1
|
Botelho MT, Umbuzeiro GDA. Designing and applying a methodology to assess sperm cell viability and DNA damage in a model amphipod. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 950:175318. [PMID: 39111426 DOI: 10.1016/j.scitotenv.2024.175318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/16/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Sperm quality is defined as the sperm cell ability to successfully fertilize eggs and allow normal embryo development. Few studies explore sperm quality using aquatic invertebrates. Parhyale hawaiensis is a marine amphipod with a circumtropical distribution and considered a model for evolution, development, and ecotoxicological studies. We aimed to develop a methodology to collect sperm cells of P. hawaiensis and evaluate their viability and DNA damage (comet assay). We directly exposed the sperm cells to different mutagenic agents to optimize/develop the protocols. Then, as a proof of concept, we exposed the males to mutagenic compounds (EMS, benzo[a]pyrene (BaP), azo and anthraquinone dyes) at non-lethal concentrations verified by the proposed viability test and analyzed their sperm cells for DNA damage (comet assay). Organisms exposed to EMS presented a clear concentration response in the DNA damage response. We also showed that BaP was able to induce a statistically significant increase in DNA damage of the sperm cells. For the two dyes, although DNA damage increased, statistically differences were not observed. We believe we successfully developed a test to detect genotoxicity of chemicals in sperm cells using an invertebrate model. The protocol for sperm cell viability needs to be further explored with different chemicals to verify its utility as a toxicity endpoint. The developed genotoxicity test has the advantages to employ organisms that are easily cultivated in reduced space, use simple laboratory resources and reduced amount of material and reagents. Positive responses with this model could be used to disclose new germ cell mutagen candidates which could be further confirmed in vertebrates' systems.
Collapse
|
2
|
Li J, Dai L, Feng Y, Cao Z, Ding Y, Xu H, Xu A, Du H. Multigenerational effects and mutagenicity of three flame retardants on germ cells in Caenorhabditis elegans. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115815. [PMID: 38091675 DOI: 10.1016/j.ecoenv.2023.115815] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/14/2023] [Accepted: 12/09/2023] [Indexed: 01/12/2024]
Abstract
Flame retardants (FRs) have raised public concerns because of their environmental persistence and negative impacts on human health. Recent evidence has revealed that many FRs exhibit reproductive toxicities and transgenerational impacts, whereas the toxic effects of FRs on germ cells remain barely explored. Here we investigated the multigenerational effects of three flame retardants (TBBPA, TCEP and TCPP) on germ cell development in Caenorhabditis elegans, and examined the germ cell mutagenicity of these FRs by using whole genome sequencing. Parental exposure to three FRs markedly increased germ cell apoptosis, and impeded oogenesis in F1-F6 offspring. In addition, the double-increased mutation frequencies observed in progeny genomes uncover the mutagenic actions of FRs on germ cells. Analysis of mutation spectra revealed that these FRs predominantly induced point mutations at A:T base pairs, whereas both small and large indels were almost unaffected. These results revealed the long-term effects of FRs on development and genomic stability of germ cells, which may pose risks to environmental organisms and human reproductive health. Taken together, our findings suggest that germ cell mutagenicity should be carefully examined for the environmental risk assessment of FRs and other emerging pollutants.
Collapse
Affiliation(s)
- Jiali Li
- Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, Anhui, China; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, CAS, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China
| | - Linglong Dai
- Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, CAS, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China; Science Island Branch, Graduate School of USTC, Hefei 230026, Anhui, China
| | - Yu Feng
- Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, CAS, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China; Science Island Branch, Graduate School of USTC, Hefei 230026, Anhui, China
| | - Zhenxiao Cao
- Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, CAS, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China; School of Environmental Science and Optoelectronic Technology, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Yuting Ding
- Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, CAS, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China; School of Environmental Science and Optoelectronic Technology, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Hao Xu
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China
| | - An Xu
- Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, CAS, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China.
| | - Hua Du
- Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, CAS, Hefei Institutes of Physical Science, CAS, Hefei 230031, Anhui, China.
| |
Collapse
|
3
|
Du H, Pan B, Alund AW, Yan J, Chen Y, Robison TW, Chen T. Evaluation of mutagenic susceptibility of different stages in germ cell development of Caenorhabditis elegans using whole genome sequencing. Arch Toxicol 2023; 97:2261-2272. [PMID: 37209179 DOI: 10.1007/s00204-023-03526-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/11/2023] [Indexed: 05/22/2023]
Abstract
In contrast to somatic mutations, mutations in germ cells affect every cell of any organism derived from the germ cell and therefore are related to numerous genetic diseases. However, there is no suitable assay to evaluate the mutagenic sensitivities of both male and female germ cells. The main type of Caenorhabditis elegans (C. elegans) is hermaphroditic, where spermatogenesis and oogenesis occur chronologically at specific stages, allowing induction of mutations in either sperm or eggs exclusively. In this study, we used the alkylating agent ethyl methanesulfonate and N-ethyl-N-nitrosourea to induce germline mutations in C. elegans at different developmental stages and analyzed mutation frequency and mutational spectrum from data gathered using next-generation sequencing (NGS) technology. Our results revealed low spontaneous mutation rates of C. elegans, along with distinct mutagenic effects elicited by the two mutagens. Our data show that the parental worms treated during germ cell mitosis, spermatogenesis, and oogenesis resulted in different mutation frequencies in their offspring, and female germ cells could be very susceptible to mutagen exposure during oogenesis. In summary, our study indicates that the use of C. elegans and its specific chronological hermaphroditism would be a promising way to explore the sensitivities of both male and female germ cells to mutagens.
Collapse
Affiliation(s)
- Hua Du
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, 230031, Anhui, People's Republic of China
| | - Bohu Pan
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Alexander W Alund
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA
- Discovery Sciences | Medicine Design, Pfizer Inc., 280 Shennecossett Rd, Groton, CT, 06340, USA
| | - Jian Yan
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| | - Ying Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| | - Timothy W Robison
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA.
| |
Collapse
|
4
|
Marchetti F, Cardoso R, Chen CL, Douglas GR, Elloway J, Escobar PA, Harper T, Heflich RH, Kidd D, Lynch AM, Myers MB, Parsons BL, Salk JJ, Settivari RS, Smith-Roe SL, Witt KL, Yauk CL, Young R, Zhang S, Minocherhomji S. Error-corrected next generation sequencing - Promises and challenges for genotoxicity and cancer risk assessment. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2023; 792:108466. [PMID: 37643677 DOI: 10.1016/j.mrrev.2023.108466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 07/12/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
Error-corrected Next Generation Sequencing (ecNGS) is rapidly emerging as a valuable, highly sensitive and accurate method for detecting and characterizing mutations in any cell type, tissue or organism from which DNA can be isolated. Recent mutagenicity and carcinogenicity studies have used ecNGS to quantify drug-/chemical-induced mutations and mutational spectra associated with cancer risk. ecNGS has potential applications in genotoxicity assessment as a new readout for traditional models, for mutagenesis studies in 3D organotypic cultures, and for detecting off-target effects of gene editing tools. Additionally, early data suggest that ecNGS can measure clonal expansion of mutations as a mechanism-agnostic early marker of carcinogenic potential and can evaluate mutational load directly in human biomonitoring studies. In this review, we discuss promising applications, challenges, limitations, and key data initiatives needed to enable regulatory testing and adoption of ecNGS - including for advancing safety assessment, augmenting weight-of-evidence for mutagenicity and carcinogenicity mechanisms, identifying early biomarkers of cancer risk, and managing human health risk from chemical exposures.
Collapse
Affiliation(s)
| | | | - Connie L Chen
- Health and Environmental Sciences Institute, Washington, DC, USA.
| | | | - Joanne Elloway
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | | | - Tod Harper
- Amgen Research, Amgen Inc, Thousand Oaks, CA, USA
| | - Robert H Heflich
- US Food and Drug Administration/National Center for Toxicological Research, Jefferson, AR, USA
| | - Darren Kidd
- Labcorp Early Development Laboratories Limited, Harrogate, North Yorkshire, UK
| | | | - Meagan B Myers
- US Food and Drug Administration/National Center for Toxicological Research, Jefferson, AR, USA
| | - Barbara L Parsons
- US Food and Drug Administration/National Center for Toxicological Research, Jefferson, AR, USA
| | | | | | | | - Kristine L Witt
- NIEHS, Division of the National Toxicology Program, Research Triangle Park, NC, USA
| | | | - Robert Young
- MilliporeSigma, Rockville, MD, USA; Current: Consultant, Bethesda, MD, USA
| | | | - Sheroy Minocherhomji
- Amgen Research, Amgen Inc, Thousand Oaks, CA, USA; Current: Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
5
|
Feng Y, Cao Z, Xu A, Du H. Evaluation of toxicity and mutagenicity of oxaliplatin on germ cells in an alternative in vivo model Caenorhabditis elegans. Food Chem Toxicol 2023:113902. [PMID: 37331561 DOI: 10.1016/j.fct.2023.113902] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
The platinum compound oxaliplatin is a widely used chemotherapeutic drug that shows a broad spectrum of activity in various human tumors. While the treatment-related side effects of oxaliplatin on directly treated individuals have been well-documented, little is known about the influence of oxaliplatin on germ cells and non-exposed progenies. Here we investigated the reproductive toxicity of oxaliplatin in a 3R-compliant in vivo model Caenorhabditis elegans, and evaluated the germ cell mutagenicity of oxaliplatin by using whole genome sequencing. Our results indicated that oxaliplatin treatment significantly disrupts development of spermatids and oocytes. By treating parental worms with oxaliplatin for three successive generations, sequencing data unveiled the clear mutagenic effects of oxaliplatin on germ cells. Analysis of genome-wide mutation spectra showed the preferentially induction of indels by oxaliplatin. In addition, we uncovered the involvement of translesion synthesis polymerase ζ in modulating mutagenic effects of oxaliplatin. These findings suggest that germ cell mutagenicity is worthy of consideration for the health risk assessment of chemotherapeutic drugs, while the combined use of alternative in vivo models and next generation sequencing technology appears to be a promising way for the preliminary safety assessment of various drugs.
Collapse
Affiliation(s)
- Yu Feng
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, CAS, Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, CAS, Hefei, 230031, Anhui, PR China; Science Island Branch, Graduate School of USTC, Hefei, 230026, Anhui, PR China
| | - Zhenxiao Cao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, CAS, Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, CAS, Hefei, 230031, Anhui, PR China; School of Environmental Science and Optoelectronic Technology, University of Science and Technology of China, Hefei, 230026, Anhui, PR China
| | - An Xu
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, CAS, Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, CAS, Hefei, 230031, Anhui, PR China.
| | - Hua Du
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, CAS, Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, Hefei Institutes of Physical Science, CAS, Hefei, 230031, Anhui, PR China.
| |
Collapse
|
6
|
Dirven Y, Eide DM, Henriksson EW, Hjorth R, Sharma AK, Graupner A, Brunborg G, Ballangby J, Boisen AMZ, Swedmark S, Gützkow KB, Olsen AK. Assessing testicular germ cell DNA damage in the comet assay; introduction of a proof-of-concept. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2023; 64:88-104. [PMID: 36629742 DOI: 10.1002/em.22527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/30/2022] [Accepted: 01/06/2023] [Indexed: 06/17/2023]
Abstract
The in vivo comet assay is widely used to measure genotoxicity; however, the current OECD test guideline (TG 489) does not recommend using the assay to assess testicular germ cells, due to the presence of testicular somatic cells. An adapted approach to specifically assess testicular germ cells within the comet assay is certainly warranted, considering regulatory needs for germ cell-specific genotoxicity data in relation to the increasing global production of and exposure to potentially hazardous chemicals. Here, we provide a proof-of-concept to selectively analyze round spermatids and primary spermatocytes, distinguishing them from other cells of the testicle. Utilizing the comet assay recordings of DNA content (total fluorescence intensity) and DNA damage (% tail intensity) of individual comets, we developed a framework to distinguish testicular cell populations based on differences in DNA content/ploidy and appearance. Haploid round spermatid comets are identified through (1) visual inspection of DNA content distributions, (2) setting DNA content thresholds, and (3) modeling DNA content distributions using a normal mixture distribution function. We also describe an approach to distinguish primary spermatocytes during comet scoring, based on their high DNA content and large physical size. Our concept allows both somatic and germ cells to be analyzed in the same animal, adding a versatile, sensitive, rapid, and resource-efficient assay to the limited genotoxicity assessment toolbox for germ cells. An adaptation of TG 489 facilitates accumulation of valuable information regarding distribution of substances to germ cells and their potential for inducing germ cell gene mutations and structural chromosomal aberrations.
Collapse
Affiliation(s)
- Yvette Dirven
- Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway
- Centre for Environmental Radioactivity (CERAD, Centre of Excellence of the Norwegian Research Council), Oslo, Norway
| | - Dag Markus Eide
- Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway
- Centre for Environmental Radioactivity (CERAD, Centre of Excellence of the Norwegian Research Council), Oslo, Norway
| | - Erika Witasp Henriksson
- Swedish Chemicals Agency, Department of Development of Legislation and Other Instruments, Unit of Proposals for Classification and Restriction, Sundbyberg, Sweden
- Swedish Chemicals Agency, Department of Development of Legislation and Other Instruments, Unit of Evaluation of Substances, Sundbyberg, Sweden
| | - Rune Hjorth
- The Danish Environmental Protection Agency, Odense, Denmark
| | - Anoop Kumar Sharma
- Technical University of Denmark, National Food Institute, Lyngby, Denmark
| | - Anne Graupner
- Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway
- Centre for Environmental Radioactivity (CERAD, Centre of Excellence of the Norwegian Research Council), Oslo, Norway
| | - Gunnar Brunborg
- Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway
- Centre for Environmental Radioactivity (CERAD, Centre of Excellence of the Norwegian Research Council), Oslo, Norway
| | - Jarle Ballangby
- Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway
- Centre for Environmental Radioactivity (CERAD, Centre of Excellence of the Norwegian Research Council), Oslo, Norway
| | | | - Stellan Swedmark
- Swedish Chemicals Agency, Department of Development of Legislation and Other Instruments, Unit of Evaluation of Substances, Sundbyberg, Sweden
| | - Kristine Bjerve Gützkow
- Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway
- Centre for Environmental Radioactivity (CERAD, Centre of Excellence of the Norwegian Research Council), Oslo, Norway
| | - Ann-Karin Olsen
- Norwegian Institute of Public Health, Division of Climate and Environmental Health, Oslo, Norway
- Centre for Environmental Radioactivity (CERAD, Centre of Excellence of the Norwegian Research Council), Oslo, Norway
| |
Collapse
|
7
|
Dos Santos A, Botelho MT, Vannuci-Silva M, Artal MC, Vacchi FI, Magalhães GR, Gomes V, Henry TB, Umbuzeiro GDA. The amphipod Parhyale hawaiensis as a promising model in ecotoxicology. CHEMOSPHERE 2022; 307:135959. [PMID: 35944683 DOI: 10.1016/j.chemosphere.2022.135959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/14/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
Near-shore marine/estuarine environments play an important role in the functioning of the marine ecosystem and are extremely vulnerable to the presence of chemical pollution. The ability to investigate the effects of pollution is limited by a lack of model organisms for which sufficient ecotoxicological information is available, and this is particularly true for tropical regions. The circumtropical marine amphipod Parhyale hawaiensis has become an important model organism in various disciplines, and here we summarize the scientific literature regarding the emergence of this model within ecotoxicology. P. hawaiensis is easily cultured in the laboratory and standardized ecotoxicity protocols have been developed and refined (e.g., miniaturized), and effects of toxicants on acute toxicity (Cd, Cu, Zn, Ag, ammonia, dyes, pesticides, environmental samples), genotoxicity as comet assay/micronuclei, and gene expression (Ag ion and Ag nanoparticles) and regeneration (pesticides) have been published. Methods for determination of internal concentrations of metals (Cu and Ag) and organic substances (synthetic dye) in hemolymph were successfully developed providing sources for the establishment of toxicokinetics models in aquatic amphipods. Protocols to evaluate reproduction and growth, for testing immune responses and DNA damage in germ cells are under way. The sensitivity of P. hawaiensis, measured as 50% lethal concentration (LC50), is in the same range as other amphipods. The combination of feasibility to culture P. hawaiensis in laboratory, the recent protocols for ecotoxicity evaluation and the rapidly expanding knowledge on its biology make it especially attractive as a model organism and promising tool for risk assessment evaluations in tropical environments.
Collapse
Affiliation(s)
- Amanda Dos Santos
- School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil; School of Technology, University of Campinas, Limeira, SP, Brazil
| | | | | | | | - Francine I Vacchi
- School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil; School of Technology, University of Campinas, Limeira, SP, Brazil; Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | | | - Vicente Gomes
- Oceanographic Institute, University of São Paulo, São Paulo, SP, Brazil
| | - Theodore Burdick Henry
- Institute of Life and Earth Sciences, School of Energy, Geoscience, Infrastructure, and Society, Heriot-Watt University, Edinburgh, Scotland, UK; Department of Forestry Wildlife and Fisheries, and Center for Environmental Biotechnology, The University of Tennessee, Knoxville, TN, USA
| | - Gisela de Aragão Umbuzeiro
- School of Technology, University of Campinas, Limeira, SP, Brazil; Institute of Biology, University of Campinas, Campinas, SP, Brazil.
| |
Collapse
|
8
|
Lupski JR. Clan genomics: From OMIM phenotypic traits to genes and biology. Am J Med Genet A 2021; 185:3294-3313. [PMID: 34405553 PMCID: PMC8530976 DOI: 10.1002/ajmg.a.62434] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/29/2021] [Accepted: 07/04/2021] [Indexed: 12/20/2022]
Abstract
Clinical characterization of a patient phenotype has been the quintessential approach for elucidating a differential diagnosis and a hypothesis to explore a potential clinical diagnosis. This has resulted in a language of medicine and a semantic ontology, with both specialty- and subspecialty-specific lexicons, that can be challenging to translate and interpret. There is no 'Rosetta Stone' of clinical medicine such as the genetic code that can assist translation and interpretation of the language of genetics. Nevertheless, the information content embodied within a clinical diagnosis can guide management, therapeutic intervention, and potentially prognostic outlook of disease enabling anticipatory guidance for patients and families. Clinical genomics is now established firmly in medical practice. The granularity and informative content of a personal genome is immense. Yet, we are limited in our utility of much of that personal genome information by the lack of functional characterization of the overwhelming majority of computationally annotated genes in the haploid human reference genome sequence. Whereas DNA and the genetic code have provided a 'Rosetta Stone' to translate genetic variant information, clinical medicine, and clinical genomics provide the context to understand human biology and disease. A path forward will integrate deep phenotyping, such as available in a clinical synopsis in the Online Mendelian Inheritance in Man (OMIM) entries, with personal genome analyses.
Collapse
Affiliation(s)
- James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
9
|
Comparison of the frequencies of ENU-induced point mutations in male germ cells and inherited germline mutations in their offspring. Genes Environ 2021; 43:43. [PMID: 34627396 PMCID: PMC8501628 DOI: 10.1186/s41021-021-00216-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/15/2021] [Indexed: 11/21/2022] Open
Abstract
Background Gene mutations induced in germ cells may be transmitted to the next generation and cause adverse effects such as genetic diseases. Certain mutations may result in infertility or death in early development. Thus, the mutations may not be inheritable. However, the extent to which point mutations in male germ cells are transmitted to the next generation or eliminated during transmission is largely unknown. This study compared mutation frequencies (MFs) in sperm of N-ethyl-N-nitrosourea (ENU)-treated gpt delta mice and de novo MFs in the whole exome/genome of their offspring. Results Male gpt delta mice were treated with 10, 30, and 85 mg/kg of ENU (i.p., weekly × 2) and mated with untreated females to generate offspring. We previously reported a dose-dependent increase in de novo MFs in the offspring estimated by whole exome sequencing (WES) (Mutat. Res., 810, 30–39, 2016). In this study, gpt MFs in the sperm of ENU-treated mice were estimated, and the MFs per reporter gene were converted to MFs per base pair. The inherited de novo MFs in the offspring (9, 26 and 133 × 10− 8/bp for 10, 30, and 85 mg/kg ENU-treated groups, respectively) were comparable to those of the converted gpt MFs in the sperm of ENU-treated fathers (6, 16, and 69 × 10− 8/bp). It indicated that the gpt MFs in the ENU-treated father’s sperm were comparable to the inherited de novo MFs in the offspring as estimated by WES. In addition, de novo MFs in the offspring of 10 mg/kg ENU-treated and control fathers were estimated by whole genome sequencing (WGS), because WES was not sufficiently sensitive to detect low background MF. The de novo MF in the offspring of the ENU-treated fathers was 6 × 10− 8/bp and significantly higher than that of the control (2 × 10− 8/bp). There were no significant differences in de novo MFs between gene-coding and non-coding regions. WGS analysis was able to detect ENU-induced characteristic de novo base substitutions at a low dose group. Conclusions Despite a difference between exome/genome and exogenous reporter genes, the results indicated that ENU-induced point mutations in male germ cells could be transmitted to the next generation without severe selection. Supplementary Information The online version contains supplementary material available at 10.1186/s41021-021-00216-z.
Collapse
|
10
|
Choudhuri S, Kaur T, Jain S, Sharma C, Asthana S. A review on genotoxicity in connection to infertility and cancer. Chem Biol Interact 2021; 345:109531. [PMID: 34058178 DOI: 10.1016/j.cbi.2021.109531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/22/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
Genotoxicity has been identified as the main cause of infertility and a variety of cancers. The mechanisms affect the structure, quality of the information or the segregation of DNA and are not inherently correlated with mutagenicity. The concept of genotoxicity, the chemical classes that cause genetic damage and the associated mechanisms of action are discussed here. Hazardous effects of pharmaceuticals, cosmetics, agrochemicals, industrial compounds, food additives, natural toxins and nanomaterials are, in large part, identified by genotoxicity and mutagenicity tests. These are critical and early steps in industrial and regulatory health assessment. Though several in vitro experiments are commonly used and approval by regulatory agencies for commercial licensing of drugs, their accuracy in human predictions for genotoxic and mutagenic effects is frequently questioned. Treatment of real and functional genetic toxicity problems depends in detail on the knowledge of mechanisms of DNA damage in the molecular, subcellular, cellular and tissue or organ system levels. Current strategies for risk assessment of human health need revisions to achieve robust and reliable results for optimizing their effectiveness. Additionally, computerized methods, neo-biomarkers leveraging '-omics' approaches, all of which can provide a convincing genotoxicity evaluation to reduce infertility and cancer risk.
Collapse
Affiliation(s)
- Sharmistha Choudhuri
- Department of Biochemistry, R. G. Kar Medical College and Hospital, Kolkata, West Bengal, India
| | - Taruneet Kaur
- Animal Biochemistry Division, National Dairy Research Institute, Karnal, Haryana, India
| | - Sapna Jain
- Multidisciplinary Clinical Translational Research, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Chandresh Sharma
- Multidisciplinary Clinical Translational Research, Translational Health Science and Technology Institute, Faridabad, Haryana, India.
| | - Shailendra Asthana
- Non-Communicable Disease, Translational Health Science and Technology Institute, Faridabad, Haryana, India.
| |
Collapse
|
11
|
Anwar S, Ansari SA, Alamri A, Alamri A, Alqarni A, Alghamdi S, Wagih ME, Ahmad A, Rengasamy KR. Clastogenic, anti-clastogenic profile and safety assessment of Camel urine towards the development of new drug target. Food Chem Toxicol 2021; 151:112131. [PMID: 33737110 DOI: 10.1016/j.fct.2021.112131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/28/2022]
Abstract
Camel Urine (CU) is composed of components that have antitumor properties and other therapeutic benefits. Regardless of short-term preliminary CU genotoxicity is reported, comprehensive genotoxic studies are limited. In this study, sensitive in vitro and in vivo genotoxic bioassays such as mitotic index (MI), chromosomal aberrations (CA), micronucleated polychromatic erythrocytes (MPE), and analysis of primary spermatocytes were employed. The adventitious roots of Allium cepa L. and mice (Mus musculus), as an experimental mammalian system, were employed to assess the MI and CA of CU induced by sodium nitrate and cyclophosphamide respectively. In contrast, other clastogenic assays were studied in mice (Mus musculus). Twenty-eight days of four repeated doses (2.5, 5, 25, and 50 mL/kg BW) of CU were tested and compared with three doses (10, 25, and 50 mg/kg BW) cyclophosphamide as a positive control and deionized water as the negative control. The results proved that cytological examination of CU was cytotoxic since a decrease in mitotic activity (16.8-1.1) was observed, since the significant reduction in cell proliferation in A. cepa L. and also in mice bone marrow cells. On the other hand, CU did not induce a clastogenic effect since no significant stickiness, fragment, multinucleoli were observed compared to the control group. Additionally, the data showed that CU decreased the CA when mice had received cyclophosphamide (25 mg BW) followed by CU doses. CU was found to be cytotoxic but no clastogenic effect. Furthermore, it possesses anticlastogenic properties. The observed results suggest that CU in whole or the metabolites present in CU could be a potent drug target. Further research is warranted to study the complete metabolites profiling and to study the molecular mechanisms.
Collapse
Affiliation(s)
- Sirajudheen Anwar
- Pharmacology and Toxicology Department, College of Pharmacy, University of Hail, Hail, 81451, Saudi Arabia.
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, PO Box 2457, Riyadh, 11451, Saudi Arabia
| | - Abdulwahab Alamri
- Pharmacology and Toxicology Department, College of Pharmacy, University of Hail, Hail, 81451, Saudi Arabia
| | - Abdulhakeem Alamri
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, Taif University, P. O. Box 11099, Taif, 21944, Saudi Arabia
| | - Aali Alqarni
- Pharmacology and Toxicology Unit, Department of Pharmaceutical Chemistry, Faculty of Clinical Pharmacy, Albaha University, Albaha, 65431, Saudi Arabia
| | - Saleh Alghamdi
- Department of Clinical Pharmacy, Faculty of Clinical Pharmacy, Albaha University, Albaha, 65431, Saudi Arabia
| | - Mohamed E Wagih
- Canadian Academy of Sciences, Toronto, Ontario, M5S1Z6, Canada
| | - Akbar Ahmad
- Genetic and Invitro Toxicology, Charles River Laboratories, Greater Chicago Area, IL, USA
| | - Kannan Rr Rengasamy
- Green Biotechnologies Research Centre of Excellence, University of Limpopo, Private Bag X1106, Polokwane, Sovenga, 0727, South Africa.
| |
Collapse
|
12
|
Bline AP, Dearfield KL, DeMarini DM, Marchetti F, Yauk CL, Escher J. Heritable hazards of smoking: Applying the "clean sheet" framework to further science and policy. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:910-921. [PMID: 33064321 PMCID: PMC7756471 DOI: 10.1002/em.22412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/26/2020] [Accepted: 10/11/2020] [Indexed: 05/06/2023]
Abstract
All the cells in our bodies are derived from the germ cells of our parents, just as our own germ cells become the bodies of our children. The integrity of the genetic information inherited from these germ cells is of paramount importance in establishing the health of each generation and perpetuating our species into the future. There is a large and growing body of evidence strongly suggesting the existence of substances that may threaten this integrity by acting as human germ cell mutagens. However, there generally are no absolute regulatory requirements to test agents for germ cell effects. In addition, the current regulatory testing paradigms do not evaluate the impacts of epigenetically mediated intergenerational effects, and there is no regulatory framework to apply new and emerging tests in regulatory decision making. At the 50th annual meeting of the Environmental Mutagenesis and Genomics Society held in Washington, DC, in September 2019, a workshop took place that examined the heritable effects of hazardous exposures to germ cells, using tobacco smoke as the example hazard. This synopsis provides a summary of areas of concern regarding heritable hazards from tobacco smoke exposures identified at the workshop and the value of the Clean Sheet framework in organizing information to address knowledge and testing gaps.
Collapse
Affiliation(s)
- Abigail P. Bline
- Fielding School of Public HealthUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | | | | | - Francesco Marchetti
- Environmental Health Science Research Bureau, Health CanadaOttawaOntarioCanada
| | - Carole L. Yauk
- Department of BiologyUniversity of OttawaOttawaOntarioCanada
| | | | | |
Collapse
|
13
|
Pligina KL, Zhanataev AK, Anisina EA, Daugel-Dauge NO, Durnev AD. Comet assay on one- and two-cell mouse embryos. Toxicol Lett 2020; 331:124-129. [PMID: 32534006 DOI: 10.1016/j.toxlet.2020.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/30/2020] [Accepted: 06/04/2020] [Indexed: 10/24/2022]
Abstract
DNA damage quantified as the comet tail length was assessed using in vitro and in vivo comet assay on one- and two-cell mouse embryos obtained by natural mating. The use of a protocol with three layers of agarose reduces the embryo loss and makes it possible to study a small number of embryos. A significantly lower level of basal, but not induced DNA damage was found in embryos with cleaved zona pellucida compared to embryos with intact zona pellucida. There were no significant differences in the length of the comet's tail between embryos lysed in different lysis solutions, both in cases of basal and induced DNA damage. A significant increase in the comet tail length was detected in one-cell embryos of mice treated with methyl methanesulfonate and etoposide compared to the control. The data show that DNA damage induced in maternal germ cells persists, which can be detected in embryos using the comet assay.
Collapse
Affiliation(s)
- Kira L Pligina
- Laboratory of Pharmacology and Mutagenesis, FSBI Zakusov Research Institute of Pharmacology, Baltiyskaya str. 8, 125315, Moscow, Russia.
| | - Aliy K Zhanataev
- Laboratory of Pharmacology and Mutagenesis, FSBI Zakusov Research Institute of Pharmacology, Baltiyskaya str. 8, 125315, Moscow, Russia.
| | - Elena A Anisina
- Laboratory of Pharmacology and Mutagenesis, FSBI Zakusov Research Institute of Pharmacology, Baltiyskaya str. 8, 125315, Moscow, Russia.
| | - Natalya O Daugel-Dauge
- Laboratory of Pharmacology and Mutagenesis, FSBI Zakusov Research Institute of Pharmacology, Baltiyskaya str. 8, 125315, Moscow, Russia.
| | - Andrey D Durnev
- Laboratory of Pharmacology and Mutagenesis, FSBI Zakusov Research Institute of Pharmacology, Baltiyskaya str. 8, 125315, Moscow, Russia.
| |
Collapse
|
14
|
Hartwig A, Arand M, Epe B, Guth S, Jahnke G, Lampen A, Martus HJ, Monien B, Rietjens IMCM, Schmitz-Spanke S, Schriever-Schwemmer G, Steinberg P, Eisenbrand G. Mode of action-based risk assessment of genotoxic carcinogens. Arch Toxicol 2020; 94:1787-1877. [PMID: 32542409 PMCID: PMC7303094 DOI: 10.1007/s00204-020-02733-2] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 03/31/2020] [Indexed: 12/16/2022]
Abstract
The risk assessment of chemical carcinogens is one major task in toxicology. Even though exposure has been mitigated effectively during the last decades, low levels of carcinogenic substances in food and at the workplace are still present and often not completely avoidable. The distinction between genotoxic and non-genotoxic carcinogens has traditionally been regarded as particularly relevant for risk assessment, with the assumption of the existence of no-effect concentrations (threshold levels) in case of the latter group. In contrast, genotoxic carcinogens, their metabolic precursors and DNA reactive metabolites are considered to represent risk factors at all concentrations since even one or a few DNA lesions may in principle result in mutations and, thus, increase tumour risk. Within the current document, an updated risk evaluation for genotoxic carcinogens is proposed, based on mechanistic knowledge regarding the substance (group) under investigation, and taking into account recent improvements in analytical techniques used to quantify DNA lesions and mutations as well as "omics" approaches. Furthermore, wherever possible and appropriate, special attention is given to the integration of background levels of the same or comparable DNA lesions. Within part A, fundamental considerations highlight the terms hazard and risk with respect to DNA reactivity of genotoxic agents, as compared to non-genotoxic agents. Also, current methodologies used in genetic toxicology as well as in dosimetry of exposure are described. Special focus is given on the elucidation of modes of action (MOA) and on the relation between DNA damage and cancer risk. Part B addresses specific examples of genotoxic carcinogens, including those humans are exposed to exogenously and endogenously, such as formaldehyde, acetaldehyde and the corresponding alcohols as well as some alkylating agents, ethylene oxide, and acrylamide, but also examples resulting from exogenous sources like aflatoxin B1, allylalkoxybenzenes, 2-amino-3,8-dimethylimidazo[4,5-f] quinoxaline (MeIQx), benzo[a]pyrene and pyrrolizidine alkaloids. Additionally, special attention is given to some carcinogenic metal compounds, which are considered indirect genotoxins, by accelerating mutagenicity via interactions with the cellular response to DNA damage even at low exposure conditions. Part C finally encompasses conclusions and perspectives, suggesting a refined strategy for the assessment of the carcinogenic risk associated with an exposure to genotoxic compounds and addressing research needs.
Collapse
Affiliation(s)
- Andrea Hartwig
- Department of Food Chemistry and Toxicology, Institute of Applied Biosciences (IAB), Karlsruhe Institute of Technology (KIT), Adenauerring 20a, 76131, Karlsruhe, Germany.
| | - Michael Arand
- Institute of Pharmacology and Toxicology, University of Zurich, 8057, Zurich, Switzerland
| | - Bernd Epe
- Institute of Pharmacy and Biochemistry, University of Mainz, 55099, Mainz, Germany
| | - Sabine Guth
- Department of Toxicology, IfADo-Leibniz Research Centre for Working Environment and Human Factors, TU Dortmund, Ardeystr. 67, 44139, Dortmund, Germany
| | - Gunnar Jahnke
- Department of Food Chemistry and Toxicology, Institute of Applied Biosciences (IAB), Karlsruhe Institute of Technology (KIT), Adenauerring 20a, 76131, Karlsruhe, Germany
| | - Alfonso Lampen
- Department of Food Safety, German Federal Institute for Risk Assessment (BfR), 10589, Berlin, Germany
| | - Hans-Jörg Martus
- Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| | - Bernhard Monien
- Department of Food Safety, German Federal Institute for Risk Assessment (BfR), 10589, Berlin, Germany
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Simone Schmitz-Spanke
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, University of Erlangen-Nuremberg, Henkestr. 9-11, 91054, Erlangen, Germany
| | - Gerlinde Schriever-Schwemmer
- Department of Food Chemistry and Toxicology, Institute of Applied Biosciences (IAB), Karlsruhe Institute of Technology (KIT), Adenauerring 20a, 76131, Karlsruhe, Germany
| | - Pablo Steinberg
- Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Haid-und-Neu-Str. 9, 76131, Karlsruhe, Germany
| | - Gerhard Eisenbrand
- Retired Senior Professor for Food Chemistry and Toxicology, Kühler Grund 48/1, 69126, Heidelberg, Germany.
| |
Collapse
|
15
|
Godschalk RWL, Yauk CL, van Benthem J, Douglas GR, Marchetti F. In utero Exposure to Genotoxicants Leading to Genetic Mosaicism: An Overlooked Window of Susceptibility in Genetic Toxicology Testing? ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:55-65. [PMID: 31743493 PMCID: PMC6973016 DOI: 10.1002/em.22347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/14/2019] [Accepted: 11/17/2019] [Indexed: 05/08/2023]
Abstract
In utero development represents a sensitive window for the induction of mutations. These mutations may subsequently expand clonally to populate entire organs or anatomical structures. Although not all adverse mutations will affect tissue structure or function, there is growing evidence that clonally expanded genetic mosaics contribute to various monogenic and complex diseases, including cancer. We posit that genetic mosaicism is an underestimated potential health problem that is not fully addressed in the current regulatory genotoxicity testing paradigm. Genotoxicity testing focuses exclusively on adult exposures and thus may not capture the complexity of genetic mosaicisms that contribute to human disease. Numerous studies have shown that conversion of genetic damage into mutations during early developmental exposures can result in much higher mutation burdens than equivalent exposures in adults in certain tissues. Therefore, we assert that analysis of genetic effects caused by in utero exposures should be considered in the current regulatory testing paradigm, which is possible by harmonization with current reproductive/developmental toxicology testing strategies. This is particularly important given the recent proposed paradigm change from simple hazard identification to quantitative mutagenicity assessment. Recent developments in sequencing technologies offer practical tools to detect mutations in any tissue or species. In addition to mutation frequency and spectrum, these technologies offer the opportunity to characterize the extent of genetic mosaicism following exposure to mutagens. Such integration of new methods with existing toxicology guideline studies offers the genetic toxicology community a way to modernize their testing paradigm and to improve risk assessment for vulnerable populations. Environ. Mol. Mutagen. 61:55-65, 2020. © 2019 The Authors. Environmental and Molecular Mutagenesis published by Wiley Periodicals, Inc. on behalf of Environmental Mutagen Society.
Collapse
Affiliation(s)
- Roger W. L. Godschalk
- Department of Pharmacology and Toxicology, School for Nutrition and Translational Research in Metabolism (NUTRIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Carole L. Yauk
- Mechanistic Studies DivisionEnvironmental Health Science and Research Bureau, Health CanadaOttawaK1A 0K9OntarioCanada
| | - Jan van Benthem
- Center for Health ProtectionNational Institute for Public Health and the Environment (RIVM)BilthovenThe Netherlands
| | - George R. Douglas
- Mechanistic Studies DivisionEnvironmental Health Science and Research Bureau, Health CanadaOttawaK1A 0K9OntarioCanada
| | - Francesco Marchetti
- Mechanistic Studies DivisionEnvironmental Health Science and Research Bureau, Health CanadaOttawaK1A 0K9OntarioCanada
| |
Collapse
|
16
|
Heflich RH, Johnson GE, Zeller A, Marchetti F, Douglas GR, Witt KL, Gollapudi BB, White PA. Mutation as a Toxicological Endpoint for Regulatory Decision-Making. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:34-41. [PMID: 31600846 DOI: 10.1002/em.22338] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/09/2019] [Accepted: 09/14/2019] [Indexed: 05/23/2023]
Abstract
Mutations induced in somatic cells and germ cells are responsible for a variety of human diseases, and mutation per se has been considered an adverse health concern since the early part of the 20th Century. Although in vitro and in vivo somatic cell mutation data are most commonly used by regulatory agencies for hazard identification, that is, determining whether or not a substance is a potential mutagen and carcinogen, quantitative mutagenicity dose-response data are being used increasingly for risk assessments. Efforts are currently underway to both improve the measurement of mutations and to refine the computational methods used for evaluating mutation data. We recommend continuing the development of these approaches with the objective of establishing consensus regarding the value of including the quantitative analysis of mutation per se as a required endpoint for comprehensive assessments of toxicological risk. Environ. Mol. Mutagen. 61:34-41, 2020. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Robert H Heflich
- U.S. Food and Drug Administration, National Center for Toxicological Research, Jefferson, Arkansas
| | | | - Andreas Zeller
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Francesco Marchetti
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| | - George R Douglas
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| | - Kristine L Witt
- National Institutes of Health, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | | | - Paul A White
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| |
Collapse
|
17
|
DeMarini DM. The mutagenesis moonshot: The propitious beginnings of the environmental mutagenesis and genomics society. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:8-24. [PMID: 31294870 PMCID: PMC6949362 DOI: 10.1002/em.22313] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 06/20/2019] [Indexed: 05/05/2023]
Abstract
A mutagenesis moonshot addressing the influence of the environment on our genetic wellbeing was launched just 2 months before astronauts landed on the moon. Its impetus included the discovery that X-rays (Muller HJ. [1927]: Science 64:84-87) and chemicals (Auerbach and Robson. [1946]: Nature 157:302) were germ-cell mutagens, the introduction of a growing number of untested chemicals into the environment after World War II, and an increasing awareness of the role of environmental pollution on human health. Due to mounting concern from influential scientists that germ-cell mutagens might be ubiquitous in the environment, Alexander Hollaender and colleagues founded in 1969 the Environmental Mutagen Society (EMS), now the Environmental Mutagenesis and Genomics Society (EMGS); Frits Sobels founded the European EMS in 1970. As Fred de Serres noted, such societies were necessary because protecting populations from environmental mutagens could not be addressed by existing scientific societies, and new multidisciplinary alliances were required to spearhead this movement. The nascent EMS gathered policy makers and scientists from government, industry, and academia who became advocates for laws requiring genetic toxicity testing of pesticides and drugs and helped implement those laws. They created an electronic database of the mutagenesis literature; established a peer-reviewed journal; promoted basic and applied research in DNA repair and mutagenesis; and established training programs that expanded the science worldwide. Despite these successes, one objective remains unfulfilled: identification of human germ-cell mutagens. After 50 years, the voyage continues, and a vibrant EMGS is needed to bring the mission to its intended target of protecting populations from genetic hazards. Environ. Mol. Mutagen. 61:8-24, 2020. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David M. DeMarini
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| |
Collapse
|
18
|
Marchetti F, Douglas GR, Yauk CL. A Return to the Origin of the EMGS: Rejuvenating the Quest for Human Germ Cell Mutagens and Determining the Risk to Future Generations. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:42-54. [PMID: 31472026 DOI: 10.1002/em.22327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/21/2019] [Accepted: 08/28/2019] [Indexed: 05/23/2023]
Abstract
Fifty years ago, the Environmental Mutagen Society (now Environmental Mutagenesis and Genomics Society) was founded with a laser-focus on germ cell mutagenesis and the protection of "our most vital assets"-the sperm and egg genomes. Yet, five decades on, despite the fact that many agents have been demonstrated to induce inherited changes in the offspring of exposed laboratory rodents, there is no consensus on whether human germ cell mutagens exist. We argue that it is time to reevaluate the available data and conclude that we already have evidence for the existence of environmental exposures that impact human germ cells. What is missing are definite data to demonstrate a significant increase in de novo mutations in the offspring of exposed parents. We believe that with over two decades of research advancing knowledge and technologies in genomics, we are at the cusp of generating data to conclusively show that environmental exposures cause heritable de novo changes in the human offspring. We call on the research community to harness our technologies, synergize our efforts, and return to our Founders' original focus. The next 50 years must involve collaborative work between clinicians, epidemiologists, genetic toxicologists, genomics experts and bioinformaticians to precisely define how environmental exposures impact germ cell genomes. It is time for the research and regulatory communities to prepare to interpret the coming outpouring of data and develop a framework for managing, communicating and mitigating the risk of exposure to human germ cell mutagens. Environ. Mol. Mutagen. 61:42-54, 2020. © 2019 Her Majesty the Queen in Right of Canada.
Collapse
Affiliation(s)
- Francesco Marchetti
- Environmental Health Science Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - George R Douglas
- Environmental Health Science Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Carole L Yauk
- Environmental Health Science Research Bureau, Health Canada, Ottawa, Ontario, Canada
| |
Collapse
|
19
|
Mišík M, Filipic M, Nersesyan A, Kundi M, Isidori M, Knasmueller S. Environmental risk assessment of widely used anticancer drugs (5-fluorouracil, cisplatin, etoposide, imatinib mesylate). WATER RESEARCH 2019; 164:114953. [PMID: 31404901 DOI: 10.1016/j.watres.2019.114953] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/31/2019] [Accepted: 08/03/2019] [Indexed: 05/21/2023]
Abstract
Anticancer drugs are among the most toxic chemicals, which are commercially produced; therefore, their release in aquatic ecosystems raised concerns in regard to potential adverse effects. This article describes the results of risk assessments concerning their environmental safety, which are based on data generated in the frame of a coordinated EU project ("Cytothreat"). Eight research institutions participated in the project and four widely used anticancer drugs with different mechanisms of therapeutic action (5-fluorouracil 5FU, cisplatin CDDP, imatinib mesylate IM and etoposide ET) were tested in a variety of indicator organisms (cyanobacteria, algae, higher plants, rotifers, crustacea, fish and also in human and fish derived cell lines) in acute/subacute/chronic toxicity assays. Furthermore, genotoxic effects in micronucleus assays, single cell gel electrophoresis experiments and γH2AX tests were studied in plants, crustacea, fish and in various cell lines. We used the results to calculate the predicted no effect concentrations (PNEC) and risk quotients (RQ) by comparing PNEC with predicted environmental concentrations (PEC values) and measured concentrations (MEC) in wastewaters. The most sensitive species in experiments concerning acute toxic and long term effects were in general crustacea (daphnids) after chronic treatment the most pronounced effects were detected with IM followed by CDDP and 5FU. Comparisons between PNEC and PEC values indicate that it is unlikely that the release of these drugs in the aquatic environments leads to adverse effects (RQ values < 1). However, when the assessments were performed with MEC found in highly contaminated municipal wastewaters and hospital effluents, RQ values were obtained which are indicative for moderate adverse effects of IM. Calculations with data from genotoxicity experiments and PEC values are indicative for increased RQ values for all compounds except ET. The most sensitive species were fish (Danio rerio) which were highly responsive towards 5FU and daphnids which were sensitive towards CDDP and IM. When environmental data (from waste waters) were used for the calculations, high RQ values (>100) were obtained for CDDP and IM. These overall conclusions were not substantially altered when the effects of other frequently used cytostatic drugs and combined effects of mixtures of anticancer drugs were taken into consideration. The results of these assessments underline the importance of efficient removal of these chemicals by improved sewage treatment strategies and the need for further investigations of adverse the long term effects of cytostatics in aquatic biota as a consequence of damage of the genetic material in highly sensitive species.
Collapse
Affiliation(s)
- Miroslav Mišík
- Institute of Cancer Research, Department of Internal Medicine I, Borschkegasse 8a, Vienna, 1090, Austria
| | - Metka Filipic
- National Institute of Biology, Department for Genetic Toxicology and Cancer Biology, Večna pot 111, SI-1000, Ljubljana, Slovenia
| | - Armen Nersesyan
- Institute of Cancer Research, Department of Internal Medicine I, Borschkegasse 8a, Vienna, 1090, Austria
| | - Michael Kundi
- Center for Public Health, Department of Environmental Health, Medical University of Vienna, Vienna, Austria
| | - Marina Isidori
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università della Campania "Luigi Vanvitelli", Via Vivaldi 43, I-81100, Caserta, Italy
| | - Siegfried Knasmueller
- Institute of Cancer Research, Department of Internal Medicine I, Borschkegasse 8a, Vienna, 1090, Austria.
| |
Collapse
|
20
|
Inselman A, Liu F, Wang C, Shi Q, Pang L, Mattes W, White M, Lyn-Cook B, Rosas-Hernandez H, Cuevas E, Lantz S, Imam S, Ali S, Petibone DM, Shemansky JM, Xiong R, Wang Y, Tripathi P, Cao X, Heflich RH, Slikker W. Dr. Daniel Acosta and In Vitro toxicology at the U.S. Food and Drug Administration's National Center for Toxicological Research. Toxicol In Vitro 2019; 64:104471. [PMID: 31628011 DOI: 10.1016/j.tiv.2019.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/01/2019] [Accepted: 03/04/2019] [Indexed: 10/25/2022]
Abstract
For the past five years, Dr. Daniel Acosta has served as the Deputy Director of Research at the National Center for Toxicological Research (NCTR), a principle research laboratory of the U.S. Food and Drug Administration (FDA). Over his career at NCTR, Dr. Acosta has had a major impact on developing and promoting the use of in vitro assays in regulatory toxicity and product safety assessments. As Dr. Acosta nears his retirement we have dedicated this paper to his many accomplishments at the NCTR. Described within this paper are some of the in vitro studies that have been conducted under Dr. Acosta's leadership. These studies include toxicological assessments involving developmental effects, and the development and application of in vitro reproductive, heart, liver, neurological and airway cell and tissue models.
Collapse
Affiliation(s)
- Amy Inselman
- Division of Systems Biology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Fang Liu
- Division of Neurotoxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Cheng Wang
- Division of Neurotoxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Qiang Shi
- Division of Systems Biology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Li Pang
- Division of Systems Biology, NCTR, FDA, Jefferson, AR 72079, USA
| | - William Mattes
- Division of Systems Biology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Matthew White
- Arkansas College of Osteopathic Medicine, Fort Smith, AR 72916, USA
| | - Beverly Lyn-Cook
- Division of Biochemical Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | | | - Elvis Cuevas
- Division of Neurotoxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Susan Lantz
- Division of Neurotoxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Syed Imam
- Division of Neurotoxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Syed Ali
- Division of Neurotoxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Dayton M Petibone
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Jennifer M Shemansky
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Rui Xiong
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Yiying Wang
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Priya Tripathi
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Xuefei Cao
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | - Robert H Heflich
- Division of Genetic and Molecular Toxicology, NCTR, FDA, Jefferson, AR 72079, USA
| | | |
Collapse
|
21
|
Hovhannisyan G, Harutyunyan T, Aroutiounian R, Liehr T. DNA Copy Number Variations as Markers of Mutagenic Impact. Int J Mol Sci 2019; 20:ijms20194723. [PMID: 31554154 PMCID: PMC6801639 DOI: 10.3390/ijms20194723] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 12/26/2022] Open
Abstract
DNA copy number variation (CNV) occurs due to deletion or duplication of DNA segments resulting in a different number of copies of a specific DNA-stretch on homologous chromosomes. Implications of CNVs in evolution and development of different diseases have been demonstrated although contribution of environmental factors, such as mutagens, in the origin of CNVs, is poorly understood. In this review, we summarize current knowledge about mutagen-induced CNVs in human, animal and plant cells. Differences in CNV frequencies induced by radiation and chemical mutagens, distribution of CNVs in the genome, as well as adaptive effects in plants, are discussed. Currently available information concerning impact of mutagens in induction of CNVs in germ cells is presented. Moreover, the potential of CNVs as a new endpoint in mutagenicity test-systems is discussed.
Collapse
Affiliation(s)
- Galina Hovhannisyan
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, 0025 Yerevan, Armenia.
| | - Tigran Harutyunyan
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, 0025 Yerevan, Armenia.
| | - Rouben Aroutiounian
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, 0025 Yerevan, Armenia.
| | - Thomas Liehr
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, D-07747 Jena, Germany.
| |
Collapse
|
22
|
Beal MA, Meier MJ, Williams A, Rowan-Carroll A, Gagné R, Lindsay SJ, Fitzgerald T, Hurles ME, Marchetti F, Yauk CL. Paternal exposure to benzo(a)pyrene induces genome-wide mutations in mouse offspring. Commun Biol 2019; 2:228. [PMID: 31240266 PMCID: PMC6586636 DOI: 10.1038/s42003-019-0476-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 05/23/2019] [Indexed: 01/14/2023] Open
Abstract
Understanding the effects of environmental exposures on germline mutation rates has been a decades-long pursuit in genetics. We used next-generation sequencing and comparative genomic hybridization arrays to investigate genome-wide mutations in the offspring of male mice exposed to benzo(a)pyrene (BaP), a common environmental pollutant. We demonstrate that offspring developing from sperm exposed during the mitotic or post-mitotic phases of spermatogenesis have significantly more de novo single nucleotide variants (1.8-fold; P < 0.01) than controls. Both phases of spermatogenesis are susceptible to the induction of heritable mutations, although mutations arising from post-fertilization events are more common after post-mitotic exposure. In addition, the mutation spectra in sperm and offspring of BaP-exposed males are consistent. Finally, we report a significant increase in transmitted copy number duplications (P = 0.001) in BaP-exposed sires. Our study demonstrates that germ cell mutagen exposures induce genome-wide mutations in the offspring that may be associated with adverse health outcomes.
Collapse
Affiliation(s)
- Marc A. Beal
- Carleton University, Ottawa, Ontario K1S 5B6 Canada
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario K1A 0K9 Canada
| | - Matthew J. Meier
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario K1A 0K9 Canada
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario K1A 0K9 Canada
| | - Andrea Rowan-Carroll
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario K1A 0K9 Canada
| | - Rémi Gagné
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario K1A 0K9 Canada
| | - Sarah J. Lindsay
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA UK
| | - Tomas Fitzgerald
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA UK
| | - Matthew E. Hurles
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA UK
| | - Francesco Marchetti
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario K1A 0K9 Canada
| | - Carole L. Yauk
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario K1A 0K9 Canada
| |
Collapse
|
23
|
Kirkland D, Uno Y, Luijten M, Beevers C, van Benthem J, Burlinson B, Dertinger S, Douglas GR, Hamada S, Horibata K, Lovell DP, Manjanatha M, Martus HJ, Mei N, Morita T, Ohyama W, Williams A. In vivo genotoxicity testing strategies: Report from the 7th International workshop on genotoxicity testing (IWGT). MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2019; 847:403035. [PMID: 31699340 DOI: 10.1016/j.mrgentox.2019.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/13/2019] [Accepted: 03/23/2019] [Indexed: 12/14/2022]
Abstract
The working group reached complete or majority agreement on many issues. Results from TGR and in vivo comet assays for 91 chemicals showed they have similar ability to detect in vivo genotoxicity per se with bacterial mutagens and Ames-positive carcinogens. TGR and comet assay results were not significantly different when compared with IARC Group 1, 2 A, and unclassified carcinogens. There were significantly more comet assay positive responses for Group 2B chemicals, and for IARC classified and unclassified carcinogens combined, which may be expected since mutation is a sub-set of genotoxicity. A liver comet assay combined with the bone marrow/blood micronucleus (MNviv) test would detect in vivo genotoxins that do not exhibit tissue-specific or site-of-contact effects, and is appropriate for routine in vivo genotoxicity testing. Generally for orally administered substances, a comet assay at only one site-of-contact GI tract tissue (stomach or duodenum/jejunum) is required. In MNviv tests, evidence of target tissue exposure can be obtained in a number of different ways, as recommended by ICH S2(R1) and EFSA (Hardy et al., 2017). Except for special cases the i.p. route is inappropriate for in vivo testing; for risk evaluations more weight should be given to data from a physiologically relevant administration route. The liver MN test is sufficiently validated for the development of an OECD guideline. However, the impact of dosing animals >6 weeks of age needs to be evaluated. The GI tract MN test shows promise but needs more validation for an OECD guideline. The Pig-a assay detects systemically available mutagens and is a valuable follow-up to in vitro positive results. A new freeze-thaw protocol provides more flexibility. Mutant reticulocyte and erythrocyte frequencies should both be determined. Preliminary data are available for the Pig-a assay in male rat germ cells which require validation including germ cell DNA mutation origin.
Collapse
Affiliation(s)
- David Kirkland
- Kirkland Consulting, PO Box 79, Tadcaster, LS24 0AS, United Kingdom.
| | - Yoshifumi Uno
- Mitsubishi Tanabe Pharma Corporation, 2-2-50, Kawagishi, Toda, Saitama, 335-8505, Japan
| | - Mirjam Luijten
- National Institute for Public Health and the Environment (RIVM), Centre for Health Protection, Bilthoven, the Netherlands
| | - Carol Beevers
- Exponent International Ltd., The Lenz, Hornbeam Park, Harrogate, HG2 8RE, United Kingdom
| | - Jan van Benthem
- National Institute for Public Health and the Environment (RIVM), Centre for Health Protection, Bilthoven, the Netherlands
| | - Brian Burlinson
- Envigo, Huntingdon, Cambridgeshire, PE28 4HS, United Kingdom
| | | | - George R Douglas
- Environmental Health Science Research Bureau, Health Canada, Ottawa, K1A 0K9, Canada
| | - Shuichi Hamada
- LSI Medience Corporation, 14-1 Sunayama, Kamisu-shi, Ibaraki, 314-0255, Japan
| | - Katsuyoshi Horibata
- National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki, Kanagawa, 210-9501, Japan
| | - David P Lovell
- St George's Medical School, University of London, London, SW17 0RE, United Kingdom
| | | | | | - Nan Mei
- US FDA, National Center for Toxicological Research, Jefferson, AR, USA
| | - Takeshi Morita
- National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki, Kanagawa, 210-9501, Japan
| | - Wakako Ohyama
- Yakult Honsha Co., Ltd., 5-11, Izumi, Kunitachi-shi, Tokyo, 186-8650, Japan
| | - Andrew Williams
- Environmental Health Science Research Bureau, Health Canada, Ottawa, K1A 0K9, Canada
| |
Collapse
|
24
|
Pacchierotti F, Masumura K, Eastmond DA, Elhajouji A, Froetschl R, Kirsch-Volders M, Lynch A, Schuler M, Tweats D, Marchetti F. Chemically induced aneuploidy in germ cells. Part II of the report of the 2017 IWGT workgroup on assessing the risk of aneugens for carcinogenesis and hereditary diseases. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2019; 848:403023. [PMID: 31708072 DOI: 10.1016/j.mrgentox.2019.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/01/2019] [Accepted: 02/20/2019] [Indexed: 12/18/2022]
Abstract
As part of the 7th International Workshops on Genotoxicity Testing held in Tokyo, Japan in November 2017, a workgroup of experts reviewed and assessed the risk of aneugens for human health. The present manuscript is one of three manuscripts from the workgroup and reports on the unanimous consensus reached on the evidence for aneugens affecting germ cells, their mechanisms of action and role in hereditary diseases. There are 24 chemicals with strong or sufficient evidence for germ cell aneugenicity providing robust support for the ability of chemicals to induce germ cell aneuploidy. Interference with microtubule dynamics or inhibition of topoisomerase II function are clear characteristics of germ cell aneugens. Although there are mechanisms of chromosome segregation that are unique to germ cells, there is currently no evidence for germ cell-specific aneugens. However, the available data are heavily skewed toward chemicals that are aneugenic in somatic cells. Development of high-throughput screening assays in suitable animal models for exploring additional targets for aneuploidy induction, such as meiosis-specific proteins, and to prioritize chemicals for the potential to be germ cell aneugens is encouraged. Evidence in animal models support that: oocytes are more sensitive than spermatocytes and somatic cells to aneugens; exposure to aneugens leads to aneuploid conceptuses; and, the frequencies of aneuploidy are similar in germ cells and zygotes. Although aneuploidy in germ cells is a significant cause of infertility and pregnancy loss in humans, there is currently limited evidence that aneugens induce hereditary diseases in human populations because the great majority of aneuploid conceptuses die in utero. Overall, the present work underscores the importance of protecting the human population from exposure to chemicals that can induce aneuploidy in germ cells that, in contrast to carcinogenicity, is directly linked to an adverse outcome.
Collapse
Affiliation(s)
- Francesca Pacchierotti
- Health Protection Technology Division, Laboratory of Biosafety and Risk Assessment, ENEA, CR Casaccia, Rome, Italy
| | - Kenichi Masumura
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, Kanagawa, Japan
| | - David A Eastmond
- Department of Molecular, Cell and System Biology, University of California, Riverside, CA, USA
| | - Azeddine Elhajouji
- Novartis Institutes for Biomedical Research, Preclinical Safety, Basel, Switzerland
| | | | - Micheline Kirsch-Volders
- Laboratory for Cell Genetics, Faculty of Sciences and Bio-Engineering, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | - Francesco Marchetti
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9, Canada.
| |
Collapse
|
25
|
Kirsch-Volders M, Pacchierotti F, Parry EM, Russo A, Eichenlaub-Ritter U, Adler ID. Risks of aneuploidy induction from chemical exposure: Twenty years of collaborative research in Europe from basic science to regulatory implications. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2018; 779:126-147. [PMID: 31097149 DOI: 10.1016/j.mrrev.2018.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/26/2018] [Indexed: 12/13/2022]
Abstract
Although Theodor Boveri linked abnormal chromosome numbers and disease more than a century ago, an in-depth understanding of the impact of mitotic and meiotic chromosome segregation errors on cell proliferation and diseases is still lacking. This review reflects on the efforts and results of a large European research network that, from the 1980's until 2004, focused on protection against aneuploidy-inducing factors and tackled the following problems: 1) the origin and consequences of chromosome imbalance in somatic and germ cells; 2) aneuploidy as a result of environmental factors; 3) dose-effect relationships; 4) the need for validated assays to identify aneugenic factors and classify them according to their modes of action; 5) the need for reliable, quantitative data suitable for regulating exposure and preventing aneuploidy induction; 6) the need for mechanistic insight into the consequences of aneuploidy for human health. This activity brought together a consortium of experts from basic science and applied genetic toxicology to prepare the basis for defining guidelines and to encourage regulatory activities for the prevention of induced aneuploidy. Major strengths of the EU research programmes on aneuploidy were having a valuable scientific approach based on well-selected compounds and accurate methods that allow the determination of precise dose-effect relationships, reproducibility and inter-laboratory comparisons. The work was conducted by experienced scientists stimulated by a fascination with the complex scientific issues surrounding aneuploidy; a key strength was asking the right questions at the right time. The strength of the data permitted evaluation at the regulatory level. Finally, the entire enterprise benefited from a solid partnership under the lead of an inspired and stimulating coordinator. The research programme elucidated the major modes of action of aneugens, developed scientifically sound assays to assess aneugens in different tissues, and achieved the international validation of relevant assays with the goal of protecting human populations from aneugenic chemicals. The role of aneuploidy in tumorigenesis will require additional research, and the study of effects of exposure to multiple agents should become a priority. It is hoped that these reflections will stimulate the implementation of aneuploidy testing in national and OECD guidelines.
Collapse
Affiliation(s)
- Micheline Kirsch-Volders
- Laboratory for Cell Genetics, Faculty of Sciences and Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium.
| | | | | | - Antonella Russo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Ursula Eichenlaub-Ritter
- Institute of Gene Technology/Microbiology, Faculty of Biology, University of Bielefeld, Bielefeld, Germany
| | | |
Collapse
|
26
|
Marchetti F, Aardema MJ, Beevers C, van Benthem J, Godschalk R, Williams A, Yauk CL, Young R, Douglas GR. Identifying germ cell mutagens using OECD test guideline 488 (transgenic rodent somatic and germ cell gene mutation assays) and integration with somatic cell testing. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 832-833:7-18. [DOI: 10.1016/j.mrgentox.2018.05.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/28/2018] [Accepted: 05/28/2018] [Indexed: 01/15/2023]
|
27
|
Webster RJ, Williams A, Marchetti F, Yauk CL. Discovering human germ cell mutagens with whole genome sequencing: Insights from power calculations reveal the importance of controlling for between-family variability. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 831:24-32. [PMID: 29875074 DOI: 10.1016/j.mrgentox.2018.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/09/2018] [Accepted: 04/10/2018] [Indexed: 12/26/2022]
Abstract
Mutations in germ cells pose potential genetic risks to offspring. However, de novo mutations are rare events that are spread across the genome and are difficult to detect. Thus, studies in this area have generally been under-powered, and no human germ cell mutagen has been identified. Whole Genome Sequencing (WGS) of human pedigrees has been proposed as an approach to overcome these technical and statistical challenges. WGS enables analysis of a much wider breadth of the genome than traditional approaches. Here, we performed power analyses to determine the feasibility of using WGS in human families to identify germ cell mutagens. Different statistical models were compared in the power analyses (ANOVA and multiple regression for one-child families, and mixed effect model sampling between two to four siblings per family). Assumptions were made based on parameters from the existing literature, such as the mutation-by-paternal age effect. We explored two scenarios: a constant effect due to an exposure that occurred in the past, and an accumulating effect where the exposure is continuing. Our analysis revealed the importance of modeling inter-family variability of the mutation-by-paternal age effect. Statistical power was improved by models accounting for the family-to-family variability. Our power analyses suggest that sufficient statistical power can be attained with 4-28 four-sibling families per treatment group, when the increase in mutations ranges from 40 to 10% respectively. Modeling family variability using mixed effect models provided a reduction in sample size compared to a multiple regression approach. Much larger sample sizes were required to detect an interaction effect between environmental exposures and paternal age. These findings inform study design and statistical modeling approaches to improve power and reduce sequencing costs for future studies in this area.
Collapse
Affiliation(s)
- R J Webster
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - A Williams
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - F Marchetti
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - C L Yauk
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada.
| |
Collapse
|
28
|
Rowan-Carroll A, Beal MA, Williams A, Marchetti F, Yauk CL. Dose-response mutation and spectrum analyses reveal similar responses at two microsatellite loci in benzo(a)pyrene-exposed mouse spermatogonia. Mutagenesis 2018; 32:463-470. [PMID: 28575466 DOI: 10.1093/mutage/gex008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/23/2017] [Indexed: 11/14/2022] Open
Abstract
Identifying chemical exposures that can cause germline mutations is important as these mutations can be inherited, impacting both individual and population health. However, germline mutations are extremely rare and difficult to detect. Chemically induced germline mutations can be detected through analysis of highly unstable tandem repeat DNA. We recently developed a single-molecule PCR (SM-PCR) approach to quantify mutations at a mouse microsatellite locus (Mm2.2.1) in sperm for such purposes. In this study, we refine this approach through the combined analysis of mouse microsatellites Mm2.2.1 and Mm19.2.3. Mice were exposed to 0, 25, 50 or 100 mg/kg/day benzo(a)pyrene (BaP) by oral gavage for 28 days and sperm sampled 42 days after the end of exposure to measure effects on dividing spermatogonia. DNA was diluted to a single genome per PCR well for amplification of microsatellites in singleplex and multiplex reactions, and alleles were sized to identify mutations using capillary electrophoresis. Analysis of ~300-500 molecules per animal at both microsatellite loci, when tested individually, showed a ~2-fold increase in mutations relative to the controls at both the 50 and 100 mg/kg/day BaP doses. Multiplex SM-PCR revealed similar increases in mutation frequencies in both microsatellites. Comparison with results from a previous lacZ mutation assay conducted on the same mice revealed that although microsatellite mutations are a sensitive endpoint for detecting changes in mutation frequencies at lower doses, they appear to be saturable and thus have a reduced dynamic range. These results confirm that BaP is a male germ cell mutagen that broadly impacts tandem repeat DNA. Likewise, addition of a second hypervariable microsatellite increases the sensitivity of this assay.
Collapse
Affiliation(s)
- Andrea Rowan-Carroll
- Mechanistic Studies Division, Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Marc A Beal
- Mechanistic Studies Division, Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Andrew Williams
- Biostatistics and Epidemiology Division, Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Francesco Marchetti
- Mechanistic Studies Division, Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Carole L Yauk
- Mechanistic Studies Division, Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| |
Collapse
|
29
|
Motoyama S, Takeiri A, Tanaka K, Harada A, Matsuzaki K, Taketo J, Matsuo S, Fujii E, Mishima M. Advantages of evaluating γH2AX induction in non-clinical drug development. Genes Environ 2018; 40:10. [PMID: 29785231 PMCID: PMC5950202 DOI: 10.1186/s41021-018-0098-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/26/2018] [Indexed: 01/17/2023] Open
Abstract
γH2AX, the phosphorylated form of a histone variant H2AX at Ser 139, is already widely used as a biomarker to research the fundamental biology of DNA damage and repair and to assess the risk of environmental chemicals, pollutants, radiation, and so on. It is also beginning to be used in the early non-clinical stage of pharmaceutical drug development as an in vitro tool for screening and for mechanistic studies on genotoxicity. Here, we review the available information on γH2AX-based test systems that can be used to develop drugs and present our own experience of practically applying these systems during the non-clinical phase of drug development. Furthermore, the potential application of γH2AX as a tool for in vivo non-clinical safety studies is also discussed.
Collapse
Affiliation(s)
- Shigeki Motoyama
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Akira Takeiri
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Kenji Tanaka
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Asako Harada
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Kaori Matsuzaki
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Junko Taketo
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Saori Matsuo
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Etsuko Fujii
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| | - Masayuki Mishima
- Research Division, Chugai Pharmaceutical Co., Ltd, Gotemba, Shizuoka Japan
| |
Collapse
|
30
|
Anwar S, Madkor HR, Ahmed N, Wagih ME. In vivo anticlastogenic effect of silymarin from milk thistle Silybum marianum L. Indian J Pharmacol 2018; 50:108-115. [PMID: 30166747 PMCID: PMC6106120 DOI: 10.4103/ijp.ijp_660_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 07/06/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Silymarin, extracted from the seeds of Silybum marianum L. (Milk thistle), is traditionally used for treating various illnesses such as diabetes, cancer, inflammation, hepatitis, liver cirrhosis, and renal problems. Acute cytotoxicity and genotoxicity studies have been reported with ambiguous outcomes; however, its relevant anticlastogenic potential is not yet evaluated. This study was aimed to evaluate in vivo subacute anticlastogenic properties of silymarin to validate its use as a medicinal agent. MATERIALS AND METHODS Silymarin was isolated from seeds of milk thistle. Various genotoxicity bioassays of silymarin were performed using mice. First, the bone marrow cell proliferation was estimated by calculating mitotic index. Second, the chromosomal abnormalities in mice bone marrow cells were studied. Third, micronucleated polychromatic erythrocytes (MPE) test and in vivo activation of sister chromatid exchanges (SCEs) were carried out in mice bone marrow cells. Finally, primary spermatocytes were analyzed to estimate genotoxic effect of silymarin on germ cells. RESULTS We found that silymarin is capable of inducing a significant increase (P ≤ 0.05) in cell proliferation of bone marrow cells. There is no increase in chromosomal aberrations following silymarin treatments. Results clearly showed that it significantly (P ≤ 0.05) decreased the MPE. Likewise, it was found to be a negative inducer of SCEs. It decreased in total abnormal metaphase, SCEs, MPE, and aberrant diakinesis. CONCLUSION The results demonstrated that silymarin has a strong anticlastogenic activity upon mice genome in somatic and germ cells, indicating its safe use as a medicinal substance. Furthermore, it is not only safe but also has protective effect from clastogens.
Collapse
Affiliation(s)
- Sirajudheen Anwar
- Department of Medicinal Chemistry, Pharmacology and Toxicology unit, College of Clinical Pharmacy, Albaha University, Albaha, Kingdom of Saudi Arabia
| | - Hafez R. Madkor
- Department of Biochemistry , Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt
| | - Nafees Ahmed
- School of Pharmacy, Monash University Malaysia, Jalan lagoon selatan, Petaling Jaya, Selangor Darul Ehsan, Malaysia
| | | |
Collapse
|
31
|
Turkez H, Arslan ME, Ozdemir O. Genotoxicity testing: progress and prospects for the next decade. Expert Opin Drug Metab Toxicol 2017; 13:1089-1098. [DOI: 10.1080/17425255.2017.1375097] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Hasan Turkez
- Faculty of Science, Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, Turkey
- Department of Pharmacy, University ‘G. d’Annunzio’, Chieti, Italy
| | - Mehmet E. Arslan
- Faculty of Science, Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, Turkey
| | - Ozlem Ozdemir
- Faculty of Science, Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, Turkey
| |
Collapse
|
32
|
Abstract
Aneugenic effects of the chemicals with antitumor activity were studied in mouse oocytes in vivo by cytogenetic analysis. In control mice, no oocytes with numerical chromosome aberrations were found. Colchicine (0.2-4 mg/kg), paclitaxel (2.5-7.5 mg/kg), and etoposide (10-60 mg/kg) produced a significant dose-dependent aneugenic effects (induction of up to 25% aneuploid oocytes) and increased the yield of oocytes arrested in the meiotic MI stage and with premature separation of sister chromatid. Paclitaxel induced up to 20% polyploid chromosomes. Doxorubicin (2.5 mg/kg), melphalan (10 mg/kg), and cisplatin (5-10 mg/kg) exhibited weak aneugenic activity (induction of up to 5% aneuploid oocytes). Cyclophosphamide (10-80 mg/kg) had minor effect on the studied parameters. Methotrexate (25-200 mg/kg) exhibited no aneugenic activity, but significantly increased the level of polyploid cells. The observed aneugenic effects included hypo- and hyperploidy in various proportions or hypoploidy, but no solely hyperhaploidy.
Collapse
|
33
|
Ji Z, LeBaron MJ. Applying the erythrocyte Pig-a assay concept to rat epididymal sperm for germ cell mutagenicity evaluation. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:485-493. [PMID: 28714084 DOI: 10.1002/em.22109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 05/18/2017] [Accepted: 05/18/2017] [Indexed: 06/07/2023]
Abstract
The Pig-a assay, a recently developed in vivo somatic gene mutation assay, is based on the identification of mutant erythrocytes that have an altered repertoire of glycosylphosphatidylinositol (GPI)-anchored cell surface markers. We hypothesized that the erythrocyte Pig-a assay concept could be applied to rat cauda epididymal spermatozoa (sperm) for germ cell mutagenicity evaluation. We used GPI-anchored CD59 as the Pig-a mutation marker and examined the frequency of CD59-negative sperm using flow cytometry. A reconstruction experiment that spiked un-labeled sperm (mutant-mimic) into labeled sperm at specific ratios yielded good agreement between the detected and expected frequencies of mutant-mimic sperm, demonstrating the analytical ability for CD59-negative sperm detection. Furthermore, this methodology was assessed in F344/DuCrl rats administered N-ethyl-N-nitrosourea (ENU), a prototypical mutagen, or clofibrate, a lipid-lowering drug. Rats treated with 1, 10, or 20 mg/kg body weight/day (mkd) ENU via daily oral garage for five consecutive days showed a dose-dependent increase in the frequency of CD59-negative sperm on study day 63 (i.e., 58 days after the last ENU dose). This ENU dosing regimen also increased the frequency of CD59-negative erythrocytes. In rats treated with 300 mkd clofibrate via daily oral garage for consecutive 28 days, no treatment-related changes were detected in the frequency of CD59-negative sperm on study day 85 (i.e., 57 days after the last dose) or in the frequency of CD59-negative erythrocytes on study day 29. In conclusion, these data suggest that the epidiymal sperm Pig-a assay in rats is a promising method for evaluating germ cell mutagenicity. Environ. Mol. Mutagen. 58:485-493, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Zhiying Ji
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, Michigan, 48674
| | - Matthew J LeBaron
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, Michigan, 48674
| |
Collapse
|
34
|
Tarazona JV, Court-Marques D, Tiramani M, Reich H, Pfeil R, Istace F, Crivellente F. Glyphosate toxicity and carcinogenicity: a review of the scientific basis of the European Union assessment and its differences with IARC. Arch Toxicol 2017; 91:2723-2743. [PMID: 28374158 PMCID: PMC5515989 DOI: 10.1007/s00204-017-1962-5] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 03/21/2017] [Indexed: 11/29/2022]
Abstract
Glyphosate is the most widely used herbicide worldwide. It is a broad spectrum herbicide and its agricultural uses increased considerably after the development of glyphosate-resistant genetically modified (GM) varieties. Since glyphosate was introduced in 1974, all regulatory assessments have established that glyphosate has low hazard potential to mammals, however, the International Agency for Research on Cancer (IARC) concluded in March 2015 that it is probably carcinogenic. The IARC conclusion was not confirmed by the EU assessment or the recent joint WHO/FAO evaluation, both using additional evidence. Glyphosate is not the first topic of disagreement between IARC and regulatory evaluations, but has received greater attention. This review presents the scientific basis of the glyphosate health assessment conducted within the European Union (EU) renewal process, and explains the differences in the carcinogenicity assessment with IARC. Use of different data sets, particularly on long-term toxicity/carcinogenicity in rodents, could partially explain the divergent views; but methodological differences in the evaluation of the available evidence have been identified. The EU assessment did not identify a carcinogenicity hazard, revised the toxicological profile proposing new toxicological reference values, and conducted a risk assessment for some representatives uses. Two complementary exposure assessments, human-biomonitoring and food-residues-monitoring, suggests that actual exposure levels are below these reference values and do not represent a public concern.
Collapse
Affiliation(s)
- Jose V Tarazona
- Pesticides Unit, European Food Safety Authority, Via Carlo Magno 1/A, 43126, Parma, Italy.
| | - Daniele Court-Marques
- Pesticides Unit, European Food Safety Authority, Via Carlo Magno 1/A, 43126, Parma, Italy
| | - Manuela Tiramani
- Pesticides Unit, European Food Safety Authority, Via Carlo Magno 1/A, 43126, Parma, Italy
| | - Hermine Reich
- Pesticides Unit, European Food Safety Authority, Via Carlo Magno 1/A, 43126, Parma, Italy
| | - Rudolf Pfeil
- Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Frederique Istace
- Pesticides Unit, European Food Safety Authority, Via Carlo Magno 1/A, 43126, Parma, Italy
| | - Federica Crivellente
- Pesticides Unit, European Food Safety Authority, Via Carlo Magno 1/A, 43126, Parma, Italy
| |
Collapse
|
35
|
Beal MA, Yauk CL, Marchetti F. From sperm to offspring: Assessing the heritable genetic consequences of paternal smoking and potential public health impacts. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2017; 773:26-50. [PMID: 28927533 DOI: 10.1016/j.mrrev.2017.04.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/05/2017] [Accepted: 04/07/2017] [Indexed: 12/16/2022]
Abstract
Individuals who smoke generally do so with the knowledge of potential consequences to their own health. What is rarely considered are the effects of smoking on their future children. The objective of this work was to review the scientific literature on the effects of paternal smoking on sperm and assess the consequences to offspring. A literature search identified over 200 studies with relevant data in humans and animal models. The available data were reviewed to assess the weight of evidence that tobacco smoke is a human germ cell mutagen and estimate effect sizes. These results were used to model the potential increase in genetic disease burden in offspring caused by paternal smoking, with specific focus on aneuploid syndromes and intellectual disability, and the socioeconomic impacts of such an effect. The review revealed strong evidence that tobacco smoking is associated with impaired male fertility, and increases in DNA damage, aneuploidies, and mutations in sperm. Studies support that these effects are heritable and adversely impact the offspring. Our model estimates that, with even a modest 25% increase in sperm mutation frequency caused by smoke-exposure, for each generation across the global population there will be millions of smoking-induced de novo mutations transmitted from fathers to offspring. Furthermore, paternal smoking is estimated to contribute to 1.3 million extra cases of aneuploid pregnancies per generation. Thus, the available evidence makes a compelling case that tobacco smoke is a human germ cell mutagen with serious public health and socio-economic implications. Increased public education should be encouraged to promote abstinence from smoking, well in advance of reproduction, to minimize the transmission of harmful mutations to the next-generation.
Collapse
Affiliation(s)
- Marc A Beal
- Carleton University, Ottawa, Ontario K1S 5B6, Canada; Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Francesco Marchetti
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario K1A 0K9, Canada.
| |
Collapse
|
36
|
Galloway SM. International regulatory requirements for genotoxicity testing for pharmaceuticals used in human medicine, and their impurities and metabolites. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:296-324. [PMID: 28299826 DOI: 10.1002/em.22077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/05/2017] [Indexed: 06/06/2023]
Abstract
The process of developing international (ICH) guidelines is described, and the main guidelines reviewed are the ICH S2(R1) guideline that includes the genotoxicity test battery for human pharmaceuticals, and the ICH M7 guideline for assessing and limiting potentially mutagenic impurities and degradation products in drugs. Key aspects of the guidelines are reviewed in the context of drug development, for example the incorporation of genotoxicity assessment into non-clinical toxicity studies, and ways to develop and assess weight of evidence. In both guidelines, the existence of "thresholds" or non-linear dose responses for genotoxicity plays a part in the strategies. Differences in ICH S2(R1) protocol recommendations from OECD guidelines are highlighted and rationales explained. The use of genotoxicity data during clinical development and in assessment of carcinogenic potential is also described. There are no international guidelines on assessment of potentially genotoxic metabolites, but some approaches to safety assessment are discussed for these. Environ. Mol. Mutagen. 58:296-324, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
|
37
|
Dearfield KL, Gollapudi BB, Bemis JC, Benz RD, Douglas GR, Elespuru RK, Johnson GE, Kirkland DJ, LeBaron MJ, Li AP, Marchetti F, Pottenger LH, Rorije E, Tanir JY, Thybaud V, van Benthem J, Yauk CL, Zeiger E, Luijten M. Next generation testing strategy for assessment of genomic damage: A conceptual framework and considerations. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:264-283. [PMID: 27650663 DOI: 10.1002/em.22045] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/08/2016] [Indexed: 06/06/2023]
Abstract
For several decades, regulatory testing schemes for genetic damage have been standardized where the tests being utilized examined mutations and structural and numerical chromosomal damage. This has served the genetic toxicity community well when most of the substances being tested were amenable to such assays. The outcome from this testing is usually a dichotomous (yes/no) evaluation of test results, and in many instances, the information is only used to determine whether a substance has carcinogenic potential or not. Over the same time period, mechanisms and modes of action (MOAs) that elucidate a wider range of genomic damage involved in many adverse health outcomes have been recognized. In addition, a paradigm shift in applied genetic toxicology is moving the field toward a more quantitative dose-response analysis and point-of-departure (PoD) determination with a focus on risks to exposed humans. This is directing emphasis on genomic damage that is likely to induce changes associated with a variety of adverse health outcomes. This paradigm shift is moving the testing emphasis for genetic damage from a hazard identification only evaluation to a more comprehensive risk assessment approach that provides more insightful information for decision makers regarding the potential risk of genetic damage to exposed humans. To enable this broader context for examining genetic damage, a next generation testing strategy needs to take into account a broader, more flexible approach to testing, and ultimately modeling, of genomic damage as it relates to human exposure. This is consistent with the larger risk assessment context being used in regulatory decision making. As presented here, this flexible approach for examining genomic damage focuses on testing for relevant genomic effects that can be, as best as possible, associated with an adverse health effect. The most desired linkage for risk to humans would be changes in loci associated with human diseases, whether in somatic or germ cells. The outline of a flexible approach and associated considerations are presented in a series of nine steps, some of which can occur in parallel, which was developed through a collaborative effort by leading genetic toxicologists from academia, government, and industry through the International Life Sciences Institute (ILSI) Health and Environmental Sciences Institute (HESI) Genetic Toxicology Technical Committee (GTTC). The ultimate goal is to provide quantitative data to model the potential risk levels of substances, which induce genomic damage contributing to human adverse health outcomes. Any good risk assessment begins with asking the appropriate risk management questions in a planning and scoping effort. This step sets up the problem to be addressed (e.g., broadly, does genomic damage need to be addressed, and if so, how to proceed). The next two steps assemble what is known about the problem by building a knowledge base about the substance of concern and developing a rational biological argument for why testing for genomic damage is needed or not. By focusing on the risk management problem and potential genomic damage of concern, the next step of assay(s) selection takes place. The work-up of the problem during the earlier steps provides the insight to which assays would most likely produce the most meaningful data. This discussion does not detail the wide range of genomic damage tests available, but points to types of testing systems that can be very useful. Once the assays are performed and analyzed, the relevant data sets are selected for modeling potential risk. From this point on, the data are evaluated and modeled as they are for any other toxicology endpoint. Any observed genomic damage/effects (or genetic event(s)) can be modeled via a dose-response analysis and determination of an estimated PoD. When a quantitative risk analysis is needed for decision making, a parallel exposure assessment effort is performed (exposure assessment is not detailed here as this is not the focus of this discussion; guidelines for this assessment exist elsewhere). Then the PoD for genomic damage is used with the exposure information to develop risk estimations (e.g., using reference dose (RfD), margin of exposure (MOE) approaches) in a risk characterization and presented to risk managers for informing decision making. This approach is applicable now for incorporating genomic damage results into the decision-making process for assessing potential adverse outcomes in chemically exposed humans and is consistent with the ILSI HESI Risk Assessment in the 21st Century (RISK21) roadmap. This applies to any substance to which humans are exposed, including pharmaceuticals, agricultural products, food additives, and other chemicals. It is time for regulatory bodies to incorporate the broader knowledge and insights provided by genomic damage results into the assessments of risk to more fully understand the potential of adverse outcomes in chemically exposed humans, thus improving the assessment of risk due to genomic damage. The historical use of genomic damage data as a yes/no gateway for possible cancer risk has been too narrowly focused in risk assessment. The recent advances in assaying for and understanding genomic damage, including eventually epigenetic alterations, obviously add a greater wealth of information for determining potential risk to humans. Regulatory bodies need to embrace this paradigm shift from hazard identification to quantitative analysis and to incorporate the wider range of genomic damage in their assessments of risk to humans. The quantitative analyses and methodologies discussed here can be readily applied to genomic damage testing results now. Indeed, with the passage of the recent update to the Toxic Substances Control Act (TSCA) in the US, the new generation testing strategy for genomic damage described here provides a regulatory agency (here the US Environmental Protection Agency (EPA), but suitable for others) a golden opportunity to reexamine the way it addresses risk-based genomic damage testing (including hazard identification and exposure). Environ. Mol. Mutagen. 58:264-283, 2017. © 2016 The Authors. Environmental and Molecular Mutagenesis Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kerry L Dearfield
- U.S. Department of Agriculture, Food Safety and Inspection Service, Washington, District of Columbia
| | - B Bhaskar Gollapudi
- Exponent® Inc, Center for Toxicology and Mechanistic Biology, Midland, Michigan
| | | | | | - George R Douglas
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Rosalie K Elespuru
- U.S. Food and Drug Administration, CDRH/OSEL DBCMS, Silver Spring, Maryland
| | - George E Johnson
- Institute of Life Science, College of Medicine, Swansea University, Swansea, SA2 8PP, United Kingdom
| | | | - Matthew J LeBaron
- The Dow Chemical Company, Molecular, Cellular, and Biochemical Toxicology, Midland, Michigan
| | - Albert P Li
- In Vitro ADMET Laboratories LLC, Columbia, Maryland
| | - Francesco Marchetti
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Lynn H Pottenger
- Formerly of The Dow Chemical Company, Toxicology & Environmental Research and Consulting now with Olin Corporation, Midland, Michigan
| | - Emiel Rorije
- National Institute for Public Health and the Environment (RIVM), Center for Safety of Substances and Products, Bilthoven, 3720 BA, The Netherlands
| | - Jennifer Y Tanir
- ILSI Health and Environmental Sciences Institute (HESI), Washington, District of Columbia
| | - Veronique Thybaud
- Sanofi, Drug Disposition, Safety and Animal Research, Vitry-sur-Seine, France
| | - Jan van Benthem
- National Institute for Public Health and the Environment (RIVM), Center for Health Protection, Bilthoven, 3720 BA, The Netherlands
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Errol Zeiger
- Errol Zeiger Consulting, Chapel Hill, North Carolina
| | - Mirjam Luijten
- National Institute for Public Health and the Environment (RIVM), Center for Health Protection, Bilthoven, 3720 BA, The Netherlands
| |
Collapse
|
38
|
Ren N, Atyah M, Chen WY, Zhou CH. The various aspects of genetic and epigenetic toxicology: testing methods and clinical applications. J Transl Med 2017; 15:110. [PMID: 28532423 PMCID: PMC5440915 DOI: 10.1186/s12967-017-1218-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/18/2017] [Indexed: 12/15/2022] Open
Abstract
Genotoxicity refers to the ability of harmful substances to damage genetic information in cells. Being exposed to chemical and biological agents can result in genomic instabilities and/or epigenetic alterations, which translate into a variety of diseases, cancer included. This concise review discusses, from both a genetic and epigenetic point of view, the current detection methods of different agents’ genotoxicity, along with their basic and clinical relation to human cancer, chemotherapy, germ cells and stem cells.
Collapse
Affiliation(s)
- Ning Ren
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China. .,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, People's Republic of China.
| | - Manar Atyah
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, People's Republic of China
| | - Wan-Yong Chen
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, People's Republic of China
| | - Chen-Hao Zhou
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, People's Republic of China
| |
Collapse
|
39
|
Stewart J. The relevance of experimental reproductive studies in safety assessment. CURRENT OPINION IN TOXICOLOGY 2017. [DOI: 10.1016/j.cotox.2017.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
40
|
White PA, Douglas GR, Phillips DH, Arlt VM. Quantitative relationships between lacZ mutant frequency and DNA adduct frequency in Muta™Mouse tissues and cultured cells exposed to 3-nitrobenzanthrone. Mutagenesis 2017; 32:299-312. [PMID: 28096451 PMCID: PMC5638019 DOI: 10.1093/mutage/gew067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/13/2016] [Indexed: 02/07/2023] Open
Abstract
The frequency of stable DNA adducts in a target tissue can be used to assess biologically effective dose; however, the utility of the metric in a risk assessment context depends on the likelihood that the DNA damage will be manifested as mutation. Previously, we employed the Muta™Mouse system to examine the induction of lacZ mutants and DNA adducts following exposure to the well-studied mutagenic carcinogen 3-nitrobenzanthrone (3-NBA). In this follow-up work, we examined the empirical relationships between total adduct frequency and mutant frequency (MF) in tissues and cultured cells following acute 3-NBA exposure. The results show a significant induction of DNA damage and lacZ mutants in liver, colon and bone marrow, as well as FE1 pulmonary epithelial cells. In contrast, lung and small intestine samples had low, but significantly elevated adduct levels, with no significant increases in lacZ MF. Additional analyses showed a significant relationship between the mutagenic efficiency of total adducts, measured as the slope of the relationships between MF and total adduct frequency, and tissue-specific mitotic index (MI). The lack of mutation response in lung, in contrast to the high in vitro MF in FE-1 lung cells, is likely related to the 100-fold difference in MI. The lack of small intestine mutagenic response may be related to limited metabolic capacity, differences in DNA repair, and /or chemically induced apoptosis that has been observed for other potent mutagens. The results indicate that interpretation of adduct frequency values in a risk assessment context can be improved by considering the MI of the target tissue; however, more generalised interpretation is hampered by tissue-specific variations in metabolic capacity and damage processing. The work provides a proof of principle regarding the use of the Muta™Mouse system to critically examine the health risks associated with tissue-specific adduct loads.
Collapse
Affiliation(s)
- Paul A White
- Environmental Health Science and Research Bureau, Health Canada, Tunney’s Pasture, Colombine Driveway, Ottawa, Ontario, Canada
| | - George R Douglas
- Environmental Health Science and Research Bureau, Health Canada, Tunney’s Pasture, Colombine Driveway, Ottawa, Ontario, Canada
| | - David H Phillips
- King’s College London, Analytical and Environmental Sciences Division, MRC-PHE Centre for Environment and Health, Franklin-Wilkins Building, London, UK
| | - Volker M Arlt
- King’s College London, Analytical and Environmental Sciences Division, MRC-PHE Centre for Environment and Health, Franklin-Wilkins Building, London, UK
| |
Collapse
|
41
|
Habas K, Brinkworth MH, Anderson D. In vitro responses to known in vivo genotoxic agents in mouse germ cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:99-107. [PMID: 28205273 DOI: 10.1002/em.22075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 01/29/2017] [Indexed: 06/06/2023]
Abstract
Genotoxic compounds have induced DNA damage in male germ cells and have been associated with adverse clinical outcomes including enhanced risks for maternal, paternal and offspring health. DNA strand breaks represent a great threat to the genomic integrity of germ cells. Such integrity is essential to maintain spermatogenesis and prevent reproduction failure. The Comet assay results revealed that the incubation of isolated germ cells with n-ethyl-n-nitrosourea (ENU), 6-mercaptopurine (6-MP) and methyl methanesulphonate (MMS) led to increase in length of Olive tail moment and % tail DNA when compared with the untreated control cells and these effects were concentration-dependent. All compounds were significantly genotoxic in cultured germ cells. Exposure of isolated germ cells to ENU produced the highest concentration-related increase in both DNA damage and gene expression changes in spermatogonia. Spermatocytes were most sensitive to 6-MP, with DNA damage and gene expression changes while spermatids were particularly susceptible to MMS. Real-time PCR results showed that the mRNA level expression of p53 increased and bcl-2 decreased significantly with the increasing ENU, 6-MP and MMS concentrations in spermatogonia, spermatocytes and spermatids respectively for 24 hr. Both are gene targets for DNA damage response and apoptosis. These observations may help explain the cell alterations caused by ENU, 6-MP and MMS in spermatogonia, spermatocytes and spermatids. Taken together, ENU, 6-MP and MMS induced DNA damage and decreased apoptosis associated gene expression in the germ cells in vitro. Environ. Mol. Mutagen. 58:99-107, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Khaled Habas
- School of Medical Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Martin H Brinkworth
- School of Medical Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| | - Diana Anderson
- School of Medical Sciences, University of Bradford, Bradford, BD7 1DP, United Kingdom
| |
Collapse
|
42
|
Masumura K, Toyoda-Hokaiwado N, Ukai A, Gondo Y, Honma M, Nohmi T. Dose-dependent de novo germline mutations detected by whole-exome sequencing in progeny of ENU-treated male gpt delta mice. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2016; 810:30-39. [DOI: 10.1016/j.mrgentox.2016.09.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 09/20/2016] [Accepted: 09/27/2016] [Indexed: 01/06/2023]
|
43
|
The Vitotox and ToxTracker assays: A two-test combination for quick and reliable assessment of genotoxic hazards. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2016; 810:13-21. [DOI: 10.1016/j.mrgentox.2016.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 12/14/2022]
|
44
|
O'Brien JM, Beal MA, Yauk CL, Marchetti F. Benzo(a)pyrene Is Mutagenic in Mouse Spermatogonial Stem Cells and Dividing Spermatogonia. Toxicol Sci 2016; 152:363-71. [PMID: 27208087 PMCID: PMC4960908 DOI: 10.1093/toxsci/kfw088] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Although many environmental agents are established male germ cell mutagens, few are known to induce mutations in spermatogonial stem cells. Stem cell mutations are of great concern because they result in a permanent increase in the number of mutations carried in sperm. We investigated mutation induction during mouse spermatogenesis following exposure to benzo(a)pyrene (BaP). MutaMouse males were given 0, 12.5, 25, 50, or 100 mg/kg bw/day BaP for 28 days by oral gavage. Germ cells were collected from the cauda epididymis and seminiferous tubules 3 days after exposure and from cauda epididymis 42 and 70 days after exposure. This design enabled targeted investigation of effects on post-spermatogonia, dividing spermatogonia, and spermatogonial stem cells, respectively. BaP increased lacZ mutant frequency (MF) in cauda sperm after exposure of dividing spermatogonia (4.2-fold at highest dose, P < .01) and spermatogonial stem cells (2.1-fold at highest dose, P < .01). No significant increases in MF were detected in cauda sperm or seminiferous tubule cells collected 3 days post-exposure. Dose-response modelling suggested that the mutational response in male germ cells to BaP is sub-linear at low doses. Our results demonstrate that oral exposure to BaP causes spermatogonial stem cell mutations, that different phases of spermatogenesis exhibit varying sensitivities to BaP, with dividing spermatogonia representing a window of peak sensitivity, and that sampling spermatogenic cells from the seminiferous tubules at earlier time-points may underestimate germ cell mutagenicity. This information is critical to optimize the use of the international test guideline for transgenic rodent mutation assays for detecting germ cell mutagens.
Collapse
Affiliation(s)
- Jason M O'Brien
- *Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Marc A Beal
- *Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Carole L Yauk
- *Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Francesco Marchetti
- *Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9, Canada
| |
Collapse
|
45
|
Luijten M, Olthof ED, Hakkert BC, Rorije E, van der Laan JW, Woutersen RA, van Benthem J. An integrative test strategy for cancer hazard identification. Crit Rev Toxicol 2016; 46:615-39. [PMID: 27142259 DOI: 10.3109/10408444.2016.1171294] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Assessment of genotoxic and carcinogenic potential is considered one of the basic requirements when evaluating possible human health risks associated with exposure to chemicals. Test strategies currently in place focus primarily on identifying genotoxic potential due to the strong association between the accumulation of genetic damage and cancer. Using genotoxicity assays to predict carcinogenic potential has the significant drawback that risks from non-genotoxic carcinogens remain largely undetected unless carcinogenicity studies are performed. Furthermore, test systems already developed to reduce animal use are not easily accepted and implemented by either industries or regulators. This manuscript reviews the test methods for cancer hazard identification that have been adopted by the regulatory authorities, and discusses the most promising alternative methods that have been developed to date. Based on these findings, a generally applicable tiered test strategy is proposed that can be considered capable of detecting both genotoxic as well as non-genotoxic carcinogens and will improve understanding of the underlying mode of action. Finally, strengths and weaknesses of this new integrative test strategy for cancer hazard identification are presented.
Collapse
Affiliation(s)
- Mirjam Luijten
- a Centre for Health Protection, National Institute for Public Health and the Environment (RIVM) , Bilthoven , the Netherlands
| | - Evelyn D Olthof
- a Centre for Health Protection, National Institute for Public Health and the Environment (RIVM) , Bilthoven , the Netherlands
| | - Betty C Hakkert
- b Centre for Safety of Substances and Products, National Institute for Public Health and the Environment (RIVM) , Bilthoven , the Netherlands
| | - Emiel Rorije
- b Centre for Safety of Substances and Products, National Institute for Public Health and the Environment (RIVM) , Bilthoven , the Netherlands
| | | | - Ruud A Woutersen
- d Netherlands Organization for Applied Scientific Research (TNO) , Zeist , the Netherlands
| | - Jan van Benthem
- a Centre for Health Protection, National Institute for Public Health and the Environment (RIVM) , Bilthoven , the Netherlands
| |
Collapse
|
46
|
Habas K, Anderson D, Brinkworth M. Detection of phase specificity of in vivo germ cell mutagens in an in vitro germ cell system. Toxicology 2016; 353-354:1-10. [PMID: 27059372 DOI: 10.1016/j.tox.2016.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/24/2016] [Accepted: 04/04/2016] [Indexed: 10/22/2022]
Abstract
In vivo tests for male reproductive genotoxicity are time consuming, resource-intensive and their use should be minimised according to the principles of the 3Rs. Accordingly, we investigated the effects in vitro, of a variety of known, phase-specific germ cell mutagens, i.e., pre-meiotic, meiotic, and post-meiotic genotoxins, on rat spermatogenic cell types separated using Staput unit-gravity velocity sedimentation, evaluating DNA damage using the Comet assay. N-ethyl-N-nitrosourea (ENU), N-methyl-N-nitrosourea (MNU) (spermatogenic phase), 6-mercaptopurine (6-MP) and 5-bromo-2'-deoxy-uridine (5-BrdU) (meiotic phase), methyl methanesulphonate (MMS) and ethyl methanesulphonate (EMS) (post-meiotic phase) were selected for use as they are potent male rodent, germ cell mutagens in vivo. DNA damage was detected directly using the Comet assay and indirectly using the TUNEL assay. Treatment of the isolated cells with ENU and MNU produced the greatest concentration-related increase in DNA damage in spermatogonia. Spermatocytes were most sensitive to 6-MP and 5-BrdU while spermatids were particularly susceptible to MMS and EMS. Increases were found when measuring both Olive tail moment (OTM) and% tail DNA, but the greatest changes were in OTM. Parallel results were found with the TUNEL assay, which showed highly significant, concentration dependent effects of all these genotoxins on spermatogonia, spermatocytes and spermatids in the same way as for DNA damage. The specific effects of these chemicals on different germ cell types matches those produced in vivo. This approach therefore shows potential for use in the detection of male germ cell genotoxicity and could contribute to the reduction of the use of animals in such toxicity assays.
Collapse
Affiliation(s)
- Khaled Habas
- Division of Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, Richmond Road, West Yorkshire BD7 1DP, UK
| | - Diana Anderson
- Division of Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, Richmond Road, West Yorkshire BD7 1DP, UK
| | - Martin Brinkworth
- Division of Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, Richmond Road, West Yorkshire BD7 1DP, UK.
| |
Collapse
|
47
|
Masumura K, Toyoda-Hokaiwado N, Ukai A, Gondo Y, Honma M, Nohmi T. Estimation of the frequency of inherited germline mutations by whole exome sequencing in ethyl nitrosourea-treated and untreated gpt delta mice. Genes Environ 2016; 38:10. [PMID: 27350829 PMCID: PMC4918133 DOI: 10.1186/s41021-016-0035-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/11/2016] [Indexed: 11/29/2022] Open
Abstract
Background Germline mutations are heritable and may cause health disadvantages in the next generation. To investigate trans-generational mutations, we treated male gpt delta mice with N-ethyl-N-nitrosourea (ENU) (85 mg/kg intraperitoneally, weekly on two occasions). The mice were mated with untreated female mice and offspring were obtained. Whole exome sequencing analyses were performed to identify de novo mutations in the offspring. Results At 20 weeks after the treatment, the gpt mutant frequencies in the sperm of ENU-treated mice were 21-fold higher than those in the untreated control. Liver DNA was extracted from six mice, including the father, mother, and four offspring from each family of the ENU-treated or untreated mice. In total, 12 DNA samples were subjected to whole exome sequencing analyses. We identified de novo mutations in the offspring by comparing single nucleotide variations in the parents and offspring. In the ENU-treated group, we detected 148 mutation candidates in four offspring and 123 (82 %) were confirmed as true mutations by Sanger sequencing. In the control group, we detected 12 candidate mutations, of which, three (25 %) were confirmed. The frequency of inherited mutations in the offspring from the ENU-treated family was 184 × 10−8 per base, which was 17-fold higher than that in the control family (11 × 10−8 per base). The de novo mutation spectrum in the next generation exhibited characteristic ENU-induced somatic mutations, such as base substitutions at A:T bp. Conclusions These results suggest that direct sequencing analyses can be a useful tool for investigating inherited germline mutations and that the germ cells could be a good endpoint for evaluating germline mutations, which are transmitted to offspring as inherited mutations. Electronic supplementary material The online version of this article (doi:10.1186/s41021-016-0035-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kenichi Masumura
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo, 158-8501 Japan
| | - Naomi Toyoda-Hokaiwado
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo, 158-8501 Japan
| | - Akiko Ukai
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo, 158-8501 Japan
| | - Yoichi Gondo
- RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074 Japan
| | - Masamitsu Honma
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo, 158-8501 Japan
| | - Takehiko Nohmi
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo, 158-8501 Japan ; Biological Safety Research Center, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo, 158-8501 Japan
| |
Collapse
|
48
|
Ates G, Raitano G, Heymans A, Van Bossuyt M, Vanparys P, Mertens B, Chesne C, Roncaglioni A, Milushev D, Benfenati E, Rogiers V, Doktorova TY. In silico tools and transcriptomics analyses in the mutagenicity assessment of cosmetic ingredients: a proof-of-principle on how to add weight to the evidence. Mutagenesis 2016; 31:453-61. [PMID: 26980085 DOI: 10.1093/mutage/gew008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Prior to the downstream development of chemical substances, including pharmaceuticals and cosmetics, their influence on the genetic apparatus has to be tested. Several in vitro and in vivo assays have been developed to test for genotoxicity. In a first tier, a battery of two to three in vitro tests is recommended to cover mutagenicity, clastogenicity and aneugenicity as main endpoints. This regulatory in vitro test battery is known to have a high sensitivity, which is at the expense of the specificity. The high number of false positive in vitro results leads to excessive in vivo follow-up studies. In the case of cosmetics it may even induce the ban of the particular compound since in Europe the use of experimental animals is no longer allowed for cosmetics. In this article, an alternative approach to derisk a misleading positive Ames test is explored. Hereto we first tested the performance of five existing computational tools to predict the potential mutagenicity of a data set of 132 cosmetic compounds with a known genotoxicity profile. Furthermore, we present, as a proof-of-principle, a strategy in which a combination of computational tools and mechanistic information derived from in vitro transcriptomics analyses is used to derisk a misleading positive Ames test result. Our data shows that this strategy may represent a valuable tool in a weight-of-evidence approach to further evaluate a positive outcome in an Ames test.
Collapse
Affiliation(s)
| | - Giuseppa Raitano
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156 Milan, Italy
| | | | - Melissa Van Bossuyt
- Unit of Toxicology, Scientific Institute of Public Health (WIV-ISP), Juliette Wytsmanstraat 14, B-1050, Brussels, Belgium
| | | | - Birgit Mertens
- Unit of Toxicology, Scientific Institute of Public Health (WIV-ISP), Juliette Wytsmanstraat 14, B-1050, Brussels, Belgium
| | - Christophe Chesne
- Biopredic International, Parc d'activité de la Bretèche Bâtiment A4, 35760 Saint Grégoire, France and
| | - Alessandra Roncaglioni
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156 Milan, Italy
| | | | - Emilio Benfenati
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa 19, 20156 Milan, Italy
| | | | - Tatyana Y Doktorova
- Unit of Toxicology, Scientific Institute of Public Health (WIV-ISP), Juliette Wytsmanstraat 14, B-1050, Brussels, Belgium
| |
Collapse
|
49
|
Marchetti F, Massarotti A, Yauk CL, Pacchierotti F, Russo A. The adverse outcome pathway (AOP) for chemical binding to tubulin in oocytes leading to aneuploid offspring. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2016; 57:87-113. [PMID: 26581746 DOI: 10.1002/em.21986] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/13/2015] [Accepted: 10/13/2015] [Indexed: 06/05/2023]
Abstract
The Organisation for Economic Co-operation and Development (OECD) has launched the Adverse Outcome Pathway (AOP) Programme to advance knowledge of pathways of toxicity and improve the use of mechanistic information in risk assessment. An AOP links a molecular initiating event (MIE) to an adverse outcome (AO) through intermediate key events (KE). Here, we present the scientific evidence in support of an AOP whereby chemicals that bind to tubulin cause microtubule depolymerization resulting in spindle disorganization followed by altered chromosome alignment and segregation and the generation of aneuploidy in female germ cells, ultimately leading to aneuploidy in the offspring. Aneuploidy, an abnormal number of chromosomes that is not an exact multiple of the haploid number, is a well-known cause of human disease and represents a major cause of infertility, pregnancy failure, and serious genetic disorders in the offspring. Among chemicals that induce aneuploidy in female germ cells, a large majority impairs microtubule dynamics and spindle function. Colchicine, a prototypical chemical that binds to tubulin and causes microtubule depolymerization, is used here to illustrate the AOP. This AOP is specific to female germ cells exposed during the periovulation period. Although the majority of the data come from rodent studies, the available evidence suggests that the MIE and KEs are conserved across species and would occur in human oocytes. The development of AOPs related to mutagenicity in germ cells is expected to aid the identification of potential hazards to germ cell genomic integrity and support regulatory efforts to protect population health.
Collapse
Affiliation(s)
- Francesco Marchetti
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| | - Alberto Massarotti
- Dipartimento Di Scienze Del Farmaco, Università Degli Studi Del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| | - Francesca Pacchierotti
- Division of Health Protection Technologies, Laboratory of Biosafety and Risk Assessment, ENEA CR Casaccia, Rome, Italy
| | | |
Collapse
|
50
|
|