Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Angiogenesis ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965173

ABSTRACT

Aortic aneurysm is characterized by a pathological dilation at specific predilection sites of the vessel and potentially results in life-threatening vascular rupture. Herein, we established a modified "Häutchen method" for the local isolation of endothelial cells (ECs) from mouse aorta to analyze their spatial heterogeneity and potential role in site-specific disease development. When we compared ECs from aneurysm predilection sites of healthy mice with adjacent control segments we found regulation of genes related to extracellular matrix remodeling, angiogenesis and inflammation, all pathways playing a critical role in aneurysm development. We also detected enhanced cortical stiffness of the endothelium at these sites. Gene expression of ECs from aneurysms of the AngII ApoE-/- model when compared to sham animals mimicked expression patterns from predilection sites of healthy animals. Thus, this work highlights a striking genetic and functional regional heterogeneity in aortic ECs of healthy mice, which defines the location of aortic aneurysm formation in disease.

2.
Am J Pathol ; 193(4): 474-492, 2023 04.
Article in English | MEDLINE | ID: mdl-36669683

ABSTRACT

The outer layer of endothelial cells (ECs), consisting of the endothelial glycocalyx (eGC) and the cortex (CTX), provides a protective barrier against vascular diseases. Structural and functional impairments of their mechanical properties are recognized as hallmarks of endothelial dysfunction and can lead to cardiovascular events, such as acute myocardial infarction (AMI). This study investigated the effects of AMI on endothelial nanomechanics and function and the use of exogenous recombinant syndecan-1 (rSyn-1), a major component of the eGC, as recovering agent. ECs were exposed in vitro to serum samples collected from patients with AMI. In addition, in situ ECs of ex vivo aorta preparations derived from a mouse model for AMI were employed. Effects were quantified by using atomic force microscopy-based nanoindentation measurements, fluorescence staining, and histologic examination of the mouse hearts. AMI serum samples damaged eGC/CTX and augmented monocyte adhesion to the endothelial surface. In particular, the anaphylatoxins C3a and C5a played an important role in these processes. The impairment of endothelial function could be prevented by rSyn-1 treatment. In the mouse model of myocardial infarction, pretreatment with rSyn-1 alleviated eGC/CTX deterioration and reduced cardiomyocyte damage in histologic analyses. However, echocardiographic measurements did not indicate a functional benefit. These results provide new insights into the underlying mechanisms of AMI-induced endothelial dysfunction and perspectives for future studies on the benefit of rSyn-1 in post-AMI treatment.


Subject(s)
Endothelial Cells , Myocardial Infarction , Animals , Mice , Endothelial Cells/pathology , Glycocalyx/pathology , Syndecan-1 , Myocytes, Cardiac , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology
3.
Cell Mol Life Sci ; 79(6): 317, 2022 May 27.
Article in English | MEDLINE | ID: mdl-35622139

ABSTRACT

Endothelial inflammation is recognized as a critical condition in the development of cardiovascular diseases. TNF-induced inflammation of endothelial cells is linked to the formation of lipid droplets, augmented cortical stiffness, and nanostructural endothelial plasma membrane remodelling, but the insight into the mechanism linking these responses is missing. In the present work, we determined the formation of lipid droplets (LDs), nanomechanical, and nanostructural responses in the model of TNF-activated vascular inflammation in the isolated murine aorta using Raman spectroscopy, fluorescence imaging, atomic force microscopy (AFM), and scanning electron microscopy (SEM). We analysed the possible role of Rac1, a major regulator of cytoskeletal organization, in TNF-induced vascular inflammation. We demonstrated that the formation of LDs, polymerization of F-actin, alterations in cortical stiffness, and nanostructural protuberances in endothelial plasma membrane were mediated by the Rac1. In particular, we revealed a significant role for Rac1 in the regulation of the formation of highly unsaturated LDs formed in response to TNF. Inhibition of Rac1 also downregulated the overexpression of ICAM-1 induced by TNF, supporting the role of Rac1 in vascular inflammation. Altogether, our results demonstrate that LDs formation, an integral component of vascular inflammation, is activated by Rac1 that also regulates nanomechanical and nanostructural alterations linked to vascular inflammation.


Subject(s)
Endothelial Cells , Endothelium, Vascular , Animals , Aorta , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Inflammation/metabolism , Lipid Droplets/metabolism , Mice
4.
Cell Mol Life Sci ; 79(5): 235, 2022 Apr 10.
Article in English | MEDLINE | ID: mdl-35397686

ABSTRACT

The contribution of the shear stress-sensitive epithelial Na+ channel (ENaC) to the mechanical properties of the endothelial cell surface under (patho)physiological conditions is unclear. This issue was addressed in in vivo and in vitro models for endothelial dysfunction. Cultured human umbilical vein endothelial cells (HUVEC) were exposed to laminar (LSS) or non-laminar shear stress (NLSS). ENaC membrane insertion was quantified using Quantum-dot-based immunofluorescence staining and the mechanical properties of the cell surface were probed with the Atomic Force Microscope (AFM) in vitro and ex vivo in isolated aortae of C57BL/6 and ApoE/LDLR-/- mice. Flow- and acetylcholine-mediated vasodilation was measured in vivo using magnetic resonance imaging. Acute LSS led to a rapid mineralocorticoid receptor (MR)-dependent membrane insertion of ENaC and subsequent stiffening of the endothelial cortex caused by actin polymerization. Of note, NLSS stress further augmented the cortical stiffness of the cells. These effects strongly depend on the presence of the endothelial glycocalyx (eGC) and could be prevented by functional inhibition of ENaC and MR in vitro endothelial cells and ex vivo endothelial cells derived from C57BL/6, but not ApoE/LDLR-/- vessel. In vivo In C57BL/6 vessels, ENaC- and MR inhibition blunted flow- and acetylcholine-mediated vasodilation, while in the dysfunctional ApoE/LDLR-/- vessels, this effect was absent. In conclusion, under physiological conditions, endothelial ENaC, together with the glycocalyx, was identified as an important shear stress sensor and mediator of endothelium-dependent vasodilation. In contrast, in pathophysiological conditions, ENaC-mediated mechanotransduction and endothelium-dependent vasodilation were lost, contributing to sustained endothelial stiffening and dysfunction.


Subject(s)
Epithelial Sodium Channels , Glycocalyx , Receptors, Mineralocorticoid , Stress, Mechanical , Acetylcholine/metabolism , Animals , Cells, Cultured , Endothelium, Vascular/metabolism , Epithelial Sodium Channels/metabolism , Glycocalyx/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mechanotransduction, Cellular , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Receptors, Mineralocorticoid/metabolism
5.
Pflugers Arch ; 474(10): 1069-1076, 2022 10.
Article in English | MEDLINE | ID: mdl-35867189

ABSTRACT

Proinflammatory cytokines target vascular endothelial cells during COVID-19 infections. In particular, the endothelial glycocalyx (eGC), a proteoglycan-rich layer on top of endothelial cells, was identified as a vulnerable, vasoprotective structure during infections. Thus, eGC damage can be seen as a hallmark in the development of endothelial dysfunction and inflammatory processes. Using sera derived from patients suffering from COVID-19, we could demonstrate that the eGC became progressively worse in relation to disease severity (mild vs severe course) and in correlation to IL-6 levels. This could be prevented by administering low doses of spironolactone, a well-known and highly specific aldosterone receptor antagonist. Our results confirm that SARS-CoV-2 infections cause eGC damage and endothelial dysfunction and we outline the underlying mechanisms and suggest potential therapeutic options.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Glycocalyx , Mineralocorticoid Receptor Antagonists , SARS-CoV-2 , Spironolactone , COVID-19/blood , COVID-19/pathology , Cytokines/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Glycocalyx/drug effects , Glycocalyx/pathology , Humans , Interleukin-6/blood , Mineralocorticoid Receptor Antagonists/pharmacology , Mineralocorticoid Receptor Antagonists/therapeutic use , Proteoglycans/analysis , Proteoglycans/blood , Spironolactone/pharmacology , Spironolactone/therapeutic use
6.
Pflugers Arch ; 474(9): 993-1002, 2022 09.
Article in English | MEDLINE | ID: mdl-35648220

ABSTRACT

Investigating atherosclerosis and endothelial dysfunction has mainly become established in genetically modified ApoE-/- or LDL-R-/- mice transgenic models. A new AAV-PCSK9DYDY mouse model with no genetic modification has now been reported as an alternative atherosclerosis model. Here, we aimed to employ this AAV-PCSK9DY mouse model to quantify the mechanical stiffness of the endothelial surface, an accepted hallmark for endothelial dysfunction and forerunner for atherosclerosis. Ten-week-old male C57BL/6 N mice were injected with AAV-PCSK9DY (0.5, 1 or 5 × 1011 VG) or saline as controls and fed with Western diet (1.25% cholesterol) for 3 months. Total cholesterol (TC) and triglycerides (TG) were measured after 6 and 12 weeks. Aortic sections were used for atomic force microscopy (AFM) measurements or histological analysis using Oil-Red-O staining. Mechanical properties of in situ endothelial cells derived from ex vivo aorta preparations were quantified using AFM-based nanoindentation. Compared to controls, an increase in plasma TC and TG and extent of atherosclerosis was demonstrated in all groups of mice in a viral load-dependent manner. Cortical stiffness of controls was 1.305 pN/nm and increased (10%) in response to viral load (≥ 0.5 × 1011 VG) and positively correlated with the aortic plaque content and plasma TC and TG. For the first time, we show changes in the mechanical properties of the endothelial surface and thus the development of endothelial dysfunction in the AAV-PCSK9DY mouse model. Our results demonstrate that this model is highly suitable and represents a good alternative to the commonly used transgenic mouse models for studying atherosclerosis and other vascular pathologies.


Subject(s)
Atherosclerosis , Proprotein Convertase 9 , Animals , Atherosclerosis/pathology , Cholesterol , Disease Models, Animal , Endothelial Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Atomic Force , Proprotein Convertase 9/genetics , Triglycerides
7.
Int J Mol Sci ; 23(18)2022 09 13.
Article in English | MEDLINE | ID: mdl-36142571

ABSTRACT

Endothelial mechanics control vascular reactivity and are regulated by the mineralocorticoid receptor (MR) and its downstream target, the epithelial Na+ channel (ENaC). Endothelial dysfunction is a hallmark of chronic kidney disease (CKD), but its mechanisms are poorly understood. We hypothesized that CKD disrupts endothelial mechanics in an MR/ENaC-dependent process. METHODS: Primary human endothelial cells were cultured with uremic serum derived from children with stage 3-5 (predialysis) CKD or adult hemodialysis (HD) patients or healthy controls. The height and stiffness of the endothelial glycocalyx (eGC) and cortex were monitored by atomic force microscopy (AFM) using an ultrasensitive mechanical nanosensor. RESULTS: In a stage-dependent manner, sera from children with CKD induced a significant increase in eGC and cortex stiffness and an incremental reduction of the eGC height. AFM measurements were significantly associated with individual pulse wave velocity and serum concentrations of gut-derived uremic toxins. Serum from HD patients increased MR expression and mechanical stiffness of the endothelial cortex, an effect reversed by MR and ENaC antagonists, decreased eNOS expression and NO bioavailability, and augmented monocyte adhesion. CONCLUSION: These data indicate progressive structural damage of the endothelial surface with diminishing kidney function and identify the MR as a mediator of CKD-induced endothelial dysfunction.


Subject(s)
Glycocalyx , Renal Insufficiency, Chronic , Adult , Child , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Glycocalyx/metabolism , Humans , Pulse Wave Analysis , Receptors, Mineralocorticoid/metabolism , Renal Insufficiency, Chronic/metabolism
8.
Am J Pathol ; 190(4): 732-741, 2020 04.
Article in English | MEDLINE | ID: mdl-32035884

ABSTRACT

The negatively charged, brush-like glycocalyx covers the surface layer of endothelial cells. This layer of membrane-bound, carbohydrate-rich molecules covers the luminal surface of the endothelium along the entire vascular tree, mostly comprising glycoproteins and proteoglycans. Together with the underlying actin-rich endothelial cortex, 50 to 150 nm beneath the plasma membrane, the endothelial glycocalyx (eGC) is recognized as a vasoprotective nanobarrier and responsive hub. Importantly, both the eGC and cortex are highly dynamic and can adapt their nanomechanical properties (ie, stiffness and height) to changes in the environment. The constant change between a soft and a stiff endothelial surface is imperative for proper functioning of the endothelium. This review defines the nanomechanical properties of the eGC and stresses the underlying mechanisms and factors leading to a disturbed structure-function relationship. Specifically, under inflammatory conditions, the eGC is damaged, resulting in enhanced vascular permeability, tissue edema, augmented leukocyte adhesion, platelet aggregation, and dysregulated vasodilation. An integrated knowledge of the relationship between the nanomechanical properties, structure, and function of the eGC might be key in understanding vascular function and dysfunction. In this context, the clinical aspects for preservation and restoration of proper eGC nanomechanics are discussed, considering the eGC as a potentially promising diagnostic marker and therapeutic target in the near future.


Subject(s)
Cell Adhesion , Endothelium, Vascular/metabolism , Glycocalyx/chemistry , Glycocalyx/physiology , Animals , Biomechanical Phenomena , Capillary Permeability , Humans
9.
Circulation ; 139(11): 1407-1421, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30586752

ABSTRACT

BACKGROUND: Arterial hypertension and its organ sequelae show characteristics of T cell-mediated inflammatory diseases. Experimental anti-inflammatory therapies have been shown to ameliorate hypertensive end-organ damage. Recently, the CANTOS study (Canakinumab Antiinflammatory Thrombosis Outcome Study) targeting interleukin-1ß demonstrated that anti-inflammatory therapy reduces cardiovascular risk. The gut microbiome plays a pivotal role in immune homeostasis and cardiovascular health. Short-chain fatty acids (SCFAs) are produced from dietary fiber by gut bacteria and affect host immune homeostasis. Here, we investigated effects of the SCFA propionate in 2 different mouse models of hypertensive cardiovascular damage. METHODS: To investigate the effect of SCFAs on hypertensive cardiac damage and atherosclerosis, wild-type NMRI or apolipoprotein E knockout-deficient mice received propionate (200 mmol/L) or control in the drinking water. To induce hypertension, wild-type NMRI mice were infused with angiotensin II (1.44 mg·kg-1·d-1 subcutaneous) for 14 days. To accelerate the development of atherosclerosis, apolipoprotein E knockout mice were infused with angiotensin II (0.72 mg·kg-1·d-1 subcutaneous) for 28 days. Cardiac damage and atherosclerosis were assessed using histology, echocardiography, in vivo electrophysiology, immunofluorescence, and flow cytometry. Blood pressure was measured by radiotelemetry. Regulatory T cell depletion using PC61 antibody was used to examine the mode of action of propionate. RESULTS: Propionate significantly attenuated cardiac hypertrophy, fibrosis, vascular dysfunction, and hypertension in both models. Susceptibility to cardiac ventricular arrhythmias was significantly reduced in propionate-treated angiotensin II-infused wild-type NMRI mice. Aortic atherosclerotic lesion area was significantly decreased in propionate-treated apolipoprotein E knockout-deficient mice. Systemic inflammation was mitigated by propionate treatment, quantified as a reduction in splenic effector memory T cell frequencies and splenic T helper 17 cells in both models, and a decrease in local cardiac immune cell infiltration in wild-type NMRI mice. Cardioprotective effects of propionate were abrogated in regulatory T cell-depleted angiotensin II-infused mice, suggesting the effect is regulatory T cell-dependent. CONCLUSIONS: Our data emphasize an immune-modulatory role of SCFAs and their importance for cardiovascular health. The data suggest that lifestyle modifications leading to augmented SCFA production could be a beneficial nonpharmacological preventive strategy for patients with hypertensive cardiovascular disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Aortic Diseases/drug therapy , Arrhythmias, Cardiac/prevention & control , Atherosclerosis/drug therapy , Cardiomegaly/prevention & control , Hypertension/drug therapy , Propionates/pharmacology , Angiotensin II , Animals , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Arrhythmias, Cardiac/immunology , Arrhythmias, Cardiac/physiopathology , Arterial Pressure/drug effects , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Cardiomegaly/immunology , Cardiomegaly/physiopathology , Disease Models, Animal , Hypertension/chemically induced , Hypertension/immunology , Hypertension/physiopathology , Male , Mice, Knockout, ApoE , Plaque, Atherosclerotic , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
10.
Pflugers Arch ; 472(4): 419-433, 2020 04.
Article in English | MEDLINE | ID: mdl-32239285

ABSTRACT

The endothelial surface is a highly flexible signaling hub which is able to sense the hemodynamic forces of the streaming blood. The subsequent mechanosignaling is basically mediated by specific structures, like the endothelial glycocalyx building the top surface layer of endothelial cells as well as mechanosensitive ion channels within the endothelial plasma membrane. The mechanical properties of the endothelial cell surface are characterized by the dynamics of cytoskeletal proteins and play a key role in the process of signal transmission from the outside (lumen of the blood vessel) to the interior of the cell. Thus, the cell mechanics directly interact with the function of mechanosensitive structures and ion channels. To precisely maintain the vascular tone, a coordinated functional interdependency between endothelial cells and vascular smooth muscle cells is necessary. This is given by the fact that mechanosensitive ion channels are expressed in both cell types and that signals are transmitted via autocrine/paracrine mechanisms from layer to layer. Thus, the outer layer of the endothelial cells can be seen as important functional mechanosensitive and reactive cellular compartment. This review aims to describe the known mechanosensitive structures of the vessel building a bridge between the important role of physiological mechanosignaling and the proper vascular function. Since mutations and dysfunction of mechanosensitive proteins are linked to vascular pathologies such as hypertension, they play a potent role in the field of channelopathies and mechanomedicine.


Subject(s)
Endothelial Cells/metabolism , Ion Channels/metabolism , Mechanotransduction, Cellular/physiology , Stress, Mechanical , Animals , Glycocalyx/metabolism , Humans , Myocytes, Smooth Muscle/metabolism
11.
Kidney Int ; 97(3): 502-515, 2020 03.
Article in English | MEDLINE | ID: mdl-32008804

ABSTRACT

Dysfunctional high-density lipoprotein (d-HDL) in chronic kidney disease is known to have a change in composition towards an endothelial-damaging phenotype, amongst others, via the accumulation of symmetric dimethylarginine. The endothelial glycocalyx, a carbohydrate-rich layer lining the endothelial luminal surface, is a first line defense against vascular diseases including atherosclerosis. Here we conducted a translational, cross-sectional study to determine the role of symmetric dimethylarginine in d-HDL as a mediator of glycocalyx damage. Using confocal and atomic force microscopy, intact HDL from healthy donors was found to maintain the glycocalyx while isolated HDL from hemodialysis patients and exogenous symmetric dimethylarginine caused significant damage to the glycocalyx in endothelial cells in vitro in a dose-dependent manner. Symmetric dimethylarginine triggered glycocalyx deterioration via molecular pathways mediated by toll-like-receptor 2 and matrix metalloprotease-9. Corresponding intravital microscopy revealed that exogenous symmetric dimethylarginine and d-HDL from hemodialysis patients caused glycocalyx breakdown, which subsequently contributed to alterations in leukocyte rolling. Biologically effective HDL, which estimates the functionality of HDL, was calculated from circulating HDL-cholesterol and symmetric dimethylarginine, as described in the literature. Biologically effective HDL was the only parameter that could independently predict glycocalyx damage in vivo. Thus, our data suggest that symmetric dimethylarginine in d-HDL mediates glycocalyx breakdown in chronic kidney disease.


Subject(s)
Glycocalyx , Renal Insufficiency, Chronic , Arginine/analogs & derivatives , Cross-Sectional Studies , Endothelial Cells , Humans , Lipoproteins, HDL
12.
Nanomedicine ; 16: 97-105, 2019 02.
Article in English | MEDLINE | ID: mdl-30550804

ABSTRACT

Endothelial dysfunction is recognized as a critical condition in the development of cardiovascular disorders. This multifactorial process involves changes in the biochemical and mechanical properties of endothelial cells leading to disturbed release of vasoprotective mediators. Hypercholesterolemia and increased stiffness of the endothelial cortex are independently shown to result in reduced release of nitric oxide and thus endothelial dysfunction. However, direct evidence linking these parameters to each other is missing. Here, a novel method combining Raman spectroscopy for biochemical analysis and Atomic Force Microscopy (AFM) for analyzing the endothelial nanomechanics was established. Using this dual approach, the same areas of native ex vivo aortas were investigated, either derived from mice with endothelial dysfunction (ApoE/LDLR-/-) or wild type mice. In particular an increased intracellular lipid content and elevated cortical stiffness/elasticity were shown in ApoE/LDLR-/- aortas, demonstrating a direct link between endothelial dysfunction, the biochemical composition and the nanomechanical properties of endothelial cells.


Subject(s)
Aorta/pathology , Apolipoproteins E/genetics , Endothelium, Vascular/pathology , Microscopy, Atomic Force/methods , Receptors, LDL/genetics , Spectrum Analysis, Raman/methods , Animals , Aorta/metabolism , Apolipoproteins E/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/metabolism
13.
Pflugers Arch ; 469(10): 1401-1412, 2017 10.
Article in English | MEDLINE | ID: mdl-28550472

ABSTRACT

High dietary salt intake may lead to vascular stiffness, which predicts cardiovascular diseases such as heart failure, and myocardial and cerebral infarctions as well as renal impairment. The vascular endothelium is a primary target for deleterious salt effects leading to dysfunction and endothelial stiffness. We hypothesize that the Ca2+- and bicarbonate-activated soluble adenylyl cyclase (sAC) contributes to Na+/K+-ATPase expression regulation in vascular endothelial cells and is an important regulator of endothelial stiffness. In vitro stimulation of vascular endothelial cells with high sodium (150 mM Na+)-induced Na+/K+-ATPase-α and Na+/K+-ATPase-ß protein expression determined by western blot. Promoter analyses revealed increased cAMP response element (CRE)-mediated Na+/K+-ATPase-α transcriptional activity under high sodium concentrations. Inhibition of sAC by the specific inhibitor KH7 or siRNA reduced the sodium effects. Flame photometry revealed increased intracellular sodium concentrations in response to high sodium stimulations, which were paralleled by elevated ATP levels. Using atomic force microscopy, a nano-technique that measures cellular stiffness and deformability, we detected significant endothelial stiffening under increased sodium concentrations, which was prevented by inhibition of sAC using KH7 and Na+/K+-ATPase using ouabain. Furthermore, analysis of primary aortic endothelial cells in an in vitro aging model revealed an impaired Na+/K+-ATPase-α sodium response and elevated intracellular sodium levels with cellular aging. We conclude that sAC mediates sodium-induced Na+/K+-ATPase expression in vascular endothelium and is an important regulator of endothelial stiffness. The reactivity of Na+/K+-ATPase-α expression regulation in response to high sodium seems to be impaired in aging endothelial cells and might be a component of endothelial dysfunction.


Subject(s)
Adenylyl Cyclases/metabolism , Endothelial Cells/metabolism , Sodium Chloride/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sodium/metabolism , Animals , Endothelium, Vascular/metabolism , Ouabain/pharmacology , Sodium Chloride, Dietary/metabolism
14.
FASEB J ; 30(1): 45-53, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26324851

ABSTRACT

Recently, a novel feedforward activation of the endothelial epithelial sodium channel (ENaC) [endothelial sodium channel (EnNaC)] by sodium was reported that counteracts ENaC function in kidney. In the absence of aldosterone, a rise in extracellular sodium (>145 mM) increases EnNaC surface abundance, thereby stiffening the cortex of vascular endothelial cells (ECs) in vitro. The latter reduces the release of NO-the hallmark of endothelial dysfunction. Here, we test whether high extracellular sodium per se increases EnNaC expression and cortical stiffness in an aldosterone synthase (Cyp11b2)-deficient (AS(-/-)) mouse model. Therefore, we employed in situ ECs of ex vivo aorta preparations from wild-type (WT) and AS(-/-). EnNaC surface expression (-16%) and cortical stiffness (-22%) were reduced in AS(-/-), compared with WT, whereas NO secretion was exclusively detectable in AS(-/-). EnNaC inhibition with benzamil decreased stiffness in both, while mineralocorticoid receptor antagonism diminished stiffness only in the WT. In the absence of aldosterone, high sodium (150 mM) increased EnNaC surface expression ex vivo (plus 19%) and cortical stiffness ex vivo (plus 41%) and in vivo (plus 44%). Application of aldosterone adjusted the stiffness of AS(-/-) to the WT level. We conclude that high sodium per se determines EnNaC expression and consequently endothelial cortical nanomechanics, thus likely contributing to endothelial dysfunction.


Subject(s)
Cytochrome P-450 CYP11B2/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Nitric Oxide/metabolism , Sodium/metabolism , Aldosterone/pharmacology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Cells, Cultured , Cytochrome P-450 CYP11B2/deficiency , Endothelial Cells/drug effects , Endothelium, Vascular/metabolism , Epithelial Sodium Channels/metabolism , Female , Male , Mice , Mice, Knockout , Mineralocorticoid Receptor Antagonists/pharmacology , Models, Animal , Up-Regulation/drug effects
15.
Pflugers Arch ; 467(3): 499-512, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25420527

ABSTRACT

High salt (NaCl) intake promotes the development of vascular diseases independent of a rise in blood pressure, whereas reduction of salt consumption has beneficial effects for the arterial system. This article summarizes our current understanding of the molecular mechanisms of high salt-induced alterations of the endothelial phenotype, the impact of the individual endothelial genotype, and the overall vascular phenotype. We focus on the endothelial Na(+) channel (EnNaC)-controlled nanomechanical properties of the endothelium, since high Na(+) leads to an EnNaC-induced Na(+)-influx and subsequent stiffening of endothelial cells. The mechanical stiffness of the endothelial cell (i.e., the endothelial phenotype) plays a crucial role as it controls the production of the endothelium-derived vasodilator nitric oxide (NO) which directly affects the tone of the vascular smooth muscle cells. In contrast to soft endothelial cells, stiff endothelial cells release reduced amounts of NO, the hallmark of endothelial dysfunction. This endothelium-born process is followed by the development of arterial stiffness (i.e., the vascular phenotype), predicting the development of vascular end-organ damage such as myocardial infarction, stroke, and renal impairment. In this context, we outline the potential clinical implication of direct (amiloride) and indirect (spironolactone) EnNaC inhibition on vascular function. However, interindividual differences exist in the response to high salt intake which involves different endothelial genotypes. Thus, selected genes and genetic variants contributing to the development of salt-induced endothelial dysfunction and hypertension are discussed. In this review, we focus on the role of salt in endothelial and vascular (dys)function and the link between salt-induced changes of the endothelial and vascular phenotype and its clinical implications.


Subject(s)
Endothelium, Vascular/metabolism , Phenotype , Sodium Chloride, Dietary/adverse effects , Sodium/metabolism , Vascular Stiffness , Animals , Endothelium, Vascular/physiology , Glycocalyx/genetics , Glycocalyx/metabolism , Humans , Sodium Chloride, Dietary/metabolism
16.
FASEB J ; 28(9): 4015-25, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24868010

ABSTRACT

Kidney epithelial sodium channels (ENaCs) are known to be inactivated by high sodium concentrations (feedback inhibition). Recently, the endothelial sodium channel (EnNaC) was identified to control the nanomechanical properties of the endothelium. EnNaC-dependent endothelial stiffening reduces the release of nitric oxide, the hallmark of endothelial dysfunction. To study the regulatory impact of sodium on EnNaC, endothelial cells (EA.hy926 and ex vivo mouse endothelium) were incubated in aldosterone-free solutions containing either low (130 mM) or high (150 mM) sodium concentrations. By applying atomic force microscopy-based nanoindentation, an unexpected positive correlation between increasing sodium concentrations and cortical endothelial stiffness was observed, which can be attributed to functional EnNaC. In particular, an acute rise in sodium concentration (+20 mM) was sufficient to increase EnNaC membrane abundance by 90% and stiffening of the endothelial cortex by 18%. Despite the absence of exogenous aldosterone, these effects were prevented by the aldosterone synthase inhibitor FAD286 (100 nM) or the mineralocorticoid receptor (MR)-antagonist spironolactone (100 nM), indicating endogenous aldosterone synthesis and MR-dependent signaling. Interestingly, in the presence of high-sodium concentrations, FAD286 increased the transcription of the MR by 69%. Taken together, a novel feedforward activation of EnNaC by sodium is proposed that contrasts ENaC feedback inhibition in kidney.


Subject(s)
Aorta/metabolism , Endothelium, Vascular/metabolism , Epithelial Sodium Channels/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Mineralocorticoid Receptor Antagonists/pharmacology , Sodium/pharmacology , Animals , Aorta/cytology , Aorta/drug effects , Blotting, Western , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cytochrome P-450 CYP11B2/antagonists & inhibitors , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Epithelial Sodium Channels/genetics , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Mice , Microscopy, Atomic Force , Microscopy, Fluorescence , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
17.
Nanomedicine ; 11(6): 1521-30, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25881741

ABSTRACT

Previous studies show that polyphenol-rich compounds can induce a swelling of the endothelial glycocalyx (eGC). Our goal was to reveal the mechanism behind the eGC-swelling. As polyphenols are potent modulators of fibrosis transmembrane conductance regulator (CFTR) Cl(-) channel, the hypothesis was tested whether polyphenol-induced increase in CFTR activity is responsible for the eGC-swelling. The impact of the polyphenols resveratrol, (-)-epicatechin, and quercetin on nanomechanics of living endothelial GM7373 cells was monitored by AFM-nanoindentation. The tested polyphenols lead to eGC-swelling with a simultaneous decrease in cortical stiffness. EGC-swelling, but not the change in cortical stiffness, was prevented by the inhibition of CFTR. Polyphenol-induced eGC-swelling could be mimicked by cytochalasin D, an actin-depolymerizing agent. Thus, in the vascular endothelium, polyphenols induce eGC-swelling by softening cortical actin and activating CFTR. Our findings imply that CFTR plays an important role in the maintenance of vascular homeostasis and may explain the vasoprotective properties of polyphenols. FROM THE CLINICAL EDITOR: Many vascular problems clinically can be attributed to a dysregulation of endothelial glycocalyx (eGC). The underlying mechanism however remains unclear. In this article, the authors used nanoindentation and showed that polyphenols could swell the endothelial glycocalyx and alter its function. This investigative method can lead to further mechanistic studies of other molecular pathways.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Endothelium, Vascular/metabolism , Glycocalyx/drug effects , Polyphenols/pharmacology , Animals , Cattle , Cell Line , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Endothelium, Vascular/cytology , Microscopy, Atomic Force
18.
Pflugers Arch ; 466(5): 851-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24046153

ABSTRACT

Once upon a time, the expression of the epithelial sodium channel (ENaC) was mainly assigned to the kidneys, colon and sweat glands where it was considered to be the main determinant of sodium homeostasis. Recent, though indirect, evidence for the possible existence of ENaC in a non-epithelial tissue was derived from the observation that the vascular endothelium is a target for aldosterone. Inhibitory actions of the intracellular aldosterone receptors by spironolactone and, more directly, by ENaC blockers such as amiloride supported this view. Shortly after, direct data on the expression of ENaC in vascular endothelium could be demonstrated. There, endothelial ENaC (EnNaC) could be defined as a major regulator of cellular mechanics which is a critical parameter in differentiating between vascular function and dysfunction. Foremost, the mechanical stiffness of the endothelial cell cortex, a layer 50-200 nm beneath the plasma membrane, has been shown to play a crucial role as it controls the production of the endothelium-derived vasodilator nitric oxide (NO) which directly affects the tone of the vascular smooth muscle cells. In contrast to soft endothelial cells, stiff endothelial cells release reduced amounts of NO, the hallmark of endothelial dysfunction. Thus, the combination of endothelial stiffness and myogenic tone might increase the peripheral vascular resistance. An elevation of arterial blood pressure is supposed to be the consequence of such functional changes. In this review, EnNaC is discussed as an aldosterone-regulated plasma membrane protein of the vascular endothelium that could significantly contribute to maintaining of an appropriate arterial blood pressure but, if overexpressed, could participate in the pathogenesis of arterial hypertension.


Subject(s)
Endothelium, Vascular/metabolism , Epithelial Sodium Channels/metabolism , Animals , Endothelium, Vascular/physiology , Epithelial Sodium Channels/genetics , Glycocalyx/metabolism , Hemodynamics , Humans , Nitric Oxide/metabolism
19.
Curr Opin Nephrol Hypertens ; 23(2): 143-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24378777

ABSTRACT

PURPOSE OF REVIEW: Approximately 20 years ago, a paradigm shift occurred questioning whether expression of the epithelial Na⁺ channel (ENaC) was mainly restricted to epithelial tissues. In this review, the recent findings of ENaC regulation, and its potential contributions to the function and dysfunction of the vasculature, is discussed. RECENT FINDINGS: Over the last few years, the expression, localization, and functional properties of ENaC have been determined in the two main vascular cell types: endothelial cells, and vascular smooth muscle cells. A chronically increased ENaC membrane abundance can lead to endothelial stiffening and to a reduced release of nitric oxide, the hallmark of endothelial dysfunction. Endothelial ENaC was shown to determine vasoconstriction by negatively modulating nitric oxide release in mesenteric arteries, likely via the PI3K/Akt signaling pathway. ENaC has therefore been recognized as a potentially important regulator of vascular nanomechanics and as a transducer of mechanical forces. SUMMARY: As ENaC expression is broader than anticipated, it has become clear that the protein may play a crucial role in the vasculature as it is located at the interface between blood and tissue, and is therefore implicated in the development of endothelial dysfunction and hypertension.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Epithelial Sodium Channels/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Sodium/metabolism , Animals , Endothelium, Vascular/physiopathology , Humans , Hypertension/metabolism , Hypertension/physiopathology , Mechanotransduction, Cellular , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stress, Mechanical
20.
FASEB J ; 27(9): 3652-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23729588

ABSTRACT

Aldosterone triggers the stiff endothelial cell syndrome (SECS), characterized by an up-regulation of epithelial sodium channels (ENaCs) and mechanical stiffening of the endothelial cell cortex accompanied by endothelial dysfunction. In vivo, aldosterone antagonism exerts sustained protection on the cardiovascular system. To illuminate the molecular mechanisms of this time-dependent effect, a study on endothelial cells in vitro and ex vivo was designed to investigate SECS over time. Endothelia (from human umbilical veins, bovine aortae, and explants of human arteries) were cultured in aldosterone-supplemented medium with or without the mineralocorticoid receptor (MR) antagonist spironolactone. MR expression, ENaC expression, cortical stiffness, and shear-mediated nitric oxide (NO) release were determined after 3 d (short term) and up to 24 d (long term). Over time, MR expression increased by 129%. ENaC expression and surface abundance increased by 32% and 42% (13.8 to 19.6 molecules per cell surface), paralleled by a 49% rise in stiffness. Spironolactone prevented this development and, after 3 wk of treatment, increased NO release by 50%. Thus, spironolactone improves endothelial function long-lastingly by preventing a time-dependent manifestation of SECS. This emphasizes the key role of vascular endothelium as a therapeutical target in cardiovascular disorders and might explain blood pressure independent actions of MR antagonism.


Subject(s)
Epithelial Sodium Channels/metabolism , Mineralocorticoid Receptor Antagonists/pharmacology , Spironolactone/pharmacology , Animals , Aorta/drug effects , Aorta/metabolism , Cattle , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Microscopy, Atomic Force , Nitric Oxide/metabolism , Umbilical Veins/drug effects , Umbilical Veins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL