1
|
Hu B, Cheng Z, Liang S. Advantages and prospects of stem cells in nanotoxicology. CHEMOSPHERE 2022; 291:132861. [PMID: 34774913 DOI: 10.1016/j.chemosphere.2021.132861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 06/13/2023]
Abstract
Nanomaterials have been widely used in many fields, especially in biomedical and stem cell therapy. However, the potential risks associated with nanomaterials applications are also gradually increasing. Therefore, effective and robust toxicology models are critical to evaluate the developmental toxicity of nanomaterials. The development of stem cell research provides a new idea of developmental toxicology. Recently, many researchers actively investigated the effects of nanomaterials with different sizes and surface modifications on various stem cells (such as embryonic stem cells (ESCs), adult stem cells, etc.) to study the toxic effects and toxic mechanisms. In this review, we summarized the effects of nanomaterials on the proliferation and differentiation of ESCs, mesenchymal stem cells and neural stem cells. Moreover, we discussed the advantages of stem cells in nanotoxicology compared with other cell lines. Finally, combined with the latest research methods and new molecular mechanisms, we analyzed the application of stem cells in nanotoxicology.
Collapse
Affiliation(s)
- Bowen Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang, 830017, China.
| | - Zhanwen Cheng
- School of Environmental Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Shengxian Liang
- Institute of Life Sciences and Green Development, College of Life Sciences, Hebei University, Baoding, 071000, China
| |
Collapse
|
2
|
Tirumala MG, Anchi P, Raja S, Rachamalla M, Godugu C. Novel Methods and Approaches for Safety Evaluation of Nanoparticle Formulations: A Focus Towards In Vitro Models and Adverse Outcome Pathways. Front Pharmacol 2021; 12:612659. [PMID: 34566630 PMCID: PMC8458898 DOI: 10.3389/fphar.2021.612659] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 08/05/2021] [Indexed: 12/18/2022] Open
Abstract
Nanotoxicology is an emerging field employed in the assessment of unintentional hazardous effects produced by nanoparticles (NPs) impacting human health and the environment. The nanotoxicity affects the range between induction of cellular stress and cytotoxicity. The reasons so far reported for these toxicological effects are due to their variable sizes with high surface areas, shape, charge, and physicochemical properties, which upon interaction with the biological components may influence their functioning and result in adverse outcomes (AO). Thus, understanding the risk produced by these materials now is an important safety concern for the development of nanotechnology and nanomedicine. Since the time nanotoxicology has evolved, the methods employed have been majorly relied on in vitro cell-based evaluations, while these simple methods may not predict the complexity involved in preclinical and clinical conditions concerning pharmacokinetics, organ toxicity, and toxicities evidenced through multiple cellular levels. The safety profiles of nanoscale nanomaterials and nanoformulations in the delivery of drugs and therapeutic applications are of considerable concern. In addition, the safety assessment for new nanomedicine formulas lacks regulatory standards. Though the in vivo studies are greatly needed, the end parameters used for risk assessment are not predicting the possible toxic effects produced by various nanoformulations. On the other side, due to increased restrictions on animal usage and demand for the need for high-throughput assays, there is a need for developing and exploring novel methods to evaluate NPs safety concerns. The progress made in molecular biology and the availability of several modern techniques may offer novel and innovative methods to evaluate the toxicological behavior of different NPs by using single cells, cell population, and whole organisms. This review highlights the recent novel methods developed for the evaluation of the safety impacts of NPs and attempts to solve the problems that come with risk assessment. The relevance of investigating adverse outcome pathways (AOPs) in nanotoxicology has been stressed in particular.
Collapse
Affiliation(s)
- Mounika Gayathri Tirumala
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pratibha Anchi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Susmitha Raja
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
3
|
Prince LM, Aschner M, Bowman AB. Human-induced pluripotent stems cells as a model to dissect the selective neurotoxicity of methylmercury. Biochim Biophys Acta Gen Subj 2019; 1863:129300. [PMID: 30742955 DOI: 10.1016/j.bbagen.2019.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/09/2019] [Accepted: 02/01/2019] [Indexed: 01/07/2023]
Abstract
Methylmercury (MeHg) is a potent neurotoxicant affecting both the developing and mature central nervous system (CNS) with apparent indiscriminate disruption of multiple homeostatic pathways. However, genetic and environmental modifiers contribute significant variability to neurotoxicity associated with human exposures. MeHg displays developmental stage and neural lineage selective neurotoxicity. To identify mechanistic-based neuroprotective strategies to mitigate human MeHg exposure risk, it will be critical to improve our understanding of the basis of MeHg neurotoxicity and of this selective neurotoxicity. Here, we propose that human-based pluripotent stem cell cellular approaches may enable mechanistic insight into genetic pathways that modify sensitivity of specific neural lineages to MeHg-induced neurotoxicity. Such studies are crucial for the development of novel disease modifying strategies impinging on MeHg exposure vulnerability.
Collapse
Affiliation(s)
- Lisa M Prince
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, United States
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, United States.
| |
Collapse
|
4
|
Gao X, Topping VD, Keltner Z, Sprando RL, Yourick JJ. Toxicity of nano- and ionic silver to embryonic stem cells: a comparative toxicogenomic study. J Nanobiotechnology 2017; 15:31. [PMID: 28399865 PMCID: PMC5387260 DOI: 10.1186/s12951-017-0265-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/03/2017] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The widespread application of silver nanoparticles (AgNPs) and silver-containing products has raised public safety concerns about their adverse effects on human health and the environment. To date, in vitro toxic effects of AgNPs and ionic silver (Ag+) on many somatic cell types are well established. However, no studies have been conducted hitherto to evaluate their effect on cellular transcriptome in embryonic stem cells (ESCs). RESULTS The present study characterized transcriptomic changes induced by 5.0 µg/ml AgNPs during spontaneous differentiation of mouse ESCs, and compared them to those induced by Ag+ under identical conditions. After 24 h exposure, 101 differentially expressed genes (DEGs) were identified in AgNP-treated cells, whereas 400 genes responded to Ag+. Despite the large differences in the numbers of DEGs, functional annotation and pathway analysis of the regulated genes revealed overall similarities between AgNPs and Ag+. In both cases, most of the functions and pathways impacted fell into two major categories, embryonic development and metabolism. Nevertheless, a number of canonical pathways related to cancer were found for Ag+ but not for AgNPs. Conversely, it was noted that several members of the heat shock protein and the metallothionein families were upregulated by AgNPs but not Ag+, suggesting specific oxidative stress effect of AgNPs in ESCs. The effects of AgNPs on oxidative stress and downstream apoptosis were subsequently confirmed by flow cytometry analysis. CONCLUSIONS Taken together, the results presented in the current study demonstrate that both AgNPs and Ag+ caused transcriptomic changes that could potentially exert an adverse effect on development. Although transcriptomic responses to AgNPs and Ag+ were substantially similar, AgNPs exerted specific effects on ESCs due to their nanosized particulate form.
Collapse
Affiliation(s)
- Xiugong Gao
- Division of Applied Regulatory Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708 USA
| | - Vanessa D. Topping
- Division of Applied Regulatory Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708 USA
| | - Zachary Keltner
- Division of Applied Regulatory Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708 USA
| | - Robert L. Sprando
- Division of Applied Regulatory Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708 USA
| | - Jeffrey J. Yourick
- Division of Applied Regulatory Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708 USA
| |
Collapse
|
5
|
|
6
|
Yao X, Yin N, Faiola F. Stem cell toxicology: a powerful tool to assess pollution effects on human health. Natl Sci Rev 2016. [DOI: 10.1093/nsr/nww089] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AbstractEnvironmental pollution is a global problem; the lack of comprehensive toxicological assessments may lead to increased health risks. To fully understand the health effects of pollution, it is paramount to implement fast, efficient and specific toxicity screening that relies on human models rather than on time-consuming, expensive and often inaccurate tests involving live animals. Human stem cell toxicology represents a valid alternative to traditional toxicity assays because it takes advantage of the ability of stem cells to differentiate into multiple cell types and tissues of the human body. Thus, this branch of toxicology provides a possibility to assess cellular, embryonic, developmental, reproductive and functional toxicity in vitro within a single system highly relevant to human physiology. In this review, we describe the development, performance and future perspectives of stem cell toxicology, with an emphasis on how it can meet the increasing challenges posed by environmental pollution in the modern world.
Collapse
Affiliation(s)
- Xinglei Yao
- Stake Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Nuoya Yin
- Stake Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Francesco Faiola
- Stake Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
7
|
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: 81] [Impact Index Per Article: 10.1] [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
|
8
|
Handral HK, Tong HJ, Islam I, Sriram G, Rosa V, Cao T. Pluripotent stem cells: An in vitro model for nanotoxicity assessments. J Appl Toxicol 2016; 36:1250-8. [PMID: 27241574 DOI: 10.1002/jat.3347] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 04/12/2016] [Accepted: 04/16/2016] [Indexed: 12/18/2022]
Abstract
The advent of technology has led to an established range of engineered nanoparticles that are used in diverse applications, such as cell-cell interactions, cell-material interactions, medical therapies and the target modulation of cellular processes. The exponential increase in the utilization of nanomaterials and the growing number of associated criticisms has highlighted the potential risks of nanomaterials to human health and the ecosystem. The existing in vivo and in vitro platforms show limitations, with fluctuations being observed in the results of toxicity assessments. Pluripotent stem cells (PSCs) are viable source of cells that are capable of developing into specialized cells of the human body. PSCs can be efficiently used to screen new biomaterials/drugs and are potential candidates for studying impairments of biophysical morphology at both the cellular and tissue levels during interactions with nanomaterials and for diagnosing toxicity. Three-dimensional in vitro models obtained using PSC-derived cells would provide a realistic, patient-specific platform for toxicity assessments and in drug screening applications. The current review focuses on PSCs as an alternative in vitro platform for assessing the hazardous effects of nanomaterials on health systems and highlights the importance of PSC-derived in vitro platforms. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Harish K Handral
- Oral Sciences, Faculty of Dentistry, National University of Singapore, Singapore
| | - Huei Jinn Tong
- Oral Sciences, Faculty of Dentistry, National University of Singapore, Singapore
| | - Intekhab Islam
- Oral Sciences, Faculty of Dentistry, National University of Singapore, Singapore
| | - Gopu Sriram
- Experimental Dermatology Laboratory, Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Vinicus Rosa
- Oral Sciences, Faculty of Dentistry, National University of Singapore, Singapore
| | - Tong Cao
- Oral Sciences, Faculty of Dentistry, National University of Singapore, Singapore.,National University of Singapore, Graduate School for Integrative Sciences and Engineering, Singapore.,Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore
| |
Collapse
|
9
|
Kim GD. Hesperidin Inhibits Vascular Formation by Blocking the AKT/mTOR Signaling Pathways. Prev Nutr Food Sci 2015; 20:221-9. [PMID: 26770908 DOI: 10.3746/pnf.2015.20.4.221] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 11/23/2015] [Indexed: 11/06/2022] Open
Abstract
Hesperidin has been shown to possess a potential inhibitory effect on vascular formation in endothelial cells. However, the fundamental mechanism for the anti-angiogenic activity of hesperidin is not fully understood. In the present study, we evaluated whether hesperidin has anti-angiogenic effects in mouse embryonic stem cell (mES)-derived endothelial-like cells, and human umbilical vascular endothelial cells (HUVECs), and evaluated their mechanism via the AKT/mammalian target of rapamycin (mTOR) signaling pathway. The endothelial cells were treated with several doses of hesperidin (12.5, 25, 50, and 100 μM) for 24 h. Cell viability and vascular formation were analyzed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and tube formation assay, respectively. Alteration of the AKT/mTOR signaling in vascular formation was analyzed by western blot. In addition, a mouse aortic ring assay was used to determine the effect of hesperidin on vascular formation. There were no differences between the viability of mES-derived endothelial-like cells and HUVECs after hesperidin treatment. However, hesperidin significantly inhibited cell migration and tube formation of HUVECs (P<0.05) and suppressed sprouting of microvessels in the mouse aortic ring assay. Moreover, hesperidin suppressed the expression of AKT and mTOR in HUVECs. Taken together, these findings suggest that hesperidin inhibits vascular formation by blocking the AKT/mTOR signaling pathways.
Collapse
Affiliation(s)
- Gi Dae Kim
- Department of Food and Nutritional Science, Kyungnam University, Gyeongnam 51767, Korea
| |
Collapse
|
10
|
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
|
11
|
Gao X, Sprando RL, Yourick JJ. Thalidomide induced early gene expression perturbations indicative of human embryopathy in mouse embryonic stem cells. Toxicol Appl Pharmacol 2015; 287:43-51. [DOI: 10.1016/j.taap.2015.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 04/23/2015] [Accepted: 05/14/2015] [Indexed: 01/22/2023]
|
12
|
Schulpen SH, Theunissen PT, Pennings JL, Piersma AH. Comparison of gene expression regulation in mouse- and human embryonic stem cell assays during neural differentiation and in response to valproic acid exposure. Reprod Toxicol 2015; 56:77-86. [DOI: 10.1016/j.reprotox.2015.06.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/01/2015] [Accepted: 06/02/2015] [Indexed: 12/15/2022]
|
13
|
Schulpen SHW, Pennings JLA, Piersma AH. Gene Expression Regulation and Pathway Analysis After Valproic Acid and Carbamazepine Exposure in a Human Embryonic Stem Cell-Based Neurodevelopmental Toxicity Assay. Toxicol Sci 2015; 146:311-20. [PMID: 25979313 DOI: 10.1093/toxsci/kfv094] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Differentiating pluripotent stem cells in vitro have proven useful for the study of developmental toxicity. Here, we studied the effects of anticonvulsant drug exposure in a human embryonic stem cell (hESC)-based neurodevelopmental toxicity test (hESTn). During neural differentiation the cells were exposed, for either 1 or 7 days, to noncytotoxic concentration ranges of valproic acid (VPA) or carbamazepine (CBZ), antiepileptic drugs known to cause neurodevelopmental toxicity. The effects observed on gene expression and correlated processes and pathways were in line with processes associated with neural development and pharmaceutical mode of action. In general, VPA showed a higher number of genes and molecular pathways affected than CBZ. The response kinetics differed between both compounds, with CBZ showing higher response magnitudes at day 1, versus VPA at day 7. With this study, we demonstrated the potential and biological relevance of the application of this hESC-based differentiation assay in combination with transcriptomics, as a tool to study neurodevelopmental toxicity.
Collapse
Affiliation(s)
- Sjors H W Schulpen
- *Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands and Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jeroen L A Pennings
- *Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands and
| | - Aldert H Piersma
- *Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands and Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
14
|
Chen X, Hansen DK, Merry G, DeJarnette C, Nolen G, Sloper D, Fisher JE, Harrouk W, Tassinari MS, Inselman AL. Developing osteoblasts as an endpoint for the mouse embryonic stem cell test. Reprod Toxicol 2015; 53:131-40. [PMID: 25929818 DOI: 10.1016/j.reprotox.2015.04.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 04/08/2015] [Accepted: 04/20/2015] [Indexed: 01/03/2023]
Abstract
The mouse Embryonic Stem cell Test (EST) using cardiomyocyte differentiation is a promising in vitro assay for detecting potential embryotoxicity; however, the addition of another differentiation endpoint, such as osteoblasts, may improve the predictive value of the test. A number of variables such as culture conditions and starting cell number were investigated. A 14 day direct plating method of D3 mouse embryonic stem cells (mESCs) was used to test the predictivity of osteoblast differentiation as an endpoint in the EST. Twelve compounds were tested using the prediction model developed in the ECVAM validation study. Eight of the compounds selected from the EST validation study served as model compounds; four additional compounds known to produce skeletal defects were also tested. Our results indicate comparable chemical classification between the validated cardiomyocyte endpoint and the osteoblast endpoint. These results suggest that differentiation to osteoblasts may provide confirmatory information in predicting embryotoxicity.
Collapse
Affiliation(s)
- Xinrong Chen
- Division of Systems Biology, National Center for Toxicological Research, Food and Drug Administration.
| | - Deborah K Hansen
- Division of Systems Biology, National Center for Toxicological Research, Food and Drug Administration.
| | - Gwenn Merry
- Division of Systems Biology, National Center for Toxicological Research, Food and Drug Administration.
| | | | - Greg Nolen
- Division of Systems Biology, National Center for Toxicological Research, Food and Drug Administration.
| | - Daniel Sloper
- Division of Systems Biology, National Center for Toxicological Research, Food and Drug Administration.
| | - J Edward Fisher
- Center for Drug Evaluation and Research, Food and Drug Administration.
| | - Wafa Harrouk
- Center for Drug Evaluation and Research, Food and Drug Administration.
| | | | - Amy L Inselman
- Division of Systems Biology, National Center for Toxicological Research, Food and Drug Administration.
| |
Collapse
|
15
|
Teng G, Zhao X, Lees-Miller JP, Belke D, Shi C, Chen Y, O’Brien ER, Fedak PW, Bracey N, Cross JC, Duff HJ. Role of Mutation and Pharmacologic Block of Human KCNH2 in Vasculogenesis and Fetal Mortality. Circ Arrhythm Electrophysiol 2015; 8:420-8. [DOI: 10.1161/circep.114.001837] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 01/20/2015] [Indexed: 11/16/2022]
Abstract
Background—
N629D
KCNH2
is a human missense long-QT2 mutation. Previously, we reported that the N629D/N629D mutation embryos disrupted cardiac looping, right ventricle development, and ablated I
Kr
activity at E9.5. The present study evaluates the role of KCNH2 in vasculogenesis.
Methods and Results—
N629D/N629D yolk sac vessels and aorta consist of sinusoids without normal arborization. Isolated E9.5 +/+ first branchial arches showed normal outgrowth of mouse ERG–positive/α-smooth muscle actin coimmunolocalized cells; however, outgrowth was grossly reduced in N629D/N629D. N629D/N629D aortas showed fewer α-smooth muscle actin positive cells that were not coimmunolocalized with mouse ERG cells. Transforming growth factor-β treatment of isolated N629D/N629D embryoid bodies partially rescued this phenotype. Cultured N629D/N629D embryos recapitulate the same cardiovascular phenotypes as seen in vivo. Transforming growth factor-β treatment significantly rescued these embryonic phenotypes. Both in vivo and in vitro, dofetilide treatment, over a narrow window of time, entirely recapitulated the N629D/N629D fetal phenotypes. Exogenous transforming growth factor-β treatment also rescued the dofetilide-induced phenotype toward normal.
Conclusions—
Loss of function of KCNH2 mutations results in defects in cardiogenesis and vasculogenesis. Because many medications inadvertently block the KCNH2 potassium current, these novel findings seem to have clinical relevance.
Collapse
Affiliation(s)
- Guoqi Teng
- From the Libin Cardiovascular Institute, Faculty of Medicine (G.T., J.P.L.-M., D.B., C.S., Y.C., E.R.O’B., P.W.F., N.B., H.J.D.) and Department of Comparative Biology and Experimental Medicine and Faculty of Veterinary Medicine (X.Z., J.C.C.), University of Calgary, Calgary, Alberta, Canada
| | - Xiang Zhao
- From the Libin Cardiovascular Institute, Faculty of Medicine (G.T., J.P.L.-M., D.B., C.S., Y.C., E.R.O’B., P.W.F., N.B., H.J.D.) and Department of Comparative Biology and Experimental Medicine and Faculty of Veterinary Medicine (X.Z., J.C.C.), University of Calgary, Calgary, Alberta, Canada
| | - James P. Lees-Miller
- From the Libin Cardiovascular Institute, Faculty of Medicine (G.T., J.P.L.-M., D.B., C.S., Y.C., E.R.O’B., P.W.F., N.B., H.J.D.) and Department of Comparative Biology and Experimental Medicine and Faculty of Veterinary Medicine (X.Z., J.C.C.), University of Calgary, Calgary, Alberta, Canada
| | - Darrell Belke
- From the Libin Cardiovascular Institute, Faculty of Medicine (G.T., J.P.L.-M., D.B., C.S., Y.C., E.R.O’B., P.W.F., N.B., H.J.D.) and Department of Comparative Biology and Experimental Medicine and Faculty of Veterinary Medicine (X.Z., J.C.C.), University of Calgary, Calgary, Alberta, Canada
| | - Chunhua Shi
- From the Libin Cardiovascular Institute, Faculty of Medicine (G.T., J.P.L.-M., D.B., C.S., Y.C., E.R.O’B., P.W.F., N.B., H.J.D.) and Department of Comparative Biology and Experimental Medicine and Faculty of Veterinary Medicine (X.Z., J.C.C.), University of Calgary, Calgary, Alberta, Canada
| | - Yongxiang Chen
- From the Libin Cardiovascular Institute, Faculty of Medicine (G.T., J.P.L.-M., D.B., C.S., Y.C., E.R.O’B., P.W.F., N.B., H.J.D.) and Department of Comparative Biology and Experimental Medicine and Faculty of Veterinary Medicine (X.Z., J.C.C.), University of Calgary, Calgary, Alberta, Canada
| | - Edward R. O’Brien
- From the Libin Cardiovascular Institute, Faculty of Medicine (G.T., J.P.L.-M., D.B., C.S., Y.C., E.R.O’B., P.W.F., N.B., H.J.D.) and Department of Comparative Biology and Experimental Medicine and Faculty of Veterinary Medicine (X.Z., J.C.C.), University of Calgary, Calgary, Alberta, Canada
| | - Paul W. Fedak
- From the Libin Cardiovascular Institute, Faculty of Medicine (G.T., J.P.L.-M., D.B., C.S., Y.C., E.R.O’B., P.W.F., N.B., H.J.D.) and Department of Comparative Biology and Experimental Medicine and Faculty of Veterinary Medicine (X.Z., J.C.C.), University of Calgary, Calgary, Alberta, Canada
| | - Nathan Bracey
- From the Libin Cardiovascular Institute, Faculty of Medicine (G.T., J.P.L.-M., D.B., C.S., Y.C., E.R.O’B., P.W.F., N.B., H.J.D.) and Department of Comparative Biology and Experimental Medicine and Faculty of Veterinary Medicine (X.Z., J.C.C.), University of Calgary, Calgary, Alberta, Canada
| | - James C. Cross
- From the Libin Cardiovascular Institute, Faculty of Medicine (G.T., J.P.L.-M., D.B., C.S., Y.C., E.R.O’B., P.W.F., N.B., H.J.D.) and Department of Comparative Biology and Experimental Medicine and Faculty of Veterinary Medicine (X.Z., J.C.C.), University of Calgary, Calgary, Alberta, Canada
| | - Henry J. Duff
- From the Libin Cardiovascular Institute, Faculty of Medicine (G.T., J.P.L.-M., D.B., C.S., Y.C., E.R.O’B., P.W.F., N.B., H.J.D.) and Department of Comparative Biology and Experimental Medicine and Faculty of Veterinary Medicine (X.Z., J.C.C.), University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
16
|
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: 0.8] [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
|
17
|
Schulpen SHW, de Jong E, de la Fonteyne LJJ, de Klerk A, Piersma AH. Distinct gene expression responses of two anticonvulsant drugs in a novel human embryonic stem cell based neural differentiation assay protocol. Toxicol In Vitro 2014; 29:449-57. [PMID: 25524013 DOI: 10.1016/j.tiv.2014.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/14/2014] [Accepted: 12/03/2014] [Indexed: 12/23/2022]
Abstract
Hazard assessment of chemicals and pharmaceuticals is increasingly gaining from knowledge about molecular mechanisms of toxic action acquired in dedicated in vitro assays. We have developed an efficient human embryonic stem cell neural differentiation test (hESTn) that allows the study of the molecular interaction of compounds with the neural differentiation process. Within the 11-day differentiation protocol of the assay, embryonic stem cells lost their pluripotency, evidenced by the reduced expression of stem cell markers Pou5F1 and Nanog. Moreover, stem cells differentiated into neural cells, with morphologically visible neural structures together with increased expression of neural differentiation-related genes such as βIII-tubulin, Map2, Neurogin1, Mapt and Reelin. Valproic acid (VPA) and carbamazepine (CBZ) exposure during hESTn differentiation led to concentration-dependent reduced expression of βIII-tubulin, Neurogin1 and Reelin. In parallel VPA caused an increased gene expression of Map2 and Mapt which is possibly related to the neural protective effect of VPA. These findings illustrate the added value of gene expression analysis for detecting compound specific effects in hESTn. Our findings were in line with and could explain effects observed in animal studies. This study demonstrates the potential of this assay protocol for mechanistic analysis of specific compound-induced inhibition of human neural cell differentiation.
Collapse
Affiliation(s)
- Sjors H W Schulpen
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands; Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands.
| | - Esther de Jong
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands; Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands
| | - Liset J J de la Fonteyne
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands
| | - Arja de Klerk
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands
| | - Aldert H Piersma
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands; Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands
| |
Collapse
|
18
|
Gao X, Yourick JJ, Sprando RL. Transcriptomic characterization of C57BL/6 mouse embryonic stem cell differentiation and its modulation by developmental toxicants. PLoS One 2014; 9:e108510. [PMID: 25247782 PMCID: PMC4172731 DOI: 10.1371/journal.pone.0108510] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/29/2014] [Indexed: 01/05/2023] Open
Abstract
The Tox21 program calls for transforming toxicology testing from traditional in vivo tests to less expensive and higher throughput in vitro methods. In developmental toxicology, a spectrum of alternative methods including cell line based tests has been developed. In particular, embryonic stem cells (ESCs) have received widespread attention as a promising alternative model for developmental toxicity assessment. Here, we characterized gene expression changes during mouse ESC differentiation and their modulation by developmental toxicants. C57BL/6 ESCs were allowed to differentiate spontaneously and RNA of vehicle controls was collected at 0, 24, 48, 72, 96, 120 and 168 h after embryoid body (EB) formation; RNA of compound-exposed EBs were collected at 24 h. Samples were hybridized to Affymetrix Mouse Gene 2.0 ST Array; using stringent cut-off criteria of Bonferroni-adjusted p<0.05 and fold change >2.0, a total of 1996 genes were found differentially expressed among the vehicle controls at different time points. Gene ontology (GO) analysis showed these regulated genes were mostly involved in differentiation-related processes such as development, morphogenesis, metabolism, cell differentiation, cell organization and biogenesis, embryonic development, and reproduction. Biomarkers of all three germ layers or of their derivative early cell types were identified in the gene list. Principal component analysis (PCA) based on these genes showed that the unexposed vehicle controls appeared in chronological order in the PCA plot, and formed a differentiation track when connected. Cultures exposed to thalidomide, monobutyl phthalate, or valproic acid deviated significantly from the differentiation track, manifesting the capacity of the differentiation track to identify the modulating effects of diverse developmental toxicants. The differentiation track defined in this study may be further exploited as a baseline for developmental toxicity testing, with compounds causing significant deviation from the differentiation track being predicted as potential developmental toxicants.
Collapse
Affiliation(s)
- Xiugong Gao
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland, United States of America
| | - Jeffrey J. Yourick
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland, United States of America
| | - Robert L. Sprando
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, Maryland, United States of America
| |
Collapse
|
19
|
Piersma AH, Ezendam J, Luijten M, Muller JJA, Rorije E, van der Ven LTM, van Benthem J. A critical appraisal of the process of regulatory implementation of novel in vivo and in vitro methods for chemical hazard and risk assessment. Crit Rev Toxicol 2014; 44:876-94. [PMID: 25058877 DOI: 10.3109/10408444.2014.940445] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Regulatory toxicology urgently needs applicable alternative test systems that reduce animal use, testing time, and cost. European regulation on cosmetic ingredients has already banned animal experimentation for hazard identification, and public awareness drives toward additional restrictions in other regulatory frameworks as well. In addition, scientific progress stimulates a more mechanistic approach of hazard identification. Nevertheless, the implementation of alternative methods is lagging far behind their development. In search for general bottlenecks for the implementation of alternative methods, this manuscript reviews the state of the art as to the development and implementation of 10 diverse test systems in various areas of toxicological hazard assessment. They vary widely in complexity and regulatory acceptance status. The assays are reviewed as to parameters assessed, biological system involved, standardization, interpretation of results, extrapolation to human hazard, position in testing strategies, and current regulatory acceptance status. Given the diversity of alternative methods in many aspects, no common bottlenecks could be identified that hamper implementation of individual alternative assays in general. However, specific issues for the regulatory acceptance and application were identified for each assay. Acceptance of one-in-one replacement of complex in vivo tests by relatively simple in vitro assays is not feasible. Rather, innovative approaches using test batteries are required together with metabolic information and in vitro to in vivo dose extrapolation to convincingly provide the same level of information of current in vivo tests. A mechanistically based alternative approach using the Adverse Outcome Pathway concept could stimulate further (regulatory) acceptance of non-animal tests.
Collapse
Affiliation(s)
- Aldert H Piersma
- RIVM, Center for Health Protection , Bilthoven , the Netherlands
| | | | | | | | | | | | | |
Collapse
|
20
|
Hong EJ, Jeung EB. Assessment of Developmental Toxicants using Human Embryonic Stem Cells. Toxicol Res 2014; 29:221-7. [PMID: 24578791 PMCID: PMC3936173 DOI: 10.5487/tr.2013.29.4.221] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 12/05/2013] [Accepted: 12/12/2013] [Indexed: 01/16/2023] Open
Abstract
Embryonic stem (ES) cells have potential for use in evaluation of developmental toxicity because they are generated in large numbers and differentiate into three germ layers following formation of embryoid bodies (EBs). In earlier study, embryonic stem cell test (EST) was established for assessment of the embryotoxic potential of compounds. Using EBs indicating the onset of differentiation of mouse ES cells, many toxicologists have refined the developmental toxicity of a variety of compounds. However, due to some limitation of the EST method resulting from species-specific differences between humans and mouse, it is an incomplete approach. In this regard, we examined the effects of several developmental toxic chemicals on formation of EBs using human ES cells. Although human ES cells are fastidious in culture and differentiation, we concluded that the relevancy of our experimental method is more accurate than that of EST using mouse ES cells. These types of studies could extend our understanding of how human ES cells could be used for monitoring developmental toxicity and its relevance in relation to its differentiation progress. In addition, this concept will be used as a model system for screening for developmental toxicity of various chemicals. This article might update new information about the usage of embryonic stem cells in the context of their possible ability in the toxicological fields.
Collapse
Affiliation(s)
- Eui-Ju Hong
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Korea
| |
Collapse
|
21
|
Jung EM, Choi YU, Kang HS, Yang H, Hong EJ, An BS, Yang JY, Choi KH, Jeung EB. Evaluation of developmental toxicity using undifferentiated human embryonic stem cells. J Appl Toxicol 2014; 35:205-18. [PMID: 24737281 DOI: 10.1002/jat.3010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 02/16/2014] [Accepted: 02/18/2014] [Indexed: 12/23/2022]
Abstract
An embryonic stem cell test (EST) has been developed to evaluate the embryotoxic potential of chemicals with an in vitro system. In the present study, novel methods to screen toxic chemicals during the developmental process were evaluated using undifferentiated human embryonic stem (hES) cells. By using surface marker antigens (SSEA-4, TRA-1-60 and TRA-1-81), we confirmed undifferentiated conditions of the used hES cells by immunocytochemistry. We assessed the developmental toxicity of embryotoxic chemicals, 5-fluorouracil, indomethacin and non-embryotoxic penicillin G in different concentrations for up to 7 days. While expressions of the surface markers were not significantly affected, the embryotoxic chemicals influenced their response to pluripotent ES cell markers, such as OCT-4, NANOG, endothelin receptor type B (EDNRB), secreted frizzled related protein 2 (SFRP2), teratocarcinoma-derived growth factor 1 (TDGF1), and phosphatase and tensin homolog (PTEN). Most of the pluripotent ES cell markers were down-regulated in a dose-dependent manner after treatment with embryotoxic chemicals. After treatment with 5-fluorouracil, indomethacin and penicillin G, we observed a remarkable convergence in the degree of up-regulation of development, cell cycle and apoptosis-related genes by gene expression profiles using an Affymetrix GeneChips. Taken together, these results suggest that embryotoxic chemicals have cytotoxic effects, and modulate the expression of ES cell markers as well as development-, cell cycle- and apoptosis-related genes that have pivotal roles in undifferentiated hES cells. Therefore, we suggest that hES cells may be useful for testing the toxic effects of chemicals that could impact the embryonic developmental stage.
Collapse
Affiliation(s)
- Eui-Man Jung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, 361-763, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
A statistical approach towards the derivation of predictive gene sets for potency ranking of chemicals in the mouse embryonic stem cell test. Toxicol Lett 2014; 225:342-9. [DOI: 10.1016/j.toxlet.2014.01.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/15/2014] [Accepted: 01/17/2014] [Indexed: 11/24/2022]
|
23
|
Jyoti S, Tandon S. Hypocapnia leads to enhanced expression of pluripotency and meso-endodermal differentiation genes in mouse embryonic stem cells. Exp Cell Res 2014; 322:389-401. [PMID: 24560741 DOI: 10.1016/j.yexcr.2014.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 01/15/2014] [Accepted: 02/07/2014] [Indexed: 11/18/2022]
Abstract
The efficient utilization of embryonic stem cells for applications like cell based therapy, transplantation and drug discovery largely depends upon the culturing conditions of these cells. In this report, we have analyzed gene, protein expression and morphological changes of embryonic stem cells when subjected to lowered CO2 levels i.e. hypocapnia. We studied the quantitative expression of pluripotent genes, Oct3/4, Nanog and Sox2 and genes involved in the differentiation to the three lineages, under varying CO2 levels. Enhanced expression of these genes was seen at cultures maintained at 1.5% CO2 as compared to those maintained at 5% CO2. The cells exposed to hypocapnic conditions when subjected to immunocytochemical analysis stained positive for Oct-3/4, Nanog and Sox2 transcription factors. Flow cytometry and western blot further showed that the pluripotent proteins in the 1.5% CO2 maintained cultures have higher levels of expression as compared to the ES cells at 5% CO2. In addition, there was enhanced differentiation particularly towards the mesodermal and endodermal lineages at cultures maintained and differentiated at 1.5% CO2 at all the time periods analyzed i.e. day 10 (5+5d), 12 (5+7d) and day 15 (5+10d). These results, which we feel are the first of their kind, indicate that lowered CO2 levels seem to be preferred for the maintenance of pluripotency and the subsequent differentiation.
Collapse
Affiliation(s)
- Saras Jyoti
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat 173234, India
| | - Simran Tandon
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat 173234, India.
| |
Collapse
|
24
|
Luft S, Pignalosa D, Nasonova E, Arrizabalaga O, Helm A, Durante M, Ritter S. Fate of D3 mouse embryonic stem cells exposed to X-rays or carbon ions. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2014; 760:56-63. [PMID: 24397998 DOI: 10.1016/j.mrgentox.2013.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 12/23/2013] [Accepted: 12/24/2013] [Indexed: 01/06/2023]
Abstract
The risk of radiation exposure during embryonic development is still a major problem in radiotoxicology. In this study we investigated the response of the murine embryonic stem cell (mESC) line D3 to two radiation qualities: sparsely ionizing X-rays and densely ionizing carbon ions. We analyzed clonogenic cell survival, proliferation, induction of chromosome aberrations as well as the capability of cells to differentiate to beating cardiomyocytes up to 3 days after exposure. Our results show that, for all endpoints investigated, carbon ions are more effective than X-rays at the same radiation dose. Additionally, in long term studies (≥8 days post-irradiation) chromosomal damage and the pluripotency state were investigated. These studies reveal that pluripotency markers are present in the progeny of cells surviving the exposure to both radiation types. However, only in the progeny of X-ray exposed cells the aberration frequency was comparable to that of the control population, while the progeny of carbon ion irradiated cells harbored significantly more aberrations than the control, generally translocations. We conclude that cells surviving the radiation exposure maintain pluripotency but may carry stable chromosomal rearrangements after densely ionizing radiation.
Collapse
Affiliation(s)
- S Luft
- Biophysics Department, GSI Helmholtz Centre for Heavy Ion Research, Planckstraße 1, 64291 Darmstadt, Germany
| | - D Pignalosa
- Biophysics Department, GSI Helmholtz Centre for Heavy Ion Research, Planckstraße 1, 64291 Darmstadt, Germany
| | - E Nasonova
- Biophysics Department, GSI Helmholtz Centre for Heavy Ion Research, Planckstraße 1, 64291 Darmstadt, Germany; Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Joliot-Curie 6, 141980 Dubna, Moscow Region, Russia
| | - O Arrizabalaga
- Biophysics Department, GSI Helmholtz Centre for Heavy Ion Research, Planckstraße 1, 64291 Darmstadt, Germany
| | - A Helm
- Biophysics Department, GSI Helmholtz Centre for Heavy Ion Research, Planckstraße 1, 64291 Darmstadt, Germany
| | - M Durante
- Biophysics Department, GSI Helmholtz Centre for Heavy Ion Research, Planckstraße 1, 64291 Darmstadt, Germany
| | - S Ritter
- Biophysics Department, GSI Helmholtz Centre for Heavy Ion Research, Planckstraße 1, 64291 Darmstadt, Germany.
| |
Collapse
|
25
|
Kim GD, Oh J, Park HJ, Bae K, Lee SK. Magnolol inhibits angiogenesis by regulating ROS-mediated apoptosis and the PI3K/AKT/mTOR signaling pathway in mES/EB-derived endothelial-like cells. Int J Oncol 2013; 43:600-10. [PMID: 23708970 DOI: 10.3892/ijo.2013.1959] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Accepted: 05/02/2013] [Indexed: 11/05/2022] Open
Abstract
Magnolol, a neolignan from the traditional medicinal plant Magnolia obovata, has been shown to possess neuroprotective, anti-inflammatory, anticancer and anti-angiogenic activities. However, the precise mechanism of the anti-angiogenic activity of magnolol remains to be elucidated. In the present study, the anti-angiogenic effect of magnolol was evaluated in mouse embryonic stem (mES)/embryoid body (EB)-derived endothelial-like cells. The endothelial-like cells were obtained by differentiation from mES/EB cells. Magnolol (20 µM) significantly suppressed the transcriptional and translational expression of platelet endothelial cell adhesion molecule (PECAM), an endothelial biomarker, in mES/EB-derived endothelial-like cells. To further understand the molecular mechanism of the suppression of PECAM expression, signaling pathways were analyzed in the mES/EB-derived endothelial-like cells. Magnolol induced the generation of reactive oxygen species (ROS) by mitochondria, a process that was associated with the induction of apoptosis as determined by positive Annexin V staining and the activation of cleaved caspase-3. The involvement of ROS generation by magnolol was confirmed by treatment with an antioxidant, N-acetyl-cysteine (NAC). NAC inhibited the magnolol-mediated induction of ROS generation and suppression of PECAM expression. In addition, magnolol suppressed the activation of MAPKs (ERK, JNK and p38) and the PI3K/AKT/mTOR signaling pathway in mES/EB-derived endothelial-like cells. Taken together, these findings demonstrate for the first time that the anti-angiogenic activity of magnolol may be associated with ROS-mediated apoptosis and the suppression of the PI3K/AKT/mTOR signaling pathway in mES/EB-derived endothelial-like cells.
Collapse
Affiliation(s)
- Gi Dae Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Gwanak-gu, Seoul 151-742, Republic of Korea
| | | | | | | | | |
Collapse
|
26
|
Abstract
The embryonic stem cell test is an animal-free alternative test method for developmental toxicity. Mouse embryonic stem cells are cultured in a hanging drop method to form embryoid bodies. These embryoid bodies, when plated on tissue culture dishes, differentiate to form contracting myocardial cell foci within 10 days. Inhibition of cardiomyocyte differentiation by test compounds serves as the end point of the assay, as monitored by counting contracting muscle foci under the microscope.
Collapse
Affiliation(s)
- Sjors H W Schulpen
- Laboratory for Health Protection Research-National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.
| | | |
Collapse
|
27
|
Dose response analysis of monophthalates in the murine embryonic stem cell test assessed by cardiomyocyte differentiation and gene expression. Reprod Toxicol 2012; 35:81-8. [PMID: 22813628 DOI: 10.1016/j.reprotox.2012.07.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 06/12/2012] [Accepted: 07/03/2012] [Indexed: 11/20/2022]
Abstract
The embryonic stem cell test (EST) is based on compound-induced inhibition of cardiomyocyte differentiation of pluripotent stem cells. We examined the use of transcriptomics to assess concentration-effect relationships and performed potency ranking within a chemical class. Three embryotoxic phthalate monoesters, monobutyl phthalate (MBuP), monobenzyl phthalate (MBzP) and mono-(2-ethylhexyl) phthalate (MEHP) and the non-embryotoxic monomethyl phthalate (MMP) were studied for their effects on gene expression. Effects on gene expression were observed at concentrations that did not inhibit cardiomyocyte differentiation or induce cytotoxicity. The embryotoxic phthalate monoesters altered the expression of 668 commonly expressed genes in a concentration-dependent fashion. The same potency ranking was observed for morphology and gene expression (MEHP>MBzP>MBuP>MMP). These results indicate that integrating transcriptomics provides a sensitive method to measure the dose-dependent effects of phthalate monoester exposure and enables potency ranking based on a common mode of action within a class of compounds. Transcriptomic approaches may improve the applicability of the EST, in terms of sensitivity and specificity.
Collapse
|
28
|
Kramer J, Bartsch M, Krug D, Klinger M, Nitschke M, Rohwedel J. Simvastatin modulates mouse embryonic stem cell-derived chondrogenesis in vitro. Toxicol In Vitro 2012; 26:1170-6. [PMID: 22771337 DOI: 10.1016/j.tiv.2012.06.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 06/22/2012] [Accepted: 06/25/2012] [Indexed: 10/28/2022]
Abstract
It has been studied in detail that cellular differentiation during chondrogenesis can be recapitulated in vitro by differentiation of embryonic stem (ES) cells as embryoid bodies (EBs). We here used this model system of cartilage development to analyze the effect of simvastatin, a potentially embryotoxic substance. Statins are a group of drugs used to treat hypercholesterolaemia. We found that simvastatin activated cartilage nodule formation during EB differentiation. Extended application of simvastatin resulted in enhanced expression of cartilage marker molecules and prolonged persistence of cartilage nodules. Expression of collagen type II was upregulated during simvastatin-induced chondrogenic ES cell differentiation as demonstrated by quantitative real time PCR. However, immunostaining for cartilage marker molecules revealed that cartilage nodules within simvastatin-treated EBs were defective, bearing cavities of cell loss. Furthermore, caspase activity was reduced in comparison to untreated controls indicating reduced apoptosis. Taken together, we may speculate that simvastatin prolongs survival of chondrocytes and disrupts cellular integrity of cartilage nodules during EB development by affecting apoptotic mechanisms. The study underlines that ES cell-derived EBs are a useful in vitro model to screen substances for their embryotoxic and teratogenic potential.
Collapse
Affiliation(s)
- J Kramer
- Medical Dept. I and Dept. of Virology and Cell Biology, University of Lübeck, 23538 Lübeck, Germany.
| | | | | | | | | | | |
Collapse
|
29
|
Kumar KK, Aboud AA, Bowman AB. The potential of induced pluripotent stem cells as a translational model for neurotoxicological risk. Neurotoxicology 2012; 33:518-29. [PMID: 22330734 DOI: 10.1016/j.neuro.2012.02.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Revised: 02/03/2012] [Accepted: 02/03/2012] [Indexed: 12/12/2022]
Abstract
An important goal of neurotoxicological research is to provide relevant and accurate risk assessment of environmental and pharmacological agents for populations and individuals. Owing to the challenges of human subject research and the real possibility of species specific toxicological responses, neuronal lineages derived from human embryonic stem cells (hESCs) and human neuronal precursors have been offered as a potential solution for validation of neurotoxicological data from model organism systems in humans. More recently, with the advent of induced pluripotent stem cell (iPSC) technology, there is now the possibility of personalized toxicological risk assessment, the ability to predict individual susceptibility to specific environmental agents, by this approach. This critical advance is widely expected to facilitate analysis of cellular physiological pathways in the context of human neurons and the underlying genetic factors that lead to disease. Thus this technology opens the opportunity, for the first time, to characterize the physiological, toxicological, pharmacological and molecular properties of living human neurons with identical genetic determinants as human patients. Furthermore, armed with a complete clinical history of the patients, human iPSC (hiPSC) studies can theoretically compare patients and at risk groups with distinct sensitivities to particular environmental agents, divergent clinical outcomes, differing co-morbidities, and so forth. Thus iPSCs and neuronal lineages derived from them may reflect the unique genetic blueprint of the individuals from which they are generated. Indeed, iPSC technology has the potential to revolutionize scientific approaches to human health. However, before this overarching goal can be reached a number of technical and theoretical challenges must be overcome. This review seeks to provide a realistic assessment of hiPSC technology and its application to risk assessment and mechanistic studies in the area of neurotoxicology. We seek to identify, prioritize, and detail the primary hurdles that need to be overcome if personalized toxicological risk assessment using patient-derived iPSCs is to succeed.
Collapse
Affiliation(s)
- Kevin K Kumar
- Department of Neurology, Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN, United States
| | | | | |
Collapse
|
30
|
Kim GD, Bae SY, Park HJ, Bae K, Lee SK. Honokiol Inhibits Vascular Vessel Formation of Mouse Embryonic Stem Cell-Derived Endothelial Cells via the Suppression of PECAM and MAPK/mTOR Signaling Pathway. Cell Physiol Biochem 2012; 30:758-70. [DOI: 10.1159/000341455] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2012] [Indexed: 01/12/2023] Open
|
31
|
Louisse J, Verwei M, Woutersen RA, Blaauboer BJ, Rietjens IMCM. Towardin vitrobiomarkers for developmental toxicity and their extrapolation to thein vivosituation. Expert Opin Drug Metab Toxicol 2011; 8:11-27. [DOI: 10.1517/17425255.2012.639762] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
32
|
Cellular reprogramming: a new technology frontier in pharmaceutical research. Pharm Res 2011; 29:35-52. [PMID: 22068279 DOI: 10.1007/s11095-011-0618-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 10/25/2011] [Indexed: 01/14/2023]
Abstract
Induced pluripotent stem cells via cellular reprogramming are now finding multiple applications in the pharmaceutical research and drug development pipeline. In the pre-clinical stages, they serve as model systems for basic research on specific diseases and then as key experimental tools for testing and developing therapeutics. Here we examine the current state of cellular reprogramming technology, with a special emphasis on approaches that recapitulate previously intractable human diseases in vitro. We discuss the technical and operational challenges that must be tackled as reprogrammed cells become incorporated into routine pharmaceutical research and drug discovery.
Collapse
|
33
|
Robinson JF, Theunissen PT, van Dartel DA, Pennings JL, Faustman EM, Piersma AH. Comparison of MeHg-induced toxicogenomic responses across in vivo and in vitro models used in developmental toxicology. Reprod Toxicol 2011; 32:180-8. [DOI: 10.1016/j.reprotox.2011.05.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 05/12/2011] [Accepted: 05/13/2011] [Indexed: 12/14/2022]
|
34
|
Anson BD, Kolaja KL, Kamp TJ. Opportunities for use of human iPS cells in predictive toxicology. Clin Pharmacol Ther 2011; 89:754-8. [PMID: 21430658 DOI: 10.1038/clpt.2011.9] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- B D Anson
- Cellular Dynamics International, Madison, Wisconsin, USA
| | | | | |
Collapse
|
35
|
Spielmann H. The EU Commission's Draft Report on Alternative (Non-animal) Methods for Cosmetics Testing: Current Status and Future Prospects — 2010: A Missed Opportunity. Altern Lab Anim 2010; 38:339-43. [DOI: 10.1177/026119291003800507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Horst Spielmann
- Department of Biology, Chemistry and Pharmacy Free University of Berlin Königin Luise Strasse 2–4 14195 Berlin, Germany
| |
Collapse
|
36
|
Cytotoxicity of 5-fluorouracil: Effect on endothelial differentiation via cell cycle inhibition in mouse embryonic stem cells. Toxicol In Vitro 2009; 23:719-27. [PMID: 19272435 DOI: 10.1016/j.tiv.2009.02.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Revised: 01/20/2009] [Accepted: 02/22/2009] [Indexed: 11/21/2022]
Abstract
Embryonic stem cells (ESCs) are known to characteristics for pluripotency and self-renewal, but the precise mechanisms of ES-derived cells to specific toxicants have not been determined. Here, we evaluated the cytotoxicity of 5-fluorouracil (5-FU) and see its effect on cell viability, proliferation, and differentiation in mouse ESC-derived endothelial differentiation. Mouse ESCs were exposed to 5-FU (10 microM) and combined with probucol (50 microM) for 24h, which is an antagonist of 5-FU. Changes in gene expression as a result of 5-FU exposure in mouse ESC-derived endothelial precursor cells (ES-EPCs) were assessed using an oligonucleotide microarray (AB1700). The expression of Oct-4 was decreased during the differentiation of mouse ESCs into endothelial cells; otherwise, the expression of PECAM was increased. Mouse ES-EPCs were shown to have a decrease in viability (49.8%) and PECAM expression, and induce G1/S phase (31.1%/60.6%) when compared with/without treatment of 5-FU. Expression of cell cycle-related proteins was increased in endothelial precursor cells exposed to 5-FU without probucol treatment. From theses results suggest that 5-FU inhibit endothelial differentiation as well as inducing the G1/S phase arrest. We propose that mouse ES-EPCs might be a useful tool for screening the cytotoxicity of compounds in endothelial cells.
Collapse
|
37
|
Kim GD, Kim GJ, Seok JH, Chung HM, Chee KM, Rhee GS. Differentiation of endothelial cells derived from mouse embryoid bodies: a possible in vitro vasculogenesis model. Toxicol Lett 2008; 180:166-73. [PMID: 18590808 DOI: 10.1016/j.toxlet.2008.05.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Revised: 05/15/2008] [Accepted: 05/23/2008] [Indexed: 10/22/2022]
Abstract
Mouse embryonic stem cells (mES cells), which are pluripotent and self-renewal cells, are derived from the inner cell mass of mouse blastocysts. The objective of this study was to construct more efficient mES cell-derived embryoid bodies (EBs) for use as a vasculogenesis model and as an in vitro vascular toxicity testing model. EBs were formed for 3 days using hanging drop cultures and plated on gelatin-coated plates in endothelial growth medium-2 (EGM-2) to promote vascular development. The differentiation of mES cell-derived EBs was confirmed by reverse transcription-polymerase chain reaction (RT-PCR), immunocytochemistry, and flow cytometry within 7 days after plating EBs. The mRNA and protein expressions of vascular endothelial growth factor receptors-2 (FLK-1), platelet endothelial cell adhesion molecule (PECAM), and vascular endothelial-cadherin (VE-cadherin) were observed in differentiated mES cells. When placed in matrigel, mES cell-derived endothelial like cells formed networks similar to vascular structures. mES cells were also exposed to 5-fluorouracil (5-FU), a strong inhibitor of vessel formation, and its cytotoxicity was determined using MTT assays. The inhibitory concentrations (IC50) of 5-FU for mES cells and C166 cells were 0.72 microM and 1.04 microM, respectively. These results demonstrate that mES cells can be used to study vasculogenesis and for cytotoxicity screening.
Collapse
Affiliation(s)
- Gi Dae Kim
- Department of Reproductive and Developmental Toxicology, National Institute of Toxicological Research, KFDA, Seoul 122-704, Republic of Korea
| | | | | | | | | | | |
Collapse
|
38
|
Woei Ng K, Speicher T, Dombrowski C, Helledie T, Haupt LM, Nurcombe V, Cool SM. Osteogenic differentiation of murine embryonic stem cells is mediated by fibroblast growth factor receptors. Stem Cells Dev 2007; 16:305-18. [PMID: 17521241 DOI: 10.1089/scd.2006.0044] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The mechanisms involved in the control of embryonic stem (ES) cell differentiation are yet to be fully elucidated. However, it has become clear that the family of fibroblast growth factors (FGFs) are centrally involved. In this study we examined the role of the FGF receptors (FGFRs 1-4) during osteogenesis in murine ES cells. Single cells were obtained after the formation of embryoid bodies, cultured on gelatin-coated plates, and coaxed to differentiate along the osteogenic lineage. Upregulation of genes was analyzed at both the transcript and protein levels using gene array, relative-quantitative PCR (RQ-PCR), and Western blotting. Deposition of a mineralized matrix was evaluated with Alizarin Red staining. An FGFR1-specific antibody was generated and used to block FGFR1 activity in mES cells during osteogenic differentiation. Upon induction of osteogenic differentiation in mES cells, all four FGFRs were clearly upregulated at both the transcript and protein levels with a number of genes known to be involved in osteogenic differentiation including bone morphogenetic proteins (BMPs), collagen I, and Runx2. Cells were also capable of depositing a mineralized matrix, confirming the commitment of these cells to the osteogenic lineage. When FGFR1 activity was blocked, a reduction in cell proliferation and a coincident upregulation of Runx2 with enhanced mineralization of cultures was observed. These results indicate that FGFRs play critical roles in cell recruitment and differentiation during the process of osteogenesis in mES cells. In particular, the data indicate that FGFR1 plays a pivotal role in osteoblast lineage determination.
Collapse
Affiliation(s)
- Kee Woei Ng
- Stem Cell and Tissue Repair Laboratory, Institute of Molecular and Cell Biology, Proteos, Singapore 138673
| | | | | | | | | | | | | |
Collapse
|
39
|
Tran DN, Ota LC, Jacobson JD, Patton WC, Chan PJ. Influence of nanoparticles on morphological differentiation of mouse embryonic stem cells. Fertil Steril 2007; 87:965-70. [PMID: 17140568 DOI: 10.1016/j.fertnstert.2006.07.1520] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Revised: 07/22/2006] [Accepted: 07/22/2006] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To assess mouse embryonic stem (ES) cell viability, growth, and differentiated morphology after exposure to different concentrations of nanoparticles. DESIGN Cell culture for 6 days. SETTING University research laboratory. ANIMALS Cryopreserved mouse ES-D3 (American Type Culture Collection, Manassas, VA) cells. INTERVENTION(S) ES cells were exposed to either 0 (control), 0.4, or 12.2 million/mL mixed-size fluorescent nanoparticles in culture (37 degrees C, 5% CO(2) in air) for 6 days. MAIN OUTCOME MEASURE(S) Cell viability and morphometric analysis were performed. RESULT(S) ES cells exposed to both concentrations of nanoparticles exhibited smaller cell surface area. The effect was not concentration dependent. In contrast, ES cell nucleus size was unaffected. The nanoparticles distributed into the cytoplasm, pseudopods, and the perinuclear region. ES cell viabilities were reduced 40% and 30% in the low versus high relative concentration, respectively. ES cells in low-concentration nanoparticles became mostly columnar and embryoid body shaped. However, in high-concentration nanoparticles, they differentiated toward fibroblast-like and less squamous types. CONCLUSION(S) The observed reduced ES cell surface area suggested disruption of cytoskeletal development but not nuclear organization by nanoparticles. The ring-like formation of nanoparticles around the nucleus and the resulting cell morphologies suggested nanoparticles may influence differentiation.
Collapse
Affiliation(s)
- Diana N Tran
- Department of Gynecology and Obstetrics, Loma Linda University School of Medicine, Loma Linda, California, USA
| | | | | | | | | |
Collapse
|
40
|
Duplomb L, Dagouassat M, Jourdon P, Heymann D. Differentiation of osteoblasts from mouse embryonic stem cells without generation of embryoid body. In Vitro Cell Dev Biol Anim 2007; 43:21-4. [PMID: 17570030 DOI: 10.1007/s11626-006-9010-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Osteoblasts are cells specialized in extracellular matrix production and mineralization. In collaboration with osteoclasts which are bone-resorbing cells, osteoblasts regulate bone homeostasis. The study of osteoblast differentiation from the earliest states of the differentiation can be performed using embryonic stem cells. Embryonic stem cells are pluripotent cells which have the capacity to give rise to all kinds of cells of the body. The main protocol to differentiate embryonic stem cells into osteoblast uses the generation of embryoid body which is a three-dimensional structure mimicking the developing embryo. Recently, it has been shown that human embryonic stem cells have the capacity to differentiate spontaneously into osteoblasts. In this manuscript, we showed that mouse embryonic stem cells have the capacity to differentiate spontaneously into osteoblasts, which can be visualized by the appearance of mineralization nodules and osteogenic markers.
Collapse
|
41
|
Choi EJ, Kim GD, Chee KM, Kim GH. Effects of hesperetin on vessel structure formation in mouse embryonic stem (mES) cells. Nutrition 2006; 22:947-51. [PMID: 16815676 DOI: 10.1016/j.nut.2006.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Revised: 04/28/2006] [Accepted: 05/01/2006] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The present study investigated the effects of hesperetin on vessel structure formation in mouse embryonic stem (mES) cells with regard to whether hesperetin acts as an antioxidant or pro-oxidant. Some flavonoids enhance antioxidant systems while increasing oxidative stress in the body. METHODS After their differentiation into endothelial-like cells for 10 d, mES cells were treated with 1 to 100 muM of hesperetin for 24 h. RESULTS Hesperetin efficiently inhibited the formation of vessel-like tubular structures consisting of platelet-endothelial cell adhesion molecule-1-immunoreactive cells and significantly (P < 0.05) increased the generation of reactive oxygen species in a concentration-dependent manner. Although glutathione (in its reduced and oxidized forms) in mES cells was not affected by hesperetin, the 8-iso-prostaglandin F2(alpha) content was decreased. In addition, cytotoxicity-induced hesperetin was not found; lactate dehydrogenase release and cell viability were determined as an index of cell damage. CONCLUSION Taken together, the present study shows that hesperetin inhibits vessel formation by pro-oxidant means and suggests its potential as an antiangiogenic agent.
Collapse
Affiliation(s)
- Eun Jeong Choi
- Plant Resources Research Institute, Duksung Women's University, Seoul, South Korea.
| | | | | | | |
Collapse
|
42
|
Greenlee AR, Kronenwetter-Koepel TA, Kaiser SJ, Liu K. Comparison of Matrigel™ and gelatin substrata for feeder-free culture of undifferentiated mouse embryonic stem cells for toxicity testing. Toxicol In Vitro 2005; 19:389-97. [PMID: 15713546 DOI: 10.1016/j.tiv.2004.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2004] [Revised: 11/11/2004] [Accepted: 11/18/2004] [Indexed: 11/18/2022]
Abstract
Murine embryonic stem (mES) cells have been used to evaluate cytotoxicity and developmental injury following exposure to embryotoxic agents. However, maintaining a homogeneous population of undifferentiated mES cells for this purpose has been complicated by the need for continuous co-culture with murine embryonic fibroblast (mEF) cells or limited passaging on plastic surfaces coated with gelatin. Here, we compare the synthetic basement membrane Matrigel with 0.1% gelatin substratum for feeder-free propagation of undifferentiated mES cells. Biomarkers of pluripotentiality, chromosome number, caspase-3 expression, and cardiomyocyte differentiation were monitored for mES cells cultured on Matrigel or 0.1% gelatin up to passage 7 (P7). Our results suggest that choice of substratum had no significant effect on population doubling time, cell viability, stage-specific embryonic antigen-1 (SSEA-1) expression, or early passage formation of beating cardiomyocytes (all P>or=0.09). In other comparisons, however, Matrigel supported significantly higher synthesis of alkaline phosphatase (7.7x10(-3)+/-0.8 vs 6.6x10(-3)+/-0.8 units/liter/cell, respectively, P=0.012), overall expression of activated caspase-3 following exposure to 5, 10, 50, 100 and 500 parts per billion (ppb) sodium arsenite (P<0.0001), and percent development to beating cardiomyocytes at P7 (P=0.01). Together, our findings suggest that Matrigel shows promise as a substrate for feeder-free propagation of undifferentiated mES cells for embryotoxicity endpoints.
Collapse
Affiliation(s)
- A R Greenlee
- Marshfield Clinic Research Foundation, 1000 North Oak Avenue, Marshfield, WI 54449, USA.
| | | | | | | |
Collapse
|
43
|
Greenlee AR, Kronenwetter-Koepel TA, Kaiser SJ, Ellis TM, Liu K. Combined effects of MatrigelTM and growth factors on maintaining undifferentiated murine embryonic stem cells for embryotoxicity testing. Toxicol In Vitro 2004; 18:543-53. [PMID: 15130612 DOI: 10.1016/j.tiv.2004.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2003] [Accepted: 01/28/2004] [Indexed: 10/26/2022]
Abstract
Undifferentiated, murine embryonic stem (mES) cells have shown promise as a substrate for identifying embryotoxic chemicals and for studying mechanisms of early developmental injury. However, long-term maintenance of mES cells in an undifferentiated state is problematic. The present study evaluates the combination of Matrigel matrix and three growth factors for this purpose. Biomarkers of mES cell pluripotency, apoptosis, chromosome number and cardiomyocyte differentiation were monitored over 119 population doublings. D3 mES cells retained undifferentiated characteristics, including sustained expression of alkaline phosphatase and stage specific embryonic antigen-1 (SSEA-1) and continued transcription of Pou5f1 (Oct-4). Cell viability remained at > or=95% and population-doubling times averaged 14.3 h over 10 weeks of observation. Caspase-3 activation, a marker of cellular death by apoptosis, was measured in early- and late-passage mES cells. Early-passage cells showed dose-responsive caspase-3 activation following exposure to sodium arsenite, whereas caspase-3 activation of late-passage cells dropped to background levels at toxicant dosages above 50 ppb. Aneuploidy and impaired differentiation into beating cardiomyocytes were noted for late-passage mES cells. Matrigel, combined with growth factors, may sustain undifferentiated mES cells. However, aneuploidy, reduced caspase-3 activation, and inability to differentiate suggests further modifications to the culture system may be needed for long-term propagation of cells for embryotoxicity endpoints.
Collapse
Affiliation(s)
- A R Greenlee
- National Farm Medicine Center, Marshfield Clinic Research Foundation, 1000 North Oak Avenue, Marshfield, WI 54449, USA.
| | | | | | | | | |
Collapse
|
44
|
zur Nieden NI, Kempka G, Ahr HJ. In vitro differentiation of embryonic stem cells into mineralized osteoblasts. Differentiation 2003; 71:18-27. [PMID: 12558600 DOI: 10.1046/j.1432-0436.2003.700602.x] [Citation(s) in RCA: 252] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Embryonic stem cells are pluripotent cells derived from the inner cell mass of mouse blastocysts that have been shown to differentiate spontaneously into cell types representing all three germ layers. This study shows that ES cells were induced to differentiate in vitro into mineralized osteoblasts under the influence of ascorbic acid, beta-glycerophosphate and 1alpha,25-OH vitamin D3. The activity of alkaline phosphatase, an early osteoblast marker, was found to be increased around day 12 of culture. Mineralized cells were clearly identified by histochemical staining, which detects mineralized calcium. The major noncollagenous component of bone matrix, osteocalcin, was localized to the mineralized cells by immunofluorescence. The expression of bone-specific genes was analyzed by real-time quantitative PCR. Osteocalcin and bone sialoprotein (BSP) were identified as early as in the fourth week of embryonic stem cell culture, both being characteristic for late stages of osteoblastic differentiation, indicating that at this time of culture the identified cells represent "mature" osteoblasts. The osteoblast-specific transcription factor Cbfa1 was induced a few days earlier. The expression of osteopontin and osteonectin, both being involved in binding calcium ions and hydroxyapatite during mineralization processes, as well as of collagen type I, representing by far the most predominant collagen in vertebrate organisms, is enhanced at the beginning of the second culture week upon addition of supplements. In the third week of culture, treated cells showed a second peak of osteopontin, osteonectin and collagen type I expression, osteopontin and osteonectin being stimulated 3-4-fold and collagen type I being induced 6-fold over control values. Alkaline phosphatase (ALP) expression was enhanced at the beginning of the third week of culture and was found to be increased again at later stages of culture at days 27-34. The in vitro differentiation of mouse embryonic stem cells into osteoblasts may provide a suitable model for studying the molecular processes of osteoblastic development in vivo.
Collapse
|
45
|
Abstract
Embryonic stem cells are pluripotent cells derived from mouse blastocysts, which have the capacity to differentiate in vitro into a wide variety of cell types. Based on this potential the embryonic stem cell test (EST) has been developed, which represents an assay system for the classification of compounds for their teratogenic potential, based on the morphological evaluation of contracting myocard cells compared to the cytotoxic effects on undifferentiated stem cells and adult 3T3 fibroblasts. To expand the EST, the quantitative expression of the alpha- and beta-myosin heavy chain (MHC) genes under the influence of test compounds was studied employing real-time TaqMan PCR analysis. The molecular evaluation of the MHC genes allows a higher sensitivity for the classification of substances and the transfer of the EST to the molecular level allows to start experimental procedures at day 9 of culture. Thus, the modulated EST holds promise as a new easily quantifiable in vitro screening assay in teratology.
Collapse
Affiliation(s)
- N I zur Nieden
- Bayer AG, Research Toxicology, 42096 Wuppertal, Germany. nicole.melzer.nm.@bayer.ag.de
| | | | | | | | | |
Collapse
|
46
|
Bremer S, Van Dooren M, Paparella M, Kossolov E, Fleischmann B, Hescheler J. Establishment of an Embryotoxicity Assay with Green Fluorescence Protein-expressing Embryonic Cell-derived Cardiomyocytes. Altern Lab Anim 1999; 27:471-84. [DOI: 10.1177/026119299902700303] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Transgenic embryonic stem cells were used to determine the embryotoxic effects of chemicals on the development of embryonic tissues. This investigation supports an ongoing validation study, aimed at reducing the time-consuming procedure currently in use, and at providing more-objective and more-detailed information on the embryotoxic potentials of chemicals. Green fluorescence protein (GFP) was used as a reporter gene and was linked to a human α-cardiac-specific promoter. The expression of GFP was switched on after specific activation of the human α-actin promoter. This permitted the easy quantification of cardiac cells by using a fluorescence-activated cell sorter (FACS). The percentage of cardiac precursor cells was calculated from the FACS-distribution pattern of cells which fluoresced versus the total number of cells. The percentage of cardiac precursor cells increased from 25% in embryoid bodies on day 3, to 86% on day 7. However, in 11-day-old embryoid bodies, the percentage decreased to 35%. Five chemicals with known embryotoxic potentials were compared with respect to the IC50 (concentration causing 50% inhibition of measured effect) values obtained by various in vitro endpoints (for example, cytotoxicity, morphology). The results showed a higher sensitivity of endpoints used for the analysis of specific effects on the selected target tissue. The data also showed the need to develop in vitro methods with specific endpoints which account for the complexity of embryotoxicology.
Collapse
Affiliation(s)
- Susanne Bremer
- ECVAM, Institute for Health & Consumer Protection, Joint Research Centre, European Commission, 21020 Ispra, Italy
| | - Maaike Van Dooren
- ECVAM, Institute for Health & Consumer Protection, Joint Research Centre, European Commission, 21020 Ispra, Italy
| | - Martin Paparella
- ECVAM, Institute for Health & Consumer Protection, Joint Research Centre, European Commission, 21020 Ispra, Italy
| | - Eugen Kossolov
- University of Cologne, Department of Neurophysiology, Robert-Koch-Strasse 39, 50931 Cologne, Germany
| | - Bernd Fleischmann
- University of Cologne, Department of Neurophysiology, Robert-Koch-Strasse 39, 50931 Cologne, Germany
| | - Juergen Hescheler
- University of Cologne, Department of Neurophysiology, Robert-Koch-Strasse 39, 50931 Cologne, Germany
| |
Collapse
|
47
|
4. Reproductive Toxicity. Hum Exp Toxicol 1997. [DOI: 10.1177/096032719701600103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
48
|
Newall D, Beedles K. The stem-cell test: an in vitro assay for teratogenic potential. Results of a blind trial with 25 compounds. Toxicol In Vitro 1996; 10:229-40. [DOI: 10.1016/0887-2333(95)00110-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/1995] [Indexed: 01/02/2023]
|
49
|
Hooghe R, Ooms D. Use of the fluorescence-activated cell sorter (FACS) for in vitro assays of developmental toxicity. Toxicol In Vitro 1995; 9:349-54. [DOI: 10.1016/0887-2333(95)00009-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/1994] [Indexed: 01/07/2023]
|
50
|
Heuer J, Graeber I, Pohl I, Spielmann H. An in vitro embryotoxicity assay using the differentiation of embryonic mouse stem cells into haematopoietic cells. Toxicol In Vitro 1994; 8:585-7. [DOI: 10.1016/0887-2333(94)90022-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|