Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Am J Clin Nutr ; 119(5): 1175-1186, 2024 May.
Article in English | MEDLINE | ID: mdl-38484976

ABSTRACT

BACKGROUND: Extracellular vesicles (EVs) are proposed to play a role in the development of cardiovascular diseases (CVDs) and are considered emerging markers of CVDs. n-3 PUFAs are abundant in oily fish and fish oil and are reported to reduce CVD risk, but there has been little research to date examining the effects of n-3 PUFAs on the generation and function of EVs. OBJECTIVES: We aimed to investigate the effects of fish oil supplementation on the number, generation, and function of EVs in subjects with moderate risk of CVDs. METHODS: A total of 40 participants with moderate risk of CVDs were supplemented with capsules containing either fish oil (1.9 g/d n-3 PUFAs) or control oil (high-oleic safflower oil) for 12 wk in a randomized, double-blind, placebo-controlled crossover intervention study. The effects of fish oil supplementation on conventional CVD and thrombogenic risk markers were measured, along with the number and fatty acid composition of circulating and platelet-derived EVs (PDEVs). PDEV proteome profiles were evaluated, and their impact on coagulation was assessed using assays including fibrin clot formation, thrombin generation, fibrinolysis, and ex vivo thrombus formation. RESULTS: n-3 PUFAs decreased the numbers of circulating EVs by 27%, doubled their n-3 PUFA content, and reduced their capacity to support thrombin generation by >20% in subjects at moderate risk of CVDs. EVs derived from n-3 PUFA-enriched platelets in vitro also resulted in lower thrombin generation, but did not alter thrombus formation in a whole blood ex vivo assay. CONCLUSIONS: Dietary n-3 PUFAs alter the number, composition, and function of EVs, reducing their coagulatory activity. This study provides clear evidence that EVs support thrombin generation and that this EV-dependent thrombin generation is reduced by n-3 PUFAs, which has implications for prevention and treatment of thrombosis. CLINICAL TRIAL REGISTRY: This trial was registered at clinicaltrials.gov as NCT03203512.


Subject(s)
Blood Coagulation , Blood Platelets , Cross-Over Studies , Extracellular Vesicles , Fatty Acids, Omega-3 , Humans , Extracellular Vesicles/metabolism , Fatty Acids, Omega-3/pharmacology , Male , Female , Middle Aged , Double-Blind Method , Blood Coagulation/drug effects , Blood Platelets/metabolism , Blood Platelets/drug effects , Dietary Supplements , Cardiovascular Diseases/prevention & control , Adult , Fish Oils/pharmacology , Fish Oils/administration & dosage , Aged , Fatty Acids/metabolism
2.
J Lipid Res ; 65(1): 100484, 2024 01.
Article in English | MEDLINE | ID: mdl-38103786

ABSTRACT

Aminophospholipids (aPL) such as phosphatidylserine are essential for supporting the activity of coagulation factors, circulating platelets, and blood cells. Phosphatidylthreonine (PT) is an aminophospholipid previously reported in eukaryotic parasites and animal cell cultures, but not yet in human tissues. Here, we evaluated whether PT is present in blood cells and characterized its ability to support coagulation. Several PT molecular species were detected in human blood, washed platelets, extracellular vesicles, and isolated leukocytes from healthy volunteers using liquid chromatography-tandem mass spectrometry. The ability of PT to support coagulation was demonstrated in vitro using biochemical and biophysical assays. In liposomes, PT supported prothrombinase activity in the presence and absence of phosphatidylserine. PT nanodiscs strongly bound FVa and lactadherin (nM affinity) but poorly bound prothrombin and FX, suggesting that PT supports prothrombinase through recruitment of FVa. PT liposomes bearing tissue factor poorly generated thrombin in platelet poor plasma, indicating that PT poorly supports extrinsic tenase activity. On platelet activation, PT is externalized and partially metabolized. Last, PT was significantly higher in platelets and extracellular vesicle from patients with coronary artery disease than in healthy controls. In summary, PT is present in human blood, binds FVa and lactadherin, supports coagulation in vitro through FVa binding, and is elevated in atherosclerotic vascular disease. Our studies reveal a new phospholipid subclass, that contributes to the procoagulant membrane, and may support thrombosis in patients at elevated risk.


Subject(s)
Coronary Artery Disease , Glycerophospholipids , Threonine/analogs & derivatives , Thromboplastin , Animals , Humans , Thromboplastin/metabolism , Phosphatidylserines/metabolism , Liposomes/metabolism , Blood Platelets/metabolism , Thrombin/metabolism
3.
Front Cardiovasc Med ; 9: 907457, 2022.
Article in English | MEDLINE | ID: mdl-35694679

ABSTRACT

Background: Extracellular vesicles (EVs) are submicron membrane-bound vesicles released from various cells, which are emerging as a potential novel biomarker in cardiovascular diseases (CVDs) due to their procoagulatory and prothrombotic properties. However, there is little information about the relationships between circulating EVs and conventional and thrombogenic risk markers of CVDs. Objective: To investigate the relationships between circulating EVs, conventional cardiovascular risk markers and thrombogenic markers in subjects with moderate risk of CVDs. Design: Subjects (n = 40) aged 40-70 years with moderate risk of CVDs were recruited and assessed for body mass index, blood pressure and plasma lipid profile, as well as platelet aggregation, clot formation, thrombin generation and fibrinolysis. Numbers of circulating EVs were assessed by Nanoparticle Tracking Analysis and flow cytometry. A range of assays were used to assess the procoagulatory activity of plasma and circulating EVs. Results: Circulating EV numbers were positively associated with body mass index, blood pressure, plasma triacylglycerol concentration and overall CVD risk. Higher circulating EV numbers were also associated with increased thrombin generation and enhanced clot formation, and EVs isolated from subjects with moderate CVD risk promoted thrombin generation ex vivo. Higher numbers of endothelial-derived EVs were associated with a greater tendency for clot lysis. Plasma triacylglycerol concentration and diastolic blood pressure independently predicted circulating EV numbers, and EV numbers independently predicted aspects of thrombin generation and clot formation and 10-year CVD risk. Conclusion: Circulating EVs were strongly associated with both conventional and thrombogenic risk markers of CVDs, and also with overall CVD risk, highlighting a potentially important role for EVs in CVDs.

4.
Proc Natl Acad Sci U S A ; 116(16): 8038-8047, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30944221

ABSTRACT

Abdominal aortic aneurysm (AAA) is an inflammatory vascular disease with high mortality and limited treatment options. How blood lipids regulate AAA development is unknown. Here lipidomics and genetic models demonstrate a central role for procoagulant enzymatically oxidized phospholipids (eoxPL) in regulating AAA. Specifically, through activating coagulation, eoxPL either promoted or inhibited AAA depending on tissue localization. Ang II administration to ApoE-/- mice increased intravascular coagulation during AAA development. Lipidomics revealed large numbers of eoxPL formed within mouse and human AAA lesions. Deletion of eoxPL-generating enzymes (Alox12 or Alox15) or administration of the factor Xa inhibitor rivaroxaban significantly reduced AAA. Alox-deficient mice displayed constitutively dysregulated hemostasis, including a consumptive coagulopathy, characterized by compensatory increase in prothrombotic aminophospholipids (aPL) in circulating cell membranes. Intravenously administered procoagulant PL caused clotting factor activation and depletion, induced a bleeding defect, and significantly reduced AAA development. These data suggest that Alox deletion reduces AAA through diverting coagulation away from the vessel wall due to eoxPL deficiency, instead activating clotting factor consumption and depletion in the circulation. In mouse whole blood, ∼44 eoxPL molecular species formed within minutes of clot initiation. These were significantly elevated with ApoE-/- deletion, and many were absent in Alox-/- mice, identifying specific eoxPL that modulate AAA. Correlation networks demonstrated eoxPL belonged to subfamilies defined by oxylipin composition. Thus, procoagulant PL regulate AAA development through complex interactions with clotting factors. Modulation of the delicate balance between bleeding and thrombosis within either the vessel wall or circulation was revealed that can either drive or prevent disease development.


Subject(s)
Aorta, Abdominal/physiopathology , Aortic Aneurysm, Abdominal , Phospholipids , Angiotensins/metabolism , Animals , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/physiopathology , Blood Coagulation Factors/genetics , Blood Coagulation Factors/metabolism , Disease Models, Animal , Female , Lipoxygenase/genetics , Lipoxygenase/metabolism , Male , Mice , Mice, Knockout, ApoE , Phospholipids/genetics , Phospholipids/metabolism
5.
JCI Insight ; 3(6)2018 03 22.
Article in English | MEDLINE | ID: mdl-29563336

ABSTRACT

Hemostatic defects are treated using coagulation factors; however, clot formation also requires a procoagulant phospholipid (PL) surface. Here, we show that innate immune cell-derived enzymatically oxidized phospholipids (eoxPL) termed hydroxyeicosatetraenoic acid-phospholipids (HETE-PLs) restore hemostasis in human and murine conditions of pathological bleeding. HETE-PLs abolished blood loss in murine hemophilia A and enhanced coagulation in factor VIII- (FVIII-), FIX-, and FX-deficient human plasma . HETE-PLs were decreased in platelets from patients after cardiopulmonary bypass (CPB). To explore molecular mechanisms, the ability of eoxPL to stimulate individual isolated coagulation factor/cofactor complexes was tested in vitro. Extrinsic tenase (FVIIa/tissue factor [TF]), intrinsic tenase (FVIIIa/FIXa), and prothrombinase (FVa/FXa) all were enhanced by both HETE-PEs and HETE-PCs, suggesting a common mechanism involving the fatty acid moiety. In plasma, 9-, 15-, and 12-HETE-PLs were more effective than 5-, 11-, or 8-HETE-PLs, indicating positional isomer specificity. Coagulation was enhanced at lower lipid/factor ratios, consistent with a more concentrated area for protein binding. Surface plasmon resonance confirmed binding of FII and FX to HETE-PEs. HETE-PEs increased membrane curvature and thickness, but not surface charge or homogeneity, possibly suggesting increased accessibility to cations/factors. In summary, innate immune-derived eoxPL enhance calcium-dependent coagulation factor function, and their potential utility in bleeding disorders is proposed.


Subject(s)
Blood Coagulation Factors/metabolism , Hemorrhage/enzymology , Hemorrhage/metabolism , Phospholipids/metabolism , Thrombin/metabolism , Adult , Aged , Aged, 80 and over , Animals , Blood Coagulation , Blood Coagulation Factors/genetics , Blood Platelets , Cardiopulmonary Bypass/adverse effects , Carrier Proteins , Cysteine Endopeptidases , Factor IX/genetics , Factor VIII/genetics , Factor VIIa/metabolism , Factor X/genetics , Hemophilia A , Hemorrhage/prevention & control , Hemostasis , Humans , Hydroxyeicosatetraenoic Acids , Lipoproteins/pharmacology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neoplasm Proteins , Surface Plasmon Resonance , Thromboplastin/antagonists & inhibitors , Thromboplastin/metabolism
6.
Sci Signal ; 10(507)2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29184033

ABSTRACT

Blood coagulation functions as part of the innate immune system by preventing bacterial invasion, and it is critical to stopping blood loss (hemostasis). Coagulation involves the external membrane surface of activated platelets and leukocytes. Using lipidomic, genetic, biochemical, and mathematical modeling approaches, we found that enzymatically oxidized phospholipids (eoxPLs) generated by the activity of leukocyte or platelet lipoxygenases (LOXs) were required for normal hemostasis and promoted coagulation factor activities in a Ca2+- and phosphatidylserine (PS)-dependent manner. In wild-type mice, hydroxyeicosatetraenoic acid-phospholipids (HETE-PLs) enhanced coagulation and restored normal hemostasis in clotting-deficient animals genetically lacking p12-LOX or 12/15-LOX activity. Murine platelets generated 22 eoxPL species, all of which were missing in the absence of p12-LOX. Humans with the thrombotic disorder antiphospholipid syndrome (APS) had statistically significantly increased HETE-PLs in platelets and leukocytes, as well as greater HETE-PL immunoreactivity, than healthy controls. HETE-PLs enhanced membrane binding of the serum protein ß2GP1 (ß2-glycoprotein 1), an event considered central to the autoimmune reactivity responsible for APS symptoms. Correlation network analysis of 47 platelet eoxPL species in platelets from APS and control subjects identified their enzymatic origin and revealed a complex network of regulation, with the abundance of 31 p12-LOX-derived eoxPL molecules substantially increased in APS. In summary, circulating blood cells generate networks of eoxPL molecules, including HETE-PLs, which change membrane properties to enhance blood coagulation and contribute to the excessive clotting and immunoreactivity of patients with APS.


Subject(s)
Blood Coagulation Factors/metabolism , Calcium/metabolism , Cell Membrane/metabolism , Hemostasis , Phospholipids/metabolism , Platelet Activation , Adult , Aged , Animals , Antiphospholipid Syndrome/blood , Antiphospholipid Syndrome/enzymology , Blood Coagulation , Cell Membrane/ultrastructure , Cohort Studies , Disease Models, Animal , Female , Humans , Hydroxyeicosatetraenoic Acids/analysis , Hydroxyeicosatetraenoic Acids/metabolism , Lipoxygenases/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Models, Theoretical , Phospholipids/analysis , Venous Thrombosis/blood , Venous Thrombosis/chemically induced , Venous Thrombosis/enzymology , beta 2-Glycoprotein I/metabolism
7.
Wellcome Open Res ; 2: 1, 2017 Jan 04.
Article in English | MEDLINE | ID: mdl-28239665

ABSTRACT

Background. The myeloid enzyme 12/15-lipoxygenase (LOX), which generates bioactive oxidized lipids, has been implicated in numerous inflammatory diseases, with several studies demonstrating an improvement in pathology in mice lacking the enzyme. However, the ability of 12/15-LOX to directly regulate B cell function has not been studied. Methods. The influence of 12/15-LOX on B cell phenotype and function, and IgM generation, was compared using wildtype (WT) and 12/15-LOX (Alox15-/-) deficient mice. The proliferative and functional capacity of splenic CD19+ B cells was measured in vitro in response to various toll-like receptor agonists. Results. WT and Alox15-/- displayed comparable responses. However in vivo, splenic B cell numbers were significantly elevated in Alox15-/- mice with a corresponding elevation in titres of total IgM in lung, gut and serum, and lower serum IgM directed against the 12/15-LOX product, 12-hydroxyeicosatetraenoic acid-phosphatidylethanolamine (HETE-PE). Discussion. Myeloid 12/15-LOX can regulate B cell numbers and innate immune antibody levels in vivo, potentially contributing to its ability to regulate inflammatory disease. Furthermore, the alterations seen in 12/15-LOX deficiency likely result from changes in the equilibrium of the immune system that develop from birth. Further studies in disease models are warranted to elucidate the contribution of 12/15-LOX mediated alterations in B cell numbers and innate immune antibody generation to driving inflammation in vivo.

8.
J Biol Chem ; 291(26): 13448-64, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27129261

ABSTRACT

Eicosanoids are important mediators of fever, pain, and inflammation that modulate cell signaling during acute and chronic disease. We show by using lipidomics that thrombin-activated human platelets generate a new type of eicosanoid that both stimulates and primes human neutrophil integrin (Mac-1) expression, in response to formylmethionylleucylphenylalanine. Detailed characterization proposes a dioxolane structure, 8-hydroxy-9,11-dioxolane eicosatetraenoic acid (dioxolane A3, DXA3). The lipid is generated in nanogram amounts by platelets from endogenous arachidonate during physiological activation, with inhibition by aspirin in vitro or in vivo, implicating cyclooxygenase-1 (COX). Pharmacological and genetic studies on human/murine platelets revealed that DXA3 formation requires protease-activated receptors 1 and 4, cytosolic phospholipase A2 (cPLA2), Src tyrosine kinases, p38 MAPK, phospholipase C, and intracellular calcium. From data generated by purified COX isoforms and chemical oxidation, we propose that DXA3 is generated by release of an intermediate from the active site followed by oxygenation at C8. In summary, a new neutrophil-activating platelet-derived lipid generated by COX-1 is presented that can activate or prime human neutrophils, suggesting a role in innate immunity and acute inflammation.


Subject(s)
Blood Platelets/enzymology , Cyclooxygenase 1/metabolism , Dioxolanes/metabolism , MAP Kinase Signaling System/physiology , Neutrophil Activation/physiology , Neutrophils/metabolism , Animals , Aspirin/pharmacology , Blood Platelets/immunology , Cyclooxygenase 1/immunology , Dioxolanes/immunology , Immunity, Innate/drug effects , Immunity, Innate/physiology , MAP Kinase Signaling System/drug effects , Macrophage-1 Antigen/immunology , Macrophage-1 Antigen/metabolism , Male , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Neutrophil Activation/drug effects , Neutrophils/immunology , Platelet Activation/drug effects , Platelet Activation/physiology
9.
FASEB J ; 27(9): 3672-82, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23729592

ABSTRACT

Mild dietary zinc deprivation in humans and rodents has little effect on blood plasma zinc levels, and yet cellular consequences of zinc depletion can be detected in vascular and other tissues. We proposed that a zinc-regulated humoral factor might mediate the effects of zinc deprivation. Using a novel approach, primary rat vascular smooth muscle cells (VSMCs) were treated with plasma from zinc-deficient (<1 mg Zn/kg) or zinc-adequate (35 mg Zn/kg, pair-fed) adult male rats, and zinc levels were manipulated to distinguish direct and indirect effects of plasma zinc. Gene expression changes were analyzed by microarray and qPCR, and incubation of VSMCs with blood plasma from zinc-deficient rats strongly changed the expression of >2500 genes, compared to incubation of cells with zinc-adequate rat plasma. We demonstrated that this effect was caused by a low-molecular-weight (∼2-kDa) zinc-regulated humoral factor but that changes in gene expression were mostly reversed by adding zinc back to zinc-deficient plasma. Strongly regulated genes were overrepresented in pathways associated with immune function and development. We conclude that zinc deficiency induces the production of a low-molecular-weight humoral factor whose influence on VSMC gene expression is blocked by plasma zinc. This factor is therefore under dual control by zinc.


Subject(s)
Zinc/blood , Zinc/pharmacology , Animals , Body Weight/drug effects , Cells, Cultured , Eating/drug effects , Immunity, Humoral/drug effects , Male , Molecular Weight , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Polymerase Chain Reaction , Rats , Zinc/deficiency
10.
Cardiovasc Res ; 99(3): 525-34, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23667188

ABSTRACT

AIMS: Dietary zinc deficiency has been associated with the development of atherosclerosis although the effects on vascular smooth muscle cells (VSMCs), important in maintaining atherosclerotic plaque integrity, are unknown. The main aim of this study was to elucidate the effect of a zinc-deficient environment on VSMCs using an in vivo model. METHODS AND RESULTS: Rats were maintained for 2 weeks on a marginally zinc-deficient diet which resulted in a significant reduction in plasma zinc levels. Large arteries from zinc-deficient rats had significantly increased apoptosis within the VSMC layers compared with arteries from rats on a zinc-adequate diet. This apoptosis occurred in parallel with a known apoptotic pathway, namely dephosphorylation of the pro-apoptotic protein Bcl-2-associated death promoter protein (BAD). Activation of extracellular signal-regulated kinase (ERK)1/2, which maintains BAD phosphorylation as a pro-survival mechanism, was decreased in arteries from zinc-deficient rats. The mechanisms of this in vivo effect were investigated in vitro. Cultured rat VSMCs incubated with plasma from zinc-deficient rats similarly resulted in increased apoptosis in parallel with BAD dephosphorylation and decreased ERK1/2 activation. Further related apoptotic mechanisms induced by plasma from zinc-deficient rats involved a prolonged rise in [Ca²âº]i leading to subsequent activation of the phosphatase calcineurin. Calcineurin activation was required to dephosphorylate BAD. In addition, an increase in oxidative stress contributed to the apoptotic effect induced by plasma from zinc-deficient rats. CONCLUSION: In conclusion, a marginally zinc-deficient diet is pro-apoptotic for VSMCs and this may contribute to cardiovascular disease.


Subject(s)
Carotid Artery Diseases/etiology , Carotid Artery Diseases/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Zinc/deficiency , Animals , Apoptosis/physiology , Calcineurin/metabolism , Calcium/metabolism , Carotid Artery Diseases/metabolism , Cells, Cultured , Disease Models, Animal , MAP Kinase Signaling System , Oxidative Stress , Phosphorylation , Rats , Zinc/blood , bcl-Associated Death Protein/metabolism
11.
Neurosci Lett ; 468(3): 326-9, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19914344

ABSTRACT

Some of the analgesic and antinociceptive properties of the endocannabinoid anandamide can be explained by modulation of voltage-activated ion channels. However, the products of anandamide metabolism by fatty acid amide hydroxylase may also contribute to the altered excitability of sensory neurones. Ethanolamine is a product of metabolism of acylethanolamines including anandamide. In this study whole cell patch clamp recording and fura-2 Ca(2+) imaging techniques were used to characterize its actions on neonatal rat cultured dorsal root ganglion neurones. Ethanolamine (1muM) increased the mean Ca(2+) transient produced by 1mM caffeine and modulated Ca(2+) transients evoked by 60mM KCl. Thapsigargicin (500nM) inhibited the ethanolamine-evoked enhancement of Ca(2+) transients evoked by depolarisation. Voltage-activated K(+) currents were evoked from a holding potential of -70mV by voltage step commands to 0mV. Acute application of 1muM ethanolamine produced irreversible current modulation. However, application of 100nM ethanolamine reversibly increased or decreased K(+) currents. These effects of ethanolamine on voltage-activated K(+) currents were not sensitive to continual application of thapsigargicin. When applied alone thapsigargicin (500nM) had no action on the mean K(+) current. In conclusion, ethanolamine may play distinct roles in the modulation of sensory neurone excitability by acting via different mechanisms to modulate K(+) channels and a component of intracellular Ca(2+) signalling. These data suggest that in a therapeutic context it may be difficult to predict the consequences of manipulating anandamide levels.


Subject(s)
Ethanolamine/metabolism , Sensory Receptor Cells/physiology , Animals , Animals, Newborn , Calcium Signaling , Cells, Cultured , Ethanolamine/pharmacology , Ganglia, Spinal/cytology , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/physiology , Potassium Chloride/pharmacology , Rats , Sensory Receptor Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...