1
|
Suazo KF, Mishra V, Maity S, Auger SA, Justyna K, Petre AM, Ottoboni L, Ongaro J, Corti SP, Lotti F, Przedborski S, Distefano MD. Improved synthesis and application of an alkyne-functionalized isoprenoid analogue to study the prenylomes of motor neurons, astrocytes and their stem cell progenitors. Bioorg Chem 2024; 147:107365. [PMID: 38636436 PMCID: PMC11653755 DOI: 10.1016/j.bioorg.2024.107365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024]
Abstract
Protein prenylation is one example of a broad class of post-translational modifications where proteins are covalently linked to various hydrophobic moieties. To globally identify and monitor levels of all prenylated proteins in a cell simultaneously, our laboratory and others have developed chemical proteomic approaches that rely on the metabolic incorporation of isoprenoid analogues bearing bio-orthogonal functionality followed by enrichment and subsequent quantitative proteomic analysis. Here, several improvements in the synthesis of the alkyne-containing isoprenoid analogue C15AlkOPP are reported to improve synthetic efficiency. Next, metabolic labeling with C15AlkOPP was optimized to obtain useful levels of metabolic incorporation of the probe in several types of primary cells. Those conditions were then used to study the prenylomes of motor neurons (ES-MNs), astrocytes (ES-As), and their embryonic stem cell progenitors (ESCs), which allowed for the identification of 54 prenylated proteins from ESCs, 50 from ES-MNs, and 84 from ES-As, representing all types of prenylation. Bioinformatic analysis revealed specific enriched pathways, including nervous system development, chemokine signaling, Rho GTPase signaling, and adhesion. Hierarchical clustering showed that most enriched pathways in all three cell types are related to GTPase activity and vesicular transport. In contrast, STRING analysis showed significant interactions in two populations that appear to be cell type dependent. The data provided herein demonstrates that robust incorporation of C15AlkOPP can be obtained in ES-MNs and related primary cells purified via magnetic-activated cell sorting allowing the identification and quantification of numerous prenylated proteins. These results suggest that metabolic labeling with C15AlkOPP should be an effective approach for investigating the role of prenylated proteins in primary cells in both normal cells and disease pathologies, including ALS.
Collapse
Affiliation(s)
- Kiall F Suazo
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Vartika Mishra
- Center for Motor Neuron Biology and Diseases, Department of Neurology. Columbia University Irving Medical Center. New York, NY 10032, USA; Department of Pathology & Cell Biology. Columbia University Irving Medical Center. New York, NY 10032, USA.
| | - Sanjay Maity
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Shelby A Auger
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Katarzyna Justyna
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Alexandru M Petre
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Linda Ottoboni
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy.
| | - Jessica Ongaro
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania P Corti
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Francesco Lotti
- Center for Motor Neuron Biology and Diseases, Department of Neurology. Columbia University Irving Medical Center. New York, NY 10032, USA; Department of Pathology & Cell Biology. Columbia University Irving Medical Center. New York, NY 10032, USA.
| | - Serge Przedborski
- Center for Motor Neuron Biology and Diseases, Department of Neurology. Columbia University Irving Medical Center. New York, NY 10032, USA; Department of Pathology & Cell Biology. Columbia University Irving Medical Center. New York, NY 10032, USA; Department of Neuroscience, Pathology, and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Mark D Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
2
|
Suazo KF, Mishra V, Maity S, Auger SA, Justyna K, Petre A, Ottoboni L, Ongaro J, Corti SP, Lotti F, Przedborski S, Distefano MD. Improved synthesis and application of an alkyne-functionalized isoprenoid analogue to study the prenylomes of motor neurons, astrocytes and their stem cell progenitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.03.583211. [PMID: 38496415 PMCID: PMC10942399 DOI: 10.1101/2024.03.03.583211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Protein prenylation is one example of a broad class of post-translational modifications where proteins are covalently linked to various hydrophobic moieties. To globally identify and monitor levels of all prenylated proteins in a cell simultaneously, our laboratory and others have developed chemical proteomic approaches that rely on the metabolic incorporation of isoprenoid analogues bearing bio-orthogonal functionality followed by enrichment and subsequent quantitative proteomic analysis. Here, several improvements in the synthesis of the alkyne-containing isoprenoid analogue C15AlkOPP are reported to improve synthetic efficiency. Next, metabolic labeling with C15AlkOPP was optimized to obtain useful levels of metabolic incorporation of the probe in several types of primary cells. Those conditions were then used to study the prenylomes of motor neurons (ES-MNs), astrocytes (ES-As), and their embryonic stem cell progenitors (ESCs), which allowed for the identification of 54 prenylated proteins from ESCs, 50 from ES-MNs and 84 from ES-As, representing all types of prenylation. Bioinformatic analysis revealed specific enriched pathways, including nervous system development, chemokine signaling, Rho GTPase signaling, and adhesion. Hierarchical clustering showed that most enriched pathways in all three cell types are related to GTPase activity and vesicular transport. In contrast, STRING analysis showed significant interactions in two populations that appear to be cell type dependent. The data provided herein demonstrates that robust incorporation of C15AlkOPP can be obtained in ES-MNs and related primary cells purified via magnetic-activated cell sorting allowing the identification and quantification of numerous prenylated proteins. These results suggest that metabolic labeling with C15AlkOPP should be an effective approach for investigating the role of prenylated proteins in primary cells in both normal cells and disease pathologies, including ALS.
Collapse
Affiliation(s)
- Kiall F Suazo
- Department of Chemistry, University of Minnesota, Minneapolis, MN USA 55455
| | - Vartika Mishra
- Center for Motor Neuron Biology and Diseases, Department of Neurology. Columbia University Irving Medical Center. New York, NY 10032
- Department of Pathology & Cell Biology. Columbia University Irving Medical Center. New York, NY 10032
| | - Sanjay Maity
- Department of Chemistry, University of Minnesota, Minneapolis, MN USA 55455
| | - Shelby A Auger
- Department of Chemistry, University of Minnesota, Minneapolis, MN USA 55455
| | - Katarzyna Justyna
- Department of Chemistry, University of Minnesota, Minneapolis, MN USA 55455
| | - Alex Petre
- Department of Chemistry, University of Minnesota, Minneapolis, MN USA 55455
| | - Linda Ottoboni
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Jessica Ongaro
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania P Corti
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Lotti
- Center for Motor Neuron Biology and Diseases, Department of Neurology. Columbia University Irving Medical Center. New York, NY 10032
- Department of Pathology & Cell Biology. Columbia University Irving Medical Center. New York, NY 10032
| | - Serge Przedborski
- Center for Motor Neuron Biology and Diseases, Department of Neurology. Columbia University Irving Medical Center. New York, NY 10032
- Department of Pathology & Cell Biology. Columbia University Irving Medical Center. New York, NY 10032
- Department of Neuroscience, Pathology, and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - Mark D Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, MN USA 55455
| |
Collapse
|
3
|
Välimäki NN, Bakreen A, Häkli S, Dhungana H, Keuters MH, Dunlop Y, Koskuvi M, Keksa-Goldsteine V, Oksanen M, Jäntti H, Lehtonen Š, Malm T, Koistinaho J, Jolkkonen J. Astrocyte Progenitors Derived From Patients With Alzheimer Disease Do Not Impair Stroke Recovery in Mice. Stroke 2022; 53:3192-3201. [DOI: 10.1161/strokeaha.122.039700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND:
Species-specific differences in astrocytes and their Alzheimer disease-associated pathology may influence cellular responses to other insults. Herein, human glial chimeric mice were generated to evaluate how Alzheimer disease predisposing genetic background in human astrocytes contributes to behavioral outcome and brain pathology after cortical photothrombotic ischemia.
METHODS:
Neonatal (P0) immunodeficient mice of both sexes were transplanted with induced pluripotent stem cell–derived astrocyte progenitors from Alzheimer disease patients carrying
PSEN1
exon 9 deletion (
P
SEN1
Δ
E
9), with isogenic controls, with cells from a healthy donor, or with mouse astrocytes or vehicle. After 14 months, a photothrombotic lesion was produced with Rose Bengal in the motor cortex. Behavior was assessed before ischemia and 1 and 4 weeks after the induction of stroke, followed by tissue perfusion for histology.
RESULTS:
Open field, cylinder, and grid-walking tests showed a persistent locomotor and sensorimotor impairment after ischemia and female mice had larger infarct sizes; yet, these were not affected by astrocytes with
P
SEN1
Δ
E
9 background. Staining for human nuclear antigen confirmed that human cells successfully engrafted throughout the mouse brain. However, only a small number of human cells were positive for astrocytic marker GFAP (glial fibrillary acidic protein), mostly located in the corpus callosum and retaining complex human-specific morphology with longer processes compared with host counterparts. While host astrocytes formed the glial scar, human astrocytes were scattered in small numbers close to the lesion boundary. Aβ (beta-amyloid) deposits were not present in
P
SEN1
ΔE
9 astrocyte-transplanted mice.
CONCLUSIONS:
Transplanted human cells survived and distributed widely in the host brain but had no impact on severity of ischemic damage after cortical photothrombosis in chimeric mice. Only a small number of transplanted human astrocytes acquired GFAP-positive glial phenotype or migrated toward the ischemic lesion forming glial scar.
P
SEN1
ΔE
9 astrocytes did not impair behavioral recovery after experimental stroke.
Collapse
Affiliation(s)
- Nelli-Noora Välimäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Abdulhameed Bakreen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Sara Häkli
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Hiramani Dhungana
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
- Neuroscience Center, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Finland (H.D., M.H.K., M.K., Š.L., J.K.)
| | - Meike H. Keuters
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
- Neuroscience Center, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Finland (H.D., M.H.K., M.K., Š.L., J.K.)
| | - Yannick Dunlop
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Marja Koskuvi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
- Neuroscience Center, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Finland (H.D., M.H.K., M.K., Š.L., J.K.)
| | - Velta Keksa-Goldsteine
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Minna Oksanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Henna Jäntti
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Šárka Lehtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
- Neuroscience Center, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Finland (H.D., M.H.K., M.K., Š.L., J.K.)
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
- Neuroscience Center, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Finland (H.D., M.H.K., M.K., Š.L., J.K.)
| | - Jukka Jolkkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland (N.-N.V., A.B., S.H., H.D., M.H.K., Y.D., M.K., V.K.-G., M.O., H.J., Š.L., T.M., J.K., J.J.)
| |
Collapse
|
4
|
Chiareli RA, Carvalho GA, Marques BL, Mota LS, Oliveira-Lima OC, Gomes RM, Birbrair A, Gomez RS, Simão F, Klempin F, Leist M, Pinto MCX. The Role of Astrocytes in the Neurorepair Process. Front Cell Dev Biol 2021; 9:665795. [PMID: 34113618 PMCID: PMC8186445 DOI: 10.3389/fcell.2021.665795] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/29/2021] [Indexed: 12/17/2022] Open
Abstract
Astrocytes are highly specialized glial cells responsible for trophic and metabolic support of neurons. They are associated to ionic homeostasis, the regulation of cerebral blood flow and metabolism, the modulation of synaptic activity by capturing and recycle of neurotransmitters and maintenance of the blood-brain barrier. During injuries and infections, astrocytes act in cerebral defense through heterogeneous and progressive changes in their gene expression, morphology, proliferative capacity, and function, which is known as reactive astrocytes. Thus, reactive astrocytes release several signaling molecules that modulates and contributes to the defense against injuries and infection in the central nervous system. Therefore, deciphering the complex signaling pathways of reactive astrocytes after brain damage can contribute to the neuroinflammation control and reveal new molecular targets to stimulate neurorepair process. In this review, we present the current knowledge about the role of astrocytes in brain damage and repair, highlighting the cellular and molecular bases involved in synaptogenesis and neurogenesis. In addition, we present new approaches to modulate the astrocytic activity and potentiates the neurorepair process after brain damage.
Collapse
Affiliation(s)
| | | | | | - Lennia Soares Mota
- Department of Pharmacology, Federal University of Goias, Goiânia, Brazil
| | | | | | - Alexander Birbrair
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Renato Santiago Gomez
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fabrício Simão
- Research Division, Vascular Cell Biology, Joslin Diabetes Center and Harvard Medical School, Boston, MA, United States
| | | | - Marcel Leist
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | | |
Collapse
|
5
|
Juneja DS, Nasuto S, Delivopoulos E. Deriving Functional Astrocytes from Mouse Embryonic Stem Cells with a Fast and Efficient Protocol. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:2994-2996. [PMID: 31946518 DOI: 10.1109/embc.2019.8857058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A growing number of studies highlight the structural and functional diversity of astrocytes throughout the central nervous system. These cells are now seen as heterogeneous as neurons and are implicated in a number of neurological and psychiatric diseases. Efficient generation of diverse subtypes of astrocytes can be a useful tool in investigating synaptogenesis and patterns of activity in developing neural networks. In this study, we developed a protocol for the fast and efficient differentiation of astrocytes from mouse embryonic stem cells, as evidenced by the upregulation of genes related to astrocytic development (Gfap, Aldh1l1). Generated astrocytes exhibit phenotypic diversity, which is demonstrated by the variant expression of markers such as GFAP, ALDH1L1, AQP4 and S100β, amongst subgroups within the same cell population. In addition, astrocytes exhibited differential calcium transients upon stimulation with ATP. Our protocol will facilitate investigations, regarding the involvement of astrocytes in the structural and functional connectivity of neural networks.
Collapse
|
6
|
Juneja DS, Nasuto S, Delivopoulos E. Fast and Efficient Differentiation of Mouse Embryonic Stem Cells Into ATP-Responsive Astrocytes. Front Cell Neurosci 2020; 13:579. [PMID: 32038173 PMCID: PMC6985097 DOI: 10.3389/fncel.2019.00579] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/16/2019] [Indexed: 12/25/2022] Open
Abstract
Astrocytes are multifunctional cells in the CNS, involved in the regulation of neurovascular coupling, the modulation of electrolytes, and the cycling of neurotransmitters at synapses. Induction of astrocytes from stem cells remains a largely underdeveloped area, as current protocols are time consuming, lack granularity in astrocytic subtype generation, and often are not as efficient as neural induction methods. In this paper we present an efficient method to differentiate astrocytes from mouse embryonic stem cells. Our technique uses a cell suspension protocol to produce embryoid bodies (EBs) that are neurally inducted and seeded onto laminin coated surfaces. Plated EBs attach to the surface and release migrating cells to their surrounding environment, which are further inducted into the astrocytic lineage, through an optimized, heparin-based media. Characterization and functional assessment of the cells consists of immunofluorescent labeling for specific astrocytic proteins and sensitivity to adenosine triphosphate (ATP) stimulation. Our experimental results show that even at the earliest stages of the protocol, cells are positive for astrocytic markers (GFAP, ALDH1L1, S100β, and GLAST) with variant expression patterns and purinergic receptors (P2Y). Generated astrocytes also exhibit differential Ca2+ transients upon stimulation with ATP, which evolve over the differentiation period. Metabotropic purinoceptors P2Y1R are expressed and we offer preliminary evidence that metabotropic purinoceptors contribute to Ca2+ transients. Our protocol is simple, efficient and fast, facilitating its use in multiple investigations, particularly in vitro studies of engineered neural networks.
Collapse
|
7
|
Sachana M, Bal-Price A, Crofton KM, Bennekou SH, Shafer TJ, Behl M, Terron A. International Regulatory and Scientific Effort for Improved Developmental Neurotoxicity Testing. Toxicol Sci 2019; 167:45-57. [PMID: 30476307 DOI: 10.1093/toxsci/kfy211] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The Organisation for Economic Co-Operation and Development (OECD) coordinates international efforts to enhance developmental neurotoxicity (DNT) testing. In most regulatory sectors, including the ones dealing with pesticides and industrial chemicals registration, historical use of the in vivo DNT test guideline has been limited. Current challenges include a lack of DNT data and mechanistic information for thousands of chemicals, and difficulty in interpreting results. A series of workshops in the last decade has paved the way for a consensus among stakeholders that there is need for a DNT testing battery that relies on in vitro endpoints (proliferation, differentiation, synaptogenesis, etc.) and is complemented by alternative species (eg, zebrafish) assays. Preferably, a battery of in vitro and alternative assays should be anchored toward mechanistic relevance for applying an integrated approach for testing and assessment (IATA) framework. Specific activities have been initiated to facilitate this OECD project: the collation of available DNT in vitro methods and their scoring for readiness; the selection of these methods to form a DNT testing battery; the generation of a reference set of chemicals that will be tested using the battery; the case studies exemplifying how DNT in vitro data can be interpreted; and the development of an OECD guidance document. This manuscript highlights these international efforts and activities.
Collapse
Affiliation(s)
- Magdalini Sachana
- Organisation for Economic Co-Operation and Development (OECD), 75775 Paris Cedex 16, France
| | - Anna Bal-Price
- European Commission Joint Research Centre, Health, Consumers and Reference Materials, Unit Chemicals Safety and Alternative Methods I-21027 Ispra (VA), Italy
| | | | - Susanne H Bennekou
- Danish Environmental Protection Agency, Haraldsgade 53, DK - 2100, Copenhagen, Denmark
| | - Timothy J Shafer
- U.S. Environmental Protection Agency (EPA), Office of Research and Development, Research Triangle Park, North Carolina 27711, USA
| | - Mamta Behl
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences Research Triangle Park, North Carolina, 27709 USA
| | - Andrea Terron
- European Food Safety Authority, Via Carlo Magno, 1A, 43126, Parma, Italy
| |
Collapse
|
8
|
Byun JS, Lee CO, Oh M, Cha D, Kim WK, Oh KJ, Bae KH, Lee SC, Han BS. Rapid differentiation of astrocytes from human embryonic stem cells. Neurosci Lett 2019; 716:134681. [PMID: 31836568 DOI: 10.1016/j.neulet.2019.134681] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 12/04/2019] [Accepted: 12/07/2019] [Indexed: 01/24/2023]
Abstract
Astrocytes are abundant cells in the brain and have vital roles in various brain functions that include biochemical support of endothelial cells, supplying nutrients to the nervous tissue, maintaining the extracellular ion balance, etc. In developing nervous tissue, the differentiation of astrocytes occurs later compared to neurons. It takes more time and more techniques to obtain mature and pure astrocytes in vitro. In this study, a protocol was developed to culture mature and pure astrocytes from human embryonic stem cells (hESCs). To obtain a high quantity and quality of differentiated astrocytes, first, we efficiently generated neural progenitor cells (NPCs) derived from hESCs through the process of embryoid body (EB) formation by adding SB431542 and LDN193189 and neurosphere step. In the astrocyte differentiation stage, the efficiency of astrocyte differentiation was increased using progenitor medium containing EGF and heparin and astrocyte defined medium containing ciliary neurotrophic factor (CNTF). The cell properties were checked with immunocytochemistry and western blot using antibodies for astrocyte-specific marker proteins. From the FACS analysis, we found that the percentage of astrocytes among the cells differentiated from NPCs was over 80%. To validate the functional properties of the astrocytes, we checked IL-6 release from the astrocytes and support of synaptic formation in a co-culture with neurons. Taken altogether, with our protocol, we obtained mature astrocytes within 4 weeks from NPCs and 6 weeks from hESCs.
Collapse
Affiliation(s)
- Jeong Su Byun
- Research Center for Biodefence, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea.
| | - Cheon Ok Lee
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea.
| | - Mihee Oh
- Research Center for Biodefence, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Dongwook Cha
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, Republic of Korea
| | - Won-Kon Kim
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, Republic of Korea
| | - Kyung-Jin Oh
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - Kwang-Hee Bae
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, Republic of Korea
| | - Sang-Chun Lee
- Research Center for Metabolic Regulation, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, Republic of Korea
| | - Baek-Soo Han
- Research Center for Biodefence, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, Republic of Korea.
| |
Collapse
|
9
|
Atkinson-Dell R, Mohamet L. Induced Pluripotent Stem Cell-Derived Astroglia: A New Tool for Research Towards the Treatment of Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1175:383-405. [PMID: 31583596 DOI: 10.1007/978-981-13-9913-8_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite over a century of research into Alzheimer's disease (AD), progress in understanding the complex aetiology has been hindered, in part, by a lack of human, disease relevant, cellular models, reflected in an inability to translate results from animal studies to successful human therapies. Induced pluripotent stem cell (iPSC) technology, in which somatic cells are reprogrammed to pluripotent stem cells, creates an ideal physiologically relevant model as they maintain the genetic identity of the donor. These iPSCs can self-renew indefinitely in vitro and have the capacity to differentiate into any cell type, opening up new discovery and therapeutic opportunities. Despite a plethora of publications indicating the generation and utility of iPSC-derived neurones for disease modelling to date, in comparison only a limited number of studies have described generation of enriched astroglia from patients with early- or late-stage onset of AD. We recently reported that iPSC-astroglia derived from these patients are capable of mimicking a wide variety of deficits in homeostatic molecular cascades, intimately associated with AD, that are routinely observed in vivo. This review examines the opportunities and limitations of this innovative technology in the context of AD modelling and uses for preclinical discovery to improve our success for an efficacious therapeutic outcome.
Collapse
Affiliation(s)
| | - Lisa Mohamet
- StrataStem Ltd., Suite 112, 4a Rylands Street, Warrington, WA1 1EN, UK.
| |
Collapse
|
10
|
Frimat JP, Luttge R. The Need for Physiological Micro-Nanofluidic Systems of the Brain. Front Bioeng Biotechnol 2019; 7:100. [PMID: 31134196 PMCID: PMC6514106 DOI: 10.3389/fbioe.2019.00100] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 04/18/2019] [Indexed: 01/09/2023] Open
Abstract
In this article, we review brain-on-a-chip models and associated underlying technologies. Micro-nanofluidic systems of the brain can utilize the entire spectrum of organoid technology. Notably, there is an urgent clinical need for a physiologically relevant microfluidic platform that can mimic the brain. Brain diseases affect millions of people worldwide, and this number will grow as the size of elderly population increases, thus making brain disease a serious public health problem. Brain disease modeling typically involves the use of in vivo rodent models, which is time consuming, resource intensive, and arguably unethical because many animals are required for a single study. Moreover, rodent models may not accurately predict human diseases, leading to erroneous results, thus rendering animal models poor predictors of human responses to treatment. Various clinical researchers have highlighted this issue, showing that initial physiological descriptions of animal models rarely encompass all the desired human features, including how closely the model captures what is observed in patients. Consequently, such animal models only mimic certain disease aspects, and they are often inadequate for studying how a certain molecule affects various aspects of a disease. Thus, there is a great need for the development of the brain-on-a-chip technology based on which a human brain model can be engineered by assembling cell lines to generate an organ-level model. To produce such a brain-on-a-chip device, selection of appropriate cells lines is critical because brain tissue consists of many different neuronal subtypes, including a plethora of supporting glial cell types. Additionally, cellular network bio-architecture significantly varies throughout different brain regions, forming complex structures and circuitries; this needs to be accounted for in the chip design process. Compartmentalized microenvironments can also be designed within the microphysiological cell culture system to fulfill advanced requirements of a given application. On-chip integration methods have already enabled advances in Parkinson's disease, Alzheimer's disease, and epilepsy modeling, which are discussed herein. In conclusion, for the brain model to be functional, combining engineered microsystems with stem cell (hiPSC) technology is specifically beneficial because hiPSCs can contribute to the complexity of tissue architecture based on their level of differentiation and thereby, biology itself.
Collapse
Affiliation(s)
- Jean-Philippe Frimat
- Neuro-Nanoscale Engineering Group, Microsystems Section & ICMS Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- Department of Neurosurgery, Maastricht University Medical Centre, School for Mental Health and Neuroscience, Eindhoven, Netherlands
| | - Regina Luttge
- Neuro-Nanoscale Engineering Group, Microsystems Section & ICMS Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
11
|
Gutbier S, Spreng AS, Delp J, Schildknecht S, Karreman C, Suciu I, Brunner T, Groettrup M, Leist M. Prevention of neuronal apoptosis by astrocytes through thiol-mediated stress response modulation and accelerated recovery from proteotoxic stress. Cell Death Differ 2018; 25:2101-2117. [PMID: 30390092 PMCID: PMC6261954 DOI: 10.1038/s41418-018-0229-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 09/12/2018] [Accepted: 10/11/2018] [Indexed: 01/04/2023] Open
Abstract
The development of drugs directly interfering with neurodegeneration has proven to be astonishingly difficult. Alternative therapeutic approaches could result from a better understanding of the supportive function of glial cells for stressed neurons. Therefore, here, we investigated the mechanisms involved in the endogenous neuro-defensive activity of astrocytes. A well-established model of postmitotic human dopaminergic neurons (LUHMES cells) was used in the absence ('LUHMES' mono-culture) or presence ('co-culture') of astrocytes. Inhibition of the LUHMES proteasome led to proteotoxic (protein aggregates; ATF-4 induction) and oxidative (GSH-depletion; NRF-2 induction) stress, followed by neuronal apoptosis. The presence of astrocytes attenuated the neuronal stress response, and drastically reduced neurodegeneration. A similar difference between LUHMES mono- and co-cultures was observed, when proteotoxic and oxidative stress was triggered indirectly by inhibitors of mitochondrial function (rotenone, MPP+). Human and murine astrocytes continuously released glutathione (GSH) into the medium, and transfer of glia-conditioned medium was sufficient to rescue LUHMES, unless it was depleted for GSH. Also, direct addition of GSH to LUHMES rescued the neurons from inhibition of the proteasome. Both astrocytes and GSH blunted the neuronal ATF-4 response and similarly upregulated NRF-1/NFE2L1, a transcription factor counter-regulating neuronal proteotoxic stress. Astrocyte co-culture also helped to recover the neurons' ability to degrade aggregated poly-ubiquitinated proteins. Overexpression of NRF-1 attenuated the toxicity of proteasome inhibition, while knockdown increased toxicity. Thus, astrocytic thiol supply increased neuronal resilience to various proteotoxic stressors by simultaneously attenuating cell death-related stress responses, and enhancing the recovery from proteotoxic stress through upregulation of NRF-1.
Collapse
Affiliation(s)
- Simon Gutbier
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
- Research Training Group RTG1331, University of Konstanz, Konstanz, Germany
| | - Anna-Sophie Spreng
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Constance, Germany
| | - Johannes Delp
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
- Research Training Group RTG1331, University of Konstanz, Konstanz, Germany
- Cooperative Doctorate College InViTe, University of Konstanz, Konstanz, Germany
| | - Stefan Schildknecht
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
| | - Christiaan Karreman
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
| | - Ilinca Suciu
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Constance, Germany
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Marcus Groettrup
- Division of Immunology, Department of Biology, University of Konstanz, D-78457, Konstanz, Germany
| | - Marcel Leist
- In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden foundation, University of Konstanz, 78457, Konstanz, Germany.
- CAAT-Europe, University of Konstanz, 78457, Konstanz, Germany.
| |
Collapse
|
12
|
Seidel F. Highlight report: Stem cell-based developmental toxicity tests. Arch Toxicol 2018; 92:3609-3610. [PMID: 30478605 DOI: 10.1007/s00204-018-2357-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 10/27/2022]
Affiliation(s)
- Florian Seidel
- IfADo, Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund, Ardeystr. 67, 44139, Dortmund, Germany.
| |
Collapse
|
13
|
Bal-Price A, Hogberg HT, Crofton KM, Daneshian M, FitzGerald RE, Fritsche E, Heinonen T, Hougaard Bennekou S, Klima S, Piersma AH, Sachana M, Shafer TJ, Terron A, Monnet-Tschudi F, Viviani B, Waldmann T, Westerink RHS, Wilks MF, Witters H, Zurich MG, Leist M. Recommendation on test readiness criteria for new approach methods in toxicology: Exemplified for developmental neurotoxicity. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2018; 35:306-352. [PMID: 29485663 DOI: 10.14573/altex.1712081] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/29/2018] [Indexed: 01/06/2023]
Abstract
Multiple non-animal-based test methods have never been formally validated. In order to use such new approach methods (NAMs) in a regulatory context, criteria to define their readiness are necessary. The field of developmental neurotoxicity (DNT) testing is used to exemplify the application of readiness criteria. The costs and number of untested chemicals are overwhelming for in vivo DNT testing. Thus, there is a need for inexpensive, high-throughput NAMs, to obtain initial information on potential hazards, and to allow prioritization for further testing. A background on the regulatory and scientific status of DNT testing is provided showing different types of test readiness levels, depending on the intended use of data from NAMs. Readiness criteria, compiled during a stakeholder workshop, uniting scientists from academia, industry and regulatory authorities are presented. An important step beyond the listing of criteria, was the suggestion for a preliminary scoring scheme. On this basis a (semi)-quantitative analysis process was assembled on test readiness of 17 NAMs with respect to various uses (e.g. prioritization/screening, risk assessment). The scoring results suggest that several assays are currently at high readiness levels. Therefore, suggestions are made on how DNT NAMs may be assembled into an integrated approach to testing and assessment (IATA). In parallel, the testing state in these assays was compiled for more than 1000 compounds. Finally, a vision is presented on how further NAM development may be guided by knowledge of signaling pathways necessary for brain development, DNT pathophysiology, and relevant adverse outcome pathways (AOP).
Collapse
Affiliation(s)
- Anna Bal-Price
- European Commission, Joint Research Centre (EC JRC), Ispra (VA), Italy
| | - Helena T Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Baltimore, MD, USA
| | - Kevin M Crofton
- National Centre for Computational Toxicology, US EPA, RTP, Washington, NC, USA
| | - Mardas Daneshian
- Center for Alternatives to Animal Testing, CAAT-Europe, University of Konstanz, Konstanz, Germany
| | - Rex E FitzGerald
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine & Heinrich-Heine-University, Düsseldorf, Germany
| | - Tuula Heinonen
- Finnish Centre for Alternative Methods (FICAM), University of Tampere, Tampere, Finland
| | | | - Stefanie Klima
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Aldert H Piersma
- RIVM, National Institute for Public Health and the Environment, Bilthoven, and Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Magdalini Sachana
- Organisation for Economic Co-operation and Development (OECD), Paris, France
| | - Timothy J Shafer
- National Centre for Computational Toxicology, US EPA, RTP, Washington, NC, USA
| | | | - Florianne Monnet-Tschudi
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland.,Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Barbara Viviani
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Tanja Waldmann
- In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Remco H S Westerink
- Neurotoxicology Research Group, Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Martin F Wilks
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland
| | - Hilda Witters
- VITO, Flemish Institute for Technological Research, Unit Environmental Risk and Health, Mol, Belgium
| | - Marie-Gabrielle Zurich
- Swiss Centre for Human Applied Toxicology, SCAHT, University of Basle, Switzerland.,Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Marcel Leist
- Center for Alternatives to Animal Testing, CAAT-Europe, University of Konstanz, Konstanz, Germany.,In vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
14
|
Chovancova P, Merk V, Marx A, Leist M, Kranaster R. Reverse-transcription quantitative PCR directly from cells without RNA extraction and without isothermal reverse-transcription: a 'zero-step' RT-qPCR protocol. Biol Methods Protoc 2017; 2:bpx008. [PMID: 32002469 PMCID: PMC6977950 DOI: 10.1093/biomethods/bpx008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/05/2017] [Accepted: 04/25/2017] [Indexed: 02/06/2023] Open
Abstract
We describe an ultra-rapid and sensitive method to quantify gene expression levels in
cultured cells. The procedure is based on reverse-transcription quantitative PCR (RT-qPCR)
directly from cells, without RNA extraction and without an isothermal
reverse-transcription step. Human neurons (Lund human mesencephalic cells) were lysed at
different stages of differentiation, and the lysates were used directly as template for
the combined RT-qPCR reaction. We detected a down-regulation of a proliferation marker and
an up-regulation of neuronal dopaminergic genes expression. We were able to detect the
reference gene target from as few as a single cell, demonstrating the application of the
method for efficient amplification from small cell numbers. The data were fully in line
with those obtained by the standard two-step RT-qPCR from the extracted total RNA. Our
‘zero-step’ RT-qPCR method proved to be simple and reliable with a total time from cell
lysis to the end of the qPCR as short as 1.5 h. It is therefore particularly suitable for
RT-qPCRs where large numbers of samples must be handled, or where data are required within
short time.
Collapse
Affiliation(s)
- Petra Chovancova
- In vitro Toxicology and Biomedicine, University of Konstanz, D-78457 Konstanz, Germany.,Konstanz Research School Chemical Biology, University of Konstanz, D-78457 Konstanz, Germany
| | - Verena Merk
- myPOLS Biotec GmbH, D-78457 Konstanz, Germany
| | - Andreas Marx
- myPOLS Biotec GmbH, D-78457 Konstanz, Germany.,Department of Chemistry, University of Konstanz, D-78457 Konstanz, Germany
| | - Marcel Leist
- In vitro Toxicology and Biomedicine, University of Konstanz, D-78457 Konstanz, Germany
| | | |
Collapse
|
15
|
Effects of the duration of hyperlipidemia on cerebral lipids, vessels and neurons in rats. Lipids Health Dis 2017; 16:26. [PMID: 28143622 PMCID: PMC5282812 DOI: 10.1186/s12944-016-0401-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 12/27/2016] [Indexed: 01/09/2023] Open
Abstract
Background The present study was designed to investigate the effects of hyperlipidemia on the cerebral lipids, vessels and neurons of rats, and to provide experimental evidence for subsequent intervention. Method One hundred adult SD rats, half of which were male and half of which were female, were randomly divided into five groups on the basis of serum total cholesterol (TC) levels. Four groups were fed a hypercholesterolemic diet (rat chow supplemented with 4% cholesterol, 1% cholic acid and 0.5% thiouracil – this is also called a CCT diet) for periods of 1 week, 2 weeks, 3 weeks and 4 weeks, respectively. A control group was included. The levels of serum lipids, cerebral lipids, free fatty acids (FFA), interleukin-6 (IL-6), interleukin-1 (IL-1), tumor necrosis factor alpha (TNF-α), vascular endothelial growth factor (VEGF), oxidized low density lipoprotein (ox-LDL), A-beta precursor proteins (APP), amyloid beta (Aβ), glial fibrillary acidic protein (GFAP) and tight junction protein Claudin-5 were measured after the experiment. The pathologic changes and apoptosis of the rat brains were evaluated. Results Compared with the control group, after 1 week of a CCT diet, the levels of serum total cholesterol (TC), triglycerides (TG), low density lipoprotein cholesterol (LDL-C) and brain triglycerides had increased by 2.40, 1.29 and 1.75 and 0.3 times, respectively. The serum high density lipoprotein cholesterol (HDL-C) had decreased by 0.74 times (P < 0.05) and the expression of IL-1, TNF-α and GFAP in the brains had increased (P < 0.05). In the second week, the expression of FFA and APP in the brains, and the amount of apoptotic neurons, had increased (P < 0.05). In the third week, the levels of VEGF, Ox-LDL and Aβ had increased, and the expression of Claudin-5 had decreased in the brains (P < 0.05). In the fourth week, the levels of TC, LDL-C and the amount of apoptotic neurons had increased (P < 0.05). The correlation analysis showed a positive correlation among FFA, TNF-α, VEGF, ox-LDL, Aβ, GFAP and neuronal apoptosis in the rat brains, and they all were negatively correlated with Claudin-5 (P < 0.05). Conclusion Hyperlipidemia may activate astrocytes by means of high levels of TG that will have direct toxic effects on the cerebral vessels and neurons by causing the secretion of TNF-α and IL-1 in the brains of rats. In the metabolic procession, brain tissue was shown to generate FFA that aggravated the biosynthesis of ox-LDL. With the extension of the duration of hyperlipidemia, high levels of cerebral TC and LDL-C were shown to aggravate the deposition of Aβ, induce the secretion of VEGF, reduce the expression of tight junction protein Claudin-5 and change the permeability of blood–brain barriers to factors that could damage cerebral vessels and neurons.
Collapse
|
16
|
Witt B, Bornhorst J, Mitze H, Ebert F, Meyer S, Francesconi KA, Schwerdtle T. Arsenolipids exert less toxicity in a human neuron astrocyte co-culture as compared to the respective monocultures. Metallomics 2017; 9:442-446. [DOI: 10.1039/c7mt00036g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
17
|
Chandrasekaran A, Avci HX, Leist M, Kobolák J, Dinnyés A. Astrocyte Differentiation of Human Pluripotent Stem Cells: New Tools for Neurological Disorder Research. Front Cell Neurosci 2016; 10:215. [PMID: 27725795 PMCID: PMC5035736 DOI: 10.3389/fncel.2016.00215] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/30/2016] [Indexed: 12/22/2022] Open
Abstract
Astrocytes have a central role in brain development and function, and so have gained increasing attention over the past two decades. Consequently, our knowledge about their origin, differentiation and function has increased significantly, with new research showing that astrocytes cultured alone or co-cultured with neurons have the potential to improve our understanding of various central nervous system diseases, such as amyotrophic lateral sclerosis, Alzheimer’s disease, or Alexander disease. The generation of astrocytes derived from pluripotent stem cells (PSCs) opens up a new area for studying neurologic diseases in vitro; these models could be exploited to identify and validate potential drugs by detecting adverse effects in the early stages of drug development. However, as it is now known that a range of astrocyte populations exist in the brain, it will be important in vitro to develop standardized protocols for the in vitro generation of astrocyte subsets with defined maturity status and phenotypic properties. This will then open new possibilities for co-cultures with neurons and the generation of neural organoids for research purposes. The aim of this review article is to compare and summarize the currently available protocols and their strategies to generate human astrocytes from PSCs. Furthermore, we discuss the potential role of human-induced PSCs derived astrocytes in disease modeling.
Collapse
Affiliation(s)
| | - Hasan X Avci
- BioTalentum LtdGödöllő, Hungary; Department of Medical Chemistry, University of SzegedSzeged, Hungary
| | - Marcel Leist
- Dorenkamp-Zbinden Chair, Faculty of Mathematics and Sciences, University of Konstanz Konstanz, Germany
| | | | - Andras Dinnyés
- BioTalentum LtdGödöllő, Hungary; Molecular Animal Biotechnology Laboratory, Szent Istvan UniversityGödöllő, Hungary
| |
Collapse
|
18
|
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: 9.0] [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
|
19
|
Aschner M, Ceccatelli S, Daneshian M, Fritsche E, Hasiwa N, Hartung T, Hogberg HT, Leist M, Li A, Mundi WR, Padilla S, Piersma AH, Bal-Price A, Seiler A, Westerink RH, Zimmer B, Lein PJ. Reference compounds for alternative test methods to indicate developmental neurotoxicity (DNT) potential of chemicals: example lists and criteria for their selection and use. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2016; 34:49-74. [PMID: 27452664 PMCID: PMC5250586 DOI: 10.14573/altex.1604201] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/09/2016] [Indexed: 11/23/2022]
Abstract
There is a paucity of information concerning the developmental neurotoxicity (DNT) hazard posed by industrial and environmental chemicals. New testing approaches will most likely be based on batteries of alternative and complementary (non-animal) tests. As DNT is assumed to result from the modulation of fundamental neurodevelopmental processes (such as neuronal differentiation, precursor cell migration or neuronal network formation) by chemicals, the first generation of alternative DNT tests target these processes. The advantage of such types of assays is that they capture toxicants with multiple targets and modes-of-action. Moreover, the processes modelled by the assays can be linked to toxicity endophenotypes, i.e. alterations in neural connectivity that form the basis for neurofunctional deficits in man. The authors of this review convened in a workshop to define criteria for the selection of positive/negative controls, to prepare recommendations on their use, and to initiate the setup of a directory of reference chemicals. For initial technical optimization of tests, a set of >50 endpoint-specific control compounds was identified. For further test development, an additional “test” set of 33 chemicals considered to act directly as bona fide DNT toxicants is proposed, and each chemical is annotated to the extent it fulfills these criteria. A tabular compilation of the original literature used to select the test set chemicals provides information on statistical procedures, and toxic/non-toxic doses (both for pups and dams). Suggestions are provided on how to use the >100 compounds (including negative controls) compiled here to address specificity, adversity and use of alternative test systems.
Collapse
Affiliation(s)
| | | | - Mardas Daneshian
- Center for Alternatives to Animal Testing-Europe (CAAT-Europe), University of Konstanz, Germany
| | - Ellen Fritsche
- Leibniz Research Institute for Environmental Medicine (IUF), Düsseldorf, Germany
| | - Nina Hasiwa
- Center for Alternatives to Animal Testing-Europe (CAAT-Europe), University of Konstanz, Germany
| | - Thomas Hartung
- Center for Alternatives to Animal Testing-Europe (CAAT-Europe), University of Konstanz, Germany.,Center for Alternatives to Animal Testing (CAAT), The Johns Hopkins University, Baltimore, MD, USA
| | - Helena T Hogberg
- Center for Alternatives to Animal Testing (CAAT), The Johns Hopkins University, Baltimore, MD, USA
| | - Marcel Leist
- Center for Alternatives to Animal Testing-Europe (CAAT-Europe), University of Konstanz, Germany.,In vitro Toxicology and Biomedicine, Dept inaugurated by the Doerenkamp-Zbinden Foundation at the University of Konstanz, Konstanz, Germany.,Konstanz Research School Chemical Biology (KoRS-CB), Konstanz University
| | - Abby Li
- Exponent Inc.,San Francisco, USA
| | - William R Mundi
- United States Environmental Protection Agency (USEPA), NHEERL, Research Triangle Park, NC, USA
| | - Stephanie Padilla
- United States Environmental Protection Agency (USEPA), NHEERL, Research Triangle Park, NC, USA
| | - Aldert H Piersma
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.,Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Anna Bal-Price
- European Commission Joint Research Centre, Institute for Health and Consumer Protection, Ispra, Italy
| | - Andrea Seiler
- Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Remco H Westerink
- Neurotoxicology Research Group, Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, The Netherlands
| | | | - Pamela J Lein
- Center for Research on Occupational and Environmental Toxicology, Oregon Health & Science University, Portland, USA.,Department of Molecular Biosciences, University of California, Davis, USA
| |
Collapse
|
20
|
Switching from astrocytic neuroprotection to neurodegeneration by cytokine stimulation. Arch Toxicol 2016; 91:231-246. [PMID: 27052459 DOI: 10.1007/s00204-016-1702-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/21/2016] [Indexed: 12/12/2022]
Abstract
Astrocytes, the largest cell population in the human brain, are powerful inflammatory effectors. Several studies have examined the interaction of activated astrocytes with neurons, but little is known yet about human neurotoxicity under such situations and about strategies of neuronal rescue. To address this question, immortalized murine astrocytes (IMA) were combined with human LUHMES neurons and stimulated with an inflammatory (TNF, IL-1) cytokine mix (CM). Neurotoxicity was studied both in co-cultures and in monocultures after transfer of conditioned medium from activated IMA. Interventions with >20 drugs were used to profile the model system. Control IMA supported neurons and protected them from neurotoxicants. Inflammatory activation reduced this protection, and prolonged exposure of co-cultures to CM triggered neurotoxicity. Neither the added cytokines nor the release of NO from astrocytes were involved in this neurodegeneration. The neurotoxicity-mediating effect of IMA was faithfully reproduced by human astrocytes. Moreover, glia-dependent toxicity was also observed, when IMA cultures were stimulated with CM, and the culture medium was transferred to neurons. Such neurotoxicity was prevented when astrocytes were treated by p38 kinase inhibitors or dexamethasone, whereas such compounds had no effect when added to neurons. Conversely, treatment of neurons with five different drugs, including resveratrol and CEP1347, prevented toxicity of astrocyte supernatants. Thus, the sequential IMA-LUHMES neuroinflammation model is suitable for separate profiling of both glial-directed and directly neuroprotective strategies. Moreover, direct evaluation in co-cultures of the same cells allows for testing of therapeutic effectiveness in more complex settings, in which astrocytes affect pharmacological properties of neurons.
Collapse
|
21
|
Kleiderman S, Sá JV, Teixeira AP, Brito C, Gutbier S, Evje LG, Hadera MG, Glaab E, Henry M, Sachinidis A, Alves PM, Sonnewald U, Leist M. Functional and phenotypic differences of pure populations of stem cell-derived astrocytes and neuronal precursor cells. Glia 2015; 64:695-715. [DOI: 10.1002/glia.22954] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 11/19/2015] [Accepted: 11/23/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Susanne Kleiderman
- The Doerenkamp-Zbinden Chair of in-Vitro Toxicology and Biomedicine/Alternatives to Animal Experimentation; University of Konstanz; Konstanz Germany
| | - João V. Sá
- Instituto de Tecnologia Química e Biológica António Xavier; Universidade Nova de Lisboa; Av. da República 2780-157 Oeiras Portugal
- IBET; Instituto de Biologia Experimental e Tecnológica; Apartado 12 2780-901 Oeiras Portugal
| | - Ana P. Teixeira
- Instituto de Tecnologia Química e Biológica António Xavier; Universidade Nova de Lisboa; Av. da República 2780-157 Oeiras Portugal
- IBET; Instituto de Biologia Experimental e Tecnológica; Apartado 12 2780-901 Oeiras Portugal
| | - Catarina Brito
- Instituto de Tecnologia Química e Biológica António Xavier; Universidade Nova de Lisboa; Av. da República 2780-157 Oeiras Portugal
- IBET; Instituto de Biologia Experimental e Tecnológica; Apartado 12 2780-901 Oeiras Portugal
| | - Simon Gutbier
- The Doerenkamp-Zbinden Chair of in-Vitro Toxicology and Biomedicine/Alternatives to Animal Experimentation; University of Konstanz; Konstanz Germany
| | - Lars G. Evje
- Department of Earth Science, University of Bergen; Allégaten 41 5007 Bergen Norway
| | - Mussie G. Hadera
- Department of Pharmacy; College of Health Sciences; Mekelle University, Tigray Ethiopia
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg; Belvaux L-4366 Luxembourg
| | - Margit Henry
- Institute of Neurophysiology and Center for Molecular Medicine, Cologne (CMMC), University of Cologne; Cologne Germany
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine, Cologne (CMMC), University of Cologne; Cologne Germany
| | - Paula M. Alves
- Instituto de Tecnologia Química e Biológica António Xavier; Universidade Nova de Lisboa; Av. da República 2780-157 Oeiras Portugal
- IBET; Instituto de Biologia Experimental e Tecnológica; Apartado 12 2780-901 Oeiras Portugal
| | - Ursula Sonnewald
- Department of Drug Design and Pharmacology; Faculty of Health and Medical Sciences; Copenhagen Denmark
- Department of Neuroscience; Norwegian University of Science and Technology; Faculty of Medicine; Trondheim Norway
| | - Marcel Leist
- The Doerenkamp-Zbinden Chair of in-Vitro Toxicology and Biomedicine/Alternatives to Animal Experimentation; University of Konstanz; Konstanz Germany
| |
Collapse
|
22
|
Chen C, Chan A, Wen H, Chung SH, Deng W, Jiang P. Stem and Progenitor Cell-Derived Astroglia Therapies for Neurological Diseases. Trends Mol Med 2015; 21:715-729. [PMID: 26443123 DOI: 10.1016/j.molmed.2015.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/04/2015] [Accepted: 09/08/2015] [Indexed: 02/07/2023]
Abstract
Astroglia are a major cellular constituent of the central nervous system (CNS) and play crucial roles in brain development, function, and integrity. Increasing evidence demonstrates that astroglia dysfunction occurs in a variety of neurological disorders ranging from CNS injuries to genetic diseases and chronic degenerative conditions. These new insights herald the concept that transplantation of astroglia could be of therapeutic value in treating the injured or diseased CNS. Recent technological advances in the generation of human astroglia from stem and progenitor cells have been prominent. We propose that a better understanding of the suitability of astroglial cells in transplantation as well as of their therapeutic effects in animal models may lead to the establishment of astroglia-based therapies to treat neurological diseases.
Collapse
Affiliation(s)
- Chen Chen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA
| | - Albert Chan
- Department of Pediatrics, University of California, Davis, CA, USA
| | - Han Wen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA
| | | | - Wenbin Deng
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA.
| | - Peng Jiang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA; Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, NE, USA.
| |
Collapse
|
23
|
Smirnova L, Hogberg HT, Leist M, Hartung T. Developmental neurotoxicity - challenges in the 21st century and in vitro opportunities. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2015; 31:129-56. [PMID: 24687333 DOI: 10.14573/altex.1403271] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 03/28/2014] [Indexed: 11/23/2022]
Abstract
In recent years neurodevelopmental problems in children have increased at a rate that suggests lifestyle factors and chemical exposures as likely contributors. When environmental chemicals contribute to neurodevelopmental disorders developmental neurotoxicity (DNT) becomes an enormous concern. But how can it be tackled? Current animal test- based guidelines are prohibitively expensive, at $ 1.4 million per substance, while their predictivity for human health effects may be limited, and mechanistic data that would help species extrapolation are not available. A broader screening for substances of concern requires a reliable testing strategy, applicable to larger numbers of substances, and sufficiently predictive to warrant further testing. This review discusses the evidence for possible contributions of environmental chemicals to DNT, limitations of the current test paradigm, emerging concepts and technologies pertinent to in vitro DNT testing and assay evaluation, as well as the prospect of a paradigm shift based on 21st century technologies.
Collapse
Affiliation(s)
- Lena Smirnova
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, USA
| | | | | | | |
Collapse
|
24
|
Using Pluripotent Stem Cells and Their Progeny as an In VitroModel to Assess (Developmental) Neurotoxicity. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2014. [DOI: 10.1002/9783527674183.ch13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
25
|
Alépée N, Bahinski A, Daneshian M, De Wever B, Fritsche E, Goldberg A, Hansmann J, Hartung T, Haycock J, Hogberg H, Hoelting L, Kelm JM, Kadereit S, McVey E, Landsiedel R, Leist M, Lübberstedt M, Noor F, Pellevoisin C, Petersohn D, Pfannenbecker U, Reisinger K, Ramirez T, Rothen-Rutishauser B, Schäfer-Korting M, Zeilinger K, Zurich MG. State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2014. [PMID: 25027500 DOI: 10.14573/altex1406111] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Integrated approaches using different in vitro methods in combination with bioinformatics can (i) increase the success rate and speed of drug development; (ii) improve the accuracy of toxicological risk assessment; and (iii) increase our understanding of disease. Three-dimensional (3D) cell culture models are important building blocks of this strategy which has emerged during the last years. The majority of these models are organotypic, i.e., they aim to reproduce major functions of an organ or organ system. This implies in many cases that more than one cell type forms the 3D structure, and often matrix elements play an important role. This review summarizes the state of the art concerning commonalities of the different models. For instance, the theory of mass transport/metabolite exchange in 3D systems and the special analytical requirements for test endpoints in organotypic cultures are discussed in detail. In the next part, 3D model systems for selected organs--liver, lung, skin, brain--are presented and characterized in dedicated chapters. Also, 3D approaches to the modeling of tumors are presented and discussed. All chapters give a historical background, illustrate the large variety of approaches, and highlight up- and downsides as well as specific requirements. Moreover, they refer to the application in disease modeling, drug discovery and safety assessment. Finally, consensus recommendations indicate a roadmap for the successful implementation of 3D models in routine screening. It is expected that the use of such models will accelerate progress by reducing error rates and wrong predictions from compound testing.
Collapse
|
26
|
Zimmer B, Pallocca G, Dreser N, Foerster S, Waldmann T, Westerhout J, Julien S, Krause KH, van Thriel C, Hengstler JG, Sachinidis A, Bosgra S, Leist M. Profiling of drugs and environmental chemicals for functional impairment of neural crest migration in a novel stem cell-based test battery. Arch Toxicol 2014; 88:1109-26. [PMID: 24691702 PMCID: PMC3996367 DOI: 10.1007/s00204-014-1231-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 03/18/2014] [Indexed: 11/28/2022]
Abstract
Developmental toxicity in vitro assays have hitherto been established as stand-alone systems, based on a limited number of toxicants. Within the embryonic stem cell-based novel alternative tests project, we developed a test battery framework that allows inclusion of any developmental toxicity assay and that explores the responses of such test systems to a wide range of drug-like compounds. We selected 28 compounds, including several biologics (e.g., erythropoietin), classical pharmaceuticals (e.g., roflumilast) and also six environmental toxicants. The chemical, toxicological and clinical data of this screen library were compiled. In order to determine a non-cytotoxic concentration range, cytotoxicity data were obtained for all compounds from HEK293 cells and from murine embryonic stem cells. Moreover, an estimate of relevant exposures was provided by literature data mining. To evaluate feasibility of the suggested test framework, we selected a well-characterized assay that evaluates ‘migration inhibition of neural crest cells.’ Screening at the highest non-cytotoxic concentration resulted in 11 hits (e.g., geldanamycin, abiraterone, gefitinib, chlorpromazine, cyproconazole, arsenite). These were confirmed in concentration–response studies. Subsequent pharmacokinetic modeling indicated that triadimefon exerted its effects at concentrations relevant to the in vivo situation, and also interferon-β and polybrominated diphenyl ether showed effects within the same order of magnitude of concentrations that may be reached in humans. In conclusion, the test battery framework can identify compounds that disturb processes relevant for human development and therefore may represent developmental toxicants. The open structure of the strategy allows rich information to be generated on both the underlying library, and on any contributing assay.
Collapse
Affiliation(s)
- B Zimmer
- Center for Stem Cell Biology, Sloan-Kettering Institute for Cancer Research, New York City, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Waldmann T, Rempel E, Balmer NV, König A, Kolde R, Gaspar JA, Henry M, Hescheler J, Sachinidis A, Rahnenführer J, Hengstler JG, Leist M. Design principles of concentration-dependent transcriptome deviations in drug-exposed differentiating stem cells. Chem Res Toxicol 2014; 27:408-20. [PMID: 24383497 PMCID: PMC3958134 DOI: 10.1021/tx400402j] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
Information on design principles
governing transcriptome changes
upon transition from safe to hazardous drug concentrations or from
tolerated to cytotoxic drug levels are important for the application
of toxicogenomics data in developmental toxicology. Here, we tested
the effect of eight concentrations of valproic acid (VPA; 25–1000
μM) in an assay that recapitulates the development of human
embryonic stem cells to neuroectoderm. Cells were exposed to the drug
during the entire differentiation process, and the number of differentially
regulated genes increased continuously over the concentration range
from zero to about 3000. We identified overrepresented transcription
factor binding sites (TFBS) as well as superordinate cell biological
processes, and we developed a gene ontology (GO) activation profiler,
as well as a two-dimensional teratogenicity index. Analysis of the
transcriptome data set by the above biostatistical and systems biology
approaches yielded the following insights: (i) tolerated (≤25
μM), deregulated/teratogenic (150–550 μM), and
cytotoxic (≥800 μM) concentrations could be differentiated.
(ii) Biological signatures related to the mode of action of VPA, such
as protein acetylation, developmental changes, and cell migration,
emerged from the teratogenic concentrations range. (iii) Cytotoxicity
was not accompanied by signatures of newly emerging canonical cell
death/stress indicators, but by catabolism and decreased expression
of cell cycle associated genes. (iv) Most, but not all of the GO groups
and TFBS seen at the highest concentrations were already overrepresented
at 350–450 μM. (v) The teratogenicity index reflected
this behavior, and thus differed strongly from cytotoxicity. Our findings
suggest the use of the highest noncytotoxic drug concentration for
gene array toxicogenomics studies, as higher concentrations possibly
yield wrong information on the mode of action, and lower drug levels
result in decreased gene expression changes and thus a reduced power
of the study.
Collapse
Affiliation(s)
- Tanja Waldmann
- Doerenkamp-Zbinden Chair for in Vitro Toxicology and Biomedicine, University of Konstanz , 78457 Konstanz, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Alépée N, Bahinski A, Daneshian M, De Wever B, Fritsche E, Goldberg A, Hansmann J, Hartung T, Haycock J, Hogberg HT, Hoelting L, Kelm JM, Kadereit S, McVey E, Landsiedel R, Leist M, Lübberstedt M, Noor F, Pellevoisin C, Petersohn D, Pfannenbecker U, Reisinger K, Ramirez T, Rothen-Rutishauser B, Schäfer-Korting M, Zeilinger K, Zurich MG. State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX 2014; 31:441-77. [PMID: 25027500 PMCID: PMC4783151 DOI: 10.14573/altex.1406111] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 06/30/2014] [Indexed: 02/02/2023]
Abstract
Integrated approaches using different in vitro methods in combination with bioinformatics can (i) increase the success rate and speed of drug development; (ii) improve the accuracy of toxicological risk assessment; and (iii) increase our understanding of disease. Three-dimensional (3D) cell culture models are important building blocks of this strategy which has emerged during the last years. The majority of these models are organotypic, i.e., they aim to reproduce major functions of an organ or organ system. This implies in many cases that more than one cell type forms the 3D structure, and often matrix elements play an important role. This review summarizes the state of the art concerning commonalities of the different models. For instance, the theory of mass transport/metabolite exchange in 3D systems and the special analytical requirements for test endpoints in organotypic cultures are discussed in detail. In the next part, 3D model systems for selected organs--liver, lung, skin, brain--are presented and characterized in dedicated chapters. Also, 3D approaches to the modeling of tumors are presented and discussed. All chapters give a historical background, illustrate the large variety of approaches, and highlight up- and downsides as well as specific requirements. Moreover, they refer to the application in disease modeling, drug discovery and safety assessment. Finally, consensus recommendations indicate a roadmap for the successful implementation of 3D models in routine screening. It is expected that the use of such models will accelerate progress by reducing error rates and wrong predictions from compound testing.
Collapse
Affiliation(s)
| | - Anthony Bahinski
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, USA
| | - Mardas Daneshian
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany
| | | | - Ellen Fritsche
- Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Alan Goldberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - Jan Hansmann
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Thomas Hartung
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany,Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - John Haycock
- Department of Materials Science of Engineering, University of Sheffield, Sheffield, UK
| | - Helena T. Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - Lisa Hoelting
- Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | | | - Suzanne Kadereit
- Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Emily McVey
- Board for the Authorization of Plant Protection Products and Biocides, Wageningen, The Netherlands
| | | | - Marcel Leist
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany,Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Marc Lübberstedt
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Fozia Noor
- Biochemical Engineering, Saarland University, Saarbruecken, Germany
| | | | | | | | | | - Tzutzuy Ramirez
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
| | | | - Monika Schäfer-Korting
- Institute for Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany
| | - Katrin Zeilinger
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Marie-Gabriele Zurich
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland,Swiss Center for Applied Human Toxicology (SCAHT), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
29
|
Human stem cell-derived spinal cord astrocytes with defined mature or reactive phenotypes. Cell Rep 2013; 4:1035-1048. [PMID: 23994478 DOI: 10.1016/j.celrep.2013.06.021] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 05/15/2013] [Accepted: 06/18/2013] [Indexed: 12/23/2022] Open
Abstract
Differentiation of astrocytes from human stem cells has significant potential for analysis of their role in normal brain function and disease, but existing protocols generate only immature astrocytes. Using early neuralization, we generated spinal cord astrocytes from mouse or human embryonic or induced pluripotent stem cells with high efficiency. Remarkably, short exposure to fibroblast growth factor 1 (FGF1) or FGF2 was sufficient to direct these astrocytes selectively toward a mature quiescent phenotype, as judged by both marker expression and functional analysis. In contrast, tumor necrosis factor alpha and interleukin-1β, but not FGFs, induced multiple elements of a reactive inflammatory phenotype but did not affect maturation. These phenotypically defined, scalable populations of spinal cord astrocytes will be important both for studying normal astrocyte function and for modeling human pathological processes in vitro.
Collapse
|
30
|
The unfolded protein response to endoplasmic reticulum stress in cultured astrocytes and rat brain during experimental diabetes. Neurochem Int 2013; 62:784-95. [PMID: 23411409 DOI: 10.1016/j.neuint.2013.02.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 01/29/2013] [Accepted: 02/04/2013] [Indexed: 12/21/2022]
Abstract
Oxidative-nitrosative stress and inflammatory responses are associated with endoplasmic reticulum (ER) stress in diabetic retinopathy, raising the possibility that disturbances in ER protein processing may contribute to CNS dysfunction in diabetics. Upregulation of the unfolded protein response (UPR) is a homeostatic response to accumulation of abnormal proteins in the ER, and the present study tested the hypothesis that the UPR is upregulated in two models for diabetes, cultured astrocytes grown in 25mmol/L glucose for up to 4weeks and brain of streptozotocin (STZ)-treated rats with diabetes for 1-7months. Markers associated with translational blockade (phospho-eIF2α and apoptosis (CHOP), inflammatory response (inducible nitric oxide synthase, iNOS), and nitrosative stress (nuclear translocation of glyceraldehyde-3-phosphate dehydrogenase, GAPDH) were not detected in either model. Nrf2 was present in nuclei of low- and high-glucose cultures, consistent with oxidative stress. Astrocytic ATF4 expression was not altered by culture glucose concentration, whereas phospho-IRE and ATF6 levels were higher in low- compared with high-glucose cultures. The glucose-regulated chaperones, GRP78 and GRP94, were also expressed at higher levels in low- than high-glucose cultures, probably due to recurrent glucose depletion between feeding cycles. In STZ-rat cerebral cortex, ATF4 level was transiently reduced at 4months, and p-IRE levels were transiently elevated at 3months. However, GRP78 and GRP94 expression was not upregulated, and iNOS, amyloid-β, and nuclear accumulation of GAPDH were not evident in STZ-diabetic brain. High-glucose cultured astrocytes and STZ-diabetic brain are relatively resistant to diabetes-induced ER stress, in sharp contrast with cultured retinal Müller cells and diabetic rodent retina.
Collapse
|
31
|
Krug AK, Kolde R, Gaspar JA, Rempel E, Balmer NV, Meganathan K, Vojnits K, Baquié M, Waldmann T, Ensenat-Waser R, Jagtap S, Evans RM, Julien S, Peterson H, Zagoura D, Kadereit S, Gerhard D, Sotiriadou I, Heke M, Natarajan K, Henry M, Winkler J, Marchan R, Stoppini L, Bosgra S, Westerhout J, Verwei M, Vilo J, Kortenkamp A, Hescheler J, Hothorn L, Bremer S, van Thriel C, Krause KH, Hengstler JG, Rahnenführer J, Leist M, Sachinidis A. Human embryonic stem cell-derived test systems for developmental neurotoxicity: a transcriptomics approach. Arch Toxicol 2012. [PMID: 23179753 PMCID: PMC3535399 DOI: 10.1007/s00204-012-0967-3] [Citation(s) in RCA: 179] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Developmental neurotoxicity (DNT) and many forms of reproductive toxicity (RT) often manifest themselves in functional deficits that are not necessarily based on cell death, but rather on minor changes relating to cell differentiation or communication. The fields of DNT/RT would greatly benefit from in vitro tests that allow the identification of toxicant-induced changes of the cellular proteostasis, or of its underlying transcriptome network. Therefore, the ‘human embryonic stem cell (hESC)-derived novel alternative test systems (ESNATS)’ European commission research project established RT tests based on defined differentiation protocols of hESC and their progeny. Valproic acid (VPA) and methylmercury (MeHg) were used as positive control compounds to address the following fundamental questions: (1) Does transcriptome analysis allow discrimination of the two compounds? (2) How does analysis of enriched transcription factor binding sites (TFBS) and of individual probe sets (PS) distinguish between test systems? (3) Can batch effects be controlled? (4) How many DNA microarrays are needed? (5) Is the highest non-cytotoxic concentration optimal and relevant for the study of transcriptome changes? VPA triggered vast transcriptional changes, whereas MeHg altered fewer transcripts. To attenuate batch effects, analysis has been focused on the 500 PS with highest variability. The test systems differed significantly in their responses (<20 % overlap). Moreover, within one test system, little overlap between the PS changed by the two compounds has been observed. However, using TFBS enrichment, a relatively large ‘common response’ to VPA and MeHg could be distinguished from ‘compound-specific’ responses. In conclusion, the ESNATS assay battery allows classification of human DNT/RT toxicants on the basis of their transcriptome profiles.
Collapse
Affiliation(s)
- Anne K. Krug
- Department of Biology, University of Konstanz (UKN), 78457 Constance, Germany
| | - Raivo Kolde
- OÜ Quretec (Qure), Limited Liability Company, 51003 Tartu, Estonia
- Institute of Computer Science, University of Tartu, 50409 Tartu, Estonia
| | - John A. Gaspar
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne (UKK), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Eugen Rempel
- Department of Statistics, TU Dortmund University , 44221 Dortmund, Germany
| | - Nina V. Balmer
- Department of Biology, University of Konstanz (UKN), 78457 Constance, Germany
| | - Kesavan Meganathan
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne (UKK), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Kinga Vojnits
- Commission of the European Communities (JRC) Joint Research Centre, 1049 Brussels, Belgium
| | - Mathurin Baquié
- Department of Pathology and Immunology, Geneva Medical Faculty, University of Geneva (UNIGE), 1211 Geneva 4, Switzerland
| | - Tanja Waldmann
- Department of Biology, University of Konstanz (UKN), 78457 Constance, Germany
| | - Roberto Ensenat-Waser
- Commission of the European Communities (JRC) Joint Research Centre, 1049 Brussels, Belgium
| | - Smita Jagtap
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne (UKK), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | | | - Stephanie Julien
- Department of Pathology and Immunology, Geneva Medical Faculty, University of Geneva (UNIGE), 1211 Geneva 4, Switzerland
| | - Hedi Peterson
- Department of Pathology and Immunology, Geneva Medical Faculty, University of Geneva (UNIGE), 1211 Geneva 4, Switzerland
| | - Dimitra Zagoura
- Commission of the European Communities (JRC) Joint Research Centre, 1049 Brussels, Belgium
| | - Suzanne Kadereit
- Department of Biology, University of Konstanz (UKN), 78457 Constance, Germany
| | - Daniel Gerhard
- Gottfried Wilhelm Leibniz University (LUH), Institute for Biostatistics, 30167 Hannover, Germany
| | - Isaia Sotiriadou
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne (UKK), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Michael Heke
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne (UKK), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Karthick Natarajan
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne (UKK), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Margit Henry
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne (UKK), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Johannes Winkler
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne (UKK), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Rosemarie Marchan
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, 44139 Dortmund, Germany
| | - Luc Stoppini
- Department of Pathology and Immunology, Geneva Medical Faculty, University of Geneva (UNIGE), 1211 Geneva 4, Switzerland
| | - Sieto Bosgra
- Nederlandse Organisatie voor Toegepast Natuurwetenschappelijk Onderzoek (TNO), 2628 VK Delft, The Netherlands
| | - Joost Westerhout
- Nederlandse Organisatie voor Toegepast Natuurwetenschappelijk Onderzoek (TNO), 2628 VK Delft, The Netherlands
| | - Miriam Verwei
- Nederlandse Organisatie voor Toegepast Natuurwetenschappelijk Onderzoek (TNO), 2628 VK Delft, The Netherlands
| | - Jaak Vilo
- OÜ Quretec (Qure), Limited Liability Company, 51003 Tartu, Estonia
- Institute of Computer Science, University of Tartu, 50409 Tartu, Estonia
| | | | - Jürgen Hescheler
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne (UKK), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Ludwig Hothorn
- Gottfried Wilhelm Leibniz University (LUH), Institute for Biostatistics, 30167 Hannover, Germany
| | - Susanne Bremer
- Commission of the European Communities (JRC) Joint Research Centre, 1049 Brussels, Belgium
| | - Christoph van Thriel
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, 44139 Dortmund, Germany
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Geneva Medical Faculty, University of Geneva (UNIGE), 1211 Geneva 4, Switzerland
| | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, 44139 Dortmund, Germany
| | - Jörg Rahnenführer
- Department of Statistics, TU Dortmund University , 44221 Dortmund, Germany
| | - Marcel Leist
- Department of Biology, University of Konstanz (UKN), 78457 Constance, Germany
| | - Agapios Sachinidis
- Center of Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne (UKK), Robert-Koch-Str. 39, 50931 Cologne, Germany
| |
Collapse
|