Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 48
Filtrer
1.
J Clin Invest ; 131(24)2021 12 15.
Article de Anglais | MEDLINE | ID: mdl-34699386

RÉSUMÉ

Chronic inflammation is a hallmark of atherosclerosis and results from an imbalance between proinflammatory and proresolving signaling. The human GPR32 receptor, together with the ALX/FPR2 receptor, transduces biological actions of several proresolving mediators that stimulate resolution of inflammation. However, since no murine homologs of the human GPR32 receptor exist, comprehensive in vivo studies are lacking. Using human atherosclerotic lesions from carotid endarterectomies and creating a transgenic mouse model expressing human GPR32 on a Fpr2×ApoE double-KO background (hGPR32myc×Fpr2-/-×Apoe-/-), we investigated the role of GPR32 in atherosclerosis and self-limiting acute inflammation. GPR32 mRNA was reduced in human atherosclerotic lesions and correlated with the immune cell markers ARG1, NOS2, and FOXP3. Atherosclerotic lesions, necrotic core, and aortic inflammation were reduced in hGPR32mycTg×Fpr2-/-×Apoe-/- transgenic mice as compared with Fpr2-/-×Apoe-/- nontransgenic littermates. In a zymosan-induced peritonitis model, the hGPR32mycTg×Fpr2-/-×Apoe-/- transgenic mice had reduced inflammation at 4 hours and enhanced proresolving macrophage responses at 24 hours compared with nontransgenic littermates. The GPR32 agonist aspirin-triggered resolvin D1 (AT-RvD1) regulated leukocyte responses, including enhancing macrophage phagocytosis and intracellular signaling in hGPR32mycTg×Fpr2-/-×Apoe-/- transgenic mice, but not in Fpr2-/-×Apoe-/- nontransgenic littermates. Together, these results provide evidence that GPR32 regulates resolution of inflammation and is atheroprotective in vivo.


Sujet(s)
Athérosclérose , Macrophages/métabolisme , Transduction du signal/génétique , Animaux , Athérosclérose/génétique , Athérosclérose/métabolisme , Athérosclérose/prévention et contrôle , Modèles animaux de maladie humaine , Acide docosahexaénoïque/génétique , Acide docosahexaénoïque/métabolisme , Femelle , Humains , Inflammation/génétique , Inflammation/métabolisme , Inflammation/prévention et contrôle , Mâle , Souris , Souris invalidées pour les gènes ApoE , Péritonite/induit chimiquement , Péritonite/génétique , Péritonite/métabolisme , Phagocytose/génétique , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme
2.
Front Cardiovasc Med ; 8: 655869, 2021.
Article de Anglais | MEDLINE | ID: mdl-33959644

RÉSUMÉ

Objectives and Aims: Vascular smooth muscle cells (VSMCs) are key constituents of both normal arteries and atherosclerotic plaques. They have an ability to adapt to changes in the local environment by undergoing phenotypic modulation. An improved understanding of the mechanisms that regulate VSMC phenotypic changes may provide insights that suggest new therapeutic targets in treatment of cardiovascular disease (CVD). The amino-acid glutamate has been associated with CVD risk and VSMCs metabolism in experimental models, and glutamate receptors regulate VSMC biology and promote pulmonary vascular remodeling. However, glutamate-signaling in human atherosclerosis has not been explored. Methods and Results: We identified glutamate receptors and glutamate metabolism-related enzymes in VSMCs from human atherosclerotic lesions, as determined by single cell RNA sequencing and microarray analysis. Expression of the receptor subunits glutamate receptor, ionotropic, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic (AMPA)-type subunit 1 (GRIA1) and 2 (GRIA2) was restricted to cells of mesenchymal origin, primarily VSMCs, as confirmed by immunostaining. In a rat model of arterial injury and repair, changes of GRIA1 and GRIA2 mRNA level were most pronounced at time points associated with VSMC proliferation, migration, and phenotypic modulation. In vitro, human carotid artery SMCs expressed GRIA1, and selective AMPA-type receptor blocking inhibited expression of typical contractile markers and promoted pathways associated with VSMC phenotypic modulation. In our biobank of human carotid endarterectomies, low expression of AMPA-type receptor subunits was associated with higher content of inflammatory cells and a higher frequency of adverse clinical events such as stroke. Conclusion: AMPA-type glutamate receptors are expressed in VSMCs and are associated with phenotypic modulation. Patients suffering from adverse clinical events showed significantly lower mRNA level of GRIA1 and GRIA2 in their atherosclerotic lesions compared to asymptomatic patients. These results warrant further mapping of neurotransmitter signaling in the pathogenesis of human atherosclerosis.

