Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Invest Ophthalmol Vis Sci ; 61(12): 6, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33027505

ABSTRACT

Purpose: Affecting children by age 3, primary congenital glaucoma (PCG) can cause debilitating vision loss by the developmental impairment of aqueous drainage resulting in high intraocular pressure (IOP), globe enlargement, and optic neuropathy. TEK haploinsufficiency accounts for 5% of PCG in diverse populations, with low penetrance explained by variable dysgenesis of Schlemm's canal (SC) in mice. We report eight families with TEK-related PCG, and provide evidence for SVEP1 as a disease modifier in family 8 with a higher penetrance and severity. Methods: Exome sequencing identified coding/splice site variants with an allele frequency less than 0.0001 (gnomAD). TEK variant effects were assayed in construct-transfected HEK293 cells via detection of autophosphorylated (active) TEK protein. An enucleated eye from an affected member of family 8 was examined via histology. SVEP1 expression in developing outflow tissues was detected by immunofluorescent staining of 7-day mouse anterior segments. SVEP1 stimulation of TEK expression in human umbilical vascular endothelial cells (HUVECs) was measured by TaqMan quantitative PCR. Results: Heterozygous TEK loss-of-function alleles were identified in eight PCG families, with parent-child disease transmission observed in two pedigrees. Family 8 exhibited greater disease penetrance and severity, histology revealed absence of SC in one eye, and SVEP1:p.R997C was identified in four of the five affected individuals. During SC development, SVEP1 is secreted by surrounding tissues. SVEP1:p.R997C abrogates stimulation of TEK expression by HUVECs. Conclusions: We provide further evidence for PCG caused by TEK haploinsufficiency, affirm autosomal dominant inheritance in two pedigrees, and propose SVEP1 as a modifier of TEK expression during SC development, affecting disease penetrance and severity.


Subject(s)
Cell Adhesion Molecules/genetics , Genes, Modifier/genetics , Hydrophthalmos/genetics , Receptor, TIE-2/genetics , Aged , Animals , Blotting, Western , Child, Preschool , Female , Gene Frequency , Genotyping Techniques , HEK293 Cells/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrophthalmos/diagnosis , Hydrophthalmos/physiopathology , Infant , Infant, Newborn , Intraocular Pressure/physiology , Male , Mice , Middle Aged , Mutation, Missense , Pedigree , Penetrance , Phosphorylation , Protein Isoforms , Receptor, TIE-2/metabolism , Exome Sequencing
2.
Asian Pac J Cancer Prev ; 16(10): 4357-61, 2015.
Article in English | MEDLINE | ID: mdl-26028099

ABSTRACT

Thunbergia Laurifolia Linn. (TL) is one of the most familiar plants in Thai traditional medicine that is used to treat various conditions, including cancer. However, the antitumor activity of TL or its constituents has never been reported at the molecular level to support the folklore claim. The present study was designed to investigate the antitumor effect of an aqueous extract of TL in human breast cancer cells and the possible mechanism(s) of action. An aqueous crude extract was prepared from dried leaves of TL. Folin-Ciocalteu colorimetric assays were used to determine the total phenolic content. Antiproliferative and cell cycle effects were evaluated in human breast adenocarcinoma MCF-7 cells by MTT reduction assay, cell growth inhibition, clonogenic cell survival, and flow cytometric analysis. Free radical generation by the extracts was detected using electron paramagnetic resonance spectroscopy. The exposure of human breast adenocarcinoma MCF-7 cells to a TL aqueous extract resulted in decreases in cell growth, clonogenic cell survival, and cell viability in a concentration-dependent manner with an IC50 value of 843 µg/ml. Treatments with extract for 24 h at 250 µg/ml or higher induced cell cycle arrest as indicated by a significant increase of cell population in the G1 phase and a significant decrease in the S phase of the cell cycle. The capability of the aqueous extract to generate radical intermediates was observed at both high pH and near-neutral pH conditions. The findings suggest the antitumor bioactivities of TL against selected breast cancer cells may be due to induction of a G1 cell cycle arrest. Cytotoxicity and cell cycle perturbation that are associated with a high concentration of the extract could be in part explained by the total phenolic contents in the extract and the capacity to generate radical intermediates to modulate cellular proliferative signals.


Subject(s)
Acanthaceae , Breast Neoplasms/drug therapy , Free Radicals/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Plant Extracts/pharmacology , S Phase Cell Cycle Checkpoints/drug effects , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Phenols/analysis , Plant Extracts/chemistry , Plant Leaves
3.
Transfusion ; 55(6): 1178-85, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25644965

ABSTRACT

BACKGROUND: The transfusion of red blood cells (RBCs) with maximum therapeutic efficacy is a major goal in transfusion medicine. One of the criteria used in determining stored RBC quality is end-of-storage hemolysis. Between donors, a wide range of hemolysis is observed under identical storage conditions. Here, a potential mechanism for this wide range is investigated. We hypothesize that the magnitude of hemolysis is a heritable trait. Also, we investigated correlations between hemolysis and RBC metabolites; this will establish pathways influencing hemolysis as future targets for genetic analysis. STUDY DESIGN AND METHODS: Units of RBCs from identical and nonidentical twins were collected and stored under standard conditions for 56 days. Hemolysis, adenosine triphosphate (ATP), and total glutathione (tGSH) were measured throughout storage. Nontargeted metabolic analyses were performed on RBCs that had been stored for 28 days. Heritability was determined by comparing values between identical and nonidentical twins. RESULTS: Hemolysis was found to be heritable (mean > 45%) throughout the storage period. Potential correlations were observed between hemolysis and metabolites from the purine metabolism, lysolipid, and glycolysis pathways. These also exhibited heritability (>20%). No correlation was found with ATP or tGSH. CONCLUSION: The susceptibility of RBCs to lysis during storage is partly determined by inheritance. We have also uncovered several pathways that are candidate targets for future genomewide association studies. These findings will aid in the design of better storage solutions and the development of donor screening tools that minimize hemolysis during storage.


Subject(s)
Blood Donors , Blood Preservation , Erythrocytes/physiology , Hemolysis/genetics , Adult , Body Height/genetics , Body Mass Index , Body Weight/genetics , Erythrocyte Indices , Erythrocytes/chemistry , Female , Hemoglobins/analysis , Humans , Leukocyte Reduction Procedures , Male , Metabolome/genetics , Polymorphism, Single Nucleotide , Time Factors , Twins, Dizygotic , Twins, Monozygotic , Young Adult
4.
Transfusion ; 54(8): 2055-63, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24601981

ABSTRACT

BACKGROUND: The degeneration of red blood cells (RBCs) during storage is a major issue in transfusion medicine. Family studies in the 1960s established the heritability of the RBC storage lesion based on poststorage adenosine triphosphate (ATP) concentrations. However, this critical discovery has not been further explored. In a classic twin study we confirmed the heritability of poststorage ATP concentrations and established the heritability of many other RBC metabolites. STUDY DESIGN AND METHODS: ATP concentrations and metabolomic profiles were analyzed in RBC samples from 18 twin pairs. On samples stored for 28 days, the heritability of poststorage ATP concentrations were 64 and 53% in CP2D- and AS-3-stored RBCs, respectively. RESULTS: Metabolomic analyses identified 87 metabolites with an estimated heritability of 20% or greater. Thirty-six metabolites were significantly correlated with ATP concentrations (p ≤ 0.05) and 16 correlated with borderline significance (0.05 ≤ p ≤ 0.10). Of the 52 metabolites that correlated significantly with ATP, 24 demonstrated 20% or more heritability. Pathways represented by heritable metabolites included glycolysis, membrane remodeling, redox homeostasis, and synthetic and degradation pathways. CONCLUSION: We conclude that many RBC metabolite concentrations are genetically influenced during storage. Future studies of key metabolic pathways and genetic modifiers of RBC storage could lead to major advances in RBC storage and transfusion therapy.


Subject(s)
Adenosine Triphosphate/blood , Blood Preservation , Erythrocytes/chemistry , Quantitative Trait, Heritable , Adenine/pharmacology , Adult , Body Mass Index , Citrates/pharmacology , Erythrocytes/drug effects , Female , Glucose/pharmacology , Glycolysis/genetics , Homeostasis/genetics , Humans , Leukocyte Reduction Procedures , Male , Metabolism/genetics , Metabolomics , Oxidation-Reduction , Phosphates/pharmacology , Sodium Chloride/pharmacology , Solutions/pharmacology , Time Factors , Twins, Monozygotic , Young Adult
5.
Antioxid Redox Signal ; 20(9): 1423-35, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-23919724

ABSTRACT

AIMS: This study determined whether acute radiation-induced liver injury seen in Sirtuin3(-/-) mice after exposure to Cs-137 γ-rays was mediated by superoxide anion (O2(•-)). RESULTS: Male wild-type (WT) and SIRT3(-/-) mice were given 2×2 Gy whole-body radiation doses separated by 24 h and livers were harvested 20 h after the second dose. Ex vivo measurements in fresh frozen liver sections demonstrated 50% increases in dihydroethidium oxidation from SIRT3(-/-) animals, relative to WT animals, before irradiation, but this increase was not detected 20 h after radiation exposure. In addition, irradiated livers from SIRT3(-/-) animals showed significant hydropic degeneration, loss of MitoTracker Green FM staining, increased immunohistochemical staining for 3-nitrotyrosine, loss of Ki67 staining, and increased mitochondrial localization of p53. These parameters of radiation-induced injury were significantly attenuated by an intraperitoneal injection of 2 mg/kg of the highly specific superoxide dismutase mimic, GC4401, 30 min before each fraction. INNOVATION: Sirtuin 3 (SIRT3) is believed to regulate mitochondrial oxidative metabolism and antioxidant defenses in response to acute radiation-induced liver injury. This work provides strong evidence for the causal role of O2(•-) in the liver injury process initiated by whole-body irradiation in SIRT3(-/-) mice. CONCLUSION: These results support the hypothesis that O2(•-) mediates acute liver injury in SIRT3(-/-) animals exposed to whole-body γ-radiation and suggest that GC4401 could be used as a radio-protective compound in vivo.


Subject(s)
Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Sirtuin 3/deficiency , Superoxides/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Chemical and Drug Induced Liver Injury/pathology , Electron Transport/radiation effects , Enzyme Activation , Ethidium/analogs & derivatives , Ethidium/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , Oxidation-Reduction , Protein Transport , Superoxide Dismutase/metabolism , Tumor Suppressor Protein p53/metabolism , Whole-Body Irradiation
6.
Radiat Res ; 180(2): 156-65, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23819597

ABSTRACT

The hypothesis that mitochondrial dysfunction and increased superoxide levels in thymocytes over expressing Bax (Lck-Bax1 and Lck-Bax38&1) contributes to lymphomagenesis after low-dose radiation was tested. Lck-Bax1 single-transgenic and Lck-Bax38&1 double-transgenic mice were exposed to single whole-body doses of 10 or 100 cGy of (137)Cs or iron ions (1,000 MeV/n, 150 keV/µm) or silicon ions (300 MeV/n, 67 keV/µm). A 10 cGy dose of (137)Cs significantly increased the incidence and onset of thymic lymphomas in female Lck-Bax1 mice. In Lck-Bax38&1 mice, a 100 cGy dose of high-LET iron ions caused a significant dose dependent acceleration of lymphomagenesis in both males and females that was not seen with silicon ions. To determine the contribution of mitochondrial oxidative metabolism, Lck-Bax38&1 over expressing mice were crossed with knockouts of the mitochondrial protein deacetylase, Sirtuin 3 (Sirt3), which regulates superoxide metabolism. Sirt3(-/-)/Lck-Bax38&1 mice demonstrated significant increases in thymocyte superoxide levels and acceleration of lymphomagenesis (P < 0.001). These results show that lymphomagenesis in Bax over expressing animals is enhanced by radiation exposure in both an LET and gender dependent fashion. These findings support the hypothesis that mitochondrial dysfunction leads to increased superoxide levels and accelerates lymphomagenesis in Lck-Bax transgenic mice.


Subject(s)
Heavy Ions/adverse effects , Linear Energy Transfer , Lymphoma/etiology , Mitochondria/radiation effects , Neoplasms, Radiation-Induced/etiology , Oxidative Stress , Sex Characteristics , Superoxides/metabolism , Thymus Neoplasms/etiology , Whole-Body Irradiation/adverse effects , bcl-2-Associated X Protein/physiology , Animals , Cesium Radioisotopes , Dose-Response Relationship, Radiation , Female , Gene Dosage , Iron , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphoma/genetics , Lymphoma/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mitochondria/metabolism , Neoplasms, Radiation-Induced/genetics , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/physiopathology , Oxidative Phosphorylation/radiation effects , Radiation Dosage , Radiation Tolerance/genetics , Recombinant Fusion Proteins/physiology , Silicon , Sirtuin 3/deficiency , Sirtuin 3/genetics , Sirtuin 3/physiology , Thymocytes/metabolism , Thymocytes/pathology , Thymocytes/radiation effects , Thymus Neoplasms/genetics , Thymus Neoplasms/physiopathology , bcl-2-Associated X Protein/genetics
7.
PLoS One ; 7(10): e48175, 2012.
Article in English | MEDLINE | ID: mdl-23118946

ABSTRACT

Increased glutathione (GSH) and thioredoxin (Trx) metabolism are mechanisms that are widely implicated in resistance of cancer cells to chemotherapy. The current study determined if simultaneous inhibition of GSH and Trx metabolism enhanced cell killing of human head and neck squamous cell carcinoma (HNSCC) cells by a mechanism involving oxidative stress. Inhibition of GSH and Trx metabolism with buthionine sulfoximine (BSO) and auranofin (AUR), respectively, induced significant decreases in clonogenic survival compared to either drug alone in FaDu, Cal-27 and SCC-25 HNSCC cells in vitro and in vivo in Cal-27 xenografts. BSO+AUR significantly increased glutathione and thioredoxin oxidation and suppressed peroxiredoxin activity in vitro. Pre-treatment with N-acetylcysteine completely reversed BSO+AUR-induced cell killing in FaDu and Cal-27 cells, while catalase and selenium supplementation only inhibited BSO+AUR-induced cell killing in FaDu cells. BSO+AUR decreased caspase 3/7 activity in HNSCC cells and significantly reduced the viability of both Bax/Bak double knockout (DKO) and DKO-Bax reconstituted hematopoietic cells suggesting that necrosis was involved. BSO+AUR also significantly sensitized FaDu, Cal-27, SCC-25 and SQ20B cells to cell killing induced by the EGFR inhibitor Erlotinib in vitro. These results support the conclusion that simultaneous inhibition of GSH and Trx metabolism pathways induces oxidative stress and clonogenic killing in HNSCCs and this strategy may be useful in sensitizing HNSCCs to EGFR inhibitors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Squamous Cell/drug therapy , Glutathione/biosynthesis , Head and Neck Neoplasms/drug therapy , Thioredoxins/biosynthesis , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Auranofin/administration & dosage , Buthionine Sulfoximine/administration & dosage , Catalase/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Erlotinib Hydrochloride , Female , Gene Knockdown Techniques , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Humans , Mice , Mice, Nude , Necrosis , Oxidation-Reduction , Oxidative Stress , Peroxiredoxins/metabolism , Quinazolines/administration & dosage , RNA, Small Interfering/genetics , Thioredoxin-Disulfide Reductase/genetics , Thioredoxin-Disulfide Reductase/metabolism , Xenograft Model Antitumor Assays
8.
PLoS One ; 6(5): e19786, 2011.
Article in English | MEDLINE | ID: mdl-21611191

ABSTRACT

Caspase-9 is a component of the apoptosome that mediates cell death following release of cytochrome c from mitochondria. Inhibition of Caspase-9 with a dominant negative construct (Casp9DN) blocks apoptosome function, promotes viability and has been implicated in carcinogenesis. Inhibition of the apoptosome in vitro impairs mitochondrial function and promotes mitophagy. To examine whether inhibition of the apoptosome would enhance mitophagy and promote oncogenesis in vivo, transgenic mice were generated that express Casp9DN in the T cell lineage. The effects of Casp9DN on thymocyte viability, mitophagy and thymic tumor formation were examined. In primary thymocytes, Casp9DN delayed dexamethasone (Dex)-induced cell death, altered mitochondrial structure, and decreased oxidant production. Transmission electron microscopy (TEM) revealed that inhibition of the apoptosome resulted in structurally abnormal mitochondria that in some cases were engulfed by double-membrane structures resembling autophagosomes. Consistent with mitochondria being engulfed by autophagosomes (mitophagy), confocal microscopy showed colocalization of LC3-GFP and mitochondria. However, Casp9DN did not significantly accelerate T-cell lymphoma alone, or in combination with Lck-Bax38/1, or with Beclin 1+/- mice, two tumor-prone strains in which altered mitochondrial function has been implicated in promoting tumor development. In addition, heterozygous disruption of Beclin 1 had no effect on T-cell lymphoma formation in Lck-Bax38/1 mice. Further studies showed that Beclin 1 levels had no effect on Casp9DN-induced loss of mitochondrial function. These results demonstrate that neither inhibition of apoptosome function nor Beclin 1 haploinsufficiency accelerate T-cell lymphoma development in mice.


Subject(s)
Autophagy , Caspase Inhibitors , Lymphoma, T-Cell/enzymology , Lymphoma, T-Cell/pathology , Mitochondria/pathology , Animals , Apoptosis Regulatory Proteins/genetics , Autophagy/drug effects , Beclin-1 , Caspases/metabolism , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Separation , Cells, Cultured , Dexamethasone/pharmacology , Gene Dosage/genetics , Genes, Dominant , Haploinsufficiency/genetics , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/ultrastructure , Reactive Oxygen Species/metabolism , Thymus Gland/pathology , bcl-2-Associated X Protein/metabolism
9.
Clin Cancer Res ; 16(2): 509-20, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20068072

ABSTRACT

PURPOSE: Pharmacologic concentrations of ascorbate may be effective in cancer therapeutics. We hypothesized that ascorbate concentrations achievable with i.v. dosing would be cytotoxic in pancreatic cancer for which the 5-year survival is <3%. EXPERIMENTAL DESIGN: Pancreatic cancer cell lines were treated with ascorbate (0, 5, or 10 mmol/L) for 1 hour, then viability and clonogenic survival were determined. Pancreatic tumor cells were delivered s.c. into the flank region of nude mice and allowed to grow at which time they were randomized to receive either ascorbate (4 g/kg) or osmotically equivalent saline (1 mol/L) i.p. for 2 weeks. RESULTS: There was a time- and dose-dependent increase in measured H(2)O(2) production with increased concentrations of ascorbate. Ascorbate decreased viability in all pancreatic cancer cell lines but had no effect on an immortalized pancreatic ductal epithelial cell line. Ascorbate decreased clonogenic survival of the pancreatic cancer cell lines, which was reversed by treatment of cells with scavengers of H(2)O(2). Treatment with ascorbate induced a caspase-independent cell death that was associated with autophagy. In vivo, treatment with ascorbate inhibited tumor growth and prolonged survival. CONCLUSIONS: These results show that pharmacologic doses of ascorbate, easily achievable in humans, may have potential for therapy in pancreatic cancer.


Subject(s)
Adenocarcinoma/pathology , Ascorbic Acid/pharmacology , Oxidative Stress/drug effects , Pancreatic Neoplasms/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Animals , Ascorbic Acid/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Cytotoxins/pharmacology , Cytotoxins/therapeutic use , Dose-Response Relationship, Drug , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Mice , Mice, Nude , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
10.
Radiat Res ; 172(6): 737-45, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19929420

ABSTRACT

Ionizing radiation induces chronic metabolic oxidative stress and a mutator phenotype in hamster fibroblasts that is mediated by H(2)O(2), but the intracellular source of H(2)O(2) is not well defined. To determine the role of mitochondria in the radiation-induced mutator phenotype, end points of mitochondrial function were determined in unstable (CS-9 and LS-12) and stable (114) hamster fibroblast cell lines derived from GM10115 cells exposed to 10 Gy X rays. Cell lines isolated after irradiation demonstrated a 20-40% loss of mitochondrial membrane potential and an increase in mitochondrial content compared to the parental cell line GM10115. Surprisingly, no differences were observed in steady-state levels of ATP (P > 0.05). Unstable clones demonstrated increased oxygen consumption (two- to threefold; CS-9) and/or increased mitochondrial electron transport chain (ETC) complex II activity (twofold; LS-12). Using Western blot analysis and Blue Native gel electrophoresis, a significant increase in complex II subunit B protein levels was observed in LS-12 cells. Furthermore, immunoprecipitation assays revealed evidence of abnormal complex II assembly in LS-12 cells. Treatment of LS-12 cells with an inhibitor of ETC complex II (thenoyltrifluoroacetone) resulted in significant decreases in the steady-state levels of H(2)O(2) and a 50% reduction in mutation frequency as well as a 16% reduction in CAD gene amplification frequency. These data show that radiation-induced genomic instability was accompanied by evidence of mitochondrial dysfunction leading to increased steady-state levels of H(2)O(2) that contributed to increased mutation frequency and gene amplification. These results support the hypothesis that mitochondrial dysfunction originating from complex II can contribute to radiation-induced genomic instability by increasing steady-state levels of reactive oxygen species.


Subject(s)
Electron Transport Complex II/metabolism , Genomic Instability , Mitochondria/enzymology , Radiation, Ionizing , Animals , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , Electrophoresis, Polyacrylamide Gel
11.
Biochem J ; 413(1): 185-91, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18352860

ABSTRACT

Chronic oxidative stress has been associated with genomic instability following exposure to ionizing radiation. However, results showing direct causal linkages between specific ROS (reactive oxygen species) and the ionizing radiation-induced mutator phenotype are lacking. The present study demonstrates that ionizing radiation-induced genomically unstable cells (characterized by chromosomal instability and an increase in mutation and gene amplification frequencies) show a 3-fold increase in steady-state levels of hydrogen peroxide, but not superoxide. Furthermore, stable clones isolated from parallel studies showed significant increases in catalase and GPx (glutathione peroxidase) activity. Treatment of unstable cells with PEG-CAT (polyethylene glycol-conjugated catalase) reduced the mutation frequency and mutation rate in a dose-dependent fashion. In addition, inhibiting catalase activity in the stable clones using AT (3-aminotriazole) increased mutation frequency and rate. These results clearly demonstrate the causal relationship between chronic oxidative stress mediated by hydrogen peroxide and the mutator phenotype that persists for many generations following exposure of mammalian cells to ionizing radiation.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/radiation effects , Genomic Instability/radiation effects , Hydrogen Peroxide/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Free Radical Scavengers/metabolism , Free Radicals/metabolism , Phenotype
12.
J Biol Chem ; 282(4): 2144-55, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17102131

ABSTRACT

Cytokine deprivation has been classically used to study molecular processes of apoptosis. Following interleukin (IL)-3 withdrawal in FL5.12 cells, Bax undergoes a conformational change that results in its mitochondria targeting, cytochrome c release, activation of caspase-9, and apoptosis. Cells overexpressing Casp9DN (dominant negative caspase-9) or treated with the caspase inhibitor Q-VD-OPh increased viability but failed to increase clonogenic survival. We find that caspase-inhibited cells had a significant fraction of viable cells (herein termed "rescued" cells) that failed to initiate cell division after IL-3 add back. The "rescued" cells had reduced mitochondrial potential, stained for active Bax, and had reduced staining with dihydroethidium, an agent sensitive to superoxide levels. Readdition of IL-3 after deprivation demonstrated that Bax activation was reversed, whereas altered 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide and dihydroethidium staining persisted for days. Furthermore, the "rescued" cells were resistant to rotenone, an inhibitor of mitochondrial respiration. The cells were highly sensitive to 2-deoxyglucose, an inhibitor of glycolysis and proposed anti-cancer agent. We conclude that the inhibition of caspase-9 allows cells to retain viability, but cells have prolonged mitochondrial dysfunction and enter a unique nondividing state that shares some properties with malignant cells.


Subject(s)
Apoptosis , B-Lymphocytes/physiology , Caspase Inhibitors , Cell Cycle , Interleukin-3 , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , B-Lymphocytes/cytology , Cell Cycle/drug effects , Cell Differentiation , Cell Line , Cytochrome c Group/physiology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Hematopoiesis , Hematopoietic System/physiology , Humans , Interleukin-3/deficiency , Interleukin-3/pharmacology , Mice , Quinolines/pharmacology , bcl-2-Associated X Protein/physiology
13.
Photochem Photobiol ; 82(6): 1634-7, 2006.
Article in English | MEDLINE | ID: mdl-16898858

ABSTRACT

Singlet oxygen is a highly reactive electrophilic species that reacts rapidly with electron-rich moieties, such as the double bonds of lipids, thiols, and ascorbate (AscH-). The reaction of ascorbate with singlet oxygen is rapid (k = 3 x 10(8) M(-1) s(-1)). Here we have investigated the stoichiometry of this reaction. Using electrodes to make simultaneous, real-time measurements of oxygen and hydrogen peroxide concentrations, we have investigated the products of this reaction. We have demonstrated that hydrogen peroxide is a product of this reaction. The stoichiometry for the reactants of the reaction (1 1O2 + 1AscH--->1H2O2 + 1dehydroascorbic) is 1:1. The formation of H2O2 results in a very different oxidant that has a longer lifetime and much greater diffusion distance. Thus, locally produced singlet oxygen with a half-life of 1 ns to 1 micros in a biological setting is changed to an oxidant that has a much longer lifetime and thus can diffuse to distant targets to initiate biological oxidations.


Subject(s)
Ascorbic Acid/chemistry , Hydrogen Peroxide/chemistry , Singlet Oxygen/chemistry , Dihematoporphyrin Ether/chemistry , Kinetics , Oxidants
14.
Free Radic Biol Med ; 40(3): 501-6, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16443165

ABSTRACT

Nitric oxide (NO*) is an effective chain-breaking antioxidant in free radical-mediated lipid oxidation (LPO). It reacts rapidly with peroxyl radicals as a sacrificial chain-terminating antioxidant. The goal of this work was to determine the minimum threshold concentration of NO* required to inhibit Fe2+ -induced cellular lipid peroxidation. Using oxygen consumption as a measure of LPO, we simultaneously measured nitric oxide and oxygen concentrations with NO* and O2 electrodes. Ferrous iron and dioxygen were used to initiate LPO in docosahexaenoic acid-enriched HL-60 and U937 cells. Bolus addition of NO* (1.5 microM) inhibited LPO when the NO* concentration was greater than 50 nM. Similarly, using (Z)-1-[N-(3-ammoniopropyl)-N-(n-propyl)amino]diazen-1-ium-1,2-diolate as a NO* donor we found that an average steady-state NO* concentration of at least 72 +/- 9 nM was required to blunt LPO. As long as the concentration of NO* was above 13 +/- 8 nM the inhibition was sustained. Once the concentration of NO* fell below this value, the rate of lipid oxidation accelerated as measured by the rate of oxygen consumption. Our model suggests that a continuous production of NO* that would yield a steady-state concentration of only 10-20 nM is capable of inhibiting Fe2+ -induced LPO.


Subject(s)
Antioxidants/pharmacology , Free Radical Scavengers/pharmacology , Nitric Oxide/pharmacology , HL-60 Cells/drug effects , Humans , Iron/pharmacology , Lipid Peroxidation/drug effects , Oxygen/metabolism , Oxygen Consumption/drug effects , U937 Cells/metabolism
15.
Resuscitation ; 60(3): 351-7, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15061157

ABSTRACT

OBJECTIVES: to demonstrate that nitric oxide (NO) contributes to free radical generation after epicardial shocks and to determinethe effect of a nitric oxide synthase (NOS) inhibitor, N(G)-nitro-L-arginine (L-NNA), on free radical generation. BACKGROUND: Free radicals are generated by direct current shocks for defibrillation. NO reacts with the superoxide (O2*-) radical to for peroxynitrite (O = NOO-), which is toxic and initiates additional free radical generation. The contribution of NO to free radical generation after defibrillation is not fully defined. METHODS AND RESULTS: Fourteen open chest dogs were studied. In the initial eight dogs, 40 J damped sinusoidal monophasic epicardial shocks was administered. Using electron paramagnetic resonance, we monitored the coronary sinus concentration of ascorbate free radical (Asc*-), a measure of free radical generation (total oxidative flux). Epicardial shocks were repeated after L-NNA, 5 mg/kg IV. In six additional dogs, immunohistochemical staining was done to identify nitrotyrosine, a marker of reactive nitrogen species-mediated injury, in post-shock myocardial tissue. Three of these dogs received L-NNA pre-shock. After the initial 40 J shock, Asc*- rose 39 +/- 2.5% from baseline. After L-NNA infusion, a similar 40 J shock caused Asc*- to increase only 2 +/- 3% form baseline (P < 0.05, post-L-NNA shock versus initial shock). Nitrotyrosine staining was more prominent in control animals than dogs receiving L-NNA, suggesting prevention of O = NOO- formation. CONCLUSION: NO contributes to free radical generation and nitrosative injury after epicardial shocks; NOS inhibitors decrease radical generation by inhibiting the production of O = NOO-.


Subject(s)
Electric Countershock , Enzyme Inhibitors/pharmacology , Free Radicals/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/physiology , Nitroarginine/pharmacology , Tyrosine/analogs & derivatives , Animals , Ascorbic Acid/metabolism , Dogs , Electron Spin Resonance Spectroscopy , Histocytochemistry , Myocardium , Nitroarginine/administration & dosage , Peroxynitrous Acid/biosynthesis , Superoxides/metabolism
16.
Resuscitation ; 59(3): 345-52, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14659604

ABSTRACT

BACKGROUND: During reperfusion of ischemic myocardium nitric oxide (NO) reacts with superoxide radicals to form cardiotoxic peroxynitrite, which causes lipid peroxidation. Our hypothesis was that infusion of a NO donor S-nitroso-N-acetylpenicillamine (SNAP) during ischemia-reperfusion would exacerbate the oxidative damage to the myocardium by increased formation of nitrogen radicals. METHODS AND RESULTS: In 19 open-chest dogs, left anterior descending (LAD) coronary occlusion (15 min)-reperfusion (15 min) sequences were created. Using electron paramagnetic resonance (EPR), we monitored the coronary sinus concentration of ascorbate free radical (Ascz*-), a measure of free radical generation (total oxidative flux). Seven control dogs (Group 1) received intravenous saline infusion during occlusion-reperfusion, while 12 dogs received SNAP infusion (Group 2: 2.5 microg/min per kg SNAP, and Group 3: 5 microg/min per kg SNAP). Left ventricular fractional area shortening was determined by echocardiography. Dogs in Group 3 receiving a high dose of SNAP (5 microg/min per kg) demonstrated a higher Ascz*- concentration increase than the control group. Percent fractional area shortening in Group 1 declined from 77+/-4.0 (baseline) to 54+/-9.0% during ischemia (P<0.05), and then fully recovered to 74+/-3.7% with reperfusion. In the SNAP-treated dogs, the percent fractional area shortening during reperfusion was significantly lower than baseline in Group 2 (55+/-3.9 vs. baseline 74+/-4.4%, P<0.05) and in Group 3 (49+/-5.0 vs. baseline 71+/-4.5%, P<0.01). In five additional dogs, nitrotyrosine immunohistochemistry showed heavy staining of the ischemic-reperfused myocardium. CONCLUSIONS: The NO donor SNAP increased free radical concentration and exacerbated myocardial oxidative damage after ischemia-reperfusion.


Subject(s)
Free Radicals/metabolism , Myocardial Ischemia/drug therapy , Myocardial Ischemia/pathology , Myocardial Reperfusion/methods , S-Nitroso-N-Acetylpenicillamine/adverse effects , S-Nitroso-N-Acetylpenicillamine/pharmacology , Animals , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Echocardiography , Electron Spin Resonance Spectroscopy , Female , Immunohistochemistry , Infusions, Intravenous , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Male , Nitric Oxide Donors/adverse effects , Nitric Oxide Donors/pharmacology , Probability , Reference Values , Risk Factors , Sensitivity and Specificity , Ventricular Function, Left , Ventricular Remodeling
17.
Resuscitation ; 57(1): 101-8, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12668306

ABSTRACT

OBJECTIVES: To demonstrate that nitric oxide (NO) contributes to free radical generation after epicardial shocks and to determine the effect of a nitric oxide synthase (NOS) inhibitor, N(G)-nitro-L-arginine (L-NNA), on free radical generation. BACKGROUND: Free radicals are generated by direct current shocks for defibrillation. NO reacts with the superoxide (O(2).(-)) radical to form peroxynitrite (O=NOO(-)), which is toxic and initiates additional free radical generation. The contribution of NO to free radical generation after defibrillation is not fully defined. METHODS AND RESULTS: Fourteen open chest dogs were studied. In the initial eight dogs, 40 J damped sinusoidal monophasic epicardial shocks was administered. Using electron paramagnetic resonance, we monitored the coronary sinus concentration of ascorbate free radical (Ascz.(-)), a measure of free radical generation (total oxidative flux). Epicardial shocks were repeated after L-NNA, 5 mg/kg IV. In six additional dogs, immunohistochemical staining was done to identify nitrotyrosine, a marker of reactive nitrogen species-mediated injury, in post-shock myocardial tissue. Three of these dogs received L-NNA pre-shock. After the initial 40 J shock, Ascz.(-) rose 39+/-2.5% from baseline. After L-NNA infusion, a similar 40 J shock caused Ascz.(-) to increase only 2+/-3% from baseline (P<0.05, post-L-NNA shock versus initial shock). Nitrotyrosine staining was more prominent in control animals than dogs receiving L-NNA, suggesting prevention of O=NOO(-) formation. CONCLUSIONS: NO contributes to free radical generation and nitrosative injury after epicardial shocks; NOS inhibitors decrease radical generation by inhibiting the production of O=NOO(-).


Subject(s)
Electric Countershock/adverse effects , Enzyme Inhibitors/pharmacology , Free Radicals/metabolism , Myocardium/pathology , Nitric Oxide Synthase/drug effects , Nitroarginine/pharmacology , Animals , Disease Models, Animal , Dogs , Electric Countershock/methods , Electron Spin Resonance Spectroscopy , Hemodynamics/physiology , Immunohistochemistry , Nitric Oxide Synthase/metabolism , Probability , Reference Values , Risk Factors , Sensitivity and Specificity , Time Factors
18.
Crit Care Med ; 31(3): 787-92, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12626985

ABSTRACT

OBJECTIVES: Some propofol emulsion formulations contain EDTA or sodium metabisulfite to inhibit microbe growth on extrinsic contamination. EDTA is not known to react with propofol formulation components; however, sulfite has been shown to support some oxidation processes and may react with propofol. This study compared the oxidation of propofol and the formation of free radicals by electron paramagnetic resonance analysis in EDTA and sulfite propofol emulsions during a simulated intensive care unit 12-hr intravenous infusion. DESIGN: Controlled laboratory study. SETTING: University laboratory. MEASUREMENTS AND MAIN RESULTS: Propofol emulsions (3.5 mL) were dripped from spiked 50-mL vials at each hour for 12 hrs. Two propofol oxidation products, identified as propofol dimer and propofol dimer quinone, were detected in sulfite and EDTA propofol emulsions; however, sulfite propofol emulsion contained higher quantities of both compounds. After initiation of the simulated infusion, the quantities of propofol dimer and propofol dimer quinone increased in the sulfite propofol emulsion, but the lower levels in the EDTA propofol emulsion remained constant. Sulfite propofol emulsion began to visibly yellow at about 6-7 hrs. The EDTA propofol emulsion remained white at all times. The absorbance spectra of the propofol dimer and propofol dimer quinone extracted from sulfite propofol emulsion showed that propofol dimer did not absorb in the visible spectrum, but the propofol dimer quinone had an absorbance peak at 421 nm, causing it to appear yellow. Electron paramagnetic resonance analysis of the propofol emulsion containing metabisulfite revealed that the sulfite propofol emulsion yielded a strong free radical signal consistent with the formation of the sulfite anion radical (SO3*-). The EDTA propofol emulsion yielded no free radical signal above background. CONCLUSION: Sulfite from the metabisulfite additive in propofol emulsion creates an oxidative environment when these emulsions are exposed to air during a simulated intravenous infusion. This oxidation results in propofol dimerization and emulsion yellowing, the latter of which is caused by the formation of propofol dimer quinone. These processes can be attributed to the rapid formation of the reactive sulfite free radical.


Subject(s)
Drug Contamination/prevention & control , Edetic Acid/chemistry , Free Radicals/analysis , Hypnotics and Sedatives/chemistry , Propofol/chemistry , Sulfites/chemistry , Chemistry, Pharmaceutical , Chromatography, High Pressure Liquid , Critical Care/methods , Dimerization , Drug Evaluation, Preclinical , Drug Stability , Edetic Acid/metabolism , Electron Spin Resonance Spectroscopy , Emulsions , Humans , Hypnotics and Sedatives/metabolism , Oxidation-Reduction , Propofol/metabolism , Quinones/analysis , Quinones/metabolism , Sulfites/metabolism , Time Factors
19.
Resuscitation ; 56(2): 199-206, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12589995

ABSTRACT

OBJECTIVE: Our objective was to determine if magnesium reduces free radicals generated by direct current countershock and preserves left ventricular contractile function. BACKGROUND: We have previously shown that magnesium reduces free radicals in a coronary occlusion-reperfusion model, and therefore also might reduce free radical generation by direct current shocks. METHODS: In eight swine weighing 18-27 kg (mean: 22 kg), using electron paramagnetic resonance, we monitored continuously the coronary sinus concentration of ascorbate free radical, a measure of free radical generation (total oxidative flux). Epicardial shocks (30 J) using a truncated exponential biphasic waveform (5/5 ms) were administered. Each animal received two shocks, one without and one with magnesium, 80 mg/min IV, beginning 10 min before the shock and continuing to 15 min after the shock. Percent fractional area shortening of the left ventricular cavity was determined by 2-dimensional echocardiography. RESULTS: Magnesium shocks resulted in a significantly lower increase in the ascorbate free radical concentration (0.6+/-4.6%) than no-magnesium shocks (16+/-3.3%, P<0.05) at 12 min after the shock. Total radical flux was reduced 72% (P<0.05), and left ventricular fractional area shortening was preserved: baseline: 69+/-2.6%, no-magnesium shocks: 41+/-2.8% (P<0.05, versus baseline) and magnesium shocks 61+/-3.7%. CONCLUSIONS: Magnesium pre-treatment reduced oxygen free radicals generated by direct current shocks; post-shock left ventricular contractile function was not impaired. Magnesium may be cardioprotective during epicardial ('surgical') defibrillation.


Subject(s)
Electric Countershock , Free Radicals/analysis , Magnesium/pharmacology , Myocardial Contraction/drug effects , Ventricular Function, Left/physiology , Animals , Disease Models, Animal , Echocardiography , Electron Spin Resonance Spectroscopy , Female , Free Radicals/blood , Male , Probability , Random Allocation , Reference Values , Swine , Ventricular Function, Left/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...