Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33903247

RESUMEN

Lanthanides are a series of critical elements widely used in multiple industries, such as optoelectronics and healthcare. Although initially considered to be of low toxicity, concerns have emerged during the last few decades over their impact on human health. The toxicological profile of these metals, however, has been incompletely characterized, with most studies to date solely focusing on one or two elements within the group. In the current study, we assessed potential toxicity mechanisms in the lanthanide series using a functional toxicogenomics approach in baker's yeast, which shares many cellular pathways and functions with humans. We screened the homozygous deletion pool of 4,291 Saccharomyces cerevisiae strains with the lanthanides and identified both common and unique functional effects of these metals. Three very different trends were observed within the lanthanide series, where deletions of certain proteins on membranes and organelles had no effect on the cellular response to early lanthanides while inducing yeast sensitivity and resistance to middle and late lanthanides, respectively. Vesicle-mediated transport (primarily endocytosis) was highlighted by both gene ontology and pathway enrichment analyses as one of the main functions disturbed by the majority of the metals. Protein-protein network analysis indicated that yeast response to lanthanides relied on proteins that participate in regulatory paths used for calcium (and other biologically relevant cations), and lanthanide toxicity included disruption of biosynthetic pathways by enzyme inhibition. Last, multiple genes and proteins identified in the network analysis have human orthologs, suggesting that those may also be targeted by lanthanides in humans.


Asunto(s)
Endocitosis/efectos de los fármacos , Elementos de la Serie de los Lantanoides/toxicidad , Saccharomyces cerevisiae/efectos de los fármacos , Fenómenos Toxicológicos/genética , Vías Biosintéticas/efectos de los fármacos , Genoma Fúngico/efectos de los fármacos , Humanos , Elementos de la Serie de los Lantanoides/farmacología , Saccharomyces cerevisiae/genética , Toxicogenética/tendencias
2.
Mutagenesis ; 35(2): 153-159, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32087008

RESUMEN

Toxicogenomics, the application of genomics to toxicology, was described as 'a new era' for toxicology. Standard toxicity tests typically involve a number of short-term bioassays that are costly, time consuming, require large numbers of animals and generally focus on a single end point. Toxicogenomics was heralded as a way to improve the efficiency of toxicity testing by assessing gene regulation across the genome, allowing rapid classification of compounds based on characteristic expression profiles. Gene expression microarrays could measure and characterise genome-wide gene expression changes in a single study and while transcriptomic profiles that can discriminate between genotoxic and non-genotoxic carcinogens have been identified, challenges with the approach limited its application. As such, toxicogenomics did not transform the field of genetic toxicology in the way it was predicted. More recently, next generation sequencing (NGS) technologies have revolutionised genomics owing to the fact that hundreds of billions of base pairs can be sequenced simultaneously cheaper and quicker than traditional Sanger methods. In relation to genetic toxicology, and thousands of cancer genomes have been sequenced with single-base substitution mutational signatures identified, and mutation signatures have been identified following treatment of cells with known or suspected environmental carcinogens. RNAseq has been applied to detect transcriptional changes following treatment with genotoxins; modified RNAseq protocols have been developed to identify adducts in the genome and Duplex sequencing is an example of a technique that has recently been developed to accurately detect mutation. Machine learning, including MutationSeq and SomaticSeq, has also been applied to somatic mutation detection and improvements in automation and/or the application of machine learning algorithms may allow high-throughput mutation sequencing in the future. This review will discuss the initial promise of transcriptomics for genetic toxicology, and how the development of NGS technologies and new machine learning algorithms may finally realise that promise.


Asunto(s)
Genómica/tendencias , Secuenciación de Nucleótidos de Alto Rendimiento/tendencias , Toxicogenética/tendencias , Toxicología/tendencias , Algoritmos , Regulación de la Expresión Génica/genética , Humanos , Aprendizaje Automático , Mutágenos/metabolismo , Mutación
3.
Toxicol Ind Health ; 33(3): 222-249, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27030242

RESUMEN

Diabetes mellitus (DM) is a group of metabolic diseases that may originate from an interaction between genetic and lifestyle risk factors. However, the possible role of occupational chemical exposures in the disease development and progression remains unclear. Therefore, this review aimed to provide a comprehensive evaluation of the relationship between occupational exposure to specific chemical substances or industrial activities and DM morbidity and mortality outcomes. Although some positive findings may support the diabetogenic role of certain pesticides and dioxins in different workplaces, the variable conditions of exposure, the lack of quantitative environmental or biological monitoring data and the different outcomes evaluated do not allow defining a specific exposure-disease causality. Therefore, further epidemiological studies will be necessary to adequately assess modes of action for different substances, dose-response relationships as well as individual susceptibility factors potentially affecting the exposure-disease continuum. Overall, this appears important to adequately assess, communicate and manage risks in occupational chemical exposure settings with the aim to protect workers and build healthier job conditions for diabetic employees.


Asunto(s)
Diabetes Mellitus/inducido químicamente , Medicina Basada en la Evidencia , Enfermedades Profesionales/inducido químicamente , Exposición Profesional/efectos adversos , Animales , Diabetes Mellitus/epidemiología , Diabetes Mellitus/genética , Diabetes Mellitus/fisiopatología , Progresión de la Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Enfermedades Profesionales/epidemiología , Enfermedades Profesionales/genética , Enfermedades Profesionales/fisiopatología , Riesgo , Medición de Riesgo , Gestión de Riesgos , Toxicogenética/métodos , Toxicogenética/tendencias
4.
Part Fibre Toxicol ; 13(1): 25, 2016 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-27169501

RESUMEN

BACKGROUND: The increasing use of engineered nanomaterials (ENMs) of varying physical and chemical characteristics poses a great challenge for screening and assessing the potential pathology induced by these materials, necessitating novel toxicological approaches. Toxicogenomics measures changes in mRNA levels in cells and tissues following exposure to toxic substances. The resulting information on altered gene expression profiles, associated pathways, and the doses at which these changes occur, are used to identify the underlying mechanisms of toxicity and to predict disease outcomes. We evaluated the applicability of toxicogenomics data in identifying potential lung-specific (genomic datasets are currently available from experiments where mice have been exposed to various ENMs through this common route of exposure) disease outcomes following exposure to ENMs. METHODS: Seven toxicogenomics studies describing mouse pulmonary responses over time following intra-tracheal exposure to increasing doses of carbon nanotubes (CNTs), carbon black, and titanium dioxide (TiO2) nanoparticles of varying properties were examined to understand underlying mechanisms of toxicity. mRNA profiles from these studies were compared to the publicly available datasets of 15 other mouse models of lung injury/diseases induced by various agents including bleomycin, ovalbumin, TNFα, lipopolysaccharide, bacterial infection, and welding fumes to delineate the implications of ENM-perturbed biological processes to disease pathogenesis in lungs. RESULTS: The meta-analysis revealed two distinct clusters-one driven by TiO2 and the other by CNTs. Unsupervised clustering of the genes showing significant expression changes revealed that CNT response clustered with bleomycin injury and bacterial infection models, both of which are known to induce lung fibrosis, in a post-exposure-time dependent manner, irrespective of the CNT's physical-chemical properties. TiO2 samples clustered separately from CNTs and disease models. CONCLUSIONS: These results indicate that in the absence of apical toxicity data, a tiered strategy beginning with short term, in vivo tissue transcriptomics profiling can effectively and efficiently screen new ENMs that have a higher probability of inducing pulmonary pathogenesis.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Exposición por Inhalación/efectos adversos , Enfermedades Pulmonares/inducido químicamente , Pulmón/efectos de los fármacos , Nanoestructuras/toxicidad , Mucosa Respiratoria/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Contaminantes Atmosféricos/química , Animales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Nanoestructuras/química , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Toxicogenética/métodos , Toxicogenética/tendencias
5.
Arch Toxicol ; 89(3): 319-25, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25586887

RESUMEN

A number of environmental toxicants affect our health through physical, biological or chemical mechanisms. There is growing evidence indicating that microRNA (miRNA) plays an important role in toxicogenomics, disease aetiology and the effect of toxicants. This article summarises recent findings on miRNAs associated with various toxicants and those targeted in the development of therapeutics. Environmental epigenetic studies have revealed the role of miRNAs in the regulation of gene activities induced by environmental changes after exposure to toxic substances. Toxicant-induced changes in miRNA expression have a potential to be informative markers in the evaluation of toxicant risks. miRNAs are now considered to be predictive biomarkers or indicators of tissue injury due to toxicant exposure; thus, miRNAs can also be utilised as therapeutic targets.


