Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 248
Filter
Add more filters

Publication year range
1.
Amino Acids ; 42(5): 1903-11, 2012 May.
Article in English | MEDLINE | ID: mdl-21614558

ABSTRACT

Hyperhomocysteinemia (HHcy) is a risk factor for vascular disease, but the underlying mechanisms remain incompletely defined. Reduced bioavailability of nitric oxide (NO) is a principal manifestation of underlying endothelial dysfunction, which is an initial event in vascular disease. Inhibition of cellular methylation reactions by S-adenosylhomocysteine (AdoHcy), which accumulates during HHcy, has been suggested to contribute to vascular dysfunction. However, thus far, the effect of intracellular AdoHcy accumulation on NO bioavailability has not yet been fully substantiated by experimental evidence. The present study was carried out to evaluate whether disturbances in cellular methylation status affect NO production by cultured human endothelial cells. Here, we show that a hypomethylating environment, induced by the accumulation of AdoHcy, impairs NO production. Consistent with this finding, we observed decreased eNOS expression and activity, but, by contrast, enhanced NOS3 transcription. Taken together, our data support the existence of regulatory post-transcriptional mechanisms modulated by cellular methylation potential leading to impaired NO production by cultured human endothelial cells. As such, our conclusions may have implications for the HHcy-mediated reductions in NO bioavailability and endothelial dysfunction.


Subject(s)
Arginine/analogs & derivatives , Endothelial Cells/metabolism , Methylation , Nitric Oxide/metabolism , S-Adenosylhomocysteine/metabolism , Arginine/metabolism , Cells, Cultured , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Hyperhomocysteinemia/metabolism , Nitric Oxide/deficiency , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Vascular Diseases/metabolism
2.
Science ; 258(5090): 1898-902, 1992 Dec 18.
Article in English | MEDLINE | ID: mdl-1281928

ABSTRACT

Nitric oxide (NO.), a potentially toxic molecule, has been implicated in a wide range of biological functions. Details of its biochemistry, however, remain poorly understood. The broader chemistry of nitrogen monoxide (NO) involves a redox array of species with distinctive properties and reactivities: NO+ (nitrosonium), NO., and NO- (nitroxyl anion). The integration of this chemistry with current perspectives of NO biology illuminates many aspects of NO biochemistry, including the enzymatic mechanism of synthesis, the mode of transport and targeting in biological systems, the means by which its toxicity is mitigated, and the function-regulating interaction with target proteins.


Subject(s)
Nitric Oxide/metabolism , Amino Acid Oxidoreductases/metabolism , Animals , Arginine/metabolism , Humans , Nitric Oxide/chemistry , Nitric Oxide/physiology , Nitric Oxide Synthase , Oxidation-Reduction
3.
Clin Pharmacol Ther ; 106(6): 1261-1267, 2019 12.
Article in English | MEDLINE | ID: mdl-31557306

ABSTRACT

Genetic variation may differentially modify drug and placebo treatment effects in randomized clinical trials. In asthma, although lung function and asthma control improvements are commonplace with placebo, pharmacogenomics of placebo vs. drug response remains unexamined. In a genomewide association study of subjective and objective outcomes with placebo treatment in Childhood Asthma Management Program of nedocromil/budesonide vs. placebo (N = 604), effect estimates for lead single nucleotide polymorphisms (SNPs) were compared across arms. The coughing/wheezing lead SNP, rs2392165 (ß = 0.94; P = 1.10E-07) mapped to BBS9, a gene implicated in lung development that contains a lung function expression quantitative trait locus. The effect was attenuated with budesonide (Pinteraction  = 1.48E-07), but not nedocromil (Pinteraction  = 0.06). The lead forced vital capacity SNP, rs12930749 (ß = -5.80; P = 1.47E-06), mapped to KIAA0556, a locus genomewide associated with respiratory diseases. The rs12930749 effect was attenuated with budesonide (Pinteraction  = 1.32E-02) and nedocromil (Pinteraction  = 1.09E-02). Pharmacogenomic analysis revealed differential effects with placebo and drug treatment that could potentially guide precision drug development in asthma.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Budesonide/therapeutic use , Nedocromil/therapeutic use , Placebo Effect , Child , Cough/genetics , Cytoskeletal Proteins/genetics , Female , Genome-Wide Association Study , Humans , Male , Microtubule-Associated Proteins/genetics , Patient Reported Outcome Measures , Pharmacogenomic Testing , Polymorphism, Single Nucleotide , Respiratory Sounds/genetics , Treatment Outcome , Vital Capacity/genetics
4.
J Clin Invest ; 76(2): 703-8, 1985 Aug.
Article in English | MEDLINE | ID: mdl-2863286

ABSTRACT

Platelet aggregation is currently felt to play an important role in the pathogenesis of ischemic vascular disorders. The smooth muscle relaxant, nitroglycerin, has been shown to inhibit platelet aggregation in vitro, but at concentrations that were felt to be unattainable in vivo. Because the in vivo action of nitroglycerin on smooth muscle cells has been shown to depend on the presence of reduced cytosolic sulfhydryl groups, the inhibitory effect of nitroglycerin on platelet aggregation was examined in the presence of the reduced thiol, N-acetylcysteine. Millimolar concentrations of N-acetylcysteine potentiated markedly the inhibitory effect of nitroglycerin on platelet aggregation induced by ADP, epinephrine, collagen, and arachidonate, decreasing the 50% inhibitory concentration (IC50) approximately 50-fold for each of these agents. Other guanylate cyclase activators inhibited ADP-induced aggregation similarly and this inhibition was likewise potentiated by N-acetylcysteine. Platelet guanosine 3',5'-cyclic monophosphate content increased fivefold in the presence of nitroglycerin and N-acetylcysteine 2 min before maximal inhibition of ADP-induced aggregation was achieved, while simultaneously measured cyclic AMP did not change relative to base-line levels. In the absence of N-acetylcysteine, nitroglycerin induced a marked decrease in platelet-reduced glutathione content as S-nitroso-thiol adducts were produced. The synthetic S-nitroso-thiol, S-nitroso-N-acetylcysteine, markedly inhibited platelet aggregation with an IC50 of 6 nM. These data show that N-acetylcysteine markedly potentiates the inhibition of platelet aggregation by nitroglycerin and likely does so by inducing the formation of an S-nitrosothiol adduct(s), which activate guanylate cyclase.


Subject(s)
Acetylcysteine/pharmacology , Nitroglycerin/pharmacology , Platelet Aggregation/drug effects , Acetylcysteine/analogs & derivatives , Adenosine Diphosphate/pharmacology , Cyclic AMP/metabolism , Drug Synergism , Enzyme Activation , Glutathione/metabolism , Guanylate Cyclase/metabolism , Humans , In Vitro Techniques , Time Factors
5.
J Clin Invest ; 82(4): 1391-7, 1988 Oct.
Article in English | MEDLINE | ID: mdl-3139714

ABSTRACT

We examined the similarities and differences in conformation between recombinant human single-chain tissue plasminogen activator (sct-PA) and two-chain tissue plasminogen activator (tct-PA), and compared these structural data with measurement of enzymatic activity. The intrinsic protein fluorescence of native tct-PA was 54% that of sct-PA. Differences in steady state protein fluorescence were also noted with denaturation of these plasminogen activators, as well as in the quenching of intrinsic fluorescence of the reduced, alkylated species by iodide. Using the chromogenic substrate H-D-isoleucyl-L-prolyl-L-arginine-p-nitroanilide (S-2288), the catalytic efficiency of sct-PA was found to be 26% that of tct-PA, and this was primarily a reflection of the difference in Km. On addition of soluble fibrin monomer prepared with the tetrapeptide glycyl-L-prolyl-L-arginyl-L-proline (GPRP), the catalytic efficiency of both species increased by 13-fold for sct-PA and by 3.5-fold for tct-PA to approximately the same value. Using the fluorophore eosin iodoacetamide covalently coupled to the single free cysteine in the molecule, Cys 83, the microenvironment of the fibrin-binding site located near this residue was studied. On addition of soluble fibrin monomer to eosin-labeled tct-PA, no effect on eosin fluorescence was noted. Eosin-labeled tct-PA had 16% less eosin fluorescence than did sct-PA and on addition of soluble fibrin monomer to eosin-labeled sct-PA, a decrease in eosin fluorescence, approaching that of eosin coupled to tct-PA, was observed. Together, these structural and kinetic data suggest that sct-PA undergoes a conformational change on binding to fibrin monomer that leads to dramatic differences in catalytic efficiency of the single-chain species. In so doing, sct-PA bound to fibrin assumes the kinetic profile of tct-PA bound to fibrin.


Subject(s)
Protein Conformation , Tissue Plasminogen Activator , Binding Sites , Enzyme Activation , Fibrin , Humans , Polylysine , Sodium Iodide , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Structure-Activity Relationship , Substrate Specificity , Tissue Plasminogen Activator/metabolism
6.
J Clin Invest ; 79(6): 1749-55, 1987 Jun.
Article in English | MEDLINE | ID: mdl-2438305

ABSTRACT

We studied the disaggregation of human platelets by tissue-type plasminogen activator (t-PA). When added to a suspension of human platelets induced to aggregate in plasma with adenosine 5'-diphosphate, t-PA promoted disaggregation of platelets over several minutes. Addition of fresh plasma or purified human fibrinogen to disaggregated platelets facilitated (reversible) aggregation and subsequent disaggregation. Aspirin treatment of platelets markedly potentiated the ability of t-PA to induce disaggregation. Disaggregation was inhibited by alpha-2-antiplasmin. Comparative analysis of the rate of proteolysis of platelet-bound fibrinogen with that of ambient plasma fibrinogen suggested that fibrinogenolysis of cohesive fibrinogen occurred more rapidly than fibrinogenolysis of ambient fibrinogen. These data demonstrate that t-PA facilitates platelet disaggregation in plasma through kinetically selective proteolysis of cohesive fibrinogen by plasmin, and suggest that thrombolytic mechanisms may serve both to remove platelets from platelet-fibrin thrombi and to disperse circulating platelet aggregates.


Subject(s)
Blood Platelets/drug effects , Platelet Aggregation/drug effects , Tissue Plasminogen Activator/pharmacology , Adenosine Diphosphate/pharmacology , Aspirin/pharmacology , Fibrin Fibrinogen Degradation Products/pharmacology , Fibrinogen/metabolism , Fibrinogen/pharmacology , Humans , Kinetics , Platelet Membrane Glycoproteins/metabolism , alpha-2-Antiplasmin/pharmacology
7.
J Clin Invest ; 81(1): 62-8, 1988 Jan.
Article in English | MEDLINE | ID: mdl-3335643

ABSTRACT

The conversion of tissue macrophages into cholesteryl ester-rich foam cells is a crucial early event in atherogenesis. We studied the platelet as a potential source of cholesterol for esterification by macrophages because (a) platelets are rich in free cholesterol, (b) they adhere to macrophages early in atherogenesis, and (c) vascular injury can induce foam cell formation in the absence of hyperlipoproteinemia. We found that washed, activated human platelets from normocholesterolemic donors stimulated cholesteryl ester formation by the human monocyte-derived cell, U-937. Platelet cholesterol, released from platelets activated with calf skin collagen, was approximately equipotent at donating cholesterol to U-937 cells for esterification as normal human low density lipoprotein cholesterol. The stimulation of cholesteryl ester formation by activated human platelets demonstrated both concentration and time dependence. When hypercholesterolemic donors were studied, it was found that increasing plasma levels of cholesterol correlated directly with the ability of these hypercholesterolemic platelets to support cholesteryl ester synthesis by U-937 cells. Cholesterol-donating activity was also found in a 1,000-g supernatants of activated platelets. These observations point to a new and potentially important role for platelets in atherogenesis and suggest a mechanism for foam cell formation in the absence of marked hypercholesterolemia.


Subject(s)
Blood Platelets/physiology , Cholesterol Esters/biosynthesis , Monocytes/metabolism , Blood Platelets/metabolism , Cell Line , Cholesterol/blood , Collagen , Humans , Lipoproteins/pharmacology , Monocytes/drug effects , Platelet Aggregation
8.
J Clin Invest ; 78(4): 1112-9, 1986 Oct.
Article in English | MEDLINE | ID: mdl-3489737

ABSTRACT

von Willebrand protein was found to promote the incorporation of platelets into evolving fibrin thrombi. Using formalin-treated or fresh platelets, both the initial rate and extent of platelet incorporation into polymerizing fibrin were dependent on von Willebrand protein. von Willebrand protein was incorporated into evolving fibrin thrombi in parallel with platelets. Soluble fibrin monomer covalently linked to acrylonitrile beads (Matrex 102) bound von Willebrand protein specifically and saturably with an apparent approximate dissociation constant (KD) of 15 micrograms/ml. Glycocalicin, the water-soluble proteolytic fragment of glycoprotein Ib, bound to fibrin monomer in this system specifically and saturably, as well, with an apparent approximate KD of 5 micrograms/ml, but only in the presence of saturating concentrations of von Willebrand protein. These data demonstrate that the initial rate and extent of platelet incorporation into evolving fibrin thrombi are dependent on von Willebrand protein; von Willebrand protein serves as a link between polymerizing fibrin and platelet surface glycoprotein Ib; and von Willebrand protein binds to fibrin monomer and is thereby able to bind to platelet surface glycoprotein Ib in the absence of ristocetin.


Subject(s)
Blood Platelets/metabolism , Fibrin/metabolism , Platelet Glycoprotein GPIb-IX Complex , von Willebrand Factor/pharmacology , Blood Platelets/drug effects , Blood Proteins/metabolism , Humans , Kinetics , Platelet Membrane Glycoproteins/metabolism , Polymers/metabolism , Solubility
9.
J Clin Invest ; 88(5): 1663-71, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1719029

ABSTRACT

Flow-mediated vasodilation is endothelium dependent. We hypothesized that flow activates a potassium channel on the endothelium, and that activation of this channel leads to the release of the endogenous nitrovasodilator, nitric oxide. To test this hypothesis, rabbit iliac arteries were perfused at varying flow rates, at a constant pressure of 60 mm Hg. Increments in flow induced proportional increases in vessel diameter, which were abolished by L,N-mono-methylarginine (the antagonist of nitric-oxide synthesis). Barium chloride, depolarizing solutions of potassium, verapamil, calcium-free medium, and antagonists of the KCa channel (charybdotoxin, iberiotoxin) also blocked flow-mediated vasodilation. Conversely, responses to other agonists of endothelium-dependent and independent vasodilation were unaffected by charybdotoxin or iberiotoxin. To confirm that flow activated a specific potassium channel to induce the release of nitric oxide, endothelial cells cultured on micro-carrier beads were added to a flow chamber containing a vascular ring without endothelium. Flow-stimulated endothelial cells released a diffusible vasodilator; the degree of vasorelaxation was dependent upon the flow rate. Relaxation was abrogated by barium, tetraethylammonium ion, or charybdotoxin, but was not affected by apamin, glybenclamide, tetrodotoxin, or ouabain. The data suggest that transmission of a hyperpolarizing current from endothelium to the vascular smooth muscle is not necessary for flow-mediated vasodilation. Flow activates a potassium channel (possibly the KCa channel) on the endothelial cell membrane that leads to the release of nitric oxide.


Subject(s)
Endothelium, Vascular/physiology , Nitric Oxide/metabolism , Potassium Channels/physiology , Vasodilator Agents/metabolism , Animals , Charybdotoxin , Female , In Vitro Techniques , Norepinephrine/pharmacology , Rabbits , Regional Blood Flow , Scorpion Venoms/pharmacology , Vasodilation , Verapamil/pharmacology
10.
J Clin Invest ; 96(1): 394-400, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7615810

ABSTRACT

GSH peroxidase (Px) catalyzes the reduction of lipid hydroperoxides (LOOH), known metabolic products of platelets and vascular cells. Because interactions between these cells are modulated by nitric oxide (NO) and LOOH inactivate NO, we investigated the effect of GSH-Px on the inhibition of platelet function by the naturally occurring S-nitrosothiol, S-nitroso-glutathione (SNO-Glu). Concentrations of SNO-Glu that alone did not inhibit platelet function (subthreshold inhibitory concentrations) were added to platelet-rich plasma together with GSH-Px (0.2-20 U/ml); this led to a dose-dependent inhibition of platelet aggregation with an IC50 of 0.6 U/ml GSH-Px. In the presence of subthreshold inhibitory concentrations of SNO-Glu, the LOOH, 5-hydroperoxy-6,8,11,14-eicosatetraenoic acid, increased platelet aggregation, an effect reversed by GSH-Px. Glutathione and SNO-Glu were equally effective as cosubstrates for GSH-Px. Incubation of SNO-Glu with GSH-Px for 1 min led to a 48.5% decrease in the concentration of SNO-Glu. Incubation of SNO-Glu with serum albumin led to the formation of S-nitroso-albumin, an effect enhanced by GSH-Px. These observations suggest that GSH-Px has two functions: reduction of LOOH, thereby preventing inactivation of NO, and metabolism of SNO-Glu, thereby liberating NO and/or supporting further transnitrosation reactions.


Subject(s)
Blood Platelets/drug effects , Glutathione Peroxidase/pharmacology , Mercaptoethanol , Nitroso Compounds/pharmacology , S-Nitrosothiols , Blood Platelets/physiology , Cyclic GMP/blood , Cysteine/analogs & derivatives , Cysteine/metabolism , Drug Synergism , Glutathione/analogs & derivatives , Glutathione/metabolism , Humans , Lipid Peroxides/pharmacology , Nitroso Compounds/metabolism , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , S-Nitrosoglutathione , Superoxide Dismutase/pharmacology
11.
J Clin Invest ; 103(3): 393-9, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9927500

ABSTRACT

Since thiols can undergo nitrosation and the cell membrane is rich in thiol-containing proteins, we considered the possibility that membrane surface thiols may regulate cellular entry of NO. Recently, protein disulfide isomerase (PDI), a protein that catalyzes thio-disulfide exchange reactions, has been found on the cell-surface membrane. We hypothesized that cell-surface PDI reacts with NO, catalyzes S-nitrosation reactions, and facilitates NO transfer from the extracellular to intracellular compartment. We observed that PDI catalyzes the S-nitrosothiol-dependent oxidation of the heme group of myoglobin (15-fold increase in the rate of oxidation compared with control), and that NO reduces the activity of PDI by 73.1 +/- 21.8% (P < 0.005). To assess the role of PDI in the cellular action of NO, we inhibited human erythroleukemia (HEL) cell-surface PDI expression using an antisense phosphorothioate oligodeoxynucleotide directed against PDI mRNA. This oligodeoxynucleotide decreased cell-surface PDI content by 74.1 +/- 9.3% and PDI folding activity by 46.6 +/- 3.5% compared with untreated or "scrambled" phosphorothioate oligodeoxynucleotide-treated cells (P < 0.0001). This decrease in cell-surface PDI was associated with a significant decrease in cyclic guanosine monophosphate (cGMP) generation after S-nitrosothiol exposure (65.4 +/- 26.7% reduction compared with control; P < 0.05), with no effect on cyclic adenosine monophosphate (cAMP) generation after prostaglandin E1 exposure. These data demonstrate that the cellular entry of NO involves a transnitrosation mechanism catalyzed by cell-surface PDI. These observations suggest a unique mechanism by which extracellular NO gains access to the intracellular environment.


Subject(s)
Nitric Oxide/metabolism , Protein Disulfide-Isomerases/metabolism , Biological Transport , Humans , Nitrosation , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Protein Disulfide-Isomerases/genetics , RNA, Messenger/genetics , Substrate Specificity , Tumor Cells, Cultured
12.
J Clin Invest ; 91(4): 1582-9, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8473501

ABSTRACT

Recent evidence suggests that sulfhydryl species can react with oxides of nitrogen under physiologic conditions and thereby stabilize endothelium-derived relaxing factor (EDRF) activity, but the presence of a specific in vivo thiol carrier for nitric oxide (NO) remains controversial. The single free sulfhydryl of serum albumin is the most abundant thiol species in plasma (approximately 0.5 mM) and is particularly reactive towards NO. To examine the potential role of serum albumin in endogenous nitric oxide metabolism, we synthesized S-nitroso-BSA (S-NO-BSA), a model S-nitroso-protein, and examined its effects on platelet function and coronary and systemic vascular tone in 16 mongrel dogs. Intravenous bolus S-NO-BSA markedly reduced mean arterial pressure in a dose-dependent manner and proved seven and a half-fold less potent than intravenous nitroglycerin and 10-fold less potent than intravenous S-nitroso-cysteine (half-maximal response of 75 nmol/kg compared to 10 and 7.5 nmol/kg, respectively; P < 0.05); when given by intravenous infusion (half-maximal response = 10 nmol/kg per min), however, S-NO-BSA and nitroglycerin were equipotent. Intravenous bolus S-NO-BSA had a greater duration of action than either nitroglycerin or S-nitroso-cysteine and produced marked prolongation of the template bleeding time associated with dose-dependent inhibition of ex vivo platelet aggregation (half-maximal response approximately 70 nmol/kg). Intracoronary S-NO-BSA increased coronary blood flow (mean +/- SEM) less effectively than nitroprusside, acetylcholine, or S-nitroso-cysteine (165% +/- 24% vs. 315% +/- 82%, 483% +/- 55%, or 475% +/- 66%, respectively; P < 0.05) although with much longer duration of action. On a molar basis, S-nitroso-cysteine proved more effective than S-nitroso-BSA, nitroprusside, or acetylcholine as an epicardial coronary vasodilator. Thus, serum albumin reacts with oxides of nitrogen to form a stable S-nitroso-thiol with properties reminiscent of authentic EDRF supporting the view that protein associated thiol may participate in the action and metabolism of EDRF.


Subject(s)
Nitric Oxide/metabolism , Nitric Oxide/physiology , S-Nitrosothiols , Serum Albumin, Bovine/metabolism , Acetylcholine/pharmacology , Animals , Bleeding Time , Blood Platelets/physiology , Coronary Vessels/drug effects , Coronary Vessels/physiology , Cysteine/analogs & derivatives , Cysteine/pharmacology , Dogs , Female , Half-Life , Male , Muscle Relaxation/drug effects , Myocardial Contraction/drug effects , Nitric Oxide/pharmacology , Nitroglycerin/pharmacology , Nitroprusside/pharmacology , Platelet Aggregation/drug effects , Serum Albumin, Bovine/pharmacology , Vasodilation/drug effects
13.
J Clin Invest ; 94(5): 1792-8, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7525649

ABSTRACT

Nitric oxide (NO) is an inhibitor of gastrointestinal smooth muscle. Model systems of the gut predict the NO will complex with biological thiol (SH) groups, yielding S-nitrosothiols (RS-NO), which may limit the propensity to form mutagenic nitrosamines. The inhibitory effects of NO and its biologically relevant adducts on sphincter of Oddi (SO) motility have been inferred from animal studies; however, their importance in regulating human SO is not known. The objectives of this study were to (a) provide histologic confirmation of nitric oxide synthase (NOS) in human SO; (b) characterize the pharmacology of S-nitroso-N-acetylcysteine (SNAC), an exemplary S-nitrosothiol, on SO motility in a rabbit model; and (c) study the effects of topical SNAC on SO motility in humans. Immunocytochemical and histochemical identification of NOS was performed in human SO. The pharmacologic response of SNAC was defined in isolated rabbit SO using a standard bioassay. Topical SNAC was then applied to the duodenal papilla in patients undergoing endoscopic retrograde cholangiopancreatography (ERCP) and biliary manometry. NOS was localized to nerve fibers and bundles of the SO in rabbits and humans. SNAC inhibited spontaneous motility (frequency and amplitude) as well as acetylcholine-induced elevations in SO basal pressure in the rabbit model. In patients undergoing ERCP and biliary manometry, topical SNAC inhibited SO contraction freqency, basal pressure, and duodenal motility. NOS is localized to neural elements in human SO, implicating a role for NO in regulating SO function. Supporting this concept, SNAC is an inhibitor of SO and duodenal motility when applied topically to humans during ERCP. Our data suggest a novel clinical approach using local NO donors to control gastrointestinal motility and regulate sphincteric function.


Subject(s)
Acetylcysteine/analogs & derivatives , Nitric Oxide/physiology , Sphincter of Oddi/drug effects , Acetylcysteine/pharmacology , Amino Acid Oxidoreductases/metabolism , Animals , Blood Pressure/drug effects , Humans , Immunohistochemistry , In Vitro Techniques , Male , Muscle Contraction/drug effects , Nitric Oxide Synthase , Rabbits , Sphincter of Oddi/physiology
14.
J Clin Invest ; 100(2): 350-6, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9218511

ABSTRACT

Vessel injury and thrombus formation are the cause of most ischemic coronary syndromes and, in this setting, activated platelets stimulate platelet recruitment to the growing thrombus. Recently, a constitutive nitric oxide synthase (NOS) has been identified in human platelets. To further define the capacity of platelets to produce nitric oxide (NO), as well as to study the role of this NO in platelet recruitment, we adapted a NO-selective microelectrode for use in a standard platelet aggregometer, thereby permitting simultaneous measurement of platelet aggregation and NO production. Treatment of platelets with the NO synthase inhibitor -NG-nitroarginine methyl ester (L-NAME), reduced NO production by 92+/-8% in response to 5 microM ADP compared to control but increased aggregation by only 15+/-2%. In contrast, L-NAME had a more pronounced effect on platelet recruitment as evidenced by a 35+/-5% increase in the extent of aggregation, a 33+/-3% decrease in cyclic GMP content, and a 31+/-5% increase in serotonin release from a second recruitable population of platelets added to stimulated platelets at the peak of NO production. To study platelet recruitment accurately, we developed an assay that monitors two platelet populations simultaneously. Nonbiotinylated platelets were incubated with L-NAME or vehicle and activated with ADP. At peak NO production, biotinylated platelets were added. As measured by three-color flow cytometry, there was a 56+/-11% increase in the number of P selectin- positive platelets in the nonbiotinylated population treated with L-NAME as compared to control. When biotinylated platelets were added to the L-NAME-treated nonbiotinylated population, the number of P selectin positive biotinylated plate-lets increased by 180+/-32% as compared to biotinylated platelets added to the control. In summary, stimulated platelets produce NO that modestly inhibits platelet activation but markedly inhibits additional platelet recruitment. These data suggest that platelet-derived NO may regulate platelet recruitment to a growing thrombus.


Subject(s)
Blood Platelets/metabolism , Nitric Oxide/physiology , Platelet Activation , Adenosine Diphosphate/pharmacology , Adult , Biotin/analogs & derivatives , Biotin/metabolism , Blood Platelets/drug effects , Blood Platelets/enzymology , Cyclic GMP/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Flow Cytometry , Humans , Microelectrodes , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , P-Selectin/metabolism , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Serotonin/metabolism , Thrombosis/metabolism
15.
J Clin Invest ; 96(6): 2630-8, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8675628

ABSTRACT

Endothelium-derived relaxing factor is important for vascular homeostasis and possesses qualities that may modulate vascular injury, including vasodilation, platelet inhibition, and inhibition of smooth muscle proliferation. S-nitrososerum albumin is a naturally occurring adduct of nitric oxide (NO) with a prolonged biologic half-life and is a potent vasodilator and platelet inhibitor. Given the avidity of serum albumin for subendothelial matrix and the antiproliferative effects of NO, we investigated the effects of locally delivered S-nitroso-bovine serum albumin (S-NO-BSA) and a polythiolated form of bovine serum albumin (pS-BSA) modified to carry several S-nitrosothiol groups (pS-NO-BSA) on neointimal responses in an animal model of vascular injury. Locally delivered S-NO-BSA bound preferentially to denuded rabbit femoral vessels producing a 26-fold increase in local concentration compared with uninjured vessels (P = 0.029). pS-NO-BSA significantly reduced the intimal/medial ratio (P = 0.038) and did so in conjunction with elevations in platelet (P < 0.001) and vascular cGMP content (P < or = 0.001). pS-NO-BSA treatment also inhibited platelet deposition (P = 0.031) after denuding injury. Comparison of BSA, S-NO-BSA, pS-NO-BSA, and control revealed a dose-response relationship between the amount of displaceable NO delivered and the extent of inhibition of neointimal proliferation at 2 wk (P < or = 0.001). Local administration of a stable protein S-nitrosothiol inhibits intimal proliferation and platelet deposition after vascular arterial balloon injury. This strategy for the local delivery of a long-lived NO adduct has potential for preventing restenosis after angioplasty.


Subject(s)
Blood Platelets/physiology , Femoral Artery/pathology , Muscle, Smooth, Vascular/pathology , Nitric Oxide/pharmacology , Serum Albumin, Bovine/pharmacology , Tunica Intima/pathology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Animals, Newborn , Aorta/drug effects , Aorta/physiology , Cattle , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Cyclic GMP/metabolism , Female , Femoral Artery/drug effects , Humans , In Vitro Techniques , Indium Radioisotopes , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Nitroso Compounds , Platelet Adhesiveness/drug effects , Rabbits , Rats , Sulfhydryl Compounds/pharmacology , Tunica Intima/cytology , Tunica Intima/drug effects
16.
J Clin Invest ; 91(1): 308-18, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8380812

ABSTRACT

Elevated levels of homocysteine are associated with an increased risk of atherosclerosis and thrombosis. The reactivity of the sulfhydryl group of homocysteine has been implicated in molecular mechanisms underlying this increased risk. There is also increasingly compelling evidence that thiols react in the presence of nitric oxide (NO) and endothelium-derived relaxing factor (EDRF) to form S-nitrosothiols, compounds with potent vasodilatory and antiplatelet effects. We, therefore, hypothesized that S-nitrosation of homocysteine would confer these beneficial bioactivities to the thiol, and at the same time attenuate its pathogenicity. We found that prolonged (> 3 h) exposure of endothelial cells to homocysteine results in impaired EDRF responses. By contrast, brief (15 min) exposure of endothelial cells, stimulated to secrete EDRF, to homocysteine results in the formation of S-NO-homocysteine, a potent antiplatelet agent and vasodilator. In contrast to homocysteine, S-NO-homocysteine does not support H2O2 generation and does not undergo conversion to homocysteine thiolactone, reaction products believed to contribute to endothelial toxicity. These results suggest that the normal endothelium modulates the potential, adverse effects of homocysteine by releasing EDRF and forming the adduct S-NO-homocysteine. The adverse vascular properties of homocysteine may result from an inability to sustain S-NO formation owing to a progressive imbalance between the production of NO by progressively dysfunctional endothelial cells and the levels of homocysteine.


Subject(s)
Blood Platelets/physiology , Endothelium, Vascular/physiology , Homocysteine/pharmacology , Nitric Oxide/physiology , Nitrogen Oxides/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Adenosine Diphosphate/pharmacology , Animals , Aorta , Blood Platelets/drug effects , Cattle , Cells, Cultured , Collagen/pharmacology , Copper/pharmacology , Cyclic GMP/blood , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Homocysteine/antagonists & inhibitors , Homocystine/pharmacology , Humans , Hydrogen Peroxide/metabolism , In Vitro Techniques , Iron/pharmacology , Kinetics , Luminescent Measurements , Magnetic Resonance Spectroscopy , Models, Cardiovascular , Nitric Oxide/pharmacology
17.
J Clin Invest ; 94(4): 1432-9, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7929818

ABSTRACT

Plasma albumin reacts with nitric oxide (NO) to form the bioactive adduct, S-nitroso-albumin (S-NO-albumin). The limited intracellular access of S-NO-albumin suggests the need for a vascular transfer mechanism of NO from a large plasma S-NO-albumin pool to effect biologic function. To study the role of low molecular weight (LMW) thiols in NO transfer in vivo, we administered intravenous S-NO-albumin (1-300 nmol/kg) to rabbits before and after an intravenous infusion of L-cysteine or N-acetyl-L-cysteine. S-NO-albumin produced dose-dependent hypotension that was significantly augmented by prior infusion of either LMW thiol. LMW thiol infusion significantly accelerated the rate of onset and reduced the duration of action of the hypotension induced by S-NO-albumin. The hemodynamic effects of S-NO-albumin after pretreatment with LMW thiols were mimicked by administration of the corresponding LMW S-nitrosothiol. The transfer of NO from albumin to L-cysteine was directly measured in rabbit plasma using a novel technique that couples high performance liquid chromatography to electrochemical detection. These data demonstrate that NO exchange between plasma protein thiol-bound NO and available LMW thiol pools (transnitrosation) occurs in vivo.


Subject(s)
Mercaptoethanol , Nitric Oxide/metabolism , Nitroso Compounds/blood , Nitroso Compounds/metabolism , S-Nitrosothiols , Serum Albumin, Bovine/metabolism , Sulfhydryl Compounds/metabolism , Acetylcysteine/administration & dosage , Acetylcysteine/analogs & derivatives , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Animals , Biological Transport , Blood Pressure/drug effects , Chromatography, High Pressure Liquid/methods , Cysteine/administration & dosage , Cysteine/analogs & derivatives , Cysteine/blood , Cysteine/metabolism , Cysteine/pharmacology , Nitrosation , Nitroso Compounds/administration & dosage , Nitroso Compounds/pharmacology , Protein Binding , Rabbits , Sulfhydryl Compounds/blood , Vascular Resistance/drug effects
18.
J Clin Invest ; 95(1): 360-6, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7529262

ABSTRACT

In vitro studies indicate that muscarinic cholinergic inhibition of beta-adrenergic cardiac responses may be modulated in part by nitric oxide (NO). To evaluate the role of NO in parasympathetic inhibition of the beta-adrenergic contractile response in vivo, we assessed the inotropic response to dobutamine before and during bilateral vagus nerve stimulation in closed-chest dogs. Dobutamine administration and vagal stimulation were repeated during intracoronary infusion of the NO synthase inhibitor NG-monomethyl-L-arginine (L-NMMA, 10 mumol/min) and again following infusion of L-arginine (100 mg/kg). In eight dogs, intracoronary dobutamine infusion at rates of 25 and 50 micrograms/min increased peak +dP/dt by 131 +/- 24 and 168 +/- 22%, respectively (P < 0.0001). Vagal stimulation (2.5 Hz) attenuated the responses to dobutamine (25 and 50 micrograms/min) by 23 +/- 4 and 21 +/- 4%, respectively (P < 0.001). L-NMMA reduced (by 44-62%; P < 0.001) and L-arginine restored vagal inhibition of the dobutamine-stimulated inotropic response. In a second group of nine dogs, dobutamine was administered systemically to assure a constant concentration in the coronary circulation. Vagal stimulation (2.5 Hz) attenuated the dobutamine-stimulated inotropic response (2.5 and 5.0 micrograms/kg per min) by 40 +/- 12% and 57 +/- 8%, respectively (P < 0.004). As with intracoronary dobutamine, L-NMMA diminished and L-arginine restored vagal inhibition of the inotropic response to dobutamine. Intracoronary infusion of atropine (12 micrograms/min) abolished the vagal inhibitory effect, and intracoronary infusion of 8-bromo-cyclic GMP (1 and 10 mM) caused a dose-dependent attenuation of the dobutamine-stimulated increase in +dP/dt. These data suggest that NO mediates, at least in part, vagal inhibition of the inotropic response to beta-adrenergic stimulation by dobutamine, and thus may play a role in normal physiologic regulation of myocardial autonomic responses.


Subject(s)
Dobutamine/pharmacology , Myocardial Contraction/physiology , Nitric Oxide/metabolism , Receptors, Adrenergic, beta/physiology , Vagus Nerve/physiology , Amino Acid Oxidoreductases/antagonists & inhibitors , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Cyclic AMP/pharmacology , Dogs , Electric Stimulation , Heart/innervation , Heart Rate , Muscarinic Antagonists , Myocardial Contraction/drug effects , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase , omega-N-Methylarginine
19.
J Clin Invest ; 97(4): 979-87, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8613552

ABSTRACT

Highly reactive oxygen species rapidly inactivate nitric oxide (NO), and endothelial product which inhibits platelet activation. We studied platelet inhibition by NO in two brothers with a cerebral thrombotic disorder. Both children had hyperreactive platelets, as determined by whole blood platelet aggregometry and flow cytometric analysis of the platelet surface expression of P-selectin. Mixing experiments showed that the patients'platelets behaved normally in control plasma; however, control platelets suspended in patient plasma were not inhibited by NO. As determined by flow cytometry, in the presence of plasma from either patient there was normal inhibition of the thrombin-induced expression of platelet surface P-selectin by prostacyclin, but not NO. Using a scopoletin assay, we measured a 2.7-fold increase in plasma H2O2 generation in one patient and a 3.4-fold increase in the second patient, both compared woth control plasma. Glutathione peroxidase (GSH-Px) activity was decreased in the patients' plasmas compared with control plasma. The addition of exogenous GSH-Px led to restoration of platelet inhibition by NO. These data show that, in these patients' plasmas, impaired metabolism of reactive oxygen species reduces the bioavailability of NO and impairs normal platelet inhibitory mechanisms. These findings suggest that attenuated NO-mediated platelet inhibition produced by increased reactive oxygen species or impaired antioxidant defense may cause a thrombotic disorder in humans.


Subject(s)
Nitric Oxide/physiology , Platelet Aggregation/drug effects , Thrombosis/etiology , Cyclic GMP/blood , Epoprostenol/physiology , Glutathione Peroxidase/blood , Humans , Hydrogen Peroxide/metabolism , Infant , Male , P-Selectin/blood , Platelet Aggregation Inhibitors/pharmacology
20.
J Clin Invest ; 93(2): 844-51, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8113416

ABSTRACT

Abnormalities in endothelium-dependent arterial relaxation develop early in atherosclerosis and may, in part, result from the effects of modified low-density lipoprotein (LDL) on agonist-mediated endothelium-derived relaxing factor (EDRF) release and EDRF degradation. alpha-Tocopherol (AT) is the main lipid-soluble antioxidant in human plasma and lipoproteins, therefore, we investigated the effects of AT on endothelium-dependent arterial relaxation in male New Zealand White rabbits fed diets containing (a) no additive (controls), (b) 1% cholesterol (cholesterol group), or 1% cholesterol with either (c) 1,000 IU/kg chow AT (low-dose AT group) or (d) 10,000 IU/kg chow AT (high-dose AT group). After 28 d, we assayed endothelial function and LDL susceptibility to ex vivo copper-mediated oxidation. Acetylcholine-and A23187-mediated endothelium-dependent relaxations were significantly impaired in the cholesterol group (P < 0.001 vs. control), but preserved in the low-dose AT group (P = NS vs. control). Compared to the control and cholesterol groups, vessels from the high-dose AT group demonstrated profound impairment of arterial relaxation (P < 0.05) and significantly more intimal proliferation than other groups (P < 0.05). In normal vessels, alpha-tocopherol had no effect on endothelial function. LDL derived from both the high- and low-dose AT groups was more resistant to oxidation than LDL from control animals (P < 0.05). These data indicate that modest dietary treatment with AT preserves endothelial vasodilator function in cholesterol-fed rabbits while a higher dose of AT is associated with endothelial dysfunction and enhanced intimal proliferation despite continued LDL resistance to ex vivo copper-mediated oxidation.


Subject(s)
Cholesterol, Dietary/pharmacology , Endothelium, Vascular/physiology , Muscle, Smooth, Vascular/physiology , Nitric Oxide/metabolism , Vasodilation/physiology , Vitamin E/pharmacology , Acetylcholine/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Calcimycin/pharmacology , Cholesterol/blood , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/ultrastructure , In Vitro Techniques , Lipoproteins/blood , Lipoproteins/drug effects , Male , Microscopy, Electron, Scanning , Muscle, Smooth, Vascular/drug effects , Nitric Oxide/physiology , Nitroprusside/pharmacology , Rabbits , Triglycerides/blood , Vasodilation/drug effects , Veins/drug effects , Veins/physiology , Vitamin E/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL