84 results on '"Venema RC"'
Search Results
2. Interaction between the 90-kDa heat shock protein and soluble guanylyl cyclase: Physiological significance and mapping of the domains mediating binding
- Author
-
Papapetropoulos, A Zhou, ZM Gerassimou, C Yetik, G and Venema, RC Roussos, C Sessa, WC Catravas, JD
- Subjects
inorganic chemicals ,polycyclic compounds ,cardiovascular system ,heterocyclic compounds - Abstract
The 90-kDa heat shock protein (hsp90) regulates the stability and function of many client proteins, including members of the NO-cGMP signaling pathway. Soluble guanylyl cyclase (sGC), an NO receptor, was recently reported to be an hsp90-interacting partner. In the present study, we show that hsp90 binds to both subunits of the most common sGC form (alpha(1)beta(1)) when these are expressed individually but only interacts with beta(1) in the heterodimeric form of the enzyme. Characterization of the region of hsp90 required to bind each subunit in immunoprecipitation experiments revealed that residues 310 to 456 of hsp90 interact with the sGC subunits. The region of beta(1) responsible for binding to hsp90 beta was mapped using in vitro binding assays and immunoprecipitation experiments and was found to lie in the regulatory domain. The physiological importance of the hsp90/sGC interaction was investigated by treating rat smooth muscle cells with the hsp90 inhibitors radicicol and geldanamycin (GA) and determining both sGC activity and protein levels. Long-term ( 24 or 48 h) inhibition of hsp90 resulted in a strong decrease of both alpha(1) and beta(1) protein levels and sGC activity. Moreover, incubation of smooth muscle cells with the proteasome inhibitor N-benzoyloxycarbonyl (Z)-Leu-Leu-leucinal (MG132) blocked the GA-induced down-regulation of sGC. We conclude that the N-terminal region of the beta(1) subunit mediates binding of the heterodimeric form of sGC to hsp90 and that this interaction involves the M domain of hsp90. Hsp90 binding to sGC regulates the pool of active enzymes by affecting the protein levels of the two subunits.
- Published
- 2005
3. TNFα reduces eNOS activity in endothelial cells through serine 116 phosphorylation and Pin1 binding: Confirmation of a direct, inhibitory interaction of Pin1 with eNOS.
- Author
-
Kennard S, Ruan L, Buffett RJ, Fulton D, and Venema RC
- Subjects
- Animals, Binding Sites, Cattle, Cells, Cultured, Endothelial Cells enzymology, Extracellular Signal-Regulated MAP Kinases metabolism, Humans, Mice, Nitric Oxide Synthase Type III metabolism, Phosphorylation, Protein Binding, Protein Interaction Domains and Motifs, Rats, Serine, Species Specificity, Endothelial Cells drug effects, Enzyme Inhibitors pharmacology, NIMA-Interacting Peptidylprolyl Isomerase metabolism, Nitric Oxide metabolism, Nitric Oxide Synthase Type III antagonists & inhibitors, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Production of NO by the endothelial nitric oxide synthase (eNOS) has a major role in blood pressure control and suppression of atherosclerosis. In a previous study, we presented evidence implicating the Pin1 prolyl isomerase in negative modulation of eNOS activity in bovine aortic endothelial cells (BAECs). Pin1 recognizes phosphoserine/phosphothreonine-proline motifs in target proteins and catalyzes prolyl isomerization at the peptide bond. In the present study, we show, first, with purified proteins, that Pin1 binds to eNOS directly via the Pin1 WW domain. Binding is enhanced by mimicking phosphorylation of eNOS at S116. Interaction of Pin1 with eNOS markedly reduces eNOS enzymatic activity. Second, in BAECs, we show that TNFα induces ERK 1/2-mediated S116 phosphorylation of eNOS, accompanied by Pin1 binding. TNFα treatment of BAECs results in a reduction in NO release from the cells in a manner that depends on the activities of both Pin1 and ERK 1/2. Evidence is also presented that this mechanism of eNOS regulation cannot occur in rat and mouse cells because there is no proline residue in the mouse and rat amino acid sequences adjacent to the putative phosphorylation site. Moreover, we find that phosphorylation of this site is not detectable in mouse eNOS., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
4. Phytonutrients Differentially Stimulate NAD(P)H:Quinone Oxidoreductase, Inhibit Proliferation, and Trigger Mitotic Catastrophe in Hepa1c1c7 Cells.
- Author
-
Jackson SJ, Singletary KW, Murphy LL, Venema RC, and Young AJ
- Subjects
- Animals, Cell Line, Hepatocytes cytology, Hepatocytes enzymology, Mice, NAD(P)H Dehydrogenase (Quinone) genetics, Apoptosis drug effects, Hepatocytes drug effects, Mitosis drug effects, NAD(P)H Dehydrogenase (Quinone) metabolism, Phytochemicals pharmacology
- Abstract
Phytonutrients have rapidly emerged as natural food chemicals possessing multifaceted biological actions that may support beneficial health outcomes. Among the vast array of phytonutrients currently being studied, sulforaphane, curcumin, quercetin, and resveratrol have been frequently reported to stimulate the expression of endogenous detoxification enzymes and may thereby facilitate the neutralization of otherwise harmful environmental agents. Some of these same phytonutrients, however, have also been implicated in disrupting normal cell proliferation and hence may possess toxic properties in and of themselves. In this study, we characterize the respective minimum threshold concentrations of the aforementioned phytonutrients in Hepa1c1c7 cells that stimulate, Nad(p)h: quinone oxidoreductase (NQO1), a key enzyme in the hepatic neutralization of menadione, other biological oxidants, and some environmental carcinogens. Moreover, our findings demonstrate that relatively low concentrations of either sulforaphane or curcumin significantly (P < .05) increase NQO1 protein expression and activity without triggering G2/M cell cycle arrest or mitotic catastrophe. The minimal quercetin concentration inducing NQO1, however, was 100-fold higher than that which disrupted mitosis. Also, while resveratrol modestly stimulated NQO1, the minimally effective resveratrol concentration concomitantly induced evidence of cellular apoptosis. Taken together, these findings indicate that only particular phytonutrients are likely efficacious in upregulating NQO1 activity without also leading to hepatic cytotoxicity.
- Published
- 2016
- Full Text
- View/download PDF
5. Amyloid β peptide-induced inhibition of endothelial nitric oxide production involves oxidative stress-mediated constitutive eNOS/HSP90 interaction and disruption of agonist-mediated Akt activation.
- Author
-
Lamoke F, Mazzone V, Persichini T, Maraschi A, Harris MB, Venema RC, Colasanti M, Gliozzi M, Muscoli C, Bartoli M, and Mollace V
- Subjects
- Acetylcysteine pharmacology, Animals, Cattle, Cells, Cultured, Dose-Response Relationship, Drug, Drug Interactions, Endothelial Cells, Endothelium, Vascular cytology, Free Radical Scavengers pharmacology, Immunoprecipitation, Phosphorylation drug effects, Serine metabolism, Signal Transduction drug effects, Vascular Endothelial Growth Factor A pharmacology, Amyloid beta-Peptides pharmacology, HSP90 Heat-Shock Proteins metabolism, Nitric Oxide metabolism, Nitric Oxide Synthase Type III metabolism, Oxidative Stress drug effects, Proto-Oncogene Proteins c-akt metabolism
- Abstract
Background: Amyloid β (Aβ)-induced vascular dysfunction significantly contributes to the pathogenesis of Alzheimer's disease (AD). Aβ is known to impair endothelial nitric oxide synthase (eNOS) activity, thus inhibiting endothelial nitric oxide production (NO)., Method: In this study, we investigated Aβ-effects on heat shock protein 90 (HSP90) interaction with eNOS and Akt in cultured vascular endothelial cells and also explored the role of oxidative stress in this process., Results: Treatments of endothelial cells (EC) with Aβ promoted the constitutive association of HSP90 with eNOS but abrogated agonist (vascular endothelial growth factor (VEGF))-mediated HSP90 interaction with Akt. This effect resulted in blockade of agonist-mediated phosphorylation of Akt and eNOS at serine 1179. Furthermore, Aβ stimulated the production of reactive oxygen species in endothelial cells and concomitant treatments of the cells with the antioxidant N-acetyl-cysteine (NAC) prevented Aβ effects in promoting HSP90/eNOS interaction and rescued agonist-mediated Akt and eNOS phosphorylation., Conclusions: The obtained data support the hypothesis that oxidative damage caused by Aβ results in altered interaction of HSP90 with Akt and eNOS, therefore promoting vascular dysfunction. This mechanism, by contributing to Aβ-mediated blockade of nitric oxide production, may significantly contribute to the cognitive impairment seen in AD patients.
- Published
- 2015
- Full Text
- View/download PDF
6. Curcumin binds tubulin, induces mitotic catastrophe, and impedes normal endothelial cell proliferation.
- Author
-
Jackson SJ, Murphy LL, Venema RC, Singletary KW, and Young AJ
- Subjects
- Animals, Antioxidants pharmacology, Cattle, Cell Cycle Checkpoints drug effects, Cell Division drug effects, Cell Line, Endothelial Cells metabolism, Heme Oxygenase-1 genetics, Heme Oxygenase-1 metabolism, Microtubules drug effects, Microtubules metabolism, Cell Proliferation drug effects, Curcumin pharmacology, Endothelial Cells drug effects, Mitosis drug effects, Tubulin metabolism
- Abstract
Curcumin, a component of turmeric spice that imparts flavor and color to curry, is thought to possess anti-inflammatory and antioxidant properties in biological tissues. However, while such efficacies have been described in the context of carcinogenesis, the impact of curcumin on normal cell cycle regulation is poorly understood. Here, we provide evidence of curcumin toxicity in proliferating bovine aortic endothelial cells, at concentrations relevant to the diet and below those previously reported in cancer models. Upon confirming curcumin's ability to upregulate hemeoxygenase-1 in a dose-dependent fashion, we found the minimally efficacious curcumin concentration to also inhibit endothelial cell DNA synthesis. Moreover, curcumin concentrations below the minimum 2 μM threshold required to induce hemeoxygenase-1 bound tubulin protein in vitro and triggered hallmark evidence of mitotic catastrophe in vivo. Concentrations as low as 0.1 μM curcumin led to disproportionate DNA segregation, karyorrhexis, and micronucleation in proliferating endothelial cells. While suggesting a mechanism by which physiological curcumin concentrations inhibit cell cycle progression, these findings describe heretofore unappreciated curcumin toxicity with potential implications for endothelial growth, development, and tissue healing., (Published by Elsevier Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
7. Calcineurin-mediated dephosphorylation of eNOS at serine 116 affects eNOS enzymatic activity indirectly by facilitating c-Src binding and tyrosine 83 phosphorylation.
- Author
-
Ruan L, Torres CM, Buffett RJ, Kennard S, Fulton D, and Venema RC
- Subjects
- Animals, COS Cells, Calcium metabolism, Cattle, Cyclosporine pharmacology, Endothelial Cells drug effects, Endothelial Cells enzymology, Endothelial Cells metabolism, Endothelium, Vascular drug effects, Endothelium, Vascular enzymology, Endothelium, Vascular metabolism, Enzyme Inhibitors pharmacology, Nitric Oxide Synthase Type III antagonists & inhibitors, Permeability drug effects, Phosphorylation, Thapsigargin pharmacology, Calcineurin metabolism, Nitric Oxide Synthase Type III metabolism, Serine metabolism, Tyrosine metabolism
- Abstract
It has been shown previously that phosphorylation of the endothelial nitric oxide synthase (eNOS) at serine 116 (S116) under basal conditions suppresses eNOS enzymatic activity in endothelial cells. It has also been shown that vascular endothelial growth factor (VEGF) treatment of endothelial cells produces a rapid S116 dephosphorylation, which is blocked by the calcineurin inhibitor, cyclosporin A (CsA). In this study, we show that activation of eNOS in response to a variety of other eNOS-activating agonists and the cytosolic calcium-elevating agent, thapsigargin also involves CsA-inhibitable S116 dephosphorylation. Studies with the purified eNOS enzyme also demonstrate that neither mimicking phosphorylation at S116 nor phosphorylation of the purified enzyme at S116 in vitro has any effect on enzymatic activity. Phospho-mimicking, however, does interfere with the interaction of eNOS with c-Src, an interaction which is known to activate eNOS by phosphorylation at tyrosine 83 (Y83). Agonist-stimulated eNOS-Src complex formation, as well as agonist-stimulated Y83 phosphorylation, are blocked by calcineurin inhibition by CsA and by a cell-permeable calcineurin inhibitory peptide. Taken together, these data suggest a mechanism of eNOS regulation whereby calcineurin-mediated dephosphorylation of eNOS at S116 affects eNOS enzymatic activity indirectly, rather than directly, by facilitating c-Src binding and Y83 phosphorylation., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
8. Expression and functional significance of NADPH oxidase 5 (Nox5) and its splice variants in human blood vessels.
- Author
-
Pandey D, Patel A, Patel V, Chen F, Qian J, Wang Y, Barman SA, Venema RC, Stepp DW, Rudic RD, and Fulton DJ
- Subjects
- Blood Vessels cytology, Cell Proliferation, Cells, Cultured, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, Humans, Isoenzymes metabolism, Membrane Proteins genetics, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular metabolism, NADPH Oxidase 5, NADPH Oxidases genetics, RNA, Messenger metabolism, Reactive Oxygen Species metabolism, Blood Vessels metabolism, Membrane Proteins metabolism, NADPH Oxidases metabolism, Signal Transduction physiology
- Abstract
The expression and functional significance of NADPH oxidase 5 (Nox5) and its five isoforms in vascular cells is poorly understood. The goal of this study was to determine whether Nox5-α, -β, -δ, -γ, and -ε (short) are expressed in human blood vessels and evaluate their respective functions. Nox5 mRNA and protein were detected in human blood vessels, cultured human vascular smooth muscle (HVSMC) and endothelium, but not fibroblasts. The most abundant isoforms were α and β, whereas δ and γ were not detected. Nox5-α and -β produced reactive oxygen species (ROS), but -δ, -γ, and -ε were not catalytically active. Coexpression of the active Nox5 isoforms with inactive Nox5 variants suppressed ROS production, and coimmunoprecipitation revealed that Nox5-β binds the inactive ε variant, which may account for reduced ROS production. In HVSMC, angiotensin II, endothelin-1 and TNF-α increased endogenous Nox5 mRNA levels, while adenovirus-mediated overexpression of Nox5 promoted p38 MAPK, JAK2, JNK, and ERK1/2 phosphorylation in endothelial cells (EC), but only increased ERK1/2 phosphorylation in HVSMC. At higher levels of Nox5, there was evidence of increased apoptosis in EC, but not in HVSMC, as detected by the presence of cleaved caspase-3 and cleaved poly(ADP-ribose)polymerase. Although catalytically inactive, Nox5-ε potently activated ERK in HVSMC, and increased expression of Nox5-ε promoted HVSMC proliferation. Nox5 is expressed in human blood vessels. The Nox5-α and -β splice variants are the major isoforms that are expressed and the only variants capable of ROS production. Nox5-ε can inhibit Nox5 activity and activate ERK and HVSMC proliferation.
- Published
- 2012
- Full Text
- View/download PDF
9. Nitric oxide reduces NADPH oxidase 5 (Nox5) activity by reversible S-nitrosylation.
- Author
-
Qian J, Chen F, Kovalenkov Y, Pandey D, Moseley MA, Foster MW, Black SM, Venema RC, Stepp DW, and Fulton DJ
- Subjects
- Animals, Blotting, Western, COS Cells, Calcium metabolism, Cell Line, Chlorocebus aethiops, Humans, Mass Spectrometry, NADPH Oxidase 5, Nitric Oxide Donors pharmacology, Phosphorylation, Reactive Oxygen Species metabolism, Membrane Proteins metabolism, NADPH Oxidases metabolism, Nitric Oxide physiology, Nitroso Compounds metabolism
- Abstract
The NADPH oxidases (Noxs) are a family of transmembrane oxidoreductases that produce superoxide and other reactive oxygen species (ROS). Nox5 was the last of the conventional Nox isoforms to be identified and is a calcium-dependent enzyme that does not depend on accessory subunits for activation. Recently, Nox5 was shown to be expressed in human blood vessels and therefore the goal of this study was to determine whether nitric oxide (NO) can modulate Nox5 activity. Endogenously produced NO potently inhibited basal and stimulated Nox5 activity and this inhibition was reversible with chronic, but not acute, exposure to L-NAME. Nox5 activity was reduced by NO donors, iNOS, and eNOS and in endothelial cells and LPS-stimulated smooth muscle cells in a manner dependent on NO concentration. ROS production was diminished by NO in an isolated enzyme activity assay replete with surplus calcium and NADPH. There was no evidence for NO-dependent changes in tyrosine nitration, glutathiolation, or phosphorylation of Nox5. In contrast, there was evidence for the increased nitrosylation of Nox5 as determined by the biotin-switch assay and mass spectrometry. Four S-nitrosylation sites were identified and of these, mutation of C694 dramatically lowered Nox5 activity, NO sensitivity, and biotin labeling. Furthermore, coexpression of the denitrosylation enzymes thioredoxin 1 and GSNO reductase prevented NO-dependent inhibition of Nox5. The potency of NO against other Nox enzymes was in the order Nox1 ≥ Nox3 > Nox5 > Nox2, whereas Nox4 was refractory. Collectively, these results suggest that endogenously produced NO can directly S-nitrosylate and inhibit the activity of Nox5., (Published by Elsevier Inc.)
- Published
- 2012
- Full Text
- View/download PDF
10. Pin1 prolyl isomerase regulates endothelial nitric oxide synthase.
- Author
-
Ruan L, Torres CM, Qian J, Chen F, Mintz JD, Stepp DW, Fulton D, and Venema RC
- Subjects
- Animals, Aorta cytology, Aorta metabolism, COS Cells, Cattle, Cells, Cultured, Chlorocebus aethiops, Endothelium, Vascular cytology, Humans, Models, Animal, NIMA-Interacting Peptidylprolyl Isomerase, Peptidylprolyl Isomerase genetics, Phosphorylation, Signal Transduction physiology, Transfection, Endothelium, Vascular metabolism, Nitric Oxide Synthase Type III metabolism, Peptidylprolyl Isomerase metabolism
- Abstract
Objective: The Pin1 prolyl isomerase acts in concert with proline-directed protein kinases to regulate function of protein substrates through isomerization of peptide bonds that link phosphoserine or phosphothreonine to proline. We sought to determine whether Pin1 interacts with endothelial nitric oxide synthase (eNOS) in endothelial cells in a manner that depends on proline-directed phosphorylation of the eNOS enzyme and whether this interaction influences basal or agonist-stimulated eNOS activity., Methods and Results: Inhibitors of the extracellular-regulated kinase (ERK) 1/2 MAP kinases inhibit proline-directed phosphorylation of eNOS at serine 116 (Ser116) in bovine aortic endothelial cells (BAECs). Moreover, eNOS and Pin1 can be coimmunoprecipitated from BAECs only when Ser116 is phosphorylated. In addition, phosphomimetic Ser116Asp eNOS, but not wild-type eNOS, can be coimmunoprecipitated with Pin1 coexpressed in COS-7 cells. Inhibition of Pin1 in BAECs by juglone or by dominant negative Pin1 increases basal and agonist-stimulated NO release from the cells, whereas overexpression of wild-type Pin1 in BAECs suppresses basal and agonist-stimulated NO production. Overexpression of wild-type Pin1 in intact aortae also reduces agonist-induced relaxation of aortic rings., Conclusions: Our results demonstrate a novel form of eNOS regulation in endothelial cells and blood vessels through Ser116 phosphorylation-dependent interaction of eNOS with Pin1.
- Published
- 2011
- Full Text
- View/download PDF
11. Activation of endothelial nitric oxide synthase by the pro-apoptotic drug embelin: Striking discrepancy between nitric oxide-mediated cyclic GMP accumulation and L-citrulline formation.
- Author
-
Schmidt K, Martens-Lobenhoffer J, Meinitzer A, Graier WF, Torres CM, Venema RC, and Mayer B
- Subjects
- Animals, Nitric Oxide metabolism, Apoptosis, Benzoquinones pharmacology, Citrulline biosynthesis, Cyclic GMP metabolism, Endothelial Cells enzymology, Nitric Oxide Synthase Type III metabolism, Signal Transduction
- Abstract
The benzoquinone derivative embelin is a multifunctional drug that not only induces apoptosis by inhibiting XIAP, the X chromosome-linked inhibitor of apoptosis protein, but also blocks nuclear factor-kappaB signaling pathways, thereby leading to down-regulation of a variety of gene products involved in tumor cell survival, proliferation, invasion, angiogenesis, and inflammation. Here, we report that embelin activates and modulates l-arginine/nitric oxide/cyclic GMP signaling in cultured endothelial cells. Embelin elicited a rapid increase of intracellular free Ca(2+), leading to activation of endothelial nitric oxide synthase (eNOS) and NO-induced cGMP accumulation. While the cGMP response was comparable to that caused by other Ca(2+)-mobilizing agents, the stimulatory effect of embelin on l-citrulline formation (approximately 4-fold) was substantially lower than that observed upon activation of eNOS with the Ca(2+) ionophore A23187 (approximately 18-fold), the receptor agonist ATP (approximately 16-fold) or the sarco-endoplasmic reticulum Ca(2+)-ATPase inhibitor thapsigargin (approximately 14-fold). The apparent discrepancy between NO/cGMP and l-citrulline formation in embelin-treated cells was not due to enhanced metabolism and/or efflux of l-citrulline, increased NO bioavailability, inhibition of cGMP hydrolysis, sensitization of soluble guanylate cyclase (sGC) to NO, or enhanced formation of a sGC/eNOS complex. Our puzzling observations suggest that embelin improves coupling of endothelial NO synthesis to sGC activation through mobilization of an as yet unrecognized signaling pathway., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
12. Inhibition of endothelial nitric oxide synthase by the lipid phosphatase PTEN.
- Author
-
Church JE, Qian J, Kumar S, Black SM, Venema RC, Papapetropoulos A, and Fulton DJ
- Subjects
- Animals, Aorta cytology, Aorta metabolism, COS Cells, Chlorocebus aethiops, Endothelial Cells metabolism, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, Humans, Nitric Oxide metabolism, Nitric Oxide Synthase Type III genetics, Nitric Oxide Synthase Type III metabolism, PTEN Phosphohydrolase genetics, Phosphorylation, Transfection, Gene Expression Regulation, Enzymologic, Nitric Oxide Synthase Type III antagonists & inhibitors, PTEN Phosphohydrolase metabolism
- Abstract
PTEN (phosphatase and tensin homologue deleted on chromosome 10) is a lipid phosphatase that functions as a negative regulator of the phosphoinositide-3-kinase (PI3K) pathway. The present study sought to examine in depth the interaction between PTEN and eNOS activity. Co-expression of eNOS and PTEN in COS-7 cells significantly decreased NO production compared to eNOS alone, while co-expression of eNOS and the dominant negative mutant PTEN(C124A) significantly increased NO production. Upon examination of the putative eNOS phosphorylation sites, phosphorylation of S116, T497, S617, S635 and S1179 was decreased by PTEN co-expression, while the dominant negative PTEN(C124A) produced an increase in phosphorylation of all sites except S116 and S635. A myristoylation-deficient eNOS construct with little dependence on phosphorylation state (G2AeNOS) was not significantly affected by co-expression with either PTEN or PTEN(C124A). Likewise, an eNOS construct with a triple phospho-null mutation (S617A, S635A and S1179A) was also unaffected by co-expression with either PTEN or PTEN(C124A). Purified PTEN or PTEN(C124A) failed to interact with purified eNOS in vitro, arguing against a direct interaction between PTEN and eNOS. When the PTEN constructs were expressed in human aortic endothelial cells (HAECs), PTEN significantly decreased NO production and PTEN(C124A) increased it, and both S617 and S1179 were altered by co-expression with the PTEN constructs. Increased expression of PTEN in endothelial cells did not influence superoxide production. We conclude that PTEN is a regulator of eNOS function both when expressed in COS-7 cells and in human endothelial cells, and does so via its effects on the PI3K/Akt pathway.
- Published
- 2010
- Full Text
- View/download PDF
13. Alterations in lung arginine metabolism in lambs with pulmonary hypertension associated with increased pulmonary blood flow.
- Author
-
Sharma S, Kumar S, Sud N, Wiseman DA, Tian J, Rehmani I, Datar S, Oishi P, Fratz S, Venema RC, Fineman JR, and Black SM
- Subjects
- Amino Acid Sequence, Amino Acids blood, Animals, Arginase metabolism, Blood Pressure, Female, Molecular Sequence Data, Nitric Oxide physiology, Pregnancy, Sheep, Arginine metabolism, Hypertension, Pulmonary metabolism, Lung metabolism, Pulmonary Circulation
- Abstract
Previous studies demonstrate impaired nitric oxide (NO) signaling in children and animal models with congenital heart defects and increased pulmonary blood flow. However, the molecular mechanisms underlying these alterations remain incompletely understood. The purpose of this study was to determine if early changes in arginine metabolic pathways could play a role in the reduced NO signaling demonstrated in our lamb model of congenital heart disease with increased pulmonary blood flow (Shunt lambs). The activities of the arginine recycling enzymes, argininosuccinate synthetase (ASS) and argininosuccinate lyase (ASL) were both decreased in lung tissues of Shunt lambs while arginase activity was increased. Associated with these alterations, lung L-arginine levels were decreased. These changes correlated with an increase in NO synthase-derived reactive oxygen species (ROS) generation. This study provides further insights into the molecular mechanisms leading to decreased NO signaling in Shunt lambs and suggests that altered arginine metabolism may play a role in the development of the endothelial dysfunction associated with pulmonary hypertension secondary to increased pulmonary blood flow.
- Published
- 2009
- Full Text
- View/download PDF
14. The hnRNA-binding proteins hnRNP L and PTB are required for efficient translation of the Cat-1 arginine/lysine transporter mRNA during amino acid starvation.
- Author
-
Majumder M, Yaman I, Gaccioli F, Zeenko VV, Wang C, Caprara MG, Venema RC, Komar AA, Snider MD, and Hatzoglou M
- Subjects
- 5' Untranslated Regions, Animals, Cationic Amino Acid Transporter 1 genetics, Cell Line, Gene Expression Regulation, Heterogeneous-Nuclear Ribonucleoprotein L genetics, Mice, Nucleic Acid Conformation, Polypyrimidine Tract-Binding Protein genetics, RNA, Messenger genetics, RNA-Binding Proteins genetics, Rats, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Ribosomes metabolism, Amino Acids metabolism, Cationic Amino Acid Transporter 1 metabolism, Heterogeneous-Nuclear Ribonucleoprotein L metabolism, Polypyrimidine Tract-Binding Protein metabolism, Protein Biosynthesis, RNA, Messenger metabolism, RNA-Binding Proteins metabolism
- Abstract
The response to amino acid starvation involves the global decrease of protein synthesis and an increase in the translation of some mRNAs that contain an internal ribosome entry site (IRES). It was previously shown that translation of the mRNA for the arginine/lysine amino acid transporter Cat-1 increases during amino acid starvation via a mechanism that utilizes an IRES in the 5' untranslated region of the Cat-1 mRNA. It is shown here that polypyrimidine tract binding protein (PTB) and an hnRNA binding protein, heterogeneous nuclear ribonucleoprotein L (hnRNP L), promote the efficient translation of Cat-1 mRNA during amino acid starvation. Association of both proteins with Cat-1 mRNA increased during starvation with kinetics that paralleled that of IRES activation, although the levels and subcellular distribution of the proteins were unchanged. The sequence CUUUCU within the Cat-1 IRES was important for PTB binding and for the induction of translation during amino acid starvation. Binding of hnRNP L to the IRES or the Cat-1 mRNA in vivo was independent of PTB binding but was not sufficient to increase IRES activity or Cat-1 mRNA translation during amino acid starvation. In contrast, binding of PTB to the Cat-1 mRNA in vivo required hnRNP L. A wider role of hnRNP L in mRNA translation was suggested by the decrease of global protein synthesis in cells with reduced hnRNP L levels. It is proposed that PTB and hnRNP L are positive regulators of Cat-1 mRNA translation via the IRES under stress conditions that cause a global decrease of protein synthesis.
- Published
- 2009
- Full Text
- View/download PDF
15. Increased nitric oxide synthase activity and Hsp90 association in skeletal muscle following chronic exercise.
- Author
-
Harris MB, Mitchell BM, Sood SG, Webb RC, and Venema RC
- Subjects
- Animals, Male, Nitric Oxide Synthase Type I, Nitric Oxide Synthase Type II metabolism, Nitric Oxide Synthase Type III, Phosphorylation, Rats, Rats, Sprague-Dawley, Up-Regulation, HSP90 Heat-Shock Proteins metabolism, Muscle Contraction, Muscle, Skeletal enzymology, Nitric Oxide Synthase metabolism, Physical Exertion
- Abstract
Exercise training results in dynamic changes in skeletal muscle blood flow and metabolism. Nitric oxide (NO) influences blood flow, oxidative stress, and glucose metabolism. Hsp90 interacts directly with nitric oxide synthases (NOS), increasing NOS activity and altering the balance of superoxide versus NO production. In addition, Hsp90 expression increases in various tissues following exercise. Therefore, we tested the hypothesis that exercise training increases Hsp90 expression as well as Hsp90/NOS association and NOS activity in skeletal muscle. Male, Sprague-Dawley rats were assigned to either a sedentary or exercise trained group (n = 10/group). Exercise training consisted of running on a motorized treadmill for 10 weeks at 30 m/min, 5% grade for 1 h. Western blotting revealed that exercise training resulted in a 1.9 +/- 0.1-fold increase in Hsp90 expression in the soleus muscle but no increase in neuronal nitric oxide synthase (nNOS), inducible nitric oxide synthase, or endothelial nitric oxide synthase (eNOS). Exercise training also resulted in a 3.4 +/- 1.0-fold increase in Hsp90 association with nNOS, a 2.3 +/- 0.4-fold increase association with eNOS measured by immunoprecipitation as well as a 1.5 +/- 0.3-fold increase in eNOS phosphorylation at Ser-1179. Total NOS activity measured by the rate of conversion of L-[(14)C]arginine to L-[(14)C]citrulline was increased by 1.42 +/- 0.9 fold in soleus muscle following exercise training compared to controls. In summary, a 10-week treadmill training program in rats results in a significant increase in total NOS activity in the soleus which may be due, in part, to increased NOS interaction with Hsp90 and phosphorylation. This interaction may play a role in altering muscle blood flow and skeletal muscle redox status.
- Published
- 2008
- Full Text
- View/download PDF
16. An essential role for SRC-activated STAT-3 in 14,15-EET-induced VEGF expression and angiogenesis.
- Author
-
Cheranov SY, Karpurapu M, Wang D, Zhang B, Venema RC, and Rao GN
- Subjects
- 8,11,14-Eicosatrienoic Acid pharmacology, Aorta cytology, Cell Movement drug effects, Cell Movement physiology, Cells, Cultured, Collagen, Drug Combinations, Endothelial Cells cytology, Endothelial Cells physiology, Gene Expression drug effects, Gene Expression physiology, Humans, Laminin, Myocytes, Smooth Muscle cytology, Myocytes, Smooth Muscle physiology, Phosphorylation drug effects, Proteoglycans, Signal Transduction drug effects, Signal Transduction physiology, Vascular Endothelial Growth Factor A metabolism, src-Family Kinases metabolism, 8,11,14-Eicosatrienoic Acid analogs & derivatives, Neovascularization, Physiologic drug effects, Neovascularization, Physiologic physiology, STAT3 Transcription Factor metabolism, Vascular Endothelial Growth Factor A genetics
- Abstract
To understand the molecular mechanisms underlying 14,15-epoxyeicosatrienoic acid (14,15-EET)-induced angiogenesis, here we have studied the role of signal transducer and activator of transcription-3 (STAT-3). 14,15-EET stimulated the tyrosine phosphorylation of STAT-3 and its translocation from the cytoplasm to the nucleus in human dermal microvascular endothelial cells (HDMVECs). Adenovirus-mediated delivery of dominant negative STAT-3 substantially inhibited 14,15-EET-induced HDMVEC migration, and tube formation and Matrigel plug angiogenesis. 14,15-EET activated Src, as measured by its tyrosine phosphorylation and blockade of its activation by adenovirus-mediated expression of its dominant negative mutant, significantly attenuated 14,15-EET-induced STAT-3 phosphorylation in HDMVECs and the migration and tube formation of these cells and Matrigel plug angiogenesis. 14,15-EET induced the expression of vascular endothelial cell growth factor (VEGF) in a time- and Src-STAT-3-dependent manner in HDMVECs. Transfac analysis of VEGF promoter revealed the presence of STAT-binding elements and 14,15-EET induced STAT-3 binding to this promoter in vivo, and this interaction was inhibited by suppression of Src-STAT-3 signaling. Neutralizing anti-VEGF antibodies completely blocked 14,15-EET-induced HDMVEC migration and tube formation and Matrigel plug angiogenesis. These results reveal that Src-dependent STAT-3-mediated VEGF expression is a major mechanism of 14,15-EET-induced angiogenesis.
- Published
- 2008
- Full Text
- View/download PDF
17. Agonist-stimulated endothelial nitric oxide synthase activation and vascular relaxation. Role of eNOS phosphorylation at Tyr83.
- Author
-
Fulton D, Ruan L, Sood SG, Li C, Zhang Q, and Venema RC
- Subjects
- Adenosine Triphosphate pharmacology, Angiopoietins pharmacology, Animals, Aorta cytology, Bradykinin pharmacology, COS Cells, Cattle, Chlorocebus aethiops, Endothelial Cells cytology, Enzyme Inhibitors pharmacology, Estrogens pharmacology, Humans, Kidney cytology, Lysophospholipids pharmacology, Male, Nitric Oxide metabolism, Nitric Oxide Synthase Type II genetics, Nitric Oxide Synthase Type II metabolism, Nitric Oxide Synthase Type III, Phosphorylation drug effects, Rats, Rats, Sprague-Dawley, Sphingosine analogs & derivatives, Sphingosine pharmacology, Thapsigargin pharmacology, Tyrosine metabolism, src-Family Kinases metabolism, Endothelial Cells enzymology, Vascular Endothelial Growth Factor A pharmacology, Vasodilation physiology
- Abstract
Tyr83 in endothelial nitric oxide synthase (eNOS) has been identified previously as a site of Src kinase-mediated phosphorylation of eNOS in bovine aortic endothelial cells (BAECs) that is phosphorylated in response to oxidant stress. In the present study, we have used a phospho-specific antibody to show that Tyr83 in eNOS is also phosphorylated in both BAECs and intact blood vessel segments in response to treatment with a variety of different eNOS-activating agonists, including thapsigargin, vascular endothelial growth factor, bradykinin, ATP, sphingosine-1-phosphate, estrogen, angiopoietin, and acetylcholine. Agonist stimulation of eNOS Tyr83 phosphorylation as well as agonist stimulation of endothelial NO release in BAECs is blocked by Src kinase inhibition by either 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo [3,4-d] pyrimidine (PP2) or by dominant negative Src. Mutation of Tyr83 to a nonphosphorylatable Phe blocks agonist stimulation of NO release from eNOS-reconstituted eNOS knockdown endothelial cells. Mutation of Tyr83 also attenuates agonist-induced relaxation of eNOS-reconstituted aortic rings from eNOS knockout mice. Phosphorylation of eNOS at Tyr83 thus appears to be a common covalent modification that is induced, not only by oxidant stress but also by other physiologically relevant extracellular signals known to be important in regulation of eNOS activity in vivo. Moreover, our results demonstrate an important role for Src-mediated phosphorylation of eNOS at Tyr83 in agonist stimulation of eNOS activation and vascular relaxation.
- Published
- 2008
- Full Text
- View/download PDF
18. Nitric oxide and superoxide generation from endothelial NOS: modulation by HSP90.
- Author
-
Sud N, Sharma S, Wiseman DA, Harmon C, Kumar S, Venema RC, Fineman JR, and Black SM
- Subjects
- Aging, Animals, Cell Separation, Cells, Cultured, Endothelial Cells cytology, Endothelial Cells drug effects, Endothelial Cells enzymology, Fetus cytology, Fetus drug effects, HSP90 Heat-Shock Proteins antagonists & inhibitors, Lung cytology, Lung drug effects, Lung enzymology, Macrolides pharmacology, Protein Binding drug effects, Pulmonary Artery cytology, Pulmonary Artery drug effects, Pulmonary Artery enzymology, Shear Strength, Sheep, HSP90 Heat-Shock Proteins metabolism, Nitric Oxide biosynthesis, Nitric Oxide Synthase Type III metabolism, Superoxides metabolism
- Abstract
Previously, we have shown that pulmonary arterial endothelial cells (PAECs) isolated from fetal lambs produce significant levels of nitric oxide (NO) but minimal superoxide upon stimulation, whereas PAECs isolated from 4-wk-old lambs produce significant amounts of both NO and superoxide. These data indicated that a certain degree of uncoupling of endothelial NO synthase (eNOS) occurs in PAECs during postnatal development. In this study, we sought to extend these studies by investigating the potential role of heat shock protein 90 (HSP90) in eNOS coupling. Western blot analyses revealed higher HSP90 expression in PAECs isolated from fetal compared with 4-wk-old lambs, whereas the analysis of recombinant human eNOS activation in vitro in the presence of HSP90 indicated that HSP90 significantly augmented NO production while inhibiting superoxide generation from eNOS. To further investigate whether HSP90 could be involved in uncoupling of eNOS in PAECs isolated from 4-wk-old lambs, we utilized an adenovirus to overexpress HSP90. We found that overexpression of HSP90 significantly increased the shear-stimulated association of HSP90 with eNOS and led to significant increases in NO production and reduced NOS-dependent superoxide generation. Conversely, the exposure of PAECs isolated from fetal lambs to the HSP90 inhibitor radicicol led to significant decreases in eNOS-HSP90 interactions, decreased shear-stimulated NO generation, and increased NOS-dependent superoxide production indicative of eNOS uncoupling. Finally, we examined eNOS-HSP90 interactions in our lamb model of pulmonary hypertension associated with increased pulmonary blood flow (shunt). Our data indicate that HSP90-eNOS interactions were decreased in shunt lambs and that this was associated with decreased NO generation and an increase in eNOS-dependent generation of superoxide. Together, our data support a significant role for HSP90 in promoting NO generation and inhibiting superoxide generation by eNOS and indicate that the disruption of this interaction may be involved in the endothelial dysfunction associated with pulmonary hypertension.
- Published
- 2007
- Full Text
- View/download PDF
19. Role of eNOS phosphorylation at Ser-116 in regulation of eNOS activity in endothelial cells.
- Author
-
Li C, Ruan L, Sood SG, Papapetropoulos A, Fulton D, and Venema RC
- Subjects
- Animals, Aorta, Thoracic enzymology, Aorta, Thoracic physiology, Cattle, Cells, Cultured, Endothelial Cells cytology, Endothelium, Vascular cytology, Humans, In Vitro Techniques, Mice, Mice, Knockout, Mutation, Nitric Oxide metabolism, Nitric Oxide Synthase Type III genetics, Phosphorylation, Vasodilation, Endothelial Cells enzymology, Nitric Oxide Synthase Type III metabolism, Serine metabolism
- Abstract
Endothelial nitric oxide synthase (eNOS) catalyzes the conversion of L-arginine to L-citrulline and nitric oxide (NO), an important modulator of vascular function. eNOS is regulated post-translationally through phosphorylation/dephosphorylation at a number of specific phosphorylation sites including Ser-116 in the bovine eNOS sequence. Whether phosphorylation of eNOS at Ser-116 in endothelial cells is stimulatory or inhibitory has not previously been definitively determined. In this study we show that mimicking phosphorylation of eNOS at Ser-116 by Asp mutation reduces basal NO release from endothelial cells. Preventing phosphorylation at this site by Ala mutation increases the amount of NO release from endothelial cells in response to agonist stimulation. In addition, mimicking phosphorylation of Ser-116 increases eNOS association with caveolin-1 and reduces the vascular reactivity of intact aortic rings. eNOS phosphorylation at Ser-116, therefore, appears to contribute to negative modulation of eNOS activity and hence to regulation of vascular tone.
- Published
- 2007
- Full Text
- View/download PDF
20. Chaperone-dependent E3 ligase CHIP ubiquitinates and mediates proteasomal degradation of soluble guanylyl cyclase.
- Author
-
Xia T, Dimitropoulou C, Zeng J, Antonova GN, Snead C, Venema RC, Fulton D, Qian S, Patterson C, Papapetropoulos A, and Catravas JD
- Subjects
- Animals, COS Cells, Chlorocebus aethiops, Molecular Chaperones metabolism, Solubility, Soluble Guanylyl Cyclase, Guanylate Cyclase metabolism, HSC70 Heat-Shock Proteins metabolism, Proteasome Endopeptidase Complex metabolism, Receptors, Cytoplasmic and Nuclear metabolism, Signal Transduction physiology, Ubiquitin metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
The nitric oxide receptor soluble guanylyl cyclase (sGC) exists in multimeric protein complexes, including heat shock protein (HSP) 90 and endothelial nitric oxide synthase. Inhibition of HSP90 by geldanamycin causes proteasomal degradation of sGC protein. In this study, we have investigated whether COOH terminus of heat shock protein 70-interacting protein (CHIP), a co-chaperone molecule that is involved in protein folding but is also a chaperone-dependent ubiquitin E3 ligase, could play a role in the process of degradation of sGC. Transient overexpression of CHIP in COS-7 cells degraded heterologous sGC in a concentration-related manner; this downregulation of sGC was abrogated by the proteasome inhibitor MG-132. Transfection of tetratricopeptide repeats and U-box domain CHIP mutants attenuated sGC degradation, suggesting that both domains are indispensable for CHIP function. Results from immunoprecipitation and indirect immunofluorescent microscopy experiments demonstrated that CHIP is associated with sGC, HSP90, and HSP70 in COS-7 cells. Furthermore, CHIP increased the association of HSP70 with sGC. In in vitro ubiquitination assays using purified proteins and ubiquitin enzymes, E3 ligase CHIP directly ubiquitinated sGC; this ubiquitination was potentiated by geldanamycin in COS-7 cells, followed by proteasomal degradation. In rat aortic smooth muscle cells, endogenous sGC was also degraded by adenovirus-infected wild-type CHIP but not by the chaperone interaction-deficient K30A CHIP, whereas CHIP, but not K30A, attenuated sGC expression in, and nitric oxide donor-induced relaxation of, rat aortic rings, suggesting that CHIP plays a regulatory role under physiological conditions. This study reveals a new mechanism for the regulation of sGC, an important mediator of cellular and vascular function.
- Published
- 2007
- Full Text
- View/download PDF
21. Heat shock protein 90 inhibitors prolong survival, attenuate inflammation, and reduce lung injury in murine sepsis.
- Author
-
Chatterjee A, Dimitropoulou C, Drakopanayiotakis F, Antonova G, Snead C, Cannon J, Venema RC, and Catravas JD
- Subjects
- Animals, Benzoquinones pharmacology, Chemokines metabolism, Cytokines metabolism, Inflammation, Lactams, Macrocyclic pharmacology, Lung metabolism, Macrolides pharmacology, Male, Mice, Mice, Inbred C57BL, NF-kappa B metabolism, Nitrates metabolism, Nitric Oxide Synthase Type II metabolism, Nitrites metabolism, Peroxidase metabolism, Respiratory Distress Syndrome etiology, Respiratory Distress Syndrome metabolism, Respiratory Distress Syndrome mortality, Sepsis complications, Survival Rate, HSP90 Heat-Shock Proteins antagonists & inhibitors, Inflammation Mediators metabolism, Respiratory Distress Syndrome prevention & control, Sepsis physiopathology
- Abstract
Rationale: Severe sepsis is the leading cause of death for patients in intensive care units. Patients with severe sepsis develop multiple organ failure, including acute lung injury (ALI), resulting from a deregulated inflammatory response. Inhibitors of the ubiquitous chaperone, heat shock protein 90 (Hsp90), block the activity of certain proinflammatory mediators in vitro. We hypothesized that Hsp90 inhibitors may ameliorate the inflammation and ALI associated with severe sepsis., Objectives: To test the hypothesis that Hsp90 inhibitors prolong survival, attenuate inflammation, and reduce lung injury in a murine model of sepsis., Methods: Male C57BL/6 mice received either one of two Hsp90 inhibitors, radicicol or 17-allylaminodemethoxygeldanamycin (17-AAG), 24, 12, 6, and 0 hours before receiving a lethal dose of endotoxin (6.75 x 10(4) endotoxin units/g body weight). Outcomes included survival and parameters of systemic inflammation (plasma neutrophil, cytokine, chemokine, and nitrite/nitrate levels), pulmonary inflammation (lung nuclear factor-kappaB and myeloperoxidase activities, inducible nitric oxide synthase expression, inducible nitric oxide synthase-Hsp90 complex formation, and leukocyte infiltration), and lung injury (pulmonary capillary leak and lung function)., Measurements and Main Results: Mice pretreated with vehicle and receiving endotoxin exhibited 100% 24-hour lethality, a dramatic increase in all parameters of systemic and pulmonary inflammation, increased capillary leak, and reduced lung function. Compared with them, mice receiving either radicicol or 17-AAG before endotoxin exhibited prolonged survival, reduced or abolished increases in systemic and pulmonary inflammatory parameters, attenuated capillary leak, and restored, normal lung function., Conclusions: Hsp90 inhibitors may offer a new pharmacological tool in the management of severe sepsis and severe sepsis-induced ALI.
- Published
- 2007
- Full Text
- View/download PDF
22. Sulforaphane suppresses angiogenesis and disrupts endothelial mitotic progression and microtubule polymerization.
- Author
-
Jackson SJ, Singletary KW, and Venema RC
- Subjects
- Angiogenesis Inhibitors therapeutic use, Animals, Anticarcinogenic Agents pharmacology, Cattle, Cell Proliferation drug effects, Cells, Cultured, Collagen administration & dosage, Dose-Response Relationship, Drug, Drug Combinations, Endothelial Cells metabolism, Female, Injections, Subcutaneous, Isothiocyanates, Laminin administration & dosage, Mice, Mice, Inbred BALB C, Microtubules metabolism, Neovascularization, Pathologic prevention & control, Proteoglycans administration & dosage, Spindle Apparatus metabolism, Subcutaneous Tissue blood supply, Sulfoxides, Thiocyanates therapeutic use, Time Factors, Tubulin Modulators therapeutic use, Vascular Endothelial Growth Factor A administration & dosage, Angiogenesis Inhibitors pharmacology, Endothelial Cells drug effects, Microtubules drug effects, Mitosis drug effects, Spindle Apparatus drug effects, Thiocyanates pharmacology, Tubulin Modulators pharmacology
- Abstract
Sulforaphane (SUL), an isothiocyanate derived from broccoli and other cruciferous vegetables, is known to induce phase II detoxification enzymes, disrupt cancer cell microtubule polymerization, and trigger cell cycle arrest in breast and colon cancer cells. Here, we provide the first evidence that SUL also acts to inhibit angiogenesis via suppression of endothelial cell proliferation. Bovine aortic endothelial (BAE) cells were exposed to concentrations of up to 15 microM SUL prior to cell cycle analysis and mitotic index quantification. Within 24 h, 15 microM SUL clearly induced G(2)/M accumulation and pre-metaphase arrest in BAE cells. Moreover, immunofluorescence tubulin staining indicated that this same SUL concentration was efficacious in not only disrupting mitotic progression, but also in perturbing normal polymerization of mitotic (and cytoplasmic) microtubules. Furthermore, daily administration of SUL (100 nmol/day, i.v. for 7 days) to female Balb/c mice bearing VEGF-impregnated Matrigel plugs strongly and significantly (P<0.05) suppressed angiogenesis progression as measured by hemoglobin concentration. Taken together, these findings suggest that the endothelial cell population is a novel target of SUL action both in vitro and in vivo. This mechanism of SUL-induced endothelial microtubule disruption and early mitotic arrest may further discern a potential role of SUL as a chemopreventive agent.
- Published
- 2007
- Full Text
- View/download PDF
23. Nitric oxide preconditioning regulates endothelial monolayer integrity via the heat shock protein 90-soluble guanylate cyclase pathway.
- Author
-
Antonova GN, Snead CM, Antonov AS, Dimitropoulou C, Venema RC, and Catravas JD
- Subjects
- Actin Cytoskeleton drug effects, Actin Cytoskeleton metabolism, Animals, Aorta drug effects, Aorta metabolism, Apoptosis drug effects, Cattle, Cells, Cultured, Cyclic GMP analogs & derivatives, Cyclic GMP metabolism, Cyclic GMP pharmacology, Cyclic GMP-Dependent Protein Kinases antagonists & inhibitors, Dose-Response Relationship, Drug, Electric Impedance, Endothelial Cells drug effects, Enzyme Inhibitors pharmacology, Guanylate Cyclase antagonists & inhibitors, Nitroprusside pharmacology, Oxadiazoles pharmacology, Quinoxalines pharmacology, Receptors, Cytoplasmic and Nuclear antagonists & inhibitors, Soluble Guanylyl Cyclase, Spermine analogs & derivatives, Spermine pharmacology, Time Factors, Endothelial Cells metabolism, Guanylate Cyclase metabolism, HSP90 Heat-Shock Proteins metabolism, Nitric Oxide metabolism, Nitric Oxide Donors pharmacology, Receptors, Cytoplasmic and Nuclear metabolism, Wound Healing drug effects
- Abstract
Large (pathological) amounts of nitric oxide (NO) induce cell injury, whereas low (physiological) NO concentrations often ameliorate cell injury. We tested the hypotheses that pretreatment of endothelial cells with low concentrations of NO (preconditioning) would prevent injury induced by high NO concentrations. Apoptosis, induced in bovine aortic endothelial cells (BAECs) by exposing them to either 4 mM sodium nitroprusside (SNP) or 0.5 mM N-(2-aminoethyl)-N-(2-hydroxy-2-nitrosohydrazino)-1,2-ethylenediamine (spermine NONOate) for 8 h, was abolished by 24-h pretreatment with either 100 microM SNP, 10 microM spermine NONOate, or 100 microM 8-bromo-cGMP (8-Br-cGMP). Repair of BAECs following wounding, measured as the recovery rate of transendothelial electrical resistance, was delayed by 8-h exposure to 4 mM SNP, and this delay was significantly attenuated by 24-h pretreatment with 100 microM SNP. NO preconditioning produced increased association and expression of soluble guanyl cyclase (sGC) and heat shock protein 90 (HSP90). The protective effect of NO preconditioning, but not the injurious effect of 4 mM SNP, was abolished by either a sGC activity inhibitor 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one (ODQ) or a HSP90 binding inhibitor (radicicol) and was mimicked by 8-Br-cGMP. We conclude that preconditioning with a low dose of NO donor accelerates repair and maintains endothelial integrity via a mechanism that includes the HSP90/sGC pathway. HSP90/sGC may thus play a role in the protective effects of NO-generating drugs from injurious stimuli.
- Published
- 2007
- Full Text
- View/download PDF
24. Effects of hsp90 binding inhibitors on sGC-mediated vascular relaxation.
- Author
-
Yetik-Anacak G, Xia T, Dimitropoulou C, Venema RC, and Catravas JD
- Subjects
- Animals, Aorta, Thoracic drug effects, Benzoquinones, Dose-Response Relationship, Drug, Guanylate Cyclase, In Vitro Techniques, Lactams, Macrocyclic, Male, Protein Binding drug effects, Rats, Rats, Sprague-Dawley, Soluble Guanylyl Cyclase, Vasodilation drug effects, Vasodilator Agents administration & dosage, Aorta, Thoracic physiology, HSP90 Heat-Shock Proteins antagonists & inhibitors, HSP90 Heat-Shock Proteins metabolism, Macrolides administration & dosage, Quinones administration & dosage, Receptors, Cytoplasmic and Nuclear metabolism, Vasodilation physiology
- Abstract
Vascular soluble guanylate cyclase (sGC) exists in multimeric complexes with endothelial nitric oxide (NO) synthase (eNOS) and heat shock protein 90 (hsp90). Whereas disruption of hsp90-eNOS complexes clearly attenuates eNOS-dependent vascular relaxation, the contribution of sGC-hsp90 complexes to eNOS- or NO donor-dependent relaxations remains unclear. Isolated rat thoracic aortic rings were preincubated with structurally diverse hsp90 binding inhibitors, radicicol (RA) or geldanamycin (GA), or vehicle for 0.5, 1, or 15 h. Preconstricted vessels were exposed to ACh, 8-bromo-cGMP (8-BrcGMP), forskolin, or one of three NO donors: nitroglycerin (NTG), sodium nitroprusside, or spermine NONOate (SNN). Both RA and GA inhibited endothelium-dependent relaxations dose dependently. Indomethacin or the antioxidant tiron did not affect the inhibition of ACh-induced relaxations by GA. Long-term (15 h) exposure to RA inhibited all NO donor-induced relaxations; however, GA inhibited SNN-induced relaxation only. The effects of GA and RA appeared to be selective because 15-h treatment with either agent did not affect forskolin-induced relaxations and only slightly decreased 8-BrcGMP-induced relaxations. Similarly to their effects on NO-donor-induced relaxation, 15-h exposure to RA, but not to GA, decreased hsp90-bound sGC protein expression and NTG-stimulated cGMP formation in aortic rings, whereas RA more than GA reduced SNN-stimulated cGMP formation. We conclude that RA, much more so than GA, selectively inhibits sGC-dependent relaxations of aortic rings by reducing sGC expression, disrupting sGC-hsp90 complex formation and decreasing cGMP formation. These studies suggest that hsp90 regulates both eNOS- and sGC-dependent relaxations.
- Published
- 2006
- Full Text
- View/download PDF
25. pH and nitric oxide synthase activity and expression in bovine aortic endothelial cells.
- Author
-
Nagy S, Harris MB, Ju H, Bhatia J, and Venema RC
- Subjects
- Animals, Aorta, Cattle, Cell Line, Cyclic GMP metabolism, Endothelium, Vascular enzymology, Immunoblotting, Endothelium, Vascular physiology, Hydrogen-Ion Concentration, Nitric Oxide Synthase metabolism
- Abstract
Aim: Nitric oxide (NO) plays an important role in the transition from intrauterine to extrauterine life. If this transition fails, a condition called persistent pulmonary hypertension of the neonate (PPHN) may develop. The current treatment modalities for this disease include induction of alkalosis by hyperventilation or alkali infusion, inhaled nitric oxide (iNO) and extracorporeal membrane oxygenation. There is evidence from animal studies that the elevated pH, not the low pCO2 is responsible for the resultant pulmonary vasodilatation. In this study, we examined the effect of pH on the activity and expression of endothelial nitric oxide synthase (eNOS) in cultured bovine aortic endothelial cells (BAEC) as a possible explanation for the pH dependent drop in pulmonary vascular resistance., Methods: BAEC were exposed to a pH gradient of 7.1-7.6 for 4 h (short-term) and 16 h (long-term). Standard Western blotting technique was used to detect expression of eNOS. Activity was measured by an indirect assay using bovine aortic smooth muscle cells (BASM) as reporter cells and measuring cGMP levels as a marker of NO production. The cells were exposed to the pH gradient for a total of 4 h and measurement were made at 30, 60 and 90 min, and 2, 3 and 4 hours., Results: eNOS activity and expression remained unchanged during the four and sixteen hours of exposure., Conclusion: In this in vitro experiment, we could not demonstrate an alkalosis-induced increase in eNOS activity and expression. The clinically observed pH dependent vasodilatation does not appear to be directly mediated through the induction of eNOS.
- Published
- 2006
- Full Text
- View/download PDF
26. Quercetin inhibits eNOS, microtubule polymerization, and mitotic progression in bovine aortic endothelial cells.
- Author
-
Jackson SJ and Venema RC
- Subjects
- Animals, Aorta, Arginine metabolism, Cattle, Cell Division drug effects, Cell Line, Citrulline metabolism, Endothelium, Vascular drug effects, Endothelium, Vascular enzymology, Microtubules drug effects, Nitric Oxide Synthase Type III drug effects, Nitric Oxide Synthase Type III genetics, Endothelium, Vascular cytology, Microtubules enzymology, Mitosis drug effects, Nitric Oxide Synthase Type III metabolism, Quercetin pharmacology
- Abstract
Quercetin (QRN), one of the most abundant flavonoids in the human diet, is a known antioxidant and inhibitor of cancer cell cycle progression. Here, we provide the first evidence that QRN inhibits angiogenesis via a mechanism involving both suppression of endothelial nitric oxide synthase (eNOS) and early M-phase cell cycle arrest. Bovine aortic endothelial (BAE) cells were exposed to doses of up to 100 micromol/L QRN and assayed for eNOS activity and phosphorylation status. Phosphorylation of eNOS at Ser 617 (bovine sequence) is thought to occur in response to Akt stimulation and to be required for eNOS activity. Together with basal eNOS activity, eNOS phosphorylation at Ser 617 and Akt Ser 473 phosphorylation were dose dependently and concomitantly suppressed by QRN within 30 min. Furthermore, although the significant (P < 0.05) inhibitory effect of a single 100 micromol/L QRN dose on eNOS activity was overcome within approximately 24 h, chronic QRN exposures (24-48 h) led to early M-phase arrest and disruption of mitotic microtubule polymerization. In vivo, QRN administered i.p. to female Balb/C mice bearing both syngeneic mammary tumors and Matrigel implants suppressed angiogenesis as measured by endothelial cell immunohistochemistry and hemoglobin concentration. Taken together, these findings suggest a dual mechanism by which QRN suppresses endothelial cell proliferation, both acutely via inhibition of eNOS Ser 617 phosphorylation, and chronically via perturbation of mitotic microtubule polymerization. This novel mechanism of QRN in endothelial cells may in part explain its inhibitory action on angiogenesis and further discern a potential role of QRN as a chemopreventive agent.
- Published
- 2006
- Full Text
- View/download PDF
27. Direct interaction of the cell division cycle 37 homolog inhibits endothelial nitric oxide synthase activity.
- Author
-
Harris MB, Bartoli M, Sood SG, Matts RL, and Venema RC
- Subjects
- Animals, Aorta, Cattle, Cells, Cultured, Endothelium, Vascular enzymology, Enzyme Activation, Enzyme Inhibitors, HSP90 Heat-Shock Proteins genetics, Nitric Oxide Synthase Type III antagonists & inhibitors, Phosphorylation, Recombinant Fusion Proteins pharmacology, Vascular Endothelial Growth Factor A pharmacology, HSP90 Heat-Shock Proteins physiology, Nitric Oxide Synthase Type III metabolism
- Abstract
Endothelial NO synthase (eNOS) via the production of NO in the endothelium plays a key role in cardiovascular biology and is tightly regulated by co- and posttranslational mechanisms, phosphorylation, and protein-protein interactions. The cell division cycle 37 homolog (Cdc37) is a key heat shock protein 90 (Hsp90) cochaperone for protein kinase clients, and Akt/Hsp90 interaction is dependent on Cdc37. Because both Hsp90 and Akt are key eNOS regulatory proteins, we hypothesized that Cdc37 interacts with eNOS as part of the regulatory complex. In the present study, we demonstrate by coimmunoprecipitation and affinity purification in bovine aortic endothelial cells (BAECs) that Cdc37 is complexed with eNOS, Hsp90, and Akt. In addition, cell fractionation data indicate that Cdc37 is found in caveolae with eNOS. Further analysis by in vitro binding assays reveals a direct interaction between purified Cdc37 and eNOS. Incubation of purified Cdc37 with purified wild-type eNOS decreases eNOS activity in vitro. Overexpression of wild-type Cdc37 in BAECs inhibits eNOS activity and NO release, whereas overexpression of S13A-Cdc37 mutant in BAECs increases eNOS activity and NO release. Taken together, these data suggest that Cdc37 has a direct regulatory interaction with eNOS and may play an important role in mediating the eNOS protein complex formation as well as subsequent eNOS phosphorylation and activation.
- Published
- 2006
- Full Text
- View/download PDF
28. Src kinase activates endothelial nitric-oxide synthase by phosphorylating Tyr-83.
- Author
-
Fulton D, Church JE, Ruan L, Li C, Sood SG, Kemp BE, Jennings IG, and Venema RC
- Subjects
- Animals, COS Cells, Calcium metabolism, Cattle, Cells, Cultured, Chlorocebus aethiops, Immunoblotting, Immunoprecipitation, Inflammation, Mutagenesis, Site-Directed, Nitric Oxide metabolism, Phosphorylation, Protein Structure, Tertiary, Pyrimidines pharmacology, Time Factors, Transfection, Tyrosine metabolism, src-Family Kinases metabolism, Nitric Oxide Synthase Type III metabolism, Tyrosine chemistry, src-Family Kinases physiology
- Abstract
The endothelial nitric-oxide synthase (eNOS) is regulated in part by serine/threonine phosphorylation, but eNOS tyrosine phosphorylation is less well understood. In the present study we have examined the tyrosine phosphorylation of eNOS in bovine aortic endothelial cells (BAECs) exposed to oxidant stress. Hydrogen peroxide and pervanadate (PV) treatment stimulates eNOS tyrosine phosphorylation in BAECs. Phosphorylation is blocked by the Src kinase family inhibitor, 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2). Moreover, eNOS and c-Src can be coimmunoprecipitated from BAEC lysates by antibodies directed against either protein. Domain mapping and site-directed mutagenesis studies in COS-7 cells transfected with either eNOS alone and then treated with PV or cotransfected with eNOS and constitutively active v-Src identified Tyr-83 (bovine sequence) as the major eNOS tyrosine phosphorylation site. Tyr-83 phosphorylation is associated with a 3-fold increase in basal NO release from cotransfected cells. Furthermore, the Y83F eNOS mutation attenuated thapsigargin-stimulated NO production. Taken together, these data indicate that Src-mediated tyrosine phosphorylation of eNOS at Tyr-83 modulates eNOS activity in endothelial cells.
- Published
- 2005
- Full Text
- View/download PDF
29. Interaction between the 90-kDa heat shock protein and soluble guanylyl cyclase: physiological significance and mapping of the domains mediating binding.
- Author
-
Papapetropoulos A, Zhou Z, Gerassimou C, Yetik G, Venema RC, Roussos C, Sessa WC, and Catravas JD
- Subjects
- Animals, Blotting, Western, Cattle, Dimerization, Guanylate Cyclase, HSP90 Heat-Shock Proteins antagonists & inhibitors, HSP90 Heat-Shock Proteins chemistry, Immunoenzyme Techniques, Immunoprecipitation, Muscle, Smooth metabolism, Nitric Oxide metabolism, Protein Binding, Rats, Receptors, Cytoplasmic and Nuclear chemistry, Receptors, Cytoplasmic and Nuclear genetics, Signal Transduction, Soluble Guanylyl Cyclase, HSP90 Heat-Shock Proteins metabolism, Receptors, Cytoplasmic and Nuclear metabolism
- Abstract
The 90-kDa heat shock protein (hsp90) regulates the stability and function of many client proteins, including members of the NO-cGMP signaling pathway. Soluble guanylyl cyclase (sGC), an NO receptor, was recently reported to be an hsp90-interacting partner. In the present study, we show that hsp90 binds to both subunits of the most common sGC form (alpha(1)beta(1)) when these are expressed individually but only interacts with beta(1) in the heterodimeric form of the enzyme. Characterization of the region of hsp90 required to bind each subunit in immunoprecipitation experiments revealed that residues 310 to 456 of hsp90 interact with the sGC subunits. The region of beta(1) responsible for binding to hsp90beta was mapped using in vitro binding assays and immunoprecipitation experiments and was found to lie in the regulatory domain. The physiological importance of the hsp90/sGC interaction was investigated by treating rat smooth muscle cells with the hsp90 inhibitors radicicol and geldanamycin (GA) and determining both sGC activity and protein levels. Long-term (24 or 48 h) inhibition of hsp90 resulted in a strong decrease of both alpha(1) and beta(1) protein levels and sGC activity. Moreover, incubation of smooth muscle cells with the proteasome inhibitor N-benzoyloxycarbonyl (Z)-Leu-Leu-leucinal (MG132) blocked the GA-induced down-regulation of sGC. We conclude that the N-terminal region of the beta(1) subunit mediates binding of the heterodimeric form of sGC to hsp90 and that this interaction involves the M domain of hsp90. Hsp90 binding to sGC regulates the pool of active enzymes by affecting the protein levels of the two subunits.
- Published
- 2005
- Full Text
- View/download PDF
30. Endostatin induces acute endothelial nitric oxide and prostacyclin release.
- Author
-
Li C, Harris MB, Venema VJ, and Venema RC
- Subjects
- Animals, Cattle, Cells, Cultured, Nitric Oxide Synthase Type III, Endostatins pharmacology, Endothelial Cells drug effects, Endothelial Cells metabolism, Epoprostenol metabolism, Nitric Oxide metabolism, Nitric Oxide Synthase metabolism
- Abstract
Chronic exposure to endostatin (ES) blocks endothelial cell (EC) proliferation, and migration and induces EC apoptosis thereby inhibiting angiogenesis. Nitric oxide (NO) and prostacyclin (PGI(2)), in contrast, play important roles in promoting angiogenesis. In this study, we examined the acute effects of ES on endothelial NO and PGI(2) production. Unexpectedly, a cGMP reporter cell assay showed that ES-induced acute endothelial NO release in cultured bovine aortic endothelial cells (BAECs). Enzyme immunoassay showed that ES also induced an acute increase in PGI(2) production in BAECs. These results were confirmed by ex vivo vascular ring studies that showed vascular relaxation in response to ES. Immunoblot analysis showed that ES stimulated acute phosphorylation of endothelial nitric oxide synthase (eNOS) at Ser116, Ser617, Ser635, and Ser1179, and dephosphorylation at Thr497 in BAECs, events associated with eNOS activation. Short-term exposure of EC to ES, therefore, unlike long-term exposure which is anti-angiogenic, may be pro-angiogenic.
- Published
- 2005
- Full Text
- View/download PDF
31. Interaction of the endothelial nitric oxide synthase with the CAT-1 arginine transporter enhances NO release by a mechanism not involving arginine transport.
- Author
-
Li C, Huang W, Harris MB, Goolsby JM, and Venema RC
- Subjects
- Adenoviridae genetics, Animals, Aorta cytology, Biological Transport drug effects, Bradykinin pharmacology, Cationic Amino Acid Transporter 1 genetics, Cationic Amino Acid Transporter 1 immunology, Cattle, Caveolin 1, Caveolins metabolism, Cells, Cultured, Endothelial Cells drug effects, Endothelial Cells enzymology, Endothelial Cells metabolism, Glycosylation, Immune Sera immunology, Immunoprecipitation, Lysine pharmacology, Mice, Nitric Oxide biosynthesis, Nitric Oxide Synthase Type II, Nitric Oxide Synthase Type III, Phosphorylation drug effects, Protein Binding, Transduction, Genetic, Arginine metabolism, Cationic Amino Acid Transporter 1 metabolism, Nitric Oxide metabolism, Nitric Oxide Synthase metabolism
- Abstract
eNOS (endothelial nitric oxide synthase) catalyses the conversion of L-arginine into L-citrulline and NO. Evidence has been presented previously that eNOS is associated with the CAT (cationic amino acid transporter)-1 arginine transporter in endothelial caveolae, and it has been proposed that eNOS-CAT-1 association facilitates the delivery of extracellular L-arginine to eNOS. Definitive proof of a protein-protein interaction between eNOS and CAT-1 is lacking, however, and it is also unknown whether the two proteins interact directly or via an adaptor protein. In the present study, we raised a polyclonal antibody against CAT-1, and show using reciprocal co-immunoprecipitation protocols that eNOS and CAT-1 do indeed form a complex in BAECs (bovine aortic endothelial cells). In vitro binding assays with GST (glutathione S-transferase)-CAT-1 fusion proteins and eNOS show that the two proteins interact directly and that no single CAT-1 intracellular domain is sufficient to mediate the interaction. Overexpression of CAT-1 in BAECs by adenoviral-mediated gene transfer results in significant increases in both L-arginine uptake and NO production by the cells. However, whereas increased L-arginine transport is reversed completely by the CAT-1 inhibitor, L-lysine, increased NO release is unaltered, suggesting that NO production in this in vitro model is independent of CAT-1-mediated transport. Furthermore, eNOS enzymic activity is increased in lysates of CAT-1-overexpressing cells accompanied by increased phosphorylation of eNOS at Ser-1179 and Ser-635, and decreased association of eNOS with caveolin-1. Taken together, these data suggest that direct interaction of eNOS with CAT-1 enhances NO release by a mechanism not involving arginine transport.
- Published
- 2005
- Full Text
- View/download PDF
32. Insulin resistance does not diminish eNOS expression, phosphorylation, or binding to HSP-90.
- Author
-
Fulton D, Harris MB, Kemp BE, Venema RC, Marrero MB, and Stepp DW
- Subjects
- Animals, Endothelium, Vascular physiology, Gene Expression Regulation, Enzymologic physiology, In Vitro Techniques, Nitric Oxide blood, Nitric Oxide Synthase Type III, Phosphorylation, Protein Processing, Post-Translational physiology, RNA, Messenger analysis, Rats, Rats, Zucker, Vasodilation physiology, HSP90 Heat-Shock Proteins metabolism, Insulin Resistance physiology, Nitric Oxide Synthase genetics, Nitric Oxide Synthase metabolism
- Abstract
Previously, using an animal model of syndrome X, the obese Zucker rat (OZR), we documented impaired endothelium-dependent vasodilation. The aim of this study was to determine whether reduced expression or altered posttranslational regulation of endothelial nitric oxide synthase (eNOS) underlies the vascular dysfunction in OZR rats. There was no significant difference in the relative abundance of eNOS in hearts, aortas, or skeletal muscle between lean Zucker rats (LZR) and OZR regardless of age. There was no difference in eNOS mRNA levels, as determined by real-time PCR, between LZR and OZR. The inability of insulin resistance to modulate eNOS expression was also documented in two additional in vivo models, the ob/ob mouse and the fructose-fed rat, and in vitro via adenoviral expression of protein tyrosine phosphatase 1B in endothelial cells. We next investigated whether changes in the acute posttranslational regulation of eNOS occurs with insulin resistance. Phosphorylation of eNOS at S632 (human S633) and T494 was not different between LZR and OZR; however, phosphorylation of S1176 was significantly enhanced in OZR. Phosphorylation of S1176 was not different in the ob/ob mouse or in fructose-fed rats. The association of heat shock protein 90 with eNOS, a key regulatory step controlling nitric oxide and aberrant O2- production, was not different between OZR and LZR. Taken together, these results suggest that changes in eNOS expression or posttranslation regulation do not underlie the vascular dysfunction seen with insulin resistance and that other mechanisms, such as altered localization, reduced availability of cofactors, substrates, and the elevated production of O2-, may be responsible.
- Published
- 2004
- Full Text
- View/download PDF
33. Acute activation and phosphorylation of endothelial nitric oxide synthase by HMG-CoA reductase inhibitors.
- Author
-
Harris MB, Blackstone MA, Sood SG, Li C, Goolsby JM, Venema VJ, Kemp BE, and Venema RC
- Subjects
- Animals, Aorta, Cattle, Cells, Cultured, Endothelium, Vascular metabolism, Enzyme Activation, Nitric Oxide metabolism, Nitric Oxide Synthase Type III, Endothelium, Vascular enzymology, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Nitric Oxide Synthase metabolism
- Abstract
3-Hydroxy-3-methylglutaryl (HMG)-CoA reductase inhibitors, statins, provide beneficial effects independent of their lipid-lowering effects. One beneficial effect appears to involve acute activation of endothelial nitric oxide (NO) synthase (eNOS) and increased NO release. However, the mechanism of acute statin-stimulated eNOS activation is unknown. Therefore, we hypothesized that eNOS activation may be coupled to altered eNOS phosphorylation. Bovine aortic endothelial cells (BAECs), passages 2-6, were treated with either lovastatin or pravastatin from 0 to 30 min. eNOS phosphorylation was examined by Western blot by use of phosphospecific antibodies for Ser-1179, Ser-635, Ser-617, Thr-497, and Ser-116. Statin stimulation of BAECs increased eNOS phosphorylation at Ser-1179 and Ser-617, which was blocked by the phosphatidylinositol 3-kinase (PI3-kinase)/Akt inhibitor wortmannin, and at Ser-635, which was blocked by the protein kinase A (PKA) inhibitor KT-5720. Statin treatment of BAECs transiently increased NO release by fourfold, measured by cGMP accumulation, and was attenuated by N-nitro-l-arginine methyl ester, wortmannin, and KT-5720 but not by mevalonate. In conclusion, these data demonstrate that eNOS is acutely activated by statins independent of HMG-CoA reductase inhibition and that in addition to Ser-1179, eNOS phosphorylation at Ser-635 and Ser-617 through PKA and Akt, respectively, may explain, in part, a mechanism by which eNOS is activated in response to acute statin treatment.
- Published
- 2004
- Full Text
- View/download PDF
34. Novel complexes of guanylate cyclase with heat shock protein 90 and nitric oxide synthase.
- Author
-
Venema RC, Venema VJ, Ju H, Harris MB, Snead C, Jilling T, Dimitropoulou C, Maragoudakis ME, and Catravas JD
- Subjects
- Animals, Aorta cytology, Benzoquinones, Cattle, Cells, Cultured, Endothelium, Vascular cytology, Enzyme Inhibitors pharmacology, Lactams, Macrocyclic, Muscle, Smooth, Vascular cytology, Nitric Oxide Donors pharmacology, Nitric Oxide Synthase Type II, Nitric Oxide Synthase Type III, Nitroprusside pharmacology, Quinones pharmacology, Rats, Rats, Sprague-Dawley, Signal Transduction drug effects, Signal Transduction physiology, Endothelium, Vascular enzymology, Guanylate Cyclase metabolism, HSP90 Heat-Shock Proteins metabolism, Muscle, Smooth, Vascular enzymology, Nitric Oxide Synthase metabolism
- Abstract
Soluble guanylate cyclase (sGC) is an important downstream intracellular target of nitric oxide (NO) that is produced by endothelial NO synthase (eNOS) and inducible NO synthase (iNOS). In this study, we demonstrate that sGC exists in a complex with eNOS and heat shock protein 90 (HSP90) in aortic endothelial cells. In addition, we show that in aortic smooth muscle cells, sGC forms a complex with HSP90. Formation of the sGC/eNOS/HSP90 complex is increased in response to eNOS-activating agonists in a manner that depends on HSP90 activity. In vitro binding assays with glutathione S-transferase fusion proteins that contain the alpha- or beta-subunit of sGC show that the sGC beta-subunit interacts directly with HSP90 and indirectly with eNOS. Confocal immunofluorescent studies confirm the subcellular colocalization of sGC and HSP90 in both endothelial and smooth muscle cells. Complex formation of sGC with HSP90 facilitates responses to NO donors in cultured cells (cGMP accumulation) as well as in anesthetized rats (hypotension). These complexes likely function to stabilize sGC as well as to provide directed intracellular transfer of NO from NOS to sGC, thus preventing inactivation of NO by superoxide anion and formation of peroxynitrite, which is a toxic molecule that has been implicated in the pathology of several vascular diseases.
- Published
- 2003
- Full Text
- View/download PDF
35. Heat-induced increases in endothelial NO synthase expression and activity and endothelial NO release.
- Author
-
Harris MB, Blackstone MA, Ju H, Venema VJ, and Venema RC
- Subjects
- Animals, Cattle, Cell Survival physiology, Cells, Cultured, Coloring Agents, Endothelium, Vascular enzymology, HSP70 Heat-Shock Proteins biosynthesis, HSP90 Heat-Shock Proteins biosynthesis, Immunoblotting, In Vitro Techniques, Male, Muscle Contraction drug effects, Muscle, Smooth, Vascular physiopathology, Nitric Oxide Synthase Type III, Phenylephrine pharmacology, Precipitin Tests, Rats, Rats, Sprague-Dawley, Trypan Blue, Vasoconstrictor Agents pharmacology, Endothelium, Vascular metabolism, Endothelium, Vascular physiology, Gene Expression Regulation, Enzymologic physiology, Heat-Shock Response physiology, Nitric Oxide metabolism, Nitric Oxide Synthase biosynthesis, Nitric Oxide Synthase metabolism
- Abstract
Endothelial nitric oxide (NO) synthase (eNOS) is regulated by heat shock protein 90 (HSP90), a heat-inducible protein; however, the effect of heat shock on eNOS expression and eNO release is unknown. Bovine aortic endothelial cells were incubated for 1 h at 37 degrees C, 42 degrees C, or 45 degrees C and cell lysates were evaluated with the use of Western blotting. We observed a 2.1 +/- 0.1-fold increase in eNOS protein content, but no change in HSP90 content, HSP70 content, or HSP90/eNOS association, 24 h after heat shock at 42 degrees C. We also observed a 7.7 +/- 1.5-fold increase in HSP70 protein content, but did not observe a change in eNOS or HSP90 24 h after heat shock at 45 degrees C. eNOS activity and maximal bradykinin-stimulated NO release was significantly increased 24 h after heat shock at 42 degrees C. Heat shock in rats (core temperature: 42 degrees C, 15 min) resulted in a significant increase in aortic eNOS, HSP90, and HSP70 protein content. The aorta from heat-shocked rats exhibited a decreased maximal contractile response to phenylephrine, which was abolished by preincubation with NG-nitro-l-arginine. We conclude that prior heat shock is a physical stimulus of increased eNOS expression and is associated with an increase in eNOS activity, agonist-stimulated NO release, and a decreased vasoconstrictor response.
- Published
- 2003
- Full Text
- View/download PDF
36. Pharmacological interference with dimerization of human neuronal nitric-oxide synthase expressed in adenovirus-infected DLD-1 cells.
- Author
-
Habisch HJ, Gorren AC, Liang H, Venema RC, Parkinson JF, Schmidt K, and Mayer B
- Subjects
- Adenoviridae physiology, Animals, COS Cells, Cells, Cultured virology, Dimerization, Humans, Nitric Oxide Synthase chemistry, Nitric Oxide Synthase Type I, Nitric Oxide Synthase metabolism
- Abstract
A recombinant adenovirus containing the cDNA of human neuronal nitric-oxide synthase (nNOS) was constructed to characterize the interaction of nNOS with N-[(1,3-benzodioxol-5-yl)methyl]-1-[2-(1H-imidazole-1-yl)pyrimidin-4-yl]-4-(methoxycarbonyl)-piperazine-2-acetamide (BBS-1), a potent inhibitor of inducible NOS dimerization [Proc Natl Acad Sci USA 97:1506-1511, 2000]. BBS-1 inhibited de novo expression of nNOS activity in virus-infected cells at a half-maximal concentration (IC(50)) of 40 +/- 10 nM in a reversible manner. Low-temperature gel electrophoresis showed that BBS-1 attenuated the formation of SDS-resistant nNOS dimers with an IC(50) of 22 +/- 5.2 nM. Enzyme inhibition progressively decreased with increasing time of addition after infection. BBS-1 did not significantly inhibit dimeric nNOS activity (IC(50) > 1 mM). Long-term incubation with BBS-1 of human embryonic kidney cells stably transfected with nNOS or endothelial NOS revealed a slow time- and concentration-dependent decrease of NOS activity with half-lives of 30 and 43 h and IC(50) values of 210 +/- 30 nM and 12 +/- 0.5 microM, respectively. These results establish that BBS-1 interferes with the assembly of active nNOS dimers during protein expression. Slow inactivation of constitutively expressed NOS in intact cells may reflect protein degradation and interference of BBS-1 with the de novo synthesis of functionally active NOS dimers. As time-dependent inhibitors of NOS dimerization, BBS-1 and related compounds provide a promising strategy to develop a new class of selective and clinically useful NOS inhibitors.
- Published
- 2003
- Full Text
- View/download PDF
37. Post-translational mechanisms of endothelial nitric oxide synthase regulation by bradykinin.
- Author
-
Venema RC
- Subjects
- Animals, Bradykinin metabolism, Caveolin 1, Caveolins metabolism, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, HSP90 Heat-Shock Proteins metabolism, Humans, Phosphorylation, Protein Binding, Receptor, Bradykinin B2, Receptors, Bradykinin metabolism, Bradykinin physiology, Endothelium, Vascular enzymology, Nitric Oxide Synthase metabolism, Protein Processing, Post-Translational
- Abstract
The endothelial nitric oxide synthase (eNOS) plays a key role in blood pressure regulation and vascular homeostasis. Among the more potent inducers of eNOS activity in vascular endothelial cells is bradykinin (BK). This brief review summarizes the current state of knowledge with regard to regulation of eNOS through several distinct molecular mechanisms, each of which acts in concert with Ca2+-calmodulin (CaM) signaling in post-translational activation of eNOS. These mechanisms include alterations in protein-protein interactions with caveolin-1, the BK B2 receptor, and heat shock protein 90 (Hsp90). In addition, BK stimulates an increase in eNOS activity through phosphorylation of the enzyme at three specific amino acid residues as well as through dephosphorylation at a fourth residue.
- Published
- 2002
- Full Text
- View/download PDF
38. Identification of regulatory sites of phosphorylation of the bovine endothelial nitric-oxide synthase at serine 617 and serine 635.
- Author
-
Michell BJ, Harris MB, Chen ZP, Ju H, Venema VJ, Blackstone MA, Huang W, Venema RC, and Kemp BE
- Subjects
- Adenosine Triphosphate pharmacology, Amino Acid Sequence, Animals, Bradykinin pharmacology, Calcium physiology, Calmodulin physiology, Cattle, Cyclic AMP-Dependent Protein Kinases metabolism, Endothelial Growth Factors pharmacology, Humans, Intercellular Signaling Peptides and Proteins pharmacology, Lymphokines pharmacology, Molecular Sequence Data, Nitric Oxide Synthase Type III, Phosphorylation, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-akt, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Nitric Oxide Synthase chemistry, Protein Serine-Threonine Kinases, Serine metabolism
- Abstract
Endothelial nitric-oxide synthase (eNOS) is regulated by signaling pathways involving multiple sites of phosphorylation. The coordinated phosphorylation of eNOS at Ser(1179) and dephosphorylation at Thr(497) activates the enzyme, whereas inhibition results when Thr(497) is phosphorylated and Ser(1179) is dephosphorylated. We have identified two further phosphorylation sites, at Ser(617) and Ser(635), by phosphopeptide mapping and matrix-assisted laser desorption ionization time of flight mass spectrometry. Purified protein kinase A (PKA) phosphorylates both sites in purified eNOS, whereas purified Akt phosphorylates only Ser(617). In bovine aortic endothelial cells, bradykinin (BK), ATP, and vascular endothelial growth factor stimulate phosphorylation of both sites. BK-stimulated phosphorylation of Ser(617) is Ca(2+)-dependent and is partially inhibited by LY294002 and wortmannin, phosphatidylinositol 3-kinase inhibitors, suggesting signaling via Akt. BK-stimulated phosphorylation of Ser(635) is Ca(2+)-independent and is completely abolished by the PKA inhibitor, KT5720, suggesting signaling via PKA. Activation of PKA with isobutylmethylxanthine also causes Ser(635), but not Ser(617), phosphorylation. Mimicking phosphorylation at Ser(635) by Ser to Asp mutation results in a greater than 2-fold increase in activity of the purified protein, whereas mimicking phosphorylation at Ser(617) does not alter maximal activity but significantly increases Ca(2+)-calmodulin sensitivity. These data show that phosphorylation of both Ser(617) and Ser(635) regulates eNOS activity and contributes to the agonist-stimulated eNOS activation process.
- Published
- 2002
- Full Text
- View/download PDF
39. Reciprocal phosphorylation and regulation of endothelial nitric-oxide synthase in response to bradykinin stimulation.
- Author
-
Harris MB, Ju H, Venema VJ, Liang H, Zou R, Michell BJ, Chen ZP, Kemp BE, and Venema RC
- Subjects
- Androstadienes pharmacology, Animals, Aorta, Calcineurin Inhibitors, Cattle, Chromones pharmacology, Cyclosporine pharmacology, Enzyme Activation, Enzyme Inhibitors pharmacology, Kinetics, Morpholines pharmacology, Nitric Oxide Synthase Type III, Phosphorylation, Phosphoserine metabolism, Phosphothreonine metabolism, Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins c-akt, Wortmannin, Bradykinin pharmacology, Endothelium, Vascular enzymology, Nitric Oxide Synthase metabolism, Protein Serine-Threonine Kinases, Proto-Oncogene Proteins metabolism
- Abstract
Endothelial nitric-oxide synthase (eNOS) is phosphorylated at Ser-1179 (bovine sequence) by Akt after growth factor or shear stress stimulation of endothelial cells, resulting in increased eNOS activity. Purified eNOS is also phosphorylated at Thr-497 by purified AMP-activated protein kinase, resulting in decreased eNOS activity. We investigated whether bradykinin (BK) stimulation of bovine aortic endothelial cells (BAECs) regulates eNOS through Akt activation and Ser-1179 or Thr-497 phosphorylation. Akt is transiently activated in BK-stimulated BAECs. Activation is blocked completely by wortmannin and LY294002, inhibitors of phosphatidylinositol 3-kinase, suggesting that Akt activation occurs downstream from phosphatidylinositol 3-kinase. BK stimulates a transient phosphorylation of eNOS at Ser-1179 that is correlated temporally with a transient dephosphorylation of eNOS at Thr-497. Phosphorylation at Ser-1179, but not dephosphorylation at Thr-497, is blocked by wortmannin and LY294002. BK also stimulates a transient nitric oxide (NO) release from BAECs with a time-course similar to Ser-1179 phosphorylation and Thr-497 dephosphorylation. NO release is not altered by wortmannin. BK-stimulated dephosphorylation of Thr-497 and NO release are blocked by the calcineurin inhibitor, cyclosporin A. These data suggest that BK activation of eNOS in BAECs primarily involves deinhibition of the enzyme through calcineurin-mediated dephosphorylation at Thr-497.
- Published
- 2001
- Full Text
- View/download PDF
40. Vascular endothelial growth factor activates STAT proteins in aortic endothelial cells.
- Author
-
Bartoli M, Gu X, Tsai NT, Venema RC, Brooks SE, Marrero MB, and Caldwell RB
- Subjects
- Animals, Aorta drug effects, Aorta metabolism, Cattle, Cells, Cultured, Endothelium, Vascular metabolism, Janus Kinase 1, Phosphorylation, Protein-Tyrosine Kinases physiology, Receptor Protein-Tyrosine Kinases physiology, Receptors, Growth Factor physiology, Receptors, Vascular Endothelial Growth Factor, STAT1 Transcription Factor, STAT3 Transcription Factor, STAT6 Transcription Factor, Tyrosine metabolism, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, DNA-Binding Proteins metabolism, Endothelial Growth Factors pharmacology, Endothelium, Vascular drug effects, Lymphokines pharmacology, Trans-Activators metabolism
- Abstract
Vascular endothelial growth factor (VEGF) intracellular signaling in endothelial cells is initiated by the activation of distinct tyrosine kinase receptors, VEGFR1 (Flt-1) and VEGFR2 (Flk-1/KDR). Because the tyrosine kinase-dependent transcription factors known as STAT (signal transducers and activators of transcription) proteins are important modulators of cell growth responses induced by other growth factor receptors, we have determined the effects VEGF of on STAT activation in BAEC (bovine aortic endothelial cells). Here, we show that VEGF induces tyrosine phosphorylation and nuclear translocation of STAT1 and STAT6. VEGF also stimulates STAT3 tyrosine phosphorylation, but nuclear translocation does not occur. We found that placenta growth factor, which selectively activates VEGFR1, has no effect on the STATs. However, upon VEGF stimulation, STAT1 associates with the VEGFR2 in a tyrosine kinase-dependent manner, indicating that VEGF-induced STAT1 activation is mediated primarily by VEGFR2. Thus, our study shows for the first time that VEGF activates the STAT pathway through VEGFR2. Because the growth-promoting activity of VEGF depends upon VEGFR2 activation, these findings suggest a role for the STATs in the regulation of gene expression associated with the angiogenic effects of VEGF.
- Published
- 2000
- Full Text
- View/download PDF
41. Bradykinin activates the Janus-activated kinase/signal transducers and activators of transcription (JAK/STAT) pathway in vascular endothelial cells: localization of JAK/STAT signalling proteins in plasmalemmal caveolae.
- Author
-
Ju H, Venema VJ, Liang H, Harris MB, Zou R, and Venema RC
- Subjects
- Active Transport, Cell Nucleus drug effects, Animals, Aorta, Cattle, Caveolae metabolism, Caveolin 1, Caveolins metabolism, Cell Nucleus drug effects, Cell Nucleus metabolism, Cells, Cultured, Endothelium, Vascular cytology, Endothelium, Vascular enzymology, Endothelium, Vascular metabolism, Enzyme Activation drug effects, Mitogen-Activated Protein Kinases metabolism, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular enzymology, Muscle, Smooth, Vascular metabolism, Phosphorylation drug effects, Phosphoserine metabolism, Phosphotyrosine metabolism, Protein Binding drug effects, Receptor, Bradykinin B2, Receptors, Bradykinin metabolism, STAT3 Transcription Factor, Bradykinin pharmacology, Caveolae drug effects, DNA-Binding Proteins metabolism, Endothelium, Vascular drug effects, MAP Kinase Signaling System drug effects, Protein-Tyrosine Kinases, Proteins metabolism, Trans-Activators metabolism
- Abstract
Bradykinin (BK) is an important physiological regulator of endothelial cell function. In the present study, we have examined the role of the Janus-activated kinase (JAK)/signal transducers and activators of transcription (STAT) pathway in endothelial signal transduction through the BK B2 receptor (B2R). In cultured bovine aortic endothelial cells (BAECs), BK activates Tyk2 of the JAK family of tyrosine kinases. Activation results in the tyrosine phosphorylation and subsequent nuclear translocation of STAT3. BK also activates the mitogen-activated p44 and p42 protein kinases, resulting in STAT3 serine phosphorylation. Furthermore, Tyk2 and STAT3 form a complex with the B2R in response to BK stimulation. Under basal conditions, Tyk2, STAT3 and the B2R are localized either partially or entirely in endothelial plasmalemmal caveolae. Following BK stimulation of BAECs, however, the B2R and STAT3 are translocated out of caveolae. Taken together, these data suggest that BK activates the JAK/STAT pathway in endothelial cells and that JAK/STAT signalling proteins are localized in endothelial caveolae. Moreover, caveolar localization of the B2R and STAT3 appears to be regulated in an agonist-dependent manner.
- Published
- 2000
- Full Text
- View/download PDF
42. Role of heat shock protein 90 in bradykinin-stimulated endothelial nitric oxide release.
- Author
-
Harris MB, Ju H, Venema VJ, Blackstone M, and Venema RC
- Subjects
- Animals, Aorta cytology, Aorta drug effects, Aorta metabolism, Benzoquinones, Cattle, Cells, Cultured, Endothelium, Vascular metabolism, Enzyme Inhibitors, HSP90 Heat-Shock Proteins antagonists & inhibitors, HSP90 Heat-Shock Proteins metabolism, Humans, Insecta, Lactams, Macrocyclic, Nitric Oxide Synthase metabolism, Nitric Oxide Synthase Type III, Protein Binding, Protein Structure, Tertiary, Quinones pharmacology, Bradykinin pharmacology, Endothelium, Vascular physiology, HSP90 Heat-Shock Proteins physiology, Nitric Oxide metabolism
- Abstract
Previously we described ENAP-1, a 90-kDa protein that is tyrosine-phosphorylated in endothelial cells in response to bradykinin (BK) stimulation and is associated with endothelial nitric oxide synthase (eNOS). Subsequently, other investigators demonstrated that eNOS interacts with heat shock protein 90 (Hsp90) following stimulation of endothelial cells with vascular endothelial growth factor (VEGF), histamine, or fluid shear stress. Therefore, we tested the hypotheses that ENAP-1 and Hsp90 are the same protein and that BK activation of eNOS is dependent on Hsp90. Immunoblotting of immunoprecipitated Hsp90 with anti-phosphotyrosine antibody shows that Hsp90 is tyrosine-phosphorylated in response to BK stimulation of bovine aortic endothelial cells (BAECs). Coimmunoprecipitation of Hsp90 with anti-eNOS antibody reveals a Hsp90-eNOS complex in endothelial cells under basal conditions that is increased following BK stimulation. Taken together with the tyrosine phosphorylation data, these data suggest that ENAP-1 is Hsp90. BK-stimulated nitric oxide (NO) release is completely blocked by pretreatment with geldanamycin, a specific inhibitor of Hsp90, illustrating the importance of the Hsp90-eNOS interaction. In vitro binding assays with Hsp90-glutathione-S-transferase fusion proteins show direct binding of eNOS with the middle domain (residues 259-615) of Hsp90.
- Published
- 2000
- Full Text
- View/download PDF
43. Ascorbic acid enhances endothelial nitric-oxide synthase activity by increasing intracellular tetrahydrobiopterin.
- Author
-
Huang A, Vita JA, Venema RC, and Keaney JF Jr
- Subjects
- Animals, Aorta, Arginine metabolism, Atrial Natriuretic Factor pharmacology, Biopterins metabolism, Biopterins pharmacology, Calcimycin pharmacology, Cattle, Cell Line, Cells, Cultured, Cyclic GMP metabolism, Endothelium, Vascular drug effects, Kinetics, Nitric Oxide metabolism, Nitric Oxide Synthase Type III, Nitroprusside pharmacology, Pteridines pharmacology, Recombinant Proteins metabolism, Spodoptera, Swine, Transfection, Ascorbic Acid pharmacology, Biopterins analogs & derivatives, Endothelium, Vascular metabolism, Nitric Oxide Synthase metabolism, Pterins
- Abstract
Ascorbic acid enhances NO bioactivity in patients with vascular disease through unclear mechanism(s). We investigated the role of intracellular ascorbic acid in endothelium-derived NO bioactivity. Incubation of porcine aortic endothelial cells (PAECs) with ascorbic acid produced time- and dose-dependent intracellular ascorbic acid accumulation that enhanced NO bioactivity by 70% measured as A23187-induced cGMP accumulation. This effect was due to enhanced NO production because ascorbate stimulated both PAEC nitrogen oxide (NO(2)(-) + NO(3)(-)) production and l-arginine to l-citrulline conversion by 59 and 72%, respectively, without altering the cGMP response to authentic NO. Ascorbic acid also stimulated the catalytic activity of eNOS derived from either PAEC membrane fractions or baculovirus-infected Sf9 cells. Ascorbic acid enhanced bovine eNOS V(max) by approximately 50% without altering the K(m) for l-arginine. The effect of ascorbate was tetrahydrobiopterin (BH(4))-dependent, because ascorbate was ineffective with BH(4) concentrations >10 microm or in PAECs treated with sepiapterin to increase intracellular BH(4). The effect of ascorbic acid was also specific because A23187-stimulated cGMP accumulation in PAECs was insensitive to intracellular glutathione manipulation and only ascorbic acid, not glutathione, increased the intracellular concentration of BH(4). These data suggest that ascorbic acid enhances NO bioactivity in a BH(4)-dependent manner by increasing intracellular BH(4) content.
- Published
- 2000
- Full Text
- View/download PDF
44. Interaction of endothelial and neuronal nitric-oxide synthases with the bradykinin B2 receptor. Binding of an inhibitory peptide to the oxygenase domain blocks uncoupled NADPH oxidation.
- Author
-
Golser R, Gorren AC, Leber A, Andrew P, Habisch HJ, Werner ER, Schmidt K, Venema RC, and Mayer B
- Subjects
- Animals, Binding Sites, Blotting, Western, Cell Line, Citrulline metabolism, Dose-Response Relationship, Drug, Humans, Kinetics, Nitric Oxide Synthase antagonists & inhibitors, Peptides metabolism, Precipitin Tests, Protein Binding, Rats, Receptor, Bradykinin B2, Recombinant Proteins metabolism, Time Factors, Endothelium, Vascular enzymology, Neurons enzymology, Nitric Oxide Synthase metabolism, Receptors, Bradykinin metabolism
- Abstract
Endothelial nitric-oxide synthase (type III) (eNOS) was reported to form an inhibitory complex with the bradykinin receptor B2 (B2R) from which the enzyme is released in an active form upon receptor activation (Ju, H., Venema, V. J., Marrero, M. B., and Venema, R. C. (1998) J. Biol. Chem. 273, 24025-24029). Using a synthetic peptide derived from the known inhibitory sequence of the B2R (residues 310-329) we studied the interaction of the receptor with purified eNOS and neuronal nitric-oxide synthase (type I) (nNOS). The peptide inhibited formation of L-citrulline by eNOS and nNOS with IC(50) values of 10.6 +/- 0.4 microM and 7.1 +/- 0.6 microM, respectively. Inhibition was not due to an interference of the peptide with L-arginine or tetrahydrobiopterin binding. The NADPH oxidase activity of nNOS measured in the absence of L-arginine was inhibited by the peptide with an IC(50) of 3.7 +/- 0.6 microM, but the cytochrome c reductase activity of the enzyme was much less susceptible to inhibition (IC(50) >0.1 mM). Steady-state absorbance spectra of nNOS recorded during uncoupled NADPH oxidation showed that the heme remained oxidized in the presence of the synthetic peptide consisting of amino acids 310-329 of the B2R, whereas the reduced oxyferrous heme complex was accumulated in its absence. These data suggest that binding of the B2R 310-329 peptide blocks flavin to heme electron transfer. Co-immunoprecipitation of B2R and nNOS from human embryonic kidney cells stably transfected with human nNOS suggests that the B2R may functionally interact with nNOS in vivo. This interaction of nNOS with the B2R may recruit the enzyme to allow for the effective coupling of bradykinin signaling to the nitric oxide pathway.
- Published
- 2000
- Full Text
- View/download PDF
45. Hyperglycemia enhances angiotensin II-induced janus-activated kinase/STAT signaling in vascular smooth muscle cells.
- Author
-
Amiri F, Venema VJ, Wang X, Ju H, Venema RC, and Marrero MB
- Subjects
- Animals, Cell Division, Cells, Cultured, Glucose pharmacology, JNK Mitogen-Activated Protein Kinases, Male, Muscle, Smooth, Vascular cytology, Phosphorylation, Rats, Rats, Sprague-Dawley, Tyrosine metabolism, Angiotensin II metabolism, Hyperglycemia metabolism, Mitogen-Activated Protein Kinases metabolism, Muscle, Smooth, Vascular metabolism, Signal Transduction
- Abstract
We have shown previously that angiotensin II (Ang II) activates the janus-activated kinase (JAK)/signal transducers and activators of transcription (STAT) pathway in vascular smooth muscle cells (VSMCs) and that activation of the JAK/STAT pathway is required for Ang II induction of VSMC proliferation. In the present study, we examined the effects of hyperglycemia (HG) on Ang II-induced JAK/STAT signaling events in cultured VSMCs. HG increases Ang II-induced JAK2 tyrosine phosphorylation and promotes a partial tyrosine phosphorylation of the enzyme under basal conditions. In addition, HG increases both basal and Ang II-induced complex formation of JAK2 with the Ang II AT(1) receptor. The extent of STAT1 and STAT3 tyrosine and serine phosphorylation are also increased under HG conditions. Furthermore, the tyrosine phosphorylation and activities of the SHP-1 and SHP-2 tyrosine phosphatases, enzymes that regulate Ang II-induced JAK2 tyrosine phosphorylation, are altered by HG. SHP-1, which is responsible for JAK2 tyrosine dephosphorylation in VSMC, is completely deactivated in HG, resulting in a prolonged duration of JAK2 phosphorylation under HG conditions. HG also enhances Ang II induction of VSMC proliferation. Taken together, these data suggest that HG augments Ang II induction of VSMC proliferation by increasing signal transduction through the JAK/STAT pathway.
- Published
- 1999
- Full Text
- View/download PDF
46. Endothelial nitric oxide synthase interactions with G-protein-coupled receptors.
- Author
-
Marrero MB, Venema VJ, Ju H, He H, Liang H, Caldwell RB, and Venema RC
- Subjects
- Animals, Bradykinin pharmacology, Calmodulin metabolism, Cells, Cultured, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Endothelium, Vascular metabolism, Enzyme Activation drug effects, Humans, Ligands, Nitric Oxide metabolism, Nitric Oxide Synthase antagonists & inhibitors, Nitric Oxide Synthase Type III, Peptide Fragments chemistry, Peptide Fragments metabolism, Peptide Fragments pharmacology, Phosphorylation drug effects, Phosphotyrosine metabolism, Protein Binding drug effects, Receptor, Angiotensin, Type 1, Receptor, Angiotensin, Type 2, Receptor, Bradykinin B2, Receptor, Endothelin B, Receptors, Angiotensin chemistry, Receptors, Bradykinin chemistry, Receptors, Endothelin chemistry, Receptors, Purinergic P2 chemistry, Receptors, Purinergic P2Y2, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Endothelium, Vascular enzymology, GTP-Binding Proteins metabolism, Nitric Oxide Synthase metabolism, Receptors, Angiotensin metabolism, Receptors, Bradykinin metabolism, Receptors, Endothelin metabolism, Receptors, Purinergic P2 metabolism
- Abstract
The endothelial nitric oxide synthase (eNOS) is activated in response to stimulation of endothelial cells by a number of vasoactive substances including, bradykinin (BK), angiotensin II (Ang II), endothelin-1 (ET-1) and ATP. In the present study we have used in vitro activity assays of purified eNOS and in vitro binding assays with glutathione S-transferase fusion proteins to show that the capacity to bind and inhibit eNOS is a common feature of membrane-proximal regions of intracellular domain 4 of the BK B2, the Ang II AT1 and the ET-1 ETB receptors, but not of the ATP P2Y2 receptor. Phosphorylation of serine or tyrosine residues in the eNOS-interacting region of the B2 receptor results in a loss of eNOS inhibition due to a decrease in the binding affinity of the receptor domain for the eNOS enzyme. Furthermore, the B2 receptor is transiently phosphorylated on tyrosine residues in cultured endothelial cells in response to BK stimulation. Phosphorylation occurs during the time in which eNOS transiently dissociates from the receptor accompanied by a transient increase in nitric oxide production. Taken together, these data support the hypotheses that eNOS is regulated in endothelial cells by reversible and inhibitory interactions with G-protein-coupled receptors and that these interactions can be modulated by receptor phosphorylation.
- Published
- 1999
47. Vascular endothelial growth factor signals endothelial cell production of nitric oxide and prostacyclin through flk-1/KDR activation of c-Src.
- Author
-
He H, Venema VJ, Gu X, Venema RC, Marrero MB, and Caldwell RB
- Subjects
- Animals, Calcium metabolism, Cattle, Cells, Cultured, Cyclic GMP metabolism, Endothelial Growth Factors pharmacology, Endothelium, Vascular drug effects, Enzyme Activation, Enzyme Inhibitors pharmacology, Genistein pharmacology, Inositol 1,4,5-Trisphosphate metabolism, Isoenzymes metabolism, Lymphokines pharmacology, Phospholipase C gamma, Placenta Growth Factor, Pregnancy Proteins pharmacology, Receptors, Vascular Endothelial Growth Factor, Signal Transduction, Type C Phospholipases metabolism, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Endothelial Growth Factors metabolism, Epoprostenol metabolism, Lymphokines metabolism, Nitric Oxide metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Growth Factor metabolism, src-Family Kinases metabolism
- Abstract
Vascular endothelial growth factor (VEGF) is a potent endothelial cell-specific mitogen that promotes angiogenesis, vascular hyperpermeability, and vasodilation by autocrine mechanisms involving nitric oxide (NO) and prostacyclin (PGI(2)) production. These experiments used immunoprecipitation and immunoassay procedures to characterize the signaling pathways by which VEGF induces NO and PGI(2) formation in cultured endothelial cells. The data showed that VEGF stimulates complex formation of the flk-1/kinase-insert domain-containing receptor (KDR) VEGF receptor with c-Src and that Src activation is required for VEGF induction of phospholipase C gamma1 activation and inositol 1,4,5-trisphosphate formation. Reporter cell assays showed that VEGF promotes a approximately 50-fold increase in NO formation, which peaks at 5-20 min. This effect is mediated by a signaling cascade initiated by flk-1/KDR activation of c-Src, leading to phospholipase C gamma1 activation, inositol 1,4,5-trisphosphate formation, release of [Ca(2+)](i) and nitric oxide synthase activation. Immunoassays of VEGF-induced 6-keto prostaglandin F(1alpha) formation as an indicator of PGI(2) production revealed a 3-4-fold increase that peaked at 45-60 min. The PGI(2) signaling pathway follows the NO pathway through release of [Ca(2+)](i), but diverges prior to NOS activation and also requires activation of mitogen-activated protein kinase. These results suggest that NO and PGI(2) function in parallel in mediating the effects of VEGF.
- Published
- 1999
- Full Text
- View/download PDF
48. Regulation of angiotensin II-induced phosphorylation of STAT3 in vascular smooth muscle cells.
- Author
-
Liang H, Venema VJ, Wang X, Ju H, Venema RC, and Marrero MB
- Subjects
- Animals, Aorta, Calcineurin metabolism, Cells, Cultured, Enzyme Inhibitors pharmacology, Janus Kinase 2, Okadaic Acid pharmacology, Oligonucleotides, Antisense pharmacology, Phosphoprotein Phosphatases metabolism, Phosphorylation, Phosphotyrosine analysis, Protein Phosphatase 2, Protein-Tyrosine Kinases antagonists & inhibitors, Protein-Tyrosine Kinases metabolism, Rats, Rats, Sprague-Dawley, STAT1 Transcription Factor, STAT3 Transcription Factor, Signal Transduction, Tacrolimus pharmacology, Tyrphostins pharmacology, Angiotensin II pharmacology, DNA-Binding Proteins metabolism, Muscle, Smooth, Vascular metabolism, Proto-Oncogene Proteins, Trans-Activators metabolism
- Abstract
Ligand binding to the angiotensin II (Ang II) AT1 receptor on vascular smooth muscle cells (VSMCs) activates the Janus-activated kinase (JAK)/signal transducers and activators of transcription (STAT) pathway. We have shown previously that the JAK2 tyrosine kinase and the Src family p59 Fyn tyrosine kinase are required for Ang II-induced STAT1 tyrosine phosphorylation in VSMCs. The mitogen-activated protein kinase phosphatase, MKP-1, is required for STAT1 tyrosine dephosphorylation. In the present study, using specific enzyme inhibitors and antisense oligonucleotides, we show that Ang II-induced tyrosine phosphorylation and nuclear translocation of STAT3 in VSMCs is mediated by p60 c-Src, whereas tyrosine dephosphorylation is mediated by calcineurin. Calcineurin is activated in response to Ang II stimulation of VSMCs and is translocated to the nucleus. In addition, we show that Ang II-induced serine phosphorylation of STAT3 in VSMCs is mediated by mitogen-activated protein kinase and that dephosphorylation is mediated by protein phosphatase 2A (PP2A). PP2A translocates to the nucleus in response to Ang II stimulation of VSMCs and forms a complex with STAT3 in an Ang II-dependent manner.
- Published
- 1999
- Full Text
- View/download PDF
49. VEGF induces nuclear translocation of Flk-1/KDR, endothelial nitric oxide synthase, and caveolin-1 in vascular endothelial cells.
- Author
-
Feng Y, Venema VJ, Venema RC, Tsai N, and Caldwell RB
- Subjects
- Animals, Aorta, Blotting, Western, Cattle, Caveolin 1, Cell Fractionation, Cell Nucleus drug effects, Cells, Cultured, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, Immunohistochemistry, Nitric Oxide Synthase Type III, Receptors, Vascular Endothelial Growth Factor, Retina, Signal Transduction drug effects, Time Factors, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Caveolins, Cell Nucleus metabolism, Endothelial Growth Factors pharmacology, Endothelium, Vascular drug effects, Lymphokines pharmacology, Membrane Proteins metabolism, Nitric Oxide Synthase metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Growth Factor metabolism
- Abstract
VEGF increases endothelial cell permeability and growth by a process requiring NOS activity. Because eNOS activity is regulated by its interaction with the caveolar structural protein caveolin-1, we analyzed VEGF effects on structural interactions between eNOS, caveolin-1 and the VEGF receptor Flk-1/KDR. Confocal immunolocalization analysis of the subcellular distribution of Flk-1/KDR, caveolin-1 and eNOS showed that VEGF stimulated the translocation of all three proteins into the nucleus. This result was confirmed by cell fractionation and immunoblotting studies showing that levels of all three proteins within the caveolar compartment declined progressively after 30 and 60 min of VEGF treatment. The pattern was reversed for nuclear fractions. Protein levels were lowest in the control cultures, but increased progressively after 30 and 60 min of treatment. Nuclear translocation of eNOS and Flk-1/KDR within caveolae may represent a mechanism for targeting NO production to the nuclear compartment where it could influence transcription factor activation., (Copyright 1999 Academic Press.)
- Published
- 1999
- Full Text
- View/download PDF
50. VEGF-induced permeability increase is mediated by caveolae.
- Author
-
Feng Y, Venema VJ, Venema RC, Tsai N, Behzadian MA, and Caldwell RB
- Subjects
- Actins metabolism, Animals, Aorta, Thoracic, Cattle, Caveolin 1, Cells, Cultured, Endothelium, Vascular metabolism, Endothelium, Vascular ultrastructure, Fluorescent Antibody Technique, Indirect, Intercellular Junctions, Membrane Proteins metabolism, NG-Nitroarginine Methyl Ester pharmacology, Nitric Oxide biosynthesis, Nitric Oxide Synthase metabolism, Nitric Oxide Synthase Type III, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Growth Factor metabolism, Receptors, Vascular Endothelial Growth Factor, Retinal Vessels, Signal Transduction, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Capillary Permeability drug effects, Caveolins, Endothelial Growth Factors pharmacology, Endothelium, Vascular drug effects, Lymphokines pharmacology
- Abstract
Purpose: To determine the cellular route by which vascular endothelial cell growth factor (VEGF) increases the permeability of cultured retinal endothelial cells and to test whether nitric oxide (NO) production by NO synthase (NOS) is involved in signaling VEGF's permeability enhancing effects., Methods: Cultured bovine retinal microvascular endothelial (BRE) cells were used for bioassay of permeability function and its ultrastructural correlates. The role of NOS activity in VEGF's permeability enhancing effects was tested with the use of an NOS inhibitor. Because activity of endothelial NOS (eNOS) is thought to be regulated by its interaction with the caveolar protein caveolin-1, structural relationships between eNOS, caveolin-1, and the VEGF receptor FIk-1/KDR were analyzed with double-label immunofluorescence and cell fractionation procedures., Results: Bioassays of permeability function and structure demonstrated that VEGF increases permeability of cultured BRE cells by an NOS-dependent process of transcytotic transport in caveolae. Double-label analysis showed that Flk-1/KDR and eNOS colocalize with caveolin-1 in plasma membrane caveolae. Cell fractionation and immunoblot analysis confirmed this effect. Densitometry showed that Flk-1/KDR, eNOS, and caveolin-1 levels were highest in caveolar fractions. Similar results were obtained in studies with bovine aortic endothelial cells., Conclusions: These results demonstrate that VEGF increases endothelial cell permeability by an eNOS-dependent mechanism of transcytosis in caveolae. Localization of Flk-1/KDR and eNOS with caveolin-1 suggests that VEGF signaling occurs within the caveolar compartment.
- Published
- 1999
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.