Asunto(s)
Exposición a Riesgos Ambientales/análisis , Contaminantes Ambientales/toxicidad , Expresión Génica/efectos de los fármacos , MicroARNs/genética , Toxicogenética , Animales , Relación Dosis-Respuesta a Droga , Humanos , Factores de Tiempo , Toxicogenética/métodos , Toxicogenética/tendencias
6.
Mutagenesis ; 29(1): 73-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24334751

RESUMEN

A number of influences including legislation, industry and academia have encouraged advances in computational toxicology and high-throughput testing to probe more broadly putative toxicity pathways. The aim of the 25th United Kingdom Mutagen Society (UKEMS) Industrial Genotoxicity Group Annual Meeting 2011 was to explore current and upcoming research tools that may provide new cancer risk estimation approaches and discuss the genotoxicity testing paradigm of the future. The meeting considered whether computer modelling, molecular biology systems and/or adverse outcome pathway approaches can provide more accurate toxicity predictions and whether high-content study data, pluripotent stem cells or new scientific disciplines, such as epigenetics and adductomics, could be integrated into the risk assessment process. With close collaboration between industry, academia and regulators next generation predictive models and high-content tools have the potential to transform genetic toxicology testing in the 21st century.


Asunto(s)
Pruebas de Mutagenicidad/métodos , Humanos , Pruebas de Mutagenicidad/normas , Pruebas de Mutagenicidad/tendencias , Toxicogenética/métodos , Toxicogenética/normas , Toxicogenética/tendencias
7.
Genetika ; 49(1): 81-93, 2013 Jan.
Artículo en Ruso | MEDLINE | ID: mdl-23662426

RESUMEN

The review highlights the history of genetic toxicology as a distinct research area, as well as the issues of genetic toxicology and development of its methodology. The strategies and testing patterns of genotoxic compounds are discussed with the purpose of identifying potential human carcinogens, as well as compounds capable of inducing heritable mutations in humans. The main achievements of genetic toxicology in the 20th century are summarized and the challenges of the 21st century are discussed.


Asunto(s)
Toxicogenética/historia , Historia del Siglo XX , Historia del Siglo XXI , Toxicogenética/tendencias
8.
Regul Toxicol Pharmacol ; 61(2): 141-53, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21840362

RESUMEN

Toxicogenomics is the application of toxicology, genetics, molecular biology and environmental health to describe the response of organisms to environmental stimuli. The field of toxicogenomics has developed over the past 15 years mainly due to advances in toxicology, molecular genetics and cell biology. Its prospective use to resolve crucial data gaps and data inconsistencies could improve risk assessment by providing additional data to increase the understanding of mechanisms and modes of action (MOA) and enhance the reliability of dose-response extrapolation. Thus, toxicogenomics holds promise for advancing the scientific basis of risk assessments. However, one of the current issues is how genomic/transcriptional data is being used to further describe a MOA for oncogenicity and, in turn, its potential uses in cancer risk assessment. This commentary identifies how toxicogenomics could be used on a case by case basis to add information to a MOA addressing both the opportunities and challenges this technology holds. In addition, some pitfalls to avoid in the generation and interpretation of toxicogenomic data and validation issues that need to be addressed before toxicogenomics can be used in the risk assessment process and regulatory decisions are discussed.


Asunto(s)
Genómica/métodos , Toxicogenética/métodos , Toxicología/métodos , Animales , Transformación Celular Neoplásica/genética , Relación Dosis-Respuesta a Droga , Genómica/tendencias , Humanos , Reproducibilidad de los Resultados , Medición de Riesgo/métodos , Medición de Riesgo/tendencias , Toxicogenética/tendencias , Toxicología/tendencias
9.
Mutat Res ; 705(3): 201-4, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20850562

RESUMEN

We identify trends over the past decades in membership in societies affiliated with the International Association of Environmental Mutagen Societies (IAEMS), and we also highlight findings in a recent review by Claxton et al. [Environ Health Perspect, in press] regarding the numbers of papers published per year using genetic toxicology assays. These analyses reveal a decline or at best a static level of membership in IAEMS-affiliated societies, as well as a decline in the number of papers published per year using genetic toxicology assays-with the exception of those using comet assays, which already have begun to plateau. In contrast, toxicogenomics and computational toxicology are becoming increasingly prominent relative to environmental mutagenesis research in most research institutes, reflecting the ascendancy of these areas of environmental toxicology. We conclude that changing the name of IAEMS and its affiliated societies to reflect these changes might enhance membership and publication by welcoming a broader range of scientists into these societies. Although various names are possible, we think that changing the name of these societies to "Environmental Genomics Society" may help to make our societies more attractive to a broader range of scientists, resulting in an increase in membership and an acceleration of the incorporation of genomic methods into environmental research.


Asunto(s)
Mutágenos , Sociedades Científicas/organización & administración , Exposición a Riesgos Ambientales , Genómica , Humanos , Cooperación Internacional , Nombres , Publicaciones , Toxicogenética/tendencias , Toxicología/tendencias
10.
Mutat Res ; 705(3): 184-200, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20399889

RESUMEN

The results of predictive toxicogenomics investigations over the past 6 years reviewed in this report have shed new light on the potential of molecular expression analysis to more properly classify both genotoxic and nongenotoxic carcinogens and to predict the carcinogenicity of untested chemicals. Predictive toxicogenomics uses global molecular expression data resulting from genomic perturbation (e.g., transcription or gene expression profiles) to predict a toxicological outcome, such as carcinogenicity. The classification of carcinogens has become an essential and highly debatable component of cancer risk assessment largely because of the default assumptions that drive regulatory decision-making regarding the presumed linearity of the dose-response curve for genotoxic carcinogens. Nongenotoxic mechanisms of carcinogenesis complicate the well-established relationship between genotoxicity and carcinogenicity and challenge the interpretation of the results of rodent carcinogenicity studies in terms of their relevance to humans. Although the number of presumed nongenotoxic rodent carcinogens has dramatically increased over the past two decades, the fact remains that more than 90% of the known human carcinogens are detected in conventional short-term tests for genotoxicity and induce tumors at multiple sites in rodents. In toxicogenomics studies, a strong DNA damage response at the gene expression level suggests direct DNA modification whereas increased expression of genes involved in cell cycle progression is more characteristic of the indirect-acting agents such as those that induce oxidative stress. Metabolism genes are prominently represented among gene expression profiles that discriminate nongenotoxic modes of action (e.g., cytotoxicity and regenerative proliferation, xenobiotic receptor agonists, peroxisome proliferator-activated receptors, or hormonal-mediated processes). The evidence accumulated to date suggests that gene expression profiles reflect underlying modes or mechanisms of action, such that they will be useful in the prediction of chemical carcinogenicity, especially in conjunction with conventional short-term tests for gene mutation, chromosomal aberration and aneuploidy.


Asunto(s)
Pruebas de Carcinogenicidad , Genómica/tendencias , Toxicogenética/tendencias , Toxicología/tendencias , Animales , Bioensayo , Carcinógenos , Daño del ADN , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Masculino , Ratones , Ratas
11.
Mutat Res ; 705(3): 172-83, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20382258

RESUMEN

Gene-environment interactions contribute to complex disease development. The environmental contribution, in particular low-level and prevalent environmental exposures, may constitute much of the risk and contribute substantially to disease. Systematic risk evaluation of the majority of human chemical exposures, has not been conducted and is a goal of regulatory agencies in the U.S. and worldwide. With the recent recognition that toxicological approaches more predictive of effects in humans are required for risk assessment, in vitro human cell line data as well as animal data are being used to identify toxicity mechanisms that can be translated into biomarkers relevant to human exposure studies. In this review, we discuss how data from toxicogenomic studies of exposed human populations can inform risk assessment, by generating biomarkers of exposure, early effect, and/or susceptibility, elucidating mechanisms of action underlying exposure-related disease, and detecting response at low doses. Good experimental design incorporating precise, individual exposure measurements, phenotypic anchors (pre-disease or traditional toxicological markers), and a range of relevant exposure levels, is necessary. Further, toxicogenomic studies need to be designed with sufficient power to detect true effects of the exposure. As more studies are performed and incorporated into databases such as the Comparative Toxicogenomics Database (CTD) and Chemical Effects in Biological Systems (CEBS), data can be mined for classification of newly tested chemicals (hazard identification), and, for investigating the dose-response, and inter-relationship among genes, environment and disease in a systems biology approach (risk characterization).


Asunto(s)
Genómica/tendencias , Medición de Riesgo/métodos , Medición de Riesgo/tendencias , Toxicogenética/tendencias , Toxicología/tendencias , Animales , Arsénico/toxicidad , Benceno/toxicidad , Exposición a Riesgos Ambientales , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Proteómica/métodos , Biología de Sistemas
12.
Mutat Res ; 705(3): 165-71, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20466069

RESUMEN

Genomics may be an effective tool in decreasing the lengthy drug development process and reducing compound attrition. It can generate specific gene expression profiles induced by chemicals that can be linked to dose and response. Toxicogenomics can identify sensitive biomarkers of early deleterious effects, distinguish genotoxic from non-genotoxic carcinogens and can provide information on the mechanism of action. It can help bridge in vitro to in vivo findings and provide context for preclinical data and thus address human health risks. Issues and shortcomings that still need to be resolved or improved for efficient incorporation of genomics in drug development and environmental toxicology research include data analysis, data interpretation tools and accessible data repositories. In addition, implementation of toxicogenomics in early screening or drug discovery phases and effective use of this information by project teams remains a challenge.


Asunto(s)
Genómica/tendencias , Toxicología/tendencias , Animales , Biomarcadores , Carcinógenos , Técnicas Químicas Combinatorias , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Perfilación de la Expresión Génica , Genómica/métodos , Humanos , Tecnología Farmacéutica/métodos , Toxicogenética/tendencias , Toxicología/métodos
13.
Planta Med ; 76(17): 2019-25, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20957595

RESUMEN

Toxicogenomics represents the integration of genomics and toxicology to investigate the interaction between genes and environmental stress in human health. It is a scientific field that studies how the genome is involved in responses to environmental stressors and toxicants. The patterns of altered gene expression that are caused by specific exposures or disease outcomes reveal how toxicants may act and cause disease. Nowadays, toxicogenomics faces great challenges in discriminating the molecular basis of toxicity. We do believe that advances in this field will eventually allow us to describe all the toxicological interactions that occur within a living system. Toxicogenomic responses of a toxic agent in one species (e.g., laboratory animals) may predict the mode of action in another species (e.g., humans) (predictive toxicology). Development and application of toxicogenomic databases and new bioinformatics tools are among the most important aspects of toxicogenomic research which will facilitate sharing and interpretation of the huge amount of biological information generated in this field. Medicinal herbs have played an important role in pharmacy from ancient to modern times. Nowadays, there is a revival of interest in medicinal plants and an increasing scientific interest in bioactive natural products. Medicinal herbs are usually considered to be nontoxic. However, the consumption of herbs could produce prominent toxic effects either due to inherent toxicity or to contaminants (heavy metals, microorganisms, pesticides, toxic organic solvents, radioactivity, etc.). Therefore, a critical assessment of their toxicity is an urgent issue. This review explores the field of toxicogenomics, pinpoints some of its research approaches and describes the challenges it faces. In particular, Chinese herbal preparations have been implicated.


Asunto(s)
Medicamentos Herbarios Chinos/efectos adversos , Plantas Medicinales , Toxicogenética/tendencias , Animales , Biología Computacional , Bases de Datos Factuales , Perfilación de la Expresión Génica , Genómica , Humanos , Medicina Tradicional China , Metabolómica , Proteómica , Control de Calidad , Toxicogenética/métodos , Toxicología
14.
Biochim Biophys Acta ; 1779(6-7): 363-75, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18515111

RESUMEN

A parallelism exists between human cytogenetics and cytogenetic toxicology. The breakthroughs, mostly coming from and used in clinical genetics, are widely used in genetic toxicology. The birth of human cytogenetics occurred in 1956 when it was published that the diploid number of chromosomes in humans is 46. The first stage in chromosome-induced mutagenesis began in 1938 when Sax published the effects of X-rays on the chromosomes of Drosophila. In 1959, the cytogenetic anomalies for Down, Klinefelter, and Turner syndromes were described, and parallelly in 1960, the first publication on chromosomal aberrations in man caused by ionizing radiation appeared. The cytogenetic analysis of chromosomal aberrations in cell cultures is considered one of the primary methods to evaluate induced mutagenesis. At the end of the 1960s, banding techniques allowed chromosomes to be individually identified, in parallel, the sister chromatid exchange analysis technology was described. Another milestone in the history of induced mutagenesis was the discovery that mutagenic agents were able to alter chromosomal division and segregation in gonads inducing meiotic nondisjunction. Here we review new approaches and applications such as biological dosimetry, translocation scoring using FISH, and micronucleus test. Chromosomal aberrations and micronucleus test are now effective cytogenetic biomarkers of early effect used as cancer predictors. Human cytogenetics has proven to be effective over its 50-year lifespan and, although each new technique that has appeared seemed to announce its end, the fact is that the current state of cytogenetics is in reality a collection of techniques that, while common, are cheap, fast, and wide-ranging. Therefore, in genotoxicology, they continue to be useful to identify mutagenic agents as well as to evaluate and analyze exposed populations.


Asunto(s)
Citogenética , Toxicogenética , Animales , Aberraciones Cromosómicas , Cromosomas Humanos/genética , Citogenética/historia , Citogenética/tendencias , Femenino , Técnicas Genéticas/historia , Técnicas Genéticas/tendencias , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Hibridación Fluorescente in Situ/historia , Hibridación Fluorescente in Situ/tendencias , Masculino , Mutagénesis , Mutación , Toxicogenética/historia , Toxicogenética/tendencias
15.
EXS ; 99: 401-52, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19157069

RESUMEN

The influence of combinatorial chemistry and high-throughput screening (HTS) technologies in the pharmaceutical industry during the last 10 years has been enormous. However, the attrition rate of drugs in the clinic due to toxicity during this period still remained 40-50%. The need for reduced toxicity failure led to the development of early toxicity screening assays. This chapter describes the state of the art for assays in the area of genotoxicity, cytotoxicity, carcinogenicity, induction of specific enzymes from phase I and II metabolism, competition assays for enzymes of phase I and II metabolism, embryotoxicity as well as endocrine disruption and reprotoxicity. With respect to genotoxicity, the full Ames, Ames II, Vitotox, GreenScreen GC, RadarScreen, and non-genotoxic carcinogenicity assays are discussed. For cytotoxicity, cellular proliferation, calcein uptake, oxygen consumption, mitochondrial activity, radical formation, glutathione depletion as well as apoptosis are described. For high-content screening (HCS), the possibilities for analysis of cytotoxicity, micronuclei, centrosome formation and phospholipidosis are examined. For embryotoxicity, endocrine disruption and reprotoxicity alternative assays are reviewed for fast track analysis by means of nuclear receptors and membrane receptors. Moreover, solutions for analyzing enzyme induction by activation of nuclear receptors, like AhR, CAR, PXR, PPAR, FXR, LXR, TR and RAR are given.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Industria Farmacéutica/métodos , Toxicogenética/métodos , Animales , Pruebas de Carcinogenicidad , Evaluación Preclínica de Medicamentos/tendencias , Industria Farmacéutica/tendencias , Humanos , Modelos Biológicos , Pruebas de Toxicidad , Toxicogenética/tendencias
16.
Curr Opin Pharmacol ; 8(5): 654-60, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18760379

RESUMEN

Productivity issues facing the pharmaceutical industry are numerous, and the current challenges come in the face of an aging population and a demand for new and better medications. These challenges call for improvement in the drug discovery and development process, which paradoxically comes on the heels of remarkable scientific advances and in an era of great opportunity in medical science. Despite these advances, the pharmaceutical industry has yet to translate breakthroughs in new technologies, including genomics, into new drug therapies for unmet medical needs. The strategic application of toxicogenomics to the earliest stages of a drug discovery program offers a valuable opportunity to identify potential safety hurdles earlier than is the norm today. We propose that using genomics predictively (in vitro to predict outcomes in vivo and short-term studies in vivo to predict safety issues in longer studies) can assist in reducing inefficiency in the current paradigm, which is still heavily weighted on traditional endpoints from lengthy in vivo studies. Implementation of these strategies will assist in solving the current pharmaceutical pipeline productivity dilemma of long cycle times and unacceptable attrition rates in the preclinical drug discovery process.


Asunto(s)
Diseño de Fármacos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Toxicogenética/tendencias , Animales , Evaluación Preclínica de Medicamentos , Predicción , Humanos , Investigación
17.
Altern Lab Anim ; 37 Suppl 1: 33-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19807202

RESUMEN

As a branch of pharmacogenomics aimed at predicting drug safety concerns, toxicogenomics drew much excitement with the emergence of technologies such as gene expression microarrays. A few years down the line, the evidence is scant that current approaches to toxicogenomics are really making an impact in areas such as preclinical toxicology. It has been argued that there needs to be a re-focus of application toward high-throughput approaches which combine the best of tissue and genomic modelling. This commentary gives a brief introduction to in vitro toxicogenomics, drawn from the perspectives of the specialist toxicogenomics company, SimuGen.


Asunto(s)
Descubrimiento de Drogas , Industria Farmacéutica/tendencias , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Toxicogenética/tendencias , Células Cultivadas , Seguridad de Productos para el Consumidor , Relación Dosis-Respuesta a Droga , Industria Farmacéutica/métodos , Ensayos Analíticos de Alto Rendimiento , Humanos , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Toxicogenética/métodos
18.
Trends Pharmacol Sci ; 40(2): 92-103, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30594306

RESUMEN

Toxicogenomics (TGx) has contributed significantly to toxicology and now has great potential to support moves towards animal-free approaches in regulatory decision making. Here, we discuss in vitro TGx systems and their potential impact on risk assessment. We raise awareness of the rapid advancement of genomics technologies, which generates novel genomics features essential for enhanced risk assessment. We specifically emphasize the importance of reproducibility in utilizing TGx in the regulatory setting. We also highlight the role of machine learning (particularly deep learning) in developing TGx-based predictive models. Lastly, we touch on the topics of how TGx approaches could facilitate adverse outcome pathways (AOP) development and enhance read-across strategies to further regulatory application. Finally, we summarize current efforts to develop TGx for risk assessment and set out remaining challenges.


Asunto(s)
Toxicogenética/métodos , Alternativas a las Pruebas en Animales , Animales , Humanos , Aprendizaje Automático , Reproducibilidad de los Resultados , Medición de Riesgo/métodos , Toxicogenética/tendencias
20.
Mutat Res ; 659(1-2): 158-65, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18342568

RESUMEN

Toxicogenomics is a field that emerged from the combination of conventional toxicology with functional genomics. In recent years, this field contributed immensely in defining adverse biological effects resulting from environmental stressors, toxins, drugs and chemicals. Through microarray technology, large-scale detection and quantification of mRNA transcripts and of microRNAs, related to alterations in mRNA stability or gene regulation became feasible. Other 'omics' technologies, notably proteomics and metabonomics soon joined in providing further fine tuning in the gathering and interpretation of toxicological data. A field that will inevitably modify the landscape for toxicogenomics is 'epigenetics', a term which refers to heritable changes in gene expression without accompanying alterations in the DNA sequence. These epigenetic changes are brought about by mechanisms such as DNA methylation, histone modifications, and non-coding RNAs in the regulation of gene expression patterns. Epigenetic mechanisms are essential in normal development and differentiation, but these can be misdirected leading to diseases, notably cancer. Indeed, there is now a mounting body of evidence that environmental exposures particularly in early development can induce epigenetic changes, which may be transmitted in subsequent generations or serve as basis of diseases developed later in life. In either way, epigenetic mechanisms will help interpret toxicological data or toxicogenomic approaches to identify epigenetic effects of environmental exposures. Thus, a full understanding of environmental interactions with the genome requires keeping abreast of epigenetic mechanisms, as well as conducting routine analysis of epigenetic modifications as part of the mechanism of actions of environmental exposure. A number of approaches are currently available to study epigenetic modifications in a gene-specific or genome-wide manner. Here we describe our approaches in studying the epigenetic modification of the tumor-suppressor gene Tslc1 (Igsf4a) in lung tumors obtained from transgenic mouse models.


Asunto(s)
Contaminantes Ambientales/toxicidad , Epigénesis Genética , Toxicogenética/tendencias , Animales , Metilación de ADN , Genes Supresores de Tumor , Humanos , Neoplasias Pulmonares/genética , Ratones , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda