Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Commun Biol ; 4(1): 1109, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34548610

ABSTRACT

The research into the pathophysiology of atherosclerosis has considerably increased our understanding of the disease complexity, but still many questions remain unanswered, both mechanistically and pharmacologically. Here, we provided evidence that the pro-oxidant enzyme Prenylcysteine Oxidase 1 (PCYOX1), in the human atherosclerotic lesions, is both synthesized locally and transported within the subintimal space by proatherogenic lipoproteins accumulating in the arterial wall during atherogenesis. Further, Pcyox1 deficiency in Apoe-/- mice retards atheroprogression, is associated with decreased features of lesion vulnerability and lower levels of lipid peroxidation, reduces plasma lipid levels and inflammation. PCYOX1 silencing in vitro affects the cellular proteome by influencing multiple functions related to inflammation, oxidative stress, and platelet adhesion. Collectively, these findings identify the pro-oxidant enzyme PCYOX1 as an emerging player in atherogenesis and, therefore, understanding the biology and mechanisms of all functions of this unique enzyme is likely to provide additional therapeutic opportunities in addressing atherosclerosis.


Subject(s)
Atherosclerosis/genetics , Carbon-Sulfur Lyases/genetics , Adult , Aged , Animals , Atherosclerosis/metabolism , Carbon-Sulfur Lyases/metabolism , Female , Humans , Inflammation/genetics , Male , Mice , Middle Aged , Oxidative Stress/genetics , Platelet Adhesiveness/genetics
2.
Nutr Metab Cardiovasc Dis ; 26(10): 922-30, 2016 10.
Article in English | MEDLINE | ID: mdl-27378395

ABSTRACT

BACKGROUND AND AIMS: Transglutaminase 2 (TGM2), a member of the transglutaminase family of enzymes, is a multifunctional protein involved in numerous events spanning from cell differentiation, to signal transduction, apoptosis, and wound healing. It is expressed in a variety of cells, macrophages included. Macrophage TGM2 promotes the clearance of apoptotic cells (efferocytosis) and emerging evidence suggests that defective efferocytosis contributes to the consequences of inflammation-associated diseases, including atherosclerotic lesion progression and its sequelae. Of interest, active TGM2 identified in human atherosclerotic lesions plays critical roles in plaque stability through effects on matrix cross-linking and TGFß activity. This study explores the mechanisms by which TGM2 controls efferocytosis in human macrophages. METHODS AND RESULTS: Herein we show that TGM2 increases progressively during monocyte differentiation towards macrophages and controls their efferocytic potential as well as morphology and viability. Two experimental approaches that took advantage of the inhibition of TGM2 activity and protein silencing give proof that TGM2 reduction significantly impairs macrophage efferocytosis. Among the mechanisms involved we highlighted a role of the receptors CD14 and SR-AI whose levels were markedly reduced by TGM2 inhibition. Conversely, CD36 receptor and αvß3 integrin levels were not influenced. Of note, lipid accumulation and IL-10 secretion were reduced in macrophages displaying defective efferocytosis. CONCLUSION: Overall, our data define a crucial role of TGM2 activity during macrophage differentiation via mechanisms involving CD14 and SR-AI receptors and show that TGM2 inhibition triggers a pro-inflammatory phenotype.


Subject(s)
Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , Lipopolysaccharide Receptors/metabolism , Macrophages/drug effects , Phagocytosis/drug effects , Serine-Arginine Splicing Factors/metabolism , Transglutaminases/antagonists & inhibitors , Apoptosis , Cadaverine/analogs & derivatives , Cadaverine/pharmacology , Cell Differentiation , Cell Shape , Cell Survival , Coculture Techniques , Cystamine/pharmacology , Dose-Response Relationship, Drug , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Interleukin-10/metabolism , Jurkat Cells , Macrophages/enzymology , Macrophages/metabolism , Phenotype , Protein Glutamine gamma Glutamyltransferase 2 , RNA Interference , Signal Transduction/drug effects , T-Lymphocytes/pathology , Time Factors , Transfection , Transglutaminases/genetics , Transglutaminases/metabolism
3.
Data Brief ; 4: 177-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26217784

ABSTRACT

This data article is referred to the research article entitled Human monocyte-derived macrophages are heterogeneous: proteomic profile of different phenotypes by Eligini et al. Eligini S., Brioschi M., Fiorelli S., Tremoli E., Banfi C., Colli S. Human monocyte-derived macrophages are heterogeneous: proteomic profile of different phenotypes. J. Proteomics 124, 2015, 112-123. Macrophages obtained in vitro from blood monocytes are largely used as surrogate model of tissue macrophages that are heterogeneous and not easy to obtain and handle. Under spontaneous differentiation in vitro, monocyte-derived macrophages (MDMs) display two dominant subsets (round and spindle) that show different transcriptional, antigenic, and functional profiles mimicking, at least in part, the heterogeneity of tissue macrophages. This article reports the nano-LC-MS(E) analysis of the proteome of round and spindle MDMs allowing a deeper comprehension of macrophage heterogeneity.

4.
J Proteomics ; 124: 112-23, 2015 Jun 21.
Article in English | MEDLINE | ID: mdl-25857278

ABSTRACT

Tissue macrophages play a key role in many aspects of human physiology and pathology. These cells are heterogeneous both in term of morphology and function. As an example, heterogeneity has been reported within the atherosclerotic lesions where distinct populations exert opposite functions driving plaque progression or stability. Tissue macrophages are not easily obtained and differentiated blood-derived monocytes are largely used as surrogate model. We previously reported that human macrophages spontaneously differentiated from adherent monocytes show two dominant subsets, distinct for morphology (spindle and round) and functions. The aim of this study was to evaluate the intracellular proteome of these two macrophage subsets by means of a microproteomic workflow properly set up to simultaneously identify and quantify proteins from a minimal number of morphotypically heterogeneous cells in culture. We report two distinct proteomic profiles that distinguish round from spindle macrophages. In particular, differential abundances were observed for proteins involved in membrane traffic regulation, lipid handling, efferocytosis, and protection against stress conditions. Results reinforce and extend previous data on the functional and antigenic profile of these macrophage phenotypes strengthening the suitability of our model to focus on macrophage heterogeneity. BIOLOGICAL SIGNIFICANCE: Tissue macrophages patrol homeostatic functions, immune surveillance, and resolution of inflammation. The spectrum of macrophage activation states is, therefore, wide and gives ground for the heterogeneity of these cells, documented in health and disease. This study provides knowledge of the distinct proteome that characterises the two dominant morphotypes (round and spindle) of human macrophages that, in our culture condition, are generated by spontaneous differentiation from blood-derived monocytes. Results extend previous data about the different antigenic, transcriptional, and functional profiles of these morphotypes and further strengthen the suitability of this in vitro model to study macrophage heterogeneity and to address the effects of environmental challenges and drugs.


Subject(s)
Blood Proteins/metabolism , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Monocytes/metabolism , Proteome/metabolism , Blood Proteins/chemistry , Cell Differentiation/physiology , Cells, Cultured , Gene Expression Profiling , Humans , Phenotype , Proteome/chemistry
5.
J Thromb Haemost ; 7(1): 206-16, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18983479

ABSTRACT

BACKGROUND: Protease-activated receptors (PARs) comprise a family of G-protein-coupled receptors with a unique proteolytic activation mechanism. PARs regulate a broad range of cellular functions and are involved in the pathogenesis of inflammatory disorders. Moreover, PAR1 and PAR2 activation in the endothelium shifts it toward a prothrombotic condition. OBJECTIVES: To assess the relevance of intracellular reactive oxygen species (ROS) in the signaling events underlying tissue factor (TF) expression elicited by PAR1 and PAR2 occupancy in endothelial cells, and to investigate their source. METHODS: Human umbilical vein endothelial cells (HUVEC) were exposed to specific PAR1 and PAR2 agonist peptides. TF expression was determined by real-time reverse transcription polymerase chain reaction analysis and measurement of procoagulant activity. ROS generation was determined by a fluorometric assay after cell loading with 2'-7'-dichlorofluorescein diacetate. RESULTS: ROS generated by the mitochondrial chain, mostly from complex III, provide a pathway through which PAR1 and PAR2 occupancy induces TF. Other sources of ROS do not participate in TF induction. Activation of both ERK1/2 and p38 MAPK is critical for mitochondrial ROS generation. In addition to these pathways shared by the two PARs, mechanisms downstream from PAR1 and PAR2 activation, different for the two receptors, also induced TF. A module that sensitively regulates PAR1 signaling and ultimately involves NF-kappaB activation has been identified. CONCLUSIONS: Our data identify ROS originating in mitochondria as key mediators of the signaling pathways triggered by PAR1 and PAR2 engagement in endothelial cells and show that downstream from receptor activation occur cascades that are mechanistically coupled to procoagulant activity.


Subject(s)
Endothelial Cells/metabolism , Mitochondria/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Thromboplastin/genetics , Blood Coagulation , Cells, Cultured , Endothelial Cells/ultrastructure , Endothelium, Vascular/cytology , Humans , Reactive Oxygen Species , Signal Transduction/physiology , Transcriptional Activation
6.
Article in English | MEDLINE | ID: mdl-17588731

ABSTRACT

UNLABELLED: The composition and incorporation of fatty acids (FA) in plasma and blood cells is the result of distinct processes: intake, metabolism and peripheral utilization. AIM OF THE STUDY: was to compare the FA profile in plasma, lipoproteins and blood cells with that in whole blood (WB) from healthy volunteers; to assess the quantitative distribution of selected FA in triacylglycerols, cholesteryl esters and phospholipids. Lipid FA profiles are comparable in plasma and lipoproteins but differ from those in blood cells. In WB, the FA profile results from the balanced proportion of FA pools in plasma and cells. The contribution of each lipid class to the total amount of FA differs among blood specimens. Phospholipids of plasma and red blood cell are the major contributors to the FA amount and profile in WB. In conclusion, the FA profile of WB reflects the FA status and WB could be an adequate specimen for the assessment of FA intakes.


Subject(s)
Blood Cells/chemistry , Fatty Acids/blood , Adult , Cholesterol Esters/chemistry , Female , Humans , Lipoproteins/chemistry , Male , Middle Aged , Reference Values , Triglycerides/chemistry
7.
Thromb Haemost ; 88(3): 524-32, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12353085

ABSTRACT

Basic and clinical evidence has provided insight into the molecular events that link inflammation and coagulation. Increased expression of tissue factor (TF) by circulating and vascular cells has been indicated as responsible for the thrombotic complications associated with acute and chronic inflammation. TF is indeed inducible in circulating and vascular cells by cytokines and bacterial lipopolysaccharide (LPS) and its expression triggers the coagulation. The cyclopentenone prostaglandins are naturally occurring prostaglandin D2 (PGD2) derivatives that comprises prostaglandin J2 (PGJ2) and its metabolites delta12-PGJ2 and 15-deoxy- delta12,14-prostaglandin J2 (15d-PGJ2). These compounds, detected in vivo in a later phase of the inflammatory response, are characterized by anti-inflammatory activity and participate to the resolution of inflammation. We have here investigated the effect of 15d-PGJ2 on TF expression in human macrophages and endothelial cells (HUVEC). Our results indicate that 15d-PGJ2 down-regulates LPS- and TNFalpha-induced TF activity, protein and mRNA through inhibition of TF gene transcription. The effect of 15d-PGJ2 is targeted to the NF-kappaB/I-kappaB pathway and to the mitogen activated protein kinase ERK1/2. A role of PPAR-gamma activation in TF inhibition by 15d-PGJ2 was excluded. We conclude that 15d-PGJ2 negatively affects TF expression in macrophages and endothelial cells through a PPARgamma-independent mechanism. This down-regulation may be crucial to limit excessive blood clotting activation in immuno-inflammatory diseases.


Subject(s)
Endothelium, Vascular/metabolism , Macrophages/metabolism , Prostaglandin D2/pharmacology , Thromboplastin/antagonists & inhibitors , Down-Regulation/drug effects , Endothelium, Vascular/cytology , Humans , Lipopolysaccharides , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Prostaglandin D2/analogs & derivatives , Thromboplastin/biosynthesis , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha , Umbilical Veins
8.
Article in English | MEDLINE | ID: mdl-12324237

ABSTRACT

Oxidative modification of low-density lipoproteins (LDLs) plays a key role in the development of atherosclerosis and the onset of coronary artery disease. LDL oxidation alters the antithrombotic balance of human endothelial cells inducing surface tissue factor (TF) pathway activity, which results in enhanced fibrin deposition. Fibrinolysis, which is strictly regulated by plasminogen activator inhibitor-1 (PAL-1) and tissue-type plasminogen activator (tPA). Is also dysregulated by LDL oxidation with a net increase in the inhibitory rate. Oxidized LDLs (oxLDLs) also affect many aspects of macrophage function linked to the inflammatory response of these cells, In particular, oxLDLs downregulate inducible cyclooxigenase (Cox-2) in human monocyte-derived macrophages exposed to bacterial lipopolysaccharide. This observation may support the hypothesis that, within atheromata, the transformation macrophages into foam cells results in the attenuation of the inflammatory response, thus contributing to the progression of athrogenesis. Among lipid constituents of oxLDLs, Ox-PAPC, a mixture of oxidized arachidonic acid-containing phospholipids, prevents Cox-2 expression, suggesting that it could be considered responsible for the biological activity of oxLDLs.


Subject(s)
Isoenzymes/metabolism , Lipoproteins, LDL/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Thrombosis/chemically induced , Arteriosclerosis/complications , Blood Coagulation/drug effects , Cyclooxygenase 2 , Foam Cells/cytology , Foam Cells/drug effects , Foam Cells/metabolism , Humans , Macrophages/cytology , Macrophages/drug effects , Macrophages/enzymology , Macrophages/metabolism , Membrane Proteins , Thrombosis/complications , Thrombosis/enzymology
9.
Br J Pharmacol ; 133(7): 1163-71, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11487528

ABSTRACT

Nitric oxide (NO) regulates cyclo-oxygenase (COX) activity in various cell systems and reports conflict in regard to its stimulatory versus inhibitory role. Incubation of human umbilical vein endothelial cells (HUVEC) with SIN-1 (3-morpholinosydnonimine), a donor of NO, resulted in a rapid and dose-dependent increase in the expression of COX-2 as analysed by Western and Northern blotting. Incubation of HUVEC with SIN-1 and interleukine (IL)-1alpha resulted in increased induction of COX-2 compared with IL-1alpha alone and corresponded to an additive effect. The COX-2 induction was dependent on a de novo synthesis since cycloheximide, an inhibitor of protein synthesis, blocked the enzyme expression. The increase in COX-2 expression was not accompanied by a corresponding change in prostaglandin (PG) production. However, the COX activity was partially recovered when immunoprecipitated COX-2 was incubated with arachidonic acid and haematin. Peroxynitrite, a highly reactive nitrogen molecule derived from the interaction of NO and superoxide anion, significantly increased COX-2 expression. Under these conditions and within the limit of detection of the antibody, selective antibody for nitrotyrosine failed to detect nitrated COX-2 in immunoprecipitated COX-2 when cells where incubated with SIN-1 or SIN-1+IL-1alpha. Ro 31-8220, a specific inhibitor of protein kinase (PK) C, blocked the induction of COX-2. Also, SB203580, the selective inhibitor of p38 MAP kinase, strongly blocked the induction of COX-2 by SIN-1 in the presence or absence of IL-1alpha, whereas the MEK-1 inhibitor, PD 98059, affected it to a lesser extent. These data demonstrate that SIN-1 induces COX-2 in HUVEC in the absence of PG formation and suggest a complex regulation of COX-2 expression and PG formation by NO in endothelial cells.


Subject(s)
Endothelium, Vascular/drug effects , Isoenzymes/metabolism , Molsidomine/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/biosynthesis , Blotting, Northern , Blotting, Western , Cell Line , Cyclooxygenase 2 , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Imidazoles/pharmacology , Interleukin-1/pharmacology , Isoenzymes/genetics , Membrane Proteins , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Molsidomine/analogs & derivatives , Nitrates/pharmacology , Precipitin Tests , Prostaglandin-Endoperoxide Synthases/genetics , Pyridines/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
10.
Arterioscler Thromb Vasc Biol ; 19(7): 1719-25, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10397690

ABSTRACT

Atherogenesis involves several aspects of chronic inflammation and wound healing. Indeed, the atheroma is considered a special case of tissue response to injury. Injurious stimuli may include lipoproteins trapped within lesions where protein and lipid moieties have undergone chemical modifications. We have studied the effect of oxidized low density lipoproteins (ox-LDL) on inducible cyclooxygenase (Cox-2) in human monocyte-derived macrophages exposed to bacterial lipopolysaccharide (LPS). Levels of both Cox-2 and constitutive cyclooxygenase (Cox-1) were assessed using Western blot analysis. Prior incubation of macrophages with ox-LDL resulted in a strong inhibition of Cox-2 induced by LPS, without effect on Cox-1. The inhibitory effect was dependent on ox-LDL concentration and its onset was early in time (already detectable 1 hour after macrophage exposure to ox-LDL). Native LDL, and other forms of modified LDL, were without effect. The inhibition was dependent on endocytosis of ox-LDL and could be reproduced using the lipid extract from ox-LDL. Lysophosphatidylcholine, 7beta-hydroxycholesterol, and 7-oxocholesterol failed to mimic the inhibition, but oxidized arachidonic acid-containing phospholipids, produced by autoxidation of 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine, markedly inhibited Cox-2. The observation that ox-LDL downregulates Cox-2 in human macrophages may explain the fact that, within atheromata, the transformation of macrophages into foam cells results in attenuation of the inflammatory response, thus contributing to progression of atherogenesis.


Subject(s)
Lipoproteins, LDL/pharmacology , Lipoxygenase/drug effects , Macrophages/enzymology , Cells, Cultured , Endocytosis , Humans , Lipopolysaccharides/pharmacology , Lipoproteins/metabolism , Lipoxygenase/biosynthesis , Lysosomes/metabolism , Macrophage Colony-Stimulating Factor/physiology
11.
Thromb Haemost ; 81(4): 576-81, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10235442

ABSTRACT

This study investigated whether spontaneous lipid enrichment of human macrophages affects their thrombogenic potential as measured by increased production of tissue factor (TF) and plasminogen activation inhibitor types 1 and 2 (PAI-1 and PAI-2). Macrophages were obtained following a 7-day culture period of monocytes, isolated from the same donor, in autologous serum (HS) or in fetal bovine serum (FBS). Those cultured in HS underwent marked lipid accumulation relative to those cultured in FBS that was accompanied by increased production of TF and PAI-1, but not of PAI-2, and decreased production of interleukin-1beta. They also contained more arachidonic and linoleic acid and lower amounts of n-3 polyunsaturated fatty acids, particularly docosahexaenoic acid (22: 6). These data indicate that the transformation of macrophages into foam cells results in an increase in their thrombogenic and antifibrinolytic potential and provide a possible explanation of the thrombotic sequelae frequently consequent on plaque fissuring and disruption.


Subject(s)
Arteriosclerosis/etiology , Foam Cells/physiology , Macrophages/physiology , Monocytes/cytology , Cell Adhesion/physiology , Cells, Cultured , Fetal Blood/chemistry , Humans , Interleukin-1/metabolism , Lipid Metabolism , Macrophage Activation/drug effects , Plasminogen Activator Inhibitor 1/metabolism , Thromboplastin/metabolism
12.
Arterioscler Thromb Vasc Biol ; 17(2): 265-72, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9081680

ABSTRACT

We examined the effect of fluvastatin, the first entirely synthetic 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor that is structurally different from other vastatins, on tissue factor (TF) expression in human macrophages spontaneously differentiated in culture from blood monocytes. Fluvastatin decreased TF activity in a dose-dependent manner (1 to 5 mumol/L) in both unstimulated and lipopolysaccharide-stimulated macrophages, and this reduction paralleled the decrease in immunologically recognized TF protein. The same results were obtained with another lipophilic vastatin, simvastatin, but not with hydrophilic pravastatin. The reduction in TF expression was also observed in macrophages enriched in cholesterol after exposure to 50 micrograms/mL acetylated low density lipoprotein. The inhibitory effect of fluvastatin on TF activity and antigen was fully reversible by coincubation with 100 mumol/L mevalonate or 10 mumol/L all-trans-geranylgeraniol but not with dolichol, farnesol, or geraniol. Suppression of TF antigen and activity was accompanied by a diminution in TF mRNA levels, which was completely prevented by mevalonate. Furthermore, fluvastatin impaired bacterial lipopolysaccharide-induced binding of c-Rel/p65 heterodimers to a kappa B site in the TF promoter, indicating that this drug influences induction of the TF gene. We conclude that lipophilic vastatins inhibit TF expression in macrophages, and because this effect is prevented by mevalonate and geranylgeraniol, a geranylgeranylated protein plays a crucial role in the regulation of TF biosynthesis. The suppression of TF in macrophages by vastatins indicates a potential mechanism by which these drugs interfere with the formation and progression of atherosclerotic plaque as well as thrombotic events in hyperlipidemic patients.


Subject(s)
Blood Cells/metabolism , Enzyme Inhibitors/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Indoles/pharmacology , Macrophages/metabolism , Thromboplastin/antagonists & inhibitors , Arteriosclerosis/prevention & control , Cells, Cultured , Cholesterol/metabolism , Fluvastatin , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Lipopolysaccharides/pharmacology , Mevalonic Acid/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-rel , RNA, Messenger/metabolism , Thromboplastin/genetics , Thrombosis/prevention & control
13.
Free Radic Biol Med ; 20(3): 271-8, 1996.
Article in English | MEDLINE | ID: mdl-8720896

ABSTRACT

Stimulated platelets secrete a variety of physiologically active substances that affect many neutrophil functions. We have examined the capacity of platelets to modulate superoxide anion generation by neutrophils. The amounts of superoxide anion produced by neutrophils in the presence of platelets were markedly enhanced when platelet-neutrophil coincubations were stimulated with agents that simultaneously activate both cell types, as the calcium ionophore A23187 and sodium arachidonate. This effect was dependent upon the number of platelets added to the incubation media and was not affected by inhibitors of arachidonic acid pathway or by preincubation of platelets with an antibody anti-P-selectin. The hypothesis of an involvement of purine nucleotides released by platelets during aggregation on the observed effect of enhancement of superoxide anion generation by neutrophils was then tested. Experimental evidence indicates that platelets release, during A23187-induced aggregation, amounts of ATP that are of the same order (5-10 microM) of those demonstrated to enhance superoxide anion generation by neutrophils. In addition, platelet lysates mimicked the effect of intact platelets in enhancing superoxide anion generation by A23187 stimulated neutrophils. Interestingly, at variance with the results obtained with intact platelets and platelet lysates, supernatants of thrombin-stimulated platelets did not increase O2.- by neutrophils. The enhancing effect of these supernatants was, however, restored when platelets were preincubated with an antibody anti P-selectin. These data indicate that platelets, through the release of purine nucleotides, enhance superoxide generation by neutrophils, thus increasing the cytotoxic potential of these cells.


