physalis angulata, Solanaceae) on phagocytosis and microaggregation by hemocytes of 5th-instar larvae of Rhodnius prolixus were investigated. In this insect, hemocyte phagocytosis and microaggregation are known to be induced by the platelet-activating factor (PAF) or arachidonic acid (AA) and regulated by phospholipase A(2) (PLA(2)) and PAF-acetyl hydrolase (PAF-AH) activities. Phagocytic activity and formation of hemocyte microaggregates by Rhodnius hemocytes were strongly blocked by oral treatment of this insect with physalin B (1mug/mL of blood meal). The inhibition induced by physalin B was reversed for both phagocytosis and microaggregation by exogenous arachidonic acid (10microg/insect) or PAF (1microg/insect) applied by hemocelic injection. Following treatment with physalin B there were no significant alterations in PLA(2) activities, but a significant enhancement of PAF-AH was observed. These results show that physalin B inhibits hemocytic activity by depressing insect PAF analogous (iPAF) levels in hemolymph and confirm the role of PAF-AH in the cellular immune reactions in R. prolixus.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Hemocytes/immunology , Insect Proteins/metabolism , Phagocytosis/drug effects , Rhodnius/enzymology , Secosteroids/pharmacology , Animals , Arachidonic Acid/pharmacology , Cell Aggregation/drug effects , Enzyme Activation/drug effects , Hemocytes/drug effects , Hemocytes/enzymology , Hemocytes/microbiology , Platelet Activating Factor/pharmacology , Rhodnius/drug effects , Rhodnius/immunology , Rhodnius/microbiology , Saccharomyces cerevisiae/physiology
18.
J Insect Physiol ; 54(2): 344-50, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18036540

ABSTRACT

The hemocytes phagocytosis in response to microorganisms may play an important role in the cellular immune responses of insects. Here, we have evaluated the effects of the platelet-activating factor (PAF) and eicosanoids in the phagocytosis of hemocyte monolayers of Rhodnius prolixus to the yeast Saccharomyces cerevisiae. Experiments showed that the phagocytosis of yeast cells by Rhodnius hemocytes is very efficient in both controls and cells treated with PAF and arachidonic acid. Phagocytosis of yeast particles is significantly blocked when the specific phopholipase A(2) inhibitor, dexamethasone, is applied on the hemocytes. By contrast, dexamethasone-pretreated hemocyte monolayers exhibit a drastic increase in the quantity of yeast cell-hemocyte internalization when the cells are treated by arachidonic acid. In addition, phagocytosis presents significant reduction in hemocyte monolayers treated with a specific PAF receptor antagonist, WEB 2086. Nevertheless, inhibition of phagocytosis with WEB 2086 is counteracted by the treatment of the hemocyte monolayers with PAF. In conclusion, phagocytosis of yeast cells by hemocytes is related to the activation of PAF receptors and eicosanoid pathways in the bloodsucking bug, R. prolixus.


Subject(s)
Hemocytes/drug effects , Hemocytes/immunology , Heteroptera/drug effects , Heteroptera/immunology , Phagocytosis/drug effects , Phospholipase A2 Inhibitors , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Azepines/pharmacology , Eicosanoids/pharmacology , Larva/drug effects , Phagocytosis/physiology , Platelet Activating Factor/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Triazoles/pharmacology
19.
Life Sci ; 81(3): 210-7, 2007 Jun 27.
Article in English | MEDLINE | ID: mdl-17588613

ABSTRACT

Tumor growth is associated with angiogenesis and inflammation and the endogenous lipid, platelet activating factor (PAF), is a pro-inflammatory and pro-angiogenic mediator. We therefore measured tumor growth, angiogenesis and inflammation in normal (WT) mice and those lacking the receptor for PAF, through gene deletion (PAFR-KO). Growth of solid tumors derived from colon 26 cells was not altered but that from Ehrlich cells was markedly (5-fold) increased in the PAFR-KO mice, relative to the WT strain. Angiogenesis, as tumor content of VEGF or hemoglobin, was increased in both tumors from the mutant strain. Inflammation, as neutrophil and macrophage accumulation and chemokine (CXCL2 and CCL2) content of tumors, was decreased or unchanged in the tumors implying an overall decrease in the inflammatory response in the PAFR-KO strain. We also assessed growth of the Ehrlich tumor in its ascites form, after i.p. injection. Here growth (ascites volume) was inhibited by about 30%, but neutrophil and macrophage numbers were increased in the ascites fluid from the PAFR-KO mice. Angiogenesis in the peritoneal wall, which is not invaded by the tumor cells, was increased but leukocyte infiltration decreased in the mutant strain. Our results show, unexpectedly, that tumor-induced angiogenesis was increased in mice lacking response to PAF, from which we infer that in normal (WT) mice, PAF is anti-angiogenic. Further, although growth was still associated with angiogenesis in PAFR-KO mice, growth was not correlated with inflammation (leukocyte accumulation).


Subject(s)
Inflammation/pathology , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Platelet Membrane Glycoproteins/physiology , Receptors, G-Protein-Coupled/physiology , Acetylglucosaminidase/metabolism , Animals , Ascites/metabolism , Carcinoma, Ehrlich Tumor/pathology , Chemokine CCL2/biosynthesis , Chemokines, CXC/biosynthesis , Inflammation/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Transplantation , Neoplasms/blood supply , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Peritoneal Cavity , Peroxidase/metabolism , Platelet Activating Factor/pharmacology , Platelet Activating Factor/physiology , Platelet Membrane Glycoproteins/genetics , Receptors, G-Protein-Coupled/genetics , Regional Blood Flow/genetics , Regional Blood Flow/physiology , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism
20.
Prostaglandins Other Lipid Mediat ; 80(3-4): 123-35, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16939877

ABSTRACT

Lipoxin A4 (LXA4) is a lipid mediator that plays an important role in the resolution of inflammation. However, the role of LXA4 and aspirin (ASA)-triggered lipoxins (ATLs) in inflammatory edema formation remains unclear. Here, we investigated the inhibitory role played by LXA4 in the carrageenan-induced and other inflammatory mediator-induced edematogenic response in mice, and also assessed the role of ATLs in the anti-edematogenic action of aspirin. Our results showed that LXA4 (1-20 ng/paw or 5 microg/kg i.p.) was effective in inhibiting carrageenan-induced paw edema from 30 min to 2 h. LXA4 (10 ng/paw) was also able to acutely inhibit PAF-, histamine-, PGE2- or bradykinin-induced paw edema, as well as the PAF-induced myeloperoxidase activity increase in the paws. Likewise, LXA4 (10 ng/cavity) also inhibited the pleural edema triggered by histamine (1h), and this response was not followed by leukocyte accumulation. Of note, the lipoxin receptor (ALX-r) antagonist Boc2 (butoxycarbonyl-Phe-Leu-Phe-Leu-Phe, 200 ng/paw) significantly reverted the anti-edematogenic effect of ASA (300 mg/kg p.o.) against carrageenan, PAF, PGE2 and BK, without affecting the anti-edematogenic action caused by indomethacin (3 mg/kg i.p.) in the carrageenan-induced paw edema. Collectively, our results demonstrate for the first time that LXA4 displays an acute and rapid onset anti-edematogenic activity that does not discriminate among different pro-inflammatory stimuli, an effect that is most likely independent of its action on the leukocyte influx. Finally, the present study demonstrates that ATLs exert a very important role in the acute anti-edematogenic action of ASA.


Subject(s)
Aspirin/pharmacology , Edema/prevention & control , Lipoxins/pharmacology , Acute Disease , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/administration & dosage , Bradykinin/administration & dosage , Bradykinin/pharmacology , Carrageenan , Dinoprostone/administration & dosage , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Edema/chemically induced , Edema/pathology , Foot/pathology , Hindlimb/drug effects , Hindlimb/pathology , Histamine/administration & dosage , Histamine/pharmacology , Indomethacin/administration & dosage , Indomethacin/pharmacology , Injections, Intraperitoneal , Leukocytes/drug effects , Leukocytes/pathology , Lipoxins/administration & dosage , Male , Mice , Neutrophil Infiltration/drug effects , Oligopeptides/administration & dosage , Oligopeptides/pharmacology , Platelet Activating Factor/administration & dosage , Platelet Activating Factor/pharmacology , Pleurisy/chemically induced , Pleurisy/pathology , Pleurisy/prevention & control , Receptors, Lipoxin/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL