You are on page 1of 12

Reproductive Toxicology 103 (2021) 159–170

Contents lists available at ScienceDirect

Reproductive Toxicology
journal homepage: www.elsevier.com/locate/reprotox

The potential of mechanistic information organised within the AOP


framework to increase regulatory uptake of the developmental
neurotoxicity (DNT) in vitro battery of assays
Magdalini Sachana a, Catherine Willett b, Francesca Pistollato c, Anna Bal-Price c, *
a
Environment Health and Safety Division, Environment Directorate, Organisation for Economic Co-Operation and Development (OECD), 75775, Paris Cedex 16, France
b
Humane Society International, 1255 23rd Street NW, Washington, DC, 20037, USA
c
European Commission Joint Research Centre (JRC), Ispra, Italy

A R T I C L E I N F O A B S T R A C T

Handling Editor: Dr. Bal-Price Anna A major challenge in regulatory developmental neurotoxicity (DNT) assessment is lack of toxicological infor­
mation for many compounds. Therefore, the Test Guidelines programme of the Organisation for Economic
Keywords: Cooperation and Development (OECD) took the initiative to coordinate an international collaboration between
In vitro developmental neurotoxicity testing diverse stakeholders to consider integration of alternative approaches towards improving the current chemical
Regulatory purposes
DNT testing. During the past few years, a series of workshops was organized during which a consensus was
Adverse outcome pathways
reached that incorporation of a DNT testing battery that relies on in vitro assays anchored to key neuro­
Integrated approaches to testing and
assessment developmental processes should be developed. These key developmental processes include neural progenitor cell
Neurodevelopmental disorders proliferation, neuronal and oligodendrocyte differentiation, neural cell migration, neurite outgrowth, synapto­
genesis and neuronal network formation, as well key events identified in the existing Adverse Outcome Pathways
(AOPs). AOPs deliver mechanistic information on the causal links between molecular initiating event, inter­
mediate key events and an adverse outcome of regulatory concern, providing the biological context to facilitate
development of Integrated Approaches to Testing and Assessment (IATA) for various regulatory purposes.
Developing IATA case studies, using mechanistic information derived from AOPs, is expected to increase sci­
entific confidence for the use of in vitro methods within an IATA, thereby facilitating regulatory uptake. This
manuscript summarizes the current state of international efforts to enhance DNT testing by using an in vitro
battery of assays focusing on the role of AOPs in informing the development of IATA for different regulatory
purposes, aiming to deliver an OECD guidance document on use of in vitro DNT battery of assays that include in
vitro data interpretation.

1. Introduction linked to triggers derived from other systemic testing related to neuro­
toxicity and/or developmental toxicity or endocrine disruption. This has
Developmental neurotoxicity (DNT) is defined as the adverse func­ historically limited the generation of detailed DNT animal data to those
tional and morphological effects induced by chemical exposure on the chemicals where testing was triggered [7].
developing nervous system of offspring that may arise from exposure in Other factors that impeded the broader use of DNT Test Guidelines
utero and during early life [1]. These effects can include motor, sensory are the complexity of the in vivo tests, the difficulty in interpreting re­
and cognitive deficits [1,2]. DNT testing for assessing chemical safety is sults and the need of significant resources, contributing to the envi­
currently performed by using the available EPA and the Organisation for ronmental release of chemicals that haven’t been ever tested for their
Economic Cooperation and Development (OECD) Test Guidelines [2,3] DNT potential [8]. In a number of workshops and scientific papers,
that rely on rodent models and measure alterations at histological, regulators, academic and industrial partners agreed on the need for
physiological and neurobehavioral levels in pups following perinatal developing more efficient and potentially more effective testing strate­
exposure [4–6]. The DNT testing requirements in the different chemical gies to assess the DNT potential of chemicals, one that relies on more
regulatory sectors across the various OECD member countries are mostly recent biological understanding and technological methods relevant to

* Corresponding author at: European Commission Joint Research Centre (JRC), Via E. Fermi 2479, 21027, Ispra, Italy.
E-mail address: Anna.PRICE@ec.europa.eu (A. Bal-Price).

https://doi.org/10.1016/j.reprotox.2021.06.006
Received 12 January 2021; Received in revised form 19 May 2021; Accepted 4 June 2021
Available online 17 June 2021
0890-6238/© 2021 The Author(s). Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
M. Sachana et al. Reproductive Toxicology 103 (2021) 159–170

DNT [9–20]. Notably, a workshop that brought together experts and synaptogenesis, synaptic pruning, myelination, and neuronal network
regulators in the field, jointly organised by the Organisation for Eco­ formation with neuronal and glial maturation [15,28]. These processes
nomic Cooperation and Development (OECD) and the European Food are tightly regulated across different brain structures at specific stages of
Safety Authority (EFSA) in 2016, led to a consensus that some in vitro brain development. These spatio-temporal specificities open up very
DNT assays [17] could be used immediately for screening of chemicals distinct developmental windows of susceptibility towards the same
and prioritization, and, following further harmonization through OECD, chemical exposure. It is well documented that disruption of these key
eventual use for hazard identification and characterization [17,21]. In neurodevelopmental processes by developmental neurotoxicants may
response to the outcome and recommendations of this workshop [17, modify neuronal/glial cell birth, structure and function leading to al­
21], in 2017, the OECD convened an international expert group to terations in neuroanatomy, neurophysiology, neurochemistry, and
develop a guidance document on the application of in vitro assays within neurobehavior, resulting in a variety of adverse outcomes [17,29].
integrated approaches for testing and assessment (IATA) to address DNT Therefore, an in vitro DNT testing battery that permits quantitative
and to assist with data interpretation of these assays and IATA. evaluation of these key neurodevelopmental processes following expo­
Two critical aspects are important when developing guidance for sure to chemical(s) was proposed in the OECD/EFSA workshop [17,30]
internationally recognised non-animal-based approaches to address and considered by the OECD project as a basis for formulating a guid­
challenging endpoints such as DNT: a) the anchoring of in vitro assays to ance document. A fundamental assumption of this approach is that any
mechanistic knowledge, and b) the integration of multiple information disturbance of these key neurodevelopmental processes can be poten­
sources not restricted to data only derived from the DNT in vitro battery tially reflected through DNT-related phenotypes in vivo. Disturbances of
(DNT-IVB). For this reason, the OECD chemical safety programme has key neurodevelopmental processes are considered as important bio­
been working over the last ten years towards the development of IATA markers of potential DNT-induced chemical effects at the cellular level.
that rely on combination of multiple layers of information (e.g., epide­ The readiness of the existing in vitro DNT assays for various regulatory
miological information, existing in vivo, in vitro, in silico and non- application has been recently evaluated and, depending on the problem
mammalian in vivo data) and that can be supported by mechanistic formulation, these assays will be incorporated into an IATA accordingly
knowledge organised according the adverse outcome pathway (AOP) [28].
framework. Currently, rodent and human neuronal and glial cell models are
The AOP framework was introduced and adopted by the OECD to available which can deliver a range of reliable in vitro assays and data
allow structured collection, organisation and evaluation of existing that permit quantitative evaluation (via concentration-response re­
scientific knowledge and that could ultimately facilitate regulatory lationships) of the impact of a compound on various stages of brain
decision-making [22–24]. This knowledge is collected and assembled development. Recently there has been an emphasis on using human in
taking into account all levels of biological organisation and covering a vitro neuronal cultures derived from neural progenitor cells (NPCs) as
wide range of data that derive from molecular, cellular, clinical and they are self-renewable and can be differentiated into several neuronal
epidemiological studies. AOPs explicitly capture the linkage of and glial cell types [28,31]. Human primary NPCs derived from brain
measurable and essential biological effects (key events) to an adverse foetal tissues or from induced pluripotent stem cells (hiPSCs) differen­
outcome (AO). The molecular initiating event (MIE) can be triggered by tiated into mixed populations of neuronal/glial cells are mainly used, as
chemical exposure, and the priority adverse outcomes are of regulatory they mimic in vitro critical brain developmental processes, including
importance, such as DNT. One of the main contributions of the OECD proliferation, migration and neuronal, astrocyte and oligodendrocyte
AOP programme and its supporting community is to build a knowl­ differentiation as well as neuronal network formation and maturation
edgebase (https://aopkb.oecd.org/ and https://aopwiki.org/) that measured by evaluation of synaptogenesis, myelination and neuronal
stores mechanistic information relevant to adverse health effects that network activity [15,28,32]. A range of in vitro test methods exists for
would support development of mechanistically-informed and studying these critical neurodevelopmental processes, whereas in vitro
human-relevant alternative assays to be used for toxicity testing within assays permitting evaluation of glia-specific processes, such as oligo­
the OECD Test Guidelines programme [22]. Another important contri­ dendrocytes differentiation, myelin formation, radial and astroglia
bution is the mechanistic-based guidance for mapping data from various development, differentiation, microglia interaction/activation, and
sources, in particular for the integration of information from a battery of functional responses to chemical exposure are still not well developed or
in vitro assays and even beyond using the IATA framework [24–26]. In missing. Taking into consideration these gaps in the current DNT-IVB,
this context, AOP-informed IATA will play a pivotal role in shifting CEFIC has recently published a call titled “Expansion of a regulatory
emphasis from traditional, current regulatory DNT toxicity testing that accepted in vitro testing battery for developmental neurotoxicity (DNT)
is entirely based on animals, to more tailored, hypothesis-driven pre­ evaluation” (http://cefic-lri.org/request-for-proposals/lri-aimt11-e
dictive approaches, taking into account mechanistic information at xpansion-of-a-regulatory-accepted-in-vitro-testing-battery-for-develo
various levels of biological organization built in AOPs relevant to DNT. pmental-neurotoxicity-dnt-evaluation/), aiming to deliver the missing in
This paper briefly summarises the current status of the OECD regu­ vitro assays that could be incorporated into the existing DNT-IVB.
latory and scientific effort to develop guidance on the development and Another step forward towards the use of alternative approaches for
implementation of a DNT-IVB, leading to improved and faster chemical DNT for regulatory purposes is the recently published EFSA report on
testing for this specific endpoint. The paper also discusses the consid­ the implementation and interpretation of an in vitro testing battery for
erations for regulatory uptake of the DNT-IVB and the overall role of the the assessment of DNT [33]. In this project, human cell-based DNT test
mechanistic information captured in AOPs, which should be incorpo­ methods with high readiness underwent a fit-for-purpose evaluation
rated into the IATA framework. taking into consideration the exposure scheme, the assay and analytical
endpoint(s) and the classification model. This in vitro testing battery,
2. Current status of the international effort to develop an OECD consisting of 5 test methods measuring 10 DNT-specific endpoints and 9
guidance document on in vitro DNT battery viability/cytotoxicity-related parameters, was challenged with 119
chemicals for which rich toxicological information was available. The
DNT is one of several toxicological (as well as medical) areas of high applied DNT testing strategy is promising since its performance
concern whose biological underpinnings are complex due to multiple demonstrated a sensitivity of 82.7 % and a specificity of 88.2 %, even
neurodevelopmental processes which take place during brain develop­ with the known battery gaps.
ment [27]. Key neurodevelopmental processes include commitment and All these research efforts aim to contribute towards the development
differentiation to neural progenitor cells followed by their proliferation, of an OECD guidance on the interpretation and use of data derived from
migration, differentiation into various neuronal and glial subtypes, the DNT-IVB that can be applied to address relevant regulatory

160
M. Sachana et al. Reproductive Toxicology 103 (2021) 159–170

questions. Additional experimental work that will feed in this interna­ pathways are structured as beginning with the molecular initiating
tional effort is expected from the US Environmental Protection Agency event (MIE), the initial perturbation triggered by a stressor (e.g.,
(US EPA) that will be made available through the US EPA’s CompTox chemical exposure), followed by subsequent perturbations of down­
Chemicals Dashboard (https://comptox.epa.gov/dashboard/che stream key events (KEs) at cellular, tissue, organ and organism levels,
mical_lists/TOXCAST) and is expected to be finalised in 2021. representing a cascade of toxicity pathways potentially resulting in
In vitro assays that are not yet part of the DNT-IVB to be included in adverse outcomes (AO) of regulatory relevance [23,34]. Individual
the OECD guidance can also be valuable and complementary to the in pathways converge or diverge to form biological networks through
vitro battery and the regulatory decisions related to DNT. However, shared key events: common key events (CKEs) shared among several key
these additional in vitro assays that, for example, might come out from pathways would be priority candidates for the development of assays to
CEFIC initiatives and other similar funding opportunities would need to query that shared network of AOPs. Therefore, application of the AOP
demonstrate validity by providing the scientific basis, reproducibility framework offers the biological context for IATA development,
and predictive capacity of the new test methods including testing of increasing scientific confidence in use of mechanistic knowledge for
accepted positive and negative DNT chemicals. Such a list of reference regulatory decision-making.
compounds will be part of the DNT-IVB guidance document and include Development of AOP-informed IATA starts with problem formula­
chemicals tested in the current battery [33]. New in vitro assays that are tion (including a description of the chemical, regulatory framework,
based on the understanding of molecular and cellular mechanisms type of decision), followed by gathering all available data relevant to the
captured in AOPs can facilitate the evolution and increase regulatory specific question, including available in vivo, epidemiological informa­
acceptance of the DNT-IVB (Fig. 1). tion and any data coming from in silico, in vitro and other non-animal
The objective of the OECD project, overseen by an OECD Expert tests [24,28,35]. The existing information is evaluated through a
Group on DNT, is to deliver a guidance document that will describe a weight of evidence (WoE) approach to decide whether it is adequate for
framework to facilitate the regulatory application of DNT in vitro data a specific regulatory conclusion. Already at this stage, the AOP frame­
from the battery through the linkage to AOPs and by applying the IATA work can be applied to organise and review the existing information in a
framework. The intention is not to replace the in vivo Test Guidelines, structured and tailored manner guided by the KE and key event rela­
but to illustrate through the guidance that there are a number of regu­ tionship (KER) descriptions and can support the WoE evaluation based
latory problem formulations, for which data from the DNT-IVB can be on expert judgment.
used for decision-making. A certain number of these problem formula­ If new data are needed to inform the decision with the necessary
tions will be captured in case studies and published together with the certainty, the AOP network can guide experimental work to deliver
OECD guidance to demonstrate the practical use of the DNT-IVB. required information. As a first tier, it is recommended that in vitro as­
says should be anchored to selected set of CKEs [15,19] identified in
3. Rational for developing an AOP-informed IATA for in vitro AOPs relevant to DNT [36–38], delivering missing data (Fig. 2). More­
DNT testing over, data from in vitro assays that allow an evaluation of the key neu­
rodevelopmental processes specific for brain development but not yet
Design of testing strategies that would sufficiently cover the breadth defined as KEs in AOPs (e.g., cell proliferation, migration, differentia­
of biological processes involved in neurodevelopmental disorders re­ tion, etc.) if needed, should also be incorporated into such IATA.
quires inclusion of a battery of in vitro assays, using a range of different Based on the fit-for-purpose principle, the combination of different in
types of test methods, that capture alterations of the key neuro­ vitro DNT assays and other non-testing methods will vary. For instance,
developmental processes (ideally in a quantitative manner). To achieve our current level of understanding supports the application of an IATA
this goal, it is useful to capture neurodevelopmental processes as AOPs, based entirely on a DNT-IVB of assays for chemical screening and pri­
since the AOP framework allows compilation, structured organisation oritization, since these decisions tolerate a higher level of uncertainty.
and evaluation of biological information demonstrating the causal links IATA for hazard identification and characterization should potentially
between steps in each process or pathway. In the AOP framework, these include highly standardized in vitro assays and potentially be combined

Fig. 1. Simplified mechanistic knowledge captured in AOPs speeds up development of relevant in vitro DNT assays to be further incorporated in the existing DNT-IVB
and insures coverage of key DNT mechanisms. Schematic representation of impaired key neurodevelopmental processes defined as key events (KEs) in DNT AOPs
(hiPSCs: human induced pluripotent stem cells; NPCs: Neural Precursor cells; MIE: Molecular Initiating Event; AO: Adverse Outcome).

161
M. Sachana et al. Reproductive Toxicology 103 (2021) 159–170

Fig. 2. Building blocks of an AOP-informed


IATA specific for DNT which integrates
multiple sources of information. KEs and
KERs defined in AOP guide reviewing of exist­
ing information and if necessary, the targeted
generation of new data using a battery of in
vitro assays anchored to key neuro­
developmental processes and key events iden­
tified in AOP(s). If required, some behavioural
tests can be performed using zebrafish and
targeted animal in vivo studies. Combination of
these approaches (including QSARs, read across
etc.) will depend on problem formulation
(screening and prioritization or hazard identi­
fication/characterization). (WoE: Weight of
Evidence; PSCs: Pluripotent Stem Cells; NPCs:
Neural Precursor Cells; QSARs: Quantitative
Structure Activity Relationship). (Modified
Fig. 2 from Bal-Price et al., 2018 [28]).

with read across and QSARs, driven always by the problem formulation. “learning and memory impairment in children/decrease of cognitive func­
In the case of a decision bearing on risk assessment, information on tion” is identified as the most common AO among the currently available
exposure and ADME will need to be incorporated into the IATA. DNT AOPs (AOP Wiki; AOP 13: https://aopwiki.org/aops/13; AOP 54:
Zebrafish is the only relatively high-throughput non-mammalian animal https://aopwiki.org/aops/54; AOP 42: https://aopwiki.org/aops/42).
model to study DNT that could potentially complement the DNT-IVB and These AOPs are triggered by various MIEs that lead to different early
provide additional data to be considered under IATA. A subgroup of the KEs, but converge on CKEs close to AOs including neuronal differenti­
OECD DNT expert group is currently working towards standardization of ation, synaptogenesis or neuronal network formation and function [32].
zebrafish protocols and testing of significant number of compounds in Interestingly, these CKEs overlap with key neurodevelopmental pro­
4–6 different laboratories. cesses which can be quantitatively measured using in vitro test methods
Taking into consideration these various regulatory applications based on human mixed neuronal glial cultures [17,33]. Therefore, the
(prioritization/screening, hazard identification/characterization and assays that permit quantitative in vitro evaluation of these CKEs are the
risk assessment), 17 in vitro DNT assays were evaluated based on the first choice candidates for inclusion in IATA DNT-IVB. The current low
(semi)-quantitative analysis of their readiness with respect to various number of DNT-AOPs concerning neurodevelopmental KEs should not
use since none of these assays had previously been validated following prohibit the use of the battery, but might foster further development of
classical EURL-ECVAM validation process. The scoring results suggest DNT AOPs.
that several assays are currently at high readiness levels. Therefore,
suggestions are made on how DNT in vitro assays may be assembled into 4. Development of case studies to support the OECD DNT
an IATA, depending on the problem formulation [28]. guidance document
The DNT-IVB (Fig. 2) proposed in the OECD guidance document is
supported by currently available DNT AOPs [19,36,37]. For example, Case studies are in development to illustrate the integration of in vitro

162
M. Sachana et al. Reproductive Toxicology 103 (2021) 159–170

data derived from the testing battery together with other information potential neurodevelopmental disorders. Therefore, further efforts are
sources by applying the IATA framework. These case studies will support needed to develop AOPs, especially for AOs with increasing prevalence,
the OECD DNT guidance by providing valuable context-specific exam­ including some of those mentioned above: autism spectrum disorder
ples. All case studies begin with problem formulations designed to (ASD); attention-deficit/hyperactivity disorder (ADHD); intellectual
address a number of regulatory needs by applying targeted DNT in vitro disability (also known as mental retardation); conduct disorders; cere­
testing. In addition, the IATA case studies aim to elucidate how mech­ bral palsy; impairments in vision and hearing; motor disorders including
anistic understanding of chemical-induced DNT that is guided in most developmental coordination disorder and stereotypic movement disor­
cases by AOPs can support the required decision-making. The case der; communication, speech and language disorders; foetal alcohol
studies will cover the following scenarios: a) evaluation of DNT alerts spectrum disorder, open neural tube defects, etc. These are complex
that derive from computational models, b) screening and prioritization diseases, where complex mechanisms and pathways are likely to be
of chemical libraries with no DNT data, c) design of screening strategies involved.
to test small chemical classes, some of which have in vivo DNT data and Having a comprehensive network of DNT AOPs that covers most
others not, and d) individual chemical hazard assessment (e.g., in case major neurodevelopmental diseases will allow the identification of a
where no DNT information exist, when the DNT data are contradictory more comprehensive set of CKEs. It would also clarify to what extent
or when a chemical is re-evaluated in light of new evidence). these diseases are covered by the existing AOPs, CKEs and associated in
It is expected that with the release of the guidance and the systematic vitro test batteries, which processes are still not being covered and for
use of the battery, additional experience will be gained and knowledge which processes assays should be developed.
will be gathered to effectively address not only the above-mentioned but The current in vivo test guidelines do not predict neuro­
also additional regulatory scenarios. New IATA case studies that might developmental disorders [41], nor do they address or provide infor­
be developed after completion of the OECD guidance document can mation on mechanisms, but rather measure the chemicals’ potential to
further enrich the regulatory application of the DNT-IVB by shedding induce DNT (in rodents). It follows that the advancements in mecha­
more light on DNT mechanistic understanding and by incorporating nistic understanding of key pathways involved in developmental
additional in vitro assays. Finally, the broader use of the DNT-IVB will neurobiology and neuropathology, combined with knowledge coming
generate data that would allow the building of new AOPs and the from studies on chemical exposure such as Neurosome (https://www.
expansion of the existing AOP network. neurosome.eu/) and the Japan Environment and Children’s Study
(https://www.env.go.jp/chemi/ceh/en/index.html), when organised
5. Mechanistic information provided by an AOP-driven IATA using the AOP framework, will further improve our ability to both un­
will improve chemical assessment derstand the effects of and to better regulate chemical exposure through
the use of alternative approaches for DNT testing.
AOP networks relevant to neurotoxicity have recently been pub­
lished [37–39], and several of the identified KEs correspond with key 6. Overview of what is covered and what is missing in current
neurodevelopmental processes. One of the first AOP networks described AOPs relevant to DNT
perturbation of thyroid hormone signalling which can lead to abnormal
brain development, cognitive impairments, and other adverse outcome Neurodevelopmental disorders are multifaceted conditions charac­
in humans and wildlife [39]. terized by impairments in cognition, learning disabilities, speech and
The AOP network published by Spinu et al. [38] covers AOPs sub­ language disorders, poor social development, behaviour and/or motor
mitted to the AOP Wiki and includes both DNT and adult neurotoxicity, skills resulting from abnormal brain development. Intellectual
since the MIE and/or some KEs could occur during the neuro­ disability, communication disorders, ASD, ADHD, epilepsy and schizo­
development stage and hence could also be potentially relevant to DNT. phrenia fall also under the umbrella of neurodegenerative diseases
This AOP network shows that currently the most common AO is defined (NDDs) where a gene-environmental interplay is involved.
as “impairment of learning and memory in children/decrease of cognitive Moreover, many symptoms are not unique to a single NDD, and
function” and can be triggered by diverse MIEs, including binding of several NDDs have clusters of symptoms in common. For example,
antagonists to NMDA receptor (AOP ID 12 and 13), binding of agonists impaired social cognition is common to ASD and schizophrenia.
to NMDA receptor (AOP ID 48), inhibition of the thyroid peroxidase Furthermore, NDDs may have diverse pathophysiology that underlies
(TPO) (AOP ID 42) or inhibition of the sodium/iodide symporter (NIS) similar clinical phenotypes, or conversely, diverse clinical outcomes
(AOP ID 54). These MIEs trigger different early KEs but, closer to AO, may result from similar pathophysiology. For example, ASD is a neu­
they converge on common KEs (CKEs), such as alteration of synapto­ rodevelopmental disease with an increasing incidence and is more
genesis and neuronal network function. The AOP network described by prevalent in males [42–44]. However, it is now clear that ASD is an
Li et al. identified additional CKEs including mitochondrial dysfunction, umbrella term for multiple disorders with overlapping clinical symp­
cell injury/neurodegeneration, altered differentiation of neural stem/­ toms, suggesting that there are shared and unique pathophysiological
precursor cells into neurons and glia cells, and decreased T4 levels in mechanisms which have yet to be identified. Therefore, developing an
neuronal tissue [37]. AOP for autism (and for many other neurodevelopmental disorders) is
In vitro batteries of assays anchored to CKEs which are triggered by challenging as there is a general lack of understanding of the potential
multiple MIEs may serve as simplification for the activation of multiple multifactorial MIE(s) that are causally responsible for triggering KEs
AOPs. By choosing those CKEs as testing priorities, the number of assays resulting in complex ASD symptoms. Nevertheless, difficulties in iden­
can be reduced while still covering multiple AOPs covering a variety of tifying MIEs for many DNT AOPs may not hinder the application of these
MIEs triggered by different classes of chemicals. This rational was AOPs to design relevant testing strategies.
applied for evaluation of DNT effects induced by mixtures of chemicals Currently, there are only a few AOPs relevant to DNT or adult
using a set of in vitro assays, anchored to CKEs of several AOPs with the neurotoxicity available that are already endorsed by Working Party of
common AO of “learning and memory impairment in children” [32,40]. National Co-ordinators of the Test Guidelines Programme and the
Although endocrine disruption-based DNT AOPs are outside of the Working Party on Hazard Assessment (https://aopkb.oecd.org/ and
scope of the OECD guidance document under development, the thyroid https://aopwiki.org) (AOP ID 12, 13, 54, 42, 48, 3, 10). Others have
disruption-triggered AOPs (e.g., AOP 54 in the AOP Wiki) contain KEs been submitted to the AOP Wiki (e.g., AOPs 17, 134, 8, 152, 281,260,
that are relevant to DNT and should be included for consideration when 161, 160, 279, 230, 231) or published in peer review journals [37,45]
designing a battery of in vitro assays. but they are still at early stages of development. There are also a fair
The available AOP network covers only a very limited range of number of AOP proposals that are relevant to DNT and have been

163
M. Sachana et al. Reproductive Toxicology 103 (2021) 159–170

included in the OECD AOP workplan (https://www.oecd.org/chemicals Table 1


afety/testing/projects-adverse-outcome-pathways.htm) and are ex­ Overview of in vitro assays covering key neurodevelopmental processes and
pected to be submitted to the AOP-Wiki and reviewed in the near future major signalling pathways involved in neurodevelopmental disorders (it in­
[46–49]. cludes some pathway published by E. Fritsche [17]).
Although there are only 7 fully OECD endorsed AOPs, the existing Key neuro- In vitro Signaling pathways References
network of AOPs covers already biological space critical to several developmental assay involved in impairment
processes available of neurodevelopmental
adverse outcomes and identifies CKEs that can contribute to both hy­
Yes/No processes
pothesis generation and the design of IATA for DNT assessment. A few
DNT AOPs have been developed referring to impairment of learning and Wnt/ β-catenin signaling,;
dorsal forebrain neural
memory/cognitive damage in children ([19,39], AOP 13, AOP 48, progenitor cells
AOP17, AOP 54, AOP 42, AOP 152; all in the AOP Wiki: https://ao (PAX6 + OTX1/2+);
pwiki.org) triggered by diverse MIEs including the interaction of telencephalon (FOXG1),
chemicals with specific receptors and enzymes. There is also an AOP the ventral telencephalon
(LHX8, LHX6, NKX2-1, [61,62]
resulting in neurological dysfunction and hearing loss (AOP 8) triggered
Differentiation of DLX1, and DLX5), the
by upregulation of thyroid hormone catabolism (https://aopwiki. Human Pluripotent hindbrain (HOXA2 and
org/wiki/index.php/Aop:8), and a few AOPs published describing Stem Cells Yes HOXB2), and the dorsal
neurological and cognitive impairment triggered by interferences with towards Neural Stem telencephalon (cortex)
thyroid hormone synthesis, metabolism and transport [46]. Cells (EMX1, EMX2, and
EOMES)
The analyses of modes-of-action of DNT compounds as well as of the
Extracellular matrix
pathways contributing to impairment of neurodevelopmental processes (ECM), CREB activity/ [63]
showed that multiple pathways involved in neurodevelopmental disor­ phosphorylation
ders merge into most of these processes. These are summarized in Notch sygnalling (Mash1,
Ngn2); NSCs maintenance [64]
Table 1 and support the concept of using neurodevelopmental processes
and differentiation
and major signalling pathways as anchors for building in vitro testing SH-group maintenance;
battery because these pathways were identified based on triggers for Redox balance; Histone
human DNT (e.g., by human DNT compounds or mutations causing acetylation/deacetylation;
[65]
neurodevelopmental syndromes) [17,21]. Prostaglandin signalling;
mTORC1-STAT3, mTOR-
Further insight into chemical modes of action could be facilitated by
GSK3ß
tools being developed by the US National Toxicology Program Inter­ activation RTKPI3K-AKT
agency Center for the Evaluation of Alternative Toxicological Methods signalling; PGE2 – wnt
(NICEATM), including the Integrated Chemical Environment (ICE) and Neural precursor Cells
signalling; BDNF-ERK- [66]
CREB, retinoic acid
the Developmental NeuroToxicity Data Integration and Visualization and
signaling
Enabling Resource (DNT-DIVER). ICE provides an interactive dashboard Neural Crest Cells: Yes
mTORC1 and mTORC2 [67]
for accessing curated in silico, in vitro and in vivo data from multiple Neurogenesis and
TH signalling [68,69]
Proliferation
sources as well as a set of computational workflows to assist with GSK3B [70]
chemical activity characterization [50]. DNT-DIVER compares results Forskolin, Indomethacin
induced increase in cAMP [66,71,72,
from a limited number of DNT-related assays for a limited number of
and activity of Protein 73]
chemicals; the potential utility of this tool will increase as assays and Kinases A/B
curated data are added [51]. Neurotrophins, RA
The most studied DNT compound is lead and indeed, there is already induced signalling through
[74,75,76]
MAPK/ERK and PI3K/Akt
an AOP endorsed by OECD working parties of the chemical safety pro­
activity
gramme where lead is described as a trigger of the MIE defined as PLCgamma1, GDNF-RET,
“binding of antagonists to NMDA receptor [52]. One of the mechanisms BDNF/TrkB, PDGFR,
triggered by lead is inhibition of BDNF-Trk signalling, causing aberrant FGFR, mTORC1 signalling;
dendritic morphology, decreased synaptogenesis and decreased MAP kinase and Reelin-
[77,78]
Dab pathways, PGE2 – wnt
neuronal network formation and function. Impairment of these key Neural Progenitor Cell
signalling; PLCgamma1-
neurodevelopmental processes may lead to decreased learning and Neuronal and Yes
dependent calcium release
Radial glia migration
memory processes in children [52,53]. During different stages of brain with activation of PKC
development, BDNF-associated signalling pathways are also affected by N-cadherin, RhoA
[79]
exposure to ethanol, another well-established DNT compound [54–56]. activation.
Integrin alpha3beta1,
Synaptogenesis is also affected by exposure to chlorpyrifos [57] and reelin
[80]
organophosphates [58,59], inducing disturbance of cAMP-CREB sig­ PDGFR-PLCγ1; BDNF/
nalling pathway, resulting in impaired axonal and dendritic TrkB activates MAPK and
differentiation. PI3K pathways and PLCγ1-
dependent IP3-mediated
Separate studies are required to link potential DNT compounds with
calcium release; GDNF-
perturbed signalling pathways described in Table 1. Such an exercise RET-mediated activation
would be beneficial to establish mechanistic understanding of DNT ef­ CaMKII; PDGFR-mediated [78]
fects induced by environmental chemicals or drugs as potential DNT Neural Migration Yes
activation of PLCγ1 with
compounds. production of intracellular
DAG gradient, ERC
Ideally, all biological pathways leading to neurological and devel­ activation, retinoic acid
opmental neurological pathologies would be known and described in the signaling, RET and JNK
form of AOPs. Without knowing the full extent of coverage, it is possible dependent migration.
that the existing AOPs cover only limited DNT-relevant biological space Reelin, ApoER2 and
[81]
VLDLR
and mechanisms of concern. Therefore, it is desirable to gather further
[82]
mechanistic understanding of neurological diseases captured as AOPs, to
(continued on next page)
explicitly describe and link to major neurodevelopmental adverse

164
M. Sachana et al. Reproductive Toxicology 103 (2021) 159–170

Table 1 (continued ) Table 1 (continued )


Key neuro- In vitro Signaling pathways References Key neuro- In vitro Signaling pathways References
developmental assay involved in impairment developmental assay involved in impairment
processes available of neurodevelopmental processes available of neurodevelopmental
Yes/No processes Yes/No processes

BDNF⁄TrkB interaction, such as INSM, GPSM2,


PI3-K, MAP-K ASPM, TRNP1,
Cdk5, Dab1, Rac1, FAK, [83] ARHGAP11B, PAX6, and
Ras, Src, and PI3K HIF1α
Calm1, Gria1 (GluA1) and [84] FGF-2 [98]
Camk4 (calmodulin- Notch, ErbB, and [99]
signaling network), Hdac2 fibroblast growth factor
and Hsbp1 (Akt1-DNA mTORC1-STAT3
transcription network), signalling, Notch
[67]
Vav3 and Ppm1a signalling; miRNA-124;
mTORC1/C2, mTORC2 signalling
prostaglandin, Histone Activation of MAPK/ERK [100,101,
acetylation/deacetylation, and increase in JAK/STAT 102]
miRNA-9, miRNA-17-92 Induction of gp130
[64,67] [103,104,
cluster, miRNA-124, receptors for JAK/STAT
105]
Notch signalling, Wnt, activity
BDNF, retinoic acid Fibronectin, BMP
[106]
signaling. Differentiation signalling
Yes
SHH and Notch pathway into Astrocytes Astrocytic expression of
activation (for the NFIAA/B and GLAST-1,
[85]
maintenance of neural S100β is an astrocyte
Differentiation and
rosette cells) progenitor marker, GFAP,
maturation of Neural [107]
Yes Wnt/β-catenin pathway, β GS, EAAT1 and EAAT2,
Stem Cells into distinct
-catenin/TCF, neurogenin [86] AQP-4, GS, GLT-1,
neuronal cell types
1 astrocytic early stage
aPKC, PRKCI and PRKCZ [87] marker: ALDH1
Wnt factors signal through LIF, STAT3, RA [108]
canonical, β-catenin CSL-dependent Notch
[109]
pathway, planar cell [88] signalling pathway
polarity pathway and Extracellular matrix
calcium pathway (ECM), fibroblast growth
Neuron maturation factor (FGF)-2, retinoic
relevant functional acid (RA), EGF
[89]
modules in protein-protein morphogenetic protein
[110,111,
interaction (PPI) network antagonists such as
112,113,
NMDA-receptor noggin, neurotrophic
114,115]
activation, BDNF-Trk factors such as
Differentiation into
signalling, calcium Yes neurotrophin (NT)-3 and
Oligodendrocytes
signalling, inositol ciliary neurotrophic
metabolism, factor, with or without
[65]
Phospholipase D activity EGF
with generation of TH signalling [68,69]
phosphatidic acid; BDNF- Signalling through RA and [116,117,
ERK-CREB/decreased p38 MAPK pathways 118]
activity/phosphorylation Induction of transcription [119,120,
Synaptogenesis Yes
ProSAP1/Shank2, MaGuK factors by Notch and Shh 121]
family, including SAP90/ CD45− /c-KIT− /CX3CR1+
PSD-95, Munc13, RIMs, cells in a PU-1, IRF-8, and
ERC/CAST, Piccolo/ [90] colony stimulating factor 1
[122]
Aczonin, and Bassoon, (CSF-1R), RFD7+) and
Shank1, Neuroligin, and monocyte-associated
GKAP markers (UCHM1+)
Differentiation
Wnt signalling through Signaling through
into No
β-catenin-dependent [91] fractalkine receptor (e.g.,
Microglia
pathways CX3CR1), cell survival
N-methyl-d-aspartate factor (CSF) 1-receptor
[123,124]
(NMDA) receptors, the (CSF1-R), the transcription
retinoic acid receptors, factors, PU.1 (SPl1) and
brain-derived interferon regulatory
neurotrophic factor [92] factor 8 (IRF8)
(BDNF), insulin-like Neregulin-1, LPA-induced
growth factor 1 (IGF-I), increase in cAMP by
Apoptosis Yes [125,126]
and basic fibroblast Differentiation activation of GPR44 and
growth factor (bFGF) into Yes GPR126
miR-132 [93] Schwann cells Neurotrophins induction
Caspase-dependent cell of specific transcription [127,128]
death, AIF, Bcl-2 and its factors by PI3K/Akt
[94,95]
related family member FGFR-mediated PLCγ1-
Bcl-xL PKC activation with
miRNA-9 [96] Outgrowth of dendrites subsequent IP3 and AA
Yes [129]
Radial glia proliferation FGF-MAPK cascade, SHH, & axons formation; NCAM/FGFR-
Yes
and migration PTEN/AKT, PDGF [97] PLCγ1 with action on
pathways, and proteins small GTPases, such as
(continued on next page)

165
M. Sachana et al. Reproductive Toxicology 103 (2021) 159–170

Table 1 (continued ) (1) a general lack of understanding of the contribution of biology,


Key neuro- In vitro Signaling pathways References including genetic and environmental factors to these disorders, (2) a
developmental assay involved in impairment lack of identification of specific pathways that might be triggered,
processes available of neurodevelopmental including potential MIEs and causally linked KEs that lead to these
Yes/No processes outcomes. The molecular and cellular mechanisms of many neuro­
Rho A, Rac1, or Cdc42; developmental disorders are largely unknown and in some cases, fea­
cytoskeleton maintenance; tures/symptoms of these diseases are overlapping since they are not
CREB and BDNF signling,
specific for one particular disease. Therefore, the complex aetiology and
MAPK activation
RYR sensitization, PIP [78] limited mechanistic understanding of neurodevelopmental disorders
metabolism, BDNF-ERK- make identification of KEs (which would be linked linearly and sup­
CREB: Prostaglandins, ported by empirical data described in KERs) extremely difficult. Addi­
cyclooxygenases, EP tional difficulty arises from the dynamic processes of brain
receptors; BDNF, ERK-
CREB
development, which can be affected differently by the same chemical,
TH signaling, [68,69] depending on the window of exposure and the susceptibility of specific
BDNF-ERK-CREB [65] brain regions and various neuronal or glial subtypes at different times
PLCγ1-dependent calcium during development.
release with activation of
[78] One advantage of focussing on AOPs describing basic neuro­
PKC, BDNF, TRPC3/6
channels, IP3 developmental processes is to make sure that the testing battery covers
Plexin B, EphA, ephrin B these fundamental processes, increasing the likelihood that any neuro­
Axon guidance Yes and Robo, regulate the [130] developmental effect will be captured, even if it cannot yet be explicitly
Rho family of GTPases described in an AOP and linked to critical KEs or AOs. Another advan­
Rho family small
guanosine triphosphatases [131]
tage of the AOP framework is that, as biological information is added
(GTPases) and AOP networks evolve, testing strategies could be designed for
TH signaling [68,69] different purposes or levels of specification (e.g., Fig. 3). A testing bat­
Interference with calcium tery that covers CKEs based on several cellular processes (e.g., mito­
signalling; miRNA-124;
[132] chondria impairment, oxidative stress, dysfunction of ion channels,
AKT signaling and miR-9/
9* receptors, changes in neurotransmitter synthesis/release, increased
ASE chemosensory proinflammatory cytokines production, etc.) could be used as a pre­
neurons lys-6 COG-1, miR-
[133,134]
cautionary screen, irrespective of identification of a particular pathway
273, DIE-1 induction of or AO. A battery of more specific CKEs could be used to identify more
ASEL identity
specific pathways or developmental stages (e.g., synaptogenesis,
Anterior–posterior axis:
[135,136] neuronal network formation and function or cell-type-specific differ­
miR-9 Hes1 (homologs)
Cortex: miR-9, FoxG1, entiation or migration), or network or pathway specific KEs could then
[137,138]
several other targets be used to identify affected pathways with increasing specificity (e.g.,
Olfactory bulb: Pax 6,
[139] specific to impairment of learning or memory, or neurodevelopmental
Neuronal miR-7◦ , etc.
subtypes
Yes
Midbrain: miR-135a disorder-specific). The current DNT-IVB included in the OECD guidance
Lmx1b. Delimiting the DV document is based on assays that mostly cover this biological space but
[140]
extent of the dopaminergic is not considered exhaustive.
progenitor pool For diseases not covered by current AOPs, such as ASD and ADHD
Midbrain–hindbrain
etc., it is important to increase the mechanistic understanding to make
boundary: miR-9, Fgfr1, [141]
Canopy, Fgf8, Her5, Her9 sure the underlying mechanisms can be covered by available testing
Retina: miR-129, miR-155, approaches. Therefore, when developing future DNT-related AOPs, the
miR-214, miR-222, Xotx2,
[142,143] systematic organization of available information and the identification
Xvsx1, let-7, miR-125,
of critical knowledge gaps are equally important. In vitro assays
miR-9 Ptrg, Lin28b
Spinal cord miR-17-3p
anchored to KEs can deliver relevant molecular and cellular information
[144,145, which would contribute to better mechanistic understanding of path­
Olig2, miR-196, Hoxb8,
146,147]
miR-9 FoxP1, OC1 ways involved, especially when using human-cell based test methods,
Inositol metabolism; PIP contributing to further AOP development relevant to neuro­
(2&3); PI3K,
developmental disorders.
Prostaglandins
(cyclooxygenases, EP
receptors); TH signalling; 7. Conclusions
Neuronal network [78,148,
EphrinA1/EphA4 - PLCγ1
formation 149]
Yes (structural synaptic
(synaptogenesis) and The AOP framework provides the opportunity for explicitly
plasticity); BDNF/TrkB -
synaptic plasticity
PLCγ1, BDNF, ERK-CREB; describing both the common and unique molecular features of the
BDNF-ERK-CREB (GABA various developmental stages of the nervous system. Developing AOP
maturation) networks reflecting the complexity of the key neurodevelopmental
TH signaling [68,69] processes that are anchored to phenotypic adverse outcomes seems a
BDNF-ERK-CREB [65]
challenging task. However, emerging experimental data can provide
significant empirical evidence and support to AOP building and reveal
outcomes not yet covered. Further development of AOPs will inform better understanding of chemicals’ involvement in manifestation of DNT
about the mechanistic space that is potentially not assessed by the cur­ relevant adverse outcomes. In parallel, epidemiological studies com­
rent proposed DNT-IVB and identify KEs as anchors for development of plemented by ‘omics’ generated data from patient samples (e.g., cere­
additional in vitro assays. brospinal fluid) or information gathered from functional MRI could
As mentioned earlier, development of AOPs for complex neuro­ potentially, in the near future, reveal reliable biomarkers of effects that
developmental disorders such as ASD or ADHD is challenging because of could further confirm and provide stronger evidence supporting the
DNT-relevant AOPs. Most importantly, studies that provide quantitative

166
M. Sachana et al. Reproductive Toxicology 103 (2021) 159–170

Fig. 3. Biological space represented as concentric spheres of increasing specificity, containing examples specific to the developing nervous system.

human data would identify factors that modify potential outcomes; interests or personal relationships that could have appeared to influence
these factors could then be described quantitatively in KER for eventual the work reported in this paper.
modelling, for example to predict susceptible individuals.
Activities for the development of DNT relevant IATAs and the use of References
DNT-IVB can draw from the mechanistic information that is structurally
stored in individual AOPs or AOP networks. Developed IATA case [1] OECD, OECD Guidelines for the Testing of Chemicals. Section 4: Health Effects.
Test No. 443: Extended One Generation Reproductive Toxicity Study, 2018. htt
studies can also lead to the generation of new mechanistic data and ps://www.oecd-ilibrary.org/environment/test-no-443-extended-one-generation-
hence development of novel AOPs, as it has happened recently in an reproductive-toxicity-study_9789264185371-en.
EFSA DNT project where the trigger-specific (deltamethrin) AOP [2] US. EPA, Health Effects Guidelines OPPTS 870.6300 Developmental
Neurotoxicity Study, Office of Prevention Pesticides and Toxic Substances, 1998.
network has been created as part of the IATA case study [33,60], and https://nepis.epa.gov/Exe/ZyNET.exe/P100IRWO.txt?ZyActionD=ZyDocumen
will soon be published. t&Client=EPA&Index=1995%20Thru%201999&Docs=&Query=&Time=&En
AOP-supported IATA can address various regulatory and non- dTime=&SearchMethod=1&TocRestrict=n&Toc=&TocEntry=&QField=&QFie
ldYear=&QFieldMonth=&QFieldDay=&UseQField=&IntQFieldOp=0&ExtQFie
regulatory problem formulations that range from chemical prioritisa­ ldOp=0&XmlQuery=&File=D%3A%5CZYFILES%5CINDEX%20DATA%5C95
tion to risk assessment. Developing IATA case studies using focused THRU99%5CTXT%5C00000034%5CP100IRWO.txt&User=ANONYMOUS&Pa
problem formulations supported by mechanistic information from ssword=anonymous&SortMethod=h%7C-&MaximumDocuments=1&FuzzyDe
gree=0&ImageQuality=r75g8/r75g8/x150y150g16/i425&Display=hpfr&De
AOPs, with clear recognition of uncertainties involved, can ultimately
fSeekPage=x&SearchBack=ZyActionL&Back=ZyActionS&BackDesc=Results%
increase the uptake and gain support from the regulatory community. 20page&MaximumPages=1&ZyEntry=1.
[3] OECD, OECD Guidelines for the Testing of Chemicals. Section 4: Health Effects.
Test No. 426: Developmental Neurotoxicity Study, 2007. https://www.oecd-ilibr
Disclaimers ary.org/environment/test-no-426-developmental-neurotoxicity-study_9789264
067394-en.
The opinions expressed and arguments employed herein are those of [4] S.L. Makris, K. Raffaele, S. Allen, W.J. Bowers, U. Hass, E. Alleva, G. Calamandrei,
L. Sheets, P. Amcoff, N. Delrue, K.M. Crofton, A retrospective performance
the authors and do not necessarily reflect the official views of the OECD assessment of the developmental neurotoxicity study in support of OECD test
or of the governments of its member countries. guideline 426, Environ. Health Perspect. 117 (1) (2009) 17–25.
[5] R. Tsuji, K.M. Crofton, Developmental neurotoxicity guideline study: issues with
methodology, evaluation and regulation, Congenit. Anom. (Kyoto) 52 (3) (2012)
Declaration of Competing Interest 122–128.
[6] C. Tohyama, Developmental neurotoxicity test guidelines: problems and
perspectives, J. Toxicol. Sci. 41 (Spec) (2016) SP69–SP79.
The authors declare that they have no known competing financial

167
M. Sachana et al. Reproductive Toxicology 103 (2021) 159–170

[7] L. Smirnova, H.T. Hogberg, M. Leist, T. Hartung, Developmental neurotoxicity - M. Sachana, T.J. Shafer, A. Terron, F. Monnet-Tschudi, B. Viviani, T. Waldmann,
challenges in the 21st century and in vitro opportunities, ALTEX 31 (2) (2014) R.H.S. Westerink, M.F. Wilks, H. Witters, M.G. Zurich, M. Leist, Recommendation
129–156. on test readiness criteria for new approach methods in toxicology: exemplified for
[8] K.M. Crofton, W.R. Mundy, T.J. Shafer, Developmental neurotoxicity testing: a developmental neurotoxicity, ALTEX 35 (3) (2018) 306–352.
path forward, Congenit. Anom. (Kyoto) 52 (3) (2012) 140–146. [29] E. Fritsche, M. Barenys, J. Klose, S. Masjosthusmann, L. Nimtz, M. Schmuck,
[9] P. Lein, P. Locke, A. Goldberg, Meeting report: alternatives for developmental S. Wuttke, J. Tigges, Development of the concept for stem cell-based
neurotoxicity testing, Environ. Health Perspect. 115 (5) (2007) 764–768. developmental neurotoxicity evaluation, Toxicol. Sci. 165 (1) (2018) 14–20.
[10] S. Coecke, A.M. Goldberg, S. Allen, L. Buzanska, G. Calamandrei, K. Crofton, [30] E. Fritsche, K.M. Crofton, A.F. Hernandez, S. Hougaard Bennekou, M. Leist,
L. Hareng, T. Hartung, H. Knaut, P. Honegger, M. Jacobs, P. Lein, A. Li, A. Bal-Price, E. Reaves, M.F. Wilks, A. Terron, R. Solecki, M. Sachana,
W. Mundy, D. Owen, S. Schneider, E. Silbergeld, T. Reum, T. Trnovec, F. Monnet- A. Gourmelon, OECD/EFSA workshop on developmental neurotoxicity (DNT): the
Tschudi, A. Bal-Price, Workgroup report: incorporating in vitro alternative use of non-animal test methods for regulatory purposes, ALTEX 34 (2) (2017)
methods for developmental neurotoxicity into international hazard and risk 311–315.
assessment strategies, Environ. Health Perspect. 115 (6) (2007) 924–931. [31] E. Fritsche, M. Barenys, J. Klose, S. Masjosthusmann, L. Nimtz, M. Schmuck,
[11] P.B. Kuegler, B. Zimmer, T. Waldmann, B. Baudis, S. Ilmjarv, J. Hescheler, S. Wuttke, J. Tigges, Current availability of stem cell-based in vitro methods for
P. Gaughwin, P. Brundin, W. Mundy, A.K. Bal-Price, A. Schrattenholz, K. developmental neurotoxicity (DNT) testing, Toxicol. Sci. 165 (1) (2018) 21–30.
H. Krause, C. van Thriel, M.S. Rao, S. Kadereit, M. Leist, Markers of murine [32] F. Pistollato, E.M. de Gyves, D. Carpi, S.K. Bopp, C. Nunes, A. Worth, A. Bal-Price,
embryonic and neural stem cells, neurons and astrocytes: reference points for Assessment of developmental neurotoxicity induced by chemical mixtures using
developmental neurotoxicity testing, ALTEX 27 (1) (2010) 17–42. an adverse outcome pathway concept, Environ. Health 19 (1) (2020) 23.
[12] K.M. Crofton, W.R. Mundy, P.J. Lein, A. Bal-Price, S. Coecke, A.E. Seiler, [33] S. Masjosthusmann, J. Blum, K. Bartmann, X. Dolde, A.K. Holzer, E.H.K. Lynn,
H. Knaut, L. Buzanska, A. Goldberg, Developmental neurotoxicity testing: C. Stürzl, N. Förster, A. Dönmez, J. Klose, M. Pahl, T. Waldmann, J.K.F. Bendt,
recommendations for developing alternative methods for the screening and I. Suciu, U. Hübenthal, A. Mosig, L. Marcel, E. Fritsche, Establishment of an a
prioritization of chemicals, ALTEX 28 (1) (2011) 9–15. Priori Protocol for the Implementation and Interpretation of an In-vitro Testing
[13] S. Kadereit, B. Zimmer, C. van Thriel, J.G. Hengstler, M. Leist, Compound Battery for the Assessment of Developmental Neurotoxicity, EFSA, 2020.
selection for in vitro modeling of developmental neurotoxicity, Front. Biosci. [34] S.W. Edwards, Y.M. Tan, D.L. Villeneuve, M.E. Meek, C.A. McQueen, Adverse
Landmark Ed. 17 (2012) 2442–2460. outcome pathways-organizing toxicological information to improve decision
[14] A.K. Bal-Price, S. Coecke, L. Costa, K.M. Crofton, E. Fritsche, A. Goldberg, making, J. Pharmacol. Exp. Ther. 356 (1) (2016) 170–181.
P. Grandjean, P.J. Lein, A. Li, R. Lucchini, W.R. Mundy, S. Padilla, A.M. Persico, [35] G. Patlewicz, C. Kuseva, A. Kesova, I. Popova, T. Zhechev, T. Pavlov, D.
A.E. Seiler, J. Kreysa, Advancing the science of developmental neurotoxicity W. Roberts, O. Mekenyan, Towards AOP application–implementation of an
(DNT): testing for better safety evaluation, ALTEX 29 (2) (2012) 202–215. integrated approach to testing and assessment (IATA) into a pipeline tool for skin
[15] A. Bal-Price, F. Pistollato, M. Sachana, S.K. Bopp, S. Munn, A. Worth, Strategies to sensitization, Regul. Toxicol. Pharmacol. 69 (3) (2014) 529–545.
improve the regulatory assessment of developmental neurotoxicity (DNT) using [36] A. Bal-Price, M.E.B. Meek, Adverse outcome pathways: application to enhance
in vitro methods, Toxicol. Appl. Pharmacol. 354 (2018) 7–18. mechanistic understanding of neurotoxicity, Pharmacol. Ther. 179 (2017) 84–95.
[16] W.R. Mundy, S. Padilla, J.M. Breier, K.M. Crofton, M.E. Gilbert, D.W. Herr, K. [37] J. Li, R. Settivari, M.J. LeBaron, M.S. Marty, An industry perspective: a
F. Jensen, N.M. Radio, K.C. Raffaele, K. Schumacher, T.J. Shafer, J. Cowden, streamlined screening strategy using alternative models for chemical assessment
Expanding the test set: Chemicals with potential to disrupt mammalian brain of developmental neurotoxicity, Neurotoxicology 73 (2019) 17–30.
development, Neurotoxicol. Teratol. 52 (Pt A) (2015) 25–35. [38] N. Spinu, A. Bal-Price, M.T.D. Cronin, S.J. Enoch, J.C. Madden, A.P. Worth,
[17] E. Fritsche, Report on Integrated Testing Strategies for the Identification and Development and analysis of an adverse outcome pathway network for human
Evaluation of Chemical Hazards Associated With the Developmental neurotoxicity, Arch. Toxicol. 93 (10) (2019) 2759–2772.
Neurotoxicity (DNT), to Facilitate Discussions at the Joint EFSA/OECD Workshop [39] P.D. Noyes, K.P. Friedman, P. Browne, J.T. Haselman, M.E. Gilbert, M.
on DNT Series on Testing and Assessment No. 261, OECD, 2017. W. Hornung, S. Barone Jr., K.M. Crofton, S.C. Laws, T.E. Stoker, S.O. Simmons, J.
[18] B.Z. Schmidt, M. Lehmann, S. Gutbier, E. Nembo, S. Noel, L. Smirnova, A. Forsby, E. Tietge, S.J. Degitz, Evaluating chemicals for thyroid disruption: opportunities
J. Hescheler, H.X. Avci, T. Hartung, M. Leist, J. Kobolak, A. Dinnyes, In vitro and challenges with in vitro testing and adverse outcome pathway approaches,
acute and developmental neurotoxicity screening: an overview of cellular Environ. Health Perspect. 127 (9) (2019) 95001.
platforms and high-throughput technical possibilities, Arch. Toxicol. 91 (1) [40] N. Davidsen, A.J. Lauvås, O. Myhre, E. Ropstad, D. Carpi, E.M. Gyves, H.
(2017) 1–33. F. Berntsen, H. Dirven, R.E. Paulsen, A. Bal-Price, F. Pistollato, Exposure to
[19] A. Bal-Price, K.M. Crofton, M. Leist, S. Allen, M. Arand, T. Buetler, N. Delrue, R. human relevant mixtures of halogenated persistent organic pollutants (POPs)
E. FitzGerald, T. Hartung, T. Heinonen, H. Hogberg, S.H. Bennekou, alters neurodevelopmental processes in human neural stem cells undergoing
W. Lichtensteiger, D. Oggier, M. Paparella, M. Axelstad, A. Piersma, E. Rached, differentiation, Reprod. Toxicol. (Elmsford, N.Y.) 100 (2021) 17–34.
B. Schilter, G. Schmuck, L. Stoppini, E. Tongiorgi, M. Tiramani, F. Monnet- [41] M. Paparella, S.H. Bennekou, A. Bal-Price, An analysis of the limitations and
Tschudi, M.F. Wilks, T. Ylikomi, E. Fritsche, International STakeholder NETwork uncertainties of in vivo developmental neurotoxicity testing and assessment to
(ISTNET): creating a developmental neurotoxicity (DNT) testing road map for identify the potential for alternative approaches, Reprod. Toxicol. (Elmsford, N.
regulatory purposes, Arch. Toxicol. 89 (2) (2015) 269–287. Y.) 96 (2020) 327–336.
[20] E. Fritsche, P. Grandjean, K.M. Crofton, M. Aschner, A. Goldberg, T. Heinonen, E. [42] K. Lyall, L. Croen, J. Daniels, M.D. Fallin, C. Ladd-Acosta, B.K. Lee, B.Y. Park, N.
V.S. Hessel, H.T. Hogberg, S.H. Bennekou, P.J. Lein, M. Leist, W.R. Mundy, W. Snyder, D. Schendel, H. Volk, G.C. Windham, C. Newschaffer, The changing
M. Paparella, A.H. Piersma, M. Sachana, G. Schmuck, R. Solecki, A. Terron, epidemiology of autism Spectrum disorders, Annu. Rev. Public Health 38 (2017)
F. Monnet-Tschudi, M.F. Wilks, H. Witters, M.G. Zurich, A. Bal-Price, Consensus 81–102.
statement on the need for innovation, transition and implementation of [43] M.E. McDonald, J.F. Paul, Timing of increased autistic disorder cumulative
developmental neurotoxicity (DNT) testing for regulatory purposes, Toxicol. incidence, Environ. Sci. Technol. 44 (6) (2010) 2112–2118.
Appl. Pharmacol. 354 (2018) 3–6. [44] C. Cheroni, N. Caporale, G. Testa, Autism spectrum disorder at the crossroad
[21] OECD, Report of the OECD/EFSA Workshop on Developmental Neurotoxicity between genes and environment: contributions, convergences, and interactions in
(DNT): the Use of Non-animal Test Methods for Regulatory Purposes, Series on ASD developmental pathophysiology, Mol. Autism 11 (1) (2020) 69.
Testing and Assessment No. 261, 2017. [45] A.P. Costa, G. Steffgen, A.C. Samson, Expressive incoherence and Alexithymia in
[22] N. Delrue, M. Sachana, Y. Sakuratani, A. Gourmelon, E. Leinala, R. Diderich, The autism Spectrum disorder, J. Autism Dev. Disord. 47 (6) (2017) 1659–1672.
adverse outcome pathway concept: a basis for developing regulatory decision- [46] A. Bal-Price, P.J. Lein, K.P. Keil, S. Sethi, T. Shafer, M. Barenys, E. Fritsche,
making tools, Altern. Lab. Anim. 44 (5) (2016) 417–429. M. Sachana, M.E.B. Meek, Developing and applying the adverse outcome
[23] OECD, Users’ Handbook Supplement to the Guidance Document for Developing pathway concept for understanding and predicting neurotoxicity,
and Assessing Adverse Outcome Pathways, 2018. https://www.oecd-ilibrary. Neurotoxicology 59 (2017) 240–255.
org/environment/users-handbook-supplement-to-the-guidance-document-for- [47] M. Barenys, I. Reverte, S. Masjosthusmann, J. Gómez-Catalán, E. Fritsche,
developing-and-assessing-adverse-outcome-pathways_5jlv1m9d1g32-en. Developmental neurotoxicity of MDMA. A systematic literature review
[24] OECD, Guidance Document for the Use of Adverse Outcome Pathways in summarized in a putative adverse outcome pathway, Neurotoxicology 78 (2020)
Developing Integrated Approaches to Testing and Assessment (IATA), 2017 209–241.
(Accessed Dec 14, 2020, http://www.oecd.org/chemicalsafety/guidance-docume [48] H. Chen, M.A. Chidboy, J.F. Robinson, Retinoids and developmental
nt-for-the-use-of-adverse-outcome-pathways-in-developing-integrated-approache neurotoxicity: utilizing toxicogenomics to enhance adverse outcome pathways
s-to-testing-and-assessment-iata-44bb06c1-en.htm. and testing strategies, Reprod. Toxicol. (Elmsford, N.Y.) 96 (2020) 102–113.
[25] M. Sachana, E. Leinala, Approaching chemical safety assessment through [49] J. Klose, J. Tigges, S. Masjosthusmann, K. Schmuck, F. Bendt, U. Hübenthal,
application of Integrated approaches to testing and assessment: combining P. Petzsch, K. Köhrer, K. Koch, E. Fritsche, TBBPA targets converging key events
mechanistic information derived from adverse outcome pathways and alternative of human oligodendrocyte development resulting in two novel AOPs, ALTEX 38
methods, Appl. In Vitro Toxicol. 3 (3) (2017) 227–233. (2) (2021) 215–234.
[26] Y. Sakuratani, M. Horie, E. Leinala, Integrated approaches to testing and [50] S. Bell, J. Abedini, P. Ceger, X. Chang, B. Cook, A.L. Karmaus, I. Lea, K. Mansouri,
assessment: OECD activities on the development and use of adverse outcome J. Phillips, E. McAfee, R. Rai, J. Rooney, C. Sprankle, A. Tandon, D. Allen,
pathways and case studies, Basic Clin. Pharmacol. Toxicol. 123 (Suppl 5) (2018) W. Casey, N. Kleinstreuer, An integrated chemical environment with tools for
20–28. chemical safety testing, Toxicol. In Vitro 67 (2020) 104916.
[27] J. Stiles, T.L. Jernigan, The basics of brain development, Neuropsychol. Rev. 20 [51] NTP, Data Release: Developmental NeuroToxicity Data Integration and
(4) (2010) 327–348. Visualization Enabling Resource (DNT-DIVER), 2018, https://doi.org/10.22427/
[28] A. Bal-Price, H.T. Hogberg, K.M. Crofton, M. Daneshian, R.E. FitzGerald, NTP-DATA-002-00062-0001-0000-1.
E. Fritsche, T. Heinonen, S. Hougaard Bennekou, S. Klima, A.H. Piersma,

168
M. Sachana et al. Reproductive Toxicology 103 (2021) 159–170

[52] M. Sachana, A. Rolaki, A. Bal-Price, Development of the Adverse Outcome [79] H. Jinnou, M. Sawada, K. Kawase, N. Kaneko, V. Herranz-Pérez, T. Miyamoto,
Pathway (AOP): chronic binding of antagonist to N-methyl-d-aspartate receptors T. Kawaue, T. Miyata, Y. Tabata, T. Akaike, J.M. García-Verdugo, I. Ajioka,
(NMDARs) during brain development induces impairment of learning and S. Saitoh, K. Sawamoto, Radial glial fibers promote neuronal migration and
memory abilities of children, Toxicol. Appl. Pharmacol. 354 (2018) 153–175. functional recovery after neonatal brain injury, Cell Stem Cell 22 (1) (2018)
[53] K.H. Stansfield, J.R. Pilsner, Q. Lu, R.O. Wright, T.R. Guilarte, Dysregulation of 128–137, e9.
BDNF-TrkB signaling in developing hippocampal neurons by Pb(2+): [80] R. Belvindrah, D. Graus-Porta, S. Goebbels, K.A. Nave, U. Müller, Beta1 integrins
implications for an environmental basis of neurodevelopmental disorders, in radial glia but not in migrating neurons are essential for the formation of cell
Toxicol. Sci. 127 (1) (2012) 277–295. layers in the cerebral cortex, J. Neurosci. 27 (50) (2007) 13854–13865.
[54] K.E. Light, Y. Ge, S.M. Belcher, Early postnatal ethanol exposure selectively [81] Y. Hirota, K. Nakajima, Control of neuronal migration and aggregation by reelin
decreases BDNF and truncated TrkB-T2 receptor mRNA expression in the rat signaling in the developing cerebral cortex, Front. Cell Dev. Biol. 5 (2017) 40.
cerebellum, Brain Res. Mol. Brain Res. 93 (1) (2001) 46–55. [82] S. Chiaramello, G. Dalmasso, L. Bezin, D. Marcel, F. Jourdan, P. Peretto,
[55] S. Shojaei, S. Ghavami, M.R. Panjehshahin, A.A. Owji, Effects of ethanol on the A. Fasolo, S. De Marchis, BDNF/ TrkB interaction regulates migration of SVZ
expression level of various BDNF mRNA isoforms and their encoded protein in the precursor cells via PI3-K and MAP-K signalling pathways, Eur. J. Neurosci. 26 (7)
Hippocampus of adult and embryonic rats, Int. J. Mol. Sci. 16 (12) (2015) (2007) 1780–1790.
30422–30437. [83] K. Khodosevich, H. Monyer, Signaling in migrating neurons: from molecules to
[56] Y. Yu, D. Xu, S. Cheng, L. Zhang, Z. Shi, J. Qin, Z. Zhang, H. Wang, Prenatal networks, Front. Neurosci. 5 (2011) 28.
ethanol exposure enhances the susceptibility to depressive behavior of adult [84] K. Khodosevich, P.H. Seeburg, H. Monyer, Major signaling pathways in migrating
offspring rats fed a high‑fat diet by affecting BDNF‑associated pathway, Int. J. neuroblasts, Front. Mol. Neurosci. 2 (2009) 7.
Mol. Med. 45 (2) (2020) 365–374. [85] Y. Elkabetz, G. Panagiotakos, G. Al Shamy, N.D. Socci, V. Tabar, L. Studer,
[57] A.S. Howard, R. Bucelli, D.A. Jett, D. Bruun, D. Yang, P.J. Lein, Chlorpyrifos Human ES cell-derived neural rosettes reveal a functionally distinct early neural
exerts opposing effects on axonal and dendritic growth in primary neuronal stem cell stage, Genes Dev. 22 (2) (2008) 152–165.
cultures, Toxicol. Appl. Pharmacol. 207 (2) (2005) 112–124. [86] Y. Hirabayashi, Y. Itoh, H. Tabata, K. Nakajima, T. Akiyama, N. Masuyama,
[58] A.A. Adigun, F.J. Seidler, T.A. Slotkin, Disparate developmental neurotoxicants Y. Gotoh, The Wnt/beta-catenin pathway directs neuronal differentiation of
converge on the cyclic AMP signaling cascade, revealed by transcriptional profiles cortical neural precursor cells, Development (Cambridge, England) 131 (12)
in vitro and in vivo, Brain Res. 1316 (2010) 1–16. (2004) 2791–2801.
[59] S.K. Verma, G. Raheja, K.D. Gill, Role of muscarinic signal transduction and CREB [87] S.M. Hapak, C.V. Rothlin, S. Ghosh, aPKC in neuronal differentiation, maturation
phosphorylation in dichlorvos-induced memory deficits in rats: an acetylcholine and function, Neuronal Signal. 3 (3) (2019). Ns20190019.
independent mechanism, Toxicology 256 (3) (2009) 175–182. [88] S.B. Rosso, N.C. Inestrosa, WNT signaling in neuronal maturation and
[60] EFSA, Public Consultation on a Draft Scientific Opinion on Development of synaptogenesis, Front. Cell. Neurosci. 7 (2013) 103.
Integrated Approaches to Testing and Assessment (IATA) on Developmental [89] Z. He, Q. Yu, Identification and characterization of functional modules reflecting
Neurotoxicity (DNT) Risk Assessment, 2021. https://www.efsa.europa.eu transcriptome transition during human neuron maturation, BMC Genomics 19 (1)
/en/consultations/call/public-consultation-draft-scientific-opinion-development. (2018) 262.
[61] N. Moya, J. Cutts, T. Gaasterland, K. Willert, D.A. Brafman, Endogenous WNT [90] S. Liebau, B. Vaida, A. Storch, T.M. Boeckers, Maturation of synaptic contacts in
signaling regulates hPSC-derived neural progenitor cell heterogeneity and differentiating neural stem cells, Stem Cells 25 (7) (2007) 1720–1729.
specifies their regional identity, Stem Cell Rep. 3 (6) (2014) 1015–1028. [91] C.W. He, C.P. Liao, C.L. Pan, Wnt signalling in the development of axon, dendrites
[62] A. Strano, E. Tuck, V.E. Stubbs, F.J. Livesey, Variable outcomes in neural and synapses, Open Biol. 8 (10) (2018).
differentiation of human PSCs arise from intrinsic differences in developmental [92] A. Kumar, H.A. LaVoie, D.J. DiPette, U.S. Singh, Ethanol neurotoxicity in the
signaling pathways, Cell Rep. 31 (10) (2020) 107732. developing cerebellum: underlying mechanisms and implications, Brain Sci. 3 (2)
[63] D.A. Brafman, Generation, expansion, and differentiation of human pluripotent (2013) 941–963.
stem cell (hPSC) derived neural progenitor cells (NPCs), Methods Mol. Biol. 1212 [93] D. Chen, S. Hu, Z. Wu, J. Liu, S. Li, The role of MiR-132 in regulating neural stem
(2015) 87–102. cell proliferation, differentiation and neuronal maturation, Cell. Physiol.
[64] I. Imayoshi, H. Shimojo, M. Sakamoto, T. Ohtsuka, R. Kageyama, Genetic Biochem. 47 (6) (2018) 2319–2330.
visualization of notch signaling in mammalian neurogenesis, Cell. Mol. Life Sci.: [94] M.A. Moskowitz, E.H. Lo, Neurogenesis and apoptotic cell death, Stroke 34 (2)
CMLS 70 (12) (2013) 2045–2057. (2003) 324–326.
[65] D.E. Ehrlich, S.A. Josselyn, Plasticity-related genes in brain development and [95] K. van Leyen, S.R. Lee, M.A. Moskowitz, E.H. Lo, Neurogenesis and apoptotic cell
amygdala-dependent learning, Genes Brain Behav. 15 (1) (2016) 125–143. death, in: D. Janigro (Ed.), The Cell Cycle in the Central Nervous System, Humana
[66] R.F. Hevner, Brain overgrowth in disorders of RTK-PI3K-AKT signaling: a mosaic Press, 2006, pp. 71–79.
of malformations, Semin. Perinatol. 39 (1) (2015) 36–43. [96] R. Petri, J. Malmevik, L. Fasching, M. Åkerblom, J. Jakobsson, miRNAs in brain
[67] D.Y. Lee, Roles of mTOR signaling in brain development, Exp. Neurobiol. 24 (3) development, Exp. Cell Res. 321 (1) (2014) 84–89.
(2015) 177–185. [97] M. Penisson, J. Ladewig, R. Belvindrah, F. Francis, Corrigendum: Genes and
[68] J. Patel, K. Landers, H. Li, R.H. Mortimer, K. Richard, Thyroid hormones and fetal Mechanisms Involved in the Generation and Amplification of Basal Radial Glial
neurological development, J. Endocrinol. 209 (1) (2011) 1–8. Cells, Front. Cell. Neurosci. 13 (2019) 462.
[69] G.M. de Escobar, M.J. Obregón, F.E. del Rey, Maternal thyroid hormones early in [98] C. Gregg, S. Weiss, Generation of functional radial glial cells by embryonic and
pregnancy and fetal brain development, best practice & research, Clin. adult forebrain neural stem cells, J. Neurosci. 23 (37) (2003) 11587–11601.
Endocrinol. Metabol. 18 (2) (2004) 225–248. [99] L. Ever, N. Gaiano, Radial’ glial’ progenitors: neurogenesis and signaling, Curr.
[70] S.G. Gonzalez Malagon, A.M. Lopez Muñoz, D. Doro, T.G. Bolger, E. Poon, E. Opin. Neurobiol. 15 (1) (2005) 29–33.
R. Tucker, H. Adel Al-Lami, M. Krause, C.J. Phiel, L. Chesler, K.J. Liu, Glycogen [100] P. Callihan, M.W. Ali, H. Salazar, N. Quach, X. Wu, S.L. Stice, S.B. Hooks,
synthase kinase 3 controls migration of the neural crest lineage in mouse and Convergent regulation of neuronal differentiation and Erk and Akt kinases in
Xenopus, Nat. Commun. 9 (1) (2018) 1126. human neural progenitor cells by lysophosphatidic acid, sphingosine 1-phos­
[71] S. Jang, H.H. Cho, Y.B. Cho, J.S. Park, H.S. Jeong, Functional neural phate, and LIF: specific roles for the LPA1 receptor, ASN Neuro 6 (6) (2014).
differentiation of human adipose tissue-derived stem cells using bFGF and [101] A. Michelucci, A. Bithell, M.J. Burney, C.E. Johnston, K.Y. Wong, S.W. Teng,
forskolin, BMC Cell Biol. 11 (2010) 25. J. Desai, N. Gumbleton, G. Anderson, L.W. Stanton, B.P. Williams, N.J. Buckley,
[72] S.S. Kim, J.M. Choi, J.W. Kim, D.S. Ham, S.H. Ghil, M.K. Kim, Y. Kim-Kwon, S. The neurogenic potential of astrocytes is regulated by inflammatory signals, Mol.
Y. Hong, S.C. Ahn, S.U. Kim, Y.D. Lee, H. Suh-Kim, cAMP induces neuronal Neurobiol. 53 (6) (2016) 3724–3739.
differentiation of mesenchymal stem cells via activation of extracellular signal- [102] T. Wang, W. Yuan, Y. Liu, Y. Zhang, Z. Wang, X. Zhou, G. Ning, L. Zhang, L. Yao,
regulated kinase/MAPK, Neuroreport 16 (12) (2005) 1357–1361. S. Feng, X. Kong, The role of the JAK-STAT pathway in neural stem cells, neural
[73] K.M. Kompisch, C. Lange, D. Steinemann, B. Skawran, B. Schlegelberger, progenitor cells and reactive astrocytes after spinal cord injury, Biomed. Rep. 3
R. Müller, U. Schumacher, Neurogenic transdifferentiation of human adipose- (2) (2015) 141–146.
derived stem cells? A critical protocol reevaluation with special emphasis on cell [103] M. Ernst, B.J. Jenkins, Acquiring signalling specificity from the cytokine receptor
proliferation and cell cycle alterations, Histochem. Cell Biol. 134 (5) (2010) gp130, Trends in genetics: TIG 20 (1) (2004) 23–32.
453–468. [104] M.F. McManus, L.C. Chen, I. Vallejo, M. Vallejo, Astroglial differentiation of
[74] M. Cargnello, P.P. Roux, Activation and function of the MAPKs and their cortical precursor cells triggered by activation of the cAMP-dependent signaling
substrates, the MAPK-activated protein kinases, Microbiol. Mol. Biol. Rev. 75 (1) pathway, J. Neurosci. 19 (20) (1999) 9004–9015.
(2011) 50–83. [105] S. Paco, M. Hummel, V. Plá, L. Sumoy, F. Aguado, Cyclic AMP signaling restricts
[75] A. Janesick, S.C. Wu, B. Blumberg, Retinoic acid signaling and neuronal activation and promotes maturation and antioxidant defenses in astrocytes, BMC
differentiation, Cell. Mol. Life Sci. 72 (8) (2015) 1559–1576. Genomics 17 (2016) 304.
[76] D.R. Kaplan, F.D. Miller, Neurotrophin signal transduction in the nervous system, [106] L. Pous, S.S. Deshpande, S. Nath, S. Mezey, S.C. Malik, S. Schildge, C. Bohrer,
Curr. Opin. Neurobiol. 10 (3) (2000) 381–391. K. Topp, D. Pfeifer, F. Fernández-Klett, S. Doostkam, D.K. Galanakis, V. Taylor,
[77] C.A. Lafourcade, T.V. Lin, D.M. Feliciano, L. Zhang, L.S. Hsieh, A. Bordey, Rheb K. Akassoglou, C. Schachtrup, Fibrinogen induces neural stem cell differentiation
activation in subventricular zone progenitors leads to heterotopia, ectopic into astrocytes in the subventricular zone via BMP signaling, Nat. Commun. 11
neuronal differentiation, and rapamycin-sensitive olfactory micronodules and (1) (2020) 630.
dendrite hypertrophy of newborn neurons, J. Neurosci. 33 (6) (2013) 2419–2431. [107] A. Chandrasekaran, H.X. Avci, M. Leist, J. Kobolák, A. Dinnyés, Astrocyte
[78] D.S. Kang, Y.R. Yang, C. Lee, S. Kim, S.H. Ryu, P.G. Suh, Roles of differentiation of human pluripotent stem cells: new tools for neurological
phosphoinositide-specific phospholipase Cγ1 in brain development, Adv. Biol. disorder research, Front. Cell. Neurosci. 10 (2016) 215.
Regul. 60 (2016) 167–173. [108] H. Asano, M. Aonuma, T. Sanosaka, J. Kohyama, M. Namihira, K. Nakashima,
Astrocyte differentiation of neural precursor cells is enhanced by retinoic acid

169
M. Sachana et al. Reproductive Toxicology 103 (2021) 159–170

through a change in epigenetic modification, Stem Cells 27 (11) (2009) [130] B.N. Patel, D.L. Van Vactor, axon guidance: the cytoplasmic tail, Curr. Opin. Cell
2744–2752. Biol. 14 (2) (2002) 221–229.
[109] W. Ge, K. Martinowich, X. Wu, F. He, A. Miyamoto, G. Fan, G. Weinmaster, Y. [131] X.B. Yuan, M. Jin, X. Xu, Y.Q. Song, C.P. Wu, M.M. Poo, S. Duan, Signalling and
E. Sun, Notch signaling promotes astrogliogenesis via direct CSL-mediated glial crosstalk of Rho GTPases in mediating axon guidance, Nat. Cell Biol. 5 (1) (2003)
gene activation, J. Neurosci. Res. 69 (6) (2002) 848–860. 38–45.
[110] B.Y. Hu, Z.W. Du, S.C. Zhang, Differentiation of human oligodendrocytes from [132] L. Stappert, B. Roese-Koerner, O. Brüstle, The role of microRNAs in human neural
pluripotent stem cells, Nat. Protoc. 4 (11) (2009) 1614–1622. stem cells, neuronal differentiation and subtype specification, Cell Tissue Res. 359
[111] S.M. Kang, M.S. Cho, H. Seo, C.J. Yoon, S.K. Oh, Y.M. Choi, D.W. Kim, Efficient (1) (2015) 47–64.
induction of oligodendrocytes from human embryonic stem cells, Stem cells [133] R.J. Johnston Jr., S. Chang, J.F. Etchberger, C.O. Ortiz, O. Hobert, MicroRNAs
(Dayton, Ohio) 25 (2) (2007) 419–424. acting in a double-negative feedback loop to control a neuronal cell fate decision,
[112] Y. Li, M. Liu, Y. Yan, S.T. Yang, Neural differentiation from pluripotent stem cells: Proc. Natl. Acad. Sci. U. S. A. 102 (35) (2005) 12449–12454.
the role of natural and synthetic extracellular matrix, World J. Stem Cells 6 (1) [134] R.J. Johnston, O. Hobert, A microRNA controlling left/right neuronal asymmetry
(2014) 11–23. in Caenorhabditis elegans, Nature 426 (6968) (2003) 845–849.
[113] F. Sher, V. Balasubramaniyan, E. Boddeke, S. Copray, Oligodendrocyte [135] B. Bonev, A. Pisco, N. Papalopulu, MicroRNA-9 reveals regional diversity of
differentiation and implantation: new insights for remyelinating cell therapy, neural progenitors along the anterior-posterior axis, Dev. Cell 20 (1) (2011)
Curr. Opin. Neurol. 21 (5) (2008) 607–614. 19–32.
[114] M. Sundberg, A. Hyysalo, H. Skottman, S. Shin, M. Vemuri, R. Suuronen, [136] B. Bonev, P. Stanley, N. Papalopulu, MicroRNA-9 modulates Hes1 ultradian
S. Narkilahti, A xeno-free culturing protocol for pluripotent stem cell-derived oscillations by forming a double-negative feedback loop, Cell Rep. 2 (1) (2012)
oligodendrocyte precursor cell production, Regen. Med. 6 (4) (2011) 449–460. 10–18.
[115] M. Sundberg, H. Skottman, R. Suuronen, S. Narkilahti, Production and isolation of [137] M. Shibata, D. Kurokawa, H. Nakao, T. Ohmura, S. Aizawa, MicroRNA-9
NG2+ oligodendrocyte precursors from human embryonic stem cells in defined modulates Cajal-Retzius cell differentiation by suppressing Foxg1 expression in
serum-free medium, Stem Cell Res. 5 (2) (2010) 91–103. mouse medial pallium, J. Neurosci. 28 (41) (2008) 10415–10421.
[116] L.J. Chew, W. Coley, Y. Cheng, V. Gallo, Mechanisms of regulation of [138] M. Shibata, H. Nakao, H. Kiyonari, T. Abe, S. Aizawa, MicroRNA-9 regulates
oligodendrocyte development by p38 mitogen-activated protein kinase, neurogenesis in mouse telencephalon by targeting multiple transcription factors,
J. Neurosci. 30 (33) (2010) 11011–11027. J. Neurosci. 31 (9) (2011) 3407–3422.
[117] S.A. Goldman, N.J. Kuypers, How to make an oligodendrocyte, Development [139] A. de Chevigny, N. Coré, P. Follert, M. Gaudin, P. Barbry, C. Béclin, H. Cremer,
(Cambridge, England) 142 (23) (2015) 3983–3995. miR-7a regulation of Pax6 controls spatial origin of forebrain dopaminergic
[118] J.D. Haines, D.L. Fulton, S. Richard, G. Almazan, p38 mitogen-activated protein neurons, Nat. Neurosci. 15 (8) (2012) 1120–1126.
kinase pathway regulates genes during proliferation and differentiation in [140] A. Anderegg, H.P. Lin, J.A. Chen, G. Caronia-Brown, N. Cherepanova, B. Yun,
oligodendrocytes, PLoS One 10 (12) (2015) e0145843. M. Joksimovic, J. Rock, B.D. Harfe, R. Johnson, R. Awatramani, An Lmx1b-
[119] S. Ahmed, H.T. Gan, C.S. Lam, A. Poonepalli, S. Ramasamy, Y. Tay, M. Tham, Y. miR135a2 regulatory circuit modulates Wnt1/Wnt signaling and determines the
H. Yu, Transcription factors and neural stem cell self-renewal, growth and size of the midbrain dopaminergic progenitor pool, PLoS Genet. 9 (12) (2013)
differentiation, Cell Adh. Migr. 3 (4) (2009) 412–424. e1003973.
[120] J. Wang, S.U. Pol, A.K. Haberman, C. Wang, M.A. O’Bara, F.J. Sim, Transcription [141] C. Leucht, C. Stigloher, A. Wizenmann, R. Klafke, A. Folchert, L. Bally-Cuif,
factor induction of human oligodendrocyte progenitor fate and differentiation, MicroRNA-9 directs late organizer activity of the midbrain-hindbrain boundary,
Proc. Natl. Acad. Sci. U. S. A. 111 (28) (2014) E2885–94. Nat. Neurosci. 11 (6) (2008) 641–648.
[121] M. Wegner, Transcriptional control in myelinating glia: the basic recipe, Glia 29 [142] S. Decembrini, D. Bressan, R. Vignali, L. Pitto, S. Mariotti, G. Rainaldi, X. Wang,
(2) (2000) 118–123. M. Evangelista, G. Barsacchi, F. Cremisi, MicroRNAs couple cell fate and
[122] D.A. Menassa, D. Gomez-Nicola, Microglial dynamics during human brain developmental timing in retina, Proc. Natl. Acad. Sci. U. S. A. 106 (50) (2009)
development, Front. Immunol. 9 (2018) 1014. 21179–21184.
[123] K.M. Lenz, L.H. Nelson, Microglia and beyond: innate immune cells as regulators [143] A. La Torre, S. Georgi, T.A. Reh, Conserved microRNA pathway regulates
of brain development and behavioral function, Front. Immunol. 9 (2018) 698. developmental timing of retinal neurogenesis, Proc. Natl. Acad. Sci. U. S. A. 110
[124] F.R.S. Lima, A.C. da Fonseca, G.P. Faria, L.G. Dubois, T.R. Alves, J. Faria, (26) (2013) E2362–70.
V. Moura Neto, The origin of microglia and the development of the brain, in: [144] N.S. Asli, M. Kessel, Spatiotemporally restricted regulation of generic motor
H. Ulrich (Ed.), Perspectives of Stem Cells, Springer, 2010, pp. 171–190. neuron programs by miR-196-mediated repression of Hoxb8, Dev. Biol. 344 (2)
[125] P. Arthur-Farraj, K. Wanek, J. Hantke, C.M. Davis, A. Jayakar, D.B. Parkinson, (2010) 857–868.
R. Mirsky, K.R. Jessen, Mouse schwann cells need both NRG1 and cyclic AMP to [145] J.A. Chen, Y.P. Huang, E.O. Mazzoni, G.C. Tan, J. Zavadil, H. Wichterle, Mir-17-
myelinate, Glia 59 (5) (2011) 720–733. 3p controls spinal neural progenitor patterning by regulating Olig2/Irx3 cross-
[126] K.R. Monk, S.G. Naylor, T.D. Glenn, S. Mercurio, J.R. Perlin, C. Dominguez, C. repressive loop, Neuron 69 (4) (2011) 721–735.
B. Moens, W.S. Talbot, A G protein-coupled receptor is essential for Schwann cells [146] G. Luxenhofer, M.S. Helmbrecht, J. Langhoff, S.A. Giusti, D. Refojo, A.B. Huber,
to initiate myelination, Science (New York, N.Y.) 325 (5946) (2009) 1402–1405. MicroRNA-9 promotes the switch from early-born to late-born motor neuron
[127] R.D. Doddrell, X.P. Dun, R.M. Moate, K.R. Jessen, R. Mirsky, D.B. Parkinson, populations by regulating Onecut transcription factor expression, Dev. Biol. 386
Regulation of Schwann cell differentiation and proliferation by the Pax-3 (2) (2014) 358–370.
transcription factor, Glia 60 (9) (2012) 1269–1278. [147] G. Otaegi, A. Pollock, J. Hong, T. Sun, MicroRNA miR-9 modifies motor neuron
[128] K.R. Monk, M.L. Feltri, C. Taveggia, New insights on Schwann cell development, columns by a tuning regulation of FoxP1 levels in developing spinal cords,
Glia 63 (8) (2015) 1376–1393. J. Neurosci. 31 (3) (2011) 809–818.
[129] D. Kiryushko, I. Korshunova, V. Berezin, E. Bock, Neural cell adhesion molecule [148] C.G. Dotti, J.A. Esteban, M.D. Ledesma, Lipid dynamics at dendritic spines, Front.
induces intracellular signaling via multiple mechanisms of Ca2+ homeostasis, Neuroanat. 8 (2014) 76.
Mol. Biol. Cell 17 (5) (2006) 2278–2286. [149] H.J. McCrea, P. De Camilli, Mutations in phosphoinositide metabolizing enzymes
and human disease, Physiology (Bethesda, Md.) 24 (2009) 8–16.

170

You might also like