Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 33(5): 6099-6114, 2019 05.
Article in English | MEDLINE | ID: mdl-30753095

ABSTRACT

Our objective was to investigate the effect of cholesterol [hypercholesterolemia and 7-ketocholesterol (7K)] on endoglin (Eng) expression and regulation with respect to endothelial or vascular dysfunction in vivo and in vitro. In vivo experiments were performed in 2-mo-old atherosclerosis-prone apolipoprotein E-deficient/LDL receptor-deficient (ApoE-/-/LDLR-/-) female mice and their wild-type C57BL/6J littermates. In in vitro experiments, human aortic endothelial cells (HAECs) were treated with 7K. ApoE-/-/LDLR-/- mice developed hypercholesterolemia accompanied by increased circulating levels of P-selectin and Eng and a disruption of NO metabolism. Functional analysis of the aorta demonstrated impaired vascular reactivity, and Western blot analysis revealed down-regulation of membrane Eng/Smad2/3/eNOS signaling in ApoE-/-/LDLR-/- mice. 7K increased Eng expression via Krüppel-like factor 6 (KLF6), liver X nuclear receptor, and NF-κB in HAECs. 7K-induced Eng expression was prevented by the treatment with 2-hydroxypropyl-ß-cyclodextrin; 8-{[5-chloro-2-(4-methylpiperazin-1-yl) pyridine-4-carbonyl] amino}-1-(4-fluorophenyl)-4, 5-dihydrobenzo[g]indazole-3-carboxamide; or by KLF6 silencing. 7K induced increased adhesion and transmigration of monocytic human leukemia promonocytic cell line cells and was prevented by Eng silencing. We concluded that hypercholesterolemia altered Eng expression and signaling, followed by endothelial or vascular dysfunction before formation of atherosclerotic lesions in ApoE-/-/LDLR-/- mice. By contrast, 7K increased Eng expression and induced inflammation in HAECs, which was followed by an increased adhesion and transmigration of monocytes via endothelium, which was prevented by Eng inhibition. Thus, we propose a relevant role for Eng in endothelial or vascular dysfunction or inflammation when exposed to cholesterol.-Vicen, M., Vitverova, B., Havelek, R., Blazickova, K., Machacek, M., Rathouska, J., Najmanová, I., Dolezelova, E., Prasnicka, A., Sternak, M., Bernabeu, C., Nachtigal, P. Regulation and role of endoglin in cholesterol-induced endothelial and vascular dysfunction in vivo and in vitro.


Subject(s)
Endoglin/metabolism , Endothelium, Vascular/metabolism , Hypercholesterolemia/metabolism , Plaque, Atherosclerotic/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/genetics , Cells, Cultured , Cholesterol/metabolism , Endoglin/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/genetics , Indazoles/pharmacology , Isonicotinic Acids/pharmacology , Kruppel-Like Factor 6/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , P-Selectin/metabolism , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/genetics , Receptors, LDL/genetics , Smad Proteins/metabolism , beta-Cyclodextrins/pharmacology
2.
Atherosclerosis ; 271: 15-25, 2018 04.
Article in English | MEDLINE | ID: mdl-29459262

ABSTRACT

BACKGROUND AND AIMS: Increased plasma levels of soluble endoglin (sEng) were detected in patients with endothelial dysfunction-related disorders and hypercholesterolemia. In this study, we hypothesized that high levels of sEng accompanied by mild hypercholesterolemia could aggravate endothelial and vessel wall dysfunction and affect endoglin/eNOS signaling in mouse aorta. METHODS: Three-month-old female transgenic mice on CBAxC57BL/6J background, with high levels of sEng (Sol-Eng+high HFD), and their littermates with low levels of sEng (Sol-Eng+low HFD), were fed a high fat diet for six months. Plasma samples were used for biochemical, ELISA and Luminex analyses of total cholesterol, sEng and inflammatory markers. Functional parameters of aorta were assessed with wire myograph 620M. Western blot analyses of membrane endoglin/eNOS signaling and endothelial dysfunction/inflammation markers in aorta were performed. RESULTS: Functional analysis of aorta showed impaired KCl induced vasoconstriction, endothelial-dependent relaxation after the administration of acetylcholine as well as endothelial-independent relaxation induced by sodium nitroprusside in the Sol-Eng+high HFD group compared to the Sol-Eng+low HFD group. Ach-induced vasodilation after administration of l-NAME was significantly higher in the Sol-Eng+high HFD group compared to the Sol-Eng+low HFD group. The expression of endoglin, p-eNOS/eNOS, pSmad2/3/Smad2/3 signaling pathway was significantly lower in the Sol-Eng+high HFD group compared to the Sol-Eng+low HFD group. CONCLUSIONS: The results indicate that long-term hypercholesterolemia combined with high levels of sEng leads to the aggravation of endothelial and vessel wall dysfunction in aorta, with possible alterations of the membrane endoglin/eNOS signaling, suggesting that high levels of soluble endoglin might be considered as a risk factor of cardiovascular diseases.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Endoglin/metabolism , Endothelium, Vascular/metabolism , Hypercholesterolemia/complications , Animals , Aorta/pathology , Aorta/physiopathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Diet, High-Fat , Disease Models, Animal , Endoglin/genetics , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Humans , Hypercholesterolemia/etiology , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Up-Regulation , Vasoconstriction , Vasodilation
3.
Heart Vessels ; 32(8): 1013-1024, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28382387

ABSTRACT

A soluble form of endoglin (sEng) released into the circulation was suggested to be a direct inducer of endothelial dysfunction, inflammation and contributed to the development of hypertension by interfering with TGF-ß signaling in cardiovascular pathologies. In the present study, we assessed the hypothesis that high sEng level-induced hypertension via a possible sEng interference with TGF-ß signaling pathways may result in inflammatory, structural or fibrotic changes in hearts of Sol-Eng+ mice (mice with high levels of soluble endoglin) fed either chow or high-fat diet. Female Sol-Eng+ mice and their age matched littermates with low plasma levels of sEng were fed either chow or high-fat diet (HFD). Heart samples were subsequently analyzed by histology, qRT-PCR and Western blot analysis. In this study, no differences in myocardial morphology/hypertrophy and possible fibrotic changes between Sol-Eng+ mice and control mice were detected on both chow and HFD. The presence of sEng did not significantly affect the expression of selected members of TGF-ß signaling (membrane endoglin, TGFßRII, ALK-5, ALK-1, Id-1, PAI-1 and activated Smad proteins-pSmad 1,5 and pSmad 2,3), inflammation, heart remodeling (PDGFb, Col1A1) and endothelial dysfunction (VCAM-1, ICAM-1) in the hearts of Sol-Eng+ mice compared to control mice on both chow and high-fat diet. High levels of soluble endoglin did not affect microscopic structure (profibrotic and degenerative cardiomyocyte changes), and specific parts of TGF-ß signaling, endothelial function and inflammation in the heart of Sol-Eng+ mice fed both chow diet or HFD. However, we cannot rule out a possibility that a long-term chronic exposure (9 months and more) to soluble endoglin alone or combined with other cardiovascular risk factors may contribute to alterations of heart function and structure in Sol-Eng+ mice, which is the topic in our lab in ongoing experiments.


Subject(s)
Cardiomyopathy, Hypertrophic/blood , Endoglin/genetics , Gene Expression Regulation , Hypertension/blood , Myocardium/metabolism , RNA/genetics , Animals , Blotting, Western , Cardiomyopathy, Hypertrophic/etiology , Cardiomyopathy, Hypertrophic/pathology , Disease Models, Animal , Endoglin/biosynthesis , Female , Gene Expression Profiling , Heart , Hypertension/complications , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/pathology , Oxidative Stress , Real-Time Polymerase Chain Reaction , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
4.
Life Sci ; 175: 52-60, 2017 Apr 15.
Article in English | MEDLINE | ID: mdl-28336397

ABSTRACT

AIMS: Endoglin is a transmembrane glycoprotein, that plays an important role in regulating endothelium. Proteolytic cleavage of membrane endoglin releases soluble endoglin (sEng), whose increased plasma levels have been detected in diseases related to the cardiovascular system. It was proposed that sEng might damage vascular endothelium, but detailed information about the potential mechanisms involved is not available. Thus, we hypothesized that sEng contributes to endothelial dysfunction, leading to a pro-inflammatory phenotype by a possible modulation of the TGF-ß and/or inflammatory pathways. MAIN METHODS: Human umbilical vein endothelial cells (HUVECs) and Human embryonic kidney cell line (HEK293T) were treated with different sEng concentration and time in order to reveal possible effect on biomarkers of inflammation and TGF-ß signaling. IL6 and NFκB reporter luciferase assays, quantitative real-time PCR analysis, Western blot analysis and immunofluorescence flow cytometry were used. KEY FINDINGS: sEng treatment results in activation of NF-κB/IL-6 expression, increased expression of membrane endoglin and reduced expression of Id-1. On the other hand, no significant effects on other markers of endothelial dysfunction and inflammation, including eNOS, peNOSS1177, VCAM-1, COX-1, COX-2 and ICAM-1 were detected. SIGNIFICANCE: As a conclusion, sEng treatment resulted in an activation of NF-κB, IL-6, suggesting activation of pro-inflammatory phenotype in endothelial cells. The precise mechanism of this activation and its consequence remains to be elucidated. A combined treatment of sEng with other cardiovascular risk factors will be necessary in order to reveal whether sEng is not only a biomarker of cardiovascular diseases, but also a protagonist of endothelial dysfunction.


Subject(s)
Endoglin/biosynthesis , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Interleukin-6/biosynthesis , NF-kappa B/biosynthesis , Signal Transduction , HEK293 Cells , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inhibitor of Differentiation Protein 1/biosynthesis , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...