Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 21(1): 30-41, 2020 01.
Article in English | MEDLINE | ID: mdl-31819254

ABSTRACT

NLRP3-inflammasome-driven inflammation is involved in the pathogenesis of a variety of diseases. Identification of endogenous inflammasome activators is essential for the development of new anti-inflammatory treatment strategies. Here, we identified that apolipoprotein C3 (ApoC3) activates the NLRP3 inflammasome in human monocytes by inducing an alternative NLRP3 inflammasome via caspase-8 and dimerization of Toll-like receptors 2 and 4. Alternative inflammasome activation in human monocytes is mediated by the Toll-like receptor adapter protein SCIMP. This triggers Lyn/Syk-dependent calcium entry and the production of reactive oxygen species, leading to activation of caspase-8. In humanized mouse models, ApoC3 activated human monocytes in vivo to impede endothelial regeneration and promote kidney injury in an NLRP3- and caspase-8-dependent manner. These data provide new insights into the regulation of the NLRP3 inflammasome and the pathophysiological role of triglyceride-rich lipoproteins containing ApoC3. Targeting ApoC3 might prevent organ damage and provide an anti-inflammatory treatment for vascular and kidney diseases.


Subject(s)
Acute Kidney Injury/immunology , Apolipoprotein C-III/immunology , Caspase 8/metabolism , Kidney Diseases/immunology , Monocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Acute Kidney Injury/pathology , Adaptor Proteins, Signal Transducing , Animals , Apolipoprotein C-III/genetics , Cell Line , Disease Models, Animal , HEK293 Cells , Humans , Inflammasomes/immunology , Inflammation/genetics , Inflammation/immunology , Kidney Diseases/pathology , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Reactive Oxygen Species/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
3.
Circ Res ; 130(1): 80-95, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34809444

ABSTRACT

BACKGROUND: The LDLR (low-density lipoprotein receptor) in the liver is the major determinant of LDL-cholesterol levels in human plasma. The discovery of genes that regulate the activity of LDLR helps to identify pathomechanisms of hypercholesterolemia and novel therapeutic targets against atherosclerotic cardiovascular disease. METHODS: We performed a genome-wide RNA interference screen for genes limiting the uptake of fluorescent LDL into Huh-7 hepatocarcinoma cells. Top hit genes were validated by in vitro experiments as well as analyses of data sets on gene expression and variants in human populations. RESULTS: The knockdown of 54 genes significantly inhibited LDL uptake. Fifteen of them encode for components or interactors of the U2-spliceosome. Knocking down any one of 11 out of 15 genes resulted in the selective retention of intron 3 of LDLR. The translated LDLR fragment lacks 88% of the full length LDLR and is detectable neither in nontransfected cells nor in human plasma. The hepatic expression of the intron 3 retention transcript is increased in nonalcoholic fatty liver disease as well as after bariatric surgery. Its expression in blood cells correlates with LDL-cholesterol and age. Single nucleotide polymorphisms and 3 rare variants of one spliceosome gene, RBM25, are associated with LDL-cholesterol in the population and familial hypercholesterolemia, respectively. Compared with overexpression of wild-type RBM25, overexpression of the 3 rare RBM25 mutants in Huh-7 cells led to lower LDL uptake. CONCLUSIONS: We identified a novel mechanism of posttranscriptional regulation of LDLR activity in humans and associations of genetic variants of RBM25 with LDL-cholesterol levels.


Subject(s)
Nuclear Proteins/metabolism , RNA Splicing , Receptors, LDL/genetics , Cholesterol/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Lipoproteins, LDL/metabolism , Liver/metabolism , Mutation , Nuclear Proteins/genetics , Receptors, LDL/metabolism , Spliceosomes/metabolism
4.
Immunity ; 38(4): 754-68, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23477738

ABSTRACT

Endothelial injury and dysfunction (ED) represent a link between cardiovascular risk factors promoting hypertension and atherosclerosis, the leading cause of death in Western populations. High-density lipoprotein (HDL) is considered antiatherogenic and known to prevent ED. Using HDL from children and adults with chronic kidney dysfunction (HDL(CKD)), a population with high cardiovascular risk, we have demonstrated that HDL(CKD) in contrast to HDL(Healthy) promoted endothelial superoxide production, substantially reduced nitric oxide (NO) bioavailability, and subsequently increased arterial blood pressure (ABP). We have identified symmetric dimethylarginine (SDMA) in HDL(CKD) that causes transformation from physiological HDL into an abnormal lipoprotein inducing ED. Furthermore, we report that HDL(CKD) reduced endothelial NO availability via toll-like receptor-2 (TLR-2), leading to impaired endothelial repair, increased proinflammatory activation, and ABP. These data demonstrate how SDMA can modify the HDL particle to mimic a damage-associated molecular pattern that activates TLR-2 via a TLR-1- or TLR-6-coreceptor-independent pathway, linking abnormal HDL to innate immunity, ED, and hypertension.


