Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
2.
Am J Physiol Heart Circ Physiol ; 281(3): H1005-14, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11514265

ABSTRACT

Prolonged infusions of 17beta-estradiol reduce fetal pulmonary vascular resistance (PVR), but the effects of endogenous estrogens in the fetal pulmonary circulation are unknown. To test the hypothesis that endogenous estrogen promotes pulmonary vasodilation at birth, we studied the hemodynamic effects of prolonged estrogen-receptor blockade during late gestation and at birth in fetal lambs. We treated chronically prepared fetal lambs with ICI-182,780 (ICI, a specific estrogen-receptor blocker, n = 5) or 1% DMSO (CTRL, n = 5) for 7 days and then measured pulmonary hemodynamic responses to ventilation with low- and high-fraction inspired oxygen (FI(O(2))). Treatment with ICI did not change basal fetal PVR or arterial blood gas tensions. However, treatment with ICI abolished the vasodilator response to ventilation with low FI(O(2)) [change in PVR -30 +/- 6% (CTRL) vs. +10 +/- 13%, (ICI), P < 0.05] without reducing the vasodilator response to ventilation with high FI(O(2)) [change in PVR, -73 +/- 3% (CTRL) vs. -77 +/- 4%, (ICI); P = not significant]. ICI treatment reduced prostacyclin synthase (PGIS) expression by 33% (P < 0.05) without altering expression of endothelial nitric oxide synthase or cyclooxygenase-1 and -2. In situ hybridization and immunohistochemistry revealed that PGIS is predominantly expressed in the airway epithelium of late gestation fetal lambs. We conclude that prolonged estrogen-receptor blockade inhibits the pulmonary vasodilator response at birth and that this effect may be mediated by downregulation of PGIS. We speculate that estrogen exposure during late gestation prepares the pulmonary circulation for postnatal adaptation.


Subject(s)
Estradiol/administration & dosage , Estrogen Antagonists/administration & dosage , Prenatal Exposure Delayed Effects , Pulmonary Circulation/drug effects , Receptors, Estrogen/antagonists & inhibitors , Animals , Animals, Newborn , Blotting, Western , Cyclooxygenase 1 , Cyclooxygenase 2 , Cytochrome P-450 Enzyme System/metabolism , Drug Administration Schedule , Estradiol/analogs & derivatives , Female , Fetus/blood supply , Fetus/drug effects , Fetus/physiology , Fulvestrant , Hemodynamics/drug effects , Infusions, Intra-Arterial/methods , Intramolecular Oxidoreductases/metabolism , Isoenzymes/metabolism , Lung/drug effects , Lung/embryology , Lung/enzymology , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Organ Specificity , Pregnancy , Prostaglandin-Endoperoxide Synthases/metabolism , Pulmonary Artery/embryology , Pulmonary Artery/physiology , Pulmonary Circulation/physiology , Pulmonary Ventilation/drug effects , Sheep , Vasodilation/drug effects , Vasodilation/physiology
3.
Nat Med ; 7(7): 853-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11433352

ABSTRACT

Atherosclerosis is the primary cause of cardiovascular disease, and the risk for atherosclerosis is inversely proportional to circulating levels of high-density lipoprotein (HDL) cholesterol. However, the mechanisms by which HDL is atheroprotective are complex and not well understood. Here we show that HDL stimulates endothelial nitric oxide synthase (eNOS) in cultured endothelial cells. In contrast, eNOS is not activated by purified forms of the major HDL apolipoproteins ApoA-I and ApoA-II or by low-density lipoprotein. Heterologous expression experiments in Chinese hamster ovary cells reveal that scavenger receptor-BI (SR-BI) mediates the effects of HDL on the enzyme. HDL activation of eNOS is demonstrable in isolated endothelial-cell caveolae where SR-BI and eNOS are colocalized, and the response in isolated plasma membranes is blocked by antibodies to ApoA-I and SR-BI, but not by antibody to ApoA-II. HDL also enhances endothelium- and nitric-oxide-dependent relaxation in aortae from wild-type mice, but not in aortae from homozygous null SR-BI knockout mice. Thus, HDL activates eNOS via SR-BI through a process that requires ApoA-I binding. The resulting increase in nitric-oxide production might be critical to the atheroprotective properties of HDL and ApoA-I.


Subject(s)
CD36 Antigens/metabolism , Lipoproteins, HDL/metabolism , Membrane Proteins , Nitric Oxide Synthase/metabolism , Receptors, Immunologic , Receptors, Lipoprotein , Animals , Base Sequence , CD36 Antigens/genetics , CD36 Antigens/physiology , CHO Cells , Cell Line, Transformed , Cricetinae , DNA Primers , Enzyme Activation , Nitric Oxide Synthase Type III , Protein Binding , Receptors, Scavenger , Reverse Transcriptase Polymerase Chain Reaction , Scavenger Receptors, Class B , Sheep
5.
J Biol Chem ; 276(29): 27071-6, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11369763

ABSTRACT

Estrogen causes rapid endothelial nitric oxide (NO) production because of the activation of plasma membrane-associated estrogen receptors (ER) coupled to endothelial NO synthase (eNOS). In the present study, we determined the role of G proteins in eNOS activation by estrogen. Estradiol-17beta (E(2), 10(-8) m) and acetylcholine (10(-5) m) caused comparable increases in NOS activity (15 min) in intact endothelial cells that were fully blocked by pertussis toxin (Ptox). In addition, exogenous guanosine 5'-O-(2- thiodiphosphate) inhibited E(2)-mediated eNOS stimulation in isolated endothelial plasma membranes, and Ptox prevented enzyme activation by E(2) in COS-7 cells expressing ERalpha and eNOS. Coimmunoprecipitation studies of plasma membranes from COS-7 cells transfected with ERalpha and specific Galpha proteins demonstrated E(2)-stimulated interaction between ERalpha and Galpha(i) but not between ERalpha and either Galpha(q) or Galpha(s); the observed ERalpha-Galpha(i) interaction was blocked by the ER antagonist ICI 182,780 and by Ptox. E(2)-stimulated ERalpha-Galpha(i) interaction was also demonstrable in endothelial cell plasma membranes. Cotransfection of Galpha(i) into COS-7 cells expressing ERalpha and eNOS yielded a 3-fold increase in E(2)-mediated eNOS stimulation, whereas cotransfection with a protein regulator of G protein signaling, RGS4, inhibited the E(2) response. These findings indicate that eNOS stimulation by E(2) requires plasma membrane ERalpha coupling to Galpha(i) and that activated Galpha(i) mediates the requisite downstream signaling events. Thus, novel G protein coupling enables a subpopulation of ERalpha to initiate signal transduction at the cell surface. Similar mechanisms may underly the nongenomic actions of other steroid hormones.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Membrane Proteins/metabolism , Nitric Oxide Synthase/metabolism , Receptors, Estrogen/metabolism , Animals , Cell Line, Transformed , Cell Membrane/metabolism , Enzyme Activation , Nitric Oxide Synthase Type III , Precipitin Tests , Signal Transduction
6.
Circ Res ; 87(11): E44-52, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-11090554

ABSTRACT

Estrogen causes nitric oxide (NO)-dependent vasodilation due to estrogen receptor (ER) alpha-mediated, nongenomic activation of endothelial NO synthase (eNOS). The subcellular site of interaction between ERalpha and eNOS was determined in studies of isolated endothelial cell plasma membranes. Estradiol (E(2), 10(-8) mol/L) caused an increase in eNOS activity in plasma membranes in the absence of added calcium, calmodulin, or eNOS cofactors, which was blocked by ICI 182,780 and ERalpha antibody. Immunoidentification studies detected the same 67-kDa protein in endothelial cell nucleus, cytosol, and plasma membrane. Plasma membranes from COS-7 cells expressing eNOS and ERalpha displayed ER-mediated eNOS stimulation, whereas membranes from cells expressing eNOS alone or ERalpha plus a myristoylation-deficient mutant eNOS were insensitive. Fractionation of endothelial cell plasma membranes revealed ERalpha protein in caveolae, and E(2) caused stimulation of eNOS in isolated caveolae that was ER-dependent; noncaveolae membranes were insensitive. Acetylcholine and bradykinin also activated eNOS in isolated caveolae. Furthermore, the effect of E(2) on eNOS in caveolae was prevented by calcium chelation. Thus, a subpopulation of ERalpha is localized to endothelial cell caveolae where they are coupled to eNOS in a functional signaling module that may regulate the local calcium environment. The full text of this article is available at http://www.circresaha.org.


Subject(s)
Caveolae/metabolism , Nitric Oxide Synthase/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Acetylcholine/pharmacology , Animals , COS Cells , Calcium/metabolism , Calmodulin/metabolism , Caveolin 1 , Caveolins/metabolism , Cell Membrane/enzymology , Cells, Cultured , Chelating Agents , Cholinergic Agents/pharmacology , Enzyme Activation/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha , Immunoblotting , Nitric Oxide Synthase Type III , Sheep , Signal Transduction/drug effects
7.
Am J Physiol Heart Circ Physiol ; 278(6): H2134-42, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10843913

ABSTRACT

Nitric oxide contributes to estrogen-mediated uterine vasodilation; however, the nitric oxide synthases (NOS) involved and their location within uterine arteries are incompletely documented. We investigated the effects of repetitive daily and acute estradiol-17beta (E(2)beta) exposure on uterine hemodynamics and NOS abundance and localization in uterine arteries from nonpregnant ovariectomized ewes receiving daily intravenous E(2)beta (1 microg/kg, n = 5) or no E(2)beta (n = 7) for 5 days to determine NOS abundance, cGMP contents, and NOS immunohistochemistry. Daily E(2)beta increased basal and E(2)beta-mediated rises in uterine blood flow (UBF) 36 and 43% (<0.01), respectively, calcium-dependent NOS activity 150% (P < 0.02) in endothelium-intact and -denuded ( approximately 40% of total NOS) arteries, and cGMP contents 39% (P < 0.05). Endothelial (eNOS) was detected in luminal endothelium, whereas neuronal NOS (nNOS) protein was only in the media. A second group of ewes received E(2)beta (1 microg/kg iv) for 4 days and acute intravenous E(2)beta (n = 8) or vehicle (n = 4) on day 5. UBF rose 5.5-fold (P < 0.001) 115 min after E(2)beta, at which time only endothelium-derived calcium-dependent NOS activity increased 30 +/- 13% (P < 0.05). Daily E(2)beta enhances basal and E(2)beta-mediated increases in UBF, which parallel increases in endothelium-derived eNOS and smooth muscle-derived nNOS. Acute E(2)beta, however, selectively increases endothelium-derived eNOS.


Subject(s)
Estradiol/pharmacology , Nitric Oxide Synthase/metabolism , Uterus/blood supply , Animals , Arteries/enzymology , Arteries/metabolism , Cyclic GMP/metabolism , Drug Administration Schedule , Estradiol/administration & dosage , Female , Injections, Intravenous , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type III , Regional Blood Flow/drug effects , Sheep , Time Factors , Up-Regulation
8.
J Biol Chem ; 275(24): 17979-85, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10849433

ABSTRACT

Endothelial nitric-oxide synthase (eNOS), a Ca(2+)/calmodulin-dependent enzyme, is critical for vascular homeostasis. While eNOS is membrane-associated through its N-myristoylation, the significance of membrane association in locating eNOS near sources of Ca(2+) entry is uncertain. To assess the Ca(2+) source required for eNOS activation, chimera containing the full-length eNOS cDNA and HA-tagged aequorin sequence (EHA), and MHA (myristoylation-deficient EHA) were generated and transfected into COS-7 cells. The EHA chimera was primarily targeted to the plasma membrane while MHA was located intracellularly. Both constructs retained enzymatic eNOS activity and aequorin-mediated Ca(2+) sensitivity. The plasma membrane-associated EHA and intracellular MHA were compared in their ability to sense changes in local Ca(2+) concentration, demonstrating preferential sensitivity to Ca(2+) originating from intracellular pools (MHA) or from capacitative Ca(2+) entry (EHA). Measurements of eNOS activation in intact cells revealed that the eNOS enzymatic activity of EHA was more sensitive to Ca(2+) influx via capacitative Ca(2+) entry than intracellular release, whereas MHA eNOS activity was more responsive to intracellular Ca(2+) release. When eNOS activation by CCE was compared with that generated by an equal rise in [Ca(2+)](i) due to the Ca(2+) ionophore ionomycin, a 10-fold greater increase in NO production was found in the former condition. These results demonstrate that EHA and MHA chimera are properly targeted and retain full functions of eNOS and aequorin, and that capacitative Ca(2+) influx is the principle stimulus for sustained activation of eNOS on the plasma membrane in intact cells.


Subject(s)
Calcium/metabolism , Nitric Oxide Synthase/metabolism , Aequorin , Animals , COS Cells , Endothelium, Vascular/enzymology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Histamine/pharmacology , Ionomycin , Luminescent Measurements , Myristic Acid/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III , Recombinant Proteins/metabolism , Thapsigargin/pharmacology , Transfection
9.
Semin Perinatol ; 24(1): 70-4, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10709864

ABSTRACT

Estrogen has a variety of effects on the vascular wall including rapid vasodilation due to the stimulation of endothelial nitric oxide synthase (eNOS). Studies in cultured endothelium indicate that the hormone cause acute, direct activation of eNOS that is unaffected by actinomycin D but fully inhibited by estrogen receptor (ER) antagonism. Overexpression of ERalpha leads to marked enhancement of the acute response to estrogen, and this process is blocked by ER antagonism and requires the ERalpha hormone binding domain. The acute response of eNOS to estrogen can also be reconstituted in COS-7 cells cotransfected with ERalpha and eNOS, but not by transfection with eNOS alone. The inhibition of calcium influx, or tyrosine kinases or mitogen-activated protein (MAP) kinase prevents eNOS stimulation by estrogen, and estrogen causes rapid ER-dependent activation of MAP kinase. Thus, the acute effect of estrogen on eNOS is mediated by ERalpha functioning in a novel, nongenomic manner to activate the enzyme via calcium-dependent, MAP kinase-dependent mechanisms.


Subject(s)
Endothelium, Vascular/physiology , Receptors, Estrogen/physiology , Animals , Humans
10.
J Biol Chem ; 275(11): 8183-9, 2000 Mar 17.
Article in English | MEDLINE | ID: mdl-10713142

ABSTRACT

Nitric oxide (NO) plays an important role in airway function, and endothelial NO synthase (eNOS) is expressed in airway epithelium. To determine the basis of cell-specific eNOS expression in airway epithelium, studies were performed in NCI-H441 human bronchiolar epithelial cells transfected with the human eNOS promoter fused to luciferase. Transfection with 1624 base pairs of sequence 5' to the initiation ATG (position -1624) yielded a 19-fold increase in promoter activity versus vector alone. No activity was found in lung fibroblasts, which do not express eNOS. 5' deletions from -1624 to -279 had modest effects on promoter activity in H441 cells. Further deletion to -248 reduced activity by 65%, and activity was lost with deletion to -79. Point mutations revealed that the GATA binding motif at -254 is mandatory for promoter activity and that the positive regulatory element between -248 and -79 is the Sp1 binding motif at -125. Electrophoretic mobility shift assays yielded two complexes with the GATA site and three with the Sp1 site. Immunodepletion with antiserum to GATA-2 prevented formation of the slowest migrating GATA complex, and antiserum to Sp1 supershifted the slowest migrating Sp1 complex. An electrophoretic mobility shift assay with H441 versus fibroblast nuclei revealed that the slowest migrating GATA complex is unique to airway epithelium. Thus, cell-specific eNOS expression in airway epithelium is dependent on the interaction of GATA-2 with the core eNOS promoter, and the proximal Sp1 binding site is also an important positive regulatory element.


Subject(s)
Nitric Oxide Synthase/genetics , Respiratory Mucosa/enzymology , Binding Sites , Bronchi/enzymology , DNA-Binding Proteins/metabolism , Endothelium, Vascular/enzymology , Erythroid-Specific DNA-Binding Factors , Fibroblasts/enzymology , GATA2 Transcription Factor , Gene Expression Regulation, Enzymologic , Genes, Reporter , Humans , Luciferases/biosynthesis , Luciferases/genetics , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type III , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , Tissue Distribution , Transcription Factors/metabolism
11.
Proc Natl Acad Sci U S A ; 97(6): 2836-9, 2000 Mar 14.
Article in English | MEDLINE | ID: mdl-10706629

ABSTRACT

Estrogen receptors (ERs) are ligand-activated transcription factors that regulate gene expression and cell growth. Two ERs now have been identified: ERalpha and the more recently discovered ERbeta. The physiological function of ERbeta remains unclear, but evidence from vascular injury studies and from ERbeta knockout mice suggests that ERbeta may be involved in the regulation of cellular proliferation. Here we show a direct and specific interaction between ERbeta and the cell cycle mitotic spindle assembly checkpoint protein, MAD2 (mitosis arrest-deficient 2). The ERbeta-MAD2 interaction was identified by screening of a yeast two-hybrid system vascular endothelial cell library with ERbeta and confirmed with glutathione S-transferase-fusion protein interaction studies. In contrast, ERalpha did not interact with MAD2 in either the two-hybrid system or in the protein-protein interaction experiments. Amino acids 173-208 in the hinge region of ERbeta were sufficient to mediate the interaction with MAD2 in the two-hybrid system and in glutathione S-transferase-fusion protein studies. These data identify a link between ERbeta and MAD2 of potential importance to regulation of the cell cycle and support a function of ERbeta distinct from the established role of ERs as transcription factors.


Subject(s)
Calcium-Binding Proteins/metabolism , Carrier Proteins , Fungal Proteins/metabolism , Receptors, Estrogen/metabolism , Amino Acid Sequence , Animals , Cell Cycle , Cell Cycle Proteins , Endothelium, Vascular/metabolism , Estrogen Receptor beta , Glutathione Transferase/metabolism , Humans , Molecular Sequence Data , Nuclear Proteins , Protein Binding , Pulmonary Artery/metabolism , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Sheep , Two-Hybrid System Techniques
12.
Am J Physiol Lung Cell Mol Physiol ; 278(1): L131-8, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10645900

ABSTRACT

Nitric oxide (NO), produced by endothelial (e) nitric oxide synthase (NOS), is a critical mediator of vascular function and growth in the developing lung. Pulmonary eNOS expression is diminished in conditions associated with altered pulmonary vascular development, suggesting that eNOS may be modulated by changes in pulmonary artery endothelial cell (PAEC) growth. We determined the effects of cell growth on eNOS expression in cultured ovine fetal PAEC studied at varying levels of confluence. NOS enzymatic activity was sixfold greater in quiescent PAEC at 100% confluence compared with more rapidly replicating cells at 50% confluence. To determine if there is a reciprocal effect of NO on PAEC growth, studies of NOS inhibition or the provision of exogenous NO from spermine NONOate were performed. Neither intervention had a discernable effect on PAEC growth. The influence of cell growth on NOS activity was unique to pulmonary endothelium, because varying confluence did not alter NOS activity in fetal systemic endothelial cells. The effects of cell growth induced by serum stimulation were also evaluated, and NOS enzymatic activity was threefold greater in quiescent, serum-deprived cells compared with that in serum-stimulated cells. The increase in NOS activity observed at full confluence was accompanied by parallel increases in eNOS protein and mRNA expression. These findings indicate that eNOS gene expression in fetal PAEC is upregulated during cell quiescence and downregulated during rapid cell growth. Furthermore, the interaction between cell growth and NO in the PAEC is unidirectional.


Subject(s)
Endothelium, Vascular/embryology , Fetus/metabolism , Nitric Oxide Synthase/metabolism , Pulmonary Artery/embryology , Animals , Cell Division/drug effects , Cell Division/physiology , Endothelium, Vascular/cytology , Immunoblotting , Nitric Oxide/pharmacology , Nitric Oxide/physiology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type III , Pulmonary Artery/cytology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sheep
13.
J Biol Chem ; 275(15): 11278-83, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10753938

ABSTRACT

Oxidized LDL (oxLDL) depletes caveolae of cholesterol, resulting in the displacement of endothelial nitric-oxide synthase (eNOS) from caveolae and impaired eNOS activation. In the present study, we determined if the class B scavenger receptors, CD36 and SR-BI, are involved in regulating nitric-oxide synthase localization and function. We demonstrate that CD36 and SR-BI are expressed in endothelial cells, co-fractionate with caveolae, and co-immunoprecipitate with caveolin-1. Co-incubation of cells with 10 microgram/ml high density lipoprotein (HDL) prevented oxLDL-induced translocation of eNOS from caveolae and restored acetylcholine-induced nitric-oxide synthase stimulation. Acetylcholine caused eNOS activation in cells incubated with 10 microgram/ml oxLDL (10-15 thiobarbituric acid-reactive substances) and blocking antibodies to CD36, whereas cells treated with only oxLDL were unresponsive. Furthermore, CD36-blocking antibodies prevented oxLDL-induced redistribution of eNOS. SR-BI-blocking antibodies were used to demonstrate that the effects of HDL are mediate by SR-BI. HDL binding to SR-BI maintained the concentration of caveola-associated cholesterol by promoting the uptake of cholesterol esters, thereby preventing oxLDL-induced depletion of caveola cholesterol. We conclude that CD36 mediates the effects of oxLDL on caveola composition and eNOS activation. Furthermore, HDL prevents oxLDL from decreasing the capacity for eNOS activation by preserving the cholesterol concentration in caveolae and, thereby maintaining the subcellular location of eNOS.


Subject(s)
Endothelium, Vascular/enzymology , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/toxicity , Membrane Proteins , Nitric Oxide Synthase/drug effects , Receptors, Immunologic , Receptors, Lipoprotein , CD36 Antigens/analysis , CD36 Antigens/physiology , Cells, Cultured , Cholesterol/metabolism , Enzyme Activation/drug effects , Humans , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Receptors, Scavenger , Scavenger Receptors, Class B
14.
J Biol Chem ; 274(45): 32512-9, 1999 Nov 05.
Article in English | MEDLINE | ID: mdl-10542298

ABSTRACT

Hypercholesterolemia-induced vascular disease and atherosclerosis are characterized by a decrease in the bioavailability of endothelium-derived nitric oxide. Endothelial nitric-oxide synthase (eNOS) associates with caveolae and is directly regulated by the caveola protein, caveolin. In the present study, we examined the effects of oxidized low density lipoprotein (oxLDL) on the subcellular location of eNOS, on eNOS activation, and on caveola cholesterol in endothelial cells. We found that treatment with 10 microgram/ml oxLDL for 60 min caused greater than 90% of eNOS and caveolin to leave caveolae. Treatment with oxLDL also inhibited acetylcholine-induced activation of eNOS but not prostacyclin production. oxLDL did not affect total cellular eNOS abundance. Oxidized LDL also did not affect the palmitoylation, myristoylation or phosphorylation of eNOS. Oxidized LDL, but not native LDL, or HDL depleted caveolae of cholesterol by serving as an acceptor for cholesterol. Cyclodextrin also depleted caveolae of cholesterol and caused eNOS and caveolin to translocate from caveolae. Furthermore, removal of oxLDL allowed eNOS and caveolin to return to caveolae. We conclude that oxLDL-induced depletion of caveola cholesterol causes eNOS to leave caveolae and inhibits acetylcholine-induced activation of the enzyme. This process may be an important mechanism in the early pathogenesis of atherosclerosis.


Subject(s)
Cell Membrane/enzymology , Endothelium, Vascular/enzymology , Lipoproteins, LDL/metabolism , Nitric Oxide Synthase/metabolism , Animals , Cell Membrane/drug effects , Cells, Cultured , Cholesterol/metabolism , Cyclodextrins/pharmacology , Endothelium, Vascular/drug effects , Enzyme Activation , Fluorescent Antibody Technique, Indirect , Lipoproteins, HDL/metabolism , Myristic Acid/metabolism , Nitric Oxide Synthase Type III , Palmitic Acid/metabolism , Phosphorylation , Swine
15.
Am J Respir Cell Mol Biol ; 21(5): 629-36, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10536122

ABSTRACT

Endothelium-derived nitric oxide (NO) generated by endothelial NO synthase (eNOS) is critically involved in pulmonary vasodilation during cardiopulmonary transition at birth. Inhaled NO therapy has recently been considered for patients with persistent pulmonary hypertension of the newborn (PPHN). To better understand the mechanisms regulating NO synthesis in the developing pulmonary circulation and the possible ramifications of NO therapy, studies were performed with early passage ovine fetal intrapulmonary artery endothelial cells (PAEC) to determine whether NO directly modulates eNOS expression. To examine the effects of exogenous NO, PAEC were treated with the NO donor spermine NONOate or the parent compound spermine. Exogenous NO caused increases in eNOS protein expression and NOS enzymatic activity that were detectable within 16 h of exposure. In contrast, the inhibition of endogenous NO production with nitro-L-arginine-methyl ester (L-NAME) caused a reduction in eNOS protein expression that was evident within 8 h. Paralleling the changes in eNOS protein, eNOS messenger RNA (mRNA) abundance was upregulated by exogenous NO and downregulated by L-NAME, suggesting that NO modulation of eNOS expression involves processes at the level of gene transcription or mRNA stability. Thus, in fetal PAEC there is positive-feedback regulation of eNOS expression by both exogenous and endogenous NO. These findings suggest that difficulties with transient effectiveness or prolonged requirements for NO therapy in certain PPHN patients are not due to declines in eNOS expression. Further, conditions such as fetal hypoxemia that impair PAEC NO production may attenuate eNOS expression through this mechanism, thereby contributing to the pathogenesis of PPHN.


Subject(s)
Endothelium, Vascular/drug effects , Nitric Oxide Synthase/biosynthesis , Nitric Oxide/pharmacology , Pulmonary Artery/enzymology , Animals , Blotting, Southern , Cells, Cultured , Endothelium, Vascular/enzymology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Immunoblotting , Mutagens/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type III , Nitrogen Oxides , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Sheep , Spermine/analogs & derivatives , Spermine/pharmacology , Time Factors , Up-Regulation/drug effects
16.
Am J Physiol ; 277(1): L106-12, 1999 07.
Article in English | MEDLINE | ID: mdl-10409236

ABSTRACT

The investigation of fetal pulmonary endothelial cell gene expression and function has been limited by the requirement for primary cells. In an effort to establish an immortalized cell line, ovine fetal pulmonary artery endothelial cells (PAECs; passage 5) were permanently transfected with the E6 and E7 open reading frames of human papillomavirus type 16, and phenotypes related to nitric oxide (NO) production were evaluated up to passage 28. Acetylated low-density lipoprotein uptake, endothelial NO synthase (eNOS) expression, and proliferation rates were unaltered by immortalization. Acetylcholine-stimulated eNOS activity was 218-255% above basal levels in immortalized cells, and this was comparable to the 250% increase seen in primary PAECs (passage 6). eNOS was also acutely activated by estradiol to levels 197-309% above basal, paralleling the stimulation obtained in primary cells. In addition, the expression of estrogen receptor-alpha, which has recently been shown to mediate the acute response in primary PAECs, was conserved. Thus fetal PAECs transfected with E6 and E7 show no signs of senescence with passage, and mechanisms of NO production, including those mediated by estradiol, are conserved. Immortalized PAECs will provide an excellent model for further studies of eNOS gene expression and function in fetal pulmonary endothelium.


Subject(s)
Cell Line, Transformed , Endothelium, Vascular/embryology , Pulmonary Artery/embryology , Animals , Cell Division/physiology , Cell Transformation, Viral/physiology , Enzyme Activation/physiology , Estrogen Receptor alpha , Fetus/cytology , Fetus/enzymology , Nitric Oxide Synthase/metabolism , Open Reading Frames/physiology , Papillomaviridae/genetics , Receptors, Estrogen/metabolism , Sheep/embryology , Transfection
17.
Early Hum Dev ; 54(3): 271-94, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10321793

ABSTRACT

The endothelium-derived vasodilator molecules prostaglandin I2 (PGI2) and nitric oxide (NO) are critically involved in the dramatic increase in pulmonary blood flow that occurs during cardiopulmonary transition at birth. Studies in animal and cell culture models have revealed that there is increased PGI2 and NO production in the pulmonary circulation of the late fetus in direct response to increased oxygenation, and that this response is unique to the pulmonary endothelium. Additional work has demonstrated that there is normally marked upregulation in the expression of the key synthetic enzymes cyclooxygenase type I and endothelial NO synthase in the lung during late gestation, thereby maximizing the capacity for vasodilator production at the time of birth. Furthermore, studies in animal models of neonatal pulmonary hypertension indicate that attenuated expression of these genes may frequently contribute to the pathogenesis of the disorder. A greater understanding of the mechanisms regulating PGI2 and NO synthesis in the developing lung will potentially lead to novel therapies for neonatal pulmonary hypertension aimed at optimizing endogenous vasodilator production.


Subject(s)
Endothelium, Vascular/metabolism , Epoprostenol/biosynthesis , Lung/blood supply , Lung/embryology , Nitric Oxide/biosynthesis , Vasodilation , Gene Expression Regulation , Gestational Age , Humans , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism
18.
Steroids ; 64(1-2): 28-34, 1999.
Article in English | MEDLINE | ID: mdl-10323670

ABSTRACT

Estrogen is an important atheroprotective molecule that causes the rapid dilation of blood vessels by stimulating endothelial nitric oxide synthase (eNOS). There is also evidence that estrogen modulates airway epithelial NO production, thereby potentially affecting bronchial hyperresponsiveness. Studies in cultured endothelial and airway epithelial cells indicate that physiologic concentrations of estrogen cause rapid direct activation of eNOS that is unaffected by actinomycin D, but fully inhibited by estrogen receptor (ER) antagonism. Overexpression of ERalpha leads to marked enhancement of the acute response to estrogen, and this process is blocked by ER antagonism, it is specific to estrogen, and it requires the ERalpha hormone binding domain. In addition, the acute response of eNOS to estrogen can be reconstituted in COS-7 cells cotransfected with wild-type ERalpha and eNOS, but not by transfection with eNOS alone. Furthermore, the inhibition of calcium influx, or tyrosine kinases or MAP kinase prevents the stimulation of eNOS by estrogen, and estrogen causes rapid ER-dependent activation of MAP kinase. These findings indicate that the acute effects of estrogen on both endothelial and airway epithelial eNOS are mediated by ERalpha functioning in a novel, nongenomic manner to activate the enzyme via calcium-dependent, MAP kinase-dependent mechanisms.


Subject(s)
Estrogens/physiology , Nitric Oxide Synthase/metabolism , Animals , COS Cells , Cell Line , Enzyme Activation , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha , Kinetics , Nitric Oxide Synthase Type III , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/physiology , Signal Transduction , Swine
19.
Am J Respir Cell Mol Biol ; 20(4): 658-66, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10100997

ABSTRACT

Nitric oxide (NO) is an important mediator of physiologic processes in the airway. Levels of exhaled NO are greatest and asthma symptoms are least in menstruating women during midcycle, when estrogen levels are highest. To better understand the role of estrogen in airway function, we tested the hypothesis that estrogen stimulates endothelial NO synthase (eNOS) in NCI-H441 human bronchiolar epithelial cells. eNOS activation was assessed by measuring conversion of [3H]L-arginine to [3H]L-citrulline in intact cells. eNOS activity rose in the presence of estradiol-17beta (E2beta), with a maximum stimulation of 243% at 10(-8) M E2beta. This response was comparable to the 201% increase elicited by the calcium (Ca2+) ionophore A23187 (10(-5) M), and was evident as early as 5 min after such treatment. Actinomycin D had no effect on the response to E2beta, and eNOS abundance was similar in control and E2beta-treated cells. E2beta-stimulated eNOS activity was dependent on the influx of extracellular Ca2+, and was completely inhibited by the estrogen receptor (ER) antagonist ICI182,780. Messenger RNA and protein for the alpha isoform of ER (ERalpha) were evident in the H441 cells, and freshly isolated ovine airway epithelial cells also coexpressed eNOS and ERalpha. These findings indicate that estrogen acutely activates existing eNOS in H441 airway epithelial cells, through a process that involves the stimulation of epithelial ER and Ca2+ influx. This process may play a role in the hormonal modulation of airway function.


Subject(s)
Bronchi/metabolism , Epithelial Cells/metabolism , Estradiol/pharmacology , Nitric Oxide Synthase/metabolism , Receptors, Estrogen/genetics , Animals , Arginine/metabolism , Bronchi/cytology , Calcimycin/pharmacology , Cell Line , Dactinomycin/pharmacology , Enzyme Activation , Epithelial Cells/cytology , Estradiol/analogs & derivatives , Estrogen Receptor alpha , Female , Fulvestrant , Humans , Kinetics , Menstruation , Nitric Oxide Synthase Type III , Reverse Transcriptase Polymerase Chain Reaction , Sheep , Uterus/metabolism
20.
Circ Res ; 84(2): 193-200, 1999 Feb 05.
Article in English | MEDLINE | ID: mdl-9933251

ABSTRACT

Prostacyclin (prostaglandin I2 [PGI2]) is a key mediator of pulmonary vascular function during early postnatal life, and its production in the pulmonary vasculature rises markedly during that period because of increasing expression of cyclooxygenase type 1 (COX-1). The postnatal rise in COX-1 may be due to the release of inhibition by glucocorticoids, since plasma glucocorticoid levels fall after birth and glucocorticoids decrease PGI2 synthesis in certain nonpulmonary cell types. We therefore studied the direct effects of dexamethasone (DEX) on COX-1 expression in early-passage ovine fetal pulmonary-artery endothelial cells (PAECs). DEX (10(-10) to 10(-6) mol/L) caused a dose-related decrease in COX-1 mRNA expression that was evident by 24 hours, was maximal at 10(-6) mol/L (50% inhibition), and was not due to changes in mRNA stability. There was a parallel decline in COX-1 protein expression. COX-1 protein rose following DEX withdrawal, and DEX blunted the stimulatory effect of 17beta-estradiol on COX-1 expression. DEX alone (10(-8) mol/L for 48 hours) caused a 93% fall in basal PGI2 production, and bradykinin- and A23187-stimulated PGI2 were diminished 96% and 94%, respectively. Similarly, PGI2 synthesis from arachidonic acid fell 86% with DEX; all of the above effects are consistent with COX-1 downregulation. The glucocorticoid receptor (GR) antagonist mifepristone (RU-486; 10(-6) mol/L) blocked the inhibitory effect of DEX, and GR expression was evident by immunoblot analysis. These findings indicate that glucocorticoids downregulate COX-1 expression and PGI2 synthesis in fetal PAECs through the activation of PAEC GR and effects on COX-1 gene transcription. This mechanism may modulate pulmonary PGI2 production in the perinatal period, and it may also play a role in the effects of glucocorticoids on the systemic circulation at a variety of ages.


Subject(s)
Dexamethasone/pharmacology , Epoprostenol/biosynthesis , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Glucocorticoids/pharmacology , Isoenzymes/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Animals , Cyclooxygenase 1 , Down-Regulation , Embryonic and Fetal Development/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/embryology , Endothelium, Vascular/metabolism , Glucocorticoids/antagonists & inhibitors , Hormone Antagonists/pharmacology , Mifepristone/antagonists & inhibitors , Mifepristone/pharmacology , Pulmonary Artery/drug effects , Pulmonary Artery/embryology , Pulmonary Artery/metabolism , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...