Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(32): E2950-7, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23878245

RESUMEN

Reactive intermediates such as reactive nitrogen species play essential roles in the cell as signaling molecules but, in excess, constitute a major source of cellular damage. We found that nitrosative stress induced by steady-state nitric oxide (NO) caused rapid activation of an ATM damage-response pathway leading to downstream signaling by this stress kinase to LKB1 and AMPK kinases, and activation of the TSC tumor suppressor. As a result, in an ATM-, LKB1-, TSC-dependent fashion, mTORC1 was repressed, as evidenced by decreased phosphorylation of S6K, 4E-BP1, and ULK1, direct targets of the mTORC1 kinase. Decreased ULK1 phosphorylation by mTORC1 at S757 and activation of AMPK to phosphorylate ULK1 at S317 in response to nitrosative stress resulted in increased autophagy: the LC3-II/LC3-I ratio increased as did GFP-LC3 puncta and acidic vesicles; p62 levels decreased in a lysosome-dependent manner, confirming an NO-induced increase in autophagic flux. Induction of autophagy by NO correlated with loss of cell viability, suggesting that, in this setting, autophagy was functioning primarily as a cytotoxic response to excess nitrosative stress. These data identify a nitrosative-stress signaling pathway that engages ATM and the LKB1 and TSC2 tumor suppressors to repress mTORC1 and regulate autophagy. As cancer cells are particularly sensitive to nitrosative stress, these data open another path for therapies capitalizing on the ability of reactive nitrogen species to induce autophagy-mediated cell death.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia/fisiología , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Autofagia/efectos de los fármacos , Western Blotting , Proteínas de Ciclo Celular/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/citología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Células HeLa , Humanos , Células MCF-7 , Ratones , Ratones Noqueados , Modelos Biológicos , Complejos Multiproteicos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico/fisiología , Donantes de Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/efectos de los fármacos , Espermina/análogos & derivados , Espermina/metabolismo , Espermina/farmacología , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
2.
J Appl Toxicol ; 35(5): 466-77, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25178734

RESUMEN

Exposure to monocyclic aromatic alkylanilines (MAAs), namely 2,6-dimethylaniline (2,6-DMA), 3,5-dimethylaniline (3,5-DMA) and 3-ethylaniline (3-EA), was significantly and independently associated with bladder cancer incidence. 3,5-DMAP (3,5-dimethylaminophenol), a metabolite of 3,5-DMA, was shown to induce an imbalance in cytotoxicity cellular antioxidant/oxidant status, and DNA damage in mammalian cell lines. This study was designed to evaluate the protective effect of ascorbic acid (Asc) against the cytotoxicity, reactive oxygen species (ROS) production, genotoxicity and epigenetic changes induced by 3,5-DMAP in AA8 Chinese Hamster Ovary (CHO) cells. In different cellular fractions, 3,5-DMAP caused alterations in the enzyme activities orchestrating a cellular antioxidant balance, decreases in reduced glutathione levels and a cellular redox ratio as well as increases in lipid peroxidation and protein oxidation. We also suggest that the cellular stress caused by this particular alkylaniline leads to both genetic (Aprt mutagenesis) and epigenetic changes in histones 3 and 4 (H3 and H4). This may further cause molecular events triggering different pathological conditions and eventually cancer. In both cytoplasm and nucleus, Asc provided increases in 3,5-DMAP-reduced glutathione levels and cellular redox ratio and decreases in the lipid peroxidation and protein oxidation. Asc was also found to be protective against the genotoxic and epigenetic effects initiated by 3,5-DMAP. In addition, Asc supplied protection against the cell cycle (G1 phase) arrest induced by this particular alkylaniline metabolite.


Asunto(s)
Aminofenoles/toxicidad , Ácido Ascórbico/farmacología , Epigénesis Genética/efectos de los fármacos , Compuestos de Anilina/toxicidad , Animales , Antioxidantes/metabolismo , Células CHO , Puntos de Control del Ciclo Celular/efectos de los fármacos , Cricetinae , Cricetulus , Daño del ADN/efectos de los fármacos , Glutatión/metabolismo , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
3.
Proc Natl Acad Sci U S A ; 109(50): 20373-8, 2012 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-23185001

RESUMEN

Melanoma patients experience inferior survival after biochemotherapy when their tumors contain numerous cells expressing the inducible isoform of NO synthase (iNOS) and elevated levels of nitrotyrosine, a product derived from NO. Although several lines of evidence suggest that NO promotes tumor growth and increases resistance to chemotherapy, it is unclear how it shapes these outcomes. Here we demonstrate that modulation of NO-mediated S-nitrosation of cellular proteins is strongly associated with the pattern of response to the anticancer agent cisplatin in human melanoma cells in vitro. Cells were shown to express iNOS constitutively, and to generate sustained nanomolar levels of NO intracellularly. Inhibition of NO synthesis or scavenging of NO enhanced cisplatin-induced apoptotic cell death. Additionally, pharmacologic agents disrupting S-nitrosation markedly increased cisplatin toxicity, whereas treatments favoring stabilization of S-nitrosothiols (SNOs) decreased its cytotoxic potency. Activity of the proapoptotic enzyme caspase-3 was higher in cells treated with a combination of cisplatin and chemicals that decreased NO/SNOs, whereas lower activity resulted from cisplatin combined with stabilization of SNOs. Constitutive protein S-nitrosation in cells was detected by analysis with biotin switch and reduction/chemiluminescence techniques. Moreover, intracellular NO concentration increased significantly in cells that survived cisplatin treatment, resulting in augmented S-nitrosation of caspase-3 and prolyl-hydroxylase-2, the enzyme responsible for targeting the prosurvival transcription factor hypoxia-inducible factor-1α for proteasomal degradation. Because activities of these enzymes are inhibited by S-nitrosation, our data thus indicate that modulation of intrinsic intracellular NO levels substantially affects cisplatin toxicity in melanoma cells. The underlying mechanisms may thus represent potential targets for adjuvant strategies to improve the efficacy of chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Óxido Nítrico/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Carcinógenos/metabolismo , Caspasa 3/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/patología , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitrosación
4.
Chem Res Toxicol ; 26(4): 538-46, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23506120

RESUMEN

One possible mechanism linking inflammation with cancer involves the generation of reactive oxygen, nitrogen, and halogen species by activated macrophages and neutrophils infiltrating sites of infection or tissue damage, with these chemical mediators causing damage that ultimately leads to cell death and mutation. To determine the most biologically deleterious chemistries of inflammation, we previously assessed products across the spectrum of DNA damage arising in inflamed tissues in the SJL mouse model nitric oxide overproduction ( Pang et al. ( 2007 ) Carcinogenesis 28 , 1807 - 1813 ). Among the anticipated DNA damage chemistries, we observed significant changes only in lipid peroxidation-derived etheno adducts. We have now developed an isotope-dilution, liquid chromatography-coupled, tandem quadrupole mass spectrometric method to quantify representative species across the spectrum of RNA damage products predicted to arise at sites of inflammation, including nucleobase deamination (xanthosine and inosine), oxidation (8-oxoguanosine), and alkylation (1,N(6)-ethenoadenosine). Application of the method to the liver, spleen, and kidney from the SJL mouse model revealed generally higher levels of oxidative background RNA damage than was observed in DNA in control mice. However, compared to control mice, RcsX treatment to induce nitric oxide overproduction resulted in significant increases only in inosine and only in the spleen. Further, the nitric oxide synthase inhibitor, N-methylarginine, did not significantly affect the levels of inosine in control and RcsX-treated mice. The differences between DNA and RNA damage in the same animal model of inflammation point to possible influences from DNA repair, RcsX-induced alterations in adenosine deaminase activity, and differential accessibility of DNA and RNA to reactive oxygen and nitrogen species as determinants of nucleic acid damage during inflammation.


Asunto(s)
Inflamación/metabolismo , ARN/metabolismo , Animales , Cromatografía Liquida , ADN/metabolismo , Daño del ADN , Modelos Animales de Enfermedad , Fenómenos Genéticos , Inosina , Riñón/metabolismo , Hígado/metabolismo , Ratones , Óxido Nítrico/metabolismo , Oxidación-Reducción , Bazo/metabolismo , Espectrometría de Masas en Tándem
5.
Chem Res Toxicol ; 25(10): 2194-202, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22971010

RESUMEN

Hypoxia-inducible factor-1α (HIF-1α) is a critical regulator of cellular responses to hypoxia. Under normoxic conditions, the cellular HIF-1α level is regulated by hydroxylation by prolyl hydroxylases (PHDs), ubiquitylation, and proteasomal degradation. During hypoxia, degradation decreases, and its intracellular level is increased. Exogenously administered nitric oxide (NO)-donor drugs stabilize HIF-1α; thus, NO is suggested to mimic hypoxia. However, the role of low levels of endogenously produced NO generated during hypoxia in HIF-1α stabilization has not been defined. Here, we demonstrate that NO and reactive oxygen species (ROS) produced endogenously by human colon carcinoma HCT116 cells are responsible for HIF-1α accumulation in hypoxia. The antioxidant N-acetyl-L-cysteine (NAC) and NO synthase inhibitor N(G)-monomethyl L-arginine (L-NMMA) effectively reduced HIF-1α stabilization and decreased HIF-1α hydroxylation. These effects suggested that endogenous NO and ROS impaired PHD activity, which was confirmed by reversal of L-NMMA- and NAC-mediated effects in the presence of dimethyloxaloylglycine, a PHD inhibitor. Thiol reduction with dithiothreitol decreased HIF-1α stabilization in hypoxic cells, while dinitrochlorobenzene, which stabilizes S-nitrosothiols, favored its accumulation. This suggested that ROS- and NO-mediated HIF-1α stabilization involved S-nitrosation, which was confirmed by demonstrating increased S-nitrosation of PHD2 during hypoxia. Our results support a regulatory mechanism of HIF-1α during hypoxia in which endogenously generated NO and ROS promote inhibition of PHD2 activity, probably by its S-nitrosation.


Asunto(s)
Neoplasias del Colon/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Hipoxia de la Célula , Colon/citología , Colon/metabolismo , Colon/patología , Neoplasias del Colon/patología , Células HCT116 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Nitrosación , Procolágeno-Prolina Dioxigenasa/metabolismo
6.
Chem Res Toxicol ; 25(4): 873-83, 2012 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-22303861

RESUMEN

Dysregulated production of nitric oxide (NO•) and reactive oxygen species (ROS) by inflammatory cells in vivo may contribute to mutagenesis and carcinogenesis. Here, we compare cytotoxicity and mutagenicity induced by NO• and ROS in TK6 and AS52 cells, delivered by two methods: a well-characterized delivery system and a novel adaptation of a system for coculture. When exposed to preformed NO•, a cumulative dose of 620 µM min reduced the viability of TK6 cells at 24 h to 36% and increased mutation frequencies in the HPRT and TK1 genes to 7.7 × 10⁻6 (p < 0.05) and 24.8 × 10⁻6 (p < 0.01), 2.7- and 3.7-fold higher than background, respectively. In AS52 cells, cumulative doses of 1700 and 3700 µM min reduced viability to 49 and 22%, respectively, and increased the mutation frequency 10.2- and 14.6-fold higher than the argon control (132 × 10⁻6 and 190 × 10⁻6, respectively). These data show that TK6 cells were more sensitive than AS52 cells to killing by NO•. However, the two cell lines were very similar in relative susceptibility to mutagenesis; on the basis of fold increases in MF, average relative sensitivity values [(MF(exp)/MF(control))/cumulative NO• dose] were 5.16 × 10⁻³ and 4.97 × 10⁻³ µM⁻¹ min⁻¹ for TK6 cells and AS52 cells, respectively. When AS52 cells were exposed to reactive species generated by activated macrophages in the coculture system, cell killing was greatly reduced by the addition of NMA to the culture medium and was completely abrogated by combined additions of NMA and the superoxide scavenger Tiron, indicating the relative importance of NO• to loss of viability. Exposure in the coculture system for 48 h increased mutation frequency in the gpt gene by more than 9-fold, and NMA plus Tiron again completely prevented the response. Molecular analysis of gpt mutants induced by preformed NO• or by activated macrophages revealed that both doubled the frequency of gene inactivation (40% in induced vs 20% in spontaneous mutants). Sequencing showed that base-substitution mutations dominated the spectra, with transversions (30-40%) outnumbering transitions (10-20%). Virtually all mutations took place at guanine sites in the gene. G:C to T:A transversions accounted for about 30% of both spontaneous and induced mutations; G:C to A:T transitions amounted to 10-20% of mutants; insertions, small deletions, and multiple mutations were present at frequencies of 0-10%. Taken together, these results indicate that cell type and proximity to generator cells are critical determinants of cytotoxic and genotoxic responses induced by NO• and reactive species produced by activated macrophages.


Asunto(s)
Especies de Nitrógeno Reactivo/toxicidad , Especies Reactivas de Oxígeno/toxicidad , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Técnicas de Transferencia de Gen , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Ratones , Pruebas de Mutagenicidad , Tasa de Mutación , Óxido Nítrico/toxicidad , Timidina Quinasa/genética
7.
Nitric Oxide ; 27(3): 161-8, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22728703

RESUMEN

Nitric oxide (NO) plays key roles in cell signaling and physiology, with diverse functions mediated by NO concentrations varying over three orders-of-magnitude. In spite of this critical concentration dependence, current approaches to NO delivery in vitro result in biologically irrelevant and poorly controlled levels, with hyperoxic conditions imposed by ambient air. To solve these problems, we developed a system for controlled delivery of NO and O(2) over large concentration ranges to mimic biological conditions. Here we describe the fabrication, operation and calibration of the delivery system. We then describe applications for delivery of NO and O(2) into cell culture media, with a comparison of experimental results and predictions from mass transfer models that predict the steady-state levels of various NO-derived reactive species. We also determined that components of culture media do not affect the steady-state levels of NO or O(2) in the device. This system provides critical control of NO delivery for in vitro models of NO biology and chemistry.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Óxido Nítrico/administración & dosificación , Oxígeno/administración & dosificación , Línea Celular Tumoral , Supervivencia Celular/fisiología , Medios de Cultivo/química , Medios de Cultivo/metabolismo , Humanos , Modelos Biológicos , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Oxígeno/química , Oxígeno/metabolismo
8.
Proc Natl Acad Sci U S A ; 106(34): 14547-51, 2009 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-19706542

RESUMEN

The transcription factor NF-E2-related nuclear factor 2 (Nrf2) regulates expression of genes that protect cells from oxidative damage. Here, we characterized nitric oxide (*NO)-induced Nrf2-Kelch-like ECH-associated protein 1 (Keap1) signaling and its role in counteracting *NO-induced apoptosis of human colon cancer HCT116 cells. Nrf2 was localized in the cytoplasm in control cells; *NO triggered its rapid nuclear accumulation, transcriptional activation, and up-regulation of HO-1, NQO1, and GCL, but not GST A4 and P1 subunits. Nrf2 accumulation in the nucleus was also associated with enhanced transcription and posttranscriptional modifications. (S)-nitrosation of Keap1 may contribute to nuclear accumulation of Nrf2 by facilitating its dissociation from Keap1, thus initiating *NO-mediated Nrf2-Keap1 signaling. *NO-mediated induction of ARE-dependent genes occurred well before apoptosis, as judged by caspase 3 activation. Collectively, these results show that the Nrf2-Keap1 signaling pathway mediates protective cellular responses to mitigate *NO-induced damage and may contribute to the relative resistance of HCT116 to *NO-induced cytotoxicity.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Óxido Nítrico/farmacología , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Citoplasma/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína 1 Asociada A ECH Tipo Kelch , Microscopía Confocal , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/genética , Nitrosación , Transporte de Proteínas/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Cancer Res ; 64(9): 3022-9, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15126337

RESUMEN

Loss of p53 function by inactivating mutations results in abrogation of NO*induced apoptosis in human lymphoblastoid cells. Here we report characterization of apoptotic signaling pathways activated by NO* in these cells by cDNA microarray expression and immunoblotting. A p53-mediated transcriptional response to NO* was observed in p53-wild-type TK6, but not in closely related p53-mutant WTK1, cells. Several previously characterized p53 target genes were up-regulated transcriptionally in TK6 cells, including phosphatase PPM1D (WIP1), oxidoreductase homolog PIG3, death receptor TNFRSF6 (Fas/CD95), and BH3-only proteins BBC3 (PUMA) and PMAIP1 (NOXA). NO* also modulated levels of several gene products in the mitochondria-dependent and death-receptor-mediated apoptotic pathways. Inhibitors of apoptosis proteins X-chromosome-linked inhibitor of apoptosis, cellular inhibitor of apoptosis protein-1, and survivin were significantly down-regulated in TK6 cells, but not in WTK1 cells. Smac release from mitochondria was induced in both cell types, but release of apoptosis-inducing factor and endonuclease G was detected only in TK6 cells. Fas/CD95 was increased, and levels of the antiapoptotic proteins Bcl-2 and Bcl-x/L were reduced in TK6 cells. Activation of procaspases 3, 8, 9, and 10, as well as Bid and poly(ADP-ribose) polymerase cleavage, were observed only in TK6 cells. NO* treatment did not alter levels of death receptors 4 and 5, Fas-associated death domain or proapoptotic Bax and Bak proteins in either cell line. Collectively, these data show that NO* exposure activated a complex network of responses leading to p53-dependent apoptosis via both mitochondrial and Fas receptor pathways, which were abrogated in the presence of mutant p53.


Asunto(s)
Apoptosis/efectos de los fármacos , Linfocitos/efectos de los fármacos , Óxido Nítrico/farmacología , Proteína p53 Supresora de Tumor/fisiología , Apoptosis/fisiología , Línea Celular , Radicales Libres/química , Radicales Libres/farmacología , Perfilación de la Expresión Génica , Humanos , Linfocitos/citología , Linfocitos/metabolismo , Linfocitos/fisiología , Mutación , Óxido Nítrico/química , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
10.
ACS Chem Biol ; 11(5): 1230-7, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-26866676

RESUMEN

Monocyclic aromatic amines are widespread environmental contaminants with multiple sources such as combustion products, pharmaceuticals, and pesticides. Their phenolic metabolites are converted intracellularly to electrophilic quinone imines upon autoxidation and can embed in the cellular matrix through a transimination reaction that leaves a redox-active residue as a substituent of lysine side-chain amino groups. To demonstrate the occurrence of this process within the cellular nucleus, Chinese hamster ovary AA8 cells were treated with the para-phenol of 3,5-dimethylamine, after which the histone proteins were isolated, derivatized, and subjected to tryptic digestion. The resulting peptides were analyzed by tandem mass spectrometry to determine which lysines were modified. Nine residues in histones H2A, H2B, and H4 were identified; these were located in histone tails, close to where DNA makes contact with the nuclear core particle, elsewhere on the protein surface, and deep within the core. Kinetics of disappearance of the modified lysines in cultured cells was determined using isotope-dilution mass spectrometry. AA8 cells were also transfected with the genetically encoded hydrogen peroxide biosensor HyPer in constructs that lead to expression of HyPer in different cellular compartments. Challenging the resulting cells with the dimethylaminophenol resulted in sustained fluorescence emission in each of the compartments, demonstrating ongoing production of H2O2. The kinetics of modified lysine loss determined by mass spectrometry was consistent with persistence of HyPer fluorescence emission. We conclude that the para-phenol of 3,5-dimethylamine can become stably integrated into the histone proteins, which are minimally repaired, if at all, and function as a persistent source of intracellular H2O2.


Asunto(s)
Histonas/metabolismo , Iminas/metabolismo , Lisina/metabolismo , Quinonas/metabolismo , Secuencia de Aminoácidos , Aminofenoles/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Histonas/química , Peróxido de Hidrógeno/metabolismo , Iminas/química , Lisina/análisis , Modelos Moleculares , Procesamiento Proteico-Postraduccional , Quinonas/química
11.
Free Radic Biol Med ; 39(11): 1489-98, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16274884

RESUMEN

Glutathione (GSH) plays an important role in cellular defense response in many in vitro and in vivo models. Here we investigated its role in NO()-induced toxicity in cell culture and mouse models. Wild-type (TK6) and p53-null (NH32) human lymphoblastoid cells were treated with NO(.) at a steady-state concentration of 0.6 muM, similar to the level estimated to occur in inflamed tissues. In both cell types, GSH was depleted by this exposure in a dose- and time-dependent manner. Contrary to expectations, prior depletion of GSH by treatment with l-buthionine-SR-sulfoximine did not potentiate NO(.)-induced cell killing or DNA deamination in TK6 cells. In activated RAW264.7 murine macrophages producing NO(.), intracellular GSH content did not change, although gamma-glutamate-cysteine ligase was upregulated. NO(.) overproduction in RcsX lymphoma-bearing SJL mice resulted in significantly elevated GSH levels in various organs. Administration of the NO(.) synthase inhibitor N-methylarginine abolished the increase in GSH in these animals. Collectively, these data indicate a multifaceted and complex involvement of GSH in responses of cells and tissues to toxic levels of NO(.). NO(.) treatment effectively depleted GSH levels in human lymphoblastoid cells, but this alteration was not a critical initiating factor for NO(.)-mediated toxicity. Murine macrophages maintained GSH homeostasis when exposed to endogenously produced NO(.). In RcsX lymphoma-bearing mice, upregulation of de novo synthesis of GSH appeared to be a response to the toxic effects of NO(.).


Asunto(s)
Glutatión/fisiología , Óxido Nítrico/toxicidad , Animales , Línea Celular , Supervivencia Celular , Humanos , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Macrófagos , Ratones , Células Tumorales Cultivadas
12.
ILAR J ; 46(3): 258-68, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15953833

RESUMEN

Antibodies are host proteins that comprise one of the principal effectors of the adaptive immune system. Their utility has been harnessed as they have been and continue to be used extensively as a diagnostic and research reagent. They are also becoming an important therapeutic tool in the clinician's armamentarium to treat disease. Antibodies are utilized for analysis, purification, and enrichment, and to mediate or modulate physiological responses. This overview of the structure and function of polyclonal and monoclonal antibodies describes features that distinguish one from the other. A limited review of their use as specific research, diagnostic, and therapeutic reagents and a list of printed and electronic resources that can be utilized to garner additional information on these topics are also included.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos/inmunología , Inmunoterapia/métodos , Anticuerpos/química , Anticuerpos/uso terapéutico , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Especificidad de Anticuerpos , Epítopos , Femenino , Humanos , Inmunoglobulinas/química , Inmunoglobulinas/inmunología , Indicadores y Reactivos
13.
Toxicol Sci ; 141(1): 300-13, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24973092

RESUMEN

Epidemiological studies have demonstrated extensive human exposure to the monocyclic aromatic amines, particularly to 3,5-dimethylaniline, and found an association between exposure to these compounds and risk for bladder cancer. Little is known about molecular mechanisms that might lead to the observed risk. We previously suggested that the hydroxylated 3,5-dimethylaniline metabolite, 3,5-dimethylaminophenol (3,5-DMAP), played a central role in effecting genetic change through the generation of reactive oxygen species (ROS) in a redox cycle with 3,5-dimethylquinoneimine. Experiments here characterize ROS generation by 3,5-DMAP exposure in nucleotide repair-proficient and -deficient Chinese hamster ovary cells as a function of time. Besides, various cellular responses discussed herein indicate that ROS production is the principal cause of cytotoxicity. Fluorescence microscopy of cells exposed to 3,5-DMAP confirmed that ROS production occurs in the nuclear compartment, as suggested by a previous study demonstrating covalent linkage between 3,5-DMAP and histones. 3,5-DMAP was also compared with 3,5-dimethylhydroquinone to determine whether substitution of one of the phenolic hydroxyl groups by an amino group had a significant effect on some of the investigated parameters. The comparatively much longer duration of observable ROS produced by 3,5-DMAP (7 vs. 1 day) provides further evidence that 3,5-DMAP becomes embedded in the cellular matrix in a form capable of continued redox cycling. 3,5-DMAP also induced dose-dependent increase of H2O2 and ·OH, which were determined as the major free radicals contributing to the cytotoxicity and apoptosis mediated via caspase-3 activation. Overall, this study provides insight into the progression of alkylaniline-induced toxicity.


Asunto(s)
Aminofenoles/toxicidad , Apoptosis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Animales , Células CHO , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Histonas/metabolismo , Microscopía Fluorescente
14.
Food Chem Toxicol ; 72: 98-110, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25014158

RESUMEN

Most common alkylanilines in the environment are 2,6-dimethylaniline (2,6-DMA), 3,5-dimethylaniline (3,5-DMA), and 3-ethylaniline (3-EA). 3,5-Dimethylaminophenol (3,5-DMAP), a metabolite of 3,5-DMA, is of particular interest, as it is potentially genotoxic. Supplementation with organic or inorganic forms of selenium (Se) may reduce toxicity following exposure to a wide variety of environmental chemicals. This study was designed to evaluate the protective effects of sodium selenite (SS) and selenomethionine (SM) at varying time points of supplementation (24 h and 72 h) against the cytotoxicity, reactive oxygen species (ROS) production, and genotoxicity of 3,5-DMAP in CHO AS52 cells. 3,5-DMAP caused dose-dependent increase of cytotoxicity, ROS production and genotoxicity, and generated free radicals in the nuclei. Thioredoxin reductase (TrxR), catalase and glutathione reductase activities, and glutathione levels were significantly lower while lipid peroxidation and protein oxidation levels were higher after 3,5-DMAP treatment in both cytoplasm and the nucleus vs. control. After 24 h, both SS and SM provided protection in antioxidant/oxidant status of the 3,5-DMAP-treated cells; however other than supplying higher glutathione peroxidase and TrxR activities, 72 h supplementation did not provide advanced improvement. Selenocompounds may be beneficial against cytotoxic and genotoxic potential of 3,5-DMAP and might protect both nucleus and cytoplasm following exposure to alkylanilines.


Asunto(s)
Compuestos de Anilina/química , Compuestos de Anilina/toxicidad , Animales , Antioxidantes/farmacología , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Línea Celular , Ensayo Cometa , Cricetinae , Daño del ADN/efectos de los fármacos , Suplementos Dietéticos , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Selenometionina/farmacología , Selenito de Sodio/farmacología , Reductasa de Tiorredoxina-Disulfuro/metabolismo
15.
Nat Nanotechnol ; 8(11): 873-80, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24185942

RESUMEN

Single-walled carbon nanotubes are particularly attractive for biomedical applications, because they exhibit a fluorescent signal in a spectral region where there is minimal interference from biological media. Although single-walled carbon nanotubes have been used as highly sensitive detectors for various compounds, their use as in vivo biomarkers requires the simultaneous optimization of various parameters, including biocompatibility, molecular recognition, high fluorescence quantum efficiency and signal transduction. Here we show that a polyethylene glycol ligated copolymer stabilizes near-infrared-fluorescent single-walled carbon nanotubes sensors in solution, enabling intravenous injection into mice and the selective detection of local nitric oxide concentration with a detection limit of 1 µM. The half-life for liver retention is 4 h, with sensors clearing the lungs within 2 h after injection, thus avoiding a dominant route of in vivo nanotoxicology. After localization within the liver, it is possible to follow the transient inflammation using nitric oxide as a marker and signalling molecule. To this end, we also report a spatial-spectral imaging algorithm to deconvolute fluorescence intensity and spatial information from measurements. Finally, we demonstrate that alginate-encapsulated single-walled carbon nanotubes can function as implantable inflammation sensors for nitric oxide detection, with no intrinsic immune reactivity or other adverse response for more than 400 days.


Asunto(s)
Técnicas Biosensibles/instrumentación , Técnicas Biosensibles/métodos , Nanotubos de Carbono/química , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacocinética , ADN/química , Inflamación/patología , Ligandos , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Óxido Nítrico/metabolismo , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Polímeros/química , Especies de Nitrógeno Reactivo/metabolismo
16.
Environ Mol Mutagen ; 53(7): 567-73, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22733615

RESUMEN

Aflatoxin B(1) (AFB(1) ) is a potent mutagen and an important risk factor for hepatocellular carcinoma (HCC) in humans. Transgenic mouse strains and cells in culture have been used to detect different types of mutations caused by AFB(1) and investigate the molecular determinants of their location and frequency. The AFB(1) mutational spectrum in the gpt gene was markedly different in AS52 cells compared with the liver in gpt delta B6C3F1 transgenic mice. The results demonstrate the importance of metabolism, chromosomal location, transcription and selection conditions on mutational spectra.


Asunto(s)
Aflatoxina B1/toxicidad , Alanina Transaminasa/genética , Mutación/efectos de los fármacos , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN/genética , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN
17.
Toxicol Sci ; 130(1): 48-59, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22831970

RESUMEN

Several alkylanilines with structures more complex than toluidines have been associated epidemiologically with human cancer. Their mechanism of action remains largely undetermined, and there is no reported evidence that it replicates that of multicyclic aromatic amines even though the principal metabolic pathways of P450-mediated hydroxylation and phase II conjugation are very similar. As a means to elucidate their mechanisms of action, lethality and mutagenicity in the adenine phosphoribosyltransferase (aprt (+/-)) gene induced in several Chinese hamster ovary cell types by 2,6- and 3,5-dimethylaniline (2,6-DMA, 3,5-DMA) and their N- and ring-hydroxyl derivatives (N-OH-2,6-DMA, N-OH-3,5-DMA, 2,6-DMAP, 3,5-DMAP) were assessed. Dose-response relationships were determined in the parental AA8 cell line, its repair-deficient UV5 subclone and other repair-deficient 5P3NAT2 or -proficient 5P3NAT2R9 subclones engineered to express mouse cytochrome P4501A2 (CYP1A2) and human N-acetyltransferase (NAT2), and also in AS52 cells harboring the bacterial guanine-hypoxanthine phosphoribosyltransferase (gpt) gene. Mutations in the gpt gene of AS52 cells were characterized and found to be dominated by G:C to A:T and A:T to G:C transitions. Separately, treatment of AS52 cells with N-OH-2,6-DMA, N-OH-3,5-DMA, 2,6-DMAP, 3,5-DMAP, and 3,5-DMAP led to intracellular production of reactive oxygen species (ROS) for at least 24h after removal of the mutagens in every case. Using the comet assay, DNA strand breaks were observed in a dose-dependent manner in AS52 cells when treated with each of the four N-OH-2,6-DMA, N-OH-3,5-DMA, 2,6-DMAP, and 3,5-DMAP derivatives. Comparative evaluation of the results indicates that the principal mechanism of mutagenic action is likely to be through redox cycling of intracellularly bound aminophenol/quinone imine structures to generate ROS rather than through formation of covalent DNA adducts.


Asunto(s)
Compuestos de Anilina/toxicidad , Mutágenos/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Compuestos de Anilina/metabolismo , Animales , Células CHO , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cricetinae , Cricetulus , Aductos de ADN , Roturas del ADN de Doble Cadena/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Pruebas de Mutagenicidad , Mutágenos/metabolismo , Mutación/efectos de los fármacos , Oxidación-Reducción
18.
Toxicol Sci ; 128(2): 326-33, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22539618

RESUMEN

Aflatoxin B (1) (AFB(1)) is a risk factor for hepatocellular carcinoma in humans. Infant, but not adult, mice are sensitive to AFB(1)-induced liver carcinogenesis; a single dose during the neonatal period leads to hepatocellular carcinoma in adulthood. Earlier work defined the mutational spectrum in the gpt gene of gpt delta B6C3F1 mice 3 weeks after exposure to aflatoxin. In the present study, we examined the gpt spectrum 10 weeks postdosing and expanded the study to examine, at 3 and 10 weeks, the spectrum at a second locus, the red/gam genes of the mouse λEG10 transgene. Whereas the gpt locus is typically used to define local base changes, the red/gam genes, via the Spi(-) assay, often are used to detect more global mutations such as large deletions and rearrangements. Three weeks after dosing with AFB(1), there was a 10-fold increase over the control in the Spi(-) mutant fraction (MF) in liver DNA; after 10 weeks, a further increase was observed. The MF in the gpt gene was also increased at 10 weeks compared with the MF at 3 weeks. No gender-specific differences were found in the Spi(-) or gpt MFs. Whereas Spi(-) mutations often signal large genetic changes, they did not in this specific case. The Spi(-) spectrum was dominated by GC to TA transversions, with one exceptionally strong hotspot at position 314. Using two genetic loci, the data show a strong preference for the induction of GC to TA mutations in mice, which is the dominant mutation seen in people exposed to aflatoxin.


Asunto(s)
Aflatoxina B1/toxicidad , Hígado/efectos de los fármacos , Aflatoxina B1/administración & dosificación , Animales , Animales Recién Nacidos , Secuencia de Bases , Cartilla de ADN , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C3H , Reacción en Cadena de la Polimerasa
19.
Toxicol Sci ; 122(1): 38-44, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21507988

RESUMEN

Exposure to genotoxic chemicals at a young age increases cancer incidence later in life. Aflatoxin B(1) (AFB(1)) is a potent genotoxin that induces hepatocellular carcinoma (HCC) in many animal species and in humans. Whereas adult mice are insensitive to aflatoxin-induced carcinogenesis, mice treated with AFB(1) shortly after birth develop a high incidence of HCC in adulthood. Furthermore, the incidence of HCC in adult male mice treated as infants is much greater than in females, reasons for which are unclear. In this study, treatment with AFB(1) produced similar levels of DNA damage and mutations in the liver of newborn male and female gpt delta B6C3F1 mice. Twenty-four hours after dosing with AFB(1) (6 mg/kg), the highly mutagenic AFB(1)-FAPY adduct was present at twice the level of AFB(1)-N(7)-guanine in liver DNA of males and females. A multiple dose regimen (3 × 2 mg/kg), while delivering the same total dose, resulted in lower AFB(1) adduct levels. Mutation frequencies in the gpt transgene in liver were increased by 20- to 30-fold. The most prominent mutations in AFB(1)-treated mice were G:C to T:A transversions and G:C to A:T transitions. At this 21-day time point, no significant differences were found in mutation frequency or types of mutations between males and females. These results show that infant male and female B6C3F1 mice experience similar amounts of DNA damage and mutation from AFB(1) that may initiate the neoplastic process. The gender difference in the subsequent development of HCC highlights the importance of elucidating additional factors that modulate HCC development.


Asunto(s)
Aflatoxina B1/metabolismo , Aflatoxina B1/toxicidad , Aductos de ADN/metabolismo , Aductos de ADN/toxicidad , Hígado/efectos de los fármacos , Factores de Edad , Animales , Animales Recién Nacidos , Secuencia de Bases , Pruebas de Carcinogenicidad , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Femenino , Guanina/metabolismo , Hígado/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Mutágenos/toxicidad , Tasa de Mutación , Análisis de Secuencia de ADN , Factores Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA