Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Eur J Prev Cardiol ; 31(7): 856-865, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38348689

ABSTRACT

AIMS: A thorough characterization of the relationship between elevated lipoprotein(a) [Lp(a)] and coronary artery disease (CAD) is lacking. This study aimed to quantitatively assess the association of increasing Lp(a) levels and CAD severity in a real-world population. METHODS AND RESULTS: This non-interventional, cross-sectional, LipidCardio study included patients aged ≥21 years undergoing angiography (October 2016-March 2018) at a tertiary cardiology centre, who have at least one Lp(a) measurement. The association between Lp(a) and CAD severity was determined by synergy between PCI with taxus and cardiac surgery (SYNTAX)-I and Gensini scores and angiographic characteristics. Overall, 975 patients (mean age: 69.5 years) were included; 70.1% were male, 97.5% had Caucasian ancestry, and 33.2% had a family history of premature atherosclerotic cardiovascular disease. Median baseline Lp(a) level was 19.3 nmol/L. Patients were stratified by baseline Lp(a): 72.9% had < 65 nmol/L, 21.0% had ≥100 nmol/L, 17.2% had ≥125 nmol/L, and 12.9% had ≥150 nmol/L. Compared with the normal (Lp(a) < 65 nmol/L) group, elevated Lp(a) groups (e.g. ≥ 150 nmol/L) had a higher proportion of patients with prior CAD (48.4% vs. 62.7%; P < 0.01), prior coronary revascularization (39.1% vs. 51.6%; P = 0.01), prior coronary artery bypass graft (6.0% vs. 15.1%; P < 0.01), vessel(s) with lesions (68.5% vs. 81.3%; P = 0.03), diffusely narrowed vessels (10.9% vs. 16.5%; P = 0.01) or chronic total occlusion lesions (14.3% vs. 25.2%; P < 0.01), and higher median SYNTAX-I (3.0 vs. 5.5; P = 0.01) and Gensini (10.0 vs. 16.0; P < 0.01) scores. CONCLUSION: Elevated Lp(a) was associated with a more severe presentation of CAD. Awareness of Lp(a) levels in patients with CAD may have implications in their clinical management.


Patients with coronary artery disease (CAD) suffer with progressive plaque buildup in the walls of coronary blood vessels, which restricts blood flow and may result in serious cardiovascular outcomes such as chest pain (angina) and heart attacks (myocardial infarction). In this study, we assessed whether elevated levels of lipoprotein(a) [Lp(a)­a lipoprotein found in blood] are associated with more severe illness. We observed that elevated Lp(a) was associated with a higher proportion of patients with prior CAD, prior interventions on coronary blood vessels, and more diseased blood vessels. These collectively form what is considered a 'severe' clinical presentation of CAD, meaning a greater likelihood of adverse clinical outcomes.


Subject(s)
Biomarkers , Coronary Angiography , Coronary Artery Disease , Lipoprotein(a) , Phenotype , Severity of Illness Index , Humans , Coronary Artery Disease/blood , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/diagnosis , Male , Lipoprotein(a)/blood , Female , Aged , Cross-Sectional Studies , Middle Aged , Biomarkers/blood , Up-Regulation , Risk Assessment , Risk Factors
2.
Circulation ; 142(8): 776-789, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32506925

ABSTRACT

BACKGROUND: Aortic valve stenosis (AVS), which is the most common valvular heart disease, causes a progressive narrowing of the aortic valve as a consequence of thickening and calcification of the aortic valve leaflets. The beneficial effects of omega-3 polyunsaturated fatty acids (n-3 PUFAs) in cardiovascular prevention have recently been demonstrated in a large randomized, controlled trial. In addition, n-3 PUFAs serve as the substrate for the synthesis of specialized proresolving mediators, which are known by their potent beneficial anti-inflammatory, proresolving, and tissue-modifying properties in cardiovascular disease. However, the effects of n-3 PUFA and specialized proresolving mediators on AVS have not yet been determined. The aim of this study was to identify the role of n-3 PUFA-derived specialized proresolving mediators in relation to the development of AVS. METHODS: Lipidomic and transcriptomic analyses were performed in human tricuspid aortic valves. Apoe-/- mice and wire injury in C57BL/6J mice were used as models for mechanistic studies. RESULTS: We found that n-3 PUFA incorporation into human stenotic aortic valves was higher in noncalcified regions compared with calcified regions. Liquid chromatography tandem mass spectrometry-based lipid mediator lipidomics identified that the n-3 PUFA-derived specialized proresolving mediator resolvin E1 was dysregulated in calcified regions and acted as a calcification inhibitor. Apoe-/- mice expressing the Caenorhabditis elegans Fat-1 transgene (Fat-1tg×Apoe-/-), which enables the endogenous synthesis of n-3 PUFA and increased valvular n-3 PUFA content, exhibited reduced valve calcification, lower aortic valve leaflet area, increased M2 macrophage polarization, and improved echocardiographic parameters. Finally, abrogation of the resolvin E1 receptor ChemR23 enhanced disease progression, and the beneficial effects of Fat-1tg were abolished in the absence of ChemR23. CONCLUSIONS: n-3 PUFA-derived resolvin E1 and its receptor ChemR23 emerge as a key axis in the inhibition of AVS progression and may represent a novel potential therapeutic opportunity to be evaluated in patients with AVS.


