ArticlePDF Available

A Return to the Origin of the EMGS: Rejuvenating the Quest for Human Germ Cell Mutagens and Determining the Risk to Future Generations

Authors:

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. This article is protected by copyright. All rights reserved.
Commentary
A Return to the Origin of the EMGS: Rejuvenating the Quest for
Human Germ Cell Mutagens and Determining the Risk to Future
Generations
Francesco Marchetti ,*George R. Douglas, and Carole L.Yauk
Environmental Health Science Research Bureau, Health Canada, Ottawa, Ontario, Canada
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, ve
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 muta-
gens exist. We argue that it is time to reevaluate the
available data and conclude that we already have
evidence for the existence of environmental expo-
sures that impact human germ cells. What is missing
are denite data to demonstrate a signicant
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 environ-
mental 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 Foundersoriginal
focus. The next 50 years must involve collaborative
work between clinicians, epidemiologists, genetic
toxicologists, genomics experts and bioinformaticians
to precisely dene how environmental exposures
impact germ cell genomes. It is time for the research
and regulatory communities to prepare to interpret
thecomingoutpouringofdataanddevelopa
framework for managing, communicating and
mitigating the risk of exposure to human germ cell
mutagens. Environ. Mol. Mutagen. 61:4254,
2020. ©2019 Her Majesty the Queen in Right of Canada
Key words: mutation; germ cell; transgene; test guideli ne; heritable; nextg eneration sequencing
ROOTS OF EMGS IN GERM CELL MUTAGENESIS
Fifty years ago, the Environmental Mutagen Society
(now Environmental Mutagenesis and Genomics Society,
EMGS) was founded with the goal of identifying and elim-
inating agents from the environment that cause mutations
in germ cells to protect the health of human populations
(DeMarini 2020). Soon after, a committee was established
to formalize and publicize the fundamental role and mis-
sion of the EMGS (Committee 17 1975). Most of the con-
ferences and publications from this period emphasized the
health hazards of germ cell mutations (e.g., Workshop on
The Evaluation of Chemical Mutagenicity Data in Rela-
tion to Population Risk, Research Triangle Park, North
Carolina, April 2628, 1973; Environmental Health Per-
spectives, Vol. 6, December 1973). Despite the main
emphasis on germ cell-mediated genetic effects, there was
one paper at the workshop (Ames 1973) that profoundly
changed the direction and intent of mutagenicity testing
from a germ cell orientation to the prediction of carcinoge-
nicity. As a result of the ground-breaking invention of the
Ames assay and the demonstrated high degree of correla-
tion between mutagenesis and carcinogenesis, the vast
majority of genetic toxicology test development and appli-
cation became centered on the relationship between muta-
tion and cancer.
Notwithstanding the increased emphasis on cancer, germ
cell mutation has remained an endpoint of regulatory con-
cern. Numerous reports have been published and regulatory
practices promulgated related to the application and use of
germ cell mutagenicity data (see review by Singer and
Yauk 2010). Indeed, there are examples of regulatory
assessment of germ cell data at a frequency similar to that
for the regulatory assessment of cancer data (Yauk et al.
2015a). Furthermore, there is renewed interest in the
Reproduced with the permission of the Minister of Health Canada
Grant sponsor: Health Canada.
*Correspondence to: Francesco Marchetti, Environmental Health Science
and Research Bureau, Health Canada, 50 Colombine Driveway, Ottawa,
ON K1A 0K9, Canada. E-mail: francesco.marchetti@canada.ca
Received 29 July 2019; Revised 21 August 2019; Accepted 28
August 2019
DOI: 10.1002/em.22327
Published online 31 August 2019 in
Wiley Online Library (wileyonlinelibrary.com).
Environmental and Molecular Mu tagenesis 61:42^54 (2020)
©2019 Her Majesty t he Queen in Right of Canada
regulatory assessment of germ cell hazard and risk because
of the wide international implementation of the Globally
Harmonized System (GHS) of classication and labelling
of chemicals (United Nations 2017), which considers germ
cell mutagenicity for hazard classication.
This commentary provides a brief summary of the issues
surrounding the use of germ cell mutagenicity data in regu-
latory hazard and risk assessment, the application of new
facilitating technologies, and the prospects for a germ cell
genotoxicity renaissance.
IMPACT OF GERM CELL MUTATIONS
The role of mutations in the carcinogenic process is
well established and thus, mutagenicity testing has been
routinely conducted in order to identify agents that are
capable of inducing mutations and may initiate cancer
(Heich et al. 2020). In this context, it should be noted that
mutation is used a proxy for the ability of a compound to
induce cancer and not as a direct determinant of cancer
development. Also, although somatic mutations are associ-
ated with a broad spectrum of human diseases (Erickson
2014; Goschalk et al., 2020), there is still limited evidence
that a somatic mutation induced by an environmental expo-
sure may result in an adverse health effect other than cancer.
The impact of mutations in germ cells is dramatically
different. A single mutation in a germ cell will produce an
individual where every cell in the organism contains that
mutation. Thus, the chance of that mutation impacting the
health of the affected individual is much larger and it is not
restricted to cancer. Indeed, the Human Gene Mutation
Database (Stenson et al. 2017) currently catalogues over
260,000 different adverse mutational events affecting
approximately 10,000 genes (accessed June 2019).
Although point mutations represent the largest group of
mutations, genomic changes affecting a few to a few thou-
sand base pairs, translocations and complex chromosomal
aberrations are also observed. The database is a critical
resource to gather insights into the landscape of mutations
that are linked to human diseases, and while it provides no
information on the causes of the mutations, it would be
hard to imagine that not a single one of those mutations
was induced by an exogenous factor.
Although germ cell mutations are relatively rare at the
individual level, they can have a profound impact when
considered at the population level. It was recently estimated
that de novo mutations contribute to developmental disor-
ders in as many as 400,000 births per year (Deciphering
Developmental Disorders Study 2017). At the population
level, even a relatively small increase in mutation rate cau-
sed by an environmental exposure can have astounding
consequences. For example, an analysis that considered
only the spectrum of mutations that are expected to be
induced by tobacco smoke and the roughly 700 genes that
have been associated so far with intellectual disability
showed that even a modest 25% increase in tobacco-
induced sperm mutations would lead to over 500,000
affected children per generation across the global popula-
tion (Beal et al. 2017). The economic consequences of
these tobacco-induced de novo mutations for the health
care system can be easily estimated in the dozens of billion
US dollars.
The above example provides just a glimpse of the socie-
tal and economic impacts of environmentally induced de
novo mutations and of the importance of identifying and
eliminating exposure to environmental agents that can
induce these effects.
THE ENIGMA OF THE EXISTENCE OF HUMAN GERM CELL
MUTAGENS
Despite many decades of research, regulatory decisions
basedongermcellmutageniceffectsarestymiedbythelack
of even one established, and widely acknowledged, human
germ cell mutagen. As noted by DeMarini (2012), this
enigma may stem from the lack of an organization that is
specically tasked with assessing the available data to deter-
mine whether the weight of evidence is sufcient to conclude
that an agent is a human germ cell mutagen (e.g., similarly to
what the International Agency for Research on Cancer
(IARC) does for carcinogens). We wholeheartedly endorse
this assessment and argue that we already have sufcient data
to say that human germ cell mutagens exist and that the cur-
rent notion to the contrary is not grounded in facts. Below,
we briey discuss the evidence that substantiates the exis-
tence of human germ cell mutagens.
Over the past 80 years, a large body of literature has
accumulated on the existence of many germ cell mutagens
in animals. These studies demonstrate that exposure of lab-
oratory rodents to genotoxic agents spanning different
modes of action not only results in the induction of genetic
damage in the germ cells themselves, but also in the trans-
mission of de novo genetic changes to the offspring.
Several reviews have been published on agents that cause
genetic damage in rodent germ cells resulting in the induc-
tion of chromosomal structural aberrations (Marchetti and
Wyrobek 2005), aneuploidy (Pacchierotti et al. 2019),
gene mutations (Marchetti et al. 2018b), dominant lethal
mutations (Green et al. 1985) and inherited mutations
(Russell 2004). The information contained in these reviews
is sufcient to identify 84 agents as rodent germ cell muta-
gens (Table I), which is likely a large underestimate of the
true number of agents that are known to be genotoxic in
rodent germ cells. Thirty-one of the mammalian germ cell
mutagens have been shown to signicantly increase
inherited genetic changes in the offspring of exposed
males (Table I). Thus, it is unquestionable that many envi-
ronmental agents when tested under controlled laboratory
Environmental and Molecular Mutagenesis. DOI 10.1002/em
43Quest for Human Germ Cell Mutagens
conditions damage the genetic material of rodent germ
cells and result in the transmission of genetic defects to
the offspring. Is it realistic then to think that none of these
agents possess these characteristics when humans are
exposed? We believe that the answer to this question is an
unequivocal noand that there are many factors that are
TABLE I. Mammalian Germ Cell Mutagens
a
Agent Germ cells
b
Embryo
c
Offspring
d
Agent Germ cells
b
Embryo
c
Offspring
d
Acrylamide M, R M M 7,12-Dimethylbenz[a]anthracene M M
Ionizing radiation H, M M H
e
, M Busulfan M M
Tobacco smoke H, M M H
f
, M Carbendazim DH, SH M
Benzo(a)pyrene M M M Chemotherapy cocktails H, M M
Bleomycin M M M Colchicine CH, DH, M M
Chlorambucil M M M Di-(2-ethylhexyl)-phthalate M M
Cyclophosphamide M M M Diepoxybutane M M
Ethyl methanesulfonate M M M Ethanol H, M M
Ethylene oxide M M, R M Hycantone methanesulfonate M R
Etoposide CH, H, M M M Nocodazole M M
Glycidamide M M M Taxol M M
Isopropyl methanesulfonate M M M Vinblastine sulfate CH, M M
Melphalan M M M 6-Mercaptopurine M
Methyl methanesulfonate M M M Captan M
Mitomycin C M M M Chloramphenicol M
N-ethyl-N-nitrosourea M M M Colcemid M
N-methyl-N-nitrosourea M M M Di-2-ethyl-hexyladipate M
Procarbazine M M M
Dihydroergotoxin
methanesulfonate M
Triethylenemelamine M M M Dimethylmyrelan M
Trophosphamide M M M Ergotamine tartrate M
1-(2-Chroethyl)
3-cycloehxyl-1-
nitrosourea R M Forfestrol tetrasodium M
1,3-Butadiene M M Fotrin M
Chlormethine M M Hexamethylphosphoramide M
Dacarbazine M M Methyl mercury chloride SH, M
Diethyl sulfate M M Methysergide hydrogen maleate M
Ifosfamide M M Mitomen M
n-Propyl methanesulfonate M M
N-methyl-N0-nitro-N-
nitrosoguanidine M
ThioTEPA M M Rubratoxin B M
Trimethyl phosphate M M Saccarin M
N-propyl-N-nitrosourea M M Triupromazine M
Trichlorfon M H
g
Tris(2-methylazidiniyl)phosphine
oxide M
1,2-Dibromo-3-chloropropane M M 4-Nitroquinoline-1-oxide M
Acrylonitrile H Griseofulvin M
Air pollution M, HG Hydroquinone M
Amsacrine M Hydroxyurea M
Benomyl M Merbarone M
Benzene H Nitrilotriacetic acid M
Chloral hydrate M Orthovanadate M
Diethylstilbestrol M Podophyllotoxin CH
Econazole M Teniposide M
Epirubicin hydrochloride M Thiabendazole M
Fenvalerate H Vincristine sulfate CH
a
Agents reported as having positive results in the following papers: Green et al. (1985); Russell (2004); Marchetti and Wyrobek (2005); Marchetti et al.
(2018a); Marchetti et al. (2018b); Pacchierotti et al. (2019). For each agent, the species used in the studies is reported: CH (Chinese hamster); DH
(Djungarian Hamster); H (Humans); HG (Herring gull); M (Mouse); R (Rat); SH (Syrian Hamster).
b
Includes results obtained with the cytogenetic or FISH analyses of either sperm or oocytes.
c
Includes results obtained with the dominant lethal assay.
d
Includes results obtained in rodents with the specic locus test, heritable translocation assay in F1 males or tandem repeats and studies with human families.
e
Dubrova et al. (1996, 2002, 2006).
f
Secretan et al. (2009).
g
Czeizel et al. (1993).
Environmental and Molecular Mutagenesis. DOI 10.1002/em
44 Marchetti et al.
responsible for the apparent lack of human germ cell
mutagens.
Undoubtedly, there are many experimental variables that
make the chance of identifying rodent germ cell mutagens
more likely than doing the same in humans. First, studies
in laboratory animals are generally conducted at dose levels
that seldom occur in human populations. These studies use
primarily acute or subacute regimens rather than the
chronic exposure that is more common in humans. Acute
exposures use much higher doses than those that can be
applied under chronic conditions and are more likely to
result in a measurable effect because the amount of DNA
lesions induced at one time can overwhelm the DNA repair
defense mechanisms (OBrien et al. 2015; Yauk et al.
2015b). Second, these experiments generally use animals
with a homogenous genetic background that is not repre-
sentative of the genetic diversity present in the human pop-
ulation. A genetically homogenous population reduces the
variability in the response and increases the statistical
power of detecting an effect. Third, exposure parameters
and experimental conditions can be very carefully con-
trolled in the laboratory to signicantly reduce confounding
factors that may affect the response. Indeed, major chal-
lenges of conducting studies on the effects of environmen-
tal chemicals in human populations are the precise
characterization of the exposure and the identication of a
true control population. Fourth, technological limitations
have contributed to the challenges of identifying human
germ cell mutagens as the most common methods used for
investigating induced mutations in rodent germ cells
(e.g., the specic locus test [SLT]; Russell et al. 1998) are
not applicable to humans. Thus, both technological and
methodological difculties associated with human studies
may be in part responsible for the current lack of consensus
on the existence of human germ cell mutagens.
Despite these caveats, rodent germ cell mutagens are pri-
mary candidates for human germ cell mutagens and,
indeed, there are several examples of agents that induce
effects in both rodent and human gametes. In his commen-
tary, DeMarini (2012) identied ionizing radiation, chemo-
therapeutic agents, tobacco smoking and air pollution as
potential human germ cell mutagens. The latter two expo-
sures constitute complex mixtures that contain dozens of
potential candidate germ cell mutagens, the most notable
being genotoxic polycyclic aromatic hydrocarbons (PAHs)
like benzo[a]pyrene (BaP). As briey summarized below,
the available human data for these agents do indeed support
their categorization as human germ cell mutagens.
Since the pioneering work of Russell et al. with the SLT
(reviewed in Russell et al. 1998), ionizing radiation has
been one of the best characterized and most studied rodent
germ cell mutagen. In the early 90s, Dubrova et al. (1996)
reported increased numbers of mutations in tandem repeat
DNA sequences (minisatellites) in the offspring of men
who had been exposed to ionizing radiation during the
Chernobyl accident. These results were highly controversial
at the time because of criticisms on the selection of concur-
rent controls. However, follow up work with better mat-
ched control populations and longitudinal study designs
conrmed the initial ndings (Dubrova 2002; Dubrova
et al. 2002, 2006) and were further supported by studies
with laboratory rodents demonstrating that ionizing radia-
tion induced the same types of mutations under controlled
laboratory conditions (Dubrova et al. 1993, 1998). The
ndings of Dubrova et al. were also criticized because of
the type of mutations that were measured. It was thought
that alterations in minisatellite sequences had no health
impact and, thus, were of little signicance for human
health. Since then, we have a much better understanding of
how duplications and deletions in noncoding genomic
regions (including tandem repeats) can impact human
health (Mirkin 2007; Mori et al. 2013; Lupski 2015). In
light of todays knowledge and understanding that point
mutations and genomic alterations in coding genes repre-
sent only a portion of the possible genetic changes with
health-related implications, we contend that a re-evaluation
of the original results of Dubrova et al. leads to the conclu-
sion that these ndings can be considered as evidence
supporting the induction of heritable de novo mutations in
humans by ionizing radiation.
Chemotherapy cocktails generally contain chemicals that
are known to be genotoxic and have been shown to induce
both chromosomal abnormalities (reviewed in Wyrobek
et al. 2005) and tandem repeat mutations (Vilarino-Guell
et al. 2003; Glen et al. 2008; Glen and Dubrova 2012) in
rodent sperm. Studies in cancer patients undergoing chemo-
therapy have also shown signicant increases in the fre-
quency of sperm carrying either aneuploidy or structural
chromosomal abnormalities (Robbins et al. 1997; Frias
et al. 2003). Although in the majority of cases these
increases were temporary with the frequencies of sperm
with chromosomal abnormalities returning to baseline
levels with increasing time from the end of chemotherapy
(Martin et al. 1997; Robbins et al. 1997; Frias et al. 2003),
a few studies reported signicantly higher levels of abnor-
mal sperm many years after chemotherapy (De Mas et al.
2001; Tempest et al. 2008). These results suggest a perma-
nent effect of the chemotherapeutic cocktails on human
stem cell spermatogonia. Importantly, these effects mirror
those observed in the sperm of laboratory animals and
demonstrate that when exposure levels are comparable,
similar effects are seen in both rodent and human germ
cells. Thus, for chemotherapeutic agents there is clear evi-
dence that results obtained in rodent germ cells are predic-
tive of similar effects in human germ cells.
Tobacco smoke is one of the most widespread environ-
mental exposures in human populations. The detrimental
effects of tobacco smoking have been known for many
decades, but in recent years there has been a renewed
appreciation for its effects in the germ cells of smokers and
Environmental and Molecular Mutagenesis. DOI 10.1002/em
45Quest for Human Germ Cell Mutagens
their potential consequences for the offspring. A recent
review on the effects of tobacco smoking identied
increases in DNA strand breaks, aneuploidy and mutations
as some of the genetic changes that are present in the
sperm of tobacco smokers (Beal et al. 2017). Also, studies
in both rodents (Yauk et al. 2007; Marchetti et al. 2011)
and humans (Linschooten et al. 2013) have consistently
reported increases in tobacco-induced tandem repeat muta-
tions. Perhaps surprisingly, there are yet no human data
showing an increase in de novo mutations in the offspring
of smokers, although whole genome analysis of cohorts of
families with smoking fathers was identied as a priority
research area (Yauk et al. 2013). Nevertheless, support for
the hereditary effects of tobacco smoke can be found in the
IARCs assessment that preconception paternal smoking is
causally linked to an increased risk of childhood leukemia
in the offspring (Secretan et al. 2009).
Among the four potential human germ cell mutagens
proposed by DeMarini, air pollution is the one with the
more limited data in humans. However, multiple studies in
rodents (Somers et al. 2002; Yauk et al. 2008) and wildlife
species (Yauk and Quinn 1996; Yauk et al. 2000) have
consistently shown the impact of air pollution on the germ
cell genome, including the demonstration that the muta-
genic effects of air pollution in germ cells reside with par-
ticulate matter (Somers et al. 2004). Importantly, all these
studies of environmentally exposed animals resulted in her-
itable tandem repeat mutations induced by ambient levels
of air pollutants. Even in the absence of direct evidence in
humans, the available animal data would be considered,
when evaluated within an IARC style assessment, to be
sufcient to conclude that air pollution is likely to be a
human germ cell mutagen.
PAHs are exemplary mutagens in tobacco smoke and air
pollution. A large body of literature demonstrates that BaP
is highly mutagenic to adult rodent germ cells. Studies
clearly show that BaP causes both tandem repeat and gene
mutations in rodent spermatogonial stem cells and differen-
tiating spermatogonia (Olsen et al. 2010; Verhofstad et al.
2011; Xu et al. 2014; OBrien et al. 2016a, b; Rowan-
Carroll et al. 2017). Moreover, a recent study applying
modern genomic technologies provides clear evidence that
BaP induced de novo mutations (both single nucleotides
and large duplications) in the offspring of exposed male
mice (Beal et al. 2019). These data also lend weight to the
relevance of tandem repeat mutations as biomarkers of
potential mutagenicity in other locations in the genome. In
parallel with this strong evidence of germ cell mutagenicity
in rodents, PAH DNA adducts are detected in human
sperm samples of individuals exposed to air pollution (Jeng
et al. 2015; Oliveri Conti et al. 2017). Sperm of human
smokers exhibit high levels of BaP DNA adducts as well
as increased burdens of bulky adducts and oxidative lesions
in general (reviewed in Beal et al. 2017). Therefore, the
presence of BaP and other bulky PAH adducts in human
sperm, and strong evidence supporting that BaP induces
mutations in rodent sperm that are transmitted to their off-
spring, strongly imply that BaP and PAHs are likely to
cause germ cell mutagenicity in humans.
Evidence for the induction of genetic damage in
human germ cells also comes from many studies that
have reported signicant increases in genetic abnormali-
ties in the sperm of workers occupationally exposed to
benzene (Xing et al. 2010; Marchetti et al. 2012), acrylo-
nitrile (Xu et al. 2003) and fenvalerate (Xia et al. 2004)
among others. When the available human germ cell data
are evaluated as a whole, the unavoidable conclusion is
that we already have sufcient data to say that human
germ cell mutagens exist. Admittedly, what is missing is
the demonstration that some of these effects observed in
human germ cells are transmitted to the next generation.
One possible such an example is the reported case of a
cluster of Down syndrome cases in a human population
linked to environmental exposure to trichlorfon (Czeizel
et al. 1993). However, this nding is outweighed by the
current failure of epidemiological studies in children of
atomic bomb (Schull 2003) and cancer (Mulvihill 2012)
survivors to demonstrate an increase in inherited diseases
in the offspring. Although these human epidemiological
studies have shortcomings (discussed in detail in Schull
2003; DeMarini 2012; Mulvihill 2012), data demonstrat-
ing the induction of de novo mutations in the offspring
of exposed parents will be necessary to change this
notion that there are no human germ cell mutagens. As
discussed later, we believe that, as a scientic commu-
nity, we are at the cusp of applying the latest genomic
approaches to demonstrate transmission of environmen-
tally induced de novo mutations to human offspring and
conclusively demonstrate the existence of human germ
cell mutagens.
ARE HUMAN GERM CELL S FUNDAMENTALLY DIFFERENT
FROM RODENT GERM CELLS?
The results obtained in rodents may not be predictive of
similar effects in humans if there are fundamental differ-
ences between human and rodent spermatogenesis or if
human germ cells have a more efcient DNA damage
response than rodent germ cells. However, there is no solid
evidence for either of these two possibilities. An extensive
description of the process of spermatogenesis in mamma-
lian species, including DNA repair competency, is outside
of the scope of this commentary and the reader is directed
to a few comprehensive reviews on the topic (Olsen et al.
2005; Amann 2008; Hermo et al. 2010). Here, we aim to
succinctly argue that there are no physiological differences
in spermatogenic processes that can be invoked to cause
differences in the response of humans and rodents to envi-
ronmental germ cell mutagens.
Environmental and Molecular Mutagenesis. DOI 10.1002/em
46 Marchetti et al.
The process of spermatogenesis has been studied in
detail in humans and in many laboratory animals. Sper-
matogenesis is remarkably similar among mammalian spe-
cies with the major difference being the time necessary to
produce mature sperm from stem cell spermatogonia,
which is species specic (Adler 1996). Furthermore, many
of the genes that are known to be essential for spermato-
genesis are conserved across species (Bonilla and Xu 2008;
White-Cooper and Bausek 2010) and defects in a particular
gene can produce the same phenotype in multiple species
(Vangompel and Xu 2011). Also conserved across species
is the fact that the DNA repair capacity is maximum in
spermatogonia and gradually disappears during the
postmeiotic phase of spermatogenesis (Olsen et al. 2005).
Data in rodents clearly show that this period is most vul-
nerable to the induction of genetic lesions that are retained
in the fertilizing sperm and that result in the formation of
de novo genetic defects in the developing embryo
(Marchetti and Wyrobek 2005).
The apparent lack of human germ cell mutagens cannot
be ascribed to a higher resistance of human germ cells to
the detrimental impact of genotoxic agents. As already dis-
cussed, exposure to comparable doses of ionizing radiation
or chemotherapeutic agents induce similar effects in both
rodent and human sperm. Furthermore, it is broadly
accepted that aging is a human germ cell mutagen. An
increase in the risk of having a child with a genetic syn-
drome as function of paternal age has been known for
decades (Crow 2000). Studies with next generation
sequencing (NGS) have unequivocally shown the presence
of a paternal age effect for the induction of de novo muta-
tions that are transmitted to the offspring (Kong et al.
2012; Francioli et al. 2015; Rahbari et al. 2016). More
recent studies have also demonstrated an increase in de
novo mutations, albeit smaller than the paternal effect, as
function of maternal age (Maretty et al. 2017; Gao et al.
2019). In these genomic studies, the stronger paternal age
effect is attributed to the continuous replication of stem cell
spermatogonia throughout the males reproductive life and
to the possibility of introducing an error during DNA repli-
cation. Interestingly, the potential contribution of an envi-
ronmental exposure in the paternal age effect is seldom
considered. For example, a study that analyzed the paternal
age effect in three human families with multiple children
showed that the slope of the paternal age effect varied
greatly among the fathers (Rahbari et al. 2016). While dif-
ferent baseline efciencies in DNA repair activities could
contribute to this variability, so could differential exposure
of the fathers to an environmental mutagen. Finally, analy-
sis of the types of de novo mutations transmitted to human
offspring show clustering of mutations (Francioli et al.
2015) as observed in the offspring of mutagen-exposed ani-
mals (Adewoye et al. 2015; Beal et al. 2019) and signatures
of mutational processes that point to DNA damage as the
source of de novo mutations (Gao et al. 2019). It is difcult
to imagine a process by which the DNA repair machinery
of human germ cells is able to distinguish between lesions
that are induced by endogenous processes vs. those induced
by exogenous agents and are able to completely repair the
latter but not the former.
In summary, there is little biological plausibility to sup-
port the notion that the response of human germ cells to an
environmental insult is fundamentally different from that
occurring in rodent germ cells, which would negate the
possibility that established rodent germ cell mutagens
would also be human germ cell mutagens.
TECHNICAL LIMITATIONS OF AVAILABLE GERM CELL
MUTATION TESTS
The search for human germ cell mutagens has been ham-
pered by the lack of efcient test methods to detect the
impact of environmental agents on the germline. Even in
laboratory rodents, the study of germ cell mutagenesis has
suffered from the limitations of available tests, such as the
need for an exceedingly large number of animals. A prime
example is the SLT developed in the late 1950s at Oak
Ridge National Laboratory. For many decades, it was the
only available test to detect de novo mutations in the
offspring of exposed mice. The SLT was instrumental in
establishing occupational exposure limits for radiation
workers (Neel and Lewis 1990) and made fundamental
contributions to the elucidation of the varying sensitivity of
the different phases of spermatogenesis to environmental
exposure (Russell 2004). However, a typical experiment
required the analysis of several hundred thousand animals,
something that would not be considered remotely feasible
today, both for nancial and ethical considerations. Other
available tests, although not requiring as many animals as
the SLT, did not provide information on the heritability of
mutations in live animals (i.e., the dominant lethal test) or
assayed only on a specic type of genetic damage
(i.e., reciprocal translocation test; results with both tests
reviewed in Marchetti and Wyrobek 2005).
Over the years, a variety of tests have been developed to
assess different types of genetic damage in germ cells and
applied to the question of environmentally induced germ
cell mutagenesis. Corollary assays from some of the current
rodent germ cell tests are available for use with human
sperm and have allowed a direct comparison of the
response to environmental insult across species. An in-
depth review of the strengths and limitations of these tests
is provided in the 2013 IWGT report on germ cell testing
(Yauk et al. 2015a). Here, we specically highlight two
approaches that represented important breakthroughs in the
eld of germ cell mutagenesis.
As discussed, the analysis of tandem repeat mutations in
sperm has been instrumental in demonstrating consistency
of effects across species and contributed to the overall
Environmental and Molecular Mutagenesis. DOI 10.1002/em
47Quest for Human Germ Cell Mutagens
weight of evidence that ionizing radiation, chemotherapeu-
tic drugs, tobacco smoking and air pollution are human
germ cell mutagens. The analysis of tandem repeat muta-
tions provided, for the rst time, an opportunity to study
heritable mutagenesis in humans using a reasonable and
manageable population size. Furthermore, the assay can be
conducted in any species, including wild animals, and is
equally applicable to the analysis of sperm of exposed
fathers and their children enabling a direct demonstration
of the heritability of these mutations. Unfortunately, uncer-
tainties over the impact of these types of mutations for
human health and a technically demanding protocol has
hampered the broad acceptance of tandem repeat assays
among the regulatory and research communities. However,
extensive efforts are ongoing to develop advanced genomic
approaches (e.g., long-read sequencing; Chaisson et al.
2019; Levy-Sakin et al. 2019) for rapidly and efciently
characterizing structural variation in the genome and it is
envisioned that these methods will become important tools
for analyzing environmentally induced structural variation
in the germ cell genome (Salk and Kennedy 2020).
A breakthrough for germ cell mutagenicity for regulatory
testing also occurred at the end of the 20th century when
transgenic rodent (TGR) models were developed (reviewed
in Lambert et al. 2005). These models have bacterial
reporter genes integrated into their genome that can be
recovered from any tissue and used in an in vitro assay to
measure mutations that were induced in vivo. The use of
TGR models for mutagenicity testing was later codied in
an Organisation for Economic Co-Operation and Develop-
ment Test Guideline (OECD 2013). TGR models provided
the opportunity to conduct in vivo mutagenicity testing
using a reasonable number of animals, in line with other
well-established genotoxicity tests for somatic cells, and to
directly compare the response in germ cells with that in
somatic tissues. This is particularly important because germ
cell mutagenicity is a health hazard criterion in the GHS
classication and labelling of chemicals (United Nations
2017) and has resulted in an increased demand for testing
in this area. The use of TGR models for germ cell muta-
genesis was recently summarized together with recommen-
dations on proper experimental design for integration with
somatic tissue mutagenicity testing (Marchetti et al.
2018a, b).
A major limitation of existing germ cell methods is the
inability to account for postfertilization events. There is a
wealth of data showing that the manifestation of damage
induced in postmeiotic germ cells into a xed genetic
change, whether point mutation or chromosomal aberra-
tion, is not completed until after the sperm fertilizes the
egg (Marchetti and Wyrobek 2005). There is also an
example of a meiotic DNA lesion that does not manifest
itself as a permanent genetic abnormality until after fertili-
zation (Marchetti et al. 2015). These data point to the
importance of the perifertilization period in determining
the amount and types of lesions that originate de novo
genetic changes, and in particular, of the role that an ef-
cient DNA repair system in the fertilized egg can play in
fundamentally mediating how much of the sperm DNA
lesions are correctly repaired (Marchetti et al. 2007;
Derijck et al. 2008). We believe that recent developments
in genomic methodologies (described later) are well posi-
tioned to ll this key research gap and also address
another current limitation of germ cell tests; i.e., the lack
of efcient and practical methods to analyze the induction
of mutations in female germ cells that has resulted in
a paucity of experimental data on female germ cell
mutagenicity.
RISK ASSESSMENT OF GERM CELL MUTATIONS OVER THE
NEXT 50 YEARS OF THE EMGS
During the rst 50 years of the EMGS, the mutagenic
activity of chemicals has been evaluated primarily in a
yes/no fashion. However, a paradigm shift is ongoing in
genetic toxicology to move away from a purely qualitative
analysis of genotoxic responses (Deareld et al. 2017). The
proposed path forward implements rened doseresponse
modeling approaches to establish point of departure (PoD)
metrics for potency comparisons, with the long-term vision
of using mutation as a PoD in human health risk assess-
ment (Johnson et al. 2015; Wills et al. 2016). The chal-
lenges of this paradigm shift and the necessary
improvement in methods to measure mutagenicity, and
how to rene risk assessment approaches to encourage
uptake by regulatory bodies, are discussed in Heich
et al. (2020).
This paradigm shift has important implications for the
risk assessment of germ cell mutations and the need to
conduct germ cell mutagenesis testing. A general assump-
tion in risk assessment is that evaluations based on
somatic cell mutation assays protect the germline by
default. Historical and experimental data support that
somatic cell mutation assays predict putative mutagenic
effects in the germline. Indeed, there is currently no evi-
dence for a unique germ cell mutagen, although caution
should be exercised in drawing broad conclusions because
chemicals that are negative in somatic cells are rarely
tested in germ cells (Yauk et al. 2015a). This new empha-
sis on doseresponse modeling requires that reliance on
somatic mutagenicity tests to predict effects in germ cells
now extends also to establishing PoDs that protect the
germline as well. However, we already know that this is
incorrect. Several examples exist demonstrating germ cell
effects at doses that do not elicit adverse effects in somatic
cells; thus, PoD based on somatic endpoints may underes-
timate the risk to the germline. For example, Witt et al.
(2003) reported strong dominant lethal effects in mice
exposed to N-hydroxyacrylamide while no signicant
Environmental and Molecular Mutagenesis. DOI 10.1002/em
48 Marchetti et al.
effects could be detected using the bone marrow micronu-
cleus (MN) analysis, one of the most commonly con-
ducted tests for regulatory purpose. Similarly, Marchetti
et al. (2011) showed induction of tandem repeat mutations
in the sperm of mice exposed to either mainstream or side-
stream tobacco smoke at doses that did not increase MN
frequencies in bone marrow. Finally, a review of the gen-
otoxicity of acrylamide showed that the response in germ
cells is stronger than the response in somatic cells
(Deareld et al. 1995).
It could be said that the above examples suffer from the
shortcoming of comparing different endpoints in germ cells
vs. somatic cells and, thus, the observed differential
responses are not a true indication of an increased sensitiv-
ity of germ cells to those agents, but simply reect method-
ological differences in the sensitivity to detect an effect.
However, even when the same endpoint is analyzed, there
are still four cases of chemicals that failed to induce MN in
the mouse bone marrow, while signicantly increasing their
frequency in spermatids (Cliet et al. 1993).
While recognizing the potential impact of induced
germline mutations on human populations, national and
international efforts have concentrated on the identica-
tion of germ cell mutagen hazards, eliminating them, or
otherwise reducing their potential exposure to human
populations (Committee 17 1975; IPCS 1985; World
Health Organization 1986). Efforts also extended to
identifying epidemiologic associations between human
male exposures and mutation-based diseases in offspring
(Narod et al. 1988) to identify potential human germline
mutagens. In recognition of such potential hazards,
national and international regulatory authorities have
established guidelines and regulations that include the
goal of identifying and controlling human exposure to
germline mutagens (Cimino 2006).
Paramount among such regulatory schemes is the inter-
national adoption of the GHS of Classication and Label-
ling of Chemicals (United Nations 2017) by most
industrialized nations. The GHS includes human germ cell
mutation as a toxicological endpoint equal to other end-
points such as cancer and reproductive effects, thus, indi-
cating its universal international acceptance. While such
regulatory schemes do establish a basis for germline hazard
identication, they do not provide a clear path to the actual
estimation of the human risk in terms of induced mutation
rate in offspring associated with such hazards. Various
germ cell risk estimation methods have relied primarily on
the use of animal studies and some form of quantitative
extrapolation to a related class of human genetically based
disease (reviewed extensively in Yauk et al. 2015a).
There are limitations to the currently available
approaches for quantitative assessment of germ cell risk
from chemical exposures. For example: (1) animal studies
do not directly measure or quantitate effects in humans,
while current methods do not routinely assess
environmentally caused de novo mutations in specic
human target genes, or groups/categories of genes; (2) there
are no accepted, or validated, allometric and exposure fac-
tors for extrapolation of animal germline mutation data to
individual humans, or human population groups, and there
is insufcient information on the means to extrapolate
mutation data in animal genes to analogous human genes
and (3) there is no accepted de minimis risk level for the
estimation of unacceptable exposure levels for germline
mutation effects to offspring.
Despite such historically based shortcomings, we are
now at a turning point facilitated by the accelerated devel-
opment of new genomic technologies that will nally
enable us to make signicant advances in the application of
genetic risk assessment in both the academic and regulatory
contexts.
THE FUTURE: NEXT GENERATIONSEQUENCING
Studies on the heritable effects of environmental agents
in humans have historically relied on measuring phenotypic
endpoints rather than detecting the underlying genetic
cause (DeMarini 2012). As not all de novo mutations have
a discernable phenotypic consequence, these approaches
were limited to measuring only dominant effects in early
life with a resulting loss of statistical power. In addition,
such approaches cannot quantify the impact of recessive
mutations on health until subsequent generations, or muta-
tions that contribute to multigenic disorders. The advent of
NGS approaches, together with great gains in bioinformat-
ics pipelines to handle, analyze and validate sequenced
data, are allowing the analysis of mutations over the entire
genome and are promising to revolutionize the way germ
cell mutagenicity is investigated. NGS complements other
genomic approaches such as array comparative genomic
hybridization to capture a variety of genomic changes that
can impact human health (Carvalho and Lupski 2016).
NGS approaches can also provide critical mechanistic
information on the types of mutations that are induced and
increase the condence that these de novo mutations are
indeed the consequence of an exogenous exposure. Impor-
tantly, these approaches are feasible with a relatively small
number of samples and are equally applicable to laboratory
animals as well as human families (Webster et al. 2018). In
fact, experiments conducted in laboratory animals can
inform studies in human families on the proper experimen-
tal design, power of analysis and confounding factors.
There are already several examples of the use of these
genomic approaches for germ cell mutagenesis studies in
rodents. Adewoye et al. (2015) reported signicant induc-
tion of copy number variants (CNVs) in the offspring of
irradiated animals. Interestingly, both irradiated spermato-
gonia and sperm caused similar levels of CNVs in off-
spring. In the same study, although there was evidence for
Environmental and Molecular Mutagenesis. DOI 10.1002/em
49Quest for Human Germ Cell Mutagens
clustering of mutations after irradiation of male germ cells,
the authors could not detect an overall signicant increase
in de novo single nucleotide variants (SNVs). However, a
recent study (Beal et al. 2019) reported signicant increases
in CNV gains and SNVs in the offspring of male mice
exposed to BaP with evidence of clustering of mutations.
The study also demonstrated that there was a signicant
increase in constitutional SNVs (i.e., present at fertilization)
when spermatogonia were exposed to BaP, while there was
a signicant increase in embryonic SNVs (i.e., originating
after fertilization) when postmeiotic germ cells were
exposed. Importantly, the types of mutations observed in
the offspring matched the expected mutational signature of
BaP and were consistent with the pattern of mutations
observed in the sperm of exposed males, conclusively dem-
onstrating that the increase in de novo mutation was linked
to the paternal exposure to BaP. Finally, there is also evi-
dence that exome sequencing may be sufcient to detect
signicant increases in de novo mutations (Masumura et al.
2016a, b). These studies found that N-ethyl-N-nitrosourea
(ENU) signicantly increased de novo mutations in the
exomes of offspring from exposed mice and that the
inherited mutations exhibited a spectrum characteristic of
ENU-induced mutations.
Together, these rodent studies: (1) provide foundational
examples for the application of genomic technologies for
investigating the induction of heritable mutations following
exposure to environmental mutagens; (2) demonstrate that
different mutagenic agents can differentially affect various
types of genetic changes; (3) provide support for the herita-
bility of mutations observed in sperm; and (4) conrm the
important role of postfertilization events in determining the
genetic load that is transmitted to the offspring.
Application of NGS to human families to study the
underlying causes of de novo mutations has focused mostly
on measuring the increase in de novo mutations as function
of paternal or maternal age. These studies are nevertheless
important because they provide information on the baseline
levels of de novo mutations and how they change as a
function of age. Clearly, paternal age is a critical con-
founding factor that must be controlled for in any study
assessing the impact of environmental exposure in the
human population. Indeed, a recent statistical power analy-
sis showed that controlling for the paternal age effect and
modeling family-to-family variability signicantly reduced
the number of families needed to detect an increase in de
novo mutations (Webster et al. 2018). The analysis also
demonstrated that inclusion of families with multiple chil-
dren confers additional sensitivity to detect an effect. For
example, sequencing of just 6 four-child families per study
group provides an 80% power to detect a 30% increase in
de novo mutations (Webster et al. 2018). These data show
the feasibility of designing studies to evaluate the effects of
environmental exposures in human populations that do not
require large cohorts and signicant nancial investment. It
is easy to envision that these types of studies will become
common over the next few years and will provide funda-
mental data on the susceptibility of human germ cells to
environmental agents. One such an example is a recent
study that demonstrated a signicant correlation between
levels of paternal exposure to dioxin and incidence of de
novo SNVs in their offspring (Ton et al. 2018).
THE NEXT 50 YEARS: A CALL TO ACTION
Fifty years ago, the founding members of the EMGS
would not have predicted that half a century would pass and
we would still be waiting for the demonstration of the exis-
tence of human germ cell mutagens. Yet, it is a testament of
the rightfulness and importance of the original mission of
the Society that the pursuit of that goal has survived. During
these 50 years, incredible progress has been made in our
understanding of the mechanisms of germ cell mutagenesis
and in the tools that we have at our disposition for fullling
the original mission of our society. It is because of the
relentless effort of a small number of EMGS members who
have kept the germ cell mutagenesis ame alive that we are
experiencing now a renaissance of the eld.
Challenges still lay ahead. First and foremost, germ cell
mutagenesis is threatenedby the current push to limit,
reduce and replace in vivo studies and the increased empha-
sis on in vitro and computational approaches to assess the
toxicological properties of chemicals. Under this paradigm,
germ cells risk may be marginalized as in vitro germ cell
systems are still in their infancy, with a consequent reliance
on somatic systems to predict effects in germ cells. Further-
more, even if in vitro germ cell systems would be fully
developed, they would just provide demonstration that a
chemical can affect the germ cell genome while contribut-
ing little to the ultimate goal of germ cell mutagenesis: the
demonstration of the heritability of environmentally
induced de novo mutations.
We now have the tools and are closer than ever before
to investigating the best possible experimental system:
ourselves. The continuing decline in the costs of NGS
approaches and the availability of new technologies
that are directly applicable to humans and that enable
the detection of rare mutations among heterogeneous
populations of cells (Salk et al. 2018) makes it inevitable
that many more agents will be demonstrated to induce
mutations in human germ cells and increase adverse health
effects in the offspring. These error-reduced sequencing
technologies can be applied in any species/tissue and are
poised to revolutionize genotoxicity testing in general
(Salk and Kennedy 2020). Such technologies provide
more opportunities for any laboratory in the world to con-
duct germ cell mutagenicity assessment. It is our hope
that with the GHS strategy, and the European Union regu-
lation for the Registration, Evaluation, Authorisation and
Environmental and Molecular Mutagenesis. DOI 10.1002/em
50 Marchetti et al.
Restriction of Chemicals requiring germ cell mutation assess-
ment, in parallel with our increasing knowledge of the major
prevalence of human genetic disorders that result from de
novo mutations and the impact of even small increases in
mutation rates on population health, more laboratories around
the world will begin to contribute data to increase our under-
standing of candidate human germ cell mutagens.
We predict that we will soon have data demonstrating an
increase in de novo mutations in the offspring of exposed par-
ents. A great challenge is to be sufciently prepared to inter-
pret the coming outpouring of data and, together with the
regulatory community, develop a framework for managing,
communicating and mitigating the risk of exposure to human
germ cell mutagens. As indicated by DeMarini (2012), it is
time for the applied genetic toxicology community to formu-
late a plan to respond to the demonstration of environmentally
induced de novo mutations in the human population. We add
our voices to his call for the establishment of an international
body that is tasked with the mandate of reviewing the avail-
able data and formally declare whether there is sufcient evi-
dence for supporting the existence of human germ cell
mutagens and evaluate the risk of exposure to these agents for
the health of human populations.
ACKNOWLEDGMENTS
We thank Drs Marc Beal and Vinita Chauhan of Health
Canada for providing comments on the manuscript before
it was submitted. Funding for this work was provided by
Health Canadas Chemicals Management Plan and the
Genomics Research and Development Initiative.
AUTHORCONTRIBUTIONS
All authors contributed to the writing of the manuscript
and all authors agree to its publication.
REFERENCES
Adewoye AB, Lindsay SJ, Dubrova YE, Hurles ME. 2015. The genome-
wide effects of ionizing radiation on mutation induction in the
mammalian germline. Nat Commun 6:6684.
Adler ID. 1996. Comparison of the duration of spermatogenesis between
male rodents and humans. Mutat Res 352:169172.
Amann RP. 2008. The cycle of the seminiferous epithelium in humans: A
need to revisit? J Androl 29:469487.
Ames BN. 1973. Carcinogens are mutagens: Their detection and classica-
tion. Environ Health Perspect 6:115118.
Beal MA, Yauk CL, Marchetti F. 2017. From sperm to offspring:
Assessing the heritable genetic consequences of paternal smoking
and potential public health impacts. Mutat Res 773:2650.
Beal MA, Meier MJ, Williams A, Rowan-Carroll A, Gagné R,
Lindsay SJ, Fitzgerald T, Hurles ME, Marchetti F, Yauk CL. 2019.
Paternal exposure to benzo(a)pyrene induces genome-wide muta-
tions in mouse offspring. Commun Biol 2:228.
Bonilla E, Xu EY. 2008. Identication and characterization of novel mam-
malian spermatogenic genes conserved from y to human. Mol
Hum Reprod 14:137142.
Carvalho CM, Lupski JR. 2016. Mechanisms underlying structural variant
formation in genomic disorders. Nat Rev Genet 17:224238.
Chaisson MJP, Sanders AD, Zhao X, Malhotra A, Porubsky D, Rausch T,
Gardner EJ, Rodriguez OL, Guo L, Collins RL, et al. 2019. Multi-
platform discovery of haplotype-resolved structural variation in
human genomes. Nat Commun 10:1784.
Cimino MC. 2006. Comparative overview of current international strate-
gies and guidelines for genetic toxicology testing for regulatory
purposes. Environ Mol Mutagen 47:362390.
Cliet I, Melcion C, Cordier A. 1993. Lack of predictivity of bone marrow
micronucleus test versus testis micronucleus test: Comparison with
four carcinogens. Mutat Res 292:105111.
Committee 17. 1975. Environmental mutagenic hazards. Science 187:
503514.
Crow JF. 2000. The origins, patterns and implications of human spontane-
ous mutation. Nat Rev Genet 1:4047.
Czeizel AE, Elek C, Gundy S, Metneki J, Nemes E, Reis A, Sperling K,
Timar L, Tusnady G, Viragh Z. 1993. Environmental trichlorfon
and cluster of congenital abnormalities. Lancet 341:539542.
De Mas P, Daudin M, Vincent MC, Bourrouillou G, Calvas P,
Mieusset R, Bujan L. 2001. Increased aneuploidy in spermatozoa
from testicular tumour patients after chemotherapy with cisplatin,
etoposide and bleomycin. Hum Reprod 16:12041208.
Deareld KL, Douglas GR, Ehling UH, Moore MM, Sega GA,
Brusick DJ. 1995. Acrylamide: A review of its genotoxicity and an
assessment of heritable genetic risk. Mutat Res 330:7199.
Deareld KL, Gollapudk BB, Bemis JC, Benz RD, Douglas GR,
Elespuru RK, Johnson GE, Kirkland DJ, LeBaron MJ, Li AP,
et al. 2017. Next generation testing strategy for assessment of
genomic damage: A conceptual framework and considerations.
Environ Mol Mutagen 58:264283.
Deciphering Developmental Disorders Study. 2017. Prevalence and archi-
tecture of de novo mutations in developmental disorders. Nature
542:433438.
DeMarini DM. 2012. Declaring the existence of human germ-cell muta-
gens. Environ Mol Mutagen 53:166172.
DeMarini DM. 2020. The mutagenesis Moonshot: The propitious begin-
ning of the Environmental Mutagenesis and Genomcs Society.
Environ Mol Mutagen 61:824.
Derijck A, van der Heijden G, Giele M, Philippens M, de Boer P. 2008.
DNA double-strand break repair in parental chromatin of mouse
zygotes, the rst cell cycle as an origin of de novo mutation. Hum
Mol Genet 17:19221937.
Dubrova YE. 2002. Nuclear weapons tests and human germline mutation
rate. Science 295:10371037.
Dubrova YE, Jeffreys AJ, Malashenko AM. 1993. Mouse minisatellite
mutations induced by ionizing radiation. Nat Genet 5:9294.
Dubrova YE, Nesterov VN, Krouchinsky NG, Ostapenko VA,
Neumann R, Neil DL, Jeffreys AJ. 1996. Human minisatellite
mutation rate after the Chernobyl accident. Nature 380:683686.
Dubrova YE, Plumb M, Brown J, Jeffreys AJ. 1998. Radiation-induced
germline instability at minisatellite loci. Int J Radiat Biol 74:
689696.
Dubrova YE, Grant G, Chumak AA, Stezhka VA, Karakasian AN. 2002.
Elevated minisatellite mutation rate in the post-Chernobyl families
from Ukraine. Am J Hum Genet 71:801809.
Dubrova YE, Ploshchanskaya OG, Kozionova OS, Akleyev AV. 2006.
Minisatellite germline mutation rate in the Techa River population.
Mutat Res 602:7482.
Environmental and Molecular Mutagenesis. DOI 10.1002/em
51Quest for Human Germ Cell Mutagens
Erickson RP. 2014. Recent advances in the study of somatic mosaicism
and diseases other than cancer. Curr Opin Genet Dev 26:7378.
Francioli LC, Polak PP, Koren A, Menelaou A, Chun S, Renkens I,
Genome of the Netherlands Consortium, van Duijn CM, Swertz M,
Wijmenga C, et al. 2015. Genome-wide patterns and properties of
de novo mutations in humans. Nat Genet 47:822826.
Frias S, Van Hummelen P, Meistrich ML, Lowe XR, Hagemeister FB,
Shelby MD, Bishop JB, Wyrobek AJ. 2003. NOVP chemotherapy
for Hodgkins disease transiently induces sperm aneuploidies asso-
ciated with the major clinical aneuploidy syndromes involving
chromosomes X, Y, 18, and 21. Cancer Res 63:4451.
Gao Z, Moorjani P, Sasani TA, Pedersen BS, Quinlan AR, Jorde LB,
Amster G, Przeworski M. 2019. Overlooked roles of DNA damage
and maternal age in generating human germline mutations. Proc
Natl Acad Sci U S A 116:94919500.
Glen CD, Dubrova YE. 2012. Exposure to anticancer drugs can result in
transgenerational genomic instability in mice. Proc Natl Acad Sci
U S A 109:29842988.
Glen CD, Smith AG, Dubrova YE. 2008. Single-molecule PCR analysis
of germ line mutation induction by anticancer drugs in mice. Can-
cer Res 68:36303636.
Goschalk RWL, Yauk CL, van Benthem J, Douglas GR, Marchetti F.
2020. In utero exposure to genotoxins leading to genetic mosai-
cism: A forgotten window of susceptibility in genetic toxicology
testing? Environ Mol Mutagen 61:5565.
Green S, Auletta A, Fabricant J, Kapp R, Manandhar M, Sheu CJ,
Springer J, Whiteld B. 1985. Current status of bioassays in
genetic toxicologyThe dominant lethal assay. A report of the
U.S. Environmental Protection Agency Gene-Tox Program. Mutat
Res 154:4967.
Heich RH, Johnson GE, Zeller A, Marchetti F, Douglas GR, Witt KL,
Gollapudi BB, White PA. 2020. Mutation as a toxicological end-
point for regulatory decision-making. Environ Mol Mutagen 61:
3441.
Hermo L, Pelletier RM, Cyr DG, Smith CE. 2010. Surng the wave,
cycle, life history, and genes/proteins expressed by testicular germ
cells. Part 1: Background to spermatogenesis, spermatogonia, and
spermatocytes. Microsc Res Tech 73:241278.
IPCS. 1985. Guidelines for the Study of Genetic Effects in Human
Populations. Geneva: World Health Organization.
Jeng HA, Pan CH, Chao MR, Lin WY. 2015. Sperm DNA oxidative dam-
age and DNA adducts. Mutat Res 794:7582.
Johnson GE, Slob W, Doak SH, Fellows MD, Gollapudi BB, Heich RH,
Rees BJ, Soeteman-Hernández LG, Verma JR, Wills JW, et al.
2015. New approaches to advance the use of genetic toxicology
analyses for human risk assessment. Toxicol Res 4:667676.
Kong A, Frigge ML, Masson G, Besenbacher S, Sulem P, Magnusson G,
Gudjonsson SA, Sigurdsson A, Jonasdottir A, Jonasdottir A, et al.
2012. Rate of de novo mutations and the importance of fathers
age to disease risk. Nature 488:471475.
Lambert IB, Singer TM, Boucher SE, Douglas GR. 2005. Detailed review
of transgenic rodent mutation assays. Mutat Res 590:1280.
Levy-Sakin M, Pastor S, Mostovoy Y, Li L, Leung AKY, McCaffrey J,
Young E, Lam ET, Hastie AR, Wong KHY, et al. 2019. Genome
maps across 26 human populations reveal population-specic pat-
terns of structural variation. Nat Commun 10:1025.
Linschooten JO, Verhofstad N, Gutzkow K, Olsen AK, Yauk C,
Oligschlager Y, Brunborg G, van Schooten FJ, Godschalk RW.
2013. Paternal lifestyle as a potential source of germline mutations
transmitted to offspring. FASEB J 27:28732879.
Lupski JR. 2015. Structural variation mutagenesis of the human genome:
Impact on disease and evolution. Environ Mol Mutagen 56:419436.
Marchetti F, Wyrobek AJ. 2005. Mechanisms and consequences of
paternally-transmitted chromosomal abnormalities. Birth Defects
Res C Embryo Today 75:112129.
Marchetti F, Essers J, Kanaar R, Wyrobek AJ. 2007. Disruption of mater-
nal DNA repair increases sperm-derived chromosomal aberrations.
Proc Natl Acad Sci U S A 104:1772517729.
Marchetti F, Rowan-Carroll A, Williams A, Polyzos A, Berndt-Weis ML,
Yauk CL. 2011. Sidestream tobacco smoke is a male germ cell
mutagen. Proc Natl Acad Sci U S A 108:1281112814.
Marchetti F, Eskenazi B, Weldon RH, Li G, Zhang L, Rappaport SM,
Schmid TE, Xing C, Kurtovich E, Wyrobek AJ. 2012. Occupational
exposure to benzene and chromosomal structural aberrations in the
sperm of Chinese men. Environ Health Perspect 120:229234.
Marchetti F, Bishop J, Gingerich J, Wyrobek AJ. 2015. Meiotic interstrand
DNA damage escapes paternal repair and causes chromosomal
aberrations in the zygote by maternal misrepair. Sci Rep 5:7689.
Marchetti F, Aardema M, Beevers C, van Benthem J, Douglas GR,
Godschalk R, Yauk CL, Young R, Williams A. 2018a. Simulation
of mouse and rat spermatogenesis to inform genotoxicity testing
using OECD test guideline 488. Corrigendum: Mutat Res, 2019,
844:69. Mutat Res 832833:1928.
Marchetti F, Aardema MJ, Beevers C, van Benthem J, Godschalk R,
Williams A, Yauk CL, Young R, Douglas GR. 2018b. Identifying
germ cell mutagens using OECD test guideline 488 (transgenic
rodent somatic and germ cell gene mutation assays) and integration
with somatic cell testing. Corrigendum: Mutat Res, 2019, 844:
7071. Mutat Res 832833:718.
Maretty L, Jensen JM, Petersen B, Sibbesen JA, Liu S, Villesen P,
Skov L, Belling K, Theil Have C, Izarzugaza JMG, et al. 2017.
Sequencing and de novo assembly of 150 genomes from Denmark
as a population reference. Nature 548:8791.
Martin RH, Ernst S, Rademaker A, Barclay L, Ko E, Summers N. 1997.
Chromosomal abnormalities in sperm from testicular cancer
patients before and after chemotherapy. Hum Genet 99:214218.
Masumura K, Toyoda-Hokaiwado N, Ukai A, Gondo Y, Honma M,
Nohmi T. 2016a. Dose-dependent de novo germline mutations
detected by whole-exome sequencing in progeny of ENU-treated
male gpt delta mice. Mutat Res 810:3039.
Masumura K, Toyoda-Hokaiwado N, Ukai A, Gondo Y, Honma M,
Nohmi T. 2016b. Estimation of the frequency of inherited germline
mutations by whole exome sequencing in ethyl nitrosourea-treated
and untreated gpt delta mice. Genes Environ 38:10.
Mirkin SM. 2007. Expandable DNA repeats and human disease. Nature
447:932940.
Mori K, Weng SM, Arzberger T, May S, Rentzsch K, Kremmer E,
Schmid B, Kretzschmar HA, Cruts M, Van Broeckhoven C, et al.
2013. The C9orf72 GGGGCC repeat is translated into aggregating
dipeptide-repeat proteins in FTLD/ALS. Science 339:13351338.
Mulvihill JJ. 2012. Preconception exposure to mutagens: Medical and
other exposures to radiation and chemicals. J Community Genet 3:
205211.
Narod SA, Douglas GR, Nestmann ER, Blakey DH. 1988. Human muta-
gens: Evidence from paternal exposure? Environ Mol Mutagen 11:
401415.
Neel JV, Lewis SE. 1990. The comparative radiation genetics of humans
and mice. Annu Rev Genet 24:327362.
OBrien JM, Beal MA, Yauk CL, Marchetti F. 2016a. Benzo(a)pyrene is
mutagenic in mouse spermatogonial stem cells and dividing sper-
matogonia. Toxicol Sci 152:363371.
OBrien JM, Beal MA, Yauk CL, Marchetti F. 2016b. Next generation
sequencing of benzo(a)pyrene-induced lacZ mutants identies a
germ cell-specic mutation spectrum. Sci Rep 6:36743.
Environmental and Molecular Mutagenesis. DOI 10.1002/em
52 Marchetti et al.
OBrien JM, Walker M, Sivathayalan A, Douglas GR, Yauk CL,
Marchetti F. 2015. Sublinear response in lacZ mutant frequency of
Muta mouse spermatogonial stem cells after low dose subchronic
exposure to N-ethyl-N-nitrosourea. Environ Mol Mutagen 56:
347355.
OECD. 2013. Test 488: Transgenic Rodent Somatic and Germ Cells Gene
Mutation Assays. Paris: OECD Publishing.
Oliveri Conti G, Calogero AE, Giacone F, Fiore M, Barchitta M,
Agodi A, Ferrante M. 2017. B(a)P adduct levels and fertility: A
crosssectional study in a Sicilian population. Mol Med Rep 15:
33983404.
Olsen AK, Lindeman B, Wiger R, Duale N, Brunborg G. 2005. How do
male germ cells handle DNA damage? Toxicol Appl Pharmacol
207:521531.
Olsen AK, Andreassen A, Singh R, Wiger R, Duale N, Farmer PB,
Brunborg G. 2010. Environmental exposure of the mouse germ
line: DNA adducts in spermatozoa and formation of de novo muta-
tions during spermatogenesis. PLoS One 5:e11349.
Pacchierotti F, Masumura K, Eastmond D, Elhajouji A, Froetschl R,
Kirsch-Volders M, Lynch AM, Schuler M, Tweats DJ,
Marchetti F. 2019. 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 heriditary diseases. Mutat
Res https://doi.org/10.1016/j.mrgentox.2019.02.004. (in press).
Rahbari R, Wuster A, Lindsay SJ, Hardwick RJ, Alexandrov LB,
Turki SA, Dominiczak A, Morris A, Porteous D, Smith B, et al.
2016. Timing, rates and spectra of human germline mutation. Nat
Genet 48:126133.
Robbins WA, Meistrich ML, Moore D, Hagemeister FB, Weier HU,
Cassel MJ, Wilson G, Eskenazi B, Wyrobek AJ. 1997. Chemother-
apy induces transient sex chromosomal and autosomal aneuploidy
in human sperm. Nat Genet 16:7478.
Rowan-Carroll A, Beal MA, Williams A, Marchetti F, Yauk CL. 2017.
Dose-response mutation and spectrum analyses reveal similar
responses at two microsatellite loci in benzo(a)pyrene-exposed
mouse spermatogonia. Mutagenesis 32:463470.
Russell LB. 2004. Effects of male germ-cell stage on the frequency,
nature, and spectrum of induced specic-locus mutations in the
mouse. Genetica 122:2536.
Russell WL, Bangham JW, Russell LB. 1998. Differential response of
mouse male germ-cell stages to radiation-induced specic-locus
and dominant mutations. Genetics 148:15671578.
Salk JJ, Kennedy SR. 2020. Next-generation genotoxicology: Using mod-
ern sequencing technologies to study carcinogens and somatic
mutagenesis. Environ Mol Mutagen 61:135151.
Salk JJ, Schmitt MW, Loeb LA. 2018. Enhancing the accuracy of next-
generation sequencing for detecting rare and subclonal mutations.
Nat Rev Genet 19:269285.
Schull WJ. 2003. The children of atomic bomb survivors: A synopsis.
J Radiol Prot 23:369384.
Secretan B, Straif K, Baan R, Grosse Y, El Ghissani F, Bouvard V,
Benbrahim-Tallaa L, Guha N, Freeman C, Galichet L, et al. 2009. A
review of human carcinogens. Part E: Tobacco smoke, areca nut,
alcohol, coal smoke, and salted sh. Lancet Oncol 10:10331034.
Singer TM, Yauk CL. 2010. Germ cell mutagens: Risk assessment chal-
lenges in the 21st century. Environ Mol Mutagen 51:919928.
Somers CM, Yauk CL, White PA, Parfett CL, Quinn JS. 2002. Air pollu-
tion induces heritable DNA mutations. Proc Natl Acad Sci U S A
99:1590415907.
Somers CM, McCarry BE, Malek F, Quinn JS. 2004. Reduction of partic-
ulate air pollution lowers the risk of heritable mutations in mice.
Science 304:10081010.
Stenson PD, Mort M, Ball EV, Evans K, Hayden M, Heywood S,
Hussain M, Phillips AD, Cooper DN. 2017. The human gene
mutation database: Towards a comprehensive repository of
inherited mutation data for medical research, genetic diagnosis and
next-generation sequencing studies. Hum Genet 136:665677.
Tempest HG, Ko E, Chan P, Robaire B, Rademaker A, Martin RH. 2008.
Sperm aneuploidy frequencies analysed before and after chemo-
therapy in testicular cancer and Hodgkins lymphoma patients.
Hum Reprod 23:251258.
Ton ND, Nakagawa H, Ha NH, Duong NT, Nhung VP, Hien LTT,
Hue HTT, Hoang NH, Wong JH, Nakano K, et al. 2018. Whole
genome sequencing and mutation rate analysis of trios with pater-
nal dioxin exposure. Hum Mutat 39:13841392.
United Nations. 2017. Globally Harmonized System of Classication and
Labelling of Chemicals (GHS), 7th revised ed. New York and
Geneva: United Nations.
Vangompel MJ, Xu EY. 2011. The roles of the DAZ family in spermato-
genesis: More than just translation? Spermatogenesis 1:3646.
Verhofstad N, van Oostrom CT, Zwart E, Maas LM, van Benthem J, van
Schooten FJ, van Steeg H, Godschalk RW. 2011. Evaluation of
benzo(a)pyrene-induced gene mutations in male germ cells.
Toxicol Sci 119:218223.
Vilarino-Guell C, Smith AG, Dubrova YE. 2003. Germline mutation
induction at mouse repeat DNA loci by chemical mutagens. Mutat
Res 526:6373.
Webster RJ, Williams A, Marchetti F, Yauk CL. 2018. Discovering human
germ cell mutagens with whole genome sequencing: Insights from
power calculations reveal the importance of controlling for
between-family variability. Mutat Res 831:2432.
White-Cooper H, Bausek N. 2010. Evolution and spermatogenesis. Philos
Trans R Soc Lond B Biol Sci 365:14651480.
Wills JW, Long AS, Johnson GE, Bemis JC, Dertinger SD, Slob W,
White PA. 2016. Empirical analysis of BMD metrics in genetic
toxicology, Part II: in vivo potency comparisons to promote reduc-
tions in the use of experimental animals for genetic toxicity assess-
ment. Mutagenesis 31:265275.
Witt KL, Hughes LA, Burka LT, McFee AF, Mathews JM, Black SL,
Bishop JB. 2003. Mouse bone marrow micronucleus test results do
not predict the germ cell mutagenicity of N-hydroxymethylacrylamide
in the mouse dominant lethal assay. Environ Mol Mutagen 41:
111120.
World Health Organization. 1986. Prevention of avoidable diseases: Mem-
orandum from a WHO meeting. Bull World Health Organ 64:
205216.
Wyrobek AJ, Schmid TE, Marchetti F. 2005. Cross-species sperm-FISH
assays for chemical testing and assessing paternal risk for chro-
mosomally abnormal pregnancies. Environ Mol Mutagen 45:
271283.
Xia Y, Bian Q, Xu L, Cheng S, Song L, Liu J, Wu W, Wang S, Wang X.
2004. Genotoxic effects on human spermatozoa among pesticide
factory workers exposed to fenvalerate. Toxicology 203:4960.
Xing C, Marchetti F, Li G, Weldon RH, Kurtovich E, Young S,
Schmid TE, Zhang L, Rappaport S, Waidyanatha S, et al. 2010.
Benzene exposure near the U.S. permissible limit is associated with
sperm aneuploidy. Environ Health Perspect 118:833839.
Xu DX, Zhu QX, Zheng LK, Wang QN, Shen HM, Deng LX, Ong CN.
2003. Exposure to acrylonitrile induced DNA strand breakage and
sex chromosome aneuploidy in human spermatozoa. Mutat Res
537:93100.
Xu G, McMahan CA, Walter CA. 2014. Early-life exposure to benzo[a]
pyrene increases mutant frequency in spermatogenic cells in adult-
hood. PLoS One 9:e87439.
Environmental and Molecular Mutagenesis. DOI 10.1002/em
53Quest for Human Germ Cell Mutagens
Yauk CL, Quinn JS. 1996. Multilocus DNA ngerprinting reveals high rate
of heritable genetic mutation in herring gulls nesting in an industrial-
ized urban site. Proc Natl Acad Sci U S A 93:1213712141.
Yauk CL, Fox GA, McCarry BE, Quinn JS. 2000. Induced minisatellite
germline mutations in herring gulls (Larus argentatus) living near
steel mills. Mutat Res 452:211218.
Yauk CL, Berndt ML, Williams A, Rowan-Carroll A, Douglas GR,
Stampi MR. 2007. Mainstream tobacco smoke causes paternal
germ-line DNA mutation. Cancer Res 67:51035106.
Yauk C, Polyzos A, Rowan-Carroll A, Somers CM, Godschalk RW,
Van Schooten FJ, Berndt ML, Pogribny IP, Koturbash I, Williams A,
et al. 2008. Germ-line mutations, DNA damage, and global
hypermethylation in mice exposed to particulate air pollution in an
urban/industrial location. Proc Natl Acad Sci U S A 105:605610.
Yauk CL, Lucas Argueso J, Auerbach SS, Awadalla P, Davis SR,
Demarini DM, Douglas GR, Dubrova YE, Elespuru RK,
Glover TW, et al. 2013. Harnessing genomics to identify environ-
mental determinants of heritable disease. Mutat Res 752:69.
Yauk CL, Aardema MJ, Benthem J, Bishop JB, Deareld KL,
DeMarini DM, Dubrova YE, Honma M, Lupski JR, Marchetti F,
et al. 2015a. Approaches for identifying germ cell mutagens:
Report of the 2013 IWGT workshop on germ cell assays. Mutat
Res 783:3654.
Yauk CL, Lambert IB, Meek ME, Douglas GR, Marchetti F. 2015b.
Development of the adverse outcome pathway "alkylation of DNA
in male premeiotic germ cells leading to heritable mutations" using
the OECDs usershandbook supplement. Environ Mol Mutagen
56:724750.
Accepted by
R. Heflich
Environmental and Molecular Mutagenesis. DOI 10.1002/em
54 Marchetti et al.
... Such pollution is known to result in respiratory illnesses in humans 12 , reduced photosynthesis and cell damage in plants 13 , higher mortality in fishes and amphibians 14 , and decreased fledgling success in birds 15 . Exposure to some pollutants can damage DNA and induce de novo mutations (hereafter simply called 'mutations') [16][17][18][19] . Although carcinogenic pollutants are known to cause somatic mutations (mutations in non-reproductive germ cell tissue), the fitness effects of these mutations and the prevalence of pollution-induced germline mutations are poorly understood outside of laboratory settings. ...
... These genotoxicants have the potential to impact the somatic cell mutation burden, contributing to the decreased health of individuals and populations 18,79 . The vast majority of mutagenicity testing is conducted in the laboratory on individual chemicals at high doses 19 , leading to a major gap in our understanding of how lifelong, low-dose exposures to mixtures of mutagens affect mutation rates and disease outcomes. Moreover, the complex interactions between sociodemographic factors and mutagenic environmental mixtures inherent to cities have yet to be explored. ...
... Laboratory and field studies suggest that exposure to many common urban pollutants can induce germline mutations. For example, over 80 chemical agents have been identified as germline mutagens in laboratory mice 19 . In humans, the best evidence of the impact of pollutants on germ cell mutagenesis comes from studies demonstrating an increased incidence of chromosomal abnormalities in human sperm 19 . ...
... Bernard Robaire, PhD, a noted researcher of male reproduction, recently remarked in a Biology of Reproduction interview that not long ago grant reviewers considered only the intrauterine environment as a major external influence on the development of offspring, with comments along the lines of "there is no way that drugs given pre-conceptionally to men could also affect their childrenthat's a maternal issue!" [3]. Further, in his opening keynote talk at the Beyond Genes conference, David DeMarini, PhD, a genetic toxicologist recently retired from the U.S. Environmental Protection Agency, explained that despite the strong likelihood of links between exposures, germ cell perturbation and offspring pathology, little attention is paid to this dimension of risk from a regulatory perspective [4][5][6][7]. In short, with the long-prevailing paradigm of "genes or somatic environment" dominating thinking about origins of human pathology, the concept of the germline exposome playing a possible role has become something of a scientific orphan. ...
... Considering that humans have such a broad variety of exposures, including stress, trauma, famine, malnutrition, diet, aging, illness, infection, pharmaceutical drugs, recreational drugs, surgery and medical procedures, smoking, pollution, endocrine disrupting chemicals, industrial chemicals, pesticides, radiation, heavy metals, wartime exposures, and more, how can we determine what exposures to germline warrant serious scientific investigation? An effort to find the needles in the haystack may be guided to a degree by the mammalian experiments, but those studies are somewhat sporadic and not at all systematic, and though high-throughput efforts may be on the horizon [6], they will miss epigenetic effects and phenotypic outcomes. This is where the work of actually surveying families comes in. ...
Article
Throughout the scientific literature, heritable traits are routinely presumed to be genetic in origin. However, as emerging evidence from the realms of genetic toxicology and epigenomics demonstrate, heritability may be better understood as encompassing not only DNA sequence passed down through generations, but also disruptions to the parental germ cells causing de novo mutations or epigenetic alterations, with subsequent shifts in gene expression and functions in offspring. The Beyond Genes conference highlighted advances in understanding these aspects at molecular, experimental and epidemiological levels. In this commentary I suggest that future research on this topic could be inspired by collecting parents’ germ cell exposure histories, with particular attention to cases of families with multiple children suffering idiopathic disorders. In so doing I focus on the endpoint of autism spectrum disorders (ASD). Rates of this serious neurodevelopment disability have climbed around the world, a growing crisis that cannot be explained by diagnostic shifts. ASD’s strong heritability has prompted a research program largely focused on DNA sequencing to locate rare and common variants, but decades of this gene-focused research have revealed surprisingly little about the molecular origins of the disorder. Based on my experience as the mother of two children with idiopathic autism, and as a research philanthropist and autism advocate, I suggest ways researchers might probe parental germ cell exposure histories to develop new hypotheses that may ultimately reveal sources of non-genetic heritability in a subset of idiopathic heritable pathologies.
... Unfortunately, to our knowledge no such studies have yet been conducted in humans, either prospectively or retrospectively. Although there is some, if insufficient, regulatory concern for pharmaceutically induced germ cell mutagesis [96], potential germline modification via other mechanisms, for any dose, combination or concentration, and during any window of exposure, is not the target of US Food and Drug Administration regulatory effort or risk assessment [33], and GA is no exception. ...
... As autism rates continue to climb, now reaching an estimated 1 in 54 US children according to the Centers for Disease Control [31], the dominance of over-simplistic assumptions about molecular heritability is thwarting desperately needed progress, and based on the literature and our own observations in the community we can think of no other germ cell exposure GA, germ cells and the missing heritability of autism | 5 that deserves more attention than GA, owing to its potent ability to disrupt the proper expression of genes important for early brain development. This work is by nature interdisciplinary, and we echo the words of renowned genetic toxicologists who recently stated, 'collaborative work between clinicians, epidemiologists, genetic toxicologists, genomics experts and bioinformaticians [is needed] to precisely define how environmental exposures impact germ cell genomes' [96]. ...
Article
Full-text available
Agents of general anesthesia (GA) are commonly employed in surgical, dental and diagnostic procedures to effectuate global suppression of the nervous system, but in addition to somatic targets, the subject’s germ cells—from the embryonic primordial stage to the mature gametes—may likewise be exposed. Although GA is generally considered safe for most patients, evidence has accumulated that various compounds, in particular the synthetic volatile anesthetic gases (SVAGs) such as sevoflurane, can exert neurotoxic, genotoxic and epigenotoxic effects, with adverse consequences for cellular and genomic function in both somatic and germline cells. The purpose of this paper is to review the evidence demonstrating that GA, and in particular, SVAGs, may in some circumstances adversely impact the molecular program of germ cells, resulting in brain and behavioral pathology in the progeny born of the exposed cells. Further, we exhort the medical and scientific communities to undertake comprehensive experimental and epidemiological research programs to address this critical gap in risk assessment.
... The ability of xenobiotics to induce mutations, encompassing everything from single nucleotide changes in the DNA sequence to large chromosomal alterations, is a required component of any comprehensive toxicology assessment submitted to regulatory agencies for review and approval of a new product [1][2][3][4]. Somatic mutagenesis is a hallmark of many human diseases including cancer [5][6][7] and certain aging syndromes [8], while DNA changes in germ cells can result in a broad spectrum of hereditary genetic diseases [9][10][11][12]. Therefore, determining whether a new drug or chemical can cause mutagenesis or genomic instability is an important component of human and environmental health risk assessment (Fig. 1). ...
Article
Full-text available
Error-corrected Next Generation Sequencing (ecNGS) is rapidly emerging as a valuable, highly sensitive and accurate method for detecting and characterising 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.
... Moreover there is increased concern of potential harmful implications of preconception exposure of gametes to genotoxic substances (Brevik et al., 2012;Marcho et al., 2020;Zhang et al., 2020), as DNA damage is implicated in a plethora of adverse effects, of particular importance in germ cells. Exposure to a genotoxic agent can lead to mutations that can be transferred to the offspring, contributing to genetic disease, and impacting the health of coming generations (Dearfield et al., 2002;Griffiths et al., 2000;Marchetti et al., 2020;Yauk et al., 2015). ...
Article
Full-text available
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) modelling 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. This article is protected by copyright. All rights reserved.
... However, it is unclear whether these traits are either exaptations or novel adaptations to the urban environment [10,17]. In fact, although most adaptations arise from selection on standing genetic variation [18], we know little about how cities influence novel mutation or their long-term effects on organism health [19], despite the importance of mutation in contributing to evolutionary potential. ...
Article
Research on the evolutionary ecology of urban areas reveals how human-induced evolutionary changes affect biodiversity and essential ecosystem services. In a rapidly urbanizing world imposing many selective pressures, a time-sensitive goal is to identify the emergent issues and research priorities that affect the ecology and evolution of species within cities. Here, we report the results of a horizon scan of research questions in urban evolutionary ecology submitted by 100 interdisciplinary scholars. We identified 30 top questions organized into six themes that highlight priorities for future research. These research questions will require methodological advances and interdisciplinary collaborations, with continued revision as the field of urban evolutionary ecology expands with the rapid growth of cities.
... One of the major current gaps of genotoxic and mutagenic assessments is the lack of protocols for germ cell evaluation, especially because the responses of somatic and germ cells to stressors may be distinct given the unique aspects of spermatogenesis and oogenesis (Ferreira and Allard, 2015;Yauk et al., 2015). In addition, mutagenic impacts on germ cells may have greater consequences than on somatic cells, since a single mutation in a germ cell will produce an individual who has this mutation in all cells (Marchetti et al., 2020). Most of the protocols to evaluate mutagenicity on germ cells were established for vertebrates, usually mice, often using many organisms in each assay. ...
Article
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.
... 34 Furthermore, SCSA has been suggested to be a suitable method to study DNA fragmentation due to chemical exposure 35 that is mutagenic to male germ cells. 36 This suggestion is supported by studies reporting that mutagenic exposure increases sperm DNA fragmentation according to both SCSA and the TUNEL assay, 37 similar to the increase seen by SCSA after tobacco smoke exposure, 38 which as mentioned has also been suggested to be mutagenic to human germ cells, [39][40][41] and reported to give rise to mutations in both sperm 42 and spermatogonial stem cells 43 of mice. Accordingly, the aim of this study was to investigate whether urinary levels of 1-OHP, 2-OHPH and cotinine, as biomarkers of exposure to PAH and nicotine, were associated with sperm DNA fragmentation in Swedish men from the general population. ...
Article
Full-text available
Background: Tobacco smoking has been reported to cause DNA fragmentation and has been suggested to cause mutations in spermatozoa. These effects have been ascribed to the action of polycyclic aromatic hydrocarbons (PAH) present in the smoke. Simultaneously, DNA fragmentation has been associated with mutagenesis. Objective: The aim of this study was to investigate whether levels of urinary biomarkers of PAH and nicotine exposure were associated with sperm DNA fragmentation. Materials/methods: In the urine of 381 men recruited from two cohorts of young men (17-21 years old) from the general Swedish population, the PAH metabolites 1-hydroxypyrene and 2-hydroxyphenanthrene, as well as the nicotine metabolite cotinine, were measured. The sperm DNA fragmentation index (DFI) was analysed using a Sperm Chromatin Structure Assay. Associations between the DFI, and PAH metabolite levels as continuous variables as well as in quartiles, were studied by general linear models adjusted for abstinence time. A similar analysis was carried out for cotinine levels, according to which the men were categorised as "non-smoking" (N = 216) and "smoking" (N = 165). Results: No association was found between levels of any of the three biomarkers and DFI, either as a continuous variable (p = 0.87 - 0.99), or when comparing the lowest and the highest quartiles (p = 0.11 - 0.61). The same was true for comparison of men categorised as non-smoking or smoking (DFI 11.1% vs 11.8%, p = 0.31). Discussion: We found no evidence of PAH or nicotine exposure to be associated with DFI, which does not exclude that these exposures may have other effects on sperm DNA. Conclusion: In these young men, levels of biomarkers of nicotine and PAH exposure were not associated with DFI. This article is protected by copyright. All rights reserved.
... Given the far-reaching health implications of germ cell mutations and other genomic changes (Stenson et al. 2017), identifying chemical agents that may produce such effects should be of high importance to regulatory agencies. However, standard toxicity testing paradigms are generally poorly designed to fully capture perturbations to germ cells that could result in heritable effects (Marchetti et al. 2020). This is in large part due to the limited window of development to which germ cells are typically exposed during testing, in addition to the fact that many assays expose only male germ cells, thus ignoring female germ 1. Somatic adverse effects from smoking are well established (e.g., cancer), but germ cell effects and effects on heritability awareness is very low 2. Reason for workshop-Examine the evidence that exposure to tobacco smoke is a heritable concern, that is, to focus on heritable effects of germ cell exposures 3. Brought together a diverse group of stakeholders (researchers, regulators, public interest groups, bioethicists) to share information 4. Goal is to provide quantitative data to model the potential risk levels of substances that induce genomic damage and contribute to human adverse health outcomes 5. ...
Article
Full-text available
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.
Article
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.
Article
Full-text available
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. This article is protected by copyright. All rights reserved.
Article
Full-text available
Mutations have a profound effect on human health, particularly through an increased risk of carcinogenesis and genetic disease. The strong correlation between mutagenesis and carcinogenesis has been a driving force behind genotoxicity research for more than 50 years. The stochastic and infrequent nature of mutagenesis makes it challenging to observe and to study. Indeed, decades have been spent developing increasingly sophisticated assays and methods to study these low frequency genetic errors, in hopes of better predicting which chemicals may be carcinogens, understanding their mode of action, and informing guidelines to prevent undue human exposure. While effective, widely used genetic selection‐based technologies have a number of limitations that have hampered major advancements in the field of genotoxicity. Emerging new tools, in the form of enhanced next generation sequencing platforms and methods, are changing this paradigm. In this review, we discuss rapidly evolving sequencing tools and technologies, such as error‐corrected sequencing and single cell analysis, that we anticipate will fundamentally reshape the field. In addition, we consider a variety emerging applications for these new technologies, including the detection of DNA adducts, inference of mutational processes based on genomic site and local sequence contexts, and evaluation of genome engineering fidelity, as well as other cutting‐edge challenges for the next 50 years of environmental and molecular mutagenesis research. This article is protected by copyright. All rights reserved.
Article
Full-text available
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.
Article
Full-text available
Significance More than three-fourths of human germline mutations are paternal in origin and their total number increases with the father’s age at conception. These observations are thought to support the textbook view that germline point mutations stem mostly from DNA replication errors. Analyzing large germline mutation datasets for humans, we find that this understanding cannot explain the observed patterns of new mutations. Instead, we show that the male mutation bias is not driven by spermatogenesis. We further find evidence that a substantial fraction of mutations are not replicative in origin and uncover a potential effect of a mother’s age on the number of mutations that happen early in the development of the embryo.
Article
Full-text available
Large structural variants (SVs) in the human genome are difficult to detect and study by conventional sequencing technologies. With long-range genome analysis platforms, such as optical mapping, one can identify large SVs (>2 kb) across the genome in one experiment. Analyzing optical genome maps of 154 individuals from the 26 populations sequenced in the 1000 Genomes Project, we find that phylogenetic population patterns of large SVs are similar to those of single nucleotide variations in 86% of the human genome, while ~2% of the genome has high structural complexity. We are able to characterize SVs in many intractable regions of the genome, including segmental duplications and subtelomeric, pericentromeric, and acrocentric areas. In addition, we discover ~60 Mb of non-redundant genome content missing in the reference genome sequence assembly. Our results highlight the need for a comprehensive set of alternate haplotypes from different populations to represent SV patterns in the genome.
Article
Full-text available
The Organisation for Economic Co-operation and Development Test Guideline (TG) 488 for the transgenic rodent (TGR) mutation assay recommends two sampling times for assessing germ cell mutagenicity following the required 28-day exposure period: 28 + > 49 days for mouse sperm and 28 + >70 days for rat sperm from the cauda epididymis, or three days (i.e., 28 + 3d) for germ cells from seminiferous tubules (hereafter, tubule germ cells) plus caudal sperm for mouse and rat. Although the latter protocol is commonly used for mutagenicity testing in somatic tissues, it has several shortcomings for germ cell testing because it provides limited exposure of the proliferating phase of spermatogenesis when mutations are fixed in the transgene. Indeed, analysis of sperm at 28 + 3d has generated negative results with established germ cell mutagens, while the analysis of tubule germ cells has generated both positive and either negative or equivocal results. The Germ Cell workgroup of the Genetic Toxicology Technical Committee of the Health and Environmental Sciences Institute modelled mouse and rat spermatogenesis to better define the exposure history of the cell population collected from seminiferous tubules. The modelling showed that mouse tubule germ cells at 28 + 3d receive, as a whole, 42% of the total exposure during the proliferating phase. This percentage increases to 99% at 28 + 28d and reaches 100% at 28 + 30d. In the rat, these percentages are 22% and 80% at 28 + 3d and 28 + 28d, reaching 100% at 28 + 44d. These results show that analysis of tubule germ cells at 28 + 28d may be an effective protocol for assessing germ cell mutagenicity in mice and rats using TG 488. Since TG 488 recommends the 28 + 28d protocol for slow dividing somatic tissues, this appears to be a better compromise than 28 + 3d when slow dividing somatic tissues or germ cells are the critical tissues of interest.
Article
A mutagenesis moonshot addressing the influence of the environment on our genetic wellbeing was launched just two months before astronauts landed on the moon. Its impetus included the discovery that X‐rays (Muller, 1927) and chemicals (Auerbach and Robson, 1947) 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 multi‐disciplinary 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. This article is protected by copyright. All rights reserved.
Article
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.
Article
2,3,7,8‐Tetrachlorodibenzo‐p‐dioxin (TCDD) or dioxin, is commonly considered the most toxic man‐made substance. Dioxin exposure impacts human health and diseases, birth defects and teratogenesis were frequently observed in children of persons who have been exposed to dioxin. However, the impact of dioxin on human mutation rate in trios has not yet been elucidated in the whole genome level. To identify and characterize the genetic alterations in the individuals exposed to dioxin, we perfomed whole genome sequencing (WGS) of nine Vietnamese trios whose fathers were exposed to dioxin. In total, 846 de novo point mutations, 26 de novo insertions and deletions, 4 de novo structural variations, and 1 de novo copy number variation were identified. The number of point mutations and dioxin concentrations were positively correlated (P‐value < 0.05). Considering the substitution pattern, the number of A > T/T > A mutation and the dioxin concentration was positively correlated (P‐value < 0.05). Our analysis also identified one possible disease‐related mutation in LAMA5 in one trio. These findings suggested that dioxin exposure might affect father genomes of trios leading to de novo mutations in their children. Further analysis with larger sample sizes would be required to better clarify mutation rates and substitution patterns in trios caused by dioxin. This article is protected by copyright. All rights reserved