Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Gen Subj ; 1861(5 Pt A): 995-999, 2017 May.
Article in English | MEDLINE | ID: mdl-28219722

ABSTRACT

We recently demonstrated that a novel storage and transport mechanism for nitric oxide (NO) mediated by glutathione-S-transferase P1 (GSTP1) and multidrug resistance protein 1 (MRP1/ABCC1), protects M1-macrophage (M1-MØ) models from large quantities of endogenous NO. This system stores and transports NO as dinitrosyl-dithiol-iron complexes (DNICs) composed of iron, NO and glutathione (GSH). Hence, this gas with contrasting anti- and pro-tumor effects, which has been assumed to be freely diffusible, is a tightly-regulated species in M1-MØs. These control systems prevent NO cytotoxicity and may be responsible for delivering cytotoxic NO as DNICs via MRP1 from M1-MØs, to tumor cell targets.


Subject(s)
Glutathione S-Transferase pi/metabolism , Killer Cells, Natural/metabolism , Macrophages/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Nitric Oxide/metabolism , Protective Agents/metabolism , Glutathione/metabolism , Humans
2.
Oncogene ; 30(22): 2504-13, 2011 Jun 02.
Article in English | MEDLINE | ID: mdl-21297667

ABSTRACT

The SET oncoprotein participates in cancer progression by affecting multiple cellular processes, inhibiting the tumor suppressor protein phosphatase 2A (PP2A), and inhibiting the metastasis suppressor nm23-H1. On the basis of these multiple activities, we hypothesized that targeted inhibition of SET would have multiple discrete and measurable effects on cancer cells. Here, the effects of inhibiting SET oncoprotein function on intracellular signaling and proliferation of human cancer cell lines was investigated. We observed the effects of COG112, a novel SET interacting peptide, on PP2A activity, Akt signaling, nm23-H1 activity and cellular migration/invasion in human U87 glioblastoma and MDA-MB-231 breast adenocarcinoma cancer cell lines. We found that COG112 interacted with SET protein and inhibited the association between SET and PP2A catalytic subunit (PP2A-c) and nm23-H1. The interaction between COG112 and SET caused PP2A phosphatase and nm23-H1 exonuclease activities to increase. COG112-mediated increases in PP2A activity resulted in the inhibition of Akt signaling and cellular proliferation. Additionally, COG112 inhibited SET association with Ras-related C(3) botulinum toxin substrate 1 (Rac1), leading to decreased cellular migration and invasion. COG112 treatment releases the SET-mediated inhibition of the tumor suppressor PP2A, as well as the metastasis suppressor nm23-H1. These results establish SET as a novel molecular target and that the inhibition of SET may have beneficial effects in cancer chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Histone Chaperones/antagonists & inhibitors , Neoplasms/drug therapy , Peptides/therapeutic use , Transcription Factors/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA-Binding Proteins , Humans , NM23 Nucleoside Diphosphate Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , rac1 GTP-Binding Protein/metabolism
3.
Oncogene ; 28(43): 3837-46, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19701246

ABSTRACT

The chemopreventative effects of dithiolethione compounds are attributed to their activation of antioxidant response elements (AREs) by reacting with the Nrf2/Keap1 protein complex. In this study, we show antiproliferative effects of the dithiolethione compound ACS-1 in human cancer cell lines (A549 and MDA-MB-231) by increasing the activity of the tumor suppressor protein phoshatase 2A (PP2A). ACS-1 inhibited epidermal growth factor (EGF)-induced cellular proliferation in a concentration- and time-dependent manner. Akt activation, as determined by serine-473 phosphorylation, was inhibited by ACS-1 in cells stimulated with either EGF or fibronectin. Furthermore, ACS-1 inhibited mammalian target of rapamycin signaling and decreased c-myc protein levels. ACS-1 did not proximally alter EGF receptor or integrin signaling, but caused a concentration-dependent increase in PP2A activity. The effect of ACS-1 on Akt activation was not observed in the presence of the PP2A inhibitor okadaic acid. ACS-1 effects on PP2A activity were independent of ARE activation and cAMP formation. In addition to ACS-1, other dithiolethione compounds showed similar effects in reducing Akt activation, suggesting that this class of compounds may have other effects beyond chemoprevention.


Subject(s)
Anticarcinogenic Agents/pharmacology , Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Lung Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
4.
Br J Pharmacol ; 151(1): 63-72, 2007 May.
Article in English | MEDLINE | ID: mdl-17351657

ABSTRACT

BACKGROUND AND PURPOSE: Angiogenesis involves multiple signaling pathways that must be considered when developing agents to modulate pathological angiogenesis. Because both cyclooxygenase inhibitors and dithioles have demonstrated anti-angiogenic properties, we investigated the activities of a new class of anti-inflammatory drugs containing dithiolethione moieties (S-NSAIDs) and S-valproate. EXPERIMENTAL APPROACH: Anti-angiogenic activities of S-NSAIDS, S-valproate, and the respective parent compounds were assessed using umbilical vein endothelial cells, muscle and tumor tissue explant angiogenesis assays, and developmental angiogenesis in Fli:EGFP transgenic zebrafish embryos. KEY RESULTS: Dithiolethione derivatives of diclofenac, valproate, and sulindac inhibited endothelial cell proliferation and induced Ser(78) phosphorylation of hsp27, a known molecular target of anti-angiogenic signaling. The parent drugs lacked this activity, but dithiolethiones were active at comparable concentrations. Although dithiolethiones can potentially release hydrogen sulphide, NaSH did not reproduce some activities of the S-NSAIDs, indicating that the dithioles regulate angiogenesis through mechanisms other than release of H(2)S. In contrast to the parent drugs, S-NSAIDs, S-valproate, NaSH, and dithiolethiones were potent inhibitors of angiogenic responses in muscle and HT29 tumor explants assessed by 3-dimensional collagen matrix assays. Dithiolethiones and valproic acid were also potent inhibitors of developmental angiogenesis in zebrafish embryos, but the S-NSAIDs, remarkably, lacked this activity. CONCLUSIONS AND IMPLICATION: S-NSAIDs and S-valproate have potent anti-angiogenic activities mediated by their dithiole moieties. The novel properties of S-NSAIDs and S-valproate to inhibit pathological versus developmental angiogenesis suggest that these agents may have a role in cancer treatment.


Subject(s)
Anethole Trithione/pharmacology , Angiogenesis Inhibitors/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Valproic Acid/pharmacology , Animals , Cell Proliferation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , HSP27 Heat-Shock Proteins , Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Phosphorylation , Solubility , Zebrafish
5.
Proc Natl Acad Sci U S A ; 103(34): 12867-72, 2006 Aug 22.
Article in English | MEDLINE | ID: mdl-16908860

ABSTRACT

Alzheimer's disease is characterized by two primary pathological features: amyloid plaques and neurofibrillary tangles. The interconnection between amyloid and tau aggregates is of intense interest, but mouse models have yet to reveal a direct interrelationship. We now show that NO may be a key factor that connects amyloid and tau pathologies. Genetic removal of NO synthase 2 in mice expressing mutated amyloid precursor protein results in pathological hyperphosphorylation of mouse tau, its redistribution to the somatodendritic compartment in cortical and hippocampal neurons, and aggregate formation. Lack of NO synthase 2 in the amyloid precursor protein Swedish mutant mouse increased insoluble beta-amyloid peptide levels, neuronal degeneration, caspase-3 activation, and tau cleavage, suggesting that NO acts at a junction point between beta-amyloid peptides, caspase activation, and tau aggregation.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Gene Deletion , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Caspase 3 , Caspases/metabolism , Disease Models, Animal , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Phosphorylation , tau Proteins/metabolism
6.
J Neurochem ; 78(5): 1126-34, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11553686

ABSTRACT

Nitric oxide (NO) is an important regulator of NMDA channel function in the CNS. Recent findings suggest that nitroxyl anion (NO(-)) may also be generated by nitric oxide synthase, which catalyzes production of NO. Using recombinant NMDA receptors (NMDA-r) transfected into human embryonic kidney cells, our data demonstrate that the nitroxyl anion donor, Angeli's salt (AS; Na(2)N(2)O(3)) dramatically blocked glycine-independent desensitization in NMDA-r containing NR1-NR2A subunits. AS did not affect glycine-dependent desensitization, calcium dependent inactivation or glutamate affinity for the NMDA-r. This effect could be mimicked by treatment with DPTA, a metal chelator and was not evident under hypoxic conditions. In contrast, receptors containing the NR1-NR2B subunits demonstrated an approximate 25% reduction in whole cell currents in the presence of AS with no apparent change in desensitization. Our data suggest that the regulation of NMDA-r function by nitroxyl anion is distinctly different from NO and may result in different cellular outcomes compared with NO.


Subject(s)
Antioxidants/metabolism , Nitrogen Oxides/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Cell Hypoxia/physiology , Cell Line , Chelating Agents/pharmacology , Glutamic Acid/pharmacology , Glycine/metabolism , Humans , Kidney/cytology , Membrane Potentials/physiology , Nitric Oxide/metabolism , Nitrites/pharmacology , Oxygen Consumption/physiology , Patch-Clamp Techniques , Pentetic Acid/pharmacology , Receptors, N-Methyl-D-Aspartate/genetics , Transfection
7.
Proc Natl Acad Sci U S A ; 98(18): 10463-8, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11517312

ABSTRACT

Nitroxyl anion (NO(-)) is the one-electron reduction product of nitric oxide (NO( small middle dot)) and is enzymatically generated by NO synthase in vitro. The physiologic activity and mechanism of action of NO(-) in vivo remains unknown. The NO(-) generator Angeli's salt (AS, Na(2)N(2)O(3)) was administered to conscious chronically instrumented dogs, and pressure-dimension analysis was used to discriminate contractile from peripheral vascular responses. AS rapidly enhanced left ventricular contractility and concomitantly lowered cardiac preload volume and diastolic pressure (venodilation) without a change in arterial resistance. There were no associated changes in arterial or venous plasma cGMP. The inotropic response was similar despite reflex blockade with hexamethonium or volume reexpansion, indicating its independence from baroreflex stimulation. However, reflex activation did play a major role in the selective venodilation observed under basal conditions. These data contrasted with the pure NO donor diethylamine/NO, which induced a negligible inotropic response and a more balanced veno/arterial dilation. AS-induced positive inotropy, but not systemic vasodilatation, was highly redox-sensitive, being virtually inhibited by coinfusion of N-acetyl-l-cysteine. Cardiac inotropic signaling by NO(-) was mediated by calcitonin gene-related peptide (CGRP), as treatment with the selective CGRP-receptor antagonist CGRP(8-37) prevented this effect but not systemic vasodilation. Thus, NO(-) is a redox-sensitive positive inotrope with selective venodilator action, whose cardiac effects are mediated by CGRP-receptor stimulation. This fact is evidence linking NO(-) to redox-sensitive cardiac contractile modulation by nonadrenergic/noncholinergic peptide signaling. Given its cardiac and vascular properties, NO(-) may prove useful for the treatment of cardiovascular diseases characterized by cardiac depression and elevated venous filling pressures.


Subject(s)
Calcitonin Gene-Related Peptide/physiology , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Nitrogen Oxides/pharmacology , Animals , Anions , Baroreflex/drug effects , Baroreflex/physiology , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Gene-Related Peptide Receptor Antagonists , Cyclic GMP/physiology , Dogs , Male , Nitrates/blood , Nitric Oxide/pharmacology , Nitrites/blood , Nitrites/pharmacology , Nitrogen Oxides/metabolism , Oxidation-Reduction , Peptide Fragments/pharmacology , Signal Transduction
9.
J Am Chem Soc ; 123(23): 5473-81, 2001 Jun 13.
Article in English | MEDLINE | ID: mdl-11389629

ABSTRACT

Diazeniumdiolate ions of structure R(2)N[N(O)NO](-) (1) are of pharmacological interest because they spontaneously generate the natural bioregulatory species, nitric oxide (NO), when dissolved in aqueous media. Here we report the kinetic details for four representative reactivity patterns: (a) straightforward dissociation of the otherwise unfunctionalized diethylamine derivative 2 (anion 1, where R = Et) to diethylamine and NO; (b) results for the zwitterionic piperazin-1-yl analogue 4, for which the protonation state of the neighboring basic amine site is an important determinant of dissociation rate; (c) data for 5, a diazeniumdiolate derived from the polyamine spermine, whose complex rate equation can include terms for a variety of medium effects; and (d) the outcome for triamine 6 (R = CH(2)CH(2)NH(3)(+)), the most stable structure 1 ion identified to date. All of these dissociations are acid-catalyzed, with equilibrium protonation of the substrate preceding release of NO. Specific rate constants and pK(a) values for 2-6 have been determined from pH/rate profiles. Additionally, a hypsochromic shift (from approximately 250 to approximately 230 nm) was observed on acidifying these ions, allowing determination of a separate pK(a) for each substrate. For 6, the pK(a) value obtained kinetically was 2-3 pK(a) units higher than the value obtained from the spectral shift. Comparison of the ultraviolet spectra for 6 at various pH values with those for O- and N-alkylated diazeniumdiolates suggests that protonation at the R(2)N nitrogen initiates dissociation to NO at physiological pH, with a second protonation (at oxygen) accounting for both the spectral change and the enhanced dissociation rate at pH <4. Our results help to explain the previously noted variability in dissociation rate of 5, whose half-life we found to increase by an order of magnitude when its concentration was raised from near-zero to 1 mM, and provide mechanistic insight into the factors that govern dissociation rates among diazeniumdiolates of importance as pharmacologic progenitors of NO.


Subject(s)
Imides/chemistry , Indoles/chemistry , Nitric Oxide/chemistry , Hydrogen-Ion Concentration , Iron/pharmacology , Kinetics , Nitrosamines/chemistry , Piperazine , Piperazines/chemistry , Spermine/chemistry
10.
Antioxid Redox Signal ; 3(2): 203-13, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11396476

ABSTRACT

The Janus face of nitric oxide (NO) has prompted a debate as to whether NO plays a deleterious or protective role in tissue injury. There are a number of reactive nitrogen oxide species, such as N2O3 and ONOO-, that can alter critical cellular components under high local concentrations of NO. However, NO can also abate the oxidation chemistry mediated by reactive oxygen species such as H2O2 and O2- that occurs at physiological levels of NO. In addition to the antioxidant chemistry, NO protects against cell death mediated by H2O2, alkylhydroperoxides, and xanthine oxidase. The attenuation of metal/peroxide oxidative chemistry, as well as lipid peroxidation, appears to be the major chemical mechanisms by which NO may limit oxidative injury to mammalian cells. In addition to these chemical and biochemical properties, NO can modulate cellular and physiological processes to limit oxidative injury, limiting processes such as leukocyte adhesion. This review will address these aspects of the chemical biology of this multifaceted free radical and explore the beneficial effect of NO against oxidative stress.


Subject(s)
Antioxidants/metabolism , Nitric Oxide/metabolism , Animals , Cytotoxicity, Immunologic , Free Radicals , Humans , Lipid Peroxidation , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism
11.
J Biol Chem ; 276(32): 30085-91, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11404354

ABSTRACT

The quintessential nitrosating species produced during NO autoxidation is N(2)O(3). Nitrosation of amine, thiol, and hydroxyl residues can modulate critical cell functions. The biological mechanisms that control reactivity of nitrogen oxide species formed during autoxidation of nano- to micromolar levels of NO were examined using the synthetic donor NaEt(2)NN(O)NO (DEA/NO), human tumor cells, and 4,5-diaminofluorescein (DAF). Both the disappearance of NO and formation of nitrosated product from DAF in aerobic aqueous buffer followed second order processes; however, consumption of NO and nitrosation within intact cells were exponential. An optimal ratio of DEA/NO and 2-phenyl-4,4,5,5-tetramethylimidazole-1-oxyl 3-oxide (PTIO) was used to form N(2)O(3) through the intermediacy of NO(2). This route was found to be most reflective of the nitrosative mechanism within intact cells and was distinct from the process that occurred during autoxidation of NO in aqueous media. Manipulation of the endogenous scavengers ascorbate and glutathione indicated that the location, affinity, and concentration of these substances were key determinants in dictating nitrosative susceptibility of molecular targets. Taken together, these findings suggest that the functional effects of nitrosation may be organized to occur within discrete domains or compartments. Nitrosative stress may develop when scavengers are depleted and this architecture becomes compromised. Although NO(2) was not a component of aqueous NO autoxidation, the results suggest that the intermediacy of this species may be a significant factor in the advent of either nitrosation or oxidation chemistry in biological systems.


Subject(s)
Nitric Oxide/chemistry , Nitrogen Oxides/chemistry , Nitrogen Oxides/pharmacology , Ascorbic Acid/metabolism , Cyclic N-Oxides/pharmacology , Fluorescein/pharmacology , Glutathione/metabolism , Humans , Imidazoles/pharmacology , Indicators and Reagents/pharmacology , Kinetics , Models, Chemical , Nitric Oxide/metabolism , Nitrogen Oxides/metabolism , Nitrosation , Oxygen/metabolism , Protein Structure, Tertiary , Reactive Oxygen Species , Spectrometry, Fluorescence , Stress, Physiological , Time Factors , Tumor Cells, Cultured
12.
J Biol Chem ; 276(31): 28799-805, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11373284

ABSTRACT

Peroxynitrite (ONOO(-)/ONOOH), the product of the diffusion-limited reaction of nitric oxide (*NO) with superoxide (O(-*)(2)), has been implicated as an important mediator of tissue injury during conditions associated with enhanced *NO and O(-*)(2) production. Although several groups of investigators have demonstrated substantial oxidizing and cytotoxic activities of chemically synthesized peroxynitrite, others have proposed that the relative rates of *NO and production may be critical in determining the reactivity of peroxynitrite formed in situ (Miles, A. M., Bohle, D. S., Glassbrenner, P. A., Hansert, B., Wink, D. A., and Grisham, M. B. (1996) J. Biol. Chem. 271, 40-47). In the present study, we examined the mechanisms by which excess O(-*)(2) or *NO production inhibits peroxynitrite-mediated oxidation reactions. Peroxynitrite was generated in situ by the co-addition of a chemical source of *NO, spermineNONOate, and an enzymatic source of O(-*)(2), xanthine oxidase, with either hypoxanthine or lumazine as a substrate. We found that the oxidation of the model compound dihydrorhodamine by peroxynitrite occurred via the free radical intermediates OH and NO(2), formed during the spontaneous decomposition of peroxynitrite and not via direct reaction with peroxynitrite. The inhibitory effect of excess O(-*)(2) on the oxidation of dihydrorhodamine could not be ascribed to the accumulation of the peroxynitrite scavenger urate produced from the oxidation of hypoxanthine by xanthine oxidase. A biphasic oxidation profile was also observed upon oxidation of NADH by the simultaneous generation of *NO and O(-*)(2). Conversely, the oxidation of glutathione, which occurs via direct reaction with peroxynitrite, was not affected by excess production of *NO. We conclude that the oxidative processes initiated by the free radical intermediates formed from the decomposition of peroxynitrite are inhibited by excess production of *NO or O(-*)(2), whereas oxidative pathways involving a direct reaction with peroxynitrite are not altered. The physiological implications of these findings are discussed.


Subject(s)
Nitrates/chemistry , Nitric Oxide/chemistry , Superoxides/chemistry , Catalase/metabolism , Glutathione/chemistry , Humans , Hypoxanthine/metabolism , Kinetics , NAD/chemistry , Nitrogen Oxides , Oxidation-Reduction , Pteridines/metabolism , Recombinant Proteins/metabolism , Rhodamines/chemistry , Spermine/analogs & derivatives , Spermine/chemistry , Superoxide Dismutase/metabolism , Xanthine Oxidase/metabolism
13.
Neurosurgery ; 48(4): 884-92; discussion 892-3, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11322449

ABSTRACT

OBJECTIVE: Deleterious effects of strokes may be ameliorated when thrombolysis (i.e., with recombinant tissue plasminogen activator) restores circulation. However, reperfusion injury, mediated by oxygen free radicals (reactive oxygen species [ROS]), may limit the benefits of recombinant tissue plasminogen activator treatment. We hypothesized that, during reperfusion, exogenous nitric oxide (NO) would reduce stroke size by quenching ROS. METHODS: To investigate this hypothesis, we used two in vivo ischemia-reperfusion models, i.e., autologous cerebral embolism in rabbits and filament middle cerebral artery occlusion in rats. Using these models, we measured ROS levels (rabbit model) and stroke volumes (rat model) in response to transient ischemia, with and without intracarotid administration of ultrafast NO donor proline NO (proliNO). RESULTS: In the rabbit cerebral embolism model, intracarotid administration of proliNO (10(-6) mol/L) (n = 6) during reperfusion decreased free radical levels from 538 +/- 86 nmol/L in the vehicle-treated group (n = 7) to 186 +/- 31 nmol/L (2,3'-dihydroxybenzoic acid; P < 0.001) and from 521 +/- 86 nmol/L (n = 7) to 201 +/- 39 nmol/L (2,5'-dihydroxybenzoic acid; P < 0.002). In the rat middle cerebral artery occlusion model, intracarotid administration of proliNO (10(-5) mol/L) (n = 10) during reperfusion reduced the brain infarction volume from 256 +/- 48 mm3 in the vehicle-treated group (n = 8) to 187 +/- 41 mm3 (P < 0.005). In both experimental groups, intracarotid infusion of proliNO did not affect regional cerebral blood flow, mean arterial blood pressure, or brain and body temperatures. CONCLUSION: The beneficial effects of early restoration of cerebral circulation after cerebral ischemia were enhanced by intracarotid infusion of proliNO, most likely because of ROS scavenging by NO. These findings suggest the possibility of preventive treatment of reperfusion injury using NO donors.


Subject(s)
Infarction, Middle Cerebral Artery/physiopathology , Intracranial Embolism/physiopathology , Nitric Oxide Donors/pharmacology , Nitric Oxide/pharmacology , Proline/pharmacology , Reactive Oxygen Species/metabolism , Reperfusion Injury/physiopathology , Animals , Brain/blood supply , Female , Infarction, Middle Cerebral Artery/pathology , Injections, Intra-Arterial , Intracranial Embolism/pathology , Male , Nitrogen Oxides , Proline/analogs & derivatives , Rabbits , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Reperfusion Injury/pathology
15.
Nitric Oxide ; 5(1): 62-71, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11178938

ABSTRACT

Numerous methods are available for measurement of nitrate (NO(-)(3)). However, these assays can either be time consuming or require specialized equipment (e.g., nitrate reductase, chemiluminescent detector). We have developed a method for simultaneous evaluation of nitrate and nitrite concentrations in a microtiter plate format. The principle of this assay is reduction of nitrate by vanadium(III) combined with detection by the acidic Griess reaction. This assay is sensitive to 0.5 microM NO(-)(3) and is useful in a variety of fluids including cell culture media, serum, and plasma. S-Nitrosothiols and L-arginine derivatives were found to be potential interfering agents. However, these compounds are generally minor constituents of biological fluids relative to the concentration of nitrate/nitrite. This report introduces a new, convenient assay for the stable oxidation products of nitrogen oxide chemistry in biological samples.


Subject(s)
Nitrates/blood , Nitrites/blood , Spectrophotometry, Ultraviolet , Ethylenediamines , Free Radical Scavengers , Kinetics , Luminescent Measurements , Sensitivity and Specificity , Sulfanilamides , Temperature , Vanadium/pharmacology
16.
Free Radic Biol Med ; 30(3): 268-76, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11165873

ABSTRACT

The physiological function of nitric oxide (NO) in the defense against pathogens is multifaceted. The exact chemistry by which NO combats intracellular pathogens such as Listeria monocytogenes is yet unresolved. We examined the effects of NO exposure, either delivered by NO donors or generated in situ within ANA-1 murine macrophages, on L. monocytogenes growth. Production of NO by the two NONOate compounds PAPA/NO (NH2(C3H6)(N[N(O)NO]C3H7) and DEA/NO (Na(C2H5)2N[N(O)NO]) resulted in L. monocytogenes cytostasis with minimal cytotoxicity. Reactive oxygen species generated from xanthine oxidase/hypoxanthine were neither bactericidal nor cytostatic and did not alter the action of NO. L. monocytogenes growth was also suppressed upon internalization into ANA-1 murine macrophages primed with interferon-gamma (INF-gamma) + tumor necrosis factor-alpha (TNF-alpha or INF-gamma + lipid polysaccharide (LPS). Growth suppression correlated with nitrite formation and nitrosation of 2,3-diaminonaphthalene elicited by stimulated murine macrophages. This nitrosative chemistry was not dependent upon nor mediated by interaction with reactive oxygen species (ROS), but resulted solely from NO and intermediates related to nitrosative stress. The role of nitrosation in controlling L. monocytogenes was further examined by monitoring the effects of exposure to NO on an important virulence factor, Listeriolysin O, which was inhibited under nitrosative conditions. These results suggest that nitrosative stress mediated by macrophages is an important component of the immunological arsenal in controlling L. monocytogenes infections.


Subject(s)
2-Naphthylamine/analogs & derivatives , Listeria monocytogenes/growth & development , Macrophages/metabolism , Macrophages/microbiology , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Oxidative Stress , 2-Naphthylamine/metabolism , Animals , Cell Line , Hydrazines/pharmacology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Listeria monocytogenes/drug effects , Mice , Mice, Knockout , Nitric Oxide/pharmacology , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type II , Nitrites/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Xanthine/metabolism , Xanthine Oxidase/metabolism
17.
J Biol Chem ; 276(3): 1720-7, 2001 Jan 19.
Article in English | MEDLINE | ID: mdl-11042174

ABSTRACT

The nitroxyl anion (NO-) is a highly reactive molecule that may be involved in pathophysiological actions associated with increased formation of reactive nitrogen oxide species. Angeli's salt (Na2N2O3; AS) is a NO- donor that has been shown to exert marked cytotoxicity. However, its decomposition intermediates have not been well characterized. In this study, the chemical reactivity of AS was examined and compared with that of peroxynitrite (ONOO-) and NO/N2O3. Under aerobic conditions, AS and ONOO- exhibited similar and considerably higher affinities for dihydrorhodamine (DHR) than NO/N2O3. Quenching of DHR oxidation by azide and nitrosation of diaminonaphthalene were exclusively observed with NO/N2O3. Additional comparison of ONOO- and AS chemistry demonstrated that ONOO- was a far more potent one-electron oxidant and nitrating agent of hydroxyphenylacetic acid than was AS. However, AS was more effective at hydroxylating benzoic acid than was ONOO-. Taken together, these data indicate that neither NO/N2O3 nor ONOO- is an intermediate of AS decomposition. Evaluation of the stoichiometry of AS decomposition and O2 consumption revealed a 1:1 molar ratio. Indeed, oxidation of DHR mediated by AS proved to be oxygen-dependent. Analysis of the end products of AS decomposition demonstrated formation of NO2- and NO3- in approximately stoichiometric ratios. Several mechanisms are proposed for O2 adduct formation followed by decomposition to NO3- or by oxidation of an HN2O3- molecule to form NO2-. Given that the cytotoxicity of AS is far greater than that of either NO/N2O3 or NO + O2, this study provides important new insights into the implications of the potential endogenous formation of NO- under inflammatory conditions in vivo.


Subject(s)
Nitrogen Oxides/chemistry , Nitrates/chemistry , Oxidation-Reduction , Rhodamines/chemistry
18.
Curr Protoc Toxicol ; Chapter 10: Unit 10.8, 2001 May.
Article in English | MEDLINE | ID: mdl-20957636

ABSTRACT

NO-derived intermediates formed under aerobic conditions may engage in complex chemical reactions with biologically important molecules. The outcomes of these reactions and their ultimate effect on biological systems depend on the selectivity of the species and the concentrations of different substances present and whether the reaction takes place in the gas or aqueous phase. In this unit conversion of two different compounds to fluorescent products is used to distinguish between oxidative and nitrosative chemistry of different reactive nitrogen oxide species.


Subject(s)
Nitric Oxide/chemistry , Reactive Nitrogen Species/chemistry , Fluorescence , Oxidation-Reduction
19.
Curr Protoc Toxicol ; Chapter 10: Unit 10.4, 2001 May.
Article in English | MEDLINE | ID: mdl-23045026

ABSTRACT

Recently fluorometric techniques have been developed to determine NO and S-nitrosothiols with improved sensitivity over spectrophotometric techniques based on similar chemistry. This unit contains protocols for detection and quantification of NO from chemical reactions and from cellular systems, and of S-nitrosothiols in chemical, biochemical, and cellular experiments.


Subject(s)
Nitric Oxide/analysis , Nitric Oxide/metabolism , Animals , Fluorometry , Humans , Kinetics , Nitrates/analysis , Nitrates/metabolism , Nitrosation , S-Nitrosothiols/analysis , S-Nitrosothiols/metabolism
20.
Br J Haematol ; 110(2): 412-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10971400

ABSTRACT

A mechanism has been proposed in which nitric oxide (NO) may bind to cysteine beta93 and be transported by haemoglobin from the lungs to the tissues and modify vascular tone. In addition, it has been reported that treatment of sickle cell anaemia blood with 80 p.p.m. NO gas in air shifts the oxygen affinity, as measured by P50 to the left. We exposed normal and sickle cell anaemia blood to 80 p.p.m. NO in air for 1 h in vitro and found no change in P50 of either normal or sickle cell blood. In addition, we exposed normal and sickle cell blood in buffer to aqueous NO (NO gas dissolved in buffer) at varying concentrations and found that the induced left shift in P50 correlates strongly and linearly with methaemoglobin formation. We also treated normal and sickle cell blood with other nitric oxide donors, such as sodium 2-(N, N-diethylamino)-diazenolate-2-oxide (DEANO), S-nitrosocysteine (CysNO) and sodium trioxodinitrate (OXINO, or Angeli's salt). In all cases, we found a dose-dependent increase in methaemoglobin that was strongly correlated with the dose-dependent P50 reduction. Our data do not support the report that low NO concentrations can selectively increase the oxygen affinity of sickle cell blood without affecting methaemoglobin levels significantly. NO, however, may have benefit in sickle cell disease by other mechanisms.


Subject(s)
Anemia, Sickle Cell/blood , Nitric Oxide Donors/pharmacology , Nitric Oxide/pharmacology , Oxygen/blood , Anemia, Sickle Cell/metabolism , Case-Control Studies , Dose-Response Relationship, Drug , Humans , Methemoglobin/biosynthesis , Methemoglobin/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...