Subject(s)
Aortic Valve Disease/metabolism , Eicosapentaenoic Acid/analogs & derivatives , Receptors, Chemokine/metabolism , Signal Transduction , Animals , Aortic Valve Disease/genetics , Eicosapentaenoic Acid/genetics , Eicosapentaenoic Acid/metabolism , Female , Humans , Male , Mice , Mice, Knockout, ApoE , Receptors, Chemokine/genetics
3.
Cells ; 9(3)2020 03 11.
Article in English | MEDLINE | ID: mdl-32168892

ABSTRACT

Aortic valve stenosis (AVS), a consequence of increased fibrosis and calcification of the aortic valve leaflets, causes progressive narrowing of the aortic valve. Proteoglycans, structural components of the aortic valve, accumulate in regions with fibrosis and moderate calcification. Particularly, proteoglycan 4 (PRG4) has been identified in fibrotic parts of aortic valves. However, the role of PRG4 in the context of AVS and aortic valve calcification has not yet been determined. Here, transcriptomics, histology, and immunohistochemistry were performed in human aortic valves from patients undergoing aortic valve replacement. Human valve interstitial cells (VICs) were used for calcification experiments and RNA expression analysis. PRG4 was significantly upregulated in thickened and calcified regions of aortic valves compared with healthy regions. In addition, mRNA levels of PRG4 positively associated with mRNA for proteins involved in cardiovascular calcification. Treatment of VICs with recombinant human PRG4 enhanced phosphate-induced calcification and increased the mRNA expression of bone morphogenetic protein 2 and the runt-related transcription factor 2. In summary, PRG4 was upregulated in the development of AVS and promoted VIC osteogenic differentiation and calcification. These results suggest that an altered valve leaflet proteoglycan composition may play a role in the progression of AVS.


Subject(s)
Aortic Valve Stenosis/blood , Aortic Valve/pathology , Aortic Valve/physiopathology , Calcinosis/blood , Constriction, Pathologic/physiopathology , Proteoglycans/metabolism , Aged , Aortic Valve Stenosis/physiopathology , Calcinosis/physiopathology , Cell Differentiation , Cells, Cultured , Humans
4.
Cardiovasc Res ; 115(10): 1557-1566, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30597013

ABSTRACT

AIMS: Vascular calcification, a marker of increased cardiovascular risk, is an active process orchestrated by smooth muscle cells. Observational studies indicate that omega-3 fatty acids protect against vascular calcification, but the mechanisms are unknown. The G-protein coupled receptor ChemR23 transduces the resolution of inflammation induced by the omega-3-derived lipid mediator resolvin E1. ChemR23 also contributes to osteoblastic differentiation of stem cells and bone formation, but its role in vascular calcification is unknown. The aim of this study was to establish the role of ChemR23 in smooth muscle cell fate and calcification. METHODS AND RESULTS: Gene expression analysis in epigastric arteries derived from patients with chronic kidney disease and vascular calcification revealed that ChemR23 mRNA levels predicted a synthetic smooth muscle cell phenotype. Genetic deletion of ChemR23 in mice prevented smooth muscle cell de-differentiation. ChemR23-deficient smooth muscle cells maintained a non-synthetic phenotype and exhibited resistance to phosphate-induced calcification. Moreover, ChemR23-deficient mice were protected against vitamin D3-induced vascular calcification. Resolvin E1 inhibited smooth muscle cell calcification through ChemR23. Introduction of the Caenorhabditis elegans Fat1 transgene, leading to an endogenous omega-3 fatty acid synthesis and hence increased substrate for resolvin E1 formation, significantly diminished the differences in phosphate-induced calcification between ChemR23+/+ and ChemR23-/- mice. CONCLUSION: This study identifies ChemR23 as a previously unrecognized determinant of synthetic and osteoblastic smooth muscle cell phenotype, favouring phosphate-induced vascular calcification. This effect may be of particular importance in the absence of ChemR23 ligands, such as resolvin E1, which acts as a calcification inhibitor under hyperphosphatic conditions.


Subject(s)
Adaptation, Physiological , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Osteogenesis , Phosphates/metabolism , Receptors, Chemokine/metabolism , Vascular Calcification/metabolism , Adaptation, Physiological/drug effects , Adult , Aged , Animals , Cadherins/genetics , Cadherins/metabolism , Cholecalciferol , Disease Models, Animal , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Female , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Osteogenesis/drug effects , Rats , Receptors, Chemokine/deficiency , Receptors, Chemokine/drug effects , Receptors, Chemokine/genetics , Signal Transduction , Vascular Calcification/chemically induced , Vascular Calcification/pathology , Vascular Calcification/prevention & control
5.
Front Pharmacol ; 9: 1327, 2018.
Article in English | MEDLINE | ID: mdl-30515096

ABSTRACT

Intimal hyperplasia remains a significant clinical problem in for example coronary artery bypass graft failure. Since omega-3 fatty acids reduce intimal hyperplasia, we hypothesized that the G protein-coupled receptor ChemR23 for the omega-3-derived pro-resolving lipid mediator resolvin E1 drives those effects. ChemR23+/+ and ChemR23-/- mice were generated with or without introduction of the Caenorhabditis elegans fat-1 transgene, which leads to an endogenous omega-3 fatty acid synthesis and thus increasing the substrate for resolvin E1 formation. ChemR23 deletion significantly increased intimal hyperplasia 28 days after ligation of the left common carotid artery. Mice expressing the fat-1 transgene showed reduced intimal hyperplasia independently of ChemR23 expression. ChemR23-/- Vascular smooth muscle cells (VSMCs) exhibited a significantly lower proliferation compared with VSMCs derived from ChemR23+/+ mice. In contrast, ChemR23-/- peritoneal macrophages had significantly higher mRNA levels of pro-inflammatory cytokines compared with ChemR23+/+ macrophages. Finally, conditioned media (CM) transfer from ChemR23-/- macrophages to VSMCs significantly increased VSMC proliferation compared with CM from ChemR23+/+ macrophages. Taken together, these results point to a dual effect of ChemR23 in resolution pharmacology by directly stimulating VSMC proliferation and at the same time suppressing macrophage-induced VSMC proliferation. In conclusion, these differential effects of ChemR23 signaling in VSMC and macrophages open up a novel notion for intimal hyperplasia pathophysiology, where ChemR23-transduced effects on the vascular wall may vary, and even be opposing, depending on the degrees of resolution of inflammation.

6.
Circulation ; 138(16): 1693-1705, 2018 10 16.
Article in English | MEDLINE | ID: mdl-29739755

ABSTRACT

BACKGROUND: In addition to enhanced proinflammatory signaling, impaired resolution of vascular inflammation plays a key role in atherosclerosis. Proresolving lipid mediators formed through the 12/15 lipoxygenase pathways exert protective effects against murine atherosclerosis. n-3 Polyunsaturated fatty acids, including eicosapentaenoic acid (EPA), serve as the substrate for the formation of lipid mediators, which transduce potent anti-inflammatory and proresolving actions through their cognate G-protein-coupled receptors. The aim of this study was to identify signaling pathways associated with EPA supplementation and lipid mediator formation that mediate atherosclerotic disease progression. METHODS: Lipidomic plasma analysis were performed after EPA supplementation in Apoe-/- mice. Erv1/Chemr23-/- xApoe-/- mice were generated for the evaluation of atherosclerosis, phagocytosis, and oxidized low-density lipoprotein uptake. Histological and mRNA analyses were done on human atherosclerotic lesions. RESULTS: Here, we show that EPA supplementation significantly attenuated atherosclerotic lesion growth induced by Western diet in Apoe-/- mice and was associated with local cardiovascular n-3 enrichment and altered lipoprotein metabolism. Our systematic plasma lipidomic analysis identified the resolvin E1 precursor 18-monohydroxy EPA as a central molecule formed during EPA supplementation. Targeted deletion of the resolvin E1 receptor Erv1/Chemr23 in 2 independent hyperlipidemic murine models was associated with proatherogenic signaling in macrophages, increased oxidized low-density lipoprotein uptake, reduced phagocytosis, and increased atherosclerotic plaque size and necrotic core formation. We also demonstrate that in macrophages the resolvin E1-mediated effects in oxidized low-density lipoprotein uptake and phagocytosis were dependent on Erv1/Chemr23. When analyzing human atherosclerotic specimens, we identified ERV1/ChemR23 expression in a population of macrophages located in the proximity of the necrotic core and demonstrated augmented ERV1/ChemR23 mRNA levels in plaques derived from statin users. CONCLUSIONS: This study identifies 18-monohydroxy EPA as a major plasma marker after EPA supplementation and demonstrates that the ERV1/ChemR23 receptor for its downstream mediator resolvin E1 transduces protective effects in atherosclerosis. ERV1/ChemR23 signaling may represent a previously unrecognized therapeutic pathway to reduce atherosclerotic cardiovascular disease.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Eicosapentaenoic Acid/pharmacology , Lipoproteins, LDL/metabolism , Macrophages/drug effects , Phagocytosis/drug effects , Plaque, Atherosclerotic , Receptors, G-Protein-Coupled/agonists , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cytochrome Reductases/genetics , Cytochrome Reductases/metabolism , Diet, Western , Disease Models, Animal , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/blood , Eicosapentaenoic Acid/metabolism , Genetic Predisposition to Disease , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Necrosis , Oxidoreductases Acting on Sulfur Group Donors , Phenotype , Receptors, Chemokine , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Signal Transduction/drug effects
7.
Sci Rep ; 7(1): 15724, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29146976

ABSTRACT

Obesity comorbidities are closely associated with chronic low-grade adipose tissue inflammation. A number of SNPs associated with inflammation has been identified, underscoring the impact of genetic determinants on this process. Here, we screened SNPs in genes with pro-inflammatory (IL-1ß, IL-6, STAT3 and JAK2), anti-inflammatory (IL-10 and SOCS3) and pro-resolving (ERV1/ChemR23) properties in 101 obese and 99 non-obese individuals. Among the SNPs analyzed, we identified that individuals carrying a C allele in the rs1878022 polymorphism of the ERV1/ChemR23 gene, which encodes for the receptor of the pro-resolving mediator RvE1, had increased ERV1/ChemR23 protein expression and reduced levels of the inflammatory cytokine IL-6 in adipose tissue. Moreover, patients carrying the C allele in homozygosity had lower plasma levels of IL-6, IFN-α2, IL-15, IL-1ra, IL-10, GM-CSF, G-CSF and VEGF and enhanced leukocyte responsiveness to RvE1. C-carriers also exhibited decreased TAG to HDL ratio, a surrogate marker of insulin resistance and a predictor of incident fatty liver. Finally, we confirmed in vivo that the ERV1/ChemR23 receptor regulates systemic and tissue inflammation since mice lacking ERV1/ChemR23 expression showed increased IL-6 levels in adipose tissue and peritoneal macrophages. Together, our study identified an ERV1/ChemR23 variant that protects patients with obesity from excessive inflammatory burden.


Subject(s)
Genetic Association Studies , Inflammation/genetics , Intra-Abdominal Fat/pathology , Obesity, Morbid/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Chemokine/genetics , Animals , Female , Gene Frequency/genetics , Homozygote , Humans , Inheritance Patterns/genetics , Interleukin-6/metabolism , Liver/pathology , Male , Mice , Middle Aged , Models, Genetic , Omentum/pathology
8.
Br J Pharmacol ; 174(22): 4043-4054, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28071789

ABSTRACT

BACKGROUND AND PURPOSE: Atherosclerosis is characterized by a chronic non-resolving inflammation in the arterial wall. Aspirin-triggered lipoxin A4 (ATL) is a potent anti-inflammatory mediator, involved in the resolution of inflammation. However, the therapeutic potential of immune targeting by means of ATL in atherosclerosis has not previously been explored. The aim of the present study was to determine the effects of ATL and its receptor Fpr2 on atherosclerosis development and progression in apolipoprotein E deficient (ApoE-/- ) mice. EXPERIMENTAL APPROACH: ApoE-/-  × Fpr2+/+ and ApoE-/-  × Fpr2-/- mice were generated. Four-week-old mice fed a high-fat diet for 4 weeks and 16-week-old mice fed chow diet received osmotic pumps containing either vehicle or ATL for 4 weeks. Atherosclerotic lesion size and cellular composition were measured in the aortic root and thoracic aorta. Lipid levels and leukocyte counts were measured in blood and mRNA was isolated from abdominal aorta and spleen. KEY RESULTS: ATL blocked atherosclerosis progression in the aortic root and thoracic aorta of ApoE-/- mice. In addition, ATL reduced macrophage infiltration and apoptotic cells in atherosclerotic lesions. The mRNA levels of several cytokines and chemokines in the spleen and aorta were reduced by ATL, whereas circulating leukocyte levels were unchanged. The ATL-induced athero-protection was absent in ApoE-/- mice lacking the Fpr2 receptor. CONCLUSION AND IMPLICATIONS: ATL blocked atherosclerosis progression by means of an Fpr2-mediated reduced local and systemic inflammation. These results suggest this anti-inflammatory and pro-resolving agent has therapeutic potential for the treatment of atherosclerosis. LINKED ARTICLES: This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.


Subject(s)
Apolipoproteins E/genetics , Aspirin/therapeutic use , Atherosclerosis/drug therapy , Lipoxins/therapeutic use , Receptors, Formyl Peptide/genetics , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aspirin/pharmacology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cytokines/genetics , Female , Lipoxins/pharmacology , Mice, Knockout
9.
Eur Heart J ; 37(47): 3532-3535, 2016 Dec 14.
Article in English | MEDLINE | ID: mdl-27091952

ABSTRACT

AIMS: Aortic valve stenosis (AS) is the most common valvulopathy and is characterized by inflammation, extracellular matrix (ECM) remodelling and calcification, causing a narrowing of the valve and the consequential obstruction of the cardiac outflow. Although intraleaflet haemorrhage is associated with AS progression, the mechanisms involved are not known. The aims of this study were to identify valvular iron in relation to pathological changes associated with AS and the effects on valvular interstitial cells (VIC) in terms of iron uptake and iron-induced responses. METHODS AND RESULTS: Valvular iron accumulation was detected by Perls' staining on aortic valve sections and shown to increase with the extent of calcification. Furthermore, qRT-PCR analysis revealed that iron-containing valve regions exhibited increased expression of genes involved in ECM remodelling and calcification. In addition, we demonstrate that iron transporters are regulated by pathways with major impact on AS and that VIC can take up and accumulate iron, which resulted in increased proliferation and decreased elastin production. CONCLUSION: Iron, which may accumulate in the aortic valve by means of intraleaflet haemorrhages, can be taken up by VIC in a pro-inflammatory environment and actively contribute to VIC proliferation, ECM remodelling and calcification. These findings suggest a possible mechanism through which iron uptake by VIC may favour AS progression.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Calcinosis , Heart Valve Diseases , Humans , Iron
10.
Vascul Pharmacol ; 73: 104-14, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26028607

ABSTRACT

Oxidized low density lipoproteins (oxLDLs) act as an etiological factor in the development of atherosclerosis by modifying the biological properties of endothelial cells through mechanisms of vascular inflammation. To deepen the oxLDL changes at cellular level, a transcriptomic analysis of human umbilical artery endothelial cells (HUAECs) treated with oxLDL was performed to identify the modified signaling pathways. Total RNA was isolated from HUAECs treated with oxLDL (100 µg/ml). Gene expression analysis was carried out using Affymetrix oligonucleotide microarrays. Biological pathway analysis was performed using Ingenuity Pathway Analysis software. Microarray assay demonstrated that oxLDL strongly affected two metabolic and signaling transduction pathways: the biosynthesis of steroids, via modification of nine genes that act sequentially in this metabolic pathway, and the Jak-Stat signaling pathway acting through STAT1 and STAT2. By means of qPCR, immunoblot, RNA interference and inhibitors we demonstrate that the mechanism used by oxLDL to activate Jak-Stat signaling pathway in artery endothelial cells is mainly mediated by STAT1. These findings provide a new mechanistic framework to better understand the effects that oxLDLs exert in artery endothelial cell gene expression and provide a source of hypothesis to understand the involvement of oxLDL in diseases in which endothelial cells play a key role, such as atherosclerosis.


Subject(s)
Human Umbilical Vein Endothelial Cells/drug effects , Janus Kinase 2/metabolism , Lipoproteins, LDL/pharmacology , STAT1 Transcription Factor/metabolism , Transcriptome/drug effects , Blotting, Western , Cells, Cultured , Gene Expression Profiling/methods , Gene Expression Regulation, Enzymologic , Gene Regulatory Networks , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Oligonucleotide Array Sequence Analysis , Protein Kinase Inhibitors/pharmacology , RNA Interference , Real-Time Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Signal Transduction/drug effects , Time Factors , Transfection
11.
Cardiovasc Res ; 106(2): 295-302, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25750192

ABSTRACT

AIMS: Atherosclerosis is a chronic inflammatory disease that is initiated by the retention and accumulation of low-density lipoprotein in the artery, leading to maladaptive response of cells from the immune system and vessel wall. Strong evidence implicates indoleamine 2,3-dioxygenase (IDO), the first and rate-limiting enzyme of the kynurenine pathway of tryptophan (Trp) degradation, with immune regulation and anti-inflammatory mechanisms in different diseases. However, the role of IDO and the endogenous degradation of Trp have never been directly examined in atherosclerosis development. We used the IDO inhibitor 1-methyl-Trp (1-MT) to determine the role of IDO-mediated Trp metabolism in vascular inflammation and atherosclerosis. METHODS AND RESULTS: Apoe(-/-) mice were treated with 1-MT in drinking water for 8 weeks. Systemic IDO inhibition led to a significant increase in atherosclerotic lesions that were ∼58 and 54% larger in the aortic arch and root, respectively. 1-MT treatment enhanced vascular inflammation, up-regulated VCAM-1 and CCL2, and increased CD68 macrophage accumulation into the plaque. Notably, the rise in VCAM-1 expression was not limited to the plaque but also found in smooth muscle cells (SMCs) of the tunica media. Furthermore, we found that IDO-dependent Trp metabolism by SMCs regulates VCAM-1 expression, and that 1-MT-induced acceleration of atherosclerosis and vascular inflammation can be reversed by exogenous administration of the Trp metabolite 3-hydroxyanthranilic acid (3-HAA). CONCLUSION: IDO-mediated Trp metabolism regulates vascular inflammation and plaque formation in hypercholesterolaemic Apoe(-/-) mice. Our data establish that this pathway plays a major role in the pathological process of atherogenesis.


Subject(s)
Atherosclerosis/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/metabolism , Tryptophan/analogs & derivatives , Animals , Apolipoproteins E/genetics , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Inflammation/drug therapy , Inflammation/metabolism , Mice, Knockout , Tryptophan/pharmacology , Tunica Media/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
13.
Cardiovasc Res ; 105(1): 65-74, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25341894

ABSTRACT

AIMS: The formyl peptide receptor (FPR) subtype FPR2/ALX transduces pro-inflammatory responses and participates in the resolution of inflammation depending on activation. The aim of the present study was to unravel the role of FPR2/ALX signalling in atherosclerosis. METHODS AND RESULTS: Expression of FPR2/ALX was analysed in 127 human carotid atherosclerotic lesions and revealed that this receptor was expressed on macrophages, smooth muscle cells (SMCs), and endothelial cells. Furthermore, FPR2/ALX mRNA levels were significantly up-regulated in atherosclerotic lesions compared with healthy vessels. In multiple regression, age, creatinine, and clinical signs of increased cerebral ischaemia were independent predictors of FPR2/ALX expression. To provide mechanistic insights into these observations, we generated Ldlr(-/-)xFpr2(-/-) mice, which exhibited delayed atherosclerosis development and less macrophage infiltration compared with Ldlr(-/-)xFpr2(+/+) mice. These findings were reproduced by transplantation of Fpr2(-/-) bone marrow into Ldlr(-/-) mice and further extended by in vitro experiments, demonstrating a lower inflammatory state in Fpr2(-/-) macrophages. FPR2/ALX expression correlated with chemo- and cytokines in human atherosclerotic lesions and leucocytes. Finally, atherosclerotic lesions in Ldlr(-/-)xFpr2(-/-) mice exhibited decreased collagen content, and Fpr2(-/-) SMCs exhibited a profile of increased collagenase and decreased collagen production pathways. CONCLUSION: FPR2/ALX is proatherogenic due to effects on bone marrow-derived cells, but promoted a more stable plaque phenotype through effects on SMCs. Taken together, these results suggest a dual role of FPR2/ALX signalling in atherosclerosis by way of promoting disease progression and but increasing plaque stability.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Aged , Aged, 80 and over , Animals , Atherosclerosis/genetics , Bone Marrow Transplantation , Carotid Artery Diseases/etiology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/metabolism , Carotid Stenosis/pathology , Collagen/metabolism , Female , Humans , Macrophages/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Formyl Peptide/deficiency , Receptors, Formyl Peptide/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, LDL/metabolism , Receptors, Lipoxin/genetics , Signal Transduction , Up-Regulation
14.
Bone ; 65: 33-41, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24815918

ABSTRACT

Osteoporosis is a multifactorial skeletal pathology with a main genetic component. To date, however, the majority of genes associated with this pathology remain unknown since genes cataloged to date only explain a part of the heritability of bone phenotypes. In the present study, we have used a genome-wide gene expression approach by means of microarrays to identify new candidate genes involved in the physiopathology of osteoporosis, using as a model the ovariectomized (OVX) mice by comparing global bone marrow gene expression of the OVX mice with those of SHAM operated mice. One hundred and eighty transcripts were found to be differentially expressed between groups. The analysis showed 23 significant regulatory networks, of which the top five canonical pathways included B-cell development, primary immunodeficiency signaling, PI3K signaling in B-cells, phospholipase C signaling, and FcgRIIB signaling in B-cells. Twelve differentially expressed genes were validated by MALDI-TOF mass spectrometry with good reproducibility. Finally, the association to bone phenotypes of SNPs in genes whose expression was increased (IL7R and CD79A) or decreased (GPX3 and IRAK3) by OVX in mice was analyzed in a cohort of 706 postmenopausal women. We detected an association of a SNP in a gene involved in the detoxification of free radicals like glutathione peroxidase 3 (GPX3) with femoral neck BMD (rs8177447, P=0.043) and two SNPs in the Ig-alpha protein of the B-cell antigen component gene (CD79A) with lumbar spine BMD (rs3810153 and rs1428922, P=0.016 and P=0.001, respectively). These results reinforce the role of antioxidant pathways and of B-cells in bone metabolism. Furthermore, it shows that a genome-wide gene expression approach in animal models is a useful method for detecting genes associated to BMD and osteoporosis risk in humans.


Subject(s)
Bone Marrow/metabolism , Gene Expression Profiling , Osteoporosis/genetics , Animals , Bone Density , Female , Humans , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide
15.
Mol Cell Endocrinol ; 365(1): 11-6, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-22982060

ABSTRACT

Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide (NO) synthase. ADMA accumulation, mainly due to a decreased dimethylarginine dimethylaminohydrolase (DDAH) activity, has been related to the development of cardiovascular diseases. We investigate whether estradiol prevents the changes induced by oxidized low density lipoprotein (oxLDL) on the DDAH/ADMA/NO pathway in human umbilical artery endothelial cells (HUAEC). HUAEC were exposed to estradiol, native LDL (nLDL), oxLDL and their combinations for 24 h. In some experiments, cells were also exposed to the unspecific estrogen receptor (ER) antagonist ICI 182780, the specific ERα antagonist MPP or specific agonists for ERα, ERß and GPER. ADMA concentration was measured by HPLC and concentration of NO by amperometry. Protein expression and DDAH activity were measured by immunoblotting and an enzymatic method, respectively. oxLDL, but not nLDL, increased ADMA concentration with a concomitant decrease on DDAH activity. oxLDL reduced eNOS protein and NO production. Estradiol alone had no effects on DDAH/ADMA/NO pathway, but increased the attenuated endothelial NO production induced by oxLDL by reduction in ADMA and preventing loss of eNOS protein levels. ICI 182780 and MPP completely abolished these effects of estradiol on oxLDL-exposed cells. ERα agonist, but not ERß and GPER agonists, mirrored estradiol effects on NO production. In conclusion, estradiol restores (1) DDAH activity, and therefore ADMA levels, and (2) NO production impaired by oxLDL in HUAEC acting through ERα.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Endothelium, Vascular/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Lipoproteins, LDL/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/agonists , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Arginine/adverse effects , Arginine/analogs & derivatives , Arginine/antagonists & inhibitors , Arginine/metabolism , Blotting, Western , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Estradiol/agonists , Estradiol/chemistry , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Estrogens, Non-Steroidal/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Lipoproteins, LDL/adverse effects , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/chemistry , Protein Stability , Protein-Arginine N-Methyltransferases/metabolism , Receptors, Estrogen/agonists , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Repressor Proteins/metabolism , Umbilical Arteries/cytology
16.
Circulation ; 124(5): 633-41, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21768538

ABSTRACT

BACKGROUND: Upregulated by atheroprotective flow, the transcription factor Krüppel-like factor 2 (KLF2) is crucial for maintaining endothelial function. MicroRNAs (miRNAs) are noncoding small RNAs that regulate gene expression at the posttranscriptional level. We examined the role of miRNAs, particularly miR-92a, in the atheroprotective flow-regulated KLF2. METHODS AND RESULTS: Dicer knockdown increased the level of KLF2 mRNA in human umbilical vein endothelial cells, suggesting that KLF2 is regulated by miRNA. In silico analysis predicted that miR-92a could bind to the 3' untranslated region of KLF2 mRNA. Overexpression of miR-92a decreased the expression of KLF2 and the KLF2-regulated endothelial nitric oxide synthase and thrombomodulin at mRNA and protein levels. A complementary finding is that miR-92a inhibitor increased the mRNA and protein expression of KLF2, endothelial nitric oxide synthase, and thrombomodulin. Subsequent studies revealed that atheroprotective laminar flow downregulated the level of miR-92a precursor to induce KLF2, and the level of this flow-induced KLF2 was reduced by miR-92a precursor. Furthermore, miR-92a level was lower in human umbilical vein endothelial cells exposed to the atheroprotective pulsatile shear flow than under atheroprone oscillatory shear flow. Anti-Ago1/2 immunoprecipitation coupled with real-time polymerase chain reaction revealed that pulsatile shear flow decreased the functional targeting of miR-92a precursor/KLF2 mRNA in human umbilical vein endothelial cells. Consistent with these findings, mouse carotid arteries receiving miR-92a precursor exhibited impaired vasodilatory response to flow. CONCLUSIONS: Atheroprotective flow patterns decrease the level of miR-92a, which in turn increases KLF2 expression to maintain endothelial homeostasis.


Subject(s)
Atherosclerosis/physiopathology , Endothelial Cells/physiology , Kruppel-Like Transcription Factors/genetics , MicroRNAs/physiology , Pulsatile Flow/physiology , 3' Untranslated Regions/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Endothelial Cells/cytology , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Homeostasis/physiology , Humans , Kruppel-Like Transcription Factors/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , RNA, Messenger/genetics , Stress, Mechanical , Thrombomodulin/genetics , Thrombomodulin/metabolism , Umbilical Veins/cytology , Vasodilation/physiology
17.
Mol Cell Endocrinol ; 335(2): 96-103, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-20615453

ABSTRACT

Migration and proliferation of endothelial cells are involved in re-endothelialization and angiogenesis, two important cardiovascular processes that are increased in response to estrogens. RhoA, a small GTPase which controls multiple cellular processes, is involved in the control of cell migration and proliferation. Our aim was to study the role of RhoA on estradiol-induced migration and proliferation and its dependence on estrogen receptors activity. Human umbilical vein endothelial cells were stimulated with estradiol, in the presence or absence of ICI 182780 (estrogen receptors antagonist) and Y-27632 (Rho kinase inhibitor). Estradiol increased Rho GEF-1 gene expression and RhoA (gene and protein expression and activity) in an estrogen receptor-dependent manner. Cell migration, stress fiber formation and cell proliferation were increased in response to estradiol and were also dependent on the estrogen receptors and RhoA activation. Estradiol decreased p27 levels, and significantly raised the expression of cyclins and CDK. These effects were counteracted by the use of either ICI 182780 or Y-27632. In conclusion, estradiol enhances the RhoA/ROCK pathway and increases cell cycle-related protein expression by acting through estrogen receptors. This results in an enhanced migration and proliferation of endothelial cells.


Subject(s)
Cell Movement/drug effects , Endothelial Cells/metabolism , Estradiol/pharmacology , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Assays , Female , Humans , Male , Transcriptional Activation/drug effects , Umbilical Cord/cytology , Up-Regulation/drug effects , rhoA GTP-Binding Protein/genetics
18.
J Mol Endocrinol ; 44(4): 237-46, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20110403

ABSTRACT

Estradiol (E(2)) acts on the endothelium to promote vasodilatation through the release of several compounds, including prostanoids, which are products of arachidonic acid metabolism. Among these, prostacyclin (PGI2) and thromboxane A2 (TXA2) exert opposite effects on vascular tone. The role of different estrogen receptors (ERs) in the PGI2/TXA2 balance, however, has not been fully elucidated. Our study sought to uncover whether E(2) enhances basal production of PGI2 or TXA2 in cultured human umbilical vein endothelial cells (HUVECs), to analyze the enzymatic mechanisms involved, and to evaluate the different roles of both types of ERs (ERalpha and ERbeta). HUVECs were exposed to E(2), selective ERalpha (1,3,5-tris(4-hydroxyphenyl)-4-propyl-1h-pyrazole, PPT) or ERbeta (diarylpropionitrile, DPN) agonists and antagonists (unspecific: ICI 182 780; specific for ERalpha: methyl-piperidino-pyrazole, MPP). PGI2 and TXA2 production was measured by ELISA. Expression of phospholipases, cyclooxygenases (COX-1 and COX-2), PGI2 synthase (PGIS), and thromboxane synthase (TXAS) was analyzed by western blot and quantitative RT-PCR. E(2) (1-100 nM) dose dependently increased PGI2 production (up to 50%), without affecting TXA2 production. COX-1 and PGIS protein and gene expressions were increased, whereas COX-2, phospholipases, and TXAS expression remained unaltered. All these effects were mediated through ERalpha, since they were produced not only in the presence of E(2), but also in that of PPT, while they were abolished in the presence of MPP. In conclusion, E(2), acting through ERalpha, up-regulates COX-1 and PGIS expression, thus directing prostanoid balance toward increased PGI2 production.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Epoprostenol/biosynthesis , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Cells, Cultured , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Epoprostenol/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Group IV Phospholipases A2/genetics , Group IV Phospholipases A2/metabolism , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Male , Thromboxane A2/metabolism
19.
PLoS One ; 4(12): e8242, 2009 Dec 14.
Article in English | MEDLINE | ID: mdl-20011585

ABSTRACT

Vascular effects of estradiol are being investigated because there are controversies among clinical and experimental studies. DNA microarrays were used to investigate global gene expression patterns in cultured human umbilical vein endothelial cells (HUVEC) exposed to 1 nmol/L estradiol for 24 hours. When compared to control, 187 genes were identified as differentially expressed with 1.9-fold change threshold. Supervised principal component analysis and hierarchical cluster analysis revealed the differences between control and estradiol-treated samples. Physiological concentrations of estradiol are sufficient to elicit significant changes in HUVEC gene expression. Notch signaling, actin cytoskeleton signaling, pentose phosphate pathway, axonal guidance signaling and integrin signaling were the top-five canonical pathways significantly regulated by estrogen. A total of 26 regulatory networks were identified as estrogen responsive. Microarray data were confirmed by quantitative RT-PCR in cardiovascular meaning genes; cyclooxygenase (COX)1, dimethylarginine dimethylaminohydrolase (DDAH)2, phospholipase A2 group IV (PLA2G4) B, and 7-dehydrocholesterol reductase were up-regulated by estradiol in a dose-dependent and estrogen receptor-dependent way, whereas COX2, DDAH1 and PLA2G4A remained unaltered. Moreover, estradiol-induced COX1 gene expression resulted in increased COX1 protein content and enhanced prostacyclin production. DDAH2 protein content was also increased, which in turn decreased asymmetric dimethylarginine concentration and increased NO release. All stimulated effects of estradiol on gene and protein expression were estrogen receptor-dependent, since were abolished in the presence of the estrogen receptor antagonist ICI 182780. This study identifies new vascular mechanisms of action by which estradiol may contribute to a wide range of biological processes.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Estradiol/pharmacology , Metabolic Networks and Pathways/drug effects , Vasodilation/drug effects , Amidohydrolases/genetics , Amidohydrolases/metabolism , Cells, Cultured , Cluster Analysis , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Metabolic Networks and Pathways/genetics , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Transforming Growth Factor beta1/metabolism , Umbilical Veins/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...