Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(2)2023 Jan 07.
Article in English | MEDLINE | ID: mdl-36674708

ABSTRACT

Periods of low energy supply are challenging conditions for organisms and cells during fasting or famine. Although changes in nutrient levels in the blood are first sensed by endothelial cells, studies on their metabolic adaptations to diminished energy supply are lacking. We analyzed the dynamic metabolic activity of human umbilical vein endothelial cells (HUVECs) in basal conditions and after serum starvation. Metabolites of glycolysis, the tricarboxylic acid (TCA) cycle, and the glycerol pathway showed lower levels after serum starvation, whereas amino acids had increased levels. A metabolic flux analysis with 13C-glucose or 13C-glutamine labeling for different time points reached a plateau phase of incorporation after 30 h for 13C-glucose and after 8 h for 13C-glutamine under both experimental conditions. Notably, we observed a faster label incorporation for both 13C-glucose and 13C-glutamine after serum starvation. In the linear range of label incorporation after 3 h, we found a significantly faster incorporation of central carbon metabolites after serum starvation compared to the basal state. These findings may indicate that endothelial cells develop increased metabolic activity to cope with energy deficiency. Physiologically, it can be a prerequisite for endothelial cells to form new blood vessels under unfavorable conditions during the process of angiogenesis in vivo.


Subject(s)
Glutamine , Starvation , Humans , Glutamine/metabolism , Amino Acids/metabolism , Glycolysis , Glucose/metabolism , Human Umbilical Vein Endothelial Cells/metabolism
2.
Atherosclerosis ; 291: 99-106, 2019 12.
Article in English | MEDLINE | ID: mdl-31706078

ABSTRACT

BACKGROUND AND AIMS: Gonadal hormones are mainly thought to account for sex and gender differences in the incidence, clinical manifestation and therapy of many cardiovascular diseases. However, intrinsic sex differences at the cellular level are mostly overlooked. Here, we assessed sex-specific metabolic and functional differences between male and female human umbilical vein endothelial cells (HUVECs). METHODS: Cellular metabolism was investigated by bioenergetic studies (Seahorse Analyser) and a metabolomic approach. Protein levels were determined by Western blots and proteome analysis. Vascular endothelial growth factor (VEGF)-stimulated cellular migration was assessed by gap closure. HUVECs from dizygotic twin pairs were used for most experiments. RESULTS: No sex differences were observed in untreated cells. However, sexual dimorphisms appeared after stressing the cells by serum starvation and treatment with VEGF. Under both conditions, female cells had higher intracellular ATP and metabolite levels. A significant decline in ATP levels was observed in male cells after serum starvation. After VEGF, the ratio of glycolysis/mitochondrial respiration was higher in female cells and migration was more pronounced. CONCLUSIONS: These results point to an increased stress tolerance of female cells. We therefore propose that female cells have an energetic advantage over male cells under conditions of diminished nutrient supply. A more favourable energy balance of female HUVECs after serum starvation and VEGF could potentially explain their stronger migratory capacity.


Subject(s)
Cell Movement , Energy Metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , Twins, Dizygotic , Angiogenesis Inducing Agents/pharmacology , Cell Movement/drug effects , Culture Media, Serum-Free/metabolism , Energy Metabolism/drug effects , Female , Humans , Male , Neovascularization, Physiologic/drug effects , Phenotype , Protein Interaction Maps , Sex Characteristics , Sex Factors , Vascular Endothelial Growth Factor A/pharmacology
3.
Cardiovasc Res ; 95(1): 97-107, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22510373

ABSTRACT

AIMS: Human relaxin-2 influences renal and cardiovascular functions. We investigated its effects on experimental endothelial dysfunction. METHODS AND RESULTS: Acetylcholine-mediated vasodilation of rat aortic rings, impaired by 48 h tumour necrosis factor-α (TNF-α) treatment, was dose-dependently improved by relaxin co-incubation, an effect sensitive to phosphatidylinositol 3-kinase (PI3K) inhibition and the glucocorticoid receptor (GR) antagonist RU-486. TNF increased endothelial nitric oxide synthase (eNOS) phosphorylation at Thr495 and decreased total eNOS expression and both basal and stimulated eNOS activity. Relaxin co-incubation did not affect eNOS expression but improved its activity via PI3K-dependent Thr495 dephosphorylation and Ser1177 phosphorylation, and additional Ser633 phosphorylation. Via GR, relaxin attenuated the TNF-related stimulation of endothelin-1 expression, superoxide and nitrotyrosine formation, and arginase II expression. Relaxin restored, via GR-CCAAT/enhancer-binding protein-ß (c/EBP-ß)-mediated promoter stimulation, the compromised expression of superoxide dismutase-1 (SOD1). In rat aortic endothelial cells, relaxin activated protein kinase B (Akt) and repressed TNF-induced nuclear factor-κB and activator protein-1. Finally, the relevance of the different findings to the model used was proved by pharmacological interventions. CONCLUSION: Relaxin improved endothelial dysfunction by promoting eNOS activity, suppressing endothelin-1 and arginase-II expression, and up-regulating SOD1 via GR, GR-c/EBP-ß, and PI3K-Akt pathways. This corroborates the notion that it functions as an endogenous and potentially therapeutic vasoprotector.


Subject(s)
Endothelium, Vascular/drug effects , Phosphatidylinositol 3-Kinases/physiology , Receptors, Glucocorticoid/physiology , Relaxin/pharmacology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Arginase/metabolism , CCAAT-Enhancer-Binding Protein-alpha/physiology , Endothelium, Vascular/physiology , Male , NADPH Oxidases/metabolism , NF-kappa B/physiology , Nitric Oxide Synthase Type III/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Superoxides/metabolism
4.
Regul Pept ; 166(1-3): 76-82, 2011 Jan 17.
Article in English | MEDLINE | ID: mdl-20851151

ABSTRACT

Increased expression of endothelial adhesion molecules, high levels of the monocyte chemoattractant protein-1 (MCP-1) and enhanced VLA4 integrin/VCAM-1 and CCR-2/MCP-1 interactions are initial steps in vascular inflammation. We sought to determine whether relaxin, a potent vasodilatory and anti-fibrotic agent, mitigates these early events compromising endothelial integrity. The effect of relaxin coincubation on the TNF-α-stimulated expression of the adhesion molecules VCAM-1, ICAM-1 and E-selectin; the MCP-1 expression by human umbilical vein endothelial cells (HUVEC) and human aortic smooth muscle cells (HAoSMC); as well as on direct monocyte-endothelium cell adhesion was quantified by ELISA or adhesion assay. CCR-2 and PECAM expression on HUVEC and THP-1 monocytes was investigated by FACS analysis. Relaxin treatment suppressed significantly TNF-α-induced upregulation of VCAM-1 and PECAM, CCR-2, and MCP-1 levels and direct monocyte adhesion to HUVEC. Our findings identify relaxin as a promising inhibitory factor in early vascular inflammation. By attenuating the upregulation of VCAM-1, key adhesion molecule in early vascular inflammation, and of MCP-1, a chemokine pivotal to monocyte recruitment, relaxin decreased initial monocyte-endothelium contact. This may be of relevance for the prevention and treatment of atherosclerosis and of other pro-inflammatory states.


Subject(s)
Inflammation/prevention & control , Relaxin/pharmacology , Aorta , Cell Adhesion/drug effects , Chemokine CCL2/biosynthesis , Down-Regulation , E-Selectin/biosynthesis , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Integrin alpha4/biosynthesis , Muscle, Smooth, Vascular/cytology , Receptors, CCR2/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Veins , Vascular Cell Adhesion Molecule-1/biosynthesis
5.
Regul Pept ; 155(1-3): 163-73, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19289144

ABSTRACT

Relaxin peptides act in brain, reproductive and cardiovascular systems, kidneys, and connective tissue through different G protein-coupled receptors. We reported that human relaxin-2 and porcine relaxin are both agonists at the human glucocorticoid receptor (GR). Here, we investigated the possible auto-regulation of relaxin-2 gene expression via recently discovered GR-binding sites in the relaxin-2 promoter. We found that porcine relaxin increased the secretion of human relaxin-like immunoreactivity in HeLa and THP-1 cells. Silencing of GR gene expression completely abolished this effect whereas transfection of wild-type GR into naturally GR-devoid HT-29 cells established relaxin sensitivity. Relaxin was shown to stimulate CAT expression driven by different deletion constructs of the 5'-flanking region of the relaxin-2 promoter. In chromatin immunoprecipitation assays, we detected both GR and relaxin binding to the relaxin-2 promoter. Gel shift assays indicated binding of relaxin-activated GR to half-GREs located between 160 and 200 bp upstream of transcription start but not to the GRE at -900 bp. Relaxin bound to human GR and displaced established GR agonists. Immunofluorescence experiments visualized nuclear co-localization of relaxin and GR in response to relaxin. In conclusion, we have identified a positive auto-regulatory loop of human relaxin-2 expression which involves GR and relaxin/GR binding to half-GREs in the relaxin-2 promoter.


Subject(s)
Gene Expression Regulation/drug effects , Promoter Regions, Genetic/genetics , Receptors, Glucocorticoid/physiology , Relaxin/pharmacology , Relaxin/physiology , Animals , Binding Sites , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Dexamethasone/pharmacology , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Glucocorticoids/pharmacology , HT29 Cells , HeLa Cells , Humans , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Relaxin/genetics , Relaxin/metabolism , Swine
6.
Regul Pept ; 154(1-3): 77-84, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19101597

ABSTRACT

The relaxin peptide family regulates diverse biological functions (central nervous processes, reproduction, cardiovascular and kidney function, and connective tissue composition) through different G protein-coupled receptors. We reported earlier that human relaxin-2 and porcine relaxin additionally interact with the human glucocorticoid receptor (GR) in an agonistic manner. Here we investigated whether the membrane receptor RXFP1 is critically involved in this pathway. We used chemically modified porcine relaxin which was biologically inactive at RXFP1. Native porcine relaxin, but not the modified peptide affected RXFP1-dependent and GR-independent readouts: ERK-1/2 and Akt phosphorylation as well as up-regulation of Akt and endothelin type-B receptor. In contrast, relaxin and modified relaxin inhibited endotoxin-stimulated secretion of TNF-alpha and IL-6 by human macrophages, an effect sensitive to the glucocorticoid receptor antagonists RU-486 and D-06. Both relaxins caused Ser(211) phosphorylation of GR, a biomarker of agonist-related receptor activation. Relaxin-induced accumulation of Ser(211)-phosphorylated GR was found in the cytoplasm and nucleus of HeLa cells, in endothelial cells, and in transfected HT-29 cells. In AP-1-luciferase assays, relaxin and modified relaxin inhibited endotoxin-induced activation of AP-1, a transcription factor essentially involved in endotoxin signaling. This study suggests that the inability of relaxin to interact with its membrane receptor does not interfere with its ability to activate GR.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Receptors, Glucocorticoid/metabolism , Relaxin/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Cyclic AMP/metabolism , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endotoxins/antagonists & inhibitors , Endotoxins/pharmacology , Female , HT29 Cells , HeLa Cells , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Mifepristone/pharmacology , Pregnancy , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , Receptors, Peptide/metabolism , Relaxin/pharmacology , Swine , Transfection , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
7.
Pharmacol Ther ; 112(1): 38-56, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16647137

ABSTRACT

The insulin-related peptide hormone relaxin (Rlx) is known as pregnancy hormone for decades. In the 1980s, researchers began to recognize the highly intriguing fact that Rlx plays a role in a multitude of physiological processes far beyond pregnancy and reproduction. So, Rlx's contribution to the regulation of vasotonus, plasma osmolality, angiogenesis, collagen turnover, and renal function has been established. In addition, the peptide has been demonstrated to represent a mediator of cardiovascular pathology. The ongoing efforts to identify Rlx receptors eventually precipitated the discovery of the G protein-coupled receptors (GPCR) LGR7 and LGR8 as membrane receptors for human Rlx-2 in 2002. This review will summarize the current state of insight into this rapidly evolving field, which has further been expanded by the discovery of GPCR135 and GPCR142 as receptors for Rlx-3. In addition, Rlx has also been shown to activate the human glucocorticoid receptor (GR). There is evidence from Rlx and Rlx receptor knockouts suggesting that LGR7 is the only relevant receptor for mouse Rlx-1 (corresponding to human Rlx-2) in vivo and that insulin-like peptide (INSL)-3 represents the physiological ligand for LGR8. Regarding Rlx signal transduction, the cyclic adenosine monophosphate (cAMP) and nitric oxide (NO) pathways will be characterized as major cascades. Investigation of downstream signaling remains an important field for future research. Finally, the current state of therapeutical strategies using Rlx in animal models as well as in humans is summarized.


Subject(s)
Relaxin/pharmacology , Animals , Humans , Relaxin/chemistry , Relaxin/metabolism , Signal Transduction
8.
Ann N Y Acad Sci ; 1041: 256-71, 2005 May.
Article in English | MEDLINE | ID: mdl-15956716

ABSTRACT

The insulin-like peptide relaxin is a central hormone of pregnancy, but it also produces antifibrotic, myocardial, renal, central nervous, and vascular effects. Recently, two G-protein-coupled receptors, LGR7 and LGR8, were identified as relaxin receptors. Prompted by reports on the immunoregulatory effects of relaxin, we investigated possible interactions with the human glucocorticoid receptor (GR). Relaxin blunted the endotoxin-induced production of inflammatory cytokines (interleukin 1 [IL-1], IL-6, and tumor necrosis factor- alpha) by human macrophages, an effect that was suppressed by the GR antagonist RU-486. In three different cell lines, relaxin induced GR activation, nuclear translocation, and DNA binding as assessed in glucocorticoid response element (GRE)-luciferase assays. Coimmunoprecipitation experiments revealed physical interaction of endogenous and exogenous relaxin with cytoplasmic and nuclear GR. Relaxin competed with GR agonists for GR binding both in vivo, in whole-cell assays, and in vitro, in fluorescence polarization assays. In LGR7- and LGR8-free cells, the relaxin-mediated activation of GR was preserved. In conclusion, relaxin acts as a GR agonist, a pathway pivotal to relaxin's effects on cytokine secretion by human macrophages. These findings may deepen our understanding of relaxin's many physiologic actions as well as our insights into general principles of hormone signaling.


Subject(s)
Receptors, Glucocorticoid/metabolism , Relaxin/metabolism , Cell Line , Cytokines/biosynthesis , Cytokines/metabolism , DNA/metabolism , Endotoxins/antagonists & inhibitors , Endotoxins/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fluorescence Polarization , Humans , Immunoprecipitation , Membrane Proteins/metabolism , Pregnancy , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Receptors, Glucocorticoid/agonists , Receptors, Peptide/metabolism , Relaxin/pharmacology , Spleen/cytology , Spleen/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism
9.
Ann N Y Acad Sci ; 1041: 441-3, 2005 May.
Article in English | MEDLINE | ID: mdl-15956743

ABSTRACT

Current findings in male relaxin knockout mice indicate antifibrotic and positive lusitropic actions of relaxin on the myocardium. We investigated in 12-month-old male spontaneously hypertensive rats (SHR) and age-matched male Wistar-Kyoto rats (WKY) whether RLX shows antihypertrophic actions as well. SHR showed left heart hypertrophy and a selective elevation of left atrial and ventricular relaxin peptide which inversely correlated with the degree of hypertrophy. In adult rat cardiomyocytes, relaxin blunted phenylephrine-induced hypertrophy by inhibiting activated ERK-1/2 kinases. In conclusion, endogenous myocardial relaxin, upregulated in left heart hypertrophy, exerts antihypertrophic effects by inhibition of activated ERK-1/2 kinases.


Subject(s)
Hypertension/metabolism , Hypertension/pathology , Hypertrophy/metabolism , Hypertrophy/pathology , Myocardium/metabolism , Myocardium/pathology , Relaxin/metabolism , Animals , Hypertension/genetics , Hypertension/physiopathology , Hypertrophy/genetics , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Relaxin/deficiency , Relaxin/genetics
10.
FASEB J ; 18(13): 1536-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15289446

ABSTRACT

The insulin-like peptide relaxin is a central hormone of pregnancy, but it also produces anti-fibrotic, myocardial, renal, central-nervous, and vascular effects. Recently, two G protein-coupled receptors, LGR7 and LGR8, have been identified as relaxin receptors. Prompted by reports on immunoregulatory effects of relaxin, we investigated possible interactions with the human glucocorticoid receptor (GR). Relaxin blunted the endotoxin-induced production of inflammatory cytokines (IL-1, IL-6, TNF-alpha) by human macrophages--an effect that was suppressed by the GR antagonist RU-486. In three different cell lines, relaxin induced GR activation, nuclear translocation, and DNA binding as assessed in GRE-luciferase assays. Co-immunoprecipitation experiments revealed physical interaction of endogenous and exogenous relaxin with cytoplasmic and nuclear GR. Relaxin competed with GR agonists for GR binding, both in vivo in whole-cell assays, and in vitro in fluorescence polarization assays. Relaxin was shown to up-regulate GR protein expression as well as the number of functionally active GR sites. In LGR7/8-free cells, the relaxin-mediated activation of GR was preserved. In conclusion, relaxin acts as GR agonist--a pathway pivotal to its effects on cytokine secretion by human macrophages. These findings may deepen our understanding of relaxin's abundant physiological actions, as well as our insights into general principles of hormone signaling.


Subject(s)
Receptors, Glucocorticoid/agonists , Relaxin/pharmacology , Active Transport, Cell Nucleus/drug effects , Binding Sites , Cell Line , Cell Nucleus/metabolism , Cytokines/biosynthesis , DNA/metabolism , Endotoxins/antagonists & inhibitors , Endotoxins/pharmacology , Female , Fluorescence Polarization , Genes, Reporter/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Immunoprecipitation , Inflammation Mediators/metabolism , Mifepristone/pharmacology , Pregnancy , Protein Binding , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Response Elements/genetics , Up-Regulation/drug effects
11.
Biochem Biophys Res Commun ; 316(3): 659-65, 2004 Apr 09.
Article in English | MEDLINE | ID: mdl-15033450

ABSTRACT

Attachment of leukocytes to the vascular endothelium and the subsequent migration of cells into the vessel wall are early events in atherogenesis. This process requires the expression of endothelial adhesion molecules. Since tea catechins are reputed to promote antiatherogenic activities, we investigated the effects of various tea catechins-i.e., epicatechin (EC), epicatechin gallate (ECG), epigallocatechin (EGC), and epigallocatechin-3-gallate (EGCG)-on cytokine-induced expression of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and endothelial leukocyte adhesion molecule-1 (E-selectin) in HUVECs by ELISA. EGCG and to a lesser extent ECG prevented the induction of VCAM-1 expression in a concentration-dependent manner after stimulation with TNF-alpha, whereas EC and EGC were without effect. EGCG also inhibited the IL-1beta-induced induction of VCAM-1 expression. Inhibition of cytokine-induced VCAM-1 expression was manifested already on the transcriptional level. Furthermore, EGCG reduced the TNF-alpha-induced adhesion of THP-1 cells to HUVECs. EGCG did not influence TNF-alpha-stimulated NF-kappaB activation.


Subject(s)
Catechin/analogs & derivatives , Catechin/pharmacology , Cytokines/biosynthesis , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Monocytes/metabolism , Tea , Vascular Cell Adhesion Molecule-1/biosynthesis , Cell Adhesion , Cell Line , Coculture Techniques , Dose-Response Relationship, Drug , E-Selectin/biosynthesis , Enzyme-Linked Immunosorbent Assay , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Interleukin-1/biosynthesis , NF-kappa B/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Umbilical Veins/cytology
12.
FASEB J ; 18(2): 272-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14769821

ABSTRACT

The ubiquitin-proteasome system is the major pathway for intracellular protein degradation in eukaryotic cells. Endothelial nitric oxide synthase (eNOS) is the key enzyme of vascular homeostasis involved in the pathophysiology of several cardiovascular diseases. The aim of our study was to investigate whether eNOS expression and activity are regulated by the proteasome. Bovine pulmonary artery endothelial cells (CPAE cells) were treated with the proteasome inhibitor MG132. MG132 (50-250 nmol/L) dose-dependently increased mRNA and protein levels of eNOS. Comparable results were obtained with other specific proteasome inhibitors, whereas the nonproteasomal calpain and cathepsin inhibitor ALLM had no effect. Efficacy of proteasome inhibition was evidenced by accumulation of poly-ubiquitinylated proteins and by measuring proteasomal activity in cell extracts. Cycloheximide prevented up-regulation of eNOS protein, indicating that post-translational stabilization of eNOS is not involved. eNOS activity was increased up to 2.8-fold (MG132 100 nmol/L, 48 h). Incubation of rat aortic rings with MG132 significantly enhanced endothelial-dependent vasorelaxation. Single MG132 treatment (100 nmol/L) induced long-term effects in CPAE cells, with increases of eNOS protein and activity for up to 10 days. Our results indicate that low-dose proteasome inhibition enhances eNOS expression and activity, and improves endothelial function.


Subject(s)
Endothelium, Vascular/drug effects , Enzyme Induction/drug effects , Leupeptins/pharmacology , Multienzyme Complexes/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Ubiquitin/antagonists & inhibitors , Ubiquitin/metabolism , Animals , Cattle , Cysteine Endopeptidases/metabolism , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiology , Male , Multienzyme Complexes/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type III , Proteasome Endopeptidase Complex , Pulmonary Artery , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Time Factors , Vasodilation
13.
Circ Res ; 92(1): 32-40, 2003 Jan 10.
Article in English | MEDLINE | ID: mdl-12522118

ABSTRACT

We have recently demonstrated that relaxin (RLX) acts as compensatory mediator in human heart failure. RLX inhibits the stimulation of endothelin-1, the most potent vasoconstrictor in heart failure. Upregulation of the endothelin type-B receptor (ET(B)), which mediates endothelin-1 clearance and endothelial release of NO, represents a pivotal mode of RLX action. However, signal transduction and abundance of this phenomenon are unknown. Therefore, we investigated RLX-induced regulation of ET(B) in human umbilical vein endothelial, epithelial (HeLa), and vascular smooth muscle cells. In human umbilical vein endothelial cells and HeLa cells, but not in human vascular smooth muscle cells, RLX upregulated ET(B) expression and activated extracellular signal-regulated kinase-1/2 (ERK-1/2) and nuclear factor-kappaB (NF-kappaB), a transcription factor. PD-98059, a selective inhibitor of the mitogen-activated protein kinase kinase-1 (MEK-1)-ERK-1/2 pathway, abolished ERK-1/2 and NF-kappaB activation and ET(B) upregulation. NF-kappaB inhibition also prevented RLX-mediated ET(B) stimulation. In NF-kappaB-luciferase reporter assays, we demonstrated complete inhibition of RLX-induced NF-kappaB activation in cells transfected with dominant-negative Raf-1, MEK-1, or ERK-1/2 constructs, whereas dominant-negative Ras had no effect. In rat aorta and mesenteric artery, RLX pretreatment, in an ET(B)-dependent fashion, mitigated the maximum contractile response to endothelin-1, by 38+/-4% and 43+/-6%, and the endothelin-1 sensitivity (-log[EC(50)]: aorta, 8.2+/-0.2 for vehicle versus 7.2+/-0.2 for RLX; mesenteric artery, 8.0+/-0.2 for vehicle versus 7.1+/-0.1 for RLX). RLX pretreatment augmented the dilator effect of the ET(B) agonist endothelin-3 by 100+/-8% and 133+/-13%. In conclusion, RLX stimulates endothelial and epithelial ET(B) via a Ras-independent Raf-1-MEK-1-ERK-1/2 pathway that activates NF-kappaB. On vascular smooth muscle cells, ET(B), a contributor to endothelin-mediated vasoconstriction, remains unaffected. This renders RLX a functional endothelin-1 antagonist.


Subject(s)
Endothelin-1/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Receptors, Endothelin/metabolism , Relaxin/pharmacology , Vasoconstriction/drug effects , Animals , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Endothelin-1/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Activators/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression/drug effects , Genes, Reporter , HeLa Cells , Humans , MAP Kinase Kinase 1 , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar , Receptor, Endothelin B , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection , Up-Regulation/drug effects , Vasoconstriction/physiology
14.
Regul Pept ; 110(1): 33-8, 2002 Dec 31.
Article in English | MEDLINE | ID: mdl-12468107

ABSTRACT

The peptide urotensin-II (U-II) has been described as most potent vasoconstrictor identified so far, but plasma values in humans and its role in cardiovascular pathophysiology are unknown. We investigated circulating urotensin-II and its potential role in human congestive heart failure (CHF). We enrolled control individuals (n=13; cardiac index [CI], 3.5+/-0.1 l/min/m2; pulmonary wedge pressure [PCWP], 10+/-1 mm Hg), patients with moderate (n=10; CI, 2.9+/-0.3 l/min/m2; PCWP, 14+/-2 mm Hg) and severe CHF (n=11; CI, 1.8+/-0.2 l/min/m2; PCWP, 33+/-2 mm Hg). Plasma levels of urotensin-II differed neither between controls, patients with moderate and severe CHF nor between different sites of measurement (pulmonary artery, left ventricle, coronary sinus, antecubital vein) within the single groups. Hemodynamic improvement by vasodilator therapy in severe CHF (CI, +78+/-3%; PCWP, -55+/-3%) did not affect circulating U-II over 24 h. Preprourotensin-II mRNA expression in right atria, left ventricles, mammary arteries and saphenous veins did not differ between controls with normal heart function and patients with end-stage CHF. In conclusion, urotensin-II plasma levels and its myocardial and vascular gene expression are unchanged in human CHF. Circulating urotensin-II does not respond to acute hemodynamic improvement. These findings suggest that urotensin-II does not play a major role in human CHF.


Subject(s)
Heart Failure/blood , Myocardium/metabolism , Urotensins/blood , Adult , Female , Gene Expression , Heart Failure/genetics , Heart Failure/physiopathology , Hemodynamics , Humans , Immunoenzyme Techniques , Male , Middle Aged , Protein Precursors/metabolism , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Urotensins/biosynthesis , Urotensins/genetics , Vasodilator Agents/therapeutic use
15.
Biochem Biophys Res Commun ; 294(2): 315-8, 2002 Jun 07.
Article in English | MEDLINE | ID: mdl-12051712

ABSTRACT

Adrenomedullin (AM) is a powerful pulmonary vasodilator with antimitogenic properties. We investigated the role of the AM receptor (AMR) and the calcitonin gene-related peptide type-1 receptor (CGRP1R) in regulating pulmonary vascular AM levels. The AMR antagonist hAM(22-52) (120 nmol/L) significantly elevated AM release compared with controls to 250% after 2 h in isolated rat lungs and to 830% after 4 h in pulmonary artery endothelial cells (PAEC). CGRP1R blockade had no effect. AMR blockade did not influence prepro-AM mRNA levels nor did inhibition of protein synthesis by cycloheximide (0.01 mg/mL) abolish the effect of the AMR antagonist. Radioligand-binding studies with PAEC membranes revealed a decrease by 44% of the AMR density in response to AMR antagonism. Altogether, the pulmonary vascular AMR represents not only a functionally active, but also a clearance receptor; its expression is constitutively stimulated by basal AM. This identifies a novel mechanism for controlling pulmonary AM levels.


Subject(s)
Endothelium, Vascular/metabolism , Lung/physiology , Pulmonary Circulation/physiology , Receptors, Peptide/metabolism , Adrenomedullin , Animals , Binding, Competitive , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Gene-Related Peptide Receptor Antagonists , Cell Membrane/metabolism , Cells, Cultured , Cycloheximide/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , In Vitro Techniques , Ligands , Lung/blood supply , Male , Peptide Fragments/pharmacology , Peptides/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Synthesis Inhibitors/pharmacology , Pulmonary Artery/cytology , Pulmonary Artery/physiology , RNA, Messenger/metabolism , Radioligand Assay , Rats , Rats, Wistar , Receptors, Adrenomedullin , Receptors, Calcitonin Gene-Related Peptide/metabolism , Receptors, Peptide/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...