Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 764
Filter
1.
Nature ; 629(8013): 910-918, 2024 May.
Article in English | MEDLINE | ID: mdl-38693263

ABSTRACT

International differences in the incidence of many cancer types indicate the existence of carcinogen exposures that have not yet been identified by conventional epidemiology make a substantial contribution to cancer burden1. In clear cell renal cell carcinoma, obesity, hypertension and tobacco smoking are risk factors, but they do not explain the geographical variation in its incidence2. Underlying causes can be inferred by sequencing the genomes of cancers from populations with different incidence rates and detecting differences in patterns of somatic mutations. Here we sequenced 962 clear cell renal cell carcinomas from 11 countries with varying incidence. The somatic mutation profiles differed between countries. In Romania, Serbia and Thailand, mutational signatures characteristic of aristolochic acid compounds were present in most cases, but these were rare elsewhere. In Japan, a mutational signature of unknown cause was found in more than 70% of cases but in less than 2% elsewhere. A further mutational signature of unknown cause was ubiquitous but exhibited higher mutation loads in countries with higher incidence rates of kidney cancer. Known signatures of tobacco smoking correlated with tobacco consumption, but no signature was associated with obesity or hypertension, suggesting that non-mutagenic mechanisms of action underlie these risk factors. The results of this study indicate the existence of multiple, geographically variable, mutagenic exposures that potentially affect tens of millions of people and illustrate the opportunities for new insights into cancer causation through large-scale global cancer genomics.


Subject(s)
Carcinoma, Renal Cell , Environmental Exposure , Geography , Kidney Neoplasms , Mutagens , Mutation , Female , Humans , Male , Aristolochic Acids/adverse effects , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/epidemiology , Carcinoma, Renal Cell/chemically induced , Environmental Exposure/adverse effects , Environmental Exposure/analysis , Genome, Human/genetics , Genomics , Hypertension/epidemiology , Incidence , Japan/epidemiology , Kidney Neoplasms/genetics , Kidney Neoplasms/epidemiology , Kidney Neoplasms/chemically induced , Mutagens/adverse effects , Obesity/epidemiology , Risk Factors , Romania/epidemiology , Serbia/epidemiology , Thailand/epidemiology , Tobacco Smoking/adverse effects , Tobacco Smoking/genetics
2.
Nutrients ; 16(7)2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38613117

ABSTRACT

The International Agency for Research on Cancer has classified the consumption of heat-processed meat as a direct human carcinogen and the consumption of red meat as a probable carcinogen. Mutagenic and carcinogenic compounds present in meat dishes include, among others, polycyclic aromatic hydrocarbons (PAHs) and heterocyclic aromatic amines (HAAs). These compounds can cause the development of gastrointestinal cancer. Oral cancer is one of the world's research priorities due to the ever-increasing incidence rate. However, the effect of diet on oral cancer is still a poorly recognized issue. The aim of this study was to assess the relationship between the risk of oral cancer and dietary ingredients with a particular emphasis on red meat and thermally processed meat. This study was conducted among patients with oral cancer in 2022 and 2023. The shortened standardized Food Frequency Questionnaire (FFQ) and a multivariate regression statistical analysis were used. The high consumption of red meat in general and thermally processed meat, especially smoked, fried, roasted and boiled, increases the risk of oral cavity cancer. Limiting the consumption of meat products and modifying the methods of preparing meat dishes may reduce exposure to carcinogenic compounds from the diet and thus reduce the risk of developing oral cancer.


Subject(s)
Mouth Neoplasms , Mutagens , Humans , Mutagens/adverse effects , Carcinogens/toxicity , Mouth Neoplasms/epidemiology , Mouth Neoplasms/etiology , Carcinogenesis , Meat/adverse effects
3.
Int J Mol Sci ; 23(7)2022 Mar 22.
Article in English | MEDLINE | ID: mdl-35408779

ABSTRACT

Transcript signatures are a promising approach to identify and classify genotoxic and non-genotoxic compounds and are of interest as biomarkers or for future regulatory application. Not much data, however, is yet available about the concordance of transcriptional responses in different cell types or tissues. Here, we analyzed transcriptomic responses to selected genotoxic food contaminants in the human p53-competent lymphoblastoid cell line TK6 using RNA sequencing. Responses to treatment with five genotoxins, as well as with four non-genotoxic liver toxicants, were compared with previously published gene expression data from the human liver cell model HepaRG. A significant overlap of the transcriptomic changes upon genotoxic stress was detectable in TK6 cells, whereas the comparison with the HepaRG model revealed considerable differences, which was confirmed by bioinformatic data mining for cellular upstream regulators or pathways. Taken together, the study presents a transcriptomic signature for genotoxin exposure in the human TK6 blood cell model. The data demonstrate that responses in different cell models have considerable variations. Detection of a transcriptomic genotoxin signature in blood cells indicates that gene expression analyses of blood samples might be a valuable approach to also estimate responses to toxic exposure in target organs such as the liver.


Subject(s)
DNA Damage , Mutagens , Blood Cells , Humans , Liver , Mutagens/adverse effects , Transcriptome
4.
Int J Mol Sci ; 23(4)2022 Feb 14.
Article in English | MEDLINE | ID: mdl-35216217

ABSTRACT

The use of in silico toxicity prediction methods plays an important role in the selection of lead compounds and in ADMET studies since in vitro and in vivo methods are often limited by ethics, time, budget and other resources. In this context, we present our new web tool VenomPred, a user-friendly platform for evaluating the potential mutagenic, hepatotoxic, carcinogenic and estrogenic effects of small molecules. VenomPred platform employs several in-house Machine Learning (ML) models developed with datasets derived from VEGA QSAR, a software that includes a comprehensive collection of different toxicity models and has been used as a reference for building and evaluating our ML models. The results showed that our models achieved equal or better performance than those obtained with the reference models included in VEGA QSAR. In order to improve the predictive performance of our platform, we adopted a consensus approach combining the results of different ML models, which was able to predict chemical toxicity better than the single models. This improved method was thus implemented in the VenomPred platform, a freely accessible webserver that takes the SMILES (Simplified Molecular-Input Line-Entry System) strings of the compounds as input and sends the prediction results providing a probability score about their potential toxicity.


Subject(s)
Carcinogens/toxicity , Drug-Related Side Effects and Adverse Reactions/prevention & control , Mutagens/adverse effects , Small Molecule Libraries/adverse effects , Small Molecule Libraries/chemistry , Computer Simulation , Machine Learning , Mutagenesis/drug effects , Quantitative Structure-Activity Relationship , Software
5.
Carcinogenesis ; 43(1): 52-59, 2022 02 11.
Article in English | MEDLINE | ID: mdl-34546339

ABSTRACT

Nucleotide excision repair (NER) is a repair mechanism that removes DNA lesions induced by UV radiation, environmental mutagens and carcinogens. There exists sufficient evidence against acetaldehyde suggesting it to cause a variety of DNA lesions and be carcinogenic to humans. Previously, we found that acetaldehyde induces reversible intra-strand GG crosslinks in DNA similar to those induced by cis-diammineplatinum(II) that is subsequently repaired by NER. In this study, we analysed the repairability by NER mechanism and the mutagenesis of acetaldehyde. In an in vitro reaction setup with NER-proficient and NER-deficient xeroderma pigmentosum group A (XPA) cell extracts, NER reactions were observed in the presence of XPA recombinant proteins in acetaldehyde-treated plasmids. Using an in vivo assay with living XPA cells and XPA-correcting XPA cells, the repair reactions were also observed. Additionally, it was observed that DNA polymerase eta inserted dATP opposite guanine in acetaldehyde-treated oligonucleotides, suggesting that acetaldehyde-induced GG-to-TT transversions. These findings show that acetaldehyde induces NER repairable mutagenic DNA lesions.


Subject(s)
Acetaldehyde/adverse effects , DNA Repair/drug effects , DNA/genetics , Mutagenesis/drug effects , DNA Damage/drug effects , DNA Repair/genetics , Fibroblasts/drug effects , Humans , Mutagenesis/genetics , Mutagens/adverse effects , Transfection/methods , Ultraviolet Rays , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum Group A Protein/genetics
6.
Genes (Basel) ; 12(12)2021 11 25.
Article in English | MEDLINE | ID: mdl-34946837

ABSTRACT

Heterocyclic amines (HCAs) are a set of food contaminants that may exert a cytotoxic effect on human peripheral blood mononuclear cells (PBMC). However, the genetic mechanism underlying the cytotoxicity of HCAs on PBMC has not been investigated. In the study, bioinformatic analysis on gene dataset GSE19078 was performed. The results of weighted correlation network analysis and linear models for microarray and RNA-seq data analysis showed that four gene modules were relevant to 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) exposure while one gene module was correlated with 2-amino-3-methyl-3H-imidazo[4,5f]quinoline (IQ) exposure. Gene functional analysis showed that the five modules were annotated mainly with mRNA transcriptional regulation, mitochondrial function, RNA catabolic process, protein targeting, and immune function. Five genes, MIER1, NDUFA4, MLL3, CD53 and CSF3 were recognized as the feature genes for each hub gene network of the corresponding gene module, and the expression of feature genes was observed with a significant difference between the PhIP/IQ samples and the other samples. Our results provide novel genes and promising mechanisms for exploration on the genetic mechanism of HCAs on PBMC.


Subject(s)
Amines/adverse effects , ELAV-Like Protein 2/genetics , Cells, Cultured , Computational Biology/methods , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/genetics , Humans , Imidazoles/adverse effects , Leukocytes, Mononuclear/drug effects , Mutagens/adverse effects , Pyridines/adverse effects , Quinolines/adverse effects , RNA, Messenger/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
7.
Food Chem Toxicol ; 158: 112579, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34597720

ABSTRACT

Paraxanthine or 1,7-dimethylxanthine is a natural dietary component and the main metabolite of caffeine in humans. A battery of toxicological studies was conducted in accordance with international guidelines to investigate mutagenicity, genotoxicity and acute and repeated-dose oral toxicity in rats of synthetic paraxanthine (ENFINITY™, Ingenious Ingredients, L.P., >99% purity). There was no evidence of mutagenicity in a bacterial reverse mutation as well as in an in vitro mammalian chromosomal aberration test. There was no evidence of genotoxicity in an in vivo mammalian erythrocyte micronucleus test as well as in an in vitro mammalian cell gene mutation test. An acute oral toxicity test resulted in a LD50 value of 1601 mg/kg bw/d. Paraxanthine did not cause mortality or toxic effects in a subacute 28-day repeated-dose oral toxicity study at daily doses of 75, 150, or 300 mg/kg bw/d (each group n = 10 per sex), administered by gavage. Paraxanthine also did not cause mortality or toxic effects in a subchronic 90-day repeated-dose oral toxicity study at daily doses of 75, 150, or 300 mg/kg bw/d (each group n = 10 per sex), administered by gavage. The no observed adverse effect level (NOAEL) determined from the 90-day study was greater than or equal to 300 mg/kg bw/d, the highest dose tested, for both male and female Wistar rats.


Subject(s)
Mutagenicity Tests , Mutagens , Theophylline , Administration, Oral , Animals , Drug-Related Side Effects and Adverse Reactions , Female , Male , Mutagens/administration & dosage , Mutagens/adverse effects , Mutagens/toxicity , Rats , Rats, Wistar , Theophylline/administration & dosage , Theophylline/adverse effects , Theophylline/toxicity
8.
Int J Mol Sci ; 22(14)2021 Jul 12.
Article in English | MEDLINE | ID: mdl-34299079

ABSTRACT

In order to tackle the study of DNA repair pathways, the physical and chemical agents creating DNA damage, the genotoxins, are frequently employed. Despite their utility, their effects are rarely restricted to DNA, and therefore simultaneously harm other cell biomolecules. Methyl methanesulfonate (MMS) is an alkylating agent that acts on DNA by preferentially methylating guanine and adenine bases. It is broadly used both in basic genome stability research and as a model for mechanistic studies to understand how alkylating agents work, such as those used in chemotherapy. Nevertheless, MMS exerts additional actions, such as oxidation and acetylation of proteins. In this work, we introduce the important notion that MMS also triggers a lipid stress that stems from and affects the inner nuclear membrane. The inner nuclear membrane plays an essential role in virtually all genome stability maintenance pathways. Thus, we want to raise awareness that the relative contribution of lipid and genotoxic stresses when using MMS may be difficult to dissect and will matter in the conclusions drawn from those studies.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , DNA Damage , Lipids/analysis , Methyl Methanesulfonate/adverse effects , Mutagens/adverse effects , Nuclear Envelope/pathology , Retinal Pigment Epithelium/pathology , DNA Repair , Hep G2 Cells , Humans , Nuclear Envelope/drug effects , Retinal Pigment Epithelium/drug effects
9.
Environ Mol Mutagen ; 62(8): 458-470, 2021 10.
Article in English | MEDLINE | ID: mdl-34331495

ABSTRACT

Urinary mutagenicity reflects systemic exposure to complex mixtures of genotoxic/carcinogenic agents and is linked to tumor development. Coal combustion emissions (CCE) and diesel engine exhaust (DEE) are associated with cancers of the lung and other sites, but their influence on urinary mutagenicity is unclear. We investigated associations between exposure to CCE or DEE and urinary mutagenicity. In two separate cross-sectional studies of nonsmokers, organic extracts of urine were evaluated for mutagenicity levels using strain YG1041 in the Salmonella (Ames) mutagenicity assay. First, we compared levels among 10 female bituminous (smoky) coal users from Laibin, Xuanwei, China, and 10 female anthracite (smokeless) coal users. We estimated exposure-response relationships using indoor air concentrations of two carcinogens in CCE relevant to lung cancer, 5-methylchrysene (5MC), and benzo[a]pyrene (B[a]P). Second, we compared levels among 20 highly exposed male diesel factory workers and 15 unexposed male controls; we evaluated exposure-response relationships using elemental carbon (EC) as a DEE-surrogate. Age-adjusted linear regression was used to estimate associations. Laibin smoky coal users had significantly higher average urinary mutagenicity levels compared to smokeless coal users (28.4 ± 14.0 SD vs. 0.9 ± 2.8 SD rev/ml-eq, p = 2 × 10-5 ) and a significant exposure-response relationship with 5MC (p = 7 × 10-4 ). DEE-exposed workers had significantly higher urinary mutagenicity levels compared to unexposed controls (13.0 ± 10.1 SD vs. 5.6 ± 4.4 SD rev/ml-eq, p = .02) and a significant exposure-response relationship with EC (p-trend = 2 × 10-3 ). Exposure to CCE and DEE is associated with urinary mutagenicity, suggesting systemic exposure to mutagens, potentially contributing to cancer risk and development at various sites.


Subject(s)
Air Pollutants, Occupational/urine , Coal/adverse effects , Mutagens/analysis , Occupational Diseases/epidemiology , Occupational Exposure/adverse effects , Smoking/urine , Vehicle Emissions/analysis , Air Pollutants, Occupational/adverse effects , China/epidemiology , Coal/analysis , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Mutagens/adverse effects , Occupational Diseases/diagnosis , Occupational Diseases/genetics , Occupational Diseases/urine , Occupational Exposure/analysis , Smoking/adverse effects
10.
Environ Mol Mutagen ; 62(5): 306-318, 2021 06.
Article in English | MEDLINE | ID: mdl-34050964

ABSTRACT

The organotypic human air-liquid-interface (ALI) airway tissue model has been used as an in vitro cell culture system for evaluating the toxicity of inhaled substances. ALI airway cultures are highly differentiated, which has made it challenging to evaluate genetic toxicology endpoints. In the current study, we assayed DNA damage with the high-throughput CometChip assay and quantified mutagenesis with Duplex Sequencing, an error-corrected next-generation sequencing method capable of detecting a single mutation per 107 base pairs. Fully differentiated human ALI airway cultures were treated from the basolateral side with 6.25 to 100 µg/mL ethyl methanesulfonate (EMS) over a period of 28 days. CometChip assays were conducted after 3 and 28 days of treatment, and Duplex Sequencing after 28 days of treatment. Treating the airway cultures with EMS resulted in time- and concentration-dependent increases in DNA damage and a concentration-dependent increase in mutant frequency. The mutations observed in the EMS-treated cultures were predominantly C → T transitions and exhibited a unique trinucleotide signature relative to the negative control. Measurement of physiological endpoints indicated that the EMS treatments had no effect on anti-p63-positive basal cell frequency, but produced concentration-responsive increases in cytotoxicity and perturbations in cell morphology, along with concentration-responsive decreases in culture viability, goblet cell and anti-Ki67-positive proliferating cell frequency, cilia beating frequency, and mucin secretion. The results indicate that a unified 28-day study can be used to measure several important safety endpoints in physiologically relevant human in vitro ALI airway cultures, including DNA damage, mutagenicity, and tissue-specific general toxicity.


Subject(s)
DNA Damage , Epithelial Cells/pathology , Ethyl Methanesulfonate/adverse effects , Mutagenesis , Mutagenicity Tests/methods , Mutation , Respiratory System/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Mutagens/adverse effects , Respiratory System/drug effects , Respiratory System/metabolism
11.
J Infect Dis ; 224(3): 415-419, 2021 08 02.
Article in English | MEDLINE | ID: mdl-33961695

ABSTRACT

Mutagenic ribonucleosides can act as broad-based antiviral agents. They are metabolized to the active ribonucleoside triphosphate form and concentrate in genomes of RNA viruses during viral replication. ß-d-N4-hydroxycytidine (NHC, initial metabolite of molnupiravir) is >100-fold more active than ribavirin or favipiravir against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with antiviral activity correlated to the level of mutagenesis in virion RNA. However, NHC also displays host mutational activity in an animal cell culture assay, consistent with RNA and DNA precursors sharing a common intermediate of a ribonucleoside diphosphate. These results indicate highly active mutagenic ribonucleosides may hold risk for the host.


Subject(s)
Antiviral Agents/pharmacology , Cytidine/analogs & derivatives , Mutagens/pharmacology , SARS-CoV-2/drug effects , Animals , Antiviral Agents/adverse effects , CHO Cells/drug effects , Cells, Cultured , Cricetulus , Cytidine/adverse effects , Cytidine/pharmacology , Dose-Response Relationship, Drug , Mutagenesis/drug effects , Mutagens/adverse effects , SARS-CoV-2/genetics , Virus Replication/drug effects
12.
Leukemia ; 35(8): 2145-2150, 2021 08.
Article in English | MEDLINE | ID: mdl-34012133

ABSTRACT

The introduction of whole genome and exome sequencing partnered with advanced bioinformatic pipelines has allowed the comprehensive characterization of mutational processes (i.e., mutational signatures) in individual cancer patients. Studies focusing on multiple myeloma have defined several mutational processes, including a recently identified mutational signature (called "SBS-MM1") directly caused by exposure to high-dose melphalan (i.e., autologous stem cell transplant). High-dose melphalan exposure increases both the overall and nonsynonymous mutational burden detected between diagnosis and relapse by ~10-20%. Nevertheless, most of these mutations are acquired within the heterochromatin and late-replicating regions, rarely involving key myeloma driver genes. In this review, we summarize key studies that made this discovery possible, and we discuss potential clinical implications.


Subject(s)
Melphalan/adverse effects , Multiple Myeloma/pathology , Mutagenesis , Mutagens/adverse effects , Mutation , Neoplasm Proteins/genetics , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics
13.
Toxicol In Vitro ; 74: 105173, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33848590

ABSTRACT

Cigarette smoking increases health risks, such as respiratory diseases and heart diseases. Despite the decline in smoking rates in some countries, millions of adults still choose to smoke cigarettes. The use of next-generation nicotine delivery devices, such as tobacco heating products (THPs), may become a potentially safer alternative to smoking. Here, we report on the development of an electrically heated THP, coded as THP COO, with three different flavored tobacco sticks. The purpose of the study was to measure the levels of a list of harmful and potentially harmful constituents (HPHCs) in the total particulate matter (TPM) generated and to conduct a set of toxicological assessments of THP COO as compared with 3R4F reference cigarette. For all 55 HPHCs identified, the levels generated by the THP tobacco sticks were significantly lower in comparison to those in 3R4F TPM. The rate of reduction of HPHCs was between 68.6% and 99.9% under Health Canada Intense (HCI) smoking regimen. Human lung cancer cells (NCI-H292) exposed to 3R4F TPM showed dose-dependent responses for most of the 15 in vitro toxicity endpoints, whereas those exposed to comparable doses of THP COO TPMs did not. Therefore, exclusive use of the THP COO products may reduce the exposure of those tested HPHCs and thus potentially reduce health risk of smoking.


Subject(s)
Electronic Nicotine Delivery Systems , Hazardous Substances/adverse effects , Hot Temperature , Smoke/adverse effects , Tobacco Products , Aerosols , Animals , CHO Cells , Cell Cycle/drug effects , Cell Line , Cell Survival/drug effects , Cricetulus , Cytokines/metabolism , Hazardous Substances/analysis , High-Throughput Screening Assays , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Micronuclei, Chromosome-Defective/chemically induced , Mutagenicity Tests , Mutagens/adverse effects , Mutagens/analysis , Reactive Oxygen Species/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Smoke/analysis
14.
Mutagenesis ; 36(1): 1-17, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33544138

ABSTRACT

In vitro test batteries have become the standard approach to determine the genotoxic potential of substances of interest across industry sectors. While useful for hazard identification, standard in vitro genotoxicity assays in 2D cell cultures have limited capability to predict in vivo outcomes and may trigger unnecessary follow-up animal studies or the loss of promising substances where animal tests are prohibited or not desired. To address this problem, a team of regulatory, academia and industry scientists was established to develop and validate 3D in vitro human skin-based genotoxicity assays for use in testing substances with primarily topical exposure. Validation of the reconstructed human skin micronucleus (RSMN) assay in MatTek Epi-200™ skin models involved testing 43 coded chemicals selected by independent experts, in four US/European laboratories. The results were analysed by an independent statistician according to predefined criteria. The RSMN assay showed a reproducibly low background micronucleus frequency and exhibited sufficient capacity to metabolise pro-mutagens. The overall RSMN accuracy when compared to in vivo genotoxicity outcomes was 80%, with a sensitivity of 75% and a specificity of 84%, and the between- and within-laboratory reproducibility was 77 and 84%, respectively. A protocol involving a 72-h exposure showed increased sensitivity in detecting true positive chemicals compared to a 48-h exposure. An analysis of a test strategy using the RSMN assay as a follow-up test for substances positive in standard in vitro clastogenicity/aneugenicity assays and a reconstructed skin Comet assay for substances with positive results in standard gene mutation assays results in a sensitivity of 89%. Based on these results, the RSMN assay is considered sufficiently validated to establish it as a 'tier 2' assay for dermally exposed compounds and was recently accepted into the OECD's test guideline development program.


Subject(s)
Animal Testing Alternatives/methods , Biological Assay/methods , DNA Damage , Laboratories/standards , Micronucleus Tests/methods , Mutagens/adverse effects , Skin/pathology , False Positive Reactions , Humans , In Vitro Techniques , Skin/drug effects , Skin/metabolism
15.
Article in English | MEDLINE | ID: mdl-33551104

ABSTRACT

Acetaldehyde (AA) has been classified as a probable human carcinogen by the International Agency for Research on Cancer (IARC, WHO) and by the US Environmental Protection Agency due to its ability to cause tumours following inhalation or alcohol consumption in animals. Humans are constantly exposed to AA through inhalation from the environment through cigarette smoke, vehicle fumes and industrial emissions as well as by persistent alcohol ingestion. Individuals with deficiencies in the enzymes that are involved in the metabolism of AA are more susceptible to its toxicity and constitute a vulnerable human population. Studies have shown that AA induces DNA damage and cytogenetic abnormalities. A study was undertaken to elucidate the clastogenic effects induced by AA and any preceding DNA damage that occurs in normal human lung fibroblasts as this will further validate the detrimental effects of inhalation exposure to AA. AA exposure induced DNA damage, involving DNA double strand breaks, which could possibly occur at the telomeric regions as well, resulting in a clastogenic effect and subsequent genomic instability, which contributed to the cell cycle arrest. The clastogenic effect induced by AA in human lung fibroblasts was evidenced by micronuclei induction and chromosomal aberrations, including those at the telomeric regions. Co-localisation between the DNA double strand breaks and telomeric regions was observed, suggesting possible induction of DNA double strand breaks due to AA exposure at the telomeric regions as a new mechanism beyond the clastogenic effect of AA. From the cell cycle profile following AA exposure, a G2/M phase arrest and a decrease in cell viability were also detected. Therefore, these effects due to AA exposure via inhalation may have implications in the development of carcinogenesis in humans.


Subject(s)
Acetaldehyde/adverse effects , Chromosome Aberrations/chemically induced , DNA Damage , Fibroblasts/pathology , Genomic Instability , Lung/pathology , Mutagens/adverse effects , Cell Survival , Fibroblasts/drug effects , Fibroblasts/metabolism , G2 Phase , Humans , Lung/drug effects , Lung/metabolism , Telomere
16.
BMC Cancer ; 21(1): 172, 2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33596864

ABSTRACT

BACKGROUND: Colibactin is a genotoxin that induces DNA double-strand breaks that may lead to carcinogenesis and is produced by Escherichia coli strains harboring the pks island. Human and animal studies have shown that colibactin-producing gut bacteria promote carcinogenesis and enhance the progression of colorectal cancer through cellular senescence and chromosomal abnormalities. In this study, we investigated the impact of prebiotics on the genotoxicity of colibactin-producing E. coli strains Nissle 1917 and NC101. METHODS: Bacteria were grown in medium supplemented with 20, 30 and 40 mg/mL of prebiotics inulin or galacto-oligosaccharide, and with or without 5 µM, 25 µM and 125 µM of ferrous sulfate. Colibactin expression was assessed by luciferase reporter assay for the clbA gene, essential for colibactin production, in E. coli Nissle 1917 and by RT-PCR in E. coli NC101. The human epithelial colorectal adenocarcinoma cell line, Caco-2, was used to assess colibactin-induced megalocytosis by methylene blue binding assay and genotoxicity by γ-H2AX immunofluorescence analysis. RESULTS: Inulin and galacto-oligosaccharide enhanced the expression of clbA in pks+ E. coli. However, the addition of 125 µM of ferrous sulfate inhibited the expression of clbA triggered by oligosaccharides. In the presence of either oligosaccharide, E. coli NC101 increased dysplasia and DNA double-strand breaks in Caco-2 cells compared to untreated cells. CONCLUSION: Our results suggest that, in vitro, prebiotic oligosaccharides exacerbate DNA damage induced by colibactin-producing bacteria. Further studies are necessary to establish whether oligosaccharide supplementation may lead to increased colorectal tumorigenesis in animal models colonized with pks+ E. coli.


Subject(s)
Carcinogenesis/pathology , Colonic Neoplasms/pathology , DNA Damage , Escherichia coli/metabolism , Mutagens/adverse effects , Oligosaccharides/pharmacology , Peptides/adverse effects , Polyketides/adverse effects , Caco-2 Cells , Carcinogenesis/chemically induced , Cellular Senescence , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Genomic Islands , Humans
17.
Chem Res Toxicol ; 34(3): 743-753, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33591737

ABSTRACT

Smoke samples from combustion of different biomass fuels were analyzed for the particulate bound Polycyclic Aromatic Hydrocarbons (PAHs) due to their carcinogenic and mutagenic nature. Out of 16 priority PAHs, 11 PAHs were detected in the emission of fuels, while the remaining 5 PAHs (chrysene, benzo[b]fluoranthene, benzo[k]fluoranthene, indeno[1,2,3-cd]pyrene, and benzo[g,h,i]perylene) were below the detection limit. The highest emission factor for the sum of all the PAHs was found for coal (353.08 mg kg-1), charcoal (27.28 mg kg-1), and the various wood types. Emission rates of total PAHs ranged from 0.37 to 5.15 mg h-1 with the highest value for bituminous coal (5.15 mg h-1) and lowest for Polyalthia longifolia (0.37 mg h-1). A cancer risk assessment was done for infants, children, and adults using the incremental lifetime cancer risk (ILCR) model via ingestion, inhalation, and dermal contact pathway. The ILCR values ranged from 10-11 to 10-6, and a higher cancer risk was observed for children and adults in comparison to infants. PAH concentrations emitted from biomass emissions shows a direct correlation with mutagenesis to humans, indicating a higher potential for the frameshift mutation as compared to base-pair mutation for dung, bituminous coal, charcoal, Dalbergia sissoo, Psidium guajava, Ziziphus mauritana, Polyalthia longifolia, and Ailanthus trithesa.


Subject(s)
Mutagens/adverse effects , Neoplasms/chemically induced , Polycyclic Aromatic Hydrocarbons/adverse effects , Biofuels , Biomass , Dose-Response Relationship, Drug , Humans , Mutation , Neoplasms/genetics , Salmonella typhimurium/drug effects
18.
Mutagenesis ; 36(1): 75-86, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33502495

ABSTRACT

Platinum-based drugs are a mainstay of cancer chemotherapy. However, their mutagenic effect can increase tumour heterogeneity, contribute to the evolution of treatment resistance and also induce secondary malignancies. We coupled whole genome sequencing with phenotypic investigations on two cell line models to compare the magnitude and examine the mechanism of mutagenicity of cisplatin, carboplatin and oxaliplatin. Cisplatin induced significantly more base substitution mutations than carboplatin or oxaliplatin when used at equitoxic concentrations on human TK6 or chicken DT40 cells, and also induced the highest number of short insertions and deletions. The analysis of base substitution spectra revealed that all three tested platinum drugs elicit both a direct mutagenic effect at purine dinucleotides, and an indirect effect of accelerating endogenous mutagenic processes, whereas the direct mutagenic effect appeared to correlate with the level of DNA damage caused as assessed through histone H2AX phosphorylation and single-cell agarose gel electrophoresis, the indirect mutagenic effects were equal. The different mutagenicity and DNA-damaging effect of equitoxic platinum drug treatments suggest that DNA damage independent mechanisms significantly contribute to their cytotoxicity. Thus, the comparatively high mutagenicity of cisplatin should be taken into account in the design of chemotherapeutic regimens.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage , Lymphocytes/pathology , Lymphoma/pathology , Mutagens/adverse effects , Animals , Carboplatin/pharmacology , Cells, Cultured , Chickens , Cisplatin/pharmacology , Humans , Lymphocytes/drug effects , Lymphoma/drug therapy , Mutagenicity Tests , Oxaliplatin/pharmacology
19.
Mutagenesis ; 36(1): 37-49, 2021 04 28.
Article in English | MEDLINE | ID: mdl-31793640

ABSTRACT

Implementation of the seventh amendment to the EU Cosmetics Directive has driven much research into suitable in vitro alternative assays to support satisfactory risk assessments. One such assay is the reconstructed skin micronucleus (RSMN) assay. First reported in 2006, further development occurred and a standard protocol was published in 2011. To evaluate and optimise the assay at Covance Laboratories, we tested nine chemicals [4-nitrophenol (4-NP), cyclohexanone (CH), 2-ethyl-1,3-hexanediol (2-EHD), methyl methansulfonate (MMS), mitomycin C (MMC), ethyl nitrosourea (ENU), benzo[a]pyrene (BaP), cyclophosphamide (CPA) and vinblastine (VIN)] using the EpiDerm™ 3D skin model (MatTek Corporation®, IVLSL, Bratislava, Slovakia) and compared the data using the standard 48-h treatment regimen and also an emerging 72-h treatment protocol. The EpiDerm™ tissue has reportedly some metabolic capacity but data using 48-h treatments has provided mixed results. Our investigations demonstrate that the two chemicals requiring metabolic activation (BaP and CPA) were negative following the 48-h protocol but were clearly positive following 72-h treatment. Furthermore, Replication Index (RI) data showed higher RI values in vehicle control treatments (indicating increased cell division) across the treatment set following 72-h treatments. A general greater magnitude of micronucleus (MN) induction was also observed following test chemical treatment. These data suggest that the 72-h treatment protocol is more suitable as a standard approach for the detection of clastogenic, aneugenic and metabolically activated chemicals in the RSMN assay. For further assay optimisation, we compare the statistical power of scoring cells from duplicate or triplicate cultures per treatment concentration and provide recommendations.


Subject(s)
Biological Assay/methods , DNA Damage , Laboratories/standards , Micronucleus Tests/methods , Micronucleus Tests/standards , Mutagens/adverse effects , Skin/pathology , Humans , Skin/drug effects , Skin/metabolism
20.
Mutagenesis ; 36(1): 63-74, 2021 04 28.
Article in English | MEDLINE | ID: mdl-31816077

ABSTRACT

In vitro genotoxicity assays utilising human skin models are becoming important tools for the safety assessment of chemicals whose primary exposure is via the dermal route. In order to explore metabolic competency and inducibility of CYP450 activating enzymes, 3D reconstructed human skin tissues were topically treated with 2-acetylaminofluorene (2-AAF) and its genotoxic metabolites, N-hydroxy-2-acetylaminofluorene (N-OH-2-AAF) and N-hydroxy-2-aminofluorene (N-OH-2-AF), which primarily cause DNA damage by forming DNA adducts. 2-AAF did not increase DNA damage measured in the reconstructed skin micronucleus (RSMN) assay when administered in multiple applications at 24 h intervals but was detected in the skin comet assay in the presence of the DNA polymerase inhibitor aphidicolin (APC). Similarly, no increase was found with N-OH-2-AAF in the RSMN assay after multiple treatments whereas a single 3 h exposure to N-OH-2-AAF caused a large dose-related increase in the skin comet assay. A significant increase in the RSMN assay was only obtained with the highly reactive N-OH-2-AF metabolite after multiple treatments over 72 h, whereas N-OH-2-AF caused a strong increase after a single 3 h exposure in the skin comet assay. In support of these results, DNA adduct formation, measured by the 32P-postlabelling assay, was examined. Adduct levels after 2-AAF treatment for 3 h were minimal but increased >10-fold after multiple exposures over 48 h, suggesting that enzyme(s) that metabolise 2-AAF are induced in the skin models. As expected, a single 3 h exposure to N-OH-2-AAF and N-OH-2-AF resulted in adduct levels that were at least 10-fold greater than those after multiple exposures to 2-AAF despite ~100-fold lower tested concentrations. Our results demonstrate that DNA damage caused by 2-AAF metabolites is more efficiently detected in the skin comet assay than the RSMN assay and after multiple exposures and enzyme induction, 2-AAF-induced DNA damage can be detected in the APC-modified comet assay.


Subject(s)
2-Acetylaminofluorene/adverse effects , DNA Adducts , DNA Damage , Micronucleus Tests/methods , Mutagens/adverse effects , Skin/pathology , Carcinogens/pharmacology , Fluorenes/adverse effects , Humans , Hydroxyacetylaminofluorene/adverse effects , Skin/drug effects , Skin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...