Research Article
More Information

Submitted: November 12, 2024 | Approved: November 14, 2024 | Published: November 15 2024

How to cite this article: Kebieche N, Ali FL, Yim S, Ali M, Lambert M, Soulimani R. Exploring Environmental Neurotoxicity Assessment Using Human Stem Cell-Derived Models. J Stem Cell Ther Transplant. 2024; 8(1): 054-068. Available from: https://dx.doi.org/10.29328/journal.jsctt.1001044

DOI: 10.29328/journal.jsctt.1001044

Copyright License: © 2024 Kebieche N, et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Keywords: Neurotoxicity; Neurodevelopmental diseases; Neurodegenerative diseases; Pesticides; Human-induced Pluripotent Stem Cells (hiPSCs); Developmental neurotoxicity; Predictive toxicology

 FullText PDF

Exploring Environmental Neurotoxicity Assessment Using Human Stem Cell-Derived Models

Narimane Kebieche1*, Farzana Liakath Ali2, Seungae Yim1, Mohamed Ali2, Claude Lambert3 and Rachid Soulimani1*

1LCOMS/Food Neurotoxicology and Bioactivity, University of Lorraine, Metz, France
2Department of Obstetrics and Gynecology, University of Chicago, USA
3Immunology Laboratory, University Hospital Saint-Etienne, France

*Address for Correspondence: Rachid Soulimani, Professor, LCOMS/Food Neurotoxicology and Bioactivity, University of Lorraine, Metz, France, Email: rachid.soulimani@univ-lorraine.fr

Narimane Kebieche, Ph.D. Candidate, 3rd year, LCOMS/Food Neurotoxicology and Bioactivity, University of Lorraine, Metz, France, Email: narimane.kebieche@univ-lorraine.fr

Neurotoxicity is increasingly recognized as a critical factor impacting long-term health, with growing evidence linking it to both neurodevelopmental and neurodegenerative diseases. Pesticides, widely used in agriculture and industry, have emerged as significant contributors to neurotoxic risk, given their capacity to disrupt key neurodevelopmental processes at low exposure levels. As conventional animal models present limitations in interspecies translation, human-derived neuron-based in vitro screening strategies are urgently needed to assess potential toxicants accurately. Human-induced pluripotent stem cells (hiPSCs) offer an innovative and scalable source for human-specific neuronal models that complement traditional animal-based approaches and support the development of predictive assays for neurotoxicity. Recent various stem cell models, including 2D cultures, 3D organoids, and microfluidic systems, are now available, advancing predictive neurotoxicology by simulating key aspects of human neural development and function. With the integration of High-Throughput (HT) and High-Content (HC) screening methodologies, these hiPSC-based systems enable efficient, large-scale evaluation of chemical effects on neural cells, enhancing our ability to detect early biomarkers of neurotoxic effects. Identifying early biomarkers of neurotoxic is essential to developing therapeutic interventions before irreversible damage occurs. This is particularly crucial in the context of developmental neurotoxicity, where early exposure to toxicants can have lifelong consequences. This review specifically presents an in-depth overview of the current progress in hiPSC-derived neural models and their applications in neurotoxicity testing, with a specific focus on their utility in assessing pesticide-induced neurotoxicity. Emphasizing future research priorities, we highlight the potential of these models to transform predictive toxicology, offering more human-relevant assessments and advancing the field toward a more precise evaluation of environmental neurotoxicants.

Human brain development involves complex processes susceptible to chemical disruption, which can lead to irreversible nervous system impairments and elevate risks of neurodevelopmental and neurodegenerative diseases [1,2]. Research indicates that the immature nervous system, particularly during fetal and early postnatal periods when the Blood-Brain Barrier (BBB) is still developing, is especially vulnerable to chemical exposure [3,4]. Evaluating neurotoxic chemicals thus requires attention to exposure timing, as critical developmental windows show heightened sensitivity to toxicants and are associated with long-lasting neurological effects [5]. There is strong evidence linking prenatal exposure to neurotoxicants with structural and functional deficits later in life, often mediated by epigenetic changes affecting brain development [6,7].

The exposome, introduced by Chris Wild in 2005, encapsulates all environmental exposures across an individual’s lifetime, advancing risk assessment and understanding gene-environment interactions [8,9]. This concept, combined with epigenetics, allows researchers to explore how environmental factors modulate gene expression through mechanisms such as DNA methylation, illuminating the pathways by which cumulative exposures during critical developmental periods can affect brain health [10].

As a response to these insights, human induced pluripotent stem cell (hiPSC)-derived neuronal cultures have emerged as practical alternatives to traditional animal models. hiPSCs replicate human physiology more closely, providing better predictions of neurotoxic effects [11-13]. Available in two-dimensional (2D), three-dimensional (3D), and microfluidic formats, hiPSC models allow human-relevant neurotoxicity assessments and hold promise for predictive screening to identify early biomarkers of neurological disorders [14,15].

This review discusses iPSC-based brain models, particularly their applications in neurotoxicity assessment, with an emphasis on pesticide effects. By examining these models, we aim to highlight their potential in identifying early biomarkers and in elucidating mechanisms underlying pesticide-induced neurotoxicity.

The relevance of stem cell models

Over the years, neurotoxicity research has utilized a range of in vitro models, each offering unique insights but also presenting limitations. Traditional models like immortalized cell lines and primary cultures have been instrumental in focusing on specific neural cell types for targeted neurotoxicology studies, but are inherently restricted by genetic modifications and short lifespans, leading to compromised physiological relevance [13,16].

Stem cells have since emerged as a versatile platform in toxicology. Offering self-renewing, pluripotent, or multipotent sources, stem cells allow for differentiation into diverse neural lineages, thus enhancing model relevance for human applications [17]. Depending on origin, these cells are categorized into Adult Stem Cells (ASCs), Fetal Stem Cells (FSCs), Embryonic Stem Cells (ESCs), and induced Pluripotent Stem Cells (iPSCs). However, ESCs, derived from early-stage embryos, offer high differentiation potential but face significant ethical constraints [18,19]. ASCs and FSCs, while ethically viable, are limited to specific lineages and lack the full pluripotency seen in ESCs [20,21].

Induced pluripotent stem cells (iPSCs) are somatic cells reprogrammed to a pluripotent state, an achievement first realized in 2006 with murine cells by Yamanaka’s team [22] and later applied to human cells [23]. The process involves introducing key transcription factors—such as c-Myc, Klf4, Sox2, and Oct3/4—using retroviral vectors, enabling iPSCs to differentiate into various cell types. With robust self-renewal and pluripotency, human iPSCs (hiPSCs) provide significant potential as human-relevant models for neurotoxicity and developmental neurotoxicity (DNT) screening [24].

Induced Pluripotent Stem Cell (iPSC) models: Advancing neurotoxicity assessment

Induced pluripotent stem cells (iPSCs) offer a promising alternative for neurotoxicity assessment, particularly given the ethical and practical limitations associated with human embryonic stem cells (hESCs). Unlike immortalized cell lines and primary cultures, iPSCs can differentiate into a range of neural cell types, including neurons and glial cells. These cells enable comprehensive DNT assessment through endpoints such as proliferation, differentiation, apoptosis, migration, neurite outgrowth, and network activity [25,26].

hiPSC-derived models address the ethical concern surrounding hESC and the limitations of interspecies extrapolation by more closely reflecting human neurodevelopment, providing ethically sound data that align with the 3R principles (Replacement, Reduction, and Refinement) [27,28]. Furthermore, these models enable the examination of gene-environment interactions, assessing how environmental factors, combined with genetic predispositions, influence neural function and contribute to pathological phenotypes [29].

For neurotoxicity assessment, iPSC models can mimic long-term exposure effects, elucidating mechanisms of diseases thought to originate from fetal exposure and revealing cumulative neurodevelopmental impacts of toxicants over time. Additionally, hiPSCs can be cryopreserved, generating diverse, consistent cell types—including neural stem cells, dopaminergic neurons, cortical glutamatergic neurons, oligodendrocytes, and astrocytes—facilitating scalability for neurotoxicity screening [30-32].

Advanced electrophysiological techniques, such as multi-well micro-electrode arrays (mwMEA) and calcium imaging, allow hiPSC-derived neurons to display mature network properties with spontaneous activity, enhanced by astrocyte co-culture [33-35]. These complex networks respond to neurotoxic compounds, providing data on excitatory and inhibitory balance crucial for toxicity studies [35-38].

Overall, hiPSC-based models hold great potential for advancing neurotoxicology, offering human-relevant data that surpasses traditional models in predicting adverse outcomes.

Platforms for neurotoxicity assessment using iPSC brain models

2D hiPSC neural differentiation models: Human-induced pluripotent stem cells (hiPSCs) serve as a versatile foundation for generating various neural cell types in 2D cultures, providing a practical framework for neurotoxicity studies. For example, hiPSCs from both healthy individuals and Parkinson’s disease patients have been differentiated into dopaminergic neurons, establishing useful models for disease and neurotoxicity screening [39]. hiPSC-derived GABAergic interneurons, crucial for maintaining neural excitation balance, are generated through targeted transcription factors, facilitating the rapid differentiation into functional neurons [40,41]. Additionally, motor neuron differentiation protocols enable the creation of spinal motor neurons that closely mimic human physiology, making them highly relevant for specific neurodegenerative studies [42,43].

Co-culturing hiPSCs-derived neurons with astrocytes enhances functional maturity in these neurons, with human astrocytes proving preferable over rodent cells to avoid interspecies variability [44]. Simplified differentiation protocols, such as those developed by Gunhanlar, et al. have refined this approach, allowing mature cortical neurons to form without astrocyte co-culture. This method consistently generated a 60:40 ratio of neurons and astrocytes, which result from a common forebrain neural progenitor and exhibit a similar neurodevelopmental process in vivo, thus providing streamlined model systems without sacrificing cellular complexity [45].

Although 2D hiPSC neural differentiation models are highly effective for rapid, HT neurotoxicity screening and allow detailed analysis of individual cell types, they inherently lack the structural complexity required to fully replicate multicellular interactions and neural network dynamics found in vivo. This limitation suggests the need for more advanced models that incorporate the intricate cellular interactions critical to accurate neurodevelopmental and neurotoxicological assessments.

Advancement from 2D to 3D models: The transition from 2D to 3D hiPSC cultures represents a significant advancement in creating in vitro models that more closely mimic the complex spatial organization of the human brain. While 2D models allow for efficient High-Throughput Screening (HTS), their limited dimensionality constrains the replication of in vivo-like cellular interactions and physiological relevance [16]. In contrast, 3D models, such as brain organoids and cortical spheroids, offer enhanced biological fidelity by fostering the development of cellular structures and interactions observed in the central nervous system [46]. These models not only support processes such as neural differentiation and synaptic connectivity but also allow for the organization of neural progenitors and mature neurons in a way that better resembles human brain architecture [46]. Therefore, 3D cellular organotypic models, including neural spheroids and organoids, have emerged as an alternative toxicity screening platform to traditional 2D in vitro and animal model approaches [47].

Through 3D induction, cell fate specification and neural network complexity are promoted, facilitating deeper insights into how chemicals may disrupt neurodevelopmental processes [48]. This shift toward three-dimensionality provides a promising bridge between in vitro studies and in vivo applicability, making 3D hiPSC models highly relevant for toxicology research that requires spatially complex cell interactions.

Efforts to further standardize and scale these models will be instrumental in expanding their accessibility for HT applications, thereby increasing the translational impact of in vitro neurotoxicity screening. Figure 1 illustrates both 2D and 3D models and their application in the field of neurotoxicity assessment.


Download Image

Figure 1: Comparison of 2D and 3D Cell Culture Models in Neurotoxicity Assessment. (A) Cell Culture Systems: (A.a) Traditional 2D cell culture on flat surfaces; (A.b) 3D cell culture systems, including scaffold-free methods (using ultra-low attachment or ECM-coated plates) and scaffold-based systems with rigid or soft biomaterials (e.g., hydrogels) that mimic ECM architecture. (B) Neurotoxicity Assessment and Chemical Screening: iPSC-derived 2D and 3D models are applied to screen and evaluate the neurotoxicity of chemicals using key neurotoxicity endpoints. (Figures created with BioRender.com).

3D hiPSC-derived brain organoids: 3D cerebral organoids derived from human pluripotent stem cells (hPSCs) represent an advanced model for studying human brain development and disease, as they partially replicate the early stages of fetal brain formation in vitro [49]. Organoids begin with embryoid body formation and progress through differentiation of the three germ layers (endoderm, mesoderm, and ectoderm), ultimately yielding structures that resemble specific brain regions, such as the forebrain, midbrain, and hindbrain, depending on differentiation cues provided [50]. Different approaches have led to region-specific organoid models, each tailored to mimic distinct aspects of brain development. For instance, forebrain organoids accurately model the ventricular zone structure with polarized SOX2-positive radial glial cells, essential for studying cortical layer formation and neuronal migration [51,52]. Midbrain organoids, replicate dopaminergic neuron populations, providing a platform for neurodegenerative disease studies [53], while cerebellar and hippocampal organoids mimic synaptic organization and connectivity relevant to neurodevelopmental disorders [54,55].

Recent advances include the development of “assembloids,” or fused organoids, where multiple brain regions are combined to investigate cellular interactions and interneuron migration. Birey, et al. fused dorsal and ventral forebrain spheroids, simulating interneuron migration as observed in vivo [56]. Another notable advancement involves vascularized organoids, which enhance viability and cellular complexity by incorporating endothelial cells to simulate the Blood-Brain Barrier (BBB) and promote nutrient exchange, a crucial factor in long-term culture viability [57,58].

These 3D organoid models provide a versatile platform for neurotoxicity testing, allowing for dynamic interaction studies and exposure assessments that reflect more accurate human brain physiology than traditional 2D cultures.

Development of iPSC Blood-Brain Barrier (BBB) models: Effective neurotoxicity assessment requires models that account for a substance’s ability to cross the Blood-Brain Barrier (BBB), a highly selective structure that controls molecular access to the Central Nervous System (CNS) [59]. The BBB comprises brain microvascular endothelial cells (BMECs), pericytes, and astrocytes, creating a barrier essential for maintaining CNS homeostasis. In recent years, several iPSC-based BBB models have emerged to replicate this structure, allowing detailed investigations into neurotoxic effects on BBB integrity and function [60,61].

Lippmann, et al. developed an iPSC-derived BMEC model that demonstrated strong barrier properties and efflux transporter activity, essential features of the in vivo BBB [62]. Co-culturing these BMECs with astrocytes has further enhanced their structural and functional integrity, achieving transendothelial electrical resistance (TEER) measurements close to in vivo values [62]. Advances have led to the development of BBB spheroids, in which BMECs, astrocytes, and pericytes self-assemble, enhancing permeability assessments in a 3D context (Urich, et al. 2013).

Organ-chip technology offers further advancements, with microfluidic devices that replicate the BBB’s selective permeability. For instance, Wang, et al. created a BBB-on-chip system using iPSC-derived BMECs and rat astrocytes, successfully demonstrating a functional barrier with dynamic media flow [63]. More recent work by Vatine, et al. established an entirely human BBB-on-chip using iPSC-derived BMECs, astrocytes, and neurons, achieving physiologically relevant TEER values and enabling targeted drug administration studies [64].

These iPSC-based BBB models are promising tools for HT neurotoxicity screening, allowing researchers to simulate the effects of chemical exposure on BBB integrity and CNS homeostasis with human-relevant insights.

Development of iPSC vascularized organoids: The complexity of brain organoids has advanced significantly with efforts to incorporate vascularization, a key factor for mimicking in vivo brain environments and enhancing organoid longevity and function. The absence of vasculature in traditional organoid models restricts nutrient and oxygen diffusion, leading to hypoxic conditions and limiting growth beyond a certain size. Vascularized organoid models overcome these limitations by supporting endothelial cell differentiation and network formation, a critical step toward creating organoids with in vivo-like properties [57].

Various approaches to vascularization have been explored, including implanting organoids into immunodeficient mice, enabling host vasculature to penetrate and support the graft [65]. Mansour, et al. demonstrated this with hiPSC-derived cerebral organoids grafted into the mouse cortex, showing axonal projection and synaptic connectivity with the host tissue over time. Alternatively, vascular networks have been induced in vitro by embedding endothelial cells within organoids or by adding endothelial growth factors, as in the study by Shi, et al. [58], where co-culturing with human umbilical vein endothelial cells led to vascularized cortical organoids capable of supporting neurogenesis.

More advanced methods involve genetic engineering for vascularization, such as Cakir, et al. who used the ETV2 gene to induce early vascular development within cerebral organoids [66]. These vascularized organoids exhibit improved neuronal activity and maturation, closely resembling the structural and functional interactions found in human brain tissue. Such models hold great promise for studying neurovascular interactions in health and disease, providing a human-relevant platform for testing drug efficacy and neurotoxicity.

The new generation of risk assessment: iPSC microfluidic chip models: Microfluidic technology, initially developed to miniaturize lab processes for analytical applications, has evolved into a robust platform for HTS screening and toxicity testing. Characterized by micron-scale channels, microfluidic systems integrate multiple experimental processes into a single device, enhancing reproducibility, reducing reagent use, and decreasing analysis time [67]. Applications in life sciences are particularly transformative, encompassing cell analysis, co-culture systems, and organ-on-chip (OoC) platforms for tissue-specific studies. OoCs, which emulate the architecture and function of human organs through 3D bioengineered constructs, improve cellular viability and offer a controlled environment for studying complex tissue interactions, such as those found in brain-on-chip models, essential for neurotoxicity studies [68].

The dynamic nature of microfluidic systems allows for real-time adjustments to culture conditions, enabling intricate toxicity and pharmacokinetic evaluations. For example, Sances, et al. demonstrated that an iPSC-derived spinal cord chip enhances neuronal activity and gene expression [69], while Liu, et al. created a 3D brain-on-chip model to assess pesticide toxicity on neuronal viability [70]. Further integration of iPSC-derived blood-BBB models has enabled studies on neurovascular interactions under continuous perfusion, enhancing BBB fidelity and drug testing relevance [71]. These findings allow simultaneous study of pharmacokinetics across interconnected tissues, promising a shift toward comprehensive toxicity assessment.

Despite these advances, challenges, including automation compatibility and scalability for HT needs remain. However, recent efforts to create automated, HT-compatible systems, such as the OrganoPlate, have demonstrated the potential of 3D microfluidic systems for screening environmental neurotoxicity at scale [72]. As microfluidic technology matures, its capacity for simulating human-specific responses in neurotoxicity studies positions it as a pivotal tool in regulatory and research applications, advancing both environmental health and personalized medicine. Table 1 summarizes the advantages and disadvantages of iPSCs-derived neural models.

Table 1: Advantages and disadvantages of iPSCs-derived neural models.
  2D 3D organoids and assemblies BBB Vascularized Microfluidics
Advantages Simple, cost-effective.
Reproducible
Highly scalable for rapid cellular, genetic, and signaling studies.
Mimic more in vivo complexity
3D cell-cell and cell-ECM interactions.
Spatial organization.  
Enables TEER (Transendothelial Electrical Resistance) measurements for barrier function evaluation.
Allow permeability studies and multi-cellular co-culture.
Enhanced physiological functionality
Presence of vascular networks.
Improved cell-cell interaction and maturation (compared to BBB)
High sensitivity and precision.
Efficient cell activity detection.
Real-time cell monitoring.
Multi-Condition Analysis.
Analytical Integration (mass spectrometry, DNA sequencing, imaging and biosensors)
Disadvantages Lacks 3D complexity
Limited cell-cell and cell-extracellular interactions (ECM)
Higher cost
(compared to 2D).
Potential for tissue necrosis (lack of homogeneous distribution of oxygen and nutrients)
Lacks vascularity
Material variability used for scaffolding
Lacks full 3D vessel structure.
Doesn’t fully replicate selective transport of in vivo BBB.
Lack fully functional vascular systems.
Require complex culture techniques
Limited size
Limited lifespan and stability
Complex device fabrication.
Issues with sterilization and fluid dynamics.
Challenges for High-Throughput (HT) applications.
High cost (compared to the other models)
Need for specialized expertise.
Applicability of hiPSC-derived neural models for in vitro neurotoxicity screening

Given rising concerns about neurotoxicity resulting from chemical exposures during critical periods of human development, there is an urgent need for efficient in vitro neurotoxicity screening systems that leverage human-derived neurons, thereby reducing reliance on interspecies extrapolation. The human brain, characterized by extensive neuronal networks, complex synaptic connectivity, and a high diversity of gene expression, may be particularly vulnerable to environmental chemicals [73]. Although recently, approximately 200 chemicals have been identified as neurotoxicants impacting human development from tens of thousands of commercial compounds, many neurotoxic substances are likely still remain unidentified [74].

The current neurotoxicity assessment framework, which is heavily reliant on animal studies, is limited by ethical concerns, low throughput, and the inability to efficiently handle large chemical libraries. These limitations highlight an essential need for human-relevant, in vitro models that can be automated for HT screening applications [75-80]. Effective in vitro models should be capable of closely mimicking the complexity of the human brain, including forming functional neural networks composed of diverse neuronal subtypes, excitatory and inhibitory pathways, and key neuroglial cells, such as astrocytes, oligodendrocytes, and microglia, to accurately reflect human neurophysiology [81,82].

hiPSC-derived neuronal and glial models are particularly promising for neurotoxicity assessment, as they allow expansion and differentiation of neural cells from an undifferentiated state in culture, providing a scalable solution for tissue-specific toxicological studies [14,30,83]. hiPSC-derived models are capable of replicating key neurodevelopmental events, from Neural Progenitor Cell (NPC) proliferation to advanced neuronal and glial maturation. This ability offers quantifiable endpoints to assess the impacts of chemical exposure. Disruptions in these processes due to chemical exposure can be measured, providing valuable readouts for in vitro toxicity screening [84]. Table 2 summarizes iPSC-derived model applications in in vitro neurotoxicity studies, with a specific focus on pesticide exposure.

Table 2: The application of in vitro iPSC-derived models for assessing pesticide-induced neurotoxicity.
iPSC model Pesticides Time of treatment Concentration Findings References
hiPSC-derived
neurons and astrocytes
Chlorpyrifos 14 days 37.1 µM, 21 µM, 0.37 µM Alterations in synapse number, neurite outgrowth, BDNF levels, and neuron/astrocyte ratios. [1]
hiPSC-NSCs DDE, HCB, α-chlordane, oxychlordane, α-HCH, β-HCH, γ-HCH, Dieldrin 3, 14 and 28 days concentrations comparable to Scandinavian human blood levels Increased NSC proliferation, and reduced synapse numbers. [2]
hiPSC-derived NSCs, neurons, and astrocytes Aldicarb, captan, carbaryl, chlorpyrifos, deltamethrin, DDT, Dieldrin, Heptachlor, lindane, permethrin, rotenone, tebuconazole 2 days Ranges from 0 µM to 1000 µM Specific inhibition or upregulation was observed in key neural markers like PAX6, OTX2, and MAP2. [3]
hiPSC-derived excitatory and inhibitory neurons 28 pesticides including rotenone and dimethoate 24h 0-20 µM Disruptions in neural network formation and function. [4]
Lineage-specific luciferase hiPSC lines Deltamethrin, dieldrin, carbaryl, and rotenone 24h 10 and 100 µM Cytotoxicity across iPSCs, NSCs, neurons, and astrocytes. [5]
hiPSC-derived neurons and astrocytes. Chlorpyrifos 3 days and 14 days. 0.49 to 500 μM. Decrease of synaptophysin levels, BDNF increased, and reduced neurite outgrowth. [6]
hiPSC-derived neurons and astrocytes. Vinclozolin 3 days and 14 days 1 to 1340 μM Increase of BDNF [7]
3D hiPSC-derived multicellular brain spheroid. Rotenone 24 h and 48 h 0 to 50 μM Induction of dopaminergic toxicity at low concentrations, affecting astrocytes at high doses; dysregulated of neural gene expression [8]
3D autism spectrum disorder (ASD) iPSC-derived brain organoid. Chlorpyrifos (CPF) 24h at 4 weeks 100µM CPF disrupted ASD metabolic biomarkers, neurotransmitters (glutamate/GABA), and dopamine level reduction. [9]
3D hiPSC-derived neurospheres Paraquat, rotenone 72h Paraquat (1µM-1000µM)
Rotenone (0.001 µM-1000µM)
Concentration-dependent cell death; neurite outgrowth and connectivity reduction. [10]

2D hiPSC-derived models and neurotoxicity assessment: The increasing prevalence of neuro-developmental disorders such as autism spectrum (ASD) disorders, Attention-deficit/hyperactivity disorder (ADHD), and neurodegenerative diseases like Parkinson’s disease has prompted interest in evaluating the neurotoxic potential of environmental pollutants, particularly pesticides [85-94].

To date, some neurotoxicity studies have been published to test the applicability of iPSC-derived models as novel models to assess chemical toxicity in vitro. In a pivotal study, Pei, et al. evaluated the neurotoxicity of an 80-compound library, including neurotoxic pesticides such as chlorpyrifos, dieldrin, and aldicarb, on iPSC-derived models (NSCs, neurons, and astrocytes). This study underscores the model’s ability to discern neural-specific toxicity patterns and highlights the high sensitivity of iPSC-derived neurons to neurotoxicants [73].

Similarly, Li, et al. conducted an HT screen of 84 compounds, including pesticides like rotenone, deltamethrin, and tebuconazole, using GFP-labeled iPSC-derived cortical and motor neurons to enable live, real-time imaging of neurite outgrowth [95]. Their findings demonstrated that pesticides such as parathion exhibited selective inhibition of neurite extension in cortical neurons, while rotenone reduced neurite length and number across both cortical and motor neurons in a dose-dependent manner [95]. By integrating HT assays with quantitative imaging analysis, this study illustrates the capacity of iPSC-derived models to not only screen compounds rapidly but also generate mechanistic insights into pesticide-induced neurotoxicity.

Further advancing the application of iPSC models, Di Consiglio, et al. examined Chlorpyrifos (CPF), an organophosphate pesticide linked to developmental neurotoxicity. The CPF exposure effects at concentrations ranging from 18.45 to 37.10 μM (IC20) over a 14-day period were evaluated using iPSC-derived NSCs undergoing differentiation toward neurons and astrocytes. Immunocytochemistry and High-Content Imaging (HCI) revealed that CPF exposure reduced the expression of synaptophysin, a pre-synaptic marker, and BDNF protein levels, both of which are critical for synapse formation and function [96]. This model demonstrated CPF’s concentration-dependent neurotoxic effects, including reduced neurite outgrowth and synapse number, highlighting its utility for studying pesticide-induced neurotoxicity. This aligns with findings from several studies [38,72,97].

Mitochondrial activity and ATP levels are key indicators of cell viability, widely used in assessing neurotoxicity in iPSCs and NPCs. Kamata, et al. employed MTS 3-(4,5-Dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium and ATP assays to screen pesticides and other developmental neurotoxicants across varying concentrations, identifying agents like captan, carbaryl, chlorpyrifos, dieldrin, deltamethrin, lindane, and rotenone as significant DNT-positives based on IC50 values [98]. The study further demonstrated that several pesticides interfere with neural differentiation markers (e.g., PAX6, OTX2, MAP2), a crucial pathway in neurodevelopment. CPF notably inhibited PAX6, emphasizing its potential to disrupt neuronal differentiation.

Moreover, assessing the effects of pesticide mixtures is critical, as environmental exposures often involve complex chemical cocktails. iPSC-derived neuronal/glial co-cultures and mathematical modeling have illuminated how chemicals with similar modes of action, such as those impacting BDNF levels, synergistically enhance neurotoxicity at human-relevant doses. Pistollato, et al. found that mixtures of BDNF-disrupting pesticides led to significant downregulation of synaptogenesis markers, particularly PSD95, suggesting synergistic impairment in neural connectivity [99]. Additionally, Davidsen, et al. highlighted the importance of synaptogenesis as a sensitive endpoint in the mixture exposures study, reinforcing iPSC-based models’ utility in capturing the intricate impacts of pesticide interactions on neurodevelopment [100].

Moreover, Park, et al. employed a high-throughput ATP assay in iPSC-derived cortical neurons to investigate pesticide-induced changes in mitochondrial function, a critical indicator of neurotoxicity [101]. This study found that pesticides, including chlorpyrifos and deltamethrin, significantly disrupted mitochondrial activity, underscoring the relevance of metabolic assays in detecting neurotoxicity at various stages of neural differentiation.

To further expand the application of 2D hiPSC-derived models in neurotoxicity assessment, recent studies have demonstrated the efficacy of these systems in evaluating pesticide-induced neural toxicity. Ishibashi, et al. developed a robust in vitro platform using hiPSC-derived dopaminergic neurons to explore the neurotoxic effects of environmental pesticides. Their finding indicated selective vulnerability of dopaminergic neurons, relevant to Parkinson’s disease [102]. This research highlights the model’s sensitivity to specific pesticides, providing a basis for understanding dopaminergic neurodegeneration pathways triggered by pesticide exposure. Bartmann, et al. advanced this field by creating a human Neural Network Formation (hNNF) assay using co-cultured hiPSC-derived excitatory and inhibitory neurons with primary human astroglia [103]. This model was screened with a library of 28 chemicals, including various pesticides, to examine neurotoxicity on network parameters such as spike, burst, and synchronization characteristics. The results demonstrated the assay’s utility in identifying neurotoxic compounds, establishing it as a valuable tool for environmental toxicology studies.

In parallel, Paul, et al. integrated epidemiological insights with a toxicity screen using hiPSC-derived dopaminergic neurons from Parkinson’s disease patients [104]. Their study identified ten pesticides with significant neurotoxic effects, particularly on neurons implicated in Parkinson’s disease pathology, offering mechanistic insights into how environmental factors might contribute to neurodegenerative processes.

Collectively, these studies validate the use of iPSC-derived 2D neural models for neurotoxicity screening, particularly in the evaluation of pesticide-induced toxicity. With integrated approaches such as High-Content imaging (HC), lineage-specific markers, and real-time monitoring, these models provide a comprehensive platform to assess the neurodevelopmental impact of pesticides and other environmental toxicants, contributing significantly to developmental neurotoxicity research and regulatory applications.

3D iPSC-derived organoids and neurotoxicity assessment: Brain-organoid technologies provide an advanced platform for studying human neural development, functionality, and associated disorders in vitro. Typically, neurotoxic substances are introduced into iPSC-derived brain organoids at defined time points, representing acute or prolonged in utero exposures. This approach allows researchers to observe key morphological, transcriptional, and functional changes, offering detailed insights into the impact of neurotoxic exposures during critical developmental windows [54,105,106]

While the Embryonic Stem cell Test (EST) remains a standard for assessing embryotoxicity [19], brain organoids derived from iPSCs enable a similar evaluation of neurotoxicants by simulating specific developmental stages and exposure scenarios.

Although no standardized DNT tests currently exist for iPSC-derived organoids, these models have already shown considerable potential in environmental neurotoxicant testing [82,107]. For instance, Pamies, et al. investigated the developmental toxicity of rotenone, a pesticide that inhibits mitochondrial Complex I, by exposing brain spheroids derived from iPSCs at varying stages of maturation [108]. The study found that early-stage BrainSpheres (2 weeks of differentiation) were significantly more vulnerable to rotenone-induced toxicity, showing increased Reactive Oxygen Species (ROS) levels and mitochondrial dysfunction. At low concentrations (1 μM), rotenone selectively affected dopaminergic neurons, while higher concentrations (25 μM) impacted astrocytes. Transcriptomic analyses highlighted activation of the p53 signaling pathway in early differentiation, with disruptions in synapse and calcium signaling pathways later stages.

Similarly, several studies have begun to explore how environmental neurotoxicants interact with genetic susceptibilities in neurodevelopmental disorders. CPF, an organophosphate pesticide associated with ASD risk, was investigated in an iPSC-derived brain organoid model with a CHD8 heterozygous knockout, a mutation implicated in ASD [109]. In this study, it was presented that CPF exposure altered glutamate/GABA balance and reduced dopamine levels pointing to altered biomarkers linked with ASD and highlighting the complex gene-environment interactions that can be studied using iPSC-derived organoids.

In 2023, Lam, et al. integrated a 3D hydrogel tissue model with iPSC-derived neurons and primary astrocytes to evaluate neural activity changes in response to pesticide exposure such as dieldrin and CPF, facilitating detailed neural network activity analyses [110]. Additionally, a study by Mariani, et al. further demonstrates the application of 3D iPSC-derived organoids in pesticide neurotoxicity studies. Human iPSC-derived organoids were used to evaluate the neurotoxic effects of neonicotinoid pesticides, revealing disruptions in neural differentiation and synaptic formation. These findings demonstrated the potential of organoid models to capture specific impacts of pesticides on neurodevelopment, particularly in pathways associated with synaptogenesis and cellular maturation [111].

Despite their promising applications, 3D organoid models still face challenges, particularly regarding scalability for HTS. The structural complexity of these models makes their adaptation to HTS formats challenging. Kobolak, et al. addressed this by developing a scalable 3D neurosphere model in a 96-well plate format that includes neurons, astrocytes, and oligodendrocytes [112]. Their model was successfully evaluated to evaluate the cytotoxicity of pesticides, specifically paraquat and rotenone, demonstrating concentration-dependent cell death with IC50 values determined for each pesticide [112]. This approach provides a scalable and efficient framework for HT 3D neurotoxicity screening, showcasing the potential of iPSC-derived organoids in assessing pesticide-induced neurotoxicity.

Enhancing functionality for neurotoxicity screening, Sirenko, et al. developed a 3D neural platform with mature cortical neurons and astrocytes optimized for measuring calcium oscillations—a sensitive biomarker for neuronal activity and toxicity [113]. This screening assay facilitated the quantitative assessment of chemical effects on Ca2+ dynamics across a library of 84 compounds, including various pesticides [113].

Altogether, 3D iPSC-derived organoid models offer a compelling alternative to animal models in neurotoxicity research, capturing human-relevant molecular, cellular, and functional responses to neurotoxicants. They enable in-depth mechanistic studies of chemical interactions, including those modulated by genetic factors, positioning them as suitable tools for advancing personalized approaches in neurotoxicity and environmental health research.

iPSC-microfluidic brain organoids and neurotoxicity assessment: Microfluidic-based brain-on-chip systems represent a significant advancement in replicating the neurovascular environment and simulating the dynamic complexity of the human brain, improving the predictive value of in vitro neurotoxicity models. By incorporating 3D cultures, microfluidic channels, and endothelial cell-derived vascular compartments to replicate the BBB, these platforms offer enhanced capabilities for assessing how neurotoxicants, particularly pesticides, affect the brain at a mechanistic level [114,115]. This model’s design, embedding brain tissue analogs such as astrocytes, pericytes, and microglia in an extracellular matrix, provides a more physiologically relevant environment for studying neurotoxicity.

Koo, et al. demonstrated the utility of a microfluidic platform by examining how organophosphate pesticides, including Diethyl Methylphosphonate (DEMP) and Dimethyl Methylphosphonate (DMMP), interact with BBB integrity and acetylcholinesterase (AChE) activity. Pesticides were tested over a 24-hour period, revealing that high concentrations of DEMP and DMMP (>10−3 M) compromised cell viability and disrupted AChE activity [116]. Residual pesticide levels in the brain compartment indicated BBB permeability, confirming this system’s capacity to model neurovascular dynamics accurately. This model also tested pesticides such as CPF, malathion, and parathion, demonstrating their capacity to penetrate the BBB, inhibit AChE, and induce cell toxicity within the brain tissue construct [117].

Recently, Amend, et al. evaluated the use of Polydimethylsiloxane (PDMS)-based organ-chip platforms for toxicokinetic assessment, particularly using the organophosphate pesticide parathion and nerve agent VX as case studies [118]. Neuronal and liver spheroids were used to monitor concentration changes in the system, revealing that high absorption of these compounds by PDMS material significantly affected bioavailability. This study emphasizes the importance of selecting appropriate materials or coatings to prevent compound loss, ensuring accurate toxicokinetic studies for organophosphates on organ-chip platforms [118].

These microfluidic brain-on-chip platforms exhibit promising correlations with in vivo toxicity data, capturing critical endpoints such as BBB penetration, AChE inhibition, and cell viability. They present a valuable tool for high-throughput neurotoxicity screening, especially in evaluating complex physiological responses to pesticides, potentially advancing personalized approaches to risk assessment in neurotoxicity research.

Opportunities and future aspects of the iPSC-based model for neurotoxicity assessment

The widespread use of chemical compounds in daily life necessitates rigorous DNT testing. Current in vitro and animal tests often face challenges in translating results to human applications due to ethical and practical limitations [119]. Chemical compounds are widely used in human life, while current DNT testing results for risk assessment from in vitro and animal tests are usually difficult to validate in humans because of a lack of adequate testing opportunities that circumvent practical and ethical constraints [119]. Consequently, advancements in hiPSCs-derived brain models have gained attention in scientific research and are used across various disciplines. Recent studies have experimentally demonstrated that hiPSCs can differentiate into various cell types, making them a valuable tool for identifying toxic substances. The effectiveness of hiPSCs in predicting neural toxicity has been confirmed by testing their response to known neural toxins like retinoic acid [14,37,95,98,99,120-128]. Additionally, several studies have shown the feasibility of using brain organoids for evaluating the potential neurotoxic effects of environmental chemicals [72,129-134]. The introduction of iPSC-derived microfluidic chip brain technology shows unprecedented advantages in neurotoxicity testing, which highly simulates the complex physiological environment of human brain cells [69,117,135-139]. These brain chips hold potential as a new chemical hazard assessment tool, allowing the development of a set of biomarkers that can be effectively detected to reliably screen hazardous chemicals and monitor brain toxicity induced by contaminants.

One of the major limitations of most current human in vitro models is the absence of vascular and glial components. It is crucial for DNT to incorporate microglia cells into in vitro models as they play an important role in the developing brain and are key for neuroinflammation, a crucial tissue response to environmental stress. In addition, astrocytes are critically involved in various central nervous system disorders, both as protectors against brain damage and as instigators of disease initiation and progression [140]. BBB is another essential component to include in neurotoxicity and DNT studies [60]. To date, advanced strategies to produce vascularized and glial in vitro models are actively being explored [69,127,132,133,135,141-147].

The prevailing view today is that a combination of epigenetic factors, environmental influences, and the interplay between genes and the environment contributes to the elevated risk of neurodevelopmental disorders such as autism. For example, Modafferi, et al. have recently addressed the complex interplay between genetic and environmental factors in ASD by using CRISPR/Cas9-engineered CHD8 heterozygous knockout and iPSCs brain organoids [109]. The special-temporal signalling may be possible by implementing microfluidic systems, growth factor gradients, and biomaterials such as ECM and BBB [148]. The combination of iPSC-in vitro models and omics data enables the building of a toxicity testing system for screening neural toxicants on a large scale before their widespread application to avoid the potential for toxicity and neurodevelopmental toxicity [113]. Furthermore, by exploring the potential of iPSC-derived models, we can establish a robust and high-throughput platform for screening environmental toxicants. By synergistically integrating functional evaluation, multi-omics data, and machine learning algorithms as well as morphological analysis, this novel platform could yield comprehensive and reliable screening capabilities for identifying environmental toxicants with unprecedented efficiency and accuracy. The incorporation of Next-Generation Sequencing (NGS) in iPSC disease modelling fulfils the need for genome sequencing with high cost-effectiveness and establishes a real-time correlation between transcriptomic profiling and phenotypic activities. It allows comparison between the sequencing outputs and the in vitro cytopathies, providing deeper insights into toxicological mechanisms [149]. HCS application approaches combining automated microscopy, quantitative image analysis, and iPSC-derived models are widely used for studying the mechanisms of compound toxicity, including developmental neurotoxicity and genotoxicity [150]. Li, et al. recently developed an HCS-based neurite outgrowth assay using iPSC-derived neurons expressing GFP. This innovation facilitates detailed live and time-lapse imaging of neurite outgrowth, enhancing the precision and depth of neurotoxicity assessments [95].

High-throughput single-cell RNA sequencing (scRNA-seq) characterizes the sequential changes of the transcriptome in individual cells. Therefore, in neurotoxicity research, scRNA seq can be employed to identify possible alterations in the developmental trajectories of neural cells at each time point of the differentiation process [151]. Multi-organoid-chip (MoC) platforms are equipped with combinations of multiple organoids, including the brain. This system can establish a complex process of chemical metabolism and responses through organs-organs interactions and, therefore, can be utilized to evaluate systemic toxicity from the dynamic process of distribution, absorption, metabolism, and excretion features of toxins in microfluidic systems [152].

Additionally, improving iPSC models is urgently needed to achieve improved reproducibility and standardization, reduced costs, increased throughput, and assay optimization. Further improvements are expected for more physiologically relevant models incorporating cellular components such as immune, glial, vascular, and genetically engineered cells. Furthermore, it would also be interesting to include iPSC-derived patient models to evaluate gene-environmental interactions (e.g., idiopathic autism) broadly. Last, the use of Artificial Intelligence (AI) emerges as a new promise to revolutionize toxicity. The DeepTox algorithm is an example that uses machine learning and the history of predefined toxicophore features to predict 12,000 environmental chemicals and drugs with high accuracy [153]. Furthermore, the advent of digital image analysis coupled with deep learning AI model provides an innovative approach to making quantification of neurotoxicity data automated and time-consuming [154].

These various models result from new technologies of exploration; although they are relevant, they do not replace the evaluations of the effects of early in utero exposures, which capture the critical processes of maternal-fetal chemical transfer, including the role of the placental barrier and maternal physiology [155]. In addition, in vivo observations are essential for tracking the emergence of sensory, behavioral, and cognitive functions at specific developmental stages, enabling the early detection of potential neurodevelopmental anomalies [156]. While in vitro models offer high-throughput and ethically favorable testing options that support the principles of the 3Rs (Refine, Reduce, Replace), real-world exposures are complex, often involving multiple agents that interact in ways difficult to isolate in a controlled environment. Therefore, an integrated approach—leveraging both in vitro technologies for molecular insights and in vivo models for comprehensive, systemic effects—holds the greatest promise for accurate neurotoxicity assessment [81]. Such a combined strategy enhances the predictive value of neurotoxicity evaluations, providing a precise and realistic understanding of how cumulative environmental exposures affect human neural development.

iPSC-derived brain models have profoundly revolutionized neurotoxicology, advancing the fields of basic research, chemical risk assessment, and translational medicine. By closely replicating human neural physiology, these models serve as a crucial bridge between fundamental research and clinical application, enabling more accurate evaluations of neurotoxicity risks associated with environmental substances and pharmaceutical compounds. Their relevance extends beyond general toxicology, providing a pathway to personalized risk assessment by evaluating individual susceptibility to neurotoxic agents. This capability facilitates precision interventions tailored to specific genetic and environmental profiles, thus advancing the field of personalized medicine.

Moreover, iPSC-derived models enable the dissection of neurotoxic mechanisms at the molecular level, underlying neurotoxic effects, which is essential for the development of targeted therapeutic interventions. Their application in drug development is equally transformative, with the potential to improve early-stage neurotoxicity detection and reduce adverse effects, thus enhancing the safety profile of candidate drugs prior to clinical trials. The advent of high-throughput, automated screening platforms based on iPSC-derived models further strengthens their utility, providing robust, scalable, and ethically sound options for screening a wide array of environmental and food-related toxicants.

The integration of these human-relevant models with in vivo approaches and advanced in silico tools provides a comprehensive framework for neurotoxicity research and risk assessment. Leveraging the predictive capabilities of these models in regulatory toxicology will be crucial for advancing human health protection and supporting personalized therapeutic strategies. This paradigm shift not only promises to safeguard public health from environmental hazards but also accelerates the transition toward individualized therapeutic treatments. By refining risk-assessment methodologies, iPSC-derived brain models stand at the forefront of innovation in toxicology and personalized medicine, underscoring their potential to revolutionize both fields and drive meaningful advances in human health protection and therapeutic development.

Author’s contribution

Narimane Kebieche: Conceptualization, Methodology, Writing. Farzana Liakath Ali and SeungAe Yim: Reviewing and Editing. Mohamed Ali: Reviewing. Rachid Soulimani. Writing and Supervision. Claude Lambert: Supervision.

Acknowledgment

This research was supported by the University of Lorraine, France.

  1. Grandjean P, Landrigan P. Developmental neurotoxicity of industrial chemicals. Lancet. 2006;368:2167–2178. Available from: https://doi.org/10.1016/s0140-6736(06)69665-7
  2. Grandjean P, Landrigan PJ. Neurobehavioural effects of developmental toxicity. Lancet Neurol. 2014;13:330–338. Available from: https://doi.org/10.1016/s1474-4422(13)70278-3
  3. Heyer DB, Meredith RM. Environmental toxicology: Sensitive periods of development and neurodevelopmental disorders. NeuroToxicol. 2017;58:23–41. Available from: https://doi.org/10.1016/j.neuro.2016.10.017
  4. Landrigan PJ, Goldman LR. Children’s vulnerability to toxic chemicals: A challenge and opportunity to strengthen health and environmental policy. Health Aff (Millwood). 2011;30:842–850. Available from: https://doi.org/10.1377/hlthaff.2011.0151
  5. Kroon T, Sierksma MC, Meredith RM. Investigating mechanisms underlying neurodevelopmental phenotypes of autistic and intellectual disability disorders: A perspective. Front Syst Neurosci. 2013;7:75. Available from: https://doi.org/10.3389/fnsys.2013.00075
  6. Harris A, Seckl J. Glucocorticoids, prenatal stress and the programming of disease. Horm Behav. 2011;59:279–289. Available from: https://doi.org/10.1016/j.yhbeh.2010.06.007
  7. Raciti M, Ceccatelli S. Epigenetic mechanisms in developmental neurotoxicity. Neurotoxicol Teratol. 2018;66:94–101. Available from: https://doi.org/10.1016/j.ntt.2017.12.002
  8. Dagnino S. Unravelling the exposome: Conclusions and thoughts for the future. In: Dagnino S, Macherone A, editors. Unraveling the Exposome. Cham: Springer International Publishing. 2018; 425–437.
  9. Wild CP. Complementing the genome with an ‘exposome’: The outstanding challenge of environmental exposure measurement in molecular epidemiology. Cancer Epidemiol Biomarkers Prev. 2005;14:1847–1850. Available from: https://doi.org/10.1158/1055-9965.epi-05-0456
  10. Feil R, Fraga MF. Epigenetics and the environment: Emerging patterns and implications. Nat Rev Genet. 2012;13:97–109. Available from: https://doi.org/10.1038/nrg3142
  11. Hartung T. On mapping the human toxome. ALTEX. 2011;28:83–93. Available from: https://doi.org/10.14573/altex.2011.2.083
  12. Hartung T. Toxicology for the twenty-first century. Nature. 2009;460:208–212. Available from: https://doi.org/10.1038/460208a
  13. Paparella M, Bennekou SH, Bal-Price A. An analysis of the limitations and uncertainties of in vivo developmental neurotoxicity testing and assessment to identify the potential for alternative approaches. Reprod Toxicol. 2020;96:327–336. Available from: https://doi.org/10.1016/j.reprotox.2020.08.002
  14. Hofrichter M, Nimtz L, Tigges J, Kabiri Y, Schröter F, Royer-Pokora B, et al. Comparative performance analysis of human iPSC-derived and primary neural progenitor cells (NPC) grown as neurospheres in vitro. Stem Cell Res. 2017;25:72–82. Available from: https://doi.org/10.1016/j.scr.2017.10.013
  15. Pamies D. Guidance document on Good Cell and Tissue Culture Practice 2.0 (GCCP 2.0). ALTEX. Epub ahead of print 2021. Available from: https://doi.org/10.14573/altex.2111011
  16. Liu W, Deng Y, Liu Y, Gong W, Deng W. Stem Cell Models for Drug Discovery and Toxicology Studies: Stem Cell Models For Drug Discovery And Toxicology. J Biochem Mol Toxicol. 2013;27:17–27. Available from: https://doi.org/10.1002/jbt.21470
  17. Biehl JK, Russell B. Introduction to Stem Cell Therapy. J Cardiovasc Nurs. 2009;24:98–103. Available from: https://doi.org/10.1097/jcn.0b013e318197a6a5
  18. Dulak J, Szade K, Szade A, Nowak W, Józkowicz A. Adult stem cells: hopes and hypes of regenerative medicine. Acta Biochim Pol. 2015;62:329–337. Available from: https://doi.org/10.18388/abp.2015_1023
  19. Seiler AEM, Spielmann H. The validated embryonic stem cell test to predict embryotoxicity in vitro. Nat Protoc. 2011;6:961–978. Available from: https://doi.org/10.1038/nprot.2011.348
  20. Jaramillo-Ferrada PA, Wolvetang EJ, Cooper-White JJ. Differential mesengenic potential and expression of stem cell-fate modulators in mesenchymal stromal cells from human-term placenta and bone marrow. J Cell Physiol. 2012;227:3234–3242. Available from: https://doi.org/10.1002/jcp.24014
  21. Visvader JE, Clevers H. Tissue-specific designs of stem cell hierarchies. Nat Cell Biol. 2016;18:349–355. Available from: https://doi.org/10.1038/ncb3332
  22. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–676. Available from: https://doi.org/10.1016/j.cell.2006.07.024
  23. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917–1920. Available from: https://doi.org/10.1126/science.1151526
  24. Shtrichman R, Germanguz I, Eldor JI. Induced pluripotent stem cells (iPSCs) derived from different cell sources and their potential for regenerative and personalized medicine. CMM. 2013;13:792–805. Available from: https://doi.org/10.2174/1566524011313050010
  25. Perrier AL, Tabar V, Barberi T, Rubio ME, Bruses J, Topf N, et al. Derivation of midbrain dopamine neurons from human embryonic stem cells. Proc Natl Acad Sci USA. 2004;101:12543–12548. Available from: https://doi.org/10.1073/pnas.0404700101
  26. Stummann T, Bremer S. The possible impact of human embryonic stem cells on safety pharmacological and toxicological assessments in drug discovery and drug development. CSCR. 2008;3:117–130. Available from: https://doi.org/10.2174/157488808784223104
  27. Hartung T. Thoughts on limitations of animal models. Parkinsonism Relat Disord. 2008;14–S83. Available from: https://doi.org/10.1016/j.parkreldis.2008.04.003
  28. Zhao Z, Zhang D, Yang F, Xu M, Zhao S, Pan T, et al. Evolutionarily conservative and non-conservative regulatory networks during primate interneuron development revealed by single-cell RNA and ATAC sequencing. Cell Res. 2022;32:425–436. Available from: https://doi.org/10.1038/s41422-022-00635-9
  29. Kumar KK, Aboud AA, Bowman AB. The potential of induced pluripotent stem cells as a translational model for neurotoxicological risk. NeuroToxicol. 2012;33:518–529. Available from: https://doi.org/10.1016/j.neuro.2012.02.005
  30. Hu B-Y, Weick JP, Yu J, Ma LX, Zhang XQ, Thomson JA, et al. Neural differentiation of human induced pluripotent stem cells follows developmental principles but with variable potency. Proc Natl Acad Sci USA. 2010;107:4335–4340. Available from: https://doi.org/10.1073/pnas.0910012107
  31. Liu Q, Pedersen OZ, Peng J, Couture LA, Rao MS, Zeng X. Optimizing dopaminergic differentiation of pluripotent stem cells for the manufacture of dopaminergic neurons for transplantation. Cytotherapy. 2013;15:999–1010. Available from: https://doi.org/10.1016/j.jcyt.2013.03.006
  32. Wang S, Bates J, Li X, Schanz S, Chandler-Militello D, Levine C, et al. Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell. 2013;12:252–264. Available from: https://doi.org/10.1016/j.stem.2012.12.002
  33. Odawara A, Katoh H, Matsuda N, Suzuki I. Physiological maturation and drug responses of human induced pluripotent stem cell-derived cortical neuronal networks in long-term culture. Sci Rep. 2016;6:26181. Available from: https://doi.org/10.1038/srep26181
  34. Odawara A, Matsuda N, Ishibashi Y, Yokoi R, Suzuki I. Toxicological evaluation of convulsant and anticonvulsant drugs in human induced pluripotent stem cell-derived cortical neuronal networks using an MEA system. Sci Rep. 2018;8:10416. Available from: https://doi.org/10.1038/s41598-018-28835-7
  35. Tukker AM, de Groot MW, Wijnolts FM, Kasteel EE, Hondebrink L, Westerink RH. Is the time right for in vitro neurotoxicity testing using human iPSC-derived neurons? ALTEX. 2016;33(3):261–271. Available from: https://doi.org/10.14573/altex.1510091
  36. Little D, Ketteler R, Gissen P, Devine MJ. Using stem cell–derived neurons in drug screening for neurological diseases. Neurobiol Aging. 2019;78:130–141. Available from: https://doi.org/10.1016/j.neurobiolaging.2019.02.008
  37. Tukker AM, Wijnolts FMJ, de Groot A, Westerink RHS. Human iPSC-derived neuronal models for in vitro neurotoxicity assessment. NeuroToxicol. 2018;67:215–225. Available from: https://doi.org/10.1016/j.neuro.2018.06.007
  38. Tukker AM, Wijnolts FMJ, de Groot A, Westerink RHS. in vitro techniques for assessing neurotoxicity using human iPSC-derived neuronal models. In: Aschner M, Costa L, editors. Cell Culture Techniques. New York, NY: Springer New York; 2019; 17–35. Available from: https://dspace.library.uu.nl/handle/1874/392153
  39. Zhang P, Xia N, Reijo Pera RA. Directed dopaminergic neuron differentiation from human pluripotent stem cells. JoVE. 2014;51737. Available from: https://doi.org/10.3791/51737
  40. Liu Y, Liu H, Sauvey C, Yao L, Zarnowska ED, Zhang SC. Directed differentiation of forebrain GABA interneurons from human pluripotent stem cells. Nat Protoc. 2013;8:1670–1679. Available from: https://doi.org/10.1038/nprot.2013.106
  41. Sun AX, Yuan Q, Tan S, Xiao Y, Wang D, Khoo AT, et al. Direct induction and functional maturation of forebrain GABAergic neurons from human pluripotent stem cells. Cell Rep. 2016;16:1942–1953. Available from: https://doi.org/10.1016/j.celrep.2016.07.035
  42. Corti S, Nizzardo M, Simone C, Falcone M, Nardini M, Ronchi D, et al. Genetic correction of human induced pluripotent stem cells from patients with spinal muscular atrophy. Sci Transl Med. 2012;4. Available from: https://doi.org/10.1126/scitranslmed.3004108
  43. Sareen D, O’Rourke JG, Meera P, Muhammad AK, Grant S, Simpkinson M, et al. Targeting RNA foci in iPSC-derived motor neurons from ALS patients with a C9ORF72 repeat expansion. Sci Transl Med. 2013;5. Available from: https://doi.org/10.1126/scitranslmed.3007529
  44. Wen Z, Nguyen HN, Guo Z, Lalli MA, Wang X, Su Y, et al. Synaptic dysregulation in a human iPS cell model of mental disorders. Nature. 2014;515:414–418. Available from: https://doi.org/10.1038/nature13716
  45. Gunhanlar N, Shpak G, van der Kroeg M, Gouty-Colomer LA, Munshi ST, Lendemeijer B, et al. A simplified protocol for differentiation of electrophysiologically mature neuronal networks from human induced pluripotent stem cells. Mol Psychiatry. 2018;23:1336–1344. Available from: https://doi.org/10.1038/mp.2017.56
  46. Clevers H. Modeling development and disease with organoids. Cell. 2016;165:1586–1597. Available from: https://doi.org/10.1016/j.cell.2016.05.082
  47. Di Lullo E, Kriegstein AR. The use of brain organoids to investigate neural development and disease. Nat Rev Neurosci. 2017;18:573–584. Available from: https://doi.org/10.1038/nrn.2017.107
  48. Chandrasekaran A, Avci HX, Ochalek A, Rösingh LN, Molnár K, László L, et al. Comparison of 2D and 3D neural induction methods for the generation of neural progenitor cells from human induced pluripotent stem cells. Stem Cell Res. 2017;25:139–151. Available from: https://doi.org/10.1016/j.scr.2017.10.010
  49. Lancaster MA, Knoblich JA. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science. 2014;345:1247125. Available from: https://doi.org/10.1126/science.1247125
  50. Elshazzly M, Lopez MJ, Reddy V, Caban O. Embryology, central nervous system. In: StatPearls. Treasure Island (FL): StatPearls Publishing; 2023. (accessed 17 June 2022). Available from: http://www.ncbi.nlm.nih.gov/books/NBK526024/
  51. Lancaster MA, Corsini NS, Wolfinger S, Gustafson EH, Phillips AW, Burkard TR, et al. Guided self-organization and cortical plate formation in human brain organoids. Nat Biotechnol. 2017;35:659–666. Available from: https://doi.org/10.1038/nbt.3906
  52. Qian X, Nguyen HN, Song MM, Hadiono C, Ogden SC, Hammack C, et al. Brain-region-specific organoids using mini-bioreactors for modeling ZIKV exposure. Cell. 2016;165:1238–1254. Available from: https://doi.org/10.1016/j.cell.2016.04.032
  53. Jo J, Xiao Y, Sun AX, Cukuroglu E, Tran HD, Göke J, et al. Midbrain-like organoids from human pluripotent stem cells contain functional dopaminergic and neuromelanin-producing neurons. Cell Stem Cell. 2016;19:248–257. Available from: https://doi.org/10.1016/j.stem.2016.07.005
  54. Fan P, Wang Y, Xu M, Han X, Liu Y. The application of brain organoids in assessing neural toxicity. Front Mol Neurosci. 2022;15:799397. Available from: https://doi.org/10.3389/fnmol.2022.799397
  55. Muguruma K, Nishiyama A, Kawakami H, Hashimoto K, Sasai Y. Self-organization of polarized cerebellar tissue in 3D culture of human pluripotent stem cells. Cell Rep. 2015;10:537–550. Available from: https://doi.org/10.1016/j.celrep.2014.12.051
  56. Birey F, Andersen J, Makinson CD, Islam S, Wei W, Huber N, et al. Assembly of functionally integrated human forebrain spheroids. Nature. 2017;545:54–59. Available from: https://doi.org/10.1038/nature22330
  57. Mansour AA, Gonçalves JT, Bloyd CW, Li H, Fernandes S, Quang D, et al. An in vivo model of functional and vascularized human brain organoids. Nat Biotechnol. 2018;36:432–441. Available from: https://doi.org/10.1038/nbt.4127
  58. Shi Y, Sun L, Wang M, Liu J, Zhong S, Li R, et al. Vascularized human cortical organoids (vOrganoids) model cortical development in vivo. PLoS Biol. 2020;18. Available from: https://doi.org/10.1371/journal.pbio.3000705
  59. Schultz L, Zurich M-G, Culot M, da Costa A, Landry C, Bellwon P, et al. Evaluation of drug-induced neurotoxicity based on metabolomics, proteomics and electrical activity measurements in complementary CNS in vitro models. Toxicol in vitro. 2015;30:138–165. Available from: https://doi.org/10.1016/j.tiv.2015.05.016
  60. Lippmann ES, Azarin SM, Kay JE, Nessler RA, Wilson HK, Al-Ahmad A, et al. Derivation of blood-brain barrier endothelial cells from human pluripotent stem cells. Nat Biotechnol. 2012;30:783–791. Available from: https://doi.org/10.1038/nbt.2247
  61. Wilson HK, Canfield SG, Hjortness MK, Palecek SP, Shusta EV. Exploring the effects of cell seeding density on the differentiation of human pluripotent stem cells to brain microvascular endothelial cells. Fluids Barriers CNS. 2015;12:13. Available from: https://doi.org/10.1186/s12987-015-0007-9
  62. Lippmann ES, Al-Ahmad A, Azarin SM, Palecek SP, Shusta EV. A retinoic acid-enhanced, multicellular human blood-brain barrier model derived from stem cell sources. Sci Rep. 2015;4:4160. Available from: https://doi.org/10.1038/srep04160
  63. Wang YI, Abaci HE, Shuler ML. Microfluidic blood–brain barrier model provides in vivo‐like barrier properties for drug permeability screening. Biotechnol Bioeng. 2017;114:184–194. Available from: https://doi.org/10.1002/bit.26045
  64. Vatine GD, Barrile R, Workman MJ, Sances S, Barriga BK, Rahnama M, et al. Human iPSC-derived blood-brain barrier chips enable disease modeling and personalized medicine applications. Cell Stem Cell. 2019;24(6):995-1005.e6. Available from: https://doi.org/10.1016/j.stem.2019.05.011
  65. Pham MT, Pollock KM, Rose MD, Cary WA, Stewart HR, Zhou P, et al. Generation of human vascularized brain organoids. NeuroReport. 2018;29:588–593. Available from: https://doi.org/10.1097/wnr.0000000000001014
  66. Cakir B, Xiang Y, Tanaka Y, Kural MH, Parent M, Kang YJ, et al. Engineering of human brain organoids with a functional vascular-like system. Nat Methods. 2019;16:1169–1175. Available from: https://doi.org/10.1038/s41592-019-0586-5
  67. Perestrelo A, Águas A, Rainer A, Forte G. Microfluidic Organ/Body-on-a-Chip devices at the convergence of biology and microengineering. Sensors. 2015;15:31142–31170. Available from: https://doi.org/10.3390/s151229848
  68. Zhuang Q-C, Ning R-Z, Ma Y, Lin J-M. Recent developments in microfluidic chip for in vitro cell-based research. Chin J Anal Chem. 2016;44:522–532. Available from: https://doi.org/10.1016/S1872-2040(16)60919-2
  69. Sances S, Ho R, Vatine G, West D, Laperle A, Meyer A, et al. Human iPSC-derived endothelial cells and microengineered organ-chip enhance neuronal development. Stem Cell Reports. 2018;10:1222–1236. Available from: https://doi.org/10.1016/j.stemcr.2018.02.012
  70. Liu L, Koo Y, Russell T, Yun Y. A three-dimensional brain-on-a-chip using human iPSC-derived GABAergic neurons and astrocytes. Methods Mol Biol. 2022;2492:117-128. Available from: https://doi.org/10.1007/978-1-0716-2289-6_6
  71. Faley SL, Neal EH, Wang JX, Bosworth AM, Weber CM, Balotin KM, et al. iPSC-derived brain endothelium exhibits stable, long-term barrier function in perfused hydrogel scaffolds. Stem Cell Reports. 2019;12:474–487. Available from: https://doi.org/10.1016/j.stemcr.2019.01.009
  72. Spijkers XM, Pasteuning-Vuhman S, Dorleijn JC, Vulto P, Wevers NR, Pasterkamp RJ. A directional 3D neurite outgrowth model for studying motor axon biology and disease. Sci Rep. 2021;11:2080. Available from: https://doi.org/10.1038/s41598-021-81335-z
  73. Pei Y, Peng J, Behl M, Sipes NS, Shockley KR, Rao MS, et al. Comparative neurotoxicity screening in human iPSC-derived neural stem cells, neurons and astrocytes. Brain Res. 2016;1638:57–73. Available from: https://doi.org/10.1016/j.brainres.2015.07.048
  74. Bose R, Spulber S, Ceccatelli S. The threat posed by environmental contaminants on neurodevelopment: What can we learn from neural stem cells? Int J Mol Sci. 2023;24:4338. Available from: https://doi.org/10.3390/ijms24054338
  75. Bal-Price AK, Coecke S, Costa L, Crofton KM, Fritsche E, Goldberg A, et al. Advancing the science of developmental neurotoxicity (DNT): Testing for better safety evaluation. ALTEX. 2012;29(2):202-215. Available from: https://doi.org/10.14573/altex.2012.2.202
  76. Bal-Price AK, Hogberg HT, Buzanska L, Lenas P, van Vliet E, Hartung T. in vitro developmental neurotoxicity (DNT) testing: Relevant models and endpoints. NeuroToxicology. 2010;31:545–554. Available from: https://doi.org/10.1016/j.neuro.2009.11.006
  77. Fritsche E, Grandjean P, Crofton KM, Aschner M, Goldberg A, Heinonen T, et al. Consensus statement on the need for innovation, transition and implementation of developmental neurotoxicity (DNT) testing for regulatory purposes. Toxicol Appl Pharmacol. 2018;354:3–6. Available from: https://doi.org/10.1016/j.taap.2018.02.004
  78. Fritsche E, Barenys M, Klose J, Masjosthusmann S, Nimtz L, Schmuck M, et al. Corrigendum to “Current availability of stem cell-based in vitro methods for developmental neurotoxicity (DNT) testing.” Toxicol Sci. 2018;165:531–531. Available from: https://doi.org/10.1093/toxsci/kfy195
  79. Fritsche E, Barenys M, Klose J, Masjosthusmann S, Nimtz L, Schmuck M, et al. Development of the concept for stem cell-based developmental neurotoxicity evaluation. Toxicol Sci. 2018;165:14–20. Available from: https://doi.org/10.1093/toxsci/kfy175
  80. Paparella M, Bennekou SH, Bal-Price A. An analysis of the limitations and uncertainties of in vivo developmental neurotoxicity testing and assessment to identify the potential for alternative approaches. Reprod Toxicol. 2020;96:327-336. Available from: https://doi.org/10.1016/j.reprotox.2020.08.002
  81. Bal-Price A, Hogberg HT, Crofton KM, Daneshian M, FitzGerald RE, Fritsche E. Recommendation on test readiness criteria for new approach methods in toxicology: Exemplified for developmental neurotoxicity. ALTEX 2018;306–352. Available from: https://doi.org/10.14573/altex.1712081
  82. Bal-Price A, Pistollato F, Sachana M, Bopp SK, Munn S, Worth A. Strategies to improve the regulatory assessment of developmental neurotoxicity (DNT) using in vitro methods. Toxicol Appl Pharmacol. 2018;354:7–18. Available from: https://doi.org/10.1016/j.taap.2018.02.008
  83. Hrstka SCL, Ankam S, Agac B, Klein JP, Moore RA, Narapureddy B, et al. Proteomic analysis of human iPSC-derived sensory neurons implicates cell stress and microtubule dynamics dysfunction in bortezomib-induced peripheral neurotoxicity. Experimental Neurology 2021;335:113520. Available from: https://doi.org/10.1016/j.expneurol.2020.113520
  84. Bal-Price A, Hogberg HT, Crofton KM, Daneshian M, FitzGerald RE, Fritsche E, et al. Recommendation on test readiness criteria for new approach methods in toxicology: Exemplified for developmental neurotoxicity. ALTEX 2018;35:306–352. Available from: https://doi.org/10.14573/altex.1712081
  85. Chen L, Yoo S-E, Na R, Liu Y, Ran Q. Cognitive impairment and increased Aβ levels induced by paraquat exposure are attenuated by enhanced removal of mitochondrial H2O2. Neurobiology of Aging 2012;33:432.e15-432.e26. Available from: https://doi.org/10.1016/j.neurobiolaging.2011.01.008
  86. De Felice A, Scattoni ML, Ricceri L, Calamandrei G. Prenatal exposure to a common organophosphate insecticide delays motor development in a mouse model of idiopathic autism. PLoS One. 2015;10(3). Available from: https://doi.org/10.1371/journal.pone.0121663
  87. Kardas F, Bayram AK, Demirci E, Akin L, Ozmen S, Kendirci M, et al. Increased serum phthalates (MEHP, DEHP) and bisphenol A concentrations in children with autism spectrum disorder: The role of endocrine disruptors in autism etiopathogenesis. J Child Neurol. 2016;31(7):629–635. Available from: https://doi.org/10.1177/0883073815609150
  88. Li G, Kim C, Kim J, Yoon H, Zhou H, Kim J. Common pesticide, dichlorodiphenyltrichloroethane (DDT), increases amyloid-β levels by impairing the function of ABCA1 and IDE: Implication for Alzheimer’s disease. J Alzheimers Dis. 2015;46(1):109–122. Available from: https://doi.org/10.3233/jad-150024
  89. Pearson BL, Simon JM, McCoy ES, Salazar G, Fragola G, Zylka MJ. Identification of chemicals that mimic transcriptional changes associated with autism, brain aging and neurodegeneration. Nat Commun. 2016;7:11173. Available from: https://doi.org/10.1038/ncomms11173
  90. Richardson JR, Taylor MM, Shalat SL, Guillot TS, Caudle WM, Hossain MM, et al. Developmental pesticide exposure reproduces features of attention deficit hyperactivity disorder. FASEB J. 2015;29(5):1960–1972. Available from: https://doi.org/10.1096/fj.14-260901
  91. Shelton JF, Geraghty EM, Tancredi DJ, Delwiche LD, Schmidt RJ, Ritz B, et al. Neurodevelopmental disorders and prenatal residential proximity to agricultural pesticides: The CHARGE study. Environ Health Perspect. 2014;122(10):1103–1109. Available from: https://doi.org/10.1289/ehp.1307044
  92. Tyler CR, Allan AM. The effects of arsenic exposure on neurological and cognitive dysfunction in human and rodent studies: A review. Curr Envir Health Rpt. 2014;1(2):132–147. Available from: https://doi.org/10.1007/s40572-014-0012-1
  93. von Ehrenstein OS, Ling C, Cui X, Cockburn M, Park AS, Yu F, Wu J, et al. Prenatal and infant exposure to ambient pesticides and autism spectrum disorder in children: Population-based case-control study. BMJ. 2019;365. Available from: https://doi.org/10.1136/bmj.l962
  94. Wang B, Du Y. Cadmium and its neurotoxic effects. Oxid Med Cell Longev. 2013;2013:1–12. Available from: https://doi.org/10.1155/2013/898040
  95. Li S, Zhang L, Huang R, Xu T, Parham F, Behl M, et al. Evaluation of chemical compounds that inhibit neurite outgrowth using GFP-labeled iPSC-derived human neurons. NeuroToxicol. 2021;83:137–145. Available from: https://doi.org/10.1016/j.neuro.2021.01.003
  96. Di Consiglio E, Pistollato F, Mendoza-De Gyves E, Bal-Price A, Testai E. Integrating biokinetics and in vitro studies to evaluate developmental neurotoxicity induced by chlorpyrifos in human iPSC-derived neural stem cells undergoing differentiation towards neuronal and glial cells. Reprod Toxicol. 2020;98:174–188. Available from: https://doi.org/10.1016/j.reprotox.2020.09.010
  97. Radio NM, Breier JM, Shafer TJ, Mundy WR. Assessment of chemical effects on neurite outgrowth in PC12 cells using high content screening. Toxicol Sci. 2008;105(1):106–118. Available from: https://doi.org/10.1093/toxsci/kfn114
  98. Kamata S, Hashiyama R, Hana-ika H, Ohkubo I, Saito R, et al. Cytotoxicity comparison of 35 developmental neurotoxicants in human induced pluripotent stem cells (iPSC), iPSC-derived neural progenitor cells, and transformed cell lines. Toxicol in vitro. 2020;69:104999. Available from: https://doi.org/10.1016/j.tiv.2020.104999
  99. Pistollato F, de Gyves EM, Carpi D, Bopp SK, Nunes C, Worth A, et al. Assessment of developmental neurotoxicity induced by chemical mixtures using an adverse outcome pathway concept. Environ Health. 2020;19:23. Available from: https://doi.org/10.1186/s12940-020-00578-x
  100. Davidsen N, Lauvås AJ, Myhre O, Ropstad E, Carpi D, Gyves EM, et al. Exposure to human relevant mixtures of halogenated persistent organic pollutants (POPs) alters neurodevelopmental processes in human neural stem cells undergoing differentiation. Reprod Toxicol. 2021;100:17–34. Available from: https://doi.org/10.1016/j.reprotox.2020.12.013
  101. Park SM, Jo NR, Lee B, Jung EM, Lee SD, Jeung EB, et al. Establishment of a developmental neurotoxicity test by Sox1-GFP mouse embryonic stem cells. Reprod Toxicol. 2021;104:96–105. Available from: https://doi.org/10.1016/j.reprotox.2021.07.004
  102. Ishibashi Y, Nagafuku N, Kanda Y, Suzuki I. Evaluation of neurotoxicity for pesticide-related compounds in human iPS cell-derived neurons using microelectrode array. Toxicol in vitro. 2023;93:105668. Available from: https://doi.org/10.1016/j.tiv.2023.105668
  103. Bartmann K, Bendt F, Dönmez A, Haag D, Keßel HE, Masjosthusmann S, et al. A human iPSC-based in vitro neural network formation assay to investigate neurodevelopmental toxicity of pesticides. ALTEX. 2023;40(3):452–470. Available from: https://doi.org/10.14573/altex.2206031
  104. Paul KC, Krolewski RC, Lucumi Moreno E, Blank J, Holton KM, Ahfeldt T, et al. A pesticide and iPSC dopaminergic neuron screen identifies and classifies Parkinson-relevant pesticides. Nat Commun. 2023;14(1):2803. Available from: https://doi.org/10.1038/s41467-023-38215-z
  105. Garcia SJ, Seidler FJ, Slotkin TA. Developmental neurotoxicity elicited by prenatal or postnatal chlorpyrifos exposure: Effects on neurospecific proteins indicate changing vulnerabilities. Environ Health Perspect. 2003;111(3):297–303. Available from: https://doi.org/10.1289/ehp.5791
  106. Tsuji R, Crofton KM. Developmental neurotoxicity guideline study: Issues with methodology, evaluation and regulation. Congenit Anomal. 2012;52(3):122–128. Available from: https://doi.org/10.1111/j.1741-4520.2012.00374.x
  107. Bal-Price A, Hogberg HT, Crofton KM, Daneshian M, FitzGerald RE, Fritsche E, et al. Recommendation on test readiness criteria for new approach methods in toxicology: Exemplified for developmental neurotoxicity. ALTEX. 2018;35(3):306–352. Available from: https://doi.org/10.14573/altex.1712081
  108. Pamies D, Block K, Lau P, Gribaldo L, Pardo CA, Barreras P, et al. Rotenone exerts developmental neurotoxicity in a human brain spheroid model. Toxicol Appl Pharmacol. 2018;354:101–114. Available from: https://doi.org/10.1016/j.taap.2018.02.003
  109. Modafferi S, Zhong X, Kleensang A, Murata Y, Fagiani F, Pamies D, et al. Gene–environment interactions in developmental neurotoxicity: A case study of synergy between chlorpyrifos and CHD8 knockout in human brainspheres. Environ Health Perspect. 2021;129(7):077001. Available from: https://doi.org/10.1289/ehp8580
  110. Lam D, Enright HA, Cadena J, George VK, Soscia DA, Tooker AC, et al. Spatiotemporal analysis of 3D human iPSC-derived neural networks using a 3D multi-electrode array. Front Cell Neurosci. 2023;17:1287089. Available from: https://doi.org/10.3389/fncel.2023.1287089
  111. Mariani A, Comolli D, Fanelli R, Forloni G, De Paola M. Neonicotinoid pesticides affect developing neurons in experimental mouse models and in human induced pluripotent stem cell (iPSC)-derived neural cultures and organoids. Cells. 2024;13(5):1295. Available from: https://doi.org/10.3390/cells13151295
  112. Kobolak J, Teglasi A, Bellak T, Janstova Z, Molnar K, Zana M, et al. Human induced pluripotent stem cell-derived 3D-neurospheres are suitable for neurotoxicity screening. Cells. 2020;9(5):1122. Available from: https://doi.org/10.3390/cells9051122
  113. Sirenko O, Parham F, Dea S, Sodhi N, Biesmans S, Mora-Castilla S, et al. Functional and mechanistic neurotoxicity profiling using human iPSC-derived neural 3D cultures. Toxicol Sci. 2019;167(1):58–76. Available from: https://doi.org/10.1093/toxsci/kfy218
  114. Campisi M, Shin Y, Osaki T, Hajal C, Chiono V, Kamm RD. 3D self-organized microvascular model of the human blood-brain barrier with endothelial cells, pericytes and astrocytes. Biomaterials. 2018;180:117–129. Available from: https://doi.org/10.1016/j.biomaterials.2018.07.014
  115. Picollet-D’hahan N, Zuchowska A, Lemeunier I, Le Gac S. Multiorgan-on-a-chip: A systemic approach to model and decipher inter-organ communication. Trends Biotechnol. 2021;39(7):788–810. Available from: https://doi.org/10.1016/j.tibtech.2020.11.014
  116. Koo Y, Hawkins BT, Yun Y. Three-dimensional (3D) tetra-culture brain on chip platform for organophosphate toxicity screening. Sci Rep. 2018;8:2841. Available from: https://doi.org/10.1038/s41598-018-20876-2
  117. Liu L, Koo Y, Akwitti C, Russell T, Gay E, Laskowitz DT, et al. Three-dimensional (3D) brain microphysiological system for organophosphates and neurochemical agent toxicity screening. PLoS One. 2019;14(11). Available from: https://doi.org/10.1371/journal.pone.0224657
  118. Amend N, Koller M, Schmitt C, Worek F, Wille T. The suitability of a polydimethylsiloxane-based (PDMS) microfluidic two compartment system for the toxicokinetic analysis of organophosphorus compounds. Toxicol Lett. 2023;388:24–29. Available from: https://doi.org/10.1016/j.toxlet.2023.10.007
  119. Smirnova L, Hogberg HT, Leist M, Hartung T. Developmental neurotoxicity – Challenges in the 21st century and in vitro opportunities. ALTEX. 2014;31(2):129–156. Available from: https://doi.org/10.14573/altex.1403271
  120. Augustyniak J, Lenart J, Zychowicz M, Lipka G, Gaj P, Kolanowska M, et al. Sensitivity of hiPSC-derived neural stem cells (NSC) to pyrroloquinoline quinone depends on their developmental stage. Toxicol in vitro. 2017;45:434–444. Available from: https://doi.org/10.1016/j.tiv.2017.05.017
  121. Pei Y, Sierra G, Sivapatham R, Swistowski A, Rao MS, Zeng X. A platform for rapid generation of single and multiplexed reporters in human iPSC lines. Sci Rep. 2015;5:9205. Available from: https://doi.org/10.1038/srep09205
  122. Pei Y, Peng J, Behl M, Sipes NS, Shockley KR, Rao MS, et al. Comparative neurotoxicity screening in human iPSC-derived neural stem cells, neurons, and astrocytes. Brain Res. 2016;1638:57–73. Available from: https://doi.org/10.1016/j.brainres.2015.07.048
  123. Pistollato F, Carpi D, Mendoza-de Gyves E, Paini A, Bopp SK, Worth A, et al. Combining in vitro assays and mathematical modelling to study developmental neurotoxicity induced by chemical mixtures. Reprod Toxicol. 2021;105:101–119. Available from: https://doi.org/10.1016/j.reprotox.2021.08.007
  124. Rana P, Luerman G, Hess D, Rubitski E, Adkins K, Somps C. Utilization of iPSC-derived human neurons for high-throughput drug-induced peripheral neuropathy screening. Toxicol in vitro. 2017;45:111–118. Available from: https://doi.org/10.1016/j.tiv.2017.08.014
  125. Shirakawa T, Suzuki I. Approach to neurotoxicity using human iPSC neurons: Consortium for safety assessment using human iPS cells. Curr Pharm Biotechnol. 2020;21(11):780–786. Available from: https://doi.org/10.2174/1389201020666191129103730
  126. Sirenko O, Parham F, Dea S, Sodhi N, Biesmans S, Mora-Castilla S, et al. Functional and mechanistic neurotoxicity profiling using human iPSC-derived neural 3D cultures. Toxicol Sci. 2019;167(1):58–76. Available from: https://doi.org/10.1093/toxsci/kfy218
  127. Tukker AM, Wijnolts FMJ, de Groot A, Westerink RHS. Applicability of hiPSC-derived neuronal cocultures and rodent primary cortical cultures for in vitro seizure liability assessment. Toxicol Sci. 2020;178(1):71–87. Available from: https://doi.org/10.1093/toxsci/kfaa136
  128. Wellens S, Dehouck L, Chandrasekaran V, Singh P, Loiola RA, Sevin E, et al. Evaluation of a human iPSC-derived BBB model for repeated dose toxicity testing with cyclosporine A as model compound. Toxicol in vitro. 2021;73:105112. Available from: https://doi.org/10.1016/j.tiv.2021.105112
  129. Slavin I, Dea S, Arunkumar P, Sodhi N, Montefusco S, Siqueira-Neto J, et al. Human iPSC-derived 2D and 3D platforms for rapidly assessing developmental, functional, and terminal toxicities in neural cells. Int J Mol Sci. 2021;22(4):1908. Available from: https://doi.org/10.3390/ijms22041908
  130. Pamies D, Wiersma D, Katt ME, Zhao L, Burtscher J, Harris G, et al. Human iPSC 3D brain model as a tool to study chemical-induced dopaminergic neuronal toxicity. Neurobiol Dis. 2022;169:105719. Available from: https://doi.org/10.1016/j.nbd.2022.105719
  131. Pomeshchik Y, Klementieva O, Gil J, Martinsson I, Hansen MG, de Vries T, et al. Human iPSC-derived hippocampal spheroids: An innovative tool for stratifying Alzheimer disease patient-specific cellular phenotypes and developing therapies. Stem Cell Rep. 2020;15(2):256–273. Available from: https://doi.org/10.1016/j.stemcr.2020.06.001
  132. Liu L, Koo Y, Russell T, Gay E, Li Y, Yun Y. Three-dimensional brain-on-chip model using human iPSC-derived GABAergic neurons and astrocytes: Butyrylcholinesterase post-treatment for acute malathion exposure. PLoS One. 2020;15(4). Available from: https://doi.org/10.1371/journal.pone.0230335
  133. Campisi M, Shin Y, Osaki T, Hajal C, Chiono V, Kamm RD. 3D self-organized microvascular model of the human blood-brain barrier with endothelial cells, pericytes, and astrocytes. Biomaterials. 2018;180:117–129. Available from: https://doi.org/10.1016/j.biomaterials.2018.07.014
  134. Lee C-T, Bendriem RM, Wu WW, Shen RF. 3D brain organoids derived from pluripotent stem cells: promising experimental models for brain development and neurodegenerative disorders. J Biomed Sci. 2017;24:59. Available from: https://doi.org/10.1186/s12929-017-0362-8
  135. Potjewyd G, Kellett KAB, Hooper NM. 3D hydrogel models of the neurovascular unit to investigate blood–brain barrier dysfunction. Neuronal Signaling. 2021;5(4). Available from: https://doi.org/10.1042/ns20210027
  136. Xiao S, Coppeta JR, Rogers HB, Isenberg BC, Zhu J, Olalekan SA, et al. A microfluidic culture model of the human reproductive tract and 28-day menstrual cycle. Nat Commun. 2017;8:14584. Available from: https://doi.org/10.1038/ncomms14584
  137. Taylor AM, Blurton-Jones M, Rhee SW, Cribbs DH, Cotman CW, Jeon NL. A microfluidic culture platform for CNS axonal injury, regeneration and transport. Nat Methods. 2005;2(9):599–605. Available from: https://doi.org/10.1038/nmeth777
  138. Bang S, Lee S, Choi N, Kim HN. Emerging brain‐pathophysiology‐mimetic platforms for studying neurodegenerative diseases: Brain organoids and brains‐on‐a‐chip. Adv Healthcare Mater. 2021;10(8):2002119. Available from: https://doi.org/10.1002/adhm.202002119
  139. Koo Y, Hawkins BT, Yun Y. Three-dimensional (3D) tetra-culture brain on chip platform for organophosphate toxicity screening. Sci Rep. 2018;8:2841. Available from: https://doi.org/10.1038/s41598-018-20876-2
  140. Lanciotti A, Brignone MS, Macioce P, Visentin S. Human iPSC-derived astrocytes: A powerful tool to study primary astrocyte dysfunction in the pathogenesis of rare leukodystrophies. Int J Mol Sci. 2021;23(1):274. Available from: https://doi.org/10.3390/ijms23010274
  141. Koo Y, Hawkins BT, Yun Y. Three-dimensional (3D) tetra-culture brain on chip platform for organophosphate toxicity screening. Sci Rep. 2018;8:2841. Available from: https://doi.org/10.1038/s41598-018-20876-2
  142. Linville RM, DeStefano JG, Sklar MB, Xu Z, Farrell AM, Bogorad MI, et al. Human iPSC-derived blood-brain barrier microvessels: validation of barrier function and endothelial cell behavior. Biomaterials. 2019;190–191:24–37. Available from: https://doi.org/10.1016/j.biomaterials.2018.10.023
  143. Middelkamp HHT, Verboven AHA, De Sá Vivas AG, Schoenmaker C, Klein Gunnewiek TM, Passier R, et al. Cell type-specific changes in transcriptomic profiles of endothelial cells, iPSC-derived neurons and astrocytes cultured on microfluidic chips. Sci Rep. 2021;11:2281. Available from: https://doi.org/10.1038/s41598-021-81933-x
  144. Popova G, Soliman SS, Kim CN, Keefe MG, Hennick KM, Jain S, et al. Human microglia states are conserved across experimental models and regulate neural stem cell responses in chimeric organoids. Cell Stem Cell. 2021;28(12):2153-2166.e6. Available from: https://doi.org/10.1016/j.stem.2021.08.015
  145. Soubannier V, Maussion G, Chaineau M, Sigutova V, Rouleau G, Durcan TM, et al. Characterization of human iPSC-derived astrocytes with potential for disease modeling and drug discovery. Neurosci Lett. 2020;731:135028. Available from: https://doi.org/10.1016/j.neulet.2020.135028
  146. Wörsdörfer P, Dalda N, Kern A, Krüger S, Wagner N, Kwok CK, et al. Generation of complex human organoid models including vascular networks by incorporation of mesodermal progenitor cells. Sci Rep. 2019;9:15663. Available from: https://doi.org/10.1038/s41598-019-52204-7
  147. Zhang Y, Sloan SA, Clarke LE, Caneda C, Plaza CA, Blumenthal PD, et al. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron. 2016;89:37–53. Available from: https://doi.org/10.1016/j.neuron.2015.11.013
  148. Nix C, Ghassemi M, Crommen J, Fillet M. Overview on microfluidics devices for monitoring brain disorder biomarkers. TrAC Trends in Analytical Chemistry. 2022;155:116693. Available from: https://scite.ai/reports/overview-on-microfluidics-devices-for-A3rxYKme
  149. Chang C-Y, Ting H-C, Liu C-A, Su HL, Chiou TW, Harn HJ, et al. Induced pluripotent stem cells: A powerful neurodegenerative disease modeling tool for mechanism study and drug discovery. Cell Transplant. 2018;27:1588–1602. Available from: https://doi.org/10.1177/0963689718775406
  150. Li S, Xia M. Review of high-content screening applications in toxicology. Arch Toxicol. 2019;93:3387–3396. Available from: https://doi.org/10.1007/s00204-019-02593-5
  151. Zhang Q, Caudle WM, Pi J, Bhattacharya S, Andersen ME, Kaminski NE, et al. Embracing systems toxicology at single-cell resolution. Curr Opin Toxicol. 2019;16:49–57. Available from: https://doi.org/10.1016/j.cotox.2019.04.003
  152. Ashammakhi N, Darabi MA, Çelebi‐Saltik B, Tutar R, Hartel MC, Lee J, et al. Microphysiological systems: Next generation systems for assessing toxicity and therapeutic effects of nanomaterials. Small Methods. 2020;4:1900589. Available from: https://doi.org/10.1002/smtd.201900589
  153. Mayr A, Klambauer G, Unterthiner T, Hochreiter S. DeepTox: Toxicity prediction using deep learning. Front Environ Sci. 2016;3. Available from: http://dx.doi.org/10.3389/fenvs.2015.00080
  154. Srivastava A, Hanig JP. Quantitative neurotoxicology: Potential role of artificial intelligence/deep learning approach. J Appl Toxicol. 2021;41:996–1006. Available from: https://doi.org/10.1002/jat.4098
  155. Myllynen P, Pasanen M, Pelkonen O. Human placenta: A human organ for developmental toxicology research and biomonitoring. Placenta. 2005;26:361–371. Available from: https://doi.org/10.1016/j.placenta.2004.09.006
  156. Rice D, Barone S. Critical periods of vulnerability for the developing nervous system: Evidence from humans and animal models. Environ Health Perspect. 2000;108:511–533. Available from: https://doi.org/10.1289/ehp.00108s3511