You are on page 1of 8

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/321913698

Teratogenesis

Article · December 2017


DOI: 10.1002/9780470015902.a0026056

CITATION READS

1 13,801

2 authors:

Neil Vargesson Lucas Rosa Fraga


University of Aberdeen Universidade Federal do Rio Grande do Sul
97 PUBLICATIONS   2,474 CITATIONS    53 PUBLICATIONS   245 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Thalidomide View project

All content following this page was uploaded by Neil Vargesson on 14 May 2019.

The user has requested enhancement of the downloaded file.


Teratogenesis Introductory article

Neil Vargesson, School of Medicine, Medical Sciences and Nutrition, Institute of Article Contents
• Introduction
Medical Sciences, University of Aberdeen, Aberdeen, UK
• How Are Teratogenic Events Studied?
Lucas Fraga, School of Medicine, Medical Sciences and Nutrition, Institute of Med- • Experimental Teratology

ical Sciences, University of Aberdeen, Aberdeen, UK • Conclusions

Online posting date: 15th December 2017

Teratogenesis is a process that causes birth defects Molecular Genetics of Human Congenital Limb Malforma-
or malformations in an embryo or foetus. Tera- tions). The causes of birth defects can be genetic, chromosomal
tology is the study of the causes and underlying or environmental (e.g. drugs), although sadly in many cases, we
mechanisms leading to birth defects or malfor- do not know the cause of the defect.
mations. These may include disorders without any Any agent that can cause a birth defect or malformation is
termed a teratogen. Many developmental biologists study ter-
obvious structural malformations, such as intellec-
atogens and how they cause malformations (teratogenesis) using
tual disabilities. A teratogen is a substance (from
animal models that display a similar defect to that seen in humans
outside the body) that causes birth defects or (Gilbert-Barness, 2010; Cassina et al., 2012; Schardein, 2000;
malformations. Examples of teratogens include Tables 1 and 2).
medicinal drugs, such as thalidomide; environ- Many factors have been linked to causing teratogenesis, which
mental toxins, for example cadmium as well as include the following:
environmental pollutants, including pesticides and
endocrine-disrupting compounds. Other causes of • Chemical agents, such as medicinal drugs, for example
thalidomide and retinoic acid; recreational drugs, for
teratogenesis include viruses, for example rubella
example alcohol and cocaine; environmental toxins, for
and Zika virus; physical compression in utero and
example heavy metals such as cadmium and environ-
poor diet. Animal models are used to study the mental pollutants, for example pesticides that are linked
mechanisms by which teratogens result in birth to reproductive and fertility malformations, for example
defects or malformations, and these studies can diethylstilbestrol in pregnancy.
also give insights into normal development. The • Infections such as rubella and Zika virus.
study and understanding of teratogenesis is also • Physical restraint or in utero damage – for example due to
essential for making safer and more targeted ther- oligohydramnios, where the amniotic fluid is lost causing the
apeutic drugs. foetus to be restrained – and clubfoot can result from this.
Amniotic band syndrome is an example of in utero physical
damage due to the wrapping of strands of the amnion around
body parts of the foetus and causing damage or, in severe
Introduction cases, death.
• Hyperthermia.
Teratology is the study of the mechanisms resulting in birth • Maternal conditions such as pre-eclampsia and gestational
defects and malformations caused by environmental factors, that diabetes.
is nongenetic. The origins of malformation have been studied
since Greek times, and the term ‘teratology’ derives from Greek The study of teratogenesis is also very important with regard
and translates broadly to the ‘study of monsters’. to drug safety – it is vitally important to ensure that drugs are
Given the incredible and complex events that underlie the safe and/or do not cause birth defects that is that they are non-
amazing journey the fertilised egg must go through to produce teratogenic. To this end, the lesson of thalidomide provides an
an embryo and then a foetus, it is amazing that any baby is born excellent example (Vargesson, 2015a; see also: Thalidomide
normally. Around 3% of births suffer from some sort of birth and Birth Defects). Briefly, thalidomide was prescribed as a non-
defects/malformations (Sadler, 2012; Schardein, 2000; also see barbiturate, nonaddictive sedative between 1957 and 1962 and
http://www.cdc.gov/ncbddd/birthdefects/features/birthdefects- given to pregnant women to prevent morning sickness. Sadly,
keyfindings.html) (see also: Birth Defects: Overview; the drug was potently teratogenic, resulting in more than 10 000
children being born with severe birth defects and countless mis-
eLS subject area: Developmental Biology carriages and stillbirths (Smithells and Newman, 1992; Varges-
How to cite: son, 2015b). The thalidomide made it clear that the effects drugs
Vargesson, Neil and Fraga, Lucas (December 2017) have upon the embryo/foetus are far from understood. Moreover,
Teratogenesis. In: eLS. John Wiley & Sons, Ltd: Chichester. the disaster confirmed that drugs can and do cross the placenta
DOI: 10.1002/9780470015902.a0026056 (Vargesson, 2013, 2015b). Widikund Lenz famously determined

