Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 732
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Chem Res Toxicol ; 35(1): 99-110, 2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-34969250

RESUMEN

Endogenous DNA lesions frequently occur due to internal effects such as oxidative stress, inflammation, endogenous alkylation, and epigenetic modifications. However, exposure to chemical toxicants from the environment, diet, or drugs can also induce significant endogenous DNA damage. The quantification of endogenous DNA damage effect markers might reflect the actual DNA damage level of chemical toxicants. Herein, we report a liquid chromatography-triple quadrupole tandem mass spectrometry (LC-QqQ MS/MS) method for simultaneous determination of eight representative endogenous DNA damage biomarkers, including five endogenous DNA damage effect markers (oxidative damage, 8-oxo-dG; lipid peroxidation, εdA and N2-Et-dG; inflammation, 5-Cl-dC; and endogenous alkylation, O6-Me-dG), and three epigenetic modifications (5-m-dC, 5-hm-dC, and N6-Me-dA). The method validation was performed, and the linear range was 0.05 pg to 2 ng (on-column), the limit of detection was 0.02 pg (on-column), and the precision, accuracy, matrix effect, and recovery were all between 85 and 115%. We then applied this method to evaluate endogenous DNA damage to human embryonic lung fibroblast cells exposed to five nitrogen mustards [NMs, i.e., HN1, HN2, HN3, chlorambucil (CB), and cyclophosphamide (CTX)], where curcumin exposure was used as a control due to its inability to induce the formation of endogenous DNA adducts. The amounts of eight DNA adducts in the low-, middle-, and high-concentration exposure groups of five NMs were almost all significantly different from those in the blank group (P < 0.05). We obtained a positive correlation between the contents of eight DNA damage biomarkers and the inhibition dose of five NMs, except for N2-Et-dG and 5-Cl-dC. Via further principal component analysis and partial least squares discriminant analysis, we clustered all NMs into three units with different cytotoxicity levels, that is, HN2 and HN1 (highly toxic), HN3 and CB (moderately toxic), and CTX (less toxic). Moreover, for the same concentration of HN1/2/3 exposure groups, as the cytotoxicity increased according to the order of HN3 < HN1 < HN2, the contents of 8-oxo-dG, 5-m-dC, 5-hm-dC, and N6-Me-dA increased, whereas the content of O6-Me-dG decreased. Therefore, the contents of these DNA damage effect markers were somewhat related to the cytotoxicity and concentration of NMs. We hope that this method will provide an alternative evaluation approach for the toxicological effects of NMs and the safety of the medication.


Asunto(s)
Compuestos de Mostaza Nitrogenada/farmacología , Biomarcadores/análisis , Cromatografía Líquida de Alta Presión , Aductos de ADN/efectos de los fármacos , Daño del ADN , Humanos , Estructura Molecular , Compuestos de Mostaza Nitrogenada/análisis , Espectrometría de Masas en Tándem
2.
Arch Toxicol ; 96(2): 511-524, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34783865

RESUMEN

Arylamine N-acetyltransferase 1 (NAT1) plays a pivotal role in the metabolism of carcinogens and is a drug target for cancer prevention and/or treatment. A protein-ligand virtual screening of 2 million chemicals was ranked for predicted binding affinity towards the inhibition of human NAT1. Sixty of the five hundred top-ranked compounds were tested experimentally for inhibition of recombinant human NAT1 and N-acetyltransferase 2 (NAT2). The most promising compound 9,10-dihydro-9,10-dioxo-1,2-anthracenediyl diethyl ester (compound 10) was found to be a potent and selective NAT1 inhibitor with an in vitro IC50 of 0.75 µM. Two structural analogs of this compound were selective but less potent for inhibition of NAT1 whereas a third structural analog 1,2-dihydroxyanthraquinone (a compound 10 hydrolysis product also known as Alizarin) showed comparable potency and efficacy for human NAT1 inhibition. Compound 10 inhibited N-acetylation of the arylamine carcinogen 4-aminobiphenyl (ABP) both in vitro and in DNA repair-deficient Chinese hamster ovary (CHO) cells in situ stably expressing human NAT1 and CYP1A1. Compound 10 and Alizarin effectively inhibited NAT1 in cryopreserved human hepatocytes whereas inhibition of NAT2 was not observed. Compound 10 caused concentration-dependent reductions in DNA adduct formation and DNA double-strand breaks following metabolism of aromatic amine carcinogens beta-naphthylamine and/or ABP in CHO cells. Compound 10 inhibited proliferation and invasion in human breast cancer cells and showed selectivity towards tumorigenic versus non-tumorigenic cells. In conclusion, our study identifies potent, selective, and efficacious inhibitors of human NAT1. Alizarin's ability to inhibit NAT1 could reduce breast cancer metastasis particularly to bone.


Asunto(s)
Arilamina N-Acetiltransferasa/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Isoenzimas/antagonistas & inhibidores , Animales , Antraquinonas/farmacología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Células CHO , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Simulación por Computador , Cricetinae , Cricetulus , Aductos de ADN/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Hepatocitos/enzimología , Humanos , Concentración 50 Inhibidora
3.
Int J Mol Sci ; 23(3)2022 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-35163790

RESUMEN

Aurora A kinase (Aurora A) is a serine/threonine kinase regulating control of multiple events during cell-cycle progression. Playing roles in promoting proliferation and inhibiting cell death in cancer cells leads Aurora A to become a target for cancer therapy. It is overexpressed and associated with a poor prognosis in ovarian cancer. Improving cisplatin therapy outcomes remains an important issue for advanced-stage ovarian cancer treatment, and Aurora A inhibitors may improve it. In the present study, we identified natural compounds with higher docking scores than the known Aurora A ligand through structure-based virtual screening, including the natural compound fangchinoline, which has been associated with anticancer activities but not yet investigated in ovarian cancer. The binding and inhibition of Aurora A by fangchinoline were verified using cellular thermal shift and enzyme activity assays. Fangchinoline reduced viability and proliferation in ovarian cancer cell lines. Combination fangchinoline and cisplatin treatment enhanced cisplatin-DNA adduct levels, and the combination index revealed synergistic effects on cell viability. An in vivo study showed that fangchinoline significantly enhanced cisplatin therapeutic effects in OVCAR-3 ovarian cancer-bearing mice. Fangchinoline may inhibit tumor growth and enhance cisplatin therapy in ovarian cancer. This study reveals a novel Aurora A inhibitor, fangchinoline, as a potentially viable adjuvant for ovarian cancer therapy.


Asunto(s)
Aurora Quinasa A/metabolismo , Bencilisoquinolinas/administración & dosificación , Cisplatino/administración & dosificación , Aductos de ADN/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Animales , Aurora Quinasa A/química , Bencilisoquinolinas/química , Bencilisoquinolinas/farmacología , Línea Celular Tumoral , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Modelos Moleculares , Simulación del Acoplamiento Molecular , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Conformación Proteica , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Carcinogenesis ; 42(5): 694-704, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33693566

RESUMEN

1,3-Butadiene (BD) is a known human carcinogen used in the synthetic polymer industry and also found in cigarette smoke, automobile exhaust and wood burning smoke. BD is metabolically activated by cytochrome P450 monooxygenases (CYP) 2E1 and 2A6 to 3,4-epoxy-1-butene (EB), which can be detoxified by GST-catalyzed glutathione conjugation or hydrolysis. We have previously observed ethnic differences in urinary levels of EB-mercapturic acids in white, Japanese American and Native Hawaiian smokers. In the present study, similar analyses were extended to urinary BD-DNA adducts. BD-induced N7-(1-hydroxy-3-buten-2-yl) guanine (EB-GII) adducts were quantified in urine samples obtained from smokers and non-smokers belonging to three racial/ethnic groups: white, Japanese American and Native Hawaiian. After adjusting for sex, age, nicotine equivalents, body mass index and batch, we found that Japanese American smokers excreted significantly higher amounts of urinary EB-GII than whites [1.45 (95% confidence interval: 1.12-1.87) versus 0.68 (95% confidence interval: 0.52-0.85) fmol/ml urine, P = 4 × 10-5]. Levels of urinary EB-GII in Native Hawaiian smokers were not different from those in whites [0.67 (95% confidence interval: 0.51-0.84) fmol/ml urine, P = 0.938]. There were no racial/ethnic differences in urinary EB-GII adduct levels in non-smokers. Racial/ethnic differences in urinary EB-GII adduct levels in smokers could not be explained by GSTT1 gene deletion or CYP2A6 enzymatic activity. Urinary EB-GII adduct levels in smokers were significantly associated with concentrations of BD metabolite dihyroxybutyl mercapturic acid. Overall, our results reveal that urinary EB-GII adducts in smokers differ across racial/ethnic groups. Future studies are required to understand genetic and epigenetic factors that may be responsible for these differences.


Asunto(s)
Butadienos/toxicidad , Citocromo P-450 CYP2A6/genética , Citocromo P-450 CYP2E1/genética , Aductos de ADN/efectos de los fármacos , Acetilcisteína/orina , Adulto , Anciano , Asiático/genética , Carcinógenos/metabolismo , Carcinógenos/toxicidad , Aductos de ADN/genética , Aductos de ADN/orina , Compuestos Epoxi/efectos adversos , Compuestos Epoxi/orina , Etnicidad/genética , Femenino , Glutatión Transferasa/genética , Humanos , Masculino , Persona de Mediana Edad , Nativos de Hawái y Otras Islas del Pacífico/genética , Humo/efectos adversos , Fumadores , Espectrometría de Masa por Ionización de Electrospray , Productos de Tabaco/efectos adversos , Emisiones de Vehículos/toxicidad , Población Blanca/genética
5.
Bioorg Med Chem Lett ; 35: 127813, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33486050

RESUMEN

Current techniques for the identification of DNA adduct-inducing and DNA interstrand crosslinking agents include electrophoretic crosslinking assays, electrophoretic gel shift assays, DNA and RNA stop assays, mass spectrometry-based methods and 32P-post-labelling. While these assays provide considerable insight into the site and stability of the interaction, they are relatively expensive, time-consuming and sometimes rely on the use of radioactively-labelled components, and thus are ill-suited to screening large numbers of compounds. A novel medium throughput assay was developed to overcome these limitations and was based on the attachment of a biotin-tagged double stranded (ds) oligonucleotide to Corning DNA-Bind plates. We aimed to detect anthracycline and anthracenedione DNA adducts which form by initial non-covalent intercalation with duplex DNA, and subsequent covalent adduct formation which is mediated by formaldehyde. Following drug treatment, DNA samples were subjected to a denaturation step, washing and then measurement by fluorescence to detect remaining drug-DNA species using streptavidin-europium. This dissociation-enhanced lanthanide fluorescent immunoassay (DELFIA) is a time-resolved fluorescence intensity assay where the fluorescence signal arises only from stabilised drug-DNA complexes. We applied this new methodology to the identification of anthracycline-like compounds with the ability to functionally crosslink double-strand oligonucleotides. The entire procedure can be performed by robotics, requiring low volumes of compounds and reagents, thereby reducing costs and enabling multiple compounds to be assessed on a single microtitre plate.


Asunto(s)
Automatización , Reactivos de Enlaces Cruzados/farmacología , Aductos de ADN/efectos de los fármacos , Desarrollo de Medicamentos , Reactivos de Enlaces Cruzados/síntesis química , Reactivos de Enlaces Cruzados/química , Relación Dosis-Respuesta a Droga , Estructura Molecular , Relación Estructura-Actividad
6.
Acta Pharmacol Sin ; 42(12): 2094-2105, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33686245

RESUMEN

Aristolochic acid I (AAI) is a well-known nephrotoxic carcinogen, which is currently reported to be also associated with hepatocellular carcinoma (HCC). Whether AAI is a direct hepatocarcinogen remains controversial. In this study we investigated the association between AAI exposure and HCC in adult rats using a sensitive rat liver bioassay with several cofactors. Formation of glutathione S-transferase placental form-positive (GST-P+) foci was used as the marker for preneoplastic lesions/clonal expansion. We first conducted a medium-term (8 weeks) study to investigate whether AAI had any tumor-initiating or -promoting activity. Then a long-term (52 weeks) study was conducted to determine whether AAI can directly induce HCC. We showed that oral administration of single dose of AAI (20, 50, or 100 mg/kg) in combination with partial hepatectomy (PH) to stimulate liver proliferation did not induce typical GST-P+ foci in liver. In the 8-week study, only high dose of AAI (10 mg · kg-1 · d-1, 5 days a week for 6 weeks) in combination with PH significantly increased the number and area of GST-P+ foci initiated by diethylnitrosamine (DEN) in liver. Similarly, only high dose of AAI (10 mg· kg-1· d-1, 5 days a week for 52 weeks) in combination with PH significantly increased the number and area of hepatic GST-P+ foci in the 52-week study. No any nodules or HCC were observed in liver of any AAI-treated groups. In contrast, long-term administration of AAI (0.1, 1, 10 mg· kg-1· d-1) time- and dose-dependently caused death due to the occurrence of cancers in the forestomach, intestine, and/or kidney. Besides, AAI-DNA adducts accumulated in the forestomach, kidney, and liver in a time- and dose-dependent manner. Taken together, AAI promotes clonal expansion only in the high-dose group but did not induce any nodules or HCC in liver of adult rats till their deaths caused by cancers developed in the forestomach, intestine, and/or kidney. Findings from our animal studies will pave the way for further large-scale epidemiological investigation of the associations between AA and HCC.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Carcinógenos/toxicidad , Carcinoma Hepatocelular/etiología , Hepatocitos/metabolismo , Neoplasias Hepáticas/etiología , Mutágenos/toxicidad , Animales , Carcinogénesis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Aductos de ADN/efectos de los fármacos , Gutatión-S-Transferasa pi/metabolismo , Neoplasias Intestinales/inducido químicamente , Intestinos/patología , Riñón/patología , Neoplasias Renales/inducido químicamente , Hígado/metabolismo , Hígado/patología , Masculino , Ratas Sprague-Dawley , Estómago/patología , Neoplasias Gástricas/inducido químicamente
7.
Arch Toxicol ; 95(8): 2839-2850, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34223934

RESUMEN

Aristolochic acids (AAs) are a family of natural compounds with AA I and AA II being known carcinogens, whose bioactivation causes DNA adducts formation. However, other congeners have rarely been investigated. This study aimed to investigate genotoxicity of AA IVa, which differs from AA I by a hydroxyl group, abundant in Aristolochiaceae plants. AA IVa reacted with 2'-deoxyadenosine (dA) and 2'-deoxyguanosine (dG) to form three dA and five dG adducts as identified by high-resolution mass spectrometry, among which two dA and three dG adducts were detected in reactions of AA IVa with calf thymus DNA (CT DNA). However, no DNA adducts were detected in the kidney, liver, and forestomach of orally dosed mice at 40 mg/kg/day for 2 days, and bone marrow micronucleus assay also yielded negative results. Pharmacokinetic analyses of metabolites in plasma indicated that AA IVa was mainly O-demethylated to produce a metabolite with two hydroxyl groups, probably facilitating its excretion. Meanwhile, no reduced metabolites were detected. The competitive reaction of AA I and AA IVa with CT DNA, with adducts levels varying with pH of reaction revealed that AA IVa was significantly less reactive than AA I, probably by hydroxyl deprotonation of AA IVa, which was explained by theoretical calculations for reaction barriers, energy levels of the molecular orbits, and charges at the reaction sites. In brief, although it could form DNA adducts in vitro, AA IVa was non-genotoxic in vivo, which was attributed to its low reactivity and biotransformation into an easily excreted metabolite rather than bioactivation.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Aductos de ADN/efectos de los fármacos , Daño del ADN/efectos de los fármacos , ADN/efectos de los fármacos , Animales , Ácidos Aristolóquicos/administración & dosificación , Ácidos Aristolóquicos/química , Carcinógenos/administración & dosificación , Carcinógenos/química , Carcinógenos/toxicidad , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Endogámicos C57BL , Pruebas de Micronúcleos , Pruebas de Mutagenicidad
8.
Carcinogenesis ; 41(2): 194-202, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-31074772

RESUMEN

Alcohol consumption is the key risk factor for the development of esophageal squamous cell carcinoma (ESCC), and acetaldehyde, a metabolite of alcohol, is an alcohol-derived major carcinogen that causes DNA damage. Aldehyde dehydrogenase2 (ALDH2) is an enzyme that detoxifies acetaldehyde, and its activity is reduced by ALDH2 gene polymorphism. Reduction in ALDH2 activity increases blood, salivary and breath acetaldehyde levels after alcohol intake, and it is deeply associated with the development of ESCC. Heavy alcohol consumption in individuals with ALDH2 gene polymorphism significantly elevates the risk of ESCC; however, effective prevention has not been established yet. In this study, we investigated the protective effects of Alda-1, a small molecule ALDH2 activator, on alcohol-mediated esophageal DNA damage. Here, we generated novel genetically engineered knock-in mice that express the human ALDH2*1 (wild-type allele) or ALDH2*2 gene (mutant allele). Those mice were crossed, and human ALDH2*1/*1, ALDH2*1/*2 and ALDH2*2/*2 knock-in mice were established. They were given 10% ethanol for 7 days in the presence or absence of Alda-1, and we measured the levels of esophageal DNA damage, represented by DNA adduct (N2-ethylidene-2'-deoxyguanosine). Alda-1 significantly increased hepatic ALDH2 activity both in human ALDH2*1/*2 and/or ALDH2*2/*2 knock-in mice and reduced esophageal DNA damage levels after alcohol drinking. Conversely, cyanamide, an ALDH2-inhibitor, significantly exacerbated esophageal DNA adduct level in C57BL/6N mice induced by alcohol drinking. These results indicate the protective effects of ALDH2 activation by Alda-1 on esophageal DNA damage levels in individuals with ALDH2 gene polymorphism, providing a new insight into acetaldehyde-mediated esophageal carcinogenesis and prevention.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Benzamidas/administración & dosificación , Benzodioxoles/administración & dosificación , Carcinogénesis/efectos de los fármacos , Neoplasias Esofágicas/prevención & control , Carcinoma de Células Escamosas de Esófago/prevención & control , Acetaldehído/metabolismo , Acetaldehído/toxicidad , Aldehído Deshidrogenasa Mitocondrial/antagonistas & inhibidores , Aldehído Deshidrogenasa Mitocondrial/genética , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/genética , Cianamida/administración & dosificación , Aductos de ADN/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Mucosa Esofágica/efectos de los fármacos , Mucosa Esofágica/patología , Neoplasias Esofágicas/etiología , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/etiología , Carcinoma de Células Escamosas de Esófago/patología , Etanol/metabolismo , Etanol/toxicidad , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones Transgénicos , Mutación , Neoplasias Experimentales/etiología , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Polimorfismo Genético , Factores de Riesgo
9.
Toxicol Appl Pharmacol ; 398: 115026, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32353386

RESUMEN

The presence of flavors is one of the commonly cited reasons for use of e-cigarettes by youth; however, the potential harms from inhaling these chemicals and byproducts have not been extensively studied. One mechanism of interest is DNA adduct formation, which may lead to carcinogenesis. We identified two chemical classes of flavors found in tobacco products and byproducts, alkenylbenzenes and aldehydes, documented to form DNA adducts. Using in silico toxicology approaches, we identified structural analogs to these chemicals without DNA adduct information. We conducted a structural similarity analysis and also generated in silico model predictions of these chemicals for genotoxicity, mutagenicity, carcinogenicity, and skin sensitization. The empirical and in silico data were compared, and we identified strengths and limitations of these models. Good concordance (80-100%) was observed between DNA adduct formation and models predicting mammalian mutagenicity (mouse lymphoma sassy L5178Y) and skin sensitization for both chemical classes. On the other hand, different prediction profiles were observed for the two chemical classes for the modeled endpoints, unscheduled DNA synthesis and bacterial mutagenicity. These results are likely due to the different mode of action between the two chemical classes, as aldehydes are direct acting agents, while alkenylbenzenes require bioactivation to form electrophilic intermediates, which form DNA adducts. The results of this study suggest that an in silico prediction for the mouse lymphoma assay L5178Y, may serve as a surrogate endpoint to help predict DNA adduct formation for chemicals found in tobacco products such as flavors and byproducts.


Asunto(s)
Aductos de ADN/efectos de los fármacos , Aromatizantes/farmacología , Nicotiana/efectos adversos , Productos de Tabaco/efectos adversos , Animales , Simulación por Computador , Sistemas Electrónicos de Liberación de Nicotina , Ratones , Mutagénesis/efectos de los fármacos , Mutágenos/efectos adversos
10.
Chem Res Toxicol ; 33(4): 852-854, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32223224

RESUMEN

Mass spectrometry-based DNA adductomics is an emerging approach for the human biomonitoring of hazardous chemicals. A mass spectral database of DNA adducts will be created for the scientific community to investigate the associations between chemical exposures, DNA damage, and disease risk.


Asunto(s)
Aductos de ADN/efectos de los fármacos , Bases de Datos de Compuestos Químicos , Contaminantes Ambientales/farmacología , Compuestos Orgánicos/farmacología , Daño del ADN , Contaminantes Ambientales/química , Humanos , Espectrometría de Masas , Compuestos Orgánicos/química
11.
Chem Res Toxicol ; 33(4): 988-998, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32174110

RESUMEN

Nitrogen mustards (NM) are an important class of chemotherapeutic drugs used in the treatment of malignant tumors. The accepted mechanism of action of NM is through the alkylation of DNA bases. NM-adducts block DNA replication in cancer cells by forming cytotoxic DNA interstrand cross-links. We previously characterized several adducts formed by reaction of bis(2-chloroethyl)ethylamine (NM) with calf thymus (CT) DNA and the MDA-MB-231 mammary tumor cell line. The monoalkylated N7-guanine (NM-G) adduct and its cross-link (G-NM-G) were major lesions. The cationic NM-G undergoes a secondary reaction through depurination to form an apurinic (AP) site or reacts with hydroxide to yield the stable ring-opened N5-substituted formamidopyrimidine (NM-Fapy-G) adduct. Both of these lesions are mutagenic and may contribute to secondary tumor development, a major clinical limitation of NM chemotherapy. We established a kinetic model with NM-treated female mice and measured the rates of formation and removal of NM-DNA adducts and AP sites. We employed liquid chromatography-mass spectrometry (LC-MS) to measure NM-G, G-NM-G, and NM-Fapy-G adducts in liver, lung, and spleen over 168 h. NM-G reached a maximum level within 6 h in all organs and then rapidly declined. The G-NM-G cross-link and NM-FapyG were more persistent with half-lives over three-times longer than NM-G. We quantified AP site lesions in the liver and showed that NM treatment increased AP site levels by 3.7-fold over the basal levels at 6 h. The kinetics of AP site repair closely followed the rate of removal of NM-G; however, AP sites remained 1.3-fold above basal levels 168 h post-treatment with NM. Our data provide new insights into NM-induced DNA damage and biological processing in vivo. The quantitative measurement of the spectrum of NM adducts and AP sites can serve as biomarkers in the design and assessment of the efficacy of novel chemotherapeutic regimens.


Asunto(s)
Aductos de ADN/química , Aductos de ADN/efectos de los fármacos , Mecloretamina/química , Mecloretamina/toxicidad , Animales , Femenino , Cinética , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Distribución Tisular
12.
Chem Res Toxicol ; 33(2): 584-593, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31885260

RESUMEN

Nitroaromatic compounds represent a major class of industrial chemicals that are also found in nature. Polycyclic derivatives are regarded as potent mutagens and carcinogens following bioactivation to produce nitrenium electrophiles that covalently modify DNA to afford N-linked C8-2'-deoxyguanosine (C8-dG) lesions that can induce frameshift mutations, especially in CpG repeat sequences. In contrast, their monocyclic counterparts typically exhibit weak mutagenicity or a lack thereof, despite also undergoing bioactivation to afford N-linked C8-dG adducts. Recently, it has been reported that cyano substitution can greatly increase the mutagenicity of nitroaniline derivatives that are components of azo dyes. The basis of this "cyano effect" may be rooted in the formation of a novel polycyclic adduct arising from initial formation of the N-linked C8-dG adduct followed by a cyclization process involving N7 of dG and the ortho-CN group of the attached C8-aryl moiety to generate a quinazolinimine ring as part of a fused tetracyclic C8,N7-dG adduct structure. The present work structurally characterizes this novel cyclic adduct using a combination of optical spectroscopies, NMR analysis, density functional theory (DFT) calculations, and molecular dynamics (MD) simulations. Our data indicate that this highly fluorescent cyclic adduct adopts the promutagenic syn conformation and can stabilize the slipped mutagenic intermediate (SMI) within the CpG repeat of the NarI sequence, which is a hotspot for frameshift mutagenesis mediated by polycyclic N-linked C8-dG adducts. In contrast, the open para-CN (4-aminobenzontrile-derived) N-linked C8-dG adduct is less likely to disrupt the canonical B-form. Together, our results provide a rationale for the potent mutagenicity of cyano-substituted nitroaniline derivatives recently reported in frameshift-sensitive tester strains.


Asunto(s)
Compuestos de Anilina/química , Compuestos de Anilina/toxicidad , Aductos de ADN/química , Aductos de ADN/efectos de los fármacos , Desoxiguanosina/análogos & derivados , Mutación del Sistema de Lectura/efectos de los fármacos , Aductos de ADN/genética , Teoría Funcional de la Densidad , Desoxiguanosina/química , Desoxiguanosina/genética , Conformación Molecular/efectos de los fármacos , Simulación de Dinámica Molecular
13.
Chem Res Toxicol ; 33(7): 1980-1988, 2020 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-32476407

RESUMEN

Our early studies demonstrated an impressive chemopreventive efficacy of dihydromethysticin (DHM), unique in kava, against tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung tumorigenesis in A/J mice in which DHM was supplemented in the diet. The current work was carried out to validate the efficacy, optimize the dosing schedule, and further elucidate the mechanisms using oral bolus dosing of DHM. The results demonstrated a dose-dependent chemopreventive efficacy of DHM (orally administered 1 h before each of the two NNK intraperitoneal injections, 1 week apart) against NNK-induced lung adenoma formation. Temporally, DHM at 0.8 mg per dose (∼32 mg per kg body weight) exhibited 100% lung adenoma inhibition when given 3 and 8 h before each NNK injection and attained >93% inhibition when dosed at either 1 or 16 h before each NNK injection. The simultaneous treatment (0 h) or 40 h pretreatment (-40 h) decreased lung adenoma burden by 49.8% and 52.1%, respectively. However, post-NNK administration of DHM (1-8 h after each NNK injection) was ineffective against lung tumor formation. In short-term experiments for mechanistic exploration, DHM treatment reduced the formation of NNK-induced O6-methylguanine (O6-mG, a carcinogenic DNA adduct in A/J mice) in the target lung tissue and increased the urinary excretion of NNK detoxification metabolites as judged by the ratio of urinary NNAL-O-gluc to free NNAL, generally in synchrony with the tumor prevention efficacy outcomes in the dose scheduling time-course experiment. Overall, these results suggest DHM as a potential chemopreventive agent against lung tumorigenesis in smokers, with O6-mG and NNAL detoxification as possible surrogate biomarkers.


Asunto(s)
Adenoma/prevención & control , Anticarcinógenos/administración & dosificación , Butanonas/toxicidad , Carcinógenos/toxicidad , Neoplasias Pulmonares/prevención & control , Nitrosaminas/toxicidad , Pironas/administración & dosificación , Administración Oral , Animales , Carcinogénesis/efectos de los fármacos , Aductos de ADN/efectos de los fármacos , Suplementos Dietéticos , Femenino , Hígado/efectos de los fármacos , Hígado/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones Endogámicos , Nicotiana
14.
Chem Res Toxicol ; 33(10): 2668-2674, 2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-32894672

RESUMEN

Inflammation is an immune response to protect against various types of infections. When unchecked, acute inflammation can be life-threatening, as seen with the current coronavirus pandemic. Strong oxidants, such as peroxynitrite produced by immune cells, are major mediators of the inflammation-associated pathogenesis. Cellular thiols play important roles in mitigating inflammation-associated macromolecular damage including DNA. Herein, we have demonstrated a role of glutathione (GSH) and other thiols in neutralizing the effect of peroxynitrite-mediated DNA damage through stable GSH-DNA adduct formation. Our observation supports the use of thiol supplements as a potential therapeutic strategy against severe COVID-19 cases and a Phase II (NCT04374461) open-label clinical trial launched in early May 2020 by the Memorial Sloan Kettering Cancer Center.


Asunto(s)
Aductos de ADN/efectos de los fármacos , ADN/efectos de los fármacos , Glutatión/farmacología , Inflamación/fisiopatología , Ácido Peroxinitroso/efectos adversos , Enfermedad Aguda , Animales , Betacoronavirus , COVID-19 , Bovinos , Infecciones por Coronavirus/tratamiento farmacológico , ADN/química , Aductos de ADN/química , Daño del ADN , Glutatión/química , Células HEK293 , Humanos , Mutágenos/química , Mutágenos/farmacología , Pandemias , Ácido Peroxinitroso/química , Neumonía Viral/tratamiento farmacológico , SARS-CoV-2 , Salmonella typhimurium/genética
15.
Mutagenesis ; 35(5): 437-443, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33511997

RESUMEN

1,3-Dichloropropene (1,3-D; CAS No. 542-75-6) is a soil fumigant used for the control of nematodes in agriculture. There is an extensive database on the genotoxicity of 1,3-D and many of the published studies are confounded by the presence of mutagenic stabilisers in the test substance. Mixed results were obtained in the in vitro assays, often due to the purity of the 1,3-D sample tested. In order to get further clarity, the mutagenic potential of 1,3-D was investigated in vivo in the transgenic Big Blue rodent models. Inhalation exposure of 150 ppm 1,3-D (×2.5 tumourigenic dose) to transgenic male B6C3F1 mice did not induce lacI mutations in either the lung (tumour target tissue) or liver. Similarly, dietary administration of 1,3-D up to 50 mg/kg/day to transgenic male Fischer 344 rats did not increase the cII mutant frequency in either the liver (tumour target) or kidney. These results, along with other available in vivo data, including the absence of DNA adducts and clastogenic/aneugenic potential, support the conclusion that 1,3-D is efficiently detoxified in vivo and, as such, does not pose a mutagenic hazard or risk.


Asunto(s)
Compuestos Alílicos/farmacología , Hidrocarburos Clorados/farmacología , Mutagénesis/efectos de los fármacos , Mutágenos/farmacología , Plaguicidas/farmacología , Compuestos Alílicos/toxicidad , Animales , Aductos de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hidrocarburos Clorados/toxicidad , Represoras Lac/genética , Ratones , Ratones Transgénicos , Pruebas de Mutagenicidad , Mutágenos/toxicidad , Mutación/efectos de los fármacos , Plaguicidas/efectos adversos , Ratas , Ratas Endogámicas F344
16.
J Child Psychol Psychiatry ; 61(11): 1253-1265, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31907931

RESUMEN

BACKGROUND: Risk for childhood psychopathology is complex and multifactorial, implicating direct and interacting effects of familial and environmental factors. The role of environmental neurotoxicants in psychiatric risk is of growing concern, including polycyclic aromatic hydrocarbons (PAH), common in air pollution. Prenatal PAH exposure is linked to adverse physical, behavioral, and cognitive outcomes as well as increasing psychiatric risk. It is unclear whether environmental exposures, like PAH, magnify the effects of exposure to early life stress (ELS), a critical risk factor for psychopathology. The current work aimed to test potential interactions between prenatal PAH exposure and psychosocial/socioeconomic stress on psychiatric symptoms in school-age children. METHODS: Data were from the Columbia Center for Children's Environmental Health Mothers and Newborns longitudinal birth cohort study. Prenatal PAH exposure was ascertained though air monitoring during pregnancy and maternal PAH-DNA adducts at delivery. Mothers reported on ELS (child age 5) and on child psychiatric symptoms across childhood (child age 5, 7, 9, and 11) using the Child Behavior Checklist (CBCL). RESULTS: Significant prenatal airborne PAH × ELS interactions (FDR-corrected) predicted CBCL Attention (ß = 0.22, t(307) = 3.47, p < .001, pfdr  = .003) and Thought Problems T-scores (ß = 0.21, t(307) = 3.29, p = .001, pfdr  = .004) at age 11 (n = 319). Relative to those with lower exposure, children with higher prenatal PAH exposure exhibited stronger positive associations between ELS and CBCL Attention and Thought Problem T-scores. This interaction was also significant examining convergent ADHD measures (Conners, DuPaul) and examining maternal PAH-DNA adducts (ß = 0.29, t(261) = 2.48, p = .01; n = 273). A three-way interaction with assessment wave indicated that the PAH × ELS interaction on Attention Problems was stronger later in development (ß = 0.03, t(1,601) = 2.19, p = .03; n = 477). CONCLUSIONS: Prenatal exposure to PAH, a common neurotoxicant in air pollution, may magnify or sustain the effects of early life psychosocial/socioeconomic stress on psychiatric outcomes later in child development. This work highlights the critical role of air pollution exposure on child mental health.


Asunto(s)
Experiencias Adversas de la Infancia/psicología , Atención/efectos de los fármacos , Salud Mental , Hidrocarburos Policíclicos Aromáticos/envenenamiento , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Psicología Infantil , Estrés Psicológico/psicología , Niño , Preescolar , Aductos de ADN/efectos de los fármacos , Femenino , Humanos , Estudios Longitudinales , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/psicología , Psicopatología
17.
Proc Natl Acad Sci U S A ; 114(16): 4207-4212, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28373545

RESUMEN

Global distribution of hepatocellular carcinomas (HCCs) is dominated by its incidence in developing countries, accounting for >700,000 estimated deaths per year, with dietary exposures to aflatoxin (AFB1) and subsequent DNA adduct formation being a significant driver. Genetic variants that increase individual susceptibility to AFB1-induced HCCs are poorly understood. Herein, it is shown that the DNA base excision repair (BER) enzyme, DNA glycosylase NEIL1, efficiently recognizes and excises the highly mutagenic imidazole ring-opened AFB1-deoxyguanosine adduct (AFB1-Fapy-dG). Consistent with this in vitro result, newborn mice injected with AFB1 show significant increases in the levels of AFB1-Fapy-dG in Neil1-/- vs. wild-type liver DNA. Further, Neil1-/- mice are highly susceptible to AFB1-induced HCCs relative to WT controls, with both the frequency and average size of hepatocellular carcinomas being elevated in Neil1-/- The magnitude of this effect in Neil1-/- mice is greater than that previously measured in Xeroderma pigmentosum complementation group A (XPA) mice that are deficient in nucleotide excision repair (NER). Given that several human polymorphic variants of NEIL1 are catalytically inactive for their DNA glycosylase activity, these deficiencies may increase susceptibility to AFB1-associated HCCs.


Asunto(s)
Aflatoxinas/toxicidad , Carcinoma Hepatocelular/prevención & control , Aductos de ADN/efectos de los fármacos , ADN Glicosilasas/fisiología , Neoplasias Hepáticas Experimentales/prevención & control , Sustancias Protectoras/farmacología , Animales , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Femenino , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Venenos/toxicidad
18.
Mol Pharmacol ; 96(4): 475-484, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31399497

RESUMEN

Topoisomerase II (TOP2) poisons are effective cytotoxic anticancer agents that stabilize the normally transient TOP2-DNA covalent complexes formed during the enzyme reaction cycle. These drugs include etoposide, mitoxantrone, and the anthracyclines doxorubicin and epirubicin. Anthracyclines also exert cell-killing activity via TOP2-independent mechanisms, including DNA adduct formation, redox activity, and lipid peroxidation. Here, we show that anthracyclines and another intercalating TOP2 poison, mitoxantrone, stabilize TOP2-DNA covalent complexes less efficiently than etoposide, and at higher concentrations they suppress the formation of TOP2-DNA covalent complexes, thus behaving as TOP2 poisons at low concentration and inhibitors at high concentration. We used induced pluripotent stem cell (iPSC)-derived human cardiomyocytes as a model to study anthracycline-induced damage in cardiac cells. Using immunofluorescence, our study is the first to demonstrate the presence of topoisomerase IIß (TOP2B) as the only TOP2 isoform in iPSC-derived cardiomyocytes. In these cells, etoposide robustly induced TOP2B covalent complexes, but we could not detect doxorubicin-induced TOP2-DNA complexes, and doxorubicin suppressed etoposide-induced TOP2-DNA complexes. In vitro, etoposide-stabilized DNA cleavage was attenuated by doxorubicin, epirubicin, or mitoxantrone. Clinical use of anthracyclines is associated with cardiotoxicity. The observations in this study have potentially important clinical consequences regarding the effectiveness of anticancer treatment regimens when TOP2-targeting drugs are used in combination. These observations suggest that inhibition of TOP2B activity, rather than DNA damage resulting from TOP2 poisoning, may play a role in doxorubicin cardiotoxicity. SIGNIFICANCE STATEMENT: We show that anthracyclines and mitoxantrone act as topoisomerase II (TOP2) poisons at low concentration but attenuate TOP2 activity at higher concentration, both in cells and in in vitro cleavage experiments. Inhibition of type II topoisomerases suppresses the action of other drugs that poison TOP2. Thus, combinations containing anthracyclines or mitoxantrone and etoposide may reduce the activity of etoposide as a TOP2 poison and thus reduce the efficacy of drug combinations.


Asunto(s)
Antraciclinas/farmacología , Aductos de ADN/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , Etopósido/farmacología , Mitoxantrona/farmacología , Cardiotoxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Aductos de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/efectos adversos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células K562 , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Inhibidores de Topoisomerasa II/farmacología
19.
Toxicol Appl Pharmacol ; 384: 114783, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31669812

RESUMEN

In a cohort of approximately 200 Bangladeshi men, equally divided into smokers and non-smokers and equally divided by exposure to high and low levels of drinking water arsenic, we examined ex vivo a series of immune markers and immune function tests in peripheral blood mononuclear cells (PBMC). These immune parameters included PBMC cell surface markers (CSM) for B, T, monocytes, and NK cells, activated T and B cell markers, cytokine production in vitro, and analysis of CD4 subsets (Th1, Th2, Treg, and Th17 cells). We found that the effects of cigarette smoke were quite different than those associated with arsenic or polycyclic aromatic hydrocarbon (PAH)-DNA adducts. Cigarette smoking was associated with a significant increase in the number of PAH-DNA adducts as well as an increase in urinary levels of 1-hydropxypyrene (1-OHP). After correcting for arsenic exposure and PAH-DNA adducts, we found that cigarette smoking was associated with an increase in the percentage of CD19+ B cells, as well as the percentage of activated B cells (CD19+, HLA-DRbright cells) found in PBMC. These findings demonstrate activation of the immune system during chronic exposure to cigarette smoke, which is a known risk factor for autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/epidemiología , Linfocitos B/inmunología , Fumar Cigarrillos/efectos adversos , Aductos de ADN/efectos de los fármacos , Antígenos HLA-DR/inmunología , Adolescente , Adulto , Anciano , Enfermedades Autoinmunes/inmunología , Linfocitos B/efectos de los fármacos , Bangladesh , Fumar Cigarrillos/sangre , Fumar Cigarrillos/inmunología , Estudios de Cohortes , Aductos de ADN/inmunología , Humanos , Masculino , Persona de Mediana Edad , Hidrocarburos Policíclicos Aromáticos/toxicidad , Factores de Riesgo , Humo/efectos adversos , Nicotiana/efectos adversos , Adulto Joven
20.
Nat Chem Biol ; 13(9): 1002-1008, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28759018

RESUMEN

Yatakemycin (YTM) is an extraordinarily toxic DNA alkylating agent with potent antimicrobial and antitumor properties and is the most recent addition to the CC-1065 and duocarmycin family of natural products. Though bulky DNA lesions the size of those produced by YTM are normally removed from the genome by the nucleotide-excision repair (NER) pathway, YTM adducts are also a substrate for the bacterial DNA glycosylases AlkD and YtkR2, unexpectedly implicating base-excision repair (BER) in their elimination. The reason for the extreme toxicity of these lesions and the molecular basis for the way they are eliminated by BER have been unclear. Here, we describe the structural and biochemical properties of YTM adducts that are responsible for their toxicity, and define the mechanism by which they are excised by AlkD. These findings delineate an alternative strategy for repair of bulky DNA damage and establish the cellular utility of this pathway relative to that of NER.


Asunto(s)
Productos Biológicos/toxicidad , Aductos de ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Indoles/toxicidad , Pirroles/toxicidad , Productos Biológicos/farmacología , Daño del ADN , Farmacorresistencia Bacteriana , Duocarmicinas , Indoles/farmacología , Estructura Molecular , Pirroles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA