Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 174
Filter
1.
Int J Mol Sci ; 21(19)2020 Sep 27.
Article in English | MEDLINE | ID: mdl-32992650

ABSTRACT

A tight junction (TJ) makes a physical barrier in the epidermal cells of skin. Ultraviolet (UV) light may disrupt the TJ barrier, but the mechanism has not been well clarified. Weak UVB (5 mJ/cm2) caused mislocalization of claudin-1 (CLDN1), a component of the TJ strand, and disruption of TJ barrier in human keratinocyte-derived HaCaT cells. The UVB-induced mislocalization of CLDN1 was inhibited by monodansylcadaverine (MDC), a clathrin-dependent endocytosis inhibitor, suggesting that UVB enhances the internalization of CLDN1. Transepidermal electrical resistance and paracellular flux of lucifer yellow, a fluorescent hydrophilic marker, were rescued by MDC. UVB changed neither the total nor phosphorylation levels of CLDN1, but it increased both mono-ubiquitination and tyrosine nitration levels of CLDN1. Fluorescence measurements revealed that UVB increased intracellular free Ca2+, nitric oxide (NO), and peroxynitrite contents, which were inhibited by Opsin2 (OPN2) siRNA, suggesting that OPN2 functions as a UVB sensor. The effects of UVB were inhibited by an antagonist of transient receptor potential type vanilloid 1 (TRPV1) and Ca2+ chelator. Both NO donor and peroxynitrite donor induced the mislocalization of CLDN1 and disruption of TJ barrier, which were rescued by a NO synthase (NOS) inhibitor and a peroxynitrite scavenger. Weak UVB irradiation induced the disruption of TJ barrier mediated by mislocalization of CLDN1 in HaCaT cells. The OPN2/TRPV1/NOS signaling pathway may be a novel target for preventing destruction of the TJ barrier by UVB irradiation.


Subject(s)
Claudin-1/metabolism , Keratinocytes/metabolism , Nitric Oxide/metabolism , Peroxynitrous Acid/biosynthesis , Signal Transduction/radiation effects , Ultraviolet Rays , Cadaverine/analogs & derivatives , Cadaverine/pharmacology , Cell Survival/radiation effects , Endocytosis/drug effects , HaCaT Cells , Humans , Nitric Oxide Synthase/metabolism , Phosphorylation/radiation effects , Signal Transduction/drug effects , TRPV Cation Channels/metabolism , Tight Junctions/metabolism , Tight Junctions/radiation effects , Ubiquitination/radiation effects
2.
Chem Commun (Camb) ; 56(18): 2759-2762, 2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32022003

ABSTRACT

We describe a colorimetric and fluorescent probe 3a to detect cellular peroxynitrite with high selectivity and sensitivity. 3a was successfully applied in the bioimaging of exogenous and endogenous peroxynitrite in living cells. The up-regulation of peroxynitrite in cancer cells and normal cells during 5-fluorouracil treatment was finally monitored.


Subject(s)
Antineoplastic Agents/pharmacology , Fluorescent Dyes/pharmacology , Fluorouracil/pharmacology , Peroxynitrous Acid/biosynthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fluorescent Dyes/chemistry , Fluorouracil/chemistry , Humans , MCF-7 Cells , Molecular Structure , Optical Imaging , Peroxynitrous Acid/chemistry , Structure-Activity Relationship
3.
J Agric Food Chem ; 67(22): 6407-6413, 2019 Jun 05.
Article in English | MEDLINE | ID: mdl-31083940

ABSTRACT

The precise cellular function of peroxynitrite (ONOO-) in biosystems remains elusive, primarily owing to being short of ultrasensitive techniques for monitoring its intracellular distribution. In this work, a novel rhodamine B cyclic 1,2-dimethylhydrazine fluorescent chemodosimeter RDMH-PN for highly specific and ultrasensitive monitoring of basal ONOO- in biosystems was rationally designed. The fluorescence titration experiments demonstrated that RDMH-PN was capable of quantitatively detecting 0-100 nM ONOO- (limit of detection = 0.68 nM). In addition, RDMH-PN has outstanding performances of ultrafast measurement, naked-eye detection, and preeminent selectivity toward ONOO- to accurately detect intracellular basal ONOO-. Finally, it has been confirmed that RDMH-PN could not only map the intracellular basal ONOO- level by inhibition tests but also trace the fluctuations of endogenous and exogenous ONOO- levels with diverse stimulations in live cells and zebrafish.


Subject(s)
Dimethylhydrazines/chemistry , Fluorescent Dyes/chemistry , Peroxynitrous Acid/analysis , Spectrometry, Fluorescence/methods , Zebrafish/metabolism , Animals , Macrophages/chemistry , Macrophages/cytology , Macrophages/metabolism , Mice , Peroxynitrous Acid/biosynthesis , RAW 264.7 Cells , Rhodamines/chemistry , Spectrometry, Fluorescence/instrumentation
4.
Georgian Med News ; (280-281): 173-178, 2018.
Article in English | MEDLINE | ID: mdl-30204120

ABSTRACT

Today we know that NO· and ONOO- are clue pathophysiological factors for progression some ischemic diseases of the central nervous system. So investigation of the antioxidants which will be able to decrease NO· and ONOO- toxicity seems to be very of current interest. The six esters and three amides of 2-(3,4-dihydro-3-oxo-2H-[1,2,4]triazino[4,3-c]quinazolin-4-yl)acetic acid were synthesized for this study, and we showed evidence of antioxidant activity of these new original derivatives. We studied the effect of 2-(3,4-dihydro-3-oxo-2H-[1,2,4]triazino[4,3-c]quinazolin-4-yl)acetic acid derivatives on superoxide dismutase activity under the condition of excessive NO· and ONOO- production. NO· induction was performed by the action of light on sodium nitroprusside Na2[Fe(NO)(CN)5]×2H2O in vitro. Also, the investigation of the substances was carried out in the brain supernatant obtained from the white Wistar rats in vivo. For nitrosative stress modeling dinitrozolic complex of Fe2+ and cysteine were utilized. Our data showed that 2-(3,4-dihydro-3-oxo-2H-[1,2,4]triazino[4,3-c]quinazolin-4-yl)acetic acid is not active compound while its esters and amides have antioxidant activity. Compound benzyl ester of this acid revealed the most effective antioxidant activity.


Subject(s)
Acetates/pharmacology , Antioxidants/pharmacology , Nitrosative Stress/drug effects , Quinazolines/pharmacology , Triazines/pharmacology , Acetates/chemical synthesis , Amides/chemical synthesis , Amides/pharmacology , Animals , Antioxidants/chemical synthesis , Brain/metabolism , Esters/chemical synthesis , Esters/pharmacology , Male , Nitric Oxide/biosynthesis , Peroxynitrous Acid/biosynthesis , Quinazolines/chemical synthesis , Rats, Wistar , Structure-Activity Relationship , Superoxide Dismutase/metabolism , Triazines/chemical synthesis , Tyrosine/analogs & derivatives , Tyrosine/metabolism
5.
J Cell Mol Med ; 22(10): 4948-4962, 2018 10.
Article in English | MEDLINE | ID: mdl-30063118

ABSTRACT

Branched-chain amino acids (BCAA: leucine, isoleucine and valine) are essential amino acids implicated in glucose metabolism and maintenance of correct brain function. Elevated BCAA levels can promote an inflammatory response in peripheral blood mononuclear cells. However, there are no studies analysing the direct effects of BCAA on endothelial cells (ECs) and its possible modulation of vascular function. In vitro and ex vivo studies were performed in human ECs and aorta from male C57BL/6J mice, respectively. In ECs, BCAA (6 mmol/L) increased eNOS expression, reactive oxygen species production by mitochondria and NADPH oxidases, peroxynitrite formation and nitrotyrosine expression. Moreover, BCAA induced pro-inflammatory responses through the transcription factor NF-κB that resulted in the release of intracellular adhesion molecule-1 and E-selectin conferring endothelial activation and adhesion capacity to inflammatory cells. Pharmacological inhibition of mTORC1 intracellular signalling pathway decreased BCAA-induced pro-oxidant and pro-inflammatory effects in ECs. In isolated murine aorta, BCAA elicited vasoconstrictor responses, particularly in pre-contracted vessels and after NO synthase blockade, and triggered endothelial dysfunction, effects that were inhibited by different antioxidants, further demonstrating the potential of BCAA to induce oxidative stress with functional impact. In summary, we demonstrate that elevated BCAA levels generate inflammation and oxidative stress in ECs, thereby facilitating inflammatory cells adhesion and endothelial dysfunction. This might contribute to the increased cardiovascular risk observed in patients with elevated BCAA blood levels.


Subject(s)
Amino Acids, Branched-Chain/metabolism , Aorta/metabolism , Endothelial Cells/drug effects , Inflammation/metabolism , Animals , Antioxidants/administration & dosage , Aorta/drug effects , E-Selectin/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Glucose/metabolism , Humans , Inflammation/genetics , Inflammation/pathology , Intercellular Adhesion Molecule-1/genetics , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , NF-kappa B/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Peroxynitrous Acid/biosynthesis , Peroxynitrous Acid/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tyrosine/analogs & derivatives , Tyrosine/biosynthesis , Tyrosine/metabolism , Vasoconstrictor Agents/administration & dosage
6.
Molecules ; 22(3)2017 Mar 08.
Article in English | MEDLINE | ID: mdl-28282895

ABSTRACT

Administration of low-dose endotoxin (lipopolysaccharide, LPS) 24 h before a lethal ischemia induces pharmacological late preconditioning. The exact mechanism of this phenomenon is not clear. Here we aimed to investigate whether low-dose LPS exerts late effects on peroxynitrite formation and activation of Akt, Erk, and STAT3 in the heart. Male Wistar rats were injected with LPS (S. typhimurium; 0.5 mg/kg i.p.) or saline. Twenty-four hours later, hearts were isolated, perfused for 10 min, and then used for biochemical analyses. LPS pretreatment enhanced cardiac formation of the peroxynitrite marker 3-nitrotyrosine. LPS pretreatment also increased cardiac levels of the peroxynitrite precursor nitric oxide (NO) and superoxide. The activities of Ca2+-independent NO synthase and xanthine oxidoreductase increased in LPS-pretreated hearts. LPS pretreatment resulted in significantly enhanced phosphorylation of STAT3 and non-significantly increased phosphorylation of Akt without affecting the activation of Erk. In separate experiments, isolated working hearts were subjected to 30 min global ischemia and 20 min reperfusion. LPS pretreatment significantly improved ischemia-reperfusion-induced deterioration of cardiac function. We conclude that LPS pretreatment enhances cardiac peroxynitrite formation and activates STAT3 24 h later, which may contribute to LPS-induced late preconditioning.


Subject(s)
Endotoxins/administration & dosage , Ischemic Preconditioning, Myocardial , Myocardial Ischemia/metabolism , Peroxynitrous Acid/biosynthesis , STAT3 Transcription Factor/metabolism , Animals , Lactate Dehydrogenases/metabolism , Lipopolysaccharides/administration & dosage , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Oxidation-Reduction , Rats , Tyrosine/analogs & derivatives , Tyrosine/biosynthesis
7.
Eur J Pharmacol ; 802: 7-19, 2017 May 05.
Article in English | MEDLINE | ID: mdl-28228357

ABSTRACT

Mammalian target of rapamycin (mTOR), a serine/threonine kinase plays an important role in various pathophysiological processes including cancer, metabolic diseases, and inflammation. Although mTOR participates in Toll-like receptor 4 signalling in different cell types, the role of this enzyme in sepsis pathogenesis and its effects on hypotension and inflammation in endotoxemic rats remains unclear. In this study we investigated the effects of mTOR inhibition on lipopolysaccharide (LPS)-induced changes on expressions and/or activities of ribosomal protein S6 (rpS6), an mTOR substrate, nuclear factor-κB (NF-κB) p65, inhibitor κB (IκB)-α, inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2 with production of nitric oxide, peroxynitrite, prostacyclin, and tumor necrosis factor (TNF)-α and activity of myeloperoxidase (MPO), which results in hypotension and inflammation. Injection of LPS (10mg/kg, i.p.) to male Wistar rats decreased blood pressure and increased heart rate that were associated with elevated nitrotyrosine, 6-keto-PGF1α, and TNF-α levels and MPO activity, and increased expressions and/or activities of rpS6, NF-κB p65, iNOS, and COX-2 and decreased expression of IκB-α in renal, cardiac, and vascular tissues. LPS also increased serum and tissue nitrite levels. Rapamycin (1mg/kg, i.p.) given one h after injection of LPS reversed these effects of LPS. These data suggest that the activation of mTOR/IκB-α/NF-κB pathway associated with vasodilator and proinflammatory mediator formation contributes to LPS-induced hypotension and inflammation.


Subject(s)
Hypotension/chemically induced , Hypotension/pathology , I-kappa B Proteins/metabolism , Lipopolysaccharides/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factor RelA/metabolism , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Arterial Pressure/drug effects , Cyclooxygenase 2/metabolism , Epoprostenol/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Heart Rate/drug effects , Hypotension/metabolism , Hypotension/physiopathology , Inflammation/chemically induced , Inflammation/pathology , Male , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Peroxidase/metabolism , Peroxynitrous Acid/biosynthesis , Rats , Rats, Wistar , Ribosomal Protein S6/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tyrosine/analogs & derivatives , Tyrosine/metabolism
8.
Free Radic Biol Med ; 102: 217-228, 2017 01.
Article in English | MEDLINE | ID: mdl-27838434

ABSTRACT

The molecular mechanisms by which the endothelial barrier becomes compromised during lipopolysaccharide (LPS) mediated acute lung injury (ALI) are still unresolved. We have previously reported that the disruption of the endothelial barrier is due, at least in part, to the uncoupling of endothelial nitric oxide synthase (eNOS) and increased peroxynitrite-mediated nitration of RhoA. The purpose of this study was to elucidate the molecular mechanisms by which LPS induces eNOS uncoupling during ALI. Exposure of pulmonary endothelial cells (PAEC) to LPS increased pp60Src activity and this correlated with an increase in nitric oxide (NO) production, but also an increase in NOS derived superoxide, peroxynitrite formation and 3-nitrotyrosine (3-NT) levels. These effects could be simulated by the over-expression of a constitutively active pp60Src (Y527FSrc) mutant and attenuated by over-expression of dominant negative pp60Src mutant or reducing pp60Src expression. LPS induces both RhoA nitration and endothelial barrier disruption and these events were attenuated when pp60Src expression was reduced. Endothelial NOS uncoupling correlated with an increase in the levels of asymmetric dimethylarginine (ADMA) in both LPS exposed and Y527FSrc over-expressing PAEC. The effects in PAEC were also recapitulated when we transiently over-expressed Y527FSrc in the mouse lung. Finally, we found that the pp60-Src-mediated decrease in DDAH activity was mediated by the phosphorylation of DDAH II at Y207 and that a Y207F mutant DDAH II was resistant to pp60Src-mediated inhibition. We conclude that pp60Src can directly inhibit DDAH II and this is involved in the increased ADMA levels that enhance eNOS uncoupling during the development of ALI.


Subject(s)
Acute Lung Injury/metabolism , Amidohydrolases/genetics , Nitric Oxide Synthase Type III/genetics , Nitric Oxide/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Amidohydrolases/metabolism , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation , Lipopolysaccharides/toxicity , Mice , Mitochondria/metabolism , Mitochondria/pathology , Mutation , Nitric Oxide Synthase Type III/metabolism , Peroxynitrous Acid/biosynthesis , Phosphorylation , Proto-Oncogene Proteins pp60(c-src)/metabolism , Superoxides/metabolism
9.
J Cardiovasc Pharmacol ; 68(5): 334-341, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27824721

ABSTRACT

Numerous studies have found that the age-associated structural and functional alterations in arteries were characterized by increased endothelial dysfunction. In this study, young (3 months), adult (9 months), and aging (20 months) male Sprague-Dawley rats were randomly divided into 6 groups, including control groups and FeTMPyP (peroxynitrite scavenger) groups receiving saline and FeTMPyP, respectively, for 5 administrations once every 3 days through intraperitoneal injection. The aged-related proteins beta-galactosidase, p53, and p16 as well as the nitrotyrosine and endothelial marker endothelial nitric oxide synthase and von Willebrand factor (vWF) in vascular tissues were measured by immunohistochemistry. Endothelium-dependent vasorelaxation and endothelium-independent vasorelaxation of rat thoracic aortas and mesenteric arteries were measured by acetylcholine and sodium nitroprusside, respectively. The amount of circulating endothelial progenitor cells (EPCs) was determined by flow cytometry. The endothelium-dependent/independent relaxation in mesenteric arteries and the amount of circulating EPCs (CD31/CD34) in peripheral blood of aging rats were reduced significantly compared with young and adult rats. Immunohistochemistry results showed that the nitrotyrosine levels and morphological damage in mesenteric arteries were increased significantly in aging rats. Adoption of peroxynitrite scavenger FeTMPyP intervention may not only improve the endothelium-dependent relaxation and the amount of circulating EPCs in aging rats but also reverse endothelial injury. In conclusion, this study demonstrates that enhanced nitrative stress may aggravate the endothelial injury and vascular dysfunction of resistance arteries in aging rats. Antiperoxynitrite treatment can ameliorate the vasorelaxation and may be involved with the protection of circulating EPCs.


Subject(s)
Aging/drug effects , Endothelial Progenitor Cells/drug effects , Endothelium, Vascular/drug effects , Peroxynitrous Acid/antagonists & inhibitors , Vascular Resistance/drug effects , Vasodilation/drug effects , Aging/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Dose-Response Relationship, Drug , Endothelial Progenitor Cells/metabolism , Endothelium, Vascular/physiology , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Metalloporphyrins/pharmacology , Organ Culture Techniques , Peroxynitrous Acid/biosynthesis , Rats , Rats, Sprague-Dawley , Treatment Outcome , Vascular Resistance/physiology , Vasodilation/physiology , Vasodilator Agents/pharmacology
10.
PLoS One ; 11(9): e0162497, 2016.
Article in English | MEDLINE | ID: mdl-27598576

ABSTRACT

Microglial priming and enhanced reactivity to secondary insults cause substantial neuronal damage and are hallmarks of brain aging, traumatic brain injury and neurodegenerative diseases. It is, thus, of particular interest to identify mechanisms involved in microglial priming. Here, we demonstrate that priming of microglia with interferon-γ (IFN γ) substantially enhanced production of reactive oxygen species (ROS) following stimulation of microglia with ATP. Priming of microglial ROS production was substantially reduced by inhibition of p38 MAPK activity with SB203580, by increases in intracellular glutathione levels with N-Acetyl-L-cysteine, by blockade of NADPH oxidase subunit NOX2 activity with gp91ds-tat or by inhibition of nitric oxide production with L-NAME. Together, our data indicate that priming of microglial ROS production involves reduction of intracellular glutathione levels, upregulation of NADPH oxidase subunit NOX2 and increases in nitric oxide production, and suggest that these simultaneously occurring processes result in enhanced production of neurotoxic peroxynitrite. Furthermore, IFNγ-induced priming of microglial ROS production was reduced upon blockade of Kir2.1 inward rectifier K+ channels with ML133. Inhibitory effects of ML133 on microglial priming were mediated via regulation of intracellular glutathione levels and nitric oxide production. These data suggest that microglial Kir2.1 channels may represent novel therapeutic targets to inhibit excessive ROS production by primed microglia in brain pathology.


Subject(s)
Interferon-gamma/pharmacology , Microglia/drug effects , Nitric Oxide Synthase Type II/genetics , Nitric Oxide/biosynthesis , Potassium Channels, Inwardly Rectifying/genetics , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Glutathione/agonists , Glutathione/antagonists & inhibitors , Glutathione/biosynthesis , Glycoproteins/pharmacology , Imidazoles/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Microglia/cytology , Microglia/metabolism , NADPH Oxidase 2 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/agonists , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Peroxynitrous Acid/agonists , Peroxynitrous Acid/antagonists & inhibitors , Peroxynitrous Acid/biosynthesis , Phenanthrolines/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/metabolism , Pyridines/pharmacology , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Signal Transduction , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Free Radic Biol Med ; 101: 284-295, 2016 12.
Article in English | MEDLINE | ID: mdl-27641237

ABSTRACT

The specific and sensitive detection of peroxynitrite (ONOO-/ONOOH) in biological systems is a great challenge due to its high reactivity towards several biomolecules. Herein, we validated the advantages of using fluorescein-boronate (Fl-B) as a highly sensitive fluorescent probe for the direct detection of peroxynitrite under biologically-relevant conditions in two different cell models. The synthesis of Fl-B was achieved by a very simply two-step conversion synthetic route with high purity (>99%) and overall yield (∼42%). Reactivity analysis of Fl-B with relevant biological oxidants including hydrogen peroxide (H2O2), hypochlorous acid (HOCl) and peroxynitrite were performed. The rate constant for the reaction of peroxynitrite with Fl-B was 1.7×106M-1s-1, a million times faster than the rate constant measured for H2O2 (k=1.7M-1s-1) and 2,700 faster than HOCl (6.2×102M-1s-1) at 37°C and pH 7.4. The reaction of Fl-B with peroxynitrite was significant even in the presence of physiological concentrations of CO2, a well-known peroxynitrite reactant. Experimental and simulated kinetic analyses confirm that the main oxidation process of Fl-B takes place with peroxynitrite itself via a direct bimolecular reaction and not with peroxynitrite-derived radicals. Fl-B was successfully applied for the detection of endogenously-generated peroxynitrite by endothelial cells and in macrophage-phagocyted parasites. Moreover, the generated data allowed estimating the actual intracellular flux of peroxynitrite. For instance, ionomycin-stimulated endothelial cells generated peroxynitrite at a rate of ∼ 0.1µMs-1, while immunostimulated macrophages do so in the order of ∼1µMs-1 inside T. cruzi-infected phagosomes. Fl-B revealed not to be toxic in concentrations up to 1mM for 24h. Cellular peroxynitrite detection was achieved by conventional laboratory fluorescence-based methods including flow cytometry and epi-fluorescence microscopy. Fl-B was shown to be more sensitive than the coumarin boronate due to a higher molar absorption coefficient and quantum yield. Overall, our results show that Fl-B is a kinetically selective and highly sensitive probe for the direct detection of cell-derived peroxynitrite.


Subject(s)
Boronic Acids/chemistry , Fluoresceins/chemistry , Fluorescent Dyes/chemical synthesis , Macrophages/metabolism , Peroxynitrous Acid/analysis , Animals , Aorta/cytology , Aorta/metabolism , Cattle , Cell Line , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hydrogen Peroxide/metabolism , Hypochlorous Acid/metabolism , Kinetics , Macrophages/cytology , Macrophages/parasitology , Mice , Oxidation-Reduction , Peroxynitrous Acid/biosynthesis , Phagocytosis/physiology , Primary Cell Culture , Sensitivity and Specificity , Trypanosoma cruzi
12.
J Am Heart Assoc ; 5(6)2016 06 13.
Article in English | MEDLINE | ID: mdl-27412900

ABSTRACT

BACKGROUND: Light chain amyloidosis (AL) is associated with high mortality, especially in patients with advanced cardiovascular involvement. It is caused by toxicity of misfolded light chain proteins (LC) in vascular, cardiac, and other tissues. There is no treatment to reverse LC tissue toxicity. We tested the hypothesis that nanoliposomes composed of monosialoganglioside, phosphatidylcholine, and cholesterol (GM1 ganglioside-containing nanoliposomes [NLGM1]) can protect against LC-induced human microvascular dysfunction and assess mechanisms behind the protective effect. METHODS AND RESULTS: The dilator responses of ex vivo abdominal adipose arterioles from human participants without AL to acetylcholine and papaverine were measured before and after exposure to LC (20 µg/mL) with or without NLGM1 (1:10 ratio for LC:NLGM1 mass). Human umbilical vein endothelial cells were exposed for 18 to 20 hours to vehicle, LC with or without NLGM1, or NLGM1 and compared for oxidative and nitrative stress response and cellular viability. LC impaired arteriole dilator response to acetylcholine, which was restored by co-treatment with NLGM1. LC decreased endothelial cell nitric oxide production and cell viability while increasing superoxide and peroxynitrite; these adverse effects were reversed by NLGM1. NLGM1 increased endothelial cell protein expression of antioxidant enzymes heme oxygenase 1 and NAD(P)H quinone dehydrogenase 1 and increased nuclear factor, erythroid 2 like 2 (Nrf-2) protein. Nrf-2 gene knockdown reduced antioxidant stress response and reversed the protective effects of NLGM1. CONCLUSIONS: NLGM1 protects against LC-induced human microvascular endothelial dysfunction through increased nitric oxide bioavailability and reduced oxidative and nitrative stress mediated by Nrf-2-dependent antioxidant stress response. These findings point to a potential novel therapeutic approach for light chain amyloidosis.


Subject(s)
Cholesterol/administration & dosage , Endothelium, Vascular/drug effects , Gangliosides/administration & dosage , Immunoglobulin Light-chain Amyloidosis/complications , Phosphatidylcholines/administration & dosage , Vascular Diseases/prevention & control , Adipose Tissue/blood supply , Arterioles/drug effects , Arterioles/physiology , Cell Survival/physiology , Drug Combinations , Endothelial Cells/metabolism , Gene Knockdown Techniques/methods , Human Umbilical Vein Endothelial Cells , Humans , Immunoglobulin Light-chain Amyloidosis/prevention & control , Male , Middle Aged , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/genetics , Nanoparticles/administration & dosage , Nitric Oxide/physiology , Nitric Oxide Synthase Type III/metabolism , Papaverine/pharmacology , Peroxynitrous Acid/biosynthesis , RNA Interference/physiology , RNA, Small Interfering/physiology , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Transfection , Vascular Diseases/physiopathology , Vasodilator Agents/pharmacology
13.
DNA Repair (Amst) ; 44: 169-177, 2016 08.
Article in English | MEDLINE | ID: mdl-27262612

ABSTRACT

Sunlight's ultraviolet wavelengths induce cyclobutane pyrimidine dimers (CPDs), which then cause mutations that lead to melanoma or to cancers of skin keratinocytes. In pigmented melanocytes, we found that CPDs arise both instantaneously and for hours after UV exposure ends. Remarkably, the CPDs arising in the dark originate by a novel pathway that resembles bioluminescence but does not end in light: First, UV activates the enzymes nitric oxide synthase (NOS) and NADPH oxidase (NOX), which generate the radicals nitric oxide (NO) and superoxide (O2(-)); these combine to form the powerful oxidant peroxynitrite (ONOO(-)). A fragment of the skin pigment melanin is then oxidized, exciting an electron to an energy level so high that it is rarely seen in biology. This process of chemically exciting electrons, termed "chemiexcitation", is used by fireflies to generate light but it had never been seen in mammalian cells. In melanocytes, the energy transfers radiationlessly to DNA, inducing CPDs. Chemiexcitation is a new source of genome instability, and it calls attention to endogenous mechanisms of genome maintenance that prevent electronic excitation or dissipate the energy of excited states. Chemiexcitation may also trigger pathogenesis in internal tissues because the same chemistry should arise wherever superoxide and nitric oxide arise near cells that contain melanin.


Subject(s)
Electrons , Melanins/chemistry , Melanoma/chemistry , Neoplasms, Radiation-Induced/chemistry , Peroxynitrous Acid/chemistry , Skin Neoplasms/chemistry , DNA Damage , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/radiation effects , Melanins/agonists , Melanins/metabolism , Melanoma/etiology , Melanoma/metabolism , Melanoma/pathology , NADPH Oxidases/metabolism , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/metabolism , Neoplasms, Radiation-Induced/pathology , Nitric Oxide/biosynthesis , Nitric Oxide/chemistry , Nitric Oxide Synthase/metabolism , Peroxynitrous Acid/biosynthesis , Pyrimidine Dimers/biosynthesis , Pyrimidine Dimers/chemistry , Skin/metabolism , Skin/pathology , Skin/radiation effects , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Sunlight/adverse effects , Superoxides/chemistry , Superoxides/metabolism , Ultraviolet Rays/adverse effects
14.
Eur J Pharmacol ; 783: 11-22, 2016 Jul 15.
Article in English | MEDLINE | ID: mdl-27118175

ABSTRACT

Diabetes mellitus is associated with reactive oxygen and nitrogen species accumulation. Behavioral stress increases nitric oxide production, which may trigger a massive impact on vascular cells and accelerate cardiovascular complications under oxidative stress conditions such as Diabetes. For this study, type-1 Diabetes mellitus was induced in Wistar rats by intraperitoneal injection of streptozotocin. After 28 days, cumulative concentration-response curves for angiotensin II were obtained in endothelium-intact carotid rings from diabetic rats that underwent to acute restraint stress for 3h. The contractile response evoked by angiotensin II was increased in carotid arteries from diabetic rats. Acute restraint stress did not alter angiotensin II-induced contraction in carotid arteries from normoglycaemic rats. However acute stress combined with Diabetes increased angiotensin II-induced contraction in carotid rings. Western blot experiments and the inhibition of nitric oxide synthases in functional assays showed that neuronal, endothelial and inducible nitric oxide synthase isoforms contribute to the increased formation of peroxynitrite and contractile hyperreactivity to angiotensin II in carotid rings from stressed diabetic rats. In summary, these findings suggest that the increased superoxide anion generation in carotid arteries from diabetic rats associated to the increased local nitric oxide synthases expression and activity induced by acute restrain stress were responsible for exacerbating the local formation of peroxynitrite and the contraction induced by angiotensin II.


Subject(s)
Angiotensin II/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/biosynthesis , Peroxynitrous Acid/biosynthesis , Stress, Psychological/metabolism , Animals , Behavior, Animal/drug effects , Carotid Arteries/drug effects , Carotid Arteries/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/psychology , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 1/psychology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Male , Nitric Oxide Synthase/chemistry , Phenylephrine/pharmacology , Phosphorylation/drug effects , Rats , Rats, Wistar , Restraint, Physical , Vasoconstriction/drug effects
15.
J Biol Chem ; 291(13): 7029-44, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26839313

ABSTRACT

Using high throughput screening-compatible assays for superoxide and hydrogen peroxide, we identified potential inhibitors of the NADPH oxidase (Nox2) isoform from a small library of bioactive compounds. By using multiple probes (hydroethidine, hydropropidine, Amplex Red, and coumarin boronate) with well defined redox chemistry that form highly diagnostic marker products upon reaction with superoxide (O2 (̇̄)), hydrogen peroxide (H2O2), and peroxynitrite (ONOO(-)), the number of false positives was greatly decreased. Selected hits for Nox2 were further screened for their ability to inhibit ONOO(-)formation in activated macrophages. A new diagnostic marker product for ONOO(-)is reported. We conclude that the newly developed high throughput screening/reactive oxygen species assays could also be used to identify potential inhibitors of ONOO(-)formed from Nox2-derived O2 (̇̄)and nitric oxide synthase-derived nitric oxide.


Subject(s)
Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Membrane Glycoproteins/antagonists & inhibitors , Molecular Probes/chemistry , NADPH Oxidases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Chromatography, High Pressure Liquid , Coumarins/chemistry , Enzyme Inhibitors/chemistry , Fluorometry , Gene Expression , HL-60 Cells , Humans , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Macrophage Activation/drug effects , Membrane Glycoproteins/metabolism , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Oxazines/chemistry , Oxidation-Reduction , Peroxynitrous Acid/antagonists & inhibitors , Peroxynitrous Acid/biosynthesis , Peroxynitrous Acid/chemistry , Phenanthridines/chemistry , Quaternary Ammonium Compounds/chemistry , Small Molecule Libraries/chemistry , Superoxides/antagonists & inhibitors , Superoxides/chemistry , Superoxides/metabolism , Tetradecanoylphorbol Acetate/pharmacology
16.
Oxid Med Cell Longev ; 2016: 9802136, 2016.
Article in English | MEDLINE | ID: mdl-26798430

ABSTRACT

Oxidative stress is involved in the development of carotid body (CB) chemosensory potentiation and systemic hypertension induced by chronic intermittent hypoxia (CIH), the main feature of obstructive sleep apnea. We tested whether peroxynitrite (ONOO(-)), a highly reactive nitrogen species, is involved in the enhanced CB oxygen chemosensitivity and the hypertension during CIH. Accordingly, we studied effects of Ebselen, an ONOO(-) scavenger, on 3-nitrotyrosine immunoreactivity (3-NT-ir) in the CB, the CB chemosensory discharge, and arterial blood pressure (BP) in rats exposed to CIH. Male Sprague-Dawley rats were exposed to CIH (5% O2, 12 times/h, 8 h/day) for 7 days. Ebselen (10 mg/kg/day) was administrated using osmotic minipumps and BP measured with radiotelemetry. Compared to the sham animals, CIH-treated rats showed increased 3-NT-ir within the CB, enhanced CB chemosensory responses to hypoxia, increased BP response to acute hypoxia, and hypertension. Rats treated with Ebselen and exposed to CIH displayed a significant reduction in 3-NT-ir levels (60.8 ± 14.9 versus 22.9 ± 4.2 a.u.), reduced CB chemosensory response to 5% O2 (266.5 ± 13.4 versus 168.6 ± 16.8 Hz), and decreased mean BP (116.9 ± 13.2 versus 82.1 ± 5.1 mmHg). Our results suggest that CIH-induced CB chemosensory potentiation and hypertension are critically dependent on ONOO(-) formation.


Subject(s)
Carotid Body/pathology , Hypertension/complications , Hypoxia/complications , Peroxynitrous Acid/biosynthesis , Animals , Azoles/pharmacology , Blood Pressure/drug effects , Carotid Body/drug effects , Carotid Body/physiopathology , Diastole/drug effects , Hypertension/pathology , Hypertension/physiopathology , Hypoxia/pathology , Isoindoles , Male , Neuronal Plasticity/drug effects , Organoselenium Compounds/pharmacology , Rats, Sprague-Dawley , Systole/drug effects , Tyrosine/analogs & derivatives , Tyrosine/metabolism
17.
Am J Physiol Lung Cell Mol Physiol ; 309(1): L53-62, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26136527

ABSTRACT

Neutrophil (PMNL) influx precedes lung macrophage (LM) influx into the lung following exposure of newborn pups to 60% O2. We hypothesized that PMNL were responsible for the signals leading to LM influx. This was confirmed when inhibition of PMNL influx with a CXC chemokine receptor-2 antagonist, SB-265610, also prevented the 60% O2-dependent LM influx, LM-derived nitrotyrosine formation, and pruning of small arterioles. Exposure to 60% O2 was associated with increased lung contents of neutrophil elastase and α-elastin, a marker of denatured elastin, and a decrease in elastin fiber density. This led us to speculate that neutrophil elastase-induced elastin fragments were the chemokines that led to a LM influx into the 60% O2-exposed lung. Inhibition of neutrophil elastase with sivelestat or elafin attenuated the LM influx. Sivelestat also attenuated the 60% O2-induced decrease in elastin fiber density. Daily injections of pups with an antibody to α-elastin prevented the 60% O2-dependent LM influx, impaired alveologenesis, and impaired small vessel formation. This suggests that neutrophil elastase inhibitors may protect against neonatal lung injury not only by preventing structural elastin degradation, but also by blocking elastin fragment-induced LM influx, thus preventing tissue injury from LM-derived peroxynitrite formation.


Subject(s)
Elastin/metabolism , Leukocyte Elastase/metabolism , Macrophages/immunology , Neutrophils/immunology , Oxygen/toxicity , Animals , Animals, Newborn , Cell Movement/immunology , Elafin/pharmacology , Elastin/immunology , Female , Glycine/analogs & derivatives , Glycine/pharmacology , Leukocyte Elastase/antagonists & inhibitors , Lung/pathology , Lung Injury/immunology , Maternal Exposure , Oxygen/pharmacology , Peroxynitrous Acid/biosynthesis , Phenylurea Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-8B/antagonists & inhibitors , Sulfonamides/pharmacology , Triazoles/pharmacology , Vascular Remodeling
18.
Free Radic Biol Med ; 87: 346-55, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26119787

ABSTRACT

Macrophage-derived nitric oxide ((•)NO) participates in cytotoxic mechanisms against diverse microorganisms and tumor cells. These effects can be mediated by (•)NO itself or (•)NO-derived species such as peroxynitrite formed by its diffusion-controlled reaction with NADPH oxidase-derived superoxide radical anion (O(2)(•-)). In vivo, the facile extracellular diffusion of (•)NO as well as different competing consumption routes limit its bioavailability for the reaction with O(2)(•-) and, hence, peroxynitrite formation. In this work, we evaluated the extent by which (•)NO diffusion to red blood cells (RBC) can compete with activated macrophages-derived O(2)(•-) and affect peroxynitrite formation yields. Macrophage-dependent peroxynitrite production was determined by boron-based probes that react directly with peroxynitrite, namely, coumarin-7-boronic acid (CBA) and fluorescein-boronate (Fl-B). The influence of (•)NO diffusion to RBC on peroxynitrite formation was experimentally analyzed in co-incubations of (•)NO and O(2)(•-)-forming macrophages with erythrocytes. Additionally, we evaluated the permeation of (•)NO to RBC by measuring the intracellular oxidation of oxyhemoglobin to methemoglobin. Our results indicate that diluted RBC suspensions dose-dependently inhibit peroxynitrite formation, outcompeting the O(2)(•-) reaction. Computer-assisted kinetic studies evaluating peroxynitrite formation by its precursor radicals in the presence of RBC are in accordance with experimental results. Moreover, the presence of erythrocytes in the proximity of (•)NO and O(2)(•-)-forming macrophages prevented intracellular Fl-B oxidation pre-loaded in L1210 cells co-cultured with activated macrophages. On the other hand, Fl-B-coated latex beads incorporated in the macrophage phagocytic vacuole indicated that intraphagosomal probe oxidation by peroxynitrite was not affected by nearby RBC. Our data support that in the proximity of a blood vessel, (•)NO consumption by RBC will limit the extracellular formation (and subsequent cytotoxic effects) of peroxynitrite by activated macrophages, while the intraphagosomal yield of peroxynitrite will remain unaffected.


Subject(s)
Nitric Oxide/metabolism , Oxidation-Reduction , Peroxynitrous Acid/metabolism , Superoxides/metabolism , Animals , Diffusion , Erythrocytes/metabolism , Kinetics , Macrophages/metabolism , Mice , Peroxynitrous Acid/biosynthesis , Phagosomes/metabolism
19.
Handb Exp Pharmacol ; 227: 103-17, 2015.
Article in English | MEDLINE | ID: mdl-25846616

ABSTRACT

A large body of evidence indicates that nitric oxide (NO) plays an important role in the processing of persistent inflammatory and neuropathic pain in the spinal cord. Several animal studies revealed that inhibition or knockout of NO synthesis ameliorates persistent pain. However, spinal delivery of NO donors caused dual pronociceptive and antinociceptive effects, pointing to multiple downstream signaling mechanisms of NO. This review summarizes the localization and function of NO-dependent signaling mechanisms in the spinal cord, taking account of the recent progress made in this field.


Subject(s)
Nitric Oxide/physiology , Pain/physiopathology , Spinal Cord/physiology , Animals , Cyclic GMP/physiology , Humans , Peroxynitrous Acid/biosynthesis , Signal Transduction
20.
Free Radic Biol Med ; 82: 114-21, 2015 May.
Article in English | MEDLINE | ID: mdl-25680284

ABSTRACT

TNF-α inhibitor reportedly protects against myocardial ischemia/reperfusion (MI/R) injury. It can also increase Notch1 expression in inflammatory bowel disease, revealing the regulation of Notch1 signaling by TNF-α inhibitor. However, the interaction between TNF-α inhibitor and Notch1 signaling in MI/R remains unclear. This study aimed to determine the involvement of TNF-α inhibitor with Notch1 in MI/R and delineate the related mechanism. Notch1-specific small interfering RNA (20 µg) or Jagged1 (a Notch ligand, 12 µg) was delivered through intramyocardial injection. Forty-eight hours after injection, mice received 30 min of myocardial ischemia followed by 3 h (for cell apoptosis and oxidative/nitrative stress) or 24h (for infarct size and cardiac function) of reperfusion. Ten minutes before reperfusion, mice randomly received an intraperitoneal injection of vehicle, etanercept, diphenyleneiodonium, 1400W, or EUK134. Finally, downregulation of Notch1 significantly reversed the alleviation of MI/R injury induced by etanercept, as evidenced by enlarged myocardial infarct size, suppressed cardiac function, and increased myocardial apoptosis. Moreover, Notch1 blockade increased the expression of inducible NO synthase (iNOS) and gp(91)(phox), enhanced NO and superoxide production, and accelerated their cytotoxic reaction product, peroxynitrite. Furthermore, NADPH inhibition with diphenyleneiodonium or iNOS suppression with 1400W mitigated the aggravation of MI/R injury induced by Notch1 downregulation in mice treated with etanercept. Additionally, either Notch1 activation with Jagged1 or peroxynitrite decomposition with EUK134 reduced nitrotyrosine content and attenuated MI/R injury. These data indicate that MI/R injury can be attenuated by TNF-α inhibitor, partly via Notch1 signaling-mediated suppression of oxidative/nitrative stress.


Subject(s)
Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Oxidative Stress/physiology , Receptor, Notch1/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Calcium-Binding Proteins/genetics , Disease Models, Animal , Down-Regulation , Enzyme Activation , Etanercept , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Myocardial Infarction/chemically induced , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/chemically induced , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , NADP/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/biosynthesis , Onium Compounds/pharmacology , Organometallic Compounds/pharmacology , Peroxynitrous Acid/biosynthesis , RNA Interference , RNA, Small Interfering , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Receptor, Notch1/biosynthesis , Receptor, Notch1/metabolism , Salicylates/pharmacology , Serrate-Jagged Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...