Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-37770142

RESUMO

N-Nitrosodiethylamine (NDEA), a well-studied N-nitrosamine, was tested in rats to compare the dose-response relationship of three genotoxicity endpoints. Mutant / mutation frequencies were determined using the transgenic rodent (TGR) gene mutation assay and error corrected next generation sequencing (ecNGS) (i.e., duplex sequencing (DS)), and genetic damage was detected by the alkaline comet assay. Big Blue® (cII Locus) animals (n = 6 per dose group) were administered doses of 0.001, 0.01, 0.1, 1, 3 mg/kg/day NDEA by oral gavage. Samples were collected for cII mutation and DS analyses following 28-days of exposure and 3 days recovery. In a separate study, male Sprague-Dawley (SD) rats (n = 6 per dose group) were administered the same doses by oral gavage for two consecutive days and then samples collected for the alkaline comet assay. A dose-related increase in mutant / mutation frequencies of the liver but not duodenum was observed using the TGR assay and DS with DS resulting in a slightly more sensitive response, with a lower benchmark dose (BMD). In addition, a dose-related increase in percent tail DNA was observed in the liver using the alkaline comet assay. Therefore, DS and comet assays showed good utility for hazard identification and dose-response analysis of a representative N-nitrosamine comparable to the TGR gene mutation assay.


Assuntos
Dietilnitrosamina , Nitrosaminas , Ratos , Animais , Masculino , Ensaio Cometa/métodos , Dietilnitrosamina/toxicidade , Roedores , Ratos Sprague-Dawley , Mutação , Animais Geneticamente Modificados , Dano ao DNA , Sequenciamento de Nucleotídeos em Larga Escala , Testes de Mutagenicidade/métodos , Relação Dose-Resposta a Droga
2.
Clin Pharmacol Ther ; 113(2): 226-245, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35388453

RESUMO

This review presents a European Federation of Pharmaceutical Industries and Association/PreClinical Development Expert Group (EFPIA-PDEG) topic group consensus on a data-driven approach to harmonized contraception recommendations for clinical trial protocols and product labeling. There is no international agreement in pharmaceutical clinical trial protocols or product labeling on when/if female and/or male contraception is warranted and for how long after the last dose. This absence of consensus has resulted in different recommendations among regions. For most pharmaceuticals, contraception recommendations are generally based exclusively on nonclinical data and/or mechanism. For clinical trials, contraception is the default position and is maintained for women throughout clinical development, whereas appropriate information can justify removing male contraception. Conversely, contraception is only recommended in product labeling when warranted. A base case rationale is proposed for whether or not female and/or male contraception is/are warranted, using available genotoxicity and developmental toxicity data. Contraception is generally warranted for both male and female subjects treated with mutagenic pharmaceuticals. We propose as a starting point that contraception is not typically warranted when the margin is 10-fold or greater between clinical exposure at the maximum recommended human dose and exposure at the no observed adverse effect level (NOAEL) for purely aneugenic pharmaceuticals and for pharmaceuticals that induce fetal malformations or embryo-fetal lethality. Other factors are discussed, including contraception methods, pregnancy testing, drug clearance, options for managing the absence of a developmental toxicity NOAEL, drug-drug interactions, radiopharmaceuticals, and other drug modalities. Overall, we present a data-driven rationale that can serve as a basis for consistent contraception recommendations in clinical trials and in product labeling across regions.


Assuntos
Anticoncepção , Indústria Farmacêutica , Gravidez , Humanos , Masculino , Feminino , Anticoncepção/efeitos adversos , Nível de Efeito Adverso não Observado , Consenso , Preparações Farmacêuticas
3.
Int J Toxicol ; 41(6): 442-454, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35989659

RESUMO

Poly(ADP-ribose) polymerase inhibitors (PARPi) are approved as monotherapies in BRCA1/2-mutated (mBRCA1/2) metastatic breast and ovarian cancers, and in advanced pancreatic and metastatic castration-resistant prostate cancers. Differential safety profiles across PARPi necessitate improved mechanistic understanding of inhibitor differences, especially with expansion of PARPi indications and drug combinations. Here, we report in vitro evaluations of PARPi (-/+ PARP trapper temozolomide, TMZ) with reference to total clinical mean concentration average or maximum (tCavg, tCmax), to elucidate contributions of primary pharmacology and structural differences to clinical efficacy and safety. In biochemical assays, rucaparib and niraparib demonstrated off-target secondary pharmacology activities, and in selectivity assays, talazoparib, olaparib, and rucaparib inhibited a broader panel of PARP enzymes. In donor-derived human bone marrow mononuclear cells, only olaparib both increased early apoptosis and decreased the cell viability half inhibitory concentration (IC50) at ≤ tCavg, whereas other PARPi only did so in the presence of TMZ. In cancer cell lines with DNA damage repair mutations, all PARPi decreased cell viability in H1048 but not TK6 cells, and only talazoparib decreased cell growth in DU145 cells at ≤ tCavg concentrations. When combined with low dose TMZ, only talazoparib left-shifted the functional consequences of PARP trapping (S-phase arrest, apoptosis, S-phase double-stranded breaks) and reduced cell viability/growth in TK6 and DU145 cell lines at ≤ tCavg, whereas the other inhibitors required high-dose TMZ. Our study suggests structural differences across PARPi may contribute to differences in PARP selectivity and off-target activities, which along with distinct pharmacokinetic properties, may influence inhibitor-specific toxicities in patients.


Assuntos
Inibidores de Poli(ADP-Ribose) Polimerases , Humanos , Masculino , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Temozolomida
4.
Int J Toxicol ; 40(3): 285-298, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33525949

RESUMO

A workshop entitled "Deriving Compound-Specific Exposure Limits for Chemicals Used in Pharmaceutical Synthesis" was held at the 2018 Genetic Toxicology Association annual meeting. The objectives of the workshop were to provide an educational forum and use case studies and live multiple-choice polling to establish the degree of similarity/diversity in approach/opinion of the industry experts and other delegates present for some of the more challenging decision points that need to be considered when developing a compound-specific exposure limit (ie, acceptable intake or permissible or permitted daily exposure). Herein we summarize the relevant background and case study information for each decision point topic presented as well as highlight significant polling responses and discussion points. A common observation throughout was the requirement for expert judgment to be applied at each of the decision points presented which often results in different reasoning being applied by the risk assessor when deriving a compound-specific exposure limit. This supports the value of precompetitive cross-industry collaborations to develop compound-specific limits and harmonize the methodology applied, thus reducing the associated uncertainty inherent in the application of isolated expert judgment in this context. An overview of relevant precompetitive cross-industry collaborations working to achieve this goal is described.


Assuntos
Exposição Ambiental/normas , Guias como Assunto , Preparações Farmacêuticas/normas , Medição de Risco/normas , Toxicologia/normas , Estudos de Casos e Controles , Tomada de Decisões , Humanos
5.
Int J Toxicol ; 40(2): 108-124, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33327828

RESUMO

There has been an increased interest in and activity for the use of peptide therapeutics to treat a variety of human diseases. The number of peptide drugs entering clinical development and the market has increased significantly over the past decade despite inherent challenges of peptide therapeutic discovery, development, and patient-friendly delivery. Disparities in interpretation and application of existing regulatory guidances to innovative synthetic and conjugated peptide assets have resulted in challenges for both regulators and sponsors. The Symposium on Development and Regulatory Challenges for Peptide Therapeutics at the 40th Annual Meeting of the American College of Toxicology held in November of 2019 focused on the following specific topics: (1) peptide therapeutic progress and future directions, and approaches to discover, optimize, assess, and deliver combination peptide therapeutics for treatment of diseases; (2) toxicological considerations to advance peptide drug-device combination products for efficient development and optimal patient benefit and adherence; (3) industry and regulatory perspectives on the regulation of synthetic and conjugated peptide products, including exploration of regulatory classifications, interpretations, and application of the existing guidances International Council for Harmonisation (ICH) M3(R2) and ICH S6(R1) in determining nonclinical study recommendations; and (4) presentation of the 2016 Health and Environmental Sciences Institute's Genetic Toxicology Technical Committee working group assessment of genotoxicity testing requirements. Perspectives were shared from industry and regulatory scientists working in the peptide therapeutics field followed by an open forum panel discussion to discuss questions drafted for the peptide therapeutics scientific community, which will be discussed in more detail.


Assuntos
Aprovação de Drogas/legislação & jurisprudência , Desenvolvimento de Medicamentos/normas , Doenças Metabólicas/tratamento farmacológico , Testes de Mutagenicidade/normas , Peptídeos/farmacologia , Peptídeos/toxicidade , Peptídeos/uso terapêutico , Aprovação de Drogas/métodos , Desenvolvimento de Medicamentos/métodos , Guias como Assunto , Humanos , Testes de Mutagenicidade/métodos , Estados Unidos , United States Food and Drug Administration/normas
6.
Nucleic Acid Ther ; 26(2): 73-85, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26978711

RESUMO

The Oligonucleotide Safety Working Group subcommittee on genotoxicity testing considers therapeutic oligonucleotides (ONs) unlikely to be genotoxic based on their properties and on the negative results for ONs tested to date. Nonetheless, the subcommittee believes that genotoxicity testing of new ONs is warranted because modified monomers could be liberated from a metabolized ON and incorporated into DNA and could hypothetically cause chain termination, miscoding, and/or faulty replication or repair. The standard test battery as described in Option 1 of International Conference on Harmonisation S2(R1) is generally adequate to assess such potential. However, for the in vitro assay for gene mutations, mammalian cells are considered more relevant than bacteria for most ONs due to their known responsiveness to nucleosides and their greater potential for ON uptake; on the other hand, bacterial assays may be more appropriate for ONs containing non-ON components. Testing is not recommended for ONs with only naturally occurring chemistries or for ONs with chemistries for which there is documented lack of genotoxicity in systems with demonstrated cellular uptake. Testing is recommended for ONs that contain non-natural chemical modifications and use of the complete drug product (including linkers, conjugates, and liposomes) is suggested to provide the most clinically relevant assessment. Documentation of uptake into cells comparable to those used for genotoxicity testing is proposed because intracellular exposure cannot be assumed for these large molecules. ONs could also hypothetically cause mutations through triple helix formation with genomic DNA and no tests are available for detection of such sequence-specific mutations across the entire genome. However, because the potential for triplex formation by therapeutic ONs is extremely low, this potential can be assessed adequately by sequence analysis.


Assuntos
Oligonucleotídeos/toxicidade , Animais , Células Cultivadas , Dano ao DNA , Avaliação Pré-Clínica de Medicamentos , Humanos , Testes de Mutagenicidade , Oligonucleotídeos/uso terapêutico
7.
Mutagenesis ; 31(4): 375-84, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27000792

RESUMO

The ICH S6(R1) recommendations on safety evaluation of biotherapeutics have led to uncertainty in determining what would constitute a cause for concern that would require genotoxicity testing. A Health and Environmental Sciences Institute's Genetic Toxicology Technical Committee Workgroup was formed to review the current practice of genotoxicity assessment of peptide/protein-related biotherapeutics. There are a number of properties of peptide/protein-related biotherapeutics that distinguish such products from traditional 'small molecule' drugs and need to be taken into consideration when assessing whether genotoxicity testing may be warranted and if so, how to do it appropriately. Case examples were provided by participating companies and decision trees were elaborated to determine whether and when genotoxicity evaluation is needed for peptides containing natural amino acids, non-natural amino acids and other chemical entities and for unconjugated and conjugated proteins. From a scientific point of view, there is no reason for testing peptides containing exclusively natural amino acids irrespective of the manufacturing process. If non-natural amino acids, organic linkers and other non-linker chemical components have already been tested for genotoxicity, there is no need to re-evaluate them when used in different peptide/protein-related biotherapeutics. Unless the peptides have been modified to be able to enter the cells, it is generally more appropriate to evaluate the peptides containing the non-natural amino acids and other non-linker chemical moieties in vivo where the cleavage products can be formed. For linkers, it is important to determine if exposure to reactive forms are likely to occur and from which origin. When the linkers are anticipated to be potential mutagenic impurities they should be evaluated according to ICH M7. If linkers are expected to be catabolic products, it is recommended to test the entire conjugate in vivo, as this would ensure that the relevant 'free' linker forms stemming from in vivo catabolism are tested.


Assuntos
Guias como Assunto , Testes de Mutagenicidade/métodos , Mutagênicos/toxicidade , Peptídeos/toxicidade , Animais , Humanos , Mutagênicos/efeitos adversos , Peptídeos/efeitos adversos , Peptídeos/uso terapêutico
8.
Regul Toxicol Pharmacol ; 72(2): 335-49, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25980641

RESUMO

The International Conference on Harmonization (ICH) M7 guidance for the assessment and control of DNA reactive impurities in pharmaceutical products includes the use of in silico prediction systems as part of the hazard identification and risk assessment strategy. This is the first internationally agreed guidance document to include the use of these types of approaches. The guideline requires the use of two complementary approaches, an expert rule-based method and a statistical algorithm. In addition, the guidance states that the output from these computer-based assessments can be reviewed using expert knowledge to provide additional support or resolve conflicting predictions. This approach is designed to maximize the sensitivity for correctly identifying DNA reactive compounds while providing a framework to reduce the number of compounds that need to be synthesized, purified and subsequently tested in an Ames assay. Using a data set of 801 chemicals and pharmaceutical intermediates, we have examined the relative predictive performances of some popular commercial in silico systems that are in common use across the pharmaceutical industry. The overall accuracy of each of these systems was fairly comparable ranging from 68% to 73%; however, the sensitivity of each system (i.e. how many Ames positive compounds are correctly identified) varied much more dramatically from 48% to 68%. We have explored how these systems can be combined under the ICH M7 guidance to enhance the detection of DNA reactive molecules. Finally, using four smaller sets of molecules, we have explored the value of expert knowledge in the review process, especially in cases where the two systems disagreed on their predictions, and the need for care when evaluating the predictions for large data sets.


Assuntos
Contaminação de Medicamentos , Mutagênicos/análise , Software , Algoritmos , Simulação por Computador , Medição de Risco
9.
J Am Chem Soc ; 136(44): 15719-29, 2014 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-25347620

RESUMO

It was by way of total synthesis that the issues concerning the stereostructure of leiodermatolide (1) have recently been solved; with the target now being unambiguously defined, the mission of synthesis changes as to secure a meaningful supply of this exceedingly scarce natural product derived from a deep-sea sponge. To this end, a scalable route of 19 steps (longest linear sequence) has been developed, which features a catalytic asymmetric propargylation of a highly enolizable ß-keto-lactone, a ring closing alkyne metathesis and a modified Stille coupling as the key transformations. Deliberate digression from this robust blueprint brought a first set of analogues into reach, which allowed the lead qualities of 1 to be assessed. The acquired biodata show that 1 is a potent cytotoxin in human tumor cell proliferation assays, distinguished by GI50 values in the ≤3 nM range even for cell lines expressing the Pgp efflux transporter. Studies with human U2OS cells revealed that 1 causes mitotic arrest, micronucleus induction, centrosome amplification and tubulin disruption, even though no evidence for direct tubulin binding has been found in cell-free assays; moreover, the compound does not seem to act through kinase inhibition. Indirect evidence points at centrosome declustering as a possible mechanism of action, which provides a potentially rewarding outlook in that centrosome declustering agents hold promise of being inherently selective for malignant over healthy human tissue.


Assuntos
Macrolídeos/síntese química , Macrolídeos/farmacologia , Macrolídeos/química , Estrutura Molecular
10.
Environ Mol Mutagen ; 55(1): 64-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23982927

RESUMO

Previous studies with TK6 cells have shown that extending the recovery period after pulse treatment allows for greater micronucleus expression for some compounds. This study explores the role of cell cycle delay in micronucleus expression after pulse treatment with three model genotoxins [mitomycin C, etoposide (ETOP), vinblastine]. Cells were treated for 4 hr and allowed to recover for 36 hr with samples removed at various time points during the recovery period and analyzed for cell cycle distribution, apoptosis and micronucleus frequency. Our results show that mitomycin C causes cell cycle delay for 20 hr after pulse treatment and cell cycle perturbation is no longer evident after 36 hr of recovery. The micronucleus frequency of cells sampled at 36 hr is doubled when compared with cells sampled at 20 hr after mitomycin C removal. When cells were treated with indirect acting genotoxins (ETOP, vinblastine), cell cycle perturbation was not observed at the 20 hr time point. Micronucleus frequency after treatment with either ETOP or vinblastine did not differ between the 20 hr and the 36 hr time point. All three compounds induced similar levels of apoptosis ranging from 4.5 to 5.6% with maximum induction occurring at the 36-hr time point. We conclude that TK6 cells exhibit extended cell cycle arrest after exposure to MMC and can go on to express micronuclei, after overcoming cell cycle arrest.


Assuntos
Ciclo Celular/genética , Testes para Micronúcleos/métodos , Mutagênicos/toxicidade , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular/efeitos dos fármacos , Etoposídeo/toxicidade , Humanos , Mitomicina/toxicidade , Vimblastina/toxicidade
11.
Mutat Res ; 746(1): 29-34, 2012 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-22445949

RESUMO

The Organization for Economic Co-operation and Development (OECD) has recently adopted Test Guideline 487 (TG487) for conducting the in vitro micronucleus (MNvit) assay. The purpose of this study is to evaluate and validate treatment conditions for the use of p53 competent TK6 human lymphoblastoid cells in a TG487 compliant MNvit assay. The ten reference compounds suggested in TG487 (mitomycin C, cytosine arabinoside, cyclophosphamide, benzo-a-pyrene, vinblastine sulphate, colchicine, sodium chloride, nalidixic acid and di(2-ethylhexyl)phthalate and pyrene) and noscapine hydrochloride were chosen for this study. In order to optimize the micronucleus response after treatment with some positive substances, we extended the recovery time after pulse treatment from 2 cell cycles recommended in TG487 to 3 cell cycles for untreated cells (40h). Each compound was tested in at least one of four exposure conditions: a 4h exposure followed by a 40h recovery, a 4h exposure followed by a 24h recovery, a 4h exposure in the presence of an exogenous metabolic activation system followed by a 40h recovery period, and a 27h continuous direct treatment. Results show that the direct acting clastogens, clastogens requiring metabolic activation and aneugens caused a robust increase in micronuclei in at least one test condition whereas the negative compounds did not induce micronuclei. The negative control cultures exhibited reproducibly low and consistent micronucleus frequencies ranging from 0.4 to 1.8% (0.8±0.3% average and standard deviation). Furthermore, extending the recovery period from 24h to 40h produced a 2-fold higher micronucleus frequency after a 4h pulse treatment with mitomycin C. In summary, the protocol described in this study in TK6 cells produced the expected result with model compounds and should be suitable for performing the MNvit assay in accordance with guideline TG487.


Assuntos
Antineoplásicos/toxicidade , Testes para Micronúcleos/métodos , Mutagênicos/toxicidade , Aneugênicos/toxicidade , Biotransformação , Linhagem Celular , Guias como Assunto , Humanos
12.
Environ Mol Mutagen ; 53(2): 94-100, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22020802

RESUMO

Cr(VI) is a human and animal carcinogen. Cr(VI) does not interact directly with DNA and thus its genotoxicity is attributed to its intracellular reduction to Cr(III) via reactive intermediates. The resulting types of DNA damage can be grouped into two categories: (1) oxidative DNA damage and (2) Cr(III)-DNA interactions. This study examines the molecular mechanism of Cr(VI) and Cr(III) genotoxicity in an intact cell. A system screening for DNA deletions (DEL assay) was used to compare induction of chromosomal rearrangements in the yeast Saccharomyces cerevisiae following Cr(VI) and Cr(III) exposure. Both forms of chromium induced DNA deletions albeit with different dose-response curves. N-acetylcysteine had a protective effect against Cr(VI) genotoxicity at high exposure doses but had no protective effect at lower doses or against Cr(III). An oxidative DNA damage repair mutant was hypersensitive to Cr(VI) only at high exposure and the mutant was not hypersensitive to Cr(III) exposure. These data imply that oxidative stress is involved in Cr(VI) genotoxicity at high exposure concentrations and not so in Cr(III). The Cr(III)-DNA interaction appears to be an important genotoxic lesion following Cr(VI) exposure at low-exposure concentrations. The CAN forward mutation assay revealed that within the concentration ranges used for this study, Cr(III) does not cause point mutations and Cr(VI) causes a mild but statistically significant increase in point mutation only at the highest concentration tested. This study reveals that DNA deletions occurring as a result of intrachromosomal homologous recombination are a useful endpoint for studying chromium genotoxicity.


Assuntos
Carcinógenos Ambientais/toxicidade , Cromo/toxicidade , Dano ao DNA/efeitos dos fármacos , Acetilcisteína/farmacologia , Animais , Carcinógenos Ambientais/metabolismo , Linhagem Celular , Cromo/metabolismo , Reparo do DNA/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Recombinação Homóloga , Taxa de Mutação , Oxirredução , Ratos
13.
J Med Chem ; 54(8): 2952-60, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21449606

RESUMO

Compound 4 (PF-04971729) belongs to a new class of potent and selective sodium-dependent glucose cotransporter 2 inhibitors incorporating a unique dioxa-bicyclo[3.2.1]octane (bridged ketal) ring system. In this paper we present the design, synthesis, preclinical evaluation, and human dose predictions related to 4. This compound demonstrated robust urinary glucose excretion in rats and an excellent preclinical safety profile. It is currently in phase 2 clinical trials and is being evaluated for the treatment of type 2 diabetes.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes/química , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Descoberta de Drogas , Inibidores do Transportador 2 de Sódio-Glicose , Animais , Área Sob a Curva , Compostos Bicíclicos Heterocíclicos com Pontes/farmacocinética , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Humanos , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Modelos Moleculares , Ratos
14.
Mutat Res ; 665(1-2): 37-43, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19427509

RESUMO

Ionizing radiation (IR) induces DNA strand breaks leading to cell death or deleterious genome rearrangements. In the present study, we examined the role of N-acetyl-L-cysteine (NAC), a clinically proven safe agent, for it's ability to protect against gamma-ray-induced DNA strand breaks and/or DNA deletions in yeast and mammals. In the yeast Saccharomyces cerevisiae, DNA deletions were scored by reversion to histidine prototrophy. Human lymphoblastoid cells were examined for the frequency of gamma-H2AX foci formation, indicative of DNA double strand break formation. DNA strand breaks were also measured in mouse peripheral blood by the alkaline comet assay. In yeast, NAC reduced the frequency of IR-induced DNA deletions. However, NAC did not protect against cell death. NAC also reduced gamma-H2AX foci formation in human lymphoblastoid cells but had no protective effect in the colony survival assay. NAC administration via drinking water fully protected against DNA strand breaks in mice whole-body irradiated with 1Gy but not with 4Gy. NAC treatment in the absence of irradiation was not genotoxic. These data suggest that, given the safety and efficacy of NAC in humans, NAC may be useful in radiation therapy to prevent radiation-mediated genotoxicity, but does not interfere with efficient cancer cell killing.


Assuntos
Acetilcisteína/farmacologia , Morte Celular/efeitos dos fármacos , Dano ao DNA , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/efeitos da radiação , Animais , Linhagem Celular , Ensaio de Unidades Formadoras de Colônias , Ensaio Cometa , Quebras de DNA , Sequestradores de Radicais Livres/farmacologia , Histonas/metabolismo , Humanos , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Linfócitos/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/genética
15.
Mutat Res ; 638(1-2): 83-9, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17963796

RESUMO

HNO is genotoxic but its mechanism is not well understood. There are many possible mechanisms by which HNO can attack DNA. Since HNO is electrophilic, it may react with exocyclic amine groups on DNA bases and through a series of subsequent reactions form a deaminated product. Alternatively, HNO may induce radical chemistry through O(2)-dependent (or possibly O(2)-independent) chemistry. In cell free systems, experiments have shown that HNO does react with DNA, resulting in base oxidation and strand cleavage. In this study, we used a whole-cell system in the yeast Saccharomyces cerevisiae to study the mechanism of HNO induced DNA damage with Angeli's salt as HNO donor. The yeast DEL assay provided a measure of intrachromosomal recombination leading to DNA deletions. We also examined interchromosomal recombination leading to genomic rearrangements and used the canavanine (CAN) assay to study induction of forward point mutations. HNO was a potent inducer of DNA deletions and recombination but it was negative for induction of point mutations. This suggests that HNO causes DNA strand breaks rather than base damage. Genotoxicity was observed under aerobic and anaerobic conditions and NAC protected against HNO induced DNA deletions. Since HNO is genotoxic under anaerobic conditions, NAC probably protected against radicals generated by HNO independent of oxygen.


Assuntos
Nitritos/toxicidade , Óxidos de Nitrogênio/toxicidade , Saccharomyces cerevisiae/genética , Deleção de Sequência , Acetilcisteína/farmacologia , Anaerobiose , DNA Fúngico/efeitos dos fármacos , Testes de Mutagenicidade/métodos , Recombinação Genética , Saccharomyces cerevisiae/efeitos dos fármacos
16.
Toxicol In Vitro ; 22(2): 296-300, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17959352

RESUMO

The estimated cancer risk from diesel exhaust particles (DEP) in the air is approximately 70% of the cancer risk from all air pollutants. DEP is comprised of a complex mixture of chemicals whose carcinogenic potential has not been adequately assessed. The polycyclic aromatic hydrocarbon quinone 9,10-phenanthrenequinone (9,10 PQ) is a major component of DEP and a suspect genotoxic agent for DEP induced DNA damage. 9,10 PQ undergoes redox cycling to produce reactive oxygen species that can lead to oxidative DNA damage. We used two systems in the yeast Saccharomyces cerevisiae to examine possible differential genotoxicity of 9,10 PQ. The DEL assay measures intra-chromosomal homologous recombination leading to DNA deletions and the CAN assay measures forward mutations leading to canavanine resistance. Cells were exposed to 9,10 PQ aerobically and anaerobically followed by DNA damage assessment. The results indicate that 9,10 PQ induces DNA deletions and point mutations in the presence of oxygen while exhibiting negligible effects anaerobically. In contrast to the cytotoxicity observed aerobically, the anaerobic effects of 9,10 PQ seem to be cytostatic in nature, reducing growth without affecting cell viability. Thus, 9,10 PQ requires oxygen for genotoxicity while different toxicities exhibited aerobically and anaerobically suggest multiple mechanisms of action.


Assuntos
DNA Fúngico/efeitos dos fármacos , DNA Fúngico/genética , Deleção de Genes , Mutagênicos/toxicidade , Mutação/efeitos dos fármacos , Fenantrenos/toxicidade , Saccharomyces cerevisiae/genética , Aerobiose , Anaerobiose , Benzoquinonas/farmacologia , Canavanina/farmacologia , Dano ao DNA/efeitos dos fármacos , Oxirredução , Saccharomyces cerevisiae/efeitos dos fármacos
17.
Cancer Res ; 66(7): 3480-4, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16585171

RESUMO

Industrial Cr(VI) emissions contaminate drinking water sources across the U.S., and many people take Cr(III) nutritional supplements. Cr(VI) is a human pulmonary carcinogen, but whether it is carcinogenic in the drinking water is not known. Due to widespread human exposure, it is imperative to determine the carcinogenic potential of Cr(VI) and Cr(III). DNA deletions and other genome rearrangements are involved in carcinogenesis. We determined the effects of Cr(VI) as potassium dichromate and Cr(III) as chromium(III) chloride on the frequencies of DNA deletions measured with the deletion assay in Saccharomyces cerevisiae and the in vivo p(un) reversion assay in C57BL/6J p(un)/p(un) mice. Exposing yeast and mice via drinking water to Cr(VI) and Cr(III) significantly increased the frequency of DNA deletions. We quantified intracellular chromium concentrations in yeast and tissue chromium concentrations in mice after exposure. Surprisingly, this revealed that Cr(III) is a more potent inducer of DNA deletions than Cr(VI) once Cr(III) is absorbed. This study concludes that both the environmental contaminant Cr(VI) and the nutritional supplement Cr(III) increase DNA deletions in vitro and in vivo, when ingested via drinking water.


Assuntos
Cloretos/toxicidade , Compostos de Cromo/toxicidade , DNA/efeitos dos fármacos , Deleção de Genes , Dicromato de Potássio/toxicidade , Animais , Testes de Carcinogenicidade , Cloretos/administração & dosagem , Compostos de Cromo/administração & dosagem , DNA/genética , DNA Fúngico/efeitos dos fármacos , DNA Fúngico/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Dicromato de Potássio/administração & dosagem , Gravidez , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Água
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...