eLS © 2017, John Wiley & Sons, Ltd. www.els.net 1


Teratogenesis

Table 1 Examples of teratogens known to cause human birth defects


Agents Malformations
Drugs
Alcohol Foetal alcohol syndrome, intrauterine growth retardation and intellectual disability
Cocaine Intrauterine growth retardation, prematurity, urogenital anomalies and brain developmental
disorders
Diethylstilbestrol (DES) Abnormalities of the reproductive tract
Retinoic acid Neural tube defects, craniofacial anomalies, cleft palate and cardiovascular anomalies
Tetracycline Teeth anomalies and damage
Thalidomide Abnormal limb development – typically shortening and loss of proximal bones, facial
anomalies, systemic anomalies including urogenital, cardiac and gastrointestinal tract
Valproic acid Craniofacial anomalies, neural tube defects and heart and skeletal defects
Warfarin Craniofacial anomalies and intellectual disability
Chemicals
Mercury Brain developmental disorders
Cadmium Limb anomalies, cardiovascular anomalies and neural tube defects
Polychlorinated biphenyls (PCBs) Intrauterine growth retardation
Infections
Cytomegalovirus Brain and sensory anomalies
HIV Growth failure, microcephaly and craniofacial defects
Rubella Intrauterine growth retardation, cardiac defects, deafness, eye defects and intellectual
disability
Zika Microcephaly
Ionising radiation Microcephaly, growth delays, skeletal development anomalies and mental retardation

Table 2 Selection of teratogens and their resulting malformations that have been studied in animals
Teratogen Models Malformations References
Thalidomide Primates, rabbits, marsupials, rodents, Limb defects, facial defects, spina Vargesson (2013)
chickens, marine fish and zebrafish bifida, microphthalmia and internal
organ abnormalities
Valproate Xenopus, zebrafish, mice and chicken Growth retardation and eyes, heart, tail Gilbert-Barness (2010); Whitsel
and neural tube defects et al. (2002)
Cadmium Rodents, zebrafish, lizards and chicken Limbs’ malformation and abnormalities Yamamoto et al. (2012);
in heart, vasculature and liver, neural Cullinane et al. (2009)
tube, somites and reproductive
system
Mercury Cats Brain damage Gilbert-Barness (2010)
Zika virus Mice and chicken Mortality, growth retardation, ocular Cugola et al. (2016); Goodfellow
malformations and microcephaly et al. (2016); Miner et al.
(2016)
Alcohol Chicken and fish Mortality, blood vessel formation Ali Laghari et al. (2015); Sylvain
inhibition, growth retardation, et al. (2010)
pericardial oedema, cyclopia and
CNS defects
Retinoic acid Chicken and fish Mortality, eye defects, facial defects Hyatt et al. (1992); Jelínek and
and heart and limb defects Kistler (1981); Larsen and
Janners (1987)

the time period that thalidomide caused damage in embryogene- nonhuman primates and rabbits (Vargesson, 2009, 2013; see also:
sis, the earlier the exposure occurred, the more severe the damage Thalidomide and Birth Defects). Furthermore, the thalidomide
(Figure 1; Vargesson, 2015b). Lenz interviewed the parents of the disaster completely changed the way drugs were tested, requir-
thalidomide damaged children and studied their medical records ing them to be screened in multiple animal models and in vitro
to deduce the time window of sensitivity (Vargesson, 2015b). It assays, which has given rise to a greater understanding of tera-
also became clear that species differences in response to drugs tology (Kelsey, 1966; Vargesson, 2015b). Today, we know that
existed, for example rodents are less sensitive to thalidomide than the placenta does allow agents, including drugs, toxins and so on,

2 eLS © 2017, John Wiley & Sons, Ltd. www.els.net


Teratogenesis

Post conception
20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36
Days
34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50
Post menstruation
External ear missing Inner ear damage
Microphthalmia
Coloboma
Thumb damage
Upper limb/s amelia
Upper limb/s phocomelia and reduction
Hip dislocation
Ear deformation
Lower limb/s amelia
Lower limb/s phocomelia and reduction
Triphalangealism

Figure 1 Outward damage caused by thalidomide exposure of human embryos at different times during pregnancy. Thalidomide caused
outward damage (e.g. to the limbs, ears and eyes) between day 20 and day 36 of embryonic development. Note that there is generally more widespread
damage the earlier the exposure to thalidomide occurs in the time-sensitive window. The action of thalidomide falls within the embryonic ‘critical period’
which is between 2 and 8 weeks of development when all the major tissues and organs are forming and these tissues are most sensitive to disruption.
Reproduced from Vargesson (2015b) © Wiley Periodicals, Inc. under the terms of the Creative Commons Attribution License.

to directly reach the embryo/foetus, which can influence/impact in 1959 by Wilson (1973 and see Cassina et al., 2012; Schardein,
development. Indeed, the field of teratology now overlaps with 2000). These include the following:
other fields of basic science, including developmental biology,
embryology and genetics. • Susceptibility to a given teratogenic agent and subsequent ter-
Teratogens can potentially harm the embryo/foetus throughout atogenesis depends on the genetic background of the embryo
the whole of development; however, the period between the sec- because embryos, such as adults, exhibit differences in the
ond and eighth weeks of development is particularly susceptible way they respond to drugs/agents and so on.
to damage as this is when the majority of the tissues and organs • Timing of exposure to the agent, typically the earlier the expo-
are forming and taking on the final, adult patterns (Figure 1). This sure occurs in embryonic development, the more severe the
period is also known as the ‘critical period’ of development, and damage, particularly between weeks 2 and 8 of development
interference in these major tissue patterning events can result in when the majority of the major tissues and organs are forming
birth defects (Sadler, 2012; Vargesson, 2015b). Before the second and being patterned.
week of development, the embryo is not supplied/connected to • Dosage and duration of exposure. Typically the occurrence
the placenta. It should be noted that exposure to some teratogens of malformations increases with increased dosage; a scale
after 8 weeks can have effects on internal organ function and mat- for each agent as the dosage increases is produced ranging
uration and on the developing brain and nervous system (Varges- from the no observable adverse effect level (NOAEL) to an
son, 2013, 2015b). Understanding how teratogenesis comes about LD100 – a dose that results in 100% lethality.
through using animal models will shed light on how birth defects • The route of administration to come into contact with the
arise and may also give insights into normal developmental pro- embryo which includes if/how the agent can cross the pla-
cesses. In addition, such information could help design better and centa.
safer drugs as determining how these events come about could • The metabolism and absorption abilities of the agent across
potentially lead to therapeutic and preventative measures. cell membrane (e.g. whether it is lipophilic or requires spe-
cific cross-membrane transporters).
• The range (and consistency) of damage the agent
How Are Teratogenic Events causes – which can vary depending on the species as
Studied? well from individual to individual.

Experimental teratology involves trying to model human condi- Various animals have been used to study teratogenic mech-
tions/malformations in animal embryos and in vitro cell culture anisms including nonhuman primates, domestic animals such
assays, where appropriate, to gain an understanding of the con- as cats and dogs and farm animals as well as developmental
dition. This methodology can also be used to test potential thera- models such as embryos of zebrafish, chickens, rodents, rabbits,
peutic strategies. Xenopus, Caenorhabditis elegans, hydra and sea urchins. The
To understand how an agent causes a teratogenic event, we need choice of animal species really depends on the teratogenic event
to be aware of several key principles which were first described being addressed. For example, studies looking at the effects of

eLS © 2017, John Wiley & Sons, Ltd. www.els.net 3


Teratogenesis

environmental toxins on the reproductive development and fertil- United Kingdom, animal use is regulated by the Government
ity utilise farmland animals such as sheep, owing to the similarity and work is licensed. In addition, a National Centre for the
in reproductive development and function to humans. Studies Replacement, Refinement and Reduction of Animals in Research
looking at drug safety and teratogenesis now more commonly (NC3Rs; www.nc3rs.org.uk/) has been established to encourage
use rodents, rabbits and in vitro cell lines. Nonhuman primates new techniques/methods that lead to the reduction/replacement
such as monkeys have also been used (Table 2; Vargesson, 2013). of animals in research. Indeed, early/initial drug testing for
These studies in mammals involve exposing pregnant animals toxicity and teratogenesis is increasingly being considered in
to agents and observing the resulting foetuses for gross mor- nonmammalian species such as chicken embryos and zebrafish
phological abnormalities (Gilbert-Barness, 2010; Cassina et al., embryos – these embryos share developmental mechanisms with
2012). Chicken and zebrafish embryos are being increasingly embryos of mammals (including humans), and similar morpho-
used to visualise and monitor the effects and actions of agents logical events produce the embryo and its complex array of tis-
on development and morphogenesis, by direct application to sues and organs. Using these embryos of lower vertebrates, it is
the embryo (Beedie et al., 2017). Today, this methodology is possible to identify drugs that have the potential to cause terato-
enhanced by utilising molecular and genetic technologies to iden- genic events, which indicates, though does not prove, that the
tify the mechanism(s) through which these agents act, such as drugs are likely to be teratogenic in mammalian species. This
screening drug-exposed embryos for gene expression changes reduces the need for screening the drugs initially to determine
(Ema et al., 2010). their actions in mammalian species, thus reducing the number of
Agents should be administered to the animal model in a experiments and the number of mammalian embryos used. Once
species-dependent manner that causes the least discomfort. For the actions of the drugs have been worked out in nonmammalian
example, in pregnant mice, the administration may be by intra- species, the drugs would then be screened in mammalian species
venous or intraperitoneal injection or by ‘gavaging’ where the to ensure that they are indeed safe and are functioning as required.
agent is administered directly into the animal’s stomach through
a tube (Gupta, 2017). Agents could be given to the animals’ food, Relevance to humans and limitations
but this method is not as reliable as the animal may not consume
all the food in one sitting, and thus the dosage received may be To prove that an agent is a human teratogen, it is required to
inaccurate. Unexpected or adverse reactions to the agent’s admin- show that the frequency of anomalies in children of mothers who
istration would bring about the termination of the experiment. were exposed to the agent is higher than the spontaneous rate of
For embryonic exposure in chickens, the agents can be applied anomalies or that children with birth defects have a confirmed
in a solution over the embryo. For marine species, the agents can history of greater maternal exposure to the agent than unaffected
be administered directly into the water in which the embryos are children, and damage shows common patterns/characteristics
bathed, and the resulting embryos can be analysed over a range between affected individuals. This can be difficult, and epidemi-
of timepoints. Following the administration of agents, animals ological records can be used to help study long-term effects
would be observed for any developmental, morphological and of the use of an agent on pregnancy outcomes (Cassina et al.,
behavioural changes. An advantage of chicken and zebrafish 2012). Epidemiology studies can also identify ethnic differences,
embryos for screening agents for potential teratogenic capabil- regional differences and seasonal differences in the occurrence
ities is that the embryos develop outside the mother’s body, so of teratogenic events. These studies provide information about
they can be visualised easily, and agents can be directly applied any possible risks of medications or other exposures in human
to the embryo and their effects observed over long time periods. pregnancies (Vargesson and Schuler-Faccini, 2016).
Understanding how a teratogenic agent causes its effect is Screening agents, whether they are drugs, toxins, pollutants
important in understanding how congenital abnormalities arise or simply screening compounds to assess potential action, can
and can shed light on the properties that are nonteratogenic, provide information on what the agent can do in vivo and can
which has the potential to develop new therapeutic drugs that give insights into the molecular pathways the agent acts through.
are safe for use by pregnant women. Today, this is particularly Such studies allow us to determine the dose response of the
relevant in Brazil, as the drug thalidomide is being used again; agent and the time of activity. Animal embryos develop in a
this time to treat a complication of leprosy and multiple myeloma very similar way to human embryos and use the same or similar
(a cancer affecting white blood cells). Sadly, a new generation genetic pathways, and so they are relevant models. Obviously, for
of thalidomide-damaged children have been born in Brazil in the study of birth defects and malformation, animal teratology
the past decade due to the drug being shared and getting into experiments allow us to replicate the conditions and study the
the hands of pregnant women (Vianna et al., 2013; Vargesson, causes and potential therapies (see the following section). So,
2013). Clearly, if a form or structural variant of thalidomide these experiments provide vital information on potential hazards,
can be experimentally identified, which retains clinical benefits mechanisms of action and contraindications about which we
without the teratogenic side effects, this may prevent birth defects would not know otherwise and shed light on what the agent might
(Vargesson, 2015b). do in humans. However, care must always be taken when trying
to extrapolate data obtained from animal species to humans.
The 3Rs Animal data can suggest that only similar effects may occur in
humans. If an agent/drug/compound produces teratogenic effects
Around the world, animal welfare in research has always been in two or more species, the probability of the agent being a
deemed important and is taken seriously. For example, in the human teratogen must be high, but the dosage still needs to be

4 eLS © 2017, John Wiley & Sons, Ltd. www.els.net


Teratogenesis

considered. For example, the thalidomide disaster taught us that ears, limbs, genitalia and internal organs including digestive tract
species differences can exist in response to drugs by animals and heart. However, the brain is largely unaffected.
versus humans – indeed, we know in human adults themselves Thalidomide teratogenicity has been extensively studied in a
that drugs can vary in activity from individual to individual. Why wide range of animal species which include nonhuman primates,
this occurs remains unclear but may be related to the levels of rabbits, marsupials, rodents, chickens, marine fish, zebrafish right
metabolic enzymes in the liver and the capacity to breakdown the way down to Hydra. Most species are sensitive, but puz-
and excrete drugs. This is why the drug testing procedures are zlingly, rodents, the preferred animal model for drug screening
so tightly regulated to ensure that those compounds that exhibit and safety, are less sensitive and are now not used to study thalido-
the desired effects are then moved into the higher species for mide teratogenicity (Vargesson, 2009, 2013). The best models of
teratogenic testing, and human clinical trials are carried out with human thalidomide embryopathy are nonhuman primates, mon-
many different conditions including age, weight, gender and so keys and macaques, and rabbits (Ema et al., 2010). However,
on tested. The thalidomide disaster brought about changes in the recently, chicken and zebrafish embryos have provided great
way we test drugs for safety and function, using multiple animal insights into the actions of thalidomide (Beedie et al., 2016; Ito
systems, in vitro systems and a range of clinical trials in humans et al., 2010; Knobloch et al., 2007; Therapontos et al., 2009;
(Kelsey, 1966; Vargesson, 2013). This system has largely been Vargesson, 2013).
very successful though it is not infallible, as was demonstrated, These studies have led to many proposed hypotheses and theo-
for example in the early 1980s with a drug called Accutane which ries of how thalidomide causes defects (Vargesson, 2009, 2013).
contains isotretinoin, which is a modification of vitamin A. This Currently favoured mechanisms of the action of the drug are
drug was used to treat severe cases of acne and sadly caused birth that it inhibits formation of new blood vessels resulting in cell
defects in some children whose mothers used the drug during death and tissue loss or that it induces reactive oxygen species
pregnancy, before being withdrawn in 1983 (Stern et al., 1984; or that it acts through binding to the protein cereblon (Ito et al.,
Gupta, 2017). 2010; Vargesson, 2009, 2013; Vargesson and Hootnick, 2017).
Cereblon is part of an ubiquitin ligase complex whose role is
to regulate signalling in the cell. Thalidomide binding to cere-
Experimental Teratology blon (Ito et al., 2010) explains thalidomide’s anti-inflammatory
and antimyeloma actions in adults (Vargesson, 2015b; see also:
Experimental teratology models birth defects in animals to under- Thalidomide and Birth Defects), but as yet it is unknown how
stand the condition and the molecular pathways impacted. Here, such an interaction leads to and results in thalidomide embryopa-
we will briefly discuss some examples of malformations that have thy and the broad range of damage the drug induces in the short
been studied in animal models and which have provided some time-sensitive window. Other studies using thalidomide-exposed
understanding of the developmental basis for the defect. We will human embryonic stem cells and thalidomide-exposed monkey
use the Zika virus and thalidomide and discuss how experimental foetuses and carrying out gene profiling screens indicate that over
modelling of the defects caused by these agents has allowed a 2000 gene profile changes occur following thalidomide exposure
better understanding of how these teratogens act (see Table 2 (reviewed in Vargesson, 2015b). Now, the major challenge is to
for some other examples of human birth defects that have been identify the temporal order in which the expression of these genes
modelled in animals and in which the mechanism underlying the is affected and the pathways affected by the drug. This should
defect has become clearer or is known). shed further light on potential therapeutic strategies and safer
drug design.
Drugs/medicines
Virus
The field of drug identification and drug safety has become cen-
trally associated with developmental teratogenesis. Obviously, Viral infections during pregnancy have been shown to cause birth
drug safety, in particular, requires that drugs are tested to ensure defects and even miscarriages. Perhaps, the most notorious and
that they are not teratogenic. Thalidomide remains the most well known is rubella (German Measles). Vaccinations to prevent
infamous drug to cause teratogenesis; however, there are many rubella virus infections are commonplace in the developed world
other drugs that are teratogenic, which include, for example where it is extremely rare to see babies born with rubella-induced
sodium valproate, alcohol and retinoic acid (Gilbert-Barness, birth defects. Rubella virus was identified as a teratogen in the
2010; Gupta, 2017; Cassina et al., 2012; Table 1), and there 1940s and contributed to changing the view that placental mem-
are some drugs suspected of causing teratogenic damage but not branes protect the embryo from harmful agents.
proven to do so, for example Primodos (Vargesson, 2016). More recently, the Zika virus has grabbed the attention of the
world (Schuler-Faccini et al., 2016). Originally discovered in
Uganda in the 1940s, the virus was thought to be harmless. Trans-
Thalidomide mitted by Aedes mosquitoes, many babies have been born with
Today, thalidomide is used to successfully treat inflammatory microcephaly (significantly small heads and brains) in Brazil. The
disorders, such as leprosy and also cancers such as multi- Zika virus is a flavivirus, a group of viruses that include dengue
ple myeloma (Vargesson, 2015a,b). In humans, thalidomide is fever, yellow fever and Nile fever and cause encephalitis (inflam-
potently teratogenic, resulting in thalidomide embryopathy – the mation of the brain) in adults, which can be life-threatening. Zika
majority of the tissues of the body can be affected, including eyes, virus infections in adults have also been linked in rare cases

eLS © 2017, John Wiley & Sons, Ltd. www.els.net 5


Teratogenesis

to a severe adult neurological syndrome, Guillain–Barre Syn- Beedie SL, Diamond AJ, Fraga LR, Figg WD and Vargesson
drome; precisely how Zika virus causes this condition remains N (2017) Vertebrate embryos as tools for anti-angiogenic drug
unclear. There were a lot of debates about whether Zika virus screening and function. Reproductive Toxicology 70: 49–59. DOI:
was the cause of microcephaly in newborn babies (Vargesson 10.1016/j.reprotox.2016.11.013.
and Schuler-Faccini, 2016). Recent work has now confirmed that Bradley MP and Nagamine CM (2017) Animal models of Zika Virus.
this is likely to be the case where calves have been born with Comparative Medicine 67 (3): 242–252.
congenital malformations following virus infection of their moth- Cassina M, Salviati L, Di Gianantonio E and Clementi M (2012)
ers (Bradley and Nagamine, 2017; Rasmussen et al., 2016). In Genetic susceptibility to teratogens: State of the art. Reproductive
addition, chicken and mouse embryos infected with Zika virus Toxicology 34: 186–191.
develop microcephaly and nerve cell loss/destruction (Goodfel- Cugola FR, Fernandes IR, Russo FB, et al. (2016) The Brazilian Zika
low et al., 2016; Miner et al., 2016; Oh et al., 2017). Recent virus strain causes birth defects in experimental models. Nature
research suggests that Zika virus prevents brain stem cells from 534: 267–271.
proliferating and forming nerves and other nervous system sup- Cullinane J, Bannigan J and Thompson J (2009) Cadmium terato-
port tissues (McGrath et al., 2017). Further research is needed genesis in the chick: period of vulnerability using the early chick
to determine whether the nerve cells are targeted directly by the culture method, and prevention by divalent cations. Reproductive
virus or if the virus has other actions which secondarily lead to Toxicology 28 (3): 335–341.
nerve cell death. This outbreak shocked the world, and with the Ema M, Ise R, Kato H, et al. (2010) Fetal malformations and
introduction of the World Health Organisation Disease Outbreak early embryonic gene expression response in cynomolgus mon-
News Service (http://www.who.int/csr/don/en/), rapid dissemina- keys maternally exposed to thalidomide. Reproductive Toxicology
tion of advice and information has occurred to reduce the spread 29 (1): 49–56.
of the virus. Gilbert-Barness E (2010) Teratogenic causes of malformations.
These two examples underline how important experimental ter- Annals of Clinical & Laboratory Science 40: 99–114.
atogenesis is, not just determining how the malformation comes Goodfellow FT, Tesla B, Simchick G, et al. (2016) Zika virus induced
about but also identifying potential molecular targets and/or mortality and microcephaly in chicken embryos. Stem Cells and
mechanisms which can shed further light on normal developmen- Development 25 (22): 1691–1697.
tal processes as well as therapeutic interventions and better/safer Gupta R (ed) (2017) Reproductive and Developmental Toxicity, 2nd
edn. Amsterdam: Elsevier.
drug design.
Hyatt GA, Schmitt EA, Marsh-Armstrong NR and Dowling JE
(1992) Retinoic acid-induced duplication of the zebrafish retina.
Conclusions Neurobiology 89: 8293–8297.
Ito T, Ando H, Suzuki T, et al. (2010) Identification of a primary
Understanding how teratogenesis comes about is essential not target of thalidomide teratogenicity. Science 327: 1345–1350.
only to try to prevent such events but also to try and identify Jelínek R and Kistler A (1981) Effect of retinoic acid upon the chick
sooner those at risk and offer therapeutic strategies. Experimental embryonic morphogenetic systems. I. The embryotoxicity dose
teratology offers insights into mechanisms that may be impaired range. Teratology 23: 191–195.
by agents such as drugs, genes or environmental influences and Kelsey FO (1966) The evolution of new drug legislation. BMQ 17
therefore sheds light on normal development. What remains clear (2): 72–81.
Knobloch J, Shaughnessy JD Jr, and Ruther U (2007) Thalidomide
is that the developing embryo is fragile, particularly in early
induces limb deformities by perturbing the Bmp/Dkk1/Wnt sig-
embryogenesis when the body plan is forming and tissues and
nalling pathway. FASEB Journal 21 (7): 1410–1421.
organs are maturing. It is essential that agents, drugs and com-
Larsen HL and Janners MY (1987) Teratogenic effects of retinoic
pounds used in the environment and workplace are tested to
acid and dimethylsulfoxide on embryonic chick wing and somite.
ensure that they are nonteratogenic or prevented from being given
Teratology 36: 313–320.
to pregnant women. Ideally, by studying teratogenic compounds,
McGrath EL, Rossi SL, Gao J, et al. (2017) Differential responses of
we can gain a better understanding of their mechanisms and pre-
human fetal brain neural stem cells to Zika virus infection. Stem
vent birth defects by either making forms of a drug with the
Cell Reports 8 (3): 715–727. DOI: 10.1016/j.stemcr.2017.01.008.
clinical benefit but not the teratogenic side effects (i.e. better drug
Miner JJ, Cao B, Govero J, Smith AM, et al. (2016) Zika virus
design) or ensuring that precautions are in place to prevent expo-
infection during pregnancy in mice causes placental damage and
sure to pregnant women.
fetal demise. Cell 165: 1081–1091.
Oh Y, Zhang F, Wang Y, et al. (2017) Zika virus directly infects
References peripheral neurons and induces cell death. Nature Neuroscience 20
(9): 1209–1212. DOI: 10.1038/nn.4612.
Ali Laghari Z, Ali Samo A, Waryani B, et al. (2015) Effects of a Rasmussen SA, Jamieson DJ, Honein MA and Petersen LR (2016)
single dose of ethanol on survival rate and angiogenesis of chick Zika virus and birth defects – reviewing the evidence for causality.
embryo. Animal and Veterinary Sciences 3: 8–11. New England Journal of Medicine 374 (20): 1981–1987.
Beedie SL, Rore HM, Barnett S, et al. (2016) In vivo screening and Sadler T (2012) Langmans Medical Embryology, 13th edn. Philadel-
discovery of novel candidate thalidomide analogs in the zebrafish phia, PA: Wolters Kluwer.
embryo and chicken embryo model systems. Oncotarget 7 (22): Schardein JL (2000) Chemically Induced Birth Defects, 3rd edn. New
33237–33245. York: Marcel Dekker.

6 eLS © 2017, John Wiley & Sons, Ltd. www.els.net


Teratogenesis

Schuler-Faccini L, Ribeiro EM, Feitosa IM, et al. (2016) Pos- Vianna F, Schuler-Faccini L, Leite JC, et al. (2013) Recognition of
sible association between Zika virus infection and micro- the phenotype of thalidomide embryopathy in countries endemic
cephaly – Brazil, 2015. Morbidity and Mortality Weekly Report 65 for leprosy: new cases and review of the main dysmorphological
(3): 59–62. findings. Clinical Dysmorphology 22: 59–62.
Smithells RW and Newman CG (1992) Recognition of thalidomide Whitsel AI, Johnson CB and Forehand CJ (2002) An in ovo chicken
defects. Journal of Medical Genetics 29 (10): 716–723. model to study the systemic and localized teratogenic effects of
Stern RS, Rosa F and Baum C (1984) Isotretinoin and pregnancy. valproic acid. Teratology 66: 153–163.
Journal of the American Academy of Dermatology 10 (5 Pt1): Wilson JG (1973) Handbook of Teratology, 1st, vols. 1–3 edn. New
851–854. York: Plenum Press.
Sylvain NJ, Brewster DL and Ali DW (2010) Zebrafish embryos Yamamoto FY, Filipak Neto F, Freitas PF, et al. (2012) Cadmium
exposed to alcohol undergo abnormal development of motor effects on early development of chick embryos. Environmental
neurons and muscle fibers. Neurotoxicology and Teratology 32: Toxicology and Pharmacology 34 (2): 548–555.
472–480.
Therapontos C, Erskine L, Gardner ER, et al. (2009) Thalidomide
induces limb defects by preventing angiogenic outgrowth during Further Reading
early limb formation. Proceedings of the National Academy of
Sciences of the United States of America 106 (21): 8573–8578. Colborn T, vom Saal FS and Soto AM (1993) Developmental effects
Vargesson N (2009) Thalidomide-induced limb defects: resolving a of endocrine-disrupting chemicals in wildlife and humans. Envi-
50-year old puzzle. BioEssays 31: 1327–1336. ronmental Health Perspectives 101 (5): 378–384.
Vargesson N (2013) Thalidomide embryopathy: an enigmatic chal- van Gelder MM, de Jong-van den Berg LT and Roeleveld N (2014a)
lenge. ISRN Developmental Biology 2013, Article ID 241016, 18 Drugs associated with teratogenic mechanisms Part II: a literature
pages. Available from: 10.1155/2013/241016. review of the evidence on human risks. Human Reproduction 29
Vargesson N (2015a) Thalidomide: The Drug with a (1): 168–183.
Dark Side but an Enigmatic Future. Available from: van Gelder MM, van Rooij IA, de Jong-van den Berg LT and
https://theconversation.com/thalidomide-the-drug-with-a-dark- Roeleveld N (2014b) Teratogenic mechanisms associated with
side-but-an-enigmatic-future-50330 prenatal medication exposure. Thérapie 69 (1): 13–24. DOI:
Vargesson N (2015b) Thalidomide-induced teratogenesis: history 10.2515/therapie/2014003.
and mechanisms. Birth Defects Research. Part C, Embryo Today Hazelden KP (2013) The developmental toxicity testing of
105 (2): 140–156. biologics. Methods in Molecular Biology 947: 31–36. DOI:
Vargesson N (2016) Is Primodos ‘The Forgotten Thalidomide’? 10.1007/978-1-62703-131-8_3.
Available from: https://theconversation.com/is-primodos- Zeller R (2010) The temporal dynamics of vertebrate limb
the-forgotten-thalidomide-50673 development, teratogenesis and evolution. Current Opin-
Vargesson N and Schuler-Faccini L (2016) Proving that ion in Genetics and Development 20 (4): 384–390. DOI:
the Zika Virus Causes Microcephaly. Available from: 10.1016/j.gde.2010.04.014.
https://theconversation.com/proving-that-the-zika-virus-causes- Zuniga A, Zeller R and Probst S (2012) The molecular basis of human
microcephaly-53716 congenital limb malformations. Wiley Interdisciplinary Reviews:
Vargesson N and Hootnick DR (2017) Arterial dysgenesis and limb Developmental Biology 1 (6): 803–822. DOI: 10.1002/wdev.59.
defects: clinical and experimental examples. Reproductive Toxicol-
ogy 70: 21–29.

eLS © 2017, John Wiley & Sons, Ltd. www.els.net 7

View publication stats

You might also like