Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
ACS Appl Mater Interfaces ; 15(37): 43880-43886, 2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37671912

ABSTRACT

Formic acid (FA) is an important C1-containing feedstock that serves as a masked source of dihydrogen gas (H2). To encourage the adoption of cleaner (noncarbonaceous) energy sources, FA detection and sensing is thus of considerable interest. Here, we examine the use of a commercially available dye, azomethine-H (Az-H), for FA sensing. Solution studies confirm that FA quenches both the absorbance and the luminescence properties of Az-H. FA was additionally found to attenuate a known Az-H (E)-to-(Z) conformational change, suggesting an Az-H/FA interaction, possibly through hydrogen bonding; this phenomenon was probed using 1H NMR spectroscopy. Moving toward a solid-state sensor, the Az-H probe was incorporated into a gelatin-based matrix. On exposure to FA, the luminescence of this system was found to increase in a FA-dependent manner, attributed to the formation of stable hydrogen-bonded structures, facilitating a (Z)-to-(E) isomerization via imine protonation, allowing for production of the more luminescent (E)-isomer. This fluorogenic signal was used as a FA sensor with an estimated detection limit of ca. 0.4 ppb FA vapor. This work constitutes an important step toward a highly sensitive FA sensor in both the solution and solid state, opening new space for the detection of organic acids in differing chemical environments.

2.
ACS Appl Mater Interfaces ; 14(41): 46562-46568, 2022 Oct 19.
Article in English | MEDLINE | ID: mdl-36194585

ABSTRACT

Microplastic (MP) pollution is a global challenge that requires immediate mitigation practices. Monitoring is crucial for quantifying MPs, but their mitigation remains very challenging due to several factors, including the lack of selective materials to specific polymers, and the low sensitivity of the current detection techniques. In this work, we introduce a novel design for the selective detection of MPs through fluorescence spectroscopy by exploiting conjugated polymer nanoparticles (CPNs). Fluorescent diketopyrrolopyrrole nanoparticles were prepared by nanoprecipitation to incorporate peripheral hyaluronic acid to increase their affinity for various plastics. The affinity of the new ligand for various types of MPs was examined through several characterization techniques, including fluorescence spectroscopy and microscopy, nanoparticle tracking analysis and computational studies. The new CPN were shown to be highly fluorescent in the presence of typically abundant MPs, achieving very strong binding constants in the picomolar range. This very strong affinity for a broad family of plastics was found to be the results of cooperative supramolecular effects and topographical affinity, as probed by advanced microscopy and in silico studies. Furthermore, the new affinity probes were shown to be highly selective for MPs, allowing for their detection in heterogeneous samples, including soil debris and other organic contaminants. The new materials design introduced in this work constitute a promising platform for the development of novel MP detection devices directly useable at the point of collection. Moreover, it opens new avenue for the mitigation of this environmental hazard through tailorable materials.


Subject(s)
Nanoparticles , Water Pollutants, Chemical , Microplastics , Plastics , Polymers/chemistry , Hyaluronic Acid , Ligands , Environmental Monitoring , Nanoparticles/chemistry , Soil , Water Pollutants, Chemical/analysis
3.
Talanta ; 237: 122981, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34736702

ABSTRACT

Here we show that the fluorescence of fluorescein isothiocyanate (FITC) is not altered by its reaction with primary amines. However, the fluorescence is rapidly quenched upon reaction with small molecular weight thiols including cysteine, glutathione, homocysteine, dithiothreitol, and sulfide. We have taken advantage of the thiol-dependent quenching of FITC to devise a sulfide specific assay by utilizing polydimethylsiloxane (PDMS) membranes that are permeable to hydrogen sulfide but not to larger charged thiols. In addition, we have discovered that the fluorescein dithiocarbamate (FDTC) formed by the reaction with sulfide can specifically react with S-nitrosothiols (RSNO) to regenerate FITC, thus serving as a specific, fluorogenic reagent to detect picomol levels of RSNO. FDTC was tested as an intracellular RSNO-sensor in germinating tomato seedlings (Solanum lycopersicum) via epifluorescence microscopy. Control plant roots exposed to FDTC showed low intracellular fluorescence which increased ∼3-fold upon exposure to extracellular S-nitrosoglutathione and ∼4-fold in the presence of N6022, a S-nitrosoglutathione reductase (GSNOR) inhibitor, demonstrating that FDTC can be used to visualize intracellular RSNO levels.


Subject(s)
Hydrogen Sulfide , S-Nitrosothiols , Fluorescein , Isothiocyanates , Nitric Oxide
4.
Antioxidants (Basel) ; 9(12)2020 Nov 30.
Article in English | MEDLINE | ID: mdl-33266126

ABSTRACT

Nitrogen remains an important macronutrient in plant root growth due to its application in amino acid production, in addition to its more elusive role in cellular signalling through nitric oxide (NO). NO is widely accepted as an important signalling oxidative radical across all organisms, leading to its study in a wide range of biological pathways. Along with its more stable NO donor, S-nitrosoglutathione (GSNO), formed by NO non-enzymatically in the presence of glutathione (GSH), NO is a redox-active molecule capable of mediating target protein cysteine thiols through the post translational modification, S-nitrosation. S-nitrosoglutathione reductase (GSNOR) thereby acts as a mediator to pathways regulated by NO due to its activity in the irreversible reduction of GSNO to oxidized glutathione (GSSG) and ammonia. GSNOR is thought to be pleiotropic and often acts by mediating the cellular environment in response to stress conditions. Under optimal conditions its activity leads to growth by transcriptional upregulation of the nitrate transporter, NRT2.1, and through its interaction with phytohormones like auxin and strigolactones associated with root development. However, in response to highly nitrosative and oxidative conditions its activity is often downregulated, possibly through an S-nitrosation site on GSNOR at cys271, Though GSNOR knockout mutated plants often display a stunted growth phenotype in all structures, they also tend to exhibit a pre-induced protective effect against oxidative stressors, as well as an improved immune response associated with NO accumulation in roots.

5.
Antioxidants (Basel) ; 8(11)2019 Nov 13.
Article in English | MEDLINE | ID: mdl-31766125

ABSTRACT

Current research has identified S-nitrosoglutathione reductase (GSNOR) as the central enzyme for regulating protein S-nitrosylation. In addition, the dysregulation of GSNOR expression is implicated in several organ system pathologies including respiratory, cardiovascular, hematologic, and neurologic, making GSNOR a primary target for pharmacological intervention. This study demonstrates the kinetic activation of GSNOR by its substrate S-nitrosoglutathione (GSNO). GSNOR kinetic analysis data resulted in nonhyperbolic behavior that was successfully accommodated by the Hill-Langmuir equation with a Hill coefficient of +1.75, indicating that the substrate, GSNO, was acting as a positive allosteric affector. Docking and molecular dynamics simulations were used to predict the location of the GSNO allosteric domain comprising the residues Asn185, Lys188, Gly321, and Lys323 in the vicinity of the structural Zn2+-binding site. GSNO binding to Lys188, Gly321, and Lys323 was further supported by hydrogen-deuterium exchange mass spectroscopy (HDXMS), as deuterium exchange significantly decreased at these residues in the presence of GSNO. The site-directed mutagenesis of Lys188Ala and Lys323Ala resulted in the loss of allosteric behavior. Ultimately, this work unambiguously demonstrates that GSNO at large concentrations activates GSNOR by binding to an allosteric site comprised of the residues Asn185, Lys188, Gly321, and Lys323. The identification of an allosteric GSNO-binding domain on GSNOR is significant, as it provides a platform for pharmacological intervention to modulate the activity of this essential enzyme.

6.
Methods ; 168: 29-34, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31278980

ABSTRACT

This study describes the theoretical basis and the methods for the facile synthesis and characterization of four fluorogenic probes, N-amido-O-aminobenzoyl-S-nitrosoglutathione (AOASNOG), N-thioamido-fluoresceinyl-S-nitroso-glutathione (TFSNOG), N,N-di(thioamido-fluoresceinyl)-cystine (DTFCys2) and N,N-di(thioamido-fluoresceinyl)-homocystine (DTFHCys2). In addition, the study describes the methodology for the application of these reagents for measuring and imaging of free thiols on cell surfaces as well as their use as pseudo substrates for the thiol reductase and S-nitrosothioldenitrosylase activities of protein disulfide isomerase (PDI) and S-nitrosothiol reductase activity of S-nitrosoglutathione reductase (GSNOR) in vitro and on live cells in culture.


Subject(s)
Disulfides/metabolism , Fluorescent Dyes/chemistry , Oxidoreductases/metabolism , S-Nitrosoglutathione/chemistry , Cell Membrane/metabolism , Humans , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Docking Simulation , Nitric Oxide/metabolism , Oxidation-Reduction , Protein Disulfide-Isomerases/metabolism , Retinal Pigment Epithelium/cytology , Sulfhydryl Compounds
7.
Biochem J ; 476(13): 1955-1956, 2019 Jul 15.
Article in English | MEDLINE | ID: mdl-31308158

ABSTRACT

Recently, Guenter Schwarz and colleagues published an elegant study in the Biochemical Journal (2019) 476, 1805-1815 which combines kinetic and spectroscopic studies with protein engineering to provide a mechanism for sulfite oxidase (SO)-catalyzed nitrite reduction that yields nitric oxide (NO). This work is noteworthy as it demonstrates that (i) for NO generation, both sulfite and nitrite must bind to the same molybdenum (Mo) center; (ii) upon sulfite reduction, Mo is reduced from +6 (MoVI) to +4 (MoIV) and MoIV reduces nitrite to NO yielding MoV; (iii) the heme moiety, linked to the Mo-center by an 11 amino acid residue tether, gets reduced by intramolecular electron transfer (IET) resulting in MoV being oxidized to MoVI; (iv) the reduced heme transfers its electron to a second nitrite molecule converting it to NO; (v) the authors demonstrate steady-state NO production in the presence of the natural electron acceptor cytochrome c; (vi) Finally, the authors use protein engineering to shorten the heme tether to reduce the heme-Mo-center distance with the aim of increasing NO production. Consequently, the rate of IET to cytochrome c is decreased but the enzymatic turnover rate for NO production is increased by ∼10-fold. This paper is unique as it provides strong evidence for a novel mechanism for steady-state NO production for human mitochondrial SO and serves as a potential template for studying NO production mechanisms in other enzymes by integrating the information gained from enzyme kinetics with EPR and UV/vis spectroscopy and protein engineering.


Subject(s)
Sulfite Oxidase , Catalysis , Humans , Kinetics , Molybdenum , Nitric Oxide , Nitrites , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors
8.
J Environ Manage ; 233: 258-263, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30580121

ABSTRACT

Phosphate (P) is a biologically important compound that is commonly incorporated into fertilizers. Wastewater from agricultural processes results in excessive accumulation of P and eutrophication of lakes. We have developed a system for the remediation, recovery, and potential reuse of P from agricultural wastewater using tomato plant roots (roots) as a capture matrix and carboxymethyl cellulose (CMC) as an eluent and enhancer of P precipitation. Untreated roots can bind up to 55.2 ±â€¯15.2 grams of P per kilogram (g/kg) of roots in comparison to the maximum 8.2 ±â€¯1.5 g/kg bound by the previously used iron-chitosan (Fe-chito). The addition of CMC enhances the precipitation of P with a clearance of 97.2% as opposed to 33.3% without CMC. On site tests show an average removal of 226.5 µg/L per day or a total of ∼28 g of P removed after 23 days. This corresponds to a 71% P removal rate.


Subject(s)
Solanum lycopersicum , Wastewater , Carboxymethylcellulose Sodium , Phosphates , Plant Roots
9.
Front Mol Biosci ; 5: 18, 2018.
Article in English | MEDLINE | ID: mdl-29541639

ABSTRACT

Despite its study since the 1960's, very little is known about the post-translational regulation of the multiple catalytic activities performed by protein disulfide isomerase (PDI), the primary protein folding catalyst of the cell. This work identifies a functional role for the highly conserved CxxC-flanking residues Lys57 and Lys401 of human PDI in vitro. Mutagenesis studies have revealed these residues as modulating the oxidoreductase activity of PDI in a pH-dependent manner. Non-conservative amino acid substitutions resulted in enzyme variants upwards of 7-fold less efficient. This attenuated activity was found to translate into a 2-fold reduction of the rate of electron shuttling between PDI and the intraluminal endoplasmic reticulum oxidase, ERO1α, suggesting a functional significance to oxidative protein folding. In light of this, the possibility of lysine acetylation at residues Lys57 and Lys401 was assessed by in vitro treatment using acetylsalicylic acid (aspirin). A total of 28 acetyllysine residues were identified, including acLys57 and acLys401. The kinetic behavior of the acetylated protein form nearly mimicked that obtained with a K57/401Q double substitution variant providing an indication that acetylation of the active site-flanking lysine residues can act to reversibly modulate PDI activity.

10.
Antioxid Redox Signal ; 27(16): 1281-1296, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28376661

ABSTRACT

AIMS: Protein succination by fumarate increases in the adipose tissue of diabetic mice and in adipocytes matured in high glucose as a result of glucotoxicity-driven mitochondrial stress. The endoplasmic reticulum (ER) oxidoreductase protein disulfide isomerase (PDI) is succinated in adipocytes that are matured in high glucose, and in this study we investigated whether succination would alter PDI oxidoreductase activity, directly linking mitochondrial stress and ER stress. RESULTS: Protein succination and the ER stress marker C/EBP homologous protein (CHOP) were diminished after pharmaceutical targeting of mitochondrial stress with the chemical uncoupler niclosamide in adipocytes matured in high-glucose concentrations. PDI was succinated by fumarate on both CXXC-containing active sites, contributing to reduced enzymatic activity. Succinated PDI decreased reductase activity in adipocytes matured in high glucose, and in db/db epididymal adipose tissue, in association with increased levels of CHOP. PDI succination was increased in fumarase knockdown adipocytes, leading to reduced PDI oxidoreductase activity, increased CHOP levels, and pro-inflammatory cytokine secretion, confirming the specific role of elevated fumarate levels in contributing to ER stress. In addition, PDI succination and ER stress were decreased, and PDI reductase activity was restored when exposure to chronic high glucose was limited, highlighting the importance of calorie restriction in the improvement of adipocyte metabolic function. INNOVATION: These experiments identify PDI succination as a novel biochemical mechanism linking altered mitochondrial metabolism to ER stress in the adipocyte during diabetes. CONCLUSION: The current study demonstrates that early biochemical changes in mitochondrial metabolism have important implications for the development of adipocyte stress. Antioxid. Redox Signal. 27, 1281-1296.


Subject(s)
Adipocytes/metabolism , Diabetes Mellitus, Experimental/metabolism , Fumarates/metabolism , Mitochondria/metabolism , Protein Disulfide-Isomerases/metabolism , 3T3-L1 Cells , Animals , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Glucose/pharmacology , Mice , Niclosamide/pharmacology , Oxidative Stress , Protein Disulfide-Isomerases/chemistry , Transcription Factor CHOP/metabolism
11.
Free Radic Biol Med ; 108: 445-451, 2017 07.
Article in English | MEDLINE | ID: mdl-28419866

ABSTRACT

S-nitrosoglutathione reductase (GSNOR) is a multifunctional enzyme. It can catalyze NADH-dependent reduction of S-nitrosoglutathione (GSNO); as well as NAD+-dependent oxidation of hydroxymethylglutathione (HMGSH; an adduct formed by the spontaneous reaction between formaldehyde and glutathione). While initially recognized as the enzyme that is involved in formaldehyde detoxification, increasing amount of evidence has shown that GSNOR also plays a significant role in nitric oxide mediated signaling through its modulation of protein S-nitrosothiol signaling. In humans, GSNOR/S-nitrosothiols have been implicated in the etiology of several diseases including lung cancer, cystic fibrosis, asthma, pulmonary hypertension, and neuronal dysfunction. Currently, it is not possible to monitor the activity of GSNOR in live cells. In this article, we present a new compound, O-aminobenzoyl-S-nitrosoglutathione (OAbz-GSNO), which acts as a fluorogenic pseudo-substrate for GSNOR with an estimated Km value of 320µM. The weak OAbz-GSNO fluorescence increases by approximately 14 fold upon reduction of its S-NO moiety. In live cell imaging studies, OAbz-GSNO is readily taken up by primary pulmonary endothelial cells and localizes to the same perinuclear region as GSNOR. The perinuclear OAbz-GSNO fluorescence increases in a time dependent manner and this increase in fluorescence is abolished by siRNA knockdown of GSNOR or by treatment with GSNOR-specific inhibitors N6022 and C3. Taken together, these data demonstrate that OAbz-GSNO can be used as a tool to monitor the activity of GSNOR in live cells.


Subject(s)
Aldehyde Oxidoreductases/metabolism , Endothelial Cells/physiology , Fluorescent Dyes/metabolism , Lung/cytology , S-Nitrosoglutathione/metabolism , Aldehyde Oxidoreductases/genetics , Animals , Cell Membrane Permeability , Cells, Cultured , Fluorescent Dyes/chemistry , Formaldehyde/chemistry , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , RNA, Small Interfering/genetics , S-Nitrosoglutathione/analogs & derivatives , S-Nitrosoglutathione/chemistry , Signal Transduction , Substrate Specificity
12.
Free Radic Biol Med ; 89: 512-21, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26453916

ABSTRACT

Hydrogen sulfide (H2S) is produced enzymatically by cystathionine ß-synthase (CBS) and cystathionine γ-lyase (CSE), as well as other enzymes in mammalian tissues. These discoveries have led to the crowning of H2S as yet another toxic gas that serves as a gasotransmitter like NO and CO. H2S is thought to exert its biological effects through its reaction with cysteine thiols in proteins, yielding sulfurated thiol (-SSH) derivatives. One of the first proteins shown to be modified by H2S was glyceraldehyde 3-phosphate dehydrogenase (GAPDH) [1] where the S-sulfuration of the active site cysteine (Cys 152) resulted in ~7-fold increase in the activity of the enzyme. In the present study we have attempted to reproduce this result with no success. GAPDH in its reduced, or hydrogen peroxide, or glutathione disulfide, or nitrosonium oxidized forms was reacted with sulfide or polysulfides. Sulfide had no effect on reduced GAPDH activity, while polysulfides inhibited GAPDH to ~42% of control. S-sulfuration of GAPDH occurred at Cys 247 after sulfide treatment, Cys 156 and Cys 247 after polysulfide treatment. No evidence of S-sulfuration at active site Cys 152 was discovered. Both sulfide and polysulfide was able to restore the activity of glutathione disulfide oxidized GAPDH, but not to control untreated levels. Treatment of glutathione disulfide oxidized GAPDH with polysulfide also produced S-sulfuration of Cys 156. Treatment of a C156S mutant of GAPDH with sulfide and polysulfide resulted in S-sulfuration of Cys 152, which also caused a decrease and not an increase in enzymatic activity. Computational chemistry shows S-sulfuration of Cys 156 may affect the position of catalytic Cys 152, raising its pKa by 0.5, which may affect the nucleophilicity of Cys 152. The current study raises significant questions about the reported ability of H2S to activate GAPDH by the sulfuration of its active site thiol, and indicates that polysulfide is a stronger protein S-sulfurating agent than sulfide.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Enzyme Activation , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Humans , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , In Vitro Techniques , Mass Spectrometry , Models, Molecular , Oxidation-Reduction , Sulfides/metabolism , Sulfides/pharmacology
13.
FEBS J ; 282(10): 2045-59, 2015 May.
Article in English | MEDLINE | ID: mdl-25754985

ABSTRACT

Cyclooxygenase-2 (COX-2) is an important contributor to ischemic brain injury. Identification of the downstream mediators of COX-2 toxicity may allow the development of targeted therapies. Of particular interest is the cyclopentenone family of prostaglandin metabolites. Cyclopentenone prostaglandins (CyPGs) are highly reactive molecules that form covalent bonds with cellular thiols. Protein disulfide isomerase (PDI) is an important molecule for the restoration of denatured proteins following ischemia. Because PDI has several thiols, including thiols within the active thioredoxin-like domain, we hypothesized that PDI is a target of CyPGs and that CyPG binding of PDI is detrimental. CyPG-PDI binding was detected in vitro via immunoprecipitation and MS. CyPG-PDI binding decreased PDI enzymatic activity in recombinant PDI treated with CyPG, and PDI immunoprecipitated from neuronal culture treated with CyPG or anoxia. Toxic effects of binding were demonstrated in experiments showing that: (a) pharmacologic inhibition of PDI increased cell death in anoxic neurons, (b) PDI overexpression protected neurons exposed to anoxia and SH-SY5Y cells exposed to CyPG, and (c) PDI overexpression in SH-SY5Y cells attenuated ubiquitination of proteins and decreased activation of pro-apoptotic caspases. In conclusion, CyPG production and subsequent binding of PDI is a novel and potentially important mechanism of ischemic brain injury. We show that CyPGs bind to PDI, cyclopentenones inhibit PDI activity, and CyPG-PDI binding is associated with increased neuronal susceptibility to anoxia. Additional studies are necessary to determine the relative role of CyPG-dependent inhibition of PDI activity in ischemia and other neurodegenerative disorders.


Subject(s)
Cyclopentanes/pharmacology , Hypoxia/metabolism , Prostaglandins/pharmacology , Protein Disulfide-Isomerases/metabolism , Cell Line , Humans , Immunoblotting
14.
Front Chem ; 2: 70, 2014.
Article in English | MEDLINE | ID: mdl-25207270

ABSTRACT

Protein disulfide isomerase (PDI), is a member of the thioredoxin superfamily of redox proteins. PDI has three catalytic activities including, thiol-disulfide oxireductase, disulfide isomerase and redox-dependent chaperone. Originally, PDI was identified in the lumen of the endoplasmic reticulum and subsequently detected at additional locations, such as cell surfaces and the cytosol. This review will provide an overview of the recent advances in relating the structural features of PDI to its multiple catalytic roles as well as its physiological and pathophysiological functions related to redox regulation and protein folding.

15.
Toxicol Appl Pharmacol ; 280(3): 389-98, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25201535

ABSTRACT

Endoplasmic reticulum (ER) stress and excessive nitric oxide production via induction of inducible nitric oxide synthase (NOS2) have been implicated in the pathogenesis of neuronal retinal cell death in ocular hypertension. Neutral sphingomyelinase (N-SMase)/ceramide pathway can regulate NOS2 expression, hence this study determined the role of selective neutral sphingomyelinase (N-SMase) inhibition on retinal NOS2 levels, ER stress, apoptosis and visual evoked potentials (VEPs) in a rat model of elevated intraocular pressure (EIOP). NOS2 expression and retinal protein nitration were significantly greater in EIOP and significantly decreased with N-SMase inhibition. A significant increase was observed in retinal ER stress markers pPERK, CHOP and GRP78 in EIOP, which were not significantly altered by N-SMase inhibition. Retinal TUNEL staining showed increased apoptosis in all EIOP groups; however N-SMase inhibition significantly decreased the percent of apoptotic cells in EIOP. Caspase-3, -8 and -9 activities were significantly increased in EIOP and returned to baseline levels following N-SMase inhibition. Latencies of all VEP components were significantly prolonged in EIOP and shortened following N-SMase inhibition. Data confirm the role of nitrative injury in EIOP and highlight the protective effect of N-SMase inhibition in EIOP via down-regulation of NOS2 levels and nitrative stress.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum Stress/physiology , Glaucoma/metabolism , Nitric Oxide Synthase Type II/metabolism , Retina/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Up-Regulation/physiology , Aniline Compounds/pharmacology , Animals , Benzylidene Compounds/pharmacology , Blotting, Western , Caspases/metabolism , Disease Models, Animal , Evoked Potentials, Visual/physiology , Glaucoma/enzymology , Immunohistochemistry , In Situ Nick-End Labeling , Intraocular Pressure/physiology , Male , Random Allocation , Rats, Wistar , Retina/enzymology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors
16.
Invest Ophthalmol Vis Sci ; 55(8): 4759-67, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24985479

ABSTRACT

PURPOSE: To investigate the effects of Bruch's membrane (BrM) neutral lipid deposition in mouse models and its significance to aging and age-related macular degeneration, it is essential to reliably detect small quantities of neutral lipids including esterified cholesterol (EC). In chorioretinal sections and BrM wholemounts, we tested a novel fluorescent cholesterol marker based on the bacterial toxin perfringolysin O (PFO) and compared results with those obtained with the classic cholesterol dye filipin. METHODS: An engineered plasmid containing the specific cholesterol binding domain (D4) of PFO fused to green fluorescent protein (GFP) was expressed in cultured E. coli, isolated, purified, and concentrated. A total of 150 BrM-choroid wholemounts and chorioretinal sections of 11- to 13-month-old ApoE(null) mice were prepared and stained with PFO/D4-GFP or filipin for EC. Samples were examined by epifluorescence microscopy. RESULTS: The fluorescence intensity of PFO/D4-GFP was strong, stable, and, if small quantities of EC were present, superior to filipin. In all specimens, we could sharply locate the PFO/D4-GFP signal to BrM. A semiquantitative evaluation of BrM lipid deposition is possible by measuring PFO/D4-GFP fluorescence intensity. CONCLUSIONS: The use of PFO/D4-GFP allowed a robust and direct detection of EC in aged murine BrM. In wholemount samples, its strong and stable fluorescence facilitated a semiquantitative evaluation of BrM-EC content over a large area. The patterns of EC deposition in murine BrM wholemounts are comparable with findings in human BrM wholemounts. Perfringolysin O/D4-GFP could be an important tool for investigating the effects of BrM lipid deposition in mouse models.


Subject(s)
Aging/metabolism , Bruch Membrane/metabolism , Cholesterol Esters/metabolism , Hemolysin Proteins , Macular Degeneration/diagnosis , Aging/pathology , Animals , Bacterial Toxins , Bruch Membrane/ultrastructure , Cells, Cultured , Clostridium perfringens , Disease Models, Animal , Feasibility Studies , Female , Humans , Macular Degeneration/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission
17.
Free Radic Biol Med ; 72: 113-23, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24742815

ABSTRACT

Endoplasmic reticulum (ER) stress and excessive nitric oxide production via the induction of inducible nitric oxide synthase (NOS2) have been implicated in the pathogenesis of ocular diseases characterized by retinal degeneration. Previous studies have revealed the sphingomyelinase/ceramide pathway in the regulation of NOS2 induction. Thus, the objective of this study was to determine the activity of the sphingomyelinase/ceramide pathway, assess nitric oxide production, and examine apoptosis in human retinal pigment epithelial (RPE) cells undergoing ER stress. Sphingomyelinase (SMase) activity; nuclear factor κB (NF-κB) activation; NOS2, nitrite/nitrate, and nitrotyrosine levels; and apoptosis were determined in cultured human RPE cell lines subjected to ER stress via exposure to tunicamycin. Induction of ER stress was confirmed by increased intracellular levels of ER stress markers including phosphorylated PKR-like ER kinase, C/EBP-homologous protein, and 78-kDa glucose-regulated protein. ER stress increased nuclear translocation of NF-κB, NOS2 expression, nitrite/nitrate levels, and nitrotyrosine formation and caused apoptosis in RPE cell lines. Inhibition of neutral SMase (N-SMase) activity via GW 4869 treatment caused a significant reduction in nuclear translocation of NF-κB, NOS2 expression, nitrite/nitrate levels, nitrotyrosine formation, and apoptosis in ER-stressed RPE cells. In conclusion, N-SMase inhibition reduced nitrative stress and apoptosis in RPE cells undergoing ER stress. Obtained data suggest that NOS2 can be regulated by N-SMase in RPE cells experiencing ER stress.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum Stress/physiology , Nitric Oxide Synthase Type II/metabolism , Retinal Pigment Epithelium/enzymology , Sphingomyelin Phosphodiesterase/metabolism , Blotting, Western , Cell Line , Flow Cytometry , Fluorescent Antibody Technique , Humans , In Situ Nick-End Labeling , Retinal Pigment Epithelium/cytology
18.
Blood ; 122(22): 3642-50, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24030382

ABSTRACT

The platelet protein disulfide isomerase called ERp57 mediates platelet aggregation, but its role in thrombus formation is unknown. To determine the specific role of platelet-derived ERp57 in hemostasis and thrombosis, we generated a megakaryocyte/platelet-specific knockout. Despite normal platelet counts and platelet glycoprotein expression, mice with ERp57-deficient platelets had prolonged tail-bleeding times and thrombus occlusion times with FeCl3-induced carotid artery injury. Using a mesenteric artery thrombosis model, we found decreased incorporation of ERp57-deficient platelets into a growing thrombus. Platelets lacking ERp57 have defective activation of the αIIbß3 integrin and platelet aggregation. The defect in aggregation was corrected by the addition of exogenous ERp57, implicating surface ERp57 in platelet aggregation. Using mutants of ERp57, we demonstrate the second active site targets a platelet surface substrate to potentiate platelet aggregation. Binding of Alexa 488-labeled ERp57 to thrombin-activated and Mn(2+)-treated platelets lacking ß3 was decreased substantially, suggesting a direct interaction of ERp57 with αIIbß3. Surface expression of ERp57 protein and activity in human platelets increased with platelet activation, with protein expression occurring in a physiologically relevant time frame. In conclusion, platelet-derived ERp57 directly interacts with αIIbß3 during activation of this receptor and is required for incorporation of platelets into a growing thrombus.


Subject(s)
Blood Platelets/enzymology , Blood Platelets/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Protein Disulfide-Isomerases/blood , Thrombosis/blood , Animals , Catalytic Domain/genetics , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Mutant Proteins/blood , Mutant Proteins/genetics , Platelet Activation/physiology , Protein Disulfide-Isomerases/deficiency , Protein Disulfide-Isomerases/genetics , Recombinant Proteins/blood , Recombinant Proteins/genetics , Thrombosis/etiology
19.
Redox Biol ; 1: 373-80, 2013.
Article in English | MEDLINE | ID: mdl-24024174

ABSTRACT

Protein disulfide isomerase (PDI) is an abundant protein primarily found in the endoplasmic reticulum and also secreted into the blood by a variety of vascular cells. The evidence obtained here, suggests that PDI could directly participate in the efflux of NO(+) from red blood cells (RBC). PDI was detected both in RBC membranes and in the cytosol. PDI was S-nitrosylated when RBCs were exposed to nitrite under ∼50% oxygen saturation but not under ∼100% oxygen saturation. Furthermore, it was observed that hemoglobin (Hb) could promote PDI S-nitrosylation in the presence of ∼600 nM nitrite. In addition, three lines of evidence were obtained for PDI-Hb interactions: (1) Hb co-immunoprecipitated with PDI; (2) Hb quenched the intrinsic PDI fluorescence in a saturable manner; and (3) Hb-Fe(II)-NO absorption spectrum decreased in a [PDI]-dependent manner. Finally, PDI was detected on the surface RBC under ∼100% oxygen saturation and released as soluble under ∼50% oxygen saturation. The soluble PDI detected under ∼50% oxygen saturation was S-nitrosylated. Based on these data it is proposed that PDI is taken up by RBC and forms a complex with Hb. Hb-Fe(II)-NO that is formed from nitrite reduction under ∼50% O2, then transfers NO(+) to either Hb-Cys ß93 or directly to PDI resulting in S-nitroso-PDI which transverses the RBC membrane and attaches to the RBC surface. When RBCs enter tissues the S-nitroso-PDI is released from the RBC-surface into the blood where its NO(+) is transferred into the endothelium thereby inducing vasodilation, suggesting local oxygen-dependent dynamic interplays between nitrite, NO and S-nitrosylation.


Subject(s)
Erythrocytes/enzymology , Nitrites/metabolism , Protein Disulfide-Isomerases/metabolism , S-Nitrosothiols/metabolism , Cells, Cultured , Erythrocytes/metabolism , Hemoglobins/metabolism , Humans , Nitric Oxide/metabolism , Oxygen/metabolism
20.
Anal Chem ; 85(7): 3638-43, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23477661

ABSTRACT

Hydrogen sulfide (H2S) has recently been recognized as an important physiologically relevant gasotransmitter. Produced by the enzymes involved in the transsulfuration pathway, cystathionine ß-synthase (CBS) and cystathionine γ-lyase (CSE), H2S has been implicated to control biological activity in virtually every organ system. In recent years it is being recognized that many commonly used H2S assays do not measure free H2S specifically and may be prone to artifacts. This has led to large variations in the reported H2S biological concentrations. In order to accurately study H2S's functions in biological systems accurate assays which measure free H2S specifically are required. In this work we present a simple microplate-based colorimetric assay for H2S gas. The underside of a 96-well microplate cover was coated with Nafion polymer doped with Ag(+) ions. H2S is a highly volatile gas, and as it is volatilized in the microplate well it reacts with Ag(+) to produce Ag2S nanoparticles, which have a strong absorbance in the low-UV range. By monitoring the absorbance change from formation of Ag2S nanoparticles, H2S production can be monitored in real time. The assay has a limit of detection (LOD) of 2.61 nmol (8.70 µM) and a liner range up to 30 nmol (100 µM). Using the assay, the KM and Vmax of recombinant CSE enzyme were determined to be 11.13 ± 0.57 mM and 0.45 ± 0.01 nmol min(-1), respectively. H2S production from mouse liver homogenate under aerobic conditions in the presence of cysteine was measured and determined to be 4.89 ± 0.19 nmol min(-1) mL(-1) homogenate. The assay is simple, low cost, and specific to free H2S gas.


Subject(s)
Colorimetry/instrumentation , Hydrogen Sulfide/analysis , Animals , Cystathionine gamma-Lyase/metabolism , Equipment Design , Fluorocarbon Polymers/chemistry , Hydrogen Sulfide/metabolism , Limit of Detection , Liver/chemistry , Liver/metabolism , Mice , Mice, Inbred BALB C , Silver/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...