1
|
Hoang P, Kowalczewski A, Sun S, Winston TS, Archilla AM, Lemus SM, Ercan-Sencicek AG, Gupta AR, Liu W, Kontaridis MI, Amack JD, Ma Z. Engineering spatial-organized cardiac organoids for developmental toxicity testing. Stem Cell Reports 2021; 16:1228-1244. [PMID: 33891865 PMCID: PMC8185451 DOI: 10.1016/j.stemcr.2021.03.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 12/16/2022] Open
Abstract
Emerging technologies in stem cell engineering have produced sophisticated organoid platforms by controlling stem cell fate via biomaterial instructive cues. By micropatterning and differentiating human induced pluripotent stem cells (hiPSCs), we have engineered spatially organized cardiac organoids with contracting cardiomyocytes in the center surrounded by stromal cells distributed along the pattern perimeter. We investigated how geometric confinement directed the structural morphology and contractile functions of the cardiac organoids and tailored the pattern geometry to optimize organoid production. Using modern data-mining techniques, we found that pattern sizes significantly affected contraction functions, particularly in the parameters related to contraction duration and diastolic functions. We applied cardiac organoids generated from 600 μm diameter circles as a developmental toxicity screening assay and quantified the embryotoxic potential of nine pharmaceutical compounds. These cardiac organoids have potential use as an in vitro platform for studying organoid structure-function relationships, developmental processes, and drug-induced cardiac developmental toxicity. Micropattern-based geometric confinement directs cardiac organoid development Cardiac organoid structure-function relationships are guided by organoid size Cardiac organoids can be used as an in vitro embryotoxicity assessment tool
Collapse
Affiliation(s)
- Plansky Hoang
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA; BioInspired Syracuse Institute for Material and Living Systems, Syracuse, NY, USA
| | - Andrew Kowalczewski
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA; BioInspired Syracuse Institute for Material and Living Systems, Syracuse, NY, USA
| | - Shiyang Sun
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA; BioInspired Syracuse Institute for Material and Living Systems, Syracuse, NY, USA
| | - Tackla S Winston
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA; BioInspired Syracuse Institute for Material and Living Systems, Syracuse, NY, USA
| | - Adriana M Archilla
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA; BioInspired Syracuse Institute for Material and Living Systems, Syracuse, NY, USA
| | - Stephanie M Lemus
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA; BioInspired Syracuse Institute for Material and Living Systems, Syracuse, NY, USA
| | | | - Abha R Gupta
- Department of Pediatrics, Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - Wenzhong Liu
- Department of Pediatrics, Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | | | - Jeffrey D Amack
- BioInspired Syracuse Institute for Material and Living Systems, Syracuse, NY, USA; Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Zhen Ma
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA; BioInspired Syracuse Institute for Material and Living Systems, Syracuse, NY, USA.
| |
Collapse
|
2
|
Ahmed U, Ahmed R, Masoud MS, Tariq M, Ashfaq UA, Augustine R, Hasan A. Stem cells based in vitro models: trends and prospects in biomaterials cytotoxicity studies. Biomed Mater 2021; 16:042003. [PMID: 33686970 DOI: 10.1088/1748-605x/abe6d8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Advanced biomaterials are increasingly used for numerous medical applications from the delivery of cancer-targeted therapeutics to the treatment of cardiovascular diseases. The issues of foreign body reactions induced by biomaterials must be controlled for preventing treatment failure. Therefore, it is important to assess the biocompatibility and cytotoxicity of biomaterials on cell culture systems before proceeding to in vivo studies in animal models and subsequent clinical trials. Direct use of biomaterials on animals create technical challenges and ethical issues and therefore, the use of non-animal models such as stem cell cultures could be useful for determination of their safety. However, failure to recapitulate the complex in vivo microenvironment have largely restricted stem cell cultures for testing the cytotoxicity of biomaterials. Nevertheless, properties of stem cells such as their self-renewal and ability to differentiate into various cell lineages make them an ideal candidate for in vitro screening studies. Furthermore, the application of stem cells in biomaterials screening studies may overcome the challenges associated with the inability to develop a complex heterogeneous tissue using primary cells. Currently, embryonic stem cells, adult stem cells, and induced pluripotent stem cells are being used as in vitro preliminary biomaterials testing models with demonstrated advantages over mature primary cell or cell line based in vitro models. This review discusses the status and future directions of in vitro stem cell-based cultures and their derivatives such as spheroids and organoids for the screening of their safety before their application to animal models and human in translational research.
Collapse
Affiliation(s)
- Uzair Ahmed
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad 38000 Punjab, Pakistan
| | | | | | | | | | | | | |
Collapse
|
3
|
Kasteel EEJ, Westerink RHS. Refining in vitro and in silico neurotoxicity approaches by accounting for interspecies and interindividual differences in toxicodynamics. Expert Opin Drug Metab Toxicol 2021; 17:1007-1017. [PMID: 33586568 DOI: 10.1080/17425255.2021.1885647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION The process of chemical risk assessment traditionally relies on animal experiments and associated default uncertainty factors to account for interspecies and interindividual differences. To work toward a more precise and personalized risk assessment, these uncertainty factors should be refined and replaced by chemical-specific adjustment factors (CSAFs). AREAS COVERED This concise review discusses alternative (in vitro/in silico) approaches that can be used to assess interspecies and interindividual differences in toxicodynamics, ranging from targeted to more integrated approaches. Although data are available on interspecies differences, the increasing use of human-induced pluripotent stem cell (hiPSC)-derived neurons may provide opportunities to also assess interindividual variability in neurotoxicity. More integrated approaches, like adverse outcome pathways (AOPs) can provide a more quantitative understanding of the toxicodynamics of a chemical. EXPERT OPINION To improve chemical risk assessment, refinement of uncertainty factors is crucial. In vitro and in silico models can facilitate the development of CSAFs, but still these models cannot always capture the complexity of the in vivo situation, thereby potentially hampering regulatory acceptance. The combined use of more integrated approaches, like AOPs and physiologically based kinetic models, can aid in structuring data and increasing suitability of alternative approaches for regulatory purposes.
Collapse
Affiliation(s)
- Emma E J Kasteel
- Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Remco H S Westerink
- Toxicology Division, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
4
|
Uçkan-Çetinkaya D, Haider KH. Induced Pluripotent Stem Cells in Pediatric Research and Clinical Translation. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
5
|
Horning KJ, Joshi P, Nitin R, Balachandran RC, Yanko FM, Kim K, Christov P, Aschner M, Sulikowski GA, Weaver CD, Bowman AB. Identification of a selective manganese ionophore that enables nonlethal quantification of cellular manganese. J Biol Chem 2020; 295:3875-3890. [PMID: 32047113 PMCID: PMC7086026 DOI: 10.1074/jbc.ra119.009781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/11/2020] [Indexed: 01/14/2023] Open
Abstract
Available assays for measuring cellular manganese (Mn) levels require cell lysis, restricting longitudinal experiments and multiplexed outcome measures. Conducting a screen of small molecules known to alter cellular Mn levels, we report here that one of these chemicals induces rapid Mn efflux. We describe this activity and the development and implementation of an assay centered on this small molecule, named manganese-extracting small molecule (MESM). Using inductively-coupled plasma-MS, we validated that this assay, termed here "manganese-extracting small molecule estimation route" (MESMER), can accurately assess Mn in mammalian cells. Furthermore, we found evidence that MESM acts as a Mn-selective ionophore, and we observed that it has increased rates of Mn membrane transport, reduced cytotoxicity, and increased selectivity for Mn over calcium compared with two established Mn ionophores, calcimycin (A23187) and ionomycin. Finally, we applied MESMER to test whether prior Mn exposures subsequently affect cellular Mn levels. We found that cells receiving continuous, elevated extracellular Mn accumulate less Mn than cells receiving equally-elevated Mn for the first time for 24 h, indicating a compensatory cellular homeostatic response. Use of the MESMER assay versus a comparable detergent lysis-based assay, cellular Fura-2 Mn extraction assay, reduced the number of cells and materials required for performing a similar but cell lethality-based experiment to 25% of the normally required sample size. We conclude that MESMER can accurately quantify cellular Mn levels in two independent cells lines through an ionophore-based mechanism, maintaining cell viability and enabling longitudinal assessment within the same cultures.
Collapse
Affiliation(s)
- Kyle J. Horning
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
| | - Piyush Joshi
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
| | - Rachana Nitin
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
| | | | - Frank M. Yanko
- School of Health Sciences, Purdue University, West Lafayette, Indiana 47907
| | - Kwangho Kim
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232,Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235
| | - Plamen Christov
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Gary A. Sulikowski
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232,Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37212
| | - C. David Weaver
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37212
| | - Aaron B. Bowman
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232,School of Health Sciences, Purdue University, West Lafayette, Indiana 47907, To whom correspondence should be addressed:
Purdue University, 550 Stadium Mall Dr., HAMP 1173A, West Lafayette, IN 47907-2051. E-mail:
| |
Collapse
|
6
|
Wages PA, Joshi P, Tallman KA, Kim HYH, Bowman AB, Porter NA. Screening ToxCast™ for Chemicals That Affect Cholesterol Biosynthesis: Studies in Cell Culture and Human Induced Pluripotent Stem Cell-Derived Neuroprogenitors. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:17014. [PMID: 31985273 PMCID: PMC7015578 DOI: 10.1289/ehp5053] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 11/20/2019] [Accepted: 11/25/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Changes in cholesterol metabolism are common hallmarks of neurodevelopmental pathologies. A diverse array of genetic disorders of cholesterol metabolism support this claim as do multiple lines of research that demonstrate chemical inhibition of cholesterol biosynthesis compromises neurodevelopment. Recent work has revealed that a number of commonly used pharmaceuticals induce changes in cholesterol metabolism that are similar to changes induced by genetic disorders with devastating neurodevelopmental deficiencies. OBJECTIVES We tested the hypothesis that common environmental toxicants may also impair cholesterol metabolism and thereby possibly contribute to neurodevelopmental toxicity. METHODS Using high-throughput screening with a targeted lipidomic analysis and the mouse neuroblastoma cell line, Neuro-2a, the ToxCast™ chemical library was screened for compounds that impact sterol metabolism. Validation of chemical effects was conducted by assessing cholesterol biosynthesis in human induced pluripotent stem cell (hiPSC)-derived neuroprogenitors using an isotopically labeled cholesterol precursor and by monitoring product formation with UPLC-MS/MS. RESULTS Twenty-nine compounds were identified as validated lead-hits, and four were prioritized for further study (endosulfan sulfate, tributyltin chloride, fenpropimorph, and spiroxamine). All four compounds were validated to cause hypocholesterolemia in Neuro-2a cells. The morpholine-like fungicides, fenpropimorph and spiroxamine, mirrored their Neuro-2a activity in four immortalized human cell lines and in a human neuroprogenitor model derived from hiPSCs, but endosulfan sulfate and tributyltin chloride did not. CONCLUSIONS These data reveal the existence of environmental compounds that interrupt cholesterol biosynthesis and that methodologically hiPSC neuroprogenitor cells provide a particularly sensitive system to monitor the effect of small molecules on de novo cholesterol formation. https://doi.org/10.1289/EHP5053.
Collapse
Affiliation(s)
- Phillip A. Wages
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Piyush Joshi
- Departments of Pediatrics, Neurology and Biochemistry, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Keri A. Tallman
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Hye-Young H. Kim
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Aaron B. Bowman
- Departments of Pediatrics, Neurology and Biochemistry, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Ned A. Porter
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
7
|
Kim TW, Che JH, Yun JW. Use of stem cells as alternative methods to animal experimentation in predictive toxicology. Regul Toxicol Pharmacol 2019; 105:15-29. [DOI: 10.1016/j.yrtph.2019.03.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/23/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022]
|
8
|
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: 0.7] [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
|
9
|
Grainger AI, King MC, Nagel DA, Parri HR, Coleman MD, Hill EJ. In vitro Models for Seizure-Liability Testing Using Induced Pluripotent Stem Cells. Front Neurosci 2018; 12:590. [PMID: 30233290 PMCID: PMC6127295 DOI: 10.3389/fnins.2018.00590] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/06/2018] [Indexed: 12/14/2022] Open
Abstract
The brain is the most complex organ in the body, controlling our highest functions, as well as regulating myriad processes which incorporate the entire physiological system. The effects of prospective therapeutic entities on the brain and central nervous system (CNS) may potentially cause significant injury, hence, CNS toxicity testing forms part of the “core battery” of safety pharmacology studies. Drug-induced seizure is a major reason for compound attrition during drug development. Currently, the rat ex vivo hippocampal slice assay is the standard option for seizure-liability studies, followed by primary rodent cultures. These models can respond to diverse agents and predict seizure outcome, yet controversy over the relevance, efficacy, and cost of these animal-based methods has led to interest in the development of human-derived models. Existing platforms often utilize rodents, and so lack human receptors and other drug targets, which may produce misleading data, with difficulties in inter-species extrapolation. Current electrophysiological approaches are typically used in a low-throughput capacity and network function may be overlooked. Human-derived induced pluripotent stem cells (iPSCs) are a promising avenue for neurotoxicity testing, increasingly utilized in drug screening and disease modeling. Furthermore, the combination of iPSC-derived models with functional techniques such as multi-electrode array (MEA) analysis can provide information on neuronal network function, with increased sensitivity to neurotoxic effects which disrupt different pathways. The use of an in vitro human iPSC-derived neural model for neurotoxicity studies, combined with high-throughput techniques such as MEA recordings, could be a suitable addition to existing pre-clinical seizure-liability testing strategies.
Collapse
Affiliation(s)
| | - Marianne C King
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - David A Nagel
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - H Rheinallt Parri
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Michael D Coleman
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Eric J Hill
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| |
Collapse
|
10
|
Barenys M, Fritsche E. A Historical Perspective on the Use of Stem/Progenitor Cell-Based In Vitro Methods for Neurodevelopmental Toxicity Testing. Toxicol Sci 2018; 165:10-13. [DOI: 10.1093/toxsci/kfy170] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Marta Barenys
- INSA·UB and Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona 08028, Spain
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf 40225, Germany
- Medical Faculty, Heinrich Heine University, Düsseldorf 40225, Germany
| |
Collapse
|
11
|
Buzanska L, Zychowicz M, Kinsner-Ovaskainen A. Bioengineering of the Human Neural Stem Cell Niche: A Regulatory Environment for Cell Fate and Potential Target for Neurotoxicity. Results Probl Cell Differ 2018; 66:207-230. [PMID: 30209661 DOI: 10.1007/978-3-319-93485-3_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human neural stem/progenitor cells of the developing and adult organisms are surrounded by the microenvironment, so-called neurogenic niche. The developmental processes of stem cells, such as survival, proliferation, differentiation, and fate decisions, are controlled by the mutual interactions between cells and the niche components. Such interactions are tissue specific and determined by the biochemical and biophysical properties of the niche constituencies and the presence of other cell types. This dynamic approach of the stem cell niche, when translated into in vitro settings, requires building up "biomimetic" microenvironments resembling natural conditions, where the stem/progenitor cell is provided with diverse extracellular signals exerted by soluble and structural cues, mimicking those found in vivo. The neural stem cell niche is characterized by a unique composition of soluble components including neurotransmitters and trophic factors as well as insoluble extracellular matrix proteins and proteoglycans. Biotechnological innovations provide tools such as a new generation of tunable biomaterials capable of releasing specific signals in a spatially and temporally controlled manner, thus creating in vitro nature-like conditions and, when combined with stem cell-derived tissue specific progenitors, producing differentiated neuronal tissue structures. In addition, substantial progress has been made on the protocols to obtain stem cell-derived cell aggregates such as neurospheres and self-assembled organoids.In this chapter, we have assessed the application of bioengineered human neural stem cell microenvironments to produce in vitro models of different levels of biological complexity for the efficient control of stem cell fate. Examples of biomaterial-supported two-dimensional and three-dimensional (2D and 3D) complex culture systems that provide artificial neural stem cell niches are discussed in the context of their application for basic research and neurotoxicity testing.
Collapse
Affiliation(s)
- Leonora Buzanska
- Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland.
| | - Marzena Zychowicz
- Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Kinsner-Ovaskainen
- European Commission, Joint Research Centre, Directorate for Health Consumers and Reference Materials, Ispra, Italy
| |
Collapse
|
12
|
Neely MD, Davison CA, Aschner M, Bowman AB. From the Cover: Manganese and Rotenone-Induced Oxidative Stress Signatures Differ in iPSC-Derived Human Dopamine Neurons. Toxicol Sci 2017; 159:366-379. [PMID: 28962525 PMCID: PMC5837701 DOI: 10.1093/toxsci/kfx145] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) is the result of complex interactions between genetic and environmental factors. Two chemically distinct environmental stressors relevant to PD are the metal manganese and the pesticide rotenone. Both are thought to exert neurotoxicity at least in part via oxidative stress resulting from impaired mitochondrial activity. Identifying shared mechanism of action may reveal clues towards an understanding of the mechanisms underlying PD pathogenesis. Here we compare the effects of manganese and rotenone in human-induced pluripotent stem cells-derived postmitotic mesencephalic dopamine neurons by assessing several different oxidative stress endpoints. Manganese, but not rotenone caused a concentration and time-dependent increase in intracellular reactive oxygen/nitrogen species measured by quantifying the fluorescence of oxidized chloromethyl 2',7'-dichlorodihydrofluorescein diacetate (DCF) assay. In contrast, rotenone but not manganese caused an increase in cellular isoprostane levels, an indicator of lipid peroxidation. Manganese and rotenone both caused an initial decrease in cellular reduced glutathione; however, glutathione levels remained low in neurons treated with rotenone for 24 h but recovered in manganese-exposed cells. Neurite length, a sensitive indicator of overall neuronal health was adversely affected by rotenone, but not manganese. Thus, our observations suggest that the cellular oxidative stress evoked by these 2 agents is distinct yielding unique oxidative stress signatures across outcome measures. The protective effect of rasagiline, a compound used in the clinic for PD, had negligible impact on any of oxidative stress outcome measures except a subtle significant decrease in manganese-dependent production of reactive oxygen/nitrogen species detected by the DCF assay.
Collapse
Affiliation(s)
- M. Diana Neely
- Department of Pediatrics
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Carrie Ann Davison
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Aaron B. Bowman
- Department of Pediatrics
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232
| |
Collapse
|
13
|
Zagoura D, Canovas-Jorda D, Pistollato F, Bremer-Hoffmann S, Bal-Price A. Evaluation of the rotenone-induced activation of the Nrf2 pathway in a neuronal model derived from human induced pluripotent stem cells. Neurochem Int 2016; 106:62-73. [PMID: 27615060 DOI: 10.1016/j.neuint.2016.09.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/25/2016] [Accepted: 09/06/2016] [Indexed: 01/21/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) are considered as a powerful tool for drug and chemical screening and development of new in vitro testing strategies in the field of toxicology, including neurotoxicity evaluation. These cells are able to expand and efficiently differentiate into different types of neuronal and glial cells as well as peripheral neurons. These human cells-based neuronal models serve as test systems for mechanistic studies on different pathways involved in neurotoxicity. One of the well-known mechanisms that are activated by chemically-induced oxidative stress is the Nrf2 signaling pathway. Therefore, in the current study, we evaluated whether Nrf2 signaling machinery is expressed in human induced pluripotent stem cells (hiPSCs)-derived mixed neuronal/glial culture and if so whether it becomes activated by rotenone-induced oxidative stress mediated by complex I inhibition of mitochondrial respiration. Rotenone was found to induce the activation of Nrf2 signaling particularly at the highest tested concentration (100 nM), as shown by Nrf2 nuclear translocation and the up-regulation of the Nrf2-downstream antioxidant enzymes, NQO1 and SRXN1. Interestingly, exposure to rotenone also increased the number of astroglial cells in which Nrf2 activation may play an important role in neuroprotection. Moreover, rotenone caused cell death of dopaminergic neurons since a decreased percentage of tyrosine hydroxylase (TH+) cells was observed. The obtained results suggest that hiPSC-derived mixed neuronal/glial culture could be a valuable in vitro human model for the establishment of neuronal specific assays in order to link Nrf2 pathway activation (biomarker of oxidative stress) with additional neuronal specific readouts that could be applied to in vitro neurotoxicity evaluation.
Collapse
Affiliation(s)
- Dimitra Zagoura
- Directorate F - Health, Consumers and Reference Materials, Joint Research Centre, Ispra, Italy
| | - David Canovas-Jorda
- Directorate F - Health, Consumers and Reference Materials, Joint Research Centre, Ispra, Italy
| | - Francesca Pistollato
- Directorate F - Health, Consumers and Reference Materials, Joint Research Centre, Ispra, Italy
| | - Susanne Bremer-Hoffmann
- Directorate F - Health, Consumers and Reference Materials, Joint Research Centre, Ispra, Italy
| | - Anna Bal-Price
- Directorate F - Health, Consumers and Reference Materials, Joint Research Centre, Ispra, Italy.
| |
Collapse
|
14
|
Singh S, Srivastava A, Kumar V, Pandey A, Kumar D, Rajpurohit CS, Khanna VK, Yadav S, Pant AB. Stem Cells in Neurotoxicology/Developmental Neurotoxicology: Current Scenario and Future Prospects. Mol Neurobiol 2015; 53:6938-6949. [DOI: 10.1007/s12035-015-9615-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/03/2015] [Indexed: 12/26/2022]
|
15
|
Oh JH, Son MY, Choi MS, Kim S, Choi AY, Lee HA, Kim KS, Kim J, Song CW, Yoon S. Integrative analysis of genes and miRNA alterations in human embryonic stem cells-derived neural cells after exposure to silver nanoparticles. Toxicol Appl Pharmacol 2015; 299:8-23. [PMID: 26551752 DOI: 10.1016/j.taap.2015.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/31/2015] [Accepted: 11/05/2015] [Indexed: 12/31/2022]
Abstract
Given the rapid growth of engineered and customer products made of silver nanoparticles (Ag NPs), understanding their biological and toxicological effects on humans is critically important. The molecular developmental neurotoxic effects associated with exposure to Ag NPs were analyzed at the physiological and molecular levels, using an alternative cell model: human embryonic stem cell (hESC)-derived neural stem/progenitor cells (NPCs). In this study, the cytotoxic effects of Ag NPs (10-200μg/ml) were examined in these hESC-derived NPCs, which have a capacity for neurogenesis in vitro, at 6 and 24h. The results showed that Ag NPs evoked significant toxicity in hESC-derived NPCs at 24h in a dose-dependent manner. In addition, Ag NPs induced cell cycle arrest and apoptosis following a significant increase in oxidative stress in these cells. To further clarify the molecular mechanisms of the toxicological effects of Ag NPs at the transcriptional and post-transcriptional levels, the global expression profiles of genes and miRNAs were analyzed in hESC-derived NPCs after Ag NP exposure. The results showed that Ag NPs induced oxidative stress and dysfunctional neurogenesis at the molecular level in hESC-derived NPCs. Based on this hESC-derived neural cell model, these findings have increased our understanding of the molecular events underlying developmental neurotoxicity induced by Ag NPs in humans.
Collapse
Affiliation(s)
- Jung-Hwa Oh
- Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea; Department of human and environmental toxicology, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Mi-Young Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahangno, Yuseong-gu, Daejeon 34141, Republic of Korea; Department of functional genomics, University of Science & Technology, 217 Gajungro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Mi-Sun Choi
- Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea
| | - Soojin Kim
- Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea
| | - A-Young Choi
- Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea
| | - Hyang-Ae Lee
- Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea; Department of human and environmental toxicology, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Ki-Suk Kim
- Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea; Department of human and environmental toxicology, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Janghwan Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahangno, Yuseong-gu, Daejeon 34141, Republic of Korea; Department of functional genomics, University of Science & Technology, 217 Gajungro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Chang Woo Song
- Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea; Department of human and environmental toxicology, University of Science & Technology, Daejeon 34113, Republic of Korea.
| | - Seokjoo Yoon
- Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea; Department of human and environmental toxicology, University of Science & Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
16
|
Ketamine causes mitochondrial dysfunction in human induced pluripotent stem cell-derived neurons. PLoS One 2015; 10:e0128445. [PMID: 26020236 PMCID: PMC4447382 DOI: 10.1371/journal.pone.0128445] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 04/27/2015] [Indexed: 02/06/2023] Open
Abstract
Purpose Ketamine toxicity has been demonstrated in nonhuman mammalian neurons. To study the toxic effect of ketamine on human neurons, an experimental model of cultured neurons from human induced pluripotent stem cells (iPSCs) was examined, and the mechanism of its toxicity was investigated. Methods Human iPSC-derived dopaminergic neurons were treated with 0, 20, 100 or 500 μM ketamine for 6 and 24 h. Ketamine toxicity was evaluated by quantification of caspase 3/7 activity, reactive oxygen species (ROS) production, mitochondrial membrane potential, ATP concentration, neurotransmitter reuptake activity and NADH/NAD+ ratio. Mitochondrial morphological change was analyzed by transmission electron microscopy and confocal microscopy. Results Twenty-four-hour exposure of iPSC-derived neurons to 500 μM ketamine resulted in a 40% increase in caspase 3/7 activity (P < 0.01), 14% increase in ROS production (P < 0.01), and 81% reduction in mitochondrial membrane potential (P < 0.01), compared with untreated cells. Lower concentration of ketamine (100 μM) decreased the ATP level (22%, P < 0.01) and increased the NADH/NAD+ ratio (46%, P < 0.05) without caspase activation. Transmission electron microscopy showed enhanced mitochondrial fission and autophagocytosis at the 100 μM ketamine concentration, which suggests that mitochondrial dysfunction preceded ROS generation and caspase activation. Conclusions We established an in vitro model for assessing the neurotoxicity of ketamine in iPSC-derived neurons. The present data indicate that the initial mitochondrial dysfunction and autophagy may be related to its inhibitory effect on the mitochondrial electron transport system, which underlies ketamine-induced neural toxicity. Higher ketamine concentration can induce ROS generation and apoptosis in human neurons.
Collapse
|
17
|
Neural Differentiation of Human Pluripotent Stem Cells for Nontherapeutic Applications: Toxicology, Pharmacology, and In Vitro Disease Modeling. Stem Cells Int 2015; 2015:105172. [PMID: 26089911 PMCID: PMC4454762 DOI: 10.1155/2015/105172] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/06/2015] [Accepted: 05/12/2015] [Indexed: 02/08/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) derived from either blastocyst stage embryos (hESCs) or reprogrammed somatic cells (iPSCs) can provide an abundant source of human neuronal lineages that were previously sourced from human cadavers, abortuses, and discarded surgical waste. In addition to the well-known potential therapeutic application of these cells in regenerative medicine, these are also various promising nontherapeutic applications in toxicological and pharmacological screening of neuroactive compounds, as well as for in vitro modeling of neurodegenerative and neurodevelopmental disorders. Compared to alternative research models based on laboratory animals and immortalized cancer-derived human neural cell lines, neuronal cells differentiated from hPSCs possess the advantages of species specificity together with genetic and physiological normality, which could more closely recapitulate in vivo conditions within the human central nervous system. This review critically examines the various potential nontherapeutic applications of hPSC-derived neuronal lineages and gives a brief overview of differentiation protocols utilized to generate these cells from hESCs and iPSCs.
Collapse
|
18
|
Pei Y, Sierra G, Sivapatham R, Swistowski A, Rao MS, Zeng X. A platform for rapid generation of single and multiplexed reporters in human iPSC lines. Sci Rep 2015; 5:9205. [PMID: 25777362 PMCID: PMC4361878 DOI: 10.1038/srep09205] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/25/2015] [Indexed: 12/20/2022] Open
Abstract
Induced pluripotent stem cells (iPSC) are important tools for drug discovery assays and toxicology screens. In this manuscript, we design high efficiency TALEN and ZFN to target two safe harbor sites on chromosome 13 and 19 in a widely available and well-characterized integration-free iPSC line. We show that these sites can be targeted in multiple iPSC lines to generate reporter systems while retaining pluripotent characteristics. We extend this concept to making lineage reporters using a C-terminal targeting strategy to endogenous genes that express in a lineage-specific fashion. Furthermore, we demonstrate that we can develop a master cell line strategy and then use a Cre-recombinase induced cassette exchange strategy to rapidly exchange reporter cassettes to develop new reporter lines in the same isogenic background at high efficiency. Equally important we show that this recombination strategy allows targeting at progenitor cell stages, further increasing the utility of the platform system. The results in concert provide a novel platform for rapidly developing custom single or dual reporter systems for screening assays.
Collapse
Affiliation(s)
- Ying Pei
- Buck Institute for Age Research, Novato, CA
| | | | | | | | | | - Xianmin Zeng
- 1] Buck Institute for Age Research, Novato, CA [2] XCell Science, Novato, CA
| |
Collapse
|
19
|
Smith I, Silveirinha V, Stein JL, de la Torre-Ubieta L, Farrimond JA, Williamson EM, Whalley BJ. Human neural stem cell-derived cultures in three-dimensional substrates form spontaneously functional neuronal networks. J Tissue Eng Regen Med 2015; 11:1022-1033. [PMID: 25712225 DOI: 10.1002/term.2001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/12/2014] [Accepted: 12/17/2014] [Indexed: 12/12/2022]
Abstract
Differentiated human neural stem cells were cultured in an inert three-dimensional (3D) scaffold and, unlike two-dimensional (2D) but otherwise comparable monolayer cultures, formed spontaneously active, functional neuronal networks that responded reproducibly and predictably to conventional pharmacological treatments to reveal functional, glutamatergic synapses. Immunocytochemical and electron microscopy analysis revealed a neuronal and glial population, where markers of neuronal maturity were observed in the former. Oligonucleotide microarray analysis revealed substantial differences in gene expression conferred by culturing in a 3D vs a 2D environment. Notable and numerous differences were seen in genes coding for neuronal function, the extracellular matrix and cytoskeleton. In addition to producing functional networks, differentiated human neural stem cells grown in inert scaffolds offer several significant advantages over conventional 2D monolayers. These advantages include cost savings and improved physiological relevance, which make them better suited for use in the pharmacological and toxicological assays required for development of stem cell-based treatments and the reduction of animal use in medical research. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Imogen Smith
- Cellular and Molecular Neuroscience Group, Department of Pharmacy, University of Reading, UK
| | - Vasco Silveirinha
- Cellular and Molecular Neuroscience Group, Department of Pharmacy, University of Reading, UK
| | - Jason L Stein
- Neurogenetics Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Luis de la Torre-Ubieta
- Neurogenetics Program, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | | - Elizabeth M Williamson
- Cellular and Molecular Neuroscience Group, Department of Pharmacy, University of Reading, UK
| | - Benjamin J Whalley
- Cellular and Molecular Neuroscience Group, Department of Pharmacy, University of Reading, UK
| |
Collapse
|
20
|
Barbosa DJ, Capela JP, de Lourdes Bastos M, Carvalho F. In vitro models for neurotoxicology research. Toxicol Res (Camb) 2015; 4:801-842. [DOI: 10.1039/c4tx00043a] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
The nervous system has a highly complex organization, including many cell types with multiple functions, with an intricate anatomy and unique structural and functional characteristics; the study of its (dys)functionality following exposure to xenobiotics, neurotoxicology, constitutes an important issue in neurosciences.
Collapse
Affiliation(s)
- Daniel José Barbosa
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - João Paulo Capela
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - Maria de Lourdes Bastos
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - Félix Carvalho
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| |
Collapse
|
21
|
Zeng X, Hunsberger JG, Simeonov A, Malik N, Pei Y, Rao M. Concise review: modeling central nervous system diseases using induced pluripotent stem cells. Stem Cells Transl Med 2014; 3:1418-28. [PMID: 25368377 DOI: 10.5966/sctm.2014-0102] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) offer an opportunity to delve into the mechanisms underlying development while also affording the potential to take advantage of a number of naturally occurring mutations that contribute to either disease susceptibility or resistance. Just as with any new field, several models of screening are being explored, and innovators are working on the most efficient methods to overcome the inherent limitations of primary cell screens using iPSCs. In the present review, we provide a background regarding why iPSCs represent a paradigm shift for central nervous system (CNS) disease modeling. We describe the efforts in the field to develop more biologically relevant CNS disease models, which should provide screening assays useful for the pharmaceutical industry. We also provide some examples of successful uses for iPSC-based screens and suggest that additional development could revolutionize the field of drug discovery. The development and implementation of these advanced iPSC-based screens will create a more efficient disease-specific process underpinned by the biological mechanism in a patient- and disease-specific manner rather than by trial-and-error. Moreover, with careful and strategic planning, shared resources can be developed that will enable exponential advances in the field. This will undoubtedly lead to more sensitive and accurate screens for early diagnosis and allow the identification of patient-specific therapies, thus, paving the way to personalized medicine.
Collapse
Affiliation(s)
- Xianmin Zeng
- XCell Science Inc., Novato, California, USA; Buck Institute for Research on Aging, Novato, California, USA; Laboratory of Stem Cell Biology, NIH Center for Regenerative Medicine, Bethesda, Maryland, USA; National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA; New York Stem Cell Foundation, New York, New York, USA
| | - Joshua G Hunsberger
- XCell Science Inc., Novato, California, USA; Buck Institute for Research on Aging, Novato, California, USA; Laboratory of Stem Cell Biology, NIH Center for Regenerative Medicine, Bethesda, Maryland, USA; National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA; New York Stem Cell Foundation, New York, New York, USA
| | - Anton Simeonov
- XCell Science Inc., Novato, California, USA; Buck Institute for Research on Aging, Novato, California, USA; Laboratory of Stem Cell Biology, NIH Center for Regenerative Medicine, Bethesda, Maryland, USA; National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA; New York Stem Cell Foundation, New York, New York, USA
| | - Nasir Malik
- XCell Science Inc., Novato, California, USA; Buck Institute for Research on Aging, Novato, California, USA; Laboratory of Stem Cell Biology, NIH Center for Regenerative Medicine, Bethesda, Maryland, USA; National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA; New York Stem Cell Foundation, New York, New York, USA
| | - Ying Pei
- XCell Science Inc., Novato, California, USA; Buck Institute for Research on Aging, Novato, California, USA; Laboratory of Stem Cell Biology, NIH Center for Regenerative Medicine, Bethesda, Maryland, USA; National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA; New York Stem Cell Foundation, New York, New York, USA
| | - Mahendra Rao
- XCell Science Inc., Novato, California, USA; Buck Institute for Research on Aging, Novato, California, USA; Laboratory of Stem Cell Biology, NIH Center for Regenerative Medicine, Bethesda, Maryland, USA; National Center for Advancing Translational Sciences, NIH, Bethesda, Maryland, USA; New York Stem Cell Foundation, New York, New York, USA
| |
Collapse
|
22
|
Kumar KK, Lowe EW, Aboud AA, Neely MD, Redha R, Bauer JA, Odak M, Weaver CD, Meiler J, Aschner M, Bowman AB. Cellular manganese content is developmentally regulated in human dopaminergic neurons. Sci Rep 2014; 4:6801. [PMID: 25348053 PMCID: PMC4210885 DOI: 10.1038/srep06801] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 10/08/2014] [Indexed: 12/29/2022] Open
Abstract
Manganese (Mn) is both an essential biological cofactor and neurotoxicant. Disruption of Mn biology in the basal ganglia has been implicated in the pathogenesis of neurodegenerative disorders, such as parkinsonism and Huntington's disease. Handling of other essential metals (e.g. iron and zinc) occurs via complex intracellular signaling networks that link metal detection and transport systems. However, beyond several non-selective transporters, little is known about the intracellular processes regulating neuronal Mn homeostasis. We hypothesized that small molecules that modulate intracellular Mn could provide insight into cell-level Mn regulatory mechanisms. We performed a high throughput screen of 40,167 small molecules for modifiers of cellular Mn content in a mouse striatal neuron cell line. Following stringent validation assays and chemical informatics, we obtained a chemical 'toolbox' of 41 small molecules with diverse structure-activity relationships that can alter intracellular Mn levels under biologically relevant Mn exposures. We utilized this toolbox to test for differential regulation of Mn handling in human floor-plate lineage dopaminergic neurons, a lineage especially vulnerable to environmental Mn exposure. We report differential Mn accumulation between developmental stages and stage-specific differences in the Mn-altering activity of individual small molecules. This work demonstrates cell-level regulation of Mn content across neuronal differentiation.
Collapse
Affiliation(s)
- Kevin K Kumar
- 1] Department of Neurology, Vanderbilt University Medical Center, Nashville, TN [2] Medical Scientist Training Program, Vanderbilt University Medical Center, Nashville, TN [3] Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Edward W Lowe
- Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN
| | - Asad A Aboud
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| | - M Diana Neely
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| | - Rey Redha
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN
| | - Joshua A Bauer
- 1] Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN [2] Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN
| | - Mihir Odak
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| | - C David Weaver
- 1] Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN [2] Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN
| | - Jens Meiler
- 1] Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN [2] Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN
| | - Michael Aschner
- Departments of Molecular Pharmacology, Neuroscience, and Pediatrics, Albert Einstein College of Medicine, Bronx NY
| | - Aaron B Bowman
- 1] Department of Neurology, Vanderbilt University Medical Center, Nashville, TN [2] Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
23
|
Induced Pluripotent Stem Cells for Disease Modeling and Drug Discovery in Neurodegenerative Diseases. Mol Neurobiol 2014; 52:244-55. [DOI: 10.1007/s12035-014-8867-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 08/14/2014] [Indexed: 12/25/2022]
|
24
|
Pistollato F, Louisse J, Scelfo B, Mennecozzi M, Accordi B, Basso G, Gaspar JA, Zagoura D, Barilari M, Palosaari T, Sachinidis A, Bremer-Hoffmann S. Development of a pluripotent stem cell derived neuronal model to identify chemically induced pathway perturbations in relation to neurotoxicity: effects of CREB pathway inhibition. Toxicol Appl Pharmacol 2014; 280:378-88. [PMID: 25150140 DOI: 10.1016/j.taap.2014.08.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 07/01/2014] [Accepted: 08/11/2014] [Indexed: 01/09/2023]
Abstract
According to the advocated paradigm shift in toxicology, acquisition of knowledge on the mechanisms underlying the toxicity of chemicals, such as perturbations of biological pathways, is of primary interest. Pluripotent stem cells (PSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), offer a unique opportunity to derive physiologically relevant human cell types to measure molecular and cellular effects of such pathway modulations. Here we compared the neuronal differentiation propensity of hESCs and hiPSCs with the aim to develop novel hiPSC-based tools for measuring pathway perturbation in relation to molecular and cellular effects in vitro. Among other fundamental pathways, also, the cAMP responsive element binding protein (CREB) pathway was activated in our neuronal models and gave us the opportunity to study time-dependent effects elicited by chemical perturbations of the CREB pathway in relation to cellular effects. We show that the inhibition of the CREB pathway, using 2-naphthol-AS-E-phosphate (KG-501), induced an inhibition of neurite outgrowth and synaptogenesis, as well as a decrease of MAP2(+) neuronal cells. These data indicate that a CREB pathway inhibition can be related to molecular and cellular effects that may be relevant for neurotoxicity testing, and, thus, qualify the use of our hiPSC-derived neuronal model for studying chemical-induced neurotoxicity resulting from pathway perturbations.
Collapse
Affiliation(s)
| | - Jochem Louisse
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Bibiana Scelfo
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Milena Mennecozzi
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Benedetta Accordi
- Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy
| | - Giuseppe Basso
- Oncohematology Laboratory, Department of Woman and Child Health, University of Padova, Padova, Italy
| | - John Antonydas Gaspar
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Dimitra Zagoura
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Manuela Barilari
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Taina Palosaari
- Institute for Health and Consumer Protection (IHCP), JRC, Ispra, Italy
| | - Agapios Sachinidis
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | | |
Collapse
|
25
|
Human Stem/Progenitor Cell-Based Assays for Neurodevelopmental Toxicity Testing. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2014. [DOI: 10.1007/978-1-4939-0521-8_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
26
|
Westerink RH. Do we really want to REACH out to in vitro? Neurotoxicology 2013; 39:169-72. [DOI: 10.1016/j.neuro.2013.10.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/02/2013] [Accepted: 10/02/2013] [Indexed: 11/24/2022]
|
27
|
Mori H, Hara M. Cultured stem cells as tools for toxicological assays. J Biosci Bioeng 2013; 116:647-52. [DOI: 10.1016/j.jbiosc.2013.05.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 05/17/2013] [Accepted: 05/20/2013] [Indexed: 12/29/2022]
|
28
|
Powers CM, Bale AS, Kraft AD, Makris SL, Trecki J, Cowden J, Hotchkiss A, Gillespie PA. Developmental neurotoxicity of engineered nanomaterials: identifying research needs to support human health risk assessment. Toxicol Sci 2013; 134:225-42. [PMID: 23708405 DOI: 10.1093/toxsci/kft109] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Increasing use of engineered nanomaterials (ENM) in consumer products and commercial applications has helped drive a rise in research related to the environmental health and safety (EHS) of these materials. Within the cacophony of information on ENM EHS to date are data indicating that these materials may be neurotoxic in adult animals. Evidence of elevated inflammatory responses, increased oxidative stress levels, alterations in neuronal function, and changes in cell morphology in adult animals suggests that ENM exposure during development could elicit developmental neurotoxicity (DNT), especially considering the greater vulnerability of the developing brain to some toxic insults. In this review, we examine current findings related to developmental neurotoxic effects of ENM in the context of identifying research gaps for future risk assessments. The basic risk assessment paradigm is presented, with an emphasis on problem formulation and assessments of exposure, hazard, and dose response for DNT. Limited evidence suggests that in utero and postpartum exposures are possible, while fewer than 10 animal studies have evaluated DNT, with results indicating changes in synaptic plasticity, gene expression, and neurobehavior. Based on the available information, we use current testing guidelines to highlight research gaps that may inform ENM research efforts to develop data for higher throughput methods and future risk assessments for DNT. Although the available evidence is not strong enough to reach conclusions about DNT risk from ENM exposure, the data indicate that consideration of ENM developmental neurotoxic potential is warranted.
Collapse
Affiliation(s)
- Christina M Powers
- Office of Research and Development, National Center for Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Human induced pluripotent stem cells and their use in drug discovery for toxicity testing. Toxicol Lett 2013; 219:49-58. [PMID: 23470867 DOI: 10.1016/j.toxlet.2013.02.020] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 02/21/2013] [Accepted: 02/23/2013] [Indexed: 12/23/2022]
Abstract
Predicting human safety risks of novel xenobiotics remains a major challenge, partly due to the limited availability of human cells to evaluate tissue-specific toxicity. Recent progress in the production of human induced pluripotent stem cells (hiPSCs) may fill this gap. hiPSCs can be continuously expanded in culture in an undifferentiated state and then differentiated to form most cell types. Thus, it is becoming technically feasible to generate large quantities of human cell types and, in combination with relatively new detection methods, to develop higher-throughput in vitro assays that quantify tissue-specific biological properties. Indeed, the first wave of large scale hiSC-differentiated cell types including patient-derived hiPSCS are now commercially available. However, significant improvements in hiPSC production and differentiation processes are required before cell-based toxicity assays that accurately reflect mature tissue phenotypes can be delivered and implemented in a cost-effective manner. In this review, we discuss the promising alignment of hiPSCs and recently emerging technologies to quantify tissue-specific functions. We emphasize liver, cardiovascular, and CNS safety risks and highlight limitations that must be overcome before routine screening for toxicity pathways in hiSC-derived cells can be established.
Collapse
|
30
|
Aboud AA, Tidball AM, Kumar KK, Neely MD, Ess KC, Erikson KM, Bowman AB. Genetic risk for Parkinson's disease correlates with alterations in neuronal manganese sensitivity between two human subjects. Neurotoxicology 2012; 33:1443-1449. [PMID: 23099318 DOI: 10.1016/j.neuro.2012.10.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 10/01/2012] [Accepted: 10/15/2012] [Indexed: 01/16/2023]
Abstract
Manganese (Mn) is an environmental risk factor for Parkinson's disease (PD). Recessive inheritance of PARK2 mutations is strongly associated with early onset PD (EOPD). It is widely assumed that the influence of PD environmental risk factors may be enhanced by the presence of PD genetic risk factors in the genetic background of individuals. However, such interactions may be difficult to predict owing to the complexities of genetic and environmental interactions. Here we examine the potential of human induced pluripotent stem (iPS) cell-derived early neural progenitor cells (NPCs) to model differences in Mn neurotoxicity between a control subject (CA) with no known PD genetic risk factors and a subject (SM) with biallelic loss-of-function mutations in PARK2 and family history of PD but no evidence of PD by neurological exam. Human iPS cells were generated from primary dermal fibroblasts of both subjects. We assessed several outcome measures associated with Mn toxicity and PD. No difference in sensitivity to Mn cytotoxicity or mitochondrial fragmentation was observed between SM and CA NPCs. However, we found that Mn exposure was associated with significantly higher reactive oxygen species (ROS) generation in SM compared to CA NPCs despite significantly less intracellular Mn accumulation. Thus, this report offers the first example of human subject-specific differences in PD-relevant environmental health related phenotypes that are consistent with pathogenic interactions between known genetic and environmental risk factors for PD.
Collapse
Affiliation(s)
- Asad A Aboud
- Vanderbilt University Medical Center, Department of Neurology and Vanderbilt Kennedy Center, Nashville, TN 37232-8552, USA
| | - Andrew M Tidball
- Vanderbilt University Medical Center, Department of Neurology and Vanderbilt Kennedy Center, Nashville, TN 37232-8552, USA; Vanderbilt Brain Institute, Nashville, TN 37232-8552, USA
| | - Kevin K Kumar
- Vanderbilt University Medical Center, Department of Neurology and Vanderbilt Kennedy Center, Nashville, TN 37232-8552, USA; Vanderbilt Brain Institute, Nashville, TN 37232-8552, USA; Vanderbilt Medical Scientist Training Program, Nashville, TN 37232-8552, USA
| | - M Diana Neely
- Vanderbilt University Medical Center, Department of Neurology and Vanderbilt Kennedy Center, Nashville, TN 37232-8552, USA; Vanderbilt Brain Institute, Nashville, TN 37232-8552, USA
| | - Kevin C Ess
- Vanderbilt University Medical Center, Department of Neurology and Vanderbilt Kennedy Center, Nashville, TN 37232-8552, USA; Vanderbilt Brain Institute, Nashville, TN 37232-8552, USA; Vanderbilt Center for Stem Cell Biology and The Department of Pediatrics, Nashville, TN 37232-8552, USA
| | - Keith M Erikson
- University of North Carolina-Greensboro, Nutrition Department, Greensboro, NC 27402-6107, USA
| | - Aaron B Bowman
- Vanderbilt University Medical Center, Department of Neurology and Vanderbilt Kennedy Center, Nashville, TN 37232-8552, USA; Vanderbilt Brain Institute, Nashville, TN 37232-8552, USA; Vanderbilt Center for Stem Cell Biology and The Department of Pediatrics, Nashville, TN 37232-8552, USA; Vanderbilt Center in Molecular Toxicology, Vanderbilt University, Nashville, TN 37232-8552, USA.
| |
Collapse
|
31
|
Schwartzer JJ, Koenig CM, Berman RF. Using mouse models of autism spectrum disorders to study the neurotoxicology of gene-environment interactions. Neurotoxicol Teratol 2012; 36:17-35. [PMID: 23010509 DOI: 10.1016/j.ntt.2012.08.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 08/28/2012] [Accepted: 08/30/2012] [Indexed: 10/27/2022]
Abstract
To better study the role of genetics in autism, mouse models have been developed which mimic the genetics of specific autism spectrum and related disorders. These models have facilitated research on the role genetic susceptibility factors in the pathogenesis of autism in the absence of environmental factors. Inbred mouse strains have been similarly studied to assess the role of environmental agents on neurodevelopment, typically without the complications of genetic heterogeneity of the human population. What has not been as actively pursued, however, is the methodical study of the interaction between these factors (e.g., gene and environmental interactions in neurodevelopment). This review suggests that a genetic predisposition paired with exposure to environmental toxicants plays an important role in the etiology of neurodevelopmental disorders including autism, and may contribute to the largely unexplained rise in the number of children diagnosed with autism worldwide. Specifically, descriptions of the major mouse models of autism and toxic mechanisms of prevalent environmental chemicals are provided followed by a discussion of current and future research strategies to evaluate the role of gene and environment interactions in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Jared J Schwartzer
- Department of Psychiatry and Behavioral Sciences, MIND Institute, University of California, Davis, Davis, CA 95618, United States.
| | | | | |
Collapse
|
32
|
|