Subject(s)
Adenosine Triphosphate/blood , Blood Platelets/physiology , Neutrophils/physiology , Superoxides/blood , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Analysis of Variance , Arachidonic Acid/pharmacology , Blood Platelets/drug effects , Calcimycin/pharmacology , Humans , In Vitro Techniques , Kinetics , Neutrophils/drug effects
14.
Am J Clin Nutr ; 61(3): 607-13, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7872228

ABSTRACT

This study addressed two questions: 1) whether a relatively low dose of n-3 fatty acid ethyl esters (n-3 FAs) administered to healthy volunteers for a prolonged period of time would exert beneficial effects on plasma lipids, platelet function, and thromboxane biosynthesis; and 2) whether a short-term loading treatment (6 wk) with 6 g n-3 FAs/d followed by 12 wk with 3 g/d results in more pronounced effects. After 6 wk treatment a reduction of plasma triglyceride concentration and an accumulation of EPA and DHA in plasma were observed. A longer period of treatment with n-3 FAs was necessary to affect platelet aggregation and thromboxane A2 biosynthesis. At 12 and 18 wk, platelet aggregation, thromboxane A2 formation, and the excretion of thromboxane metabolites in urine were reduced, particularly in subjects who received 6 g n-3 FAs/d during the initial 6 wk. After treatment ended, triglyceride and thromboxane A2 biosynthesis returned to baseline values within 4 wk, whereas platelet aggregation remained impaired for > or = 14 wk. The longlasting impairment in platelet aggregation was accompanied by the retention of n-3 FAs in platelet phospholipids.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Administration, Oral , Adult , Dose-Response Relationship, Drug , Fatty Acids, Omega-3/blood , Female , Humans , Lipoproteins/blood , Male , Platelet Aggregation Inhibitors/blood , Thromboxanes/biosynthesis , Triglycerides/blood
15.
Arterioscler Thromb ; 14(10): 1600-8, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7918310

ABSTRACT

n-3 Fatty acids are known to influence several functions of monocytes, including adhesion, cytokine synthesis, and superoxide generation. Monocytes express tissue factor, a membrane-bound glycoprotein, that acts as a catalyst in the coagulation cascade. In this study we evaluated the effects of administration of n-3 fatty acid ethyl esters to healthy volunteers and to hypertriglyceridemic patients on tissue factor activity (TF activity) in adherent monocytes. n-3 Fatty acids containing 75% eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) (ratio of EPA to DHA, 1.34) were administered (3 g/d) to normal volunteers for 18 weeks. In addition, the effects of this treatment were evaluated in 30 hypertriglyceridemic patients for 24 weeks by using a double-blind, placebo-controlled study. TF activity in adherent monocytes was evaluated with a one-stage clotting assay. Plasma and monocyte fatty acid compositions were determined by gas-liquid chromatography. In healthy volunteers, n-3 fatty acids significantly reduced TF activity in adherent monocytes either in the unstimulated condition or after exposure to endotoxin. The inhibitory effect was observed after 12 weeks of treatment and was more pronounced after 18 weeks (> 70%, P < .001 versus baseline). Concomitantly, levels of EPA and DHA increased in plasma and monocyte lipids. Interestingly, after stopping treatment, monocyte TF activity remained inhibited for at least 14 weeks. Treatment with n-3 fatty acids for 24 weeks also resulted in a significant reduction of TF activity in adherent monocytes from hypertriglyceridemic patients (-31% and -40% in unstimulated and endotoxin-stimulated cells; P < .05 versus baseline).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Hypertriglyceridemia/blood , Monocytes/physiology , Adult , Cell Adhesion , Double-Blind Method , Fatty Acids/blood , Female , Humans , Male , Placebos , Reference Values , Thromboplastin/metabolism
17.
Biochim Biophys Acta ; 1210(1): 55-62, 1993 Dec 02.
Article in English | MEDLINE | ID: mdl-8257719

ABSTRACT

Eight normal volunteers (four men and four women) were treated with 3 x 1 g capsules of n-3 fatty acid ethyl esters for a period of 18 weeks, followed by a 24 week washout. Fatty acids of plasma, platelets, monocytes and red blood cells were analyzed at 0, 6, 12 and 18 weeks of treatment and at 4, 14 and 24 weeks of washout. During treatment, accumulation of EPA in plasma and cells was almost maximal at 6 weeks, whereas that of DHA reached a peak at 18 weeks. Arachidonic acid declined somewhat at 12 weeks in plasma and more markedly at 18 weeks in red blood cells and monocytes. During washout, EPA returned rapidly toward pretreatment values in all compartments, but it remained significantly higher in plasma and platelets at the end of washout. DHA declined more slowly, maintaining higher than basal values in plasma and platelets and lower than basal in red blood cells, at the end of washout. Rebound increments of AA occurred in plasma. Finally, the plasma levels of AA, but not those of the n-3 fatty acids, were more markedly modified in males than in females. The presented results suggest interactions between circulating fatty acids in the different compartment after n-3 FA administration, and indicate that very long washouts are necessary for a complete recovery from the induced fatty acid modifications.


Subject(s)
Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/analogs & derivatives , Fatty Acids/blood , Adult , Eicosapentaenoic Acid/pharmacology , Female , Humans , Male , Middle Aged , Reference Values
18.
J Lab Clin Med ; 121(3): 406-14, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8383163

ABSTRACT

In recent years the interactions between lipoxygenase enzymes present in platelets and leukocytes, resulting in the transcellular biosynthesis of eicosanoids, have been discovered and their relevance in thrombotic and inflammatory disorders recognized. However, attention has focused on the synthesis of novel products, not normally formed by the single-cell population. Less information is available on the changes in hydroxyacid formation during platelet-neutrophil interactions. In this study, we evaluated the quantitative changes of the levels of 12- and 5-hydroxyeicosatetraenoic acids, leukotriene B4, and thromboxane B2 occurring during the incubation of platelet-neutrophil mixed suspensions stimulated with the calcium ionophore A23187. Cell-to-cell interaction resulted in quantitative changes of the level of hydroxyacids; in mixed platelet-neutrophil suspensions, the levels of 12-hydroxyeicosatetraenoic acid were significantly increased with respect to those measured in platelets incubated alone. The amount of 5-hydroxyeicosatetraenoic acid formed by neutrophils was significantly decreased by the presence of platelets in the incubation medium. In addition, a slight but significant reduction in leukotriene B4 synthesis was observed. Concomitantly with these changes, the formation of thromboxane B2 by platelets was modified, indicating that--besides the lipoxygenase pathway--the arachidonic acid metabolism by cyclooxygenase is affected. Our study demonstrates that in mixed platelet-neutrophil suspensions profound quantitative modifications in hydroxyacid and thromboxane synthesis occur, indicating that the overall balance of arachidonic acid products may be altered in pathologic conditions in which early events of multicellular origin have been recognized.


Subject(s)
Blood Platelets/metabolism , Cell Communication/physiology , Hydroxyeicosatetraenoic Acids/biosynthesis , Leukotriene B4/biosynthesis , Neutrophils/metabolism , Thromboxane B2/biosynthesis , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid , Blood Platelets/physiology , Humans , Neutrophils/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...