Subject(s)
Atherosclerosis/immunology , Hypertension/immunology , Kidney Diseases/immunology , Lipoproteins, HDL/metabolism , Toll-Like Receptor 2/metabolism , Adult , Animals , Arginine/analogs & derivatives , Arginine/chemistry , Arterial Pressure , Child , Endothelium , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Lipoproteins, HDL/chemistry , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Signal Transduction , Superoxides/metabolism , Toll-Like Receptor 2/genetics , Wound Healing
5.
Arterioscler Thromb Vasc Biol ; 41(10): e468-e479, 2021 10.
Article in English | MEDLINE | ID: mdl-34407633

ABSTRACT

Objective: ApoM enriches S1P (sphingosine-1-phosphate) within HDL (high-density lipoproteins) and facilitates the activation of the S1P1 (S1P receptor type 1) by S1P, thereby preserving endothelial barrier function. Many protective functions exerted by HDL in extravascular tissues raise the question of how S1P regulates transendothelial HDL transport. Approach and Results: HDL were isolated from plasma of wild-type mice, Apom knockout mice, human apoM transgenic mice or humans and radioiodinated to trace its binding, association, and transport by bovine or human aortic endothelial cells. We also compared the transport of fluorescently-labeled HDL or Evans Blue, which labels albumin, from the tail vein into the peritoneal cavity of apoE-haploinsufficient mice with (apoE-haploinsufficient mice with endothelium-specific knockin of S1P1) or without (control mice, ie, apoE-haploinsufficient mice without endothelium-specific knockin of S1P1) endothelium-specific knockin of S1P1. The binding, association, and transport of HDL from Apom knockout mice and human apoM-depleted HDL by bovine aortic endothelial cells was significantly lower than that of HDL from wild-type mice and human apoM-containing HDL, respectively. The binding, uptake, and transport of 125I-HDL by human aortic endothelial cells was increased by an S1P1 agonist but decreased by an S1P1 inhibitor. Silencing of SR-BI (scavenger receptor BI) abrogated the stimulation of 125I-HDL transport by the S1P1 agonist. Compared with control mice, that is, apoE-haploinsufficient mice without endothelium-specific knockin of S1P1, apoE-haploinsufficient mice with endothelium-specific knockin of S1P1 showed decreased transport of Evans Blue but increased transport of HDL from blood into the peritoneal cavity and SR-BI expression in the aortal endothelium. Conclusions: ApoM and S1P1 promote transendothelial HDL transport. Their opposite effect on transendothelial transport of albumin and HDL indicates that HDL passes endothelial barriers by specific mechanisms rather than passive filtration.


Subject(s)
Apolipoproteins M/metabolism , Atherosclerosis/metabolism , Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Biological Transport , Cattle , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Permeability , Plaque, Atherosclerotic , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Sphingosine-1-Phosphate Receptors/genetics
6.
Int J Mol Sci ; 23(16)2022 Aug 22.
Article in English | MEDLINE | ID: mdl-36012766

ABSTRACT

High-density lipoprotein (HDL) is a mixture of complex particles mediating reverse cholesterol transport (RCT) and several cytoprotective activities. Despite its relevance for human health, many aspects of HDL-mediated lipid trafficking and cellular signaling remain elusive at the molecular level. During HDL's journey throughout the body, its functions are mediated through interactions with cell surface receptors on different cell types. To characterize and better understand the functional interplay between HDL particles and tissue, we analyzed the surfaceome-residing receptor neighborhoods with which HDL potentially interacts. We applied a combination of chemoproteomic technologies including automated cell surface capturing (auto-CSC) and HATRIC-based ligand-receptor capturing (HATRIC-LRC) on four different cellular model systems mimicking tissues relevant for RCT. The surfaceome analysis of EA.hy926, HEPG2, foam cells, and human aortic endothelial cells (HAECs) revealed the main currently known HDL receptor scavenger receptor B1 (SCRB1), as well as 155 shared cell surface receptors representing potential HDL interaction candidates. Since vascular endothelial growth factor A (VEGF-A) was recently found as a regulatory factor of transendothelial transport of HDL, we next analyzed the VEGF-modulated surfaceome of HAEC using the auto-CSC technology. VEGF-A treatment led to the remodeling of the surfaceome of HAEC cells, including the previously reported higher surfaceome abundance of SCRB1. In total, 165 additional receptors were found on HAEC upon VEGF-A treatment representing SCRB1 co-regulated receptors potentially involved in HDL function. Using the HATRIC-LRC technology on human endothelial cells, we specifically aimed for the identification of other bona fide (co-)receptors of HDL beyond SCRB1. HATRIC-LRC enabled, next to SCRB1, the identification of the receptor tyrosine-protein kinase Mer (MERTK). Through RNA interference, we revealed its contribution to endothelial HDL binding and uptake. Furthermore, subsequent proximity ligation assays (PLAs) demonstrated the spatial vicinity of MERTK and SCRB1 on the endothelial cell surface. The data shown provide direct evidence for a complex and dynamic HDL receptome and that receptor nanoscale organization may influence binding and uptake of HDL.


Subject(s)
Lipoproteins, HDL , Vascular Endothelial Growth Factor A , Humans , Ligands , Lipoproteins, HDL/metabolism , Receptors, Cell Surface , Receptors, Scavenger , Vascular Endothelial Growth Factor A/metabolism , c-Mer Tyrosine Kinase
7.
J Proteome Res ; 20(11): 4974-4984, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34677978

ABSTRACT

High-density lipoprotein (HDL) is a heterogeneous mixture of blood-circulating multimolecular particles containing many different proteins, lipids, and RNAs. Recent advancements in mass spectrometry-based proteotype analysis show promise for the analysis of proteoforms across large patient cohorts. In order to create the required spectral libraries enabling these data-independent acquisition (DIA) strategies, HDL was isolated from the plasma of more than 300 patients with a multiplicity of physiological HDL states. HDL proteome spectral libraries consisting of 296 protein groups and more than 786 peptidoforms were established, and the performance of the DIA strategy was benchmarked for the detection of HDL proteotype differences between healthy individuals and a cohort of patients suffering from diabetes mellitus type 2 and/or coronary heart disease. Bioinformatic interrogation of the data using the generated spectral libraries showed that the DIA approach enabled robust HDL proteotype determination. HDL peptidoform analysis enabled by using spectral libraries allowed for the identification of post-translational modifications, such as in APOA1, which could affect HDL functionality. From a technical point of view, data analysis further shows that protein and peptide quantities are currently more discriminative between different HDL proteotypes than peptidoforms without further enrichment. Together, DIA-based HDL proteotyping enables the robust digitization of HDL proteotypes as a basis for the analysis of larger clinical cohorts.


Subject(s)
Lipoproteins, HDL , Proteomics , Humans , Mass Spectrometry , Peptides/analysis , Proteome/analysis
8.
J Lipid Res ; 61(4): 492-504, 2020 04.
Article in English | MEDLINE | ID: mdl-31907205

ABSTRACT

Loss of pancreatic ß-cell mass and function as a result of sustained ER stress is a core step in the pathogenesis of diabetes mellitus type 2. The complex control of ß-cells and insulin production involves hedgehog (Hh) signaling pathways as well as cholesterol-mediated effects. In fact, data from studies in humans and animal models suggest that HDL protects against the development of diabetes through inhibition of ER stress and ß-cell apoptosis. We investigated the mechanism by which HDL inhibits ER stress and apoptosis induced by thapsigargin, a sarco/ER Ca2+-ATPase inhibitor, in ß-cells of a rat insulinoma cell line, INS1e. We further explored effects on the Hh signaling receptor Smoothened (SMO) with pharmacologic agonists and inhibitors. Interference with sterol synthesis or efflux enhanced ß-cell apoptosis and abrogated the anti-apoptotic activity of HDL. During ER stress, HDL facilitated the efflux of specific oxysterols, including 24-hydroxycholesterol (OHC). Supplementation of reconstituted HDL with 24-OHC enhanced and, in cells lacking ABCG1 or the 24-OHC synthesizing enzyme CYP46A1, restored the protective activity of HDL. Inhibition of SMO countered the beneficial effects of HDL and also LDL, and SMO agonists decreased ß-cell apoptosis in the absence of ABCG1 or CYP46A1. The translocation of the SMO-activated transcription factor glioma-associated oncogene GLI-1 was inhibited by ER stress but restored by both HDL and 24-OHC. In conclusion, the protective effect of HDL to counter ER stress and ß-cell death involves the transport, generation, and mobilization of oxysterols for activation of the Hh signaling receptor SMO.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Lipoproteins, HDL/pharmacology , Smoothened Receptor/metabolism , Animals , Cell Line , Cholesterol/metabolism , Hedgehog Proteins/metabolism , Homeostasis/drug effects , Insulin-Secreting Cells/metabolism , Rats , Signal Transduction/drug effects , Triglycerides/metabolism
9.
J Lipid Res ; 59(11): 2188-2201, 2018 11.
Article in English | MEDLINE | ID: mdl-30173145

ABSTRACT

Clear-cell renal cell carcinomas (ccRCCs) are characterized by inactivation of the von Hippel-Lindau (VHL) gene and intracellular lipid accumulation by unknown pathomechanisms. The immunochemical analysis of 356 RCCs revealed high abundance of apoA-I and apoB, as well as scavenger receptor BI (SR-BI) in the ccRCC subtype. Given the characteristic loss of VHL function in ccRCC, we used VHL-defective and VHL-proficient cells to study the potential influence of VHL on lipoprotein uptake. VHL-defective patient-derived ccRCC cells and cell lines (786O and RCC4) showed enhanced uptake as well as less resecretion and degradation of radio-iodinated HDL and LDL (125I-HDL and 125I-LDL, respectively) compared with the VHL-proficient cells. The ccRCC cells showed enhanced vascular endothelial growth factor (VEGF) and SR-BI expression compared with normal kidney epithelial cells. Uptake of 125I-HDL and 125I-LDL by patient-derived normal kidney epithelial cells as well as the VHL-reexpressing ccRCC cell lines, 786-O-VHL and RCC4-O-VHL cells, was strongly enhanced by VEGF treatment. The knockdown of the VEGF coreceptor, neuropilin-1 (NRP1), as well as blocking of SR-BI significantly reduced the uptake of lipoproteins into ccRCC cells in vitro. LDL stimulated proliferation of 786-O cells more potently than 786-O-VHL cells in a NRP1- and SR-BI-dependent manner. In conclusion, enhanced lipoprotein uptake due to increased activities of VEGF/NRP1 and SR-BI promotes lipid accumulation and proliferation of VHL-defective ccRCC cells.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Receptors, Scavenger/metabolism , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Apolipoproteins/genetics , Apolipoproteins/metabolism , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Blotting, Western , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Kidney Neoplasms/genetics , Lipoproteins , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Receptors, Scavenger/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
10.
Arterioscler Thromb Vasc Biol ; 37(5): 794-803, 2017 05.
Article in English | MEDLINE | ID: mdl-28360088

ABSTRACT

OBJECTIVE: Low- and high-density lipoproteins (LDL and HDL) must pass the endothelial layer to exert pro- and antiatherogenic activities, respectively, within the vascular wall. However, the rate-limiting factors that mediate transendothelial transport of lipoproteins are yet little known. Therefore, we performed a high-throughput screen with kinase drug inhibitors to identify modulators of transendothelial LDL and HDL transport. APPROACH AND RESULTS: Microscopy-based high-content screening was performed by incubating human aortic endothelial cells with 141 kinase-inhibiting drugs and fluorescent-labeled LDL or HDL. Inhibitors of vascular endothelial growth factor (VEGF) receptors (VEGFR) significantly decreased the uptake of HDL but not LDL. Silencing of VEGF receptor 2 significantly decreased cellular binding, association, and transendothelial transport of 125I-HDL but not 125I-LDL. RNA interference with VEGF receptor 1 or VEGF receptor 3 had no effect. Binding, uptake, and transport of HDL but not LDL were strongly reduced in the absence of VEGF-A from the cell culture medium and were restored by the addition of VEGF-A. The restoring effect of VEGF-A on endothelial binding, uptake, and transport of HDL was abrogated by pharmacological inhibition of phosphatidyl-inositol 3 kinase/protein kinase B or p38 mitogen-activated protein kinase, as well as silencing of scavenger receptor BI. Moreover, the presence of VEGF-A was found to be a prerequisite for the localization of scavenger receptor BI in the plasma membrane of endothelial cells. CONCLUSIONS: The identification of VEGF as a regulatory factor of transendothelial transport of HDL but not LDL supports the concept that the endothelium is a specific and, hence, druggable barrier for the entry of lipoproteins into the vascular wall.


Subject(s)
Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Scavenger Receptors, Class B/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/enzymology , High-Throughput Screening Assays/methods , Humans , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Transport , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Scavenger Receptors, Class B/genetics , Signal Transduction/drug effects , Transfection , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Eur Heart J ; 38(20): 1597-1607, 2017 May 21.
Article in English | MEDLINE | ID: mdl-28379378

ABSTRACT

AIMS: The vascular effects of high-density lipoproteins (HDL) differ under certain clinical conditions. The composition of HDL is modified in patients with chronic kidney disease (CKD). As a consequence, uremic HDL induces endothelial dysfunction. We have previously shown that accumulation of symmetric dimethylarginine (SDMA) in HDL causes these adverse effects of HDL in CKD. The aim of the study is to determine the impact of the accumulation of SDMA on the association between HDL and mortality. METHODS AND RESULTS: Mortality, renal function, serum SDMA and HDL-cholesterol (HDL-C) were assessed in the LURIC study including 3310 subjects undergoing coronary angiography. All-cause mortality was 30.0% during median follow-up of 9.9 years. Serum SDMA levels significantly predicted all-cause and cardiovascular mortality, and were significantly correlated with SDMA accumulation in HDL. Notably, higher serum SDMA was independently associated with lower cholesterol efflux (P = 0.004) as a measure of HDL functionality. In subjects with low SDMA levels, higher HDL-C was associated with significantly lower mortality. In contrast, in subjects with high SDMA, HDL-C was associated with higher mortality. These findings were confirmed in 1424 participants of the MONICA/KORA S3 cohort. Of note, we derived an algorithm allowing for calculation of biologically effective HDL-C' based on measured HDL-C and SDMA. We corroborated these clinical findings with invitro evidence showing that SDMA accumulation abolishes the anti-inflammatory and regenerative properties of HDL. CONCLUSION: The data identify SDMA as a marker of HDL dysfunction. These findings highlight on the pivotal role of SDMA accumulation in HDL as a mediator of pre-mature cardiovascular disease in patients with CKD.


Subject(s)
Arginine/analogs & derivatives , Cardiovascular Diseases/etiology , Lipoproteins, HDL/metabolism , Renal Insufficiency, Chronic/mortality , Aged , Arginine/metabolism , Biomarkers/metabolism , Cardiovascular Diseases/mortality , Female , Glomerular Filtration Rate/physiology , Humans , Male , Middle Aged , Prognosis , Renal Insufficiency, Chronic/complications , Risk Factors
12.
Biochim Biophys Acta ; 1861(2): 98-107, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26577406

ABSTRACT

High density lipoprotein (HDL) and its main protein component apolipoprotein A-I (ApoA-I) have multiple anti-atherogenic functions. Some of them are exerted within the vessel wall, so that HDL needs to pass the endothelial barrier. To elucidate their itinerary through endothelial cells (ECs), we labelled ApoA-I and HDL either fluorescently or with 1.4 nm nanogold and investigated their cellular localization by using immunofluorescent microscopy (IFM) and electron microscopy (EM). HDL as well as ApoA-I is taken up by ECs into the same route of intracellular trafficking. Time kinetics and pulse chase experiments revealed that HDL is trafficked through different vesicles. HDL partially co-localized with LDL, albumin, and transferrin. HDL did not co-localize with clathrin and caveolin-1. Fluorescent HDL was recovered at small proportions in early endosomes and endosome to trans-golgi network vesicles but not at all in recycling endosomes, in late endosomes or lysosomes. EM identified HDL mainly in large filled vesicles which however upon IFM did not colocalize with markers of multivesicular bodies or autophagosomes. The uptake or cellular distribution of HDL was altered upon pharmacological interference with cytochalasine D, colchicine and dynasore. Blockage of fluid phase uptake with Amiloride or EIPA did not reduce the uptake of HDL. Neither did we observe any co-localization of HDL with dextran as the marker of fluid phase uptake. In conclusion, HDL and ApoA-I are internalized and trafficked by endothelial cells through a non-classical endocytic route.


Subject(s)
Apolipoprotein A-I/metabolism , Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , Transport Vesicles/metabolism , trans-Golgi Network/metabolism , Animals , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Biological Transport , Cattle , Caveolin 1/metabolism , Clathrin/metabolism , Colchicine/pharmacology , Cytochalasin D/pharmacology , Endocytosis , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fluorescent Dyes , Gold , Hydrazones/pharmacology , Kinetics , Lipoproteins, LDL/metabolism , Nanostructures/chemistry , Primary Cell Culture , Serum Albumin/metabolism , Transferrin/metabolism , Transport Vesicles/chemistry , Transport Vesicles/drug effects , trans-Golgi Network/chemistry , trans-Golgi Network/drug effects
13.
Curr Opin Lipidol ; 27(3): 264-73, 2016 06.
Article in English | MEDLINE | ID: mdl-27031272

ABSTRACT

PURPOSE OF REVIEW: The clinical utility of HDLs has been scrutinized upon the publication of Mendelian randomization studies showing no effect of HDL-cholesterol (HDL-C) modifying variants on cardiovascular disease (CVD) outcome. The failures of randomized controlled HDL-C-directed intervention trials have further fueled this skepticism. This general criticism originates from oversimplification that has equated 'HDL-C' with 'HDL' and misconceived both as the 'good cholesterol'. RECENT FINDINGS: HDL particles are heterogeneous and carry hundreds of different lipids, proteins, and microRNAs. Many of them but not cholesterol, that is, HDL-C, contributes to the multiple protective functions of HDLs that probably evolved to manage potentially life-threatening crises. Inflammatory processes modify the composition of HDL particles as well as their individual protein and lipid components, and, as a consequence, also their functionality. Gain of dominant-negative functions makes dysfunctional HDL a part rather than a solution of the endangering situation. Quantification of HDL particle numbers, distinct proteins or lipids, and modifications thereof as well as bioassays of HDL functionality are currently explored toward their diagnostic performance in risk prediction and monitoring of treatment response. SUMMARY: Any successful clinical exploitation of HDLs will depend on the identification of the most relevant (dys)functions and their structural correlates. Stringent or prioritized structure-(dys)function relationships may provide biomarkers for better risk assessment and monitoring of treatment response. The most relevant agonists carried by either functional or dysfunctional HDLs as well as their cellular responders are interesting targets for drug development.


Subject(s)
Lipoproteins, HDL/metabolism , Animals , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cholesterol, HDL/metabolism , Humans , Signal Transduction
14.
Circulation ; 131(10): 871-81, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25673670