4.
Vasc Med ; 26(1): 3-10, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33350884

RÉSUMÉ

Interleukin (IL) 6 contributes to atherosclerotic plaque development through IL6 membrane-bound (IL6R and gp130) and soluble (sIL6R and sgp130) receptors. We investigated IL6 receptor expression in carotid plaques and its correlation with circulating IL6 and soluble receptor levels. Plasma samples and carotid plaques were obtained from 78 patients in the Biobank of Karolinska Endarterectomies study. IL6, sIL6R, and sgp130 were measured in plasma and IL6, IL6R, sIL6R, GP130, and sGP130-RAPS (sGP130) gene expression assessed in carotid plaques. Correlations between plaque IL6 signaling gene expression and plasma levels were determined by Spearman's correlation. Differences in plasma and gene expression levels between patients with (n = 53) and without (n = 25) a history of a cerebral event and statin-treated (n = 65) and non-treated (n = 11), were estimated by Kruskal-Wallis. IL6 and its receptors were all expressed in carotid plaques. There was a positive, borderline significant, moderate correlation between plasma IL6 and sIL6R and the respective gene expression levels (rho 0.23 and 0.22, both p = 0.05). IL6R expression was higher in patients with a history of a cerebrovascular event compared to those without (p = 0.007). Statin-treated had higher IL6R, sIL6R, and sGP130 expression levels and plasma sIL6R compared to non-treated patients (all p < 0.05). In conclusion, all components of the IL6 signaling pathways are expressed in carotid artery plaques and IL6 and sIL6R plasma levels correlate moderately with IL6 and sIL6R. Our data suggest that IL6 signaling in the circulation might mirror the system activity in the plaque, thus adding novel perspectives to the role of IL6 signaling in atherosclerosis.


Sujet(s)
Artères carotides/métabolisme , Sténose carotidienne/métabolisme , Récepteur gp130 de cytokines/métabolisme , Interleukine-6/métabolisme , Plaque d'athérosclérose , Récepteurs à l'interleukine-6/métabolisme , Sujet âgé , Marqueurs biologiques/métabolisme , Artères carotides/chirurgie , Sténose carotidienne/sang , Sténose carotidienne/génétique , Sténose carotidienne/thérapie , Études transversales , Récepteur gp130 de cytokines/sang , Récepteur gp130 de cytokines/génétique , Endartériectomie carotidienne , Femelle , Humains , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/usage thérapeutique , Interleukine-6/sang , Interleukine-6/génétique , Mâle , Récepteurs à l'interleukine-6/sang , Récepteurs à l'interleukine-6/génétique , Transduction du signal
5.
Sci Rep ; 10(1): 19108, 2020 11 05.
Article de Anglais | MEDLINE | ID: mdl-33154409

RÉSUMÉ

The Caspase activation and recruitment domain 8 (CARD8) protein is a component of innate immunity and overexpression of CARD8 mRNA was previously identified in atherosclerosis. However, very little is known about the regulation of CARD8 in endothelial cells and atherosclerosis. The aim of this study was to investigate CARD8 in the regulation of cytokine and chemokine expression in endothelial cells. Sections of human atherosclerotic lesions and non-atherosclerotic arteries were immunostained for CARD8 protein. Expression of CARD8 was correlated to mediators of inflammation in atherosclerotic lesions using Biobank of Karolinska Endarterectomies microarray data. The CARD8 mRNA was knocked-down in human umbilical vein endothelial cells (HUVECs) in vitro, followed by quantitative RT-PCR analysis and OLINK Proteomics. Endothelial and smooth muscle cells in arterial tissue expressed CARD8 and CARD8 correlated with vWF, CD163 and the expression of inflammatory genes, such as CXCL1, CXCL6 and PDGF-A in plaque. Knock-down of CARD8 in HUVECs significantly altered proteins involved in inflammatory response, such as CXCL1, CXCL6, PDGF-A, MCP-1 and IL-6. The present study suggest that CARD8 regulate the expression of cytokines and chemokines in endothelial cells and atherosclerotic lesions, suggesting that CARD8 plays a significant role in endothelial activation.


Sujet(s)
Athérosclérose/métabolisme , Protéines adaptatrices de signalisation CARD/métabolisme , Artères carotides/métabolisme , Cellules endothéliales/métabolisme , Endothélium vasculaire/métabolisme , Inflammation/métabolisme , Protéines tumorales/métabolisme , Athérosclérose/chirurgie , Artériopathies carotidiennes/métabolisme , Artériopathies carotidiennes/chirurgie , Chimiokines/métabolisme , Cytokines/métabolisme , Endartériectomie carotidienne , Humains , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/chirurgie
6.
Cells ; 9(7)2020 07 16.
Article de Anglais | MEDLINE | ID: mdl-32708790

RÉSUMÉ

Calcific aortic valve stenosis (CAVS) is a common age-related disease characterized by active calcification of the leaflets of the aortic valve. How innate immune cells are involved in disease pathogenesis is not clear. In this study we investigate the role of the pattern recognition receptor Toll-like receptor 7 (TLR7) in CAVS, especially in relation to macrophage subtype. Human aortic valves were used for mRNA expression analysis, immunofluorescence staining, or ex vivo tissue assays. Response to TLR7 agonist in primary macrophages and valvular interstitial cells (VICs) were investigated in vitro. In the aortic valve, TLR7 correlated with M2 macrophage markers on mRNA levels. Expression was higher in the calcified part compared with the intermediate and healthy parts. TLR7+ cells were co-stained with M2-type macrophage receptors CD163 and CD206. Ex vivo stimulation of valve tissue with the TLR7 ligand imiquimod significantly increased secretion of IL-10, TNF-α, and GM-CSF. Primary macrophages responded to imiquimod with increased secretion of IL-10 while isolated VICs did not respond. In summary, in human aortic valves TLR7 expression is associated with M2 macrophages markers. Ex vivo tissue challenge with TLR7 ligand led to secretion of immunomodulatory cytokine IL-10. These results connect TLR7 activation in CAVS to reduced inflammation and improved clearance.


Sujet(s)
Sténose aortique/métabolisme , Valve aortique/anatomopathologie , Calcinose/métabolisme , Macrophages/métabolisme , Récepteur de type Toll-7/métabolisme , Valve aortique/métabolisme , Marqueurs biologiques/métabolisme , Cellules cultivées , Cytokines/métabolisme , Humains , Imiquimod/pharmacologie , Ligands , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme , Récepteur de type Toll-7/agonistes
7.
Circ Res ; 126(5): 571-585, 2020 02 28.
Article de Anglais | MEDLINE | ID: mdl-31893970

RÉSUMÉ

RATIONALE: PCSKs (Proprotein convertase subtilisins/kexins) are a protease family with unknown functions in vasculature. Previously, we demonstrated PCSK6 upregulation in human atherosclerotic plaques associated with smooth muscle cells (SMCs), inflammation, extracellular matrix remodeling, and mitogens. OBJECTIVE: Here, we applied a systems biology approach to gain deeper insights into the PCSK6 role in normal and diseased vessel wall. METHODS AND RESULTS: Genetic analyses revealed association of intronic PCSK6 variant rs1531817 with maximum internal carotid intima-media thickness progression in high-cardiovascular risk subjects. This variant was linked with PCSK6 mRNA expression in healthy aortas and plaques but also with overall plaque SMA+ cell content and pericyte fraction. Increased PCSK6 expression was found in several independent human cohorts comparing atherosclerotic lesions versus healthy arteries, using transcriptomic and proteomic datasets. By immunohistochemistry, PCSK6 was localized to fibrous cap SMA+ cells and neovessels in plaques. In human, rat, and mouse intimal hyperplasia, PCSK6 was expressed by proliferating SMA+ cells and upregulated after 5 days in rat carotid balloon injury model, with positive correlation to PDGFB (platelet-derived growth factor subunit B) and MMP (matrix metalloprotease) 2/MMP14. Here, PCSK6 was shown to colocalize and cointeract with MMP2/MMP14 by in situ proximity ligation assay. Microarrays of carotid arteries from Pcsk6-/- versus control mice revealed suppression of contractile SMC markers, extracellular matrix remodeling enzymes, and cytokines/receptors. Pcsk6-/- mice showed reduced intimal hyperplasia response upon carotid ligation in vivo, accompanied by decreased MMP14 activation and impaired SMC outgrowth from aortic rings ex vivo. PCSK6 silencing in human SMCs in vitro leads to downregulation of contractile markers and increase in MMP2 expression. Conversely, PCSK6 overexpression increased PDGFBB (platelet-derived growth factor BB)-induced cell proliferation and particularly migration. CONCLUSIONS: PCSK6 is a novel protease that induces SMC migration in response to PDGFB, mechanistically via modulation of contractile markers and MMP14 activation. This study establishes PCSK6 as a key regulator of SMC function in vascular remodeling. Visual Overview: An online visual overview is available for this article.


Sujet(s)
Myocytes du muscle lisse/métabolisme , Proprotein convertases/génétique , Serine endopeptidases/génétique , Remodelage vasculaire , Animaux , Artères carotides/métabolisme , Artères carotides/anatomopathologie , Mouvement cellulaire , Prolifération cellulaire , Cellules cultivées , Mâle , Matrix metalloproteinases/génétique , Matrix metalloproteinases/métabolisme , Souris , Souris de lignée C57BL , Myocytes du muscle lisse/physiologie , Polymorphisme de nucléotide simple , Proprotein convertases/métabolisme , Protéines proto-oncogènes c-sis/métabolisme , Rats , Rat Sprague-Dawley , Serine endopeptidases/métabolisme , Transcriptome
8.
Atherosclerosis ; 292: 215-223, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31606133

RÉSUMÉ

BACKGROUND AND AIMS: Endothelin-1 (ET-1) and arginase are both suggested to be involved in the inflammatory processes and development of endothelial dysfunction in atherosclerosis. However, information regarding the roles of ET-1 and arginase, as well as the interactions between the two in human atherosclerosis, is scarce. We investigated the expression of ET-1 and its receptors, ETA and ETB, as well as arginase in human carotid atherosclerotic plaques and determined the functional interactions between ET-1 and arginase in endothelial cells and THP-1-derived macrophages. METHODS: Carotid plaques and blood samples were retreived from patients undergoing surgery for symptomatic or asymptomatic carotid stenosis. Plaque gene and protein expression was determined and related to clinical characteristics. Functional interactions between ET-1 and arginase were investigated in endothelial cells and THP-1 cells. RESULTS: Expression of ET-1 and ETB receptors was increased in plaques from patients with symptomatic carotid artery disease. ET-1 was co-localized with arginase 1 and arginase 2 in the necrotic core, together with macrophage markers CD163 and CD68. Arginase 2, ET-1 and ETB receptors were expressed in endothelial cells as well as in smooth muscle cells in the fibrous cap. ET-1 increased arginase 2 mRNA expression and arginase activity in endothelial cells and arginase activity in macrophages. Moreover, ET-1 stimulated formation of reactive oxygen species (ROS) in THP-1-derived macrophages via an arginase-dependent mechanism. CONCLUSIONS: This is the first study that demonstrates co-localization of ET-1 and arginase 2 in human atherosclerotic plaques. ET-1 stimulated arginase 2 expression and activity in endothelial cells, as well as arginase activity and ROS formation in macrophages via an arginase-dependent mechanism. These results indicate an important interaction between the ET pathway and arginase in human atherosclerotic plaques.


Sujet(s)
Arginase/physiologie , Endothéline-1/physiologie , Plaque d'athérosclérose/métabolisme , Récepteur de l'endothéline de type B/physiologie , Arginase/biosynthèse , Cellules cultivées , Cellules endothéliales , Endothéline-1/biosynthèse , Humains
9.
JACC Basic Transl Sci ; 4(3): 304-317, 2019 Jun.
Article de Anglais | MEDLINE | ID: mdl-31312755

RÉSUMÉ

CANTOS (Canakinumab Antiinflammatory Thrombosis Outcome Study) confirmed interleukin (IL)-1ß as an appealing therapeutic target for human atherosclerosis and related complications. However, there are serious gaps in our understanding of IL-1 production in atherosclerosis. Herein the authors show that complex plaques, or plaques derived from patients with suboptimally controlled hyperlipidemia, or on no or low-intensity statin therapy, demonstrated higher recruitable IL-1ß production. Generation of mature IL-1ß was matched by IL-1α release, and both were attenuated by inhibition of NLR family pyrin domain containing 3 or caspase. These findings support the inflammasome as the main pathway for IL-1α/ß generation in atherosclerosis and high-intensity lipid-lowering therapies as primary and additional anti-IL-1-directed therapies as secondary interventions in high-risk patients.

10.
Eur Heart J ; 40(30): 2495-2503, 2019 08 07.
Article de Anglais | MEDLINE | ID: mdl-31081038

RÉSUMÉ

AIMS: Radiotherapy-induced cardiovascular disease is an emerging problem in a growing population of cancer survivors where traditional treatments, such as anti-platelet and lipid-lowering drugs, have limited benefits. The aim of the study was to investigate vascular inflammatory patterns in human cancer survivors, replicate the findings in an animal model, and evaluate whether interleukin-1 (IL-1) inhibition could be a potential treatment. METHODS AND RESULTS: Irradiated human arterial biopsies were collected during microvascular autologous free tissue transfer for cancer reconstruction and compared with non-irradiated arteries from the same patient. A mouse model was used to study the effects of the IL-1 receptor antagonist, anakinra, on localized radiation-induced vascular inflammation. We observed significant induction of genes associated with inflammasome biology in whole transcriptome analysis of irradiated arteries, a finding supported by elevated protein levels in irradiated arteries of both, pro-caspase and caspase-1. mRNA levels of inflammasome associated chemokines CCL2, CCL5 together with the adhesion molecule VCAM1, were elevated in human irradiated arteries as was the number of infiltrating macrophages. A similar pattern was reproduced in Apoe-/- mouse 10 weeks after localized chest irradiation with 14 Gy. Treatment with anakinra in irradiated mice significantly reduced Ccl2 and Ccl5 mRNA levels and expression of I-Ab. CONCLUSION: Anakinra, administered directly after radiation exposure for 2 weeks, ameliorated radiation induced sustained expression of inflammatory mediators in mice. Further studies are needed to evaluate IL-1 blockade as a treatment of radiotherapy-induced vascular disease in a clinical setting.


Sujet(s)
Artérite/prévention et contrôle , Antagoniste du récepteur à l'interleukine-1/pharmacologie , Interleukine-1/antagonistes et inhibiteurs , Lésions radiques expérimentales/prévention et contrôle , Radiothérapie/effets indésirables , Animaux , Artérite/étiologie , Chimiokine CCL2/métabolisme , Femelle , Humains , Interleukine-1/métabolisme , Souris , Souris de lignée C57BL , Tumeurs/radiothérapie , Lésions radiques expérimentales/métabolisme
11.
Eur Heart J ; 40(4): 372-382, 2019 01 21.
Article de Anglais | MEDLINE | ID: mdl-30452556

RÉSUMÉ

Aims: The E3-ligase CBL-B (Casitas B-cell lymphoma-B) is an important negative regulator of T cell activation that is also expressed in macrophages. T cells and macrophages mediate atherosclerosis, but their regulation in this disease remains largely unknown; thus, we studied the function of CBL-B in atherogenesis. Methods and results: The expression of CBL-B in human atherosclerotic plaques was lower in advanced lesions compared with initial lesions and correlated inversely with necrotic core area. Twenty weeks old Cblb-/-Apoe-/- mice showed a significant increase in plaque area in the aortic arch, where initial plaques were present. In the aortic root, a site containing advanced plaques, lesion area rose by 40%, accompanied by a dramatic change in plaque phenotype. Plaques contained fewer macrophages due to increased apoptosis, larger necrotic cores, and more CD8+ T cells. Cblb-/-Apoe-/- macrophages exhibited enhanced migration and increased cytokine production and lipid uptake. Casitas B-cell lymphoma-B deficiency increased CD8+ T cell numbers, which were protected against apoptosis and regulatory T cell-mediated suppression. IFNγ and granzyme B production was enhanced in Cblb-/-Apoe-/- CD8+ T cells, which provoked macrophage killing. Depletion of CD8+ T cells in Cblb-/-Apoe-/- bone marrow chimeras rescued the phenotype, indicating that CBL-B controls atherosclerosis mainly through its function in CD8+ T cells. Conclusion: Casitas B-cell lymphoma-B expression in human plaques decreases during the progression of atherosclerosis. As an important regulator of immune responses in experimental atherosclerosis, CBL-B hampers macrophage recruitment and activation during initial atherosclerosis and limits CD8+ T cell activation and CD8+ T cell-mediated macrophage death in advanced atherosclerosis, thereby preventing the progression towards high-risk plaques.


Sujet(s)
Athérosclérose/étiologie , Lymphocytes T CD8+/immunologie , Lymphome B/complications , Macrophages/anatomopathologie , Protéine oncogène v-cbl/métabolisme , Plaque d'athérosclérose/étiologie , Animaux , Apoptose , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Modèles animaux de maladie humaine , Humains , Lymphome B/métabolisme , Lymphome B/anatomopathologie , Macrophages/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Phénotype , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/anatomopathologie
12.
Circ Genom Precis Med ; 11(9): e002115, 2018 09.
Article de Anglais | MEDLINE | ID: mdl-30354329

RÉSUMÉ

BACKGROUND: Atherosclerosis is a chronic inflammatory disease in part caused by lipid uptake in the vascular wall, but the exact underlying mechanisms leading to acute myocardial infarction and stroke remain poorly understood. Large consortia identified genetic susceptibility loci that associate with large artery ischemic stroke and coronary artery disease. However, deciphering their underlying mechanisms are challenging. Histological studies identified destabilizing characteristics in human atherosclerotic plaques that associate with clinical outcome. To what extent established susceptibility loci for large artery ischemic stroke and coronary artery disease relate to plaque characteristics is thus far unknown but may point to novel mechanisms. METHODS: We studied the associations of 61 established cardiovascular risk loci with 7 histological plaque characteristics assessed in 1443 carotid plaque specimens from the Athero-Express Biobank Study. We also assessed if the genotyped cardiovascular risk loci impact the tissue-specific gene expression in 2 independent biobanks, Biobank of Karolinska Endarterectomy and Stockholm Atherosclerosis Gene Expression. RESULTS: A total of 21 established risk variants (out of 61) nominally associated to a plaque characteristic. One variant (rs12539895, risk allele A) at 7q22 associated to a reduction of intraplaque fat, P=5.09×10-6 after correction for multiple testing. We further characterized this 7q22 Locus and show tissue-specific effects of rs12539895 on HBP1 expression in plaques and COG5 expression in whole blood and provide data from public resources showing an association with decreased LDL (low-density lipoprotein) and increase HDL (high-density lipoprotein) in the blood. CONCLUSIONS: Our study supports the view that cardiovascular susceptibility loci may exert their effect by influencing the atherosclerotic plaque characteristics.


Sujet(s)
Athérosclérose/génétique , Maladies cardiovasculaires/génétique , Locus génétiques/génétique , Prédisposition génétique à une maladie/génétique , Polymorphisme de nucléotide simple , Protéines adaptatrices du transport vésiculaire/génétique , Sujet âgé , Allèles , Artériopathies carotidiennes/génétique , Femelle , Régulation de l'expression des gènes , Génotype , Protéines HMG/génétique , Humains , Mâle , Adulte d'âge moyen , Plaque d'athérosclérose/génétique , Protéines de répression/génétique
13.
JACC Basic Transl Sci ; 3(4): 464-480, 2018 Aug.
Article de Anglais | MEDLINE | ID: mdl-30175270

RÉSUMÉ

Clinical tools to identify individuals with unstable atherosclerotic lesions are required to improve prevention of myocardial infarction and ischemic stroke. Here, a systems-based analysis of atherosclerotic plaques and plasma from patients undergoing carotid endarterectomy for stroke prevention was used to identify molecular signatures with a causal relationship to disease. Local plasma collected in the lesion proximity following clamping prior to arteriotomy was profiled together with matched peripheral plasma. This translational workflow identified biliverdin reductase B as a novel marker of intraplaque hemorrhage and unstable carotid atherosclerosis, which should be investigated as a potential predictive biomarker for cardiovascular events in larger cohorts.

14.
Mol Ther Methods Clin Dev ; 10: 17-28, 2018 Sep 21.
Article de Anglais | MEDLINE | ID: mdl-30003117

RÉSUMÉ

Variants in the PLPP3 gene encoding for lipid phosphate phosphohydrolase 3 have been associated with susceptibility to atherosclerosis independently of classical risk factors. PLPP3 inactivates lysophosphatidic acid (LPA), a pro-inflammatory, pro-thrombotic product of phospholipase activity. Here we performed the first exploratory analysis of PLPP3, LPA, and LPA receptors (LPARs 1-6) in human atherosclerosis. PLPP3 transcript and protein were repressed when comparing plaques versus normal arteries and plaques from symptomatic versus asymptomatic patients, and they were negatively associated with risk of adverse cardiovascular events. PLPP3 localized to macrophages, smooth muscle, and endothelial cells (ECs) in plaques. LPAR 2, 5, and especially 6 showed increased expression in plaques, with LPAR6 localized in ECs and positively correlated to PLPP3. Utilizing in situ mass spectrometry imaging, LPA and its precursors were found in the plaque fibrous cap, co-localizing with PLPP3 and LPAR6. In vitro, PLPP3 silencing in ECs under LPA stimulation resulted in increased expression of adhesion molecules and cytokines. LPAR6 silencing inhibited LPA-induced cell activation, but not when PLPP3 was silenced simultaneously. Our results show that repression of PLPP3 plays a key role in atherosclerosis by promoting EC activation. Altogether, the PLPP3 pathway represents a suitable target for investigations into novel therapeutic approaches to ameliorate atherosclerosis.

15.
Circulation ; 138(16): 1693-1705, 2018 10 16.
Article de Anglais | MEDLINE | ID: mdl-29739755

RÉSUMÉ

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.


Sujet(s)
Aorte/effets des médicaments et des substances chimiques , Maladies de l'aorte/prévention et contrôle , Athérosclérose/prévention et contrôle , Acide eicosapentanoïque/pharmacologie , Lipoprotéines LDL/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Phagocytose/effets des médicaments et des substances chimiques , Plaque d'athérosclérose , Récepteurs couplés aux protéines G/agonistes , Animaux , Aorte/métabolisme , Aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/métabolisme , Maladies de l'aorte/anatomopathologie , Athérosclérose/génétique , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Cytochrome reductases/génétique , Cytochrome reductases/métabolisme , Régime occidental , Modèles animaux de maladie humaine , Acide eicosapentanoïque/analogues et dérivés , Acide eicosapentanoïque/sang , Acide eicosapentanoïque/métabolisme , Prédisposition génétique à une maladie , Humains , Macrophages/métabolisme , Macrophages/anatomopathologie , Mâle , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Nécrose , Oxidoreductases acting on sulfur group donors , Phénotype , Récepteurs aux chimiokines , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs aux lipoprotéines LDL/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
16.
Circ Res ; 122(10): 1385-1394, 2018 05 11.
Article de Anglais | MEDLINE | ID: mdl-29618596

RÉSUMÉ

RATIONALE: Regulatory T (Treg) cells suppress immune responses and have been shown to attenuate atherosclerosis. The Treg cell lineage-specification factor FOXP3 (forkhead box P3) is essential for Treg cells' ability to uphold immunologic tolerance. In humans, FOXP3 exists in several different isoforms, however, their specific role is poorly understood. OBJECTIVE: To define the regulation and functions of the 2 major FOXP3 isoforms, FOXP3fl and FOXP3Δ2, as well as to establish whether their expression is associated with the ischemic atherosclerotic disease. METHODS AND RESULTS: Human primary T cells were transduced with lentiviruses encoding distinct FOXP3 isoforms. The phenotype and function of these cells were analyzed by flow cytometry, in vitro suppression assays and RNA-sequencing. We also assessed the effect of activation on Treg cells isolated from healthy volunteers. Treg cell activation resulted in increased FOXP3 expression that predominantly was made up of FOXP3Δ2. FOXP3Δ2 induced specific transcription of GARP (glycoprotein A repetitions predominant), which functions by tethering the immunosuppressive cytokine TGF (transforming growth factor)-ß to the cell membrane of activated Treg cells. Real-time polymerase chain reaction was used to determine the impact of alternative splicing of FOXP3 in relation with atherosclerotic plaque stability in a cohort of >150 patients that underwent carotid endarterectomy. Plaque instability was associated with a lower FOXP3Δ2 transcript usage, when comparing plaques from patients without symptoms and patients with the occurrence of recent (<1 month) vascular symptoms including minor stroke, transient ischemic attack, or amaurosis fugax. No difference was detected in total levels of FOXP3 mRNA between these 2 groups. CONCLUSIONS: These results suggest that activated Treg cells suppress the atherosclerotic disease process and that FOXP3Δ2 controls a transcriptional program that acts protectively in human atherosclerotic plaques.


Sujet(s)
Épissage alternatif , Facteurs de transcription Forkhead/génétique , Plaque d'athérosclérose/métabolisme , Lymphocytes T régulateurs/métabolisme , Amaurose fugace/métabolisme , Amaurose fugace/anatomopathologie , Encéphalopathie ischémique/métabolisme , Encéphalopathie ischémique/anatomopathologie , Cellules cultivées , Facteurs de transcription Forkhead/physiologie , Régulation de l'expression des gènes , Vecteurs génétiques/pharmacologie , Humains , Cellules Jurkat , Plaque d'athérosclérose/immunologie , Plaque d'athérosclérose/anatomopathologie , Isoformes de protéines/génétique , Réaction de polymérisation en chaine en temps réel , Protéines recombinantes/métabolisme , Lymphocytes T régulateurs/anatomopathologie , Transcription génétique
17.
Eur Heart J ; 39(24): 2301-2310, 2018 06 21.
Article de Anglais | MEDLINE | ID: mdl-29562241

RÉSUMÉ

Aims: Identification and treatment of the rupture prone atherosclerotic plaque remains a challenge for reducing the burden of cardiovascular disease. The interconnection of metabolic and inflammatory processes in rupture prone plaques is poorly understood. Herein, we investigate associations between metabolite profiles, inflammatory mediators and vulnerability in carotid atherosclerotic plaques. Methods and results: We collected 159 carotid plaques from patients undergoing endarterectomy and measured 165 different metabolites in a targeted metabolomics approach. We identified a metabolite profile in carotid plaques that associated with histologically evaluated vulnerability and inflammatory mediators, as well as presence of symptoms in patients. The distinct metabolite profiles identified in high-risk and stable plaques were in line with different transcription levels of metabolic enzymes in the two groups, suggesting an altered metabolism in high-risk plaques. The altered metabolic signature in high-risk plaques was consistent with a change to increased glycolysis, elevated amino acid utilization and decreased fatty acid oxidation, similar to what is found in activated leucocytes and cancer cells. Conclusion: These results highlight a possible key role of cellular metabolism to support inflammation and a high-risk phenotype of atherosclerotic plaques. Targeting the metabolism of atherosclerotic plaques with novel metabolic radiotracers or inhibitors might therefore be valid future approaches to identify and treat the high-risk atherosclerotic plaque.


Sujet(s)
Acides aminés/métabolisme , Artériopathies carotidiennes/métabolisme , Acides gras/métabolisme , Glucose/métabolisme , Plaque d'athérosclérose/métabolisme , Sujet âgé , Artériopathies carotidiennes/chirurgie , Endartériectomie carotidienne , Femelle , Glycolyse , Humains , Inflammation , Mâle , Métabolomique , Adulte d'âge moyen , Oxydoréduction , Plaque d'athérosclérose/chirurgie , Analyse en composantes principales , Pronostic
18.
Cardiovasc Res ; 113(1): 30-39, 2017 01.
Article de Anglais | MEDLINE | ID: mdl-27864310

RÉSUMÉ

AIMS: Processes in the development of atherosclerotic lesions can lead to plaque rupture or erosion, which can in turn elicit myocardial infarction or ischaemic stroke. The aims of this study were to determine whether Toll-like receptor 7 (TLR7) gene expression levels influence patient outcome and to explore the mechanisms linked to TLR7 expression in atherosclerosis. METHODS AND RESULTS: Atherosclerotic plaques were removed by carotid endarterectomy (CEA) and subjected to gene array expression analysis (n = 123). Increased levels of TLR7 transcript in the plaques were associated with better outcome in a follow-up study over a maximum of 8 years. Patients with higher TLR7 transcript levels had a lower risk of experiencing major cardiovascular and cerebrovascular events (MACCE) during the follow-up period after CEA (hazard ratio: 2.38, P = 0.012, 95% CI 1.21-4.67). TLR7 was expressed in all plaques by T cells, macrophages and endothelial cells in capillaries, as shown by immunohistochemistry. In short-term tissue cultures, ex vivo treatment of plaques with the TLR7 ligand imiquimod elicited dose-dependent secretion of IL-10, TNF-α, GM-CSF, and IL-12/IL-23p40. This secretion was blocked with a TLR7 inhibitor. Immunofluorescent tissue analysis after TLR7 stimulation showed IL-10 expression in T cells, macrophages and vascular smooth muscle cells. TLR7 mRNA levels in the plaques were correlated with IL-10 receptor (r = 0.4031, P < 0.0001) and GM-CSF receptor A (r = 0.4354, P < 0.0001) transcripts. CONCLUSION: These findings demonstrate that TLR7 is abundantly expressed in human atherosclerotic plaques. TLR7 ligation elicits the secretion of pro-inflammatory and anti-inflammatory cytokines, and high TLR7 expression in plaques is associated with better patient outcome, suggesting that TLR7 is a potential therapeutic target for prevention of complications of atherosclerosis.


Sujet(s)
Artères carotides/métabolisme , Artériopathies carotidiennes/métabolisme , Angiopathies intracrâniennes/métabolisme , Cardiopathies/métabolisme , Plaque d'athérosclérose , Récepteur de type Toll-7/métabolisme , Sujet âgé , Artères carotides/effets des médicaments et des substances chimiques , Artères carotides/anatomopathologie , Artériopathies carotidiennes/génétique , Artériopathies carotidiennes/anatomopathologie , Artériopathies carotidiennes/chirurgie , Études cas-témoins , Cellules cultivées , Angiopathies intracrâniennes/génétique , Angiopathies intracrâniennes/anatomopathologie , Angiopathies intracrâniennes/prévention et contrôle , Cytokines/métabolisme , Survie sans rechute , Endartériectomie carotidienne , Femelle , Cardiopathies/génétique , Cardiopathies/anatomopathologie , Cardiopathies/prévention et contrôle , Humains , Médiateurs de l'inflammation/métabolisme , Estimation de Kaplan-Meier , Macrophages/métabolisme , Mâle , Adulte d'âge moyen , ARN messager/génétique , ARN messager/métabolisme , Facteurs de risque , Transduction du signal , Lymphocytes T/métabolisme , Facteurs temps , Récepteur de type Toll-7/effets des médicaments et des substances chimiques , Récepteur de type Toll-7/génétique , Transcriptome , Résultat thérapeutique
19.
Circ Res ; 120(4): 633-644, 2017 Feb 17.
Article de Anglais | MEDLINE | ID: mdl-27895035

RÉSUMÉ

RATIONALE: In the search for markers and modulators of vascular disease, microRNAs (miRNAs) have emerged as potent therapeutic targets. OBJECTIVE: To investigate miRNAs of clinical interest in patients with unstable carotid stenosis at risk of stroke. METHODS AND RESULTS: Using patient material from the BiKE (Biobank of Karolinska Endarterectomies), we profiled miRNA expression in patients with stable versus unstable carotid plaque. A polymerase chain reaction-based miRNA array of plasma, sampled at the carotid lesion site, identified 8 deregulated miRNAs (miR-15b, miR-29c, miR-30c/d, miR-150, miR-191, miR-210, and miR-500). miR-210 was the most significantly downregulated miRNA in local plasma material. Laser capture microdissection and in situ hybridization revealed a distinct localization of miR-210 in fibrous caps. We confirmed that miR-210 directly targets the tumor suppressor gene APC (adenomatous polyposis coli), thereby affecting Wnt (Wingless-related integration site) signaling and regulating smooth muscle cell survival, as well as differentiation in advanced atherosclerotic lesions. Substantial changes in arterial miR-210 were detectable in 2 rodent models of vascular remodeling and plaque rupture. Modulating miR-210 in vitro and in vivo improved fibrous cap stability with implications for vascular disease. CONCLUSIONS: An unstable carotid plaque at risk of stroke is characterized by low expression of miR-210. miR-210 contributes to stabilizing carotid plaques through inhibition of APC, ensuring smooth muscle cell survival. We present local delivery of miR-210 as a therapeutic approach for prevention of atherothrombotic vascular events.


Sujet(s)
microARN/administration et posologie , microARN/biosynthèse , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/thérapie , Animaux , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Athérosclérose/thérapie , Sténose carotidienne/métabolisme , Sténose carotidienne/anatomopathologie , Sténose carotidienne/thérapie , Cellules cultivées , Études de cohortes , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Humains , Microdissection au laser/méthodes , Mâle , Souris , Souris de lignée C57BL , Souris knockout , microARN/analyse , Plaque d'athérosclérose/anatomopathologie , Rats , Rat Sprague-Dawley , Accident vasculaire cérébral/métabolisme , Accident vasculaire cérébral/anatomopathologie , Accident vasculaire cérébral/prévention et contrôle
20.
Arterioscler Thromb Vasc Biol ; 36(9): 1947-61, 2016 09.
Article de Anglais | MEDLINE | ID: mdl-27470516

RÉSUMÉ

OBJECTIVE: Key augmented processes in atherosclerosis have been identified, whereas less is known about downregulated pathways. Here, we applied a systems biology approach to examine suppressed molecular signatures, with the hypothesis that they may provide insight into mechanisms contributing to plaque stability. APPROACH AND RESULTS: Muscle contraction, muscle development, and actin cytoskeleton were the most downregulated pathways (false discovery rate=6.99e-21, 1.66e-6, 2.54e-10, respectively) in microarrays from human carotid plaques (n=177) versus healthy arteries (n=15). In addition to typical smooth muscle cell (SMC) markers, these pathways also encompassed cytoskeleton-related genes previously not associated with atherosclerosis. SYNPO2, SYNM, LMOD1, PDLIM7, and PLN expression positively correlated to typical SMC markers in plaques (Pearson r>0.6, P<0.0001) and in rat intimal hyperplasia (r>0.8, P<0.0001). By immunohistochemistry, the proteins were expressed in SMCs in normal vessels, but largely absent in human plaques and intimal hyperplasia. Subcellularly, most proteins localized to the cytoskeleton in cultured SMCs and were regulated by active enhancer histone modification H3K27ac by chromatin immunoprecipitation-sequencing. Functionally, the genes were downregulated by PDGFB (platelet-derived growth factor beta) and IFNg (interferron gamma), exposure to shear flow stress, and oxLDL (oxidized low-density lipoprotein) loading. Genetic variants in PDLIM7, PLN, and SYNPO2 loci associated with progression of carotid intima-media thickness in high-risk subjects without symptoms of cardiovascular disease (n=3378). By eQTL (expression quantitative trait locus), rs11746443 also associated with PDLIM7 expression in plaques. Mechanistically, silencing of PDLIM7 in vitro led to downregulation of SMC markers and disruption of the actin cytoskeleton, decreased cell spreading, and increased proliferation. CONCLUSIONS: We identified a panel of genes that reflect the altered phenotype of SMCs in vascular disease and could be early sensitive markers of SMC dedifferentiation.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Autoantigènes/métabolisme , Protéines de liaison au calcium/métabolisme , Artériopathies carotidiennes/métabolisme , Protéines du cytosquelette/métabolisme , Protéines de filaments intermédiaires/métabolisme , Protéines à domaine LIM/métabolisme , Protéines des microfilaments/métabolisme , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Plaque d'athérosclérose , Cytosquelette d'actine/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Animaux , Apolipoprotéines E/déficit , Apolipoprotéines E/génétique , Athérosclérose/génétique , Athérosclérose/métabolisme , Athérosclérose/anatomopathologie , Autoantigènes/génétique , Protéines de liaison au calcium/génétique , Artères carotides/métabolisme , Artères carotides/anatomopathologie , Artères carotides/physiopathologie , Artériopathies carotidiennes/génétique , Artériopathies carotidiennes/anatomopathologie , Artériopathies carotidiennes/physiopathologie , Lésions traumatiques de l'artère carotide/génétique , Lésions traumatiques de l'artère carotide/métabolisme , Études cas-témoins , Dédifférenciation cellulaire , Cellules cultivées , Protéines du cytosquelette/génétique , Modèles animaux de maladie humaine , Régulation négative , Études d'associations génétiques , Humains , Protéines de filaments intermédiaires/génétique , Protéines à domaine LIM/génétique , Mâle , Souris knockout , Protéines des microfilaments/génétique , Adulte d'âge moyen , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/physiopathologie , Myocytes du muscle lisse/anatomopathologie , Néointima , Phénotype , Interférence par ARN , Rat Sprague-Dawley , Transduction du signal , Facteurs temps , Transfection , Vasoconstriction
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...