Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Sci Rep ; 8(1): 791, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29335435

ABSTRACT

Maternal depressive symptoms during pregnancy predict increased psychiatric problems in children. The underlying biological mechanisms remain unclear. Hence, we examined whether alterations in the morphology of 88 term placentas were associated with maternal depressive symptoms during pregnancy and psychiatric problems in 1.9-3.1-years old (Mean = 2.1 years) toddlers. Maternal depressive symptoms were rated biweekly during pregnancy with the Center of Epidemiological Studies Depression Scale (n = 86). Toddler psychiatric problems were mother-rated with the Child Behavior Checklist (n = 60). We found that higher maternal depressive symptoms throughout pregnancy [B = -0.24 Standard Deviation (SD) units: 95% Confidence Interval (CI) = -0.46; -0.03: P = 0.03; Mean difference = -0.66 SDs; 95% CI = -0.08; -1.23: P = 0.03; between those with and without clinically relevant depressive symptoms] were associated with lower variability in the placental villous barrier thickness of γ-smooth muscle actin-negative villi. This placental morphological change predicted higher total (B = -0.34 SDs: 95% CI = -0.60; -0.07: P = 0.01) and internalizing (B = -0.32 SDs: 95% CI = -0.56; -0.08: P = 0.01) psychiatric problems in toddlers. To conclude, our findings suggest that both maternal depressive symptoms during pregnancy and toddler psychiatric problems may be associated with lower variability in the villous membrane thickness of peripheral villi in term placentas. This lower heterogeneity may compromise materno-fetal exchange, suggesting a possible role for altered placental morphology in the fetal programming of mental disorders.


Subject(s)
Depression/pathology , Mental Disorders/diagnosis , Mothers/psychology , Placenta/pathology , Actins/metabolism , Adult , Child, Preschool , Chorionic Villi/metabolism , Female , Humans , Infant , Male , Mother-Child Relations , Pre-Eclampsia/diagnosis , Pregnancy , Pregnancy Complications , Psychiatric Status Rating Scales
2.
Thromb Haemost ; 113(2): 329-37, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25354586

ABSTRACT

Carbon monoxide (CO) is a gaseous autacoid known to positively regulate vascular tone; however, its role in angiogenesis is unknown. The aim of this study was to investigate the effect of CO on angiogenesis and vascular endothelial growth factor (VEGF) receptor-2 phosphorylation. Human umbilical vein endothelial cells (HUVECs) were cultured on growth factor-reduced Matrigel and treated with a CO-releasing molecule (CORM-2) or exposed to CO gas (250 ppm). Here, we report the surprising finding that exposure to CO inhibits vascular endothelial growth factor (VEGF)-induced endothelial cell actin reorganisation, cell proliferation, migration and capillary-like tube formation. Similarly, CO suppressed VEGF-mediated phosphorylation of VEGFR-2 at tyrosine residue 1175 and 1214 and basic fibroblast growth factor- (FGF-2) and VEGF-mediated Akt phosphorylation. Consistent with these data, mice exposed to 250 ppm CO (1h/day for 14 days) exhibited a marked decrease in FGF-2-induced Matrigel plug angiogenesis (p<0.05). These data establish a new biological function for CO in angiogenesis and point to a potential therapeutic use for CO as an anti-angiogenic agent in tumour suppression.


Subject(s)
Carbon Monoxide/chemistry , Neovascularization, Pathologic , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Movement , Cell Proliferation , Collagen/chemistry , Drug Combinations , Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells , Humans , Immunoprecipitation , Laminin/chemistry , Mice, Inbred C57BL , Organometallic Compounds/chemistry , Phosphorylation , Proteoglycans/chemistry , Recombinant Proteins/chemistry , Tyrosine/chemistry
4.
Circulation ; 127(25): 2514-22, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23704251

ABSTRACT

BACKGROUND: The exact etiology of preeclampsia is unknown, but there is growing evidence of an imbalance in angiogenic growth factors and abnormal placentation. Hydrogen sulfide (H2S), a gaseous messenger produced mainly by cystathionine γ-lyase (CSE), is a proangiogenic vasodilator. We hypothesized that a reduction in CSE activity may alter the angiogenic balance in pregnancy and induce abnormal placentation and maternal hypertension. METHODS AND RESULTS: Plasma levels of H2S were significantly decreased in women with preeclampsia (P<0.01), which was associated with reduced placental CSE expression as determined by real-time polymerase chain reaction and immunohistochemistry. Inhibition of CSE activity by DL-propargylglycine reduced placental growth factorproduction from first-trimester (8-12 weeks gestation) human placental explants and inhibited trophoblast invasion in vitro. Knockdown of CSE in human umbilical vein endothelial cells by small-interfering RNA increased the release of soluble fms-like tyrosine kinase-1 and soluble endoglin, as assessed by enzyme-linked immunosorbent assay, whereas adenoviral-mediated CSE overexpression in human umbilical vein endothelial cells inhibited their release. Administration of DL-propargylglycine to pregnant mice induced hypertension and liver damage, promoted abnormal labyrinth vascularization in the placenta, and decreased fetal growth. Finally, a slow-releasing H2S-generating compound, GYY4137, inhibited circulating soluble fms-like tyrosine kinase-1 and soluble endoglin levels and restored fetal growth in mice that was compromised by DL-propargylglycine treatment, demonstrating that the effect of CSE inhibitor was attributable to inhibition of H2S production. CONCLUSIONS: These results imply that endogenous H2S is required for healthy placental vasculature and that a decrease in CSE/H2S activity may contribute to the pathogenesis of preeclampsia.


Subject(s)
Cystathionine gamma-Lyase/metabolism , Hydrogen Sulfide/antagonists & inhibitors , Hydrogen Sulfide/metabolism , Hypertension/etiology , Placenta Diseases/etiology , Pre-Eclampsia/metabolism , Pregnancy Complications, Cardiovascular/etiology , Adolescent , Adult , Alkynes/adverse effects , Alkynes/pharmacology , Animals , Antigens, CD/metabolism , Cells, Cultured , Disease Models, Animal , Endoglin , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Fetal Development/drug effects , Fetal Development/physiology , Glycine/adverse effects , Glycine/analogs & derivatives , Glycine/pharmacology , Humans , Hypertension/chemically induced , Hypertension/metabolism , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , Neovascularization, Physiologic/physiology , Organ Culture Techniques , Organothiophosphorus Compounds/pharmacology , Placenta/drug effects , Placenta/metabolism , Placenta/physiopathology , Placenta Diseases/metabolism , Placenta Diseases/physiopathology , Placenta Growth Factor , Pre-Eclampsia/physiopathology , Pregnancy , Pregnancy Complications, Cardiovascular/metabolism , Pregnancy Complications, Cardiovascular/physiopathology , Pregnancy Proteins/metabolism , Pregnancy, Animal , Receptors, Cell Surface/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Young Adult
5.
Nat Commun ; 3: 972, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22828632

ABSTRACT

VEGF-A activity is tightly regulated by ligand and receptor availability. Here we investigate the physiological function of heterodimers between VEGF receptor-1 (VEGFR-1; Flt-1) and VEGFR-2 (KDR; Flk-1) (VEGFR(1-2)) in endothelial cells with a synthetic ligand that binds specifically to VEGFR(1-2). The dimeric ligand comprises one VEGFR-2-specific monomer (VEGF-E) and a VEGFR-1-specific monomer (PlGF-1). Here we show that VEGFR(1-2) activation mediates VEGFR phosphorylation, endothelial cell migration, sustained in vitro tube formation and vasorelaxation via the nitric oxide pathway. VEGFR(1-2) activation does not mediate proliferation or elicit endothelial tissue factor production, confirming that these functions are controlled by VEGFR-2 homodimers. We further demonstrate that activation of VEGFR(1-2) inhibits VEGF-A-induced prostacyclin release, phosphorylation of ERK1/2 MAP kinase and mobilization of intracellular calcium from primary endothelial cells. These findings indicate that VEGFR-1 subunits modulate VEGF activity predominantly by forming heterodimer receptors with VEGFR-2 subunits and such heterodimers regulate endothelial cell homeostasis.


Subject(s)
Endothelial Cells/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoprecipitation , Protein Multimerization , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
6.
Eur Heart J ; 33(9): 1150-8, 2012 May.
Article in English | MEDLINE | ID: mdl-21411816

ABSTRACT

AIMS: Endothelial dysfunction is a hallmark of preeclampsia. Desensitization of the phosphoinositide 3-kinase (PI3K)/Akt pathway underlies endothelial dysfunction and haeme oxygenase-1 (HO-1) is decreased in preeclampsia. To identify therapeutic targets, we sought to assess whether these two regulators act to suppress soluble endoglin (sEng), an antagonist of transforming growth factor-ß (TGF-ß) signalling, which is known to be elevated in preeclampsia. METHODS AND RESULTS: Vascular endothelial growth factor-A (VEGF-A), fibroblast growth factor (FGF-2), angiopoietin-1 (Ang-1), and insulin, which all activate the PI3K/Akt pathway, inhibited the release of sEng from endothelial cells. Inhibition of the PI3K/Akt pathway, by overexpression of phosphatase and tensin homolog (PTEN) or a dominant-negative isoform of Akt (Akt(dn)) induced sEng release from endothelial cells and prevented the inhibitory effect of VEGF-A. Conversely, overexpression of a constitutively active Akt (Akt(myr)) inhibited PTEN and cytokine-induced sEng release. Systemic delivery of Akt(myr) to mice significantly reduced circulating sEng, whereas Akt(dn) promoted sEng release. Phosphorylation of Akt was reduced in preeclamptic placenta and this correlated with the elevated level of circulating sEng. Knock-down of Akt using siRNA prevented HO-1-mediated inhibition of sEng release and reduced HO-1 expression. Furthermore, HO-1 null mice have reduced phosphorylated Akt in their organs and overexpression of Akt(myr) failed to suppress the elevated levels of sEng detected in HO-1 null mice, indicating that HO-1 is required for the Akt-mediated inhibition of sEng. CONCLUSION: The loss of PI3K/Akt and/or HO-1 activity promotes sEng release and positive manipulation of these pathways offers a strategy to circumvent endothelial dysfunction.


Subject(s)
Antigens, CD/metabolism , Heme Oxygenase-1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pre-Eclampsia/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cell Surface/metabolism , Angiopoietin-1/metabolism , Animals , Endoglin , Endothelium, Vascular/metabolism , Female , Fibroblast Growth Factor 2/metabolism , Humans , Insulin/metabolism , Mice , Mice, Inbred C57BL , PTEN Phosphohydrolase/metabolism , Phosphorylation , Pregnancy , RNA, Messenger/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Umbilical Veins/metabolism , Vascular Endothelial Growth Factor A/metabolism
7.
Am J Obstet Gynecol ; 206(3): 253.e10-5, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22197494

ABSTRACT

OBJECTIVE: Soluble vascular endothelial growth factor receptor-1 (also know as soluble fms-like tyrosine kinase [sFlt]-1) is a key causative factor of preeclampsia. Resveratrol, a plant phytoalexin, has antiinflammatory and cardioprotective properties. We sought to determine the effect of resveratrol on sFlt-1 release. STUDY DESIGN: Human umbilical vein endothelial cells, transformed human trophoblast-8 (HTR/SVneo)-8/SVneo trophoblast cells, or placental explants were incubated with cytokines and/or resveratrol. Conditioned media were assayed for sFlt-1 by enzyme-linked immunosorbent assay and cell proteins used for Western blotting. RESULTS: Resveratrol inhibited cytokine-induced release of sFlt-1 from normal placental explants and from preeclamptic placental explants. Preincubation of human umbilical vein endothelial cells or HTR-8/SVneo cells with resveratrol abrogated sFlt-1 release. Resveratrol prevented the up-regulation of early growth response protein-1 (Egr-1), a transcription factor necessary for induction of the vascular endothelial growth factor receptor-1 gene and caused up-regulation of heme oxygenase-1, a cytoprotective enzyme found to be dysfunctional in preeclampsia. CONCLUSION: In summary, resveratrol can inhibit sFlt-1 release and up-regulate heme oxygenase-1; thus, may offer therapeutic potential in preeclampsia.


Subject(s)
Enzyme Inhibitors/pharmacology , Placenta/drug effects , Placenta/enzymology , Stilbenes/pharmacology , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Cells, Cultured , Cytokines/pharmacology , Early Growth Response Protein 1/biosynthesis , Female , Heme Oxygenase-1/biosynthesis , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Pre-Eclampsia/enzymology , Pregnancy , Resveratrol , Up-Regulation , Vascular Endothelial Growth Factor Receptor-1/metabolism
8.
Vasc Cell ; 3(1): 15, 2011 Jul 13.
Article in English | MEDLINE | ID: mdl-21752276

ABSTRACT

BACKGROUND: The negative feedback system is an important physiological regulatory mechanism controlling angiogenesis. Soluble vascular endothelial growth factor (VEGF) receptor-1 (sFlt-1), acts as a potent endogenous soluble inhibitor of VEGF- and placenta growth factor (PlGF)-mediated biological function and can also form dominant-negative complexes with competent full-length VEGF receptors. METHODS AND RESULTS: Systemic overexpression of VEGF-A in mice resulted in significantly elevated circulating sFlt-1. In addition, stimulation of human umbilical vein endothelial cells (HUVEC) with VEGF-A, induced a five-fold increase in sFlt-1 mRNA, a time-dependent significant increase in the release of sFlt-1 into the culture medium and activation of the flt-1 gene promoter. This response was dependent on VEGF receptor-2 (VEGFR-2) and phosphoinositide-3'-kinase signalling. siRNA-mediated knockdown of sFlt-1 in HUVEC stimulated the activation of endothelial nitric oxide synthase, increased basal and VEGF-induced cell migration and enhanced endothelial tube formation on growth factor reduced Matrigel. In contrast, adenoviral overexpression of sFlt-1 suppressed phosphorylation of VEGFR-2 at tyrosine 951 and ERK-1/-2 MAPK and reduced HUVEC proliferation. Preeclampsia is associated with elevated placental and systemic sFlt-1. Phosphorylation of VEGFR-2 tyrosine 951 was greatly reduced in placenta from preeclamptic patients compared to gestationally-matched normal placenta. CONCLUSION: These results show that endothelial sFlt-1 expression is regulated by VEGF and acts as an autocrine regulator of endothelial cell function.

9.
Development ; 138(10): 2111-20, 2011 May.
Article in English | MEDLINE | ID: mdl-21521739

ABSTRACT

Endothelial tip cells guide angiogenic sprouts by exploring the local environment for guidance cues such as vascular endothelial growth factor (VegfA). Here we present Flt1 (Vegf receptor 1) loss- and gain-of-function data in zebrafish showing that Flt1 regulates tip cell formation and arterial branching morphogenesis. Zebrafish embryos expressed soluble Flt1 (sFlt1) and membrane-bound Flt1 (mFlt1). In Tg(flt1(BAC):yfp) × Tg(kdrl:ras-cherry)(s916) embryos, flt1:yfp was expressed in tip, stalk and base cells of segmental artery sprouts and overlapped with kdrl:cherry expression in these domains. flt1 morphants showed increased tip cell numbers, enhanced angiogenic behavior and hyperbranching of segmental artery sprouts. The additional arterial branches developed into functional vessels carrying blood flow. In support of a functional role for the extracellular VEGF-binding domain of Flt1, overexpression of sflt1 or mflt1 rescued aberrant branching in flt1 morphants, and overexpression of sflt1 or mflt1 in controls resulted in short arterial sprouts with reduced numbers of filopodia. flt1 morphants showed reduced expression of Notch receptors and of the Notch downstream target efnb2a, and ectopic expression of flt4 in arteries, consistent with loss of Notch signaling. Conditional overexpression of the notch1a intracellular cleaved domain in flt1 morphants restored segmental artery patterning. The developing nervous system of the trunk contributed to the distribution of Flt1, and the loss of flt1 affected neurons. Thus, Flt1 acts in a Notch-dependent manner as a negative regulator of tip cell differentiation and branching. Flt1 distribution may be fine-tuned, involving interactions with the developing nervous system.


Subject(s)
Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-1/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Base Sequence , Blood Vessels/embryology , Blood Vessels/metabolism , Cell Differentiation , Gene Expression Regulation, Developmental , Macrophages/metabolism , Nervous System/embryology , Nervous System/metabolism , Oligodeoxyribonucleotides, Antisense/genetics , Receptors, Notch/metabolism , Signal Transduction , Solubility , Tissue Distribution , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/genetics , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
10.
Hypertension ; 56(3): 477-83, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20696992

ABSTRACT

Angiopoietin-1 (Ang-1) is an angiogenic growth factor that activates Tie-2 and integrins to promote vessel wall remodeling. The recent finding of the potential proatherogenic effects of Ang-1 prompted us to investigate whether Ang-1 promotes monocyte chemotaxis, endothelial binding, and transendothelial migration, key events in the progression of atherosclerosis. Here, we show that Ang-1 induces chemotaxis of monocytes in a manner that is independent of Tie-2 and integrin binding but dependent on phosphoinositide 3-kinase and heparin. In addition, Ang-1 promoted phosphoinositide 3-kinase-dependent binding of monocytes to endothelial monolayers and stimulated transendothelial migration. Fluorescence-activated cell sorting analysis showed that exogenous Ang-1 adheres directly to monocytes as well as to human umbilical endothelial cells, but neither Tie-2 mRNA nor protein were expressed by primary monocytes. Although Ang-1 binding to human umbilical endothelial cells was partially Tie-2 and integrin dependent, Ang-1 binding to monocytes was independent of these factors. Finally, preincubation of monocytes with soluble heparin abrogated Ang-1 binding to monocytes and migration, and partially prevented Ang-1 binding to human umbilical endothelial cells. In summary, Ang-1 induces chemotaxis of monocytes by a mechanism that is dependent on phosphoinositide 3-kinase and heparin but independent of Tie-2 and integrins. The ability of Ang-1 to recruit monocytes suggests it may play a role in inflammatory angiogenesis and may promote atherosclerosis.


Subject(s)
Angiopoietin-1/pharmacology , Chemotaxis, Leukocyte/drug effects , Integrins/metabolism , Monocytes/drug effects , Receptor, TIE-2/metabolism , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Angiopoietin-2/pharmacology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Heparin/metabolism , Heparin/pharmacology , Humans , Monocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism
11.
Hypertension ; 56(1): 172-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20479331

ABSTRACT

Preeclampsia is a hypertensive disorder of pregnancy caused by abnormal placental function, partly because of chronic hypoxia at the utero-placental junction. The increase in levels of soluble vascular endothelial growth factor receptor 1, an antiangiogenic agent known to inhibit placental vascularization, is an important cellular factor implicated in the onset of preeclampsia. We investigated the ligand urotensin II (U-II), a potent endogenous vasoconstrictor and proangiogenic agent, for which levels have been reported to increase in patients with preeclampsia. We hypothesized that an increased sensitivity to U-II in preeclampsia might be achieved by upregulation of placental U-II receptors. We further investigated the role of U-II receptor stimulation on soluble vascular endothelial growth factor receptor 1 release in placental explants from diseased and normal patients. Immunohistochemistry, real-time PCR, and Western blotting analysis revealed that U-II receptor expression was significantly upregulated in preeclampsia placentas compared with controls (P<0.01). Cellular models of syncytiotrophoblast and vascular endothelial cells subjected to hypoxic conditions revealed an increase in U-II receptor levels in the syncytiotrophoblast model. This induction is regulated by the transcriptional activator hypoxia-inducible factor 1alpha. U-II treatment is associated with increased secretion of soluble vascular endothelial growth factor receptor 1 only in preeclamptic placental explants under hypoxia but not in control conditions. Interestingly, normal placental explants did not respond to U-II stimulation.


Subject(s)
Endothelium, Vascular/metabolism , Placenta/metabolism , Pre-Eclampsia/genetics , RNA/genetics , Receptors, G-Protein-Coupled/genetics , Up-Regulation , Vascular Endothelial Growth Factor Receptor-1/metabolism , Blotting, Western , Cells, Cultured , Endothelium, Vascular/physiopathology , Female , Humans , Immunohistochemistry , Placenta/pathology , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy , Receptors, G-Protein-Coupled/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Vasodilation/physiology
12.
Hypertension ; 55(3): 689-97, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20124108

ABSTRACT

The proteinase-activated receptor 2 (PAR-2) expression is increased in endothelial cells derived from women with preeclampsia, characterized by widespread maternal endothelial damage, which occurs as a consequence of elevated soluble vascular endothelial growth factor receptor-1 (sVEGFR-1; commonly known as sFlt-1) in the maternal circulation. Because PAR-2 is upregulated by proinflammatory cytokines and activated by blood coagulation serine proteinases, we investigated whether activation of PAR-2 contributed to sVEGFR-1 release. PAR-2-activating peptides (SLIGRL-NH(2) and 2-furoyl-LIGRLO-NH(2)) and factor Xa increased the expression and release of sVEGFR-1 from human umbilical vein endothelial cells. Enzyme-specific, dominant-negative mutants and small interfering RNA were used to demonstrate that PAR-2-mediated sVEGFR-1 release depended on protein kinase C-beta(1) and protein kinase C-epsilon, which required intracellular transactivation of epidermal growth factor receptor 1, leading to mitogen-activated protein kinase activation. Overexpression of heme oxygenase 1 and its gaseous product, carbon monoxide, decreased PAR-2-stimulated sVEGFR-1 release from human umbilical vein endothelial cells. Simvastatin, which upregulates heme oxygenase 1, also suppressed PAR-2-mediated sVEGFR-1 release. These results show that endothelial PAR-2 activation leading to increased sVEGFR-1 release may contribute to the maternal vascular dysfunction observed in preeclampsia and highlights the PAR-2 pathway as a potential therapeutic target for the treatment of preeclampsia.


Subject(s)
Endothelial Cells/metabolism , ErbB Receptors/metabolism , Pre-Eclampsia/metabolism , Receptor, PAR-2/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Carbon Monoxide/metabolism , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/physiology , Endothelial Cells/cytology , Factor Xa/metabolism , Female , Gene Expression/physiology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kidney/cytology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Pregnancy , Protein Kinase C/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptor, PAR-2/agonists , Simvastatin/pharmacology , Solubility , Umbilical Veins/cytology , Vascular Endothelial Growth Factor Receptor-1/genetics
13.
J Cell Mol Med ; 14(6B): 1857-67, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19538465

ABSTRACT

Preeclampsia (PE) is characterized by widespread endothelial damage with hypertension, proteinuria, glomeruloendotheliosis and elevated soluble Flt-1 (sFlt-1), a natural occurring antagonist of vascular endothelial growth factor (VEGF). Cancer patients receiving anti-VEGF therapy exhibit similar symptoms. We suggested that a decrease in circulating sFlt-1 would alleviate the symptoms associated with PE. Adenoviral (Adv) overexpression of sFlt-1 induced proteinuria, caused glomerular damage and increase in blood pressure in female Balb/c mice. Circulating level of sFlt-1 above 50 ng/ml plasma induced severe vascular damage and glomerular endotheliosis. Albumin concentration in urine was elevated up to 30-fold, compared to control AdvGFP-treated animals. The threshold of kidney damage was in the range of 20-30 ng/ml sFlt-1 in plasma (8-15 ng/ml in urine). Co-administration of AdvsFlt-1 with AdvVEGF to neutralize circulating sFlt-1 resulted in more than a 70% reduction in free sFlt-1 in plasma, more than 80% reduction in urine and rescued the damaging effect of sFlt-1 on the kidneys. This demonstrates that below a critical threshold sFlt-1 fails to elicit damage to the fenestrated endothelium and that co-expression of VEGF is able to rescue effects mediated by sFlt-1 overexpression.


Subject(s)
Pre-Eclampsia/blood , Vascular Endothelial Growth Factor Receptor-1/blood , Animals , Blood Pressure/drug effects , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Glomerular Filtration Rate/drug effects , Heart Rate/drug effects , Kidney/drug effects , Kidney/pathology , Mice , Pre-Eclampsia/physiopathology , Pregnancy , Protein Binding/drug effects , Solubility/drug effects , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
Biochem Soc Trans ; 37(Pt 6): 1237-42, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19909254

ABSTRACT

Pre-eclampsia, a pregnancy-specific multi-organ syndrome characterized by widespread endothelial damage, is a new risk factor for cardiovascular disease. No therapies exist to prevent or treat this condition, even to achieve a modest improvement in pregnancy length or birth weight. Co-administration of soluble VEGFR-1 [VEGF (vascular endothelial growth factor) receptor-1; more commonly known as sFlt-1 (soluble Fms-like tyrosine kinase-1)] and sEng (soluble endoglin) to pregnant rats elicits severe pre-eclampsia-like symptoms. These two anti-angiogenic factors are increased dramatically prior to the clinical onset of pre-eclampsia and are quite possibly the 'final common pathway' responsible for the accompanying signs of hypertension and proteinuria as they can be reversed by VEGF administration in animal models. HO-1 (haem oxygenase-1), an anti-inflammatory enzyme, and its metabolite, CO (carbon monoxide), exert protective effects in several organs against oxidative stimuli. In a landmark publication, we showed that the HO-1 pathway inhibits sFlt-1 and sEng in cultured cells and human placental tissue explants. Both CO and NO (nitric oxide) promote vascular homoeostasis and vasodilatation, and activation of VEGFR-1 or VEGFR-2 induced eNOS (endothelial nitric oxide synthase) phosphorylation, NO release and HO-1 expression. Our studies established the HO-1/CO pathway as a negative regulator of cytokine-induced sFlt-1 and sEng release and eNOS as a positive regulator of VEGF-mediated vascular morphogenesis. These findings provide compelling evidence for a protective role of HO-1 in pregnancy and identify it as a target for the treatment of pre-eclampsia. Any agent that is known to up-regulate HO-1, such as statins, may have potential as a therapy. Any intervention achieving even a modest prolongation of pregnancy or amelioration of the condition could have a significant beneficial health impact worldwide.


Subject(s)
Heme Oxygenase-1/metabolism , Pre-Eclampsia/physiopathology , Vascular Endothelial Growth Factor A/metabolism , Animals , Carbon Monoxide/metabolism , Carbon Monoxide/therapeutic use , Endothelium, Vascular/metabolism , Female , Homeostasis , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Pre-Eclampsia/drug therapy , Pre-Eclampsia/prevention & control , Pregnancy , Randomized Controlled Trials as Topic , Rats , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-1/metabolism
15.
Circ Res ; 104(12): 1333-6, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19461044

ABSTRACT

Atherosclerosis is promoted by a combination of hypercholesterolemia and vascular inflammation. The function of Angiopoietin (Ang)-2, a key regulator of angiogenesis, in the maintenance of large vessels is unknown. A single systemic administration of Ang-2 adenovirus (AdAng-2) to apoE(-/-) mice fed a Western diet significantly reduced atherosclerotic lesion size ( approximately 40%) and oxidized LDL and macrophage content of the plaques. These beneficial effects were abolished by the inhibition of nitric oxide synthase (NOS). In endothelial cells, endothelial NOS activation per se inhibited LDL oxidation and Ang-2 stimulated NO release in a Tie2-dependent manner to decrease LDL oxidation. These findings demonstrate a novel atheroprotective role for Ang-2 when endothelial cell function is compromised and suggest that growth factors, which stimulate NO release without inducing inflammation, could offer atheroprotection.


Subject(s)
Angiopoietin-2/metabolism , Apolipoproteins E , Atherosclerosis/metabolism , Lipoproteins, LDL/metabolism , Receptor, TIE-2/metabolism , Adenoviridae , Angiopoietin-2/genetics , Animals , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Female , Lipoproteins, LDL/genetics , Male , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Oxidation-Reduction , Receptor, TIE-2/genetics , Transduction, Genetic , Vasculitis/genetics , Vasculitis/metabolism , Vasculitis/prevention & control
16.
Arterioscler Thromb Vasc Biol ; 27(12): 2619-26, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17901375

ABSTRACT

OBJECTIVE: Tie2 and its ligands, the angiopoietins (Ang), are required for embryonic and postnatal angiogenesis. Previous studies have demonstrated that Tie2 is proteolytically cleaved, resulting in the production of a 75-kDa soluble receptor fragment (sTie2). We investigated mechanisms responsible for Tie2 shedding and its effects on Tie2 signaling and endothelial cellular responses. METHODS AND RESULTS: sTie2 bound both Ang1 and Ang2 and inhibited angiopoietin-mediated Tie2 phosphorylation and antiapoptosis. In human umbilical vein endothelial cells, Tie2 shedding was both constitutive and induced by treatment with PMA or vascular endothelial growth factor (VEGF). Constitutive and VEGF-inducible Tie2 shedding were mediated by PI3K/Akt and p38 MAPK. Tie2 shedding was blocked by pharmacological inhibitors of either PI3K or Akt as well as by overexpression of the lipid phosphatase PTEN. In contrast, sTie2 shedding was enhanced by overexpression of either dominant negative PTEN, which increased Akt phosphorylation, or constitutively active, myristoylated Akt. CONCLUSIONS: These findings demonstrate that VEGF regulates angiopoietin-Tie2 signaling by inducing proteolytic cleavage and shedding of Tie2 via a novel PI3K/Akt-dependent pathway. These results suggest a previously unrecognized mechanism by which VEGF may inhibit vascular stabilization to promote angiogenesis and vascular remodeling.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Physiologic , Peptide Fragments/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, TIE-2/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Animals , Apoptosis , Cells, Cultured , Chromones/pharmacology , Dipeptides/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Imidazoles/pharmacology , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/metabolism , Mice , Morpholines/pharmacology , NIH 3T3 Cells , Neovascularization, Physiologic/drug effects , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Peptide Fragments/blood , Peptide Fragments/genetics , Peptide Hydrolases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyridines/pharmacology , Receptor, TIE-2/blood , Receptor, TIE-2/genetics , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Transfection , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Circulation ; 115(13): 1789-97, 2007 Apr 03.
Article in English | MEDLINE | ID: mdl-17389265

ABSTRACT

BACKGROUND: Preeclampsia is characterized clinically by hypertension and proteinuria. Soluble Flt-1 (sFlt-1; also known as soluble vascular endothelial growth factor receptor-1 [VEGFR-1]) and soluble endoglin (sEng) are elevated in preeclampsia, and their administration to pregnant rats elicits preeclampsia-like symptoms. Heme oxygenase-1 (HO-1) and its metabolite carbon monoxide (CO) exert protective effects against oxidative stimuli. Thus, we hypothesized that HO-1 upregulation may offer protection against preeclampsia by inhibiting sFlt-1 and sEng release. METHODS AND RESULTS: Preeclamptic villous explants secreted high levels of sFlt-1 and sEng. Adenoviral overexpression of HO-1 in endothelial cells inhibited VEGF-mediated sFlt-1 release and interferon-gamma- and tumor necrosis factor-alpha-induced sEng release, whereas HO-1 inhibition potentiated sFlt-1 and sEng production from endothelial cells and placental villous explants. Consistent with these findings, mice lacking HO-1 produced higher levels of sFlt-1 and sEng compared with wild-type mice. Using selective ligands (VEGF-E and placental growth factor) and a receptor-specific inhibitor (SU-1498), we demonstrated that VEGF-induced sFlt-1 release was VEGFR-2 dependent. Furthermore, CO-releasing molecule-2 (CORM-2) or CO decreased sFlt-1 release and inhibited VEGFR-2 phosphorylation. Treatment of endothelial cells with statins upregulated HO-1 and inhibited the release of sFlt-1, whereas vitamins C and E had no effect. CONCLUSIONS: The present study demonstrates that the HO-1/CO pathway inhibits sFlt-1 and sEng release, providing compelling evidence for a protective role of HO-1 in pregnancy, and identifies HO-1 as a novel target for the treatment of preeclampsia.


Subject(s)
Antigens, CD/physiology , Heme Oxygenase (Decyclizing)/physiology , Heme Oxygenase-1/physiology , Pre-Eclampsia/metabolism , Receptors, Cell Surface/physiology , Vascular Endothelial Growth Factor Receptor-1/physiology , Animals , Antioxidants/pharmacology , Carbon Monoxide/pharmacology , Cell Hypoxia , Cells, Cultured/drug effects , Culture Media, Serum-Free/pharmacology , Endoglin , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Female , Genetic Vectors , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1/deficiency , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Interferon-gamma/pharmacology , Mice , Mice, Knockout , Organ Culture Techniques , Organometallic Compounds/pharmacology , Oxidative Stress , Placenta/pathology , Placenta Growth Factor , Pre-Eclampsia/pathology , Pregnancy , Pregnancy Proteins/pharmacology , RNA, Small Interfering/pharmacology , Rats , Recombinant Fusion Proteins/physiology , Solubility , Swine , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/physiology
18.
PLoS One ; 1: e25, 2006 Dec 20.
Article in English | MEDLINE | ID: mdl-17183652

ABSTRACT

BACKGROUND: Free nitric oxide (NO) reacts with sulphydryl residues to form S-nitrosothiols, which act as NO reservoirs. We sought to determine whether thiol-preserving agents and antioxidants, such as dithiothreitol (DTT) and vitamin C, induce NO release from S-nitrosylated proteins in endothelial cell cultures to promote angiogenesis. METHODOLOGY/PRINCIPAL FINDINGS: NO release was measured directly in cell supernatants using a Sievers NO Analyser, and in vitro angiogenesis was assessed by quantifying capillary-like tube network formation of porcine aortic endothelial cells (PAEC) on growth factor-reduced Matrigel. Incubation of PAEC with DTT or vitamin C significantly increased NO release in a concentration-dependent manner. However, the nitric oxide synthase (NOS) inhibitors, L-NNA and L-NIO, had no effect on DTT- or vitamin C-induced NO release, and there was no concomitant increase in the phosphorylation of endothelial NOS at serine-1177 following DTT or vitamin C treatment. DTT and vitamin C increased capillary-like tube network formation by nine- and two-fold, respectively, and the addition of copper ions doubled the effect of vitamin C. Surprisingly, DTT maintained endothelial tube networks for up to one month under serum-free conditions, and selective inhibitors of guanylyl cyclase (ODQ) and PKG (KT-5823) blocked this, demonstrating the requirement of cyclic GMP and PKG in this process. CONCLUSIONS/SIGNIFICANCE: Both DTT and vitamin C are capable of releasing sufficient NO from S-nitrosothiols to induce capillary morphogenesis. This study provides the first evidence that increased denitrosylation leads to increased bioavailability of NO, independent of NOS activity, to promote sustained angiogenesis.


Subject(s)
Neovascularization, Physiologic , Nitric Oxide/metabolism , S-Nitrosothiols/metabolism , Animals , Ascorbic Acid/pharmacology , Carbazoles/pharmacology , Cells, Cultured , Culture Media, Serum-Free , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/metabolism , Dithiothreitol/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Neovascularization, Physiologic/drug effects , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/chemistry , Nitric Oxide Synthase Type III/metabolism , Nitroarginine/pharmacology , Ornithine/analogs & derivatives , Ornithine/pharmacology , Oxadiazoles/pharmacology , Phosphorylation , Quinoxalines/pharmacology , Swine
19.
Circ Res ; 99(7): 715-22, 2006 Sep 29.
Article in English | MEDLINE | ID: mdl-16946136

ABSTRACT

Vascular endothelial growth factor-A (VEGF) is critical for angiogenesis but fails to induce neovascularization in ischemic tissue lesions in mice lacking endothelial nitric oxide synthase (eNOS). VEGF receptor-2 (VEGFR-2) is critical for angiogenesis, although little is known about the precise role of endothelial VEGFR-1 and its downstream effectors in this process. Here we have used a chimeric receptor approach in which the extracellular domain of the epidermal growth factor receptor was substituted for that of VEGFR-1 (EGLT) or VEGFR-2 (EGDR) and transduced into primary cultures of human umbilical vein endothelial cells (HUVECs) using a retroviral system. Activation of HUVECs expressing EGLT or EGDR induced rapid phosphorylation of eNOS at Ser1177, release of NO, and formation of capillary networks, similar to VEGF. Activation of eNOS by VEGFR-1 was dependent on Tyr794 and was mediated via phosphatidylinositol 3-kinase, whereas VEGFR-2 Tyr951 was involved in eNOS activation via phospholipase Cgamma1. Consistent with these findings, the VEGFR-1-specific ligand placenta growth factor-1 activated phosphatidylinositol 3-kinase and VEGF-E, which is selective for VEGFR-2-activated phospholipase Cgamma1. Both VEGFR-1 and VEGFR-2 signal pathways converged on Akt, as dominant-negative Akt inhibited the NO release and in vitro tube formation induced following activation of EGLT and EGDR. The identification Tyr794 of VEGFR-1 as a key residue in this process provides direct evidence of endothelial VEGFR-1 in NO-driven in vitro angiogenesis. These studies provide new sites of modulation in VEGF-mediated vascular morphogenesis and highlight new therapeutic targets for management of vascular diseases.


Subject(s)
Endothelium, Vascular/metabolism , Neovascularization, Physiologic/physiology , Nitric Oxide/physiology , Vascular Endothelial Growth Factor Receptor-1/physiology , Amino Acid Sequence , Cells, Cultured , Enzyme Activation/physiology , Humans , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tyrosine , Vascular Endothelial Growth Factor Receptor-1/genetics
20.
Biochem Biophys Res Commun ; 345(4): 1275-82, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16725109

ABSTRACT

Vascular endothelial growth factor-A (VEGF), which binds to both VEGF receptor-1 (Flt1) and VEGFR-2 (KDR/Flk-1), requires nitric oxide (NO) to induce angiogenesis in a cGMP-dependent manner. Here we show that VEGF-E, a VEGFR-2-selective ligand stimulates NO release and tube formation in human umbilical vein endothelial cells (HUVEC). Inhibition of phospholipase Cgamma (PLCgamma) with U73122 abrogated VEGF-E induced endothelial cell migration, tube formation and NO release. Inhibition of endothelial nitric oxide synthase (eNOS) using l-NNA blocked VEGF-E-induced NO release and angiogenesis. Pre-incubation of HUVEC with the soluble guanylate cyclase inhibitor, ODQ, or the protein kinase G (PKG) inhibitor, KT-5823, had no effect on angiogenesis suggesting that the action of VEGF-E is cGMP-independent. Our data provide the first demonstration that VEGFR-2-mediated NO signaling and subsequent angiogenesis is through a mechanism that is dependent on PLCgamma but independent of cGMP and PKG.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Endothelial Cells/drug effects , Nitric Oxide Synthase/metabolism , Signal Transduction/drug effects , Viral Proteins/pharmacology , Animals , Carbazoles/pharmacology , Cell Movement/drug effects , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Humans , Indoles/pharmacology , Neovascularization, Physiologic/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitroarginine/pharmacology , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/metabolism , Phosphorylation/drug effects , Pyrrolidinones/pharmacology , Swine , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...