ABSTRACT

BACKGROUND: Roux-en-Y gastric bypass (RYGB) reduces body weight and cardiovascular mortality in morbidly obese patients. Glucagon-like peptide-1 (GLP-1) seems to mediate the metabolic benefits of RYGB partly in a weight loss-independent manner. The present study investigated in rats and patients whether obesity-induced endothelial and high-density lipoprotein (HDL) dysfunction is rapidly improved after RYGB via a GLP-1-dependent mechanism. METHODS AND RESULTS: Eight days after RYGB in diet-induced obese rats, higher plasma levels of bile acids and GLP-1 were associated with improved endothelium-dependent relaxation compared with sham-operated controls fed ad libitum and sham-operated rats that were weight matched to those undergoing RYGB. Compared with the sham-operated rats, RYGB improved nitric oxide (NO) bioavailability resulting from higher endothelial Akt/NO synthase activation, reduced c-Jun amino terminal kinase phosphorylation, and decreased oxidative stress. The protective effects of RYGB were prevented by the GLP-1 receptor antagonist exendin9-39 (10 µg·kg(-1)·h(-1)). Furthermore, in patients and rats, RYGB rapidly reversed HDL dysfunction and restored the endothelium-protective properties of the lipoprotein, including endothelial NO synthase activation, NO production, and anti-inflammatory, antiapoptotic, and antioxidant effects. Finally, RYGB restored HDL-mediated cholesterol efflux capacity. To demonstrate the role of increased GLP-1 signaling, sham-operated control rats were treated for 8 days with the GLP-1 analog liraglutide (0.2 mg/kg twice daily), which restored NO bioavailability and improved endothelium-dependent relaxations and HDL endothelium-protective properties, mimicking the effects of RYGB. CONCLUSIONS: RYGB rapidly reverses obesity-induced endothelial dysfunction and restores the endothelium-protective properties of HDL via a GLP-1-mediated mechanism. The present translational findings in rats and patients unmask novel, weight-independent mechanisms of cardiovascular protection in morbid obesity.


Subject(s)
Body Weight/physiology , Endothelium, Vascular/physiology , Glucagon-Like Peptide 1/physiology , Lipoproteins, HDL/physiology , Obesity/surgery , Weight Loss/physiology , Adult , Animals , Antioxidants/physiology , Case-Control Studies , Cells, Cultured , Diet, High-Fat/adverse effects , Disease Models, Animal , Endothelium, Vascular/pathology , Female , Gastric Bypass , Humans , Male , Nitric Oxide/physiology , Obesity/physiopathology , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/physiology , Rats , Rats, Wistar , Signal Transduction , Treatment Outcome
15.
Handb Exp Pharmacol ; 224: 337-66, 2015.
Article in English | MEDLINE | ID: mdl-25522994

ABSTRACT

Reduced plasma levels of HDL-C are associated with an increased risk of CAD and myocardial infarction, as shown in various prospective population studies. However, recent clinical trials on lipid-modifying drugs that increase plasma levels of HDL-C have not shown significant clinical benefit. Notably, in some recent clinical studies, there is no clear association of higher HDL-C levels with a reduced risk of cardiovascular events observed in patients with existing CAD. These observations have prompted researchers to shift from a cholesterol-centric view of HDL towards assessing the function and composition of HDL particles. Of importance, experimental and translational studies have further demonstrated various potential antiatherogenic effects of HDL. HDL has been proposed to promote macrophage reverse cholesterol transport and to protect endothelial cell functions by prevention of oxidation of LDL and its adverse endothelial effects. Furthermore, HDL from healthy subjects can directly stimulate endothelial cell production of nitric oxide and exert anti-inflammatory and antiapoptotic effects. Of note, increasing evidence suggests that the vascular effects of HDL can be highly heterogeneous and HDL may lose important anti-atherosclerotic properties and turn dysfunctional in patients with chronic inflammatory disorders. A greater understanding of mechanisms of action of HDL and its altered vascular effects is therefore critical within the context of HDL-targeted therapies.


Subject(s)
Arteries/metabolism , Cardiovascular Diseases/metabolism , Cholesterol, HDL/metabolism , Animals , Apoptosis , Arteries/pathology , Biomarkers/metabolism , Cardiovascular Diseases/blood , Cardiovascular Diseases/pathology , Cardiovascular Diseases/prevention & control , Cholesterol, HDL/blood , Cholesterol, HDL/chemistry , Cholesterol, HDL/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Lipoproteins, LDL/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Predictive Value of Tests , Prognosis , Protective Factors , Protein Conformation , Risk Assessment , Risk Factors , Structure-Activity Relationship
16.
J Am Soc Nephrol ; 25(11): 2658-68, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24854267

ABSTRACT

Endothelial dysfunction begins in early CKD and contributes to cardiovascular mortality. HDL is considered antiatherogenic, but may have adverse vascular effects in cardiovascular disease, diabetes, and inflammatory conditions. The effect of renal failure on HDL properties is unknown. We studied the endothelial effects of HDL isolated from 82 children with CKD stages 2-5 (HDL(CKD)), who were free of underlying inflammatory diseases, diabetes, or active infections. Compared with HDL from healthy children, HDL(CKD) strongly inhibited nitric oxide production, promoted superoxide production, and increased vascular cell adhesion molecule-1 expression in human aortic endothelial cells, and reduced cholesterol efflux from macrophages. The effects on endothelial cells correlated with CKD grade, with the most profound changes induced by HDL from patients on dialysis, and partial recovery observed with HDL isolated after kidney transplantation. Furthermore, the in vitro effects on endothelial cells associated with increased aortic pulse wave velocity, carotid intima-media thickness, and circulating markers of endothelial dysfunction in patients. Symmetric dimethylarginine levels were increased in serum and fractions of HDL from children with CKD. In a longitudinal follow-up of eight children undergoing kidney transplantation, HDL-induced production of endothelial nitric oxide, superoxide, and vascular cell adhesion molecule-1 in vitro improved significantly at 3 months after transplantation, but did not reach normal levels. These results suggest that in children with CKD without concomitant disease affecting HDL function, HDL dysfunction begins in early CKD, progressing as renal function declines, and is partially reversed after kidney transplantation.


Subject(s)
Cholesterol, HDL/blood , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/mortality , Vascular Diseases/blood , Vascular Diseases/mortality , Adolescent , Arginine/analogs & derivatives , Arginine/blood , Biomarkers/blood , Child , Cholesterol, LDL/blood , Endothelium, Vascular/metabolism , Female , Humans , Kidney Transplantation/mortality , Male , Nitric Oxide/metabolism , Renal Dialysis/mortality , Renal Insufficiency, Chronic/surgery , Triglycerides/blood , Vascular Diseases/surgery
17.
Eur Heart J ; 35(43): 3021-32, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-24658767

ABSTRACT

AIMS: Cardiovascular events remain the leading cause of death in Western world. Atherosclerosis is the most common underlying complication driven by low-density lipoproteins (LDL) disturbing vascular integrity. Carbamylation of lysine residues, occurring primarily in the presence of chronic kidney disease (CKD), may affect functional properties of lipoproteins; however, its effect on endothelial function is unknown. METHODS AND RESULTS: Low-density lipoprotein from healthy donors was isolated and carbamylated. Vascular reactivity after treatment with native LDL (nLDL) or carbamylated LDL (cLDL) was examined in organ chambers for isometric tension recording using aortic rings of wild-type or lectin-like-oxidized LDL receptor-1 (LOX-1) transgenic mice. Reactive oxygen species (ROS) and nitric oxide (NO) production were determined using electron spin resonance spectroscopy. The effect of LDL-carbamyl-lysine levels on cardiovascular outcomes was determined in patients with CKD during a median follow-up of 4.7 years. Carbamylated LDL impaired endothelium-dependent relaxation to acetylcholine or calcium-ionophore A23187, but not endothelium-independent relaxation to sodium nitroprusside. In contrast, nLDL had no effect. Carbamylated LDL enhanced aortic ROS production by activating NADPH-oxidase. Carbamylated LDL stimulated endothelial NO synthase (eNOS) uncoupling at least partially by promoting S-glutathionylation of eNOS. Carbamylated LDL-induced endothelial dysfunction was enhanced in LOX-1 transgenic mice. In patients with CKD, LDL-carbamyl-lysine levels were significant predictors for cardiovascular events and all-cause mortality. CONCLUSIONS: Carbamylation of LDL induces endothelial dysfunction via LOX-1 activation and increased ROS production leading to eNOS uncoupling. This indicates a novel mechanism in the pathogenesis of atherosclerotic disease which may be pathogenic and prognostic in patients with CKD and high plasma levels of cLDL.


Subject(s)
Endothelium, Vascular/physiopathology , Lipoproteins, LDL/physiology , Acetylcholine/pharmacology , Analysis of Variance , Animals , Aorta/physiology , Cardiovascular Diseases/physiopathology , Enzyme Inhibitors/pharmacology , Healthy Volunteers , Humans , In Vitro Techniques , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase Type III/metabolism , Onium Compounds/pharmacology , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Scavenger Receptors, Class E/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
18.
J Lipid Res ; 55(8): 1730-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24950692

ABSTRACT

Sphingosine-1-phosphate (S1P) mediates several cytoprotective functions of HDL. apoM acts as a S1P binding protein in HDL. Erythrocytes are the major source of S1P in plasma. After glomerular filtration, apoM is endocytosed in the proximal renal tubules. Human or murine HDL elicited time- and dose-dependent S1P efflux from erythrocytes. Compared with HDL of wild-type (wt) mice, S1P efflux was enhanced in the presence of HDL from apoM transgenic mice, but not diminished in the presence of HDL from apoM knockout (Apom(-/-)) mice. Artificially reconstituted and apoM-free HDL also effectively induced S1P efflux from erythrocytes. S1P and apoM were not measurable in the urine of wt mice. Apom(-/-) mice excreted significant amounts of S1P. apoM was detected in the urine of mice with defective tubular endocytosis because of knockout of the LDL receptor-related protein, chloride-proton exchanger ClC-5 (Clcn5(-/-)), or the cysteine transporter cystinosin. Urinary levels of S1P were significantly elevated in Clcn5(-/-) mice. In contrast to Apom(-/-) mice, these mice showed normal plasma concentrations for apoM and S1P. In conclusion, HDL facilitates S1P efflux from erythrocytes by both apoM-dependent and apoM-independent mechanisms. Moreover, apoM facilitates tubular reabsorption of S1P from the urine, however, with no impact on S1P plasma concentrations.


Subject(s)
Apolipoproteins M/metabolism , Erythrocytes/metabolism , Kidney Tubules/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Apolipoproteins M/genetics , Lipoproteins, HDL/genetics , Lipoproteins, HDL/metabolism , Lysophospholipids/genetics , Mice , Mice, Knockout , Sphingosine/genetics , Sphingosine/metabolism
19.
Circulation ; 127(8): 891-904, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23349247

ABSTRACT

BACKGROUND: Endothelial dysfunction and injury are thought to play an important role in the progression of coronary artery disease (CAD). High-density lipoprotein from healthy subjects (HDL(Healthy)) has been proposed to exert endothelial antiapoptotic effects that may represent an important antiatherogenic property of the lipoprotein. The present study therefore aimed to compare effects of HDL(CAD) and HDL(Healthy) on the activation of endothelial anti- and proapoptotic pathways and to determine which changes of the lipoprotein are relevant for these processes. METHODS AND RESULTS: HDL was isolated from patients with stable CAD (HDL(sCAD)), an acute coronary syndrome (HDL(ACS)), and healthy subjects. HDL(Healthy) induced expression of the endothelial antiapoptotic Bcl-2 protein Bcl-xL and reduced endothelial cell apoptosis in vitro and in apolipoprotein E-deficient mice in vivo. In contrast, HDL(sCAD) and HDL(ACS) did not inhibit endothelial apoptosis, failed to activate endothelial Bcl-xL, and stimulated endothelial proapoptotic pathways, in particular, p38-mitogen-activated protein kinase-mediated activation of the proapoptotic Bcl-2 protein tBid. Endothelial antiapoptotic effects of HDL(Healthy) were observed after inhibition of endothelial nitric oxide synthase and after delipidation, but not completely mimicked by apolipoprotein A-I or reconstituted HDL, suggesting an important role of the HDL proteome. HDL proteomics analyses and subsequent validations and functional characterizations suggested a reduced clusterin and increased apolipoprotein C-III content of HDL(sCAD) and HDL(ACS) as mechanisms leading to altered effects on endothelial apoptosis. CONCLUSIONS: The present study demonstrates for the first time that HDL(CAD) does not activate endothelial antiapoptotic pathways, but rather stimulates potential endothelial proapoptotic pathways. HDL-proteome remodeling plays an important role for these altered functional properties of HDL. These findings provide novel insights into mechanisms leading to altered vascular effects of HDL in coronary disease.


Subject(s)
Apoptosis/physiology , Coronary Artery Disease/metabolism , Endothelium, Vascular/metabolism , Lipoproteins, HDL/antagonists & inhibitors , Lipoproteins, HDL/physiology , Proteome/physiology , Signal Transduction/physiology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apoptosis/genetics , Coronary Artery Disease/pathology , Endothelium, Vascular/pathology , Female , Flow Cytometry/methods , Humans , Lipoproteins, HDL/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Proteome/genetics , Signal Transduction/genetics
20.
Circulation ; 127(11): 1229-40, e1-21, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23410942

ABSTRACT

BACKGROUND: Reactive oxygen species are major determinants of vascular aging. JunD, a member of the activated protein-1 family of transcription factors, is emerging as a major gatekeeper against oxidative stress. However, its contribution to reactive oxygen species homeostasis in the vasculature remains unknown. METHODS AND RESULTS: Endothelium-dependent vasorelaxation was impaired in young and old JunD(-/-) mice (6 and 22 months old) compared with age-matched wild-type mice. JunD(-/-) mice displayed an age-independent decline in endothelial nitric oxide release and endothelial nitric oxide synthase activity and increased mitochondrial superoxide formation and peroxynitrite levels. Furthermore, vascular expression and activity of the free radical scavengers manganese and extracellular superoxide dismutase and aldehyde dehydrogenase 2 were reduced, whereas the NADPH oxidase subunits p47phox, Nox2, and Nox4 were upregulated. These redox changes were associated with premature vascular aging, as shown by reduced telomerase activity, increased ß-galactosidase-positive cells, upregulation of the senescence markers p16(INK4a) and p53, and mitochondrial disruption. Interestingly, old wild-type mice showed a reduction in JunD expression and transcriptional activity resulting from promoter hypermethylation and binding with tumor suppressor menin, respectively. In contrast, JunD overexpression blunted age-induced endothelial dysfunction. In human endothelial cells, JunD knockdown exerted a similar impairment of the O2(-)/nitric oxide balance that was prevented by concomitant NADPH inhibition. In parallel, JunD expression was reduced in monocytes from old versus young healthy subjects and correlated with mRNA levels of scavenging and oxidant enzymes. CONCLUSIONS: JunD provides protection in aging-induced endothelial dysfunction and may represent a novel target to prevent reactive oxygen species-driven vascular aging.


Subject(s)
Aging/physiology , Endothelium, Vascular/physiopathology , Gene Deletion , Oxidative Stress/physiology , Proto-Oncogene Proteins c-jun/deficiency , Animals , Endothelium, Vascular/metabolism , Homeostasis/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/physiology , Models, Animal , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/physiology , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL