Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
J Biol Chem ; 299(12): 105385, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37890780

RESUMEN

Cancer is a genetic disease requiring multiple mutations for its development. However, many carcinogens are DNA-unreactive and nonmutagenic and consequently described as nongenotoxic. One of such carcinogens is nickel, a global environmental pollutant abundantly emitted by burning of coal. We investigated activation of DNA damage responses by Ni and identified this metal as a replication stressor. Genotoxic stress markers indicated the accumulation of ssDNA and stalled replication forks, and Ni-treated cells were dependent on ATR for suppression of DNA damage and long-term survival. Replication stress by Ni resulted from destabilization of RRM1 and RRM2 subunits of ribonucleotide reductase and the resulting deficiency in dNTPs. Ni also increased DNA incorporation of rNMPs (detected by a specific fluorescent assay) and strongly enhanced their genotoxicity as a result of repressed repair of TOP1-DNA protein crosslinks (TOP1-DPC). The DPC-trap assay found severely impaired SUMOylation and K48-polyubiquitination of DNA-crosslinked TOP1 due to downregulation of specific enzymes. Our findings identified Ni as the human carcinogen inducing genome instability via DNA-embedded ribonucleotides and accumulation of TOP1-DPC which are carcinogenic abnormalities with poor detectability by the standard mutagenicity tests. The discovered mechanisms for Ni could also play a role in genotoxicity of other protein-reactive carcinogens.


Asunto(s)
Carcinógenos , Replicación del ADN , Níquel , Nucleótidos , Humanos , Carcinógenos/toxicidad , ADN/metabolismo , Daño del ADN , Reparación del ADN , Replicación del ADN/efectos de los fármacos , ADN-Topoisomerasas de Tipo I/metabolismo , Níquel/toxicidad , Saccharomyces cerevisiae/metabolismo , Nucleótidos/biosíntesis
2.
Toxicol Appl Pharmacol ; 477: 116696, 2023 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-37734571

RESUMEN

Inhalation exposure to hexavalent chromium is known to cause lung cancer and other pulmonary toxicity. Cellular metabolism of chromium(VI) entering cells as chromate anion produces different amounts of reactive Cr(V) intermediates and finally yields Cr(III). Direct reduction of Cr(VI) by ascorbate (Asc), the dominant metabolic reaction in vivo but not in standard cell cultures, skips production of Cr(V) but still permits extensive formation of Cr-DNA damage. To understand the importance of different forms of biological injury in Cr(VI) toxicity, we examined activation of several protein- and DNA damage-sensitive stress responses in human lung cells under Asc-restored conditions. We found that Asc-restored cells suppressed upregulation of oxidant-sensitive stress systems by Cr(VI) but showed a strong activation of the apical DNA damage-responsive kinase ATR. ATR signaling was triggered in late S phase and persisted upon entry of cells into G2 phase. Inhibition of ATR prevented the establishment of late-S and G2 cell cycle checkpoints and did not lead to a compensatory activation of a related kinase ATM. Inactivation of ATR also strongly impaired viability of Cr(VI)-treated lung cells including stem-like cells and revealed a significant formation of toxic Cr-DNA damage at low Cr(VI) doses. Our findings identified a major Cr(VI) resistance mechanism involving sensing of Cr-DNA damage by ATR in late S phase and a subsequent establishment of protective cell cycle checkpoints.

3.
Toxicol Appl Pharmacol ; 445: 116041, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35504338

RESUMEN

Transcription factors HIF1 and HIF2 are central regulators of physiological responses to hypoxia and important for normal functioning of tissue stem cells and maintenance of healthy microvasculature. Even modest decreases in HIF activity exert detrimental effects in tissues although it is unclear what factors can directly impair HIF functions. We hypothesized that the presence of functionally important, large intrinsically disordered regions in HIFα subunits of HIF1/2 could make them structurally vulnerable to protein-damaging conditions. We found that common protein-damaging agents such as endogenous/exogenous aldehydes (formaldehyde, acetaldehyde), moderate heat shock and the environmental toxicant cadmium cause inactivation of HIF1 and HIF2 due to structural damage to HIFα subunits. Aldehydes triggered a rapid and selective depletion of HIF1α and HIF2α, which occurred as a result of enhanced binding of Pro-hydroxylated/VHL-ubiquitinated HIFα by 26S proteasomes. In the absence of proteasomal degradation, aldehyde-damaged HIF1 and HIF2 were transactivation defective and HIFα subunits became insoluble/denatured when their VHL-mediated ubiquitination was blocked. Protein damage by heat shock and cadmium resulted in the insolubility of Pro-nonhydroxylated HIFα. Thus, VHL-dependent ubiquitination of damaged HIFα also acts as means to maintain their solubility, permitting capture by proteasomes. The observed control of HIFα stability at the point of proteasome binding may extend to several posttranslational modifications that occur in the conformationally flexible regions of these proteins. Our findings revealed vulnerability of HIF1 and HIF2 to direct inactivation by protein-damaging agents, which helps understand their tissue injury mechanisms and favorable responses of hypoxic tumors to hyperthermia.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Cadmio , Aldehídos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cadmio/metabolismo , Humanos , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ubiquitinación
4.
Chem Res Toxicol ; 35(10): 1690-1700, 2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-35948068

RESUMEN

Cellular homeostasis is continuously challenged by damage from reactive oxygen species (ROS) and numerous reactive electrophiles. Human cells contain various protective systems that are upregulated in response to protein damage by electrophilic or oxidative stress. In addition to the NRF2-mediated antioxidant response, ROS and reactive electrophiles also activate HSF1 and HIF1 that control heat shock response and hypoxia response, respectively. Here, we review chemical and biological mechanisms of activation of these three transcription factors by ROS/reactive toxicants and the roles of their gene expression programs in antioxidant protection. We also discuss how NRF2, HSF1, and HIF1 responses establish multilayered cellular defenses consisting of largely nonoverlapping programs, which mitigates limitations of each response. Some innate immunity links in these stress responses help eliminate damaged cells, whereas others suppress deleterious inflammation in normal tissues but inhibit immunosurveillance of cancer cells in tumors.


Asunto(s)
Antioxidantes , Factor 2 Relacionado con NF-E2 , Antioxidantes/metabolismo , Factores de Transcripción del Choque Térmico/metabolismo , Humanos , Sistema Inmunológico/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Oxidantes , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
5.
Arch Toxicol ; 95(12): 3623-3631, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34596731

RESUMEN

Ascorbate has many biological activities that involve fundamental cellular functions such as gene expression, differentiation, and redox homeostasis. Biochemically, it serves as a cofactor for a large family of dioxygenases (> 60 members) which control transcription, formation of extracellular matrix, and epigenetic processes of histone and DNA demethylation. Ascorbate is also a major antioxidant acting as a very effective scavenger of primary reactive oxygen species. Reduction of Fe(III) by ascorbate is important for cellular uptake of iron via DMT1. Cell culture models are extensively used in toxicology and pharmacology for mechanistic studies of nutrients, drugs and other xenobiotics. High-throughput screens in vitro, such as a large-scale Tox21 program in the US, offers opportunities to assess hazardous properties of a vast and growing number of industrial chemicals. However, cells in typical cultures are severely deficient in ascorbate, raising concerns about their ability to accurately recapitulate toxic and other responses in vivo. Scarcity of ascorbate and a frequently unrecognized use of media with its thiol substitute alters stress sensitivity of cells in different directions. Remediation of ascorbate deficiency in tissue culture restores the physiological state of many cellular processes and it should improve a currently limited toxicity predictability of in vitro bioassays.


Asunto(s)
Antioxidantes/farmacología , Deficiencia de Ácido Ascórbico/fisiopatología , Ácido Ascórbico/farmacología , Animales , Antioxidantes/metabolismo , Ácido Ascórbico/metabolismo , Células Cultivadas , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Técnicas In Vitro , Pruebas de Toxicidad/métodos
6.
Chem Res Toxicol ; 33(10): 2515-2526, 2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-33001635

RESUMEN

Vitamin C (ascorbic acid) is a water-soluble antioxidant and a cofactor for a large number of enzymes. It is present in all tissues and especially abundant in corneal epithelium, stem cells, and neurons. Although similar to thiols in its ability to react with many reactive oxygen species (ROS), ascorbate is much better (>100× faster) than glutathione at scavenging of primary ROS (superoxide radical and singlet oxygen). Ascorbate appears to be especially important for elimination of O2•- in the nucleus which contains little or no SOD activity. Cofactor functions of ascorbate involve the maintenance of activity of Fe(II)/2-oxoglutarate-dependent dioxygenases via reduction of Fe(III). The most prominent activity of ascorbate-dependent dioxygenases in the cytoplasm is hydroxylation of prolines in proteins involved in the formation of extracellular matrix and regulation of metabolism and hypoxia responses. In the nucleus, ascorbate is important for oxidative demethylation of 5-methylcytosine in DNA (by TET proteins) and removal of methyl groups from histone lysines (by JmjC demethylases). Differentiation and other cellular reprograming processes involving DNA demethylation are especially sensitive to ascorbate insufficiency. High doses of vitamin C alone or in combinations with drugs produced cancer-suppressive effects which involved redox, immune, and epigenetic mechanisms. Solutions to vitamin C deficiency in cultured cells are discussed to improve the physiological relevance of in vitro models. An abundance of vitamin C in rodents limits their ability to fully recapitulate human sensitivity to adverse health effects of malnutrition and xenobiotics, including neurotoxicity, lung injury, and intergenerational and other epigenetic effects.


Asunto(s)
Ácido Ascórbico/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Animales , Ácido Ascórbico/química , Núcleo Celular/química , Citoplasma/química , ADN/metabolismo , Humanos
7.
Chem Res Toxicol ; 32(7): 1318-1319, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31046246

RESUMEN

N-Acetylcysteine is a commonly used antioxidant that is broadly effective despite its limited reactive oxygen species (ROS) reactivity. Chemoprotection by N-acetylcysteine frequently results from inactivation of primary toxicants or reactive electrophiles arising as metabolites or lipid peroxidation products. ROS are linked to the development of many human diseases and biological injury by numerous xenobiotics. Oxidative damage is the first mechanism that is often tested for toxicants. There is also a frequent projection of the established ROS mechanism for one member to a broader group to which this chemical belongs. However, the biological significance of oxidative processes is not always easy to establish, as oxidants could be a cause or result of cellular injury. The role of ROS is tested through genetic manipulations of oxidative stress-protective proteins and addition of small antioxidants. In general, genetic approaches produce protective effects weaker than those of small antioxidants, which can reflect different anti-ROS specificity. Another possibility is that chemical antioxidants have ROS-unrelated chemoprotective activities.


Asunto(s)
Acetilcisteína/metabolismo , Depuradores de Radicales Libres/metabolismo , Acetilcisteína/química , Aldehídos/química , Aldehídos/metabolismo , Disulfuros/química , Disulfuros/metabolismo , Depuradores de Radicales Libres/química , Metales Pesados/química , Metales Pesados/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/química , Especies Reactivas de Oxígeno/metabolismo
8.
Molecules ; 24(3)2019 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-30695991

RESUMEN

Ascorbic acid (vitamin C) has been gaining attention as a potential treatment for human malignancies. Various experimental studies have shown the ability of pharmacological doses of vitamin C alone or in combinations with clinically used drugs to exert beneficial effects in various models of human cancers. Cytotoxicity of high doses of vitamin C in cancer cells appears to be related to excessive reactive oxygen species generation and the resulting suppression of the energy production via glycolysis. A hallmark of cancer cells is a strongly upregulated aerobic glycolysis, which elevates its relative importance as a source of ATP (Adenosine 5'-triphosphate). Aerobic glycolysis is maintained by a highly increased uptake of glucose, which is made possible by the upregulated expression of its transporters, such as GLUT-1, GLUT-3, and GLUT-4. These proteins can also transport the oxidized form of vitamin C, dehydroascorbate, permitting its preferential uptake by cancer cells with the subsequent depletion of critical cellular reducers as a result of ascorbate formation. Ascorbate also has a potential to affect other aspects of cancer cell metabolism due to its ability to promote reduction of iron(III) to iron(II) in numerous cellular metalloenzymes. Among iron-dependent dioxygenases, important targets for stimulation by vitamin C in cancer include prolyl hydroxylases targeting the hypoxia-inducible factors HIF-1/HIF-2 and histone and DNA demethylases. Altered metabolism of cancer cells by vitamin C can be beneficial by itself and promote activity of specific drugs.


Asunto(s)
Ácido Ascórbico/farmacología , Ácido Ascórbico/uso terapéutico , Neoplasias/terapia , Animales , Terapia Combinada , Sinergismo Farmacológico , Humanos , Hipoxia/tratamiento farmacológico , Hipoxia/genética , Hipoxia/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Resultado del Tratamiento
9.
Toxicol Appl Pharmacol ; 355: 238-246, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30006243

RESUMEN

DNA double-strand breaks (DSBs) are a highly toxic form of DNA damage produced by a number of carcinogens, drugs, and metabolic abnormalities. Involvement of DSBs in many pathologies has led to frequent measurements of these lesions, primarily via biodosimetry of S139-phosphorylated histone H2AX (γ-H2AX). However, γ-H2AX is also induced by some non-DSB conditions and abundantly formed in apoptosis, raising concerns about the overestimation of potential genotoxic agents and accuracy of DSB assessments. DSB-triggered γ-H2AX undergoes RNF168-mediated K13/K15 monoubiquitination, which is rarely analyzed in DSB/genotoxicity studies. Here we identified critical methodological factors that are necessary for the efficient detection of mono- (ub1) and diubiquitinated (ub2) γ-H2AX. Using optimized technical conditions, we found that γ-H2AX-ub1 was a predominant form of γ-H2AX in three primary human cell lines containing mechanistically distinct types of DSBs. Replication stress-associated DSBs also triggered extensive formation of γ-H2AX-ub1. For DSBs induced by oxidative damage or topoisomerase II, both γ-H2AX and γ-H2AX-ub1 showed dose-dependent increases whereas γ-H2AX-ub2 plateaued at low levels of breaks. Despite abundance of γ-H2AX, γ-H2AX-ub1,2 formation was blocked in apoptosis, which was associated with proteolytic cleavage of RNF168. Chromatin damage also caused only the production of γ-H2AX but not its ub1,2 forms. Our results revealed a major contribution of ubiquitinated forms to the overall γ-H2AX response and demonstrated the specificity of monoubiquitinated γ-H2AX as a biodosimeter of non-apoptotic DSBs.


Asunto(s)
Biomarcadores/análisis , Roturas del ADN de Doble Cadena/efectos de los fármacos , Histonas/metabolismo , Ubiquitinación/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular , Cromatina/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/metabolismo , Relación Dosis-Respuesta a Droga , Técnicas de Silenciamiento del Gen , Respuesta al Choque Térmico , Histonas/genética , Humanos , Estrés Oxidativo/efectos de los fármacos , Células Madre
10.
Chem Res Toxicol ; 31(11): 1172-1184, 2018 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-30362728

RESUMEN

Epidemiological studies in chromate production have established hexavalent chromium as a potent lung carcinogen. Inhalation of chromium(VI) most often occurs in mixtures with other metals as among stainless steel welders, which is the largest occupational group with Cr(VI) exposure. Surprisingly, carcinogenicity of Cr(VI)-containing welding fumes is moderate and not consistently higher than that of Cr-free welding. Here, we investigated interactions between chromate and three other metal ions [Fe(III), Mn(II), Ni(II)] that are typically released from stainless steel welding particles. In human lung epithelial cells with physiological levels of ascorbate and glutathione, Cr(VI) was by far the most cytotoxic metal in single exposures. Coexposure with Fe(III) suppressed cytotoxicity and genotoxicity of Cr(VI), which resulted from a severe inhibition of Cr uptake by cells and required extracellular ascorbate/glutathione. Chemically, detoxification of Cr(VI) occurred via its rapid extracellular reduction by Fe(II) that primarily originated from ascorbate-reduced Fe(III). Glutathione was a significant contributor to reduction of Cr(VI) by Fe only in the presence of ascorbate. We further found that variability in Cr(VI) metabolism among common cell culture media was caused by their different Fe content. Ni(II) and Mn(II) had no detectable effects on metabolism, cellular uptake or cytotoxicity of Cr(VI). The main biological findings were confirmed in three human lung cell lines, including stem cell-like and primary cells. We discovered extracellular detoxification of carcinogenic chromate in coexposures with Fe(III) ions and identified the underlying chemical mechanism. Our findings established an important case when exposure to mixtures causes inactivation of a potent human carcinogen.


Asunto(s)
Cromo/química , Gases/química , Hierro/química , Metales/química , Soldadura , Contaminantes Ocupacionales del Aire/química , Contaminantes Ocupacionales del Aire/toxicidad , Ácido Ascórbico/química , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cromo/metabolismo , Cromo/toxicidad , Daño del ADN/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Glutatión/química , Humanos , Oxidación-Reducción
11.
Nucleic Acids Res ; 44(1): 198-209, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26420831

RESUMEN

Many carcinogens damage both DNA and protein constituents of chromatin, and it is unclear how cells respond to this compound injury. We examined activation of the main DNA damage-responsive kinase ATM and formation of DNA double-strand breaks (DSB) by formaldehyde (FA) that forms histone adducts and replication-blocking DNA-protein crosslinks (DPC). We found that low FA doses caused a strong and rapid activation of ATM signaling in human cells, which was ATR-independent and restricted to S-phase. High FA doses inactivated ATM via its covalent dimerization and formation of larger crosslinks. FA-induced ATM signaling showed higher CHK2 phosphorylation but much lower phospho-KAP1 relative to DSB inducers. Replication blockage by DPC did not produce damaged forks or detectable amounts of DSB during the main wave of ATM activation, which did not require MRE11. Chromatin-monitoring KAT5 (Tip60) acetyltransferase was responsible for acetylation and activation of ATM by FA. KAT5 and ATM were equally important for triggering of intra-S-phase checkpoint and ATM signaling promoted recovery of normal human cells after low-dose FA. Our results revealed a major role of the KAT5-ATM axis in protection of replicating chromatin against damage by the endogenous carcinogen FA.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Cromatina/genética , Cromatina/metabolismo , Daño del ADN/efectos de los fármacos , Replicación del ADN , Formaldehído/toxicidad , Histona Acetiltransferasas/metabolismo , Carcinógenos/toxicidad , Línea Celular , Activación Enzimática/efectos de los fármacos , Humanos , Lisina Acetiltransferasa 5 , Fase S , Transducción de Señal/efectos de los fármacos
12.
Am J Pathol ; 186(11): 2857-2868, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27639166

RESUMEN

Endogenous and exogenous formaldehyde (FA) has been linked to cancer, neurotoxicity, and other pathophysiologic effects. Molecular and cellular mechanisms that underlie FA-induced damage are poorly understood. In this study, we investigated whether proteotoxicity is an important, unrecognized factor in cell injury caused by FA. We found that irrespective of their cell cycle phases, all FA-treated human cells rapidly accumulated large amounts of proteins with proteasome-targeting K48-linked polyubiquitin, which was comparable with levels of polyubiquitination in proteasome-inhibited MG132 controls. Both nuclear and cytoplasmic proteins were damaged and underwent K48-polyubiquitination. There were no significant changes in the nonproteolytic K63-polyubiquitination of soluble and insoluble cellular proteins. FA also rapidly induced nuclear accumulation and Ser326 phosphorylation of the main heat shock-responsive transcription factor HSF1, which was not a result of protein polyubiquitination. Consistent with the activation of the functional heat shock response, FA strongly elevated the expression of HSP70 genes. In contrast to the responsiveness of the cytoplasmic protein damage sensor HSF1, FA did not activate the unfolded protein response in either the endoplasmic reticulum or mitochondria. Inhibition of HSP90 chaperone activity increased the levels of K48-polyubiquitinated proteins and diminished cell viability after FA treatment. Overall, our results indicate that FA is a strong proteotoxic agent, which helps explain its diverse pathologic effects, including injury in nonproliferative tissues.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Formaldehído/efectos adversos , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Factores de Transcripción/metabolismo , Ubiquitinación/efectos de los fármacos , Línea Celular , Proteínas de Unión al ADN/genética , Retículo Endoplásmico/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/genética , Factores de Transcripción del Choque Térmico , Humanos , Leupeptinas/efectos adversos , Lisina/metabolismo , Fosforilación , Poliubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Transcripción/genética
13.
Toxicol Appl Pharmacol ; 331: 94-100, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28552779

RESUMEN

Nickel is a human carcinogen that acts as a hypoxia mimic by activating the transcription factor HIF-1α and hypoxia-like transcriptomic responses. Hypoxia and elevated HIF-1α are typically associated with drug resistance in cancer cells, which is caused by increased drug efflux and other mechanisms. Here we examined the role of HIF-1α in uptake of soluble Ni(II) and Ni(II)-induced cell fate outcomes using si/shRNA knockdowns and gene deletion models. We found that HIF-1α had no effect on accumulation of Ni(II) in two transformed (H460, A549) and two normal human cell lines (IMR90, WI38). The loss of HIF-1α also produced no significant impact on p53-dependent and p53-independent apoptotic responses or clonogenic survival of Ni(II)-treated transformed cells. In normal human cells, HIF-1α enhanced the ability of Ni(II) to inhibit cell proliferation and cause a permanent growth arrest (senescence). Consistent with its growth-suppressive effects, HIF-1α was important for upregulation of the cell cycle inhibitors p21 (CDKN1A) and p27 (CDKN1B). Irrespective of HIF-1α status, Ni(II) strongly increased levels of MYC protein but did not change protein expression of the cell cycle-promoting phosphatase CDC25A or the CDK inhibitor p16. Our findings indicate that HIF-1α limits propagation of Ni(II)-damaged normal cells, suggesting that it may act in a tumor suppressor-like manner during early stages of Ni(II) carcinogenesis.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Senescencia Celular/fisiología , Inhibidores de Crecimiento/toxicidad , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Níquel/toxicidad , Células A549 , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Senescencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores de Crecimiento/farmacología , Humanos , Níquel/farmacología
14.
Chem Res Toxicol ; 30(9): 1720-1729, 2017 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-28759204

RESUMEN

Inhalation of soluble chromium(VI) is firmly linked with higher risks of lung cancer in humans. However, comparative studies in rats have found a high lung tumorigenicity for moderately soluble chromates but no tumors for highly soluble chromates. These major species differences remain unexplained. We investigated the impact of extracellular reducers on responses of human and rat lung epithelial cells to different Cr(VI) forms. Extracellular reduction of Cr(VI) is a detoxification process, and rat and human lung lining fluids contain different concentrations of ascorbate and glutathione. We found that reduction of chromate anions in simulated lung fluids was principally driven by ascorbate with only minimal contribution from glutathione. The addition of 500 µM ascorbate (∼rat lung fluid concentration) to culture media strongly inhibited cellular uptake of chromate anions and completely prevented their cytotoxicity even at otherwise lethal doses. While proportionally less effective, 50 µM extracellular ascorbate (∼human lung fluid concentration) also decreased uptake of chromate anions and their cytotoxicity. In comparison to chromate anions, uptake and cytotoxicity of respirable particles of moderately soluble CaCrO4 and SrCrO4 were much less sensitive to suppression by extracellular ascorbate, especially during early exposure times and in primary bronchial cells. In the absence of extracellular ascorbate, chromate anions and CaCrO4/SrCrO4 particles produced overall similar levels of DNA double-stranded breaks, with less soluble particles exhibiting a slower rate of breakage. Our results indicate that a gradual extracellular dissolution and a rapid internalization of calcium chromate and strontium chromate particles makes them resistant to detoxification outside the cells, which is extremely effective for chromate anions in the rat lung fluid. The detoxification potential of the human lung fluid is significant but much lower and insufficient to provide a threshold-type dose dependence for soluble chromates.


Asunto(s)
Cromatos/toxicidad , Pulmón/efectos de los fármacos , Animales , Ácido Ascórbico/química , Compuestos de Calcio/química , Compuestos de Calcio/metabolismo , Compuestos de Calcio/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cromatos/química , Cromatos/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , Glutatión/química , Humanos , Pulmón/patología , Oxidación-Reducción , Ratas , Estroncio/química , Estroncio/metabolismo , Estroncio/toxicidad
15.
Toxicol Appl Pharmacol ; 286(2): 135-41, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25817892

RESUMEN

Formaldehyde (FA) is a human carcinogen with numerous sources of environmental and occupational exposures. This reactive aldehyde is also produced endogenously during metabolism of drugs and other processes. DNA-protein crosslinks (DPCs) are considered to be the main genotoxic lesions for FA. Accumulating evidence suggests that DPC repair in high eukaryotes involves proteolysis of crosslinked proteins. Here, we examined a role of the main cellular proteolytic machinery proteasomes in toxic responses of human lung cells to low FA doses. We found that transient inhibition of proteasome activity increased cytotoxicity and diminished clonogenic viability of FA-treated cells. Proteasome inactivation exacerbated suppressive effects of FA on DNA replication and increased the levels of the genotoxic stress marker γ-H2AX in normal human cells. A transient loss of proteasome activity in FA-exposed cells also caused delayed perturbations of cell cycle, which included G2 arrest and a depletion of S-phase populations at FA doses that had no effects in control cells. Proteasome activity diminished p53-Ser15 phosphorylation but was important for FA-induced CHK1 phosphorylation, which is a biochemical marker of DPC proteolysis in replicating cells. Unlike FA, proteasome inhibition had no effect on cell survival and CHK1 phosphorylation by the non-DPC replication stressor hydroxyurea. Overall, we obtained evidence for the importance of proteasomes in protection of human cells against biologically relevant doses of FA. Biochemically, our findings indicate the involvement of proteasomes in proteolytic repair of DPC, which removes replication blockage by these highly bulky lesions.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Formaldehído/toxicidad , Fase G2/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Quinasas/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Daño del ADN/efectos de los fármacos , Humanos , Hidroxiurea/farmacología , Fosforilación , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos
16.
Chem Res Toxicol ; 27(5): 843-51, 2014 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-24646070

RESUMEN

Cr(VI) genotoxicity is caused by products of its reductive metabolism inside the cells. Reactive oxygen species (ROS) and Cr(V,IV) intermediates are potential sources of oxidative damage by Cr(VI). Here, we investigated seven fluorescent probes for the detection of ROS and non-ROS oxidants in Cr(VI) reactions with its main reducers. We found that Cr(V)-skipping metabolism of Cr(VI) by ascorbate in vitro gave no responses with all tested dyes, indicating nonreactivity of Cr(IV) and absence of ROS. Cr(VI) reduction with glutathione (GSH) or Cys strongly enhanced the fluorescence of dichlorofluorescein (DCF) and dihydrorhodamine 123 (DHR123) but produced minimal fluorescence with dihydroethidium and no increases with aminophenylfluorescein and CellRox Green, Orange, and Red. Several tests showed that Cr(VI)-thiol reactions lacked ROS and that Cr(V) caused oxidation of DCF and DHR123. DCF reacted only with free Cr(V), whereas DHR123 detected both the free Cr(V) and Cr(V)-GSH complex. We estimated that Cr(VI)-GSH reactions generated approximately 75% Cr(V)-GSH and 25% free Cr(V), whereas Cys reactions appeared to produce only free Cr(V). DHR123 measurements in H460 cells showed that reduction of Cr(VI) was complete within 20 min postexposure, but it lasted at least 1 h without GSH. Cells with restored ascorbate levels exhibited no DCF or DHR123 oxidation by Cr(VI). Overall, our results demonstrated that Cr(VI) metabolism with its biological reducers lacked ROS and that DHR123 and DCF responses were indicators of total and free Cr(V), respectively. CellRox dyes, dihydroethidium and aminophenylfluorescein, are insensitive to Cr(V,IV) and can be used for monitoring ROS during coexposure to Cr(VI) and oxidants.


Asunto(s)
Cromatos/metabolismo , Colorantes Fluorescentes/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ácido Ascórbico/análisis , Ácido Ascórbico/metabolismo , Línea Celular , Cromatos/análisis , Cromatos/toxicidad , Células Epiteliales/metabolismo , Fluoresceínas/análisis , Fluoresceínas/metabolismo , Colorantes Fluorescentes/análisis , Glutatión/análisis , Glutatión/metabolismo , Humanos , Oxidación-Reducción , Especies Reactivas de Oxígeno/análisis , Rodaminas/análisis , Rodaminas/metabolismo
17.
Toxicol Appl Pharmacol ; 269(3): 233-9, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23566959

RESUMEN

Hypoxia mimic nickel(II) is a human respiratory carcinogen with a suspected epigenetic mode of action. We examined whether Ni(II) elicits a toxicologically significant activation of the tumor suppressor p53, which is typically associated with genotoxic responses. We found that treatments of H460 human lung epithelial cells with NiCl2 caused activating phosphorylation at p53-Ser15, accumulation of p53 protein and depletion of its inhibitor MDM4 (HDMX). Confirming the activation of p53, its knockdown suppressed the ability of Ni(II) to upregulate MDM2 and p21 (CDKN1A). Unlike DNA damage, induction of GADD45A by Ni(II) was p53-independent. Ni(II) also increased p53-Ser15 phosphorylation and p21 expression in normal human lung fibroblasts. Although Ni(II)-induced stabilization of HIF-1α occurred earlier, it had no effect on p53 accumulation and Ser15 phosphorylation. Ni(II)-treated H460 cells showed no evidence of necrosis and their apoptosis and clonogenic death were suppressed by p53 knockdown. The apoptotic role of p53 involved a transcription-dependent program triggering the initiator caspase 9 and its downstream executioner caspase 3. Two most prominently upregulated proapoptotic genes by Ni(II) were PUMA and NOXA but only PUMA induction required p53. Knockdown of p53 also led to derepression of antiapoptotic MCL1 in Ni(II)-treated cells. Overall, our results indicate that p53 plays a major role in apoptotic death of human lung cells by Ni(II). Chronic exposure to Ni(II) may promote selection of resistant cells with inactivated p53, providing an explanation for the origin of p53 mutations by this epigenetic carcinogen.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasa 3/efectos de los fármacos , Caspasa 9/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Pulmón/efectos de los fármacos , Níquel/toxicidad , Proteína p53 Supresora de Tumor/efectos de los fármacos , Western Blotting , Caspasa 3/fisiología , Caspasa 9/fisiología , Línea Celular , Daño del ADN/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Pulmón/citología
18.
Chem Res Toxicol ; 26(10): 1591-8, 2013 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-24059640

RESUMEN

Hexavalent chromium is a human carcinogen activated primarily by direct reduction with cellular ascorbate and to a lesser extent, by glutathione. Cr(III), the final product of Cr(VI) reduction, forms six bonds allowing intermolecular cross-linking. In this work, we investigated the ability of Cr(VI) to cause interstrand DNA cross-links (ICLs) whose formation mechanisms and presence in human cells are currently uncertain. We found that in vitro reduction of Cr(VI) with glutathione showed a sublinear production of ICLs, the yield of which was less than 1% of total Cr-DNA adducts at the optimal conditions. Formation of ICLs in fast ascorbate-Cr(VI) reactions occurred during a short reduction interval and displayed a linear dose dependence with the average yield of 1.3% of total adducts. In vitro production of ICLs was strongly suppressed by increasing buffer molarity, indicating inhibitory effects of ligand-Cr(III) binding on the formation of cross-linking species. The presence of ICLs in human cells was assessed from the impact of ICL repair deficiencies on Cr(VI) responses. We found that ascorbate-restored FANCD2-null and isogenic FANCD2-complemented cells showed similar cell cycle inhibition and toxicity by Cr(VI). XPA-null cells are defective in the repair of Cr-DNA monoadducts, but stable knockdowns of ERCC1 or XPF in these cells with extended time for the completion of cross-linking reactions did not produce any sensitization to Cr(VI). Our results together with chemical and steric considerations of Cr(III) reactivity suggest that ICL generation by chromate is probably an in vitro phenomenon occurring at conditions permitting the formation of Cr(III) oligomers.


Asunto(s)
Cromo/química , ADN/química , Ácido Ascórbico/química , Ácido Ascórbico/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Cromo/metabolismo , Cromo/toxicidad , ADN/metabolismo , Aductos de ADN/química , Aductos de ADN/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endonucleasas/antagonistas & inhibidores , Endonucleasas/genética , Endonucleasas/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Glutatión/química , Glutatión/metabolismo , Humanos , Oxidación-Reducción , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína de la Xerodermia Pigmentosa del Grupo A/genética , Proteína de la Xerodermia Pigmentosa del Grupo A/metabolismo
19.
Adv Pharmacol ; 96: 25-46, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36858775

RESUMEN

Hexavalent chromium is a firmly established human carcinogen with documented exposures in many professional groups. Environmental exposure to Cr(VI) is also a significant public health concern. Cr(VI) exists in aqueous solutions as chromate anion that is unreactive with DNA and requires reductive activation inside the cells to produce genotoxic and mutagenic effects. Reduction of Cr(VI) in cells is nonenzymatic and in vivo principally driven by ascorbate with a secondary contribution from nonprotein thiols glutathione and cysteine. In addition to its much faster rate of reduction, ascorbate-driven metabolism avoids the formation of Cr(V) which is the first intermediate in Cr(VI) reduction by thiols. The end-product of Cr(VI) reduction is Cr(III) which forms several types of Cr-DNA adducts that are collectively responsible for all mutagenic and genotoxic effects in Cr(VI) reactions with ascorbate and thiols. Some Cr(V) forms can react with H2O2 to produce DNA-oxidizing peroxo species although this genotoxic pathway is suppressed in cells with physiological levels of ascorbate. Chemical reactions of Cr(VI) with ascorbate or thiols lack directly DNA-oxidizing metabolites. The formation of oxidative DNA breaks in early studies of these reactions was caused by iron contamination. Production of Cr(III)-DNA adducts in cells showed linear dose-dependence irrespective of the predominant reduction pathway and their processing by mismatch repair generated more toxic secondary genetic lesions in euchromatin. Overall, Cr(III)-DNA adduction is the dominant pathway for the formation of genotoxic and mutagenic DNA damage by carcinogenic Cr(VI).


Asunto(s)
Carcinógenos , Aductos de ADN , Humanos , Peróxido de Hidrógeno , Daño del ADN , Ácido Ascórbico
20.
Mutagenesis ; 27(4): 437-43, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22241526

RESUMEN

Cultured human cells are invaluable biological models for mechanistic studies of genotoxic chemicals and drugs. Continuing replacement of animals in toxicity testing will further increase the importance of in vitro cell systems, which should accurately reproduce key in vivo characteristics of toxicants such as their profiles of metabolites and DNA lesions. In this work, we examined how a common severe deficiency of cultured cells in ascorbate (Asc) impacts the formation of oxidative DNA damage by hexavalent chromium (chromate). Cr(VI) is reductively activated inside the cells by both Asc and small thiols but with different rates and spectra of intermediates and DNA adducts. We found that Cr(VI) exposure of H460 human lung epithelial cells in standard culture (<0.01 mM cellular Asc) induced biologically significant amounts of oxidative DNA damage. Inhibition of oxidative damage repair in these cells by stable XRCC1 knockdown strongly enhanced cytotoxic effects of Cr(VI) and led to depletion of cells from G(1) and accumulation in S and G(2) phases. However, restoration of physiological levels of Asc (≈ 1 mM) completely eliminated Cr(VI) hypersensitivity of XRCC1 knockdown. The induction of chromosomal breaks assayed by the micronucleus test in Asc-restored H460, primary human lung fibroblasts, and CHO cells was also unaffected by the XRCC1 status. Centromere-negative (clastogenic) micronuclei accounted for 80-90% of all Cr(VI)-induced micronuclei. Consistent with the micronuclei results, Asc-restored cells also showed no increase in the levels of poly(ADP-ribose), which is a biochemical marker of single-stranded breaks. Asc had no effect on cytotoxicity of O(6)-methylguanine, a lesion produced by direct DNA alkylation. Overall, our results indicate that the presence of physiological levels of Asc strongly suppresses pro-oxidant pathways in Cr(VI) metabolism and that the use of standard cell cultures creates a distorted profile of its genotoxic properties.


Asunto(s)
Ácido Ascórbico/farmacología , Carcinógenos Ambientales/efectos adversos , Ciclo Celular/efectos de los fármacos , Cromo/efectos adversos , Daño del ADN/efectos de los fármacos , Pulmón/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Antioxidantes/farmacología , Western Blotting , Muerte Celular/efectos de los fármacos , Células Cultivadas , Cromatografía Líquida de Alta Presión , Cricetinae , Cricetulus , Aductos de ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Glutatión/metabolismo , Guanina/análogos & derivados , Guanina/farmacología , Humanos , Pulmón/citología , Pruebas de Micronúcleos , Oxidación-Reducción , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA