1
|
Üremiş MM, Üremiş N, Gül M, Gül S, Çiğremiş Y, Durhan M, Türköz Y. Acrylamide, Applied During Pregnancy and Postpartum Period in Offspring Rats, Significantly Disrupted Myelination by Decreasing the Levels of Myelin-Related Proteins: MBP, MAG, and MOG. Neurochem Res 2024; 49:617-635. [PMID: 37989894 DOI: 10.1007/s11064-023-04053-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 08/06/2023] [Accepted: 10/19/2023] [Indexed: 11/23/2023]
Abstract
Acrylamide (ACR) is a colorless, odorless, and water-soluble solid molecule. In addition to being an important industrial material, ACR is found in fried and baked carbohydrate-rich foods. ACR is regarded as a typical axonal neurotoxin that induces neuropathy. The brain is protected from oxidative damage by vitamin E, which is regarded as the most powerful fat-soluble antioxidant vitamin. This study aimed to reveal the toxic effect of ACR on the development of myelin in the brain at the molecular level and to examine whether Vitamin E has a neuroprotective effect on the harmful effect of ACR. The study was started by dividing 40 pregnant rats into 4 groups and after lactation, the study was continued with offspring rats (females and males offspring rats) from each group. Offspring rats were equally divided into Control, Vitamin E, ACR, ACR + Vitamin E groups. Following the ACR administration, the Water Maze test was applied to evaluate cognitive function. To evaluate the level of demyelination and remyelination, MBP, MAG, and MOG proteins and mRNA levels were performed. In addition, the degeneration of myelin and glial cells was examined by immunohistochemistry and electron microscopic analysis. Analysis results showed that ACR administration decreased gene and protein levels of myelin-related proteins MBP, MAG, and MOG. The findings were confirmed by histopathological, immunohistochemical, and microscopic examinations. The application of vitamin E improved this negative effect of ACR. It has been observed that ACR may play a role in the pathogenesis of myelin-related neurodegenerative diseases by causing demyelination during gestation, lactation, and post-lactation. In addition, it has been understood that vitamin E supports myelination as a strong neuroprotective vitamin against the toxicity caused by ACR. Our research results suggest that acrylamide may play a role in the etiopathogenesis of demyelinating diseases such as multiple sclerosis in humans since fast-food-type nutrition is very common today and people are chronically exposed to acrylamide.
Collapse
Affiliation(s)
- Muhammed Mehdi Üremiş
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| | - Nuray Üremiş
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey
| | - Mehmet Gül
- Department of Histology and Embryology, Medical Faculty, Inonu University, Malatya, Turkey
| | - Semir Gül
- Department of Histology and Embryology, Medical Faculty, Inonu University, Malatya, Turkey
| | - Yılmaz Çiğremiş
- Department of Medical Biology and Genetics, Medical Faculty, Inonu University, Malatya, Turkey
| | - Merve Durhan
- Department of Medical Biology and Genetics, Medical Faculty, Inonu University, Malatya, Turkey
| | - Yusuf Türköz
- Department of Medical Biochemistry, Medical Faculty, Inonu University, Malatya, Turkey.
| |
Collapse
|
2
|
Samrani LMM, Dumont F, Hallmark N, Bars R, Tinwell H, Pallardy M, Piersma AH. Nervous system development related gene expression regulation in the zebrafish embryo after exposure to valproic acid and retinoic acid: A genome wide approach. Toxicol Lett 2023; 384:96-104. [PMID: 37451652 DOI: 10.1016/j.toxlet.2023.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/04/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
The evaluation of chemical and pharmaceutical safety for humans is moving from animal studies to New Approach Methodologies (NAM), reducing animal use and focusing on mechanism of action, whilst enhancing human relevance. In developmental toxicology, the mechanistic approach is facilitated by the assessment of predictive biomarkers, which allow mechanistic pathways perturbation monitoring at the basis of human hazard assessment. In our search for biomarkers of maldevelopment, we focused on chemically-induced perturbation of the retinoic acid signaling pathway (RA-SP), a major pathway implicated in a plethora of developmental processes. A genome-wide expression screening was performed on zebrafish embryos treated with two teratogens, all-trans retinoic acid (ATRA) and valproic acid (VPA), and a non-teratogen reference compound, folic acid (FA). Each compound was found to have a specific mRNA expression profile with 248 genes commonly dysregulated by both teratogenic compounds but not by FA. These genes were implicated in several developmental processes (e.g., the circulatory and nervous system). Given the prominent response of neurodevelopmental gene sets, and the crucial need to better understand developmental neurotoxicity, our study then focused on nervous system development. We found 62 genes that are potential early neurodevelopmental toxicity biomarker candidates. These results advance NAM-based safety assessment evaluation by highlighting the usefulness of the RA-SP in providing early toxicity biomarker candidates.
Collapse
Affiliation(s)
- Laura M M Samrani
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Université Paris-Saclay, Inflammation, Microbiome and Immunosurveillance, INSERM, Faculté Pharmacie, 91104 Orsay, France; Institute for Risk Assessment Sciences (IRAS), Utrecht University, the Netherlands.
| | | | | | | | | | - Marc Pallardy
- Université Paris-Saclay, Inflammation, Microbiome and Immunosurveillance, INSERM, Faculté Pharmacie, 91104 Orsay, France
| | - Aldert H Piersma
- Centre for Health Protection National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, the Netherlands
| |
Collapse
|
3
|
Hinojosa MG, Johansson Y, Cediel-Ulloa A, Ivanova E, Gabring N, Gliga A, Forsby A. Evaluation of mRNA markers in differentiating human SH-SY5Y cells for estimation of developmental neurotoxicity. Neurotoxicology 2023; 97:65-77. [PMID: 37210002 DOI: 10.1016/j.neuro.2023.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/05/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
Current guidelines for developmental neurotoxicity (DNT) evaluation are based on animal models. These have limitations so more relevant, efficient and robust approaches for DNT assessment are needed. We have used the human SH-SY5Y neuroblastoma cell model to evaluate a panel of 93 mRNA markers that are frequent in Neuronal diseases and functional annotations and also differentially expressed during retinoic acid-induced differentiation in the cell model. Rotenone, valproic acid (VPA), acrylamide (ACR) and methylmercury chloride (MeHg) were used as DNT positive compounds. Tolbutamide, D-mannitol and clofibrate were used as DNT negative compounds. To determine concentrations for exposure for gene expression analysis, we developed a pipeline for neurite outgrowth assessment by live-cell imaging. In addition, cell viability was measured by the resazurin assay. Gene expression was analyzed by RT-qPCR after 6 days of exposure during differentiation to concentrations of the DNT positive compounds that affected neurite outgrowth, but with no or minimal effect on cell viability. Methylmercury affected cell viability at lower concentrations than neurite outgrowth, hence the cells were exposed with the highest non-cytotoxic concentration. Rotenone (7.3nM) induced 32 differentially expressed genes (DEGs), ACR (70µM) 8 DEGs, and VPA (75µM) 16 DEGs. No individual genes were significantly dysregulated by all 3 DNT positive compounds (p<0.05), but 9 genes were differentially expressed by 2 of them. Methylmercury (0.8nM) was used to validate the 9 DEGs. The expression of SEMA5A (encoding semaphorin 5A) and CHRNA7 (encoding nicotinic acetylcholine receptor subunit α7) was downregulated by all 4 DNT positive compounds. None of the DNT negative compounds dysregulated any of the 9 DEGs in common for the DNT positive compounds. We suggest that SEMA5A or CHRNA7 should be further evaluated as biomarkers for DNT studies in vitro since they also are involved in neurodevelopmental adverse outcomes in humans.
Collapse
Affiliation(s)
- M G Hinojosa
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Y Johansson
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - A Cediel-Ulloa
- Department of Organismal Biology, Environmental Toxicology, Uppsala University, 752 36, Uppsala, Sweden
| | - E Ivanova
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - N Gabring
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - A Gliga
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, 171 77, Sweden
| | - A Forsby
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| |
Collapse
|
4
|
Lauvås AJ, Lislien M, Holme JA, Dirven H, Paulsen RE, Alm IM, Andersen JM, Skarpen E, Sørensen V, Macko P, Pistollato F, Duale N, Myhre O. Developmental neurotoxicity of acrylamide and its metabolite glycidamide in a human mixed culture of neurons and astrocytes undergoing differentiation in concentrations relevant for human exposure. Neurotoxicology 2022; 92:33-48. [PMID: 35835329 DOI: 10.1016/j.neuro.2022.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/29/2022] [Accepted: 07/08/2022] [Indexed: 11/19/2022]
Abstract
Neural stem cells (NSCs) derived from human induced pluripotent stem cells were used to investigate effects of exposure to the food contaminant acrylamide (AA) and its main metabolite glycidamide (GA) on key neurodevelopmental processes. Diet is an important source of human AA exposure for pregnant women, and AA is known to pass the placenta and the newborn may also be exposed through breast feeding after birth. The NSCs were exposed to AA and GA (1 ×10-8 - 3 ×10-3 M) under 7 days of proliferation and up to 28 days of differentiation towards a mixed culture of neurons and astrocytes. Effects on cell viability was measured using Alamar Blue™ cell viability assay, alterations in gene expression were assessed using real time PCR and RNA sequencing, and protein levels were quantified using immunocytochemistry and high content imaging. Effects of AA and GA on neurodevelopmental processes were evaluated using endpoints linked to common key events identified in the existing developmental neurotoxicity adverse outcome pathways (AOPs). Our results suggest that AA and GA at low concentrations (1 ×10-7 - 1 ×10-8 M) increased cell viability and markers of proliferation both in proliferating NSCs (7 days) and in maturing neurons after 14-28 days of differentiation. IC50 for cell death of AA and GA was 5.2 × 10-3 M and 5.8 × 10-4 M, respectively, showing about ten times higher potency for GA. Increased expression of brain derived neurotrophic factor (BDNF) concomitant with decreased synaptogenesis were observed for GA exposure (10-7 M) only at later differentiation stages, and an increased number of astrocytes (up to 3-fold) at 14 and 21 days of differentiation. Also, AA exposure gave tendency towards decreased differentiation (increased percent Nestin positive cells). After 28 days, neurite branch points and number of neurites per neuron measured by microtubule-associated protein 2 (Map2) staining decreased, while the same neurite features measured by βIII-Tubulin increased, indicating perturbation of neuronal differentiation and maturation.
Collapse
Affiliation(s)
- Anna Jacobsen Lauvås
- Department of Chemical Toxicology, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Malene Lislien
- Department of Chemical Toxicology, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Jørn Andreas Holme
- Department of Chemical Toxicology, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Hubert Dirven
- Department of Chemical Toxicology, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Ragnhild Elisabeth Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Inger Margit Alm
- Department of Chemical Toxicology, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Jill Mari Andersen
- Department of Chemical Toxicology, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Ellen Skarpen
- Core Facility for Advanced Light Microscopy, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Vigdis Sørensen
- Core Facility for Advanced Light Microscopy, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Peter Macko
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | - Nur Duale
- Department of Chemical Toxicology, Norwegian Institute of Public Health (NIPH), Oslo, Norway
| | - Oddvar Myhre
- Department of Chemical Toxicology, Norwegian Institute of Public Health (NIPH), Oslo, Norway.
| |
Collapse
|
5
|
A bioinformatics framework for targeted gene expression assay design: Application to in vitro developmental neurotoxicity screening in a rat model. Regul Toxicol Pharmacol 2022; 133:105211. [PMID: 35724854 DOI: 10.1016/j.yrtph.2022.105211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/05/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022]
Abstract
Brain development involves a series of intricately choreographed neuronal differentiation and maturation steps that are acutely vulnerable to interferences from chemical exposures. Many genes involved in neurodevelopmental processes show evolutionarily conserved expression patterns in mammals and may constitute useful indicators/biomarkers for the evaluation of potential developmental neurotoxicity. Based on these premises, this study developed a bioinformatics framework to guide the design of a gene expression-based in vitro developmental neurotoxicity assay targeting evolutionary conserved genes associated with neuronal differentiation and maturation in rat cerebellar granule cells (CGCs). Rat, mouse and human genes involved in neurodevelopment and presenting one-to-one orthology were selected and orthologous exons within these genes were identified. PCR primer sets were designed within these orthologous exons and their specificity was evaluated in silico. The performance and specificity of rat, mouse and human PCR primer sets were then confirmed experimentally. Finally, RT-qPCR analyses in CGCs exposed in vitro to well-known neurotoxicants (Chlorpyrifos and Chlorpyrifos oxon) uncovered perturbations of expression levels for most of the selected genes. This bioinformatics framework for gene and target sequence selection may facilitate the identification of transcriptional biomarkers for developmental neurotoxicity assays and the comparison of gene expression data across experimental models from different mammalian species.
Collapse
|
6
|
Establishment of a 13 genes-based molecular prediction score model to discriminate the neurotoxic potential of food relevant-chemicals. Toxicol Lett 2021; 355:1-18. [PMID: 34748853 DOI: 10.1016/j.toxlet.2021.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 08/30/2021] [Accepted: 10/26/2021] [Indexed: 11/23/2022]
Abstract
Although many neurotoxicity prediction studies of food additives have been developed, they are applicable in a qualitative way. We aimed to develop a novel prediction score that is described quantitatively and precisely. We examined cell viability, reactive oxygen species activity, intracellular calcium and RNA transcription level of potential prediction related genes to develop a high-throughput neurotoxicity test method in vitro to screen the neurotoxicity of hazardous factors in food using AI-based machine learning. We trained artificial intelligence models (random forest and neural network) to predict neurotoxicity precisely, establishing a universal classification assessment score (CA-Score) that relies on the expression status of only 13 of prediction related genes. The CA-Score system is almost universally applicable to food risk factors (p<0.05) in a manner independent of platform (microarray or RNA sequencing) by being compared with cut-off value 23.487 to judge whether it's neurotoxic or not. We finally validated our prediction with the external validation of CA-Score on neural precursor cells derived from embryonic stem cells. Therefore, we draw a conclusion that the AI-based machine learning including neural network and random forest is likely to provide a useful tool for large-scale screening of neurotoxicity in food risk factors.
Collapse
|
7
|
Kumar D, Baligar P, Srivastav R, Narad P, Raj S, Tandon C, Tandon S. Stem Cell Based Preclinical Drug Development and Toxicity Prediction. Curr Pharm Des 2021; 27:2237-2251. [PMID: 33076801 DOI: 10.2174/1381612826666201019104712] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/22/2020] [Indexed: 01/09/2023]
Abstract
Stem cell based toxicity prediction plays a very important role in the development of the drug. Unexpected adverse effects of the drugs during clinical trials are a major reason for the termination or withdrawal of drugs. Methods for predicting toxicity employ in vitro as well as in vivo models; however, the major drawback seen in the data derived from these animal models is the lack of extrapolation, owing to interspecies variations. Due to these limitations, researchers have been striving to develop more robust drug screening platforms based on stem cells. The application of stem cells based toxicity testing has opened up robust methods to study the impact of new chemical entities on not only specific cell types, but also organs. Pluripotent stem cells, as well as cells derived from them, can be evaluated for modulation of cell function in response to drugs. Moreover, the combination of state-of-the -art techniques such as tissue engineering and microfluidics to fabricate organ- on-a-chip, has led to assays which are amenable to high throughput screening to understand the adverse and toxic effects of chemicals and drugs. This review summarizes the important aspects of the establishment of the embryonic stem cell test (EST), use of stem cells, pluripotent, induced pluripotent stem cells and organoids for toxicity prediction and drug development.
Collapse
Affiliation(s)
- Dhruv Kumar
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh 201313, India
| | - Prakash Baligar
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh 201313, India
| | - Rajpal Srivastav
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201313, India
| | - Priyanka Narad
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201313, India
| | - Sibi Raj
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh 201313, India
| | - Chanderdeep Tandon
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh 201313, India
| | - Simran Tandon
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida, Uttar Pradesh 201313, India
| |
Collapse
|
8
|
Davidsen N, Lauvås AJ, Myhre O, Ropstad E, Carpi D, Gyves EMD, Berntsen HF, Dirven H, Paulsen RE, Bal-Price A, Pistollato F. 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. [PMID: 33333158 PMCID: PMC7992035 DOI: 10.1016/j.reprotox.2020.12.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 12/03/2020] [Accepted: 12/11/2020] [Indexed: 12/11/2022]
Abstract
Halogenated persistent organic pollutants (POPs) like perfluorinated alkylated substances (PFASs), brominated flame retardants (BFRs), organochlorine pesticides and polychlorinated biphenyls (PCBs) are known to cause cancer, immunotoxicity, neurotoxicity and interfere with reproduction and development. Concerns have been raised about the impact of POPs upon brain development and possibly neurodevelopmental disorders. The developing brain is a particularly vulnerable organ due to dynamic and complex neurodevelopmental processes occurring early in life. However, very few studies have reported on the effects of POP mixtures at human relevant exposures, and their impact on key neurodevelopmental processes using human in vitro test systems. Aiming to reduce this knowledge gap, we exposed mixed neuronal/glial cultures differentiated from neural stem cells (NSCs) derived from human induced pluripotent stem cells (hiPSCs) to reconstructed mixtures of 29 different POPs using concentrations comparable to Scandinavian human blood levels. Effects of the POP mixtures on neuronal proliferation, differentiation and synaptogenesis were evaluated using in vitro assays anchored to common key events identified in the existing developmental neurotoxicity (DNT) adverse outcome pathways (AOPs). The present study showed that mixtures of POPs (in particular brominated and chlorinated compounds) at human relevant concentrations increased proliferation of NSCs and decreased synapse number. Based on a mathematical modelling, synaptogenesis and neurite outgrowth seem to be the most sensitive DNT in vitro endpoints. Our results indicate that prenatal exposure to POPs may affect human brain development, potentially contributing to recently observed learning and memory deficits in children.
Collapse
Affiliation(s)
- Nichlas Davidsen
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| | - Anna Jacobsen Lauvås
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| | - Oddvar Myhre
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| | - Erik Ropstad
- Department of Production Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Donatella Carpi
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | - Hanne Friis Berntsen
- Department of Production Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway; National Institute of Occupational Health, Oslo, Norway
| | - Hubert Dirven
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| | - Ragnhild E Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Anna Bal-Price
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | |
Collapse
|
9
|
Lindeman B, Johansson Y, Andreassen M, Husøy T, Dirven H, Hofer T, Knutsen HK, Caspersen IH, Vejrup K, Paulsen RE, Alexander J, Forsby A, Myhre O. Does the food processing contaminant acrylamide cause developmental neurotoxicity? A review and identification of knowledge gaps. Reprod Toxicol 2021; 101:93-114. [PMID: 33617935 DOI: 10.1016/j.reprotox.2021.02.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/11/2021] [Accepted: 02/16/2021] [Indexed: 12/15/2022]
Abstract
There is a worldwide concern on adverse health effects of dietary exposure to acrylamide (AA) due to its presence in commonly consumed foods. AA is formed when carbohydrate rich foods containing asparagine and reducing sugars are prepared at high temperatures and low moisture conditions. Upon oral intake, AA is rapidly absorbed and distributed to all organs. AA is a known human neurotoxicant that can reach the developing foetus via placental transfer and breast milk. Although adverse neurodevelopmental effects have been observed after prenatal AA exposure in rodents, adverse effects of AA on the developing brain has so far not been studied in humans. However, epidemiological studies indicate that gestational exposure to AA impair foetal growth and AA exposure has been associated with reduced head circumference of the neonate. Thus, there is an urgent need for further research to elucidate whether pre- and perinatal AA exposure in humans might impair neurodevelopment and adversely affect neuronal function postnatally. Here, we review the literature with emphasis on the identification of critical knowledge gaps in relation to neurodevelopmental toxicity of AA and its mode of action and we suggest research strategies to close these gaps to better protect the unborn child.
Collapse
Affiliation(s)
- Birgitte Lindeman
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Ylva Johansson
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Mathilda Andreassen
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Trine Husøy
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Hubert Dirven
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Tim Hofer
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Helle K Knutsen
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Ida H Caspersen
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Kristine Vejrup
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Ragnhild E Paulsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Jan Alexander
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Anna Forsby
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Oddvar Myhre
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway.
| |
Collapse
|
10
|
Masood MI, Naseem M, Warda SA, Tapia-Laliena MÁ, Rehman HU, Nasim MJ, Schäfer KH. Environment permissible concentrations of glyphosate in drinking water can influence the fate of neural stem cells from the subventricular zone of the postnatal mouse. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 270:116179. [PMID: 33348142 DOI: 10.1016/j.envpol.2020.116179] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/03/2020] [Accepted: 11/26/2020] [Indexed: 06/12/2023]
Abstract
The developing nervous system is highly vulnerable to environmental toxicants especially pesticides. Glyphosate pesticide induces neurotoxicity both in humans and rodents, but so far only when exposed to higher concentrations. A few studies, however, have also reported the risk of general toxicity of glyphosate at concentrations comparable to allowable limits set up by environmental protection authorities. In vitro data regarding glyphosate neurotoxicity at concentrations comparable to maximum permissible concentrations in drinking water is lacking. In the present study, we established an in vitro assay based upon neural stem cells (NSCs) from the subventricular zone of the postnatal mouse to decipher the effects of two maximum permissible concentrations of glyphosate in drinking water on the basic neurogenesis processes. Our results demonstrated that maximum permissible concentrations of glyphosate recognized by environmental protection authorities significantly reduced the cell migration and differentiation of NSCs as demonstrated by the downregulation of the expression levels of the neuronal ß-tubulin III and the astrocytic S100B genes. The expression of the cytoprotective gene CYP1A1 was downregulated whilst the expression of oxidative stresses indicator gene SOD1 was upregulated. The concentration comparable to non-toxic human plasma concentration significantly induced cytotoxicity and activated Ca2+ signalling in the differentiated culture. Our findings demonstrated that the permissible concentrations of glyphosate in drinking water recognized by environmental protection authorities are capable of inducing neurotoxicity in the developing nervous system.
Collapse
Affiliation(s)
- Muhammad Irfan Masood
- Division of Bioorganic Chemistry, School of Pharmacy, Saarland University, Saarbrücken, D-66123, Germany; Working Group Enteric Nervous System, University of Applied Sciences Kaiserslautern, Campus Zweibrücken, 66482, Germany; Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, 54000, Pakistan.
| | - Mahrukh Naseem
- Department of Zoology, University of Balochistan, Quetta, 87550, Pakistan
| | - Salam A Warda
- Working Group Enteric Nervous System, University of Applied Sciences Kaiserslautern, Campus Zweibrücken, 66482, Germany
| | | | - Habib Ur Rehman
- Department of Physiology, University of Veterinary and Animal Sciences, Lahore, 54000, Pakistan
| | - Muhammad Jawad Nasim
- Division of Bioorganic Chemistry, School of Pharmacy, Saarland University, Saarbrücken, D-66123, Germany
| | - Karl Herbert Schäfer
- Working Group Enteric Nervous System, University of Applied Sciences Kaiserslautern, Campus Zweibrücken, 66482, Germany; Department of Pediatric Surgery Mannheim, University Medicine Mannheim, University of Heidelberg, Mannheim, 68167, Germany.
| |
Collapse
|
11
|
Hogberg HT, de Cássia da Silveira E Sá R, Kleensang A, Bouhifd M, Cemiloglu Ulker O, Smirnova L, Behl M, Maertens A, Zhao L, Hartung T. Organophosphorus flame retardants are developmental neurotoxicants in a rat primary brainsphere in vitro model. Arch Toxicol 2021; 95:207-228. [PMID: 33078273 PMCID: PMC7811506 DOI: 10.1007/s00204-020-02903-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/02/2020] [Indexed: 12/26/2022]
Abstract
Due to regulatory bans and voluntary substitutions, halogenated polybrominated diphenyl ether (PBDE) flame retardants (FR) are increasingly substituted by mainly organophosphorus FR (OPFR). Leveraging a 3D rat primary neural organotypic in vitro model (rat brainsphere), we compare developmental neurotoxic effects of BDE-47-the most abundant PBDE congener-with four OPFR (isopropylated phenyl phosphate-IPP, triphenyl phosphate-TPHP, isodecyl diphenyl phosphate-IDDP, and tricresyl phosphate (also known as trimethyl phenyl phosphate)-TMPP). Employing mass spectroscopy-based metabolomics and transcriptomics, we observe at similar human-relevant non-cytotoxic concentrations (0.1-5 µM) stronger developmental neurotoxic effects by OPFR. This includes toxicity to neurons in the low µM range; all FR decrease the neurotransmitters glutamate and GABA (except BDE-47 and TPHP). Furthermore, n-acetyl aspartate (NAA), considered a neurologic diagnostic molecule, was decreased by all OPFR. At similar concentrations, the FR currently in use decreased plasma membrane dopamine active transporter expression, while BDE-47 did not. Several findings suggest astrogliosis induced by the OPFR, but not BDE-47. At the 5 µM concentrations, the OPFR more than BDE-47 interfered with myelination. An increase of cytokine gene and receptor expressions suggests that exposure to OPFR may induce an inflammatory response. Pathway/category overrepresentation shows disruption in 1) transmission of action potentials, cell-cell signaling, synaptic transmission, receptor signaling, (2) immune response, inflammation, defense response, (3) cell cycle and (4) lipids metabolism and transportation. Taken together, this appears to be a case of regretful substitution with substances not less developmentally neurotoxic in a primary rat 3D model.
Collapse
Affiliation(s)
- Helena T Hogberg
- Center for Alternatives To Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Rita de Cássia da Silveira E Sá
- Center for Alternatives To Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Physiology and Pathology, Federal University of Paraíba, João Pessoa, Brazil
| | - Andre Kleensang
- Center for Alternatives To Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Mounir Bouhifd
- Center for Alternatives To Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ozge Cemiloglu Ulker
- Center for Alternatives To Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Toxicology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Lena Smirnova
- Center for Alternatives To Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Mamta Behl
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, USA
| | - Alexandra Maertens
- Center for Alternatives To Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Liang Zhao
- Center for Alternatives To Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas Hartung
- Center for Alternatives To Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- CAAT-Europe, University of Konstanz, Konstanz, Germany
| |
Collapse
|
12
|
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. [PMID: 33011216 PMCID: PMC7772889 DOI: 10.1016/j.reprotox.2020.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/17/2020] [Accepted: 09/24/2020] [Indexed: 12/19/2022]
Abstract
Human iPSC-derived NSCs undergoing differentiation possess some metabolic competence. CPF entered the cells and was biotrasformed into its two main metabolites (CPFO and TCP). After repeated exposure, very limited bioaccumulation of CPF was observed. Treatment with CPF decreased neurite outgrowth, synapse number and electrical activity. Treatment with CPF increased BDNF levels and the percentage of astrocytes.
For some complex toxicological endpoints, chemical safety assessment has conventionally relied on animal testing. Apart from the ethical issues, also scientific considerations have been raised concerning the traditional approach, highlighting the importance for considering real life exposure scenario. Implementation of flexible testing strategies, integrating multiple sources of information, including in vitro reliable test methods and in vitro biokinetics, would enhance the relevance of the obtained results. Such an approach could be pivotal in the evaluation of developmental neurotoxicity (DNT), especially when applied to human cell-based models, mimicking key neurodevelopmental processes, relevant to human brain development. Here, we integrated the kinetic behaviour with the toxicodynamic alterations of chlorpyrifos (CPF), such as in vitro endpoints specific for DNT evaluation, after repeated exposure during differentiation of human neural stem cells into a mixed culture of neurons and astrocytes. The upregulation of some cytochrome P450 and glutathione S-transferase genes during neuronal differentiation and the formation of the two major CPF metabolites (due to bioactivation and detoxification) supported the metabolic competence of the used in vitro model. The alterations in the number of synapses, neurite outgrowth, brain derived neurotrophic factor, the proportion of neurons and astrocytes, as well as spontaneous electrical activity correlated well with the CPF ability to enter the cells and be bioactivated to CPF-oxon. Overall, our results confirm that combining in vitro biokinetics and assays to evaluate effects on neurodevelopmental endpoints in human cells should be regarded as a key strategy for a quantitative characterization of DNT effects.
Collapse
Affiliation(s)
- Emma Di Consiglio
- Istituto Superiore di Sanità, Environment and Health Department, Mechanisms, Biomarkers and Models Unit, Rome, Italy
| | | | | | - Anna Bal-Price
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Emanuela Testai
- Istituto Superiore di Sanità, Environment and Health Department, Mechanisms, Biomarkers and Models Unit, Rome, Italy
| |
Collapse
|
13
|
Shafer TJ, Brown JP, Lynch B, Davila-Montero S, Wallace K, Friedman KP. Evaluation of Chemical Effects on Network Formation in Cortical Neurons Grown on Microelectrode Arrays. Toxicol Sci 2020; 169:436-455. [PMID: 30816951 DOI: 10.1093/toxsci/kfz052] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Thousands of chemicals to which humans are potentially exposed have not been evaluated for potential developmental neurotoxicity (DNT), driving efforts to develop a battery of in vitro screening approaches for DNT hazard. Here, 136 unique chemicals were evaluated for potential DNT hazard using a network formation assay (NFA) in cortical cells grown on microelectrode arrays. The effects of chemical exposure from 2 h postplating through 12 days in vitro (DIV) on network formation were evaluated at DIV 5, 7, 9, and 12, with cell viability assessed at DIV 12. Only 82 chemicals altered at least 1 network development parameter. Assay results were reproducible; 10 chemicals tested as biological replicates yielded qualitative results that were 100% concordant, with consistent potency values. Toxicological tipping points were determined for 58 chemicals and were similar to or lower than the lowest 50% effect concentrations (EC50) for all parameters. When EC50 and tipping point values from the NFA were compared to the range of potencies observed in ToxCast assays, the NFA EC50 values were less than the lower quartile for ToxCast assay potencies for a subset of chemicals, many of which are acutely neurotoxic in vivo. For 13 chemicals with available in vivo DNT data, estimated administered equivalent doses based on NFA results were similar to or lower than administered doses in vivo. Collectively, these results indicate that the NFA is sensitive to chemicals acting on nervous system function and will be a valuable contribution to an in vitro DNT screening battery.
Collapse
Affiliation(s)
- Timothy J Shafer
- Integrated Systems Toxicology Division, NHEERL, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Jasmine P Brown
- Integrated Systems Toxicology Division, NHEERL, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711.,Graduate Program in Public Health, University of Michigan, Ann Arbor, MI
| | - Brittany Lynch
- Tandon School of Engineering, New York University, Brooklyn, New York 11201
| | - Sylmarie Davila-Montero
- Department of Electrical and Computer Engineering, Michigan State University, E. Lansing, Michigan 48824
| | - Kathleen Wallace
- Integrated Systems Toxicology Division, NHEERL, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Katie Paul Friedman
- National Center for Computational Toxicology, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| |
Collapse
|
14
|
Ben Bacha A, Norah AO, Al-Osaimi M, Harrath AH, Mansour L, El-Ansary A. The therapeutic and protective effects of bee pollen against prenatal methylmercury induced neurotoxicity in rat pups. Metab Brain Dis 2020; 35:215-224. [PMID: 31625070 DOI: 10.1007/s11011-019-00496-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 09/12/2019] [Indexed: 12/14/2022]
Abstract
The current study evaluated the protective and therapeutic potency of bee pollen in ameliorating the toxic effects of methylmercury (MeHg), by measuring certain biochemical parameters related to neurotransmission, neuroinflammation, apoptosis, and glutamate excitotoxicity in the male neonate brain. Healthy, pregnant female rats (N = 40) were randomly divided into 5 groups, each comprising10 male neonates, as follows: (i) neonates delivered by control mothers; (ii) neonates delivered by MeHg-treated mothers who received 0.5 mg/kg BW/day MeHg via drinking water from gestational day 7 till postnatal day 7; (iii) neonates delivered by bee pollen treated mothers who received 200-mg/kg BW bee pollen from postnatal day 0 for 4 weeks; (iv) protective group of neonates delivered by MeHg and bee pollen-treated mothers, who continued to receive bee pollen until day 21 at the same dose, and (v) therapeutic group of neonates delivered by MeHg- treated mothers followed by bee pollen treatment, wherein they received 200-mg/kg BW bee pollen from postnatal day 0 for 4 weeks. Selected biochemical parameters in brain homogenates from each group were measured. MeHg-treated groups exhibited various signs of brain toxicity, such as a marked reduction in neurotransmitters (serotonin (5-HT), nor-adrenalin (NA), dopamine (DA)) and gamma aminobutyric acid (GABA) and elevated levels of interferon gamma (IFN-γ), caspase-3, and glutamate (Glu). Bee pollen effectively reduced the neurotoxic effects of MeHg. Minimal changes in all measured parameters were observed in MeHg-treated animals compared to the control group. Therefore, bee pollen may safely improve neurotransmitter defects, inflammation, apoptosis, and glutamate excitotoxicity.
Collapse
Affiliation(s)
- Abir Ben Bacha
- Biochemistry Department, Science College, King Saud University, P.O Box 22452, Riyadh, 11495, Saudi Arabia.
- Laboratory of Plant Biotechnology Applied to Crop Improvement, Faculty of Science of Sfax, University of Sfax, Sfax, Tunisia.
| | - Al-Orf Norah
- Biochemistry Department, Science College, King Saud University, P.O Box 22452, Riyadh, 11495, Saudi Arabia
- The Materials Science Research Institute, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - May Al-Osaimi
- Biochemistry Department, Science College, King Saud University, P.O Box 22452, Riyadh, 11495, Saudi Arabia
| | - Abdel Halim Harrath
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Lamjed Mansour
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Laboratory, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Sachana M, Bal-Price A, Crofton KM, Bennekou SH, Shafer TJ, Behl M, Terron A. International Regulatory and Scientific Effort for Improved Developmental Neurotoxicity Testing. Toxicol Sci 2019; 167:45-57. [PMID: 30476307 DOI: 10.1093/toxsci/kfy211] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The Organisation for Economic Co-Operation and Development (OECD) coordinates international efforts to enhance developmental neurotoxicity (DNT) testing. In most regulatory sectors, including the ones dealing with pesticides and industrial chemicals registration, historical use of the in vivo DNT test guideline has been limited. Current challenges include a lack of DNT data and mechanistic information for thousands of chemicals, and difficulty in interpreting results. A series of workshops in the last decade has paved the way for a consensus among stakeholders that there is need for a DNT testing battery that relies on in vitro endpoints (proliferation, differentiation, synaptogenesis, etc.) and is complemented by alternative species (eg, zebrafish) assays. Preferably, a battery of in vitro and alternative assays should be anchored toward mechanistic relevance for applying an integrated approach for testing and assessment (IATA) framework. Specific activities have been initiated to facilitate this OECD project: the collation of available DNT in vitro methods and their scoring for readiness; the selection of these methods to form a DNT testing battery; the generation of a reference set of chemicals that will be tested using the battery; the case studies exemplifying how DNT in vitro data can be interpreted; and the development of an OECD guidance document. This manuscript highlights these international efforts and activities.
Collapse
Affiliation(s)
- Magdalini Sachana
- Organisation for Economic Co-Operation and Development (OECD), 75775 Paris Cedex 16, France
| | - Anna Bal-Price
- European Commission Joint Research Centre, Health, Consumers and Reference Materials, Unit Chemicals Safety and Alternative Methods I-21027 Ispra (VA), Italy
| | | | - Susanne H Bennekou
- Danish Environmental Protection Agency, Haraldsgade 53, DK - 2100, Copenhagen, Denmark
| | - Timothy J Shafer
- U.S. Environmental Protection Agency (EPA), Office of Research and Development, Research Triangle Park, North Carolina 27711, USA
| | - Mamta Behl
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences Research Triangle Park, North Carolina, 27709 USA
| | - Andrea Terron
- European Food Safety Authority, Via Carlo Magno, 1A, 43126, Parma, Italy
| |
Collapse
|
16
|
Iron Sulfide Minerals as Potential Active Capping Materials for Mercury-Contaminated Sediment Remediation: A Minireview. SUSTAINABILITY 2019. [DOI: 10.3390/su11061747] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Several innovative approaches have been proposed in recent years to remediate contaminated sediment to reduce human health and environmental risk. One of the challenges of sediment remediation stems from its unfeasible high cost, especially when ex situ strategies are selected. Therefore, in situ methods such as active capping have been emerging as possible options for solving sediment problems. Active capping methods have been extensively tested in field-scale sediment remediation for organic pollutants (e.g., PCBs, PAHs, DDT) contamination with good sequestration efficiency; however, these methods have not been widely tested for control of heavy metal pollutants, such as mercury (Hg). In this review, the potentials of using iron sulfide minerals to sequestrate Hg were discussed. Iron sulfide minerals are common in the natural environment and have shown good effectiveness in sequestrating Hg by adsorption or precipitation. Iron sulfides can also be synthesized in a laboratory and modified to enhance their sequestration ability for Hg. Some of the potential advantages of iron sulfides are pointed out here. Additional tests to understand the possibility of applying iron sulfides as active caps to remediate complicated environment systems should be conducted.
Collapse
|
17
|
Valand R, Magnusson P, Dziendzikowska K, Gajewska M, Wilczak J, Oczkowski M, Kamola D, Królikowski T, Kruszewski M, Lankoff A, Mruk R, Marcus Eide D, Sapierzyński R, Gromadzka-Ostrowska J, Duale N, Øvrevik J, Myhre O. Gene expression changes in rat brain regions after 7- and 28 days inhalation exposure to exhaust emissions from 1st and 2nd generation biodiesel fuels - The FuelHealth project. Inhal Toxicol 2018; 30:299-312. [DOI: 10.1080/08958378.2018.1520370] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Renate Valand
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Pål Magnusson
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Katarzyna Dziendzikowska
- Faculty of Human Nutrition and Consumer Science, Warsaw University of Life Sciences, Warsaw, Poland
| | - Malgorzata Gajewska
- Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Jacek Wilczak
- Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Michał Oczkowski
- Faculty of Human Nutrition and Consumer Science, Warsaw University of Life Sciences, Warsaw, Poland
| | - Dariusz Kamola
- Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Tomasz Królikowski
- Faculty of Human Nutrition and Consumer Science, Warsaw University of Life Sciences, Warsaw, Poland
| | - Marcin Kruszewski
- Institute of Nuclear Chemistry and Technology, Warsaw, Poland
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Lublin, Poland
| | - Anna Lankoff
- Institute of Nuclear Chemistry and Technology, Warsaw, Poland
- Jan Kochanowski University, Kielce, Poland
| | - Remigiusz Mruk
- Faculty of Production Engineering, Warsaw University of Life Sciences, Warsaw, Poland
| | - Dag Marcus Eide
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Rafał Sapierzyński
- Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | | | - Nur Duale
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Johan Øvrevik
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Oddvar Myhre
- Division of Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
18
|
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. [PMID: 29476865 PMCID: PMC6095942 DOI: 10.1016/j.taap.2018.02.008] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/05/2018] [Accepted: 02/13/2018] [Indexed: 01/23/2023]
Abstract
Currently, the identification of chemicals that have the potential to induce developmental neurotoxicity (DNT) is based on animal testing. Since at the regulatory level, systematic testing of DNT is not a standard requirement within the EU or USA chemical legislation safety assessment, DNT testing is only performed in higher tiered testing triggered based on chemical structure activity relationships or evidence of neurotoxicity in systemic acute or repeated dose toxicity studies. However, these triggers are rarely used and, in addition, do not always serve as reliable indicators of DNT, as they are generally based on observations in adult rodents. Therefore, there is a pressing need for developing alternative methodologies that can reliably support identification of DNT triggers, and more rapidly and cost-effectively support the identification and characterization of chemicals with DNT potential. We propose to incorporate mechanistic knowledge and data derived from in vitro studies to support various regulatory applications including: (a) the identification of potential DNT triggers, (b) initial chemical screening and prioritization, (c) hazard identification and characterization, (d) chemical biological grouping, and (e) assessment of exposure to chemical mixtures. Ideally, currently available cellular neuronal/glial models derived from human induced pluripotent stem cells (hiPSCs) should be used as they allow evaluation of chemical impacts on key neurodevelopmental processes, by reproducing different windows of exposure during human brain development. A battery of DNT in vitro test methods derived from hiPSCs could generate valuable mechanistic data, speeding up the evaluation of thousands of compounds present in industrial, agricultural and consumer products that lack safety data on DNT potential.
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission, Joint Research Centre (JRC), Ispra, Italy.
| | | | - Magdalini Sachana
- Organisation for Economic Co-operation and Development (OECD), 2 rue André Pascal, 75775 Paris, Cedex 16, France
| | | | - Sharon Munn
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Andrew Worth
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| |
Collapse
|
19
|
Myhre O, Låg M, Villanger GD, Oftedal B, Øvrevik J, Holme JA, Aase H, Paulsen RE, Bal-Price A, Dirven H. Early life exposure to air pollution particulate matter (PM) as risk factor for attention deficit/hyperactivity disorder (ADHD): Need for novel strategies for mechanisms and causalities. Toxicol Appl Pharmacol 2018; 354:196-214. [PMID: 29550511 DOI: 10.1016/j.taap.2018.03.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/14/2018] [Accepted: 03/12/2018] [Indexed: 12/11/2022]
Abstract
Epidemiological studies have demonstrated that air pollution particulate matter (PM) and adsorbed toxicants (organic compounds and trace metals) may affect child development already in utero. Recent studies have also indicated that PM may be a risk factor for neurodevelopmental disorders (NDDs). A pattern of increasing prevalence of attention deficit/hyperactivity disorder (ADHD) has been suggested to partly be linked to environmental pollutants exposure, including PM. Epidemiological studies suggest associations between pre- or postnatal exposure to air pollution components and ADHD symptoms. However, many studies are cross-sectional without possibility to reveal causality. Cohort studies are often small with poor exposure characterization, and confounded by traffic noise and socioeconomic factors, possibly overestimating the study associations. Furthermore, the mechanistic knowledge how exposure to PM during early brain development may contribute to increased risk of ADHD symptoms or cognitive deficits is limited. The closure of this knowledge gap requires the combined use of well-designed longitudinal cohort studies, supported by mechanistic in vitro studies. As ADHD has profound consequences for the children affected and their families, the identification of preventable risk factors such as air pollution exposure should be of high priority.
Collapse
Affiliation(s)
- Oddvar Myhre
- Department of Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway.
| | - Marit Låg
- Department of Air pollution and Noise, Norwegian Institute of Public Health, Oslo, Norway
| | - Gro D Villanger
- Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | - Bente Oftedal
- Department of Air pollution and Noise, Norwegian Institute of Public Health, Oslo, Norway
| | - Johan Øvrevik
- Department of Air pollution and Noise, Norwegian Institute of Public Health, Oslo, Norway
| | - Jørn A Holme
- Department of Air pollution and Noise, Norwegian Institute of Public Health, Oslo, Norway
| | - Heidi Aase
- Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | - Ragnhild E Paulsen
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Norway
| | - Anna Bal-Price
- European Commission, Joint Research Centre, Ispra, Italy
| | - Hubert Dirven
- Department of Toxicology and Risk Assessment, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
20
|
Pamies D, Block K, Lau P, Gribaldo L, Pardo CA, Barreras P, Smirnova L, Wiersma D, Zhao L, Harris G, Hartung T, Hogberg HT. Rotenone exerts developmental neurotoxicity in a human brain spheroid model. Toxicol Appl Pharmacol 2018; 354:101-114. [PMID: 29428530 DOI: 10.1016/j.taap.2018.02.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 01/22/2018] [Accepted: 02/02/2018] [Indexed: 12/21/2022]
Abstract
Growing concern suggests that some chemicals exert (developmental) neurotoxicity (DNT and NT) and are linked to the increase in incidence of autism, attention deficit and hyperactivity disorders. The high cost of routine tests for DNT and NT assessment make it difficult to test the high numbers of existing chemicals. Thus, more cost effective neurodevelopmental models are needed. The use of induced pluripotent stem cells (iPSC) in combination with the emerging human 3D tissue culture platforms, present a novel tool to predict and study human toxicity. By combining these technologies, we generated multicellular brain spheroids (BrainSpheres) from human iPSC. The model has previously shown to be reproducible and recapitulates several neurodevelopmental features. Our results indicate, rotenone's toxic potency varies depending on the differentiation status of the cells, showing higher reactive oxygen species (ROS) and higher mitochondrial dysfunction during early than later differentiation stages. Immuno-fluorescence morphology analysis after rotenone exposure indicated dopaminergic-neuron selective toxicity at non-cytotoxic concentrations (1 μM), while astrocytes and other neuronal cell types were affected at (general) cytotoxic concentrations (25 μM). Omics analysis showed changes in key pathways necessary for brain development, indicating rotenone as a developmental neurotoxicant and show a possible link between previously shown effects on neurite outgrowth and presently observed effects on Ca2+ reabsorption, synaptogenesis and PPAR pathway disruption. In conclusion, our BrainSpheres model has shown to be a reproducible and novel tool to study neurotoxicity and developmental neurotoxicity. Results presented here support the idea that rotenone can potentially be a developmental neurotoxicant.
Collapse
Affiliation(s)
- David Pamies
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Katharina Block
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Pierre Lau
- European Commission, Joint Research Centre, European Reference Laboratory - European Centre for the Validation of Alternative Methods (EURL ECVAM), Via Enrico Fermi 2749, Ispra, VA 21027, Italy
| | - Laura Gribaldo
- European Commission, Joint Research Centre, European Reference Laboratory - European Centre for the Validation of Alternative Methods (EURL ECVAM), Via Enrico Fermi 2749, Ispra, VA 21027, Italy
| | - Carlos A Pardo
- Department of Neurology, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD 21287, USA
| | - Paula Barreras
- Department of Neurology, Johns Hopkins University, 600 N Wolfe Street, Baltimore, MD 21287, USA
| | - Lena Smirnova
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Daphne Wiersma
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Liang Zhao
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, 650 Orleans Street, CRB1, Rm 464, Baltimore, MD 21287, USA
| | - Georgina Harris
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Thomas Hartung
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA; University of Konstanz, CAAT-Europe, Universitätsstr. 10, Konstanz 78464, Germany
| | - Helena T Hogberg
- Center for Alternative to Animal Testing (CAAT), Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
21
|
Attoff K, Gliga A, Lundqvist J, Norinder U, Forsby A. Whole genome microarray analysis of neural progenitor C17.2 cells during differentiation and validation of 30 neural mRNA biomarkers for estimation of developmental neurotoxicity. PLoS One 2017; 12:e0190066. [PMID: 29261810 PMCID: PMC5738075 DOI: 10.1371/journal.pone.0190066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/07/2017] [Indexed: 01/01/2023] Open
Abstract
Despite its high relevance, developmental neurotoxicity (DNT) is one of the least studied forms of toxicity. Current guidelines for DNT testing are based on in vivo testing and they require extensive resources. Transcriptomic approaches using relevant in vitro models have been suggested as a useful tool for identifying possible DNT-generating compounds. In this study, we performed whole genome microarray analysis on the murine progenitor cell line C17.2 following 5 and 10 days of differentiation. We identified 30 genes that are strongly associated with neural differentiation. The C17.2 cell line can be differentiated into a co-culture of both neurons and neuroglial cells, giving a more relevant picture of the brain than using neuronal cells alone. Among the most highly upregulated genes were genes involved in neurogenesis (CHRDL1), axonal guidance (BMP4), neuronal connectivity (PLXDC2), axonogenesis (RTN4R) and astrocyte differentiation (S100B). The 30 biomarkers were further validated by exposure to non-cytotoxic concentrations of two DNT-inducing compounds (valproic acid and methylmercury) and one neurotoxic chemical possessing a possible DNT activity (acrylamide). Twenty-eight of the 30 biomarkers were altered by at least one of the neurotoxic substances, proving the importance of these biomarkers during differentiation. These results suggest that gene expression profiling using a predefined set of biomarkers could be used as a sensitive tool for initial DNT screening of chemicals. Using a predefined set of mRNA biomarkers, instead of the whole genome, makes this model affordable and high-throughput. The use of such models could help speed up the initial screening of substances, possibly indicating alerts that need to be further studied in more sophisticated models.
Collapse
Affiliation(s)
- Kristina Attoff
- Department of Neurochemistry, Stockholm University, Stockholm, Sweden
- * E-mail:
| | - Anda Gliga
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jessica Lundqvist
- Department of Neurochemistry, Stockholm University, Stockholm, Sweden
- Swetox, Karolinska Institutet, Unit of Toxicology Sciences, Södertälje, Sweden
| | - Ulf Norinder
- Swetox, Karolinska Institutet, Unit of Toxicology Sciences, Södertälje, Sweden
| | - Anna Forsby
- Department of Neurochemistry, Stockholm University, Stockholm, Sweden
- Swetox, Karolinska Institutet, Unit of Toxicology Sciences, Södertälje, Sweden
| |
Collapse
|
22
|
Bal-Price A, Meek MEB. Adverse outcome pathways: Application to enhance mechanistic understanding of neurotoxicity. Pharmacol Ther 2017; 179:84-95. [PMID: 28529068 PMCID: PMC5869951 DOI: 10.1016/j.pharmthera.2017.05.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent developments have prompted the transition of empirically based testing of late stage toxicity in animals for a range of different endpoints including neurotoxicity to more efficient and predictive mechanistically based approaches with greater emphasis on measurable key events early in the progression of disease. The adverse outcome pathway (AOP) has been proposed as a simplified organizational construct to contribute to this transition by linking molecular initiating events and earlier (more predictive) key events at lower levels of biological organization to disease outcomes. As such, AOPs are anticipated to facilitate the compilation of information to increase mechanistic understanding of pathophysiological pathways that are responsible for human disease. In this review, the sequence of key events resulting in adverse outcome (AO) defined as parkinsonian motor impairment and learning and memory deficit in children, triggered by exposure to environmental chemicals has been briefly described using the AOP framework. These AOPs follow convention adopted in an Organization for Economic Cooperation and Development (OECD) AOP development program, publically available, to permit tailored application of AOPs for a range of different purposes. Due to the complexity of disease pathways, including neurodegenerative disorders, a specific symptom of the disease (e.g. parkinsonian motor deficit) is considered as the AO in a developed AOP. Though the description is necessarily limited by the extent of current knowledge, additional characterization of involved pathways through description of related AOPs interlinked into networks for the same disease has potential to contribute to more holistic and mechanistic understanding of the pathophysiological pathways involved, possibly leading to the mechanism-based reclassification of diseases, thus facilitating more personalized treatment.
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission Joint Research Centre, Directorate F - Health, Consumers and Reference Materials, Ispra, Italy.
| | - M E Bette Meek
- McLaughlin Centre for Risk Science, University of Ottawa, Ottawa, Canada
| |
Collapse
|
23
|
|
24
|
Antunes Dos Santos A, Appel Hort M, Culbreth M, López-Granero C, Farina M, Rocha JBT, Aschner M. Methylmercury and brain development: A review of recent literature. J Trace Elem Med Biol 2016; 38:99-107. [PMID: 26987277 PMCID: PMC5011031 DOI: 10.1016/j.jtemb.2016.03.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 03/02/2016] [Indexed: 02/02/2023]
Abstract
Methylmercury (MeHg) is a potent environmental pollutant, which elicits significant toxicity in humans. The central nervous system (CNS) is the primary target of toxicity, and is particularly vulnerable during development. Maternal exposure to MeHg via consumption of fish and seafood can have irreversible effects on the neurobehavioral development of children, even in the absence of symptoms in the mother. It is well documented that developmental MeHg exposure may lead to neurological alterations, including cognitive and motor dysfunction. The neurotoxic effects of MeHg on the developing brain have been extensively studied. The mechanism of toxicity, however, is not fully understood. No single process can explain the multitude of effects observed in MeHg-induced neurotoxicity. This review summarizes the most current knowledge on the effects of MeHg during nervous system development considering both, in vitro and in vivo experimental models. Considerable attention was directed towards the role of glutamate and calcium dyshomeostasis, mitochondrial dysfunction, as well as the effects of MeHg on cytoskeletal components/regulators.
Collapse
Affiliation(s)
| | - Mariana Appel Hort
- Institute of Biological Sciences, Federal University of Rio Grande, Campus Carreiros, Rio Grande do Sul, Brazil
| | - Megan Culbreth
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Caridad López-Granero
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Joao B T Rocha
- Department of Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
25
|
Brown JP, Hall D, Frank CL, Wallace K, Mundy WR, Shafer TJ. Editor's Highlight: Evaluation of a Microelectrode Array-Based Assay for Neural Network Ontogeny Using Training Set Chemicals. Toxicol Sci 2016; 154:126-139. [PMID: 27492221 DOI: 10.1093/toxsci/kfw147] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Thousands of compounds in the environment have not been characterized for developmental neurotoxicity (DNT) hazard. To address this issue, methods to screen compounds rapidly for DNT hazard evaluation are necessary and are being developed for key neurodevelopmental processes. In order to develop an assay for network formation, this study evaluated effects of a training set of chemicals on network ontogeny by measuring spontaneous electrical activity in neural networks grown on microelectrode arrays (MEAs). Rat (0-24 h old) primary cortical cells were plated in 48 well-MEA plates and exposed to 6 compounds: acetaminophen, bisindolylmaleimide-1 (Bis-1), domoic acid, mevastatin, sodium orthovanadate, and loperamide for a period of 12 days. Spontaneous network activity was recorded on days 2, 5, 7, 9, and 12 and viability was assessed using the Cell Titer Blue assay on day 12. Network activity (e.g. mean firing rate [MFR], burst rate [BR], etc), increased between days 5 and 12. Random Forest analysis indicated that across all compounds and times, temporal correlation of firing patterns (r), MFR, BR, number of active electrodes and % of spikes in a burst were the most influential parameters in separating control from treated wells. All compounds except acetaminophen (≤ 30 µM) caused concentration-related effects on one or more of these parameters. Domoic acid and sodium orthovanadate altered several of these parameters in the absence of cytotoxicity. Although cytotoxicity was observed with Bis1, mevastatin, and loperamide, some parameters were affected by these compounds at concentrations below those resulting in cytotoxicity. These results demonstrate that this assay may be suitable for screening of compounds for DNT hazard identification.
Collapse
Affiliation(s)
| | - Diana Hall
- NHEERL, US EPA, Research Triangle Park, NC, USA
| | | | | | | | | |
Collapse
|
26
|
Schmidt BZ, Lehmann M, Gutbier S, Nembo E, Noel S, Smirnova L, Forsby A, Hescheler J, Avci HX, Hartung T, Leist M, Kobolák J, Dinnyés A. In vitro acute and developmental neurotoxicity screening: an overview of cellular platforms and high-throughput technical possibilities. Arch Toxicol 2016; 91:1-33. [PMID: 27492622 DOI: 10.1007/s00204-016-1805-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 07/07/2016] [Indexed: 01/03/2023]
Abstract
Neurotoxicity and developmental neurotoxicity are important issues of chemical hazard assessment. Since the interpretation of animal data and their extrapolation to man is challenging, and the amount of substances with information gaps exceeds present animal testing capacities, there is a big demand for in vitro tests to provide initial information and to prioritize for further evaluation. During the last decade, many in vitro tests emerged. These are based on animal cells, human tumour cell lines, primary cells, immortalized cell lines, embryonic stem cells, or induced pluripotent stem cells. They differ in their read-outs and range from simple viability assays to complex functional endpoints such as neural crest cell migration. Monitoring of toxicological effects on differentiation often requires multiomics approaches, while the acute disturbance of neuronal functions may be analysed by assessing electrophysiological features. Extrapolation from in vitro data to humans requires a deep understanding of the test system biology, of the endpoints used, and of the applicability domains of the tests. Moreover, it is important that these be combined in the right way to assess toxicity. Therefore, knowledge on the advantages and disadvantages of all cellular platforms, endpoints, and analytical methods is essential when establishing in vitro test systems for different aspects of neurotoxicity. The elements of a test, and their evaluation, are discussed here in the context of comprehensive prediction of potential hazardous effects of a compound. We summarize the main cellular characteristics underlying neurotoxicity, present an overview of cellular platforms and read-out combinations assessing distinct parts of acute and developmental neurotoxicology, and highlight especially the use of stem cell-based test systems to close gaps in the available battery of tests.
Collapse
Affiliation(s)
- Béla Z Schmidt
- BioTalentum Ltd., Gödöllő, Hungary.,Stem Cell Biology and Embryology Unit, Department of Development and Regeneration, Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium
| | - Martin Lehmann
- BioTalentum Ltd., Gödöllő, Hungary.,Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Simon Gutbier
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Constance, Germany
| | - Erastus Nembo
- BioTalentum Ltd., Gödöllő, Hungary.,Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Sabrina Noel
- Louvain Centre for Toxicology and Applied Pharmacology, Université Catholique de Louvain, Brussels, Belgium
| | - Lena Smirnova
- Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Anna Forsby
- Swedish Toxicology Research Center (Swetox), Södertälje, Sweden.,Department of Neurochemistry, Stockholm University, Stockholm, Sweden
| | - Jürgen Hescheler
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Hasan X Avci
- BioTalentum Ltd., Gödöllő, Hungary.,Department of Medical Chemistry, University of Szeged, Szeged, Hungary
| | - Thomas Hartung
- Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Marcel Leist
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Constance, Germany
| | | | - András Dinnyés
- BioTalentum Ltd., Gödöllő, Hungary. .,Molecular Animal Biotechnology Laboratory, Szent István University, Gödöllő, 2100, Hungary.
| |
Collapse
|
27
|
Methylmercury-induced developmental toxicity is associated with oxidative stress and cofilin phosphorylation. Cellular and human studies. Neurotoxicology 2016; 59:197-209. [PMID: 27241350 DOI: 10.1016/j.neuro.2016.05.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/13/2016] [Accepted: 05/27/2016] [Indexed: 01/04/2023]
Abstract
Environmental exposure to methylmercury (MeHg) during development is of concern because it is easily incorporated in children's body both pre- and post-natal, it acts at several levels of neural pathways (mitochondria, cytoskeleton, neurotransmission) and it causes behavioral impairment in child. We evaluated the effects of prolonged exposure to 10-600nM MeHg on primary cultures of mouse cortical (CCN) and of cerebellar granule cells (CGC) during their differentiation period. In addition, it was studied if prenatal MeHg exposure correlated with altered antioxidant defenses and cofilin phosphorylation in human placentas (n=12) from the INMA cohort (Spain). Exposure to MeHg for 9days in vitro (DIV) resulted in protein carbonylation and in cell death at concentrations ≥200nM and ≥300nM, respectively. Exposure of CCN and CGC to non-cytotoxic MeHg concentrations for 5 DIV induced an early concentration-dependent decrease in cofilin phosphorylation. Furthermore, in both cell types actin was translocated from the cytosol to the mitochondria whereas cofilin translocation was found only in CGC. Translocation of cofilin and actin to mitochondria in CGC occurred from 30nM MeHg onwards. We also found an increased expression of cortactin and LIMK1 mRNA in CGC but not in CCN. All these effects were prevented by the antioxidant probucol. Cofilin phosphorylation was significantly decreased and a trend for decreased activity of glutathione reductase and glutathione peroxidase was found in the fetal side of human placental samples from the highest (20-40μg/L) MeHg-exposed group when compared with the low (<7μg/L) MeHg-exposed group. In summary, cofilin dephosphorylation and oxidative stress are hallmarks of MeHg exposure in both experimental and human systems.
Collapse
|
28
|
Jyoti S, Tandon S. Impact of homeopathic remedies on the expression of lineage differentiation genes: an in vitro approach using embryonic stem cells. HOMEOPATHY 2015; 105:148-59. [PMID: 27211322 DOI: 10.1016/j.homp.2015.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 10/01/2015] [Accepted: 11/19/2015] [Indexed: 11/18/2022]
Abstract
BACKGROUND Well-documented studies of the potential effects and safety of homeopathic medicines in pregnancy are required. In this study, specific genes were studied which could serve as biomarkers for specification of three lineages to predict the safety of homeopathic remedies using mouse embryonic stem (ES) cells. Thus, the present work was to study the effects of homeopathic remedies taken during pregnancy using ES cells as the model. METHODS Mouse ES cells were exposed to 30C potency of Nux Vomica and Sepia, which are homeopathic medicines prescribed for the management of pregnancy related symptoms. Cytotoxicity studies were done using a modified Embryonic Stem cell test (EST). The expression levels of key genes and proteins were analyzed using real time polymerase chain reaction and immunocytochemistry, respectively. RESULTS Homeopathic treatment led to modulations in the expression of certain lineage specific genes but this difference was not significant with respect to solvent control and showed normal differentiation as demonstrated by the expression of α/β MHC and α-actinin proteins in the differentiated ES cells. CONCLUSIONS Our study for the first time has shown the feasibility of using ES cells in the developmental toxicity testing of remedies. The results suggest that they are not associated with developmental toxicity.
Collapse
Affiliation(s)
- Saras Jyoti
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, 173234, India.
| | - Simran Tandon
- Amity University Uttar Pradesh, Sector 125, Noida, 201313, UP, India.
| |
Collapse
|
29
|
Qasemian Lemraski M, Soodi M, Fakhr Taha M, Zarei MH, Jafarzade E. Study of lead-induced neurotoxicity in neural cells differentiated from adipose tissue-derived stem cells. Toxicol Mech Methods 2015; 25:128-35. [DOI: 10.3109/15376516.2014.997949] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
30
|
Jyoti S, Tandon S. Genetic basis for developmental toxicity due to statin intake using embryonic stem cell differentiation model. Hum Exp Toxicol 2015; 34:965-84. [PMID: 25712412 DOI: 10.1177/0960327114564795] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The in utero environment is a key factor controlling the fate of the growing embryo. The deleterious effects of statins during the fetal development are still not very well understood. Data from animal studies and retrospective studies performed in pregnant women give conflicting reports. In this study, using in vitro differentiation model of embryonic stem cells, which mimic the differentiation process of the embryo, we have systematically exposed the cells to lipophilic statins, simvastatin, and atorvastatin at various doses and at critical times during differentiation. The analysis of key genes controlling the differentiation into ecto-, meso- and endodermal lineages was assessed by quantitative polymerase chain reaction. Our results show that genes of the mesodermal lineage were most sensitive to statins, leading to changes in the transcript levels of brachyury, Flk-1, Nkx2.5, and α/β-myosin heavy chain. In addition, changes to endodermal marker α-fetoprotein, along with ectodermal Nes and Neurofilament 200 kDa, imply that during early differentiation exposure to these drugs leads to altered signaling, which could translate to the congenital abnormalities seen in the heart and limbs.
Collapse
Affiliation(s)
- S Jyoti
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Biotechnology & Bioinformatics, Solan, India
| | - S Tandon
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Biotechnology & Bioinformatics, Solan, India
| |
Collapse
|
31
|
Smirnova L, Hogberg HT, Leist M, Hartung T. Developmental neurotoxicity - challenges in the 21st century and in vitro opportunities. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2015; 31:129-56. [PMID: 24687333 DOI: 10.14573/altex.1403271] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 03/28/2014] [Indexed: 11/23/2022]
Abstract
In recent years neurodevelopmental problems in children have increased at a rate that suggests lifestyle factors and chemical exposures as likely contributors. When environmental chemicals contribute to neurodevelopmental disorders developmental neurotoxicity (DNT) becomes an enormous concern. But how can it be tackled? Current animal test- based guidelines are prohibitively expensive, at $ 1.4 million per substance, while their predictivity for human health effects may be limited, and mechanistic data that would help species extrapolation are not available. A broader screening for substances of concern requires a reliable testing strategy, applicable to larger numbers of substances, and sufficiently predictive to warrant further testing. This review discusses the evidence for possible contributions of environmental chemicals to DNT, limitations of the current test paradigm, emerging concepts and technologies pertinent to in vitro DNT testing and assay evaluation, as well as the prospect of a paradigm shift based on 21st century technologies.
Collapse
Affiliation(s)
- Lena Smirnova
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, USA
| | | | | | | |
Collapse
|
32
|
Barbosa DJ, Capela JP, de Lourdes Bastos M, Carvalho F. In vitro models for neurotoxicology research. Toxicol Res (Camb) 2015; 4:801-842. [DOI: 10.1039/c4tx00043a] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
The nervous system has a highly complex organization, including many cell types with multiple functions, with an intricate anatomy and unique structural and functional characteristics; the study of its (dys)functionality following exposure to xenobiotics, neurotoxicology, constitutes an important issue in neurosciences.
Collapse
Affiliation(s)
- Daniel José Barbosa
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - João Paulo Capela
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - Maria de Lourdes Bastos
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - Félix Carvalho
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| |
Collapse
|
33
|
An integrated approach for detecting embryotoxicity and developmental toxicity of environmental contaminants using in vitro alternative methods. Toxicol Lett 2014; 230:356-67. [DOI: 10.1016/j.toxlet.2014.01.037] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 12/10/2013] [Accepted: 01/27/2014] [Indexed: 11/19/2022]
|
34
|
Early life arsenic exposure and brain dopaminergic alterations in rats. Int J Dev Neurosci 2014; 38:91-104. [DOI: 10.1016/j.ijdevneu.2014.08.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/09/2014] [Accepted: 08/23/2014] [Indexed: 11/21/2022] Open
|
35
|
Estevan C, Fuster E, Del Río E, Pamies D, Vilanova E, Sogorb MA. Organophosphorus pesticide chlorpyrifos and its metabolites alter the expression of biomarker genes of differentiation in D3 mouse embryonic stem cells in a comparable way to other model neurodevelopmental toxicants. Chem Res Toxicol 2014; 27:1487-95. [PMID: 25137620 DOI: 10.1021/tx500051k] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
There are discrepancies about whether chlorpyrifos is able to induce neurodevelopmental toxicity or not. We previously reported alterations in the pattern of expression of biomarker genes of differentiation in D3 mouse embryonic stem cells caused by chlorpyrifos and its metabolites chlorpyrifos-oxon and 3,5,6-trichloro-2-pyridinol. Now, we reanalyze these data comparing the effects on these genes with those caused in the same genes by retinoic acid, valproic acid, and penicillin-G (model compounds considered as strong, weak, and non-neurodevelopmental toxicants, respectively). We also compare the effects of chlorpyrifos and its metabolites on the cell viability of D3 cells and 3T3 mouse fibroblasts with the effects caused in the same cells by the three model compounds. We conclude that chlorpyrifos and its metabolites act, regarding these end-points, as the weak neurodevelopmental toxicant valproic acid, and consequently, a principle of caution should be applied avoiding occupational exposures in pregnant women. A second independent experiment run with different cellular batches coming from the same clone obtained the same result as the first one.
Collapse
Affiliation(s)
- Carmen Estevan
- Unidad de Toxicología y Seguridad Química, Instituto de Bioingeniería, Universidad Miguel Hernández de Elche , Avenida de la Universidad s/n, 03202-Elche, Spain
| | | | | | | | | | | |
Collapse
|
36
|
Changes in miRNA Expression Profiling during Neuronal Differentiation and Methyl Mercury-Induced Toxicity in Human in Vitro Models. TOXICS 2014. [DOI: 10.3390/toxics2030443] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
37
|
Pamies D, Hartung T, Hogberg HT. Biological and medical applications of a brain-on-a-chip. Exp Biol Med (Maywood) 2014; 239:1096-1107. [PMID: 24912505 DOI: 10.1177/1535370214537738] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The desire to develop and evaluate drugs as potential countermeasures for biological and chemical threats requires test systems that can also substitute for the clinical trials normally crucial for drug development. Current animal models have limited predictivity for drug efficacy in humans as the large majority of drugs fails in clinical trials. We have limited understanding of the function of the central nervous system and the complexity of the brain, especially during development and neuronal plasticity. Simple in vitro systems do not represent physiology and function of the brain. Moreover, the difficulty of studying interactions between human genetics and environmental factors leads to lack of knowledge about the events that induce neurological diseases. Microphysiological systems (MPS) promise to generate more complex in vitro human models that better simulate the organ's biology and function. MPS combine different cell types in a specific three-dimensional (3D) configuration to simulate organs with a concrete function. The final aim of these MPS is to combine different "organoids" to generate a human-on-a-chip, an approach that would allow studies of complex physiological organ interactions. The recent discovery of induced pluripotent stem cells (iPSCs) gives a range of possibilities allowing cellular studies of individuals with different genetic backgrounds (e.g., human disease models). Application of iPSCs from different donors in MPS gives the opportunity to better understand mechanisms of the disease and can be a novel tool in drug development, toxicology, and medicine. In order to generate a brain-on-a-chip, we have established a 3D model from human iPSCs based on our experience with a 3D rat primary aggregating brain model. After four weeks of differentiation, human 3D aggregates stain positive for different neuronal markers and show higher gene expression of various neuronal differentiation markers compared to 2D cultures. Here we present the applications and challenges of this emerging technology.
Collapse
Affiliation(s)
- David Pamies
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA; University of Konstanz, POB 600, Konstanz 78457, Germany
| | - Thomas Hartung
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA; University of Konstanz, POB 600, Konstanz 78457, Germany
| | - Helena T Hogberg
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA; University of Konstanz, POB 600, Konstanz 78457, Germany
| |
Collapse
|
38
|
Verrotti A, Scaparrotta A, Cofini M, Chiarelli F, Tiboni GM. Developmental neurotoxicity and anticonvulsant drugs: a possible link. Reprod Toxicol 2014; 48:72-80. [PMID: 24803404 DOI: 10.1016/j.reprotox.2014.04.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/05/2014] [Accepted: 04/18/2014] [Indexed: 01/16/2023]
Abstract
In utero exposure to antiepileptic drugs (AEDs) may affect neurodevelopment causing postnatal cognitive and behavioral alterations. Phenytoin and phenobarbital may lead to motor and learning dysfunctions in the pre-exposed children. These disorders may reflect the interference of these AEDs with the development of hippocampal and cerebellar neurons, as suggested by animal studies. Exposure to valproic acid may result in inhibition of neural stem cell proliferation and/or immature neuron migration in the cerebral cortex with consequent increased risk of neurodevelopmental impairment, such as autistic spectrum disorders. A central issue in the prevention of AED-mediated developmental effects is the identification of drugs that should be avoided in women of child-bearing potential and during pregnancy. The aim of this review is to explore the possible link between AEDs and neurodevelopmental dysfunctions both in human and in animal studies. The possible mechanisms underlying this association are also discussed.
Collapse
Affiliation(s)
- A Verrotti
- Department of Pediatrics, University of Perugia, Italy
| | - A Scaparrotta
- Department of Medicine and Aging Sciences, University "G. d'Annunzio" of Chieti-Pescara, Italy
| | - M Cofini
- Department of Pediatrics, University of Perugia, Italy
| | - F Chiarelli
- Department of Medicine and Aging Sciences, University "G. d'Annunzio" of Chieti-Pescara, Italy
| | - G M Tiboni
- Department of Medicine and Aging Sciences, University "G. d'Annunzio" of Chieti-Pescara, Italy.
| |
Collapse
|
39
|
Dueñas-Gonzalez A, Alatorre B, Gonzalez-Fierro A. The impact of DNA methylation technologies on drug toxicology. Expert Opin Drug Metab Toxicol 2014; 10:637-646. [PMID: 24660662 DOI: 10.1517/17425255.2014.889682] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Drug toxicology is central to drug development. Despite improvements in our understanding of molecular and cell biology, high attrition rates in drug development continue, speaking to the difficulties of developing unequivocal methods to predict the efficacy and safety of drugs. AREAS COVERED In this review, the authors provide a short overview of the 'omics' technologies that have been applied to drug toxicology, with an emphasis on a whole-genome DNA methylation analysis. Preliminary results from DNA methylation analysis technologies that may help in predicting response and efficacy of a drug are discussed. EXPERT OPINION Currently, we cannot fully contextualize the application of epigenetics to the field of drug toxicology, as there are still many challenges to overcome before DNA methylation-based biomarkers can be effectively used in drug development. Comprehensive whole-genome DNA methylation methods for a unbiased analysis based on either microarray or next-generation sequencing need to be evaluated in drug toxicology in an intensive and systematic manner. Additionally, robust analysis systems need to be developed to decode the large amounts of data generated by whole-genome DNA methylation analyses as well as protocol standardization for reproducibility to develop meaningful databases that can be applied to drug toxicology.
Collapse
Affiliation(s)
- Alfonso Dueñas-Gonzalez
- Universidad Nacional Autonoma de Mexico UNAM/Instituto Nacional de Cancerologia, Unidad de Investigacion Biomedica en Cancer, Insituto de Investigaciones Biomedicas , San Fernando 22, Tlalpan, Mexico City , Mexico
| | | | | |
Collapse
|
40
|
|
41
|
Toxicity of TDCPP and TCEP on PC12 cell: changes in CAMKII, GAP43, tubulin and NF-H gene and protein levels. Toxicol Lett 2014; 227:164-71. [PMID: 24717766 DOI: 10.1016/j.toxlet.2014.03.023] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 03/27/2014] [Accepted: 03/28/2014] [Indexed: 11/21/2022]
Abstract
TDCPP and TCEP are two major types of organophosphorus flame retardants (OPFRs) that are bioaccumulative and persistent in the environment. The toxicity effects of TDCPP and TCEP on PC12 cell are not well understood. In the present study, we investigated morphology, viability and apoptosis in cultured PC12 cells in response to TDCPP and TCEP. The mRNA and protein expression levels of CAMKII, GAP43, tubulin and NF-H were quantified in PC12 cells treated with varying concentrations of the two agents. Results indicate that, upon treatment with the two OPFRs, cell growth decreased, apoptosis increased, morphology was altered and significant changes were found in the gene and protein levels. Treatment with TDCPP caused a reduction in the levels of each of the six proteins studied and in the gene levels of GAP43, NF-H and the two tubulins, but it resulted in an increase in CAMKII gene levels. Treatment with TCEP resulted in similar changes in gene levels to TDCPP and led to decreases in the protein levels of GAP43 and the tubulins while increasing the CAMKII and NF-H protein levels. These results suggest that changes in the gene and protein levels of the regulatory proteins (CAMKII, GAP43) and the structural proteins (tubulin, NF-H) are due to different mechanisms of the toxins, and these proteins may be useful biomarkers for the cytotoxicity and neurotoxicity.
Collapse
|
42
|
Zimmer B, Pallocca G, Dreser N, Foerster S, Waldmann T, Westerhout J, Julien S, Krause KH, van Thriel C, Hengstler JG, Sachinidis A, Bosgra S, Leist M. Profiling of drugs and environmental chemicals for functional impairment of neural crest migration in a novel stem cell-based test battery. Arch Toxicol 2014; 88:1109-26. [PMID: 24691702 PMCID: PMC3996367 DOI: 10.1007/s00204-014-1231-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 03/18/2014] [Indexed: 11/28/2022]
Abstract
Developmental toxicity in vitro assays have hitherto been established as stand-alone systems, based on a limited number of toxicants. Within the embryonic stem cell-based novel alternative tests project, we developed a test battery framework that allows inclusion of any developmental toxicity assay and that explores the responses of such test systems to a wide range of drug-like compounds. We selected 28 compounds, including several biologics (e.g., erythropoietin), classical pharmaceuticals (e.g., roflumilast) and also six environmental toxicants. The chemical, toxicological and clinical data of this screen library were compiled. In order to determine a non-cytotoxic concentration range, cytotoxicity data were obtained for all compounds from HEK293 cells and from murine embryonic stem cells. Moreover, an estimate of relevant exposures was provided by literature data mining. To evaluate feasibility of the suggested test framework, we selected a well-characterized assay that evaluates ‘migration inhibition of neural crest cells.’ Screening at the highest non-cytotoxic concentration resulted in 11 hits (e.g., geldanamycin, abiraterone, gefitinib, chlorpromazine, cyproconazole, arsenite). These were confirmed in concentration–response studies. Subsequent pharmacokinetic modeling indicated that triadimefon exerted its effects at concentrations relevant to the in vivo situation, and also interferon-β and polybrominated diphenyl ether showed effects within the same order of magnitude of concentrations that may be reached in humans. In conclusion, the test battery framework can identify compounds that disturb processes relevant for human development and therefore may represent developmental toxicants. The open structure of the strategy allows rich information to be generated on both the underlying library, and on any contributing assay.
Collapse
Affiliation(s)
- B Zimmer
- Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York City, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Balmer NV, Leist M. Epigenetics and transcriptomics to detect adverse drug effects in model systems of human development. Basic Clin Pharmacol Toxicol 2014; 115:59-68. [PMID: 24476462 DOI: 10.1111/bcpt.12203] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 01/16/2014] [Indexed: 01/01/2023]
Abstract
Prenatal exposure to environmental chemicals or drugs has been associated with functional or structural deficits and the development of diseases in later life. For example, developmental neurotoxicity (DNT) is triggered by lead, and this compound may predispose to neurodegenerative diseases in later life. The molecular memory for such late consequences of early exposure is not known, but epigenetic mechanisms (modification of the chromatin structure) could take this role. Examples and underlying mechanisms have been compiled here for the field of DNT. Moreover, we addressed the question as to what readout is suitable for addressing drug memory effects. We summarize how complex developmental processes can be modelled in vitro by using the differentiation of human stem cells. Although cellular models can never replicate the final human DNT phenotype, they can model the adverse effect that a chemical has on key biological processes essential for organ formation and function. Highly information-rich transcriptomics data may inform on these changes and form the bridge from in vitro models to human prediction. We compiled data showing that transcriptome analysis can indicate toxicity patterns of drugs. A crucial question to be answered in our systems is when and how transcriptome changes indicate adversity (as opposed to transient adaptive responses), and how drug-induced changes are perpetuated over time even after washout of the drug. We present evidence for the hypothesis that changes in the histone methylation pattern could represent the persistence detector of an early insult that is transformed to an adverse effect at later time-points in life.
Collapse
Affiliation(s)
- Nina V Balmer
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | | |
Collapse
|
44
|
Hogberg HT, Bressler J, Christian KM, Harris G, Makri G, O'Driscoll C, Pamies D, Smirnova L, Wen Z, Hartung T. Toward a 3D model of human brain development for studying gene/environment interactions. Stem Cell Res Ther 2013; 4 Suppl 1:S4. [PMID: 24564953 PMCID: PMC4029162 DOI: 10.1186/scrt365] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
This project aims to establish and characterize an in vitro model of the developing human brain for the purpose of testing drugs and chemicals. To accurately assess risk, a model needs to recapitulate the complex interactions between different types of glial cells and neurons in a three-dimensional platform. Moreover, human cells are preferred over cells from rodents to eliminate cross-species differences in sensitivity to chemicals. Previously, we established conditions to culture rat primary cells as three-dimensional aggregates, which will be humanized and evaluated here with induced pluripotent stem cells (iPSCs). The use of iPSCs allows us to address gene/environment interactions as well as the potential of chemicals to interfere with epigenetic mechanisms. Additionally, iPSCs afford us the opportunity to study the effect of chemicals during very early stages of brain development. It is well recognized that assays for testing toxicity in the developing brain must consider differences in sensitivity and susceptibility that arise depending on the time of exposure. This model will reflect critical developmental processes such as proliferation, differentiation, lineage specification, migration, axonal growth, dendritic arborization and synaptogenesis, which will probably display differences in sensitivity to different types of chemicals. Functional endpoints will evaluate the complex cell-to-cell interactions that are affected in neurodevelopment through chemical perturbation, and the efficacy of drug intervention to prevent or reverse phenotypes. The model described is designed to assess developmental neurotoxicity effects on unique processes occurring during human brain development by leveraging human iPSCs from diverse genetic backgrounds, which can be differentiated into different cell types of the central nervous system. Our goal is to demonstrate the feasibility of the personalized model using iPSCs derived from individuals with neurodevelopmental disorders caused by known mutations and chromosomal aberrations. Notably, such a human brain model will be a versatile tool for more complex testing platforms and strategies as well as research into central nervous system physiology and pathology.
Collapse
Affiliation(s)
- Helena T Hogberg
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA and University of Konstanz, POB 600, 78457 Konstanz, Germany
| | - Joseph Bressler
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA and University of Konstanz, POB 600, 78457 Konstanz, Germany
- Hugo Moser Institute at the Kennedy Krieger, Johns Hopkins University, Bloomberg School of Public Health, 707 N. Broadway, Baltimore, MD 21205, USA
| | - Kimberly M Christian
- Institute for Cell Engineering, Department of Neurology, Johns Hopkins University, School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Georgina Harris
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA and University of Konstanz, POB 600, 78457 Konstanz, Germany
| | - Georgia Makri
- Institute for Cell Engineering, Department of Neurology, Johns Hopkins University, School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Cliona O'Driscoll
- Hugo Moser Institute at the Kennedy Krieger, Johns Hopkins University, Bloomberg School of Public Health, 707 N. Broadway, Baltimore, MD 21205, USA
| | - David Pamies
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA and University of Konstanz, POB 600, 78457 Konstanz, Germany
| | - Lena Smirnova
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA and University of Konstanz, POB 600, 78457 Konstanz, Germany
| | - Zhexing Wen
- Institute for Cell Engineering, Department of Neurology, Johns Hopkins University, School of Medicine, 733 N. Broadway, Baltimore, MD 21205, USA
| | - Thomas Hartung
- Center for Alternatives to Animal Testing, Johns Hopkins University, Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA and University of Konstanz, POB 600, 78457 Konstanz, Germany
| |
Collapse
|
45
|
Influence of prenatal exposure to environmental pollutants on human cord blood levels of glutamate. Neurotoxicology 2013; 40:102-10. [PMID: 24361731 DOI: 10.1016/j.neuro.2013.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 12/09/2013] [Accepted: 12/10/2013] [Indexed: 11/21/2022]
Abstract
Some chemicals released into the environment, including mercury and some organochlorine compounds (OCs), are suspected to have a key role on subclinical brain dysfunction in childhood. Alteration of the glutamatergic system may be one mechanistic pathway. We aimed to determine whether mercury and seven OCs, including PCBs 138, 153, and 180, DDT and DDE, hexachlorobenzene (HCB), and beta-hexachlorocyclohexane (β-HCH) influence the cord levels of two excitatory amino acids, glutamate and aspartate. Second, we evaluated if this association was mediated by glutamate uptake measured in human placental membranes. The study sample included 40 newborns from a Spanish cohort selected according to cord mercury levels. We determined the content of both amino acids in cord blood samples by means of HPLC and assessed their associations with the contaminants using linear regression analyses, and the effect of the contaminants on glutamate uptake by means of [(3)H]-aspartate binding in human placenta samples. PCB138, β-HCH, and the sum of the three PCBs and seven OCs showed a significant negative association with glutamate levels (decrease of 51, 24, 56 and 54%, respectively, in glutamate levels for each 10-fold increase in the contaminant concentration). Mercury did not show a significant correlation neither with glutamate nor aspartate levels in cord blood, however a compensatory effect between T-Hg and both PCB138, and 4,4'-DDE was observed. The organo-metallic derivative methylmercury completely inhibited glutamate uptake in placenta while PCB138 and β-HCH partially inhibited it (IC50 values: 4.9±0.8 μM, 14.2±1.2 nM and 6.9±2.9 nM, respectively). We conclude that some environmental toxicants may alter the glutamate content in the umbilical cord blood, which might underlie alterations in human development.
Collapse
|
46
|
Scientific Opinion on the developmental neurotoxicity potential of acetamiprid and imidacloprid. EFSA J 2013. [DOI: 10.2903/j.efsa.2013.3471] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
47
|
Kang SJ, Jeong SH, Kim EJ, Park YI, Park SW, Shin HS, Son SW, Kang HG. Toxic effects of methylmercury, arsanilic acid and danofloxacin on the differentiation of mouse embryonic stem cells into neural cells. J Vet Sci 2013; 15:61-71. [PMID: 24136205 PMCID: PMC3973767 DOI: 10.4142/jvs.2014.15.1.61] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 10/06/2013] [Indexed: 11/20/2022] Open
Abstract
This study was performed to assess the neurotoxic effects of methylmercury, arsanilic acid and danofloxacin by quantification of neural-specific proteins in vitro. Quantitation of the protein markers during 14 days of differentiation indicated that the mouse ESCs were completely differentiated into neural cells by Day 8. The cells were treated with non-cytotoxic concentrations of three chemicals during differentiation. Low levels of exposure to methylmercury decreased the expression of GABAA-R and Nestin during the differentiating stage, and Nestin during the differentiated stage. In contrast, GFAP, Tuj1, and MAP2 expression was affected only by relatively high doses during both stages. Arsanilic acid affected the levels of GABAA-R and GFAP during the differentiated stage while the changes of Nestin and Tuj1 were greater during the differentiating stage. For the neural markers (except Nestin) expressed during both stages, danofloxacin affected protein levels at lower concentrations in the differentiated stage than the differentiating stage. Acetylcholinesterase activity was inhibited by relatively low concentrations of methylmercury and arsanilic acid during the differentiating stage while this activity was inhibited only by more than 40 μM of danofloxacin in the differentiated stage. Our results provide useful information about the different toxicities of chemicals and the impact on neural development.
Collapse
Affiliation(s)
- Seok-Jin Kang
- Toxicology and Residue Chemistry Division, Animal and Plant Quarantine Agency, Anyang 430-824, Korea
| | | | | | | | | | | | | | | |
Collapse
|
48
|
miRNA expression profiling in a human stem cell-based model as a tool for developmental neurotoxicity testing. Cell Biol Toxicol 2013; 29:239-57. [PMID: 23903816 DOI: 10.1007/s10565-013-9250-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 06/20/2013] [Indexed: 10/26/2022]
Abstract
The main aim of this study was to evaluate whether microRNA (miRNA) profiling could be a useful tool for in vitro developmental neurotoxicity (DNT) testing. Therefore, to identify the possible DNT biomarkers among miRNAs, we have studied the changes in miRNA expressions in a mixed neuronal/glial culture derived from carcinoma pluripotent stem cells (NT2 cell line) after exposure to methyl mercury chloride (MeHgCl) during the process of neuronal differentiation (2-36 days in vitro (DIV1)). The neuronal differentiation triggered by exposure to retinoic acid (RA) was characterized in the control culture by mRNA expression analysis of neuronal specific markers such as MAP2, NF-200, Tubulin βIII, MAPT-tau, synaptophysin as well as excitatory (NMDA, AMPA) and inhibitory (GABA) receptors. The results obtained from the miRNA expression analysis have identified the presence of a miRNA signature which is specific for neural differentiation in the control culture and another for the response to MeHgCl-induced toxicity. In differentiated neuronal control cultures, we observed the downregulation of the stemness phenotype-linked miR-302 cluster and the overexpression of several miRNAs specific for neuronal differentiation (e.g. let-7, miR-125b and miR-132). In the cultures exposed to MeHgCl (400 nM), we observed an overexpression of a signature composed of five miRNAs (miR-302b, miR-367, miR-372, miR-196b and miR-141) that are known to be involved in the regulation of developmental processes and cellular stress response mechanisms. Using gene ontology term and pathway enrichment analysis of the validated targets of the miRNAs deregulated by the toxic treatment, the possible effect of MeHgCl exposure on signalling pathways involved in axon guidance and learning and memory processes was revealed. The obtained data suggest that miRNA profiling could provide simplified functional evaluation of the toxicity pathways involved in developmental neurotoxicity in comparison with the transcriptomics studies.
Collapse
|
49
|
Croes K, De Coster S, De Galan S, Morrens B, Loots I, Van de Mieroop E, Nelen V, Sioen I, Bruckers L, Nawrot T, Colles A, Den Hond E, Schoeters G, van Larebeke N, Baeyens W, Gao Y. Health effects in the Flemish population in relation to low levels of mercury exposure: from organ to transcriptome level. Int J Hyg Environ Health 2013; 217:239-47. [PMID: 23920476 DOI: 10.1016/j.ijheh.2013.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 04/29/2013] [Accepted: 06/04/2013] [Indexed: 11/30/2022]
Abstract
Due to possible health risks, quantification of mercury accumulation in humans was included in the Flemish biomonitoring programmes FLEHS I (2002-2006) and FLEHS II (2007-2011). The general objective of FLEHS I was to assess regional exposure levels in order to link possible differences in these internal exposure levels to different types of local environmental pressure. Therefore, Hg and MMHg (methylmercury) were only measured in pooled blood samples per region and per age class. In FLEHS II, mercury concentrations were measured in hair of each participant. About 200 adolescents and 250 mothers (reference group) and two times 200 adolescents (2 hotspots) were screened. The main objectives of the FLEHS II study were: (1) to determine reference levels of mercury in hair for Flanders; (2) to assess relations between mercury exposure and possible sources like fish consumption; (3) to assess dose-effect relations between mercury exposure and health effect markers. The results showed that mercury concentrations in the Flemish population were rather low compared to other studies. Mercury levels in the Flemish populations were strongly related to the age of the participants and consumption of fish. Significant negative associations were observed between mercury in hair and asthma, having received breast feeding as a newborn, age at menarche in girls, allergy for animals and free testosterone levels. Significant correlations were also observed between mercury in hair and genes JAK2, ARID4A, Hist1HA4L (boys) and HLAdrb5, PIAS2, MANN1B1, GIT and ABCA1 (girls).
Collapse
Affiliation(s)
- Kim Croes
- Free University of Brussels (VUB), Department of Analytical and Environmental Chemistry (ANCH), Pleinlaan 2, 1050 Brussels, Belgium
| | - Sam De Coster
- Ghent University Hospital, Study Centre for Carcinogenesis and Primary Prevention of Cancer, De Pintelaan 185, 9000 Ghent, Belgium
| | - Sandra De Galan
- Free University of Brussels (VUB), Department of Analytical and Environmental Chemistry (ANCH), Pleinlaan 2, 1050 Brussels, Belgium
| | - Bert Morrens
- University of Antwerp, Faculty of Political and Social Sciences, Department of Sociology, Sint Jacobstraat 2, 2000 Antwerp, Belgium
| | - Ilse Loots
- University of Antwerp, Faculty of Political and Social Sciences, Department of Sociology, Sint Jacobstraat 2, 2000 Antwerp, Belgium
| | - Els Van de Mieroop
- Provincial Institute for Hygiene, Kronenburgstraat 45, 2000 Antwerp, Belgium
| | - Vera Nelen
- Provincial Institute for Hygiene, Kronenburgstraat 45, 2000 Antwerp, Belgium
| | - Isabelle Sioen
- Ghent University, Department of Public Health, UZ-2 Blok A, De Pintelaan 185, 9000 Ghent, Belgium; Research Foundation - Flanders, Egmontstraat 5, 1000 Brussels, Belgium
| | - Liesbeth Bruckers
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Martelarenlaan 42, 3500 Hasselt, Belgium
| | - Tim Nawrot
- School of Public Health, Occupational & Environmental Medicine, K.U. Leuven, Herestraat 49 (O&N 706), 3000 Leuven, Belgium; Centre for Environmental Sciences, Hasselt University, Agoralaan Gebouw D, 3590 Diepenbeek, Belgium
| | - Ann Colles
- Flemish Institute for Technological Research (VITO), Environmental Health and Risk, Boeretang 200, 2400 Mol, Belgium
| | - Elly Den Hond
- Flemish Institute for Technological Research (VITO), Environmental Health and Risk, Boeretang 200, 2400 Mol, Belgium
| | - Greet Schoeters
- Flemish Institute for Technological Research (VITO), Environmental Health and Risk, Boeretang 200, 2400 Mol, Belgium
| | - Nicolas van Larebeke
- Ghent University Hospital, Study Centre for Carcinogenesis and Primary Prevention of Cancer, De Pintelaan 185, 9000 Ghent, Belgium
| | - Willy Baeyens
- Free University of Brussels (VUB), Department of Analytical and Environmental Chemistry (ANCH), Pleinlaan 2, 1050 Brussels, Belgium
| | - Yue Gao
- Free University of Brussels (VUB), Department of Analytical and Environmental Chemistry (ANCH), Pleinlaan 2, 1050 Brussels, Belgium.
| |
Collapse
|
50
|
A human pluripotent carcinoma stem cell-based model for in vitro developmental neurotoxicity testing: effects of methylmercury, lead and aluminum evaluated by gene expression studies. Int J Dev Neurosci 2013; 31:679-91. [PMID: 23501475 DOI: 10.1016/j.ijdevneu.2013.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 01/24/2013] [Accepted: 03/03/2013] [Indexed: 12/23/2022] Open
Abstract
The major advantage of the neuronal cell culture models derived from human stem cells is their ability to replicate the crucial stages of neurodevelopment such as the commitment of human stem cells to the neuronal lineage and their subsequent stages of differentiation into neuronal and glial-like cell. In these studies we used mixed neuronal/glial culture derived from the NTERA-2 (NT-2) cell line, which has been established from human pluripotent testicular embryonal carcinoma cells. After characterization of the different stages of cell differentiation into neuronal- and glial-like phenotype toxicity studies were performed to evaluate whether this model would be suitable for developmental neurotoxicity studies. The cells were exposed during the differentiation process to non-cytotoxic concentrations of methylmercury chloride, lead chloride and aluminum nitrate for two weeks. The toxicity was then evaluated by measuring the mRNA levels of cell specific markers (neuronal and glial). The results obtained suggest that lead chloride and aluminum nitrate at low concentrations were toxic primarily to astrocytes and at the higher concentrations it also induced neurotoxicity. In contrast, MetHgCl was toxic for both cell types, neuronal and glial, as mRNA specific for astrocytes and neuronal markers were affected. The results obtained suggest that a neuronal mixed culture derived from human NT2 precursor cells is a suitable model for developmental neurotoxicity studies and gene expression could be used as a sensitive endpoint for initial screening of potential neurotoxic compounds.
Collapse
|